Magnetic Resonance of Myelination and Myelin Disorders 3rd Ed (LG PDF) - M. Van Der Knaap, J. Valk (Springer, 2005) WW

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 1097

M.S. van der Knaap, J.

Valk
Magnetic Resonance of Myelination and Myelin Disorders
Third Edition
Marjo S. van der Knaap
Jaap Valk

Magnetic Resonance
of Myelination
and Myelin Disorders
Third Edition

With 647 Figures in 3873 parts

With contributions by:


F. Barkhof R. van den Berg
V. Gieselmann J.M.C. van Dijk
G.J. Lycklama à Nijeholt R.J. Vermeulen
E. Morava R.J.A. Wanders
P.J.W. Pouwels R.A. Wevers
J.A.M. Smeitink

123
Marjo S. van der Knaap, MD, PhD
Department of Child Neurology
VU University Medical Center
De Boelelaan 1117
1081 HV Amsterdam
The Netherlands

Jaap Valk, MD, PhD


Department of Radiology
VU University Medical Center
De Boelelaan 1117
1081 HV Amsterdam
The Netherlands

Third Edition
ISBN-10 3-540-22286-3 Springer Berlin Heidelberg New York
ISBN-13 978-3-540-22286-6 Springer Berlin Heidelberg New York
Second Edition
ISBN 3-540-59277-6 Springer Berlin Heidelberg New York

Library of Congress Control Number: 2004117334

This work is subject to copyright. All rights are reserved, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting,
reproduction on microfilm or in any other way, and storage in data banks. Duplication of this publication
or parts thereof is permitted only under the provision of the German Copyright Law of September 9,
1965, in its current version, and permission for use must always be obtained from Springer-Verlag.
Violations are liable to prosecution under the German Copyright Law.
Springer is a part of Springer Science+Business Media
springeronline.com
© Springer-Verlag Berlin Heidelberg, 1989, 1995, 2005
Printed in Germany
The use of designations, trademarks, etc. in this publication does not imply, even in the absence of a
specific statement, that such names are exempt from the relevant protective laws and regulations and
therefore free for general use.
Product liability: The publisher can not guarantee the accuracy of any information about dosage and
application contained in this book. In every individual case the user must check such information by
consulting the relevant literature.
Editor: Dr. Ute Heilmann, Heidelberg, Germany
Desk editor: Dörthe Mennecke-Bühler, Heidelberg, Germany
Production: PRO EDIT GmbH, Heidelberg, Germany
Cover-Design: Frido Steinen-Broo, Pau, Spain
Typesetting and Reproduction: AM-productions GmbH, Wiesloch, Germany
Printing and Binding: Stürtz GmbH, Würzburg, Germany
Printed on acid-free paper 21/3151Di - 5 4 3 2 1 0
Preface

Preface to the Third Edition Our thanks go to our colleagues at the VU Univer-
sity Medical Center and to those in other hospitals
Reading through the prefaces of the two previous edi- who referred their patients to us. We are indebted to
tions, we can say that much of what was said there still all colleagues who allowed us to use their MR images,
holds. At the same time, however, much has changed. published or unpublished, making it possible for us to
There has been immense progress in the technical present illustrations of nearly all known white matter
possibilities of magnetic resonance and in the know- disorders. Two colleagues were particularly helpful
ledge of genetic defects, biochemical abnormalities, and provided us with essential and unpublished fig-
and cellular processes underlying myelin disorders. ures: our friends Susan Blaser, from the Hospital for
This immense progress has prompted us to embark Sick Children in Toronto, and Zoltán Patay, from the
upon the enormous task of rewriting the previous King Faisal Hospital in Riyadh.
edition and adding 40 chapters. In doing so we have Many people at the VU University Medical Center
tried to cover most white matter disorders, hereditary have been of great technical help to us in producing
and acquired, and to present a collection of images to high quality images and in providing secretarial
illustrate the field to the fullest possible extent. This assistance. The contributions of these people are
edition will therefore be more complete than the pre- mentioned separately in the acknowledgements.
vious ones. The number of illustrations has increased Our special thanks go to patients with white mat-
considerably. This was necessary to reflect not only ter disorders and their families. They came to see us
the typical patterns of a disease, but to show also the and were willing to work with us and to go through
variability that exists in some disorders. The best ex- the procedure of diagnostic testing, including MR
ample of this is found in Alexander disease. Genetic examinations. Many patients and families were also
verification now makes it possible to recognize very willing to participate in our research projects to
different patterns of imaging abnormalities, all relat- advance the understanding of white matter disorders.
ed to a defect in the same gene. Today’s increased in- Patients with white matter disorders are the focus of
sight into disease classification based on increased our work. They are our most important collaborators.
knowledge of related genes and proteins is best re- Often they are children. To show our gratitude to
flected in the chapter on congenital muscular dystro- them, we have decided that all profits of this book will
phies. go to the Foundation for Children with White Matter
This is the first time that we have invited a number Disorders.
of experts in special fields to write or co-write a chap-
ter, in order to assure the highest level of scientific Amsterdam, May 2005
accuracy. To assemble the knowledge presented in
this work we have also harvested the literature, prof- M.S. van der Knaap
iting from the work and discoveries of many others. J. Valk
VI Preface

Preface to the Second Edition been derived both from MRI and from MR spec-
troscopy (MRS). This prompted us to review the clin-
The first edition of this book was well received by ical, laboratory, biochemical, and pathological data on
readers and reviewers and we are very grateful for the this subject in order to integrate all available informa-
positive reactions. We were convinced then, and even tion and to provide improved insights into normal
more now, that MRI and MRS have much to offer in and disordered myelin and myelination. We will show
diagnosis, therapy monitoring and research of hered- how the synthesis of all available information con-
itary and acquired myelin disorders. tributes to the interpretation of MR images.
In the last few years, a great deal of new information Following a brief historical review of the increas-
has become available concerning the genetic basis of ing knowledge on myelin and myelin disorders, we
inborn errors of metabolism and neurodegenerative propose a new classification of myelin disorders
disorders, the role of subcellular structures, the en- based on the subcellular localization of the enzymat-
zyme biochemistry, the pathophysiological mecha- ic defects as far as the inborn errors of metabolism
nisms of posthypoxic-ischemic cerebral damage, and are concerned. This classification serves as a guide
the inflammatory processes in infectious and inflam- throughout the book. All items of the classification
matory disorders. MR images of many rare disorders will be discussed and, whenever relevant and possi-
have become available, either in our own experience ble, illustrated by MR images.
or published by other groups. MR spectroscopy could We are aware of the fact that in a number of myelin
confirm its role in certain clinical applications. Be- disorders MRI is not a part of the usual diagnostic
cause of these developments, it was necessary for us to work up because a definite diagnosis is reached by
rewrite the book almost completely. In some fields other means, such as biochemical investigations of
developments are so fast that we have not have caught blood and urine, enzyme assessment or detection of
all the latest developments. The pattern of the new specific antibodies. However, in many disorders MRI
approaches has, however, been established, making may facilitate a rapid diagnosis and early instigation
the assimilation of newly available information easy. of treatment, thus preventing structural cerebral
We are extremely grateful for the help of colleagues damage. In other cases the role of MRI is to visualize
to make this book as complete as possible. The posi- the extent of brain damage and give an indication of
tive reactions of those from whom we requested MR the prognosis. In disorders which present in a non-
pictures or other forms of support were of enormous specific way, for instance with behavioral problems or
encouragement to us during our efforts to complete learning difficulties, MRI can be one of the first-line
this project. investigations. It is important to be acquainted with
We hope this work will be as warmly welcomed by the various MRI patterns of the myelin disorders, as
our colleagues as the first edition. an early diagnosis may be of major importance in
young families with a view to the provision of ade-
Amsterdam, January 1995 quate genetic counseling.
MRS has been of limited clinical importance until
M.S. van der Knaap now, and its application in patients only has a short
J. Valk history. We do, however, expect it to be a promising
technique in the field of myelin and myelin disorders
in clinical as well as in basic, experimental research
Preface to the First Edition and have, therefore, devoted a separate chapter to this
subject.
Magnetic resonance imaging (MRI) is now consid- This volume was written by a neuroradiologist and
ered to be the imaging modality of choice for the ma- a neurologist/child neurologist. It is the product of
jority of disorders affecting the central nervous sys- close cooperation, animated discussions, strong argu-
tem. This is particularly true for gray and white mat- ments, restructuring, rewriting, and editing, in which
ter disorders, thanks to the superb soft tissue contrast they had an equal share. If the reader finds value in
in MRI which allows gray matter, unmyelinated, and this monograph, it is because of this dual effort.
myelinated white matter to be distinguished and their
respective disorders identified. The present book is Amsterdam and Utrecht, March 1989
devoted to the disorders of myelin and myelination.
A growing amount of detailed in vivo information J. Valk
about myelin, myelination, and myelin disorders has M.S. van der Knaap
Acknowledgements

The preparation of this book was a project of several Annuska Houtappels, who digitized older films and
years and could not have been concluded successfully helped us improve the quality of the images.
without the support and collaboration of many peo- Excellent secretarial help was provided by Sigrid
ple. Thanks to all. Bruinsma, who single-handedly took care of the ref-
Special thanks go to our colleagues: Jeroen Ver- erence section. Staff members of the VUMC Library
meulen and Leo Smit, pediatric neurologists, and Els van Deventer, Linda Glas, Margreet Bosshardt, and
Frederik Barkhof and Jonas Castelijns, neuroradiolo- Cisca Frederiks were very helpful in providing us
gists, at the VU University Medical Center (VUMC) with the necessary literature.
in Amsterdam; Martin Heitbrink and Bart Wiarda, Technical support and guidance with computer
radiologists at the Medical Center Alkmaar; and Erik programs and settings were provided by the Depart-
Veldhuizen, radiologist at the MRI center, Amster- ment of Informatics of the VUMC. We are grateful for
dam, for their continuous support during this en- their kind and prompt assistance. Special thanks go to
deavor. We are grateful to the MRI technicians at the Michiel Sprenger, Guido Zonneveld, and Peter Theijs-
VUMC, who guaranteed the quality of the MR exam- meijer.
inations and had the patience and empathy to deal We acknowledge the continuous friendly and
with very sick children and their parents. We want to encouraging support of the editorial staff of Springer-
mention especially the help of Karin Barbiers and Er- Verlag, Dr. Ute Heilmann and Mrs Dörthe Mennecke-
win Kist, who headed this team and carried out the re- Bühler.
trieval of older examinations to the Image Manage-
ment System. Amsterdam, May 2005
We received great support from the audiovisual
center at the VUMC. We are especially indebted M.S. van der Knaap
to Daan van Eijndhoven, Rene den Engelsman, and J. Valk
Contents

1 Myelin and White Matter . . . . . . . . . 1 33 Trichothiodystrophy


2 Classification of Myelin Disorders . . . . 20 with Photosensitivity . . . . . . . . . . . 268
3 Selective Vulnerability . . . . . . . . . . 25 34 Pelizaeus–Merzbacher Disease
4 Myelination and Retarded Myelination 37 and X-linked Spastic
Paraplegia Type 2 . . . . . . . . . . . . . 272
5 Lysosomes and Lysosomal Disorders . . 66
35 18q– Syndrome . . . . . . . . . . . . . . . 281
6 Metachromatic Leukodystrophy. . . . . 74
36 Phenylketonuria . . . . . . . . . . . . . . 284
7 Multiple Sulfatase Deficiency . . . . . . 82
37 Glutaric Aciduria Type 1 . . . . . . . . . . 294
8 Globoid Cell Leukodystrophy
(Krabbe Disease) . . . . . . . . . . . . . . 87 38 Propionic Acidemia . . . . . . . . . . . . 300
9 GM1 Gangliosidosis . . . . . . . . . . . . 96 39 Nonketotic Hyperglycinemia . . . . . . 306
10 GM2 Gangliosidosis . . . . . . . . . . . . 103 40 Maple Syrup Urine Disease . . . . . . . . 311
11 Fabry Disease . . . . . . . . . . . . . . . . 112 41 3-Hydroxy 3-Methylglutaryl-CoA
Lyase Deficiency . . . . . . . . . . . . . . 321
12 Fucosidosis . . . . . . . . . . . . . . . . . 119
42 Canavan Disease . . . . . . . . . . . . . . 326
13 Mucopolysaccharidoses . . . . . . . . . 123
43 L-2-Hydroxyglutaric Aciduria . . . . . . 334
14 Free Sialic Acid Storage Disorder . . . . 133
44 D-2-Hydroxyglutaric Aciduria . . . . . . 338
15 Neuronal Ceroid Lipofuscinoses . . . . 137
45 Hyperhomocysteinemias . . . . . . . . . 342
16 Adult Polyglucosan Body Disease . . . . 147
46 Urea Cycle Defects . . . . . . . . . . . . . 360
17 Peroxisomes
and Peroxisomal Disorders . . . . . . . . 151 47 Serine Synthesis Defect Caused
by 3-Phosphoglycerate
18 Peroxisome Biogenesis Defects . . . . . 154
Dehydrogenase Deficiency . . . . . . . . 369
19 Peroxisomal D-Bifunctional
48 Molybdenum Cofactor
Protein Deficiency . . . . . . . . . . . . . 167
Deficiency and Isolated Sulfite
20 Peroxisomal Acyl-CoA Oxidase Deficiency. . . . . . . . . . . . . 372
Oxidase Deficiency. . . . . . . . . . . . . 172
49 Galactosemia . . . . . . . . . . . . . . . . 377
21 X-linked Adrenoleukodystrophy . . . . 176
50 Sjögren–Larsson Syndrome . . . . . . . 383
22 Refsum Disease . . . . . . . . . . . . . . . 191
51 Lowe Syndrome . . . . . . . . . . . . . . 387
23 Mitochondria
52 Wilson Disease . . . . . . . . . . . . . . . 392
and Mitochondrial Disorders. . . . . . . 195
53 Menkes Disease . . . . . . . . . . . . . . 400
24 Mitochondrial Encephalopathy
with Lactic Acidosis 54 Fragile X Premutation . . . . . . . . . . . 406
and Stroke-like Episodes . . . . . . . . . 204 55 Hypomelanosis of Ito . . . . . . . . . . . 409
25 Leber Hereditary Optic Neuropathy . . 212 56 Incontinentia Pigmenti . . . . . . . . . . 412
26 Kearns–Sayre Syndrome . . . . . . . . . 215 57 Alexander Disease . . . . . . . . . . . . . 416
27 Mitochondrial Neurogastrointestinal 58 Giant Axonal Neuropathy . . . . . . . . . 436
Encephalomyopathy. . . . . . . . . . . . 221 59 Megalencephalic Leukoencephalo-
28 Leigh Syndrome and Mitochondrial pathy with Subcortical Cysts . . . . . . . 442
Leukoencephalopathies . . . . . . . . . 224 60 Congenital Muscular Dystrophies . . . . 451
29 Pyruvate Carboxylase Deficiency . . . . 245 61 Myotonic Dystrophy Type I . . . . . . . . 469
30 Multiple Carboxylase Deficiency . . . . 248 62 Myotonic Dystrophy Type 2 . . . . . . . 473
31 Cerebrotendinous Xanthomatosis . . . 252 63 X-linked Charcot–Marie–Tooth
32 Cockayne Syndrome . . . . . . . . . . . . 259 Disease . . . . . . . . . . . . . . . . . . . 476
X Contents

64 Oculodentodigital Dysplasia . . . . . . 479 86 Whipple Disease . . . . . . . . . . . . . . 658


65 Leukoencephalopathy 87 Toxic Encephalopathies . . . . . . . . . . 664
with Vanishing White Matter . . . . . . . 481 88 Iatrogenic Toxic Encephalopathies . . . 679
66 Aicardi–Goutières Syndrome . . . . . . 496 89 Central Pontine and Extrapontine
67 Leukoencephalopathy Myelinolysis . . . . . . . . . . . . . . . . 684
with Calcifications and Cysts . . . . . . . 505 90 Hypernatremia . . . . . . . . . . . . . . . 690
68 Leukoencephalopathy with Brain Stem 91 Marchiafava–Bignami Syndrome . . . . 695
and Spinal Cord Involvement 92 Posterior Reversible
and Elevated White Matter Lactate . . . 510 Encephalopathy Syndrome . . . . . . . 699
69 Hypomyelination with Atrophy 93 Langerhans Cell Histiocytosis . . . . . . 709
of the Basal Ganglia and Cerebellum . . 519
94 Post-Hypoxic–Ischemic Damage . . . . 714
70 Hereditary Diffuse Leukoencephalo-
95 Post-Hypoxic–Ischemic
pathy with Neuroaxonal Spheroids . . . 526
Leukoencephalopathy of Neonates . . . 718
71 Dentatorubropallidoluysian Atrophy . 530
96 Neonatal Hypoglycemia . . . . . . . . . 749
72 Cerebral Amyloid Angiopathy . . . . . . 535
97 Delayed Posthypoxic
73 Cerebral Autosomal Dominant Leukoencephalopathy . . . . . . . . . . 755
Arteriopathy with Subcortical Infarcts
98 White Matter Lesions of the Elderly . . . 759
and Leukoencephalopathy . . . . . . . . 541
99 Subcortical Arteriosclerotic
74 Cerebral Autosomal Recessive
Encephalopathy . . . . . . . . . . . . . . 767
Arteriopathy with Subcortical Infarcts
and Leukoencephalopathy . . . . . . . . 549 100 Vasculitis . . . . . . . . . . . . . . . . . . 773
75 Polycystic Lipomembranous 101 Leukoencephalopathy
Osteodysplasia with Sclerosing and Dural Venous Fistula . . . . . . . . . 801
Leukoencephalopathy 102 Leukoencephalopathy after Chemo-
(Nasu-Hakola Disease). . . . . . . . . . . 552 therapy and/or Radiotherapy . . . . . . 808
76 Pigmentary Orthochromatic 103 Gliomatosis Cerebri . . . . . . . . . . . . 818
Leukodystrophy . . . . . . . . . . . . . . 557 104 Diffuse Axonal Injury . . . . . . . . . . . 823
77 Adult-Onset Autosomal Dominant 105 Wallerian Degeneration
Leukoencephalopathies . . . . . . . . . 559 and Myelin Loss Secondary
78 Inflammatory to Neuronal and Axonal
and Infectious Disorders . . . . . . . . . 561 Degeneration . . . . . . . . . . . . . . . . 832
79 Multiple Sclerosis . . . . . . . . . . . . . 566 106 Diffusion-Weighted Imaging . . . . . . 839
80 Acute Disseminated Encephalomyelitis 107 Magnetization Transfer Imaging . . . . 854
and Acute Hemorrhagic 108 Magnetic Resonance Spectroscopy:
Encephalomyelitis . . . . . . . . . . . . . 604 Basic Principles, and Application
81 Acquired Immunodeficiency in White Matter Disorders. . . . . . . . . 859
Syndrome . . . . . . . . . . . . . . . . . . 616 109 Pattern Recognition
82 Progressive Multifocal in White Matter Disorders. . . . . . . . . 881
Leukoencephalopathy . . . . . . . . . . 628
References . . . . . . . . . . . . . . . . . . . . . 905
83 Brucellosis . . . . . . . . . . . . . . . . . 635
84 Subacute Sclerosing Panencephalitis . 640 Subject Index . . . . . . . . . . . . . . . . . . 1075
85 Congenital and Perinatal
Cytomegalovirus Infection . . . . . . . . 645
Contributors

F. Barkhof, MD PhD R. van den Berg, MD PhD


Department of Radiology Department of Radiology
and MR Center for MS Research VU University Medical Center, Amsterdam
VU University Medical Center and Department of Radiology
Amsterdam, The Netherlands Leiden University Medical Center
Leiden, The Netherlands
V. Geiselmann, PhD
Institut fur Physiologische Chemie M.S. van der Knaap, MD PhD
Rheinische Friedrich-Wilhelms-Universität Department of Child Neurology
Bonn, Germany VU University Medical Center
Amsterdam, The Netherlands
G.J. Lycklama à Nijeholt, MD PhD
Department of Radiology J.M.C. van Dijk, MD PhD
VU University Medical Center Department of Neurosurgery
Amsterdam, The Netherlands Leiden University Medical Center
Leiden, The Netherlands
E. Morava, MD
Nijmegen Center for Mitochondrial Disorders R.J. Vermeulen, MD PhD
and Department of Pediatrics Department of Child Neurology
University Medical Center Nijmegen VU University Medical Center
Nijmegen, The Netherlands Amsterdam, The Netherlands

P.J.W. Pouwels, PhD R.J.A. Wanders, PhD


Department of Clinical Physics and Informatics Department of Clinical Chemistry
VU University Medical Center and Department of Pediatrics
Amsterdam, The Netherlands Academic Medical Center
Amsterdam, The Netherlands
J.A.M. Smeitink, MD PhD
Nijmegen Center for Mitochondrial Disorders R.A. Wevers, PhD
and Department of Pediatrics Laboratory of Pediatrics and Neurology
University Medical Center Nijmegen University Medical Center Nijmegen St Radboud
Nijmegen, The Netherlands Nijmegen, The Netherlands

J. Valk, MD PhD
Department of Radiology
VU University Medical Center
Amsterdam, The Netherlands
List of Abbreviations

ACE angiotensin converting enzyme CAMFAK cataracts–microcephaly–failure


ACTH adrenocorticotropic hormone to thrive–kyphoscoliosis (syndrome)
AD Alexander disease CD Canavan disease; cluster determinant
ADC apparent diffusion coefficient Cho choline
ADEM acute disseminated encephalomyelitis CIPO chronic intestinal pseudo-obstruction
ADP adenosine diphosphate CIS clinically isolated symptom
AD PEO autosomal dominant progressive CK creatine kinase
external ophthalmoplegia CMD congenital muscular dystrophy
AHEM acute hemorrhagic encephalomyelitis CMT Charcot–Marie–Tooth disease
AIDS acquired immunodeficiency syndrome CMTX X-linked form of CMT
ALD adrenoleukodystrophy CMV cytomegalovirus
ALDP ALD protein CNP 2’3’-cyclic nucleotide
ALL acute lymphocytic leukemia 3’-phosphodiesterase
AMN adrenomyeloneuropathy CNS central nervous system
ANCAs anti-neutrophil cytoplasm antibodies COFS cerebro-oculofacioskeletal (syndrome)
ANCL adult neuronal ceroid lipofuscinosis COX cytochrome-c oxidase
(or Kufs disease) CPEO chronic progressive external
AP4 2-amino-4-phosphonobutyrate ophthalmoplegia
APLA anti-phospholipid antibodies CPM central pontine myelinolysis
APBD adult polyglucosan body disease CPSD carbamyl phosphate synthetase
apoE apolipoprotein E deficiency
APP amyloid precursor protein CPT carnitine palmitoyl transferase
aPTT activated partial thromboplastin time Cr creatine
ASLD argininosuccinate lyase deficiency CREST calcinosis, Raynaud syndrome,
ASSD argininosuccinate synthetase deficiency esophageal problems, sclerodactylia,
ATP adenosine triphosphate and telangiectasia (syndrome)
BAEP brain stem auditory evoked potential CS Cockayne syndrome; concentric
BCNU bis-chloroethyl-nitrosourea sclerosis (or Baló disease)
BDNF brain-derived neurotrophic factor CSF cerebrospinal fluid
bFGF basic fibroblast growth factor CSI chemical shift imaging
BIDS brittle hair, impaired intelligence, CT computed tomography/tomogram
decreased fertility, short stature CTX cerebrotendinous xanthomatosis
(syndrome) DAB diaminobenzidine
BMAA β-N-methylamino-L-alanine DAGC dystrophin-associated
BOMAA β-N-oxalylmethylamino-L-alanine glycoprotein complex
BPD D-bifunctional protein deficiency DAI diffuse axonal injury
CAA cerebral amyloid angiopathy DAVF cranial dural arteriovenous fistula
CACH childhood ataxia with central nervous DHAPAT dihydroxyacetonephosphate
system hypomyelination acyltransferase
CACT mitochondrial carnitine/acylcarnitine DM 1 myotonic dystrophy type 1
transporter DM 2 myotonic dystrophy type 2
CADASIL cerebral autosomal dominant DNA deoxyribonucleic acid
arteriopathy with subcortical infarcts DNC deoxynucleotide carrier
and leukoencephalopathy dNTP deoxyribonucleoside triphosphate
cANCA cytoplasmic form of ANCA DOA dominant optic atrophy
CARASIL cerebral autosomal recessive DOPA dihydroxyphenylalanine
arteriopathy with subcortical infarcts DPHL delayed posthypoxic
and leukoencephalopathy leukoencephalopathy
DRPLA dentatorubropallidoluysian atrophy
XIV List of Abbreviations

DS diffuse sclerosis (or Schilder disease) HCHWA-D Dutch type of hereditary cerebral
DSA digital subtraction angiography hemorrhage with amyloidosis
DTI diffusion tensor imaging HDL high-density lipoproteins
DWI diffusion-weighted imaging HDLS hereditary diffuse leukoencephalopathy
EAA excitatory amino acid with spheroids
EAE experimental allergic encephalomyelitis 5HIAA 5-hydroxyindoleacetic acid
ECD ethyl cysteinate dimer HIV-1 human immunodeficiency virus type 1
ECG electrocardiography/electrocardiogram HLA human leukocyte antigen
EDSS Expanded Disability Status Scale HMG-CoA 3-hydroxy-3-methylglutaryl-
EEG electroencephalogram coenzyme A
EGF epidermal growth factor HMI hypomelanosis of Ito
eIF eukaryotic initiation factor HMPAO hexamethylpropyleneamine oxime
ELISA enzyme-linked immunosorbent assay HSP hereditary spastic paraplegia; heat
EMG electromyogram shock protein
EPI echo planar imaging HTLV human T-cell lymphotropic virus
EPM extrapontine myelinolysis HUS hemolytic–uremic syndrome
EPMR progressive epilepsy HVA homovanillic acid
with mental retardation IBIDS ichthyosis, brittle hair, impaired
ERG electroretinography/electroretinogram intelligence, decreased fertility,
FA fractional anisotropy short stature (syndrome)
FAD flavin adenine dinucleotide IFN interferon
FADH2 flavin adenine dinucleotide, reduced Ig immunoglobulin
FCMD Fukuyama congenital muscular IGF insulin-like growth factor
dystrophy INCL infantile neuronal ceroid lipofuscinosis
FD Fabry disease (or Santavuori disease)
FISH fluorescent in situ hybridization IP incontinentia pigmenti
FLAIR fluid-attenuated inversion recovery IQ intelligence quotient
FSE fast spin echo IR inversion recovery
FSH follicle-stimulating hormone IRD infantile Refsum disease
5-FU 5-fluorouracil ISIS image-selective in vivo spectroscopy
FvLINCL Finnish variant of late-infantile ISSD severe infantile sialic acid
neuronal ceroid lipofuscinosis storage disease
GA gestational age IVL intravascular lymphomatosis
GABA γ-aminobutyric acid JNCL juvenile neuronal ceroid lipofuscinosis
GAMT guanidinoacetate methyltransferase (or Spielmeyer–Vogt disease, or Batten
GAN giant axonal neuropathy disease)
GDP guanosine diphosphate KA kainate
GE gradient echo kDa kiloDalton
GEF guanine-nucleotide exchange factor KSS Kearns–Sayre syndrome
GFAP glial fibrillary acidic protein LAMP lysosome-associated membrane protein
GIP general insertion protein LBSL leukoencephalopathy with brain stem
GLD globoid cell leukodystrophy and spinal cord involvement and elevat-
Glx glutamine, glutamate, GABA ed white matter lactate
GOM granular osmiophilic material LCC leukoencephalopathy with calcifications
GRACILE growth retardation, aminoaciduria, and cysts
cholestasis, iron overload, lactic LCH Langerhans cell histiocytosis
acidosis, and early death (syndrome) LDL low-density lipoproteins
GROD granular osmiophilic deposits LGMD limb girdle muscular dystrophy
GTE glyceryl trierucate LH luteinizing hormone
GTO glyceryl trioleate LHON Leber hereditary optic neuropathy
GTP guanosine triphosphate LINCL late-infantile neuronal ceroid
GVHD graft-versus-host disease lipofuscinosis (or Jansky–
HAART highly active/aggressive Bielschowsky disease)
anti-retroviral treatment MAG myelin-associated glycoprotein
HABC hypomyelination with atrophy MAP microtubule-associated protein
of the basal ganglia and cerebellum MBS Marchiafava–Bignami syndrome
MBP myelin basic protein
List of Abbreviations XV

MCE multicystic encephalopathy NCL neuronal ceroid lipofuscinosis


MD Menkes disease; myotonic dystrophy nDNA nuclear DNA
MDC1A merosin-deficient congenital NKH nonketotic hyperglycinemia
muscular dystrophy NMDA N-methyl-D-aspartate
MEB muscle–eye–brain disease NMO neuromyelitis optica (or Devic disease)
MELAS mitochondrial encephalomyopathy, NRTI nucleoside analogue reverse
lactic acidosis, and stroke-like episodes transcriptase inhibitor
MEPOP mitochondrial encephalomyopathy NT neurotrophin
with sensorimotor polyneuropathy, OCRL oculocerebrorenal syndrome of Lowe
ophthalmoplegia, and pseudo- ODDD oculodentodigital dysplasia
obstruction OGIMD oculogastrointestinal muscular
MERRF myoclonic epilepsy dystrophy
with ragged red fibers OHS occipital horn syndrome
MHC major histocompatibility complex OMgp oligodendrocyte myelin glycoprotein
MHPG 3-methoxy-4-hydroxyphenylglycol ONMR onychotrichodysplasia, neutropenia,
MICS microcephaly–intracranial mental retardation (syndrome)
calcifications syndrome OSP oligodendrocyte-specific protein
MIL multifocal inflammatory OTCD ornithine transcarbamylase deficiency
leukoencephalopathy PACNS primary angiitis of the CNS
mIns myo-inositol PAF platelet activating factor
MLC megalencephalic leukoencephalopathy PAN polyarteritis nodosa
with subcortical cysts pANCA perinuclear form of ANCA
MLD metachromatic leukodystrophy PAS periodic acid–Schiff
MNGIE mitochondrial neurogastrointestinal PCD pyruvate carboxylase deficiency
encephalomyopathy PCr phosphocreatine
MOBP myelin-associated oligodendrocytic PCR polymerase chain reaction
basic protein PDE phosphodiesters
MOG myelin oligodendrocyte glycoprotein PDGF platelet-derived growth factor
MOM mitochondrial outer membrane PDHc pyruvate dehydrogenase complex
MOSP myelin-/oligodendrocyte-specific PEP processing enhancing protein
protein PET positron emission tomography
MPP mitochondrial processing peptidase Pi inorganic phosphate
MPS mucopolysaccharidoses; mucopoly- PIBIDS photosensitivity, ichthyosis, brittle hair,
saccharidoses impaired intelligence, decreased
MPTP methylphenyltetrahydropyridine fertility, short stature (syndrome)
MR magnetic resonance PIP2 phosphatidylinositol 4,5-biphosphate
MRA magnetic resonance angiography PKU phenylketonuria
MRI magnetic resonance imaging PLOSL polycystic lipomembranous osteo-
mRNA messenger RNA dysplasia with sclerosing leuko-
MRS magnetic resonance spectroscopy encephalopathy
MS multiple sclerosis PLP proteolipid protein
MSD multiple sulfatase deficiency PMD Pelizaeus–Merzbacher disease;
MSUD maple syrup urine disease proximal myotonic dystrophy
MT magnetization transfer PME phosphomonoesters
mtDNA mitochondrial DNA PML progressive multifocal leuko-
MTI magnetization transfer imaging encephalopathy
MTR magnetization transfer ratio PMP peroxisomal membrane protein
NAA N-acetylaspartate PNS peripheral nervous system
NAAG N-acetylaspartyl glutamate POLD pigmentary orthochromatic
NAD nicotinamide adenine dinucleotide leukodystrophy
NADH nicotinamide adenine dinucleotide, POLIP polyneuropathy, ophthalmoplegia,
reduced leukoencephalopathy, and intestinal
NALD neonatal adrenoleukodystrophy pseudo-obstruction
NARP neurogenic muscle weakness, ataxia, PPAR peroxisome proliferator activating
and retinitis pigmentosa receptor
NAWM normal-appearing white matter ppm parts per million
NBCA n-butyl cyanoacrylate
XVI List of Abbreviations

PPRE peroxisome proliferator response T Tesla


element TE toxic encephalopathy; echo time
PPT1 palmitoyl protein thioesterase 1 TI inversion time
PRES posterior reversible encephalopathy TNF-α tumor necrosis factor-alpha
syndrome TORCH toxoplasmosis, rubella, cytomegalo-
PRESS point-resolved spectroscopy virus, herpes simplex
PROMM proximal myotonic myopathy TPP1 tripeptidyl peptidase 1
PTS peroxisome targeting signals TR repetition time
PVA polyvinyl alcohol tRNA transfer RNA
PVL periventricular leukomalacia TSD Tay–Sachs disease
QA quisqualate TSE turbo spin echo
RCDP rhizomelic chondrodysplasia punctata TTD trichothiodystrophy with photo-
RD Refsum disease sensitivity
RF radiofrequency TTP thrombotic thrombocytopenic purpura
RNA ribonucleic acid TvLINCL Turkish variant of late-infantile
RPLS reversible posterior leukoencephalo- neuronal ceroid lipofuscinosis
pathy syndrome TYROBP TYRO protein tyrosine kinase
RPR rapid plasma reagin (test) binding protein
RR relapsing remitting UDP uridine diphosphate
rRNA ribosomal RNA US ultrasound/ultrasonography
RXR retinoic acid receptor UV ultraviolet
SAE subcortical arteriosclerotic V-CAM cellular adhesion molecules
encephalopathy VDAC voltage-dependent,
SAP sphingolipid activator protein anion-selective channel
SCA spinocerebellar ataxia VDRL Venereal Disease Research Laboratory
SCL subcortical leukomalacia (test)
SD Salla disease VEGF vascular endothelial growth factor
SE spine echo VEP visual evoked potential
SIBIDS osteosclerosis, ichthyosis, brittle hair, VLA-4 very late antigen 4
impaired intelligence, decreased VLCFA very-long-chain fatty acids
fertility, short stature (syndrome) vLINCL variant late-infantile neuronal ceroid
SLE systemic lupus erythematosus lipofuscinosis
SLS Sjögren–Larsson syndrome VMA vanillyl mandelic acid
SP secondary progressive VWM vanishing white matter
SPECT single photon emission computed WD Wilson disease
tomography WM white matter
SPG2 spastic paraparesis type 2 WWS Walker–Warburg syndrome
SSEP somatosensory evoked potential XALD X-linked adrenoleukodystrophy
SSPE subacute sclerosing panencephalitis XP xeroderma pigmentosum
STEAM stimulated-echo acquisition mode ZS Zellweger syndrome
STIR short tau inversion recovery
Chapter 1

Myelin and White Matter

1.1 Introduction some myelin is present around intracortical and in-


tranuclear fibers. The myelin content of the thalamus
Myelin makes up most of the substance of the white and the globus pallidus is relatively high.
matter in the central nervous system (CNS). It is
also present in large quantities in the peripheral ner-
vous system (PNS). In both the CNS and the PNS, 1.2 Morphology of Myelin
myelin is essential for normal functioning of the
nerve fibers. Myelin is a spiral membranous structure that is tight-
The white matter in the CNS is composed of a vast ly wrapped around axons. It has a very high lipid con-
number of axons, which are ensheathed with myelin, tent and is soluble in fat solvents. Hence, when ordi-
which is responsible for the white color. Besides nary paraffin sections of the brain are prepared for
myelinated axons, white matter contains many cells of light microscopic examination, most of the myelin
the neuroglia type, but no cell bodies of neurons. The dissolves away. After staining, the sites where myelin
axons it contains originate from neuronal cell bodies was present appear as round spaces that are empty
in gray matter structures. except that each has a little round dot in the center,
There are two main types of macroglia in the white which represents a cross section of the axon. By
matter: astrocytes and oligodendrocytes. Among the means of fixatives that make myelin insoluble, it is
many putative functions of glial cells, it is proposed possible to demonstrate it in paraffin sections. Osmic
that they contribute to the structural and nutritive acid fixes myelin so that it does not dissolve in paraf-
support of neurons, regulate the extracellular envi- fin sections. Osmic acid itself stains myelin black.
ronment of ions and transmitters, guide migrating When examined under very low power, the white
neurons during development, and play an important matter appears black (Fig. 1.1). If the white matter is
part in repair and regeneration. The best known examined under high power the myelin will be seen
function of glial cells is the ensheathment of axons to be arranged in small rings around each nerve fiber.
with myelin by oligodendrocytes. There are several myelin stains that can be used once
Gray matter contains the nerve cell bodies with the tissue has been fixed by some other means. Com-
their extensive dendritic arborization. The myelin monly used stains include hematoxylin, Luxol fast
content of gray matter structures is much lower, but blue, and Oil-Red-O.

Fig. 1.1. T2-weighted MR image compared with a


postmortem section prepared with a myelin stain,
illustrating the capability of MRI to reflect histology
2 Chapter 1 Myelin and White Matter

Fig. 1.3.
A micelle

Fig. 1.4.
A lipid bilayer

Fig. 1.2. Electron micrograph of white matter with myelin sheaths

The information derived from light microscopic soluble in water, but soluble in oil. Conversely, hy-
investigations is limited and is inadequate when more drophilic substances are insoluble in oil, but soluble
detailed information about myelin structure is re- in water. In an aqueous environment, the amphipa-
quired. Analysis of the structure of myelin began in thic character of the lipids favors aggregation into mi-
the 1930s, stimulated by polarization-microscope celles or a molecular bilayer. In a micelle (Fig. 1.3), the
studies and X-ray diffraction work, which led to the hydrophobic regions of the amphipathic molecules
suggestion that the myelin sheath was made up of are shielded from water, while the hydrophilic polar
layers or lamellae. The lamellar structure was con- groups are in direct contact with water. The stability
firmed by electron microscopic studies. In electron of this structure lies in the fact that significant free
micrographs myelin is seen as a series of alternating energy is required to transfer a nonpolar molecule
dark and less dark lines separated by unstained from a nonpolar medium to water. Likewise, a great
zones. These lines are wrapped spirally around the deal of energy is required to transfer a polar moiety
axon (Fig. 1.2). The evidence available from studies from water to a nonpolar medium. Thus, the micelle
using polarized light, X-ray diffraction and electron provides a minimal energy configuration and is ac-
microscopy led to the current view of myelin as a sys- cordingly thermodynamically stable. The molecular
tem of condensed plasma membranes with alternat- bilayer, the basic structure of plasma cell membranes,
ing protein-lipid-protein-lipid-protein lamellae as also satisfies the thermodynamic requirements of
the repeating subunit. amphipathic molecules in an aqueous environment.
Plasma membranes are composed predominantly A bilayer exists as a sheet in which the hydrophobic
of lipids and proteins, and also contain carbohydrate regions of the lipids are protected from the water
components. The lipid elements of the membranes while the hydrophilic regions are immersed in water
are phospholipids, glycolipids, and cholesterol. A (Fig. 1.4).As the structure of the bilayer is an inherent
common property of these lipids is that they are am- part of the amphipathic character of the lipid mole-
phipathic. This means that the lipid molecules con- cules, the formation of lipid bilayers is essentially a
tain both hydrophobic and hydrophilic regions, cor- self-assembly process.
responding to the nonpolar tails and the polar head In comparison with other molecular bilayers, the
groups, respectively. Hydrophobic substances are in- myelin bilayer is unique in having a very high lipid
1.2 Morphology of Myelin 3

Fig. 1.5. Membrane split open to


demonstrate the layers.The lipid
bilayer is interrupted by proteins
embedded in this layer. Glycoprotein
chains rise from the surface of the
membrane

content and containing chiefly saturated fatty acids are asymmetrically distributed across the membrane
with an extraordinarily long chain length. This fatty bilayer and the protein composition on the inside is
acid composition leads to a closely packed, highly sta- different from that on the outside of the bilayer.
ble membrane structure. The presence of unsaturated On electron microscopic examination, a plasma
fatty acids in a bimolecular leaflet leads to a more membrane is shown as a three-layered structure and
loosely packed, less stable structure, as unsaturated consists of two dark lines separated by a lighter inter-
fatty acid chains have a kinked, hook-like configura- val. It is also revealed that the plasma membrane is
tion. Lipids containing such unsaturated fatty acids not symmetrical in form: the dark line adjacent to the
cannot approach neighboring molecules as closely as cytoplasm is denser than the leaflet on the outside.
saturated lipids can, since the latter are rod-like struc- From both X-ray diffraction and electron micro-
tures. There will be much less total interaction be- scope data it can be seen that the smallest radial sub-
tween the tails of an unsaturated lipid and a neigh- unit that can be called myelin is a five-layered struc-
boring molecule than between the tails of two satu- ture of protein-lipid-protein-lipid-protein (Fig. 1.6).
rated lipids, and the resulting binding forces will be The repeat distance is 160–180 Å. The dark lines seen
much smaller. Lipids containing long-chain fatty in electron microscopic studies represent the protein
acids are more tightly held in a membrane structure layers and the unstained zones, the lipids. The uneven
than those containing shorter chain fatty acids, since staining of the protein layers results from the way the
with increasing length of the hydrocarbon chain the myelin sheath is generated from the plasma mem-
binding interactions between the lipid molecules be- brane. The less dark lines (so-called intraperiod
come stronger. It has also been suggested that very- lines) represent the closely apposed outer protein
long-chain fatty acids can form complexes by inter- coats of the original cell membrane. The dark lines
digitation of the hydrocarbon tail on one side with the (so-called major dense lines) are the fused inner pro-
hydrocarbon tail of a lipid on the opposite side of the tein coats of the cell membrane. High-magnification
bimolecular leaflet. Such complexes would contribute electron micrographs show that the intraperiod line
to the stability of the myelin membrane. If this lipid is double in nature (Fig. 1.6).
composition is changed, as is the case in a number of The myelin sheath is not continuous along the en-
demyelinating disorders, it is clear that the stability of tire length of axons, but axons are covered by seg-
the myelin membrane may be diminished. ments of myelin, which are separated by small regions
The bimolecular lipid structure allows for interac- of uncovered axon, the nodes of Ranvier. The myelin
tion of amphipathic proteins with the membrane. lamellae terminate as they approach the node. The re-
These proteins form an integral part of the membrane, gion where the lamellae terminate is known as the
with hydrophilic regions protruding from the inner paranode. Electron micrographs of longitudinal sec-
and outer faces of the membrane and connected by a tions of paranodal regions show that the major dense
hydrophobic region traversing the hydrophobic core lines open up and loop back upon themselves, enclos-
of the bilayer. In addition, there are peripheral pro- ing cytoplasm within the loop (Fig. 1.7). In that part
teins, which do not interact directly with the lipids in of the paranode most distant from the node, the in-
the bilayer, but are bound to the hydrophilic regions of nermost lamellae of the myelin terminate first, and
specific integral proteins. Thus, the cell membrane is a succeeding turns of the spiral of lamellae then over-
bimolecular lipid leaflet coated with proteins on both lap and project beyond the ones lying beneath. Thus,
sides (Fig. 1.5). There is inside-outside asymmetry of the outermost lamella overlaps all the others and ter-
the lipids. In addition, integral and peripheral proteins minates nearest the node, so that the myelin sheath
4 Chapter 1 Myelin and White Matter

Fig. 1.6. The electron microscopic


picture of a myelin sheath (upper left)
reveals the five-layered structure of
myelin with major dense lines and
intraperiod lines. A higher magnifica-
tion of two myelin lamellae (lower left)
shows the periodicity of myelin even
more clearly. On the right, a schematic
representation of an electron micro-
scopic picture of a myelin sheath
surrounding an axon (A) demonstrates
major dense lines (md) and intraperi-
od lines (ip)

Fig. 1.7. Node of Ranvier, where the


nerve fiber between two myelinated
segments is bare.The outer myelin
layers envelope the inner layer and
cover these at the nodal junctions

gradually becomes thinner with increasing proximity and myelin sheaths can be observed. In the gray mat-
to the node. ter they aggregate closely around neuronal cell bod-
Schmidt-Lantermann clefts such as are described ies, where they are called satellite oligodendrocytes.
in the PNS are rare in the CNS. These are funnel- PNS myelin is formed by Schwann cells. The CNS
shaped clefts within myelin sheaths. They contain cy- myelin membranes originate from and are part of the
toplasm and extend from the soma of the myelin- oligodendroglial cell membrane. The oligodendro-
forming cell to the inner end of the myelin sheath. In cytes form flat cell processes, which are wrapped
a transverse section of a myelin sheath they appear as around the nerve axon in a spiral fashion (Fig. 1.8).
islands of cytoplasm between openings of the major
dense lines.
There is considerable variation in the number of
myelin lamellae in the sheaths surrounding different
axons. Generally, the larger the diameter of the axon
the thicker its myelin sheath. In addition to this direct
relationship between axon size and myelin thickness,
the lengths of internodal segments also vary with the
size of the axon: the larger the nerve fiber, the greater
the internodal length.

1.3 Oligodendrocytes

Oligodendrocytes are the key cells in myelination of


the CNS. They are cells of moderate size with a small
number of short, branched processes. They are the
predominant type of neuroglia in white matter and
are frequently found interposed between myelinated Fig. 1.8. Diagram showing the axon being rolled in the myelin
axons. Actual connections between oligodendrocytes sheath
1.4 Astrocytes 5

Fig. 1.9. Impression of the three-


dimensional structure of oligodendro-
cytes with their plasma membrane
extensions as myelin sheaths covering
the axons that cross their region

With the exception of the outer and lateral loops of 1.4 Astrocytes
the flat cell processes, the cellular cytoplasm disap-
pears from these processes and the remaining cell Astrocyte functions have long been a subject of de-
membranes condense into a compact structure in bate. Their major role has long been thought to be a
which each membrane is closely apposed to the adja- sort of skeletal function, providing packing for other
cent one. If myelin were unrolled from the axon it CNS components. It is becoming increasingly clear
would be a flat, spade-shaped sheet surrounded by a that astrocytes are of fundamental importance in
tube containing cytoplasm. maintaining the structural and functional integrity of
Although the myelin sheath is an extension of the neural tissue.
oligodendroglial cell membrane, the chemical com- A well-known function of astrocytes is concerned
position of myelin is quite different from that of the with repair. When damage is sustained, astrocytes
oligodendroglial cell membrane. The oligoden- proliferate, become larger, and accumulate glycogen
droglial cell membrane is transformed into myelin in and filaments. This state of gliosis can be total, in
processes of modification and differentiation. which case all other elements are lost, leaving a glial
On the same axon, adjacent myelin segments be- scar, or occur against a background of regenerating or
long to different oligodendrocytes. A single oligoden- normal CNS parenchyma. Following demyelination,
drocyte provides the myelin for many internodal seg- astrocytes synthesize growth factors thought to be in-
ments of different axons simultaneously. One oligo- volved in myelin repair.Astrocytes may also phagocy-
dendrocyte can be responsible for the production and tose debris in some conditions.
maintenance of up to 40 nerve fibers (Fig. 1.9). This Astrocytes are involved in transport and in main-
has implications for disease conditions and repara- taining the blood–brain and CSF–brain barriers.
tive processes, as the destruction of even only a few End-feet of astrocytes form part of these barriers in
oligodendrocytes can have an extensive demyelinat- perivascular and subpial regions. Endothelial tight
ing effect. junctions form the primary seal of the blood–brain
Together with the Schwann cells of the PNS, oligo- barrier. The role of astrocytes in the blood–brain bar-
dendrocytes are unique in their ability to produce rier is less well defined. They are physically separated
vast amounts of a characteristic unit membrane. The from endothelial cells by the basal lamina and do not
ratio between cell body surface membrane and contribute directly to the physical barrier. Perivascu-
myelin membrane is estimated at 1:620 in the case of lar astroglial end-feet contain many transport pro-
oligodendrocytes. The deposition and maintenance teins, including transporters of monocarboxylates,
of such large expanses of membrane require optimal glucose, and glutamate, as well as water. Aquaporin-4
coordination of the synthesis of its various lipid and is the only known water channel in the brain and has
protein components and their interaction to ensure a localization in the astroglial end-feet.
production of a stable membrane on the one hand Astrocytes play a part in the process of myelin de-
and a well-regulated and controlled breakdown and position. They promote the adhesion of oligodendro-
replacement of spent components needed to support cyte processes to axons and stimulate myelin forma-
the myelin membrane on the other. tion by local secretion of different growth factors. As-
6 Chapter 1 Myelin and White Matter

trocytes and neurons are the sources of platelet-de- CNS white matter is half myelin and half non-
rived growth factor (PDGF), which promotes oligo- myelin on a dry weight basis. Owing to the high
dendrocyte progenitors to proliferate, migrate, and myelin content, white matter has a relatively low wa-
differentiate. Astrocytes release basic fibroblast ter content and a high lipid content. The water content
growth factor (bFGF), which promotes oligoden- of white matter is 72% and that of gray matter 82%.
droglial differentiation. Extension of oligodendro- The nonmyelin portion of white matter contains
cyte processes, a critical early step in myelin forma- about 80% water.
tion, is facilitated by astrocytic bFGF. Insulin-like Myelin is mainly responsible for the gross chemical
growth factor I (IGF-I), which plays a crucial role in differences between white and gray matter. Myelin is
oligodendrocyte development and myelin formation, rich in all lipid classes, although nonpolar lipids and
is produced by various cells, including astrocytes. It glycolipids (galactolipids) are particularly well re-
acts as an oligodendrocyte mitogen and a differentia- presented. The lipids of CNS myelin are composed of
tion and survival factor and is one of the main regu- 25–28% cholesterol, 27–30% galactolipid, and 40–
lators of the amount of myelin production.Astrocytes 45% phospholipid when expressed as percentages of
express neurotrophin-3 (NT-3), which promotes pro- total lipid weight. When lipid data are expressed as
liferation of oligodendroglial precursors and oligo- molar ratios, CNS myelin preparations contain cho-
dendrocyte survival. There is evidence that NT-3 in lesterol, phospholipid and galactolipid in a ratio vary-
combination with brain-derived neurotrophic factor ing between 4:3:2 and 4:4:2.
(BDNF) can induce proliferation of endogenous The biochemical composition of mature gray and
oligodendrocyte progenitors and the subsequent white matter is shown in Table 1.1. With respect to
myelination of regenerating axons. Astrocytes and white matter, separate figures are given for the myelin
oligodendrocytes communicate via gap junction-me- and nonmyelin portions, CNS white matter being half
diated contacts. myelin and half nonmyelin on a dry weight basis. In
Astrocytes have a role in the conduction of nerve Table 1.1 the lipid figures are expressed as percent-
impulses. Astrocytes and axons have an intimate rela- ages of total lipid weight. Since the water content and
tionship at the node of Ranvier. Perinodal astrocytes the dry weight lipid content of gray matter and white
and nodal parts of the axon have a high concentration matter, myelin and nonmyelin, differ widely, the fig-
of sodium channels, indicating specialization of as- ures expressed in this way give no direct information
trocyte function at these sites. about lipid concentration in either dry or wet tissue.
Synthesis of the neurotransmitters glutamate and However, from the data presented, these concentra-
GABA (gamma aminobutyric acid) can originate ei- tions can be calculated.
ther from glutamine or from α-ketoglutarate or an- When the lipid compositions of gray and white
other tricarboxylic acid cycle intermediate plus an matter are compared, the most conspicuous differ-
amino acid as a donor of the amino group. Neurons ence to emerge is that white matter is relatively rich in
lack the enzymes glutamine synthetase and pyruvate galactolipids and relatively poor in phospholipids.
carboxylase, which are present exclusively in astro- Galactolipids (galactocerebroside and sulfatide) con-
cytes. Astrocyte processes in perisynaptic regions stitute 25–30% of the lipids in white matter, whereas
take up the excitatory neurotransmitter glutamate they account for only 5–10% of those in gray matter.
from the synapse and recycle it to its precursor gluta- Phospholipids account for two-thirds of the total
mine. Therefore, astrocytes are important in the syn- lipids in gray matter, but less than half those in white
thesis and recycling of some neurotransmitters and matter. There are, strictly speaking, no myelin-specif-
protect neurons from excitotoxicity. ic lipids that are not found elsewhere in the brain.
However, the most specific distinguishing feature of
myelin lipids is the high cerebroside content, and
cerebroside can be considered the most typical
1.5 Biochemical Composition myelin lipid. During development, the concentration
of Mature Myelin and White Matter of cerebroside in brain is directly proportional to the
amount of myelin present.
The most conspicuous feature of the composition of Ethanolamine phosphoglyceride in plasmalogen
myelin as opposed to other membranes is the high ra- form (plasmenylethanolamine) is the major myelin
tio of lipid to protein. It is one of the most lipid-rich phospholipid. Approximately 80% of the ethanol-
membranes, lipids making up 70–80% lipid by dry amine phosphoglycerides of myelin and white matter
weight. In comparison with other membranes, the are present in plasmalogen form, and only a small
protein concentration of 20–30% is low. For example, proportion are formed by phosphatidylethanol-
the concentration of protein in liver cell membranes amine. Conversely, the plasmalogens, which comprise
is 60%. Myelin is a relatively dehydrated structure, nearly one-third of the total phospholipids, are main-
containing only 40% water. ly of the ethanolamine type with lesser amounts of
1.5 Biochemical Composition of Mature Myelin and White Matter 7

Table 1.1. Composition of human CNS gray matter, white matter, myelin portion and nonmyelin portion of whole white matter.
From Norton and Cammer (1984)
Gray matter White matter Myelin Nonmyelind

Watera 82 72 44 82
Total proteinb 55.3 39.0 30.0 62.2
Total lipidb 32.7 54.9 70.0 41.2
Cholesterol 22.0 27.5 27.7 14.6
Glycolipids 7.3 26.4 27.5 28.2
Cerebroside 5.4 19.8 22.7 19.9
Sulfatide 1.7 5.4 3.8 7.7
Phospholipids 69.5 45.9 43.1 51.9
Ethanolamine PG 22.7 14.9 15.6 6.8
Choline PG 26.7 12.8 11.2 16.5
Serine PG 8.7 7.9 4.8 20.4
Inositol PG 2.7 0.9 0.6 1.0
Sphingomyelin 6.9 7.7 7.9 5.6
Plasmalogensc 8.8 11.2 12.3 9.2

a
Percentage of total brain weight
b Figures for total protein and total lipid are percentages of dry weight; all others are percentages of total lipid weight
c Plasmalogens are primarily ethanolamine phosphoglycerides
d Figures for bovine brain, which are thought to be in close agreement with those for human brain (Norton and Autilio 1966)

PG phosphoglycerides

plasmenylserine. Phosphatidylcholine is the major times that in gray matter. The proteins are encoded by
choline phosphoglyceride; only traces of choline the same gene and are formed by alternative splicing
phosphoglyceride have the plasmalogen form. of the primary gene transcript. The proteins differ by
Gangliosides are minor myelin lipids and make up a hydrophilic peptide 35 amino acids in length, whose
only 0.3–0.7% of total myelin lipids. They are local- presence generates PLP. DM 20 is predominant in ear-
ized mainly in neuronal membranes, and gray matter ly development, whereas PLP is the major protein in
is 10 times as rich in gangliosides as in white matter. mature myelin. The proteins are very hydrophobic.
Gangliosides are complex sialic acids containing There are multiple isoforms of myelin basic pro-
glycosphingolipids. GM1, a monosialoganglioside, is tein (MBP), arising from different patterns of splicing
the major myelin ganglioside accounting for about of the primary gene transcript. The heterogeneity is
70 mol % of the total myelin ganglioside content. increased further by various posttranslational modi-
Within the CNS, the ganglioside GM4 (sialogalacto- fications. Myelin basic proteins account for 30–35%
sylceramide) is probably specific for myelin and of the total myelin protein. MBP contains no extensive
oligodendroglia. It is a derivative of cerebroside. regions of hydrophobic residues and is hydrophilic.
Myelin lipids contain somewhat different fatty acid MBP is the antigen which, when injected into an ani-
constituents than other membranes. Characteristic of mal, elicits a cellular immune response, producing the
myelin are α-hydroxy fatty acids in cerebrosides and CNS autoimmune disease called experimental aller-
sulfatides and high amounts of long-chain fatty acids gic encephalomyelitis.
in the different lipid classes. There are monounsatu- There are several CNS myelin glycoproteins:
rated fatty acids, but only low amounts of polyunsat- myelin-associated glycoprotein (MAG), myelin/oligo-
urated fatty acids. dendrocyte glycoprotein (MOG), and oligodendro-
Table 1.2 shows the chemical structures of the cyte-myelin glycoprotein (OMgp). These are high-
main lipid constituents of myelin. molecular-weight proteins. They are quantitatively
The protein composition of myelin is simpler than minor myelin components: MAG accounts for about
that of other membranes. Proteolipid protein and 1% of total protein and MOG, for 0.05%.
myelin basic protein encompass approximately 60– Other minor myelin proteins are oligodendrocyte-
80% of the total protein. Most myelin proteins are specific protein (OSP), which is a tight junction
unique to myelin. protein, myelin-associated oligodendrocytic basic
Proteolipid protein (PLP) and its isoform DM 20 protein (MOBP), a small basic protein distributed
make up about 50% of the total protein in CNS throughout compact myelin, and myelin/oligoden-
myelin. Their concentration in white matter is about 5 drocyte-specific protein (MOSP), which is located on
8 Chapter 1 Myelin and White Matter

Table 1.2. Structure of the important myelin lipids


Cerebroside sphingosine galactose

fatty acid

Sulfatide sphingosine galactose sulfate

fatty acid

Phosphatidylethanolamine fatty acid


glycerol fatty acid
phosphate ethanolamine

Phosphatidylcholine = lecithin fatty acid


glycerol fatty acid
phosphate choline

Phosphatidylserine fatty acid


glycerol fatty acid
phosphate serine

Phosphatidylinositol fatty acid


glycerol fatty acid
phosphate inositol

Ethanolamine plasmalogens *fatty acid


glycerol fatty acid
phosphate ethanolamine

Sphingomyelin sphingosine fatty acid

phosphate choline

GM3 ganglioside N-acylsphingosine

glucose

galactose N-acetylneuraminic acid

GM2 ganglioside N-acylsphingosine

glucose
galactose N-acetylneuraminic acid

N-acetylgalactosamine

GM1 ganglioside N-acylsphingosine

glucose

galactose N-acetylneuraminic acid

N-acetylgalactosamine

galactose

Sphingolipids of myelin are formed from sphingosine. N-acylsphingosine is termed ceramide. A phosphorylcholine group at-
tached to ceramide forms sphingomyelin; glucose or galactose in glycosidic linkage forms cerebroside (most often: galactosylce-
ramide). When the glucose or galactose is esterified with sulfate, sulfatide is formed. Phosphoglycerides contain two fatty acids
in ester linkage at the α and β position of glycerol and at the α’ position a phosphate group to which the moiety definitive of the
class is linked. For example, a choline group defines phosphatidylcholine. The plasmalogens are similarly formed, except that at
the α position of the glycerol there is a 1:2 unsaturated ether structure (*). Gangliosides are synthesized from N-acylsphingosine
by stepwise addition of sugars and N-acetylneuraminic acid.
1.6 Molecular Architecture of Myelin 9

the extracellular surface of oligodendrocytes and (four transmembrane domains). The hydrophobic
myelin. transmembrane segments are linked by hydrophilic
Highly purified myelin contains a number of en- portions on both sides of the membrane. This means
zymes. Two of these enzymes, 2’,3’-cyclic nucleotide that the protein has domains in both the intraperiod
3’-phosphodiesterase (CNP) and a cholesterol ester and the major dense lines. Probably both isoforms,
hydrolase, are found at much higher specific activities PLP and DM 20, are involved in stabilizing the in-
in myelin than in brain homogenates. It appears traperiod line. Their role is described as that of ‘ad-
that these enzymes are fairly myelin specific, and are hesive struts’ or ‘spacers,’ maintaining a set distance
probably also present in oligodendroglial mem- between apposed lamellae. DM 20 is the major proteo-
branes. Many other enzymes are found that are not lipid protein in early development, whereas PLP is
specific to myelin but also present in other brain frac- the major product in mature myelin. It is believed that
tions. The exact function of the two enzymes is not DM 20 has a still unidentified regulatory role in early
known. In particular, their contribution to the metab- oligodendrocyte progenitor development and differ-
olism of myelin constituents is not known. CNP cat- entiation, and that PLP plays a part later on in oligo-
alyzes the hydrolysis of several 2’,3’-cyclic nucleotide dendrocyte function, in the proper formation of the
monophosphates, all of which are converted to the intraperiod line of myelin during its final elaboration
corresponding 2’-isomer. The substrates of the en- and compaction.
zyme are not present in nervous tissue. CNP is one of Myelin basic protein is an extrinsic protein located
the proteins formerly called Wolfgram proteins, a het- on the cytoplasmic face of the myelin membranes at
erogeneous group of high-molecular-weight myelin the major dense lines. It probably stabilizes the major
proteins named after the investigator who first sug- dense lines by keeping the cytoplasmic faces of the
gested that myelin contained proteins other than pro- myelin lamellae in close apposition. Myelin-associat-
teolipid protein and myelin basic protein. ed oligodendrocytic basic protein is another small
basic protein distributed throughout compact myelin
at the major dense lines. There is evidence that MOBP
1.6 Molecular Architecture of Myelin reinforces the apposition of the cytoplasmic faces of
the myelin sheath.
The currently accepted view of the myelin structure is Gangliosides are located almost entirely on the
that of a double lipid bilayer, each coated on both external surface of membranes. They may have an im-
sides with protein. The resulting repeating subunit portant role in cell surface recognition and signal
consists of radial protein-lipid-protein-lipid-protein transduction processes such as those that occur dur-
lamellae. Some proteins are fully or partially embed- ing myelination.
ded in the bilayer, and others are attached to the sur- Myelin glycoproteins are transmembrane proteins
face by weaker linkages. with the polypeptide extending through the lipid bi-
Both proteins and lipids have an asymmetrical dis- layer and the glycosylated portion of the molecule ex-
tribution. Galactolipids, cholesterol, phosphatidyl- posed on the outer surface of the bilayer. They are all
choline, and sphingomyelin are preferentially located implicated in recognition and cell–cell interactions.
in the former extracellular half of the bilayer (in- MAG is one of these proteins. Its external region
traperiod line). Ethanolamine plasmalogen and contains immunoglobulin-like domains. Thus, MAG
myelin basic protein are preferentially located in the is a member of the immunoglobulin superfamily. It is
former cytoplasmic half of the bilayer. concentrated in the inner periaxonal membrane of
Membranes are fluid structures. Lipid molecules the myelin sheath and absent from the compact mul-
diffuse rapidly in the plane of the membrane, as do tilamellar myelin sheath. The exposed, periaxonal po-
proteins, unless anchored by specific interactions. sition is compatible with its postulated involvement
The spontaneous rotation of lipids from one side of in oligodendrocyte–axon interaction, including
the membrane to the other is a very slow process. The maintenance of the structural integrity of the glia–
transition of a molecule from one membrane surface axon adhesion in mature myelin. The observation
to the other is called transverse diffusion, or flip-flop. that the protein can be detected at the very earliest
In view of the asymmetry of lipids in the bilayer, the stages of myelination has led to the hypothesis that
transverse mobility must be limited. The diffusion the protein may also play a role in mediating the
within the plane of the membrane is referred to as oligodendrocyte-axon recognition events that pre-
lateral diffusion. cede myelination and specify the initial path of
Proteolipid protein consists of alternating hydro- myelin deposition.
philic and hydrophobic sequences with four stretches MOG is another of the myelin glycoproteins. It
of hydrophobic residues that are of sufficient length also belongs to the immunoglobulin superfamily. The
to span the lipid bilayer. It is an integral membrane protein is located at the outermost layer of the myelin
protein that passes through the bilayer four times sheath and the oligodendrocyte plasma membrane.
10 Chapter 1 Myelin and White Matter

The function of this glycoprotein is unknown. It may quence, substances transported to the plasma mem-
be involved in the adhesion between neighboring brane via vesicles end up in the extracellular leaflet of
myelinated fibers and function as glue in the mainte- the myelin sheath. MAG resembles proteolipid pro-
nance of axon bundles in the CNS. MOG may also be tein as far as the site of synthesis and transport to the
a cell surface receptor that transduces signals from plasma membrane are concerned.
the external milieu to the inside of the oligodendro- The same two mechanisms of synthesis and trans-
cyte or myelin sheath. port can be distinguished for myelin lipids, i.e., the
The enzyme CNP is found in myelin and oligoden- routes of PLP and MBP, respectively. The endoplasmic
drocytes. Within the myelin sheath it is localized reticulum is the site of synthesis of phosphatidyl-
on the cytoplasmic side of noncompact regions, e.g., choline and cholesterol. The Golgi apparatus is the
periaxonally and in the paranodal loops. CNP is site of synthesis of cerebroside, sulfatide, sphin-
essential for axonal survival but not for myelin as- gomyelin, and gangliosides. The lipids are transport-
sembly. ed from the Golgi apparatus to the plasma membrane
by a vesicle-mediated process. Expression on the cell
surface occurs by fusion of the vesicles with the plas-
1.7 Myelinogenesis ma membrane. The lipids are located predominantly
in the extracellular leaflet of the myelin lamellae. In
The time-course of the appearance of newly synthe- contrast, the myelin phospholipids that predominant-
sized lipids and proteins in myelin indicates that ly reside on the inner leaflet, including phos-
myelin is not laid down as a unit. Different compo- phatidylserine and ethanolamine plasmalogens, are
nents are synthesized and processed in different cel- synthesized in the superficial cytoplasmic channels
lular compartments, are transported to the sites of of the myelin sheath and rapidly enter compact
myelin formation by different mechanisms, and show myelin, possibly with phospholipid transfer proteins
different rates of entry into the myelin sheath. For ex- as carriers. Several other phospholipids are also syn-
ample, MBP enters the myelin sheath with almost no thesized in the superficial cytoplasmic channels.
lag after synthesis, whereas proteolipid protein enters After reaching the outermost myelin layers, sub-
myelin with a lag-time of 30–40 min following syn- stances penetrate to the deepest layers over a period
thesis. Once protein synthesis is stopped with cyclo- of a few days. This movement of substances from out-
heximide, the entry of MBP is halted immediately, but er to inner layers occurs at rates consistent with later-
proteolipid protein continues to be incorporated into al diffusion along the spirally wound bilayer.
myelin for 30 min. These data indicate that MBP and
PLP are assembled by different mechanisms, with
PLP taking a longer and more circuitous route 1.8 Regulation
through the cytoplasm. Lipids also continue to be in- of Myelinogenesis
corporated into myelin for 4 h after protein synthesis
has stopped. Elaboration of the myelin sheath involves a precisely
MBP is synthesized on free polyribosomes near ordered sequence of events beginning with the initial
the plasma membrane or the adjacent myelin sheath. ensheathment of the axon, proceeding to formation
The myelin membrane is surrounded by and infiltrat- of multiple loose wrappings and eventually com-
ed with cytoplasmic channels, called the outer loops paction to form the mature multilamellar myelin
and longitudinal incisures of Schmidt-Lantermann, sheath. These processes imply a temporally regulated
respectively. Myelin basic protein mRNA is trans- program of gene expression in the oligodendrocyte
located from the nucleus to the myelin membrane via to ensure that the appropriate biochemical compo-
these cytoplasmic channels. MBP synthesized here is nents are synthesized in the appropriate proportions
rapidly sequestered into the myelin sheath and ap- at each stage of myelinogenesis. Just before the onset
pears in the cytoplasmic leaflet of compact myelin of rapid myelin membrane synthesis the expression
(major dense lines). mRNAs for several other myelin of genes of myelin proteins is sharply up-regulat-
proteins follow similar trafficking pathways. ed. There is evidence of a coordinated mechanism
Proteolipid protein and DM 20 are synthesized on for synchronous activation of the myelin protein
polyribosomes bound to the endoplasmic reticulum. genes. This period of sharp up-regulation of ex-
The nascent protein is inserted into the endoplasmic pression of myelin genes is the most vulnerable part
reticulum and passes through the Golgi apparatus to of the myelination process and is called the critical
the plasma membrane and myelin sheath via vesicu- period.
lar transport. Inclusion in the plasma membrane Apparently, there are both tissue-specific and
occurs by fusion of the vesicles with the plasma mem- stage-specific mechanisms controlling myelin genes.
brane. The inside of the vesicle after fusion becomes Myelin genes are only expressed in oligodendrocytes
the outside of the plasma membrane. As a conse- and Schwann cells. The expression of the genes is de-
1.8 Regulation of Myelinogenesis 11

velopmentally regulated and is probably intimately ticularly and disproportionately in the amount of
associated with the stage of differentiation of these myelin produced per oligodendrocyte. Thyroid hor-
cells. Control mechanisms are active at the transcrip- mone also has an effect on myelinogenesis. Hypo-
tional level. Regulatory regions, including the pro- thyroidism during early development leads to hy-
moter regions, have been identified for myelin pro- pomyelination, whereas hyperthyroidism accelerates
tein genes. Key sites for tissue-specific expression of myelination. Steroids have a complex influence. None
myelin proteins are clustered near the promoter re- of the myelin protein genes is transcriptionally regu-
gions, and within these clusters are several motifs that lated by steroids, but steroids probably act at the post-
may be involved in coordinating the regulation of translational level, stimulating the translation of MBP
myelin-specific genes. The alternative splicing pat- and PLP mRNAs and inhibiting the translation of
terns produced from the primary myelin protein CNP mRNA.
transcripts are also developmentally regulated. The The importance of iron in myelination has been
splicing patterns for the different proteins have been examined. Iron and the iron mobilization protein
shown to change in the course of development. transferrin are localized in oligodendrocytes, and
In both the CNS and the PNS, glial cells are influ- may participate in the formation and/or maintenance
enced to produce myelin by both neuronal targets of myelin by complexing with enzymes involved in
that they ensheathe and by a range of hormones and the synthesis of myelin components.
growth factors produced by neurons and astrocytes. Myelination is vulnerable to undernourishment. If
There is a continuous oligodendrocyte-neuron-astro- there is undernourishment during the critical period
cyte interaction in the process of myelination and just prior to the onset of rapid myelin synthesis,
myelin maintenance. myelination is more severely reduced than total brain
Proliferation of oligodendrocyte precursor cells weight, whereas the number of oligodendrocytes is
depends on electrical activity of neurons. Oligoden- unaltered. The hypomyelination is permanent. Severe
drocyte number is also dependent on number of undernutrition during the critical period leads to de-
axons. Differentiation of oligodendroglia has been creased levels of IGFs and a failure in up-regulation of
shown to depend heavily on the presence and the myelin genes.
integrity of axons. Gene expression for myelin con- Successful myelination is also dependent on func-
stituents is modulated by the presence of axons. tion. It is known that myelination is diminished by
Within oligodendrocytes, proteins are produced that preventing the conduction of impulses in a nerve. Im-
are thought to be involved in the induction of myeli- pulse conduction is a stimulus to myelination. Prema-
nation (e.g., glia-specific surface receptors for differ- ture activity accelerates myelination. Hypermyelina-
entiation signals), in the initial deposition of the tion has incidentally been noticed in cerebral anom-
myelin sheath (e.g., axon-glial adhesion molecules), alies, supposedly via the stimulus of epilepsy. It has
and in its wrapping and compaction around the nerve been shown that oligodendrocyte progenitor cells
axon (e.g., structural proteins of compact myelin). A express adenosine receptors, which are activated in
minimal axonal diameter is important for the initia- response to action potential firing. Action potential
tion of myelination. Final myelin sheath thickness is firing leads to the nonsynaptic release of several sub-
also related to axonal size. This match is reached by stances from axons, including ATP and adenosine.
local control mechanisms. Therefore, a single oligo- Adenosine acts as a potent neuroglial transmitter to
dendrocyte can be associated with several axons of inhibit oligodendrocyte progenitor cell proliferation,
different sizes, the myelin sheaths being thicker for stimulate differentiation, and promote the formation
larger axons. Larger axons also have longer inter- of myelin.
nodes. After formation the myelin sheath and the axon re-
Astrocytes are essential in the process of myelina- main mutually dependent. The myelin sheath needs
tion and myelin maintenance. They produce trophic an intact axon, as demonstrated by the studies on wal-
factors, including PDGF, bFGF, IGF-I, and NT-3. These lerian degeneration. On the other hand, for mainte-
factors promote proliferation, migration and differ- nance of the normal structure and function the axon
entiation of oligodendrocyte progenitors, extension requires an intact myelin sheath. Normal astrocytes
of oligodendrocyte processes, adhesion of oligoden- are essential for an intact myelin-axon unit.
drocyte processes to axons, myelin formation and Since myelin, once deposited, is a relatively stable
myelin maintenance. substance metabolically, it is relatively invulnerable
Hormones have a dramatic effect on myelinogene- to adverse external factors. Generalized vulnerability
sis. A deficiency of growth hormone during the criti- of myelin to noxious agents and adverse influences is
cal period leads to hypomyelination. Most of the ef- likely to be confined to the period just before and dur-
fects of growth hormone are mediated by IGF-I. Ad- ing active myelination.
ministration of this substance in early development
leads to an increase in all brain constituents, but par-
12 Chapter 1 Myelin and White Matter

1.9 Myelination of the Nervous System In the 6th month of gestation myelination pro-
ceeds rapidly cephalad in the medial lemniscus and
Myelination of each of the multiple connecting fiber spinothalamic tracts in the brain stem tegmentum.
systems of the CNS takes place at a different time in Myelin begins to appear in the statoacoustic tectum
early development. Some fiber systems start to myeli- and tegmentum and the lateral lemniscus for the con-
nate halfway through gestation or later and rapidly duction of acoustic stimuli. Myelin is seen in the in-
attain their maximal degree of myelination, whereas ner, vestibulocerebellar part of the inferior cerebellar
other systems attain their maximal degree of myeli- peduncle.
nation only slowly. It is, therefore, not correct to refer In the 7th month of gestation myelination is still
to myelination as a singular process. There is a largely confined to structures outside the dien-
marked, temporal diversity in topographic patterns cephalon and cerebral hemispheres. Progress of
of myelination throughout the last half of gestation myelination is seen in the optic nerve, optic chiasm
and during the first 2 postnatal years. Thus, at any and tracts, inferior cerebellar peduncle, the parasagit-
time in the early development of the human brain tal part of the cerebellum, the descending trigeminal
there are multiple separate or intermixed regions tract, superior cerebellar peduncle, capsule of the red
of unmyelinated, partly myelinated, or completely nucleus, capsule of the inferior olivary nucleus,
myelinated tracts. vestibulospinal, reticulospinal and tectospinal de-
Myelination of the nervous system follows a fixed scending tracts to the spinal cord and posterior limb
pattern consisting of ordered sequences of myelinat- of the internal capsule.
ing systems apparently governed by some rules: In the eighth month of gestation, myelination
1. The first rule, probably governing all other rules, starts in the corpus striatum (in particular globus
is that tracts in the nervous system become myeli- pallidus), anterior limb of the internal capsule, sub-
nated at the time they become functional. cortical white matter of the post- and precentral gyri,
2. Most tracts become myelinated in the direction of rostral part of the optic radiation as well as corti-
the impulse conduction. cospinal tracts in midbrain and pons, transpontine
3. Myelination starts in the PNS before it starts in the fibers, middle cerebellar peduncles and cerebellar
CNS. hemispheres.
4. Myelination in central sensory areas tends to pre- In the ninth month of gestation, myelination con-
cede myelination in central motor areas. tinues in the thalamus (in particular ventrolateral nu-
5. Myelination in the brain occurs earlier in areas of cleus), putamen, central part of the corona radiata,
primary function than in association areas. distal part of the optic radiation, acoustic radiation,
6. Roughly speaking, myelination progresses from anterior commissure, midportion of the corpus callo-
caudal (spinal cord) to rostral parts (brain) and sum and fornix.
spreads from central (diencephalon, pre- and However, in a child born at term, most of the struc-
postcentral gyri) to peripheral parts of the brain. tures and tracts mentioned are not fully myelinated
However, there are many exceptions to this rule. and, in fact, in some myelination has just started.
Apart from some myelin in the central tracts of the
It is important to note that the times mentioned be- corona radiata connected with the pre- and postcen-
low for myelination of the different tracts and struc- tral gyri, and the primary optic and acoustic radia-
tures of the brain are only generalizations and ap- tions, the cerebral hemispheres are still largely un-
proximations. In the first place, there is a considerable myelinated. During the first postnatal year, myelin
degree of normal variation. Secondly, the onset of spreads throughout the entire brain. By the postnatal
myelination is difficult to define. It can be defined as age of 12 weeks myelination is well advanced in the
the first myelin tube found on light microscopic corona radiata, the optic radiation and the corpus cal-
examination, as the appearance of the first myelin losum, but the frontal and temporal white matter are
lamella on ultrastructural examination, or as the first still largely unmyelinated. By the age of about 8
evidence of the presence of myelin constituents in im- months, the adult state is foreshadowed in that none
munological investigations. of the fiber systems is still completely devoid of
In the 4th month of gestation myelin is first seen in myelin sheaths. Myelin sheaths are still sparse in the
the anterior motor roots and soon appears in the pos- temporal and frontal areas. It is not until the end of
terior roots. the second postnatal year that an advanced state of
In the 5th month of gestation myelination starts in myelination is seen in all subcortical areas. Histologi-
the dorsal columns of the spinal cord and the anteri- cally, myelination reaches completion in early adult-
or and lateral spinothalamic tracts for conduction of hood.
somatesthetic stimuli.
1.10 Compositional Changes in the Developing Brain 13

1.10 Compositional Changes ter at different ages is shown in Table 1.3. A major
in the Developing Brain change is an increase in total lipid content, with a rel-
ative increase in glycolipids. One of these, cerebro-
The DNA content of brain is considered to be a reli- side, is usually considered to be a marker for myelin
able indicator of cell number. The period of cellular as it is deposited at the same rate in the brain as
proliferation can, therefore, be followed by measuring myelin. However, cerebroside is not restricted to
the amount of DNA per brain volume. In human brain myelin and as much as 30% of it may be present in
two major periods of cell proliferation have been de- membranes other than myelin. There is a relative de-
tected by measuring DNA levels. The first period be- crease (but absolute increase) in phospholipids in the
gins at 15–20 weeks of gestation and corresponds to white matter, which were relatively high in concentra-
neuroblast proliferation. The second period begins at tion in unmyelinated white matter and are relatively
25 weeks of gestation and continues into the 2nd year low in concentration in myelin. The relative contribu-
of postnatal life. This latter period corresponds to tion of cholesterol to total lipids remains constant,
multiplication of glial cells and includes a second but the absolute cholesterol content of white matter
wave of neuronogenesis, producing mainly cerebellar increases with deposition of myelin. The changes in
neurons. gray matter composition are much less important.
The ratio of protein to DNA indicates cell size. This Myelin deposition in gray matter is minor.
ratio increases after neuronal division ends, reflecting The changes in white matter composition are not
in part the arborization of neuronal processes. The caused only by glial cell proliferation, growth of axons
maximum ratio of protein to DNA is reached at and dendrites, and myelin deposition, but also by
2 years of age. some changes in myelin composition. The composi-
The outgrowth of neuronal axons and dendrites tion of the myelin first deposited is somewhat differ-
results in a rapid increase in total ganglioside content ent from that in adults. The most important changes
in the brain. Increasing lipid content indicates mem- are an increase in cholesterol and glycolipids as a pro-
brane formation with, in particular, an increase in portion of total lipid and a decrease in phospholipids.
quantity of axonal, dendritic, and myelin membranes. In the immature brain significant amounts of glucose
The increasing lipid content is associated with a con- are present in the glycolipids, whereas in a mature
comitant decrease in water content. The most rapid brain glycolipids are present mainly as galactolipids.
increase in lipid content of the brain begins after the In contrast to the modest decrease in total phospho-
period of greatest increase of DNA and protein and is lipids, more marked variations in the relative contri-
closely related to the onset of myelination. bution of individual phospholipids are found. Sphin-
At birth, cerebral hemispheric white matter con- gomyelin and ethanolamine phosphoglycerides in-
tains very little myelin and the white matter composi- crease, whereas choline phosphoglycerides decline.
tion of neonates is very different from the composi- The molar ratio of galactolipids and choline phos-
tion of mature myelinated white matter. There is an phoglycerides appears to be a sensitive marker of
important overall decrease in water content of the myelin maturation.
brain after birth and the change in water content is In human unmyelinated white matter much of the
larger for white matter than for gray matter. The wa- cholesterol present is esterified. The same is true for
ter content of neonatal gray matter is about 89% and cholesterol in newly formed myelin. During myelin
of neonatal unmyelinated white matter about 87%, maturation there is a decrease in the amount of cho-
whereas the water content of adult gray matter is esti- lesterol esters, and in adult white matter cholesterol is
mated to be 82% and of adult myelinated white mat- present almost entirely in the free form. The ratio of
ter 72%. The lipid composition of cerebral white mat- cholesterol to phospholipids in myelin increases after

Table 1.3. Lipid composition of human brain during development. From Svennerholm (1963)
Lipid composition of (frontal) cerebral cortex Lipid composition of (frontal) cerebral white matter

Age 2 months 1 year 5 years 2 months 1 year 5 years


Total lipidsa 28.4 31.3 29.5 29.5 49.6 58.2
Cholesterolb 21.5 19.8 19.3 26.4 25.0 24.4
b
Phospholipids 76.8 7.6 75.6 66.1 53.4 49.8
Glycolipidsb 1.8 2.6 4.1 7.5 21.6 25.8
a Expressed as percentage of dry weight
b Expressed as percentage of total lipid weight
14 Chapter 1 Myelin and White Matter

birth and reaches the adult value at about 5 years of 1.12 Aging of Myelin
age. The ratio of galactolipids to phospholipids reach-
es the adult value at about the same time. With increasing age, human brain weight decreases
During development the ganglioside composition and water content increases. Levels of DNA and num-
of myelin becomes simplified. The polysialoganglio- bers of neurons in the cerebral cortex decrease signif-
sides decline and the monosialoganglioside GM1 con- icantly with aging. Little change is found in some re-
tent approaches about 90% of the total gangliosides gions, including the brain stem.
with increasing age. The total ganglioside content Multiple morphological changes take place with
remains constant. increasing age. The most prominent neuronal
Maturation of myelin is accompanied by an in- changes are the appearance of senile plaques (areas of
crease in hydroxy fatty acids and saturated and mo- degenerating neuronal processes, reactive nonneu-
nounsaturated fatty acids. ronal cells, and amyloid), increasing deposits of lipo-
Maturation of myelin is also accompanied by fuscin, and areas of neurofibrillary tangles. Synapses
changes in the proteins. As the brain matures there is and dendrites are lost with aging. Neurotransmitter
a change in occurrence of the major isoforms of the systems are also affected by aging. Acetylcholin-
major myelin proteins. For example, initially, early in esterase, choline acyltransferase, tyrosine hydroxy-
myelination, DM 20 is the principal isoform, whereas lase, DOPA decarboxylase, and glutamic acid de-
in adult brain DM 20 is present at much lower levels carboxylase, enzymes involved in cholinergic, and
than the isoform PLP. With advancing development dopaminergic and GABA-ergic transmission, respec-
the contribution of PLP and MBP to myelin proteins tively, show appreciable decreases.
shows a relative increase, whereas the high-molecu- The total myelin content of white matter is reduced
lar-weight proteins decrease. in old age. Low myelin concentrations of white matter
On the whole, the differences in chemical compo- most probably reflect the continuous loss of neurons
sition of immature myelin and adult myelin are not with degeneration of axons and of the myelin sheaths.
striking, which suggests that only subtle remodeling The lipid composition of myelin is quite constant dur-
of myelin occurs in humans once myelination has ing aging, with the possible exception of galac-
started. The major difference between white matter tolipids, which tend to decline. Some differences are
early in life and in adult life seems to be the quantity seen in the fatty acid composition of myelin phospho-
of myelin rather than its quality. glycerides and cerebrosides during aging. Myelin pro-
teins do not undergo distinct quantitative changes in
their relative proportions during old age.
1.11 Myelin Turnover

The principal features of myelin metabolism are its 1.13 Function of Myelin
high rate of synthesis during the active stages of
myelination, when each oligodendroglial cell makes Nerve fibers transmit information to other nerve
more than three times its own weight of myelin per fibers and to receptors of effector organs. The infor-
day, and its relative metabolic stability after the com- mation is transmitted via an electric impulse called
pletion of myelination. Individual components turn the action potential, which is conducted in an all-or-
over at quite different rates. There are conflicting da- none way, i.e., the impulse is propagated or not. More
ta about the precise half-lives of the various myelin detailed information is provided by temporal and
lipids and proteins. This is understandable, since spatial summation of many action potentials within
there are several variables in the experimental design one nerve. Myelin plays an important role in the im-
that have considerable influence on the observed, pulse propagation. It is an insulator, but more impor-
real or apparent, half-lives. However, some general tant is its function to facilitate conduction in axons.
conclusions can be formulated. The concept of rela- In a resting nerve fiber, polarization of the mem-
tive long-term metabolic stability of most myelin brane exists: the inside is charged negatively com-
components has been confirmed. Some components pared with the outside. In an excited area the situa-
do turn over much faster than others, and all compo- tion is reversed: the inside is charged positively com-
nents show both a slow- and a fast-turnover compo- pared with the outside. This is called membrane de-
nent. The data indicate that newly formed myelin is polarization. There is a difference in potential
catabolized faster than old myelin. Hence, myelin that between excited and adjacent resting fiber sections
has been deposited early in life appears to have a owing to the inversion of polarization in the excited
higher metabolic stability than newly synthesized area. In an effort to compensate this difference in po-
myelin. tential, local circuits of currents flow into the active
region of the axonal membrane through the axon and
out through the adjacent, polarized sections of the
1.14 Myelin Disorders: Definitions 15

Fig. 1.10. Because of the myelin


sheath, the conduction in a myelinated
nerve fiber is saltatory, jumping from
node to node

membrane. These local circuits depolarize the adja- and more like unmyelinated fibers while with longer
cent section of the membrane. As soon as this depo- internodal distances the current density at the next
larization reaches the threshold of excitation, an ac- node of Ranvier becomes smaller. Consequently,
tion potential arises. These local circuits depolarize there is an optimal ratio of internode distance to
the adjacent section of membrane in continuous se- axon diameter. With increasing temperature conduc-
quential fashion. Of course, the local circuits do not tion velocity increases, reaching a maximum at about
only flow in the direction of the impulse conduction. 42 °C and decreasing thereafter.
However, they cause no renewed excitation in the
membrane that has just been excited because a tem-
porary state of inexcitability, called the refractory pe- 1.14 Myelin Disorders: Definitions
riod exists, which ensures that the fiber conducts the
action potential in one direction and does not remain ‘Demyelination’ means, literally: loss of myelin and
permanently excited. In unmyelinated fibers impuls- the literal interpretation of ‘demyelinating disorders’
es are propagated in this way, and the entire mem- is: disorders characterized by loss of myelin. The term
brane surface needs to be successively excited when demyelination is commonly used to indicate the
an action potential travels along it. process of losing myelin, which is caused by primary
In myelinated fibers, the excitable axonal mem- involvement of oligodendroglia or myelin mem-
brane is only exposed to the extracellular space at the branes. Myelin loss that is secondary to axonal loss
nodes of Ranvier. In the area of the node of Ranvier, and simultaneous loss of axons and myelin sheaths is
the axon is rich in sodium channels. The remainder of not usually included under the heading of demyelina-
the axolemma is covered by the myelin sheath, which tion.
has a much higher resistance and much lower capac- However, there is considerable confusion about the
itance than the axonal membrane. When the mem- meaning of the terms demyelination and demyelinat-
brane at the node is excited, the local circuit generat- ing disorders. Sometimes demyelination is used to
ed cannot flow through the high-resistance sheath, mean all conditions in which loss of myelin occurs,
and therefore flows out through the next node of Ran- irrespective of whether the myelin membrane was
vier and depolarizes the membrane there (Fig. 1.10). primarily affected or was broken down secondary to
In this so-called saltatory conduction, the impulse or at the same time as axonal loss. This is probably
jumps from node to node, whereby the conduction partly because it is not always clear whether the loss
velocity is considerably increased. Saltatory nerve of myelin is primary or secondary in nature. The mu-
conduction is not only faster, but it also saves energy tual dependence of axons and myelin sheaths is an
because only parts of the membrane need to depolar- important factor in this respect. Demyelination will
ize and repolarize for impulse conduction. For con- eventually lead to axonal loss, and in the end axonal
duction velocities in unmyelinated fibers equivalent degeneration will lead to loss of myelin. Hence, using
to those in the fastest conducting myelinating fibers, histological examination it may be very difficult to
impossibly large unmyelinated fibers and energy ex- differentiate between primary and secondary myelin
penditures several orders of magnitude greater would loss. Another confusing factor is that some disorders
be required. show evidence of simultaneous primary neuronal de-
There are several factors that influence conduction generation and primary demyelination. The random
velocity. Conduction velocity increases with increas- use of related terms, such as dysmyelination, myelino-
ing fiber diameter as a consequence of the smaller in- clastic disorders, white matter disorders, leuko-
ternal resistance, leading to an increased flow of cur- encephalopathies and leukodystrophies add to the
rent and thus shortening the time necessary for the confusion.
excitation of the adjacent membrane section or the Poser (1957) introduced the concept of ‘dysmyeli-
next node of Ranvier. Increase in myelin thickness, nation.’ He proposed dividing the disorders charac-
which accompanies increase in fiber diameter, also terized by primary myelin loss into ‘myelinoclastic
increases conduction velocity, mainly as the result of disorders’ and ‘dysmyelinating disorders’ (1961,
a change in myelin sheath capacitance. The intern- 1978). He considered the myelinoclastic disorders to
odal distance influences conduction velocity. With be the true demyelinating disorders, in which the
shorter internodal distances, the fibers behave more myelin sheath is destroyed after having been normal-
16 Chapter 1 Myelin and White Matter

ly constituted. Examples are multiple sclerosis and myelin membrane itself. ‘Demyelinating disor-
acute disseminated encephalomyelitis. The dysmyeli- ders’ are conditions characterized by demyelina-
nating disorders comprise those disorders in which tion. Examples: metachromatic leukodystrophy,
“myelin is not formed properly, or in which myelin multiple sclerosis.
formation is delayed or arrested, or in which the – ‘Hypomyelination’ is reserved for conditions with
maintenance of already formed myelin is disturbed.” a significant permanent deficit in myelin deposit-
Examples are metachromatic leukodystrophy and ed. The most extreme variant of hypomyelination
adrenoleukodystrophy. The idea behind the concept is amyelination. Example: Pelizaeus-Merzbacher
of dysmyelinating and myelinoclastic disorders is to disease.
distinguish between inherited disorders, especially – ‘Dysmyelination’, as the literal translation of the
inborn errors of metabolism, leading to disturbed name implies, is reserved for conditions in which
myelination and myelin loss, and acquired disorders the process of myelination is disturbed, leading to
characterized by primary myelin loss. However, the abnormal, patchy, irregular myelination, some-
definition of dysmyelinating disorders, as formulated times but not necessarily combined with myelin
by Poser, does not exclude all acquired disorders. loss. Examples: some amino acidopathies, dam-
There are many conditions characterized by a distur- aged structure of unmyelinated white matter after
bance of myelination, and most of these are caused by perinatal hypoxia or encephalitis.
external factors. Moreover, myelin may have been – ‘Retarded myelination’ is reserved for disorders
constituted normally in inherited disorders, only to in which the deposition of myelin is delayed,
be lost after many years. but progressing. Examples: inborn errors of me-
There are several definitions of the term ‘leukody- tabolism with early onset, malnutrition, hydro-
strophy.’ Seitelberger (1984) defines leukodystrophies cephalus.
as degenerative demyelinating processes caused by – ‘Myelin disorders’ comprise all the above-men-
metabolic disorders. Morell and Wiesmann (1984) tioned conditions.
state that leukodystrophies are disorders affecting – ‘White matter disorders’ and ‘leukoencephalo-
primarily oligodendroglial cells or myelin. The disor- pathies’ can be defined as all conditions in which
ders have to be of endogenous origin with a pattern predominantly or exclusively white matter is af-
compatible with genetic transfer of a metabolic fected. Either myelin or a combination of myelin
defect. The clinical criterion is a steadily progressive and other white matter components is involved.
deterioration of function. Menkes (1990) defines Hence, white matter disorders comprise all myelin
leukodystrophies as a group of genetically transmit- disorders, but also, for instance, white matter infec-
ted diseases in which abnormal metabolism of myelin tions and infarctions, which may affect various
constituents leads to progressive demyelination. white matter components nonselectively.
Common concepts in these definitions are demyeli- – ‘Gray matter disorders’ comprise all disorders in
nation and inborn errors of metabolism. Heritability which neurons and axons are predominantly or
is implied. As such, the leukodystrophies are identical exclusively affected.
with inherited demyelinating disorders.
The terms ‘white matter disorders’ and ‘leukoen-
cephalopathies’ comprise all disorders that selective- 1.15 Levels of Myelin Involvement
ly or predominantly involve the white matter of the
CNS, irrespective of the underlying pathophysiologic Both inherited and acquired myelin disorders can
mechanism and histopathologic basis. ‘White matter arise at the level of the myelin membranes or the
disorders’ is a literal translation of leukoencephalo- oligodendroglial cells. As a consequence, the process-
pathies. Sometimes these terms are used as if they are es of myelin build-up, maintenance, and turnover
interchangeable with ‘demyelinating disorders,’ but may be disturbed.
usually they are used in the context of a wider range The processes of myelin build-up and deposition
of disorders, characterized by either primary myelin are highly complex and require the expression of
loss or nonselective damage to myelin, axons and sup- many genes, the presence of many substances, the
portive tissue of the white matter. For instance, when activity of many enzymes, optimal coordination of
the terms white matter disorder and leukoen- processes within the oligodendrocytes, and optimal
cephalopathy are applied in elderly people, ischemic cooperation with the environment. Complex and dy-
white matter lesions are also implied, which do not in- namic processes are particularly vulnerable, and the
volve or do not only involve a selective loss of myelin. process of active myelination is easily disturbed.
In this book the following definitions are used: Some inborn errors of metabolism lead to a shortage
– ‘Demyelination’ is reserved for the process of of myelin components, and as a consequence to a dis-
myelin loss caused by primary and selective ab- turbance of the process of myelination. An example is
normality of either oligodendroglia or of the found in Pelizaeus-Merzbacher disease. Acquired dis-
1.16 Biochemical Changes Related to Demyelination 17

orders, such as hormonal imbalances and severe mal- ondary to adverse factors in the internal or external
nutrition, can also lead to disturbed myelin build-up. environment.
A disturbance of myelin maintenance and turn- Biochemical analysis of myelin and white matter
over can lead to demyelination. In some inborn errors demonstrates an abnormal composition of the same
of metabolism, the basic enzymatic defect involves type in many demyelinating disorders of diverse
the breakdown of one of the myelin components. This etiology. The concept of the nonspecific process of
component is trapped in the myelin sheath, and its myelin breakdown suggests that when maintenance
concentration increases gradually. Finally, the myelin of normal myelin is no longer possible it follows a
composition is altered to such a degree that the stereotyped route to complete destruction, largely
stability is lost, leading to demyelination. Examples irrespective of the initiating causes. The etiological
are metachromatic leukodystrophy and globoid cell factors can be wallerian degeneration, infections such
leukodystrophy. Of the acquired demyelinating disor- as subacute sclerosing panencephalitis, or intoxica-
ders, toxic disorders in particular can lead to a distur- tions with such agents as triethyltin, but also inherit-
bance of myelin maintenance and turnover. Myelin is ed metabolic diseases, e.g., X-linked adrenoleukodys-
rich in lipids and has a long half-life. Consequently, trophy, Canavan disease, and many other demyelinat-
lipophilic substances easily accumulate in myelin, ing diseases. In inherited diseases affecting myelin
disturbing the stability of the myelin membrane and metabolism, biochemical analysis often reveals cer-
leading to demyelination. tain abnormalities superimposed on the nonspecific
The myelin membrane may be intact and normal compositional abnormalities. These abnormalities
in appearance, biochemical composition, and func- are specific for a particular disorder or type of disor-
tion until it is attacked from the outside. This appears ders. For instance, an elevation of the very long-chain
to be the case in several acquired demyelinating fatty acids of the cholesterol esters is specific for a
disorders, including inflammatory processes (e.g., subgroup of peroxisomal disorders, including X-
multiple sclerosis, acute disseminated encephalomye- linked adrenoleukodystrophy. An elevation of sul-
litis), metabolic disturbances (e.g., central pontine fatide is found in the white matter of patients with
myelinolysis, Marchiafava-Bignami syndrome) and metachromatic leukodystrophy. The specific bio-
hypoxia (delayed posthypoxic demyelination). chemical abnormalities of myelin in the various dis-
Demyelinating disorders can also arise at the level orders are discussed in separate chapters. Here we
of the oligodendrocytes. Damage to oligodendrocytes will limit our discussion to the nonspecific myelin ab-
can lead to disturbances of myelin build-up, main- normalities. It should, however, be kept in mind that
tenance, and turnover. In some inborn errors of the degree of abnormality varies considerably among
metabolism storage of unwanted material occurs, different diseases and among different cases of the
ultimately leading to dysfunction and death of oligo- same disease depending on the stage of disease.
dendrocytes. In globoid cell leukodystrophy the toxic In degenerating myelin, the proportion of total
substance psychosine is thought to lead to oligoden- protein to total lipid is not usually dramatically
droglial cell death and myelin loss. In acquired de- altered, but the proportions of individual lipids are
myelinating disorders, selective oligodendroglial cell abnormal. The amount of galactolipids is decreased,
death can also occur. This is the case, for instance, in and cerebroside is usually much more severely affect-
progressive multifocal leukoencephalitis, in which ed than sulfatide. Moderate decreases of ethanol-
viral infection of oligodendrocytes is present. amine phosphoglycerides (mostly plasmalogen) are
Of course, in many disorders more than one mech- common. The amount of unesterified cholesterol is
anism of myelin affection is involved. In disturbances increased, often strikingly so, constituting almost half
of myelin build-up the myelin that is laid down may or even more than half the total lipid content, in con-
have an abnormal composition and configuration. trast to approximately 27% in normal myelin. No
Delayed myelination, dysmyelination, and early de- esterified cholesterol is found in the degenerating
myelination can occur at the same time. In other dis- myelin sheath. Such abnormal myelin is an interme-
orders, oligodendroglial cell death and myelin break- diate form between normal myelin and completely
down independent of oligodendroglial cell death catabolized myelin. The abnormalities are a result of
occur simultaneously. partial degradation.
The compositional changes in white matter as a
whole depend primarily on the extent of myelin loss
1.16 Biochemical Changes Related and only secondarily on changes in myelin com-
to Demyelination position. Typical white matter changes are increased
water content and reduced lipid-to-protein ratios,
Demyelinating disorders can be subdivided into two with specific decreases in such major myelin con-
large categories: inherited disorders due to an inborn stituents as cholesterol, cerebroside, sulfatide, and
error of metabolism and acquired disorders sec- ethanolamine phosphoglycerides. In addition, there
18 Chapter 1 Myelin and White Matter

is an increase in cholesterol esters in whole white of the axon. Even with time, there is no restitution of
matter in a number of diseases, but not in all. The the normal axon-to-myelin ratio. The new myelin
fatty acid composition of these esters is different from sheath in itself is normal with normal lamellar peri-
that of the small amount of esters normally present in odicity.
white matter, but closely resembles the fatty acids Remyelination also occurs when the demyelinated
linked to the 2-position in phosphatidylcholine. It is lesion was depleted of oligodendrocytes. The neces-
assumed that these esters come from myelin choles- sary supply of oligodendrocytes is provided by prolif-
terol and phosphoglyceride fatty acids. The presence eration of remaining, mature oligodendrocytes and
of cholesterol esters is taken as evidence of an active possibly also by proliferation of progenitor cells fol-
phagocytosis of myelin and, as such, as an indicator of lowed by differentiation into myelinating oligoden-
active demyelination, but the absence of cholesterol drocytes. It is often found that axons tend to be re-
esters does not mean that there is no active demyeli- myelinated in clusters, suggesting that a single oligo-
nation. The presence of cholesterol esters is reflected dendrocyte myelinates many axons in the vicinity.
in sudanophilia on histological examination. It is Remyelination among the demyelinating disorders
probable that the mechanism of breakdown is slight- is variable. The most successful examples of remyeli-
ly different in sudanophilic myelin destruction and nation are found in those conditions in which de-
nonsudanophilic breakdown. myelination has occurred rapidly, irrespective of
whether the condition is acute and monophasic or re-
lapsing and remitting. Remyelination is much more
1.17 Demyelination: Loss of Function limited in demyelinating disorders with a protracted,
chronic course. The presence of additional axonal
In normal myelinated nerve fibers, conduction is damage has an adverse effect on potential remyelina-
saltatory and internodal conduction time is fairly reg- tion. Some local factors, when present, may stimulate
ular. The conduction in demyelinated axons differs remyelination. There is evidence that epidermal
dramatically from that in normal fibers. The impulse growth factor, interleukin-2, immunoglobulins,
conduction may be either saltatory or continuous. If platelet-derived growth factor and insulin growth
the impulse conduction remains saltatory, the intern- factors may stimulate survival and proliferation of
odal conduction time varies widely from internode to oligodendrocytes and remyelination. In contrast, the
internode. The internodal conduction time is pro- presence of T-CD4+ immune cells interferes with
longed by increased leakage of current between the remyelination.
nodes and by depression of excitability of the nodal
membrane. There is, therefore, a decreased current
generation capacity and an increased threshold for 1.19 Retarded Myelination
excitation. In demyelinated fibers, a very slow contin-
uous conduction (about 5% of the conduction veloc- The process of myelination is both complex and pro-
ity of normal fibers) may be seen over short stretch- tracted. This means that the process is vulnerable to
es. A so-called safety factor for impulse conduction adverse factors over a long period of time, namely
can be calculated. If the required minimum is not from the second half of gestation up to the end of the
reached, impulse propagation is blocked. Further- 1st or 2nd year of life. Many stress factors that act on
more, the refractory period of demyelinated fibers is the incompletely myelinated brain and interfere with
increased, which leads to failure to transmit high-fre- the process of myelination do not have such a pro-
quency trains of impulses. foundly adverse effect on the mature brain. For in-
It is clear that demyelination, depending on its ex- stance, in the mature brain in which myelination is
tent and severity, can lead to serious loss of function. complete, stress factors such as malnutrition or hor-
However, damage to neurons, although not as promi- monal imbalances will not appreciably reduce the
nent as destruction of myelin, may also play a part in amount of myelin.
the functional deficit. Especially in inborn errors of Well-known factors potentially leading to retarda-
metabolism, substances may also accumulate in the tion of myelination include malnutrition, hormonal
membranes of axons, and in this way axonal dysfunc- imbalances (growth hormone deficiency, hypothy-
tion may arise, contributing to the functional loss. roidism, hypocortisolism, hypercortisolism), prena-
tal exposure to toxins (alcohol, anticonvulsants),
chromosomal abnormalities, pre- and postnatal as-
1.18 Remyelination phyxia, cerebral infections, hydrocephalus, and in-
born errors of metabolism with early onset.
Remyelination in the CNS is possible. Remyelinated It is important to realize that myelination is depen-
fibers can be recognized because the internodes are dent on normal function and interaction of oligoden-
too short and the myelin sheath is too thin for the size drocytes, neurons, and astrocytes and that retarda-
1.19 Retarded Myelination 19

tion of myelination can be related to dysfunction of disease, Alpers disease, infantile neuronal ceroid lipo-
oligodendroglia and myelin, dysfunction of astro- fuscinosis, infantile GM1 gangliosidosis, and infantile
cytes, or neuronal dysfunction. Cerebral infections GM2 gangliosidosis, all of which are neuronal disor-
and perinatal asphyxia may lead to disturbance of ders, myelination is severely retarded and the white
myelination through white matter damage or through matter looks severely abnormal on MRI, whereas in
neuronal damage. In addition, inborn errors of the later onset variants of neuronal ceroid lipofusci-
metabolism may disturb the process of myelination nosis, GM1 gangliosidosis and GM2 gangliosidosis
either directly, at the level of the oligodendrocyte or these white matter abnormalities do not occur.
myelin sheath, or indirectly, at the level of the astro- It has been demonstrated that myelination is an
cyte or neuron. It is important to realize that a distur- expression of the functional maturity of the brain.
bance of myelination may also be seen in neuronal Retarded myelination is an expression of immaturity
disorders with early onset. For instance, in Menkes or dysfunction.
Chapter 2

Classification of Myelin Disorders

The history of classifications of myelin disorders rical distribution were observed than in the familial
shows how each classification reflects the state of sci- cases described up to that time. Schilder considered
entific development of its time. A revised classifica- that this disease was a nosological and histological
tion based on the most recent scientific insights is entity related to multiple sclerosis and thought there
proposed at the end of this chapter. were acute and chronic variants of diffuse sclerosis
Interest in CNS myelin dates back to the nineteenth just as there were acute and chronic types of multiple
century. In 1854, Virchow was the first to suggest the sclerosis.
name ‘myelin’ when he described the sheaths around Since Schilder’s time a number of familial neuro-
axons in the CNS. It is not certain when Schwann logical disorders have been recognized, which were
(1810–1882) first described the cells since named af- histologically characterized by diffuse demyelination
ter him, which supply the myelin sheaths around the and again presented under the heading of diffuse
peripheral nerve fibers. In 1878, Ranvier described sclerosis. In 1916, Krabbe described a familial infan-
the nodes that have since been given his name in his tile form of diffuse sclerosis.Another familial variant,
“Leçons sur l’histologie du système nerveux.” He be- with a later onset and a less rapid progression, was re-
lieved that the nodes prevented the essentially liquid ported in 1925 by Scholz and in 1928 by Bielschowsky
myelin from flowing to the bottom of the nerve fiber and Henneberg. Scholz noted that in this case the
(axon). But despite this conviction, he showed consid- myelin breakdown products did not show the usual
erable insight into the functional role of the myelin (orthochromatic) staining properties, but stained
sheath, both as an insulator and as a facilitatory agent metachromatically.
in CNS functions. It was not until 1960–1961 that the In 1921, Neubürger drew attention to the fact that
role of the oligodendrocyte in the formation of the term diffuse sclerosis was being applied to sever-
myelin in the CNS became clear, and this was due to al very different disease entities, and he proposed a
the work of Bunge. distinction between inflammatory and degenerative
During the nineteenth century and early twentieth forms. In 1928, Bielschowsky and Henneberg suggest-
century, important progress was made in the clinical ed the name ‘hereditary progressive leukodystro-
and histological description of several demyelinating phies’ for the degenerative forms of diffuse sclerosis
disorders. Multiple sclerosis was recognized as a clin- and devised the following classification, based on the
ical disease entity, and the characteristic histological time of onset of the disease and its clinical course:
abnormalities, in the form of multiple demyelinated, 1. Infantile type of Krabbe
sclerotic plaques within otherwise normal white mat- 2. Subacute juvenile type of Scholz
ter, were described. Prominent names in this develop- 3. Chronic type of Pelizaeus-Merzbacher
ment are Carswell (1838), Cruveilhier (1835–1842)
and Charcot (1868). Hallervorden (1940) recognized that there were en-
In 1897, Heubner described a rare neurological dogenous and exogenous factors causing diffuse de-
disease in children, using the name diffuse sclerosis myelination and that a distinction was possible be-
as opposed to multiple sclerosis. The disease was his- tween disorders in which demyelination is invariably
tologically characterized by diffuse demyelination of present and forms a specific part of the disease and
the cerebral white matter and eventual striking hard- disorders in which demyelination occurs occasional-
ening of the white matter. Since that time, the term ly and is nonspecific. He proposed a more extended
‘diffuse sclerosis’ has commonly been used to de- classification based on these subdivisions:
scribe cerebral diseases with diffuse demyelination
and sclerotic hardening of the cerebral white matter. I. Endogenous central demyelination
Pelizaeus in 1899 and Merzbacher in 1910 reported on A. Specific demyelinating diseases
a chronic progressive familial type of diffuse sclerosis. a. Diffuse sclerosis of Krabbe and Scholz
In 1912, Schilder described a nonfamilial case of b. Pelizaeus-Merzbacher disease
more acute diffuse cerebral demyelination in a child, B. Nonspecific occasional demyelination
and he suggested the name encephalitis periaxialis e.g. Tay-Sachs disease
diffusa rather than diffuse sclerosis. In this case, more II. Exogenous central demyelination
prominent signs of inflammation and a less symmet- A. Specific demyelinating diseases
Classification of Myelin Disorders 21

a. Inflammatory types: and Suzuki were the first to propose a deficiency of


– Disseminated sclerosis galactocerebrosidase as the underlying biochemical
(= multiple sclerosis) cause of this disease. Advances in histochemistry also
– Diffuse sclerosis (Schilder) made it possible to discover the basis of metachro-
– Concentric sclerosis (Balò) matic leukodystrophy. Metachromasia had already
– Neuromyelitis optica (Devic) been found by Alzheimer in 1910, by Scholz in 1925,
– Encephalomyelitis disseminata and later by Von Hirsch and Peiffer (1955 and 1957).
– Infectious encephalitis Edgar (1955) pointed out that this condition was
b. Toxic-metabolic types: characterized by a remarkable elevation of white mat-
– Funicular myelosis ter hexosamine. In 1964, Austin et al. demonstrated a
(= vitamin B12 deficiency) decrease in arylsulfatase A activity in metachromatic
– Marchiafava-Bignami disease leukodystrophy.
B. Nonspecific occasional demyelination The enzyme defects of an increasing number of
a. Disturbances of blood flow, e.g. subcortical hereditary diseases were detected, whereas in other
atherosclerosis (= Binswanger disease) cases the precise enzyme defect could not yet be dis-
b. Edema covered but typical biochemical abnormalities char-
c. Toxic processes (carbon monoxide) acteristic for the diseases could be demonstrated. The
d. Tumors increased insight into hereditary metabolic disorders
and the ongoing ability to distinguish different hered-
Until that time, distinctions between different dis- itary and acquired demyelinating disorders on the ba-
eases had been based on neuropathological and clin- sis of a combination of clinical, histological and bio-
ical aspects of different demyelinating disorders. chemical data, were reflected in the classification pro-
From about this time onwards, histochemical meth- posed by Raine (1984). Raine distinguished five main
ods and chemical analyses became increasingly im- categories:
portant. The classification proposed by Blackwood in I Acquired inflammatory and infectious diseases of
1957 is a reflection of this development. It is based not myelin
only on morphological but also on histochemical dif- 1. Multiple sclerosis
ferences between various subgroups of diffuse sclero- 2. Multiple sclerosis variants
sis: (Schilder, Balò, Devic)
I Disseminated sclerosis (= multiple sclerosis) 3. Acute disseminated encephalomyelitis
II Diffuse demyelinating cerebral sclerosis 4. Acute hemorrhagic leukoencephalopathy
1. With replacement of myelin by sudanophilic 5. Progressive multifocal leukoencephalopathy
lipid II Hereditary metabolic disorders of myelin
a. With large bilateral cerebral plaques 1. Metachromatic leukodystrophy
b. With concentric demyelination (Balò type) 2. Globoid cell leukodystrophy (Krabbe)
2. a. With replacement of myelin by metachro- 3. Adrenoleukodystrophy
matic PAS-positive lipid (Norman type or 4. Refsum disease
Scholz type) 5. Pelizaeus-Merzbacher disease
b. With associated degeneration of interfasci- 6. Dysmyelinogenetic leukodystrophy
cular oligodendroglia (Greenfield type) (Alexander)
3. With replacement of myelin by nonmetachro- 7. Spongy degeneration (Canavan)
matic PAS-positive lipid (globoid cell or Krabbe 8. Phenylketonuria
type) III Acquired toxic-metabolic diseases of myelin
1. Hexachlorophene neuropathy
Meanwhile, insight into normal biochemistry and in- 2. Hypoxic encephalopathy
to mechanisms of biochemical derangement was IV Nutritional diseases of myelin
growing. The concept of hereditary inborn errors of 1. Vitamin B12 deficiency
metabolism caused by an enzyme defect leading to 2. Central pontine myelinolysis
dysfunction and breakdown of myelin started to 3. Marchiafava-Bignami disease
emerge. Fölling (1934) reported 10 patients in the V Traumatic diseases of myelin
same family with mental retardation and phenyl- 1. Edema
pyruvic acid in their urine. Jervis discovered in 1947 2. Compression
that the underlying metabolic defect in phenylke- 3. Barbotage
tonuria is a deficiency of phenylalanine hydroxylase. 4. Pressure release
In 1955, Diezel found that the lipids stored in the
globoid cells in Krabbe disease have very similar In this classification, four of the five categories in-
properties to those of cerebroside. In 1970, Suzuki volve acquired demyelinating disorders, and only one
22 Chapter 2 Classification of Myelin Disorders

involves hereditary demyelinating disorders. The log- disorders remain for which the primary defect is
ical continuation of this development is a refinement largely or completely unknown.
of the classification of hereditary demyelinating dis- An important point is that with increasing scientif-
orders. For instance, in some diseases the inborn er- ic insight the difference between ‘primary demyeli-
ror affects the metabolism of amino acids, and in oth- nating disorders’ or ‘myelin disorders’ and ‘primary
er diseases it affects the lipid metabolism. A further neuronal or axonal degenerative disorders’ is becom-
subdivision can be made among the disorders of lipid ing less clear. It is evident now that in a classic ‘prima-
metabolism according to the type of lipids involved. ry demyelinating disorder’ such as multiple sclerosis,
In 1987, Poser proposed a classification of hereditary early and important axonal damage and loss occurs.
myelin disorders based on the biochemical group of Some disorders, such as vanishing white matter, are
compounds whose metabolism is disturbed. He dis- characterized by serious loss of both axons and
tinguished six categories: myelin sheaths, and it may be that neither of them is
1. Disorders of glycosphingolipid metabolism really ‘primary.’ Several ‘primary neuronal disorders’
a. Ganglioside: GM1 and GM2 gangliosidoses, with infantile onset are accompanied by prominent
hematoside sphingolipodystrophy white matter abnormalities, which are not seen in the
b. Sulfatide: metachromatic leukodystrophy later onset forms of the same disorders. This is the
c. Galactocerebroside: case, for instance, in infantile GM2 gangliosidosis, in-
globoid cell leukodystrophy fantile GM1 gangliosidosis, and infantile neuronal
2. Disorders of phosphosphingolipid metabolism ceroid lipofuscinosis. We agree with Hallervorden
a. Sphingomyelin: Niemann-Pick disease and Poser that these disorders must have a place in a
3. Disorders of fatty acid metabolism classification of myelin disorders, just as they also be-
a. Adrenoleukodystrophy long in a classification of neuronal disorders. Because
4. Disorders of amino acid metabolism of the difficulties in distinguishing ‘primary neu-
a. Phenylalanine: phenylketonuria ronal/axonal disorders’ from ‘primary myelin disor-
b. Branched-chain amino acids: ders,’ we use the neutral word ‘leukoencephalo-
maple syrup urine disease pathies’ to comprise all disorders that predominantly
c. Many other amino acidopathies affect the white matter of the CNS, irrespective of
5. Multiple abnormalities whether or not the white matter abnormalities are the
a. Mucosulfatidosis result of a primary abnormality of myelin.
6. Unknown abnormalities We propose the following classification of leuko-
a. Idiopathic spongy sclerosis (Canavan) encephalopathies:
b. Fibrinoid leukodystrophy (Alexander) I Hereditary disorders
c. Pelizaeus-Merzbacher disease 1. Lysosomal storage disorders
d. Idiopathic sudanophilic leukodystrophy a. Metachromatic leukodystrophy
b. Multiple sulfatase deficiency
An important development during the last few c. Globoid cell leukodystrophy
decades concerns the knowledge of subcellular struc- (Krabbe disease)
tures, their role in normal metabolism and the conse- d. GM1 gangliosidosis
quences of their dysfunction. Major subcellular struc- e. GM2 gangliosidosis
tures are the nucleus, lysosomes, mitochondria, per- f. Fabry disease
oxisomes, cytoplasm matrix, smooth and rough en- g. Fucosidosis
doplasmic reticulum, Golgi apparatus, ribosomes, h. Mucopolysaccharidoses
and microtubules. i. Sialic acid storage disorders
Demyelinating disorders have been described as j. Neuronal ceroid lipofuscinoses
resulting from nuclear, lysosomal, mitochondrial, k. Polyglucosan body disease
peroxisomal, and cytoplasmic enzyme dysfunctions. 2. Peroxisomal disorders
Classification of hereditary demyelinating disorders a. Peroxisome biogenesis defects
according to the subcellular localization of the under- b. Bifunctional protein deficiency
lying metabolic defect stresses the clinical, biochemi- c. Acyl-CoA oxidase deficiency
cal, and neuropathological similarities within one d. X-linked adrenoleukodystrophy
category and the differences between the different and adrenomyeloneuropathy
categories. For the same reason, it is preferable to e. Refsum disease
classify the acquired demyelinating disorders accord- 3. Mitochondrial dysfunction
ing to their underlying causes into noninfectious–in- with leukoencephalopathy
flammatory, infectious–inflammatory, toxic–meta- a. Mitochondrial myopathy encephalopathy,
bolic, hypoxic–ischemic and traumatic. A number of lactic acidosis, and stroke-like episodes
(MELAS)
Classification of Myelin Disorders 23

b. Leber hereditary optic neuropathy v. Hypomyelination with atrophy of the basal


c. Kearns-Sayre syndrome ganglia and cerebellum
d. Mitochondrial neurogastrointestinal w. Hereditary diffuse leukoencephalopathy
encephalomyopathy (MNGIE) with neuroaxonal spheroids
e. Leigh syndrome and mitochondrial x. Dentatorubropallidoluysian atrophy
leukoencephalopathies y. Amyloid angiopathy
f. Pyruvate carboxylase deficiency z. Cerebral autosomal dominant arteriopathy
g. Multiple carboxylase deficiency with subcortical infarcts and leukoen-
h. Cerebrotendinous xanthomatosis cephalopathy (CADASIL)
4. Nuclear DNA repair defects aa. Cerebral autosomal recessive arteriopathy
a. Cockayne syndrome with subcortical infarcts
b. Trichothiodystrophy with photosensitivity and leukoencephalopathy (CARASIL)
5. Defects in genes encoding myelin proteins bb. Nasu-Hakola disease
a. Pelizaeus-Merzbacher disease cc. Pigmentary orthochromatic
b. 18q– syndrome leukodystrophy
6. Disorders of amino acid and organic acid me- dd. Adult autosomal dominant
tabolism leukoencephalopathies
a. Phenylketonuria II Acquired myelin disorders
b. Glutaric aciduria type 1 1. Noninfectious-inflammatory disorders
c. Propionic acidemia a. Multiple sclerosis and variants
d. Nonketotic hyperglycinemia b. Acute disseminated encephalomyelitis
e. Maple syrup urine disease and acute hemorrhagic encephalomyelitis
f. 3-Hydroxy 3-methylglutaryl-CoA lyase 2. Infectious-inflammatory disorders
deficiency a. Subacute HIV encephalitis
g. Canavan disease b. Progressive multifocal leukoencephalitis
h. L-2-Hydroxyglutaric aciduria c. Brucellosis
i. D-2-Hydroxyglutaric aciduria d. Subacute sclerosing panencephalitis
j. Hyperhomocysteinemias e. Congenital cytomegalovirus infection
k. Urea cycle defects f. Whipple disease
l. Serine synthesis defects g. Other infections
7. Miscellaneous 3. Toxic-metabolic disorders
a. Sulfite oxidase deficiency and molybdenum a. Toxic leukoencephalopathies
cofactor deficiency (endogenous and exogenous toxins)
b. Galactosemia b. Central pontine and extrapontine
c. Sjögren-Larsson syndrome myelinolysis
d. Lowe syndrome c. Salt intoxication
e. Wilson disease d. Marchiafava-Bignami syndrome
f. Menkes disease e. Vitamin B12 deficiency, folate deficiency
g. Premutation fragile X f. Malnutrition
h. Hypomelanosis of Ito g. Paraneoplastic syndromes
i. Incontinentia pigmenti h. Posterior reversible encephalopathy
j. Alexander disease syndrome
k. Giant axonal neuropathy 4. Hypoxic–ischemic disorders
l. Megalencephalic leukoencephalopathy a. Posthypoxic–ischemic leukoencephalo-
with subcortical cysts pathy of neonates
m. Congenital muscular dystrophies b. Delayed posthypoxic–ischemic
n. Myotonic dystrophy type I leukoencephalopathy
o. Proximal myotonic dystrophy c. Subcortical arteriosclerotic encephalopathy
p. X-linked Charcot-Marie-Tooth disease (Binswanger disease)
q. Oculodigitodental dysplasia d. Vasculitis
r. Vanishing white matter e. Vasculopathy of other origin
s. Aicardi-Goutières syndrome and variants 5. Traumatic disorders
t. Leukoencephalopathy with calcifications and a. Diffuse axonal injury
cysts
u. Leukoencephalopathy with involvement of The category of so-called cytoplasmic enzyme defi-
brain stem and spinal cord and elevated ciencies is not listed in this classification. The ratio-
white matter lactate nale is that such a disease category would represent a
24 Chapter 2 Classification of Myelin Disorders

very heterogeneous group of disorders as the cyto- Over the years, this classification has been modi-
plasm contains enzymes of many different biochemi- fied repeatedly. The basic defects of a steadily increas-
cal pathways. This is why it is preferable in this case to ing number of hereditary myelin disorders have been
make a subdivision according to the specific metabol- elucidated, and the number of ‘unknown’ disorders is
ic pathway involved. However, the group of amino aci- decreasing. Even the present classification of leuko-
dopathies and organic acidopathies is heterogeneous, encephalopathies is provisional and will have to be
as some of the enzymes concerned are in fact mito- adapted in the future to take account of expanding
chondrial or peroxisomal. In view of the relative ho- scientific insights. The structure of the proposed clas-
mogeneity in clinical presentation, diagnostic tests, sification allows easy integration of further informa-
and treatment strategies, we prefer to place them in tion.
one category, which is also in keeping with general
practice.
Chapter 3

Selective Vulnerability

Within the context of this book, attention is paid to Table 3.1. Hierarchy of selective vulnerability to hypoxic–is-
the concept of selective vulnerability, for two reasons. chemic conditions for different CNS structures
In the first place, the recognition of patterns of selec- Structure Hierarchy of vulnerability
tive vulnerability contributes to the understanding of
pathogenetic mechanisms of cerebral damage in the Hippocampus CA1 > CA4 > CA3 > granule cells
Cerebellum Purkinje cells > stellate or basket cells
different disorders. In the second place, the recogni- > granule cells > Golgi cells
tion of patterns of selective vulnerability is of practi- Striatum Small to medium-sized neurons
cal value and contributes to the diagnostic specificity > large neurons
of MRI interpretation. The concept of MRI pattern Neocortex Layers 3, 5, 6> layers 2, 4
recognition is based on the concept of selective vul-
nerability.
Spielmeyer (1925), Meyer (1936), Vogt and Vogt Vogt (1937) suggested the physicochemical proper-
(1937) and Scholz (1953) introduced the concept that, ties of specific neurons as the reason for the unequal
apart from the distribution of infarctions in vascular vulnerability to disease and introduced the term ‘top-
territories and border zones, specific brain regions istic areas.’ The ‘pathoclisis’ of a region is determined
may be more vulnerable to ischemic injury than oth- by specific chemical and physical properties, which
ers. Spielmeyer tried to find an explanation for this are also the essence of the specific function of that
difference by suggesting that variations in the local region. Meyer (1936) stated that it was too simple to
vascular supply facilitated vascular insufficiency in assume that only the physicochemical or local vascu-
such areas as the hippocampus. As we now know, lar factors were involved, and that other factors
structures of the CNS have different degrees of sensi- should also be taken into consideration. Such factors,
tivity to oxygen deprivation. Of the cellular elements according to him, are the nature of the noxious agent,
of the CNS, the neurons are the most vulnerable, fol- the ‘porte d’entrée,’ the path of distribution, and de-
lowed by oligodendroglia, astroglia and, finally, velopmental factors.
endothelial cells. Within the group of neurons, Meyer initiated a discussion about the pathophys-
some neuronal cell types are more vulnerable to hy- iology of the selective involvement of the basal gan-
poxic–ischemic damage than others. Structures more glia in some disorders. He drew attention to the selec-
liable to damage by hypoxia–ischemia are the hip- tive involvement of the globus pallidus in carbon
pocampus, the Purkinje cells of the cerebellum, the monoxide intoxication (Fig. 3.1), which is more con-
striatum, and the neocortex. Within these structures stant than the involvement of other structures, such
there is a further order of sensitivity among the dif- as the pars compacta of the substantia nigra, cornu
ferent cell types, as indicated in Table 3.1. Vogt and ammonis, Purkinje cell layer of the cerebellum, and

Fig. 3.1. Carbon monoxide intoxica-


tion in a 42-year-old male patient.The
T2-weighted images show the hyper-
intense lesion in the globus pallidus
26 Chapter 3 Selective Vulnerability

Fig. 3.2. Pyruvate dehydrogenase


complex deficiency in an 8-year-old
boy.The images depict selective
involvement of the globus pallidus
and the substantia nigra, the latter
probably due to transsynaptic
degeneration

Fig. 3.3. Kernicterus in a 1-week-old


neonate.The upper row shows a
proton density and a T2-weighted
image at the level of the basal ganglia.
The lower row shows a FLAIR and
a T1-weighted image at the same
level.The images show the typical
involvement of the globus pallidus
and the pulvinar.The globus pallidus
lesions are hardly seen on the T2-
weighted image, but the T2-weighted
image shows the abnormal signal of
the pulvinar more clearly than the
other images. It is not completely clear
why the globus pallidus has a high
signal on the T1-weighted image

cerebral white matter. However, the globus pallidus zymes,’ showing that awareness of something like the
may also be selectively involved in respiratory failure, mitochondrial system already existed.
mitochondrial defects (Fig. 3.2), kernicterus (Fig. 3.3), This discussion was renewed recently by Johnston
and intoxications with ether, potassium cyanide, and and Hoon (1999), who compared three conditions in
dinitrobenzol (leading to methemoglobinemia). children: pyruvate dehydrogenase complex deficien-
Meyer, who was well aware of the differences of these cy with a Leigh-like presentation and lesions in the
conditions, suggested the common factor responsible basal ganglia (Fig. 3.2); kernicterus with abnormali-
for the involvement of the globus pallidus was inter- ties prominently involving the globus pallidus but
ference with oxygen transport by either severe hypox- also involving the nucleus subthalamicus (Fig. 3.3);
ia or anemia or ‘inhibition of the respiratory en- and posthypoxic-ischemic encephalopathy caused by
Selective Vulnerability 27

Fig. 3.4. T2-weighted series depicting


the late pattern of acute profound
ischemia in a term neonate.There is
a triangular gliosis in the perirolandic
white matter, with focal ulegyria of the
cortex. In addition, there are lesions
in the dorsal part of the putamen,
the ventrolateral part of the thalamus,
and the dentate nucleus

acute profound asphyxia in a term neonate with basal ing. It is a fact, for instance, that in inherited mito-
ganglia abnormalities typically located in the dorsal chondrial encephalopathies with cellular energy fail-
part of the putamen and the ventrolateral part of the ure the putamen and caudate nucleus are usually
thalamus (Fig. 3.4). The authors try to explain the dif- preferentially affected and not the globus pallidus
ference in location of the lesion by arguing that the (Fig. 3.7). With this observation, it is questionable
neuronal circuit involved in asphyxia is different from whether the preferential involvement of the globus
that involved in mitochondrial disorders and kern- pallidus in carbon monoxide intoxications can be
icterus. They suggest that bilirubin toxicity is affect- blamed on mitochondrial dysfunction.
ing mitochondria in the globus pallidus, in this way It is clear that hypoxia–ischemia, some toxic sub-
providing a link with mitochondrial respiratory chain stances, some metabolites increased to toxic levels in
disorders. They reason that, on the other hand, gluta- inborn errors of metabolism, hypoglycemia, and
mate toxicity in particular affects the putamen, thala- some nutritional deficiencies (e.g. thiamine deficien-
mus, and cerebral cortex in hypoxic–ischemic condi- cy) all interfere with mitochondrial function. In more
tions. general terms, regardless of its cause, energy deple-
In an earlier edition of this book (1995) we made tion will lead to failure of mitochondrial oxidative
the same observation as Johnston and Hoon in 1999: phosphorylation,ATP depletion, accumulation of glu-
carbon monoxide intoxication affects the globus pal- tamate and other excitatory amino acids, opening of
lidus preferentially and most consistently, whereas ion channels, accumulation of Ca2+ in the cell, activa-
hypoxia in cases of near-drowning or strangulation tion of polyunsaturated fatty acid cascades and, final-
preferentially involves the putamen and caudate nu- ly, cell death. It would be logical if all forms of cerebral
cleus (Figs. 3.5, 3.6), layers 3, 5 and 6 of the cerebral energy failure, either caused by hypoxia–ischemia,
cortex, and Purkinje cells in the cerebellar cortex. In hypoglycemia, primary mitochondrial dysfunction
carbon monoxide intoxication we assumed a mito- or deficiencies, intoxications and inborn errors of
chondrially mediated effect on the globus pallidus. metabolism mediated through mitochondrial dys-
However, our hypotheses are apparently too simple, function, would lead to selective involvement of the
and important details of the pathophysiology of the same brain structures. This, however, is not true. It is
development of brain lesions elude our understand- correct that sometimes the pattern of abnormalities
28 Chapter 3 Selective Vulnerability

Fig. 3.5. A 3-year-old boy suffered


near-drowning and prolonged
attempts at resuscitation. Not only
the striatum is involved, but also the
globus pallidus, the thalamus, the
hippocampus, and tracts in the brain
stem. In addition, cortical laminar
necrosis is seen in the higher slices

Fig. 3.6. A young woman has been


anoxic for at least 3 min during resus-
citation.The FLAIR images show
that the lesions are confined to
the striatum

Fig. 3.7. Leigh syndrome in an


8-month-old boy, caused by the
Leigh/NARP mutation with a high
percentage of heteroplasmy.There
are lesions in the putamen and
caudate nucleus
Selective Vulnerability 29

Fig. 3.8. Leigh syndrome related to a


complex I deficiency in a 3-year-old
boy.The MR pattern shows symmetri-
cal lesions in the wall of the third ven-
tricle, the dorsal part of the midbrain,
and the dentate nucleus.The mamillary
bodies are not affected

Fig. 3.9. Pattern of thiamine deficien-


cy, which is very similar to that of Leigh
syndrome (cf. Fig. 3.8). An important
difference is that in thiamine deficien-
cy the mamillary bodies are involved in
most cases, whereas in Leigh syndrome
they are not

in Leigh syndrome (Fig. 3.8) and the pattern of thi- Apart from mitochondrially mediated disorders,
amine deficiency (Fig. 3.9) are very similar, although other conditions may involve the basal ganglia pre-
the lesions in the mamillary bodies, typical for thi- dominantly or selectively. This is the case in many
amine deficiency, are consistently lacking in Leigh inborn errors of metabolism other than mitochon-
syndrome, but most different causes of energy failure driopathies, in hepatic failure (Fig. 3.14), and in clas-
lead to very different patterns of abnormalities sic Creutzfeldt-Jakob disease (Fig. 3.15). The explana-
(Figs. 3.1, 3.2, 3.4–3.13). The biochemical explanation tions must be different from those so far proposed.
for the differences in selectively involved structures is In glutaric aciduria type I there is typically in-
unclear, and apparently our present models are too volvement of the putamen and caudate nucleus, but
simplistic. not of the cortical layers and Purkinje cells. The neo-
30 Chapter 3 Selective Vulnerability

Fig. 3.10. Typical involvement of the


parieto-occipital region in a neonate
with hypoglycemia; in the worst cases
this leads to multicystic encephalo-
pathy restricted to this region. Selec-
tive vulnerability of this region for
hypoglycemia disappears with age,
indicating the influence of morpho-
logic and biochemical maturation of
the brain on the pattern of resulting
damage

Fig. 3.11. Series of T2-weighted


images showing the classic triad
of late sequelae of recurrent or pro-
longed partial hypoxia–ischemia in
a preterm neonate: periventricular
leukomalacia.There is a periventri-
cular rim of signal abnormality; the
ventricles have an irregular border,
especially in the trigonum and occipi-
tal horns; and there is loss of white
matter volume, the sulci in the pari-
eto-occipital region abutting the
ventricular walls

striatal dysfunction and degeneration in glutaric native metabolites. Methylmalonate has been impli-
aciduria type I was demonstrated to be at least partly cated in inhibition of respiratory chain complex II,
related to N-methyl-D-aspartate receptor-mediated and it also inhibits the tricarboxylic cycle (Okun et al,
neurotoxicity of the endogenously accumulating 2002).
3-hydroxyglutarate (Kolker et al. 2002). Preference for the (neo)striatum in Creutzfeldt-
In methylmalonic academia, as in carbon monox- Jakob disease is partly due to the local severe loss of
ide intoxication, there is preferential involvement of parvalbumin-positive GABA-ergic inhibitory neu-
the globus pallidus. In this disease the selective lesion rons. As in patients with liver failure and hepato-
is due to accumulation of methylmalonate and alter- cerebral syndromes, the striatum has a high signal
Selective Vulnerability 31

Fig. 3.12. In Kearns-Sayre syndrome,


the subcortical white matter and
the globus pallidus are preferentially
affected

Fig. 3.13. In mitochondrial


encephalopathy with lactic acidosis
and stroke-like episodes (MELAS)
the lesions have an infarct-like
appearance on MR images, as shown
on these transverse FLAIR images in
a 9-year-old girl, but do not, as a rule,
respect vascular territories. (Courtesy
of Dr. M. Heitbrink and Dr. B. Wiarda,
Department of Radiology, Medical
Center Alkmaar, The Netherlands)

Fig. 3.14. IR images of a 3-year-old


boy with hepatic failure, showing the
effects of T1 shortening of the basal
ganglia. Because of the T1 shortening,
the basal ganglia can no longer be
distinguished from the surrounding
white matter

intensity on both T1- and T2-weighted images. In Scholz (1953, 1963) stated that under specific
Creutzfeldt-Jakob disease this could be related to a pathophysiological conditions focal ischemic brain
higher concentration of manganese (Guentchev et al. injury could be attributed to peculiarities of the vas-
1999). cular anatomy, while in other conditions the pattern
In intoxications with methylene dioxymetham- of brain damage could only be explained by the
phetamine (MDMA, or ‘ecstasy’) the lesions in the unique properties of the cells themselves. One of the
globus pallidus are due to severe brain dopaminergic most important observations he made was the influ-
neurotoxicity combined with less severe serotonergic ence of the nature of the insult on the resulting dam-
neurotoxicity (Reneman 2001; Ricaurte et al. 2002). age to the CNS. In acute obstruction of cerebral ves-
32 Chapter 3 Selective Vulnerability

Fig. 3.15. Involvement of the basal


ganglia is seen in sporadic ’classic’
Creutzfeldt-Jakob disease.The puta-
men and caudate nucleus have a high
signal on the T2-weighted image,
whereas the globus pallidus has a
high signal on the T1-weighted image

Fig. 3.16. In progressive multifocal


leukoencephalopathy the white
matter is predominantly involved,
to a much greater extent than the
gray matter.This results in a sharp
demarcation of white from gray
matter when the lesion abuts the
cortex, as shown in these images

sels, he found experimentally that “the cerebral and of activity than white matter and will as a rule be
cerebellar cortical layers, especially the Purkinje cells, damaged first and most severely. In infants, active-
were destroyed, whereas the shorter the duration of ly myelinating zones have a high activity and are,
ischemia the greater the possibility for selection to therefore, liable to damage. In term neonates with
take place and for the innate characteristics of the acute profound asphyxia, lesions can be seen in
structures to be expressed in a special pattern of mor- primary myelinating zones in the cerebral cortex,
phological alterations.” This, in fact, also seems to be subcortical tracts, and basal ganglia (Fig. 3.4).
true for other conditions, such as toxic encephalo- 2. Specific chemical affinity contributes to selective
pathies and inherited metabolic disorders. vulnerability. It has been known for a long time
We could add that the “innate characteristics” that certain areas in the brain are especially liable
mentioned by Scholz should not be seen as a static to damage by certain toxic agents. Hexachloro-
concept, but rather as a condition that is the product phene intoxication involves myelin sheaths exclu-
of genetic endowment, stage of development, interac- sively. Hexachlorophene encephalopathy, induced
tion with other structures in neuronal functional cir- in preterm neonates by washing them for antisep-
cuits, and dependence on their functional activity tic reasons with hexachlorophene-containing so-
and the local condition at the time of the insult. lutions, causes a myelinopathy with splitting of the
There are several pathophysiological mechanisms myelin lamellae and intramyelinic vacuole forma-
that help to explain selective vulnerability of certain tion. Vacuolating myelinopathy in the neonate
brain areas relative to others: always has a special distribution, irrespective of its
1. Level of activity is an important factor in selective cause, related to the distribution of myelinated
vulnerability. Energy depletion by hypoxia–is- versus unmyelinated areas. A clear example is
chemia, toxic influences, and metabolic derange- found in maple syrup urine disease. Intoxication
ments will have the greatest effect on structures with triethyltin, cuprizone, toxic heroin, or poi-
with the highest oxygen demand and chemical soned cocaine also leads to myelin splitting and
turnover. Gray matter in adults has a higher level vacuolation. Furthermore, lipophilic substances
Selective Vulnerability 33

generally accumulate preferentially in myelin. Or- lation by excess excitatory amino acids (glutamate
ganic solvents such as are used by painters lead to and aspartate) has recently been recognized as a
irregular, patchy demyelination and can cause so- final common pathway for inflicting injury upon
called house-painter’s dementia or an organic the CNS. Many conditions can lead to an abnormal
psychiatric syndrome. Demyelination, loss of accumulation of excitatory amino acids. The pref-
gray–white matter distinction, and signal changes erential distribution of lesions by this mechanism
in the basal ganglia have been described in will basically be in areas with the highest density
toluene sniffers. Heavy metal poisoning also of related receptors.
shows selective affinity for certain brain regions, 6. The density of mitochondria and varying percent-
as seen for example in Wilson disease (neostria- ages of mutated mitochondrial DNA have been
tum, mesencephalon, dentatorubral tracts, nucle- suggested as explanations for the selective in-
us dentatus), lead encephalopathy (cerebellar volvement of CNS structures in mitochondrial en-
white matter in adults, cortical neurons in infants cephalopathies. Some reports have linked the per-
and children), and mercury poisoning in Minama- centage of mutated mitochondrial DNA in the
ta disease (occipital and parietal cortex). basal ganglia in MELAS to local levels of lactate in
3. Accumulation and/or deficiency of substances MRS (Dubeau et al. 2000).
have different effects on different areas of the 7. Antigen–antibody reactions may be at the root of
brain. In inborn errors of metabolism, the selec- selective CNS lesions. This is the case in a number
tion of primary targets and the pattern of spread of the paraneoplastic and parainfectious lesions
of the lesions may be influenced by these factors. of the brain. Antibodies against tumor antigens
Differences in selective vulnerability may be ex- may, for example, cross-react with similar anti-
plained by differences in residual activity of en- bodies on Purkinje cells. Paraneoplastic CNS dis-
zymes in the various cells, in importance of the orders such as limbic encephalitis and brain stem
enzyme function missing, in effects of the accu- encephalitis may be caused by this mechanism. In
mulation of abnormal breakdown compounds parainfectious disorders, for example those relat-
(psychosine in Krabbe disease), in sensitivity to ed to Mycoplasma pneumoniae infections, the
lack of substances that are not formed, and pres- same mechanism may play a part and lead to
ence of other factors within the cell with synergis- myelin damage in patients with acute disseminat-
tic or antagonistic effects. It is often very difficult, ed encephalomyelitis.
if not impossible, to define the factors responsible 8. Bacterial, viral, or fungal infection may involve
for well-known patterns of selective involvement specific structures in the brain. For example, pro-
in inborn errors of metabolism. Shortage of di- gressive multifocal leukencephalitis is an infec-
etary nutrients may also lead to selective damage. tion of the oligodendrocyte, and thus predomi-
Malnutrition of infants in the 1st year of life leads nantly involves white matter (Fig. 3.16). In other
to delayed myelination. Cobalamin deficiency in disorders the porte d’entrée may be responsible
subacute combined tract degeneration leads to in- for the localization of the lesion, e.g., in herpes
volvement of specific areas in the brain and spinal simplex encephalitis (Fig. 3.17).
cord. 9. Hyper- or hypo-osmolar conditions may cause
4. Patterns of selective vulnerability may be related white matter lesions in specific brain areas. In
to distribution of neurotransmitter systems. In sodium intoxication in infants, unmyelinated ar-
some inborn errors of metabolism, some neu- eas appear to be most severely involved, probably
rodegenerative disorders and some toxic–meta- because of the lower ‘resistance’ to the shifts of
bolic encephalopathies, selective vulnerability water. In central pontine myelinolysis the central
may result from interference with a neurotrans- part of the pons is mainly involved, for unknown
mitter system. For instance, inborn errors of reasons.
GABA metabolism have been described that lead 10. Another important factor is the nature (severity,
to dysfunction of structures in which GABA- duration, recurrence) of the noxious event. This
ergic neurotransmission is important. In Segawa can be seen in neonates with perinatal asphyxia.
syndrome, hereditary progressive dystonia The pattern of brain damage in chronic or recur-
with marked diurnal variation, disturbances of rent partial hypoxia-ischemia is very different
dopaminergic neurotransmission cause nigrostri- from the pattern observed in acute profound
atal dysfunction. In hyperammonemia, impair- hypoxia-ischemia. Whereas the first leads to
ment of the neurotransmission by glutamate oc- periventricular leukomalacia (Fig. 3.11), the sec-
curs. ond leads mainly to lesions in the basal ganglia,
5. The density of synapses for excitatory amino acids thalamus, and perirolandic cortex (Fig. 3.4).
determines the sensitivity to adverse effects of 11. Trans-synaptic degeneration illustrates that the
these substances. Excitotoxicity due to overstimu- function of the brain depends on functional cir-
34 Chapter 3 Selective Vulnerability

Fig. 3.17. Predominant but not


exclusive involvement of temporal
lobes is seen in herpes simplex infec-
tions, shown here in a FLAIR and a
T1-weighted image with contrast
(upper row) and two trace diffusion-
weighted images (b=1000) (lower
row).The ADC values in the affected
areas are low (–40 % relative to
normal appearing tissue)

cuits, so that damage to one part of the circuit in- white matter is involved, probably due to a com-
fluences function, and possibly the structure of bination of hypomyelination and white matter
other parts of the circuit. This was established degeneration. These diseases primarily involve
long time ago by Guillain and Mollaret for hyper- neurons and axons, and later onset variants lead
trophic olivary degeneration. The Guillain-Mol- to degeneration of gray matter structures only.
laret triangle consists of the connections of three However, if the disease has its onset before com-
nuclei: nucleus ruber, nucleus dentatus, and nu- pletion of myelination, the white matter of the
cleus olivary inferior. Disruption of this circuit CNS is also severely affected. It may be that the
by trauma, tumor, or surgery leads to lesions in complex process of myelin deposition, which re-
the inferior olivary nucleus. It is important to rec- quires joint activity and cross-talk of oligoden-
ognize this as a product of trans-synaptic degen- drocytes, axons, and astrocytes, is disturbed be-
eration, and not, in tumor cases, as recurrent or cause of axonal abnormalities. We found evi-
multifocal tumor. Another example is substantia dence for apoptosis of oligodendrocytes in infan-
nigra degeneration secondary to lesions in the tile GM1 gangliosidosis, and this may be the
globus pallidus or secondary lesions in the limbic primary problem, but it may also be that this is
system when one part is affected (Fig. 3.2). the consequence of the failed collaboration and
12. In neonates and infants the developmental stage subsequently in itself a cause for further myelin
is also of great importance in determination of loss.
which areas will be affected by a noxious agent.
The patterns of hypoxic–ischemic lesions in Understanding mechanisms of selective vulnerability
preterm and term neonates are well described contributes to the understanding of patterns of cere-
and show the changes in vulnerability, which de- bral involvement as shown by MRI. On the other
pend on developmental factors. Another example hand, MRI gives us insight into patterns of selective
is found in several neuronal storage disorders, vulnerability and into what we understand about
such as GM1 gangliosidosis, GM2 gangliosidis them and what we do not. In disorders of quite differ-
and neuronal ceroid lipofuscinosis. It is only in ent origins, some final common pathways may
the early-infantile onset variants that the cerebral explain similarities in image abnormalities. On the
Selective Vulnerability 35

Fig. 3.18. Urea cycle disorder and


encephalopathy related to a metabolic
decompensation with hyper-
ammonemia in a 6-year-old boy.
Images show asymmetrical infarct-like
lesions involving the frontal lobes,
predominantly affecting the left hemi-
sphere.The distribution of the lesions
does not correspond to a vascular terri-
tory, and the lesions differ in some
aspects from those seen in infarctions,
especially with respect to the cortical
involvement

other hand, even in disorders that have rather obvious stant: the central white matter is involved first, includ-
similarities as far as pathogenesis is concerned, strik- ing periventricular white matter and corpus callo-
ing differences in image abnormalities are sometimes sum, and the demyelination proceeds centrifugally
observed, indicating the inadequacy of our present from there. The arcuate fibers are the last to be in-
understanding of pathogenetic and pathoplastic volved. An explanation proposed for this feature is
mechanisms. that in these disorders abnormal substances accumu-
An illustration of this inadequacy is provided by late in membranes, leading to a progressively altered
the involvement of cerebral structures in hyperam- myelin composition and progressively unstable
monemia. Impaired function of the urea cycle en- myelin membrane that is liable to breakdown. As the
zymes leads to hyperammonemia and raised concen- arcuate fibers are the last to myelinate they contain
trations of glutamine in the brain. Hyperammonemia the youngest myelin, which is altered least. However,
can also be caused by hepatic failure of quite different some amino acidopathies and organic acidopathies
origins (acquired, other inborn errors of metabolism) primarily involve the arcuate fibers and the demyeli-
and still leads to an encephalopathy. MRS has re- nation progresses in a centripetal way. It is obvious
vealed high concentrations of glutamine in the brain that the biochemical abnormalities and interactions
in hyperammonemia, irrespective of its origin. On the are completely different in the latter disorders than in
basis of similarities in the pathogenesis of en- lysosomal disorders: interference with energy metab-
cephalopathy in these disorders, a similar clinical pic- olism, lack of normal myelin components, and pres-
ture and similar MR images might be expected. How- ence of toxic metabolites are important in amino aci-
ever, the expressions of these disorders, clinically and dopathies and organic acidopathies. However, it is
on MRI, are very different. In urea cycle disorders, still difficult to explain why the arcuate fibers are first
large cerebral lesions involving cortex and white affected. MRI shows an involvement of the periven-
matter are seen, with asymmetrical distribution and tricular white matter in the occipital lobe and the
asymmetrical neurological signs and symptoms splenium of the corpus callosum in the early phases
(Fig. 3.18). In encephalopathy due to hepatic failure of most patients with the cerebral form of X-linked
related to an acquired disease, brain involvement and adrenoleukodystrophy. The disease spreads in a
neurological symptomatology are symmetrical. MRI frontal direction. The reason for this occipital prefer-
shows a symmetrical T1 shortening of the basal gan- ence is unclear. It is noteworthy that in about 10% of
glia, including globus pallidus, putamen, caudate nu- patients with cerebral X-linked adrenoleukodystro-
cleus and other central gray matter structures phy the pattern is reversed, starting in the frontal
(Fig. 3.14). This means that other factors in addition lobes and the genu of the corpus callosum and pro-
to hyperammonemia must be involved. gressing towards the dorsal parts of the brain. The
Selective vulnerability, and thus MRI pattern reason for the frontal predominance in these patients
recognition, is not a static concept; it also refers to dy- is even less clear. The cerebral involvement in X-
namic changes. It has become clearer with MRI than linked adrenoleukodystrophy tends to be symmetri-
it ever was with neuropathology that among the white cal, as in the lysosomal disorders, but there are excep-
matter disorders there are great differences in early tions in which one side of the brain is far more severe-
involvement of brain structures and spread in the ly involved than the other. It would be worth knowing
course of time. In the lysosomal storage disorders in- whether the areas primarily involved are more vul-
volving the white matter, especially the sphingolipi- nerable to the disease (and if so, why?) or whether the
doses, the temporal progression is remarkably con-
36 Chapter 3 Selective Vulnerability

areas that are primarily spared are more resistant to lesion. The lesions in urea cycle disorders are not me-
the process (and if so, why?). diated by vascular changes, but are probably the result
Symmetry of cerebral involvement can be expect- of a direct toxic effect in the involved area, mediated
ed to be a general rule in inborn errors of metabo- by the presence of elevated levels of glutamine. The
lism, toxic encephalopathies, and neurodegenerative asymmetrical and focal nature of the lesions is how-
disorders, as toxic influences, genetic factors, and de- ever unexplained, however. Another well-known ex-
ficiencies of essential substances are thought to be ample of asymmetry is found in MELAS, a mitochon-
similar for the left and right sides of the brain. In a drial disorder. The lesion usually has a cortical pre-
number of disorders, however, asymmetrical involve- dominance and is restricted to one part of the brain,
ment is the rule. An example is found once more in again simulating an infarction (Fig. 3.13). However,
urea cycle disorders. In all urea cycle disorders, meta- the lesion is not located in a vascular territory or a
bolic derangement is characterized by the accumula- border zone area. Here too, there is no explanation for
tion of urea precursors, notably ammonium, and by the asymmetry.
increased glutamine in the brain. During episodes of The basis of this book is the recognition of MR pat-
metabolic derangement focal brain lesions occur, terns that allow differentiation between disease cate-
which are seen on MRI to be strikingly asymmetrical gories and between disease entities. Knowledge of
(Fig. 3.18). The large lesions can involve an entire which structures are selectively damaged by a specif-
hemisphere or a large part of it. Such an MRI pattern ic disease lies at the basis of this approach. MRI has an
may be misinterpreted as an infarction, because the edge on histopathology, because it gives us the privi-
distribution of the lesion may suggest the involve- lege of seeing patterns in an early stage of disease and
ment of one or more vascular territories. The MRI allows us to follow their development. This has not
characteristics of the lesion, however, are different only helped in pattern recognition of classified disor-
from those of infarctions, even though both show low ders, but also helped us to recognize unfamiliar pat-
signal intensities on T1-weighted images and high terns and to define new entities.
signal intensities on T2-weighted images. These dif- In all the chapters concerning specific disorders,
ferences can be described by the way in which gray one section is devoted to the description of MRI pat-
and white matter are simultaneously involved, the terns of that specific disorder; another section is de-
way in which the whole affected area is swollen and voted to the description of pathogenetic mechanisms.
demarcated from the rest of the brain, the slightly in- As far as possible, these two will be linked. A separate
homogeneous change in signal intensity, and the of- chapter will deal with the principles of MRI pattern
ten ‘unusual’ vascular territory that is occupied by the recognition.
Chapter 4

Myelination and Retarded Myelination

4.1 Myelination cerebellar vermis, inferior cerebellar peduncles, vesti-


bular nuclei, superior cerebellar peduncles and their
It was Flechsig (1920) who originally put forward the decussation, dentate nucleus, medial longitudinal fas-
view that the degree of myelination of the CNS might ciculus, medial geniculate bodies, subthalamic nuclei,
be correlated with functional capacity. In his theory inferior olivary nuclei and ventrolateral nuclei of
he stated that myelination started in projection path- thalamus. Myelin can also be seen in the fasciculus
ways before association pathways, in peripheral gracilis and cuneatus and in their nuclei (Counsell et
nerves before central pathways, and in sensory areas al. 2002; Sie et al. 1997).
before motor ones. Although he modified his theory In the period between 30 and 36 weeks of gestation
slightly in response to his critics, he continued to the quantity of myelin in the aforementioned struc-
maintain that fibers always myelinated in the same tures increases, but from weeks 30–36 of gestation no
order: first the afferent (sensory), then the efferent myelin is seen in any new sites on MRI. This is not in
(motor), then the association fibers. agreement with histological descriptions of the
The histological study of fetal development has myelin process. Reasons for this are the lower sensi-
confirmed that myelination proceeds systematically tivity of MRI to small quantities of myelin, also the
and, in nerve pathways with several neurons, in the reason for a time-lag in myelination timetables be-
order of conduction of the impulse. The first signs of tween histology and MRI, and the higher spatial res-
myelination appear in the column of Burdach at the olution of histological methods. At a GA of 36 weeks
gestational age (GA) of 16 weeks, becoming stronger evidence of the presence of myelin appears on T1-
from the 24th week onward. The column of Goll starts weighted images in the posterior limb of the internal
to myelinate at 23 weeks of gestation. Cerebellar tracts capsules (with higher intensity in the area of the cor-
start to myelinate at about 20 weeks of gestation, and ticospinal tracts) and in the tracts from and to the
the amount of myelin at birth is considerable. Pyra- precentral and postcentral gyri in the corona radiata.
midal tracts start to myelinate at 36 weeks at the level In the period between 37 and 42 weeks of gestation
of the pons, but at birth the amount of myelin is still myelination of these tracts also becomes visible on
small. In other tracts, for example, the rubrospinal T2-weighted images. At this time myelination is visi-
tracts, the pattern of the pyramidal tract is followed. ble in the tegmentum pontis but not in the basis pon-
In a term neonate at a GA of 40 weeks myelin stains tis. Myelin now also appears in the lateral geniculate
reveal myelin in the medulla oblongata, in the central bodies and in the optic tracts, chiasm, and nerves. The
parts of the cerebellar white matter, in the cerebellar myelin density in the basal ganglia and corticospinal
peduncles and the vermis, in the medial lemniscus tracts increases, and myelin shows up in the optic ra-
and fasciculus medialis longitudinalis in the pons and diation.
mesencephalon, in the posterior limb of the internal During the 1st month after birth, myelination pro-
capsule, spreading into the globus pallidus and thala- gresses rapidly. It becomes more prominent in the ar-
mus and, in the thalamocortical connections in the eas mentioned above. The pattern of myelin presence
centrum semiovale, upwards to the parasagittal parts in the cerebellum changes (see below). On T1-weight-
of the postcentral gyrus and backwards into the optic ed MR images myelin becomes visible in the rest of
radiation. Paul Flechsig’s lithographs (1920) demon- the striatum and caudate nucleus. Myelin in the optic
strate this myelination pattern beautifully (Fig. 4.1). pathways becomes more prominent, and myelin is al-
Several authors, including Keene and Hewer (1931) so present in cortical layers of the primary motor and
and Yakovlev and Lecours (1967), have published dia- sensory cortex and in the hippocampus and parahip-
grams of the progress of myelination (Fig. 4.2). pocampal gyrus. With increasing myelination in the
MRI is unique in making it possible to visualize the occipital and parietal lobes, the splenium of the cor-
progress of myelination in vivo in astonishing detail. pus callosum starts to myelinate. From the 3rd or 4th
It is now possible to describe the state of myelination month onward myelination proceeds in the frontal
in preterm and term neonates in detail and to follow direction, and from the 4th to 5th month onward,
this process through up to full maturation. also in the temporal direction. The anterior limb of
In the preterm child with a GA of less than the internal capsule shows myelination from the 3rd
30 weeks the following structures show myelination: to 4th month onward, proceeding in the 5th month to-
38 Chapter 4 Myelination and Retarded Myelination

Fig. 4.1. Lithograph in left upper row


is reproduced from work of Paul
Flechsig (1920), who used refined
histological techniques to depict
ongoing myelination in the brain.
Progress of myelination of a young
infant is presented here. Note that
myelin (dark in the image) is already
circling around the temporal horn to
reach the hippocampus and parahip-
pocampal gyrus. Also note myelina-
tion of the auditory pathway in the
superior temporal gyrus.The two
T2-weighted coronal MR images show
the same features in vivo. Myelination
in this case is somewhat further
advanced than on the lithograph,
already spreading towards the
parietal U fibers

wards the genu of the corpus callosum. At 6 months tion can be followed for much longer on T2-weighted
myelination starts to spread in the frontal lobes, and than on T1-weighted images. On T2-weighted images
on the T1-weighted images the pattern of myelination myelination does not reach the arcuate fibers in the
is seen to be more or less complete at about 8 months. frontal and temporal areas before the 12th–14th and
The corpus callosum reflects the myelination of the 14th–18th months, respectively.
parts it connects. T1-weighted images show myelin in Myelination is not an all-or-none process. Myeli-
the splenium at 3 months and in the genu at about nated white matter gradually replaces the unmyeli-
6 months of age; T2-weighted images show this nated white matter. In T2-weighted series unmyelinat-
4–6 weeks later. ed white matter has higher signal intensity than gray
It should be clear that we are describing ’apparent’ matter. With ongoing myelination, white matter
myelination, i.e. myelination as it appears on T1- or becomes darker, and eventually it can no longer be
T2-weighted images, which is dependent on pulse se- differentiated from gray matter. This transition or
quences and field strength. The apparent progress in ’cross-over’ period is reached in the parietal and
myelination on T2-weighted images lags behind that occipital areas between 8 and 10 months after birth.
seen on T1-weighted images. Because of this, myelina- After this period myelinated white matter has lower
4.2 MRI Pulse Sequences 39

Fig. 4.2. Classic diagram of progression of


myelination as conceived by Yakovlev and
Lecours. Most of the structures mentioned
can also be made visible on MRI. Appearance
of myelination on MR images is 1 or 2 weeks
behind this schedule with conventional MR
techniques. From Yakovlev and Lecours
(1967), with permission

signal intensity than gray matter on T2-weighted 4.2 MRI Pulse Sequences
images in this region. The frontal and temporal lobes
show this cross-over at 12–14 and 14–18 months, T1-weighted spin echo (SE) or inversion recovery (IR)
respectively. T1-weighted images show the same re- and T2-weighted SE sequences are complementary. In
versal in signal as the T2-weighted images, but in the the first 6 months of life, T1-weighted images show to
reverse direction. Unmyelinated white matter has a better advantage which areas contain myelin, while
lower signal than gray matter, whereas myelinated T2-weighted SE images, with the proper pulse se-
white matter has a higher signal. Because of the pref- quence, differentiate partially myelinated from non-
erential T1-shortening effect of myelin, which exceeds myelinated and completely myelinated white matter
the T2-shortening effect, the signal reversal on T1- more adequately. In MRI, pulse sequences should be
weighted images occurs weeks to months before it chosen so that the contrast/noise ratio is as high as
occurs on the T2-weighted images. This implies possible and the differences between tissues are max-
that there is a phase, for the cerebral hemispheric imal. It is, therefore, useful to consider the T1 and T2
white matter mainly in the second half of the 1st year values of gray matter and of unmyelinated and myeli-
of life, in which white matter structures have a higher nated white matter in order to make an adequate
signal than gray matter on both T1- and T2-weighted choice. Holland et al. (1987) found that at a field
images. strength of 0.35 T, T1 of white matter is 1615±120 ms
40 Chapter 4 Myelination and Retarded Myelination

(mean±standard deviation) at birth, 1150±60 ms at netization transfer component present in multislice


6 months and 580±50 ms at 1 year; T2 of white matter conventional SE images is increased in FSE and influ-
is 91±6 ms at birth, 64±6 ms at 6 months, 57±5 ms at enced by the number of slices. From this it will be
1 year and 53±3 ms at 3 years. For gray matter the clear that MR images made with FSE sequences can-
corresponding T1 measurements are 1590±60 ms at not be compared with conventional SE images. The
birth, 1300±70 ms at 6 months and 890±75 ms at influence of the FSE techniques on the estimation of
1 year, and the corresponding T2 measurements the progress of myelination is probably negligible, but
88±8 ms at birth, 67±7 ms at 6 months, 69±3 ms at the influence on the depiction of disease conditions
1 year and 62±3 ms at 3 years. The changes observed should be considered, especially when the abnormal-
with increasing age can be explained by a decreasing ities are subtle. In examination of neonates it is im-
water content and increasing myelin content. Protons possible to be sure in advance whether abnormalities
in the myelin membrane are less mobile, water con- will be found.
tent is lower, and T1 and T2 are, therefore, shorter in T1-weighted SE or IR series are of great importance
myelinated areas. for following the spurt of myelination during the first
A long TR, long TE sequence is advantageous to 6°months after birth. They demonstrate the progres-
allow full benefit of the T2 differences between gray sion of myelination beautifully, but they are less reli-
matter, unmyelinated white matter and myelinated able than T2-weighted images for assessing the quan-
white matter and to minimize T1 effects, which coun- tity of myelin deposited. The difference in T1 between
teract the T2 effects. We use a 3000/120 SE series, gray matter and unmyelinated white matter at birth
which shows unmyelinated white matter with high gives the unmyelinated white matter a darker appear-
signal intensity, gray matter and partially myelinated ance than the cortex. With ongoing myelination the
white matter with intermediate signal intensity, and white matter will become brighter than the cortex.
myelinated areas with low signal intensity. In our SE Between these structures, again, a cross-over or con-
3000/120 series, the transition or ’isointense’ pattern trast inversion takes place. In T1-weighted images,
between gray and white matter is reached in the pari- however, this is a less striking event than in T2-
etal and occipital lobes in normal children by the age weighted images. In premature neonates the unmyeli-
of 6–9 months.With a different pulse sequence, for in- nated white matter appears much darker than the rim
stance SE 2000/80, this transition phase may be of cortical gray matter. Even at 40°weeks of GA this
7–12 months. Because of the heavier T2-weighting in difference is still present, although less marked. After
our series, the T2-decay trajectories traverse each that, the unmyelinated white matter rapidly changes
other at a somewhat steeper angle and the transition its signal and becomes almost isointense with gray
period is, therefore, more clearly marked and of matter. The white matter structures in which myelin
shorter duration. is advancing stand out as very bright and attract most
Fast or turbo spin echo (FSE or TSE) sequences are attention in the assessment of myelination age. A
time-saving procedures and are frequently used in comparison of T1- and T2-weighted images makes it
imaging of neonates and infants, but they have impor- clear that the T1-weighted images are more sensitive
tant disadvantages. Instead of one ’projection’ or line indicators of the presence of myelin, even when it is
in the k-space (the virtual spatial frequency space present only in small amounts. Partially myelinated
from where the image is reconstructed by Fourier structures, which are isointense or even still mildly
transformation), FSE sequences generate multiple hyperintense relative to gray matter on T2-weighted
spin echoes from a single RF excitation, which are images, are already white on T1-weighted images. The
close enough together (echo spacing in the order of reason for the higher myelin sensitivity of T1- than of
15 ms) to provide multiple ’projections’ or lines in the T2-weighted images can be found in a number of fac-
k-space. Imaging time is shortened by a factor of the tors at the molecular level. The special structure of
number of echoes collected in the train. SEs are, the myelin membrane with a lipid : protein ratio of
therefore, acquired at slightly different TEs, which 70 : 30 (dry weight) and a cholesterol content of 30%
will affect the resulting image. The repetition of refo- is important. The construction of lipid layers separat-
cusing 180° pulses improves local magnetic homo- ed by a 40-molecule thick layer of water, into which
geneity, thus diminishing magnetic susceptibility ef- the hydrophilic phosphate polar groups of lipids and
fects. On FSE and TSE images hemorrhages and calci- the cholesterol hydroxyls project, creates a unique
fications will consequently be less conspicuous. Be- lipid–water interfacial interaction seven times as in-
cause of the short time interval between refocusing tense as that at a typical protein–lipid interface. There
pulses, the apparent T2 of adipose tissue becomes is a field-dependent cross-relaxation between protons
longer and the fat signal will be less suppressed than of myelin water and protons of myelin lipid, a phe-
on conventional T2-weighted SE images. Fat will, nomenon known as magnetization transfer. The ap-
therefore, appear bright on heavily T2-weighted FSE parent effect of these conditions is a shortening of T1
images. Other factors influence FSE images. The mag- induced by myelin, which is more pronounced than
4.3 Diffusion-weighted Imaging and Diffusion Tensor Imaging 41

would be expected to result from deposition of mem- Table 4.1. Apparent diffusion coefficient (ADC) (×10–3 mm 2/s)
branous structures alone. and fractional anisotropy (FA) (103) in term neonates
Fluid-attenuated inversion recovery (FLAIR) im- Region ADC FA
ages use a combination of an IR pulse with an inver-
sion time chosen to suppress the water signal, fol- Frontal white matter 1.62–1.73 190–210
lowed by a heavily T2-weighted FSE. Because the Posterior limb internal capsule 1.06–1.01 410–500
water signal is suppressed, abnormalities will stand Occipital cortex 1.15–1.20 150–180
out clearly. FLAIR pulses are not very useful in the Occipital white matter 1.58–1.69 330–370
estimation of progress of myelination. The difference Corpus callosum
between gray, unmyelinated and myelinated white Genu 1.22–1.37
matter is much less on FLAIR images than on conven- Splenium 1.17–1.32
tional T1- and T2-weighted images. It is probably the Thalamus 1.23–1.08
suppression of the water signal in the water-rich envi- Mesencephalon (tegmentum) 0.99–1.09 420–480
ronment of the neonatal brain that is responsible for Pons, anterior 1.13–1.27
this diminished contrast, since the disappearance of
Pons, posterior 0.94–1.06
’free’ water in the brain of neonates and its replace-
ment by myelinated structures play a major part in
the maturation process.
images. In contrast with ADC maps, FA images show
excellent gray–white matter contrast. In clinical
4.3 Diffusion-weighted Imaging practice, estimation of ADC and FA of different brain
and Diffusion Tensor Imaging structures makes it possible to quantitate the
progress of brain maturation, including myelination,
Diffusion-weighted imaging (DWI) and diffusion and to visualize the formation of white matter tracts.
tensor imaging (DTI) use the microscopic movement In addition, DWI offers anisotropic diffusion maps
of water molecules and the relative loss of phase of that can be displayed separately for each diffusion
protons caused by this movement in different brain gradient direction or as an averaged ADC map, the
structures as a means of image contrast. Parameters so-called trace map.
that are frequently used are the apparent diffusion co- Measurements of ADC are available for different
efficient (ADC) of tissues and the relative anisotropy ages from the fetal period to adult age. ADC values
of brain structures, usually expressed as fractional decrease in between these points in time and develop-
anisotropy (FA). ment, from 1.50–1.95 × 10–3 mm2/s for white matter in
The relative anisotropy is practically zero in gray the fetal brain to about 0.87–0.95 × 10–3 mm2/s in the
matter in term-born neonates and in adults: diffusion mature brain. Maturity in this respect is reached at
in gray matter is isotropic. However, at a GA of the age of approximately 2 years. For the basal ganglia
26 weeks cortical anisotropy is not zero, because at the corresponding data are, respectively, 1.56 × 10–3
that time the cortical cyto-architecture is dominated mm2/s and 0.79 × 10-3 mm2/s. More detailed data are
by radial glial fibers and radially oriented apical den- available for term neonates (see Table 4.1). ADC and
drites of the pyramidal cells. This structure is dis- FA values are different for different brain structures,
rupted in time by the addition of basal dendrites and depending on their structure and stage of develop-
thalamocortical efferents. ment.
White matter ADC and FA depend on stage of mat- ADC values decrease with ongoing maturation and
uration. During the early development of the brain FA values rise. This process is exponential. The de-
ADC values are high, and they subsequently fall. This crease in ADC and increase in FA are fast in the first
is because of the initial high water content and the 3–6 months, followed by a slower further decrease
subsequent overall decrease in water content together and increase, respectively. White matter anisotropy
with the development of more densely packed struc- increases over the next 6 months, leveling in the 2nd
tures with a high density of membranes that hinder year of life. The numbers in Table 4.1 are only guide-
free water movement. FA is initially low and increases lines.ADC and FA values reported from different cen-
rapidly. The increase in FA can be observed even in ters can differ substantially. Data given here are in the
the premyelination state. The increase in FA during same order of magnitude as those reported by Neill et
early development is not only the result of the pro- al. (1998).
gressing myelination; other contributing factors are Detailed measurements of diffusion tensor charac-
the number of microtubule-associated proteins in ax- teristics related to brain maturation have been taken
ons, axon caliber changes, and a significant increase and have shown regional differences running parallel
in the number of glia. More densely packed structures to the progress of myelination as seen on convention-
have higher FA. Both ADC and FA can be displayed in al MR images (Mukherjee et al. 2002). Unfortunately,
42 Chapter 4 Myelination and Retarded Myelination

Table 4.2. Magnetization transfer ratio (%) during develop- Table 4.3. Signal intensity of central white matter relative to
ment white matter on long TR SE images

Projection fibers (pyramidal tracts) At 1 month 23–25 % Short TE Long TE


At 3 months 25–27 % Stage Age MWM MWM
At 6 months 31–33 %
At 20 months 34–37 % I 1st month ↑ =/↓
Association fibers At 1 month 19–21 % II 2nd months =/↓ ↓/↓↓
At 3 months 22–25 % III 3rd–6th months ↓ ↓↓
At 6 months 28–30 %
At 20 months 29–33 % IV 7th–9th month ↓ ↓↓
Commissural fibers At 1 month 23–24 % V >9th month ↓ ↓↓
At 3 months 24–25 %
At 6 months 29–33 % ↑↑ hyperintense, ↑ slightly hyperintense, = isointense, ↓ slight-
At 20 months 34–37 % ly hypointense, ↓↓ hypointense, SE spin echo, MWM myelinat-
ed white matter, TE echo time

the reported values refer to ’eigenvalues’ of the diffu- Timetables can be based upon the visual inspection
sion tensor and are not directly comparable to ADC of changes on T1- and T2-weighted images. T1-weight-
and FA values. ed images are very useful in the first 6 months of life,
as discussed above, whereas T2- weighted images pro-
vide more useful information after 6 months. Combi-
4.4 Magnetization Transfer nation of T1 and T2 data has distinct advantages, espe-
cially in cases with delayed or distorted myelination.
With magnetization transfer (MT) quantitative infor- At term birth, T1-weighted images show evidence
mation about the condition of brain tissue can be ob- of myelination in the medulla spinalis, cerebellar
tained by estimating MT ratios (MTRs), either voxel white matter, dorsal part of the pons, mesencephalon,
based and displayed as maps, or measured globally posterior limb of the internal capsula (in particular
and displayed as MTR histograms. It has been shown the area of the corticospinal tracts), and the postcen-
that MTR changes in infants correlate with changes in tral parasagittal areas, as a continuation of the long
the degree of myelination (Van Buchem et al. 2001). ascending spinocortical tracts. The optic radiation
MTR can be used to quantitate the progress of myeli- becomes myelinated soon after birth. The splenium of
nation and monitor the regional development. MTR the corpus callosum is myelinated in the 3rd month,
values (%) show an increase over time (Rademacher the truncus in the 4th and 5th month, and the genu in
et al. 1999) (Table 4.2). the 5th and 6th months. In the 3rd and 4th months
Gray matter structures also show an increase in myelination spreads to the anterior limb of the inter-
MTR over time; these changes, however, are much less nal capsule. From the parietal parasagittal area,
impressive (Rademacher et al. 1999). myelination starts to spread in anterior and posterior
It is possible to use MTR whole-brain histograms directions. After this stage further distinction on T1-
to monitor the progress of brain maturation. This will weighted images becomes difficult.
possibly develop into a method that will allow easy es- Additional T2-weighted images can be used to re-
timation of retarded maturation and be a good tool fine the assessment of myelination. Myelination of the
for individual follow-up studies (Van Buchem et al. central parts of the brain can be distinguished from
2001). the hemispheric white matter, making it possible to
compose a timetable based on signal intensities with
five steps of progression (see Tables 4.3, 4.4).
4.5 Myelination: Timetables Some markers are useful in daily practice. On long
TR, long TE SE images, the splenium of the corpus
In daily practice it is useful to have a timetable of nor- callosum has a low signal by 6 months of age, the genu
mal progress of myelination at hand. Even with a con- at 8 months of age. The cross-over in the occipital
siderable variation, it is evidently possible to provide lobe, when gray and white matter have a uniform and
a time scale for normal development and to assess re- indistinguishable intermediate signal intensity (are
liably significant delay in myelination. There are sev- isointense), occurs at about 7–9 months. At about
eral approaches to making a timetable and each one 9 months the ‘adult’ contrast between gray and white
has its own advantages and disadvantages. The meth- matter starts to emerge in the occipital lobes. The an-
ods can be subdivided in nonquantitative (visual in- terior limb of the internal capsule is myelinated on
spection), semi-quantitative (ratios) and quantitative the heavily T2-weighted sequence at 8–11 months. At
(absolute values). 12 months the frontal white matter starts to myeli-
4.5 Myelination: Timetables 43

Table 4.4. Signal intensities of peripheral white matter relative to white matter on long TR SE images
Short TE Long TE

stage age UWM MWM UWM MWM

I 1st month ↓ ↑ ↑ =
II 2nd month = = ↑↑ =
III 3rd-6th month ↑ = ↑↑ =
IV 7th-9th month = =
V >9th month ↓ ↓↓

↑↑ hyperintense; ↑ slightly hyperintense; = isointense; ↓ slightly hypointense; ↓↓ hypointense; MWM myelinated white matter;
UWM unmyelinated white matter; TE echo time

nate; it should be nearly complete at 14 months of age. darkening of the rim around the dentate nucleus in
The temporal lobe is the last to myelinate; this occurs the first weeks of life; of the second marker, the exten-
between 14 and 18 months of age. The U-fibers of the sion of myelin into the cerebellar folia; and of the
cerebral hemispheric white matter become fully third marker, the cross-over in signal intensities be-
myelinated between 18 and 24 months. tween the corticospinal tracts and the substantia ni-
Use of marker sites can be helpful, especially for re- gra. When automatic scaling is used, which is usually
search purposes, to provide the necessary detail and the case on MR systems, it proves difficult to identify
quantitation. An approach defining specific targets the five stages as described by these authors. Usually,
for myelination, and scoring in a large population the however, three stages of maturation can be distin-
time of onset and completion of myelination of such guished in the posterior fossa. The structures in-
targets, leads to normal values with definition of nor- volved in the recognition of these three stages on T2-
mal variation. weighted transverse images are: the basis pontis, the
In a semi-quantitative way the progress of myeli- tegmentum pontis, the middle cerebellar peduncle,
nation can be assessed by calculating a ratio between the dentate nucleus, the peridentate white matter, the
the averaged signal intensity of a chosen region of in- corpus medullare cerebelli, and the white matter ex-
terest and dividing that by the averaged signal inten- tending into the cerebellar folia (Fig. 4.3). In stage 1
sity of a fully myelinated structure. For this purpose (<1 month after term) the basis pontis is not myeli-
the posterior limb of the internal capsule is often cho- nated, there is some myelin in the tegmentum of the
sen, because myelination is complete at that structure pons and in the middle cerebellar peduncles, and the
at a GA of 44 weeks. A ratio so obtained is indepen- nucleus dentatus has a high signal intensity and is
dent of type of equipment and field strength. In surrounded by a rim of lower signal intensity, fol-
myelin disorders that also affect the posterior limb of lowed by a high signal of the cerebellar white matter.
the internal capsule, of course, this does not apply. In stage 2 (>1 months, <3–4 months) myelination
The head of the caudate nucleus has also been used as starts to appear in the basis pontis; the tegmentum is
a reference. still darker, however, and the dentate nucleus starts to
Some groups have used this more refined method appear darker. In stage 3 (>3–4 months), tegmentum
of marker sites in combination with estimation of and basis pontis are now approximately as dark as
time of contrast cross-over between structures and each other; the dentate nucleus and the cerebellar
used this approach to look at the detailed progress of white matter appear completely dark and isointense
myelination in the cerebellum and brain stem in the with the middle cerebellar peduncle. After comple-
first months of life. Martin et al. (1990) took as target tion of these stages, myelination starts to extend to-
areas the cerebellar hemispheres, dentate nucleus, wards the cerebellar folia, gradually shaping the ’ar-
nucleus ruber, middle cerebellar peduncle, corpus bor vitae’ of the cerebellum.
medullare cerebelli, pontine tegmentum, basis pontis, We could make the assessment more detailed and
medial lemniscus, and corticospinal tracts. They de- add more structures: pyramidal tracts, medial lem-
fined five stages of progress of cerebellar myelination, niscus, and medial longitudinal fasciculus, structures
depending on the relative signal intensities of these that can be distinguished in good-quality images of
regions. Landmarks of these studies used for time es- the brain stem and cerebellum. A similar diagram
timates were again the gradually darker appearance could be produced for the mesencephalic structures,
of myelinated areas on T2-weighted images, the fur- where the signal intensity, of the red nucleus, the sub-
ther extension of myelination towards the subcortical stantia nigra, the corticospinal tracts, medial lemnis-
structures, and inversion of contrast between struc- cus, and inferior calicles changes with time, depend-
tures. An example of the first marker is the gradual ing on the pulse sequence used.
44 Chapter 4 Myelination and Retarded Myelination

Fig. 4.3. Myelination


of the posterior fossa
(nd nucleus dentatus;
mcp middle cere-
bellar peduncle;
V vermis cerebelli;
1 peridentate
white matter;
2 corpus medullare;
3 peripheral white
matter; IV fourth
ventricle)

Fig. 4.4. Myelination and gyration are both part of the matu- 27 weeks there are still remains of the germinal matrix, but it is
ration process of the brain. These axial T2-weighted images less conspicuous. At 35 weeks of gestation myelination has
obtained in infants with gestational ages (GAs) of 23, 27, and started in the posterior limb of the internal capsule. (The dark
35 weeks demonstrate brain maturation in that period. At dot represents the corticospinal motor tract.) The gyral devel-
23 weeks of gestation the germinal matrix is visible at the opment over this period is beautifully shown in these images.
trigonum, frontal horns and caudothalamic notch. The brain From Childs and Ramenghi et al. (2001), with permission
surface is still smooth. The sylvian fissure is hardly visible. At
4.6 Gyration 45

Quantitative measurements can be used to assess


myelination of the brain in general, and in different
structures in particular. Examples are measurements
of T1, T2, ADC, FA, and MTR.
It is clear that myelination is not the only concern
as far as development of the CNS is concerned. Pro-
gression of gyration should be included in an inven-
tory of brain maturation. For premature children
such a maturation index has been proposed (Childs et
al. 2001), which provides a standardized method of
assessing cerebral maturation. Four parameters are
assessed in this scale: cortical folding, myelination,
germinal matrix distribution, and glial cell migration
(see also Fig. 4.4).

4.6 Gyration

The brain of premature children and neonates is im- Fig. 4.5. A–D. Patterns of sulcus configuration for gyral devel-
mature, not only in myelination but also in develop- opment score 2 (A or B), score 3 (C), and score 4 (D). From Van
ment in gyri (gyration). Histopathological, intrauter- der Knaap et al. (1996), with permission
ine ultrasound and MR studies have yielded insight
into the development of some major fissures, which
provide landmarks of gyral development: the inter- ent brain areas using a five-point scoring system
hemispheric fissure (before 15 weeks of GA), the pari- (Fig. 4.5): (1) The surface is smooth without gyri and
eto-occipital fissure, the calcarine fissure, the central sulci or there is, at most, some undulation of the cor-
rolandic sulcus (visible at a GA of 23–25 weeks), and tical surface area. (2) Width of the gyri is greater than
the development of the insula, visible at a GA of the depth of the sulci. (3) Width of the gyri is equal
18 weeks, with overriding frontal, temporal and pari- to the depth of the sulci. (4) Width of the gyri is less
etal opercula at 28–29 weeks’ gestation (Chi et al. than the depth of the sulci. (5) Gyri and sulci are
1977). branched. Seven cortical areas were studied separate-
For MRI, insight into gyral development from the ly: (1) the frontal lobe minus the area of the central
age of 26–28 weeks gestation is most important, as sulcus, (2) the area of the central sulcus, (3) the pari-
preterm infants often come for MRI. In premature in- etal lobe minus the area of the central sulcus, (4) the
fants the cerebral cortex is still entirely or relatively occipital lobe minus the medial area, (5) the medial
smooth and lacking in sulci, depending on the post- occipital area, (6) the posterior part of the temporal
conceptional age of the infant. Over time, shallow sul- lobe, and (7) the anterior part of the temporal lobe.
ci develop in an ordered sequence. The sulci increase The five stages of gyration distinguished are shown in
in number and become deeper. The gyri become in- Figs. 4.6–4.10. The ages of the children ranged from
creasingly branched. Most of the process of a conver- 30 to 42 weeks. At all ages the development of the
sion of a smooth, lissencephalic brain into a nearly rolandic area and the medial part of the occipital
fully developed cortical gyral pattern occurs between lobe (areas 2 and 5) was most advanced. In the pari-
26 and 44 weeks of gestation. The mature pattern of etal area, occipital area and posterior temporal area
gyri and sulci is normally reached at the age of (areas 3, 4, and 6), an intermediate rate of gyral devel-
3 months after term. In our study of gyral develop- opment was found. Gyral development was slowest
ment in preterm and term neonates (Van der Knaap and latest in the frontal and anterior temporal areas
et al. 1996) gyral development was graded for differ- (areas 1 and 7).
46 Chapter 4 Myelination and Retarded Myelination

Fig. 4.6. Sagittal T1-weighted, and coronal and transverse bordering gyrus. The frontal and temporal cortical surface is
T2-weighted images in a preterm infant at a GA of 30 weeks, smooth; the cortex is slightly undulating in the posterior area.
showing stage 1 gyration. The depth of the central, parieto- From Van der Knaap et al. (1996), with permission
occipital and calcarine sulci is about equal to the width of the
4.6 Gyration 47

Fig. 4.7. Preterm infant at a GA of 32 weeks. Sagittal T1- and the bordering gyri. Compared with stage 1 sulci are better
coronal and transverse T2-weighted images show gyration defined and increased in number in the remaining areas. From
stage 2. The central and calcarine sulci are now deeper than Van der Knaap et al. (1996), with permission
48 Chapter 4 Myelination and Retarded Myelination

Fig. 4.8. Gyration at a GA of 36 weeks. Sagittal and coronal defined and more numerous.The depth of the sulci is equal or
T1- and transverse T2-weighted images demonstrate stage 3 greater than the width of the gyri in most areas. From Van der
gyration. The central sulcus and sulci of the medial occipital Knaap et al. (1996), with permission
area are now becoming branched. Sulci are becoming better
4.6 Gyration 49

Fig. 4.9. Gyration at a GA of 39 weeks. Sagittal and coronal The sulci have a closed form in most areas. The depth of the
T1- and transverse T2-weighted images depict stage 4 gyra- majority of sulci is greater than that of the bordering gyri.From
tion.The central and sulci of the medial occipital area are now Van der Knaap et al. (1996), with permission
branched. The number of sulci and gyri has increased again.
50 Chapter 4 Myelination and Retarded Myelination

Fig. 4.10. Gyration at a GA of 42 weeks, depicted on sagittal T1-weighted, and coronal and transverse T2-weighted images, show-
ing stage 5 gyration. Branching of sulci is now seen in all areas. From Van der Knaap et al. (1996), with permission
4.8 Iconography of Myelination and Gyration 51

4.7 Delayed Myelination, Irregular Irregular myelination with local or generalized


Myelination, Hypomyelination, hypermyelination, or myelination not following the
and Arrest of Myelination normal routes of progress, is rare, but is seen occa-
sionally. Hypermyelination, or advanced myelination,
Once MRI criteria for normal progress of myelination has been observed in patients with Sturge-Weber
have been established, it is possible to diagnose delays syndrome. It has been suggested that epileptic
in this process. If it is true that myelination expresses seizures may stimulate myelination. However, ad-
functional maturity a correlation between delay in vanced myelination or hypermyelination is not seen
myelination and delayed development of psychomo- in most patients with infantile forms of epilepsy. Lo-
tor functions can be expected. Roughly speaking, this cal hypermyelination in the basal ganglia is manifest
appears to be the case. We have been able to confirm histologically as the so-called status marmoratus, a
it in a group of children with hydrocephalus, in whom late sequela of perinatal hypoxia. In this case the
MRI and neuropsychological data were obtained be- myelination does not involve the proper targets and
fore and twice after shunting. There was a strong cor- does not occur around axons but around astrocytic
relation between (a) the progress of myelination as extensions. Because of the low signal intensity of the
compared with the normal myelination standard and basal ganglia on T2-weighted images and the dark ap-
(b) the progress of mental development as compared pearance of myelin in this sequence, MRI has so far
with the normal developmental standard. It is impor- not succeeded in identifying this condition.
tant to follow up the progress of myelination in any Hypomyelination or arrest of myelination occurs
child in whom a delay is suspected, to see whether, in Pelizaeus-Merzbacher disease, a disorder of proteo-
and if so when, the child catches up with normal lipid protein synthesis, one of the major myelin pro-
myelination. It might be assumed that a longer delay teins. In this disorder no myelin, or only very little, is
in the restoration of the normal pattern would coin- produced. In Salla disease, a lysosomal storage disor-
cide with a poorer prognosis. der, and DNA repair disorders such as Cockayne syn-
There are many possible causes for a delay in drome and trichothiodystrophy with sun hypersensi-
myelination: hypoxia–ischemia, congenital infec- tivity hypomyelination is also present. To establish a
tions, congenital malformations, chromosomal ab- secure diagnosis of retarded or arrested myelination,
normalities, congenital heart failure, postnatal infec- at least two observations sufficiently far apart are
tions, hydrocephalus, hypothyroidism, hypercorti- necessary.
solism, hypocortisolism, fetal intoxications, malnutri-
tion, and inborn errors of metabolism. The delay is
usually bilateral and symmetrical, but unilateral delay 4.8 Iconography of Myelination
is seen in cases with hemimegalencephaly, unilateral and Gyration
porencephalic cysts, cerebral hemiatrophy, or unilat-
eral periventricular leukomalacia. Illustrations in this chapter show the progress of
The critical period in myelin development was ini- gyration (Figs. 4.4–4.10) and myelination in normal
tially thought to coincide with the proliferation of neonates and infants (Figs. 4.11–4.23). Many exam-
myelin-forming cells, rather than with the period of ples of disturbances of myelination are found in the
membrane accumulation. The mechanism of ’stunt- other chapters in this book. In Table 4.4 the myelina-
ing’ of oligodendroglial proliferation as a cause of hy- tion of some important structures on MRI is indicat-
pomyelination has been under discussion, because in ed. In some cases a more detailed look at structures in
animal research no major deficits of oligodendro- relation to their surroundings is useful, in order to see
cytes could ever be established, except in severely how contrast changes over time. The structures in the
starved animals. Therefore the induction of myelin posterior fossa are a good example (Fig. 4.3). We also
membrane formation, rather than cell proliferation, include an example of diffusion-weighted imaging in
seems to be the actual critical event. Damage in criti- estimating the progress of myelination (Fig. 4.24).
cal periods is often limited to areas in which myelina-
tion is beginning at that time. This knowledge is help-
ful in establishing the time of insult in infants and
children.
52 Chapter 4 Myelination and Retarded Myelination

Fig. 4.11. Myelination at a GA of 32 weeks. The sagittal T1- dorsal part of the pons, the mesencephalon, and the corpus
weighted series (upper row) shows the features of the prema- medullare of the cerebellum. The transverse T1-weighted se-
ture brain nicely: lack of gyration in the frontal areas, with ries shows the same features and gives a good impression of
some gyration in the parietal and occipital lobes.The midsagit- the high water content of the unmyelinated white matter
tal image shows myelin present in the medulla oblongata, the
4.8 Iconography of Myelination and Gyration 53

Fig. 4.12. Myelination at a GA of 39 weeks. A sagittal T1- losum is still thin and also unmyelinated. From the basal gan-
weighted SE series is shown from right to left.In the brain stem, glia, myelinated white matter tracts can be followed towards
the basis pontis is still not myelinated (arrow). The corpus cal- the post-rolandic gyrus (arrows)
54 Chapter 4 Myelination and Retarded Myelination

Fig. 4.13. Myelination 2 weeks after birth at term, as seen on a lar peduncles, optic tracts, posterior limb of the internal cap-
T1-weighted transverse inversion recovery (IR) series. Myeli- sule, white matter tracts in the basal ganglia and ascending
nation is seen in the medulla oblongata, middle cerebellar tracts towards the post-rolandic gyrus. Note in the upper im-
peduncle, tegmentum pontis (especially medial lemniscus, ages that cortical gray matter is also myelinated
arrows), colliculus inferior, decussation of the superior cerebel-
4.8 Iconography of Myelination and Gyration 55

Fig. 4.14. T2-weighted transverse series of myelination will give clues to the age of myelination. On T2-weighted
2 weeks after birth at term for comparison.Cerebellar myelina- images the tegmentum pontis (arrow) and mesencephalon
tion is still in stage 1: the hilus of the dentate nucleus is bright; are darker than the ventral pons. Myelin can also be seen in
the dentate nucleus is surrounded by a dark band (arrow), the superior vermis, posterior limb of the internal capsule,
again followed by bright cerebellar white matter. Contrast in- basal ganglia and ascending tracts into the post-rolandic
version of these structures during the progress of myelination gyrus (arrows)
56 Chapter 4 Myelination and Retarded Myelination

Fig. 4.15. In the posterior fossa T2-weighted images show basis pontis and tegmentum pontis, although much less pro-
that cerebellar myelination has progressed to stage 2 in this nounced than before. In the mesencephalon, the pyramidal
2-month-old infant.The bright ring around the dentate nucle- tracts and decussation of the superior cerebellar peduncles
us has disappeared, but the peripheral white matter of the can be identified
cerebellum is still bright.There is still a difference between the
4.8 Iconography of Myelination and Gyration 57

Fig. 4.16. IR images at 3 months. The myelinated structures ed. The optic tract is myelinated, as is the optic radiation. The
can easily be identified. Note the beginning of myelination in posterior limb of the internal capsule is fully myelinated at the
the pyramidal tracts in the mesencephalon (large white arrow) postnatal age of 2 weeks. Myelin has now spread to the pre-
and the strongly myelinated decussation of the superior cere- central gyrus and will advance dorsally and ventrally to myeli-
bellar peduncles (small black arrow). The colliculus inferior nate the occipital, the frontal and, finally, the temporal lobes
(black arrow) and the auditory tracts are also clearly myelinat-
58 Chapter 4 Myelination and Retarded Myelination

Fig. 4.17. At the age of 5 months the genu of the corpus callosum starts to myelinate. On IR images myelination will soon appear
to be complete.T2-weighted images will then be more useful in providing information about maturation of the brain
4.8 Iconography of Myelination and Gyration 59

Fig. 4.18. T2-weighted series at 4 months of age. In the pons, superior cerebellar peducles, the colliculus inferior (arrow), the
basis and tegmentum have a low signal; the medial lemniscus pyramidal tracts,the corpus mamillare and the optic tract have
has an even lower signal (arrow), as do the middle cerebellar a low signal. The posterior limb of the internal capsule is also
peduncles. The corpus medullare of the cerebellum is myeli- dark (arrows). A difference is visible between the unmyelinat-
nated, but myelination is not yet extending towards the cor- ed white matter in the frontal and temporal regions and the
tex. At the level of the mesencephalon, the decussation of the occipital and parietal region where myelination has started
60 Chapter 4 Myelination and Retarded Myelination

Fig. 4.19. T2-weighted coronal images at the age of 4 months, showing the difference between still unmyelinated white matter
in the frontal and temporal lobe and the more advanced myelination posteriorly
4.8 Iconography of Myelination and Gyration 61

Fig. 4.20. Myelination at 7–8 months of age. On the T2- gray and white matter in the occipital and parietal areas has
weighted images the central parts are now myelinated,includ- started; there is little contrast between gray and white matter.
ing the genu of the corpus callosum. The crossover between In the frontal and temporal regions this is not yet the case
62 Chapter 4 Myelination and Retarded Myelination

Fig. 4.21. Myelination at 12–13 months. The adult contrast is now emerging in all lobes except the temporal lobe, the latest to
myelinate.The T2-weighted series shows that the spread of myelin into the arcuate fibers is still not complete
4.8 Iconography of Myelination and Gyration 63

Fig. 4.22. Adult pattern of myelination on T2-weighted images in a 5-year-old child.The temporal lobes now also show the adult
gray–white matter contrast
64 Chapter 4 Myelination and Retarded Myelination

Fig. 4.23. Adult pattern of myelination on T1-weighted (IR) images.These images were taken from a 5-year-old boy

Fig. 4.24. Diffusion-weighted-


imaging (DWI) and diffusion
tensor imaging (DTI) allow further
refinement and quantitation of
the progress of myelination.These
images depict single-shot EPI with
single diffusion gradient in slice,
read or phase direction at b=1000,
showing anisotropy of myelinated
fibers depending on the gradient
direction in a baby boy 3 months
of age
4.8 Iconography of Myelination and Gyration 65

Fig. 4.25. Images demonstrating evolution of FA over time, measured with 12+1 diffusion gradient settings

Table 4.5. Myelination on MRI: chronological table (WM white matter). From:Yakovlev and Lecours (1967), with permission
Regions of CNS Fetal age (weeks) Postnatal age (weeks) Postnatal age (months)

24 28 32 36 40 4 8 12 16 20 6 9 12 >12
Cerebellar peduncles + + ++ +++ +++ +++ +++ +++ +++ +++ +++ +++ Further re-
Tegmentum pontis + + + ++ ++ +++ +++ +++ +++ +++ +++ finement of
Basis pontis + + ++ +++ +++ +++ +++ myelination
in subcortical
Medial lemniscus + ++ ++ +++ +++ +++ +++ +++
arcuate fibers
Pyramidal tracts + + + + + + ++ ++ +++ continues for
Optic nerve + ++ ++ +++ +++ +++ +++ +++ +++ several years
Optic radiation + + ++ +++ +++ +++ +++ +++
Internal capsule,
posterior limb + ++ +++ +++ +++ +++ +++ +++ +++ +++ +++
Internal capsule,
anterior limb + + ++ +++ +++ +++
Corpus callosum splenium + + ++ ++ ++ +++ +++
Corpus callosum genu + + ++ +++
Parieto-occipital WM + ++ ++ +++ +++ +++ +++ +++
Frontal WM + ++
Temporal WM +
Chapter 5

Lysosomes and Lysosomal Disorders


R.A. Wevers, V. Gieselmann

5.1 Lysosomal Biogenesis their site of synthesis. The signal that specifies the
and Biochemical Functions destination of each nascent protein resides in its se-
quence or spatial structure. The cellular transport
Lysosomes are hydrolase-rich organelles surrounded machinery recognizing these signals distributes the
by membranes and with an acidic interior milieu. proteins to the diverse cellular compartments. Some
They are present in almost all types of body cells. signal peptides direct proteins specifically into the
Their number varies greatly, depending on cell type nucleus, mitochondria, or peroxisomes. Membrane,
and function. They display considerable structural secretory, or lysosomal proteins are also sorted
heterogeneity and appear in all shapes, sizes, and den- initially via signal peptides into the lumen of the
sities. They have been given their name because they endoplasmic reticulum. Here the lysosomal enzyme
are small bodies (soma = body) containing various proteins undergo glycosylation, as do most of the
enzymes that are hydrolytic (lysis = dissolution). secretory and plasma membrane proteins. The glyco-
These hydrolases catalyze reactions in which macro- sylation step involves the transfer of a large oligosac-
molecules and macromolecular structures are de- charide with high mannose content to selected as-
graded into smaller components. Among the more paragine residues of the nascent protein. Subsequent-
than 50 different lysosomal enzymes so far identified ly, the signal peptide is cleaved, the protein folds, and
are proteases, nucleases, glycosidases, lipases, phos- the processing of the asparagine-linked oligosaccha-
pholipases, sulfatases, and phosphatases. The variety ride begins. From the endoplasmic reticulum the pro-
of enzymes enables the lysosome to digest almost all teins travel via vesicular transport to the Golgi appa-
types of biological macromolecules, such as proteins, ratus. In the cis compartment of the Golgi complex,
polysaccharides, lipids, and nucleic acids. The low- oligosaccharide side chains of lysosomal enzymes are
molecular components released are transported to phosphorylated and thus acquire mannose-6-phos-
the cytoplasm to be reused. For this purpose the lyso- phate moieties. In contrast, oligosaccharide side
somal membrane contains various transporters to chains of secretory and membrane proteins are
translocate amino acids, sugars, and possibly nu- trimmed and remodeled further to yield complex-
cleotides into the cytoplasm. The lysosomal mem- type side chains. The synthesis of the mannose-6-
brane separates the hydrolytic enzymes from the cy- phosphate residues is initiated by a phosphotrans-
toplasm to prevent uncontrolled lysis of cytoplasmic ferase, which specifically recognizes lysosomal en-
components. The acidic interior of lysosomes pro- zymes. Recognition does not occur by way of a signal
vides a favorable environment for the digestive activ- peptide but is mediated by a spatial signal depending
ities of the enzymes: oligomeric proteins dissociate on the three-dimensional structure of the enzymes.
into monomers, proteins dissociate away from the Given the structural diversity of lysosomal enzymes
protecting membrane, and stabilizing complexes be- the precise nature of the signal shared by all enzymes
come split. The low pH is generated by a complex is still a mystery. So far, only surface-located lysine
multisubunit ATP-dependent proton pump. Several residues seem to be an essential common component
subunits of this proton pump are found on the cy- of this topogenic signal.
tosolic side of the lysosomal membrane, and others Phosphorylated lysosomal enzymes then proceed
are integral lysosomal membrane proteins. Further- through the remainder of the Golgi complex (from
more, the lysosomal membrane harbors various pro- cis- through medial to trans-Golgi). In the trans-Gol-
teins, with highly glycosylated intralysosomal do- gi network (TGN) they bind to mannose-6-phosphate
mains (e.g. LAMP1 and 2). The high carbohydrate receptors, which segregate the enzymes into distinct
content is thought to protect the lysosomal mem- transport vesicles away from secretory and cell sur-
brane from hydrolytic attack by the enzymes. face proteins. There are two mannose-6-phosphate
Lysosomal enzymes, along with secretory proteins receptors, which bind different but overlapping sets of
and plasma membrane proteins, are synthesized on enzymes. Once lysosomal enzymes are bound, the
membrane-bound polyribosomes on the rough en- mannose-6-phosphate receptor–enzyme complexes
doplasmic reticulum. An important question is how are collected in clathrin-coated pits, which bud off to
proteins, which are destined for specific intracellular form coated vesicles. Most of these transport vesicles
compartments, are targeted at their destination from deliver the complexes to acidic early endosomes, but
5.1 Lysosomal Biogenesis and Biochemical Functions 67

complexes do also arrive at late endosomes. The low and almost all undergo proteolytic processing. The
pH in the endosomes causes lysosomal hydrolases pre-piece is the signal sequence, which is cleaved
and receptors to dissociate. After dissociation, man- immediately after transport into the endoplasmic
nose-6-phosphate receptors are retrieved from this reticulum. With the exception of aspartylgluco-
compartment and returned to the TGN, whereas lyso- saminidase, which is already processed in the endo-
somal enzymes are delivered to mature lysosomes. plasmic reticulum, the pro-piece is cleaved later in en-
Thus, receptors do not occur in lysosomes. Their ab- dosomal compartments. Cleavage is completed after
sence is an important histochemical feature of lyso- arrival of the enzymes in the lysosomes. For lysoso-
somes and differentiates them from late endosomes. mal proteases, cleavage of proenzymes is accompa-
Mannose-6-phosphate receptors also cycle between nied by activation of the enzymes. Prior to arrival in
the endosomal compartment and the plasma mem- the lysosomes the pro-piece keeps the proteases in an
brane. One of the receptors can bind lysosomal en- inactive state. In lysosomal enzymes other than pro-
zymes at the plasma membrane and mediate their en- teases, however, the biological significance of this
docytosis and subsequent delivery to lysosomes. This proteolytic processing is poorly understood.
is probably a recapture mechanism, since depending Some enzymes involved in the degradation of
on cell type, 5–40% of newly synthesized lysosomal sphingolipids need the assistance of enzymatically
enzymes escape receptor binding in the TGN and are inactive activator proteins for hydrolysis of their sub-
secreted. A proportion of these enzymes bind to the strates. So far, five different activator proteins encod-
mannose-6-phosphate receptors on the plasma mem- ed by two different genes have been identified. One
brane and are recaptured, internalized, and delivered gene codes for the GM2 ganglioside activator protein
to lysosomes. Sorting signals within the cytoplasmic only, whereas the other encodes a precursor protein
tails of the receptors are crucial for their correct in- that harbors four different but homologous sphin-
tracellular trafficking. golipid activator proteins (SAPs). The mature SAPs A,
Although the mannose-6-phosphate recognition B, C, and D – also called saposins – are generated from
pathway is a major route for targeting soluble lysoso- this precursor via proteolytic processing. They act on
mal enzymes, there is evidence for an alternative different enzymes and facilitate the degradation of
mechanism, independent of mannose-6-phosphate, various sphingolipids. They also differ in their mode
and localizing soluble acid hydrolases to lysosomes. of action. GM2-activator protein and SAP-B bind the
Although it seems likely that this pathway is also re- lipid substrates and present them to the respective
ceptor mediated, attempts to demonstrate this recep- enzymes, whereas SAP-C activates the enzyme direct-
tor have so far been unsuccessful. Only in cases of ly.
activator proteins – see below – has the multiligand Lysosomes are the final destination of endocytic,
receptor sortilin been shown to be involved in lysoso- autophagic and phagocytic routes. The endosomal
mal trafficking independent of mannose-6-phos- membranous network connects the lysosomes to the
phate. Golgi apparatus and the plasma membrane. Early en-
Lysosomal membrane glycoproteins travel the dosomes start to accumulate internal membranes,
same route as soluble enzymes from the rough endo- and as this accumulation proceeds they mature into
plasmic reticulum via the Golgi apparatus and endo- late endosomes. Since late endosomes are rich in lu-
somes to lysosomes. However, the transport of lyso- minal membranes they are also referred to as multi-
somal membrane glycoproteins to lysosomes is inde- vesicular bodies (MVBs) or multivesicular endo-
pendent of the mannose-6-phosphate receptor sys- somes. Lipid and protein composition of these lumi-
tem, depending rather on signals in their cytoplasmic nal membranes differs from that of early endosomes,
portion. An example is the classic lysosomal marker suggesting a specific partitioning event during their
enzyme acid phosphatase. It is synthesized as a trans- generation. Thus, for some proteins it has been shown
membrane precursor protein with a large luminal do- that tagging with ubiquitin directs them through this
main and a short cytoplasmic tail. After reaching the luminal compartment for lysosomal degradation,
TGN the enzyme precursor is repeatedly recycled be- whereas other proteins seem to be quite stable in
tween the cell surface and the endosomal compart- these membranes. This endocytotic lysosomal route
ment before reaching the lysosome. After its delivery can also be used to terminate growth factor receptor
to the lysosome, acid phosphatase undergoes proteo- signaling, a process that is crucial for cellular regula-
lytic processing of the membrane-anchoring domain, tion. Thus, ligand activation of epidermal growth fac-
resulting in conversion to a soluble form. Sorting tor receptor does not only activate downstream sig-
signals for this mannose-6-phosphate receptor-inde- naling pathways, but also induces endocytosis. Endo-
pendent pathway reside in the short cytoplasmic tail cytosed receptors may be cycled back to the plasma
of the acid phosphatase precursor. membrane for continuous signaling or can be deliv-
In addition to oligosaccharide processing, lysoso- ered to the lysosome for degradation, resulting in sig-
mal hydrolases are synthesized as pre-proenzymes, nal termination. Thus, the balance between recycling
68 Chapter 5 Lysosomes and Lysosomal Disorders

and lysosomal delivery has a key role in regulation of 5.2 The Pathobiochemistry
the signal intensity of at least some tyrosine kinase of Lysosomal Disease in Humans
receptors.
The MVBs/late endosomes can fuse homotypical- More than 45 different lysosomal diseases are cur-
ly, but they also fuse with lysosomes, forming a hybrid rently known in man (Table 5.1). They can be caused
organelle. The dense lysosomes can be regarded as by defects in the genes of individual lysosomal hydro-
storage granules of hydrolytic enzymes, which fuse lases, activator proteins, transporters, lysosomal
with late endosomes to perform their hydrolytic task membrane proteins, or enzymes modifying lysoso-
on the late endosome contents. During this process mal hydrolases. In general a profound deficiency with
continuous condensation occurs to recover lyso- residual activity of the respective protein <5% of nor-
somes from this hybrid organelle. mal is found in tissues of affected persons. Since un-
Autophagy is the process by which the cell se- degraded substrates cannot leave the lysosomes, or
questers parts of its own cytoplasm, often containing only very slowly, these organelles are converted into
entire organelles. In the first step, called autophagic storage granules, which steadily increase in size and
sequestration, a cytoplasmic membrane, which is number.
probably derived from the endoplasmic reticulum, Over the years several classes of lysosomal diseases
envelops a region of cytoplasm in a closed vacuole have been unraveled. In Table 5.1 and in the text be-
called an autophagosome. Through fusion, the se- low we have classified the lysosomal diseases primar-
questered material is transferred to lysosomes. The ily on the basis of functional characteristics, resulting
lysosomal membrane protein LAMP2 seems to be in seven main groups. Furthermore, we have taken ac-
essential for the maturation of autophagosomes. In count of well-established clinical entities that are of-
normal cells this process is important because of its ten based on the nature of the accumulating com-
participation in cell renewal and turnover of worn- pound.
out cell constituents. In secretory cells there is a spe-
cial kind of autophagy, called crinophagy. It occurs by
way of direct fusion between secretory granules and 5.2.1 Defects in Individual Hydrolases
lysosomes and results in the destruction of excess se-
cretory material. Alternatively, in chaperone-mediat- The molecular basis of most currently known lysoso-
ed autophagy proteins can be unfolded in the cyto- mal diseases is a deficiency of an individual hydrolase
plasm and transported directly through the lysoso- in the lysosome. These hydrolases have a role in the
mal membrane. catabolism of different molecular species, such as
Finally, phagocytosis is the process by which cells lipids, mucopolysaccharides, and glycoproteins. This
internalize large particles, such as bacteria. Thus, has led to a classification according to the biochemi-
phagocytosis is particularly active in neutrophils and cal nature of the substrate accumulating in a particu-
macrophages. After internalization the interior of a lar disease (see also Table 5.1).
phagosome initially resembles the extracellular mi- In many lysosomal diseases lipid species accumu-
lieu. However, phagosomes may fuse with endosomes late. Examples of lipids that are involved are sphin-
and slowly acquire the characteristics of late endo- gomyelin (Niemann-Pick A and B), glucosylceramide
somes and lysosomes. In this context it is important (Gaucher), galactosylceramide and psychosine
to note that lysosomes also generate peptides via hy- (Krabbe), globotriaosylceramide (Fabry), ceramide
drolysis of phagocytosed material to load MHCII (Farber), sulfatides (metachromatic leukodystrophy),
molecules. Thus, lysosomes have an essential role in gangliosides (GM1 and GM2 gangliosidoses), choles-
the immune system, maintaining the health of cells teryl esters, and triglycerides (Wolman). This group
and the body’s defense against foreign invaders. of diseases can be referred to collectively as the lipi-
Apart from the catabolic functions, lysosomes doses. Figure 5.1 illustrates the sequential action of
have also been shown to play an essential part in the the various enzymes involved in the pathway of lyso-
repair of plasma membrane defects. In wounded cells somal lipid metabolism.
lysosomes can fuse with the defective plasma mem- In a second subgroup acid mucopolysaccharides,
brane via a calcium-triggered exocytotic process. also called glycosaminoglycans, accumulate. These
Lysosomes can thus serve as a reservoir allowing for consist of long chains of repeating disaccharide units.
rapid provision of membrane lipids in the case of Different subspecies exist, which are characterized by
extended defects that cannot be compensated by lipid the composition of their repeating disaccharide units.
biosynthesis within an appropriate time period. This Heparan sulfate, dermatan sulfate, chondroitin sul-
clearly demonstrates that lysosomes also have ana- fate, keratan sulfate, and hyaluronan are representa-
bolic functions. tives of the mucopolysaccharide family. The mu-
copolysaccharides are degraded in the lysosome. De-
fects in the enzymes involved in this catabolism cause
5.2 The Pathobiochemistry of Lysosomal Disease in Humans 69

Table 5.1. A review of the lysosomal storage disorders, with the defective enzymes and proteins in each
A. Defects in individual lysosomal hydrolases
1. Lipidoses
a. Metachromatic leukodystrophy Arylsulfatase A
b. Globoid cell leukodystrophy (Krabbe disease) Galactocerebrosidase
c. GM1 gangliosidosis β-Galactosidase
d. GM2 gangliosidoses:
Tay Sachs disease Hexosaminidase A
Sandhoff disease Hexosaminidase A and B
e. Gaucher disease β-Glucosidase
f. Fabry disease α-Galactosidase A
g. Farber disease Acid ceramidase
h. Niemann-Pick disease (types A and B) Sphingomyelinase
i. Wolman disease and cholesterol ester storage disease Acid lipase
2. Mucopolysaccharidoses
a. Hurler disease and Scheie disease (I) α-Iduronidase
b. Hunter disease (II) Iduronate sulfatase
c. Sanfilippo disease (IIIA-D) a. Heparin sulfamidase
b. N-Acetyl α-glucosaminidase
c. α-Glucosaminide N-acetyltransferase
d. N-Acetylglucosamine 6-sulfatase
d. Morquio disease (IV) Galactose 6-sulfate sulfatase
e. Maroteaux-Lamy disease (VI) Arylsulfatase B
f. Sly disease (VII) β-Glucuronidase
g. Hyaluronidase deficiency (IX) Hyaluronidase
3. Disorders of glycoprotein degradation
a. Sialidosis Neuraminidase
b. Fucosidosis α-Fucosidase
c. Mannosidosis (α and β) α− and β-Mannosidase
d. Aspartylglycosaminuria Aspartylglucosaminidase
4. Glycogen storage disorders
a. Pompe disease α-Glucosidase
5. Neuronal ceroid lipofuscinoses
a. Infantile Finnish type NCL (Santavuori disease) Palmitoyl thioesterase
b. Late-infantile NCL (Jansky-Bielschowsky disease) Tripeptidyl peptidase1
6. Non classifiable
a. Pyknodysostosis Cathepsin K
b. Schindler disease N-Acetyl α-galactosaminidase
B. Defects in activator proteins
1. Lipidoses
a. GM2 activator protein deficiency GM2 Activator protein
b. Saposin B deficiency Metachromatic leukodystrophy variant
c. Saposin C deficiency Gaucher disease variant
d. Prosaposin deficiency
C. Defects in the postsynthetic modification of lysosomal proteins
1. Mucolipidoses
a. I cell disease (mucolipidosis II) N-Acetylglucosaminylphosphotransferase
b. Pseudo-Hurler polydystrophy (mucolipidosis III) N-Acetylglucosaminylphosphotransferase
2. Non-classifiable
a. Multiple sulfatase deficiency FGly-Generating enzyme
D. Defects in structural lysosomal proteins
1. Glycogen storage disorders
a. Danon disease LAMP2
E. Defect in a protective protein
a. Galactosialidosis Cathepsin A
F. Defects in transport and trafficking of substrates
a. Cystinosis Cystinosin
b. Salla disease Sialin
c. Niemann-Pick disease type C NPC1 or NPC2
G. Non-classifiable
a. Mucolipidosis IV Mucolipin 1
70 Chapter 5 Lysosomes and Lysosomal Disorders

Fig. 5.1. Disorders in lysosomal


lipid metabolism

a group of diseases collectively referred to as the mu- The NCLs collectively constitute the most common
copolysaccharidoses. group of neurodegenerative diseases in childhood,
When glycoproteins are degraded in the lysosome, with an estimated total incidence in the U.S. of
breakdown of the glycan part requires a set of glycosi- 1:12 500. All NCL forms share unifying pathomor-
dases. Degradation of the glycans involves their se- phologic features. In two subtypes the primary defect
quential action. If one of these enzymes is deficient was found in a soluble lysosomal enzyme. The en-
various oligosaccharides deriving from the glycan zymes involved are protein thioesterase (PPT) and
accumulate in tissues and body fluids. The material tripeptidyl-peptidase 1 (TPP1). The PPT enzyme re-
accumulating in the lysosomes is disease specific. As moves fatty acids from several S-acetylated proteins.
a group, these diseases can be referred to as glycopro- The function of TPP1, a serine protease, is the re-
tein degradation disorders. moval of N-terminal tripeptides from substrates with
The breakdown of glycogen, a glucose polymer, free amino termini. The in vivo substrates of PPT and
requires lysosomal α-glucosidase enzymatic activity. TPP1 remain unknown. It may turn out that some of
Defects in this enzyme lead to Pompe disease with the remaining eight genetic NCL forms are also lyso-
massive glycogen storage in muscle and liver in the somal diseases.Another lysosomal enzyme, cathepsin
infantile-onset form of the disease. This disease is one D, is for instance involved in an ovine neurodegener-
of the family of glycogen storage disorders. ative disease with ultrastructural features closely re-
A last group in this category is formed by the neu- sembling human NCL. The human counterpart of this
ronal ceroid lipofuscinoses (NCL). To date, eight main disease has not yet been identified.
genetic forms are generally accepted to occur in man.
5.2 The Pathobiochemistry of Lysosomal Disease in Humans 71

Although the classification of lysosomal disorders fect in the targeting of lysosomal enzymes towards
according to storage compounds appears straightfor- the lysosome. This causes an intracellular deficiency
ward, it is important to realize that lysosomal en- of many lysosomal enzymes. The diagnosis can be
zymes are frequently specific not for a certain sub- confirmed by abnormally high enzymatic activity of
strate, but for a component that may occur in differ- many lysosomal enzymes in blood plasma. Because of
ent substrates. Thus, glycosidases are specific for par- the mistargeting, these enzymes are directed out of
ticular sugar residues and the geometry of their the cell and end up in the blood plasma. Defects in the
linkage. The respective sugar and its linkage can phosphotransferase cause mucolipidosis II (inclusion
occur both in lipids and in glycosaminoglycans. body or I cell disease) and III. Patients with mucolipi-
Degradation of both is affected in the case of enzyme doses II and III share clinical symptoms and bio-
deficiency, and both lipids and glycosaminoglycans chemical characteristics with patients who have a
accumulate. Thus, in some diseases the classification mucopolysaccharidosis or a sphingolipidosis. Glyco-
is somewhat artificial. lipids as well as mucopolysaccharides accumulate in
In Schindler disease the defective enzyme, α-N- lysosomes in these diseases. Recently the primary
acetylgalactosaminidase, is involved in various path- defect in mucolipidosis IV was found to be in the
ways, so that this disease cannot be assigned unam- MCOLN1 gene encoding for a protein, mucolipin 1.
biguously to any one of the above groups. It exerts an The function of the protein has not yet been fully
action in the catabolism of various glycoconjugates characterized, and this disorder is therefore nonclas-
with terminal α-N-acetylgalactosaminyl residues. sifiable (group G in Table 5.1).
Deficiency of the enzyme results in accumulation of Among the lysosomal storage disorders multiple
glycopeptides, glycosphingolipids, and oligosaccha- sulfatase deficiency is particularly interesting. In this
rides in many tissues. disorder the activity of all lysosomal and nonlysoso-
The defective enzyme in pyknodysostosis has been mal sulfatases is reduced. Since sulfate groups occur
found in cathepsin K, a lysosomal protease. This in many different molecules a complex mixture of
enzyme is highly expressed in osteoclasts. The defect compounds accumulates. The enzymatic activity de-
leads to a reduced capacity of this cell to remove pends on a formylglycine residue (FGly) in the active
organic bone matrix, thus causing defective bone center of all sulfatases. This amino acid residue is gen-
growth and remodeling. This explains why the pa- erated by a posttranslational modification from a cys-
tients suffer predominantly from skeletal, orthopedic, teine residue. Patients with multiple sulfatase defi-
craniofacial, and dental abnormalities. ciency have defects in the SUMF1 gene. The protein
product of the SUMF1 gene is the FGly-generating en-
zyme (=FGE) localized in the lumen of the endoplas-
5.2.2 Defects in Activator Proteins mic reticulum. The function of this enzyme is to gen-
erate the formylglycine residue in the catalytic center
Some enzymes require the presence of activator pro- of the sulfatases. When this modifying reaction is de-
teins or saposins for their catalytic function inside fective the sulfatases remain inactive. This causes ac-
the lysosome. Examples are sphingomyelinase, aryl- cumulation of the various substrates. Therefore sev-
sulfatase A, and α- and β-galactosidase. Since defects eral compounds could be identified as storage mater-
in activator proteins affect the degradation of sphin- ial. The diagnosis can be established biochemically at
golipids only, all activator protein deficiencies are the enzyme level by measuring various sulfatases in
lipidoses. The clinical signs and symptoms frequent- leukocytes, or preferably in fibroblasts.
ly resemble those found in patients in whom the same
glycolipid accumulates as the result of deficiency of
hydrolase activated by the respective saposins (e.g., 5.2.4 Defects in Structural
saposin B deficiency causes a variant form of meta- Lysosomal Proteins
chromatic leukodystrophy).
Lysosomes have several structural proteins. Examples
of such ubiquitous, highly glycosylated integral mem-
5.2.3 Defects in the Postsynthetic brane proteins are LAMP1 and LAMP2 (LAMP =
Modification of Lysosomal Proteins lysosome-associated membrane protein). They ac-
count for about 50% of the protein content of the
As outlined earlier, all soluble lysosomal enzymes are lysosomal membrane. Recently the primary defect of
N-glycosylated and their oligosaccharide side chains Danon disease has been assigned to the LAMP2 gene.
receive mannose-6-phosphate residues, which are a This gene encodes LAMP2, which is thought to be a
lysosomal targeting signal, in the Golgi apparatus. structural protein in the lysosome. This X-linked dis-
Defects in a phosphotransferase initiating the synthe- ease is characterized by lysosomal glycogen storage
sis of mannose-6-phosphate residues result in a de- leading to cardiomyopathy and myopathy in patients
72 Chapter 5 Lysosomes and Lysosomal Disorders

with normal α-glucosidase activity. It is not fully clear known function that is thought to work in a coordi-
how a defect in this protein can lead to accumulation nate fashion with NPC1 protein.
of glycogen. It can be anticipated that several new
lysosomal diseases in this subgroup will be found in
the future. 5.3 Clinical Features and Diagnosis

A full survey of clinical symptomatology of lysosomal


5.2.5 Defects in a Protective Protein diseases is beyond the scope of this chapter. A few
characteristics or general features can clearly be un-
Galactosialidosis is caused by a defect in cathepsin A. derstood from the molecular basis of the diseases.
This protein has a dual function: it is not only a pro- Storage material often gives rise to organomegaly, for
tease, but also a protective protein. It combines with instance of liver or spleen.Another characteristic that
neuraminidase and β-galactosidase in an early may occur is the loss of acquired mental or motor
biosynthetic compartment. By virtue of this associa- skills in the course of time, which is due to an increase
tion the complex is correctly delivered to the lyso- in storage material with time. Some clinical features,
somes. In the lysosome, cathepsin A protects the neu- such as a cherry red spot in the retina or downward
raminidase and β-galactosidase against rapid prote- gaze paralysis, may be highly suggestive for lysosomal
olysis and inactivation. In the case of cathepsin A disease, and in some cases even pathognomonic for a
deficiency both enzymes are rapidly degraded and specific disease. The same holds in the case of evi-
thus deficient. Sialyloligosaccharides accumulate in dence for storage material in body fluids or tissues.
the lysosomes of affected patients and are also excret- Vacuolization may occur in peripheral white blood
ed in the urine. cells. The finding of sea blue histiocytes in bone mar-
row should also be followed up with a thorough work-
up for lysosomal diseases. As most cell types in the
5.2.6 Defects in Transport and Trafficking human body contain lysosomes, many tissues or cell
of Substrates types can be involved in lysosomal diseases. Often
these diseases affect the CNS, resulting in neurode-
Lysosomal degradation of macromolecules leads to generative disease. Others, such as Morquio and
the formation of smaller molecules, which generally Pompe disease, leave the brain relatively unaffected.
are exported from the lysosome towards the cyto- In general, the lysosomal diseases are multisystem
plasm. Some molecules require specific carriers to diseases. Pyknodysostosis is an example of a disease
leave the lysosome. Defects in such carriers lead to ac- in which the molecular defect, the deficiency of
cumulation of the molecule involved within the lyso- cathepsin K, seems to interfere predominantly with
some. Examples of such diseases are cystinosis and the function of only one cell type. Dysfunction of the
Salla disease. Cystinosis is characterized by intralyso- osteoclast causes the clinical features of this disease.
somal storage of the amino acid cystine and is caused Most lysosomal diseases show significant clinical
by defective carrier-mediated transport of cystine heterogeneity. The onset of clinical signs and symp-
across the lysosomal membrane. The protein in- toms can occur in any decade of life, and even before
volved is cystinosin. In Salla disease intralysosomal birth. Hydrops fetalis has been observed as a presen-
storage of sialic acid occurs, caused by a defect in its tation form in several lysosomal diseases. β-Glu-
transport across the lysosomal membrane by the curonidase deficiency is an example of a disease that
transporter sialin. can present as early as this. The time of presentation
Niemann-Pick disease type C is a lipid trafficking can vary rather widely within one disease. Pompe dis-
disorder. The majority of patients have mutations in ease, for example, can have an early infantile onset: in
the gene coding for the NPC1 protein. The postulated such patients the course of the disease is invariably
role for this protein involves modulation of the vesic- very severe and most of them die before the age of
ular trafficking of cholesterol and glycolipids. Several 6 months. Other patients with Pompe disease have
lipids (sphingomyelin, phospholipids, glycolipids, adult-onset forms of the disease and have milder
and unesterified cholesterol) are stored in excess in symptoms. The concept for our understanding of this
the liver and spleen of these patients. Foam cells or variability in clinical presentation lies in the residual
sea blue histiocytes may be found in many tissues of activity of the enzyme in a specific patient. However,
affected patients. Primarily, the diagnosis requires the with the methodology currently available it is not
demonstration of excess cholesterol in fibroblasts possible to predict the disease course from the resid-
with the so-called filipin staining test. In a small sub- ual activity in leukocytes or fibroblasts.
group of patients with Niemann-Pick type C disease In recent years numerous mutations of genes for
there are mutations in another gene coding for the proteins that are deficient in lysosomal storage dis-
NPC2 protein, a soluble lysosomal enzyme with un- eases have been described, leading to a better under-
5.3 Clinical Features and Diagnosis 73

standing of the biochemical consequences of muta- diagnosis. An increased concentration of urinary


tions. Severe mutations that truncate the protein or mucopolysaccharides can be found in the mu-
shift the reading frame thereby alter the primary copolysaccharidoses. Subsequent electrophoresis of
structure of the protein so that it has no residual bio- mucopolysaccharides will show which subtype of the
logical activity. Such mutations almost always result mucopolysaccharides the patient cannot adequately
in a severe clinical phenotype. In lysosomal storage degrade. Defects in mucopolysaccharide catabolism
diseases missense mutations are the most frequent. can affect the breakdown of heparan sulfate, der-
Often missense mutations lead to misfolded enzymes, matan sulfate, chondroitin sulfate, keratan sulfate, or
which are not transported to the lysosomes but are hyaluronan. The result of electrophoresis will give a
retained and degraded in the endoplasmic reticulum. clue to the defective enzyme. Abnormal urinary
Alternatively, defective enzymes may still be sorted oligosaccharides are present in the disorders of glyco-
properly but become rapidly degraded on arrival in protein degradation shown in Table 5.1. Thin layer
the lysosome. Mutations can be located in the active chromatography of oligosaccharides is also diagnos-
center of an enzyme or indirectly influence the cat- tic in infantile Pompe disease, where a tetraglucoside
alytic activity of the enzyme. In some cases a combi- deriving from glycogen accumulates in the urine. In
nation of these effects is the cause of enzyme defi- late-onset Pompe disease this tetraglucoside is gener-
ciency. Missense mutations can influence the catalyt- ally not found in the urine and other techniques are
ic activity of the enzyme as badly as truncating muta- necessary to establish the diagnosis. In some diseases,
tions. When they occur in less relevant parts of the then, accumulating material cannot be detected in the
coding region they may allow residual activity, result- urine, and for some diseases the techniques that
ing in a milder clinical phenotype. It is not always would be required to diagnose them at the metabolite
possible to show a clear genotype–phenotype rela- level are too time consuming. This is the case for the
tion. Most lysosomal diseases have an autosomal lipidoses, defects in activator proteins, and the NCLs,
recessive mode of inheritance. Few diseases have an for instance. In such cases direct enzyme analysis in
X-chromosomal inheritance. Fabry, Danon, and leukocytes is often used as a first step in establishing
Hunter diseases are examples. Males have more se- the diagnosis. Cultured skin fibroblasts can generally
vere clinical symptoms, as they only have one X chro- also be used to confirm the diagnosis. A further test
·
mosome. However, female carriers (=XX) can also that may be relevant to the demonstration of lysoso-
have clinical symptoms of the disease because of un- mal involvement is measurement of the activity of
even X inactivation (lyonization). chitotriosidase. This enzyme is secreted by activated
To confirm a clinical suspicion of a lysosomal dis- macrophages. In several lysosomal diseases increased
ease at the biochemical level various approaches can chitotriosidase activity can be used as a nonspecific
be used as diagnostic strategy. In some lysosomal dis- diagnostic marker.
eases undegraded substrates can be found in the The chapters below discuss only those lysosomal
urine. Investigations of urine samples are therefore storage disorders that are accompanied by a white
often used as a first step towards establishing the matter disorder.
Chapter 6

Metachromatic Leukodystrophy

6.1 Clinical Features velop. On rare occasions the clinical picture is domi-
and Laboratory Investigations nated by extrapyramidal features. Clinical symptoms
of a peripheral neuropathy are often lacking, and
Metachromatic leukodystrophy (MLD) is an autoso- deep tendon reflexes are usually brisk. Optic atrophy
mal recessive progressive disorder. Its incidence is develops. Seizures occur in about 50% of the patients.
estimated to be 1:40,000. The disease can be divided Eventually complete tetraplegia with decerebration
into three subtypes: the late infantile (40% of the pa- posture, brain stem dysfunction, and profound de-
tients with MLD), juvenile (40%), and adult (20%) mentia evolves. Death usually occurs 5–10 years after
variants. This subdivision is based on the age at onset, onset.
the duration, and the clinical picture of the disease.As Some authors prefer to divide the juvenile variant
a rule, only one variant of MLD occurs within one into two subgroups. An early juvenile variant has its
family, although exceptions have been reported. onset between 4 and 6 years. The clinical symptoma-
The age of onset in the late infantile variant varies tology resembles that of the late infantile variant,
between 6 months and 3 years. Of the different sub- showing gait disturbance and other motor dysfunc-
types, this variant is the one that shows the greatest tion as early manifestations. The late juvenile variant
uniformity with regard to clinical picture and course has its onset between 6 and 16 years. In the clinical
of disease. Most children learn to sit and walk with symptomatology, behavioral abnormalities, poor
support normally, but show a delay in walking unaid- school performance, and language regression pre-
ed. The first symptom is usually an unsteady gait due dominate as early abnormalities.
to muscle hypotonia. There are signs of a progressive The adult form usually reveals itself between 16
polyneuropathy, ending in a generalized flaccid pare- and 30 years. Onset of the disease at 60 years or later
sis of the arms and legs and a loss of tendon reflexes. has also been described. Most patients experience a
The neuropathy may be painful. Cerebellar ataxia is gradual decline in intellectual abilities. At onset the
also often an early sign. Nystagmus is usually present. clinical picture is often dominated by emotional labil-
Speech development is disturbed and dysarthria be- ity, behavioral abnormalities, or psychiatric symp-
comes manifest. Mental development stagnates and toms such as delusions and hallucinations. It is not
regression occurs. Gradually, involvement of the uncommon for the patient to be treated initially for
pyramidal system causes the flaccid paresis to be su- schizophrenia or a psychotic depression. After sever-
perseded by a spastic tetraplegia with abnormal re- al months or years progressive spastic paresis of the
flexes, such as extensor plantar reflexes, but deep ten- arms and legs develops, with increased tendon reflex-
don reflexes are absent. Bulbar and pseudobulbar es and extensor plantar reflexes. Cerebellar ataxia and
symptoms develop, leading to feeding difficulties. such extrapyramidal features as choreiform move-
Speech is affected, and the child eventually becomes ments and dystonia may be present. Signs of periph-
mute. Optic atrophy with impaired vision is present. eral neuropathy are often absent, although flaccid
Epileptic seizures occur in about 25% of the children. tetraparesis may occur in the terminal stage. Optic at-
Eventually the child becomes blind and completely rophy and signs of bulbar dysfunction may appear.
tetraplegic in a decerebrate state without purposeful Epileptic seizures are rare. A state of severe dementia
movements. This final stage may last for several years. gradually develops. The patient loses contact with the
Death usually occurs about 5 years after the onset of surroundings and lies in a decorticate or decerebrate
clinical symptoms. posture; eventually a persistent vegetative state is
The age of onset in the juvenile variant ranges from reached. The duration of the disease varies from a few
4 to 16 years. The disease occurs in apparently years to 15 years and longer. Although a rapid deteri-
healthy, intellectually normal children. Early signs are oration is seen in some patients, in most cases pro-
a gradual deterioration in school performance, lan- gression is slow over a period of years. Another, more
guage regression, and clumsiness. There are usually rare presentation in adults is dominated by signs of a
also emotional and behavioral disturbances. These peripheral neuropathy.
symptoms may be present for several months and up CSF protein is elevated in the late infantile form
to a year before the onset of other neurological signs. and in most juvenile cases. In the adult form, CSF pro-
A spastic paresis and cerebellar ataxia gradually de- tein this is less often the case. The EEG is normal at
6.2 Pathology 75

the beginning of the disease. At later stages it shows Initially, the involvement of the white matter in
nonspecific abnormalities in the form of slowing of MLD is patchy, but after some time all the white mat-
the background pattern, often together with paroxys- ter is affected, often in a symmetrical fashion, so that
mal or epileptiform activity. In the infantile and juve- the demyelinated lesions of the two hemispheres have
nile variants, the conduction velocity of peripheral a butterfly configuration. Sometimes, in cases of long
nerves is markedly reduced. Particularly in adults, duration, the white matter is reduced to a narrow
however, the nerve conduction velocity may be nor- strip 1–2 cm in diameter and shrinkage of the white
mal. matter has led to enlargement of the ventricles. De-
In all patients urinary sulfatide excretion is in- myelination occurs predominantly in the cerebral
creased. The activity of arylsulfatase A in urine and in hemispheres, especially in the centrum semiovale.
peripheral leukocytes is low, but may be normal in ex- Demyelination tends to be most intense in the
ceptional cases of activator deficiency (see below). In periventricular area, diminishing towards the surface;
all cases of MLD a decreased catabolism of exogenous the arcuate fibers are relatively spared. The internal
sulfatide by cultured fibroblasts can be demonstrated. capsule, cerebral peduncles in the midbrain, pyrami-
In this test sulfatide is radiolabeled and added to cul- dal tracts in the pons, and pyramids in the medulla
tured fibroblasts. The uptake of label by cells with are severely affected, but other brain stem tracts are
subsequent metabolism is measured at frequent in- usually only moderately or slightly demyelinated. The
tervals. This procedure is not performed routinely, cerebellar white matter can also be demyelinated, but
but only in specific situations. The definitive diagno- usually less so than the cerebral white matter.
sis of MLD is based on the determination of the activ- Microscopic examination shows demyelination
ity of arylsulfatase A in peripheral leukocytes or fi- with paucity or complete loss of myelin from affected
broblasts. The diagnostic reliability of this test is areas. Axons are relatively spared, but their density is
hampered by the relatively frequent occurrence of so- reduced in severely affected areas. There is prolifera-
called pseudodeficiency. In this condition low aryl- tion of astrocytes with fibrous gliosis. At the edge of
sulfatase A activity is found without associated clini- affected areas and scattered throughout the lesions
cal signs of MLD (see also section 6.4). For this rea- there are macrophages that contain the specific
son, sulfatide excretion in urine should be used to degradation products. Usually oligodendroglia are
confirm the diagnosis of MLD. It is increased in all absent from lesions and are reduced in number even
cases of MLD, but normal in pseudodeficiency. DNA in areas where the myelin is still intact. No inflamma-
analysis is another option. tory cells are present in the lesions.
In cases in which symptoms are strongly sugges- MLD is characterized by the deposition of
tive of MLD and the enzyme activity in peripheral metachromatically staining material in the white
leukocytes is normal, deficiency of arylsulfatase A ac- matter. The term ‘metachromasia’ designates the phe-
tivator protein can be surmised. Additional tests then nomenon of certain cationic dyes changing their col-
include the measurement of urinary sulfatides and or from blue to pink or brown when bound to certain
the assessment of fibroblast sulfatide catabolism. A anionic groups present in several organic com-
sural nerve biopsy can be performed to demonstrate pounds. In MLD, sulfatides are the organic com-
the deposition of metachromatic material. pounds responsible for the metachromasia. The
Determination of the enzyme activity in cultured metachromatic material is mainly stored in the cyto-
chorionic villi or cultured amniotic fluid cells allows plasm of the proliferated glial cells and macrophages,
prenatal diagnosis. Assessment of sulfatide catabo- although some is also found in the oligodendroglia
lism can be performed in amniotic fluid cells or fetal cells, in the neurons of cranial nerve nuclei, basal gan-
fibroblasts. DNA-based prenatal diagnosis is also an glia, and spinal cord, and seemingly extracellularly as
option. The detection of heterozygotes by determin- free granules in the white matter. The greatest densi-
ing enzyme activity in leukocytes and DNA tech- ty of metachromatic deposits is seen in macrophages
niques facilitates genetic counseling. in perivascular spaces.
The cerebral cortex is relatively intact. Loss of neu-
rons is slight or absent. The cortical neurons contain
6.2 Pathology no metachromatic material. The cerebellar cortex is
normal or may show a diffuse loss of granular cells. A
Gross inspection of the brain reveals quite a firm con- decrease in the number of Purkinje cells may occur.
sistency in most cases. The brain may be enlarged and In certain areas, metachromatic granular material is
heavier than normal, but in later stages reduced size stored in the neuronal perikarya, although rarely in
is usually found. The cut surface shows discoloration large amounts. Neuronal metachromatic deposits are
of the white matter. preferentially found in the globus pallidus, thalamus,
76 Chapter 6 Metachromatic Leukodystrophy

subthalamic nucleus, hypothalamus, geniculate nu- 6.4 Pathogenetic Considerations


cleus, amygdala, and dentate nucleus. The cerebral
and cerebellar cortex, claustrum, and caudate nucle- MLD is a sphingolipidosis caused by deficient activi-
us, and also certain brain stem nuclei, tend to be ty of the lysosomal enzyme arylsulfatase A (= cere-
spared. broside-3-sulfate sulfatase = cerebroside-3-sulfate-3-
Electron microscopy demonstrates inclusions sulfohydrolase = sulfatide sulfatase). This enzyme
bounded by a membrane of lysosomal origin. The catalyzes the hydrolysis of sulfatide, the sulfate ester
morphological organization of the material varies in of cerebroside. Desulfation is the first step in the
appearance, probably because of a difference in the metabolic degradation of sulfatides. Following this
lipid composition or in the physicochemical state of sulfate cleavage, cerebroside is then degraded by cere-
the lipids. Prismatic and tuffstone-like profiles are broside galactosidase.
characteristic. In addition to the inclusions that stain The three clinical forms of the disease (late infan-
metachromatically on light microscopic examination, tile, juvenile, and adult onset) can be explained in part
lamellar inclusions are present in glial cells formed by different levels of residual enzyme activity. The
from fragments of degenerated myelin. The ‘extracel- arylsulfatase A gene, ARSA, is located on chromo-
lular’ deposits of metachromatic material described some 22q13.3. Many different mutations have been
in light microscopic studies appear to be cytoplasmic found in the MLD gene. There is evidence of a geno-
processes containing inclusions in electron mi- type–phenotype correlation in MLD. Some mutations
croscopy. have been exclusively associated with either the in-
In MLD the peripheral nerves are affected by seg- fantile-onset or the later onset variants of the disease.
mental demyelination. Signs of remyelination may be Patients with two mutations that cause complete loss
found. Metachromatic material is present in Schwann of enzyme activity always suffer from the early-onset
cells and macrophages. form of the disease; patients with two mutations that
Not only are there neuropathological changes in lead to a low residual enzyme activity usually have the
MLD, but also visceral abnormalities. Outside the adult-onset form of the disease, whereas patients who
CNS, metachromatic material is found in the liver, are compound heterozygous for ‘severe’ and ‘mild’
spleen and lymph nodes, gallbladder, pancreas, kid- mutations usually have intermediate phenotypes.
neys, adrenal glands, ovaries, ganglion cells of the Individuals have been found with low (approxi-
retina, and leukocytes of peripheral blood and bone mately 10–20%) arylsulfatase A activity but no clini-
marrow. Storage in these organs is limited to certain cal abnormalities. This is called arylsulfatase A pseu-
cell types. Visceral accumulations are not accompa- dodeficiency. Individuals who are compound het-
nied by further morphological changes or obvious erozygotes for the pseudodeficiency (PD) mutation
clinical dysfunction. Although MLD becomes mani- and an MLD mutation have 6–10% residual arylsulfa-
fest only with neurological abnormalities, it is clear tase A activity but do not develop MLD. Thus, arylsul-
that it is a generalized metabolic disorder. fatase A activities only slightly higher than those
encountered in patients with two mild mutations
(2–5%) are sufficient to sustain a normal phenotype.
6.3 Chemical Pathology Pseudodeficiency for arylsulfatase A causes diag-
nostic problems Pseudodeficiency is related to two
Biochemical analysis of the white matter shows a common polymorphisms, which are usually found
greatly increased amount of sulfatide with a concomi- together. These mutations can reduce arylsulfatase A
tant decrease in cerebroside as the major chemical activity to 10% of its control level, but do not lead to
abnormality. Whereas in normal white matter the ra- clinical symptoms. The allele frequency for pseudo-
tio of cerebroside to sulfatide is approximately 4 : 1, in deficiency is much higher (7–15%) than the MLD
MLD it can be below 1. There is not only evidence that allele frequency (0.5%). Because homozygous pseudo-
the membrane-bound deposits seen in this disease deficiency is frequent (0.5–2% of the population), it is
contain sulfatide, but also that myelin, which still ap- not uncommon for patients with pseudodeficiency
pears normal ultrastructurally, has an abnormally and neurological symptoms of unknown origin to be
high sulfatide content. Other biochemical changes in misdiagnosed as having MLD. There are also serious
the white matter are a consequence of loss of myelin problems with prenatal diagnosis of MLD in families
with a decrease in cholesterol, phospholipid, and gly- in which the parents carry an MLD allele and a pseudo-
colipids other than sulfatides. There is no increase in deficiency allele: MLD and pseudodeficiency cannot
cholesterol esters. There are relatively few chemical be distinguished on the basis of enzyme activity de-
changes in gray matter. terminations using artificial substrates. Measurement
of sulfatide excretion in urine and assays measuring
the in vivo degradation of sulfatide in cultured fibro-
6.4 Pathogenetic Considerations 77

blasts allow the distinction between MLD and pseudo- branes of organelles. Sulfatide is predominantly pre-
deficiency. Both are abnormal in MLD and normal in sent in membranes of myelin-producing cells and in
pseudodeficiency. Demonstration of metachromatic myelin. The amount of this substance normally pre-
material in a sural nerve biopsy is also diagnostic for sent in membranes of other organs is much lower. As
MLD.Alternatively, the pseudodeficiency allele can be a result of the block in catabolism, sulfatides accumu-
determined directly by DNA techniques, on the basis late in tissues that normally synthesize them. In the
of knowledge of the underlying sequence alterations. first place, they accumulate in membranes of myelin-
A problem encountered with this technique, however, producing cells and myelin sheaths, and within the
is that given the high frequency of the pseudodefi- lysosomes of these cells. The membrane build-up is
ciency allele, MLD mutations may also occur within basically normal in MLD. It is the membrane turnover
the pseudodeficiency allele, rendering it nonfunc- that is abnormal. Sulfatide cannot be degraded and is
tional. The frequency of MLD mutations in the nor- trapped in the membrane. Simultaneously, the cere-
mal and the pseudodeficiency allele is similar, so that broside content decreases as the conversion of sul-
0.5% of the pseudodeficiency alleles harbors an MLD fatide to cerebroside is impeded.
mutation. This finding calls for caution in the diagno- A number of pathogenetic mechanisms have been
sis of pseudodeficiency by DNA tests detecting the invoked to explain the demyelination in MLD. One of
mutations of the pseudodeficiency allele. There is them is the fact that the myelin composition in MLD
evidence that pseudodeficiency mutations may con- becomes increasingly abnormal and that therefore
tribute to the phenotype when concurrent with dis- myelin possibly becomes increasingly unstable. As
ease causing mutations. Mutations that cause the lat- soon as the disturbance of the normal physicochemi-
er onset forms of MLD when they occur in the normal cal stability reaches a critical point, demyelination
arylsulfatase A gene may lead to a more severe form starts. Another explanation proposed is that lysoso-
when they occur in the pseudodeficiency gene. mal storage of sulfatides in oligodendroglia and
A small subgroup of MLD patients are not defi- Schwann cells leads to cellular dysfunction and death,
cient in arylsulfatase A, but in an activator protein resulting in loss of all myelin sheaths maintained by
that is essential for the enzymatic action of arylsulfa- these cells. Indeed, changes in the subcellular or-
tase A. The gene for the precursor of this protein, ganelles, especially an increase in the numbers of
PSAP, is located on chromosome 10q22.1. This gene lysosomes of these cells, have been observed before
has been shown to code for a large precursor the detection of any morphological abnormalities in
polypeptide, prosaposin, which is processed to yield the myelin sheaths associated with them. A third
four different activator proteins. Sphingolipid activa- mechanism that has been proposed is that sulfogalac-
tor protein B, also called saposin B, SAP-B, or SAP-1, tosylsphingosine, a compound closely related to the
activates the hydrolysis of sulfatide by arylsulfatase A. cytotoxic compound galactosylsphingosine or psy-
In addition, it activates the hydrolysis of GM1-gan- chosine in globoid cell leukodystrophy, might accu-
glioside and globotriaosyl ceramide by β-galactosi- mulate and cause the death of oligodendrocytes and
dase and α-galactosidase, respectively. The protein Schwann cells. However, there is no evidence for the
interacts with the substrate and solubilizes it for en- enzymatic conversion of sulfatide to sulfogalactosyl-
zymatic hydrolysis. The deficiency of SAP-B causes a sphingosine, and the concentration of this substance
disease clinically resembling juvenile or late infantile is not elevated in MLD.
MLD, but with histochemical and ultrastructural The sulfatide accumulation in other organs (kid-
evidence of storage of gangliosides and other glyco- ney, liver, pancreas, adrenal, gallbladder and intestinal
sphingolipids. The diagnosis is established in these tract) does not lead to impairment of functions. Only
cases by revealing metachromatic material in sural the gallbladder shows progressive functional impair-
nerve biopsy, by finding an increased urinary sul- ment attributable to sulfatide accumulation, but
fatide excretion, and by demonstrating deficient gallbladder disease does not contribute to the fatal
turnover of sulfatide in the loading test in cultured outcome of MLD. The tolerance of these tissues
fibroblasts, all in the presence of normal arylsulfatase for sulfatides may be related to the fact that these
A activity. It can be shown in cultured fibroblasts that organs have an excretory function and can discharge
the defect in sulfatide catabolism can be corrected by the accumulating lipid from the cell into urine, bile,
adding activator protein. Deficient turnover of the or other fluids. Another important factor is that the
other glycosphingolipids can also be shown in load- sulfatide content of the cellular membranes in these
ing tests in fibroblasts. organs is normally much lower than that of the
Sulfatides are membrane lipids. They are impor- myelin membrane, which has a remarkably high
tant constituents of cell membranes, including myelin content of galactosphingolipids (cerebroside and sul-
sheaths. Within the cell they are present in the mem- fatide).
78 Chapter 6 Metachromatic Leukodystrophy

6.5 Therapy 6.6 Magnetic Resonance Imaging

Various forms of therapy have been attempted in an The CT scan findings in MLD are symmetrical, dif-
effort to alter the natural course of the disease, but fuse decreases in the density of cerebral white matter,
with little success. Diets low in vitamin A or in sulfur with little evidence of cerebral atrophy until the later
(both substances are necessary for the synthesis of stages. Hypodensity of the cerebellar white matter
sulfatide) have failed to have any favorable effects. has been observed less frequently. No contrast en-
After intravenous and intrathecal infusion of arylsul- hancement has been found.
fatase A the enzyme does not enter the brain, and no Probably the first abnormalities to be noted on
clinical benefit has been seen. Increasing numbers of MRI are in the corpus callosum (Fig. 6.1). Subse-
patients have received hematopoietic stem cell trans- quently, MRI discloses periventricular white matter
plants in attempts to correct their low cerebral aryl- abnormalities, with a more or less symmetrical distri-
sulfatase A levels and repair or retard their CNS dete- bution. The white matter lesions are highly confluent.
rioration. The results appear to depend largely on the In later onset cases involvement is often predomi-
stage of disease at transplantation and the rate of dis- nantly frontal, whereas in early-onset cases occipital
ease progression. The results of transplantation in the predominance can be observed. However, in all vari-
presymptomatic or early symptomatic stage are bet- ants the cerebral white matter tends to become dif-
ter than those of transplantation in the fully sympto- fusely affected. The arcuate fibers are relatively
matic stage of the disease with decreasing verbal and spared, but become involved in the later stages. Typi-
performance IQs. The chances are better in the late- cally, a pattern of radiating stripes with a signal inten-
onset cases, in which disease progression is slower. sity closer to normal is seen within the abnormal
Stabilization and reversal of MRI abnormalities have cerebral white matter (Fig. 6.2). On microscopic ex-
been described, as has clinical improvement or ar- amination, this radiating pattern is explained by the
rested progression. However, slow progression de- accumulation of products of myelin breakdown in
spite successful engraftment has also been observed. perivascular macrophages and some sparing of
The chances of stabilization or improvement have to myelin sheaths. This pattern is not evident in all cases
be weighed against the possibility of major complica- (Figs. 6.3, 6.4), and it is lacking especially in far ad-
tions and death during or after the transplantation. vanced cases with serious white matter atrophy
The general experience is that hematopoietic stem (Fig. 6.5). The corpus callosum is invariably affected,
cell transplantation has little or no beneficial effect on connecting the lesions from both sides. Cerebral
the polyneuropathy. Gene therapy still has to be test- white matter atrophy occurs in advanced stages
ed in the clinical situation. (Fig. 6.5). The posterior limb of the internal capsule
becomes involved. Brain stem lesions are observed

Fig. 6.1. MRI in a 5 1/2-year-old boy, who was diagnosed with should have been accepted as a warning against this inter-
metachromatic leukodystrophy (MLD) and extensive white pretation. Courtesy of Dr. M.A.A.P Willemsen, Department of
matter abnormalities 3 years later.The areas of higher signal in Pediatric Neurology, University Medical Center Nijmegen, The
the deep parietal white matter are often seen as a nonspecific Netherlands
finding, but the lesion in the splenium of the corpus callosum
6.6 Magnetic Resonance Imaging 79

Fig. 6.2. A case of juvenile MLD in a 6-year-old girl. The cere- weighted images, high on T1-weighted images). The corpus
bral white matter is diffusely involved, with sparing of the U callosum, posterior limb of the internal capsule, and pyramidal
fibers. Within the affected white matter a pattern of radiating tracts in the brain stem are involved
stripes is seen of more normal signal intensity (low on T2-

bilaterally in the pyramidal tracts, especially in the The condition has to be differentiated from
more advanced cases (Fig. 6.4). Some patients show globoid cell leukodystrophy (Krabbe disease), the
involvement of the cerebellar white matter. No con- cerebral form of X-linked adrenoleukodystrophy, and
trast enhancement is seen. Gray matter lesions are inflammatory white matter disorders (acute demyeli-
never conspicuous. Subtle abnormalities in signal nating encephalomyelitis). The same pattern of radi-
intensity and atrophy of the basal nuclei have been ating lines with a more normal signal intensity with-
observed. in extensive confluent white matter abnormalities can
80 Chapter 6 Metachromatic Leukodystrophy

Fig. 6.3. A case of juvenile MLD in an 8-year-old girl.There is no evident stripe-like pattern

Fig. 6.4. An 18-year-old female patient with adolescent-onset MLD. Note the corticospinal tract involvement

be seen in globoid cell leukodystrophy, and in these er been observed in MLD. Contrast enhancement is
cases the MRI abnormalities are indistinguishable generally present in the acute stage of inflammatory
from those in MLD. In early-onset globoid cell disorders. It is also present in most patients with pa-
leukodystrophy, the cerebral atrophy is usually more tients with cerebral X-linked leukodystrophy. In cere-
pronounced and there are typically areas with in- bral X-linked adrenoleukodystrophy an occipital pre-
creased density on CT in the cerebellum, thalamus, ponderance occurs most frequently, whereas MLD
internal capsule, and corona radiata, which have nev- tends to have a frontal preponderance in juvenile and
6.6 Magnetic Resonance Imaging 81

Fig. 6.5. In this MR series of a 29-year-old male with adult-on- nance of the abnormalities.The U fibers are still largely spared.
set MLD, extensive periventricular white matter abnormalities Note the involvement of the corticospinal tracts in this case
are seen, with serious atrophy. There is some frontal predomi- too

adult variants. The two zones typically present in


cerebral X-linked adrenoleukodystrophy lesions are
not present in MLD. In inflammatory disorders the
demyelinating process is usually not perfectly sym-
metrical and the cortex is often not completely pre-
served.
Chapter 7

Multiple Sulfatase Deficiency

7.1 Clinical Features facial dysmorphism, corneal clouding, hepatospleno-


and Laboratory Investigations megaly, dwarfism, severe dysostosis multiplex, and
hirsutism are present, but no ichthyosis, no retinal de-
Multiple sulfatase deficiency (MSD) is a very rare dis- generation, no deafness, and no progressive demen-
order with an autosomal recessive mode of inheri- tia. Some patients are macrocephalic, but they are
tance. It occurs with a prevalence of about 1 in 1.4 mil- rarely hydrocephalic. Some suffer from cardiac valvu-
lion births. The disease combines the features of lar involvement.
metachromatic leukodystrophy and mucopolysac- A rare juvenile type of MSD has been reported
charidosis. It is also called mucosulfatidosis, Austin with onset in childhood. The disease is characterized
variant or variant O. Three different types of MSD by short stature, ichthyosis, hepatomegaly, moderate
have been described: a neonatal form, an early-child- dysostosis multiplex and slowly progressive neuro-
hood form and a very rare juvenile form. logical abnormalities consisting of dementia, ataxia,
The early-childhood form is the usual, or classic, quadriplegia, retinal degeneration, and blindness.
form of MSD. The clinical features are those of infan- Corneal clouding is not present.
tile metachromatic leukodystrophy with mild signs of Additional investigations show changes on bone
mucopolysaccharidosis. Early development can be X-ray, such as a J-shaped sella turcica, structural
normal or delayed. Affected children usually acquire changes in vertebral bodies, scoliosis, gibbus, flared
the ability to stand and to say a few words, but their ribs, broad phalanges, abnormal metacarpals and, in
development is less well advanced in the presympto- the Saudi variant, synostosis of cranial sutures. Large
matic period than that of children with infantile basophilic to azurophilic granules are seen in lym-
metachromatic leukodystrophy. During the 2nd year phocytes of bone marrow and peripheral blood.
of life the children develop signs of a progressive en- There is an increased urinary content of sulfatide and
cephalopathy with loss of acquired abilities, progres- mucopolysaccharides, including dermatan sulfate
sive dementia, spasticity, microcephaly, blindness, and heparan sulfate. CSF protein is increased. Nerve
hearing loss, and difficulties in swallowing. Tendon conduction velocity is slowed.
reflexes are variable. In the final stages there is often Diagnosis is established by demonstrating a defi-
areflexia caused by the peripheral neuropathy. Fea- ciency of arylsulfatases A, B, and C and a number
tures similar to those of mucopolysaccharidosis may of sulfatases necessary in the degradation of mu-
occur early or later in the course of the disease. These copolysaccharides. Prenatal diagnosis can be made by
include ichthyosis, mild coarsening of the facial fea- demonstrating markedly reduced activities of sulfa-
tures, hepatosplenomegaly, stiff joints, growth retar- tases in cultured amniotic cells or chorionic villi.
dation, and skeletal anomalies. Hydrocephalus may DNA-based diagnostic procedures are also possible.
occur. There is no corneal clouding. Optic atrophy,
cherry-red macula, and retinal degeneration also oc-
cur in some cases. Death usually occurs when patients 7.2 Pathology
are between 10 and 18 years of age.
The neonatal form presents at birth with severe Pathological findings in MSD are those that might
features suggestive of mucopolysaccharidosis, such be expected in a combination of metachromatic
as facial dysmorphism, short neck, corneal clouding, leukodystrophy and mucopolysaccharidosis. There is
ichthyosis, cardiac valvular involvement, hepato- widespread neuronal lipid storage in the cortex and
splenomegaly, and severe dysostosis multiplex. subcortical gray matter structures. The storage is
The children affected have macrocephaly caused lysosomal. The neuronal deposits are finely granular
by hydrocephalus. They have signs of a severe en- and PAS positive, with slight reddish to purple
cephalopathy, with early death, usually before the end metachromasia. On electron microscopy, neuronal in-
of the 1st year of life. A Saudi variant of the neonatal clusions range in configuration from zebra bodies,
type of MSD has been described, with mild to moder- which are typically seen in mucopolysaccharidoses,
ate mental retardation, cranial synostosis and conse- to membranous cytoplasmic bodies such as are seen
quent deformities, and cervical cord compression or in neuronal gangliosidoses. There is loss of neurons
transection due to vertebral abnormalities. Severe in cerebral and cerebellar cortex and cortical atrophy.
7.6 Magnetic Resonance Imaging 83

The white matter shows demyelination with deposi- from the lack of a posttranslational modification that
tion of metachromatic material, oligodendroglial is common to all sulfatases. The reduced activity of
loss, relative axonal preservation, and marked gliosis. the enzymes is caused by a defect in the modification
The U fibers are relatively spared. Mucopolysaccha- of a conserved cysteine residue to form formyl-
rides are stored in perivascular mesenchymal tissue, glycine. The modification of the cysteine residue ap-
which may lead to the formation of macroscopically pears to be a prerequisite for the catalytic activity of
visible pseudocystic cavities. Meninges are thickened sulfatases.
and cloudy. Obliteration of the subarachnoid spaces The gene mutated in MSD is SUMF1, which is
can lead to hydrocephalus. Abnormalities in periph- located on chromosome 3p26. SUMF1, standing for
eral nerves are typical of metachromatic leukodystro- sulfatase-modifying factor 1, encodes the protein
phy. formylglycine-generating enzyme (FGE).

7.3 Chemical Pathology 7.5 Therapy

Chemical analysis of the brain has shown increases in No effective form of treatment is known.
sulfatide, sulfated steroids (e.g. cholesterol sulfate),
gangliosides, and mucopolysaccharides. The lipids
stored in the white matter are predominantly sul- 7.6 Magnetic Resonance Imaging
fatides, while those stored in the gray matter are
predominantly gangliosides and mucopolysaccha- Imaging findings are scarce in MSD. In the classic ear-
rides. ly-childhood variant images are indistinguishable
from those of infantile and juvenile metachromatic
leukodystrophy with severe cerebral white matter ab-
7.4 Pathogenetic Considerations normalities. The corpus callosum and long corti-
cospinal tracts are also involved. The cerebellar white
In MSD activity of all known sulfatases is found to be matter can be abnormal, but is not in all cases. In
reduced, including arylsulfatase A (metachromatic some patients a pattern of radiating stripes of more
leukodystrophy), arylsulfatase B = N-acetylgalac- normal signal intensity is seen within the affected
tosamine-4-sulfatase (Maroteaux-Lamy syndrome), cerebral white matter (Fig. 7.1). In these patients the
arylsulfatase C = steroid sulfatase (X-linked ichthyo- images are indistinguishable from those seen in
sis), N-acetylgalactosamine-6-sulfate sulfatase (Mor- metachromatic leukodystrophy. However, these
quio syndrome type A), heparan sulfate sulfatase stripes are not seen in all patients (Fig. 7.2). In the lat-
(Sanfilippo A), iduronate-2-sulfate sulfatase (Hunter er stages of the disease severe white matter atrophy is
syndrome), N-acetylglucosamine-6-sulfate sulfatase seen (Fig. 7.2).
(Sanfilippo D), and some other sulfatases. The resid- In the neonatal form of MSD more variable white
ual activities of the sulfatases vary considerably in fi- matter involvement has been described. Myelination
broblast lines from different patients. The lysosomal is delayed. In addition, scattered, small high-signal-
storage of sulfatides, mucopolysaccharides and sul- intensity spots are sometimes seen in the white
fated steroids is a direct consequence of the enzyme matter, which are more suggestive of mucopolysac-
deficiencies mentioned. The accumulation of gan- charidosis than of metachromatic leukodystrophy
gliosides may be caused by inhibition of other lysoso- (Fig. 7.3). In some of the patients, white matter abnor-
mal hydrolases due to the present lysosomal storage. malities are more extensive and confluent. Severe,
It is probable that the different phenotypic expres- highly confluent white matter disease can also be
sions of MSD are due to differences in relative resid- found. Dilatation of the ventricular system and
ual activity of the various enzymes. prominence of the sulci are possible findings. En-
The genes coding for the various deficient sulfa- larged perivascular spaces within the white matter
tases are intact. The rate of synthesis of various sulfa- can be expected.At the cranio–cervical junction signs
tases is normal in MSD, but they are degraded at an of cervical cord compression may be found, which is
enhanced rate. The deficiency of the sulfatases results caused by atlantoaxial abnormalities.
84 Chapter 7 Multiple Sulfatase Deficiency

Fig. 7.1. A 5-year-old male patient with classic MSD. The im- signal intensity. Within the brain stem the corticospinal tracts
ages are indistinguishable from those seen in metachromatic are involved. Courtesy of Dr. G. Mancini and Dr. H. Stroink,
leukodystrophy. The cerebral white matter is diffusely abnor- Sophia Children’s Hospital, Erasmus Medical Center, Rotter-
mal and contains countless radiating stripes of more normal dam, The Netherlands (see also Mancini et al. 2001)
7.6 Magnetic Resonance Imaging 85

Fig. 7.2. A 7-year-old boy with MSD in an advanced stage. Note the serious white matter atrophy. Courtesy of Dr. S. Blaser,
Department of Diagnostic Imaging, Hospital for Sick Children, Toronto
86 Chapter 7 Multiple Sulfatase Deficiency

Fig. 7.3. A 4.5-year-old child with the Saudi variant of MSD.


Note the multiple small high signal intensity spots bilaterally
in the centrum semiovale. From Aqeel et al. (1992), with per-
mission
Chapter 8

Globoid Cell Leukodystrophy: Krabbe Disease

8.1 Clinical Features blind and make no voluntary movements. Touching


and Laboratory Investigations may elicit primitive generalized reflex movements.
The children lack sucking abilities and have to be fed
Globoid cell leukodystrophy (GLD), also known as by nasogastric tube. The muscular tone often changes
Krabbe disease, is a progressive white matter disorder again. The opisthotonic posture disappears and is fol-
with autosomal recessive inheritance. A number of lowed by a tendency to hypotonicity. The children be-
clinical types can be distinguished, which differ in age come cachectic, and if they do not die of an intercur-
of onset and in the rate of clinical deterioration. The rent respiratory infection or of aspiration they suc-
early infantile or classic type is the most common. Its cumb gradually. Death occurs between 5 months and
incidence is estimated at 1–2 in 100 000. The other 3 years of age.
types are: congenital, late-infantile, juvenile, and ado- In rare cases, GLD is already manifest during the
lescent-adult. As a rule only one variant of GLD oc- neonatal period in the form of unspecific feeding dif-
curs within one family, although exceptions have ficulties, irritability, twitchiness, or respiratory prob-
been reported. lems. Both floppy and hypertonic neonatal variants
Clinical features and course of disease are fairly have been described.
uniform in early-infantile GLD. During the first few The later onset forms of GLD are clinically much
months of life the infants are healthy and their psy- more heterogeneous and progress more slowly. A
chomotor development is normal. The onset of clini- subdivision into late-infantile onset (6 months to
cal symptoms occurs between 1 and 6 months of age. 3 years), juvenile onset (4–10 years) and adolescent–
Hagberg et al. (1969) distinguish three clinical stages. adult onset has been proposed, but the subdivision is
Stage I is characterized by hyperirritability and peri- arbitrary. In most children the onset of disease occurs
ods of crying, particularly when the infants are han- before the age of 5 years; adolescent–adult onset is
dled and nursed. They seem to be extremely sensitive rare. There is no significant difference in symptoma-
to light and noise and often have excessive startle re- tology according to the age at onset. The initial signs
sponses. Periods of fever often occur without signs of in the late-onset forms of GLD are quite variable:
infection. The muscular tone increases.At the onset of spastic paraparesis, hemiparesis, cerebellar ataxia,
the disease the deep tendon reflexes are normal. isolated visual failure (as a result of either optic atro-
There is a stagnation of mental and motor develop- phy or bilateral involvement of the optic radiations),
ment, soon followed by regression. In sporadic cases dystonia, epileptic seizures, psychosis, and mental de-
convulsions, hemiplegia, or predominant signs of pe- terioration. Of these signs, increasing difficulties in
ripheral neuropathy are the presenting abnormali- walking caused by spasticity or ataxia and isolated
ties. Within 2–4 months of onset most patients reach visual failure are the most frequent first manifesta-
stage II. This stage comprises the subsequent period tions. Irritability is not infrequently noted early in the
of rapid and severe motor and mental deterioration. disease. Peripheral neuropathy as an isolated present-
A constant opisthotonus and a hypertonic flexion of ing sign of the disease is a rare variant that can occur
the arms and extension of the legs are present. The in late-onset types but has also been reported in in-
deep tendon reflexes can no longer be evoked. There fants. The rate of progression and duration of the dis-
are extensor plantar signs. Abilities previously ease are quite variable. Some patients, and especially
achieved are lost. At the same time the general condi- those younger than 3 years, have a very rapid course
tion deteriorates, with more frequent hyperpyretic of disease, becoming bedridden, tetraparetic, and de-
periods, often accompanied by intense sweating. Pul- mented in less than 3 months. Most patients, especial-
monary infections are common. Myoclonic irregular ly those with later onset, have a more chronic course.
jerks of the arms and legs, startle myoclonus, hyper- Death occurs after a highly variable period ranging
tonic fits, and other atypical seizures occur, as do typ- from 18 months to more than 14 years. Bronchopul-
ical, generalized tonic-clonic seizures and infantile monary infections are the most frequent cause of
spasms. The signs of visual failure and optic atrophy death.
begin to appear. Stage III is the vegetative, burned-out In early-onset GLD an abnormally high protein
stage. The children seem to have little or no mental level is always found in the CSF while the cell count
activity and lie in a decerebrate posture. They are is normal. Only occasionally are there more than
88 Chapter 8 Globoid Cell Leukodystrophy: Krabbe Disease

10 cells/ml, the majority of these being mononuclear served, but axons are relatively preserved in compar-
leukocytes. The elevated CSF protein level is less con- ison with the degree of myelin loss. There is an early
stant in later onset forms of GLD, being found in and marked loss of oligodendrocytes through apop-
about 50% of the patients. In early-onset GLD, the tosis. There is diffuse fibrillary gliosis throughout the
EEG is normal at the beginning of the disease, some- white matter with proliferation of astrocytes. In the
times even after some months of manifest clinical central nuclei of the brain moderate neuronal loss can
symptoms. Abnormal findings tend to appear at the be observed, and sometimes intense gliosis.
end of stage I. The pattern then becomes increasingly The pathognomonic feature of GLD is the accumu-
abnormal, characterized by markedly slow activity of lation of globoid cells. These cells are multinucleated
high voltage. In the terminal stage the background giant cells with ballooned cytoplasm containing fine-
activity is frequently interrupted by numerous fast, ly fibrillary to granular material. Their cytoplasm
sharp-wave activities, episodic dysrhythmias, or stains eosinophilic in HE, moderately positive in PAS,
epileptiform episodes. Sometimes a picture of hyps- and negative or faintly positive in Sudan Black prepa-
arrhythmia is found. Nerve conduction velocity is rations. There are also smaller rounded cells with sin-
markedly reduced in all patients with early-onset gle nuclei and accumulation of identical material in
GLD. In later onset forms the reduction in nerve con- their cytoplasm. These are called epithelioid cells by
duction velocity is more variably present; conduction some, and are included with the globoid cells by
velocity can also be normal. others. Globoid cells are present as single cells or,
The diagnosis of GLD is established by showing a more often, as groups of cells in the affected white
deficiency of galactocerebroside β-galactosidase matter. They accumulate around blood vessels. They
(galactocerebrosidase) in white blood cells or cul- are not only present within the affected white matter,
tured fibroblasts. It is not possible to distinguish be- but can also accumulate in central gray matter struc-
tween early-onset GLD and late-onset variants by tures. There is considerable variation in the distribu-
comparing residual enzyme activities. There is a con- tion and number of globoid cells in individual cases,
siderable overlap between the normal range and the depending on the duration of the disease and the in-
carrier range of enzyme activity, so that carrier test- tensity of the histopathological process. They are low-
ing is often unreliable. In addition, very low levels of er in number in patients with long-standing disease
galactocerebrosidase activity (less than 10%) have who have burned-out pathology. At an ultrastructur-
been found in healthy people. The presence of poly- al level the globoid cells appear to contain specific
morphic amino acid changes in the enzyme causes straight and twisted tubular-type inclusions. The
this wide range of activities. In some families addi- tubular inclusions are usually dispersed throughout
tional studies, such as substrate loading tests in cul- the cytoplasm, and rarely membrane bound. The
tured skin fibroblasts, are necessary. DNA analysis straight tubules are angular or crystalloid in cross
may help in solving the problems with diagnosis. GLD section, while the twisted tubules are rectangular or
can be diagnosed prenatally by assaying the enzyme oval. Sometimes myelin figures and lipid droplets are
activity in chorionic villi or amniotic fluid cells. DNA found in the cytoplasm of globoid cells. Globoid cells
confirmation is an option. stem from phagocytic mesenchymal cells.
In GLD the peripheral nerves are also involved in
the disease process. Light microscopic examination
8.2 Pathology reveals segmental demyelination and variable axonal
degeneration in early-onset forms. In adult patients,
Gross examination of the brain reveals a moderate to myelin sheaths have been reported to be abnormally
marked reduction in size. On sectioning, the cortex thin for the fiber diameter, suggesting hypomyelina-
appears to be relatively spared but there is a marked tion rather than demyelination. Usually no typical
reduction in the amount of white matter, which shows globoid cells are found. Electron microscopy shows
a brownish discoloration. that Schwann cells and endoneurial macrophages
Microscopic examination confirms that there is lit- contain cytoplasmic inclusions similar to the inclu-
tle or no involvement of cortical gray matter. The sions in the globoid cells of the CNS.
cerebral, cerebellar, and brain stem white matter
shows diffuse demyelination throughout the brain,
although there are regional variations in intensity. 8.3 Chemical Pathology
There is a distinct tendency for the U fibers to be pre-
served. The extent to which tracts in the brain stem Chemical analysis of the brain shows that the gray
and cord are affected varies. Long tracts, in particular matter is only moderately abnormal, while severe
the corticospinal and spinocerebellar tracts, and the changes are present in the white matter. The white
dorsal columns are usually severely damaged. In the matter has an increased water content, and its lipid
areas of demyelination axonal degeneration is ob- content is greatly diminished.Although both cerebro-
8.4 Pathogenetic Considerations 89

side and sulfatide are greatly decreased, as might be strate specificity explains why psychosine accumu-
expected from the myelin loss, there is a significant lates to give high levels in GLD brain, whereas cere-
change in the cerebroside-to-sulfatide ratio, from the broside, while moderately increased in relative con-
normal value of 4 :1 to as much as 10 : 1. Proportional centration, is decreased in absolute concentration.
to the total loss of myelin there is a decrease in the Cerebroside is a constituent almost exclusively of
white matter content of total lipids, cholesterol, gly- oligodendrocytes, Schwann cells, and myelin sheaths.
colipids, and ethanolamine phosphoglycerides. There Metabolism of cerebroside is closely related to the
is a relative increase (though an absolute decrease) metabolism of myelin. In immature brains, prior to
of other phospholipids, such as choline phosphogly- myelination cerebroside is practically absent, and
cerides, serine phosphoglycerides, and sphingomyelin. lack of galactocerebrosidase is therefore of little con-
As a rule, no elevation of cholesterol esters is found. sequence. As soon as myelination begins the normal
Galactosylsphingosine, also called psychosine, accu- turnover of myelin starts. This coincides with a rapid
mulates in considerable amounts in the white matter rise of galactocerebrosidase activity in normal brain.
of GLD patients. Concentrations of up to 100 times In GLD brains cerebroside from catabolized myelin
the normal value have been found. Psychosine is not cannot be disposed of adequately because of the lack
normally detectable in gray matter, but is elevated in of the enzyme. GM1-ganglioside β-galactosidase may
GLD patients, though not nearly to the same extent as be responsible for part of the cerebroside breakdown
in white matter. and prevent real accumulation of cerebroside. Excess
Analysis of isolated myelin in GLD reveals a rela- of cerebroside in phagocytic cells leads to the trans-
tively normal lipid composition. The cerebroside con- formation of these cells into globoid cells. In experi-
tent of myelin is not elevated. A relatively high con- mental studies the relationship between cerebroside
centration of cerebroside is found in globoid cell-en- and globoid cells has been confirmed. Intracerebral
riched fractions from the diseased white matter. injection of cerebroside into rat brain elicits a globoid
There is cerebroside storage in globoid cells, while cell reaction, whereas the injection of many other
there is none in the white matter as a whole or in the substances does not lead to this response.
myelin membrane. Psychosine is also a substrate for the deficient en-
zyme galactocerebrosidase. It is formed from UDP-
galactose and sphingosine through the action of
8.4 Pathogenetic Considerations UDP-galactose : ceramide galactosyl transferase. It
cannot be disposed of in GLD, because it is a very
The primary defect in GLD is a deficiency of galacto- poor substrate for GM1-ganglioside β-galactosidase.
sylceramidase, also called galactocerebroside β- Psychosine is a highly cytotoxic substance. There is
galactosidase or galactocerebrosidase. This is a lyso- evidence that accumulated psychosine leads to pro-
somal enzyme that catalyzes the first step of cerebro- duction of cytokines and inducible nitric oxide syn-
side (= galactosyl ceramide) degradation and splits thase expression and contributes to apoptosis. It is
cerebroside into galactose and ceramide. Sulfatide is probable that psychosine generated within oligoden-
also normally degraded through cerebroside into ce- drocytes during the period of active myelination
ramide and galactose. The gene coding for galacto- accumulates until it reaches a toxic level. Apoptosis is
cerebrosidase, GALC, is localized on chromosome selectively induced in oligodendrocytes because psy-
14q31. Many different mutations in GALC have been chosine formation occurs primarily in these cells.
identified in GLD. There is no clear genotype–pheno- This explains the early and marked loss of oligoden-
type correlation. One might anticipate that the differ- drocytes, resulting in loss of the myelin sheaths main-
ence in phenotypic expression in GLD would result tained by these cells. In GLD brains the concentration
from different levels of residual enzyme activity, but of psychosine correlates well with the severity of
the currently available assay procedures reveal no pathological changes. When the stage of massive
consistent difference in residual enzyme activity death of oligodendroglial cells is reached, rapid
between early- and late-onset forms. myelin breakdown occurs, contributing more cere-
Mammalian tissues contain two genetically dis- broside, which may enhance the globoid cell reaction.
tinct lysosomal β-galactosidases with different, The death of oligodendroglia prevents further myeli-
though overlapping, substrate specificities: galacto- nation.
cerebroside β-galactosidase (galactocerebrosidase) The sequence of events is well illustrated in the
and GM1-ganglioside β-galactosidase. It has been Twitcher mutant mouse, which has the same defect as
demonstrated that under certain assay conditions is present in the human disease. Initial myelin devel-
GM1-ganglioside β-galactosidase can also hydrolyze opment is normal, highlighting the point that myelin
cerebroside. Psychosine (= galactosyl sphingosine) is build-up is essentially normal in GLD. Subsequently,
hydrolyzed by galactocerebrosidase, but not by GM1- there is a declining rate of myelination, followed by
ganglioside β-galactosidase. This difference in sub- demyelination and the appearance of globoid cells. It
90 Chapter 8 Globoid Cell Leukodystrophy: Krabbe Disease

is myelin turnover that brings the enzymatic defect to more slowly, and to a lesser degree. Sulfatide is
expression. trapped in the myelin membrane and its content in-
The difference in tissue reaction between CNS and creases until the composition of the myelin sheath is
PNS is difficult to explain. Phagocytic cells in the PNS so abnormal that the normal stability of the mem-
contain abnormal inclusions similar to those in the brane is lost and demyelination ensues.
brain, and it is not clear why they are not transformed Sphingolipid activator proteins, i.e., saposins A, B,
to the typical multinucleated globoid cells. The al- C, and D, are small glycoproteins derived from a com-
most obligatory involvement of the CNS and the vari- mon precursor protein, prosaposin. These activator
able involvement of the PNS, particularly in later proteins are required for degradation of sphin-
onset forms of GLD, suggests that oligodendrocytes golipids. Total deficiency of all saposins and specific
are more vulnerable to psychosine than are Schwann deficiencies of saposins B and C are associated with
cells. known human diseases. No specific saposin A or D
In the brain there is a regional variation in vulner- deficiency is known in humans. It has recently been
ability to pathologic changes. In particular, the U demonstrated that saposin A is indispensable for the
fibers are typically spared. As the process of myelin degradation of cerebroside by galactocerebrosidase.
build-up is normal in GLD and the defect involves A transgenic mouse with saposin A deficiency has
myelin turnover and breakdown, the areas of the been shown to develop a demyelinating disease simi-
brain that myelinate last can be expected to be in- lar to, but milder than, that seen in the Twitcher
volved in later stages of the disease than are the early- mouse. Saposin A deficiency can be anticipated
myelinating areas. The U fibers are the last to myeli- among human patients with a late-onset slow-de-
nate, which may explain their relative preservation. In myelination disease without galactocerebrosidase
addition, in animal research the in vivo turnover rate deficiency.
of cerebroside appears to be higher in the areas of the
white matter that are consistently more severely
affected in GLD; this may furnish another part of the 8.5 Therapy
explanation for regional variation in white matter
involvement. To date, early-onset GLD has invariably proved fatal.
GLD is a primary myelin disorder, and there are no The problem with early-onset forms of GLD is that
morphological or functional abnormalities outside neuropathological changes are already present in the
the nervous system. This is explained by the fact that second trimester of pregnancy and that any form of
cerebroside is present almost exclusively in nervous treatment is applied to an already damaged nervous
tissue, the amounts in organs outside the nervous sys- system. Hematopoietic stem cell transplantation to
tem being quantitatively negligible. infantile patients who are already clinically sympto-
Although GLD and metachromatic leukodystro- matic has not resulted in beneficial effects. Reports on
phy are both caused by a genetic defect in the catabo- hematopoietic stem cell transplantation very soon
lism of myelin constituents and initial myelin build- after birth, before the onset of any clinical symptoms,
up is normal in both disorders, there are interesting are appearing, and the results are promising but must
dissimilarities between GLD and metachromatic be confirmed in larger studies. Hematopoietic stem
leukodystrophy. Metachromatic leukodystrophy is cell transplantation is now performed in increasing
characterized by extreme storage of sulfatide, where- numbers of patients with later onset forms. Stabiliza-
as GLD hardly deserves the name ‘storage disorder.’ In tion and improvement of clinical symptoms and MRI
metachromatic leukodystrophy the composition of abnormalities have been reported. Early treatment,
the myelin membrane is altered, with excessive sul- while the intellect is still intact and the motor disabil-
fatide within the myelin sheath. In GLD, myelin is ities are only mild, is essential.
compositionally normal, and the relative rise in cere- Another therapeutic approach is one of substrate-
broside relative to other lipids in the analysis of whole reduction therapy. L-Cycloserine is an inhibitor of
white matter appears to be related to an excess of sphingosine synthesis by irreversible inhibition of
cerebroside in globoid cells. It is likely that these dif- 3-ketodyhydrosphingosine synthase. Administration
ferences can be explained by a difference in the rate of of L-cycloserine to animals leads to a reduction in the
oligodendroglial cell death. In GLD these cells disap- production of brain cerebroside, sulfatide, and gan-
pear rapidly, early on in the local disease process. This gliosides. Although the level of psychosine has not
early cell death is probably caused by psychosine tox- been measured, it is likely that it is also lowered, since
icity. With oligodendroglial cell death myelin mem- psychosine is made up of sphingosine and galactose.
branes are lost simultaneously, before the defect in Thus, L-cycloserine lowers the levels of substrates that
turnover can trap cerebroside or other abnormal con- are not digested in GLD. Twitcher mice treated with
stituents within the myelin sheath. In metachromatic L-cycloserine have milder disease and live longer, but
leukodystrophy oligodendroglial cells disappear later, only if the treatment is started early. If combined with
8.6 Magnetic Resonance Imaging 91

bone marrow transplantation, L-cycloserine improves extent of white matter abnormalities. Signal abnor-
the outcome further. The problem with L-cycloserine malities in the hilus of the dentate nucleus, cerebellar
is that cerebroside and sulfatide are essential for nor- hemispheric white matter, and pyramidal tracts of the
mal myelin structure and function. L-Cycloserine is brain stem in infants only a few months of age should
toxic in higher doses. In addition, L-cycloserine has suggest the possibility of GLD. The areas of high den-
not yet been tested in humans. Gene therapy also has sity on CT may not be so clearly abnormal on MRI.
not yet progressed beyond the animal experimental These areas may have a relatively low or high signal
stage. intensity on T2-weighted images and a normal, high,
or low signal intensity on T1-weighted images. With
progression of the disease, the subcortical white
8.6 Magnetic Resonance Imaging matter also becomes involved and global atrophy
ensues. An unusual finding reported in a few GLD
In cases of early-onset GLD, the CT findings are quite infants is optic nerve enlargement. This is related to
characteristic. The abnormalities seen are related to the presence of a very high number of globoid cells. In
the stage of the disease. In stage I there are either no many patients MRI and CT provide complementary
changes at all, or symmetrically increased density in information, CT showing the characteristic hyper-
the thalami, corona radiata and, less often, posterior densities and MRI showing the extent of the demyeli-
limb of the internal capsule, caudate nucleus, globus nation.
pallidus, and putamen. Sometimes more extensive In cases of late-onset GLD, hyperdensities in the
hyperdense areas are noted, including the brain stem, thalamus and basal ganglia have been found on CT.
cerebellum, dentate nucleus, optic radiation, central There are white matter abnormalities, and these may
subcortical white matter, and cortical gray matter in part have a high density on CT. Both CT and MRI
(Fig. 8.1). Pathologically, the hyperdensity on CT cor- show a predominant involvement of the parieto-oc-
relates with a higher concentration of globoid cells cipital periventricular white matter with extensions
and proliferating glia. In stages II and III, symmetri- in the temporal direction and associated involvement
cal low-density areas in the periventricular white of the splenium of the corpus callosum (Fig. 8.3). MRI
matter and corpus medullare of the cerebellum be- may show a pattern of radiating stripes of more near-
come obvious. The low density of the white matter is ly normal signal intensity within the abnormal cere-
relatively less marked than in other leukodystrophies. bral white matter. The U fibers are relatively spared
The low density represents demyelination and in- (Fig. 8.3). The posterior limb of the internal capsule
creased water content. The severe fibrous astrogliosis and pyramidal tracts in the brain stem are involved.
that occurs in GLD may be responsible for the less in- The cerebellar white matter is spared. More extensive
tense hypodensity. In stage III diffuse brain atrophy, white matter lesions affecting the whole centrum
both central and peripheral, is observed. Sulci are semiovale probably represent a more advanced stage
widened, the head of the caudate nuclei is flattened, of disease. In the later stages atrophy becomes obvi-
and the ventricles, including the third, are widened as ous.
a result of atrophy of the thalamus. Furthermore, adults may exhibit signal changes re-
MRI confirms the presence of periventricular stricted to the corticospinal tracts, extending from
white matter abnormalities with relative sparing of the motor cortex through the posterior limb of the in-
the U fibers in early-onset GLD (Fig. 8.2). The corpus ternal capsule into the pyramidal tracts of the brain
callosum is affected, connecting the lesions of both stem, in either a symmetrical or an asymmetrical or
sides.A radiating pattern with stripes of more normal unilateral distribution (Fig. 8.4). However, some adult
signal intensity is often observed within the cerebral patients have a normal brain MRI for a long time.
white matter (Fig. 8.2), but this pattern is not obliga- Some adults present with signs of an isolated poly-
tory. The stripes correlate with perivascular deposits neuropathy. If they exhibit white matter abnor-
of globoid cells in microscopic examination of the malities on cerebral MRI as described above, this
brain. The posterior limb of the internal capsule, cere- should lead to inclusion of GLD in the differential
bellar white matter, hilus of the dentate nucleus, and diagnosis.
pyramidal tracts in the brain stem are involved Variable, but at most subtle, enhancement has been
(Fig. 8.2). In fact, involvement of the brain stem and described in GLD cases and may involve the nor-
cerebellum occurs early, before supratentorial white mal–abnormal white matter junction at the border
matter involvement. MRI can be deceptively normal between the deep white matter and the U fibers, and
in the initial stages of early-infantile GLD, which is also the corpus callosum, cranial nerves, and spinal
probably largely related to the still immature myeli- nerve roots. However, in most cases no enhancement
nation preventing recognition of the presence and is seen.
92 Chapter 8 Globoid Cell Leukodystrophy: Krabbe Disease

Fig. 8.1. CT findings in a 4-month-old boy with early-infantile the internal capsule, thalamus, and central part of the corona
GLD.The series demonstrates the hyperdensities in the prima- radiata. At this stage the CT appearance is almost diagnostic,
ry myelination zones: brain stem, cerebellum, posterior limb of and more characteristic than the MRI findings

Fig. 8.2. Early-infantile GLD in a 6-month-old girl. The three hemispheres. Cavitary lesions are visible in the posterior limb
upper rows show transverse T2-weighted images. The periven- of the internal capsule. The cerebellar white matter and the
tricular white matter has a higher signal intensity than the hilus of the dentate nucleus are prominently involved. The
directly subcortical white matter, although the latter is still T1-weighted transverse images (lowest row) confirm the stripe-
unmyelinated.A stripe-like pattern is seen in the affected cere- like pattern in the abnormal white matter
bral white matter.There is a significant atrophy of the cerebral
8.6 Magnetic Resonance Imaging 93

Fig. 8.2.
94 Chapter 8 Globoid Cell Leukodystrophy: Krabbe Disease

Fig. 8.3. Late-infantile GLD in a girl aged 4 years.There are ex- terior limb of the internal capsule and corticospinal tracts in
tensive, symmetrical white matter abnormalities, predomi- the brain stem are affected.The cerebellar white matter is nor-
nantly involving the posterior part of the cerebral hemi- mal
spheres. There is a subtle pattern of radiating stripes. The pos-

The pattern of symmetrical and extensive involve- ter. Patients with early-onset GLD usually have more
ment of the cerebral white matter, corpus callosum, pronounced cerebral atrophy than patients with ear-
internal capsule, and brain stem tracts is indistin- ly-onset metachromatic leukodystrophy. In patients
guishable from the pattern observed in many with later onset metachromatic leukodystrophy, the
metachromatic leukodystrophy patients, particularly white matter abnormalities often have a predomi-
when radiating stripes of more normal signal intensi- nantly frontal location, whereas in patients with later
ty are seen within the abnormal cerebral white mat- onset GLD the parieto-occipital white matter is usual-
8.6 Magnetic Resonance Imaging 95

Fig. 8.4. Adult GLD in a female patient 25 years of age. Note as is the splenium. Courtesy of Dr. D. Loes and Dr. C. Peters, Uni-
the selective involvement of the corticospinal tracts. The con- versity of Minnesota, USA
necting tracts through the corpus callosum are also affected,

ly predominantly involved. The hyperdense areas ter and a rim of contrast enhancement in between,
seen on CT in GLD are lacking in metachromatic which is so characteristic of X-linked adrenoleukody-
leukodystrophy. strophy, has never been described in GLD, although
The CT scan findings with high density in the contrast enhancement involving the splenium of the
thalamus and basal ganglia are reminiscent of the CT corpus callosum has been described in late-onset
findings in early-onset GM1 and GM2 gangliosidoses. GLD patients.
The MRI findings are, however different (see related The pattern with selective involvement of the cor-
chapters). ticospinal tracts may resemble the pattern seen in
The pattern with parieto-occipital predominance amyotrophic lateral sclerosis. However, the signal
seen in later onset GLD patients resembles the pattern changes are less pronounced in the latter. In adreno-
seen in cerebral X-linked adrenoleukodystrophy. myeloneuropathy a similar pattern of MRI abnormal-
However, the pattern consisting of two zones of dif- ities can be observed, and this disease should be ex-
ferent signal change within the abnormal white mat- cluded by means of the appropriate laboratory tests.
Chapter 9

GM1 Gangliosidosis

9.1 Clinical Features In type 2 GM1 gangliosidosis the clinical course is


and Laboratory Investigations slower and the onset insidious. The children initially
appear normal. The first clinical signs begin between
GM1 gangliosidosis is an autosomal recessive disor- 6 months (late-infantile form) and 2 years (juvenile
der of GM1 metabolism, resulting in variable neural form) of age. They usually consist of developmental
and visceral accumulations. Three forms can be dis- arrest and gait disturbances. The subsequent course is
tinguished: infantile or type 1 GM1 gangliosidosis, ju- characterized by progressive dementia, lethargy,
venile or type 2 GM1 gangliosidosis, and adult, chron- epilepsy, an abnormally pronounced startle response
ic or type 3 GM1 gangliosidosis. to noise, spastic tetraplegia, cerebellar ataxia, loss
Type 1 infantile GM1 gangliosidosis presents at or of speech, and extrapyramidal features, such as
soon after birth, signs being poor sucking and feed- choreoathetosis. Blindness is also a feature, but starts
ing. The child is hypotonic and hypoactive and soon late in the course. No retinal degeneration or cherry-
develops facial and peripheral edema. At times red spots are seen. The patients are not characterized
neonatal ascites and hydrocele are seen, and some- by organomegaly or coarsening of facial features sug-
times generalized edema. There are characteristic gestive of mucopolysaccharidosis. Skeletal deformi-
coarse facial features similar to those found in mu- ties are relatively mild. The average life span varies
copolysaccharidoses. Facial abnormalities include between 3 and 10 years. Death is usually caused by
frontal bossing, wide and depressed nasal bridge, long recurrent bronchopneumonia.
philtrum, and large low-set ears. The gums and Type 3 GM1 gangliosidosis is the adult or chronic
tongue may appear hypertrophied. The cornea is variant. The clinical signs usually emerge in the sec-
clear. Hepatomegaly is present, and the spleen is often ond decade of life, but onset in the first decade has
also enlarged. The dysmorphic features and he- also been described. Gait disturbance and speech dis-
patosplenomegaly gave this disease its other name: turbance are early signs of the disease. Extrapyra-
pseudo-Hurler disease. The skin is usually thick and midal features are usually most prominent and
rough. Exceptional cases of angiokeratoma corporis can take the form of slowly progressive dystonia,
diffusum have been described. Failure to thrive and choreoathetotic movements, facial grimacing, ble-
severe psychomotor retardation are early signs of the pharospasm, dysarthria, rigidity, parkinsonism with
disease. Bilateral, cherry-red spots are found on the immobile face, bradykinesia, and typical gait abnor-
maculae in about half the patients. Early blindness malities. In rare cases ataxia, pyramidal signs, mild
occurs, which is cortical or retinal in origin. The child intellectual impairment, and seizures occur. Bony ab-
remains hypoactive and is weak. Noise frequently normalities are minimal, if present. Cherry-red spots,
provokes an exaggerated startle response. Movements visceromegaly, and facial dysmorphism do not occur.
are poorly coordinated. Reflexes are hyperactive. In type 1 GM1 gangliosidosis vacuolated lympho-
Macrocephaly can develop, but is less marked than in cytes are found in the peripheral blood smear, but
Tay-Sachs disease. As the child gets older, broadness these are not present in types 2 and 3. Large, foamy
of the hands and shortness of the fingers become sea-blue histiocytes are present in the bone marrow
apparent. Joints become stiff. The wrist and ankle in type 1; these cells are fewer in number in types 2
joints are often enlarged, but not tender. Flexion con- and 3. Rectal biopsy shows neuronal lipidosis in
tractures frequently occur at elbows and knees. Meissner’s plexus. Ultrastructurally, membranous
Kyphoscoliosis is frequently found. After a year, cytoplasmic bodies similar to those seen in GM2 gan-
neurological deterioration is rapid, with epileptic gliosidosis are seen within neurons. The membra-
seizures, progressive spasticity, and finally decere- nous cytoplasmic bodies consist of spirally wound
brate rigidity, deafness, blindness, and loss of social lamellae enclosed within a membrane of lysosomal
contact. In rare cases cardiomyopathy with cardiac origin. Other inclusions are more pleiomorphic in
failure occurs. Flexion contractures of the arms and nature. Evidence of neuronal lipidosis on rectal
legs may become extremely severe. Respiratory prob- biopsy can be found in all types of GM1 gangliosido-
lems are common, with frequent infections. Bron- sis. Clear vacuoles can also be found in visceral histi-
chopneumonia is a frequent cause of death, which ocytes and parenchymal cells of visceral organs and
usually occurs between 12 and 24 months.
9.3 Chemical Pathology 97

in epithelial cells in all types, but are less abundant in is degeneration and loss of neurons. Storage bodies
the later onset forms. are also present in glia.
Radiological abnormalities in type 1 may be mini- Electron microscopy demonstrates that the neu-
mal at birth but become progressively more pro- ronal inclusion bodies are identical to the membra-
nounced with time. By 6 months there is usually little nous cytoplasmic bodies seen in GM2 gangliosidosis.
difficulty in identifying them. The abnormalities in- They consist of spirally wound membranous lamellae
clude kyphoscoliosis and hypoplasia and beaking of enclosed within a limiting membrane of lysosomal
one or more vertebrae. The long bones are wide in the origin.
center, tapering to both ends. There is generalized The white matter in type 1 GM1 gangliosidosis is
rarefaction of the cortex of most bones. With increas- gliotic, and there is a diffuse and profound paucity of
ing age the externally thickened cortical wall is re- myelin. Axons are relatively preserved. Where pre-
moved by expansion of the medullary cavity. The sent, myelin is structurally normal, but the thickness
metacarpals are wedge-shaped, being expanded dis- of most myelin sheaths is reduced. The early myeli-
tally and constricted proximally. The sella turcica is nating structures have a myelin content that is normal
shoe shaped, shallow, and elongated. In type 2, radio- for age, whereas the myelin content of later myelinat-
logical changes are mild and involve mainly the verte- ing structures is much lower than expected. Oligo-
bral bodies. In type 3 the radiological changes are dendrocytes are decreased in number, and apoptosis
minimal or absent. Flattening of vertebral bodies may of these cells can be observed. The white matter ab-
be seen. normalities are probably the result of a combination
In types 1 and 2 the EEG shows progressive deteri- of disturbed myelination and myelin loss.
oration, but epileptic discharges are rare. The ERG In type 1 GM1 gangliosidosis visceral storage is
remains normal. found. The liver is enlarged, and storage material is
Biochemical investigations disclose abnormal uri- present in hepatocytes and in histiocytes in liver
nary oligosaccharide excretion. The concentration of sinusoids. The renal glomerular epithelium shows
the urinary oligosaccharides appears to correlate marked vacuolization of the cytoplasm. The spleen,
with the severity of the disease, the excretion being lymph nodes, thymus, and intestinal mucosa contain
highest in type 1 and lowest in type 3 GM1 gangliosi- many foamy histiocytes. Large foamy histiocytes are
dosis. The CSF protein is normal. In types 1 and 2 an present in bone marrow aspirates. Skin biopsies show
increased concentration of GM1 ganglioside may be foamy vacuolization of sweat gland epithelium, histi-
found in plasma and CSF. ocytes, fibroblasts, and endothelium cells. The vac-
Demonstration of a deficiency of β-galactosidase uoles appear empty. The stored oligosaccharides in
activity in leukocytes or cultured fibroblasts is the visceral organs are extremely soluble in water and are
most effective means of establishing the diagnosis. It lost on fixation.
is important also to analyze neuraminidase activity In types 2 and 3 GM1 gangliosidosis identical neu-
in order to exclude galactosialidosis. A decreased hy- ronal storage is seen. In type 3 the neuronal storage is
drolysis rate of GM1 ganglioside can be demonstrated present predominantly in the basal ganglia. White
in cultured skin fibroblasts. DNA analysis for diag- matter is either not affected at all, or only to a mini-
nostic purposes is possible. Prenatal diagnosis is pos- mal extent. The visceral storage varies considerably.
sible by enzyme analysis in cultured amniotic fluid There may be no storage at all, or sparse histiocytes
cells or chorionic villi or by DNA analysis. may be seen in spleen and liver. There may be foam
cells in bone marrow and lymphatic tissue, and vac-
uolization of hepatic and renal cells similar in form to
9.2 Pathology but less severe than in type 1.

External examination of the brain in type 1 GM1


gangliosidosis usually reveals no abnormalities, 9.3 Chemical Pathology
though sometimes some cortical atrophy is detect-
ed. The consistency of the white matter may be in- Gangliosides are glycosphingolipids that contain
creased. sialic acid in their oligosaccharide chain. In type 1
Light microscopy reveals neuronal storage through- GM1 gangliosidosis pronounced accumulation of the
out the nervous system, mainly in cerebral and cere- normal monosialoganglioside GM1 occurs, accompa-
bellar cortex, but also in basal ganglia, brain stem, nied by a minor accumulation of its asialo derivative
spinal cord, and Meissner’s plexus. The neurons have GA1 and of other minor glycolipids and glycopep-
ballooning, foamy cytoplasm, and the nucleus is dis- tides. The total ganglioside content of the brain is
placed to the periphery. Accumulation of storage ma- increased, with an approximately 10-fold increase of
terial in proximal nerve cell processes results in the GM1 in gray matter and a 2-fold increase in white
formation of meganeurites and megadendrites. There matter. In gray matter GM1 ganglioside constitutes
98 Chapter 9 GM1 Gangliosidosis

70–90% of total ganglioside, instead of the normal glycolipids. Visceral accumulation of oligosaccha-
about 25%. The level of total lipid in gray matter is rides is minor.
slightly decreased, mainly due to a moderate decrease
of phospholipids and glycolipids. In white matter a
marked decrease of major myelin constituents is 9.4 Pathogenetic Considerations
found, such as cholesterol, phospholipids, cerebro-
sides, sulfatides, and proteolipid protein. Marked GM1 gangliosidosis is caused by a deficiency of the
increases in free fatty acids and in cholesterol esters lysosomal degradative enzyme acid β-galactosidase.
are found in most cases. The white matter chemical This enzyme is an acid hydrolase catalyzing cleavage
abnormalities are compatible with moderately severe of terminal β-linked galactose from a variety of
myelin destruction. In the myelin membrane itself substrates, including GM1 ganglioside, its asialo de-
the concentration of GM1 ganglioside is also several rivative GA1, lactosylceramide, galactose-containing
times the normal concentration. Other abnormalities oligosaccharides, and the mucopolysaccharide ker-
in the composition of isolated myelin are a very atan sulfate. The gene coding for β-galactosidase,
high concentration of cholesterol, a low level of glyco- GLB1, is located on chromosome 3p21.33. Activity of
lipids, especially cerebroside, and a low concentration β-galactosidase is also dependent on a functional
of phospholipids, especially ethanolamine phospho- protein, called protective protein/cathepsin A. This
lipids. These myelin abnormalities represent the protein is encoded by PPGB, a gene located on chro-
transitional state of myelin undergoing nonspecific mosome 20q13.1. Protective protein associates with
breakdown. two enzymes, β-galactosidase and neuraminidase, in
The neuronal inclusion bodies, the so-called mem- an early biosynthetic compartment, and by virtue of
branous cytoplasmic bodies, have an extremely high the association the two enzymes are correctly routed
ganglioside content. GM1 accounts for approximately to the lysosome and protected against rapid break-
95% of the total ganglioside content. Other compo- down by intralysosomal proteases. Some hydrolytic
nents are proteolipid protein, phospholipids, and enzymes need nonenzymic factors (activator pro-
cerebroside. One of the glycolipids, cerebroside, con- teins) for degradation of sphingolipids in the lyso-
sists mainly of glucocerebroside, which is an unusual some. Substrates cleaved by β-galactosidase differ in
cerebral constituent after the infantile period. their requirement of such activator proteins. Sphin-
There is a 20- to 50-fold accumulation of GM1 gan- golipid activator protein 1 (called SAP-B or saposin
glioside in liver and spleen. Furthermore, there is vis- B) is required for the cleavage of GM1 ganglioside by
ceral accumulation of galactose-containing oligosac- β-galactosidase. SAP-B acts on GM1 as a kind of solu-
charides, which by far exceeds the accumulation of bilizer. SAP-B shares a common larger precursor pro-
gangliosides. Some oligosaccharide fractions contain tein, called prosaposin, with some other SAPs. Pro-
sialic acid. The oligosaccharide accumulation rather saposin is encoded by a gene on chromosome
than the ganglioside storage is the chief cause of the 10q22.1, PSAP. SAP-B not only activates β-galactosi-
visceral histiocytic vacuolation. Storage of galactose- dase to cleave GM1 ganglioside, but also activates the
containing, partially degraded, derivatives of keratan cleavage of sulfatide by arylsulfatase A and globo-
sulfate has been demonstrated in liver and brain. triaosylceramide by α-galactosidase.
In type 2 GM1 gangliosidosis the concentration of Two diseases have been recognized for deficiency
GM1 ganglioside and its asialo derivative are moder- of β-galactosidase: GM1 gangliosidosis, a neuro-
ately elevated in the brain, to a considerably lesser de- degenerative disorder with visceral involvement,
gree than in type 1 GM1 gangliosidosis. The myelin and mucopolysaccharidosis type IV B, also called
lipids, such as cholesterol, phospholipids, sulfatide, Morquio B disease, a generalized bone disease. Mole-
and cerebroside, are much closer to normal than in cular analysis has confirmed allelic mutations of the
type 1 GM1 gangliosidosis. Visceral accumulation of same gene in these two diseases with diverse pheno-
oligosaccharides is less marked. typic expressions. It has been suggested that this mul-
In type 3 GM1 gangliosidosis the accumulation of tiplicity of phenotypes can be explained by different
GM1 ganglioside in the brain is more focal. Accumu- alterations in the catalytic activity of the mutant
lation is most marked in the putamen and caudate enzyme, which differentially alters its activity on a
nucleus, where GM1 accounts for 50% or more of all variety of substrates. There is evidence that patients
gangliosides, whereas GM1 accounts for about 30% of with Morquio disease type B retain a higher catalytic
all gangliosides in the white matter and only a slight activity for GM1 ganglioside than for oligosaccha-
increase, if any, in the proportion of GM1 ganglioside rides and mucopolysaccharides. The phenotypic vari-
is noted in the cerebral cortex. Abnormal accumula- ability in clinical symptomatology of GM1 gangliosi-
tion of asialo GM1 is only noted in the basal ganglia. dosis may be explained in the same way. The early in-
There are no abnormalities in the concentrations of fantile form is characterized by neurological dysfunc-
other lipids, such as cholesterol, phospholipids, and tion in combination with bony abnormalities and
9.4 Pathogenetic Considerations 99

visceral storage. In later onset forms, progressive neu- of the membrane and reduce its fluidity. GM1 ganglio-
rological symptoms are present but visceral and bony side, specifically, has a pronounced effect on the re-
abnormalities are minimal or absent. Differences in duction in membrane fluidity as the carbohydrate
residual enzyme activity, different types of mutations moieties of GM1 reduce the rotational freedom with-
affecting different catalytic functions of the enzyme, in the hydrophobic regions of the membrane. The
different rates of turnover of the enzyme and sub- increased cholesterol content also contributes to the
strates in brain, viscera, and bone may be responsible reduction of membrane fluidity. The altered mem-
for these phenotypic variations. An attractive hypo- brane fluidity may influence the synaptic transmis-
thesis is that in some cases the mutant enzyme pos- sion and the activity of membrane-bound enzymes.
sesses about the same low residual activity for each GM1 gangliosidosis is also characterized by inappro-
natural substrate (infantile type), whereas the mutant priate proliferation of secondary neurites and aber-
enzyme possesses significantly different residual rant formation of synapses. This abnormal sprouting
activities for different natural substrates in other cas- may lead to changes in neuronal connectivity, result-
es (juvenile and adult types). ing in specific functional impairment. Evidence has
Apart from GM1 gangliosidosis and Morquio dis- been found for neurotransmitter dysfunction with
ease type B, β-galactosidase deficiency occurs in a disturbed neurotransmitter release and re-uptake
number of disorders caused by different gene muta- and for specific dysfunction of cholinergic and
tions: variant forms of metachromatic leukodystro- GABAergic neurons.
phy, galactosialidosis, and I cell disease. Deficiency of The later onset forms of GM1 gangliosidosis have
the protective protein caused by a protective protein more focal neuronal pathology, which is especially
gene mutation leads to galactosialidosis. Deficiency marked in basal ganglia and the spinal cord, whereas
of the protective protein gives rise to a secondary in the infantile form there is more generalized neu-
deficiency of both β-galactosidase and neuramini- ronal pathology, which is especially pronounced in
dase activity. SAP-B deficiency results in a form of cerebral and cerebellar cortex. We can only speculate
metachromatic leukodystrophy, with a variable con- on the explanation for this phenomenon. The regula-
comitant accumulation of gangliosides and other tion of substrate and enzyme synthesis and turnover
glycosphingolipids in addition to sulfatide storage. may not be identical in different types of cells and
In I-cell disease, or mucolipidosis III, deficiency of may not be the same at all ages, which would change
β-galactosidase is caused by a defect in posttrans- the distribution of cells in which saturation of the
lational processing of the enzyme molecule. residual enzyme occurs earliest and is most promi-
In GM1 gangliosidosis, GM1 ganglioside and its nent.
asialo derivative accumulate in lysosomes owing to Significant white matter changes are only present
the impairment of normal degradation. GM1 is a nor- in type 1 GM1 gangliosidosis. There is a severe myelin
mal component of cellular membranes, and its con- deficiency, which is caused mainly by a disturbance of
tent is especially high in neuronal plasma mem- myelinogenesis with seriously delayed and arrested
branes, particularly in the regions of nerve endings myelination. Myelin deposition occurs in close col-
and dendrites. GM1 gangliosides act as binding mole- laboration between axons, oligodendrocytes, and
cules for toxins and hormones and are involved in cell astrocytes. Altered neuronal/axonal membrane prop-
differentiation and cell–cell interaction. They stimu- erties and disturbed oligodendroglial-axonal com-
late neurite outgrowth and enhance the action of munication may be at the basis of the disturbed
nerve growth factor. In GM1 gangliosidosis accumula- myelination process. Disturbances of oligoden-
tion occurs predominantly in neurons, resulting in droglial maturation and myelin production and early
neuronal dysfunction and eventually neuronal cell oligodendroglial cell death through apoptosis are
death. There are several factors, which may contribute contributing factors. However, there is also a compo-
to neuronal dysfunction and death. The accumulated nent of myelin loss. The myelin loss may be secondary
GM1 ganglioside and its asialo derivative are relative- to degeneration of neurons as well as primary owing
ly insoluble in water and aggregate within lamellated to oligodendroglial cell death, altered myelin compo-
membranous bodies in lysosomes. Expanding lyso- sition, and myelin instability.
somes with increasing amounts of stored products Galactose-containing oligosaccharides accumulate
may disturb intracellular transport, in this way dis- in the viscera, particularly in type 1 GM1 gangliosido-
turbing cellular metabolism. Leakage of toxic in- sis. These oligosaccharides are derived from the in-
tralysosomal products of enzymes into the cytoplasm complete degradation of glycoproteins in lysosomes.
during the process of intralysosomal storage may The accumulation of these water-soluble compounds
cause damage. Accumulation of GM1 ganglioside in is responsible for the cytoplasmic vacuolation of vis-
the neuronal membrane results in alterations of ceral cells, the foamy histiocytosis in bone marrow,
membrane structure. Gangliosides contain long, satu- and the vacuoles in circulating lymphocytes. Storage
rated fatty acids, which increase the packing density of galactose-containing, partially degraded deriva-
100 Chapter 9 GM1 Gangliosidosis

tives of keratan sulfate occurs in liver, spleen, brain, intensity within a patient’s abnormal cerebral white
and bone, especially in type 1. Storage of these com- matter, which we found to be related to a higher
pounds in bones is responsible for the bony deformi- myelin content in perivascular regions. The brain
ties. stem, and sometimes the posterior part of the corpus
callosum, are better myelinated, but the cerebellar
white matter can be myelin deficient (Fig. 9.1). The
9.5 Therapy basal ganglia and thalamus display subtle signal
changes on MRI, with a slightly increased or decreased
At present there is no effective treatment for GM1 signal intensity of the thalamus and a slightly in-
gangliosidosis. Hematopoietic stem cell transplanta- creased signal intensity of the basal ganglia on T2-
tion can be attempted, especially in the later onset weighted images and a variably increased signal in-
forms, but insufficient data are as yet available to tensity of the thalamus on T1-weighted images (Fig.
assess its potential. In addition, there is evidence in 9.1, upper row). The pattern closely resembles the
animal experiments that the low molecular com- imaging findings in early-onset GM2 gangliosidosis.
pound 1-deoxygalactonojirimycin may increase β- In type 2 GM1 gangliosidosis, progressive atrophy
galactosidase activity. The compound passes through of the cerebral hemispheres has been described with
the blood–brain barrier. Human studies are in enlargement of the ventricular system and subarach-
progress. Gene therapy is still at the level of experi- noid space and atrophy of the cerebellum and brain
mental research. stem (Fig. 9.2). There are subtle white matter signal
abnormalities, which are commonly seen in neuronal
degenerative disorders and are secondary to loss
9.6 Magnetic Resonance Imaging of neurons and axons and their myelin sheaths
(Fig. 9.2). The CT images that have been published
In type 1 GM1 gangliosidosis CT of the brain typical- suggest that the density of the thalamus and basal
ly shows a slightly increased density of the thalamus ganglia might possibly be mildly increased.
(Fig. 9.1). MRI shows diffuse white matter abnormal- In type 3 GM1 gangliosidosis, elevated T2 signal
ities, partly but not only explained by severely delayed and atrophy may be found bilaterally in the caudate
and disturbed myelination. The signal abnormalities nucleus and putamen. Hypointensity of the globus
are more pronounced than would be expected if they pallidus on T2-weighted images has also been report-
were due merely to delayed myelination (Fig. 9.1). ed. In addition, cerebral atrophy and slight white mat-
White matter gliosis and some myelin destruction ter signal changes secondary to neuronal degenera-
add to the signal changes. We observed a very subtle tion can be found. The images are identical to those of
pattern of radiating lines with a more normal signal adult GM2 gangliosidosis.
9.6 Magnetic Resonance Imaging 101

Fig. 9.1. CT scan and MR images in a 7-month-old boy with The cerebral and cerebellar white matter has a diffusely dis-
type 1 GM1 gangliosidosis. Note the high density of the thala- tributed high signal on T2-weighted images, which is higher
mus on CT.The thalamus and basal ganglia are diffusely slight- than is compatible with hypomyelination only
ly abnormal on both T1- and T2-weighted MR images (first row).
102 Chapter 9 GM1 Gangliosidosis

Fig. 9.2. Sagittal T1-weighted and axial T2-weighted images in T2-weighted images, and the cerebral hemispheres are mildly
a 9-year-old boy with type 2 GM1 gangliosidosis.The thick skull atrophic, suggestive of a primary neuronal degenerative
indicates a long-standing disease process. The cerebral white process.The globus pallidus has a low signal
matter has a slightly higher signal than normal on these
Chapter 10

GM2 Gangliosidosis

10.1 Clinical Features acteristic sign in TSD is megalencephaly, which usu-


and Laboratory Investigations ally becomes prominent at about 2 years of age. By the
age of 2 most patients are completely paralyzed,
GM2 gangliosidoses are inherited disorders of GM2 demented, blind, and deaf with frequent seizures.
ganglioside metabolism. Its inheritance is autosomal Decerebrate posturing may be present. Most patients
recessive. There are three major, biochemically dis- die of bronchopneumonia and emaciation. Death
tinct types: B, O, and AB. Among the B and O types, usually occurs between 2 and 3 years of age, survival
infantile, juvenile, and adult forms can be distin- after the age of 4 being rare.
guished; the AB variant is known only as an infantile The clinical features of SD are similar to those of
form. Infantile type B is the classic Tay-Sachs disease TSD, with the exception of hepatosplenomegaly,
(TSD), and infantile type O is the same as Sandhoff which does not occur in TSD. Occasionally there are
disease (SD). bony deformities similar to those associated with in-
TSD is common in Ashkenazi Jews of eastern Euro- fantile GM1 gangliosidosis. Infantile GM2 gangliosi-
pean origin. In the United States carrier the frequen- dosis type AB is also clinically similar to TSD. These
cy is 1 in 30 among Ashkenazi Jews and only 1 in 380 disorders have no racial predilection.
among other groups. TSD infants seem normal at In addition to the severe infantile forms of GM2
birth, and their early development apparently follows gangliosidosis, later onset forms are known. The so-
a normal pattern. The disease begins at the end of the called juvenile form usually has its onset between 2
first 6 months of life. An exaggerated startle response and 6 years of age. The adult, or rather chronic, form,
is often the earliest symptom, although it is frequent- has its onset between the end of the 1st decade and
ly only recognized in retrospect. It is provoked by the 3rd decade of life. Even later onset has been de-
sudden noise and consists in extension, abduction, scribed. However, the age of onset is difficult to deter-
and elevation of the arms. Listlessness and irritability mine because of the very slow progression of the dis-
usually occur early in the course of disease. Gradual- ease. While the juvenile form has no ethnic predilec-
ly, psychomotor retardation and deterioration with tion, the adult B form is more frequent among Ashke-
loss of skills becomes evident. After 6 months of age nazi Jews than in other ethnic groups. The main
the patient’s vision noticeably deteriorates and hypo- systems affected in the juvenile and adult variants are
tonic motor weakness becomes obvious. Affected in- the cerebellum, the pyramidal cells, the lower motor
fants may crawl, sit unaided, and pull themselves up neurons and, less frequently, the basal ganglia. Atypi-
to a standing position but do not usually manage to cal spinocerebellar ataxia syndromes are common as
walk. By 1 year of age the deterioration of mental and modes of presentation of late-onset GM2 gangliosido-
motor capacities is obvious. The children no longer sis. They are characterized by slowly progressive atax-
sit, hold, or transfer objects; they lose interest in their ia, spasticity, dysarthria, and muscle atrophy. Such
surroundings and usually lie placidly in bed. In the cases have been diagnosed as atypical variants of
2nd year hypotonic motor weakness progresses, and Friedreich ataxia, however, usually without sensory
by the end of the 2nd year generalized flaccid paraly- involvement. In some patients additional abnormali-
sis has developed. The tendon reflexes are increased ties in the form of supranuclear or internuclear oph-
at all stages, and plantar responses may be extensor. thalmoplegia and sensory neuropathy have been de-
In the later stages of the disease signs of spasticity, scribed, but these are rare.Another relatively frequent
dystonia, rigidity, chorea and athetosis may be vari- presentation is as motor neuron disease. Clinical fea-
ably present. At the end of the 1st year of life most tures include weakness, cramps, proximal muscle
children are blind. Ophthalmoscopic examination re- wasting, and fasciculations. This clinical picture
veals a cherry-red spot in one or both maculae in closely resembles the Kugelberg-Welander pheno-
about 90% of the patients. Optic atrophy is also seen. type of spinal muscular atrophy or bulbospinal neu-
Feeding becomes a problem in the 2nd year because ronopathy. Amyotrophic lateral sclerosis-like syn-
of ineffective swallowing. Seizures are rare before the dromes present with involvement of both lower and
age of 1 year, but frequent thereafter. The epileptic upper motor neurons. Apart from paresis, atrophy
manifestations may consist in tonic-clonic seizures, and fasciculations, high reflexes, and extensor plantar
myoclonic epilepsy, and also gelastic epilepsy. A char- reflexes are found. Upper limb postural tremor may
104 Chapter 10 GM2 Gangliosidosis

occur, as in other disorders of the lower motor neu- tested with the conventional, nonsulfated synthetic
ron. Various extrapyramidal features have been de- substrate, but a profound deficiency of hexosamini-
scribed in late-onset GM2 gangliosidosis, either in dase A activity is found on testing with the natural
isolation or in combination with the more common substrate GM2 ganglioside or a sulfated synthetic sub-
motor neuron and cerebellar syndromes. Dystonia, strate. Prenatal diagnosis of these variants of GM2
rigidity, choreiform movements, and athetoid postur- gangliosidosis is possible in the first trimester of
ing have been noted.Another clinical characteristic of pregnancy by enzyme analysis in cultured amniotic
late-onset GM2 gangliosidosis is the high incidence of fluid cells or chorionic villi.
recurrent psychosis. In addition, psychic changes in- In the case of type AB, the activities of hex-
clude anxiety, depression, insomnia, aggressiveness, osaminidase A and B are found to be normal, since in
severe behavioral problems, and disintegration of the this type the defect is a deficiency of the GM2 activa-
personality. The psychic changes may precede all tor protein. In type AB the diagnosis requires either
other manifestations or may appear later. Neurovege- the demonstration of accumulating GM2 ganglioside
tative disorders are common and take the form of in the presence of normal hexosaminidase A and B
sweating impairment, loss of libido, impaired esoph- activities or the demonstration of the GM2 activator
agus motility, fixed cardiac frequency, and orthostat- protein deficiency. GM2 ganglioside accumulation
ic hypotension. Intellectual deterioration is frequent. can be demonstrated in brain biopsy tissue or alter-
Epilepsy may occur, but is not obligatory. Blindness native sources of nervous tissue (rectum, conjuncti-
occurs late in the course of the disease. On ophthal- va), and probably also in CSF, although the sensitivity
moscopic examination, optic atrophy and retinitis and specificity of the latter test is not known. The de-
pigmentosa may be seen at that time, but a cherry-red ficiency of GM2 activator protein can be demonstrat-
spot is not a consistent finding and appears late if at ed by feeding radiolabeled GM2 ganglioside to cul-
all. In the juvenile variant death occurs between 5 and tured fibroblasts and correcting the disturbed degra-
15 years of age, often secondary to bronchopneumo- dation of this substance by the addition of purified
nia. Patients with the adult form usually live for some GM2 activator protein to the culture medium. The
decades. expression level of GM2 activator protein can also be
In the infantile variants, EEG is either normal or assessed in fibroblasts.
shows slight changes during the 1st year of life. In the DNA analysis is possible for all variants of GM2
2nd year there are paroxysmal discharges of high- gangliosidosis. If the mutations responsible are found
voltage, slow-wave activity with single and multiple in a family, carrier testing and prenatal diagnosis
spikes and sharp wave complexes. In the vegetative become more reliable. Pseudodeficiency may occur,
state of the disease there is a marked decrease in spike and DNA analysis helps to ensure the presence of a
discharges. These findings are not specific for infan- benign pseudodeficiency allele. Accurate and inex-
tile GM2 gangliosidosis. In later onset variants the pensive screening tests are available for detection of
EEG shows variable, nonspecific findings. Nerve con- GM2 gangliosidosis carriers. Enzymatic tests are
duction velocities are usually normal in the first stage used, which determine total serum hexosaminidase
of the disease and then decline. EMG shows fascicula- and hexosaminidase A activity; the leukocyte hex-
tions, especially in the proximal muscles, and signs of osaminidase assay is used for confirmation. Nowa-
loss of motor units with collateral reinnervation. days screening for common mutations is preferred in
Muscle biopsy shows signs of neurogenic atrophy populations with a high carrier frequency for certain
with type grouping and increased connective tissue. mutations.
Sural nerve biopsy demonstrates decreased fiber den-
sity. A histogram of counted nerve fibers shows a
decrease in the number of large myelinated fibers and 10.2 Pathology
an increase in small myelinated fibers, indicating
active regeneration. Rectal biopsy reveals swollen In infantile GM2 gangliosidoses the gross changes in
ganglion cells with vacuolated cytoplasm. Ultrastruc- the brain vary with the length of the patient’s life. The
turally, the ganglion cells contain membranous cyto- weight and volume of the brain increase massively
plasmic bodies, which are typically found in neurons during the 2nd year of life. The brain frequently
in GM2 gangliosidosis. weighs over 2000 g (normal weight 1000 g). Enlarge-
A definitive diagnosis is established by assaying ment of the brain causes the gyri to become broad-
hexosaminidase A and B in serum, leukocytes, or cul- ened. The cerebellum, however, is usually atrophic.
tured skin fibroblasts. In the case of variant B, hex- On sectioning, the cut surface is abnormally firm. The
osaminidase A is deficient. In the case of variant O, hemispheric white matter may be gelatinous with
both hexosaminidase A and hexosaminidase B are local cavitation. The ventricles are variably enlarged.
deficient. In the case of variant B1, the activities of Light microscopy shows ubiquitous involvement of
hexosaminidase A and B are found to be normal when the nerve cells throughout the brain, with a predilec-
10.3 Chemical Pathology 105

tion for the neurons in the cerebral hemispheres over the lateral columns and in the pyramidal tracts, but
the ganglion cells of the motor cranial nerves or oth- they are normally myelinated. Microscopic examina-
er brain stem nuclei. There is a diffuse disturbance of tion of the retina reveals extensive degeneration and
the cytoarchitecture of the gray matter with a reduc- loss of ganglion cells. The cytoplasm of the remaining
tion in the number of nerve cells, an unusual increase cells is filled with lipid material similar to that seen in
in size of the remaining neurons, and a concomitant the neurons of the brain. These changes are particu-
augmentation in the number of glial elements. The larly conspicuous in the area of the macula.
neurons are large and distorted as a result of the de- Electron-microscopic studies have shown that the
position of lipid material. They have a distended, cytoplasm of the distended neurons contains so-
rounded outline; their nuclei are displaced to the called membranous cytoplasmic bodies. These are
circumference of the cell and are often shrunken and membrane-bound structures, which contain closely
pyknotic. Cortical neuronal cells have swellings in the packed lamellae, frequently arranged concentrically
proximal axon segment or in the apical dendrite, re- in a regular fashion. The lipid material, which is seen
sulting in so-called meganeurites. As the disease pro- under light microscopy, is located in these membra-
gresses, the neurons gradually disappear. There is a nous cytoplasmic bodies. They occupy a considerable
decrease in the number of axons seen within the proportion of the nerve cell cytoplasm. Their accre-
white matter of the brain, which parallels the process tion within the neuronal cytoplasm causes the enor-
of degeneration of the cerebral cortical nerve cells. mous ballooning of the cell and the displacement of
With progression of the disease there are profound the nucleus to the periphery. Accumulation of these
disturbances in myelination, with evidence of addi- storage bodies in proximal nerve processes leads to
tional myelin loss. The myelin deficiency may be very the formation of meganeurites and megadendrites. It
extensive. In some patients the myelin deficiency is has been shown that these storage bodies are lysoso-
seen predominantly in the centrum semiovale with mal in origin. They are also found in axons and glial
sparing of the subcortical U fibers, but in most cells. In glial cells the deposits are more pleomorphic
patients it involves almost the entire white matter, than in neurons.
including the U fibers. The internal capsule is usually Especially in SD, extraneuronal storage of lipids is
well preserved. The preserved myelin sheaths fre- found. Cells containing stored material are found in
quently appear thinner than normal. Complete ab- the spleen, in renal tubular cells, and in liver cells. The
sence of myelin throughout the hemispheric white deposited material appears to be similar to that of the
matter can occur if the patient survives for a long neurons.
time. The white matter changes cannot be attributed In juvenile and adult GM2 gangliosidoses patho-
to wallerian degeneration only. There is evidence for logical changes predominantly affect the anterior
an additional role of both failure of myelination and horn cells of the spinal cord, the cerebellar cortical
active demyelination: the severity of myelin loss is of- neurons, brain stem nuclei, and basal ganglia. In these
ten greater than the axonal loss, and the tendency to areas prominent neuronal storage and degeneration
softening and cavitation in the most severely affected are present. The cerebral cortex is less severely or
areas is consistent with active demyelination and not minimally involved. This is the reverse of what occurs
with wallerian degeneration only. As the disease pro- in infantile gangliosidoses. The cerebellum is atroph-
gresses, the glial reaction increases and eventually ic. Slight diffuse myelin loss within the cerebral and
large numbers of microglia can be observed as well as cerebellar white matter may be observed.
numerous proliferating astrocytes. The glial cells are
swollen and filled with large globules. The contents of
these glial cells show similar properties to those ob- 10.3 Chemical Pathology
served in neurons. The cerebellum shows extensive
degenerative changes. Narrowing or reduction in size GM2 ganglioside is accumulated in abnormally large
of the cerebellar folia is associated with decreased amounts in GM2 gangliosidoses. In the brain, the con-
numbers of cells in the cerebellar cortex. The Purkin- centration of gangliosides is 100–300 times that in
je cells show extensive damage and those remaining normal brain. The storage patterns of the ganglio-
are filled with the same material that is present in the sides exhibit some characteristic differences in the
neurons of the cerebral cortex. The neurons of the three variants of GM2 gangliosidosis. In all cases the
cerebellar nuclei also show the typical ballooning due accumulation of the ganglioside GM2 is most pro-
to deposition of lipids. The spinal cord neurons un- nounced. It is accompanied by minor storage of its
dergo changes similar to those seen elsewhere in the sialic acid-free derivative, GA2. Variant 0 is character-
CNS. The neurons of the anterior horns are more in- ized by the fact that the nervous tissue contains – in
tensely affected than those of the posterior and later- relative terms – the lowest amount of GM2 and the
al horns. The spinal cord white matter frequently highest amount of GA2. Variant B and variant AB dif-
shows rarefaction of the nerve fibers, particularly in fer from each other in the extent to which GM2 and
106 Chapter 10 GM2 Gangliosidosis

GA2 are accumulated, the accumulation being higher GM2 ganglioside: an activator protein. This activator
in the AB variant. The gangliosides are mainly stored protein is termed GM2 activator protein (GM2AP) or
in the neuronal cells, but the ganglioside concentra- sphingolipid activator protein 3 (SAP-3). The GM2
tion of white matter is also increased. In late-onset activator protein has an isoenzyme specificity for
forms of GM2 gangliosidosis, cerebral levels of GM2 hexosaminidase A, and not for hexosaminidase B or
and GA2 are markedly increased above normal, but S. Interaction of the activator protein with GM2 gan-
not to the extent seen in infantile forms. A regional glioside or related compounds results in the forma-
variation in ganglioside accumulation in the brain tion of a water-soluble dimer. The activator–lipid
can be seen, depending on the variation of neuronal complex binds to a specific recognition site of hex-
storage in the different types of GM2 gangliosidosis. osaminidase A in such a way that the glycosidic bond
Some 30–40% of the lysosomal inclusion bodies is positioned at the active site in the α-subunit. Thus,
consist of GM2 ganglioside. Other components are the GM2 activator functions as a transport protein
proteolipid protein, cholesterol, phospholipids, and rather than as an activator of the enzyme.
glycolipids. The different types of GM2 gangliosidosis are char-
Except for a high concentration of GM2 ganglio- acterized by the isoenzyme, which is missing. In type
side, the change in chemical composition of the white B, there is a deficiency of hexosaminidase A (iso-
matter is nonspecific and reflects the extent of myelin enzyme αβ) resulting from mutations in the gene
deficit. The main findings are decreases in proteolipid encoding the α-chain, HEXA, located on chromo-
protein, total lipids, glycolipids, and phospholipids some 15q23–24. In type O, both hexosaminidase A
and the presence of significant amounts of choles- (αβ) and hexosaminidase B (ββ) are deficient. This
terol esters as a sign of active myelin breakdown. is the result of mutations in the gene encoding the
In the B variant and the AB variant, GM2 ganglio- β-chain on chromosome 5q13, HEXB. Type AB is
side is not stored in large amounts outside the ner- caused by a deficiency of the GM2 activator protein,
vous system. In the O variant there is an extensive encoded by a gene located on chromosome 5q31.3–
storage of globoside in the visceral organs, besides 33.1, GM2A. A special variant of GM2 gangliosidosis
storage of GM2 and GA2 ganglioside. The level of glo- has been described, the B1 variant, which is allelic to
boside is approximately normal in the visceral organs the B variant. In the B1 variant, a mutation affects a
in the B variant and the AB variant. specific α-chain site to which the activator–substrate
complex binds. The mutant enzyme has an almost
normal activity towards substrates that are split at the
10.4 Pathogenetic Considerations active site located on the β-subunit (including non-
sulfated synthetic substrates). It is virtually inactive
Gangliosides are glycosphingolipids, which contain towards the substrates that are exclusively or prefer-
sialic acid in their oligosaccharide chain. GM2 gan- entially cleaved at the active site of the α-subunit
gliosidosis is caused by a deficient activity of the (GM2 ganglioside and also synthetic substrates con-
lysosomal enzyme β-hexosaminidase, also called taining a sulfate group). The B1 mutation appears to
GM2 gangliosidase or β-N-acetylgalactosaminidase. be rare in the homozygous form, but may be more
This enzyme hydrolyzes the terminal N-acetylgalac- commonly encountered in the B/B1 compound het-
tosamine from the ganglioside GM2. Hexosaminidase erozygous form.
is composed of two subunits. The α- and the β-chain The time of onset and clinical severity of the dis-
can associate in different combinations to produce ease are related to the rate of ganglioside accumula-
isoenzymes of different structure and catalytic activ- tion, which is inversely related to the residual activity
ity. Isoenzyme αβ is called hexosaminidase A, isoen- of hexosaminidase in the patient’s tissues. The vari-
zyme ββ hexosaminidase B, isoenzyme αα hex- able residual enzyme activities among infantile, juve-
osaminidase S. Hexosaminidase A cleaves the sub- nile, and adult-onset GM2 gangliosidosis patients are
strates ganglioside GM2, the asialo derivative GA2, related to different mutations present either in the
globoside, neutral oligosaccharides, and negatively homozygous or the compound heterozygous state. In
charged substrates, such as terminal β-linked N- its homozygous state the most common mutation in
acetylglucosamine-6-sulfate contained in keratan sul- the α-subunit gene causes a total absence of hex-
fate, chondroitin sulfate, and dermatan sulfate. Hex- osaminidase A and leads to the severe infantile form
osaminidase B has an overlapping substrate specifici- of the disease, TSD. In contrast, adult α-subunit muta-
ty and cleaves GA2, globoside, and neutral oligosac- tions cause a severe, but not complete, deficiency
charides. Hexosaminidase B does not possess any of hexosaminidase A. Both the infantile and adult
significant ganglioside GM2-cleaving activity. Hex- α-subunit mutations occur with enhanced frequency
osaminidase S has only negligible catalytic activity. among Ashkenazi Jews. Compound heterozygotes
Apart from the α- and β-chains of hexosaminidase, a carrying an infantile and an adult α-subunit mutation
third protein is necessary for in vivo catabolism of on homologous chromosomes have adult-onset GM2
10.5 Therapy 107

gangliosidosis. The patients who are homozygous for different locations are preferentially involved in dif-
the B1 mutation generally belong to the juvenile cate- ferent variants of the disease. It may have something
gory. The clinical severity in compound heterozygotes to do with the relative contribution of pathogenetic
depends on the other allele. When the other allele is mechanisms mentioned in each particular variant. In
totally inactive a late-infantile phenotype results. addition, the regulation of substrate and enzyme syn-
Compound heterozygosity in which the other allele thesis and turnover may not be identical in different
carries an adult GM2 gangliosidosis mutation is re- types of cells and may not be the same over the years,
sponsible for the patients with a chronic form of B1 altering the distribution of cells in which saturation
variant with survival into the third decade of life. For of the residual enzyme occurs most prominently. Im-
the β-subunit gene too, different mutations have been pairment of cellular functions can occur at different
identified and variations in residual enzyme activity threshold values of accumulated gangliosides in dif-
appear to explain the different clinical phenotypes. ferent types of cells at different times.
Gangliosides are typical components of the outer The white matter disease in infantile forms of GM2
leaflet of plasma membranes and are particularly gangliosidosis can be explained by a combination of
abundant in the neuronal plasma membranes. An hypomyelination, myelin loss secondary to wallerian
accumulation of these lipids will therefore occur pre- degeneration, and primary demyelination. The hy-
dominantly in neurons. The accumulation of lipids pomyelination may be secondary to neuronal dys-
occurs primarily inside the lysosomes, where they fail function, as a normal neuron–myelin interaction is
to be broken down in the absence of adequate hex- necessary for normal myelin deposition. The de-
osaminidase activity. The accumulating amphipathic myelination might be explained by altered myelin
lipids will precipitate and form lamellar structures. composition, structure, and stability. The myelin
Although the stored compounds are normal, non- membrane fluidity is decreased by the increased con-
toxic, components of the cell, their excessive storage tent of GM2 ganglioside. GM2 gangliosides contain
will interfere with normal cell function. In cells with long, saturated fatty acid moieties, which increase the
extreme storage mechanical destruction of the neu- packing density of the lipid matrix, resulting in
rons may occur. Undegraded storage material is not reduced fluidity.
completely confined to the lysosomes, but can to
some extent be recycled and reach other compart-
ments, such as the Golgi apparatus and plasma mem- 10.5 Therapy
brane via normal membrane flow. This may lead to
changes in the content and pattern of gangliosides in Treatment in GM2 gangliosidosis is largely restricted
the neuronal plasma membrane. Gangliosides are im- to supportive care and management of intercurrent
plicated in cell–cell communication and recognition problems.Attempts at enzyme replacement have been
phenomena including dendrotogenesis and synapto- made by intravenous, intrathecal, and intraventricu-
genesis. Presence of abnormalities in gangliosides in lar injection of hexosaminidase preparations; these
neuronal plasma membranes interferes with the es- attempts have been unsuccessful. It has been suggest-
tablishment of proper connections and leads to aber- ed that hematopoietic stem cell transplantation might
rant synaptogenesis. Inappropriate proliferation of be successful in halting the disease, but the results so
secondary neurites, a tremendous increase in synap- far have been disappointing. This form of treatment
tic spines on neurons, and formation of meganeurites would have a better chance in the later onset and
and megadendrites occurs. Increased ganglioside slower variants of the disease. Substrate deprivation is
content in plasma membranes results in markedly re- another option. This method uses a specific inhibitor
duced membrane fluidity. Evaluation of neurotrans- of glycolipid biosynthesis to partially reduce the syn-
mitter metabolism has shown reduced high-affinity thesis of the unwanted products. The feasibility of
uptake of glutamate, GABA, and norepinephrine by this approach is presently being tested with N-
synaptosomes. Other studies have suggested abnor- butyldeoxynojirimycin. Oral administration of the
mal calcium homeostasis and interference with sec- compound has been shown to result in the reduced
ond messenger systems. storage of glycolipid in multiple organs, including the
In the infantile form, mechanical storage is respon- brain, and an improved clinical course in mice with
sible for the megalencephaly and may be a major GM2 gangliosidosis. The combination of hematopoi-
cause of neuronal dysfunction and death. In the late- etic stem cell transplantation and substrate depriva-
onset forms the lipid accumulation is much less pro- tion worked even better. The efficacy of the approach
nounced and the other mechanisms mentioned may in humans has to be verified. Gene therapy is still in
be more important in explaining the neuronal dys- the experimental stage.
function. It is difficult to explain why neurons from
108 Chapter 10 GM2 Gangliosidosis

10.6 Magnetic Resonance Imaging similarly high density on CT, the T2 hyperintense sig-
nal abnormalities in the thalami and basal ganglia are
A characteristic abnormality in infantile GM2 gan- lacking, and the white matter disease does not spare
gliosidosis is a homogeneously and symmetrically in- the corpus callosum. The images in GM2 gangliosido-
creased density within the thalami on CT scan sis are indistinguishable from those seen in GM1 gan-
(Fig. 10.1). Sometimes, the caudate nucleus, putamen, gliosidosis.
and globus pallidus are also hyperdense (Fig. 10.1). In late-onset GM2 gangliosidosis, CT and MRI
Thalami have a low or mixed low and high signal in- show cerebral and cerebellar atrophy, generally in
tensity on T2-weighted MR images. They have a high combination with slight white matter signal changes
signal on T1-weighted images. In addition, MRI shows (Figs. 10.3, 10.4). These abnormalities are consistent
high signal intensity abnormalities on T2-weighted with primary neuronal degeneration. Considering
images in the caudate nucleus, globus pallidus, and the histopathological findings, one might expect ab-
putamen on both sides (Fig. 10.2). These nuclei have a normalities in signal intensity on MR images of basal
low or mixed low and high signal intensity on T1- ganglia (Fig. 10.3.).
weighted images. The appearance of the cerebral A highly unusual patient has been reported by
white matter is at first suggestive of delayed myelina- Nassogne et al. (2003): this child presented with pro-
tion, but over time the signal intensity becomes more gressive cerebellar ataxia and Babinski signs at the
markedly abnormal, suggesting a combination of dis- age of 3 years. MRI revealed asymmetrical lesions in
turbed and abnormal myelination and myelin loss. the brain stem and middle cerebellar peduncles with
The corpus callosum is well myelinated and intact. some mass effect. The lesions had a high signal on T2-
The cerebellar white matter may also be insufficient- weighted images and a low signal on T1-weighted im-
ly myelinated and become more deeply abnormal in ages, and did not enhance after contrast. The slight
the course of the disease. In later stages cerebral and mass effect suggested a tumoral or inflammatory
cerebellar atrophy ensues. process.A stereotactic biopsy was performed, and mi-
The finding of a high density of the thalamus on croscopy revealed evidence of lipid storage in neu-
CT scans and low signal intensity of the thalamus on rons and glial cells. Enzymatic analysis revealed a de-
T2-weighted MR images is also seen in globoid cell ficiency of hexosaminidase A, indicative of variant B
leukodystrophy (Krabbe disease). However, in the lat- of GM2 gangliosidosis.
ter disease many more brain structures may show a

Fig. 10.1. The CT scan of a 12-month-


old girl with SD (left) shows the hyper-
density of the thalamus on both sides.
From Brismar et al. (1990), with per-
mission.The CT scan of a 5-year-old
child with TSD (right) shows hyperden-
sity of thalamus, globus pallidus, puta-
men, and caudate nucleus, together
with some diffuse white matter hypo-
density and cerebral atrophy. From
Fukumizu et al. (1992), with permis-
sion
10.6 Magnetic Resonance Imaging 109

Fig. 10.2. The T2-weighted images of an 18-month-old girl mal.The T1-weighted images show that the basal ganglia have
with TSD show abnormal thalami, the signal intensity being an abnormally low signal intensity, whereas the thalamus has
too low. The caudate nucleus, globus pallidus, and putamen an abnormally high signal. The signal intensity of the cerebral
have a high signal intensity.The caudate nucleus has a slightly white matter is inhomogeneous on the T1-weighted images,
swollen aspect. The signal intensity of the cerebral white mat- high in some parts and low in others, suggestive of a combina-
ter is diffusely abnormally high,except for the corpus callosum tion of hypomyelination and myelin loss
and internal capsule.The cerebellar white matter is also abnor-
110 Chapter 10 GM2 Gangliosidosis

Fig. 10.3. T2-weighted images in a 5-year-old boy with juve- tion. The basal ganglia also have a slightly abnormal signal.
nile GM2 gangliosidosis show that the cerebral white matter is Courtesy of Dr. P.G. Barth, Department of Child Neurology,
slightly abnormal, suggestive of underlying axonal degenera- Academic Medical Center, Amsterdam, The Netherlands
10.6 Magnetic Resonance Imaging 111

Fig. 10.4. T2-weighted images in a 9-year-old girl with juve- white matter, as seen in neuronal degenerative disorders.
nile GM2 gangliosidosis show a picture of advanced cerebral Courtesy of Dr. S. Blaser, Department of Diagnostic Imaging,
atrophy. There are mild signal abnormalities in the cerebral Hospital for Sick Children, Toronto
Chapter 11

Fabry Disease

11.1 Clinical Features lenticular deposits, periorbital edema, and retinal


and Laboratory Investigations edema are less frequent. The corneal and lenticular
opacities do not impair the visual acuity. Central reti-
Fabry disease (FD) is an X-linked recessive disorder. nal artery occlusion may cause acute blindness.
The onset of clinical symptoms usually occurs during With increasing age, the major morbid symptoms
childhood or adolescence, but may be as late as the result from the cardiovascular system. Cardiac dis-
third or fourth decade. Early manifestations consist of ease is characterized by mitral insufficiency or aortic
episodic pain in the extremities and a telangiectatic stenosis, left ventricular hypertrophy, hypertrophic
scaly maculopapular rash called angiokeratoma cor- obstructive cardiomyopathy, dysrhythmias, angina
poris diffusum. The angiokeratomas usually appear pectoris, myocardial ischemia and infarction, and
between the ages of 7 and 10 years and consist of dark congestive heart failure. Cardiac disease is worsened
red to black nonblanching macules and papules that by systemic hypertension caused by renal vascular
range in size from punctate to 4 mm in diameter. They disease.
increase in number with time. They have a predilec- Cerebrovascular disease results mainly from small
tion for the genitals, the upper thighs, and the lower vessel involvement with transient ischemic attacks, is-
trunk, and spare the face, scalp, palms, and soles. chemic infarctions, and cerebral hemorrhage. Clinical
Pain is a predominant symptom of FD, both chron- symptomatology includes hemiplegia, hemianesthe-
ic acral paresthesias with burning discomfort in the sia, aphasia, and seizures. Dementia, personality
hands and feet and episodes of excruciating pain, es- changes, and psychosis may appear in older patients.
pecially of the extremities. The pain crises, which last Less prominent clinical signs and symptoms in-
from minutes to several days, are often accompanied clude sensorineural hearing loss, tinnitus, delayed
by fever and an elevated erythrocyte sedimentation puberty, short stature, and dysmorphic facial features
rate. These episodes can easily be mistaken for with thickening of the lips and nasolabial folds.
rheumatic fever. The attacks are triggered by exercise, Chronic airway obstruction may lead to decreased
temperature change, fatigue, and emotional stress. pulmonary function and, rarely, pulmonary insuffi-
The painful crises usually persist throughout life and ciency.
are often the most debilitating aspect of the disorder. Over the years, the patients develop symptoms of
Gastrointestinal symptoms consist of recurrent chronic progressive renal failure with proteinuria,
bouts of abdominal pain, described as colic with lymphedema, other signs of tubular dysfunction,
burning pain, located in the mid and lower abdomen. renal hypertension, and uremia. The median survival
Nausea and vomiting are common. These symptoms of affected males is 50 years. The causes of death are
tend to occur after meals and as a result patients are predominantly cerebrovascular disease and renal fail-
often afraid to eat. These complaints frequently lead ure, unless chronic dialysis or renal transplantation is
to diagnosis as appendicitis or renal colic. Episodic performed.
diarrhea may occur, leading to diagnosis as colitis. Atypical variants have been described with mild
Many patients are underweight. single symptomatology at ages when patients with
Loss of autonomic nerve function leads to hypo- the classical disease are severely affected. Cardiac
hidrosis, which begins at puberty and progresses to involvement can be the sole manifestation of the dis-
an absence of sweating by the third decade of life. ease in some patients.
Hypohidrosis results in temperature intolerance and In heterozygous female patients, clinical expres-
overheating during the summer months. It con- sion is variable. Especially with increasing age, symp-
tributes to the elevation in body temperature during a toms of the disease may become manifest.About 70%
crisis. Saliva and tear formation is also decreased. of carrier females experience some form of neuro-
Ophthalmological abnormalities occur in more pathic pain. The typical whorled corneal opacities
than 90% of the patients and often predate skin can be found in 70–80% of the female carriers. An-
changes. Whorled corneal opacities (cornea verticil- giokeratomas and hypohidrosis are found in some of
lata), posterior linear lenticular cataracts with narrow the females. Gastrointestinal complaints are present
wavy spokes, and dilatation and tortuosity of con- in about 50%. Renal and cardiac disease, if present, is
junctival and retinal vessels are frequent; anterior usually milder than in affected males. Transient is-
11.3 Chemical Pathology 113

chemic attacks and cerebral infarction may occur, but Apart from vascular endothelium, lipid accumula-
much less frequently than in affected males. The me- tion also occurs in the leptomeninges and the choroid
dian survival of carrier females is 70 years, which is an stroma, in astrocytes, and in neurons in layers V and
approximate reduction of 15 years from the general VI of the cerebral cortex, hypothalamus, amygdala,
population. subiculum, entorhinal cortex, substantia nigra, peri-
Maximal motor and sensory nerve conduction ve- aqueductal gray, dorsal motor nucleus of the vagus,
locities are usually normal. In contrast, quantitative midline raphe nuclei in the medulla oblongata, inter-
assessment of thermal thresholds often reveals small- mediolateral cell column, dorsal root ganglia, anteri-
fiber neuropathy. The amplitudes of the motor and or horn of the spinal cord, dorsal horn of the spinal
sensory nerve action potentials are also decreased. cord, Meissner’s and Auerbach’s plexuses, and periph-
The diagnosis of FD is established by demonstra- eral autonomic ganglia. Neurons in other areas of the
tion of a deficiency of α-galactosidase A activity in CNS, including thalamus, subthalamic nucleus, cau-
plasma, leukocytes, urine, cultured skin fibroblasts, or date nucleus, putamen, red nucleus, and cerebellar
single hair roots. Plasma, urine sediment, and cells cortex and nuclei do not show signs of lipid accumu-
can also be assayed for their content of accumulated lation. Neurons of the more superficial layers of the
globotriaosylceramide. The biochemical identifica- cortex contain some lipid but are relatively spared.
tion of female carriers is less reliable because of ran- Quantitative studies of peripheral sensory neurons
dom X-chromosomal inactivation. In female het- and spinal ganglia have shown preferential loss of
erozygotes 25–40% have levels of α-galactosidase A small myelinated and unmyelinated fibers as well as
within the normal range. High-performance liquid small cell bodies of spinal ganglia.
chromatography of urinary sediment glycolipids is a Many organs other than the CNS are also affected.
more sensitive test for the detection of carriers. DNA FD is characterized by widespread deposits of
analysis permits precise heterozygote detection and lipids, occurring predominantly in the lysosomes of
is preferred in families in which the molecular defect endothelial, perithelial, and smooth muscle cells of
has been identified. blood vessels and, to a lesser degree, histiocytes
Prenatal diagnosis can be made by fetal sex deter- and reticular cells of connective tissue. The deposits
mination and enzyme assay in cultured amniotic flu- are also present in epithelial cells of the cornea,
id cells or chorionic villi. In some pregnancies in in renal glomeruli and tubuli, and in cardiac muscle
which the fetus is a female carrier, enzyme activity fibers. The dermal lesions are dilated capillaries
may be very low or deficient in cultured amniotic flu- measuring 0.5–2 mm and underlying a hyperkera-
id cells if they are derived from only a few cell clones totic skin.
which predominantly express the mutant X chromo- Histochemical studies of the deposits show that
some. Without knowledge of the sex of the fetus, this they are PAS-positive, positive with Sudan black,
result can be misinterpreted as indicating an affected show birefringence, and stain with Luxol fast blue.
male fetus. In chorionic villi, an intermediate enzyme Electron microscopic examination reveals that the in-
activity could either indicate an unaffected female clusions are intralysosomal and composed of tightly
carrier or could result from maternal tissue contami- packed lipid lamellae, which may be concentric or
nation of the sample from an affected male pregnan- parallel.
cy. Hence, it is necessary to combine enzyme analysis
with chromosome analysis. DNA analysis is the other
option for prenatal diagnosis in families in which the 11.3 Chemical Pathology
molecular defect has been identified.
FD is characterized by the accumulation of neutral
glycosphingolipids, in particular globotriaosylce-
11.2 Pathology ramide (= trihexosylceramide), galabiosylceramide
(= digalactosylceramide), and, to a lesser extent, oth-
The pathology of the nervous system is mostly relat- er galactolipids. In the CNS an increased content of
ed to vascular changes. Pathological lipid storage globotriaosylceramide is found in FD patients, but no
occurs in vascular endothelium throughout the brain evidence has been found of an increase in galabiosyl-
and spinal cord, leading to thickening of vessel walls ceramide. The most dramatic increases in globotriao-
and obstruction of vessels resulting in infarcts. Small sylceramide are found in the dorsal root ganglia,
vessels are particularly involved. Lacunar infarcts are choroid plexus, and leptomeninges, where they can
seen in the basal nuclei and central white matter. exceed 150-fold. Furthermore, globotriaosylceramide
Larger infarcts may also be seen. The blood vessels of is present in CSF of FD patients, in contrast to normal
the peripheral nerves are involved as well. CSF.
114 Chapter 11 Fabry Disease

11.4 Pathogenetic Considerations mote neuronal absorption from blood, but not all in-
volved neuronal groups are in such areas. Absorption
The basic defect in FD is deficient activity of α-galac- of globotriaosylceramide from the CSF into adjacent
tosidase A, the lysosomal enzyme responsible for the neurons could be a possible mechanism. Many of the
hydrolysis of terminal α-galactosyl residues from gly- involved neuronal groups are located adjacent to the
colipids and glycoproteins. The gene encoding for the CSF. There is, however, selective sparing of neighbor-
enzyme is GALA and is localized on the long arm of ing neuronal groups similarly exposed to CSF. Selec-
the X chromosome (Xq22.11). Different mutations tive uptake and transfer of globotriaosylceramide by
have been identified. In male patients with classical neurons could play a role.
FD (classical hemizygotes) there is no detectable Although structural compromise to the cerebral,
enzyme activity and either no detectable enzyme pro- renal, and cardiac arterial vasculature is believed to
tein or normal or decreased amounts of enzyme pro- play a major role in the ischemic events in FD, there is
tein. In the latter case, presumably, the enzyme is also evidence for increased endothelium-mediated
altered and kinetically defective. In mild, atypical vascular reactivity and hyperdynamic cerebral circu-
hemizygotes, some residual enzyme activity is found. lation. Patients with FD have been found to have in-
Females have a variable clinical phenotype related to creased cerebral blood flow velocities. It is presently
random X inactivation. unclear how the disturbance in regulation of the vas-
Glycosphingolipids are important constituents of cular tone contributes to the ischemic incidents. The
plasma cell membranes and of some intracellular white matter involvement in FD is of hypoxic–is-
membranes including lysosomal membranes. Defi- chemic origin, related to small vessel disease, and not
ciency of α-galactosidase A leads to progressive accu- demyelinating in nature.
mulation of neutral glycosphingolipids with terminal The correlation of neurological complaints and
α-galactosyl residues in lysosomes. The highest in- lipid storage is hampered by the presence of a combi-
crease is found in globotriaosylceramide and digalac- nation of neuronal storage, angiopathic infarcts in
tosylceramide. FD hemizygotes and heterozygotes nervous tissue, and deposition of glycosphingolipids
who have blood group B or AB also accumulate B and in end-organs such as the sweat glands in the skin.
B1 glycosphingolipids, which are normal human ery- The episodic limb pain typical of FD has been as-
throcyte antigens. Another neutral glycosphingolipid cribed to dorsal root ganglia neuropathy, peripheral
that can accumulate in FD is the P1 blood group anti- small-fiber neuropathy, involvement of substantia
gen. gelatinosa neurons, and peripheral nerve ischemia
There are several factors that may contribute to the due to involvement of the vasa nervorum. Autonomic
phenotypic expression. First of all, there is the type of dysfunction could arise from involvement of the
mutation. The blood group may also have an effect on autonomic nervous system at either central or pe-
the severity of the disease. It has often been reported ripheral level, but anhidrosis could also be explained
that patients with blood group types B and BA are by dysfunction of sweat glands. The episodic fever
more severely affected than patients with blood may be related to lesions of the hypothalamus and to
group types O and A. This can be due to the fact that the inability to sweat. The clinical correlate of the
patients with blood groups B and BA accumulate the cerebral neuronal glycosphingolipid deposition is
erythrocyte antigens B and B1. In female hetero- unclear. Psychosis, personality changes, and dementia
zygotes, the phenotype is also influenced by the pat- have been described in FD but are not prominent
tern of X chromosome inactivation. phenomena. Seizures are rare. Apparently the accu-
The pattern of glycosphingolipid accumulation in mulation of glycosphingolipids in neurons is a prob-
FD differs from that in other glycosphingolipidoses. lem of lesser importance than their accumulation in
There is a very special cellular and tissue distribution endothelial cells, producing occlusive angiopathy and
of accumulated glycosphingolipids with particular cerebral infarction.
involvement of vascular endothelium, smooth mus-
cles, and neurons. Within the nervous system the pat-
tern of involved neurons is also very special. A num- 11.5 Therapy
ber of explanations have been given for this distribu-
tion of neuronal storage. Site-specific differences in Recently, a breakthrough in the treatment of FD pa-
globotriaosylceramide metabolism have been pro- tients has been accomplished by the successful pro-
posed. Absorption of high levels of globotriaosylce- duction of recombinant α-galactosidase A. The first
ramide from blood has been suggested as the source results of trials of enzyme replacement therapy are
of glycosphingolipids in cells, as concentrations of very promising. Intravenous administration every
this substance are much elevated in the blood of other week was well tolerated and cleared the globo-
patients with FD. Anatomical location in areas of triaosylceramide deposits in the vascular endotheli-
reduced blood–brain barrier could potentially pro- um of the kidney, heart, and skin and led to a decrease
11.6 Magnetic Resonance Imaging 115

in globotriaosylceramide concentration in urine sed- reported in several patients, in others no positive ef-
iment and plasma. The treatment led to significant fect could be demonstrated. Significant lipid deposi-
improvement in renal function, cardiac function, car- tion and allograft dysfunction have been reported
diac conduction, and hearing. Clinically, the patients several years after transplantation. Several patients
had less neuropathic complaints. Patients with FD who underwent successful engraftment died 10–15
have elevated cerebral blood flow velocities. These ve- years later from complications of cardiac disease.
locities improved significantly with enzyme replace-
ment therapy. Similar positive results have been ob-
served in affected carrier FD females. 11.6 Magnetic Resonance Imaging
Certain missense mutations produce catalytically
active mutant α-galactosidase A that is unstable and MRI in FD changes with the course of time. Increas-
rapidly degraded. Reversible competitive inhibitors ing with age, 20–30% of the patients have a high sig-
of α-galactosidase A, such as 1-deoxygalactonojir- nal in the pulvinar on T1-weighted images, whereas
imycin and galactose, have been shown to be able susceptibility-weighted T2* studies demonstrate low
to increase or stabilize the activity of the residual signal intensity in the more severe cases, suggesting
mutant α-galactosidase A. Intravenous infusions of mineralization of the pulvinar (Fig. 11.1). This has
galactose every other day led to an increase in α- been confirmed by CT. CT may show more extensive
galactosidase A activity in circulating lymphocytes calcium deposits, particularly in more severely affect-
and endomyocardial cells and improved cardiac func- ed patients, involving the cerebral cortical–subcorti-
tion in an FD patient with cardiomyopathy as the on- cal junction, globus pallidus, pulvinar, and cerebellar
ly clinical manifestation. These findings suggest that corticomedullary junction. In cases of early and mild
the administration of competitive inhibitors as chem- cerebral involvement, multiple bilateral lacunar in-
ical chaperones at subinhibitory intracellular concen- farcts are seen (Figs. 11.1 and 11.2). These may occur
trations may be efficacious in the treatment of partic- anywhere in the brain, in both gray and white matter
ular variants of FD. structures. The number of small infarcts spread over
Substrate deprivation is a rational therapy for FD. the brain may become very high. In some patients an
This approach is based on the inhibition of an earlier additional small rim of periventricular signal abnor-
step in the synthesis of the accumulating glycosphin- mality is seen (Fig. 11.3). In older patients extensive
golipids. Globotriaosylceramide contains glucosylce- confluent periventricular white abnormalities are
ramide as its base cerebroside. D-threo-1-ethylene- seen in combination with small lacunar infarcts
dioxyphenyl-2-palmitoylamino-3-pyrrolidinopropan- elsewhere in the brain, especially the basal nuclei
ol is a potent inhibitor of glucosylceramide synthase. (Fig. 11.4). The pattern of extensive and confluent
Administration of the compound to FD knock-out white matter involvement may resemble a demyeli-
mice led to a concentration-dependent decrease in nating disease. However, the presence of additional
globotriaosylceramide levels in kidney, liver, and small lesions in the basal ganglia and brain stem
spleen. The effects on brain levels were less profound. should suggest the possibility of an underlying vascu-
The results of human studies are still awaited. lar disorder. This pattern closely resembles the pat-
Gene therapy for FD is in the experimental stage. tern of Binswanger disease. The anterior temporal ab-
Transplantation of genetically corrected bone mar- normalities typically seen in CADASIL are lacking.
row cells in α-galactosidase-A-deficient mice in- Large infarcts in the territories of the major cerebral
creased α-galactosidase A activity and decreased arteries and cerebral hemorrhages may also occur.
globotriaosylceramide storage in all organs except Ectatic vessels are apparent in some patients. Dilata-
the brain. tion of the ventricles and cortical sulci may occur in
Symptomatic care is important with regard to severe disease. If present, the hyperintensity of the
cardiac, pulmonary, and neurological manifestations. pulvinar on T1-weighted images should suggest FD.
The pain in FD can in many cases be reduced by In female heterozygotes, MRI is often normal.
antiepileptic drugs, such as phenytoin, carbamaze- However, multiple small lesions in the deep white
pine, gabapentin, and lamotrigine. Renal insufficien- matter, thalamus, and basal ganglia as well as more
cy requires chronic hemodialysis and/or renal trans- confluent periventricular white matter abnormalities
plantation. In addition to correcting the chronic renal in combination with lacunar infarctions elsewhere in
failure, kidney transplantation provides a source of the brain may occur, but usually at a later age than in
α-galactosidase A. Although a transient or sustained affected males. Hyperintensity of the pulvinar on T1-
biochemical and/or clinical improvement has been weighted images may occur as well.
116 Chapter 11 Fabry Disease

Fig. 11.1. A 42-year-old male patient with Fabry disease. The weighted images, a low signal on susceptibility-weighted T2*
T2-weighted (FSE) images (second row) show two lacunar images (first row, middle and right). Courtesy of Dr. R. Schiff-
infarcts. The CT scan demonstrates calcium deposits in the mann, Developmental and Metabolic Neurology Branch,
pulvinar (first row, left). The pulvinar has a high signal on T1- National Institutes of Health, Bethesda, Maryland, USA
11.6 Magnetic Resonance Imaging 117

Fig. 11.2. FLAIR images in a 44-year-old male patient with mann, Developmental and Metabolic Neurology Branch,
Fabry disease show multiple small infarcts and a rim of high National Institutes of Health, Bethesda, Maryland, USA
signal surrounding the ventricles. Courtesy of Dr. R. Schiff-

Fig. 11.3. FLAIR images and a T1-weighted image (second row, right. The T1-weighted image shows a high signal in the pulv-
right) of a 57-year-old male patient with Fabry disease, show- inar. Courtesy of Dr. R. Schiffmann, Developmental and Meta-
ing multiple infarctions which are partially confluent. There is bolic Neurology Branch, National Institutes of Health, Bethes-
a cystic lesion in the medial cerebral artery territory on the da, Maryland, USA
118 Chapter 11 Fabry Disease

Fig. 11.4. Sagittal T2-weighted images in a 58-year-old male same level (third row, right) shows that part of the lesions are
patient with Fabry disease (first row) reveal lesions in the brain cystic. In addition, lesions are present in the brain stem, cere-
stem and corpus callosum. The axial FLAIR images demon- bellar white matter, and basal ganglia. Courtesy of Dr. R. Schiff-
strate extensive, irregular lesions in the periventricular white mann, Developmental and Metabolic Neurology Branch,
matter. Comparing the FLAIR and T2-weighted image at the National Institutes of Health, Bethesda, Maryland, USA
Chapter 12

Fucosidosis

12.1 Clinical Features the mutations responsible in the family at risk have
and Laboratory Investigations been identified.

Fucosidosis is a very rare, autosomal recessive neuro-


visceral storage disorder. Two clinical variants have 12.2 Pathology
been described, but in fact the two types represent a
continuous clinical spectrum. Mild and severe forms The brain may be enlarged or small, depending on the
can occur within the same family. stage of the disease. The most striking feature is dif-
Type I manifests before the end of the first year of fuse neuronal ballooning and neuronal loss. The cy-
life with frequent respiratory infections. From the age toplasm of remaining neurons is packed with small
of about 1 year, mental and motor regression occurs. vacuoles. These neuronal changes are seen every-
Initially, the children show signs of hypotonic weak- where in the gray matter. There are prominent white
ness, but later hypertonia and spasticity develop. matter abnormalities, variably described as deficient
Seizures may occur. In the end stage decorticate and myelination or demyelination. The white matter is
decerebrate postures are seen. The children’s facial deficient in myelin and gliotic.
appearance resembles that seen in mucopolysaccha- Electron microscopy demonstrates that the vac-
ridosis type I, with coarse features and a protruding uoles present in brain cells are membrane-bound.
tongue. Other similarities between these disorders Many vacuoles contain two different components,
are growth retardation and a mild to moderate dysos- moderately electron-dense reticular materials and
tosis multiplex. Hepatosplenomegaly, cardiomegaly, parallel lamellae. The vacuoles are present in neu-
and anhidrosis with a marked increase in the sodium rons, astrocytes, and oligodendroglia.
chloride content of sweat are present. Death occurs in Enlargement of many internal organs is found, in-
the first decade of life. cluding liver, spleen, heart, pancreas, thymus, thyroid,
Type II has a milder and more prolonged course. and kidneys. Marked vacuolar storage is present in
Mental retardation becomes evident between the ages hepatocytes, Kupffer cells, and bile duct epithelium.
of 1 and 2 years. The coarse facies, growth retarda- The gall bladder may be “strawberry-like” and non-
tion, and skeletal deformities are very similar to those functioning and the adrenals may be small and at-
seen in mucopolysaccharidosis type I. Angioker- rophic. Granulovacuolar storage is seen in almost all
atoma corporis diffusum is a very special characteris- organs, including kidney, spleen, lymph nodes, lungs,
tic of this type and is identical to that occurring in heart, endocrine glands, and sweat glands. In addi-
Fabry disease. The content of sodium chloride in tion, vacuoles are present in vascular endothelial
sweat is normal, although anhidrosis may be present. cells, fibroblasts, bone marrow cells, and circulating
The patients suffer from similar but milder neurolo- lymphocytes.
gical signs than in type I. These patients survive much
longer and often reach adulthood.
In the urine of patients with fucosidosis, an in- 12.3 Pathogenetic Considerations
creased level of fucose-containing glycoconjugates is
found, including fuco-oligosaccharides and fucogly- Fucosidosis is caused by a deficiency of the lysosomal
copeptides. There is evidence that fucosidosis types I enzyme acidic a-L-fucosidase. This enzyme hy-
and II can be distinguished by the pattern of urinary drolyzes a-fucose from glycolipids and glycoproteins.
excretion. Cytoplasmic vacuolation in circulating Fucose is a normal sugar constituent of many tissue
lymphocytes is common in type I, less frequent in mucopolysaccharides, plasma glycoproteins, and tis-
type II. Definite diagnosis is established by the sue mucolipids. In fucosidosis, tissues store fucose-
demonstration of a deficiency of the lysosomal en- rich glycolipids, sphingolipids, glycoproteins, oligo-
zyme a-fucosidase in leukocytes, cultured fibro- saccharides, and mucopolysaccharides. The major
blasts, or other tissue cells. Prenatal detection is pos- part of the stored material consists of ceramide con-
sible by assessing enzyme activity in amniotic fluid taining fucose and other hexoses. This material is
cells. DNA-based prenatal diagnosis is possible when derived from secretor antigens and blood group anti-
120 Chapter 12 Fucosidosis

gens, which are fucose-rich glycolipids and glycopro- 12.5 Magnetic Resonance Imaging
teins. In the liver, there is a major accumulation of gly-
colipids. These are only present to a minor extent in CT scan findings include hypodensity of the cerebral
the brain, where oligosaccharides predominate as white matter and globus pallidus. Mildly increased
storage material. density of the thalami has been reported.
The gene encoding a-fucosidase, FUCA1, is locat- The MR images obtained in a fucosidosis patient
ed on chromosome 1 at position 1p34. The observed show symmetrical white matter abnormalities. In
clinical variability among patients cannot be ex- some patients, the white matter signal behavior on
plained by the nature of the mutations in FUCA1 and T1- and T2-weighted images is suggestive of moderate
must be secondary to unknown factors. A region hypomyelination: the white matter is mildly hyperin-
showing homology to the fucosidase gene was identi- tense on both T1- and T2-weighted images (Fig. 12.1).
fied on chromosome 2 and designated FUCA1P. In other patients, however, the white matter has in
FUCA1P does not encode for fucosidase enzyme some parts a higher signal intensity on T2-weighted
activity and is a so-called pseudogene. Because of the images than is usual for hypomyelination, and its sig-
wide variation in clinical severity of fucosidosis, even nal on T1-weighted images is low in places, suggesting
within families, it is of interest to know whether the myelin loss (Fig. 12.2). The subcortical U fibers, inter-
FUCA1P gene could encode for a protein product that nal, external, and extreme capsules, and cerebellar
might affect fucosidase enzyme activity. A third gene, white matter also have an abnormal signal intensity.
FUCA2 on chromosome 6, is thought to regulate the The corpus callosum is normal in signal, but may be
fucosidase activity, although it does not encode for thin. The cerebral white matter may have a decreased
the fucosidase enzyme. volume with enlarged CSF spaces. The internal
medullary laminae of the thalamus and in some cas-
es the lateral and medial medullary laminae of the
12.4 Therapy globus pallidus and hypothalamus have a high signal
on T2-weighted images. The globus pallidus, thala-
To date therapy has been entirely supportive. Hemato- mus, and substantia nigra tend to have an abnormal-
poietic stem cell transplantation has been performed ly low signal on T2-weighted images. Cerebral and
in fucosidase-deficient animals. Following successful cerebellar atrophy may be seen and may be marked,
engraftment, increased levels of fucosidase activity especially in older patients.
were found in leukocytes, plasma, and neural and The involvement of a combination of cerebral
visceral tissue. The enzyme reaches viscera and hemispheric white matter, globus pallidus, and thala-
peripheral nerves rapidly via phagocytes, but it takes mus is reminiscent of Canavan disease and later-on-
months to achieve substantial levels of enzyme activ- set variants of maple syrup urine disease, but in the
ity in the CNS. Long-term engraftment from an early latter two conditions the white matter is swollen,
age reduced the severity and slowed the progression whereas it is reduced in volume in fucosidosis. The
of clinical neurological disease in these animals; images show resemblance with the pattern observed
transplantation after the onset of clinical signs was in infantile GM1 and GM2 gangliosidoses, although
not effective. Hematopoietic stem cell transplantation the hypointensity of central brain nuclei on T2-
has been performed in a few patients and the results weighted images is not seen in the latter disorders.
are promising in those undergoing the transplanta-
tion early. At present, it is still uncertain whether ear-
ly treatment will completely prevent the clinical signs
of disease.
12.5 Magnetic Resonance Imaging 121

Fig. 12.1. The cerebral white matter in this 4-year-old patient signal on T1-weighted images and low signal on T2-weighted
with fucosidosis has a high signal on both T1- and T2-weighted images. Courtesy of Dr. S. Blaser, Department of Diagnostic
images, suggestive of moderate hypomyelination. The globus Imaging, Hospital for Sick Children, Toronto
pallidus, thalamus, and substantia nigra have a strikingly high
122 Chapter 12 Fucosidosis

Fig. 12.2. Part of the cerebral white matter in this 7-year-old abnormalities in the internal laminae of the thalamus. Also
patient with fucosidosis has a high signal on both T1- and note the very low signal of the globus pallidus, thalamus, and
T2-weighted images, suggestive of moderate hypomyelina- substantia nigra on T2-weighted images. Courtesy of Dr.
tion, but parts of the deep cerebral white matter have a S. Blaser, Department of Diagnostic Imaging, Hospital for Sick
higher signal on T2-weighted images and a low signal on Children, Toronto
T1-weighted images, suggestive of myelin loss. Note the signal
Chapter 13

Mucopolysaccharidoses

13.1 Clinical Features Most children with Hurler syndrome manage to walk,
and Laboratory Investigations but develop only limited language skills. They have a
characteristic appearance. Prominent features are
The mucopolysaccharidoses (MPS) constitute a fami- relative macrocephaly, a prominent forehead, coarse
ly of heritable disorders caused by the deficiency of facial features, hypertelorism, flat nasal bridge,
specific lysosomal enzymes involved in the degrada- prominent bushy eyebrows, thick and dry hair, hir-
tion of mucopolysaccharides (glycosaminoglycans). sutism, thick skin, enlarged tongue, hypertrophic
The mucopolysaccharidoses are classified into six gums, short and broad hands with stubby fingers,
groups, which are further subdivided on the basis short and broad feet, exaggerated lumbar lordosis,
of genetic, biochemical, and clinical findings (Table thoracic kyphosis, and a protuberant abdomen,
13.1). These disorders share a number of characteris- frequently with umbilical and inguinal hernias. On
tic clinical features, although there is considerable physical examination hepatosplenomegaly is found.
variability among the MPS types and within one type Patients experience increasing joint stiffness and lim-
of MPS. itation of joint mobility, which may begin in early in-
MPS type I, inherited as an autosomal recessive fancy. The preferentially affected joints are shoulders,
disease, can be divided into three subtypes: Hurler fingers, and wrists. Deformities become apparent
syndrome, Scheie syndrome, and Hurler–Scheie syn- with claw hands and flexion contractures of elbows
drome. The clinical phenotype in Hurler–Scheie syn- and knees. Vision becomes impaired due to progres-
drome is intermediate between the severe presenta- sive corneal clouding. Glaucoma, optic atrophy, and
tion of Hurler syndrome and the mild presentation of pigmentary retinal degeneration can contribute to
Scheie syndrome. the loss of vision. The majority of children have some
Children with Hurler syndrome (MPS I H) appear degree of hearing loss, usually caused by a combina-
normal at birth. They may be unusually large in in- tion of conductive and sensorineural problems. Most
fancy, but subsequent growth is retarded, finally re- patients have recurrent upper respiratory tract infec-
sulting in dwarfism. During the first year of life men- tions, copious nasal discharge, and ear infections.
tal retardation becomes evident, and after several Valvular heart disease is common. Occasionally
years this is followed by progressive deterioration. patients have progressive communicating hydro-

Table 13.1. Classification of the mucopolysaccharidoses


Number Eponym Enzyme deficiency Urinary glycosaminoglycan

MPS I H Hurler a-L-Iduronidase DS, HS


MPS I S Scheie a-L-Iduronidase DS, HS
MPS I H/S Hurler–Scheie a-L-Iduronidase DS, HS
MPS II Hunter Iduronate sulfatase DS, HS
MPS III A Sanfilippo A Heparan N-sulfatase HS
MPS III B Sanfilippo B a-N-Acetylglucosaminidase HS
MPS III C Sanfilippo C acetyl-CoA: a-glucosaminide acyltransferase HS
MPS III D Sanfilippo D N-Acetylglucosamine 6-sulfatase HS
MPS IV A Morquio A Galactose 6-sulfatase KS, C-6-S
MPS IV B Morquio B b-Galactosidase KS
MPS V No longer used – –
MPS VI Maroteaux–Lamy N-Acetylgalactosamine 4-sulfatase DS
MPS VII Sly b-Glucuronidase DS, HS, C-4-S, C-6-S
MPS VIII No longer used – –

DS, dermatan sulfate; HS, heparan sulfate; KS, keratan sulfate; C-4-S, chondroitin-4-sulfate; C-6-S, chondroitin-6-sulfate.
124 Chapter 13 Mucopolysaccharidoses

cephalus caused by dysfunction of arachnoid villi, ored patches over the lower angle of the scapulae and
and some patients have overt signs of increased in- sometimes over the pectoralis area, in the neck and
tracranial pressure. Signs of spinal cord compression on the lateral sides of upper arms and thighs. Progres-
may occur but are infrequent. sive neurological impairment dominates the course
Not all mentioned signs and symptoms are obliga- of the disease. By the age of 6 years, developmental
tory and there is considerable clinical variability. skills begin to plateau and to regress. By the age of 10
However, progressive physical and neurological dete- years, 90% of the patients are bedridden. The neuro-
rioration is the rule. Patients with Hurler syndrome logical deterioration may be worsened by progressive
rarely survive beyond the age of 16 years. Obstructive communicating hydrocephalus. This problem usually
airway disease caused by deposition of mucopolysac- arises between 7 and 10 years. Spinal cord compres-
charides in soft tissue, respiratory infections, and car- sion may contribute to the neurological deterioration.
diac disease are the usual causes of death. Upper and lower respiratory tract disease is common.
Scheie syndrome (MPS I S) represents a mild vari- Tracheal stenosis may occur. Ear infections and pro-
ant of Hurler syndrome. Intelligence is normal. Com- gressive hearing impairment occur in most patients.
mon abnormalities are restricted mobility of joints, There is a high incidence of hepatosplenomegaly and
development of claw hands, hirsutism, variable deaf- inguinal and umbilical hernias. Chronic and in-
ness, corneal clouding, and aortic valve disease. tractable diarrhea is a troublesome problem in many
Stature is normal. Glaucoma and pigmentary retinal of the patients. Cardiac disease is present with valvu-
degeneration may occur, which together with corneal lar dysfunction, myocardial thickening, cardiac fail-
opacities lead to impaired vision. Neurological prob- ure, pulmonary hypertension, coronary artery nar-
lems may occur in the form of carpal tunnel syn- rowing, and myocardial infarction. In end-stage dis-
drome and cervical cord compression by the thick- ease convulsions may occur. Respiratory problems in
ened dura. Psychosis has been described in adults the form of infection or obstruction or cardiac prob-
with Scheie syndrome. First abnormalities are usual- lems superimposed on a condition of emaciation are
ly noted in the second half of the first decade of life, the usual causes of death. Death usually occurs
and the disease is slowly progressive. between 8 and 15 years of age.
Hurler–Scheie syndrome (MPS I H/S) represents a In the mild variant of Hunter syndrome, onset of
variant of intermediate severity with onset of clinical disease is usually between 2 and 6 years. Coarse facial
signs and symptoms between 3 and 8 years. Most pa- appearance is the commonest presenting feature. In-
tients have normal or near-normal intelligence, but telligence is preserved and there are no behavioral
slowly progressive loss of mental capacities may problems. There are, however, obvious somatic prob-
occur. Coarsening of facial features, corneal clouding, lems: hepatosplenomegaly, cardiac symptoms (pri-
joint stiffness, and valvular cardiac disease are fre- marily valvular in origin), upper and lower respirato-
quent. Cervical spinal cord compression may occur. ry tract disease, tracheal stenosis, and inguinal and
Development of communicating hydrocephalus is umbilical herniae. Diarrhea is less frequent than in
rare. Psychosis may occur in adulthood. Most patients severe Hunter syndrome. Hearing impairment is
survive well into adulthood. Cardiac complications common. Subtle corneal opacities have been found.
and upper airway obstruction are the most common Retinal degeneration is much less marked than in
causes of death. severe Hunter syndrome. The picture of chronic
MPS type II or Hunter syndrome is an X-linked papilledema has been observed in about 60% of the
disorder with two clinical phenotypes, a severe type patients, probably due to the deposition of glyco-
(MPS II A) and a mild type (MPS II B). The mild type saminoglycans within the sclerae. The occurrence of
is rarer than the severe type. The most important dis- hydrocephalus is exceptional. Head growth is, how-
tinguishing feature is the presence or absence of men- ever, abnormally rapid with evident macrocephaly.
tal deterioration. In the severe type progressive intel- Cervical myelopathy may occur secondary to at-
lectual decline occurs, whereas intellectual perfor- lantoaxial subluxation and dural thickening. Carpal
mance remains normal or relatively normal in the tunnel syndrome is common. Death usually occurs in
mild type. The age at onset of the severe form is usu- early adulthood, although survival into the fifth and
ally between 1 and 4 years of age. Early abnormalities sixth decade has been described. Death results pri-
are coarse facial features, enlargement of the tongue, marily from cardiac problems, respiratory infections,
growth retardation, mental retardation, severe behav- and upper respiratory airway obstruction.
ioral problems, joint stiffness, gibbus formation, and Female patients with MPS II are extremely rare
skeletal deformities. Head growth is abnormally and are explained by unbalanced expression of the
rapid initially, but slows down after several years. mutant X chromosome.
Retinal degeneration may occur, but there is no Sanfilippo syndrome comprises four different
corneal clouding. Occasionally characteristic skin diseases, MPS III A, B, C, and D, which are caused by
changes are found. They consist of pebbly, ivory-col- different enzyme deficiencies. Clinically, however,
13.1 Clinical Features and Laboratory Investigations 125

they are indistinguishable. Sanfilippo syndrome pre- rough and discolored. Facial features are usually
sents with great inter- and intrafamilial heterogene- coarse and the mouth wide. Corneal opacities may be
ity. The clinical characteristics of the disease are rela- present but are usually mild. Hepatomegaly, cardiac
tively mild somatic features and severe, progressive valvular abnormalities, and inguinal and umbilical
mental deficiency. Early psychomotor development is hernias may occur. Intelligence is usually normal or
usually slightly or moderately delayed. Speech devel- just below normal. Neurological complaints are
opment, in particular, is often slow and poor. Intellec- caused by compression of the medulla or spinal cord
tual deterioration is usually evident by school age. due to atlantoaxial subluxation and diffuse thicken-
Dementia occurs early and progresses rapidly in ing of the cervical dura. The signs of cervical myelo-
some patients, but is more gradual in others. Behav- pathy are usually slowly progressive, but occasionally
ioral disturbances are often dramatic with extreme acute tetraplegia occurs. Cardiac valvular disease and
restlessness and hyperkinesia. Some patients are cervical myelopathy contribute to death, which usual-
withdrawn and lose contact with their environment. ly occurs in late childhood or early adulthood. Sur-
Aggression is often present in stressful situations. vival into the fourth and the fifth decade has been
Most of the children are too mentally disabled to described. As a rule, MPS IV B has a later onset and
attend school, but some are able to attend primary slower course than MPS IV A, but severe forms of
school. Speech deteriorates, becomes slurred, the MPS IV B and mild forms of MPS IV A have been
patient begins to stutter and eventually loses speech described. A variant of MPS IV B has been described
altogether. Motor functions are less frequently and with progressive mental handicap.
less severely affected, but in some patients the gait be- Maroteaux–Lamy syndrome or MPS VI clinically
comes unstable with frequent falling. Facial changes resembles Hurler disease, but intelligence is pre-
are mild or absent in most of the patients. Skeletal in- served. The disease usually presents in the third year
volvement is minimal, with only mild dysostosis mul- of life and is characterized by growth retardation,
tiplex, kyphosis, scoliosis, and usually normal stature coarse facial features similar to but milder than those
for age. Joint stiffness and contractures are mild seen in Hurler syndrome, corneal clouding, joint con-
and rarely cause loss of function. Some patients are tractures, claw hand deformities, kyphosis, protru-
macrocephalic, but most older patients have a normal sion of the sternum, hepatosplenomegaly, umbilical
head circumference and may even be microcephalic. and inguinal hernias, and mild hirsutism. Nerve
Hepatomegaly is usual in younger patients, but is entrapment syndromes may occur, in particular
less frequent in older patients. Splenomegaly is rare. carpal tunnel syndrome. Myelopathy secondary to
Other features that can be found are inguinal and thickening of the cervical dura occurs frequently with
umbilical herniae, coarse hair, hirsutism, and sen- the insidious development of spastic tetraparesis.
sorineural hearing loss. Neurological findings are in- Exceptional cases with mental retardation have been
consistent; they may include hypotonia, hypertonia, described. Cardiac valvular dysfunction resulting in
hyporeflexia, hyperreflexia, tetraparesis, and muscu- cardiac failure is the most common cause of death. In
lar atrophy. Corneas are usually not clouded, but pig- the severe forms death usually occurs in the second or
mentary degeneration of the retina may occur. Some third decade, but patients with milder variants of the
patients develop epilepsy. Many patients develop disease have a longer life expectancy.
swallowing difficulties with time, necessitating tube Sly syndrome or MPS VII is also characterized by
feeding at later stages. Infections and unexplained considerable clinical variation. In the severe neonatal
diarrhea are frequent problems. Cachexia and aspira- form, hydrops fetalis and dysostosis multiplex are
tion pneumonia are common causes of death. Death prominent early features; the course is rapidly fatal.
usually occurs in the second or third decade. The other end of the clinical spectrum is formed by
Morquio syndrome is characterized by marked patients with very mild clinical symptomatology, with
skeletal involvement and preserved intellectual ca- normal intelligence, normal height, absence of coarse
pacities. Two types can be distinguished, MPS IV A facial features, and minimal skeletal abnormalities.
and MPS IV B, characterized by different enzyme de- The classical form of the disease is characterized by
ficiencies. Presenting features of Morquio syndrome coarse facial features, hepatosplenomegaly, diastasis
are growth retardation with dwarfism, short neck and recti, umbilical and inguinal herniae, thoracolumbar
trunk, pigeon breast deformity, kyphosis, hyperlor- gibbus, short stature, metatarsus adductus, and vari-
dosis, scoliosis, genua valga, valgus deformity of the ably present corneal clouding. Respiratory infections
elbow, and ulnar deviation and broadening of the are frequent. Early psychomotor development is nor-
wrist. The tone appears to be decreased due to liga- mal, but after 2 or 3 years of life retardation becomes
mentous laxity. Decreased joint mobility may occur in evident. Retardation is usually moderate, but severe
the large joints. The teeth are usually widely spaced mental deficiency has also been reported. Epilepsy is
and there are numerous minute pits in the abnormal- rare.
ly thin enamel, causing the surface of the teeth to be
126 Chapter 13 Mucopolysaccharidoses

Radiographs show skeletal changes in all MPS onic villus cells. Prenatal diagnosis poses a problem
variants, but the dysostosis multiplex varies in sever- in Hunter syndrome because of the X-linked mode of
ity. Typical findings are cortical sclerosis and thicken- inheritance.When the cells obtained are derived from
ing of the skull, which may involve the base and the only a few cell clones which predominantly express
vault. The pituitary fossa is often elongated and the mutant X chromosome, very low enzyme activity
J-shaped. Teeth are widely spaced. The head is often may be detected in a carrier female fetus. Hence, sex
scaphocephalic and there is early closure of cranial determination is essential in prenatal diagnosis of
sutures, in particular the sagittal and lambdoid Hunter syndrome. In families in which the molecular
sutures. Orbits are shallow. Basilar impression may defect has been elucidated, DNA-based prenatal diag-
occur. In MPS IV odontoid dysplasia is a universal nosis is an option.
finding with a tendency to atlantoaxial subluxation.
Various types of vertebral dysplasia are seen. Clavi-
cles are short and stubby. There is anterior flaring of 13.2 Pathology
the ribs, hypoplasia of the inferior portion of the iliac
bones, flared iliac wings, oblique acetabular margins, Mental deficiency is an important clinical character-
and a valgus deformity of the hips. Long bones are istic of MPS I H, MPS II, MPS III, and MPS VII. Neu-
short and broad with metaphyseal and epiphyseal de- ropathological findings in the syndromes causing
formities. Cortical margins are wavy and scalloped. mental deficiency are similar. In particular, neuronal
Metacarpals and phalanges are widened and short- storage is confined to the MPS variants with intellec-
ened. tual problems.
The MPS variants were originally classified ac- The skull is sometimes grossly thickened. The lep-
cording to the types of glycosaminoglycans excreted tomeninges are thickened and opalescent. There is a
in the urine in addition to consideration of clinical marked increase in connective tissue elements and
features. In MPS I and II dermatan sulfate and he- there are numerous mononuclear cells containing
paran sulfate are excreted, but inheritance is autoso- large cytoplasmic vacuoles. These cells stain positive
mal recessive in MPS I and X-linked in MPS II. MPS for glycosaminoglycans. The blood vessels running
III is associated with excretion of heparan sulfate. over the surface of the brain are prominent and
MPS IV is characterized by excretion of keratan sul- enveloped by the thickened leptomeninges, which
fate, MPS VI by excretion of dermatan sulfate. MPS extend deep into the brain parenchyma.
VII is associated with excretion of dermatan sulfate, The weight of the brain is usually at the upper lim-
heparan sulfate, and chondroitin sulfate.A problem in it of normal or slightly increased. The external sur-
the diagnosis of MPS IV is that keratosulfaturia can face is normal or the gyri are slightly atrophic. At the
be fairly easily missed. Special sensitive techniques cut surfaces increased perivascular spaces with in-
are required for the detection of urinary keratan sul- creased volumes of connective tissue are evident.
fate excretion. An additional problem of diagnosis in Hydrocephalus is a common finding caused by im-
MPS IV is that keratan sulfate excretion may be di- paired circulation of CSF through the subarachnoid
minished both early and late in the disease process. spaces or dysfunction of the arachnoid granulations.
Peripheral blood cells may also show changes re- Within the brain there are two main pathological
lated to the specific type of MPS. Alder–Reilly granu- features which may occur in MPS: increase in perivas-
lation, a coarse reddish-violet granulation present in cular connective tissue, and neuronal storage.
neutrophils in blood films stained with May–Grün- The neuronal changes are ubiquitous but their ex-
wald–Giemsa or Wright stain, is found in MPS VI and tent varies in different parts of the brain. Generally,
MPS VII. Vacuolated lymphocytes may be present in the large nerve cells in the cerebral cortex and brain
MPS IV B. Occasionally vacuolated lymphocytes with stem are the most severely affected. The cytoplasm of
basophilic inclusions can be seen in any of the MPS neurons is distended by an excessive amount of accu-
variants. Metachromatic inclusions in lymphocytes mulated material, which stains positively with vari-
are most prominent in MPS III. On the whole, how- ous Sudan dyes and PAS, corresponding to the pres-
ever, vacuolation of lymphocytes is not usually ence of gangliosides.Variable numbers of neurons are
prominent in MPS. Bone marrow aspirates will reveal in various stages of degeneration and shrinkage. Loss
the presence of storage cells. of nerve cells is usually mild. Electron microscopic
Definitive diagnosis of the MPS is established by examination of neurons shows several types of inclu-
enzyme assays. It is important to realize that not all sions. The most characteristic are the zebra bodies.
MPS patients have glycosaminoglycan elevations in Zebra bodies are single membrane-bound vacuoles
urine. Therefore, enzyme assays should be performed filled with stacked transverse lamellae, separated at
in all cases with strong clinical suspicion. Prenatal intervals by larger clear spaces. In rare instances the
diagnosis is possible for all MPS variants with en- lamellae have a concentric arrangement and look
zyme assays on cultured amniotic fluid cells or chori- very similar to the membranous cytoplasmic bodies
13.4 Pathogenetic Considerations 127

seen in the gangliosidoses. Granular inclusions are In MPS I, MPS II, and MPS III, chemical studies
less common than zebra bodies. Transitional forms demonstrate that in neuronal perikarya glycosamino-
between zebra bodies and bodies with granular mate- glycans and gangliosides are increased. Ganglioside
rial may occur. Inclusions resembling lipofuscin storage involves the gangliosides GM2, GM3, and GD3.
granules are infrequent. All inclusion bodies have These substances together amount to 65% of the gan-
acid phosphatase activity, indicating their lysosomal gliosides stored in neurons. The ganglioside storage is
origin. In MPS the formation of meganeurites has comparable in magnitude to the amounts stored in
been reported as well as the formation of ectopic the gangliosidoses.
secondary neurites. These are identical to those
described in GM1 and GM2 gangliosidoses.
The cerebral white matter and basal ganglia are 13.4 Pathogenetic Considerations
characterized by perivascular lacunation. Radially
oriented, round to oval cystic white matter abnormal- Proteoglycans are complex molecules consisting of
ities are found at cut surfaces. On microscopic exam- long sulfated polysaccharide chains with up to 100
ination, the adventitia of vessels is abnormally thick sugar residues covalently linked to a protein core.
and consists of a delicate fibrous network with nu- Glycosaminoglycans, formerly called mucopolysac-
merous large cells containing large clear inclusions charides, are degradation products derived by proteo-
caused by storage of glycosaminoglycans. On electron lytic removal of the protein core of proteoglycans.
microscopy they appear empty except for a variable Many enzymes are necessary in the intralysosomal
amount of granular dispersed material. A few lamel- stepwise degradation of each of the glycosaminogly-
lar lipid inclusions resembling the zebra bodies are cans dermatan sulfate, heparan sulfate, keratan sul-
also observed. Loss of myelin may occur around the fate, and chondroitin sulfate. In deficiencies of the re-
cysts but is not conspicuous. On occasion, focal areas lated enzymes the undegraded or partially degraded
of demyelination have been described. Some oligo- glycosaminoglycans are stored in the lysosomes.
dendrocytes contain abnormal, clear inclusions. The respective enzyme deficiencies in the var-
In visceral organs a variable degree of cellular vac- ious forms of MPS are listed in Table 13.1. a-L-
uolation is seen caused by glycosaminoglycan accu- Iduronidase, the enzyme that is deficient in MPS I, is
mulation. Hepatocytes and Kupffer cells store gly- encoded by the gene IDUA, located on chromosome
cosaminoglycans, in particular in MPS I, II, and III, in 4p16.3. It hydrolyzes terminal a-L-iduronate residues
which hepatic fibrosis may occur. In addition to gly- from dermatan sulfate and heparan sulfate. Iduronate
cosaminoglycans, Kupffer cells may store ganglio- sulfatase, the enzyme that is deficient in MPS II, is en-
sides. Glycosaminoglycan storage is found in lymph coded by the gene IDS, located on chromosome Xq28.
nodes, spleen, kidney, and the heart. Cardiac valvular It removes a sulfate group from L-iduronate present in
dysfunction is caused by the presence of large foamy dermatan sulfate and heparan sulfate. In a concerted
cells and an increase of connective tissue. Fibroblasts action, the four enzymes related to MPS III accom-
in skin, cornea, and conjunctiva, endothelial cells of plish removal of the variably substituted a-linked
the vascular system, smooth muscle cells, skeletal glucosamine residues from heparan sulfate. Heparan
muscle cells, macrophages, epithelial cells of distal N-sulfatase, the enzyme that is deficient in MPS III A,
and collecting tubules of the kidney, chondrocytes, is encoded by the gene MPS3A, located on chromo-
osteoblasts, and periosteal cells of bone and cartilage some 17q25.3. It removes sulfate groups linked to the
show glycosaminoglycan storage. There are irregular- amino group of glucosamine. The enzyme is impor-
ities in enchondral ossification with variations in size tant in the breakdown of heparan sulfate. The enzyme
and shape of the diaphyses of the long bones, pe- is also called sulfamate sulfohydrolase or sulfamidase.
riosteal fibrosis, and various degrees of fibrosis and a-N-Acetylglucosaminidase, the enzyme deficient in
lipid storage in marrow tissue. MPS III B, is encoded by the gene NAGLU, located on
chromosome 17q21. It removes N-acetylglucosamine
residues in heparan sulfate. Acetyl-CoA:a-glu-
13.3 Chemical Pathology cosamide acetyltransferase, the enzyme that is defi-
cient in MPS III C, is encoded by the gene MPS3C,
Chemical analysis of the brain and leptomeninges located on chromosome 14. It catalyzes the acylation
reveals a highly increased concentration of gly- of glucosamine amino groups that have become ex-
cosaminoglycans. The level of glycosaminoglycans is posed by the action of heparan N-sulfatase. After the
much higher in the meninges than in brain tissue, acylation of glucosamine amino groups, a-N-acetyl-
where the largest amounts of glycosaminoglycans are glucosaminidase removes the N-acetylglucosamine
found in vascular and perivascular tissue. In MPS I group. N-acetyl-glucosamine 6-sulfatase is deficient
and MPS II, it is mainly dermatan sulfate that is stored, in MPS III D; the gene, GNS or G6S, is located on chro-
whereas in MPS III it is mainly heparan sulfate. mosome 12q14. This enzyme desulfates 6-sulfated N-
128 Chapter 13 Mucopolysaccharidoses

acetylglucosamine residues of heparan sulfate and MPS I, II, and III. Intralysosomal storage of ganglio-
keratan sulfate. Since hexosaminidase A can bypass sides leads to the formation of zebra bodies and
the block in the degradation of keratan sulfate, only membranous cytoplasmic bodies, similar to those
the block in the degradation of heparan sulfate is im- seen in GM1 and GM2 gangliosidoses. The question is
portant. Galactose 6-sulfatase, the enzyme deficient what causes the storage of gangliosides. Apart from
in MPS IV A, is encoded by GALNS, located on chro- b-galactosidase, none of the enzymes involved in
mosome 16q24.3. It cleaves sulfate from 6-sulfated MPS plays a role in ganglioside breakdown. It has
galactose residues of keratan sulfate and 6-sulfated N- been demonstrated, however, that the activity of sev-
acetylgalactosamine residues of chondroitin 6-sul- eral additional lysosomal enzymes is reduced in the
fate. b-Galactosidase, the enzyme deficient in MPS IV MPS, probably as a result of inhibition by the accumu-
B, is encoded by a gene GLB1 located on chromosome lating glycosaminoglycans, and this decreased en-
3p21.33. It removes galactose residues of keratan sul- zyme activity may lead to ganglioside storage. The
fate. N-acetylgalactosamine 4-sulfatase, also called accumulation of gangliosides is probably responsible
arylsulfatase B, is deficient in MPS VI. It is encoded by for the formation of meganeurites and ectopic sec-
the gene ARSB, located on chromosome 5q13–14. It ondary neurites, as these are also seen in GM1 and
hydrolyzes the sulfate groups in the 4-position of N- GM2 gangliosidoses. It is striking that the ganglioside
acetylgalactosamine residues in dermatan sulfate and storage is only found in MPS subtypes characterized
chondroitin 4-sulfate. Urinary chondroitin 4-sulfate by mental deficiency. These phenomena are most
is not elevated in MPS VI, probably because the enzy- probably related.
matic block is bypassed by the action of lysosomal There is a striking clinical variability within all
hyaluronidase. b-Glucuronidase, the enzyme defi- MPS subtypes. An example is found in MPS I, in
cient in MPS VII, is encoded by the gene GUSB, locat- which Hurler syndrome is the most severe variant,
ed on chromosome 7q21.11. It removes b-glucuronate Scheie syndrome the mild variant, and the Hurler-
residues present in dermatan sulfate, heparan sulfate, Scheie syndrome in between. The Scheie syndrome
and chondroitin sulfate. was previously classified as a separate disease entity,
The same enzyme deficiency underlies MPS IV B MPS V, until it became known that deficiency of the
and GM1 gangliosidosis. b-Galactosidase hydrolyzes same enzyme underlies both Hurler syndrome and
terminal b-linked galactose residues found in GM1 Scheie syndrome. It is probable that the clinical
ganglioside, glycoproteins, and oligosaccharides, as heterogeneity is caused by the presence of different
well as keratan sulfate. Deficiency of enzyme activity mutant alleles, the Hurler patients having two severe
toward all substrates causes GM1 gangliosidosis. A mutations and the Scheie patients two mild muta-
mutation that predominantly impairs catalytic activ- tions. It is likely that the type of mutation determines
ity towards keratan sulfate results in MPS IV B. the residual activities of the mutant enzymes, which
In the absence of specific enzymes, nondegraded are associated with wide clinical variation within as
or partially degraded glycosaminoglycans accumu- well as between clinical subgroups. Environmental
late in lysosomes and are partially excreted in urine. factors and modifying genes may also play a role in
Glycosaminoglycans normally constitute the “ground clinical severity, being in particular responsible for
substance” of connective tissue. They are attached to intrafamilial variability. A special phenomenon is the
protein in proteoglycans, the macromolecular forms rare occurrence of clinical disease in female MPS II
in which they exist in connective tissue. In MPS main- carriers, caused by unbalanced inactivation of the
ly bone and connective tissue are affected, causing the normal X chromosome.
most characteristic clinical signs and symptoms:
growth retardation, dysostosis multiplex, coarse
facial features, joint stiffness, corneal opacities, valvu- 13.5 Therapy
lar heart disease, and upper airway narrowing. In the
CNS storage of glycosaminoglycans also occurs in Hematopoietic stem cell transplantation presently
connective tissue elements. Thickening of the lep- represents the best therapeutic option in most MPS
tomeninges may lead to hydrocephalus and to com- variants, if performed early in the course of the dis-
pression of the spinal cord in the cervical region, ease. In MPS I H, early transplantation with success-
resulting in cervical myelopathy. Subluxation of the ful engraftment can preserve intellectual function
odontoid process can contribute to spinal cord com- and prevent or improve the systemic manifestations
pression. Storage in perineural tissue may lead to en- of the disease. Depending on the stage in which the
trapment neuropathy. Within the brain, glycosamino- transplantation is performed, mental decline is pre-
glycans are stored in the tissue around vessels. vented or arrest or slowing of the mental regression is
In addition to glycosaminoglycans, there is evi- achieved. Progressive hydrocephalus requiring shunt-
dence of accumulation of gangliosides (GM2, GM3, ing does not occur after transplantation. Hepato-
and GD3) in the brain of MPS patients, especially in splenomegaly, joint stiffness, upper airway obstruc-
13.6 Magnetic Resonance Imaging 129

tion, and cardiac problems resolve or improve. How- carditis prophylaxis should be advised for MPS pa-
ever, hematopoietic stem cell transplantation does tients with valvular abnormalities. Progressive hydro-
not reverse the progression of the skeletal abnormal- cephalus may require ventriculoperitoneal shunting.
ities in MPS I H. Hematopoietic stem cell transplanta- Carpal tunnel syndrome is a common complication
tion also leads to improvement in MPS I H/S and MPS in MPS patients and surgical decompression should
I S, but these patients may be better candidates for en- be performed early, before permanent nerve damage
zyme replacement therapy. Hematopoietic stem cell occurs. Cervical fusion to prevent atlantoaxial sub-
transplantation in MPS II A and B may lead to im- luxation is performed to prevent or treat cervical
provements with respect to organomegaly, airway spinal cord compression, especially in MPS IV. Anes-
obstruction, and cardiac function. However, cognitive thesia poses a particular problem in MPS. Atlantoax-
decline occurs despite early successful transplanta- ial instability requires careful positioning and avoid-
tion in MPS II A. In MPS II B cognitive function re- ance of hyperextension of the neck. Another problem
mains intact, as expected on the basis of the natural may be difficulty in maintaining an adequate airway
history of the disorder. As in MPS II, hematopoietic during anesthesia and postoperative airway obstruc-
stem cell transplantation is able to effectively treat the tion. Sudden cardiovascular collapse may be caused
somatic aspects of MPS III, but the progressive neu- by a combination of valvular disease, myocardial
rocognitive and behavioral deterioration that causes thickening, systemic and pulmonary hypertension,
the dominant problems in the clinical picture of MPS and narrowing of coronary arteries, all contributing
III is not halted. For this reason, hematopoietic stem to congestive heart failure.
cell transplantation is generally not recommended
for MPS III. Hematopoietic stem cell transplantation
is unable to ameliorate the severe skeletal abnormali- 13.6 Magnetic Resonance Imaging
ties which dominate the clinical phenotype in MPS
IV, and this treatment is therefore not recommended The MRI abnormalities found in MPS vary greatly in
for MPS IV. In MPS VI, hematopoietic stem cell trans- severity from absent or negligible to severe, with a
plantation leads to resolution of hepatosplenomegaly marked variation among sibs. However, in them-
and airway obstruction, prevents further cardiopul- selves, the abnormalities are fairly homogeneous.
monary deterioration, and improves joint mobility. Over the years many patients develop white matter
The skeletal abnormalities, however, do not benefit abnormalities. These consist of multiple small spot-
from the treatment. The experience in MPS VII is lim- like lesions dispersed in the white matter, with a
ited due to the rarity of the disease, but there is evi- predilection for the parietal and occipital white mat-
dence for beneficial effects on neurocognitive, motor, ter. The signal intensity follows the signal intensity of
and pulmonary outcome. CSF, indicative of the cystic nature of the lesions. The
Enzyme replacement therapy has been applied so cystic areas often have a radial orientation from the
far only in MPS I. Administration of recombinant subependymal region toward the cortex (Figs. 13.1
a-L-iduronidase leads to a decrease of the hepato- and 13.2). Punched-out cystic areas are often also pre-
splenomegaly, increased growth rate in prepubertal sent in the corpus callosum, best visualized on the
patients, improved joint mobility, and decreased air- sagittal images (Figs. 13.1 and 13.2). These cystic
way obstruction. This mode of treatment is consid- white matter lesions represent the perivascular lacu-
ered for other MPS variants as well. Although sys- nae seen on histopathological examination. In excep-
temic improvement is likely, no beneficial effects on tional cases, the thalamus and basal ganglia have a
the neurological manifestations is expected. honeycomb-like appearance, related to highly en-
Various forms of in vivo and ex vivo gene therapy larged perivascular spaces in these areas. In addition,
are being studied in animal models. T2-weighted and FLAIR images may show multifocal
Symptomatic treatment is very important in MPS. smaller and larger hyperintense areas, the signal in-
Corneal transplantation can be performed in cases of tensity of which does not follow that of CSF. These
corneal clouding; however, poor vision caused by reti- areas may become extensive and confluent, and prob-
nal degeneration or optic atrophy cannot be reversed. ably reflect gliosis, which is also seen on histopatho-
Hearing aids may be helpful in cases of significant logical examination. The white matter abnormalities
hearing loss. Exercise to optimize joint mobility can be progressive on follow-up MRI. MRI and CT
should be started early. Airway obstruction may be have demonstrated that white matter abnormalities
alleviated by tonsillectomy, adenoidectomy, and, if may occur in all MPS variants. MRI may show a delay
necessary, tracheobronchial stent insertion. Some- in myelination in young children.
times a tracheostomy is required. Cardiac evaluation Another frequent observation consists of ventricu-
at regular intervals with echocardiography is impor- lar enlargement, with or without accompanying en-
tant. Cardiac valve replacement is occasionally per- largement of subarachnoid spaces. Enlargement of
formed in cases of valvular disease. Bacterial endo- CSF spaces may occur in all MPS variants. The ven-
130 Chapter 13 Mucopolysaccharidoses

Fig. 13.1. A 3-year-old boy with Hurler syndrome (MPS I). images confirm the enlarged perivascular spaces and show
The sagittal T1-weighted images show the radial stripes of more extensive areas of signal abnormality in the region of
enlarged perivascular spaces, most prominent in the parietal the perivascular spaces. Courtesy of Dr. S. Blaser, Department
region and also seen in the corpus callosum. The T2-weighted of Diagnostic Imaging, Hospital for Sick Children, Toronto

tricular enlargement is of variable severity and is in Fig. 13.2. A 4-year-old boy with Hunter syndrome (MPS II).The
some cases progressive. Some of the patients appear sagittal and axial T1-weighted images show the radial stripes
to have enlarged CSF spaces on the basis of diffuse of enlarged perivascular spaces, most prominent in the pari-
atrophy of the brain parenchyma. In many cases, how- etal region and the corpus callosum. The T2-weighted images
ever, the enlargement of the CSF spaces is caused by confirm the enlarged perivascular spaces and show more ex-
hydrocephalus as a consequence of disturbed CSF re- tensive signal abnormalities in the region of the perivascular
sorption. Signs of hydrocephalus are upward bulging spaces. Courtesy of Dr. S. Blaser, Department of Diagnostic
Imaging, Hospital for Sick Children, Toronto
13.6 Magnetic Resonance Imaging 131

Fig. 13.2.
132 Chapter 13 Mucopolysaccharidoses

Fig. 13.3. Spinal MRI of a 5-year-old boy with Maroteaux– spinal cord. The cervical vertebrae show platyspondyly. In the
Lamy syndrome (MPS VI) shows abnormalities of the cranio- lower part of the vertebral column, T12, L1, L2, and L3 are dys-
cervical junction, with deposits of mucopolysaccharide mate- morphic with agenesis of the anterior parts of the vertebrae,
rial around the dens, narrowing the spinal canal. The T2- resulting in local kyphosis
weighted images show no free CSF space around the cervical

Fig. 13.4. A 5-year-old boy with


Morquio syndrome (MPS IV).The mid-
sagittal images show the intradural
mucopolysaccharide deposits, leading
to severe narrowing of the arachnoid
space at the level of the craniocervical
junction with compression of the
spinal cord. Courtesy of Dr. P.Tortori
Donati, Department of Pediatric
Neuroradiology, G. Gaslini Children’s
Hospital, Genoa, Italy
Chapter 14

Free Sialic Acid Storage Disorder

14.1 Clinical Features there is a phenotypic continuum between ISSD and


and Laboratory Investigations SD.
Light microscopic examination of blood smears
The sialic acid storage disorders include severe infan- and bone marrow specimens reveal vacuolated lym-
tile sialic acid storage disease (ISSD) and a milder phocytes in almost all ISSD patients and in most SD
variant, Salla disease (SD). Intermediate variants have patients; young SD patients in particular may not
also been reported. have them. Electron microscopy of skin or conjuncti-
ISSD is a rare disorder, presenting in the neonatal val biopsy reveals vacuoles in many cell types includ-
period with coarse facial features, hepatospleno- ing fibroblasts, smooth muscle cells, perineural cells,
megaly, and often ascites or hydrops. Cardiomegaly and Schwann cells. These vacuoles are bound by a
and heart failure may also be present. The patients single membrane and prove to be lysosomes. Most of
usually display generalized hypotonia. The subse- them seem empty, but some contain small amounts of
quent clinical course is invariably characterized by fibrillogranular material, membrane fragments, and
failure to thrive and grossly delayed development. occasional dark globules.
Most patients have hypopigmented skin and fair hair. Dysostosis multiplex is found in about half of the
Albinoid fundi and optic atrophy may be present. patients with ISSD. Skeletal abnormalities are rare in
Mild hypertrophy of the tongue and gums may be SD and may include ovoid deformation of the verte-
seen. A nephrotic syndrome may complicate the bral bodies and a thickened calvarium of the skull. In
course of the disease. Spastic tetraparesis develops SD EEG initially shows a slowing of background
with increased muscle tone and hyperactive tendon activity; in some patients epileptic activity is seen.
reflexes. Seizures may occur. There are recurrent res- With increasing age a gradual decrease in amplitude
piratory tract infections. Hypothyroidism has been occurs; in adults a low-voltage EEG is a consistent
reported infrequently. The age at death varies from finding. Motor and sensory nerve condition velocities
soon after birth to 5 years of age. are reduced in about half of the SD patients. So-
SD occurs with a relatively high frequency in Fin- matosensory evoked potentials are abnormal in the
land. Pregnancy and the perinatal period are un- majority of SD patients, but visual and brain stem
eventful. The first clinical signs usually appear at 6–9 auditory evoked potentials are usually normal. The
months of age and include hypotonia, ataxia, and nys- most prominent changes in nerve conduction and
tagmus. Gradually the patients develop spasticity. evoked responses are seen in the patients with the
Many develop signs of athetosis. Motor development most severe disease.
is delayed, and about 30% of the patients never walk Demonstration of increased urinary excretion of
without support. Speech development is also delayed, free sialic acid and sialic acid accumulation in cul-
and the speech is dysarthric. The nystagmus disap- tured fibroblasts is the mainstay of the laboratory
pears. Most patients acquire a divergent squint. Many diagnosis in both ISSD and SD. The levels of sialic
patients are growth-retarded with a height below the acid excretion vary widely among patients. The level
second percentile. Mental development is delayed reflects more or less the severity of the clinical dis-
from early on, and most adults are severely mentally ease. Confirmation of the diagnosis by DNA analysis
handicapped. Facial features may become coarse late is possible.
in the course of the disease. Epileptic seizures may oc- Prenatal diagnosis is possible by assay of free
cur. Rarely, endocrine disturbances have been report- sialic acid in a chorionic villus biopsy or by molecular
ed, including growth hormone deficiency and hypo- studies. In all families in which the mutation is
gonadotropic hypogonadism. The clinical course in known, the latter option is the most reliable, in par-
SD patients is often static for many years, which de- ticular in SD, in which the increase in sialic acid in
lays evaluation for a metabolic disorder. The life span chorionic villi of an affected fetus is less pronounced
is relatively long. Most patients die in their thirties, than in ISSD. The use of cultured amniotic fluid cells
but death in the seventies is also known to occur. is less appropriate because the elevation of the free
A few patients have been reported with a pheno- sialic acid content in these cells may be only moder-
type intermediate between SD and ISSD, and in fact ate.
134 Chapter 14 Free Sialic Acid Storage Disorder

14.2 Pathology 14.4 Pathogenetic Considerations

In ISSD, the brain has a firmer consistency at autopsy. Free sialic acid storage disorders are autosomal reces-
There is some atrophy of the brain which a marked sive lysosomal storage disorders characterized by the
dilatation of the lateral ventricles. On sectioning, the accumulation of the acid monosaccharide sialic acid
white matter is firm and reduced in volume. in lysosomes, caused by a defective efflux of sialic acid
Histological examination shows clear vacuoles in from the lysosomes. The basic defect in both ISSD
neurons and astrocytes at all levels of the CNS. The and SD concerns the lysosomal free sialic acid trans-
stromal cells of the choroid plexus also contain these porter. The related gene SLC17A5 is located on chro-
vacuoles. There may be neuronal loss in some areas. mosome 6q14–15. The gene has also been designated
Axonal spheroids are present in all gray matter struc- AST (for anion and sugar transporter). This gene en-
tures. The white matter is poorly myelinated and codes the protein sialin, a predicted integral lysoso-
severely gliotic. The cerebellar white matter is also mal membrane protein.
poorly myelinated and may show microcalcifications. The anion and sugar transporter does not only
Axonal spheroids are present in the cerebral and cere- transport sialic acid. The transporter carries acid
bellar white matter. monosaccharides, but also aliphatic nonsugar mono-
In SD, there is some external atrophy of the brain at and dicarboxylates. So, N-substituted acid monosac-
autopsy. On sectioning, a marked reduction in white charides (N-acetylneuraminic acid and N-glycolyl-
matter volume is found, whereas the cortex and basal neuraminic acid), glucuronic acids (the class of hex-
ganglia are macroscopically normal. The corpus cal- oses with a carboxyl on C-6), and glucoaldonic acids
losum is very thin. The cerebellum is atrophic with a as well as lactic acid and a-ketoglutarate are all trans-
markedly reduced white matter volume. Brain stem ported by the same carrier. Additionally, a large num-
and spinal cord may appear thinner than normal. ber of organic anions bind to the carrier, inhibiting
Histological examination reveals storage of large transport function.
amounts of lipofuscin in the perikarya of neurons in Both ISSD and SD patients have mutations in the
the cerebral cortex, thalamus, basal ganglia, brain AST gene. Almost all Finnish SD patients have the
stem nuclei, cerebellar cortex, and spinal cord. There same missense mutation in both alleles. The patients
may be extensive loss of Purkinje cells in the cerebel- who are compound heterozygous for the Finnish
lum. The cerebral and to a lesser extent the cerebellar mutation and another mutation usually have a more
white matter demonstrate severe myelin paucity with severe SD phenotype (intermediate phenotype). A
a marked loss of axons as well, accompanied by pro- variety of mutations, including deletions, insertions,
nounced astrogliosis. The remaining axons frequent- missense, and nonsense mutations, are found in ISSD
ly show spheroids. patients. These findings suggest that there is some
On electron microscopy, a single membrane binds genotypic–phenotypic correlation.
the cellular inclusions, indicative of lysosomes. They Sialic acids constitute a family of over 30 com-
are filled with sparse fibrillogranular material and pounds derived from neuraminic acid. In humans,
occasionally small neutral lipid droplets. N-acetylneuraminic acid is the predominant sialic
The sural nerve is normal on light microscopic ex- acid, referred to simply as “sialic acid.” Normally, a
amination, but electron microscopy shows vacuolar small portion of total sialic acid is free in tissues and
inclusions in the cytoplasm of Schwann cells. body fluids. Most sialic acid is bound to glycoconju-
In ISSD widespread storage of fibrillogranular ma- gates, providing these macromolecules with a nega-
terial within large vacuoles is found in virtually all tively charged terminal sugar that serves many func-
tissues of the body, including skin, conjunctiva, liver, tions. No biological role is attributed to free sialic
kidney, myocardium, and bone marrow. Vacuolated acid. Free sialic acid is produced in the lysosome by
lymphocytes can also been seen in blood. In SD the the degradation of sialylated oligosaccharides by
storage is less impressive. neuraminidase. Sialic acid is removed from lyso-
somes for further metabolism by a specific mem-
brane transporter system.
14.3 Chemical Pathology As a result of the defective transport of free sialic
acid from lysosomes, patients store it excessively in
The material stored in ISSD and SD consists almost many types of tissues and cultured fibroblasts and ex-
exclusively of N-acetylneuraminic acid. Studies of crete large amounts in urine. Free sialic acid storage
various tissues including the brain give identical disorders must be discriminated from other forms of
results. sialic acid storage, namely sialidosis, galactosialido-
14.6 Magnetic Resonance Imaging 135

sis, and sialuria. In sialidosis and galactosialidosis, normal or moderately increased in size and the sub-
bound sialic acid accumulates as the terminal sugar of arachnoid spaces are prominent. The corpus callo-
complex oligosaccharides, which remain undegraded sum is thin (Fig. 14.1). The brain stem and cerebellar
as a result of deficient lysosomal sialidase (neu- white matter are usually better myelinated, although
raminidase) or as a result of a defect in protective hypomyelination of the cerebellar white matter has
protein. Sialuria results from the lack of feedback in- also been observed (Fig. 14.1). The internal capsule is
hibition of the enzyme epimerase, which is involved myelinated to a variable extent. In older patients vari-
in the biosynthesis of sialic acid form N-acetylman- able atrophy of the cerebellum and brain stem is
nosamine. found. There is a correlation between the imaging
The pathophysiology of the myelin deficiency is findings and the clinical phenotype: better myelina-
unknown in ISSD and SD. Widespread storage of sial- tion is seen in patients with milder clinical symp-
ic acid in neurons and astrocytes may lead to dys- toms.
function of these cells from early on. Myelin is de- The images in ISSD and SD are similar to those in
posited in a close collaboration and interdependence Pelizaeus–Merzbacher disease. Proton MRS, however,
of neurons, oligodendrocytes, and astrocytes. This distinguishes ISSD and SD from Pelizaeus–Merz-
collaboration may be severely disturbed in the sialic bacher disease by showing elevations of sialic acid.
acid storage disorders, leading to a profound defi- Sialic acid (= N-acetylneuraminic acid) co-resonates
ciency of myelin. with N-acetylaspartate. The result is a very high
“NAA” peak at 2.02 ppm in a patient with a Peli-
zaeus–Merzbacher-like MRI. NAA is not equally high
14.5 Therapy in Pelizaeus–Merzbacher disease.

There is no causal treatment for sialic acid storage


disorders. For both ISSD and SD, treatment is only
supportive.

14.6 Magnetic Resonance Imaging Fig. 14.1. The sagittal images in a 7-year-old girl with Salla
disease demonstrate the thin corpus callosum and cerebellar
In ISSD MRI is characterized by a severe deficiency in atrophy. The axial T2-weighted images show that the cerebral
myelin and a reduction in volume of the cerebral white matter has a high signal intensity throughout, consis-
white matter, with enlargement of the lateral ventri- tent with myelin deficiency. The internal capsule is also hy-
cles and in particular the subarachnoid spaces. The pomyelinated.The cerebellar white matter appears to contain
sylvian fissure may be wide open. more myelin, although still less than normal. The brain stem is
In SD MRI show a similarly serious myelin defi- myelinated best. Courtesy of Dr. J. Østergaard, Department of
ciency, but the reduction in cerebral white matter vol- Pediatrics, and Dr. T. Christensen, MRI Research Center, Univer-
ume is less severe (Fig. 14.1). The lateral ventricles are sity Hospital of Aarhus, Denmark. (Fig. 14.1 see next page)
136 Chapter 14 Free Sialic Acid Storage Disorder

Fig. 14.1.
Chapter 15

Neuronal Ceroid Lipofuscinoses

15.1 Clinical Features with mental retardation (EPMR), and variant forms
and Laboratory Investigations of late-infantile, juvenile, and adult NCL with granu-
lar osmiophilic deposits (see Table 15.1). The classifi-
The neuronal ceroid lipofuscinoses (NCL), often col- cation of NCL is based on the age at onset, clinical
lectively called Batten disease, constitute a group of course of the disease, ultrastructural findings and,
progressive neurodegenerative disorders character- most of all, genetic defect.
ized by accumulation of ceroid lipopigment in lyso- INCL is the earliest and most severe form of NCL.
somes in neurons and other cell types. They have an The disease occurs most frequently in Finland, the
autosomal recessive mode of inheritance. Nine types incidence there being 7:100,000. Early psychomotor
are presently distinguished: infantile NCL (INCL or development is normal until the age of 6–18 months.
Santavuori disease), late-infantile NCL (LINCL or Many children manage to stand up and to speak sin-
Jansky–Bielschowsky disease), the Finnish variant of gle words, but few learn to walk alone. Then rapid
late-infantile NCL (FvLINCL), variant late-infantile psychomotor deterioration sets in. Head growth
NCL (vLINCL), Turkish variant of late-infantile NCL slows down. The children develop muscular hypoto-
(TvLINCL), juvenile NCL (JNCL, Spielmeyer–Vogt nia, microcephaly, ataxia, choreoathetosis, stereo-
disease or Batten disease), adult NCL (ANCL or Kufs typed hand movements, myoclonic jerks, epilepsy,
disease), Northern epilepsy or progressive epilepsy irritability, and visual failure. Most are restless and

Table 15.1. Classification of neuronal ceroid lipofuscinoses


Disease Eponym Deposits Storage material Gene Gene Gene product
location symbol

Infantile form (INCL) Santavuori Granular SAPs A and D 1p32 CLN1 Palmitoyl protein
osmiophilic thioesterase 1
deposits
Variant late-infantile, Granular SAPs A and D 1p32 CLN1 Palmitoyl protein
juvenile, and adult osmiophilic thioesterase 1
forms with GROD deposits
Late-infantile form Jansky- Curvilinear Subunit c of 11p15 CLN2 Tripeptidyl
(LINCL) Bielschowsky bodies ATP synthase peptidase 1
Juvenile form (JNCL) Spielmeyer- Fingerprint bodies Subunit c of 16p12 CLN3 Membrane protein
Vogt/Batten and rectilinear ATP synthase of unknown
profiles dolichols, SAPs, function
amyloid-b peptide
Adult form (ANCL) Kufs Curvilinear bodies, Subunit c ? CLN4 ?
fingerprint bodies, of ATP synthase,
rectilinear profiles SAPs,
amyloid-b peptide
Finnish variant Curvilinear bodies, Subunit c of 13q22 CLN5 Soluble protein
late-infantile (FvLINCL) fingerprint bodies, ATP synthase, of unknown
rectilinear profiles some SAPs A function
and D
Variant late-infantile Curvilinear bodies, Subunit c of 15q21–23 CLN6 Membrane protein
fingerprint bodies, ATP synthase of unknown
rectilinear profiles function
Turkish variant Curvilinear bodies, ? ? CLN7 ?
late-infantile (TvLINCL) fingerprint bodies,
rectilinear profiles
Northern epilepsy Rectilinear profiles Subunit c of 8p23 CLN8 Membrane protein
(EPMR) ATP synthase SAPs, of unknown
amyloid-b peptide function
138 Chapter 15 Neuronal Ceroid Lipofuscinoses

sleep poorly. The stereotypical hand movements re- TvLINCL has its onset between 1 and 7 years of age.
semble those observed in Rett syndrome. Visual fail- The disease is predominantly found within the Turk-
ure becomes apparent at between 12 and 20 months. ish population. Seizures and motor problems are ear-
Most patients are blind before the age of 2. Slow or ab- ly features. Visual impairment has a variable age of
sent pupillary responses, optic atrophy, and macular onset and is the leading symptom in some patients.
and retinal degeneration without pigment aggrega- Subsequently, motor and cognitive deterioration
tions can be found on ophthalmological examination. occurs.
Partial and generalized seizures and, occasionally, a JNCL is the most common type of NCL in the
Lennox–Gastaut-like epileptic syndrome are seen. world, with the highest incidence in North European
Between 24 and 36 months most children have be- countries (0.5:100,000), in particular in Finland
come bedridden. They are grossly mentally retarded (5:100,000). The first clinical symptom is visual fail-
and do not perform voluntary movements. Opistho- ure starting at the age of 4–8 years. Subsequently,
tonus, decorticate posturing, and hyperexcitability to mental slowing, epilepsy, and motor disturbances
any kind of stimulation increasing the myoclonic occur. The disease progresses at a slow pace. Long pe-
jerks are characteristically present. Flexion contrac- riods of ostensible arrest, sometimes even improve-
tures are common.After a number of years the hyper- ment, are common. Ophthalmological examination
excitability ceases. The final vegetative state is domi- reveals macular degeneration and pigmentary retinal
nated by extensor posturing, unresponsiveness, degeneration. Blindness is present by the age of 6–14
blindness, gross microcephaly, stimulus-sensitive years. Seizures increase in frequency and severity and
myoclonus, and seizures. Death ensues before the age are usually of a generalized and complex partial
of 15, usually between 8 and 11 years. nature. Mental deterioration with a widening gap be-
LINCL has its onset between 2 and 4 years of age. tween JNCL patients and classmates become obvious
Usually epilepsy is the first symptom. Often several after the age of 12 years. Patients are unable to follow
types of seizures occur, including generalized a regular school program and begin to lose acquired
tonic–clonic seizures, myoclonic jerks, absences, and cognitive skills. A speech impediment becomes no-
drop attacks. As disease progresses, myoclonia be- ticeable, in that the tone is monotonous. Patients also
comes the most predominant type. Onset of epilepsy demonstrate echolalia and perseveration. Motor skills
is soon followed by mental and motor deterioration. are lost. Rigidity, hypokinesia, and dystonia are fre-
Ataxia, especially truncal ataxia, hypotonia, and quently observed and movements are slow. Intention
dysarthria, occur. Speech diminishes and disappears tremor, tremor at rest, and myoclonias are often not-
within a year. Typically, visual failure appears after the ed. Spasticity does not set in until a late stage and is
neurological symptoms and progression is slow. never severe. Many teenagers become depressed.
Blindness usually occurs at the age of 6 years. Fundus- Some manifest restlessness, rage, physical violence,
copic examination may initially be normal, but with- insomnia, and visual and auditory hallucinations
in 2 years after onset macular degeneration and reti- with paranoid delusions. Chewing and swallowing
nal pigmentation are seen. Progression of disease is become more difficult, eventually necessitating nasal
otherwise rapid with precipitous regression of mental tube feeding. Myoclonia may become severe. Life
and motor faculties. The initial hypotonia gives way expectancy is 18–25 years. Some patients have a so-
to spasticity, painful flexor spasms, and severe flexion called delayed-classic JNCL variant, which progresses
contractures. Excessive drooling and difficulty with less rapidly and leads to death after the age of 25. In
swallowing occur. The final stage of the disease is one addition, there are patients with so-called protracted
of unresponsiveness, myoclonic hyperexcitability, JNCL. In these patients the first symptom, progressive
and blindness. Death occurs between 10 and 15 years. visual loss, occurs at the same time as in classic JNCL,
The most important clinical difference between but mental and motor symptoms become manifest
classic LINCL and FvLINCL is the later onset of clini- later, usually in the third or fourth decade of life.
cal symptomatology in FvLINCL, usually between 4.5 These patients have a different mutation in CLN3,
and 7 years. The first symptoms are mental deteriora- with milder clinical phenotypes.
tion, clumsiness, and visual failure. Other symptoms, ANCL usually starts around the age of 25–30 years.
including epilepsy, ataxia and myoclonia, develop Two main clinical subtypes are usually distinguished,
between the age of 7 and 10 years. although there is considerable overlap. Clinical phe-
Incidence and prevalence figures for vLINCL, notype A is characterized by behavioral changes,
caused by CLN6 mutations, are not known.Visual loss dementia, progressive myoclonus epilepsy, cerebellar
and seizures may occur initially, but in some children ataxia, and dysarthria. Vision is normal and there are
with the onset of the disease after 4 years, symptoms no signs of optic atrophy, macular degeneration, or
may start with epilepsy, ataxia, and myoclonus. Devel- pigmentary retinal degeneration. In the course of the
opmental regression occurs. disease seizures become intractable. Signs of involve-
ment of the pyramidal, extrapyramidal and lower
15.1 Clinical Features and Laboratory Investigations 139

motor neuron systems are absent or seen only termi- no abnormal photic response. ERG becomes absent
nally. Clinical phenotype B is characterized by behav- by the age of 5–7 years. The amplitudes of the VEP
ioral changes, dementia, and motor abnormalities. and SSEP are reduced. In ANCL, the resting EEG is
Cerebellar or extrapyramidal features are prominent, abnormal, but nonspecifically so. An intense photo-
the latter most frequently taking the form of a hyper- paroxysmal response and an unusual sensitivity to
kinetic movement disorder, rarely parkinsonism. low-frequency photic stimulation is of more diagnos-
Tic-like facial dyskinesias may be present. Pyramidal tic significance. ERG and VEP are normal. Giant
signs are rarely prominent. Visual failure and retinal short-latency SSEPs can be observed. They are com-
abnormalities are not present. Seizures are rare and mon to many forms of progressive myoclonus epilep-
may only occur late in the course of the disease. In sy, and are of limited value in the differential diagno-
both phenotypes death usually occurs about 12 years sis. In EPMR, EEG shows progressive slowing of the
after the onset of the disease, ranging from a few years background activity, impaired reactivity to eye open-
to over four decades. ing, and a disappearance of specific sleep patterns. In
Northern epilepsy, also called progressive epilepsy the fully developed disease, there is hardly any differ-
with mental retardation (EPMR), is an autosomal ence between waking and sleep recordings. The inter-
recessive disease with onset between 5 and 10 years of ictal epileptic activity is relatively scanty.
age with generalized tonic–clonic seizures. In a mi- Ultrastructural findings are also important in the
nority of the patients complex partial seizures also diagnosis of NCL. Skin, rectum, skeletal muscle, and
occur during the first few years. The seizures increase conjunctiva biopsy are mostly used for this purpose.
in frequency, reaching a maximum at puberty. After Peripheral lymphocytes can also be examined, in par-
puberty, the frequency of the seizures declines in a ticular in JNCL. In this disorder peripheral lympho-
manner unrelated to changes in medication. After 35 cytes can already be shown to contain vacuoles by
years of age, many patients are almost seizure-free. means of light microscopy. Vacuolated lymphocytes
Mental development is initially normal, but begins to on light microscopy are not a feature of the other NCL
deteriorate 2–5 years after the onset of the epilepsy. forms. Electron microscopic examination of lympho-
Deterioration continues during adulthood despite cytes can be very useful in the diagnosis and subclas-
good epilepsy control. sification of NCL variants. A granular matrix indi-
Findings of repeated neurophysiological examina- cates INCL; curvilinear profiles indicate LINCL; fin-
tions are important in establishing the diagnosis. In gerprint profiles indicate JNCL. The biopsied tissues
INCL, EEG shows characteristic disappearance of examined by electron microscopy show characteris-
sleep spindles from the age of 1.3 years onwards; sleep tic, membrane-bound deposits in vascular endotheli-
spindles are absent by 2.0 years at the latest.When the um, sweat gland epithelium, smooth muscle cells,
patient is 1.5–2 years old, the EEG slows and begins to neurons, and in lymphocytes. In INCL, granular
attenuate, becoming isoelectric at the age of 3–4 osmiophilic deposits are the predominant type of in-
years. There is no abnormal photic response. ERG ab- clusions. In LINCL, FvLINCL, vLINCL, and TvLINCL
normalities, especially a marked loss in amplitude, curvilinear and fingerprint bodies and rectilinear
are early findings and may precede impairment of profiles are seen. In JNCL, fingerprint bodies are most
vision. ERG is usually negative at 12 months of age. numerous. In EPMR rectilinear profiles are found. In
VEP is markedly reduced in amplitude and becomes ANCL either granular osmiophilic deposits or finger-
negative. It has been demonstrated that the SSEP be- print and curvilinear structures are seen. In ANCL, a
comes negative at an early stage, even before the ERG brain biopsy containing full-thickness cortex may be
and VEP become negative. In LINCL, EEG already necessary to demonstrate the disease.
shows typical large polyspikes to a low rate of photic For INCL and LINCL and their variants, definite
stimulation or single flashes at an early stage of the diagnosis is established by the demonstration of defi-
disease. ERG is negative by the age of 3–4 years. cient activity of palmitoyl protein thioesterase 1 and
Other very typical findings are giant VEPs and SSEPs. tripeptidyl peptidase, respectively. Enzyme assays are
In FvLINCL, the latency between onset of the first relatively simple and rapid and should therefore be
clinical symptoms of the disease and the appearance applied liberally in NCL patients to detect possible
of the typical neurophysiological signs, including the INCL and LINCL variants. DNA confirmation is an
negative ERG, the giant VEPs and SSEPs, and the EEG option in CLN1, CLN2, CLN3, CLN5, CLN6, and CLN8.
with spikes to a low rate of photic stimulation, is Prenatal diagnosis is possible using enzyme or
longer than in LINCL. These characteristic abnormal- molecular studies, with the exception of ANCL and
ities appear several years after the onset of clinical TvLINCL. Prenatal diagnosis using electron mi-
symptoms, which is different from classic LINCL. In croscopy of chorionic villus specimens and amniotic
vLINCL the results of neurophysiological studies re- fluid cells, searching for the characteristic inclusions
semble those of classic LINCL. In JNCL, EEG findings in affected fetuses, is an option in these cases but
are progressively abnormal, but not specific. There is requires special expertise.
140 Chapter 15 Neuronal Ceroid Lipofuscinoses

15.2 Pathology cells, cells of the exocrine and endocrine pancreas,


fibroblasts, and about 10% of lymphocytes.
The hallmarks of neuropathological findings in NCL In LINCL, neuropathological changes are less
consist of neuronal lipofuscin storage and neuronal severe. The external atrophy is variable and some-
loss. The storage leads to displacement of the nuclei times more pronounced in the cerebellum than in the
and distention of the proximal axon segment, but cerebral hemispheres. Neuronal storage, neuronal
usually not to ballooning of the cells. On light mi- loss, and mild to moderate cortical astrocytosis are
croscopy the storage material is pale yellow with found, but neurons remain present, even in older pa-
hematoxylin–eosin stains, slightly pigmented and tients. There is also neuronal storage and neuronal
granular. It is strongly PAS-positive, and stains with loss in the basal ganglia, thalamus, and substantia
Sudan dyes and with many other stains for lipofuscin. nigra, but neurons in the hypothalamus, brain stem
Examination with ultraviolet light reveals bright yel- nuclei other than the substantia nigra, and spinal
low autofluorescence. The deposits are readily shown cord are relatively preserved. There may be some loss
by an acid phosphatase reaction, indicative of their of myelin in the white matter, but this is not marked.
lysosomal location. The ultrastructural appearance of The white matter fibrillary gliosis may be more im-
the storage material varies depending on the form of pressive than the myelin loss. In electron microscopy
NCL. the stored material mostly has the appearance of
In INCL atrophy of the brain is exceedingly severe, curvilinear profiles, but may also take the form of
affecting cerebral hemispheres and cerebellum. The fingerprint profiles. Curvilinear profiles consist of
brain stem and spinal cord are relatively spared. thin stacks of short, curved lamellae forming little
Microscopic findings change with age. Up to the age arcs or semicircles. Accumulations of these are mem-
of about 2.5 years, neuronal storage is observed with brane-bound. These curvilinear bodies are also wide-
slight to moderate cortical neuronal loss, intense fib- spread outside the nervous system, similar to what is
rillary astrocytosis, and presence of macrophages. seen in INCL. Although no vacuoles containing lym-
The white matter shows mild changes. From about 2.5 phocytes are seen on light microscopy, curvilinear
to 4 years of age, neurons become grossly depleted. bodies may be shown in lymphocytes on electron
There is massive cortical macrophagocytosis and microscopy.
astrocytosis. The white matter shows moderate to The most important pathological difference be-
severe loss of myelin and significant astrogliosis. The tween classic LINCL and FvLINCL is the presence of
astrocytes are hypertrophic and contain storage ma- not only curvilinear bodies but also fingerprints
terial similar to that of nerve cells. Above the age of and rectilinear profiles at electron microscopy in
about 4 years, the atrophic cortex is entirely depleted FvLINCL. Fingerprint profiles are formed by groups
of nerve cells and consists of a spongy network of of parallel, paired lines, each pair of lines separated by
fibrillary astrocytes and capillaries with some macro- a lucent space. The lines tend to be equidistant,
phages. The white matter shows complete loss of straight or curved. Groups of lines form whorl-like
myelin and axons, also in the subcortical U fibers, and patterns of fingerprints. Rectilinear profiles represent
severe fibrillary astrogliosis. Only a very few myeli- short, oligolamellar stacks with a prevailing straight
nated nerve fibers are still present. At this stage the course. They differ from curvilinear profiles in shape,
cerebellar cortex also shows complete atrophy. Most width, and distinctness of the internal lines. In
subcortical nuclei show florid neuronal storage, loss vLINCL, the neuronal and visceral storage is similar
of neurons, macrophagocytosis, and astrocytosis, in distribution to that in classic LINCL and consists of
although the primary motor and sensory nuclei of a mixture of rectilinear, curvilinear, and fingerprint
brain stem and spinal cord are remarkably resistant. profiles.
The stored material in INCL has a similar ultrastruc- Diffuse cerebral and cerebellar atrophy is also seen
tural appearance wherever it is found. The substance in JNCL, the degree being proportional to the dura-
is osmiophilic and consists of globules with a granu- tion of the disease. The cortical neurons show signs of
lar matrix, present either singly or as aggregates of storage and degeneration. Astrogliosis is usually
globules. The single globules and the aggregates are mild. The white matter shows little evidence of myelin
stored in larger vesicles surrounded by a unit mem- loss. In electron microscopy of stored material, the
brane of lysosomal origin. The deposits have been fingerprint pattern predominates, but may be ad-
designated granular osmiophilic deposits (GROD) mixed with curvilinear and rectilinear profiles. The
and can be found in neurons, ependymal cells, deposits are membrane-bound. In JNCL, too, the stor-
choroid plexus epithelium, astrocytes, macrophages, age bodies are widespread in other tissues than the
Schwann cells, smooth muscle cells, renal glomerular nervous system. A small proportion of the lympho-
endothelium, renal distal tubular epithelium, Kupffer cytic vacuoles seen on light microscopy are shown on
cells, vascular endothelium, germinal epithelium of electron microscopy to contain fragments of finger-
the testis, sweat gland epithelium, thyroid follicle print profiles.
15.4 Pathogenetic Considerations 141

In ANCL, variable cerebral atrophy is found. Neu- Protein is the major component of intralysosomal
ronal storage and neuronal loss is found in the cere- storage material, which accumulates in all forms of
bral cortex (sometimes confined to layers III and V), NCL. Over 40–60% of the dry weight of isolated stor-
the cerebellar cortex, and central nuclei. On electron age material is protein. In INCL the saposins A and B
microscopy fingerprint profiles, curvilinear bodies, constitute a major proportion of the accumulated
or granular osmiophilic deposits are found. These protein. In LINCL, variants of LINCL, JNCL, and
storage bodies can also be found in several nonneur- ANCL, subunit c of the mitochondrial ATP synthetase
al tissues. constitutes a considerable proportion (up to 85% in
In EPMR, most neurons have intracytoplasmic LINCL, about 20% in JNCL) of the accumulated pro-
accumulations of autofluorescent storage material. In tein. The protein molecules are intact and there is no
electron microscopy, the storage material is mem- evidence of any abnormality in amino acid se-
brane-bound and consists of rectilinear profiles. quences. Dolichols, which are unesterified alcohols,
All infantile and childhood forms of NCL have a represent up to 2% of the dry weight of storage bod-
severe, progressive retinopathy. Ganglion cells show ies. The dolichols are largely in the form of dolichyl
signs of lipofuscin pigment storage, similar to neu- pyrophosphoryl oligosaccharides.
rons in the brain. The same pigment is present in the
cells of the bipolar layer. Most conspicuous is the de-
generation and loss of photoreceptor cells, spreading 15.4 Pathogenetic Considerations
from the macula outwards. Severe atrophy of all reti-
nal layers develops, with narrowing of small retinal NCL represents a group of unrelated disorders which
vessels and displacement of melanin-containing pig- share a number of clinical, biochemical, and histo-
mented cells into the atrophic retina through the ex- pathological characteristics. Although NCL variants
ternal limiting membrane. These displaced melanin- are lysosomal storage disorders, they differ from the
containing cells may also harbor NCL-type-specific classic lysosomal storage diseases. The accumulated
lipopigments. On electron microscopy the storage material consists mainly of proteins and not of lipids,
material has the appearance specific for the form of making NCL a proteinosis rather than lipidosis.
NCL. The term “lipopigment” is a general one given to
yellow-brown pigments that stain with lipid stains
and fluoresce under ultraviolet light. The prototype
15.3 Chemical Pathology pigment is lipofuscin (age pigment), ubiquitously
present in cells of aged individuals. The term “ceroid”
Analysis of brain biopsy tissue from INCL cases has been used to describe any abnormal lipopigment,
shows lipid disturbance in the cerebral cortex, but a including the lipopigment of NCL. The nature of the
more severe reduction in myelin lipids in the cerebral fluorophores in the NCL-specific ceroid has been
white matter. At autopsy, when the end stage of the studied but is still not clear.
disease is reached, the cerebral and cerebellar tissues At present there is evidence for at least eight genes
are extremely lipid-poor. The concentrations of cho- that are related to NCL. Currently, six of them have
lesterol and phospholipids in cerebral gray matter are been identified and characterized: CLN1, CLN2,
reduced to about 40% of those in age-matched con- CLN3, CLN5, CLN6, and CLN8. Two of these genes,
trols. In cerebral white matter the lipid changes are CLN1 and CLN2, encode lysosomal enzymes, palmi-
even more pronounced. The concentration of choles- toyl protein thioesterase 1 and tripeptidyl peptidase
terol is reduced to about 10% of the control value, that 1, respectively. CLN3, CLN6, and CLN8 encode pro-
of phospholipids to about 20%, and the concentra- teins of predicted transmembrane topology, but their
tions of cerebrosides and sulfatides to values of often function is as yet unknown. CLN5 is probably a solu-
less than 1%. These results demonstrate an extreme ble lysosomal protein. The genes associated with
reduction of myelin lipids, and in the terminal stage TvLINCL and ANCL are at present unknown. It is im-
of INCL all attempts to isolate myelin from cerebral portant to realize that one clinical type of NCL is not
hemispheres and cerebellum fail. The ganglioside automatically equivalent to one genetic type. Some
concentration and pattern of individual gangliosides adult NLC patients have been shown to have a defect
are the same in gray and white cerebral matter, which in CLN1. It cannot be assumed that all remaining
means a more severe reduction in gangliosides for ANCL patients have a defect in CLN4; there may be
gray matter (to about 15% of control value) than for more than one related gene. Likewise, Turkish pa-
white matter (to about 40% of control value). Lipid tients with a late-infantile onset of disease, who have
changes are much less pronounced in brain stem and the other NCL genes excluded, do not necessarily
spinal cord. The other forms of NCL show only minor share a defect in the same gene (CLN7); again there
lipid alterations. may be more than one related gene.
142 Chapter 15 Neuronal Ceroid Lipofuscinoses

Palmitoyl protein thioesterase 1 (PPT1) is a lysoso- metabolic needs of the cell. TPP1 could provide
mal enzyme that cleaves fatty acyl moieties (usually tripeptides for subsequent action of dipeptidyl pepti-
palmitate) from cysteine residues of proteins. Palmi- dases and dipeptidases. The specificity and substrate
toylation and depalmitoylation is a reversible and dy- range of TPP1 is still under investigation. TPP1 has
namic process, which may regulate the function of been shown to degrade small peptides with an ex-
numerous proteins of the CNS that are implicated in tended N-terminal domain. TPP1 appears to be in-
important processes such as neuronal transmission, volved in the initial lysosomal degradation of subunit
signaling pathways, and protein transport. Potential c of ATP synthase, a very hydrophobic protein that
impairment of these processes by nonfunctional accumulates in LINCL. The decreased TPP1 activity
PPT1 could induce a deleterious effect on the func- may be directly responsible for the accumulation of
tioning of neurons and neuronal death. PPT1 has subunit c of mitochondrial ATP synthase and un-
been shown to depalmitate several neurospecific pep- digested small peptides. Whether and how subunit c
tides in vitro, supporting a specific role of PPT1 in the accumulation is associated with progressive neuronal
CNS. PPT1 may also have a role in the protection of degeneration and death remains unknown. Storage of
neurons against apoptosis. Recent evidence indicates lesser amounts of subunit c is observed in other
that in neurons PPT1 is localized in synaptosomes forms of NCL, mucopolysaccharidosis types I, II, and
and synaptic vesicles. The precise mechanisms lead- III, mucolipidosis type I, GM1 and GM2 gangliosi-
ing to lysosomal storage of mainly saposins (sphin- doses, and Niemann–Pick disease types A and C; the
golipid activator proteins, SAPs) A and D remain elu- cause of this is unknown. So, the significance of the
sive. SAPs A, B, C, and D are small glycoproteins accumulation of subunit c in the pathophysiology of
derived from a precursor protein, prosaposin. SAPs NCL is unclear.
are lysosomal proteins that facilitate the actions of CLN3, the gene for JNCL, encodes a putative inte-
several lysosomal hydrolases engaged in the degrada- gral membrane protein. The subcellular localization
tion of sphingolipids. There is no evidence that SAPs of the protein remains a subject of controversy. Avail-
are palmitoylated and could serve as substrates for able evidence indicates that CLN3 protein is traf-
PPT1.Accumulation of SAPs is not restricted to INCL. ficked through the endoplasmic reticulum and Golgi
SAPs are also found in variable amounts among to the lysosome, where it is localized in the lysosomal
subunit c of ATP synthase in other NCL variants and membrane. It may also be present at the plasma
they are found in GM1 gangliosidosis, GM2 gangliosi- membrane. Subunit c of mitochondrial ATP synthase
dosis, type A Niemann–Pick disease, metachromatic constitutes part of the protein content of the storage
leukodystrophy, and globoid cell leukodystrophy. The material. Other identified constituents of the stored
highest levels of SAPs are found in lysosomal storage material include phosphorylated dolichols, small
disorders in which the stored lipids bind to SAPs. amounts of SAPs, amyloid-b peptide, and mannose-
Although the lysosomal accumulation of SAPs may 6-phosphate glycoproteins. Newly synthesized lyso-
represent an epiphenomenon of no or minor signifi- somal enzymes are post-translationally modified by
cance, it is possible that the dysregulation of SAP me- the carbohydrate residue mannose 6-phosphate. This
tabolism plays a role in the pathogenesis of INCL. The residue serves as a recognition signal for mannose
severe paucity of myelin in INCL is striking and not 6-phosphate receptors of proteins that are destined
shared by the other NCL variants. The myelin defi- for lysosomes, where the recognition signal is re-
ciency is more severe than can be explained by the moved. In JNCL the levels of mannose 6-phosphate
neuronal loss. There is probably a component of glycoproteins are significantly elevated.
hypomyelination. Early onset neuronal/axonal dys- CLN5, the gene for FvLINCL, encodes a protein
function may interfere with the complex interaction which was formerly predicted to be a membrane pro-
of oligodendrocytes, neurons/axons, and astrocytes tein, but more recent evidence suggests it is a soluble
that is necessary for myelin to be produced and de- lysosomal protein. There is evidence for interactions
posited. A similarly severe myelin deficiency is seen of the CLN5 protein with the CLN2 and CLN3 pro-
in other neuronal storage disorders with infantile teins. In contrast, there is no evidence for interaction
onset, including infantile-onset GM1 and GM2 gan- between the CLN1 protein and the CLN2, CLN3, and
gliosidoses. CLN5 proteins. This is in line with the fact that the
Tripeptidyl peptidase 1 (TPP1), also called pep- storage material in INCL is different from the materi-
statin-insensitive proteinase, is a lysosomal enzyme al in other NCL variants. The lysosomal storage mate-
with aminopeptidase activity, removing tripeptides rial consists mainly of subunit c of mitochondrial
from the N-terminus of proteins. Efficient protein ATP synthase and small amounts of SAPs A and D.
degradation in lysosomes depends on concerted ac- CLN6, associated with vLINCL, codes for a trans-
tion of endopeptidases and exopeptidases, which pro- membrane protein localized to the endoplasmic
vide dipeptides and free amino acids to be exported reticulum. Its function is unknown, but there is evi-
to the cytoplasm and further used according to the
15.6 Magnetic Resonance Imaging 143

dence that defects in the protein lead to lysosomal fect. Nasogastric tube or, preferably, gastrostomy tube
dysfunction. feeding is important in the later stages of the disease
CLN8, associated with Northern epilepsy, also and has prolonged and improved the quality of life.
encodes a putative membrane protein. The CLN8 pro- Hematopoietic stem cell transplantation has been
tein is nonlysosomal. Most CLN8 protein is localized unsuccessful in halting the progression of the disease.
in the endoplasmic reticulum with a proportion in
the endoplasmic reticulum–Golgi intermediate com-
partment. It is involved in the early secretory path- 15.6 Magnetic Resonance Imaging
way. It is proposed that the CLN8 protein may play a
role in the transport of proteins across the endoplas- In INCL, CT scan of the brain shows severe atrophy,
mic reticulum membrane. The storage material con- more severe in the cerebral hemispheres than in the
tains subunit c of mitochondrial ATP synthase, SAPs, cerebellum. Some atrophy of the brain stem can also
and amyloid-b peptide. be seen. The ventricles and subarachnoid spaces are
Many different mutations have been identified in enlarged. The entire white matter is hypodense and
the NCL-associated genes. The age at onset of the dis- reduced in volume. The cortex is abnormally thin,
ease is no longer the most important criterion for the whereas the volume of the basal ganglia is not re-
subclassification of patients. Mutations in CLN1 have duced, except in the oldest patients. The calvarial
been found to be associated with infantile, late-infan- bone is thickened in patients over 2 years of age. MRI
tile, juvenile, and, rarely, also adult onset of the dis- is already abnormal in an early stage of disease in
ease. Mutations in CLN2 may give rise to late-infantile INCL, before the appearance of clinical symptoms.
but also to juvenile disease onset. Late-infantile onset With increasing age there is increasing atrophy. The
of the disease has been found in mutations of CLN1, white matter has a high signal intensity on T2-weight-
CLN2, CLN5, and CLN6, and will be for CLN7. There ed images in all stages of the disease (Fig. 15.1), but
is evidence for a genotype–phenotype correlation. more seriously in the later stages (Fig. 15.2). The sig-
Mutations that abolish all function of the encoded nal intensity is highest in the periventricular area.
protein may lead to a more severe phenotype than The corpus callosum and internal capsule are myeli-
mutations with some residual function of the protein. nated normally. With increasing age the loss in white
Mutations leading to intracellular misrouting of the matter volume becomes extreme (Fig. 15.2). The ap-
encoded protein may be associated with a more pearance of the white matter abnormalities suggests
severe phenotype. a combination of seriously delayed and disturbed
myelination, increasingly severe gliosis, and some
myelin loss. In the end only a thin cerebral mantle is
15.5 Therapy left with highly abnormal signal intensity of the white
matter throughout, compatible with complete ab-
Treatment continues to be supportive only. Anticon- sence of myelin and severe gliosis (Fig. 15.2). The cor-
vulsants are important and need to be tailored to the tex becomes increasingly thin. The basal ganglia and,
patient, seizure type, and side effects. Tegretol and in particular, the thalamus have a low signal intensity
lamotrigine may be helpful in the setting of behav- on T2-weighted images from early on (Figs. 15.1 and
ioral disturbances. In the management of INCL, irri- 15.2). The pattern of a strikingly low signal of the
tability, sleeping disorders, choreoathetosis, and late thalamus within cerebral white matter with a high
pains have responded best to baclofen, tizanidine, signal intensity is quite characteristic of INCL. Initial-
levomepromazine, and benzodiazepines. Only a few ly, the volume of the thalamus and basal ganglia is
children with LINCL show significant irritability and relatively normal, but subsequently the central nuclei
sleeping disturbances. Behavioral problems, depres- also become severely atrophic.
sion, and psychosis are often present in JNCL. Mean- In the NCL variants of later onset, CT shows pro-
ingful hobbies and individual, not too demanding gressive atrophy involving cerebral hemispheres and
teaching are of basic importance in the management cerebellum. The atrophy involves both gray and white
of these problems. Medication is necessary periodi- matter (Figs. 15.3 and 15.4). In LINCL cerebellar
cally.Valproate and benzodiazepines can be of help in atrophy is more severe than cerebral atrophy. MRI in
coping with the sleeping problems. Severe behavioral LINCL, FvINCL, vLINCL, and TvLINCL shows a rim of
problems and psychotic symptoms can be treated mildly increased signal intensity around the lateral
with haloperidol, levomepromazine, and benzodi- ventricles and in the posterior limb of the internal
azepines. In the bedridden stage, motor restlessness capsule (Fig. 15.3). Additionally, a decrease in signal
or panic attacks can be alleviated with baclofen and intensity is seen in the thalami and, to a lesser degree,
tizanidine. Antiepileptics, neuroleptics, antidepres- sometimes the basal ganglia on T2-weighted images.
sants, and anxiolytics have been used to try to sup- In JNCL abnormalities appear later and initial MRI
press involuntary movements, but with no clear ef- may be normal. Apart from progressive atrophy, most
144 Chapter 15 Neuronal Ceroid Lipofuscinoses

Fig. 15.1. MRI in a 21-month-old girl with INCL shows cerebral myelination. The corpus callosum has a low signal intensity,
atrophy with enlarged ventricles and subarachnoid spaces. consistent with a higher myelin content. The thalami have a
The white matter has a higher signal intensity than gray low signal intensity
matter in most areas, consistent with disturbed and delayed


severe in the cerebellum, the rim of mildly increased
signal intensity in the periventricular white matter Fig. 15.3. MRI in a 4-year-old Turkish boy with TvLINCL reveals
and posterior limb of the internal capsule may also be a rim of slightly increased signal intensity around the lateral
seen in JNCL. In ANCL, CT and MRI show atrophy. ventricles.The cerebellum is slightly atrophic
White matter abnormalities have not been reported.
In early EPMR MRI is normal, but it shows atrophy in
later stages of the disease.
15.6 Magnetic Resonance Imaging 145

Fig. 15.2. A 6-year-old child with


INCL. Note the severe generalized
atrophy, and the low signal intensity of
the thalami and basal ganglia on these
mildly T2-weighted images.
The white matter has a high signal
intensity throughout, consistent with
absence of myelin and presence of
gliosis. Courtesy of Drs. P. Santavuori
and S.L. Vanhanen, Helsinki, Finland
146 Chapter 15 Neuronal Ceroid Lipofuscinoses

Fig. 15.4. MRI in the same boy with TvLINCL, now 8 years old. matter has a slightly elevated signal intensity, as seen in neu-
Note the serious generalized atrophy involving cerebral hemi- ronal degenerative disorders
spheres, basal ganglia, and cerebellum. The cerebral white
Chapter 16

Adult Polyglucosan Body Disease

16.1 Clinical Features On microscopy, the most striking finding is the


and Laboratory Investigations presence of many polyglucosan bodies throughout
the nervous system. Most of them appear spherical,
Adult polyglucosan body disease (APBD) is a rare but there are also elongated thread-like forms, espe-
autosomal recessive neurological disorder with an cially in the gray matter. They vary in size from 1 to
onset in the fifth to seventh decade of life. The clinical 25 mm, even up to 50 mm; the usual size is around
features consist of pyramidal tract signs, peripheral 10 mm. The bodies are variably positive on staining
neuropathy with motor deficits, and usually pro- with PAS, silver proteinate (Bodian, Bielschowsky),
nounced distal sensory loss predominantly involving iodine stains, and alcian blue, but negative with Sudan
the lower limbs, hyper- or hypoactive reflexes, and black, luxol fast blue, and Congo red. With hema-
urinary incontinence due to a neurogenic bladder. toxylin–eosin they show varying degrees of basophil-
Most patients develop dementia, especially in the lat- ia. Those in the white matter are usually more strong-
er stages of the disease. Cerebellar ataxia, extrapyra- ly basophilic than those in the cortex. Their location
midal movement abnormalities, and seizures may is intra-axonal and within astrocytic processes. Elec-
occur. Some patients have predominantly signs of up- tron microscopy shows that polyglucosan bodies con-
per and lower motor dysfunction, prominent wasting sist of non-membrane-bound, about 8-nm-wide
of muscles, and fasciculations, suggesting a diagnosis branching filaments, associated to some extent with
of amyotrophic lateral sclerosis. The clinical course is haphazardly distributed granular and amorphous,
progressive. The interval between onset of symptoms densely osmiophilic condensations. These are some-
and death ranges from 3 to 21 years. times clumped together into much larger dense gran-
CSF protein levels are usually elevated. Neurophys- ular structures, which tend to aggregate in the center
iological studies show signs of an axonal sensorimo- of the bodies to form round, target-like dense areas.
tor peripheral neuropathy with normal to moderate- Many of the polyglucosan bodies can be identified as
ly slowed nerve conduction and signs of denervation being located within axons. The surrounding myelin
in electromyography. SSEP studies show prolonged sheaths are usually thinner than would be expected
central conduction times. In the context of the above for the caliber of the axon. Polyglucosan bodies are
clinical symptoms, the demonstration of polyglu- not seen in neuronal cell bodies.
cosan bodies in sural nerve biopsy or axillary skin Within the cerebral hemispheric white matter
biopsy confirms the diagnosis, especially if they are there are extensive, ill-defined areas of myelin pallor,
numerous. In some patients, especially patients of which involve the cerebral and cerebellar white mat-
Ashkenazi Jewish ethnic background, deficiency of ter bilaterally and symmetrically. Myelin loss is in-
glycogen branching enzyme has been demonstrated complete and more marked around the ventricles
in leukocytes and peripheral nerve biopsies. Howev- than in the subcortical white matter. In the affected
er, not all patients are deficient in this enzyme despite areas, axons are relatively spared. In addition, small
the otherwise indistinguishable clinical and histo- necrotic and even cystic lesions are frequently pre-
pathological findings. Prenatal diagnosis is possible sent in the subcortical white matter, globus pallidus,
in families in which deficiency of glycogen branching putamen, dentate nucleus, and the pons.
enzyme or the underlying genetic defect has been Examination of the sural nerve reveals the pres-
demonstrated. ence of numerous intra-axonal polyglucosan bodies
in both unmyelinated and myelinated axons. The
overlying myelin sheaths in myelinated axons are
16.2 Pathology thin. The nerve may otherwise be normal or show
moderate diffuse loss of both small and large myeli-
At autopsy mild external atrophy with prominence of nated axons. Onion bulbs may be found, suggesting
sulci and widening of the lateral ventricles may be demyelination and remyelination.
present. The spinal cord is atrophic. On sectioning, Polyglucosan bodies may also be found in other
poorly demarcated areas of discoloration are seen organs, such as the heart, liver, kidneys, lungs, skele-
within the white matter of the brain. tal, and smooth muscles. The presence of polyglu-
148 Chapter 16 Adult Polyglucosan Body Disease

cosan bodies in myoepithelial cells of apocrine glands Glycogen branching enzyme activity can be either
facilitates diagnosis by axillary skin biopsy. deficient or normal in patients with APBD. Most pa-
tients of Ashkenazi Jewish ethnic background have a
deficiency of this enzyme, but so do some patients of
16.3 Chemical Pathology other ethnic background. It is likely that APBD is ge-
netically heterogeneous and that the disease may
Polyglucosan bodies are composed principally of glu- have more than one biochemical etiology resulting in
cose polymers, with a small but variable component similar phenotypes.
of phosphate and sulfate groups and less than 5% Polyglucosan bodies are composed principally of
protein. glucose polymers and occur in various conditions.
The polyglucosan bodies of APBD greatly resemble
and may be identical to corpora amylacea, which ac-
16.4 Pathogenetic Considerations cumulate in the CNS with normal aging, Lafora bod-
ies present in Lafora body disease, and Bielschowsky
Type IV glycogen storage disease is a rare autoso- bodies. Corpora amylacea are seen in a characteristic
mal recessive disorder representing 0.3% of all cases topography in the healthy nervous system with aging.
of glycogenosis of any type. The disease is caused by They develop in astrocytic processes and are mainly
a deficiency of the glycogen branching enzyme. distributed subpially, subependymally, and perivas-
Glycogen branching enzyme is encoded by the gene cularly, although intra-axonal corpora amylacea may
GBE, which is located on chromosome 3p14. There is also occur. Lafora bodies are found in association
evidence for the existence of different isoenzymes, with Lafora body disease, a progressive myoclonus
probably generated by alternative splicing. The en- epilepsy. Lafora bodies appear in neuronal perikarya
zyme is involved in glycogen synthesis. Deficiency and processes, especially in the cerebral cortex, thala-
leads to accumulation of polysaccharides, which are mus, globus pallidus, substantia nigra, and dentate
resistant to amylase digestion and have longer outer nucleus. Bielschowsky bodies are found in the peri-
chains and shorter branch points than normal glyco- karya and processes of neurons. They differ from
gen. Lafora bodies in their much more limited distribu-
Glycogen storage disease type IV is highly hetero- tion and pleiomorphic appearance. They are usually
geneous as to clinical symptoms and age of onset. The only found in the external segment of the globus pal-
latter varies between the early neonatal period lidus. They may represent a variant of Lafora body
through childhood to late adulthood. The classical disease. Besides these specific conditions, polyglu-
type is characterized by early onset and rapidly pro- cosan bodies may be seen as a nonspecific phenome-
gressive hepatosplenomegaly, progressive cirrhosis non within the CNS in olivopontocerebellar atrophy
with portal hypertension, ascites, esophageal varices, and amyotrophic lateral sclerosis, and in peripheral
and death between 3 and 5 years due to liver failure, nerves in exceptional cases of diabetes mellitus, mo-
unless liver transplantation is performed. Nonpro- tor neuropathy, familial spastic paraparesis, and spin-
gressive hepatic variants have also been described. ocerebellar syndromes, as well as in normal aging.
Patients with neuromuscular involvement have a my- It is unclear how the polyglucosan bodies arise in
opathy with or without cardiomyopathy, neuropathy, APBD. They may represent systemic glycogen storage
and liver cirrhosis. The age at onset varies from due to deficiency of glycogen branching enzyme or
neonatal to adulthood. Neonatal variants presenting another enzyme related to glycogen metabolism. The
with pronounced hypotonia are extremely rare. They relationship with the CNS structural damage and dys-
resemble patients with the Werdnig–Hoffmann type function is unclear. It is conceivable that there is an
of spinal muscular atrophy. They may have addition- underlying metabolic defect, which damages neurons
al cardiomyopathy or hepatosplenomegaly. Presenta- by itself and as an independent side effect leads to the
tion with hydrops fetalis is also very rare. APBD char- production of polyglucosan bodies. The growth of
acterized by late onset and slowly progressive severe polyglucosan bodies within axons could also become
neurological symptoms can also be an expression of deleterious for those axons, at the very least impeding
glycogen branching enzyme deficiency. or blocking axonal flow.
The phenotypic variability and tissue specificity
described above may be due to particular mutations
resulting in selective tissue-based expression or defi- 16.5 Therapy
ciency of isoenzymes. This hypothesis does not, how-
ever, explain the differences in phenotypic expression There is at present no effective causal treatment for
between patients with identical genotype. APBD.
16.6 Magnetic Resonance Imaging 149

16.6 Magnetic Resonance Imaging callosum (Fig. 16.1). The signal change is often most
pronounced within the periventricular white matter,
MRI in patients with APBD shows extensive, often ill- gradually becoming more normal in the deep white
defined abnormalities within the cerebral and cere- matter, and merging with normal white matter in the
bellar white matter (Figs. 16.1 and 16.2). The abnor- U fibers. In other patients, however, the white matter
malities are most prominent in the periventricular abnormalities are better demarcated (Figs. 16.1 and
region and tend to spare the U fibers and the corpus 16.2). In later stages, the corpus callosum and U fibers

Fig. 16.1. FLAIR images in a 52-year-old female patient with U-fibers, where only small foci of abnormal signal are seen.
APBD. Polyglucosan bodies were found in the sural nerve Note the cerebellar and brain stem involvement. Courtesy of
biopsy. There are confluent periventricular white matter ab- Dr. B. Barcelos da Nóbrega, Department of Radiology, School of
normalities with relative sparing of the deep white matter and Medicine, Federal University of Goias, Brazil
150 Chapter 16 Adult Polyglucosan Body Disease

Fig. 16.2. FLAIR images in the 49-year-old sister of the patient temporal white matter is prominently involved. Note the cere-
in Fig. 16.1. Her disease is radiologically more advanced. There bellar and brain stem involvement. Courtesy of Dr. B. Barcelos
are confluent signal abnormalities in the periventricular white da Nóbrega, Department of Radiology, School of Medicine,
matter with some additional focal spots. The U fibers are rela- Federal University of Goias, Brazil
tively preserved. The corpus callosum has become thin. The

become involved as well (Fig. 16.2). The cerebral respect to both the white matter abnormalities and
white matter may be decreased in volume with the atrophy. Within the brain stem signal abnormali-
widening of the lateral ventricles and subarachnoid ties may be seen in the long tracts (Figs. 16.1 and
spaces and thinning of the corpus callosum. The 16.2). The cerebellum, brain stem, and spinal cord
frontal lobe tends to be involved more seriously, with may be atrophic.
Chapter 17

Peroxisomes and Peroxisomal Disorders


R.J.A. Wanders

17.1 Peroxisomal Biogenesis ing consequences of a defect in peroxisome biogene-


and Biochemical Functions sis as in Zellweger syndrome. In the last few years
much has been learned about the biogenesis and
Peroxisomes belong to the family of intracellular or- metabolic functions of peroxisomes. With respect to
ganelles, are present in virtually all eukaryotic cells peroxisome biogenesis, it is now clear that peroxiso-
with the sole exception of erythrocytes, and contain a mal proteins are synthesized on free polyribosomes
fine granular matrix surrounded by a single mem- and are targeted to pre-existing peroxisomes by
brane. Identification of peroxisomes in cells is greatly virtue of specific targeting signals contained within
helped by the fact that peroxisomes stain dark when the amino acid sequences of the different peroxiso-
incubated in a medium containing diaminobenzidine mal proteins. Peroxisomal matrix proteins contain
(DAB) and hydrogen peroxide (H2O2) at alkaline pH one of two different peroxisome targeting signals
thanks to the present of catalase in peroxisomes. The (PTS), the first of which, PTS1, is used by most of the
availability of specific antibodies raised against per- peroxisomal matrix proteins. The PTS1 involves a
oxisomal matrix as well as membrane proteins, some C-terminal tripeptide of the sequence serine–lysine–
of which are commercially available, has allowed leucine (SKL) or a variant thereof. The second per-
unequivocal identification of peroxisomes using oxisomal targeting signal, PTS2, has been found in far
immunohistochemical, immunofluorescence, and fewer peroxisomal proteins and involves a stretch of
immuno-electron microscopic methods. These meth- nine amino acids, of which amino acids 1, 2, 8, and 9
ods have been used to study peroxisomes in different are essential. The following consensus sequence has
cells and have shown that the morphology of peroxi- been found for the PTS2 motif (R/K)–(L/V/I)–
somes varies among different cell types, ranging from XXXXX–(H/Q)–(L/A), in which X may be any amino
round to oval structures to elaborate tubular or retic- acid. In human beings the PTS2 has only been identi-
ular structures. Furthermore, the size and abundance fied in three peroxisomal proteins. Peroxisomal
of peroxisomes also vary widely, ranging from very membrane proteins lack both a PTS1 and a PTS2 sig-
small so-called microperoxisomes in fibroblasts nal, implying the existence of additional signals,
(<0.1 mm) to large peroxisomes in hepatocytes which remain ill defined. The identification of the
(>1.0 mm). A striking feature of peroxisomes, at least PTS1 and PTS2 targeting signals was soon followed
in rodents, is their inducibility by a variety of dietary, by the discovery of soluble receptors, called Pex5p
hormonal, and other physiological effectors includ- and Pex7p, which recognize the PTS1 and PTS2 sig-
ing fibrates such as clofibrate, bezafibrate, and phe- nals respectively. These two receptors pick up PTS1 or
nofibrate. Fibrates exert their effect on peroxisomes PTS2 proteins in the cytosol and direct these proteins
via activation of one of the peroxisome proliferator to the peroxisomal membrane, where a complex net-
activating receptors (PPARs), PPAR-a, which forms a work of proteins takes care of the transport of the
heterodimer with the retinoic acid receptor (RXR) PTS1 and PTS2 proteins across the peroxisomal
and activates the transcription of a range of different membrane to end up in the peroxisome interior. On
genes containing a peroxisome proliferator response the basis of studies in yeasts, many of the proteins in-
element (PPRE) in their promoter regions. Several volved in correct transmembrane transport of PTS1
genes coding for different peroxisomal enzyme activ- and PTS2 proteins have been identified. Proteins that
ities contain such a PPRE, which explains their are essential for peroxisome biogenesis have been
inducibility by fibrates and other PPAR-a ligands. termed peroxins and are written as “Pex” followed by
Although fibrates are effective hypolipidemic agents a number and a small “p,” e.g., Pex5p, Pex7p. The cor-
in humans as well, their effect on peroxisomes in responding genes are called PEX genes, so that PEX5
humans in terms of peroxisome induction and upreg- codes for Pex5p, PEX7 for Pex7p, and so on. So far, 32
ulation of peroxisomal enzyme activities remains to different peroxins have been identified in different
be determined. species. Since peroxisome biogenesis is so markedly
Although long regarded as relatively insignificant conserved among different species ranging from
organelles with only a minor role in cellular metabo- yeast to plants and humans, most of the peroxins have
lism, it is now clear that peroxisomes are essential homologues in all the different species. So far 14
subcellular organelles, as exemplified by the devastat- different PEX genes have been identified in humans.
152 Chapter 17 Peroxisomes and Peroxisomal Disorders

As anticipated, mutations in any of these PEX genes and di- and trihydroxycoprostanoic acid, two bifunc-
will give rise to a defect in peroxisome biogenesis and tional proteins, and two peroxisomal thiolases. With
underlie the disorders of peroxisome biogenesis. respect to the bifunctional proteins, current knowl-
Originally, peroxisomes were thought to play a role edge holds that the second, more recently identified
in hydrogen peroxide (H2O2) metabolism only. Per- bifunctional protein, named D-bifunctional protein,
oxisomes contain a variety of H2O2 -generating oxi- is the single enzyme involved in the oxidation of C26:0,
dases and catalase, which cleaves H2O2 to H2O and pristanic acid, as well as the two cholestanoic acids,
oxygen, thus constituting a simple respiratory path- whereas the two thiolases both play a part in the oxi-
way. In addition, peroxisomes carry out a series of dation of these compounds. Apart from the differ-
other metabolic functions, the most important of ences in terms of enzymes involved in b-oxidation in
which are fatty acid b-oxidation, ether phospholipid mitochondria and peroxisomes, there are a number
biosynthesis, fatty acid a-oxidation, glyoxylate de- of other differences including the transport of fatty
toxification, and L-pipecolic acid oxidation. There is acids across the peroxisomal versus mitochondrial
much confusion about the role of peroxisomes in iso- membrane. Mitochondrial entrance of long-chain
prenoid biosynthesis including cholesterol forma- fatty acids is mediated by a carnitine cycle involving
tion, for which reason we have decided to leave this carnitine palmitoyl transferase 1 (CPT1), mitochon-
potential function of peroxisomes out. drial carnitine/acylcarnitine transporter (CACT),
Fatty acid b-oxidation in peroxisomes involves a and carnitine palmitoyl transferase 2 (CPT2), where-
sequence of four reactions just as in mitochondria, in as transport of fatty acids across the peroxisomal
which an acyl-CoA ester undergoes dehydrogenation, membrane is probably mediated by membrane pro-
hydration, dehydrogenation again, and thiolytic teins belonging to the family of ABC transporters, of
cleavage. Apart from this similarity, peroxisomal and which four have been identified in peroxisomal mem-
mitochondrial b-oxidation differ in many respects, branes. One of these four is called adrenoleukodys-
including the oxidation of different substrates in the trophy protein (ALDP), which is deficient in X-linked
two organelles. Mitochondria are able to oxidize adrenoleukodystrophy and probably catalyzes the
short-, medium-, and long-chain fatty acids and are transport of very-long-chain fatty acids in their
responsible for the oxidation of the bulk of dietary CoA-ester form. The end products of peroxisomal
fatty acids. Peroxisomes, on the other hand, are not so b-oxidation, including acetyl-CoA, propionyl-CoA,
important energetically, but are important for the and medium-chain acyl-CoAs, are shuttled to mito-
degradation of a specific subset of fatty acids includ- chondria in the form of carnitine esters for final oxi-
ing very-long-chain fatty acids, pristanic acid, and dation to CO2 and water inside the mitochondria. The
di- and trihydroxycholestanoic acid. The latter two peroxisomal b-oxidation system does not only act on
acids are formed in the liver from cholesterol and are saturated fatty acids, but it is also capable of cata-
the direct precursors of the primary bile acid cheno- lyzing chain shortening of monounsaturated and
deoxycholic acid and cholic acid, respectively. The polyunsaturated fatty acids. To this end, peroxisomes
difference between the functional roles of mitochon- are equipped with enzymes such as 2,4-dienoyl-CoA
drial and peroxisomal b-oxidation is exemplified by reductase and D3, D2-enoyl-CoA isomerase.
the different clinical signs and symptoms of patients The second important function of peroxisomes
affected by a defect in mitochondrial versus peroxiso- has to do with ether phospholipids. Ether phospho-
mal b-oxidation. Another major function of the per- lipids are a special class of phospholipids which differ
oxisomal b-oxidation system concerns the “bio- from the regular, better-known diacylphospholipids
synthesis” of polyunsaturated fatty acids including in one major respect, which is the occurrence of
docosahexaenoic acid (C22:6 w-3). Recent studies have an ether linkage rather than an ester linkage at the
clearly shown that the formation of C22:6 w-3 from sn-1 position of the glycerol backbone. Two groups
linolenic acid (C18:3 w-3) involves active participation of ether phospholipids can be distinguished with
of the peroxisomal b-oxidation system at the level of either an 1-O-alkyl or an 1-O-alk-1-enyl-linkage. The
the conversion of C24:6 w-3 to C22:6 w-3. latter phospholipids (1-O-alk-1-enyl-2-acyl phospho-
With respect to the enzymes involved in the b-oxi- glycerides) with an a-, b-unsaturated ether bond
dation of all these compounds, it was long thought are also known by their trivial name plasmalogens.
that a single set of enzymes, including acyl-CoA Platelet activating factor (PAF; 1-O-alkyl-2-acetyl-
oxidase, L-bifunctional protein with enoyl-CoA hy- glycerophosphocholine) is the best-known ether
dratase and 3-hydroxyacyl-CoA dehydrogenase ac- phospholipid. Plasmalogens constitute 5–20% of
tivities, and peroxisomal thiolase would catalyze the the phospholipids of cell membranes and are present
b-oxidation of all these compounds. It now turns out, at high concentrations in the brain, especially in
however, that peroxisomes harbor two acyl-CoA oxi- myelin. PAF induces both platelet and leukocyte
dases, one for straight-chain fatty acids like C26:0, and aggregation and degranulation and is important in
one for branched-chain fatty acids like pristanic acid several disease processes including inflammation and
17.2 Peroxisomal Disorders 153

anaphylaxis. Peroxisomes are involved in the intro- orders. Although different classifications have been
duction of ether bonds in ether phospholipids, since proposed through the years, most authors now use a
the first two enzymes involved in ether phospholipid classification in two distinct categories: the disorders
biosynthesis are localized in peroxisomes. These of peroxisome biogenesis, and the single peroxisomal
enzymes are dihydroxyacetonephosphate acyltrans- enzyme deficiencies.
ferase (DHAPAT) and alkyldihydroxyacetonephos- 1. Peroxisome biogenesis disorders
phate synthase (alkyl-DHAP synthase), whereas all (a) Cerebrohepatorenal syndrome
subsequent steps are catalyzed by enzymes in the (Zellweger syndrome)
endoplasmic reticulum. (b) Neonatal adrenoleukodystrophy
The third important function of peroxisomes is (c) Infantile Refsum disease
fatty acid a-oxidation. Fatty acids such as phytanic (d) Rhizomelic chondrodysplasia punctata type 1
acid (3,7,11,15-tetramethylhexadecanoic acid) with a 2. Single peroxisomal enzyme deficiencies
methyl group at the 3-position cannot undergo direct (a) X-linked adrenoleukodystrophy
b-oxidation. Instead, 3-methyl branched-chain fatty (b) Acyl-CoA oxidase deficiency
acids like phytanic acid first undergo a-oxidation in (c) D-Bifunctional protein deficiency
which the terminal carboxyl group is removed as CO2. (d) 2-Methylacyl-CoA racemase deficiency
The pathway of phytanic acid a-oxidation has long (e) Adult Refsum disease
remained mysterious but has been resolved in recent (f) Rhizomelic chondrodysplasia punctata type 2
years. It is now clear that phytanic acid is first con- (DHAPAT deficiency)
verted into its CoA-ester followed by a series of en- (g) Rhizomelic chondrodysplasia punctata type 3
zymes including phytanoyl-CoA hydroxylase, 2-hy- (alkyl-DHAP synthase deficiency)
droxyphytanoyl-CoA lyase, and pristanal dehydroge- (h) Acatalasemia
nase, which are all peroxisomal enzymes, to produce (i) Mulibrey nanism
CO2 and pristanic acid (2,4,6,10-tetramethylpentade-
canoic acid). Pristanic acid can then be activated via The first category contains peroxisomal disorders in
an intraperoxisomal acyl-CoA synthetase (VLCS) to which peroxisome biogenesis is deficient. In Zell-
produce pristanoyl-CoA followed by b-oxidation. weger syndrome (ZS), neonatal adrenoleukodystro-
Detoxification of glyoxylate via the peroxisomal phy (NALD), and infantile Refsum disease (IRD),
enzyme alanineglyoxylate aminotransferase (AGT) is together called Zellweger spectrum, both the PTS1
another major function of peroxisomes. Deficiency of and PTS2 pathways are affected, which is associated
AGT as in hyperoxaluria type 1 leads to the accumu- with the generalized loss of peroxisomal functions,
lation of glyoxylate, which is subsequently converted whereas in the other type of peroxisome biogenesis
into glycolate and oxalate, which precipitates as calci- disorder, with rhizomelic chondrodysplasia punctata
um oxalate in tissues. (RCDP) type 1 as sole representative, peroxisome bio-
A final important function of peroxisomes is the genesis is only partially defective. In RCDP type 1
degradation of L-pipecolic acid via a peroxisomal only the PTS2 pathway is blocked as a consequence of
oxidase called L-pipecolate oxidase. Pipecolic acid is a a nonfunctional PTS2-receptor (Pex7p) encoded by
degradation product of L-lysine, which is normally PEX7. In contrast to RCDP type 1, which is related to
degraded by the saccharopine pathway. However, a single gene defect, the Zellweger spectrum disor-
L-lysine may also be degraded via the L-pipecolic acid ders are genetically heterogeneous, with 11 different
pathway, which may be especially important in brain genes identified so far: PEX1, PEX2, PEX3, PEX5,
since the activity of the saccharopine pathway is low PEX6, PEX10, PEX12, PEX13, PEX16, PEX19, and
in brain. In the L-pipecolic acid pathway, L-pipecolic PEX26. In 65% of Zellweger spectrum patients, PEX1
acid is produced from L-lysine via two enzymatic is the gene involved, which is explained, at least in
steps, and is then oxidized by L-pipecolate oxidase, a part, by the occurrence of two frequent mutations in
peroxisomal enzyme, at least in humans. The struc- the PEX1 gene. As a consequence, PTS2 proteins like
ture and function of the L-pipecolic acid pathway peroxisomal thiolase, alkyldihydroxyacetonephos-
remain incompletely understood. phate synthase, and phytanoyl-CoA hydroxylase are
not properly targeted to peroxisomes, which explains
a selective loss of peroxisomal functions (plasmalo-
17.2 Peroxisomal Disorders gen biosynthesis and phytanic acid a-oxidation).
In the second category, peroxisome biogenesis is
Disturbance of normal peroxisomal function is asso- intact, but the basic problem is at the level of one of
ciated with far-reaching and devastating conse- the enzymes or transporters involved in each of the
quences. Zellweger syndrome is generally considered different metabolic pathways contained in peroxi-
to be the prototype of the group of peroxisomal dis- somes.
Chapter 18

Peroxisome Biogenesis Defects

18.1 Clinical Features ZS. Widely patent fontanels are uncommon. The af-
and Laboratory Investigations fected children show feeding problems, failure to
thrive, hepatomegaly, and sometimes jaundice. Fur-
Peroxisome biogenesis disorders are genetically het- thermore, the disease is characterized by seizures,
erogeneous diseases with an autosomal recessive sensorineural hearing loss, decreased vision with
mode of inheritance. They include Zellweger syn- nystagmus, optic atrophy or dysplasia, and pigmen-
drome (ZS, also called cerebrohepatorenal syn- tary retinal degeneration. The posterior eye segment
drome), neonatal adrenoleukodystrophy (NALD), abnormalities are identical to those of ZS, but anteri-
and infantile Refsum disease (IRD). The clinical pic- or segment abnormalities are lacking. Macrocephaly
tures of these disorders show similarities, but an im- may be present. Within the first year of life severe de-
portant difference is a difference in severity, the clin- velopmental retardation becomes apparent, although
ical course being most severe in ZS and mildest in most infants reach some milestones before neurolo-
IRD. Exceptional patients present with a still milder gical deterioration occurs. The age at which regres-
phenotype. sion begins varies from 12 months to more than
After birth children with ZS show profound mus- 7 years. Progressive neurological dysfunction is char-
cular hypotonia. Most patients lie motionless with acterized by cerebellar ataxia, spasticity of the arms
weak or absent Moro reflex, tendon reflexes, and and legs with truncal hypotonia, increased deep ten-
sucking and swallowing reflexes. Tube feeding is nec- don reflexes, extensor plantar reflexes, and sensory
essary. Typically, the children have craniofacial dys- defects. If not present from birth onwards, seizures
morphism with a high and bulging forehead, flat usually occur in this period. Visual dysfunction pro-
occiput, upslanting palpebral fissures, puffy eyelids, gresses to blindness. Adrenal insufficiency is rarely
hypoplastic supraorbital ridges, and a low and broad clinically manifest. Exceptional patients are initially
nasal bridge with hypertelorism and epicanthus entirely normal or close to normal with onset of rapid
folds, giving them a mongoloid appearance. In addi- neurological deterioration in the second year of life.
tion, Brushfield spots, peripheral pigmentary The course of disease is more protracted in NALD
retinopathy, optic nerve dysplasia or hypoplasia, than in ZS, death occurring between the ages of 1.5
glaucoma, corneal clouding, cataracts, low-set mal- and more than 10 years.
formed ears, high arched palate, micrognathia, and IRD is the mildest variant of the three mentioned
widely patent sutures and fontanels are present. disorders. The disease is usually not manifest at birth
Macrocephaly may be present. Some children have a but presents itself within the first 6 months of life
cleft soft palate. The children have a severe visual with psychomotor retardation, minor facial dysmor-
and hearing deficit. Nystagmus is often present. He- phism, mild hypotonia, sensorineural deafness,
patomegaly, prolonged neonatal or later-onset and visual impairment with retinal pigmentary de-
icterus, and hemorrhages due to hypoprothrombine- generation, optic dysplasia, and nystagmus. Hepato-
mia are common. Limb anomalies include cubitus megaly and failure to thrive with growth retardation
valgus, camptodactyly, single transverse palmar are common. Seizures occur but epilepsy is not as
creases, and talipes equinovarus. Failure to thrive and severe as in ZS. Many patients are able to sit and walk
severe psychomotor retardation are conspicuous. independently after several years, whereas others
Seizures are frequent. Cardiac defects are not fre- never acquire these abilities. The gait is usually ataxic
quent, but ventricular septum defect, patent ductus and broad-based, and cognitive function in the se-
arteriosus, and patent foramen ovale may occur. verely retarded range. Life expectancy is considerably
Cryptorchidism is frequently observed in boys, cli- longer than in ZS and NALD, up to more than 2
toromegaly and labial hypoplasia in girls. About 90% decades.
of the patients die within the first year of life, death Milder variants of peroxisome biogenesis defects
occurring in the majority within the first few months. with later onset and more protracted disease course
Most children with NALD have neurological ab- have been described. One of the mildest variants
normalities at birth, but some are initially near-nor- reported concerns a family in whom adults with a
mal. Hypotonia is moderate to severe, reflexes are hy- normal or borderline intelligence only demonstrate
poactive. Craniofacial dysmorphism is milder than in sensorineural hearing loss and retinitis pigmentosa,
18.2 Pathology 155

leading to a diagnosis of Usher syndrome (Raas- ently normal adrenal function is not incompatible
Rothschild et al. 2002). with the diagnosis. In IRD adrenal function is nor-
In ZS, laboratory investigations may reveal many mal.
different, in themselves nonspecific biochemical ab- X-ray examinations in ZS often reveal calcific stip-
normalities, such as hyperbilirubinemia, elevated liv- pling of bony epiphyses. Stippled, irregular calcifica-
er enzymes, hypoprothrombinemia, reduced albumin tion of particularly the patellae, greater trochanters,
level, hypocarnitinemia, hypocholesterolemia, gener- triradiate cartilages, acetabulum, scapula, and ster-
alized amino aciduria, and elevated CSF protein. num is seen in 50–70% of the ZS patients. Ultrasound
These abnormalities are not necessarily all present. may detect multiple small renal cortical cysts, but
Elevated serum iron, iron saturation, and transferrin they are often difficult to find. In NALD and IRD no
may be found, but these findings are inconsistent and calcific stippling of bony epiphyses is present. No
transient. As a rule, an abnormally low cortisol re- renal cysts are found. ERG is extinguished at a very
sponse to ACTH stimulation is found despite normal early age in ZS and the EEG is highly abnormal with
basal cortisol level. More specific abnormalities are epileptic discharges. BAEP shows reduced responses.
directly related to generalized deficiency of peroxiso- In NALD and IRD the ERG becomes extinguished and
mal function. Plasma levels of very long-chain fatty BAEP is abnormal. Nerve conduction velocity may be
acids are increased with an elevation of the C26:C22 decreased in NALD; it is normal in IRD.
and C24:C22 fatty acid ratio. Saturated as well as mo- Prenatal diagnosis can be performed with the help
nounsaturated and polyunsaturated very-long-chain of various biochemical investigations in cultured
fatty acids are increased. Plasma pipecolic acid and chorionic villus cells and amniocytes. DNA tech-
phytanic acid may be normal initially but increase niques can be applied in families with known muta-
with age. Plasma dicarboxylic acids are raised.Abnor- tions.
mal bile acids such as dihydroxycholestanoic acid and
trihydroxycholestanoic acid are elevated. In urine
elevated levels of dicarboxylic acids, dihydroxyc- 18.2 Pathology
holestanoic acid, and trihydroxycholestanoic acid are
present. In platelets and red blood cells a deficiency of Brain weight in ZS is normal or may exceed normal.
the peroxisomal enzyme dihydroxyacetone phos- External examination reveals abnormalities in the
phate acyltransferase can be shown. The plasmalogen cerebral convolutional pattern with areas of pachy-
content of red blood cells is decreased in the first few gyria and polymicrogyria. Polymicrogyria is typical-
months of life. The plasmalogen level of the red blood ly present in the opercular regions of the frontal, pari-
cells increases with age and may be normal in pa- etal, and temporal lobes and within the insular re-
tients who are 4 months or older. The synthesis of gion, with an increased number of gyri and decreased
platelet activating factor by leukocytes is deficient. amplitude of the gyri. In the superior region, over the
The activity of dihydroxyacetone phosphate acyl- frontoparietal convexities, the polymicrogyric cortex
transferase can be shown to be deficient in leukocytes merges with a pachygyric cortex, where convolutions
and thrombocytes. In cultured fibroblasts a de- are abnormally broad and reduced in number. There
creased content of plasmalogen and increased levels is a failure of full opercularization of the insula and
of very-long-chain fatty acids can be demonstrated. the sylvian fissure is abnormally vertical in orienta-
The b-oxidation of very-long-chain fatty acids, the de tion. Otherwise the gyral pattern of the cerebral
novo biosynthesis of plasmalogens, and the activity hemispheres is normal. Often the cerebellum is hy-
of dihydroxyacetone phosphate acyltransferase, alkyl poplastic and the cerebellar cortex has areas of
dihydroxyacetone phosphate synthase, and phytanic polymicrogyria. External appearance of the brain
acid oxidation can be shown to be deficient in fibro- stem is normal.
blasts. Finally, in fibroblasts catalase activity is not On sectioning the gyral abnormalities are con-
found in organelles, but in the cellular cytoplasm. firmed with moderate thickening of the cortical plate
Laboratory abnormalities are essentially the same in the areas of abnormal gyration. The lateral ventri-
in NALD and IRD, but milder than in ZS. In ZS the cles are mildly enlarged and have a mildly colpo-
accumulation of very-long-chain fatty acids includes cephalic configuration. There is a moderate symmet-
the saturated and monounsaturated C26 fatty acid and rical decrease in volume of the white matter. Olfacto-
is associated with a decrease in the C22 saturated fatty ry bulbs and tracts and optic nerves are thin, as is
acid concentration. In NALD the rise in very-long- the corpus callosum. Periventricular subependymal
chain fatty acids is not associated with an increase in cysts, so-called germinolytic cysts, are often present
monounsaturated C26 fatty acid and the C22 fatty acid over the heads of the caudate nucleus. In the brain
is on average higher than normal. In NALD, mild stem, hypoplasia and dysplasia of the inferior olives is
adrenal insufficiency and low cortisol response in apparent.
ACTH stimulation is usually found, although appar-
156 Chapter 18 Peroxisome Biogenesis Defects

Microscopic examination reveals that the cerebral lack the usual delicately convoluted pattern. The
cortex has a normal cytoarchitectonic pattern in the brain stem is poorly myelinated. A diffuse increase in
normally convoluted areas. The cytoarchitecture of the number of astrocytes is found in gray and white
polymicrogyric and pachygyric cortex is abnormal matter and lipid-laden foamy cells are present. The
and there are heterotopias in the subcortical white cerebellum is often hypoplastic and its convolutional
matter under the regions of cortical abnormality. The pattern abnormal. Areas of polymicrogyria are com-
migrational abnormality has principally affected mon and microscopic examination shows that corti-
neurons destined for the outer cortical layers. Many cal lamination is markedly abnormal at these points.
cells normally found in the outer cortical layers (lay- Many heterotopic Purkinje cells are present in the
ers II and III) are distributed in heterotopic positions subcortical white matter. A marked increase in astro-
within the deep cortical layers (layers V and VI) and cytes is present in the cerebellar cortex, and many
in the white matter below the cortex. However, some glial cells and macrophages have vacuolated foamy
of the neurons destined for the deep cortical layers cytoplasm filled with lipid droplets. The changes in
are in their normal laminar positions. The difference the cerebellar white matter are similar to those in the
between the pachygyric and polymicrogyric cortices white matter of the cerebral hemispheres. The dentate
is related to differences in intracortical cell patterns. nuclei may be hypoplastic and dysplastic, showing
The outer cortical layers are relatively more cellular in the same lack of convolutions as observed in the infe-
polymicrogyric cortex than in pachygyric cortex, rior olivary nuclei.
whereas the reverse is true with regard to the deeper Electron microscopy reveals that the lipid-laden
cortical layers. Within the cortex a marked astrocytic cells contain typical trilamellar inclusions along with
gliosis is present. Within the proliferated astrocytes, heterogeneous material. These inclusions are in-
an excess of sudanophilic lipid material is seen. PAS- tralysosomal. The trilamellar structures are com-
positive deposits are seen in the cortex of some of the posed of two parallel electron-dense lines separated
patients related to glycogen deposition in the cyto- by an electron-lucent zone. They are identical to those
plasm and nucleoplasm of neurons and astrocytes. encountered in other peroxisomopathies and may
Clusters of unusual histiocytes and multinucleated contain very-long-chain fatty acids. Structurally ab-
giant cells have been described with cytoplasm that is normal mitochondria have been reported in some
in part homogeneously PAS-positive and in part cases.
foamy and sudanophilic. Following birth hepatic cirrhosis may develop
The white matter is characterized by a profound rapidly, although not invariably. The liver is enlarged
deficiency of myelin and presence of gliosis. Myelin in most ZS patients. Histological findings vary from
deficiency is caused by delayed and disturbed myeli- near-normal to diffusely abnormal, dependent on the
nation; active loss of myelin cannot be demonstrated. age of the patient. In the first 2 months of life, micro-
Throughout the white matter many astrocytes are scopic abnormalities are absent or mild and include
present which are hypertrophic and contain sudano- fibrosis, cholestasis, and intrahepatic bile duct hy-
philic lipid material in their cytoplasm. Lipid deposits poplasia. In older patients fibrosis and distortion of
are also seen in histiocytes and macrophages. No liver architecture is more severe, ending in micro-
perivascular infiltration with inflammatory cells is nodular cirrhosis. Inconstant excess of hemosiderin
present. The white matter abnormality is usually dif- in hepatocytes and in particular in Kupffer cells and
fuse and generalized. In some cases the periventricu- macrophages may be related to age, with the greatest
lar white matter is most severely involved with no prominence between 5 and 18 weeks of age. In excep-
myelin left in that area. Oligodendrocytes are reduced tional cases, accumulation of glycogen is found. On
in number. There is a decrease of axons in the white ultrastructural examination, no peroxisomes are seen
matter corresponding to an area of pachygyria or in hepatocytes, where they are normally found in
polymicrogyria. The ventricles are partially denuded abundance. Invariably, abnormal mitochondrial mor-
of ependyma and regionally the ependyma is a pseu- phology is seen. Intralysosomal trilamellar inclusions
dostratified columnar epithelium resembling the epi- are present in Kupffer cells and macrophages. In the
thelium seen in midfetal life. In many patients germi- kidney multiple small cysts are present in the cortex,
nolytic cysts are present in subependymal areas. especially in the subcapsular region. The average di-
The basal ganglia have a normal configuration, but ameter is 3 mm, with some reaching 8 mm. They are
at microscopic examination changes similar in nature predominantly of tubular, occasionally of glomerular
to those in the cortex are seen, with an increase in origin. Ultrastructural examination reveals no perox-
astrocytes and the presence of lipid-laden cells. The isomes in renal tubular epithelium, where they are
optic nerves, chiasm, and optic tracts may show dif- normally found in large numbers. Adrenal glands are
fuse deficiency of myelin and presence of gliosis. The either normal in size or small. The medulla is unre-
brain stem is normal in architecture, except for hy- markable. In the zona reticularis and inner fascicula-
poplasia and dysplasia of the inferior olives, which ta large striated cells are seen with dense inclusions.
18.3 Pathogenetic Considerations 157

On ultrastructural examination, these cells contain phages not seen in ZS. The storage cells harbor tri-
trilaminar inclusions. Hyperplasia of pancreatic islets lamellar inclusions and heterogeneous material. They
and pancreatic fibrosis have been observed in isolat- are seen in multiple sites of the reticuloendothelial
ed patients. In muscle tissue myopathic changes and system and are not confined to sites of active degen-
presence of abnormal mitochondria are occasionally eration such as the CNS. They occur in liver, spleen,
observed. lungs, lymph nodes, and gastrointestinal mucosa.
In NALD brains, the gyrational abnormalities are In IRD no malformations of the cerebral cortex are
much milder than in ZS. Areas of polymicrogyria and present and no neuronal heterotopias within the
pachygyria, in particular in the area of the sylvian white matter. The white matter may be hypoplastic.
fissure, and a few islands of heterotopic neurons are White matter changes are mild. Myelin content is
seen within the white matter. The cerebral cortex is diminished, but there are no signs of active demyeli-
otherwise normal. Few cerebellar heterotopias are nation. In the areas of myelin deficiency and gliosis,
seen. Inferior olives may be dysplastic. one finds macrophages surrounding vessels and con-
White matter degenerative changes are, however, taining trilaminar lamellae. There may be cerebellar
much more severe in NALD than in ZS. Diffuse de- atrophy with reduced numbers of granular cells.
myelination involves cerebellar white matter, brain In the liver in IRD fibrotic changes may be present.
stem, and cerebral hemispheres. Demyelination oc- Trilamellar lipid inclusions are seen in macrophages,
curs in zoned lesions in X-linked adrenoleukodystro- Kupffer cells, and hepatocytes. Peroxisomes are ab-
phy, but is more diffuse in NALD. The process is most sent or reduced in number and size. Mitochondria
severe in the cerebellar white matter, the pyramidal have abnormal morphology. No cortical renal cysts
tracts in the internal capsule, brain stem and spinal are seen. No adrenal degeneration is present. Foamy
cord, and in the parieto-occipital region. The cerebral histiocytes may be seen in multiple organs.
arcuate fibers are relatively spared. In the areas of
demyelination, the axons are relatively intact, but if
demyelination is severe, axons may also be destroyed 18.3 Pathogenetic Considerations
and cavitation may occur. In the affected areas gliosis
is present and there is an accumulation of lipids, The assembly of peroxisomes requires the interaction
predominantly in histiocytes and macrophages, but of a set of biogenesis proteins, peroxins, which are en-
little, if any, in astrocytes. Perivascular infiltration coded by PEX genes. Peroxisomal matrix proteins are
with mononuclear inflammatory cells may be pre- synthesized on free polyribosomes and are directed
sent, but is then less severe than in X-linked adreno- to the peroxisome by specific targeting signals, PTS1
leukodystrophy. On ultrastructural examination tri- and PTS2. Most peroxisomal proteins use PTS1;
lamellar inclusions have been found in vacuoles with- phytanoyl-CoA hydroxylase, alkyldihydroxyacetone
in histiocytes and macrophages in areas of demyeli- phosphate synthase, and peroxisomal thiolase are
nation. A polyneuropathy with thin myelin sheaths the only three peroxisomal enzymes that use a PTS2
and trilamellar inclusions in Schwann cells and fibro- targeting sequence. PTS1 consists of a C-terminal
blasts has been found in some of the patients. tripeptide, SKL, that is recognized by the PTS1 recep-
Liver disease in NALD is less severe than in ZS. He- tor. There is a third mechanism for importation of
patic fibrosis and micronodular cirrhosis may occur, peroxisomal membrane proteins (PMPs).
but are not obligatory. Evidence of glycogen deposits In peroxisome biogenesis disorders there is a
may be present. Mitochondria have been described as defect in peroxisomal membrane synthesis or the
either normal or abnormal in morphology. In most matrix protein import. Complementation studies by
cases, absence or a marked reduction in number and somatic cell fusion studies have been extremely
size of peroxisomes has been reported, but enlarged important in the elucidation of the basic defects in
hepatic peroxisomes have also been found. Histio- peroxisome biogenesis defects. In complementation
cytes and macrophages that contain lipid storage ma- studies cultured skin fibroblasts from different pa-
terial are present and may be abundant. On ultra- tients are fused and the resulting multinucleated cells
structural examination these cells are shown to con- are collected. Complementation is said to have oc-
tain the typical trilamellar inclusions. No renal corti- curred when the multinucleated cells show a restora-
cal cysts are present. The adrenal cortex is atrophic tion of function or structural features that were defi-
and contains ballooned, lipid-laden cells. Electron cient in the unfused cell lines. Eleven different com-
microscopy reveals trilamellar inclusions in adreno- plementation groups have so far been identified for
cortical cells and in macrophages. Myopathic changes ZS, NALD, and IRD, and the underlying genes for the
and mitochondrial abnormalities have been found in complementation groups are: PEX1 (complementa-
muscle tissue. tion group 1 or E), PEX2 (group 10 or F), PEX3 (group
NALD is characterized by a generalized, systemic 12 or G), PEX5 (group 2), PEX6 (group 4 or C), PEX10
infiltration of many tissues by lipid-laden macro- (group 7 or B), PEX12 (group 3), PEX13 (group 13 or
158 Chapter 18 Peroxisome Biogenesis Defects

H), PEX16 (group 9 or D), PEX19 (group 14 or J), and largely empty. They lack most of the peroxisomal ma-
PEX26 (group 8 or G). Almost all complementation trix proteins; they contain the unprocessed precursor
groups are associated with more than one clinical form of thiolase, unprocessed acyl-CoA oxidase, and
phenotype and often with all three. Rhizomelic chon- residual dihydroxyacetone phosphate acyltransferase
drodysplasia punctata patients belong to a separate activity. Some catalase has also been found in the
complementation group (complementation group interior of peroxisomal remnant structures.
11), related to the gene PEX7. The absence of normal peroxisomes is associated
PEX1, located on chromosome 7q21–22, encodes with defective function of multiple peroxisomal en-
Pex1p, a protein of the AAA ATPase family involved in zymes. Several of the enzyme proteins, which are nor-
peroxisome matrix protein import (AAA stands for mally located in the peroxisomal matrix, are free in
ATPases associated with diverse cellular activities). the cytosol and are stable and biologically active. This
PEX2, located on chromosome 8q21.1, encodes Pex2p, is the case with catalase, D-amino acid oxidase, ala-
an integral peroxisomal membrane protein involved nine glyoxylate aminotransferase, polyamine oxidase
in matrix protein import. PEX3, located on chromo- and L-a-hydroxy acid oxidase. In contrast, other per-
some 6q23–24, encodes Pex3p, a peroxisomal mem- oxisomal enzymes are synthesized normally, but are
brane protein factor for the proper localization of unstable in the cytosol, are rapidly degraded, and
peroxisomal membrane proteins and involved in their enzyme activities are decreased. This is the case
peroxisomal membrane biogenesis. PEX5, located on with the peroxisomal b-oxidation enzyme proteins,
chromosome 12p13, encodes the PTS1 receptor, in- alkyldihydroxyacetone phosphate synthase and dihy-
volved in peroxisome matrix protein import. PEX6, droxyacetone phosphate acyltransferase, and the
located on chromosome 6p21.1, encodes Pex6p, a pro- plasmalogen synthesizing enzymes.
tein of the AAA ATPase family involved in peroxi- In the peroxisome biogenesis disorders, mitochon-
some matrix protein import. PEX10, located on chro- drial abnormalities are also found. Mitochondria are
mosome 1p36, encodes Pex10p, an integral peroxiso- often morphologically abnormal. It is generally as-
mal membrane protein involved in peroxisome ma- sumed that the mitochondrial abnormalities are sec-
trix protein import. PEX12, located on chromosome ondary. There is a metabolic interdependence of
17q11–12, encodes Pex12p, an integral peroxisomal mitochondria and peroxisomes. Participation in fatty
membrane protein involved in matrix protein import. acid metabolism is a property of both organelles. Per-
PEX13, located on chromosome 2p14–16, encodes oxisomes shorten very-long-chain fatty acids prior to
Pex13p, an integral peroxisomal membrane protein their oxidation by mitochondria. Defects in peroxiso-
involved in matrix protein import. PEX16, located on mal b-oxidation lead to accumulation of long-chain
chromosome 11p12–2, encodes Pex16p, an integral fatty acyl-CoAs, which have regulatory effects on a
peroxisomal membrane protein involved in peroxiso- number of mitochondrial enzymes. The mitochon-
mal membrane biogenesis. PEX19, located on chro- drial abnormality may contribute to the clinical dis-
mosome 1q22, encodes a peroxisomal membrane ease. In some patients muscle pathology with abnor-
protein receptor involved in membrane biogenesis. mal mitochondria is observed, suggesting a mito-
PEX26 encodes Pex26p, a peroxisomal membrane chondrial myopathy. However, no changes in lactate,
protein involved in peroxisome matrix protein im- pyruvate, 3-hydroxy butyrate, and acetoacetate have
port. It recruits Pex1p and Pex6p AAA ATPase com- ever been reported in patients, indicating that in vivo
plexes to peroxisomes. the proposed mitochondrial dysfunction is usually of
Complementation group 1 is the largest and con- no or only minor importance.
tains about 65% of the patients. The related clinical The peroxisome biogenesis defects are histopatho-
phenotype is highly variable and covers the entire logically characterized by a combination of malfor-
clinical spectrum from ZS to NALD to IRD. There is mative and degenerative abnormalities. The dysonto-
some genotype–phenotype correlation. Patients with genetic or malformative changes include facial dys-
two null mutations have a more severe phenotype morphia, renal cortical cysts, gray matter migrational
than patients with a residual function of Pex1p. disturbances, and abnormalities in myelin deposi-
With defects in PEX3, PEX16, and PEX19 no perox- tion. The degenerative, regressive changes include the
isomal membrane structures are present at all. In the pigmentary retinal degeneration, the liver fibrosis
other complementation groups, the defect involves and cirrhosis, the adrenal cortical atrophy, and
peroxisomal protein import but not the synthesis of storage, demyelination, and neuronal degeneration.
peroxisomal membranes, and in cells belonging to In ZS the dysontogenetic abnormalities predominate
these complementation groups remnant peroxisomal in the CNS. In NALD there are some dysontogenetic
membrane structures are present, also called peroxi- changes, but CNS pathology is dominated by degen-
somal ghosts. These structures can be demonstrated eration and storage phenomena. In IRD both are mild
in cultured fibroblasts by using antibodies to peroxi- or absent in the nervous system.
somal membrane proteins. These structures are
18.4 Therapy 159

Mechanisms which interfere with migration in ZS acids in the process of myelin breakdown. These may
and NALD do so to a partial degree only, as some neu- be immunogenic and elicit an inflammatory re-
rons are in their normal position and only some of sponse, as seen in X-linked adrenoleukodystrophy.
the neurons of a given class fail to complete their ZS, NALD, and IRD are all characterized by the
migrations. Neuronal migration is not disturbed in presence of trilamellar inclusions in lysosomes, the
rhizomelic chondrodysplasia punctata. This observa- amount of which increases with age. They consist of
tion makes a disturbance of plasmalogen synthesis or cholesterol-bound very-long-chain fatty acids. The
phytanic acid oxidation improbable as the cause of storage is, as a rule, more abundant in macrophages
the migrational defect. The disturbance of neuronal than in parenchymal cells.
migration in patients with D-bifunctional protein de- Hepatic fibrosis and cirrhosis are probably related
ficiency directs attention to the very-long-chain fatty to abnormal bile acid oxidation. Bile acid intermedi-
acid and bile acid abnormalities. Neuronal migra- ates such as trihydroxycholestanoic acid with known
tional abnormalities may reflect the effect of accumu- hepatic toxicity may be important pathogenetically
lated very-long-chain fatty acids, since elevations for both the development of bile duct paucity and
consistently accompany the peroxisomal disorders hepatocellular injury.
with disturbed migration. The exception is X-linked The adrenal cortex is affected in all peroxisomal
adrenoleukodystrophy, in which very-long-chain fat- disorders with an elevation of very-long-chain fatty
ty acids are elevated and no migrational disturbance acids. The increase in these fatty acids causes an in-
is present. However, in X-linked adrenoleukodystro- crease in membrane viscosity in adrenocortical cells,
phy the increase in very-long-chain fatty acids is not which in turn results in a decreased number of
as severe as in ZS or NALD, and is more restricted. In hormone receptor sites, subsequently leading to a
ZS, saturated, monounsaturated and polyunsaturated decreased ability to respond to ACTH. Adrenal insuf-
fatty acids are increased. In NALD and IRD, saturated ficiency followed by atrophy is due to the lack of
and monounsaturated fatty acids are elevated, where- response to ACTH.
as in X-linked adrenoleukodystrophy, only saturated
fatty acids are elevated. A possible role of accumula-
tion of bile acid intermediates must also be consid- 18.4 Therapy
ered. It is hypothesized that an accumulating sub-
stance interferes with the cell adhesion molecule in- In treating patients with a disorder of peroxisome
teractions and linkages, which are necessary for nor- biogenesis, first of all supportive care is essential. In
mal migration of neurons along radial glial fibers. In addition, it would seem rational to try to compensate
ZS, ependymal abnormalities are found which are as far as possible for the biochemical abnormalities
qualitatively similar but quantitatively less extensive that have been brought about by the peroxisomal dys-
than those found in classical lissencephaly. The ab- function. Treatment would include oral supplementa-
normal ependyma may be a primary factor in the tion of ether lipids and bile salts and dietary restric-
pathogenesis of migrational disturbances. tion of very-long-chain fatty acids and phytanic acid.
The white matter abnormalities vary among the The treatment has so far not resulted in definite clin-
disorders of peroxisomal biogenesis from predomi- ical improvement or prolongation of life. Treatment
nantly deficient and disturbed myelination in ZS to with docosahexaenoic acid ethyl ester has been advo-
predominant demyelination in NALD and variable cated on the basis of improvement of biochemical
white matter gliosis in IRD. In ZS myelin is severely parameters and some neurological improvement in
deficient, but no signs of active demyelination are treated children. The rationale of the treatment is the
seen; in NALD the process of myelination is initially observation of severe docosahexaenoic acid deficien-
relatively normal, but demyelination follows. The dif- cy in tissues of children with disorders of peroxiso-
ference between the two may be related to the degree mal biogenesis, while this substance is known to be an
of abnormality of membrane composition. Abnormal important constituent of brain membrane phospho-
membrane composition is related to decreased avail- lipid and of photoreceptor cells. So far, the results of a
ability of plasmalogens and accumulation of very- controlled trial are lacking.
long-chain fatty acids and phytanic acid in the vari- Administration of clofibrate fails to induce liver
ous membrane lipids. The more severe abnormality peroxisomes in ZS patients. Administration of 4-
in ZS results in disturbed myelin formation, whereas phenylbutyrate, a human peroxisome proliferator,
in NALD myelin is laid down but subsequently bro- increased the number of peroxisomes in fibroblasts of
ken down as a consequence of increasing instability. patients with a peroxisome biogenesis disorder (Wei
In IRD the biochemical abnormalities are mildest and et al. 2000). In NALD and IRD fibroblasts, but not in
white matter pathology only mild or minor. The in- ZS fibroblasts, there was an increase in very-long-
flammatory response in NALD may be related to the chain fatty acid b-oxidation and plasmalogen con-
liberation of lipids containing very-long-chain fatty centrations, and a decrease in very-long-chain fatty
160 Chapter 18 Peroxisome Biogenesis Defects

acid concentrations. These data suggest that pharma- pachygyric cortex in the frontoparietal region
cological agents that induce peroxisome proliferation (Figs. 18.1 and 18.2). The pachygyric cortex is visual-
may have a therapeutic potential in the treatment of ized as broad convolutions of mildly thickened cor-
patients with milder variants of peroxisome biogene- tex. The cortex bordering the interhemispheric fis-
sis defects. sure and the occipital cortex are relatively normal.
The problem of any therapeutic trial in the disor- The gyral abnormalities are very extensive and seri-
ders of peroxisomal biogenesis is that the dysontoge- ous in exceptional cases (Fig. 18.1). Small dots of ec-
netic abnormalities cannot be changed by treatment topic gray matter may also be seen under the cortex
and that only the degenerative changes acquired post- and in the subependymal region (Figs. 18.2 and 18.3).
natally can, hopefully, be prevented. The ventricular system is mildly enlarged and tends
to have a primitive form with mildly enlarged occipi-
tal horns, which have a squared-off configuration.
18.5 Magnetic Resonance Imaging The ventricles are rarely markedly enlarged. Germi-
nolytic cysts are often seen in the caudatothalamic
In ZS the migrational derangement is well depicted groove in the early stages (Figs. 18.1 and 18.3). The
by MRI. A very characteristic abnormality is the peri- corpus callosum is thin. The width of the white mat-
sylvian polymicrogyria, which appears as a thickened ter is reduced. Myelination is delayed and may be
cortical mantle consisting of many little dots patchy, consistent with a disturbed myelination.
(Figs. 18.1–18.3). The dots are cross-sections of the In NALD, CT has been shown to reveal progressive
microgyri. The polymicrogyric cortex merges with white matter hypodensities, particularly in the peri-

Fig. 18.1. Sagittal (first row) and axial (second row) T2-weighted images in a neonate with ZS. The lateral aspects of the brain are
diffusely pachygyric and polymicrogyric. The sylvian fissure is wide open. Note the huge subependymal cysts on both sides.
(Courtesy of Dr. M.A. Breukels, Department of Pediatrics, and Drs. J.P. Westerhof and F. Kok, Department of Radiology, Elkerliek
Hospital, Helmond, The Netherlands)
18.5 Magnetic Resonance Imaging 161

Fig. 18.2. Axial T2-weighted images in a 3-month-old boy whereas polymicrogyria is present in the perisylvian region.
with a PEX1 defect and the typical clinical course of ZS.The im- There are several minor neuronal heterotopias in the periven-
ages show some ventricular enlargement. Myelination is tricular region.The cerebellar vermis is hypoplastic
sparse. In the frontal area the gyri are too coarse (pachygyria),

ventricular area, centrum semiovale, and cerebellum. In IRD MRI is normal in some patients, but is ab-
The presence of extensive contrast enhancement in normal in others. MRI does not show migrational ab-
centrum semiovale, internal capsules, and cerebral normalities. Symmetrical abnormalities in signal in-
peduncles has been reported. MRI may either show tensity may be seen in the hilus of the dentate nuclei
evidence of polymicrogyria in the area of the sylvian and peridentate cerebellar white matter (Fig. 18.6).
fissure (Fig. 18.4) or fail to show evidence of gyral These seem to be the first and sometimes the only ab-
abnormalities (Fig. 18.5). When demyelination starts, normalities present. Patchy, ill-defined abnormalities
the earliest changes are seen in the cerebellum, in- may be seen in the periventricular cerebral white
volving both the hilus of the dentate nucleus and the matter (Fig. 18.6). The abnormal white matter merges
peridentate white matter (Figs. 18.4 and 18.5). Other into the normal white matter without sharp demarca-
structures that become involved are brain stem tracts, tion. The corpus callosum and posterior limb of the
in particular the pyramidal tracts, the posterior limb internal capsule may also be involved. There is no
of the internal capsule, and the posterior cerebral contrast enhancement. The white matter abnormali-
white matter more than the anterior white matter ties may be slowly progressive over time, not neces-
(Figs. 18.4 and 18.5). On sequential MRI, the abnor- sarily associated with clinical decline. Profound cere-
malities are rapidly progressive. bral and cerebellar atrophy may occur (Fig. 18.7).
162 Chapter 18 Peroxisome Biogenesis Defects

Fig. 18.3. A 1-week-old baby boy with ZS, related to a PEX1 show mildly enlarged lateral ventricles and absence of a large
mutation. The upper row (T1-weighted sagittal images) shows part of the vermis with an abnormally shaped fourth ventricle.
a thin corpus callosum, a hypoplastic vermis, and a sub- The gyral deformity is most marked in the region of the sylvian
ependymal cyst in the thalamocaudate notch. The sylvian fis- fissure, with a combination of pachygyria and polymicrogyria.
sure has a more vertical orientation than normal and is bor- The left image of the third row shows a germinolytic cyst over
dered by polymicrogyric cortex. The axial T2-weighted images the caudate nucleus and some blood in the occipital horns
18.5 Magnetic Resonance Imaging 163

Fig. 18.4. A 2-year-old boy with a PEX5 defect and progressive and the posterior cerebral white matter.The cortex in the area
neurological deterioration with death at the age of 2.8 years. of the sylvian fissure appears mildly dysplastic with evidence
The images show white matter abnormalities in the hilus of of some polymicrogyria. Courtesy of Dr. P.G. Barth [Barth et al.
the dentate nucleus, the cerebellar hemispheric white matter, 2001 (patient 3), 2004 (patient 4), with permission]
164 Chapter 18 Peroxisome Biogenesis Defects

Fig. 18.5. A 2.5-year old girl with a PEX5 defect and progres- dal tracts of the brain stem, and the posterior and central cere-
sive neurological deterioration who died at the age of 3 years. bral white matter. There is no evidence of cortical dysplasia.
The images show signal abnormalities in the hilus of the den- Courtesy of Dr. P.G. Barth [Barth et al. 1990, 2004 (patient 1),
tate nucleus, the cerebellar hemispheric white matter, pyrami- with permission]
18.5 Magnetic Resonance Imaging 165

Fig. 18.6. A 10.5-year-old boy with a PEX1 defect and a stable rior cerebral white matter, and splenium of the corpus callo-
clinical course. Note the signal abnormalities in the hilus of the sum. Apart from the hilus of the dentate nucleus, the signal
dentate nucleus, the corticospinal tracts at the level of the changes are mild and poorly demarcated. Courtesy of Dr. P.G.
midbrain, the posterior limb of the internal capsule, the poste- Barth [Barth et al. 2004 (patient 13), with permission]
166 Chapter 18 Peroxisome Biogenesis Defects

Fig. 18.7. T2-weighted images of a 14-year-old boy with a white matter, extending into the arcuate fibers.The hilus of the
PEX1 defect and a very slow downhill course.The images show dentate nucleus is abnormal in signal.Courtesy of Dr.P.G.Barth
dilated ventricles and cerebral and cerebellar atrophy and [Barth et al. 2004 (patient 11), with permission]
widespread signal abnormalities in the atrophic cerebral
Chapter 19

Peroxisomal D-Bifunctional Protein Deficiency

19.1 Clinical Features demonstrated and mutations are found in the D-BP
and Laboratory Investigations gene. Prenatal diagnosis is possible by measuring D-
bifunctional protein activity in chorionic villus sam-
A limited number of patients have been described ples and, if the molecular defect has been resolved in
with isolated D-bifunctional protein deficiency the index patient, by DNA analysis.
(BPD), a disease with an autosomal recessive mode of
inheritance. The children show severe CNS involve-
ment with profound hypotonia from birth onwards. 19.2 Pathology
There are minimal spontaneous movements and the
neonatal reflexes are depressed. Feeding problems are Postmortem examination of the brain reveals the
usually prominent. In most children dysmorphic brain to be relatively large. A combination of mal-
features are present with high forehead, large open formative and destructive abnormalities is usually
fontanel, flat nasal bridge, low-set ears, high-arched found. Polymicrogyria is present in most but not all
palate, and micrognathia, similar to the facial charac- patients, in particular involving the lateral aspects of
teristics seen in patients with generalized peroxiso- the brain in the area of the sylvian fissure. Scattered
mal dysfunction. Some infants have macrocephaly. heterotopic neurons may be found in the centrum
Joint contractures may be present. Most patients semiovale, the subcortical white matter, and cerebel-
suffer from severe epilepsy from the beginning and lar white matter. Mild dysplasia of the inferior olivary
seizures are often uncontrollable. Psychomotor de- nucleus is noted. In addition, lack of stainable myelin
velopment is severely delayed and most patients is found in the cerebral and cerebellar white matter,
fail to acquire any significant developmental mile- probably partially related to a disturbance of myeli-
stones. Some patients have ocular abnormalities, nation, but there is also evidence of active demyelina-
including glaucoma, optic atrophy, pigmentary reti- tion. The extent of both components also depends on
nal degeneration, and cataract. Auditory dysfunction the age at death. In infants of a few months the myelin
is present in some. Hepatomegaly is found in some of content of the brain may be normal. In older infants,
the patients. Adrenocortical insufficiency is manifest an evident deficiency of myelin is seen with relative
in few children. Death occurs most often between sparing of the arcuate fibers. The active demyelina-
4 and 12 months of age, but some patients survive a tion is most prominent in the cerebellar and occipital
few years. white matter. In the areas of active demyelination,
EEG is severely abnormal with epileptic dis- axons are relatively spared. Lipid-filled, foamy macro-
charges. BAEP findings are often abnormal. Nerve phages, occasionally striated, and astrocytosis are
conduction velocities are normal. Skeletal X-ray sur- seen in the areas of demyelination, cerebral cortex,
vey is often normal, but calcific stippling of some and basal nuclei. The macrophages are located in the
joints may be seen. Renal ultrasound is normal. perivascular spaces. Cystic degeneration of the
Laboratory investigations reveal signs of adrenal periventricular white matter has been observed. In
insufficiency in some of the patients. In all patients the spinal cord the anterior and lateral corticospinal
very-long-chain fatty acids are elevated in plasma and tracts and dorsal spinocerebellar tracts show loss of
fibroblasts. The plasma levels of bile acid intermedi- axons and myelin.
ates (dihydroxycholestanoic acid and trihydroxy- In liver, mild fibrosis is found. Electron microscopy
cholestanoic acid) and pristanic acid may also be of liver shows abundant peroxisomes. Additionally,
elevated, but not in all patients.When pristanic acid is adrenocortical atrophy is found with presence of
elevated, phytanic acid is usually also elevated, lipid-containing, ballooned, striated cells. Electron
although much less seriously and with an increase in microscopy reveals trilamellar lipid inclusions in
pristanic to phytanic acid ratio. Plasma pipecolic acid these cells. Minute glomerular cysts may be seen in
and the de novo plasmalogen synthesis in fibroblasts the kidney, but not in all cases.
are normal. A D-bifunctional protein deficiency is
168 Chapter 19 Peroxisomal D-Bifunctional Protein Deficiency

19.3 Pathogenetic Considerations 19.4 Therapy

D-bifunctional protein is involved in the b-oxida- No specific treatment is possible. Supportive care is
tion process of both very-long-chain fatty acids, important.
branched-chain fatty acids, like pristanic acid, and the
oxidation of bile acid precursors. D-bifunctional pro-
tein has two catalytic activities: enoyl-CoA hydratase 19.5 Magnetic Resonance Imaging
and 3-hydroxyacyl-CoA dehydrogenase activity. In
some patients (group 2A), D-bifunctional protein is A number of CT scans have been reported. Most were
completely absent, which is associated with the loss of described as normal, some as showing white matter
activity of both the enoyl-CoA hydratase and 3-hy- hypodensity and slight enlargement of the occipital
droxyacyl-CoA dehydrogenase components. In group horns of the ventricular system.
2B patients, enoyl-CoA hydratase activity is deficient, MRI shows a combination of polymicrogyria,
whereas in group 2C 3-hydroxyacyl-CoA dehydro- especially over the lateral aspects of the brain in the
genase activity is deficient. Group A and group C area of the sylvian fissure, and white matter abnor-
patients accumulate very-long-chain fatty acids, malities (Fig. 19.1). The white matter abnormalities
branched-chain fatty acids, and abnormal bile acid consist of delayed and disturbed myelination
intermediates. Group B patients have elevated very- (Fig. 19.2), while in particular in the older infants
long-chain fatty acids and branched-chain fatty acids, with more advanced myelination active demyelina-
but bile intermediates are normal, due to the normal tion occurs (Figs. 19.3 and 19.4). The demyelination is
3-hydroxyacyl-CoA dehydrogenase activity. most pronounced in brain stem tracts and occipital
In patients with BPD, mutations have been found and cerebellar white matter (Fig. 19.4).
in the gene D-BP, located on chromosome 5q23.1. The The combination of gyrational abnormalities and
patient formerly classified as a thiolase-deficient pa- deficient myelination is also seen in Zellweger syn-
tient (Goldfischer et al. 1986) also has a defect in the drome. The combination of gyrational abnormalities
D-BP gene (Ferdinandusse et al. 2002). and demyelination with predilection for the cerebel-
It is striking that a disease caused by an isolated lar and occipital white matter is also seen in neonatal
deficiency of one of the peroxisomal b-oxidation adrenoleukodystrophy. In BPD no inflammatory
enzymes can have clinical symptomatology that is in- reaction is seen in the area of active demyelination.
distinguishable from that of peroxisome biogenesis In conformity with this observation, no contrast en-
defects. This observation indicates the pathophysio- hancement was found on the CT of the child who had
logical significance of accumulation of very-long- active demyelination at autopsy, unlike the situation
chain fatty acids and abnormal bile acid intermedi- in neonatal adrenoleukodystrophy, where extensive
ates. Evidently, the relationship between biochemical contrast enhancement is seen.
abnormalities and clinical symptomatology requires
further elucidation.
19.5 Magnetic Resonance Imaging 169

Fig. 19.1. Baby boy, 3 weeks old, with BPD. The sagittal T1- lateral ventricles. Myelination is compatible with a neonatal
weighted images show a germinolytic cyst in the thalamocau- stage. These images are indistinguishable from those seen in
date notch and polymicrogyria in the perisylvian region. The Zellweger syndrome. Courtesy of Dr. D. Holder, Department of
axial T2-weighted images confirm the perisylvian polymicro- Pediatric Neurology, Cincinnati Children’s Hospital, Cincinnati
gyria. There are possibly some small heterotopias around the
170 Chapter 19 Peroxisomal D-Bifunctional Protein Deficiency

Fig. 19.2. Follow-up MRI of the same boy with BPD at the age the lesions in the hilus of the dentate nucleus. Courtesy of
of 6 months. The polymicrogyria in the perisylvian region is Dr. D. Holder, Department of Pediatric Neurology, Cincinnati
again visualized. The brain stem is now well myelinated, but Children’s Hospital, Cincinnati
there is little myelin in the supratentorial white matter. Note

Fig. 19.3. Follow-up MRI of the same boy with BPD at the age affected. Courtesy of Dr. D. Holder, Department of Pediatric
of 22 months. Demyelination has started in the pyramidal Neurology, Cincinnati Children’s Hospital, Cincinnati
tracts of the brain stem. The cerebellar white matter is also
19.5 Magnetic Resonance Imaging 171

Fig. 19.4. Follow-up MRI of the same boy with BPD at the age white matter are also involved. Courtesy of Dr. D. Holder,
of 26 months, shortly before he died. The process of demyeli- Department of Pediatric Neurology, Cincinnati Children’s Hos-
nation now involves the brain stem more extensively. The pital, Cincinnati
posterior limb of the internal capsule and posterior cerebral
Chapter 20

Peroxisomal Acyl-CoA Oxidase Deficiency

20.1 Clinical Features acyl-CoA oxidase in fibroblasts. DNA confirmation


and Laboratory Investigations by showing mutations in the acyl-CoA oxidase gene is
possible. Prenatal diagnosis is possible using the
A small number of patients has been described with same techniques.
isolated peroxisomal acyl-CoA oxidase deficiency.
This autosomal recessive disease has also been re-
ferred to as pseudo-neonatal adrenoleukodystrophy 20.2 Pathology
(pseudo-NALD), because the clinical features are very
similar to those of neonatal adrenoleukodystrophy. No postmortem examination of the brain has been
The disease had its onset in the neonatal period with performed in patients with pseudo-NALD. Liver tis-
hypotonia and seizures. Neonatal reflexes may be ab- sue investigations show peroxisomes to be present in
sent. Craniofacial dysmorphism may be present and normal or increased numbers and to be increased in
include hypertelorism, epicanthus, low nasal bridge, size.
low-set ears, and polydactyly, but some children have
normal features. Hepatomegaly is present in some but
not all patients. Psychomotor development is retard- 20.3 Pathogenetic Considerations
ed, but several motor milestones can be reached and
children may achieve walking although delayed. They The biochemical findings of an isolated accumulation
may learn to speak a few words. Subsequently, neuro- of VLCFA in the absence of abnormal bile acid inter-
logical deterioration sets in, usually at the age of mediates are consistent with an isolated deficiency of
1–3 years, with regression of motor abilities. The hy- fatty acyl-CoA oxidase. Bile acid intermediates have
potonia gradually changes into hypertonia with hy- their own CoA oxidase, whereas bifunctional protein
perreflexia and positive Babinski signs. The epilepsy and thiolase are active for all substances b-oxidized in
may become very severe with almost continuous peroxisomes. The gene encoding acyl-CoA oxidase or
epileptic seizures. Sensorineural hearing deficit be- palmitoyl-CoA oxidase, ACOX1, is located on chro-
comes apparent. Whereas ophthalmological exami- mosome 17q25.
nation initially reveals normal pupillary light re- Two peroxisomal disorders, peroxisomal acyl-CoA
sponses and normal fundi, increasing abnormalities oxidase deficiency (pseudo-NALD) and X-linked
are subsequently noted with nystagmus, strabismus, adrenoleukodystrophy, are characterized by an isolat-
optic atrophy, tapetoretinal degeneration, and absent ed accumulation of VLCFA. It is striking that both dis-
pupillary light responses. After a few years a vegeta- orders may lead to demyelination with inflammation,
tive state is reached, followed by death. as suggested by the contrast enhancement on neu-
Neurophysiological investigations reveal increas- roimaging. There are, however, also important differ-
ing EEG abnormalities with epileptic discharges; the ences. Patients with acyl-CoA oxidase deficiency
ERG may become flattened, and VEP may become al- display clinical abnormalities from birth on, while
most entirely absent. Motor and sensory nerve con- the clinical symptoms start much later in X-linked
duction velocities are normal. Skeletal X-ray exami- adrenoleukodystrophy. In acyl-CoA oxidase deficien-
nation and ultrasound of the kidneys are normal. cy, the b-oxidation of C22 unsaturated fatty acids by
Laboratory investigations show signs of mild liver chain-shortening and the b-oxidation of dicarboxylic
dysfunction. Serum cortisol level is low, with an in- acids are deficient in addition to the oxidation of
creased ACTH value. In serum and fibroblasts, very- VLCFA, such as C24:0 and C26:0 fatty acids. In
long-chain fatty acids (VLCFA) are elevated, but no X-linked adrenoleukodystrophy only the b-oxidation
increase is found in plasma levels of phytanic acid, of the VLCFA is deficient. These differences may be
pristanic acid, pipecolic acid, and bile acids like dihy- important in explaining the clinical differences.
droxycholestanoic acid and trihydroxycholestanoic
acid. In fibroblasts, VLCFA b-oxidation is seriously
deficient, whereas de novo plasmalogen biosynthesis
and other peroxisomal parameters are normal. The
diagnosis is confirmed by measuring the activity of
20.5 Magnetic Resonance Imaging 173

Fig. 20.1. A 3-year-old female patient with isolated peroxiso- white matter. The pyramidal tracts in the posterior limb of the
mal acyl-CoA oxidase deficiency.Note the signal abnormalities internal capsule are affected. From Suzuki et al. (2002), with
in the corticospinal tracts in the brain stem and the cerebellar permission

20.4 Therapy MRI soon after the onset of neurological deteriora-


tion shows signal abnormalities in the cerebellar
No specific treatment is available. Supportive care is white matter, brain stem tracts, and middle cerebellar
important. peduncles (Figs. 20.1 and 20.2). On follow-up, the
pyramidal tracts at higher levels of the brain stem and
the posterior limb of the internal capsule become
20.5 Magnetic Resonance Imaging abnormal in signal. Subsequently signal changes
develop in the periventricular parieto-occipital white
In several patients CT scan of the brain was per- matter and the splenium of the corpus callosum
formed at birth and found to be unremarkable. No ev- (Figs. 20.2 and 20.3), spreading outward and forward,
idence of cortical malformation was seen, although finally involving also the frontal white matter. If
minor or mild abnormalities in gyration and hetero- contrast is administered, enhancement is expected. No
topias can easily be missed on CT. Repeat CT after the evidence of a migrational defect has been reported.
onset of neurological deterioration reveals symmetri-
cal white matter hypodensities in the centrum semio-
vale and the occipital area with contrast enhancement
of the border of the lesions. The CT findings are rem-
iniscent of those reported in neonatal adrenoleuko-
dystrophy.
174 Chapter 20 Peroxisomal Acyl-CoA Oxidase Deficiency

Fig. 20.2. The first two rows show the images of a male patient the same patient at the age of 7 years and 11 months.The pos-
with peroxisomal acyl-CoA oxidase deficiency at the age of 3 terior limb of the internal capsule, parieto-occipital white mat-
years and 5 months. There are extensive signal abnormalities ter, and splenium of the corpus callosum now display promi-
in brain stem tracts, middle cerebellar peduncles, and cerebel- nent signal abnormalities. From Suzuki et al. (2002), with per-
lar white matter.There are incipient signal changes in the pari- mission
eto-occipital white matter. The third row shows the images of
20.5 Magnetic Resonance Imaging 175

Fig. 20.3. Female patient with peroxisomal acyl-CoA oxidase splenium of the corpus callosum are affected. In addition,
deficiency at the age of 3 years and 3 months. Tracts in the there is some generalized cerebral atrophy.The images resem-
brain stem (especially the corticospinal tracts), cerebellar ble those of the cerebral form of X-linked adrenoleukodystro-
white matter,hilus of the dentate nucleus,posterior limb of the phy. From Suzuki et al. (2002), with permission
internal capsule, thalamus, parieto-occipital white matter, and
Chapter 21

X-Linked Adrenoleukodystrophy

21.1 Clinical Features gait, loss of vision, and impaired auditory discrimina-
and Laboratory Investigations tion. The course of the disease is relentlessly progres-
sive and spastic tetraplegia and dementia become
X-linked adrenoleukodystrophy (XALD) is a geneti- manifest in months. Decreased vision is caused by op-
cally determined disorder that mainly involves the tic atrophy or bilateral occipital white matter lesions
adrenal cortex and the CNS. Inheritance is X-linked or, more often, a combination of the two. Initially,
recessive. The disease has a wide phenotypic variabil- neurological findings are often asymmetrical with
ity. The rapidly progressive childhood cerebral form hemiparesis or hemianopia. Frequently noted subse-
accounts for about one-third of the cases. Adreno- quent problems are dysarthria, dysphagia, and hear-
myeloneuropathy (AMN) has a later onset and slower ing loss. Cerebellar ataxia or sensory disturbances
progression and accounts for about 40–45% of the may be present, but are not usually prominent. There
patients. The relative frequency of the Addison-only are no clinical signs of peripheral nerve dysfunction.
form varies with age and accounts for up to 50% of Progressive dementia occurs. Epileptic seizures occur
the cases in childhood. Less frequent variants include and are often multifocal in origin. The pace of deteri-
the adolescent and the adult cerebral forms. In addi- oration is variable. In the final stage a spastic quadri-
tion, there are patients with unusual presentation and plegia is present and a variable degree of decorticate
asymptomatic patients, a common observation below posturing. The affected boys are blind, deaf, and
the age of 4 years and very rare above the age of 40. mute. A vegetative state or death is reached in 1–5
These different phenotypes may occur all within one years. Most patients die within 2 or 3 years after the
affected family. Even identical twins may display dif- onset of neurological symptoms, but some live for
ferent phenotypes. Females may also be affected, es- many years in a vegetative state. However, some pa-
pecially when they become older. The minimum fre- tients stabilize for years in an earlier stage of the dis-
quency of males with the defect has been estimated to ease.
be 1:42,000 in the USA, whereas the minimum fre- The adolescent (onset at 10–21 years) and adult
quency of males with the defect and female carriers is (onset after 21 years) cerebral forms resemble the
estimated to be 1:16,800 (Bezman et al. 2001). childhood cerebral form, except for the later onset.
In the childhood cerebral form of XALD, the age at Just like the childhood from, the adolescent and adult
onset of neurological symptoms is usually between 5 cerebral forms have a rapidly progressive course. The
and 9 years. Features of adrenal insufficiency may oc- disease is often misdiagnosed. It may present as a psy-
cur before overt neurological symptoms or may fol- chosis or dementing illness, or as a single focal brain
low. In some cases a diagnosis of Addison disease is lesion that can be mistaken for a tumor.
made 1–3 years before any neurological disorder is The usual age at onset of AMN is within the third
evident on the basis of increased pigmentation, fa- or fourth decade, but ranges from 14 to 60 years. Most
tigue, episodes of vomiting, or catastrophic reactions patients with an adult onset form of XALD have
to intercurrent infections. In others, neurological de- AMN. Neurological deficits are primarily due to a
terioration may continue for some years without en- myelopathy and to a lesser extent a neuropathy. Af-
docrine symptoms, and sophisticated investigations fected males have a slowly progressive paraparesis,
are needed to reveal evidence of adrenocortical dys- disturbed vibration sense of the legs, voiding distur-
function. Adrenocortical dysfunction is present in at bances, and a variable degree of sexual dysfunction.
least 80% of the patients. Signs of a distal polyneuropathy may also be found.
The earliest neurological symptoms are often Approximately two-thirds of the patients have overt
vague and frequently consist of behavioral changes. or biochemical signs of adrenocortical insufficiency,
The changes vary from withdrawn to bizarre hyper- which may precede or follow neurological dysfunc-
active and aggressive behavior and are often accom- tion. Twenty percent of the patients have signs of hy-
panied by poor school performance. Many boys re- pogonadism with infertility. Many patients have
ceive psychiatric treatment until deteriorating learn- scanty scalp hair. The disease is slowly progressive
ing capabilities and other neurological symptoms over decades.About half of the patients with AMN de-
force recognition of the organic nature of the disease. velop some cerebral involvement, with usually mild
Early neurological abnormalities are disturbances of cognitive dysfunction. Visual memory, spatial cogni-
21.1 Clinical Features and Laboratory Investigations 177

tion, and psychomotor speed are affected most. Psy- corticosteroids and 17-oxysteroids may be reduced.
chological dysfunction may also occur, with emotion- Further evidence for primary adrenocortical insuffi-
al disturbances and depression. About 20% of the pa- ciency is found in impaired cortisol responsiveness to
tients with AMN develop a rapidly progressive cere- adrenocorticotropic hormone (ACTH) in the pres-
bral white matter involvement as seen in the cerebral ence of elevated baseline ACTH levels. Evidence of
forms of XALD. primary testicular insufficiency is provided by low
In the Addison-only form no neurological dys- testosterone levels and elevated luteinizing hormone
function is found. However, all patients are at risk of (LH) or follicle stimulating hormone (FSH) levels.
developing overt neurological symptoms sooner or CSF protein is elevated in the majority of the sympto-
later. Some patients develop Addison disease in child- matic patients, sometimes combined with an eleva-
hood and neurological abnormalities only arise in tion of g-globulin level or moderate increase in lym-
adulthood. phocytes.
The asymptomatic group includes males in whom Diagnosis of XALD depends upon the demonstra-
the typical biochemical abnormalities of XALD are tion of abnormally high levels of saturated very-long-
found and who are completely healthy. Most of the chain fatty acids (VLCFA) in plasma, cultured skin
males included in this group are small boys who were fibroblasts, or tissue. For routine purposes plasma
investigated because of clinically evident XALD in an is used. The concentration of C26:0 fatty acids is in-
older brother. The group also includes adolescents vestigated as well as the ratios of C26:0/C22:0 and
and a few older males. The oldest asymptomatic male C24:0/C22:0 fatty acids. In over 90% of XALD patients
ever mentioned was 62 years old. all three parameters are abnormally elevated. In a mi-
There are a few patients with unusual forms of nority of the patients only one or two of the three pa-
XALD. Patients have been reported with predomi- rameters are abnormal. VLCFA analysis and C26:0
nantly cerebellar ataxia or a spinocerebellar syn- fatty acid b-oxidation measurements in cultured skin
drome. One patient, aged 57 years, has been reported fibroblasts are used for definite diagnosis. Patients
who showed rapid neuropsychiatric deterioration with XALD already show the characteristic elevations
and signs of cerebral demyelination at the site of a se- in blood VLCFA during the first 2 weeks of life and
vere cerebral contusion suffered several months pre- even in cord blood.
viously. Cerebral XALD presents rarely as an acute Neurophysiological investigations are often used
encephalopathy with seizures, status epilepticus, to establish the extent of disease in XALD. Nerve con-
headache, vomiting, lowering of consciousness, or duction studies usually show a normal conduction
even coma. Papilledema may be seen. The presence of velocity in cerebral forms, but may also show a de-
fever may suggest encephalitis. After this acute creased velocity. In AMN the conduction velocity is
episode, the patient recovers with temporary en- decreased. In cerebral forms of ALD the EEG is as a
cephalopathic signs, which resolve in the course of rule abnormal, although nonspecifically, with diffuse
days and weeks. slowing of the rhythm and a maximum usually in the
Female XALD carriers may also develop clinical posterior regions. Evoked potential studies may show
problems. Most women below the age of 30 years are abnormalities reflecting the central white matter in-
uninvolved. The percentage of females with neurolog- volvement.
ical problems increases with age. Approximately 50% Investigation of the level of VLCFA is also a sensi-
of the female carriers of 40 years and older display tive test in carrier detection: 85% of heterozygotes
signs of a mild myelopathy with increased tendon re- can be identified when the results of VLCFA assays in
flexes and distal sensory changes in the legs, but no or plasma and cultured fibroblasts are combined. Con-
mild disability. About 15% of the females of 40 years sequently, about 15% of the female carriers have false
or older have an AMN-like clinical phenotype, but negative test results. Monoclonal antibodies have
with later onset and milder symptomatology. Cere- been raised against the XALD gene product (ALD
bral involvement and adrenocortical insufficiency are protein, ALDP) and use of these antibodies in im-
very rare among female carriers at all ages. Cerebral munofluorescence studies of cultured skin fibroblasts
involvement is exceedingly rare in childhood and or leukocytes may aid in the identification of het-
then probably related to skewed, highly unfortunate X erozygous females, in particular when VLCFA con-
inactivation or a partial deletion of one X chromo- centrations in plasma and fibroblasts are normal. In
some and a mutated gene on the other chromosome. 70–80% of the affected kindreds affected boys and
Some older female carriers have mild intellectual im- men lack ALDP immunoreactivity; female carriers in
pairment, which is only detected by detailed psycho- these kindreds show a mixture of positive and nega-
logical tests. An adult female has been described with tive immunoreactivity in their cells. Antenatal diag-
a lethal cerebral leukodystrophy. nosis can be established by measuring VLCFA in cul-
Laboratory testing usually reveals signs of adreno- tured amniocytes and chorionic villi, by measuring
cortical dysfunction. Urinary excretion of 17-hydroxy- the activity of peroxisomal b-oxidation, and im-
178 Chapter 21 X-Linked Adrenoleukodystrophy

munofluorescence analysis of ALPD. DNA techniques marked perivascular mononuclear cell infiltration.
can also be used for carrier detection and prenatal This zone contains many large ballooned macro-
diagnosis. A problem in genetic counseling and pre- phages laden with lipids. There are many preserved
natal diagnosis is that the demonstration of the bio- demyelinated axons and little myelin remains. The
chemical defect does not provide information about large central area is destroyed and burnt out. There is
whether the patient will develop severe childhood no evidence of an active process. Axons, myelin
XALD or the milder AMN. sheaths, and oligodendroglia are absent. There are
few lymphocytes and only occasional macrophages
surrounding blood vessels. This area is filled with a
dense mesh of glial fibrils and scattered astrocytes.
21.2 Pathology Sometimes cavitation or calcium depositions are seen
in this area.
At autopsy the surface of the brain of a patient with In the brain stem and spinal cord degeneration of
cerebral XALD is either normal or shrunken, depend- the tracts appears to proceed at the same rate
ing on the degree of tissue loss. The central white mat- throughout, with no evidence of a dying-back phe-
ter is grayish and indurated, sometimes cystic and nomenon. Here too the demyelinating process occurs
cavitated. The thickness of the cortex is normal. The in a continuous fashion. No small independent foci of
atrophic external appearance is, if present, secondary demyelination are seen. Perivascular accumulations
to loss of white matter. In cases of extensive loss of of inflammatory cells may well be noted.
myelin the ventricular system is enlarged. The cytoarchitecture of the cerebral cortex is nor-
The pathological abnormalities are the same for mal. Only in more advanced cases may neuronal loss
childhood, adolescent, and adult cerebral XALD and and gliosis be seen, especially in the deeper cortical
consist of widespread demyelination of the white layers. The cerebral cortical damage is mainly found
matter. The demyelinating lesion constitutes one in the occipital region where the demyelinating lesion
large area extending across the corpus callosum and may not spare the U fibers and may be contiguous
involving both hemispheres. In most cases of cerebral with the deep layers of the cortex.
XALD the demyelinating process starts in the spleni- Electron microscopic examinations show that
um of the corpus callosum and spreads bilaterally to many macrophages and microglia contain distinctive
the occipital region. Gradually the process spreads cytoplasmic inclusions, consisting of linear lamellae.
outwards and forwards as a confluent lesion until An individual lamella has a trilaminar structure, con-
most of the cerebral white matter is involved. The sisting of paired electron-dense leaflets separated by
frontal white matter is generally affected less severely an electron-lucent space. These trilamellar structures
and often asymmetrically. The subcortical U fibers are often closely associated with lipid droplets. They
are preserved until a far-advanced stage, and usually are not found in oligodendroglia or astrocytes. In
the U fibers in the occipital area are affected before addition, macrophages contain myelin debris.
those elsewhere. Other areas of the brain that are usu- Microscopic examination of peripheral nerves
ally heavily involved are the fornix, the hippocampal reveals either no abnormalities or demyelination. On
commissure, the posterior limb of the internal cap- ultrastructural examination abnormal cytoplasmic
sule, the lateral two-thirds of the cerebral peduncles inclusions may be seen in Schwann cells and in en-
including the occipitoparietotemporopontine and doneurial macrophages. These inclusions have the
pyramidal tracts, and the lateral lemniscus. In some characteristic linear, trilamellar appearance.
patients the cerebellar white matter is involved, some- Pathological studies in AMN demonstrate bilater-
times the cerebellar peduncles as well. In a smaller al, usually symmetrical, long tract degeneration in the
portion of the patients with cerebral XALD the de- spinal cord with most prominent involvement of the
myelination starts bilaterally in the frontal area, also corticospinal and dorsal tracts. The distribution of
involving the anterior part of the corpus callosum. In the degeneration of these tracts conforms to a dying-
these cases the anterior limb of the internal capsule back pattern in that the greatest loss of myelinated
and the medial third of the cerebral peduncles con- fibers is found in the cervical dorsal tracts and the
taining the frontopontine tracts are affected. In a few lumbar corticospinal tracts. Axonal loss is equal to or
patients the demyelinating lesions are highly asym- greater than myelin loss. The above findings provide
metrical. strong evidence for primary axonal degeneration.
Within the white matter lesion three zones can be There are no inflammatory cells. Microglia are in-
distinguished on histopathological examination. The creased in numbers and appear the dominant re-
outer zone shows evidence of active destruction of sponding cells. Dorsal ganglion cells are not de-
myelin with axonal sparing. Scattered PAS-positive creased in numbers but are decreased in size. Periph-
and sudanophilic macrophages are present. The eral nerves display both axonopathic and myelino-
middle zone shows signs of active inflammation with pathic features. In Schwann cells and macrophages
21.4 Pathogenetic Considerations 179

the typical trilamellar inclusions are present. There for regions with different stages of myelin breakdown
are no inflammatory cells. in the cerebral form of XALD. In morphologically
The cerebral involvement in AMN is highly vari- normal white matter, subtle changes in lipid composi-
able. In most AMN patients, patchy, poorly defined, tion are found. Phospholipids are increased, whereas
small areas of demyelination are scattered through- galactolipids and cholesterol are slightly decreased.
out the cerebral hemispheres, with relative or total ax- Only traces of cholesterol esters are found in histolog-
onal sparing, activation of microglia, presence of stri- ically intact white matter. The fatty acid composition
ated macrophages with trilamellar inclusions, and ab- of cholesterol esters, cerebroside, and sulfatide in in-
sence of inflammatory cells. In some patients, moder- tact white matter is relatively normal, whereas phos-
ate diffuse demyelination is observed in the cerebral pholipids in the same area contain increased VLCFA,
white matter and to a lesser extent the cerebellar with the most striking increase in VLCFA in phos-
white matter, with sparing of the U fibers and without phatidylcholine. A moderate increase in VLCFA is
inflammation. Still others display inflammatory de- seen in gangliosides. The area of active demyelination
myelinating lesions with axonal loss, qualitatively shows major changes in lipid composition. The water
similar to cerebral XALD but much more localized. content is increased, the amount of total lipids is de-
Some AMN patients develop the cerebral form of creased, and there is a large increase in cholesterol es-
XALD with rapidly progressive inflammatory de- ters, whereas unesterified cholesterol is severely de-
myelination. In addition to these inflammatory de- creased. Galactolipids are decreased, whereas phos-
myelinating lesions, AMN patients also demonstrate pholipids are stable as a proportion of total lipids.
noninflammatory, bilateral, fairly symmetrical le- The fatty acid composition of cerebroside and sul-
sions with comparable loss of axons and myelin fatide shows a slight elevation in VLCFA, whereas the
sheaths, involving most often the brain stem corti- VLCFA content of phospholipids, gangliosides, and
cospinal and spinocerebellar tracts, medial and later- cholesterol esters is greatly increased. The most strik-
al lemnisci, cerebellar peduncles, posterior limb of ing rise in VLCFA among the phospholipids is seen in
the internal capsule, and the optic radiations. phosphatidylcholine and sphingomyelin. In the area
The adrenal glands in XALD show gross atrophy of of gliosis, the amount of remaining lipids is small, and
the cortex, the medulla being normal. The zona retic- the water content is high. The amount of galactolipids
ularis and zona fasciculata are particularly affected is relatively very low, whereas phospholipids and cho-
with ballooned cortical cells, in which a striated ap- lesterol are low in absolute content but constitute a
pearance of the cytoplasm may be seen. These striat- relatively normal proportion of total lipids. Choles-
ed cells are specific for XALD. Ultrastructurally the terol esters are present in small but measurable
striations are shown to consist of linear, trilamellar amounts. Significant amounts of triglycerides and
accumulations within the adrenal cortical cell cyto- free fatty acids can also be measured. In gliotic tissue
plasm. Lymphocytic infiltrates are found in a minori- the VLCFA content of cerebroside, sulfatide, and
ty of the patients. Light microscopic examination of phospholipids is barely elevated, whereas the VLCFA
the testis often reveals no abnormalities, although ful- content of gangliosides and cholesterol esters is mild-
ly developed Leydig cells may be lacking. Interstitial ly to markedly elevated.
cells, presumptive Leydig cell precursors, may contain The adrenal and testicular content of cholesterol
the characteristic trilamellar profiles. In all tissues, esters is abnormally high. The cholesterol esters
the morphology of peroxisomes is normal. contain an abnormally elevated amount of saturated
VLCFA.

21.3 Chemical Pathology


21.4 Pathogenetic Considerations
XALD is a lipidosis, in which accumulation of saturat-
ed VLCFA occurs in all tissues, especially in CNS XALD is caused by mutations in the gene ABCD1,
white matter, peripheral nerve, adrenal cortex, and which is located on chromosome Xq28 and encodes
testis. Substantial quantities of these VLCFA are de- ALDP, a peroxisomal ATP-binding cassette trans-
posited as cholesterol esters, which appear as the membrane transporter. A great number of different
characteristic lamellated cytoplasmic inclusions. mutations have been identified. No consistent corre-
These VLCFA vary in chain length from C23 to C32 lation between genotype and phenotype has been
with a peak at C25–C26. Several other lipids also con- demonstrated.
tain an increased percentage of saturated VLCFA. The disease is biochemically characterized by ele-
These increases in VLCFA are found in cerebral vated VLCFA due to reduced VLCFA b-oxidation. The
XALD, AMN, and female carriers. first step in the b-oxidation of VLCFA is conversion of
The changes in lipid composition of myelin and fatty acid to fatty acyl-CoA, catalyzed by the enzyme
whole white matter have been investigated separately VLCFA-CoA synthase or ligase, present on the perox-
180 Chapter 21 X-Linked Adrenoleukodystrophy

isomal membrane. The enzyme is more specifically myelin antibodies in serum of XALD patients. Tumor
called lignoceroyl- or hexacosanoyl-CoA synthase or necrosis factor-alpha (TNF-a), a proinflammatory
ligase. The relation between ALDP and VLCFA-CoA cytokine thought to be responsible for tissue damage
ligase is still elusive.ALDP is not necessary for the im- in inflammatory brain disorders including multiple
port of this enzyme into peroxisomes or the import of sclerosis, is expressed in astrocytes and macrophages
VLCFA into peroxisomes. The reason for impaired at the active edge of the lesion. Reactive astrocytes,
VLCFA b-oxidation in XALD has yet to be found. A macrophages, and T lymphocytes are the most preva-
recent hypothesis assumes that ALDP facilitates the lent cellular elements. Most lymphocytes are CD8-
interaction between peroxisomes and mitochondria, positive cytotoxic T cells. CD1 molecules, which play
and that ALDP deficiency leads to impaired b-oxida- major roles in lipid antigen presentation, have been
tion in mitochondria. The repeated observation of found to be present, most conspicuously in the acute
structural mitochondrial abnormalities in XALD tis- inflammatory lesions. It is hypothesized that the pri-
sues, including lipid inclusions in mitochondria, is an mary biochemical abnormality in XALD, i.e. the ab-
argument in favor of this hypothesis. normal accumulation of VLCFA, leads to membrane
The impaired degradation of VLCFA leads to en- instability and breakdown, leading to liberation of
richment of these fatty acids in various lipids at the VLCFA-containing moieties, which may be antigenic
expense of the normally degraded short-chain fatty and elicit an immune response. Not only peptide anti-
acids. The accumulating fatty acids are saturated and gens, but also lipid antigens may play a key role in the
have a chain length varying between C24 (lignoceroyl pathogenesis of inflammatory demyelination in cere-
acid) and C32 with a peak at C26 (hexacosanoic acid). bral XALD. Lipids containing an abnormally high
VLCFA are derived both from the diet and from en- proportion of VLCFA may stimulate nearby astro-
dogenous synthesis by a microsomal system that cytes, microglia, and macrophages to initiate a cyto-
elongates long-chain fatty acids. There is evidence kine cascade resulting in further myelin destruction
that in XALD, not only b-oxidation of VLCFA is de- by T cells, B cells, and complement. This two-stage hy-
creased, but fatty acid chain elongation activity is al- pothesis explains why the zone of active inflamma-
so elevated, contributing to the accumulation of VL- tion in cerebral XALD is found behind the zone of ac-
CFA. It has been shown that the addition of monoun- tive demyelination. This location of the inflammation
saturated fatty acids to culture medium has a dramat- contrasts with the situation in multiple sclerosis, in
ic effect in lowering the content of VLCFA in XALD which the inflammation is most intense at the edges
fibroblasts. These monounsaturated fatty acids ap- of the lesion, with little or no inflammatory response
pear to inhibit the synthesis of VLCFA without having in the inner zones of the lesion. Others hypothesize
any effect on the degradation of VLCFA. that accumulation of VLCFA in membrane domains
The exact mechanisms by which nervous tissue associated with signal transduction pathways may
damage occurs are still unknown. There is a funda- trigger inflammatory processes through activation of
mental difference between the inflammatory de- microglia and astrocytes, resulting in loss of myelin.
myelinative lesions seen in the white matter of Adrenal cortical cells, Leydig cells, and Schwann
patients with cerebral XALD and the axonopathic cells are also involved in the disease process. They ac-
lesions seen in patients with AMN and AMN-like phe- cumulate VLCFA incorporated in cholesterol esters in
notypes. Both biochemical and immunological mech- the form of lamellar cytoplasmic inclusions. A cyto-
anisms may be important in the pathogenesis of the toxic pathogenesis has been proposed for the adren-
lesions. The greater length of the aliphatic chain caus- al, testicular, and Schwann cell lesions. Furthermore,
es VCLFA to be extremely insoluble. Abnormally high it has been shown that accumulation of VLCFA in
VLCFA levels alter membrane physiological proper- adrenal cortical cell membranes leads to increased
ties and functions. Increasing levels of VLCFA in membrane microviscosity and can interfere with
membranes may result in instability and breakdown ACTH responsiveness. Impaired ACTH receptor
of the membranes, leading to noninflammatory function has been found, probably contributing to the
myelin loss and axonal degeneration, as seen in AMN. adrenocortical insufficiency. It is noteworthy that the
The destruction of adrenocortical cells, Leydig cells, destruction of these cells is accompanied by little or
and Schwann cells is also noninflammatory and may no inflammation.
be related to the toxic effects of VLCFA. The rapidly All phenotypic variants of XALD have the same
progressive demyelination with a marked inflamma- basic defect and all variants may occur within the
tory reaction suggests additional immunological same family. No differences in fatty acid abnormality
pathogenetic mechanisms in cerebral XALD. Further could be established in fibroblasts, erythrocyte mem-
evidence for a role of immunological mechanisms is branes, or blood in repeated investigations. There is
found in the occasional presence of signs of intrathe- evidence for an autosomal modifier gene, which
cal immunoglobulin production, increased levels of would explain the phenotypic variability of XALD
IgA and IgG in XALD tissues, and high levels of within pedigrees. This modifier gene probably modu-
21.5 Therapy 181

lates the immune response, considering the marked (GTE; erucic acid is a C22 monounsaturated fatty
difference in immune reaction between cerebral acid), popularly referred to as Lorenzo’s oil, can lead
XALD and AMN. It is likely that environmental fac- to complete normalization of blood levels of VLCFA.
tors may also influence the phenotype, as disparate Moderate reduction in platelet count occurs as a side
phenotypes can be seen in sets of identical twins. effect in 40% of the patients but does not lead to he-
Many female heterozygotes suffer from a late-on- morrhages. This treatment does not alter the clinical
set, slowly progressive neurological disorder. Inacti- disease course in patients in whom the neurological
vation of one of the two X chromosomes in female so- deterioration has already set in. However, there is in-
matic cells is considered to be a random process. creasing evidence that, depending on the degree of
However, there is some evidence that, in XALD carri- treatment compliance and decrease in VLCFA, the
er females, selection mechanisms favor cells express- treatment has a protective effect when started in neu-
ing the mutant allele rather than cells expressing the rologically intact patients. Treated adequately, a
normal allele. This observation may explain the rela- smaller number of boys with the biochemical defect
tively frequent occurrence of symptoms in female develop the aggressive cerebral form of the disease,
carriers. There is also evidence that skewing of X in- and the onset of neurological symptoms is delayed.
activation is an important factor in determining the Unfortunately, dietary treatment is not an absolute
severity of disease manifestations in female XALD preventive, and patients may still develop cerebral
carriers. XALD despite perfect compliance and normalization
of plasma VLCFA levels.
Another important cornerstone in the treatment of
21.5 Therapy cerebral XALD is bone marrow transplantation. The
outcome is highly dependent on the degree of cere-
First and foremost, family counseling, carrier detec- bral involvement at the time of the transplantation. If
tion, and prenatal diagnosis are important in prevent- applied early in the course of the disease, bone mar-
ing the occurrence of further cases in known families. row transplants may prevent further deterioration
Symptomatic care is essential. Hormonal substitu- and may even lead to improvement of cerebral lesions
tion is necessary to correct the adrenocortical insuffi- on MRI. In the brain, the donor-derived cells are mi-
ciency, if present. Testosterone administration can be croglia, and it is assumed that the beneficial effect of
of help in AMN patients who have testicular insuffi- bone marrow transplantation is that donor microglia
ciency. Psychiatric care may be necessary, in particu- metabolize VLCFA, thus reducing their levels in the
lar in adult patients with cerebral XALD. brain. The observation that improvement appears to
An important difference between cerebral XALD commence only 6 months after transplantation is
and the more benign AMN is in the presence or ab- compatible with the slow turnover of microglia. The
sence of an inflammatory response.Various modes of therapy is not recommended for patients with rapid-
immunosuppression and immunomodulation have ly advancing or severe cerebral involvement, because
been tried in order to influence the inflammatory these patients do not tolerate the temporary worsen-
reaction, including the use of steroids, b-interferon, ing associated with the procedure or the delay in on-
cyclophosphamide, pentoxifylline, thalidomide, cy- set of beneficial effect. Generally, an IQ of 80 (perfor-
closporine, plasmapheresis, and high-dose intra- mance IQ appears to be especially important) is con-
venous immune globulin, and unfortunately so far sidered the limit because outcome in patients with a
none of these has been effective. They do not influ- performance IQ below 80 is poor. In these patients
ence the course of the neurological disease. Inciden- transplantation may even lead to rapid worsening
tally, improvement under steroids has been reported, and sometimes an early death. Delayed stabilization
but this effect may be temporary. in a poor neurological condition or vegetative state
Therapeutic strategies aiming to lower VLCFA may also occur. Bone marrow transplantation is also
have been the cornerstone of therapeutic strategies not recommended for asymptomatic patients, be-
for a long time. Dietary restriction of saturated cause of the over 50% chance that they will not devel-
VLCFA is insufficient to lower blood VLCFA. Mono- op the serious cerebral form of the disease, and the
unsaturated fatty acids compete with saturated fatty high mortality associated with bone marrow trans-
acids for the microsomal fatty acid elongation system. plantation. Transplantation is also not recommended
Diets enriched in monounsaturated fatty acids lead to in stable patients, because the duration of the stable
decreased synthesis of VLCFA and are more effective period is unclear and may be many years. Unfortu-
in lowering blood VLCFA. The best results are ob- nately, the prognosis of XALD cannot be predicted by
tained by combining the two. Diets restricted in satu- VLCFA measurement, mutation analysis, or the ex-
rated VLCFA combined with the use of a 4:1 mixture pression of the disease in relatives. It is presently not
of glyceryl trioleate (GTO; oleic acid is a C18 mo- possible to predict whether a young asymptomatic
nounsaturated fatty acid) and glyceryl trierucate boy is destined for a severe phenotype, for which
182 Chapter 21 X-Linked Adrenoleukodystrophy

bone marrow transplantation should be performed, cipital white matter. The arcuate fibers are relatively
or a mild phenotype. For this reason, monitoring of or completely spared, which is most easily seen on T1-
the disease on MRI and using neuropsychological weighted images. In most patients two zones can be
tests from the presymptomatic stage onwards is ex- distinguished, the anterior zone where demyelination
tremely important. As soon as lesions appear, and ap- advances being less severely affected than the posteri-
pear to be progressive, bone marrow transplantation or zone. After administration of contrast, a rim of en-
should be attempted. Extensive screening of family hancement can be seen surrounding the most severe-
members should be performed as soon as the diagno- ly affected area, separating this area from the less se-
sis of XALD or AMN has been established in one pa- verely affected and normal white matter (Fig. 21.1).
tient and HLA typing should initiated in all persons These three zones are in accordance with the three
who are potential future candidates for bone marrow zones recognized histologically. The outer and ad-
transplantation in order to search for a suitable donor vancing zone is in the process of active demyelination
and be prepared for transplantation. Bone marrow without inflammation. The middle zone shows signs
transplantation is presently not performed in adult of prominent inflammation. The innermost and most
patients with AMN. This is because the morbidity and posterior region is completely demyelinated and
mortality of bone marrow transplantation are higher burnt out. In this latter area, cavitation and calcifica-
in adults than in children, whereas AMN is a relative- tion may be present. The calcium deposits are best
ly slow disease that may take decades before death en- seen on CT (Fig. 21.2). The lesions are essentially
sues. At present, it is not clear whether bone marrow symmetrical, but are often not so in detail. Demyeli-
transplantation at a young age for incipient cerebral nation advances in the frontal direction. Structures
XALD will prevent the development of AMN in adult- affected relatively early on are the lateral and medial
hood. geniculate bodies, in some cases the lateral-inferior
Promising alternative therapies have been pro- part of the thalamus, the posterior limb of the inter-
posed, but no definitive data on their clinical effects nal capsule, and the external capsule. The frontal lobe
are as yet available. 4-Phenylbutyrate up-regulates the is affected last and more variably. In the end, almost
expression of a protein referred to as ALDR, which is all cerebral white matter is affected. In the end-stage,
a protein with a high homology with ALDP and may serious white matter atrophy is seen. The cerebellum
be able to substitute at least in part for its function. is not usually involved early in the course of disease,
The gene encoding this protein is called ABCD2. in contrast to the brain stem. Typically, the involved
There is evidence that the drug may lead to lowering tracts are the occipitoparietotemporopontine and
of plasma and tissue VLCFA. Lovastatin also induces pyramidal tracts, the brachium of the inferior collicu-
the increased expression of ALDR. It improves the ca- lus, the brachium of the superior colliculus, and the
pacity of XALD fibroblasts to metabolize VLCFA and lateral lemniscus. The frontopontine tracts are pre-
normalizes VLCFA in plasma of XALD patients. Lo- served. This MRI pattern is present in about 80% of
vastatin may also inhibit inducible nitric oxide syn- patients with childhood cerebral XALD and has a
thase and proinflammatory cytokines. However, sim- lower percentage in older patients. The overall fre-
vastatin, an analogue of lovastatin with similar phar-
macokinetics and effects on plasma VLCFA, failed to
decrease VLCFA tissue content. 䊳

Ultimately, gene therapy may hold the greatest Fig. 21.1. An 8-year-old boy with cerebral XALD. The T2-
hope for the treatment of XALD patients. One possi- weighted axial images show the characteristic pattern of sym-
ble strategy would be bone marrow ablation followed metrical lesions in the parieto-occipital white matter. The im-
by autologous transplantation with genetically cor- ages show that the lesion contains three zones. The inner and
rected hematopoietic cells of the patient’s own bone more posterior zone has a slightly higher signal than the out-
marrow. Transplantation and oligodendrocytes or er and more anterior zone; there is a rim of low signal in be-
pluripotent stem cells to repair damaged tissue are tween. The contrast-enhanced T1-weighted images confirm
other options. the two zones, the inner and more posterior zone having the
lowest signal, whereas the outer and more anterior zone has a
less low signal. The rim in between shows prominent contrast
21.6 Magnetic Resonance Imaging enhancement. The three zones of the disorder can be visual-
ized this way: the outer and more anterior zone being inactive
In imaging the brain of patients with XALD, different and completely demyelinated,zone 2 showing marked inflam-
patterns can be detected. The most commonly occur- mation and advanced demyelination, and zone 3 representing
ring pattern in cerebral XALD consists of predomi- the start of demyelination. Zones 1 and 3 can usually be distin-
nantly parieto-occipital white matter abnormalities guished on T2-weighted images. The splenium of the corpus
(Fig. 21.1). The lesion starts in the splenium of the callosum is affected and the lateral part of the midbrain as well
corpus callosum and spreads out into the parieto-oc- as the corticospinal tracts in the pons
21.6 Magnetic Resonance Imaging 183

Fig. 21.1.
184 Chapter 21 X-Linked Adrenoleukodystrophy

Fig. 21.2. CT scan of an 8-year-old boy


with cerebral XALD shows calcium
deposits in the lesion

quency of this pattern among patients with cerebral adult patient suffered a severe cerebral contusion in
XALD is about 65–70%. the left temporal area after which the course was
The pattern of cerebral XALD may be reversed, downhill. Demyelination started in the area of the
starting bilaterally in the frontal area with concomi- contusion, and proceeded to affect both hemispheres,
tant involvement of rostrum and genu of the corpus but asymmetry remained. Marked contrast enhance-
callosum (Fig. 21.3). In such cases the anterior limb of ment was seen and extensive inflammation on brain
the internal capsule is involved instead of the posteri- biopsy.
or limb, and the frontopontine brain stem tracts in- In general, patients with the parieto-occipital pat-
stead of the occipitoparietotemporopontine tracts. tern and those with the frontal pattern have rapid dis-
Again, there are three distinguishable zones with con- ease progression if the abnormalities are present at an
trast enhancement of the middle zone. This pattern is early age and if contrast enhancement is present. Pa-
most commonly seen in teenagers and adolescents tients with both frontal and parieto-occipital abnor-
with cerebral XALD. The overall frequency among malities have the most rapid disease progression. Pa-
patients with cerebral XALD is about 15%. tients with predominant involvement of frontopon-
In a few patients, there is concomitant but inde- tine or corticospinal projection fibers or predomi-
pendent frontal and parieto-occipital white matter nant involvement of the cerebellar white matter
involvement. This pattern is rare and occurs mainly generally have much slower disease progression.
in children. The primary finding in AMN consists of spinal
In some patients the frontopontine or corti- cord atrophy. In half of the symptomatic AMN pa-
cospinal projection fibers are involved without in- tients, abnormalities are noted on brain MRI. This
volvement of the periventricular or deep white mat- percentage depends on age: it is lower among the
ter. In this pattern lesions are initially seen in the in- younger AMN patients and higher among the older
ternal capsule and brain stem tracts. With progres- AMN patients. Involvement of corticopontine and
sion of disease abnormalities most often develop in corticospinal projection fibers, described above, is
cerebellar peduncles, the splenium of the corpus cal- most commonly seen. However, AMN patients may
losum, and optic radiation. Contrast enhancement also develop much more extensive white matter ab-
may be present. This pattern is seen in 10–15% of all normalities, involving mainly the parieto-occipital
cerebral XALD patients. It is mainly seen in adult white matter and splenium of the corpus callosum,
XALD patients with clinical signs of AMN (Fig. 21.4), the frontal white matter and anterior part of the cor-
but has also been reported in children with mainly pus callosum, or the cerebellar white matter.
pyramidal signs. Only a few female heterozygotes show abnormali-
Another pattern consists of symmetrical involve- ties on MRI of the brain and spinal cord, even when
ment of cerebellar white matter, middle cerebellar pe- they display an AMN-like clinical phenotype. The
duncles, and brain stem tracts. This pattern is rare pattern of full-blown cerebral demyelination is very
and is mainly seen in adolescents. rare.
In exceptional cases patients have unilateral dis- Apart form its role in diagnostics, MRI has a major
ease initially and bilateral but markedly asymmetri- role in monitoring of the disease (Figs. 21.6 and 21.7).
cal disease in later stages (Fig. 21.5). The presence of Monitoring is an essential part of treatment, first of
two zones in the lesion on unenhanced images and all to determine which patients are eligible for a par-
contrast enhancement of the rim in between these ticular treatment and secondly to monitor treatment
zones are helpful diagnostic clues. One asymptomatic effects. At present, there are two cornerstones in the
21.6 Magnetic Resonance Imaging 185

Fig. 21.3. A boy with cerebral XALD. The first three rows show pontine tracts at the level of the midbrain.The three zones are
images at the age of 9 years. Note the frontal predominance of visible, although less clearly than in Fig. 21.1, with enhance-
the lesions with involvement of the genu of the corpus callo- ment of the middle zone on the T1-weighted images. (Con-
sum and the anterior limb of the internal capsule.The U fibers tinue see next page)
are relatively spared. Also note the involvement of the fronto-

treatment of XALD: dietary treatment and bone mar- indicated for patients in whom cerebral demyelina-
row transplantation. Dietary treatment should be ap- tion is starting, especially young patients without
plied in all asymptomatic patients in an attempt to AMN, but not for patients with a normal MRI or pa-
delay and, hopefully, prevent the onset of clinical tients with entirely stable MRI abnormalities. Loes et
symptoms. Unfortunately, dietary treatment is not an al. (1994 and 2003) developed a 34-point scoring
absolute preventive. Bone marrow transplantation is method for the grading of MRI abnormalities. It has
186 Chapter 21 X-Linked Adrenoleukodystrophy

Fig. 21.3. (continued). A boy with cerebral XALD. The second with spread over almost the entire brain. Only the parieto-oc-
three rows show the follow-up MRI, obtained 1.5 years later. cipital region is partially spared. The cerebellar white matter
The images demonstrate serious progression of the disease remains intact

been found that the Loes score has predictive value. demyelination; in the age group between 7 and 10
With a score higher than 3, almost all patients wors- years, 10% will develop rapidly progressive cerebral
en, irrespective of age; only 10% remain neurologi- demyelination; while in the age group above 10 years,
cally stable.With a score of 1–3, 60% of patients wors- rapidly progressive cerebral demyelination is rare
en, irrespective of age, but survival is longer. If the and AMN is more likely. Contrast enhancement also
Loes score is below 1, prognosis depends on the age of has predictive value: absence of enhancement is asso-
the patient: among patients aged between 3 and 7 ciated with stable disease in 80–85% of the patients,
years, 30% will develop rapidly progressive cerebral whereas clear contrast enhancement is an indication
21.6 Magnetic Resonance Imaging 187

Fig. 21.4. A 45-year-old man with an AMN clinical phenotype. splenium of the corpus callosum, and the posterior limb of the
The T2-weighted images (upper two rows) show the involve- internal capsule. Some enhancement occurs after injection of
ment of the cerebellar white matter, the brain stem tracts, the contrast (third row)

of active inflammatory demyelination and is associat- reflects the three zones of white matter involvement,
ed with progression in 85–90% of the patients. Diffu- the lower ratios being present in the central burnt-out
sion tensor imaging may be more sensitive than con- region. Normal-appearing white matter has a normal
ventional MRI and show abnormalities in areas that magnetization transfer ratio. The findings at MRS of
are not (yet) abnormal on conventional images. The the brain also have predictive value, in particular the
potential role of magnetization contrast imaging in findings just outside the visible lesion. If the N-acety-
the monitoring of XALD patients is presently not laspartate:choline ratio is decreased, this is evidence
clear. The decrease in magnetization transfer ratios for impending demyelination in the area and disease
188 Chapter 21 X-Linked Adrenoleukodystrophy

Fig. 21.5. Adult male patient with cerebral XALD. The MRI of of the right side of the brain. Still the MR characteristics sug-
the first row was obtained when he was 29 years old; the MRI gest cerebral XALD with a zoned lesion, the middle zone hav-
of the second and third rows when he was 31 years old. The ing a low signal on the T2-weighted images. The zone are par-
process started unilaterally and progressed to a bilateral but ticularly prominent on the T1-weighted images
highly asymmetrical disease with predominant involvement

progression. A normal N-acetylaspartate:choline ra- the protocol.As soon as abnormalities are found, a re-
tio provides evidence for a stable situation. For these peat MRI is obtained after 2–3 months to document
reasons, combined MRI and MRS studies every 6 progression. If progression of abnormalities is
months are recommended for asymptomatic XALD demonstrated and the Loes score becomes 3 or high-
boys up to the age of 10 years. If abnormalities are er, bone marrow transplantation is performed as soon
seen on MRI, contrast-enhanced images are added to as possible. Between the ages of 10 and 20 years, the
21.6 Magnetic Resonance Imaging 189

Fig. 21.6. This 18-year-old boy had learning and behavioral pus callosum is involved. The entire cerebral white matter ap-
problems from early on. At the age of 15 years he experienced pears slightly elevated in signal, with a loss of contrast be-
an encephalitis-like episode from which he fully recovered. tween white and gray matter.There is some contrast enhance-
Since then he has been clinically stable with a normal neuro- ment at the rim of the lesion. In view of the static nature of the
logical examination. Between the ages of 15 and 18 years MRI disease, hematopoietic stem transplantation has not been
findings have remained unchanged with frontal white matter considered
abnormalities and white matter atrophy. The genu of the cor-

frequency of MRI and MRS is decreased to once a


year. After bone marrow transplantation, various out-
comes are seen. In some patients MRI abnormalities
regress or disappear; in others the abnormalities be-
come stable. Unfortunately, bone marrow transplan-
tation is not a great success in all patients and further
deterioration may also occur.
190 Chapter 21 X-Linked Adrenoleukodystrophy

Fig. 21.7. This boy is the younger


brother of the patient shown in
Fig. 21.1. He was diagnosed with
the biochemical defect of XALD after
his brother was diagnosed with the
disease. He was monitored by MRI
and MRS every 6 months and normal
results were found (upper row, left).
When he was 6 years old a lesion was
found in the splenium of the corpus
callosum (upper row, right), which had
increased in size after 3 months (mid-
dle row, left). At that time contrast
enhancement of the lesion was found
(middle row, right). Bone marrow trans-
plantation was performed soon after
without major complications.The first
MRI after the procedure showed some
further progression of the lesion, but
the lesion has been stable since and he
is now 9 years old (third row, left).There
is at most minimal contrast enhance-
ment remaining. He is clinically intact
Chapter 22

Refsum Disease

22.1 Clinical Features There is a progressive lowering and loss of deep re-
and Laboratory Investigations flexes. The plantar responses are as a rule flexor or ab-
sent. Over a period of years the muscular weakness
Refsum disease (RD), also called heredopathia atacti- can become widespread and disabling, involving not
ca polyneuritiformis and hereditary sensory and mo- only distal but also proximal musculature. Acute ex-
tor neuropathy type IV, is a rare disorder with an au- acerbations are observed frequently in RD patients.
tosomal recessive mode of inheritance, characterized When these happen, the weakness is not limited to
by accumulation of phytanic acid. The age at onset of distal muscles of the extremities, but proximal mus-
clinical signs and symptoms varies from early child- cles are also paretic and cranial nerves may be in-
hood to the fifth decade. The onset is insidious and volved. Periods of exacerbations may be followed by
may be difficult to determine precisely. Most patients remissions in which the acutely developed weakness
have clear-cut manifestations before the age of 20 disappears.
years. Symptoms may be precipitated by infections Cerebellar ataxia forms the third major manifesta-
and low caloric intake. Dramatic exacerbations and tion of RD. Its onset is later in the course of the dis-
remissions of symptoms may also occur sponta- ease. The ataxia is more marked than can be ex-
neously, without obvious antecedent cause. The main plained by the degree of weakness and sensory loss
clinical features are visual disturbances, peripheral present. Nystagmus may be present as a sign of cere-
polyneuropathy, and cerebellar ataxia. Other fre- bellar dysfunction.
quently observed abnormalities are anosmia, cardiac Cardiomyopathy is present in most patients, as
problems, skin changes, sensorineural deafness, and demonstrated by cardiac enlargement, tachycardia,
skeletal abnormalities. Psychoses, particularly of a conduction disturbances, and ECG abnormalities.
paranoid type, occur more frequently than one would Sudden death is common during acute exacerbations
expect on the basis of coincidence. and is probably caused by cardiac arrhythmias.
Initial visual disturbances include night blindness, Skin changes vary from a dry, scaly skin to a condi-
which may be present for years before the diagnosis is tion of full-blown ichthyosis. The skin abnormalities
established. Gradually, concentric constriction of the change rapidly with the clinical state and are well cor-
visual fields develops and finally only a tubular field related with plasma phytanic acid level. The skin
of vision remains. Central vision may be intact or on- problems tend to be more severe in children than in
ly minimally disturbed for years. In some cases optic adults.
atrophy, cataract, or vitreous opacities occur, con- Bony deformities are frequently present. The skele-
tributing to visual failure and blindness. The fundus- tal manifestations include abnormalities of the
copic appearances are variable, also depending on the metacarpal and metatarsal bones, which may be short
stage of disease. Typical pigmentary retinal degener- or elongated; pes cavus; syndactyly; hammer toes;
ation is rare, and usually pigmentation looks like fine, and epiphyseal dysplasia of the shoulders, elbows,
small granules or has a salt-and-pepper appearance. and knees.
In the majority of the patients pigmentation occurs in Renal dysfunction with amino aciduria and hy-
the peripheral retina; macular pigmentation is excep- pokalemia may occur.
tional. In rare cases, no pigmentary degeneration is The signs and symptoms of RD can be divided ac-
seen on funduscopy. The pupils are often small, and cording to their liability to rapid change following
reaction to light, convergence, and accommodation is changes in blood phytanic acid levels. Bony abnor-
often minimal or absent. malities are stationary. Visual disturbances, anosmia,
Chronic progressive polyneuropathy is the second and sensorineural deafness develop gradually, do not
major manifestation of RD. The visual symptoms worsen rapidly on increasing phytanic acid levels,
sometimes precede the polyneuropathy by several and do not improve on lowering of phytanic acid lev-
years. When the polyneuropathy develops gradually, els; only stabilization at the same level of impairment
it is usually symmetrical and initially distal, causing can be achieved by low blood phytanic acid levels. Pe-
muscular weakness, atrophy, cutaneous hypesthesia, ripheral neuropathy, cerebellar ataxia, and cardiomy-
painful paresthesia, and disturbed position sense. Pe- opathy are slowly progressive but liable to deteriorate
ripheral nerves are sometimes palpably enlarged. rapidly if phytanic acid levels rise. The component of
192 Chapter 22 Refsum Disease

rapid deterioration responds well to lowering of phy- In the brain stem variable demyelination occurs
tanic acid levels, while the more chronic component mainly affecting the pontocerebellar tracts, medial
responds slowly and often incompletely. Skin abnor- lemniscus, olivocerebellar tracts, cerebellar pedun-
malities are closely related to actual blood phytanic cles, and corticospinal tracts. Axons are relatively
acid levels. spared. A variable number of fat-filled macrophages
The clinical diagnosis of RD is not easy as no and hypertrophic astrocytes are present. Diffuse loss
symptom is pathognomonic of the disease and the of neurons in the inferior olivary nuclei is commonly
various signs and symptoms may develop in succes- observed.
sion at different times. The demonstration of exces- The cerebellar cortex is usually normal, although
sive amounts of phytanic acid in serum is the most some neuronal loss may occur. The white matter
valuable aid in the diagnosis of RD. Phytanic acid is within and surrounding the dentate nucleus is affect-
not normally present in detectable amounts, and the ed. Lipid-laden macrophages and hypertrophic astro-
absence of phytanic acid in the serum of an untreated cytes are present. Considerable loss of neurons is pre-
patient suspected of suffering from RD makes the sent within the dentate nucleus.
diagnosis highly improbable. In fact, so far no false In the leptomeninges surrounding the spinal cord
negatives have been reported. However, elevated phy- large amounts of fat are present. Sudanophilic gran-
tanic acid is not specific for the disease, since it may ules of variable size are deposited in endothelial cells,
also accumulate in several other genetic diseases, histiocytes, and macrophages. Within the spinal cord
including peroxisomal biogenesis defects and rhizo- there is marked demyelination of the posterior
melic chondrodysplasia punctata type 1. columns and demyelination of less severity in both
The protein in CSF is increased to levels between 1 spinocerebellar tracts. Retrograde changes and loss of
and 7 g/l or even higher. The cell count is normal. motor neurons are observed in the anterior horns at
Neurophysiological studies show a greatly reduced all levels. In the spinal roots, especially at the level of
motor and sensory nerve conduction velocity and the cauda equina, severe demyelination is observed
signs of denervation and reinnervation in the EMG. together with complete destruction of some axons
The nerve conduction velocity improves in conjunc- and axonal swellings. Onion-bulb formations are pre-
tion with clinical improvement. The ERG shows ab- sent.
sent or reduced reaction. ECG may reveal a prolonged The peripheral nerves, both somatic and autonom-
QT segment and a widened QRS complex. When the ic, show macroscopic thickening. The changes in the
urinary sediment of a patient with RD is stained for peripheral nerves are constant, but their intensity
lipids, large amounts of fatty material can be detect- varies greatly between cases. Nerve hypertrophy is
ed. usually most conspicuous in the lumbar and brachial
A deficiency of phytanic acid a-oxidation can be plexuses. The hypertrophy is often irregular, forming
shown in cultured fibroblasts. In RD patients the rate localized swellings. Histological study of these
of a-oxidation of phytanic acid is less than 5% of nor- swellings indicates that myelinated nerve fibers are
mal, whereas in parents of RD patients the oxidation reduced in number, and Schwann cell processes have
rate is about 50% of normal, indicating a heterozy- given rise to typical onion-bulb formations. Many un-
gous state. Phytanic a-oxidation activity can be deter- myelinated axons are present within the onion bulbs.
mined in cultured amniotic cells, allowing prenatal The myelin sheaths are often abnormally thin and of
diagnosis. DNA-based prenatal diagnosis is possible unequal thickness, and segmental demyelination has
in families in which the mutations responsible have been found. Axonal destruction is also present. In
been identified. some onion bulbs the whorls are closely packed, but
in others they are more loosely disposed and separat-
ed by large extracellular spaces of variable width. In
22.2 Pathology the cytoplasm of the whorl-forming Schwann cells,
several types of inclusion can be seen with help of
In RD the site of major involvement is the PNS, electron microscopy: large crystalline inclusions and
whereas the CNS shows more subtle abnormalities. rounded osmiophilic bodies which are probably lipid
On gross examination of the brain, the leptomeninges in nature.
appear thickened and the cerebellum may appear In the kidney, fat is accumulated in large amounts
moderately atrophic. On microscopic examination, in the epithelial cells of the convoluted tubules. In the
deposition of fat is noted in the leptomeninges, liver, fat is stored in mesenchymal and parenchymal
ependymal cells, choroid plexus epithelium, and cells cells. There are no or only slight signs of fibrosis. Per-
of the globus pallidus. The cerebral hemispheres are oxisomes are morphologically normal in RD.
otherwise intact with normal cytoarchitecture of the
cortex.
22.4 Pathogenetic Considerations 193

22.3 Chemical Pathology family showed linkage to the PAHX locus on chromo-
some 10p13 (Tranchant et al. 1993; Nadal et al. 1995).
RD is a disorder of phytanic acid metabolism. Phy- In another patient with clinical findings of mental
tanic acid is a C20 multibranched fatty acid. It has retardation and abnormally short metatarsals and
been isolated in the brain and has been found to be metacarpals, but no other signs of classic RD, both
present in much larger amounts in the white matter phytanic acid and pipecolic acid were elevated
than in the gray matter. The presence of substantial (Baumgartner et al. 2000). Liver peroxisomes were
portions of phytanic acid can be demonstrated with- reduced in number, had a larger average size, and
out any appreciable alteration in total amounts of fat. lacked catalase. This patient was homozygous for a
Analysis of the lipid composition of the CNS reveals frameshift mutation in PAHX (Baumgartner et al.
that the proportions of the remaining lipids are near- 2000).
ly normal. Phytanic acid is mainly present in phos- RD is genetically heterogeneous and some patients
phoglycerides, with higher proportions of phytanic have mutations in the gene PEX7, located on chromo-
acid in the choline phosphoglycerides than in the oth- some 6q21–22.2. Mutations in the latter gene are
er phosphoglycerides. Lower concentrations are pre- known to be associated with rhizomelic chondrodys-
sent in the galactolipids. Phytanic acid accumulates plasia punctata (RCDP). This is another peroxisomal
mainly in myelin and is found in higher concentra- disorder characterized by proximal shortening of the
tions in the choline phosphoglycerides from myelin extremities, contractures, growth failure, cataracts,
than from gray matter or whole white matter. The mental retardation, and usually early death. RCDP is
proportions of phytanic acid in peripheral nerve genetically heterogeneous with three distinct forms.
myelin are even higher. A defect in the de novo plasmalogen biosynthesis is
In several organs other than the nervous system the central biochemical feature of RCDP. Only RCDP
there are accumulations of neutral lipids, especially in type 1, which is caused by mutations in PEX7, is also
liver and spleen. A high proportion of the fatty acids characterized by elevated phytanic acid. Mutations in
consists of phytanic acid, incorporated in cholesterol PEX7 lead to a defective PTS2 receptor, resulting in
esters and triglycerides. The heart muscle and blood defective import of peroxisomal proteins with a PTS2
also contain a large amount of phytanic acid. targeting signal. Phytanoyl-CoA hydroxylase, alkyl-
dihydroxyacetonephosphate synthase, and peroxiso-
mal thiolase are the three peroxisomal enzymes
22.4 Pathogenetic Considerations with a PTS2 targeting sequence. Alkyl-dihydroxyace-
tonephosphate synthase is involved in plasmalogen
Phytanic acid is a C20 multibranched fatty acid: biosynthesis. In patients with a defect in PEX7, phy-
3,7,11,15-tetramethylhexadecanoic acid. The com- tanoyl-CoA hydroxylase, alkyl-dihydroxyacetone-
mon pathway for fatty acid degradation is formed by phosphate synthase, and peroxisomal thiolase are
the b-oxidation pathway in mitochondria and perox- deficient. Patients with RCDP types 2 and 3 have an
isomes. The presence of a 3-methyl group in phytan- isolated deficiency of either alkyl-dihydroxyace-
ic acid prevents it from undergoing b-oxidation. In- tonephosphate synthase or dihydroxyacetonephos-
stead, phytanic acid undergoes a-oxidation to pris- phate acyltransferase, both resulting in a defect in
tanic acid in peroxisomes and pristanic acid is further plasmalogen biosynthesis. While patients with muta-
degraded by b-oxidation. Phytanic acid is converted tions in PHYH have an isolated phytanic acid a-oxi-
into phytanoyl-CoA by acyl-CoA synthase. Phy- dation deficiency, patients with mutations in PEX7
tanoyl-CoA hydroxylase, a peroxisomal matrix pro- have a deficiency of both phytanic acid a-oxidation
tein, catalyzes the conversion of phytanoyl-CoA into and de novo plasmalogen biosynthesis. It is remark-
2-hydroxyphytanoyl-CoA, which is subsequently able that mutations in PEX7 can give rise to both
converted into pristanal, pristanic acid, and finally RCDP and RD.
pristanoyl-CoA, a substrate for further b-oxidation. The genetic heterogeneity of RD is further empha-
The basic defect in RD is a defect in a-oxidation of sized by the finding of a 2-methylacyl-CoA racemase
phytanic acid in peroxisomes. In about 50% of the RD deficiency in some patients with retinitis pigmentosa
patients the defect involves the enzyme phytanoyl- and a late-onset sensory neuropathy (Ferdinandusse
CoA hydroxylase, resulting in an excessive accumula- et al. 2000). In these patients both phytanic acid and
tion of phytanic acid in blood and various tissues. pristanic acid are elevated, in contrast to true RD pa-
The gene encoding this enzyme, PHYH or PAHX, is tients in whom only phytanic acid is elevated.
located on chromosome 10p13. Many different muta- Phytanic acid is of exogenous, dietary origin.
tions have been identified in RD patients. A number There is no evidence of any endogenous synthesis. It
of atypical RD cases have been related to a defect in is consumed by humans in considerable quantities in
the same gene. In one RD family, not only phytanic dairy products, ruminant animal fats and meats, and
acid, but also pipecolic acidemia was elevated. This certain fish. Phytol can be metabolized by humans to
194 Chapter 22 Refsum Disease

phytanic acid, but most phytol in the diet, present in During conditions of increased mobilization of body
green vegetables, is bound to chlorophyll and little is fat, phytanic acid is liberated from body stores and
absorbed. Hence, the contribution of phytol to the the serum concentration increases even when intake
phytanic acid concentration in humans is low. In view of phytanic acid is low. During periods of signifi-
of the large intake and the presence of only trace cant weight loss and catabolic conditions related to
amounts in normal human tissue, the catabolic path- febrile infections, serious worsening of neurological
way that disposes of phytanic acid must be very effi- or myocardial pathology may occur, even resulting in
cient. death.
The accumulation of large amounts of this unusu- Strict diet and supply of sufficient energy to main-
al fatty acid in various tissues and lipid classes in RD tain a near-constant body weight lead to a gradual
is probably the cause of a series of symptoms, both drop in serum phytanic acid levels to normal or
acute and chronic. Phytanic acid is readily incorpo- slightly increased levels. Treated RD patients show
rated into phospholipids. It is probable that the incor- clinical improvement. They no longer have apparent-
poration of phytanic acid into lipids of the myelin ly spontaneous relapses. The improvement in partic-
sheaths leads to a less stable myelin structure. The ular relates to the signs and symptoms that change
methyl branching of phytanic acid probably disrupts rapidly: neuropathy, cerebellar ataxia, cardiomyopa-
the packing of the hydrocarbon tails in the bimolecu- thy, and ichthyosis. Problems such as visual distur-
lar lipid leaflet due to steric hindrance, and in this way bances, anosmia, and deafness usually stabilize and
destabilizes the myelin. Once phytanic acid levels do not deteriorate any further. Some clinical prob-
reach a critical point, myelin dissolution occurs. The lems are due to irreversible damage and are not influ-
phytanic acid levels are higher in peripheral nerve enced favorably by lowering phytanic acid levels. Ear-
myelin than in CNS myelin and demyelination starts ly diagnosis and treatment are important to prevent
earlier and is more severe in the PNS than in the CNS. irreversible handicaps. The advice to ensure sufficient
A question is why the concentration of phytanic acid energy provision applies particularly at the start of
is higher in myelin of the PNS than in myelin of the treatment, when the serum phytanic acid levels are
CNS. The turnover rate of myelin is higher in periph- still very high. Later, when the phytanic acid content
eral nerves than in brain and spinal cord. Phytanic of the serum has been reduced to safer levels, a grad-
acid may, therefore, accumulate more easily in the ual weight reduction is acceptable. Dietary treatment
peripheral nerves. Another possibility is that the is life-long.
blood–nerve barrier in the PNS is less restrictive than Plasmapheresis provides the means to lower
the blood–brain barrier in the CNS, allowing a more serum phytanic levels rapidly. Very high serum levels
rapid increase in the phytanic acid content of periph- are toxic and may precipitate life-threatening symp-
eral nerves. The distribution of demyelination in the toms. In these circumstances, plasmapheresis can
CNS, with preferential involvement of brain stem and be used as an emergency measure. Furthermore,
cerebellum, remains to be explained. plasmapheresis has been shown to be an excellent
Phytanic acid is an effective regulator of gene ex- supplement to dietary treatment. When applied with
pression by virtue of its capacity to activate certain a low frequency, it can compensate for a less strict
nuclear receptors affecting nuclear signal transduc- dietary regimen. Dialysis is ineffective, as plasma
tion pathways. It is likely that this effect is important phytanic acid is bound to lipoproteins.
in the pathophysiology of the disease in RD.

22.6 Magnetic Resonance Imaging


22.5 Therapy
There are no reports on MRI findings in RD, probably
As the origin of phytanic acid is exclusively exoge- because of the rarity of the disease and the fact that
nous, further accumulation of the substance can be the site of major involvement is the PNS rather than
prevented by a strict diet that is low in phytanic acid. the CNS. In relation to histopathological findings,
There is no need to restrict phytol. Furthermore, the MRI signs of demyelination can be expected to occur
diet should contain enough energy to prevent consid- mainly in brain stem tracts, cerebellar peduncles, and
erable weight loss.An appreciable quantity of phytan- the cerebellar white matter.
ic acid is present in hepatic lipid and body fat stores.
Chapter 23

Mitochondria and Mitochondrial Disorders


E. Morava, J.A.M. Smeitink

23.1 Mitochondrial Structure The pyruvate dehydrogenase complex is located at


and Function the inner border of the inner mitochondrial mem-
brane.
Mitochondria are membranous organelles that pro- Fatty acids with more than eight or ten carbon
vide the energy necessary for the different cell func- atoms require a specific carrier system to enter the
tions. They are called mitochondria because of their mitochondrial matrix space (Fig. 23.1). They are acti-
threadlike appearance (Greek mitos = thread) on light vated to acyl-CoA by acyl-CoA ligase (= acyl-CoA
microscopy. On electron microscopy they appear as synthetase) on the mitochondrial outer membrane.
vesicles bounded by two membranes. The inner Fatty acyl-CoA is subsequently converted to fatty acyl
membrane is thrown into folds that project like carnitine in the intermembrane space by carnitine
shelves into the mitochondria. These projections are palmitoyl transferase 1 (CPT 1), which is located in
called cristae. Mitochondria consist of four compart- the outer mitochondrial membrane. Fatty acyl carni-
ments: the outer membrane, the intermembrane tine is then translocated across the inner mitochon-
space, the inner membrane, and the mitochondrial drial membrane in exchange for free carnitine, a reac-
matrix. Mitochondria vary considerably in size in any tion catalyzed by carnitine:acylcarnitine translocase,
one cell type, but most have a diameter of between 0.1 located in the inner mitochondrial membrane. On the
and 1.0 mm. In different cell types the size, shape, and inner surface of the inner membrane a second carni-
number of cristae vary considerably. Most cells con- tine palmitoyl transferase, CPT 2, converts fatty acyl
tain many mitochondria, the actual number differing carnitine to acyl-CoA and free carnitine. Shorter-
in relation to the energy requirements of the type of chain fatty acids (ten or fewer carbons) enter the mi-
cell. Mitochondria are dynamic organelles. They un- tochondria independently of the carnitine-requiring
dergo frequent fission and fusion or branching, and transport system, and are activated by short- and
they may alter their size and location in the cell under medium-chain acyl-CoA ligase to the respective acyl-
different conditions. CoA esters in the mitochondrial matrix. Acyl-CoA is
The main role of mitochondria is to synthesize the primary substrate for mitochondrial b-oxidation.
adenosine triphosphate (ATP), the universal source of This b-oxidation spiral involves four successive reac-
energy for the cell. Mitochondria convert the energy tions, mediated by acyl-CoA dehydrogenase, 2-enoyl-
derived from oxidation of substrates into the high- CoA hydratase, 3-hydroxyacyl-CoA dehydrogenase,
energy bond of ATP, which is then transported into and 3-ketoacyl-CoA thiolase. The products of each
the cytosol in exchange for adenosine diphosphate turn of the spiral are acetyl-CoA and acyl-CoA, now
(ADP). The process of production and storage of en- two carbons shorter, with conversion of flavin ade-
ergy by mitochondria is called oxidative phosphoryla- nine dinucleotide (FAD) to FADH2 and NAD+ to
tion. In addition to this process, mitochondria also NADH + H+. An acyl-CoA can recycle through b-oxi-
perform many other functions, including the first two dation spirals as many times as it can yield acetyl-
reactions of the urea cycle, the synthesis of ketone CoA fragments. With each turn of the spiral, as the
bodies, and propionate metabolism. acyl-CoA becomes shorter, it encounters enzymes
Pyruvate and fatty acids are the most important with different substrate specificities. An example of
substrates for energy production, although amino this is the series of chain-length-specific acyl-CoA
acids may also contribute in certain conditions, for dehydrogenases: long-chain acyl-CoA dehydrogenase
instance during fasting. mediates the reaction for acyl-CoA compounds from
Pyruvate represents the metabolic end point of 8 carbons to 18 carbons; medium-chain acyl-CoA de-
glycolysis, which occurs in the cytoplasm and yields a hydrogenase from 4 to 12 carbons; and short-chain
small amount of ATP. Pyruvate is carried across the acyl-CoA dehydrogenase from 4 to 6 carbons. Fatty
mitochondrial membrane into the mitochondrial acids with double bonds require additional enzymes,
matrix space by monocarboxylate translocase. Subse- d3, d2-enoyl-CoA isomerase and 2,4-dienoyl-CoA re-
quently it is oxidatively decarboxylated by the pyru- ductase. In the liver, the metabolic end product of
vate dehydrogenase complex (PDHc), which produces each cycle of b-oxidation, acetyl-CoA, is primarily
CO2 and acetyl-CoA, while reducing one nicoti- converted to ketone bodies, while in other tissues
namide adenine dinucleotide (NAD+) to NADH + H+. acetyl-CoA is completely oxidized via the citric acid
196 Chapter 23 Mitochondria and Mitochondrial Disorders

Fig. 23.1. Import and b-oxidation of fatty acids in mitochondria

Fig. 23.2. Citric acid cycle in the mitochondrial matrix


23.1 Mitochondrial Structure and Function 197

Fig. 23.3. Electron (e) transport chain in mitochondrial inner membrane

cycle. Electrons from the acyl-CoA dehydrogenase- reductase) oxidizes NADH to NAD+ and the electrons
mediated reactions are transferred to electron trans- of the hydrogen are transferred to ubiquinone (CoQ)
fer flavoprotein to the electron transport chain. to yield ubiquinol (reduced CoQ). Complex II (succi-
Therefore, acetyl-CoA is produced from different nate ubiquinone reductase) accepts reducing equiva-
sources. Carboxylation of pyruvate yields oxalo- lents from succinate and also passes electrons down
acetate and decarboxylation of pyruvate yields the chain to ubiquinone to yield ubiquinol. The elec-
acetyl-CoA. Acetyl-CoA is produced in the b-oxida- trons from ubiquinol are transferred to complex III
tion of fatty acids. It can also be derived from break- (ubiquinol: cytochrome-c reductase), subsequently to
down of certain amino acids (for instance, leucine cytochrome c, then to complex IV (cytochrome-c ox-
and isoleucine). Acetyl-CoA and oxaloacetate con- idase), and finally to oxygen, which combines with
dense to form citrate. Citrate is decarboxylated in the protons to form water. The energy released in the
citric acid cycle (also called Krebs cycle, tricarboxylic electron transport is used to pump hydrogen ions out
acid cycle, or TCA cycle, see Fig. 23.2), yielding CO2 of the mitochondrial inner membrane through com-
and reducing equivalents in the form of NADH + H+ plexes I, III, and IV. The resulting electrochemical gra-
and FADH2. Certain amino acids can be converted dient is exploited by complex V (ATP synthetase =
to citric acid cycle intermediates, such as glutamate, ATPase). Complex V allows some of the protons to
aspartate, alanine, proline, and glutamine. flow back into the mitochondria and uses the energy
The reducing equivalents NADH and FADH2, pro- so generated for the synthesis of ATP from ADP and
duced by b-oxidation of fatty acids and the citric acid inorganic phosphate. ATP formed in the matrix space
cycle, are reoxidized to NAD+ and FAD by the respira- is then transported out of the mitochondria by ade-
tory chain. This oxidation sequence is tightly coupled nine nucleotide translocase in exchange for ADP. The
to the phosphorylation of ADP to yield ATP. This production and storage of energy by mitochondrial
process of so-called oxidative phosphorylation is re- oxidative phosphorylation is a very efficient process
sponsible for the main production of ATP in most with a high ATP yield. The rate of mitochondrial sub-
cells. strate oxidation is finely geared to the needs of the
Oxidative phosphorylation (Fig. 23.3) is achieved cell. The main control mechanism is the ratio of ATP
by five multisubunit enzyme complexes (complexes to ADP. ADP is an activator of mitochondrial respira-
I–V), all located within the mitochondrial inner tion.
membrane. Complexes I–IV constitute the electron Each complex of the respiratory chain is made up
transport chain (or respiratory chain). Complex I of a number of protein components. Complex I con-
(NADH ubiquinone reductase = NADH-coenzyme Q tains 46 polypeptides and has as prosthetic groups
198 Chapter 23 Mitochondria and Mitochondrial Disorders

flavin mononucleotide and several nonheme functions. The genetic code used in the mitochondria
iron–sulfur clusters. Complex II consists of 4 sub- differs from the universal code, making nDNA and
units. Complex III is composed of 11 subunits includ- mtDNA reciprocally untranslatable. The rate of spon-
ing cytochrome b, cytochrome c1, and a nonheme iron taneous mutations of mtDNA genes is much higher
protein. Complex IV is composed of 13 different than that of nDNA genes and repair mechanisms are
protein units, 2 cytochromes (a and a3) and two limited and less efficient.
copper atoms. Complex V is composed of 16 different The entire mitochondrial genome of each individ-
polypeptides. Two small electron carriers, ubi- ual, either male or female, is inherited from the moth-
quinone (coenzyme Q10) and cytochrome c (a low er, although the possibility is not excluded that a
molecular weight hemoprotein) act as shuttles be- small contribution of the mitochondrial genotype
tween the complexes. may be of paternal origin. This is due to the fact that
during egg fertilization the sperm cell contributes al-
most no cytoplasm to the zygote.
23.2 Mitochondrial Genetics The replication and expression of mtDNA are con-
trolled by nuclear genes. The nuclear genome further-
Human cells possess two different genomes: nuclear more encodes for most of the mitochondrial proteins
DNA (nDNA), a 3¥109-base-pair-long genome, pre- and cooperates with the mitochondrial genome in the
sent in two copies in each cell, and mitochondrial assembly of the multisubunit enzyme complexes of
DNA (mtDNA), a 16,569-base pair-long genome, pre- the oxidative phosphorylation apparatus. The D-loop
sent in 2–10 copies per mitochondrion. Mitochondria region is an important area of interaction between
are dependent upon the coordinated expression of nDNA and mtDNA.
these two parallel genetic systems. The structural proteins of the oxidative phospho-
Each mitochondrion contains on average five mi- rylation (complexes I–V) are all dual-coded, except
tochondrial genomes and each cell contains hundreds for complex II. Most mitochondrial membrane and
to thousands of mitochondria. Consequently, there matrix proteins are coded by nuclear genes and syn-
are hundreds or thousands of copies of mtDNA in thesized in the cellular cytoplasm on free polyribo-
each cell. In normal individuals all of these copies of somes and have to be imported from the cytoplasm
mtDNA are identical (homoplasmy), but in disease into the mitochondria through a complicated translo-
there may be more than one distinct population of cation machine, which is under the control of the nu-
mtDNA (heteroplasmy), one being normal and the clear genome. In addition, nDNA encodes several fac-
other being mutant. During cell division mitochon- tors controlling mtDNA replication, transcription
dria (and mtDNA) are randomly distributed between and translation.
daughter cells. As a consequence, the proportion of Most nuclear-encoded mitochondrial proteins
mutant genomes may shift in daughter cells. destined for the inner three mitochondrial compart-
mtDNA consists of two complementary strands: ments (the intermembrane space, the inner mem-
one filament is rich in guanine nucleotide residues, brane, and the mitochondrial matrix) are synthesized
while the other is rich in cytosine residues. They are as larger precursors containing an amino-terminal
conventionally called heavy (H) and light (L) strands, extension (presequence). The presequences are the
respectively. mtDNA contains only 37 genes and targeting signals of the precursor proteins and direct
codes for different proteins of the respiratory chain these proteins to mitochondria. The presequences
and the RNAs (transfer and ribosomal RNAs) neces- consist of 20–80 amino acid residues. There is no ap-
sary for expression of these genes (13 messenger parent sequence identity among the different mito-
RNAs [mRNAs], 22 transfer RNAs [tRNAs], and 2 ri- chondrial protein presequences, but a characteristic
bosomal RNAs [rRNAs]). The 13 mRNAs specify as common to all of them is a relatively high content
many polypeptides of the respiratory chain: 7 sub- of positively charged and nonpolar residues and
units of complex I, the cytochrome b subunit of com- an almost complete absence of negatively charged
plex III, 3 subunits of complex IV and 2 subunits of residues. This finding suggests that the specificity of
complex V. the presequences resides in a structural configuration
In mtDNA, the gene organization is highly com- rather than a particular biochemical motif. Most of
pact; all of the coding sequences are contiguous with the presequences probably adopt an a-helical struc-
each other and there are no introns. The only noncod- ture. Most mitochondrial proteins, in particular ma-
ing stretch of DNA is the displacement loop (D loop), trix proteins, contain their targeting signal in a prese-
a region of 1123 base pairs that contains the origin of quence, but there are a few exceptions. A few proteins
replication of the H strand and the promoters for do not have presequences and apparently contain a
L- and H-strand transcription. The D loop is an im- targeting signal in their mature form.
portant area of interaction of mtDNA with nuclear- Presequences are recognized by specific receptors
encoded proteins regulating mtDNA housekeeping on the mitochondrial surface (a special class of mito-
23.3 Mitochondrial Disorders 199

chondrial outer membrane or MOM proteins). The dent on the membrane potential. In addition to
receptors directly interact with another protein, the VDAC, other types of channels are present in the out-
general insertion protein (GIP), and thereby donate er membrane. Probably the outer membrane has sev-
the precursor proteins to this membrane insertion eral permeability pathways differing in selectivity,
site. The further transport of precursor proteins oc- regulation, and function.
curs through contact sites, where the mitochondrial
outer and inner membranes are closely apposed. The
inner membrane has a translocation channel distinct 23.3 Mitochondrial Disorders
from, but in dynamic interaction with, the transloca-
tion channel of the outer membrane. Import across Mitochondrial disorders are the most common in-
the inner mitochondrial membrane requires ATP and born errors of metabolism, with an estimated inci-
the presence of a membrane potential across this dence of 1 per 10,000 live births. They present as an
membrane. extremely heterogeneous group of disorders with
The configuration of proteins is important in the variable age of onset, progression, and severity. Con-
process of import across the mitochondrial mem- sidering the central role of mitochondria in cellular
brane. Completely folded polypeptides are unable to metabolism, it is not surprising that mitochondrial
traverse the membrane. Premature folding in the cel- diseases are often multiorgan disorders with predom-
lular cytoplasm of the precursor proteins is prevent- inant involvement of brain and muscles. Mitochondr-
ed by stabilization of the proteins in a translocation- ial disorders can follow either a maternal, autosomal,
competent, unfolded conformation with help of so- or X-linked mode of inheritance.
called molecular chaperones. Molecular chaperones Many mitochondrial disorders follow maternal in-
do not form part of the final folded or assembled pro- heritance. A mother carrying a mtDNA mutation will
tein structure but prevent nondesired folding and in- transmit it to all her children, male and female, but
teractions of the substrate protein along its transport, only her daughters will pass it on to their children. At
folding, and assembly pathways. Appropriate peptide a clinical level maternal transmission may be difficult
folding is attained after the chaperone has released to detect. Due to heteroplasmy, unequal mitotic segre-
the polypeptide substrate. The majority of the cur- gation, and threshold effect, different individuals in
rently identified molecular chaperones belong to the the matrilinear lineage may differ in symptomatology
class of so-called “heat shock proteins.” A different and in organ involvement; some may even be asymp-
kind of molecular chaperones involved in mitochon- tomatic. In most cases of mitochondrial genomic de-
drial precursor proteins consists of the so-called “pre- fects, heteroplasmy is present, with occurrence of
sequence binding factors.” These proteins, through both wild type and mutant mtDNA. At cell division
their interaction with the presequences of mitochon- mitochondria and mtDNA are haphazardly distrib-
drial precursors, are also essential in the import path- uted between the daughter cells and consequently the
way of proteins into mitochondria. proportion of mutant genomes may differ between
After import across the mitochondrial membrane, daughter cells. The percentage of mutant DNA versus
presequences are proteolytically removed in the mi- normal DNA may, therefore, be very different in dif-
tochondrial matrix. This cleavage is necessary for fur- ferent children of the same mother, and in different
ther assembly of the newly imported polypeptides in- tissues in the same person, and may alter in the course
to functional proteins and complexes. Two different of time. Whether or not the mtDNA mutation is actu-
proteins are required for full protease activity: mito- ally expressed is largely determined by the relative
chondrial processing peptidase (MPP) and process- proportion of normal versus mutant genomes in a
ing enhancing protein (PEP). The MPP component given tissue. A minimum critical number of mutant
contains the catalytic activity, which is stimulated by DNA is necessary to impair energy metabolism se-
PEP. After that, the proteins are sorted to their respec- verely enough to cause dysfunction of that particular
tive mitochondrial subcompartments. organ or tissue. This phenomenon is known as the
The outer and inner mitochondrial membranes threshold effect. The number of affected mitochon-
differ in permeability. The transport of metabolites dria and cells needed to cause organ dysfunction
and inorganic ions across the inner membrane is varies from tissue to tissue depending on the vulner-
highly regulated. This relative impermeability is im- ability of that particular tissue to impairments of ox-
portant in maintaining a membrane potential and pH idative phosphorylation. The relative reliance of tis-
gradient across the membrane necessary for oxida- sues on oxidative phosphorylation energy decreases
tive phosphorylation. The permeability of the outer in the following order: CNS, skeletal muscle, heart,
membrane is less restricted. The outer membrane kidney, and liver. The presence of a mutation in a par-
contains pore-forming proteins called porins or volt- ticular percentage of mitochondrial genomes may
age-dependent, anion-selective channels (VDAC). lead to signs of encephalopathy and/or myopathy,
Whether the pore is open or partially closed is depen- without any sign of dysfunction of other organs. The
200 Chapter 23 Mitochondria and Mitochondrial Disorders

metabolic vulnerability may also vary in the same tis- 3. Disorders with indirect involvement of the respi-
sue with time and according to functional demands. ratory chain
As the proportion of mutant mitochondrial genomes (a) Defects of mitochondrial protein importation
may shift in daughter cells, this may also be the cause (b) Defects in mitochondrial motility
of a change in phenotype. In general, there is a decline (c) Neurodegenerative disorders
in oxidative phosphorylation capacity with age. The
most likely mechanism for this phenomenon is the For the clinician, the genetic origin of the mitochon-
accumulation of damage to mtDNA in the face of in- drial defect is the most important item in the classifi-
sufficient ability to repair DNA alterations. This phe- cation, recognizing two major groups: mtDNA defects
nomenon may explain the late age of onset of clinical (point mutations in structural proteins of the respira-
signs and symptoms in some patients, and the in- tory chain, point mutations in tRNAs and rRNAs, and
crease in severity of the disease with age. major mtDNA rearrangements) and nDNA defects
The mitochondrial genome depends heavily on the (defects in structural oxidative phosphorylation pro-
nuclear genome, which encodes several factors in- teins, defects in genes encoding assembly factors, de-
volved in mtDNA replication, transcription, and fects in genes involved in the biogenesis of the oxida-
translation. Faulty communications between nuclear tive phosphorylation system, and defects in the main-
and mitochondrial genomes may lead to either multi- tenance of mtDNA).
ple mtDNA deletions or mtDNA depletion. These dis- In a simplified view, a positive family history sug-
orders are transmitted by mendelian inheritance, be- gestive of maternal inheritance may help in the early
cause the primary genetic defect resides in nDNA. diagnosis in mtDNA defects, but the clinical presenta-
The nuclear genome also contains the genes encoding tion may be extremely variable in different family
structural mitochondrial proteins, most of the respi- members. In many affected individuals the disease
ratory chain proteins, and the proteins necessary for becomes evident only in early adulthood and may be
the proper assembly of the oxidative phosphorylation organ-specific. Mitochondrial deletions, however, are
complexes. most often sporadic, the symptoms may appear early,
The classification of these pheno- and genotypi- and the family history is negative.
cally heterogeneous diseases is very difficult. Classifi- In nDNA defects, the family history is noncontrib-
cation may be based on the clinical presentation, on utory in most cases, the parents are healthy, the symp-
the age of presentation and possible progression, or toms frequently arise soon after birth, and when pro-
on the underlying biochemical or genetic defects. gressive, the disease may be fatal in early childhood.
A previous classification by DeVivo (1993) is based on Sibs may be affected to a variable degree (genetic and
a genetic framework. The major subdivision is into environmental factors), but the clinical variability is
nDNA defects, mtDNA defects, and intergenomic usually much less marked than in mtDNA defects.
signaling defects.
This original classification developed further with
time, e.g., with the description of defects in assembly 23.3.1 Mitochondrial DNA Defects
factors, defects in mtDNA maintenance (replication
control), and defects in oxidative phosphorylation A variety of defects in mtDNA can be distinguished:
system biogenesis. The classification of DiMauro and point mutations, deletions, and duplications. The DNA
Schon (2003) has incorporated these recent insights: defects may involve genes coding for proteins of the
1. Respiratory chain disorders due to defects in respiratory chain (structural proteins) or genes cod-
mtDNA ing for tRNA or rRNA. mtDNA depletion is related to
(a) Mutations in protein synthesis genes a nDNA defect and is included under nuclear defects
(b) Mutations in protein coding genes of mtDNA maintenance.
2. Respiratory chain disorders due to defects in In cases of mutations involving genes encoding
nDNA structural proteins of the respiratory chain, biochemi-
(a) Mutations in structural components of the res- cal analysis reveals a defect restricted to one respira-
piratory chain tory enzyme complex (complex I, II, II, IV, or V). In
(b) Mutations in ancillary proteins of the respira- such patients the primary biochemical analysis,
tory chain demonstrating a usually partial isolated complex de-
(c) Defects in intergenomic signaling affecting ficiency, leads to targeted mutation analysis of partic-
respiratory function ular mitochondrial structural genes. Different point
(d) Defects of the membrane lipid milieu mutations in genes coding for components of com-
(e) Defects in mitochondrial motility, fusion, and plex I have been observed in exercise intolerance,
fission Leber hereditary optic neuropathy (LHON), Leigh
23.3 Mitochondrial Disorders 201

syndrome, Leigh-like syndrome, progressive epilepsy, analysis reveals decreased activity of complexes I, III,
adult-onset dystonia associated with neurological and IV, individually or in combination. The percent-
and ophthalmological symptoms, and in mitochon- age of deleted mtDNA is similar in muscles of both
drial encephalomyopathy, lactic acidosis, and stroke- CPEO and KSS patients, but deletions are restricted to
like episodes (MELAS) syndrome. mtDNA mutations skeletal muscle in CPEO and widely distributed in ex-
involving components of complex III have been re- tramuscular tissue in KSS. In Pearson syndrome high
ported in patients with progressive exercise intoler- amounts of mtDNA defects are present in bone mar-
ance, proximal limb weakness with myoglobinuria, row precursor cells. The proportion probably falls
Parkinsonism, and LHON. In complex IV defects, with age if the patient survives; however, repeated in-
structural mutations have been detected in patients vestigation has shown an increased proportion of
presenting with isolated motor neuron disease, Leigh- deleted mtDNA in muscle tissue. Most (or all) of the
like syndrome, MELAS, proximal myopathy with lac- cases associated with single DNA deletions are spo-
tic acidosis, and LHON. In patients with neurogenic radic. The deletion probably occurs after egg fertiliza-
muscle weakness, ataxia, and retinitis pigmentosa tion and owing to unequal mitotic segregation is pre-
(NARP) and Leigh syndrome, common mutations are sent in only some of the cells. The replication of delet-
known in a gene coding for an ATP synthetase (com- ed mtDNA occurs more rapidly and, in consequence,
plex V) subunit. positive selection of respiratory-deficient cells may
A number of point mutations in tRNA and rRNA occur in tissues with rapid cell turnover.
genes have been identified. Each tRNA mutation is Duplications of mtDNA are very rare in man. Spo-
generally associated with a distinctive phenotype, al- radic cases of KSS with heteroplasmic mtDNA dupli-
though some phenotypic overlap between the differ- cations have been described.
ent tRNA defects is common. Well-known clinical The relationship between mtDNA changes, bio-
phenotypes include myoclonic epilepsy with ragged chemical defects of the respiratory chain, and clinical
red fibers (MERRF), MELAS, Leigh syndrome and phenotype remains difficult to understand (genetic
variable combinations of myopathy, cardiomyopathy, and phenotypic heterogeneity). One point mutation
chronic progressive external ophthalmoplegia can be associated with different clinical phenotypes
(CPEO), diabetes mellitus, and hearing loss. Further- (for instance, the same mutation is present in both
more, a few rRNA mutations have been also described NARP and some cases of Leigh syndrome; the same
with a very similar phenotype of CNS involvement, mutation in a tRNA gene is found in MELAS, in ma-
deafness, neuropathy, and diabetes mellitus. Despite ternally inherited myopathy and cardiomyopathy,
the overlap in the clinical symptoms mentioned, the and in maternally inherited diabetes mellitus). The
association of different tRNA mutations with clinical phenotype related to the same abnormality in
syndromes is surprisingly specific considering that mtDNA may also vary within a single kindred. The
these mutations all act on the same mitochondrial same disease can be caused by different point muta-
function, i.e., the ability of mitochondria to translate tions (see Chap. 25). Combined features of different
their own genes. Since mutant tRNA is unavailable for syndromes have been observed in one patient (KSS
translation, all mitochondrially encoded proteins are combined with MELAS; MERRF with MELAS; Pear-
present in decreased amounts. Among the different son syndrome progressing to KSS). Part of the rela-
clinical phenotypes the biochemical abnormalities tionship between genotype and phenotype can be
are virtually indistinguishable, revealing multiple explained by heteroplasmy with different percentages
partial complex defects. of mutant mtDNA in different tissues and changes
Large, single deletions of a substantial proportion of percentages over the course of time.
of mtDNA have been described in CPEO, Pearson syn-
drome, and Kearns–Sayre syndrome (KSS). These
syndromes are clinically overlapping. Muscle weak- 23.3.2 Nuclear DNA Defects
ness and chronic progressive external ophthalmople-
gia are also present in KSS. Many patients with Pear- Nuclear genes responsible for oxidative phosphoryla-
son syndrome die in early childhood from a combina- tion defects can be categorized structurally and func-
tion of pancreatic, hepatic, renal, and bone marrow tionally into genes encoding structural components
insufficiency with pancytopenia, but the few patients of the oxidative phosphorylation system, genes en-
who improve and survive develop KSS and histologi- coding assembly factors of the oxidative phosphory-
cal signs of a mitochondrial myopathy in later child- lation complexes, genes involved in the biogenesis of
hood. The deletions encompass many of the genes en- the oxidative phosphorylation system, and genes in-
coding for proteins of the respiratory chain, and also volved in the maintenance of the mtDNA.
several tRNAs. This explains the decrease in presence Among the defects in structural components of the
of all mitochondrial translation products demon- oxidative phosphorylation system, complex I deficien-
strated by immunological analyses. Biochemical cy is the most common. In general, isolated complex I
202 Chapter 23 Mitochondria and Mitochondrial Disorders

deficiency is one of the most frequent disturbances of with SURF1 mutation, but milder neurological in-
the oxidative phosphorylation system and often fol- volvement with a malabsorption syndrome and cases
lows an autosomal recessive inheritance. The clinical with a leukoencephalopathy have been described in
presentation in the majority of cases is of a Leigh or a association with SURF1 mutations as well. Mutations
Leigh-like syndrome, a leukodystrophy, or a car- in two other COX assembly genes, SCO1 and SCO2,
diomyopathy. Mutations have been identified in seven frequently present with hypertrophic cardiomyopa-
of the 39 nuclear genes encoding structural subunits thy and encephalopathy, sometimes combined with
of complex I. Complex II comprises four nuclear en- hepatic failure. The COX10 and COX15 genes are in-
coded subunits. It has a dual function as an enzyme volved in the synthesis of heme A, a prosthetic group
complex of the oxidative phosphorylation system and of COX. Mutations in these genes result in a clinical
as an essential enzyme of the citric acid cycle. It con- manifestation comparable to that of SCO2 mutations.
sists of a flavoprotein (active site with covalently A novel mutation has been recently described in the
bound FAD) and an iron–sulfur protein, and is an- ATP12 assembly gene of complex V with severe mi-
chored to the membrane by two transmembrane pro- crocephaly, hepatomegaly, and early death.
teins. A mutation in the flavoprotein coding gene of a As to defects in the biogenesis of the oxidative phos-
patient with Leigh syndrome was the first mutation phorylation system, oxidative phosphorylation defi-
identified in a nuclear gene causing oxidative phos- ciencies can caused by defects in the import of nu-
phorylation deficiency. Mutations in the genes encod- clear-encoded mitochondrial proteins into the mito-
ing the iron–sulfur protein and the anchor proteins chondrion. A mutation in the gene for subunit 8a of
cause a different phenotype of hereditary paragan- the translocase of the inner mitochondrial mem-
gliomas and/or pheochromocytomas. Complex III brane, the TIMM8A or DDP1 gene, causes human dys-
transfers electrons from the ubiquinone pool to cy- tonia deafness syndrome, also known as Mohr–Tra-
tochrome c, and is composed of 11 subunits, of which jenberg syndrome, a progressive neurodegenerative
ten are nuclear-encoded. Complex III deficiency is of- disorder. Friedreich ataxia, an autosomal recessive
ten associated with encephalomyopathy or cardiomy- disorder, is caused by disturbances of the iron–sulfur
opathy. However, in the patient with the first mutation cluster formation, related to an abnormal expansion
described in a nuclear-encoded subunit of complex of a GAA repeat in the first intron of the frataxin gene,
III (an enzyme-binding protein), no psychomotor re- which encodes a mitochondrial protein of unknown
tardation or neurological impairment was detected, function. The defect in frataxin results in oxidative
but hypoglycemia and lactic acidemia were. To date damage of the highly sensitive iron–sulfur protein
no mutations have been reported in the structural nu- complexes I, II, III, and the Krebs cycle enzyme aconi-
clear components of complex IV and complex V. Most tase. Hereditary spastic paraplegia (HSP) is a geneti-
syndromes with isolated complex IV deficiency are cally heterogeneous neurodegenerative disorder
caused by mutations in genes encoding proteins in- characterized by progressive spasticity and weakness
volved in the proper assembly of the complex. of the legs.A subtype of HSP is related to mutations in
Assembly factors are proteins which mediate the SPG7. Patients with this subtype show typical signs of
process of assemblage of subunits and intermediate mitochondrial disease (ragged red fibers and COX-
complexes into fully assembled oxidative phosphory- negative fibers). The spastic paraplegia gene SPG7 en-
lation complexes, and have chaperone-like functions. codes a nuclear-encoded mitochondrial metallopro-
A few assembly factors have been identified for com- tease protein, which has both proteolytic and chaper-
plex I and III, and many for complex IV. Mutations of one-like activities at the inner mitochondrial mem-
the only known assembly factor gene responsible for brane. The oxidative phosphorylation system may be
isolated complex III deficiency (Rieske iron–sulfur disturbed not only by defects in factors which are di-
subunit) are associated with neonatal proximal tubu- rectly part of the system or involved in its biogenesis,
lopathy, hepatic involvement and encephalopathy, but also by defects in factors that indirectly con-
and with GRACILE syndrome (growth retardation, tribute to its function, such as alterations in the lipid
amino aciduria, cholestasis, iron overload, lactic aci- composition of the inner membrane (Barth syn-
dosis, and early death), a fatal metabolic disorder with drome). Dominant optic atrophy (DOA) is caused by
iron overload. Complex IV (COX) is composed of 13 defects in the OPA1 gene, which encodes a mitochon-
structural subunits, ten of which are encoded by the drial GTPase that is important for the formation and
nucleus. A large number of accessory factors are nec- maintenance of the mitochondrial network.
essary for the assembly and maintenance of the active Mutations in nuclear genes involved in mtDNA
complex, and mutations in several of these factors maintenance cause disorders that clinically resemble
have been described. The first gene encoding an as- disorders caused by mtDNA mutations. This is under-
sembly factor known to be responsible for COX defi- standable since the nuclear gene defect causes sec-
ciency is the SURF1 gene. Leigh syndrome is the most ondary mtDNA loss (mtDNA depletion) or formation
common clinical manifestation observed in patients of multiple mtDNA deletions. However, these disor-
23.3 Mitochondrial Disorders 203

ders follow a mendelian inheritance pattern. Multiple or childhood progressive encephalomyopathy. Two
deletions of the mtDNA have been described in a genes involved in mitochondrial deoxyribonucleo-
number of clinical syndromes. The most frequently side metabolism, TK2, encoding thymidine kinase 2,
described is autosomal dominant progressive exter- and DGUOK, encoding deoxyguanosine kinase, have
nal ophthalmoplegia (AD PEO), for which three dif- been associated with a myopathic form and a hepa-
ferent disease genes have been identified: ANT1, en- toencephalopathic form of mtDNA depletion syn-
coding the adenine nucleotide translocator or drome, respectively. Thymidine kinase 2 and de-
ADP/ATP translocator; POLG1, encoding the catalyt- oxyguanosine kinase are both enzymes of the “sal-
ic subunit of the mtDNA-specific polymerase g; and vage pathway,” which is the main supply of deoxyri-
C10ORF2 encoding the Twinkle protein, a putative bonucleoside triphosphates (dNTPs) for mtDNA
mtDNA helicase. For defects in POLG1, mutations synthesis. The link between these salvage pathway en-
with a recessive mode of inheritance have also been zymes and mtDNA depletion suggests their involve-
reported. The mtDNA depletion syndrome causes a ment in the maintenance of balanced mitochondrial
rapidly fatal mitochondrial disorder in infancy. dNTP pools. Deficiency of thymidine phosphorylase
Quantitative analysis of the mtDNA reveals a severe causes multiple mtDNA deletions and/or mtDNA de-
depletion, varying from 50% up to 98% as compared pletion leading to mitochondrial neurogastrointesti-
to normal controls. Depletion of mtDNA correlates nal encephalomyopathy (MNGIE). Recently, muta-
with the presence of multiple respiratory chain de- tions in a mitochondrial deoxynucleotide carrier
fects. Clinically three syndromes are distinguished: (DNC) have been shown to cause failure of deoxynu-
fatal infantile hepatopathy, congenital myopathy cleotide transport across the inner mitochondrial
with or without nephropathy, and later-onset infantile membrane in Amish congenital microcephaly.
Chapter 24

Mitochondrial Encephalopathy with Lactic Acidosis


and Stroke-like Episodes

24.1 Clinical Features blown MELAS picture. Short stature, sensorineural


and Laboratory Investigations hearing loss, mild myopathy, cardiomyopathy, dia-
betes mellitus, and seizures without stroke or learn-
Mitochondrial myopathy, encephalopathy, lactic aci- ing problems may be the only clinical manifestation
dosis, and stroke-like episodes constitute the MELAS of disease.
acronym. The disease shows maternal inheritance Some MELAS patients have manifestations which
with considerable intrafamilial variation in expres- are typical of other mitochondrial syndromes. Pa-
sion of the disease. The age at onset varies between 3 tients with MELAS–MERFF overlap have myoclonic
months and 40 years, but in most cases first signs and epilepsy and the associated EEG features in combina-
symptoms occur before adulthood. Early develop- tion with clinical symptoms seen in MELAS, includ-
ment is normal in the majority of patients. The first ing stroke-like episodes. LHON–MELAS overlap
manifestations of disease usually belong to the group leads to features typical of Leber hereditary optic
of general features of encephalomyopathies. Growth neuropathy and stroke-like episodes.
disturbance and epileptic seizures are the most fre- On laboratory examination, most patients have
quent first symptoms. The disease is progressive with lactic acidosis, although in some resting serum lactate
increasing symptomatology. Learning disabilities, is normal. CSF lactate is often also elevated. CSF pro-
cognitive regression, exercise intolerance, and limb tein is often found to be elevated. EMG may show a
weakness are frequent manifestations of the disease. myopathic pattern. There may be neurophysiological
The myopathic features are rarely very prominent in evidence of a mixed axonal and demyelinating sen-
MELAS. Stroke-like episodes are rarely early signs of sorimotor neuropathy. Some patients have ECG
the disease but have occurred before the age of 40 abnormalities with evidence of cardiomyopathy,
years in almost all patients. The stroke-like events Wolff–Parkinson–White abnormality, or conduction
give rise to both reversible and permanent neurolog- block. In most patients ragged red fibers are found on
ical deficits. Hemiparesis and hemianopia or cortical muscle biopsy.
blindness are seen most frequently. Seizures are Antenatal diagnosis is very difficult in defects of
common during the strokes. Episodic migrainous mitochondrial DNA. Because of heteroplasmy, a
headaches with nausea and vomiting are common chorionic villus sample may not be representative of
and often precede the stroke-like episodes. In some the level of mutant mitochondrial DNA in the embryo
patients focal seizures progress to epilepsia partialis and it is therefore difficult to predict the future phe-
continua. All patients eventually develop cognitive notype.
impairment. In those with early-onset neurological
impairment, development is generally delayed from
early on in life, whereas in patients with later-onset 24.2 Pathology
impairment, the rapidity of disease progression and
the number of cerebral infarcts have a direct impact On light microscopic examination of muscle biopsies,
on the presence and severity of cognitive impairment. application of the modified Gomori trichrome stain
Sensorineural hearing loss is frequent. Delayed often reveals the presence of ragged red fibers. With
puberty, infertility, and hypothalamic hypogonadism this stain the abnormal fibers demonstrate a mottled
may be present. Diabetes mellitus, growth hormone and irregular appearance with red-staining peripher-
deficiency, hypothyroidism, hypoparathyroidism, and al and intermyofibrillar zones. Histochemistry for
hyperaldosteronism may occur. Less frequent find- oxidative enzymes, such as NADH dehydrogenase,
ings, noted in less than half of the patients, include succinate dehydrogenase, and cytochrome c oxidase,
myoclonus, cerebellar ataxia, episodic coma, pe- may yield abnormal staining patterns. On electron
ripheral neuropathy, pigmentary retinopathy, oph- microscopy, the ragged red fibers display large aggre-
thalmoplegia, ptosis, optic atrophy, hypertrophic gates of mitochondria, generally under the sarcolem-
cardiomyopathy, electrocardiographic evidence of ma, but also between myofibrils. The mitochondria
pre-excitation, cardiac conduction block, and are frequently abnormal in size and structure. The
nephropathy. In relatives of typical MELAS patients a mitochondrial cristae are often increased in number
“partial” syndrome may be seen instead of the full- and irregularly oriented. The mitochondria may con-
24.4 Therapy 205

tain different abnormal inclusions such as crystalline found in patients with chronic progressive external
or paracrystalline structures or globular bodies. ophthalmoplegia and other neurological complaints,
Postmortem examination of the brain of deceased but without stroke-like episodes.
MELAS patients often reveals atrophy on external ex- In MELAS, an isolated defect of complex I or com-
amination. The cerebellum, too, may have an atroph- bined defects in complexes I, III, and IV are found in
ic aspect. Major blood vessels are normal. Microscop- muscle mitochondria. The presence of a mutant tRNA
ic examination reveals areas of extensive cortical lam- disturbs the mitochondrial gene translation machin-
inar necrosis, usually in the occipital and posterior ery and leads to a decrease in all mitochondrially
temporal areas. The cortical lesions usually have an encoded proteins. The mechanism by which the
asymmetrical distribution and are not related to vas- tRNAleu(UUR) gene mutation interferes with protein
cular supply areas. The three deepest cortical layers synthesis remains unclear, but defects in tRNAleu(UUR)
are affected most severely. The gyral crests and sulcal function, rRNA to mRNA ratio, impaired incorpora-
depths are equally affected. The severity of the corti- tion of leucine into mitochondrial translation prod-
cal damage varies from fractional neuronal loss to ucts, and defects in rRNA processing are viable alter-
microcystic destruction. Gliosis is present. The corti- natives.
cal damage may have a spongiform aspect. In the area Among MELAS patients and their maternal rela-
of cortical damage, there is a proliferation of capillar- tives there is a clear relationship between percentage
ies. The lesions vary in age, the patient often having of mutant mitochondrial DNA and severity of dis-
lesions of different ages. The adjacent subcortical ease. This is clearer for the level of mutant mitochon-
white matter is involved with loss of myelin and pres- drial DNA in muscle than for that in blood. Asympto-
ence of fibrous gliosis. The white matter damage may matic relatives have the lowest percentage, oligo-
be spongiform. The periventricular white matter is symptomatic relatives an intermediate percentage,
usually not involved. However, diffuse white matter and MELAS patients the highest. Among patients it
gliosis and atrophy may also be found. Calcium de- has been found that the higher the percentage of mu-
posits are frequently present in the globus pallidus, tant DNA is, the earlier the symptoms of stroke-like
less frequently in the caudate nucleus, lateral thala- attacks appear.
mus, dentate nucleus, subthalamic nucleus, substantia The cause of the large cerebral lesions is a matter of
nigra, and red nucleus. The calcium granules are debate. One hypothesis is that the relationship be-
mostly deposited in and around capillary and small tween mitochondrial dysfunction and cerebral
arterial walls. Otherwise the basal nuclei are intact pathology is explained by a mitochondrial vasculopa-
and no neuronal loss is seen. Within the cerebellum thy of small arteries, observed in the “infarcted” ar-
pathological changes consist of some variable loss of eas. There is a marked increase in number of mito-
Purkinje cells and granule cells. The dendrites of chondria in endothelial cells, smooth muscle cells,
Purkinje cells may show remarkable segmented and pericytes of arterioles. The mitochondria are ab-
swelling, which is called cactus. On electron mi- normal and enlarged. These vascular abnormalities
croscopy, accumulation of mitochondria, neurofila- may result in decreased blood supply. However, in
ments, and membrane complex is seen in the region SPECT and PET scan studies, evidence of patency of
of cactus formation. Brain stem and spinal cord may blood vessels is found in the acute stage of the infarct
also contain spongiform lesions. Electron microscopy with the presence of a well-preserved blood flow.
reveals abnormal accumulation of mitochondria in SPECT studies have repeatedly shown focal hyperper-
endothelial and smooth muscle cells of small arteries. fusion before and during the stroke in MELAS and
The major cerebral arteries are normal. focal hypoperfusion in the chronic, atrophic stage.
Hence, it is more probable that the metabolic de-
mands exceed the potential of energy provision by
24.3 Pathogenetic Considerations the disturbed oxidative phosphorylation, leading to
cell damage and edema.
Two heteroplasmic point mutations have been discov-
ered in most cases of MELAS. In 80% of the patients
a mutation is present in nucleotide 3243 of mitochon- 24.4 Therapy
drial DNA, which affects a nucleotide position in the
dihydrouridine loop of the tRNA specific to leucine Various therapeutic strategies have been used in
(UUR codon) (tRNAleu(UUR)). In addition, 7.5% of MELAS. In particular, administration of cofactors is
the patients have another mutation in the same often employed in an attempt to ameliorate the course
tRNAleu(UUR) gene at nucleotide 3271. The remainder of disease. Riboflavin is a precursor of both flavin
of MELAS patients have other point mutations in the monophosphate and flavin adenine dinucleotide
mitochondrial DNA; few have a deletion within the (FAD), which are part of NADH-coenzyme Q reduc-
mitochondrial DNA. The 3243 mutation has also been tase (complex I) and of complex II. Nicotinamide is a
206 Chapter 24 Mitochondrial Encephalopathy with Lactic Acidosis and Stroke-like Episodes

precursor of NAD. Coenzyme Q10 is given to substi- in progressive atrophy with enlargement of the ven-
tute and supplement for endogenous coenzyme Q. tricular system and subarachnoid spaces (Fig. 24.2).
Idebenone, a quinone compound similar to coen- Cerebellar atrophy may be prominent.
zyme Q10, has the theoretical advantage of crossing Different MRI techniques can be of advantage in
the blood–brain barrier. Vitamin C and vitamin K3 MELAS. Gradient-echo techniques are more sensitive
(menadione) are given to bridge a defect in the elec- to calcium than other techniques. FLAIR images are
tron transport chain, as they accept and transport superior in showing small cortical lesions (Fig. 24.2).
electrons. These drugs are given alone or in varying These lesions may arise without associated clinical
combinations. In patients with MELAS variable im- symptoms of stroke. Diffusion-weighted imaging and
provement has been reported, ranging from none to mapping ADC values are of particular interest during
remarkable, but evidence for a consistent beneficial the acute stages of a stroke-like episode (Figs. 24.3
effect is lacking. Dichloroacetate inhibits the pyruvate and 24.4). The results reported so far are conflicting.
dehydrogenase specific kinase, thereby activating the A study with sequential imaging from day 3 up to day
pyruvate dehydrogenase complex and reducing lac- 25 shows that lesions in the acute phase may show
tate levels. Oral creatine supplementation has been normal or elevated ADC values in MELAS (Yoneda et
reported to lead to clinical improvement in incidental al. 1999). Normal or elevated ADC values have also
patients. Intravenous administration of L-arginine been reported in several other, less systematic studies.
during acute stroke has also been reported to lead to This, of course, is contrary to what is seen in ischemic
improvement. infarctions. However, we and others (Wang et al. 2003)
Symptomatic treatment of the neurological handi- have found strongly reduced ADC values in acute le-
cap, epilepsy, and endocrine dysfunction is impor- sions, suggesting cytotoxic edema. It remains unclear
tant. why ADC values are raised in the acute lesion in some
patients with MELAS. It has been suggested that it is
because of the presence of vasogenic edema, but this
24.5 Magnetic Resonance Imaging explanation seems too simple. There is evidence for
hyperperfusion in acute MELAS lesions. Both this hy-
In MELAS CT often shows the presence of calcium perperfusion with more prominent presence of blood
deposits in the globus pallidus and caudate nucleus in the voxels and the disruption of the blood–brain
(Fig. 24.1). Sometimes the calcium deposits are more barrier may affect the ADC measurements. Perhaps
extensive and also affect the putamen and thalamus. the conflicting findings described are also reflected in
During the acute phase of stroke-like episodes, one or the observation that some of the lesions in MELAS
more large hypodense areas are seen. The areas are disappear after a couple of weeks, whereas others per-
swollen. They have, as a rule, an asymmetric distribu- sist or result in focal atrophy. It seems a reasonable as-
tion. sumption that there are differences between subtypes
In MRI the calcium deposits in the basal nuclei are of MELAS lesions, reflected in differences in MR mea-
more difficult to see (Fig. 24.1). MRI shows the precise surements. A more systematic MR approach, follow-
distribution of the lesions occurring during the up, and analysis could probably better document
stroke-like episodes. From MRI it is clear that the cor- these differences.
tex is often more severely involved than the underly- Cerebral angiography, xenon-enhanced computed
ing white matter and that the periventricular white tomography, and SPECT demonstrate patency of ves-
matter is most often preserved (Figs. 24.1 and 24.2). It sels in MELAS and, in fact, vasodilation. The regional
is also the cortex that enhances after contrast injec- cerebral blood flow is increased in the area of an
tion (Fig. 24.1). The lesions are variable in size, some- acute lesion (hyperperfusion). This has been docu-
times small, often large, sometimes single, often mul- mented in SPECT studies with 99mTc-ethyl cysteinate
tiple, and usually asymmetrical (Fig. 24.2). The distri-
bution of the lesions does not follow vascular supply
or vascular border zones. The occipital and posterior
temporal areas are preferentially involved (Figs. 24.1 䊳

and 24.2). Other areas that may be involved are the Fig. 24.1. A 21-year-old female patient with MELAS and the
thalamus, basal ganglia, and brain stem. Diffuse cere- 3243 mitochondrial DNA mutation. Note the calcium deposits
bellar involvement has also been described. In the in the globus pallidus on CT (second row, left). The globus pal-
acute stage the lesions are often swollen. The lesions lidus has a low signal on T2-weighted SE images. There is a
may increase in size over days. In the course of a few large lesion in the left temporo-occipital region, involving the
weeks, the lesions may either resolve or leave behind cortex and to a lesser extent the subcortical white matter. A
an area of atrophy and altered signal intensity, in par- small lesion in the body of the caudate nucleus is seen on the
ticular in the cortex (Fig. 24.2). Over the years MRI right. After contrast administration (third and fourth row) it is
may show “migrating infarcts” that leave their traces mainly the affected cortex that enhances
24.5 Magnetic Resonance Imaging 207

Fig. 24.1.
208 Chapter 24 Mitochondrial Encephalopathy with Lactic Acidosis and Stroke-like Episodes

Fig. 24.2.
24.5 Magnetic Resonance Imaging 209

Fig. 24.2. A male patient with MELAS and the 3243 mitochon- showing the cortical lesions. At the age of 12.5 years he had an
drial DNA mutation underwent MRI at the ages of 9 years (first episode of dysphasia and right-sided hemiplegia and hemi-
row), 11 years (second and third rows), 12.5 years (fourth row), anopia. The MRI showed a large lesion in the left temporo-oc-
and 15.5 years (fifth row). At 9 years, he had seizures and some cipital region, involving both cortex and white matter. In addi-
learning problems, but was otherwise normal. At 11 years, he tion,multiple old,small cortical lesions were seen.At 15.5 years
had seizures and migrainous headaches and was evidently de- the patient had serious dementia and dysphasia. MRI showed
menting. He had not had any stroke-like episodes. The MRI marked generalized cerebral atrophy as well as focal atrophy
showed multiple small cortical lesions and generalized cere- of the previously infarcted region
bral atrophy.The FLAIR images (third row) were much better at

dimer (ECD) and 99mTc-D,L-hexamethylpropylene- phases of moyamoya disease. The distribution of


amine oxime (HMPAO). The increased uptake of lesions in patients with moyamoya syndrome can be
these substances in the lesion area also suggests a the same as in MELAS. The abnormal vessels in
breakdown of the blood–brain barrier. Unfortunately, moyamoya syndrome, usually already seen on con-
perfusion studies have not been performed systemat- ventional MR images, but more clearly on MR angio-
ically in a large group of MELAS patients, and no re- graphy, will suggest the latter diagnosis. MELAS also
ports are available concerning MR perfusion studies has to be differentiated from vasculitic disorders
of acute lesions. Especially, no data are available link- presenting with multiple infarctions. In a case of
ing perfusion studies with ADC maps. predominantly temporal location of the acute lesion,
Proton MRS shows low N-acetylaspartate in acute herpes simplex encephalitis may be suspected. The
lesions and highly elevated lactate. The decrease in raised serum lactate suggests mitochondrial dysfunc-
N-acetylaspartate is at least partially reversible, and tion rather than herpes encephalitis, and the calcium
in the chronic stage lactate is usually normal or only deposits which are often present in the basal ganglia
mildly elevated. Elevated lactate may also be seen in in MELAS patients may also help in differentiation.
areas that are not clearly abnormal on MR images, but Other multifocal infectious and inflammatory lesions
the elevation is usually much more striking in acute should be considered as well. Single and multiple
and subacute lesions. large areas of abnormal signal intensity in the cortex
The obvious task for MR is to help distinguish be- and white matter are also seen in urea cycle defects. In
tween ischemic infarctions and the infarct-like le- MRS, however, findings are very different, with raised
sions in MELAS. In the younger group it may be im- lactate in MELAS and raised glutamine in urea cycle
portant to differentiate the infarct-like lesions of defects. Assessment of blood lactate and ammonia
MELAS from the ischemic infarctions in the early levels shows a similar difference.
210 Chapter 24 Mitochondrial Encephalopathy with Lactic Acidosis and Stroke-like Episodes

Fig. 24.3. Conventional and diffusion-weighted MRI of the extent of the lesion.The diffusion-weighted images show high
patient in Fig. 24.2 at the age of 12.5 years.To be more precise, signal in the lesion with an area of lower signal in the occipital
the stroke-like episode started with dysphasia,followed after 6 white matter.The ADC maps show that the temporal lesion has
days by development of right-sided hemiplegia and hemi- a high ADC, whereas most of the occipital lesion has a low
anopia. The MRI was obtained 12 days after the onset of the ADC.The difference in ADC may be related to the different age
episode. Displayed are T2-weighted SE images (first row), trace of the lesions. It could also be that the temporal lesion is more
diffusion-weighted images with a b value of 1000 (second row), edematous, as suggested by the T2-weighted image
and ADC maps (third row). The T2-weighted images show the
24.5 Magnetic Resonance Imaging 211

Fig. 24.4. Conventional and diffusion-weighted MRI of a 13- occipital lesion. On the trace diffusion-weighted image with a
year-old MELAS patient with the 3243 mitochondrial DNA b value of 1000 (middle) the lesion has a high signal, whereas
mutation. The MRI was obtained 3 days after the onset of the lesion has a low signal on the ADC map (right). The mea-
right-sided hemianopia. The FLAIR image (left) shows a large sured ADC is very low
Chapter 25

Leber Hereditary Optic Neuropathy

25.1 Clinical Features perience more severe neurological problems. An ex-


and Laboratory Investigations trapyramidal movement disorder with bilateral le-
sions of the basal ganglia on MRI has been reported
Leber hereditary optic neuropathy (LHON) is a ma- in several patients. Association with a Leigh-like en-
ternally inherited disorder that causes acute or suba- cephalopathy with brain stem and basal ganglia in-
cute loss of bilateral central vision. The onset is usu- volvement on MRI has also been observed. The most
ally asymmetrical, but the interval between involve- frequently observed association is with a multiple
ment of the two eyes is usually less than a few months. sclerosis-like disease, and multiple studies have
Monocular involvement is extremely rare. Men are af- shown that the association is more than coincidental.
fected much more frequently than women and the age Episodic neurological abnormalities occur with par-
at onset is usually lower in males than in females. The tial or complete recovery. Apart from optic neuropa-
male preponderance ranges from 80% to 90% in thy, frequently observed signs include spasticity, cere-
most white pedigrees to approximately 60% in Japan- bellar ataxia, sensory disturbances, vertigo, diplopia,
ese families. The onset of visual loss typically occurs internuclear ophthalmoplegia, and urgency of mic-
between the ages of 15 and 35 years, with an age range turition. The clinical features are indistinguishable
of 5–70 years. In most patients visual acuities deteri- from those of multiple sclerosis, except for their oc-
orate to worse than 2/20 in the course of several currence in the context of a family history of LHON.
months, stabilizing thereafter. The loss of vision is Presence of severe and bilateral visual signs justifies
usually permanent, but spontaneous improvement or considering the possibility of LHON in otherwise
recovery can occur. Transient worsening of vision typical multiple sclerosis patients.
with exercise or heat may occur (Uhthoff ’s symp- Laboratory tests are of limited use in LHON. Fluo-
tom).Visual field defects are typically central or ceco- rescein angiography is helpful for the illustration and
central, but may also be bitemporal. Headache, eye confirmation of the LHON funduscopic features.VEP
discomfort, and flashes of light may occur at the time studies confirm the absence of any response or pres-
of vision loss. Color vision is affected severely, often ence of responses with prolonged latencies and de-
early on in the course of vision loss. creased amplitudes. ERG is typically normal. In pa-
The first sign of the disease, peripapillary telang- tients in whom the disease course resembles that of
iectatic microangiopathy, is ophthalmoscopically vis- multiple sclerosis, CSF analysis may reveal an eleva-
ible in the presymptomatic stage. At the onset of visu- tion of the IgG index and presence of oligoclonal
al loss, the optic disc is swollen and there is marked bands. ECG may reveal conduction abnormalities. In
dilatation and tortuosity of the peripapillary retinal the past, a definitive diagnosis depended on a positive
vessels. However, vessels do not leak fluorescein on family history, age at onset of the vision loss, and the
fluorescein angiography. The typical ophthalmoscop- characteristic circumpapillary microangiopathy of
ic findings may also be present in asymptomatic fam- the optic disc in the acute stage. Demonstration of a
ily members, but some patients with LHON never ex- point mutation in mitochondrial DNA in affected in-
hibit these characteristics, even if examined at the dividuals confirms the diagnosis; DNA confirmation
time of acute vision loss. After the development of vi- of the diagnosis can also be obtained in atypical or
sual loss, a capillary-poor retina with attenuated arte- sporadic cases.
rioles and a pale optic disc remain.
In the majority of patients with LHON, visual dys-
function is the only significant manifestation of the 25.2 Pathology
disease. In some pedigrees additional cardiac con-
duction abnormalities occur, in particular pre-excita- In LHON severe axonal degeneration with myelin loss
tion syndromes such as Wolff–Parkinson–White syn- of the central part of the optic nerve and pregenicu-
drome. Minor neurological abnormalities such as hy- late pathway is found. The nature of the cerebral mul-
perreflexia, Babinski signs, mild ataxia, or peripheral tiple sclerosis-like white matter lesions has not yet
neuropathy have been reported. Several pedigrees been investigated.
have been described in which some of the patients ex-
25.5 Magnetic Resonance Imaging 213

25.3 Pathogenetic Considerations suggested but never proven. The male preponderance
and earlier onset for males suggest a recessive suscep-
A number of mitochondrial DNA mutations have tibility gene on the X chromosome acting in synergy
been described in association with LHON. The muta- with the mitochondrial DNA mutation. Affected fe-
tions can be distinguished into high-risk mutations, males heterozygous for the X-chromosomal suscepti-
called class I or primary mutations, and low-risk mu- bility gene may be involved because of unfortunate X
tations, called class II or secondary mutations. The chromosome inactivation.
most frequent class I mutations include a mutation at Almost all class I mutations alter mitochondrial
nucleotide position 11778 in the mitochondrial gene DNA-encoded subunits of complex I. The LHON mu-
encoding subunit 4 of complex I, present in 50% of tations do not alter the amount, assembly, and stabil-
European patients to 95% of Asian LHON patients; a ity of complex I. However, various mutations have
mutation at nucleotide position 3460 in the gene en- been shown to affect complex I function, resulting in
coding subunit 1 of complex I, present in about 15% altered electron transport capacity, respiration rate,
of European patients; a mutation at position 14484 in and ATP production. It is suggested that the decline in
the gene encoding for subunit 6 of complex I, present oxidative phosphorylation capacity caused by a com-
in about 15% of European patients; and a mutation at bination of influences (class I and class II mutations,
position 14459 in the gene encoding for subunit 6 of X-linked nuclear factor, other genetic factors, envi-
complex I. In addition, several class II mutations are ronmental factors, effects of aging) may result in op-
known, the pathogenic role of which is much less tic atrophy, cardiac conduction abnormalities, and
clear. Generally, only one class I mutation occurs in a neurological disease. With regard to those pedigrees
LHON pedigree and individuals harboring this muta- with LHON and neurological disease, it would appear
tion have a relatively high probability of vision loss. that certain mitochondrial DNA mutations may be
Class I mutations are not observed in the normal pop- particularly responsible. The mutation at position
ulation. Class II mutations occur at a much higher fre- 14459 is frequently associated with a combination of
quency among LHON patients than among the nor- LHON and dystonia. Females with the 11778 muta-
mal population.When present in LHON patients, they tion are especially likely to develop clinical and neu-
occur in combination with other class I or II muta- roimaging findings indistinguishable from multiple
tions. They may serve as additional predisposing or sclerosis.
exacerbating genetic factors that increase the proba-
bility of expressing LHON. It is probably the synergis-
tic effect of multiple mitochondrial DNA mutations 25.4 Therapy
that, by their accumulative effect on the oxidative
phosphorylation, produces a pathogenic reduction in No effective treatment to prevent or halt LHON has as
ATP-generating capacity. yet been found.
For all class I mutations variable penetrance is ob-
served among apparently homoplastic LHON pedi-
grees. About 50% of males and 10% of females har- 25.5 Magnetic Resonance Imaging
boring a pathogenic mutation actually develop
LHON. In spite of sharing the same mutations, males In patients with LHON, optic nerve abnormalities can
have a 4–5 times higher risk of being affected than fe- be shown with STIR (short tau inversion recovery) se-
males. About 15% of the patients are heteroplasmic quences. Increased signal intensity of the mid and
for a class I mutation, and in about 35% of the fami- posterior intraorbital section of the optic nerve is
lies at least one heteroplasmic individual can be iden- seen, sparing the anterior portion. In most patients
tified, but heteroplasmy is not sufficient to explain the brain MRI is normal. In LHON associated with dysto-
observed phenotypic variation. Therefore, class I mu- nia, bilateral putamen lesions have been found. In pa-
tations appear to be necessary but not sufficient for tients with a Leigh-like presentation, additional brain
the clinical manifestation of LHON.Additional genet- stem lesions are found. Cerebellar atrophy has also
ic (nuclear or mitochondrial), environmental, or been reported. In patients with a multiple sclerosis-
physiological factors may play a significant role in the like disease, multiple small white matter lesions are
expression of LHON. Both internal and external envi- seen as in multiple sclerosis (Fig. 25.1). The lesions
ronmental factors may play a role. Systemic illnesses, are found in the periventricular and deep white mat-
nutritional deficiencies, and toxins that stress or in- ter of the cerebral hemispheres, in the brain stem, and
hibit the body’s mitochondrial respiratory capacity in the cerebellum. The periventricular white matter is
could conceivably initiate or increase phenotypic ex- predominantly involved. On the basis of MRI only,
pression of the disease. Adverse effects of tobacco differentiation between LHON and multiple sclerosis
smoke (cyanide present in tobacco smoke inhibits cy- is not possible. In MRS we found no elevation of lac-
tochrome-c oxidase activity) and alcohol have been tate in LHON.
214 Chapter 25 Leber Hereditary Optic Neuropathy

Fig. 25.1. Male patient, 43 years of age, with LHON.The transverse proton density series shows a white rim around the ventricles,
involvement of the corpus callosum, and some isolated spots in the centrum semiovale.The ventricles are slightly enlarged
Chapter 26

Kearns–Sayre Syndrome

26.1 Clinical Features velop aplastic anemia without other signs of Pearson
and Laboratory Investigations syndrome.
Laboratory investigations in KSS almost invariably
Kearns–Sayre syndrome (KSS) is a rare, sporadic dis- reveal an increased CSF protein level, usually above
order that affects males and females equally. Disease the level of 1 g/l. Blood and CSF lactate and pyruvate
onset is before the age of 20 years. The sequence of are usually elevated. Signs of variable endocrine dys-
manifestations is not constant, but the signs and function are frequently found. EMG may reveal signs
symptoms in themselves are consistent. Early devel- of a myopathy. ECG shows evidence of a disturbance
opment is normal. Ptosis and chronic progressive of cardiac conduction. Echocardiography and chest
external ophthalmoplegia are usually the initial signs. X-ray may show cardiomegaly.
Apart from progressive external ophthalmoplegia the Biochemical analysis of muscle tissue reveals de-
typical clinical triad includes pigmentary degenera- creased activity of respiratory complexes I, III, and IV,
tion of the retina and cardiac conduction block. The individually or in combination. The diagnosis is con-
fine salt-and-pepper type of atypical retinitis pig- firmed by demonstrating a heteroplasmic, single,
mentosa is usually associated with good visual func- large-scale deletion of mitochondrial DNA or, rarely,
tion and follows a benign course. Incidentally, choroi- a mitochondrial DNA duplication.
deremia is present instead of pigmentary retinopathy.
The most common cardiac conduction blocks are
complete atrioventricular block, bundle branch 26.2 Pathology
blocks, and fascicular blocks. Other frequently noted
signs are short stature due to progressive growth fail- On light microscopic examination of muscle biopsies,
ure, delayed psychomotor development, sensorineur- application of the modified Gomori trichrome stain
al hearing loss, cerebellar ataxia, proximal myopathy, reveals the presence of ragged red fibers. With this
cardiomyopathy, and sensory neuropathy. Less fre- stain the abnormal fibers demonstrate a mottled and
quent are pyramidal dysfunction and dementia. irregular appearance with red-staining peripheral
Seizures may occur. Endocrine disease is often pre- and intermyofibrillar zones. Histochemistry for ox-
sent and may include primary gonadal failure, idative enzymes such as NADH dehydrogenase, succi-
delayed puberty, diabetes mellitus, growth hormone nate dehydrogenase, and cytochrome-c oxidase may
deficiency, hyperaldosteronism, hypothyroidism, hy- yield abnormal staining patterns. On electron mi-
poparathyroidism, ACTH deficiency, and primary croscopy, the ragged red fibers display large aggre-
adrenal insufficiency. Hypomagnesemia may occur. gates of mitochondria, generally under the sarcolem-
Some patients have a mottled, hyperpigmented skin. ma, but also between myofibrils. The mitochondria
Cardiac arrhythmias and congestive cardiomyopathy are frequently abnormal in size and structure. The
may be the cause of death. Cardiogenic embolism and mitochondrial cristae are often increased in number
stroke may also occur and contribute to the disability and irregularly oriented. The mitochondria may con-
and the risk of death. tain different abnormal inclusions such as crystalline
Incidental patients have been reported who suf- or paracrystalline structures or globular bodies.
fered from Pearson syndrome in infancy, followed lat- In KSS the main finding on postmortem examina-
er by KSS. Pearson syndrome comprises refractory tion is a spongy state of the cerebral white matter due
sideroblastic anemia requiring transfusions, throm- to splitting of myelin sheaths. The axons are general-
bocytopenia, neutropenia, pancreatic insufficiency, ly preserved. The white matter changes are diffusely
and hepatic dysfunction. Onset is in infancy and most observed in the frontal, parietal, temporal, and occip-
patients die in early childhood from a combination of ital lobes of both hemispheres. It is the hemispheric
pancreatic, hepatic, renal, and bone marrow insuffi- white matter that is involved, whereas the corpus cal-
ciency. Some children survive the infantile period and losum and internal capsule tend to be preserved. The
no longer need repeated blood transfusions. These U fibers are preferentially affected. The cerebral cor-
patients subsequently developed KSS syndrome. Inci- tex is intact. The basal ganglia are involved, in partic-
dental KSS patients have also been reported who de- ular the globus pallidus and caudate nucleus, but the
216 Chapter 26 Kearns–Sayre Syndrome

putamen, thalamus, hypothalamus, subthalamic nu- In all patients the deletions are heteroplasmic. Part
clei, and substantia nigra may also be affected. The of the relationship between genotype and phenotype
basal ganglia lesions are characterized by loss of can be explained by heteroplasmy with different per-
nerve cells, spongiosis, gliosis, and capillary prolifer- centages of mutant mitochondrial DNA in different
ation.Within the globus pallidus perivascular deposi- tissues and changes of percentages with course of
tions of calcium and intracellular depositions of iron time.An increase with time of the mutated mitochon-
may be seen. The same type of depositions may be drial DNA fraction has been reported, paralleling the
seen within the caudate nucleus, dentate nucleus, and progression of the disease. In patients surviving Pear-
the putamen. Within the cerebellar cortex, loss of son syndrome it is likely that the patient initially had
Purkinje cells may be seen. The cerebellar white mat- a high percentage of mitochondrial DNA with a dele-
ter displays spongiosis and gliosis. In the brain stem tion in blood cells. The spontaneous recovery indi-
spongiosis and gliosis have been reported in white cates that selection favoring normal cells may occur
matter structures, in the pontine and midbrain in vivo. The percentage of deleted mitochondrial
tegmentum, red nucleus, substantia nigra, and other DNA is similar in muscles of both CPEO and KSS pa-
gray matter nuclei. Within the spinal cord vacuolation tients, but deletions are restricted to skeletal muscle
of tracts,in particular the dorsal columns,may be seen. in CPEO and widely distributed in extramuscular tis-
Cardiomyopathy with ragged red fibers may be sues in KSS. In Pearson syndrome mitochondrial
found. The pancreas may display fatty infiltration. DNA defects are present in high amounts in all tissue,
The adrenals may be atrophic and small. Glomerular in particular in blood and bone marrow. The propor-
and tubular abnormalities may be found in the kid- tion in blood and bone marrow probably falls with
neys. The lobules of the testes may be atrophic with age if the patient survives, whereas conversely an in-
marked overgrowth of fibrous tissue. crease in proportion of deleted mitochondrial DNA
has been shown in repeated investigation of muscle
tissue. In KSS patients the deletion is usually present
26.3 Pathogenetic Considerations in only a small proportion of peripheral blood cells.
The deletions are sporadic. The deletions are not
In most KSS patients large deletions of mitochondri- present in mothers of patients or in children of affect-
al DNA are present. Large deletions of a substantial ed mothers. Apparently, the deletions occur in the
proportion of mitochondrial DNA have been de- oocyte or zygote and affect somatic rather than germ
scribed in chronic progressive external ophthalmo- cells. An alternative explanation for not finding the
plegia (CPEO), Pearson syndrome, and KSS. These deletion in offspring of KSS mothers could be that
syndromes are clinically overlapping: muscle weak- oocytes containing mitochondrial DNA deletions
ness and chronic progressive external ophthalmople- may not be viable for gametogenesis and fertilization.
gia form part of the symptomatology in KSS. By means of unequal mitotic segregation the deletion
Although the size and the position of the large dele- is spread among part of the cells of the body of the
tions ranges from 1.8 to 8 kilobases, the same 4.977- embryo and fetus. Replication of genomes containing
kilobase deletion is known to occur most often and the deletion occurs more rapidly, and as a conse-
is therefore known as “the common 5-kb deletion.” quence selection of respiratory-deficient cells may
Most deletions are flanked by direct repeats, suggest- occur in the rapid-turnover tissues.
ing that homologous recombination or slip-replica- KSS is considered a clinical diagnosis and multiple
tion may be responsible for the deletion. Despite the KSS patients have been reported with other mito-
deletion, the mitochondrial genomes are usually com- chondrial DNA abnormalities than those described
petent for replication and transcriptionally active. above, including point mutations, multiple deletions,
Most frequently, the deleted segment encompasses and mitochondrial DNA depletion. It is striking that
some of the genes encoding the 13 proteins of the res- as a rule these patients do not have the same abnor-
piratory chain, but also several genes encoding tRNAs. malities on MRI of the brain as are observed in pa-
This explains the decrease in all mitochondrial trans- tients with classical KSS. In most of these patients
lation products demonstrated in immunological and MRI of the brain is normal.
biochemical analyses, leading to respiratory chain
dysfunction. Biochemical analysis reveals decreased
activity of complexes I, III, and IV, individually or in 26.4 Therapy
combination. Although large single deletions of mito-
chondrial DNA were initially thought to underlie KSS, Symptomatic treatment is important in KSS. Cardiac
the genetic abnormality is more complex. Mitochon- conduction defects often require medication or the
drial DNA duplications appear frequently to be pre- implantation of a pacemaker. Endocrine dysfunction
sent as well, and there may be multiple deletions. can be treated adequately.
26.5 Magnetic Resonance Imaging 217

Fig. 26.1. Male patient, 29 years old, with KSS.The CT scan reveals calcium in the head and tail of the caudate nucleus and in the
globus pallidus.There is some generalized atrophy

Administration of cofactors has often been em- MRI often shows a characteristic pattern with
ployed in an attempt to ameliorate the course of dis- symmetrical lesions of the globus pallidus and thala-
ease. Coenzyme Q10, cytochrome c, flavin adenine mus and subcortical white matter abnormalities
dinucleotide, riboflavin, thiamine, nicotinamide, vita- (Figs. 26.2–26.4). Other central nuclei that are often
min C, and vitamin K3 (menadione) have been given involved include the caudate nucleus, substantia ni-
alone or in varying combinations and variable im- gra, and red nuclei. The putamen is usually less
provement has been reported. However, none of these prominently involved. The white matter abnormali-
has led to a consistent improvement of ocular move- ties are symmetrical and tend to involve all subcorti-
ments, CNS symptoms, and cardiac function. cal white matter in a patchy or confluent way, sparing
the periventricular white matter and corpus callo-
sum, although in our experience the splenium of the
corpus callosum is usually affected. The cerebellar
26.5 Magnetic Resonance Imaging white matter may also be involved. Often extensive
brain stem abnormalities are seen, in particular in-
In KSS CT scan often reveals calcium deposits in the volving the brain stem tegmentum. Cerebellar and to
globus pallidus and caudate nucleus in addition to a lesser extent cerebral atrophy may occur.
low density of the cerebral white matter and progres- The full-blown pattern in KSS is diagnostic. How-
sive atrophy (Fig. 26.1). In some patients the calcium ever, the pattern develops over time and in the earlier
deposits are more widespread and also occur in the stages only limited abnormalities in some of the
cerebral white matter. In the absence of calcium depo- structures may be seen. In these patients, the differen-
sition, low density of the globus pallidus may be ap- tial diagnosis is longer. MRS shows elevated lactate in
parent. KSS, which may aid the diagnosis.
218 Chapter 26 Kearns–Sayre Syndrome

Fig. 26.2. A 15-year-old male patient with KSS. There are malities in the thalamus and globus pallidus. It is not clear
extensive cerebral white matter abnormalities with predomi- to what extent the posterior limb of the internal capsule is
nant involvement of the deep and subcortical white matter involved. Note the brain stem abnormalities, predominantly
and sparing of the periventricular white matter and corpus involving the midbrain, pontine tegmentum, and medulla.The
callosum, except for the splenium.There are mild signal abnor- cerebellar white matter is also involved
26.5 Magnetic Resonance Imaging 219

Fig. 26.3. A 21-year-old female patient with KSS. The white nal changes in the globus pallidus, thalamus, and posterior
matter abnormalities are less impressive and appear to involve limb of the internal capsule. Note the brain stem and cerebel-
the deep white matter with relative sparing of the U fibers and lar white matter abnormalities. Courtesy of Dr. T. van Laar and
complete sparing of a periventricular rim.The splenium of the Dr. J.C.H van Oostrom, Department of Neurology, University
corpus callosum is prominently involved. There are subtle sig- Hospital Groningen, The Netherlands
220 Chapter 26 Kearns–Sayre Syndrome

Fig. 26.4. The MRI of a 13-year-old female KSS patient displays ities in the thalamus, globus pallidus, and corticospinal tracts
more diffuse, mild signal changes in the cerebral white matter. in the posterior limb of the internal capsule, midbrain, and
The subcortical white matter in the parietal and occipital pons. The pontine tegmentum, cerebellar white matter, and
region has a more pronounced signal abnormality than the medulla also contain signal abnormalities. Courtesy of Dr. S.
remainder of the cerebral white matter. The sparing of the Blaser, Department of Diagnostic Imaging, Hospital for Sick
periventricular white matter is less clear. Again, the splenium Children, Toronto, Canada
of the corpus callosum is involved. There are signal abnormal-
Chapter 27

Mitochondrial Neurogastrointestinal
Encephalomyopathy

27.1 Clinical Features Some patients have an abnormal ECG with evidence
and Laboratory Investigations of conduction disturbances. Laboratory studies show
various mitochondrial abnormalities in skeletal mus-
Mitochondrial neurogastrointestinal encephalomy- cle, including ragged red fibers with ultrastructurally
opathy (MNGIE) is a multisystem mitochondrial dis- abnormal mitochondria, muscle fibers with increased
ease which has also been described under a number succinate dehydrogenase stain, cytochrome-c oxi-
of other acronyms: myoneurogastrointestinal en- dase-negative fibers, and decreased activities of respi-
cephalopathy (also MNGIE); polyneuropathy, oph- ratory chain enzymes. In addition, multiple mito-
thalmoplegia, leukoencephalopathy, and intestinal chondrial DNA deletions or mitochondrial DNA de-
pseudo-obstruction (POLIP); oculogastrointestinal pletion or both are found.
muscular dystrophy (OGIMD); mitochondrial en- The diagnosis is confirmed by demonstrating
cephalomyopathy with sensorimotor polyneur- elevated thymidine levels in plasma and deficient
opathy, ophthalmoplegia, and pseudo-obstruction thymidine phosphorylase activity in leukocytes.
(MEPOP); and chronic intestinal pseudo-obstruction DNA-based confirmation of the diagnosis is also pos-
(CIPO) with myopathy and ophthalmoplegia. sible.
The disease has an autosomal recessive mode of
inheritance. In the majority of the patients symptoms
have their onset between the first and fifth decade, 27.2 Pathology
with a mean of 19 years and extremes of 5 months and
45 years. Initial symptoms are most frequently gas- In MNGIE cerebral gray matter structures are intact.
trointestinal, ocular, or both. Gastrointestinal signs Myelin pallor is found in the cerebral hemispheres,
include dysphagia, borborygmi, abdominal pain, extending from the periventricular area into the arcu-
cramps, recurrent nausea, vomiting, diarrhea, malab- ate fibers and into the internal capsule. The white
sorption, diverticulosis, and pseudo-obstruction. In matter is also pale in axonal preparations, but myelin
most patients, delayed gastric emptying and dys- pallor is relatively more marked. The corpus callosum
motility of small intestine, esophagus, and/or phar- is well myelinated. There are no signs of active myelin
ynx are found. The gastrointestinal dysmotility is breakdown, no necrosis, and no significant astroglio-
caused by visceral myopathy and visceral neuropathy. sis. Electron microscopy provides some evidence of
Some patients have liver problems, which may end in loosening and thinning of myelin sheaths. Similar
liver cirrhosis. Most often the liver problems are at- changes are present in the cerebellar white matter.
tributable to parenteral nutrition. Ocular signs in- Myelin pallor is also seen in the central part of the
clude chronic progressive external ophthalmoplegia, pons. The optic nerves show myelin pallor, some vac-
ptosis, and pigmentary retinopathy. Other common uolation, and some axonal loss. In the cranial nerves
clinical features include thin body habitus, short myelin pallor and endoneurial fibrosis are seen. In the
stature, tinnitus, sensorineural hearing loss, sensori- spinal cord, tracts are either normal or also show
motor peripheral neuropathy, and myopathy. Limb some myelin pallor and vacuolation, in particular in-
weakness is either distal, proximal, or diffuse.Areflex- volving the dorsal columns. Within the spinal roots
ia may be present. There is rarely evidence of CNS in- marked endoneurial fibrosis and myelin pallor are
volvement. Mental retardation is rare. The average life present, the changes being more severe in the dorsal
expectancy in MNGIE is 38 years, with a range of than in the anterior roots. Within peripheral nerves
26–58 years. variable combinations of scanty presence of myelin
Laboratory investigations may reveal elevated sheaths, abnormal internodal length, (rarely) onion
blood lactate and pyruvate. CSF protein level is often bulbs, axonal loss, excessive endoneurial fibrosis, and
raised. Nerve conduction velocity is moderately to perineural thickening are seen. Thus, the neuropathy
markedly decreased and EMG reveals signs of dener- displays mixed features of segmental demyelination
vation. These findings are consistent with a combina- and axonal degeneration. Visceral nerves show axon-
tion of demyelination and axonal loss. In addition, al loss.
some myopathic changes may be present in the EMG.
222 Chapter 27 Mitochondrial Neurogastrointestinal Encephalomyopathy

27.3 Pathogenetic Considerations 27.4 Therapy

MNGIE is related to mutations in the thymidine phos- No successful treatment is available. The use of coen-
phorylase gene, TP, located on chromosome 22q13.32- zyme Q, vitamin C, thiamin, riboflavin, and other
qter. Thymidine phosphorylase catalyzes the re- vitamins does not halt progression of the disease.
versible phosphorolysis of the nucleoside thymidine Treatment in MNGIE is largely supportive. Manage-
to thymine and 2-deoxy D-ribose 1-phosphate. The en- ment of the gastrointestinal and nutritional problems
zyme is likely to have an important role in nucleoside is important. Parenteral nutrition may be necessary.
homeostasis by regulating the availability of thymi- Pharmacotherapy and celiac plexus neurolysis may
dine for DNA synthesis. The forward reaction, conver- achieve symptomatic pain relief in selected cases.
sion of thymidine to thymine, is favored under physi-
ological conditions, and a defect in thymidine phos-
phorylase leads to accumulation of thymidine. In nor- 27.5 Magnetic Resonance Imaging
mal cells, thymidine is either degraded to thymine by
thymidine phosphorylase or salvaged to deoxythymi- In MNGIE CT reveals diffuse hypodensity of cerebral
dine monophosphate by thymidine kinase and reuti- and cerebellar white matter. MRI shows diffuse high
lized for DNA synthesis through nucleotide pools. Be- signal intensity of cerebral and cerebellar white mat-
cause the thymidine salvage pathway is the only avail- ter on T2-weighted and FLAIR images, and usually
able metabolic pathway for thymidine in MNGIE pa- but not invariably sparing of the U fibers and corpus
tients, a large amount of thymidine should be salvaged callosum (Figs. 27.1 and 27.2). The thalami and basal
to deoxythymidine monophosphate and subsequently ganglia may display patchy signal abnormalities
converted to deoxythymidine triphosphate. The accu- (Fig. 27.1), but are spared in other patients (Fig. 27.2).
mulation of thymidine is therefore likely to alter nu- The internal capsule, external capsule, brain stem,
cleoside and nucleotide pools. Mitochondria have sep- and middle cerebellar peduncles may be involved as
arate and independently regulated nucleotide pools well. In some patients the white matter abnormalities
and distinct thymidine kinase. Mitochondria depend on MRI are more limited in extent and involve the
more on the thymidine salvage pathway than on the de periventricular white matter most prominently.
novo synthetic pathway. Mitochondrial DNA con-
stantly replicates, even in quiescent cells. Therefore a
constant supply of nucleotides is essential for the
maintenance of the mitochondrial genome. Because of
these unique properties of mitochondria, the imbal-
ance in nucleotide pools probably affects mitochon-
drial DNA more adversely than nuclear DNA, impair-
ing mitochondrial DNA replication, repair, or both,
and resulting in mitochondrial DNA depletion and
multiple deletions.
Thymidine phosphorylase also has extracellular
functions and is involved in angiogenesis and cell
trophism. Thymidine phosphorylase is also called en-
dothelial cell growth factor 1, because of its angio-
genetic properties, and gliostatin, to denote its in-
hibitory effects on glial cell proliferation. MNGIE pa-
tients do not have vascular problems, suggesting that
decreased thymidine phosphorylase activity does not
interfere with normal angiogenesis.
The nature of the nervous system pathology in
MNGIE is enigmatic. In both the CNS and PNS,
myelin and axons are involved, but myelin more so
than axons. It is remarkable that in PNS histopatho-
logical abnormalities are associated with evident
signs of dysfunction (peripheral polyneuropathy, vis-
ceral neuropathy), whereas patients rarely have overt Fig. 27.2. A 17-year-old patient with MNGIE. Note the diffuse-
signs of CNS dysfunction. This and the nature of the ly high signal intensity of the cerebral white matter on this T2-
histopathological findings constitute arguments for weighted MR image. The corpus callosum, internal capsule,
abnormal myelination (dysmyelination) rather than and optic radiation are spared. From Simon et al. (1990), with
demyelination. permission
27.5 Magnetic Resonance Imaging 223

Fig. 27.1. The FLAIR images of a 47-year-old patient with thalami, and brain stem contain areas of abnormal signal. The
MNGIE show diffuse signal abnormalities of the cerebral white cerebellar white matter is not affected. From Labauge et al.
matter with sparing of the corpus callosum.The basal ganglia, (2002), with permission
Chapter 28

Leigh Syndrome
and Mitochondrial Leukoencephalopathies

28.1 Clinical Features quinone oxidoreductase) deficiency, complex II (suc-


and Laboratory Investigations cinate dehydrogenase, succinate:ubiquinone oxido-
reductase) deficiency, complex IV (cytochrome c oxi-
Leigh syndrome, also called subacute necrotizing dase) deficiency, and defects in subunit 6 of ATP syn-
encephalomyelopathy, is a neurodegenerative disor- thase (complex V). Most of these defects may also
der mainly occurring in infancy and childhood. The lead to extensive leukoencephalopathy. A few patients
disease often starts before 1 year of age and leads to with Leigh syndrome harbor a point mutation in the
death in months or years. Juvenile and adult-onset tRNA gene encoding lysine, usually associated with
forms have also been described. In most cases the dis- MERRF (myoclonus epilepsy and ragged red fibers),
ease has an autosomal recessive inheritance; in some or have a mitochondrial DNA depletion.
cases inheritance is maternal or X-linked. Both sexes Pyruvate dehydrogenase complex deficiency results
are affected, but among infants there is a 3:2 male in a wide spectrum of neurological disorders. Patients
predominance. The course can be acute, subacute, may have a neonatal or early-infantile-onset severe
episodic, or chronically progressive. Generally, the encephalopathy with profound lactic acidosis and
later the onset, the slower the progression of the dis- early death. Some patients have a neurodegenerative
ease. course of the disease with an infantile or childhood
Although Leigh syndrome is a multisystem disor- onset and milder lactic acidosis (often typical of
der, the clinical picture is dominated by signs of CNS Leigh encephalopathy). At the mild end of the spec-
dysfunction. In patients with neonatal and infantile trum patients have mild, intermittent ataxia and nor-
onset, frequent signs are respiratory problems (irreg- mal intelligence. In patients with pyruvate dehydro-
ular respiration, apnea, sighing, and hyperventila- genase complex deficiency, worsening may be pro-
tion), ocular abnormalities (strabismus, bizarre eye voked by infections. Peripheral neuropathy has been
movements, external ophthalmoplegia, ptosis, optic reported in patients with a Leigh-like presentation
atrophy, nystagmus, loss of vision, impaired pupillary and patients with intermittent ataxia. Patients with a
reaction, retinal pigmentary degeneration), hypoto- neonatal presentation often have dysmorphic fea-
nia, pyramidal signs (spastic paresis, hyperreflexia, tures, including a broad nasal bridge, upturned nose,
extensor plantar reflexes), weakness, easy fatigability, micrognathia, low-set and posteriorly rotated ears,
and feeding problems (anorexia, difficulty in swal- short fingers and arms, simian creases, hypospadias,
lowing or sucking, vomiting, weight loss, and retard- and anteriorly placed anus. Most patients have a de-
ed growth). Episodes of lethargy, seizures, deafness, fect in the E1a subunit of the pyruvate dehydrogenase
renal tubular dysfunction, and cardiac problems (car- complex, with an X-linked mode of inheritance.
diomyopathy and disturbances of cardiac rhythm Females can also be symptomatic, depending on the
with periods of tachycardia and bradycardia) may al- pattern of X inactivation. In fact, the number of
so be present. The same problems are frequent in lat- affected females is approximately equal to the num-
er-onset forms of the disease, in addition to mental ber of affected males, consistent with a high rate of
and motor retardation or deterioration, exercise in- manifestations of the disease in heterozygous fe-
tolerance, cerebellar signs (ataxia, dysarthria), and males. Male patients tend to have a more severe phe-
extrapyramidal signs (rigidity, hypokinesia, chorea, notype, but symptomatic females may also have a
athetosis, myoclonus, tremor, ballismus). Sometimes neonatal-onset devastating encephalopathy. Most de-
there are signs of a peripheral polyneuropathy. In cas- fects in the E1a gene seem to originate in the germline.
es of acute onset, coma and convulsions, sometimes Isolated complex I deficiency most often leads to
status epilepticus, may dominate the clinical picture. clinical symptoms in the neonatal or infantile period
Causes of death are neurogenic disturbances of respi- or early childhood, but onset may also be later. There
ration, status epilepticus, sudden coma, pneumonia, are a great variety of clinical presentations. Often
hyperpyrexia, and cardiac problems. complex I deficiency is a multisystem disorder with
Leigh syndrome is caused by a number of inborn fatal outcome. Up to 50% of patients with complex I
errors of energy metabolism. Frequent causes are deficiency present with Leigh syndrome or Leigh-like
pyruvate dehydrogenase complex deficiency, com- disease. Other commonly observed phenotypes are
plex I (NADH coenzyme Q reductase, NADH:ubi- cardiomyopathy, fatal infantile lactic acidosis, macro-
28.2 Pathology 225

cephaly with leukoencephalopathy, unspecified en- Since Leigh syndrome is a serious condition, pre-
cephalopathy, and myopathy. natal diagnosis is important for families. In most
Complex II deficiency may lead to Leigh syndrome, cases of pyruvate dehydrogenase complex deficiency,
diffuse leukoencephalopathy, late-onset ataxia and the defect is in the E1a subunit and the condition is
optic atrophy, myopathy with exercise intolerance, X-linked. Mutation analysis is the preferred method
and isolated cardiomyopathy. for prenatal diagnosis. Affected male fetuses are like-
Cytochrome-c oxidase deficiency is associated with ly to have a phenotype similar to that of previous
a wide range of clinical phenotypes including Leigh affected male siblings. The problem is that the clinical
syndrome, leukoencephalopathy, unspecified en- presentation of an affected female cannot be predict-
cephalopathy, fatal infantile lactic acidosis, hyper- ed, since there is no way of assessing the X chromo-
trophic cardiomyopathy and myopathy, isolated my- some inactivation pattern in the fetal brain. In all
opathy, reversible cytochrome-c oxidase deficiency mitochondrial defects with an autosomal recessive
confined to skeletal muscle, motor neuron disease, mode of inheritance, DNA-based prenatal diagnosis
spinocerebellar syndrome, myoglobinuria, hepatic is possible as soon as the basic defect is known in the
failure, ketoacidotic coma, and renal tubulopathy. family. Prenatal diagnosis may also be considered if
Mutations in the mitochondrial gene encoding the complex I deficiency is expressed in both skeletal
subunit 6 of ATP synthase are associated with two muscle and skin fibroblasts to rule out tissue speci-
main phenotypes: the NARP syndrome (NARP stand- ficity and if no mitochondrial DNA defects have been
ing for neurogenic weakness, ataxia, and retinitis pig- established or suspected. The situation is much more
mentosa, or neuropathy, ataxia, and retinitis pigmen- complicated for mitochondrial DNA mutations. The
tosa) and maternally inherited Leigh syndrome. mother carrying a mitochondrial DNA mutation
Other clinical features of NARP include mental retar- conveys the mutation to all her offspring. The clinical
dation, dementia, seizures, behavioral problems, sen- phenotype of these children is mainly determined by
sorineural deafness, and proximal muscle weakness. the load of mutated mitochondrial DNA in different
The severity of the phenotype is correlated with the tissues. As a consequence of the heteroplasmy, a
load of the heteroplasmic mutation. Symptoms usual- chorionic villus sample may not be representative of
ly appear when mutant mitochondrial DNA exceeds the level of mutant mitochondrial DNA in the em-
60%; retinal-dystrophy-related visual loss is the most bryo. In addition, the level of mutant mitochondrial
prevalent symptom in the 60–75% range of mutant DNA may be different for different tissues within the
mitochondrial DNA; full-blown NARP syndrome embryo and change over time.A reliable prediction of
usually occurs at between 75% and 90% heteroplas- the phenotype is therefore impossible. The mutations
my; whereas Leigh syndrome usually occurs at mu- in the ATP synthase 6 gene are an exception. The dis-
tant mitochondrial DNA levels above 90%. In a few tribution of mutant load among tissues is generally
patients retinal dysfunction occurs at mutant loads uniform in patients, lacking the skewed segregation
even lower than 60% and manifests in an age-related seen in other mitochondrial DNA mutations, and
fashion. Mutant loads tend to increase from mother to there is a good genotype–phenotype correlation.
child, most frequently with a very rapid “leap” toward These factors make it possible to provide reliable
mutant homoplasmy. In some families, the mutation genetic counseling and prenatal diagnosis.
can only be demonstrated in the patient and not in
the mother, other children, or maternal relatives, sug-
gesting the possibility of a de novo mutation during 28.2 Pathology
oogenesis or after fertilization, or a germline muta-
tion in the mother. The brunt of histopathological abnormalities in Leigh
Laboratory investigations in Leigh syndrome re- syndrome is borne by the central gray matter. The
veal blood levels of lactate and pyruvate to be typical- most consistent site of lesions is the brain stem gray
ly but not invariably elevated. In CSF, lactate and matter. The lesions are usually bilateral, although not
pyruvate are usually elevated. CSF protein is in- necessarily symmetrical. They are sharply delineated
creased in about half of the patients. EEG shows nor- and not confined to the gray matter structures but of-
mal findings or nonspecific abnormalities including ten spread into the white matter. Preferential sites of
diffuse or focal slowing and epileptic phenomena. affection are the periaqueductal region and brain
EMG is either normal or shows signs of denervation stem tegmentum, posterior colliculi, substantia nigra,
or signs of a myopathy. Nerve conduction velocity is floor of the fourth ventricle, red nuclei, inferior oli-
either normal or reduced. On biochemical analysis of vary nuclei, dentate nuclei, putamen, caudate nucleus,
intact mitochondria in muscle biopsy tissue, variable and globus pallidus. Thalamus, hypothalamus, and
defects are encountered. Analysis of mitochondrial subthalamic nuclei may also be involved, but less
DNA may reveal mutations, whereas in some other often. In the spinal cord, lesions are mainly located in
cases defects in nuclear genes are encountered. the anterior horns, dorsal columns, and pyramidal
226 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

tracts. Lesions rarely occur in the cerebral or cerebel- (NADH coenzyme Q reductase) deficiency, complex
lar cortex or mammillary bodies. Exceptional cases II (succinate dehydrogenase) deficiency, complex IV
with predominant cerebral and cerebellar cortical (cytochrome-c oxidase) deficiency, and subunit 6 of
damage have been described. Microscopic examina- ATP synthase (complex V) deficiency. Rarely, patients
tion of the lesions shows a marked sponginess with with Leigh syndrome harbor a point mutation in the
loosening and rarefaction of the neuropil. There is a tRNA gene encoding lysine, usually associated with
characteristic intense capillary proliferation.Astrocy- MERRF (myoclonus epilepsy and ragged red fibers),
tosis and microglial proliferation are present and or mitochondrial DNA depletion.
macrophages may occur. Nerve cells are remarkably The pyruvate dehydrogenase multienzyme com-
well preserved, although there may be some nerve cell plex catalyzes the thiamine-dependent oxidative de-
loss. The spongy lesions contain numerous vacuoles, carboxylation of pyruvate to acetyl CoA in the mito-
enclosed by single or double membranes. Myelin chondrial matrix. The complex contains three catalyt-
splitting may contribute to the vacuolation. Cavita- ic components: pyruvate dehydrogenase (E1), di-
tion and tissue collapse is the end result in most hydrolipoyl transacetylase (E2), and dihydrolipoyl
severe lesions. dehydrogenase (E3); two regulatory components,
In the majority of the cases the white matter is well E1-kinase and E1-phosphatase; and protein X, a dihy-
preserved, but sometimes there is also extensive drolipoamide dehydrogenase binding protein, neces-
involvement of cerebral and cerebellar white matter, sary for proper interaction between the E2 and E3
often with sparing of the corpus callosum and inter- components. The E1 subunit is a heterotetramer com-
nal capsule. The white matter abnormalities are char- posed of two a and two b subunits. The E1 subunit
acterized by sponginess, deficient myelin formation, contains a thiamin pyrophosphate binding site that is
myelin loss, abundant presence of lipid-laden shared by the a and b subunits. The gene for the E1a
macrophages, marked capillary proliferation, promi- subunit, PDHA1, is located on the X chromosome.
nent gliosis, and eventually also axonal loss. In all There is an autosomal counterpart located on chro-
areas there is a gradient of damage, so that myelin mosome 4. The E1b subunit gene, PDHB, is located on
sheaths and dendrites degenerate before axons and chromosome 3. The E2 subunit is encoded by a gene
cell bodies do. In some patients the affected white on chromosome 3. The gene for the E3 subunit, PHE3,
matter is partially cystic, sometimes even extensively is located on chromosome 7. The gene for protein X,
cavitated. The optic nerves and tracts are often affect- PDX1, is located on chromosome 11. Pyruvate dehy-
ed by demyelination and gliosis. drogenase deficiencies are most often associated with
In patients with a neonatal-onset encephalopathy mutations in the gene that encodes the E1a subunit of
related to pyruvate dehydrogenase complex deficien- the complex. Primary defects in the other genes en-
cy, neuropathological findings are dominated by coding other subunits of the complex are very rare.
signs of dysgenesis of the CNS, with agenesis of the Consequently, transmission of pyruvate dehydroge-
corpus callosum, dilatation of the ventricular system, nase deficiency occurs most often in an X-linked
dysplasia and ectopia of the inferior olivary nuclei, fashion, but sometimes in an autosomal recessive
dysplasia of the dentate nuclei, absence or hypoplasia fashion.
of the medullary pyramids, periventricular neuronal Complex I (NADH coenzyme Q reductase, NADH:
heterotopias, and delayed or deficient myelination. ubiquinone oxidoreductase) represents the largest
Degenerative findings, which may also be present, in- complex of the mitochondrial electron transfer chain
clude gliosis and cystic degeneration of the white and consists of at least 35 nuclear-encoded subunits
matter and vascular proliferation in the white matter (NDUFV1–3, NDUFA1–10, NDUFAB1, NDUFB1–10,
and striatum. In patients with pyruvate dehydroge- NDUFS1–8, NDUFC1–2, and a 17.2-kDa subunit), and
nase complex deficiency and intermittent ataxia, neu- 7 mitochondrial-encoded subunits (ND1–6, ND4L).
ropathological findings consist of atrophy of cerebel- The overall function of the complex is to pass elec-
lar structures. trons from NADH to ubiquinone while pumping hy-
In the sural nerve, signs of demyelination and re- drogen ions out of the mitochondrial matrix into the
myelination have been found as well as loss of myeli- inner membrane space. Complex I deficiency has an
nated and unmyelinated axons. Ragged red fibers are autosomal recessive or maternal mode of inheritance.
found in muscle tissue of some patients. Complex II, or succinate:ubiquinone oxidoreduc-
tase, consists of a flavoprotein and an iron–sulfur pro-
tein, which together constitute the soluble enzyme
28.3 Pathogenetic Considerations succinate dehydrogenase. In addition, there are two
membrane-anchored proteins. It catalyzes the oxida-
Leigh syndrome is caused by a number of inborn er- tion of succinate to fumarate in the Krebs cycle and
rors of energy metabolism. Frequent causes are pyru- carries electrons to the ubiquinone pool of the respi-
vate dehydrogenase complex deficiency, complex I ratory chain. All four subunits of the complex are en-
28.4 Therapy 227

coded by nuclear DNA. Complex II deficiencies have drome. These mutations have a maternal mode of in-
an autosomal recessive mode of inheritance. heritance.
Complex IV, or cytochrome-c oxidase, catalyzes A few patients with Leigh syndrome were found to
the transfer of electrons from reduced cytochrome c harbor a point mutation in the mitochondrial tRNA
to molecular oxygen. It is composed of 13 subunits, 10 gene for lysine,A8344G or G8363A, usually associated
of which are encoded by nuclear genes and 3 by mito- with MERRF (myoclonus epilepsy and ragged red
chondrial genes. The catalytic core of the complex fibers).
consists of the proteins COX I, COX II, and COX III, All defects underlying Leigh syndrome affect ener-
encoded by the mitochondrial genes COI, COII, and gy metabolism. There is a striking clinical and mor-
COIII, respectively. Heteroplasmic mutations in these phological similarity between Leigh syndrome and
three genes have been found in cytochrome c defi- thiamine deficiency (beriberi). Thiamine is part of
ciency. Additionally, mutations in the mitochondrial the pyruvate dehydrogenase, ketoglutarate dehydro-
tRNA gene for tryptophan and the tRNA gene for genase, and branched-chain keto acid dehydrogenase
isoleucine may underlie a selective cytochrome-c complexes, and deficiency leads to a disturbance in
oxidase deficiency. In these rare patients with a muta- oxidation of pyruvate and consequently to energy
tion in mitochondrial DNA, the disease has a mater- failure. The only histopathological differences be-
nal mode of inheritance. Autosomal recessive inheri- tween thiamine deficiency and Leigh syndrome are
tance is much more common for cytochrome-c oxi- that in thiamine deficiency the mammillary bodies
dase deficiency. Despite intensive studies, mutations are mostly involved and the substantia nigra is
have not been identified in any of the 10 nuclear-en- not, whereas in Leigh syndrome it is the substantia
coded genes in patients with an autosomal recessive nigra that is often involved and the mammillary
cytochrome-c oxidase deficiency. In order to assem- bodies rarely are. These differences, however, are not
ble a functional cytochrome-c oxidase complex, addi- absolute.
tional nuclear-encoded proteins, assembly factors, are
necessary, including SURF1, SCO1, SCO2, COX10,
COX11, COX15, and COX17. Mutations in SURF1, 28.4 Therapy
SCO1, SCO2, COX10, and COX15 have been identified
in patients with cytochrome-c oxidase deficiency. The Therapeutic success is limited in Leigh syndrome. An
genetic defect in the majority of the patients with important problem is that most of the brain damage
cytochrome-c oxidase deficiency is still unknown. is irreversible. Supportive care is important. Other
Mutations in SURF1 most often lead to Leigh syn- possible therapeutic strategies include the removal of
drome, but may also lead to a severe leukoen- toxic metabolites, administration of artificial electron
cephalopathy. Mutations in COX10 have been de- acceptors, administration of cofactors, administra-
scribed in association with leukoencephalopathy and tion of radical scavengers, and dietary interventions.
tubulopathy. However, mutations in SURF1 and L-carnitine supplementation may have a nonspe-
COX10 may also be associated with other phenotypes. cific beneficial effect, in particular if toxic organic
Recently, mutations in the gene LRPPRC have been acid intermediates are present. Dichloroacetate has
found in patients with cytochrome-c oxidase defi- been used to lower blood and CSF lactate levels. Vari-
ciency and Leigh syndrome (the so-called French– able favorable results have been reported following
Canadian subtype). The precise function of LRPPRC the use of riboflavin (vitamin B2), nicotinamide,
protein is presently not known. coenzyme Q10, vitamin C and menadione (vitamin
Defects in subunit 6 of ATP synthase (complex V of K3) in respiratory chain defects.
the respiratory chain) are most frequently caused by Thiamine treatment is effective in some patients
the T8993G mutation in the mitochondrial DNA. The with pyruvate dehydrogenase complex deficiency.
same mutation is associated with NARP. Defective The most rational therapeutic strategy in pyruvate
catalytic properties of the enzyme complex may re- dehydrogenase complex deficiency is the use of a ke-
sult from an impairment of the proton transport or togenic diet. Oxidation of fatty acids and ketone bod-
from impaired coupling of proton translocation with ies provides alternative sources of acetyl-CoA not de-
ATP synthesis. There is a clear-cut reduction in the rived from pyruvate. This acetyl-CoA enters the citric
rate of ATP synthesis in patient cells with a high load acid cycle, thus bypassing the block at the level of
of mutant mitochondrial DNA. The T8993C mutation pyruvate dehydrogenase. Dichloroacetate, a structur-
is observed less frequently and seems to be associat- al analogue of pyruvate, inhibits E1 kinase, thereby
ed with a somewhat less severe phenotype and a less keeping any residual E1 in its active, dephosphorylat-
severe reduction in the rate of ATP synthesis. The ed, form. Despite these therapeutic interventions, the
T9176C and T8851C mutation in the same gene have outcome in patients with serious neurological disease
also been found in some patients with Leigh syn- is generally poor.
228 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

28.5 Magnetic Resonance Imaging behind it. In other neonates, these dysgenetic changes
are not seen, but the white matter has an abnormal
Irrespective of the underlying defect, the most com- and swollen appearance and there may be multiple
monly reported abnormalities on CT and MRI in subependymal cysts. In patients with episodic cere-
Leigh syndrome involve the basal nuclei and brain bellar ataxia, cerebellar atrophy may be seen.
stem (Figs. 28.1, 28.3, 28.9, 28.10, 28.14–28.16). The Patients with isolated complex I deficiency may
putamen and caudate nucleus are the most frequent- present with extensive cerebral leukoencephalopathy
ly affected, but the globus pallidus, subthalamic nu- with initial swelling of the abnormal white matter and
cleus, dentate nucleus, substantia nigra, tegmentum followed by macrocystic degeneration (Figs. 28.4 and
of the pons, periaqueductal gray, red nucleus, the 28.5). Focal areas of contrast uptake may be present
medulla, and other brain stem structures are also fre- (Figs. 28.5). A periventricular rim of white matter is
quently involved. The colliculi, thalamus, hypothala- sometimes spared. The corpus callosum is often in-
mus, and cortex are less often involved. Although the volved, usually most seriously in its posterior part
lesions are often symmetrical, they may also be asym- (Figs. 28.5). The cerebral or cerebellar cortex may be
metrical. These lesions may be swollen during the abnormal in signal with subsequent development of
acute stage, may improve and become smaller, or may cortical atrophy (Fig. 28.5). There may also be lesions
develop into focal atrophy or cystic lesions. During in the basal ganglia, thalamus, cerebellum, and brain
the acute stage, contrast enhancement may be pre- stem. In other patients there are cerebral white matter
sent. Generalized cerebral and cerebellar atrophy has abnormalities but they are less impressive and not
also been reported, and some of these patients devel- cystic.
op secondary subdural effusions. Depending on the In complex II deficiency extensive cerebral white
onset of the clinical symptomatology, myelination matter abnormalities may occur in the presence or
may be delayed or totally deficient. absence of basal ganglia and thalamus lesions
Incidentally, focal or, more often, diffuse white (Figs. 28.6–28.8). The U fibers tend to be spared. The
matter abnormalities are seen on MRI, involving the corpus callosum may be involved in the process, often
cerebral white matter and often also the cerebellar more seriously in its posterior part (Figs. 28.6 and
white matter (Figs. 28.4–28.8 and 28.10–28.13). In 28.7). Focal areas of contrast enhancement may be
some patients, the white matter disease is most present (Fig. 28.6). The abnormal white matter may
prominent in the subcortical white matter; in some be partially cystic (Figs. 28.6 and 28.8). Additional
patients the white matter disease is mainly periven- brain stem white and gray matter lesions may be pre-
tricular with sparing of the U fibers, whereas in other sent. The cerebellar white matter may also be in-
patients the cerebral white matter abnormalities are volved.
diffuse. Small cysts (or, in some patients, large cysts) In cytochrome-c oxidase deficiency, diffuse or less
may develop within the abnormal white matter extensive leukoencephalopathy has been reported in-
(Figs. 28.4–28.6, 28.8, 28.10, 28.12, and 28.13) and volving cerebral white matter (Figs. 28.10–28.13). The
their presence is particularly suggestive of a mito- corpus callosum may also be involved (Figs. 28.10,
chondrial disease. The cysts are generally well delin- 28.12, and 28.13). The internal capsule, cerebellar
eated. This is in contrast with the diffuse melting away white matter, and U fibers are more variably affected.
pattern of cystic degeneration usually seen in vanish- The abnormal white matter may contain small cysts
ing white matter disease. Foci of contrast enhance- (Figs. 28.10, 28.12, and 28.13), sometimes many cysts.
ment within the abnormal white matter is another The basal ganglia, thalamus, subthalamic nucleus,
feature suggestive of a mitochondrial disorder and is, dentate nucleus, and brain stem may be normal or
for instance, not seen in vanishing white matter dis- contain lesions (Figs. 28.10 and 28.11). After contrast,
ease (Figs. 28.5, 28.6, and 28.13). The concomitant focal enhancement may be seen, probably occurring
presence of Leigh-like gray matter lesions is also sug- in necrotic lesions (Fig. 28.13).
gestive of a mitochondrial disease. In Leigh syndrome and NARP related to a muta-
The reported white matter abnormalities for each tion in the mitochondrial gene encoding the ATP syn-
basic defect are specified below: thase 6 subunit, leukoencephalopathy has not been
Pyruvate dehydrogenase complex deficiency is reported.
incidentally associated with diffuse leukoencephalo- Proton MRS of the brain usually reveals elevated
pathy. Patients with neonatal presentation of pyru- lactate in the brain of patients with Leigh syndrome,
vate dehydrogenase complex deficiency usually have most prominently in lesion areas. In diffuse cerebral
a distinct MRI pattern (Fig. 28.2) with signs of dysge- white matter abnormalities with cystic degeneration,
nesis of the brain, including hypoplasia or agenesis of the presence of highly elevated lactate is an argument
the corpus callosum and highly dilated lateral ventri- in favor of an underlying mitochondrial defect and
cles and third ventricle with a normal fourth ventri- against vanishing white matter disease. In pyruvate
cle. The cerebellum may be small with a cystic space dehydrogenase complex deficiency, additionally ele-
28.5 Magnetic Resonance Imaging 229

Fig. 28.1. An 8-year-old boy with pyruvate dehydrogenase of the corpus callosum, globus pallidus, substantia nigra, and
complex deficiency and episodic neurological deterioration dentate nucleus. The globus pallidus lesions are a typical fea-
with Leigh-like features.There are lesions in the posterior part ture of pyruvate dehydrogenase complex deficiency

vated pyruvate may be detectable at 2.37 ppm. In suc-


cinate dehydrogenase deficiency, highly elevated suc-
cinate is seen within the abnormal white matter in ad-
dition to the elevated lactate. Succinate is represented
by a resonance at 2.40 ppm. More details and illustra-
tions are found in Chap. 108.
230 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

Fig. 28.2. An 8-month-old girl with pyruvate dehydrogenase a very thin cerebral mantle, absence of the corpus callosum, a
complex deficiency and neonatal presentation. She has a cyst in the left frontal area, and a small cerebellum
severely dysgenetic brain with highly dilated lateral ventricles,

Fig. 28.3. A 3-year-old boy with isolated complex I deficiency,


related to mutations in the NDUFS7 subunit, and a Leigh-like
presentation.The T2-weighted (first and second row) and FLAIR
images (third row) show lesions in the medial thalamus, mid-
brain (including the periaqueductal gray), dentate nucleus,
and medulla. Sagittal T1-weighted images (fourth row) without
(left) and with contrast (middle and right) show enhancement
of small spots within the areas of abnormal signal
28.5 Magnetic Resonance Imaging 231

Fig. 28.3.
232 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

Fig. 28.4. A 1-year-old girl with complex I deficiency related


to a mutant NDUFV1 subunit of mitochondrial complex I
(Schuelke et al. 1999). Clinically she has a severe encephalo-
pathy and macrocephaly. The MRI reveals a diffuse leuko-
encephalopathy with macrocystic degeneration. Courtesy of
Dr. J.A.M. Smeitink, Department of Department of Pediatrics,
Nijmegen Center for Mitochondrial Disorders, University Hos-
pital Nijmegen, Nijmegen, The Netherlands
28.5 Magnetic Resonance Imaging 233

Fig. 28.5. A 19-month-old boy with isolated complex I defi- fusely abnormal. The T1-weighted images without (third row,
ciency. There are extensive, partially focal, but largely conflu- middle) and with contrast (third row, right) demonstrate that
ent cerebral white matter abnormalities. The internal capsule there are foci of contrast enhancement within the abnormal
is spared, while the periventricular rim is partially spared. The white matter, most of all the posterior part of the corpus callo-
corpus callosum is severely involved.The coronal FLAIR image sum. From Wolf et al. (2003), with permission; also courtesy of
(third row, left) shows that the abnormal white matter is par- Dr. W. Evert, Children’s Hospital Offenbach, and Dr. N. Rilinger,
tially cystic. The sagittal (first row, left) and transverse (second Central Institute for Diagnostic and Interventional Radiology,
row, right) T2-weighted images and the coronal FLAIR image Offenbach, Germany
(third row, left) demonstrate that the cerebellar cortex is dif-
234 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

Fig. 28.6.
28.5 Magnetic Resonance Imaging 235

Fig. 28.6. (continued). A 2-year-old boy with complex II defi- the corpus callosum and cerebral white matter are partially
ciency. The are extensive cerebral white matter abnormalities cystic. There is a slit in the corpus callosum over its entire
with sparing of the U fibers. The corpus callosum, posterior length. The T1-weighted images without (fourth row) and with
limb of the internal capsule, and corticospinal tracts in the contrast (fifth row) demonstrate that there are foci of contrast
brain stem are also involved. The sagittal T1-weighted image uptake in the abnormal white matter. From Brockmann et al.
and the transverse proton density images (first row) reveal that (2002), with permission
236 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

Fig. 28.7. An 11-month-old girl with complex II deficiency. cerebellar peduncles,and lateral parts of the pons.Myelination
There are extensive signal abnormalities in the periventricular is delayed for the age of the child. From Brockmann et al.
and deep cerebral white matter and corpus callosum. There (2002), with permission
are signal abnormalities in the cerebellar white matter, middle

Fig. 28.8. A 1-year-old girl with complex II deficiency. The ed cysts within the abnormal cerebral white matter, best seen
cerebral white matter is diffusely abnormal. The corpus callo- on the sagittal T1-weighted images (first row) and axial FLAIR
sum,posterior limb of the corpus callosum,corticospinal tracts images (fourth row). Courtesy of Dr. A. Bizzi, Department of
in the brain stem, middle cerebellar peduncles, and cerebellar Neuroradiology, and Dr. G. Uziel, Department of Child Neuro-
white matter are also involved. There are small, well-delineat- logy, Istituto Nazionale Neurologico C. Besta, Milan, Italy
28.5 Magnetic Resonance Imaging 237

Fig. 28.8.
238 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

Fig. 28.9. A 3-year-old boy with cytochrome-c oxidase defi- and multiple nuclei in the medulla. From Rossi et al. (2003),
ciency related to SURF1 mutations.There are signal abnormal- with permission
ities in the posterior part of the putamen, the subthalamic nu-
cleus, substantia nigra, dentate nucleus, periaqueductal gray,
28.5 Magnetic Resonance Imaging 239

Fig. 28.10. A 2.5-year-old girl with cytochrome-c oxidase defi- sule, corpus callosum, and the cerebral hemispheric white
ciency related to SURF1 mutations.There are signal abnormal- matter, posterior more than anterior.The abnormal white mat-
ities in the subthalamic nucleus, substantia nigra, periaque- ter contains small cysts. From Rahman et al. (2001), with per-
ductal gray,dentate nucleus,posterior limb of the internal cap- mission
240 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

Fig. 28.11. A 1.5-year-old boy with cytochrome-c oxidase terior limb of the internal capsule. Courtesy of Dr. M. Savoiardo,
deficiency. There are signal abnormalities in the subthalamic Department of Neuroradiology, and Dr. G. Uziel, Department
nucleus, dentate nucleus, substantia nigra, and nuclei in the of Child Neurology, Istituto Nazionale Neurologico C. Besta,
medulla. In addition, there are white matter abnormalities Milan, Italy
involving the periventricular parieto-occipital region and pos-
28.5 Magnetic Resonance Imaging 241

Fig. 28.12. Girl with cytochrome-c oxidase deficiency.The first veals atrophy of the affected white matter. The coronal FLAIR
MRI was obtained when she was aged 2.5 years (first row). images (third row) confirm the partial cystic degeneration.The
These proton density images show extensive signal abnormal- deep cerebellar white matter is also affected.Courtesy of Dr.M.
ities involving all cerebral white matter including the corpus Savoiardo, Department of Neuroradiology, and Dr. G. Uziel, De-
callosum, but sparing the U fibers. The images suggest some partment of Child Neurology, Istituto Nazionale Neurologico
cystic degeneration, with a lower signal in some white matter C. Besta, Milan, Italy
areas. Follow-up MRI 10 years later (second and third row) re-
242 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

Fig. 28.13. Boy with isolated complex I deficiency. The first white matter is partially involved (first row, right). The second
MRI (first row; second row, left and middle) was obtained at MRI (second row, right, and third row) was obtained after 3
presentation at 3 years of age. Note the extensive signal ab- months and shows progression of the white matter abnormal-
normalities in the posterior cerebral white matter and posteri- ities. Only the U fibers are still spared, more so in the anterior
or part of the corpus callosum. The abnormal white matter is than the posterior part of the brain. The corpus callosum is
partially cystic. There are foci of contrast enhancement within now involved throughout. From Topçu et al. (2000), with per-
the abnormal white matter (second row, middle).The cerebellar mission
28.5 Magnetic Resonance Imaging 243

Fig. 28.14. An 8-month-old boy with Leigh syndrome related putamen, globus pallidus, medial thalamus, substantia nigra,
to a mutation in the mitochondrial gene encoding the ATP and periaqueductal gray. Myelination is delayed, but there are
synthase 6 subunit. There are lesions in the caudate nucleus, no white matter lesions
244 Chapter 28 Leigh Syndrome and Mitochondrial Leukoencephalopathies

Fig. 28.15. A 2.5-year-old girl with Leigh syndrome related to an ATP synthase 6 subunit mutation. There is severe cerebral and
cerebellar atrophy. Myelination is delayed. Faint lesions are visible in the basal ganglia, midbrain, and pontine tegmentum

Fig. 28.16. A 38-year-old man with NARP and an ATP synthase pallidus. In addition, the cerebellum is atrophic. There are no
6 subunit mutation. There are lesions in the substantia nigra, white matter lesions
caudate nucleus, putamen, and, most prominently, the globus
Chapter 29

Pyruvate Carboxylase Deficiency

29.1 Clinical Features onyl-CoA carboxylase is normal, excluding multiple


and Laboratory Investigations carboxylase deficiency. DNA confirmation is possi-
ble. Prenatal diagnosis can be performed by enzyme
Pyruvate carboxylase deficiency (PCD) is a rare dis- assessment in chorionic villi samples or cultured am-
order with autosomal recessive inheritance. General- niocytes. In families with known mutations in the
ly, three clinical and biochemical phenotypes of pyruvate carboxylase gene, prenatal diagnosis using
isolated pyruvate carboxylase deficiency can be dis- DNA techniques is also possible.
tinguished. The group A or North American form or
infantile form is less severe than the group B pheno-
type. Patients become symptomatic between 2 and 5 29.2 Pathology
months of age with developmental delay, failure to
thrive, apathy, hypotonia, spasticity, ataxia, nystag- In all reported cases, whether of the North American
mus, and convulsions. Episodes of vomiting, tachy- or the French phenotype, the brunt of abnormalities
pnea, tachycardia, ataxia, and lactic acidosis occur, is borne by the white matter of the CNS. The brain
precipitated by metabolic or infectious stress. Some may be swollen and the white matter may be friable
patients die in the infantile period, while most of the and edematous. The hemispheric white matter is
survivors are grossly retarded. The group B or French often grossly cystic, the cysts being located either in
form or neonatal form presents in the neonatal peri- the periventricular or in the deep white matter.
od with severe lactic acidemia and is clinically char- Subependymal cysts may also occur. Cystic degenera-
acterized by failure to thrive, anorexia, vomiting, tion tends to be symmetrical, but is not always sym-
hepatomegaly, weak cry, convulsions, stupor, hypore- metrical in detail. On microscopic examination dif-
activity, irritability, hypotonia, tachypnea, dyspnea, fuse myelin paucity, sponginess, and gliosis of the
respiratory failure, and subsequently hypertonia, ex- white matter are found, involving cerebral and cere-
trapyramidal tract signs, and severely retarded devel- bellar white matter and sometimes also the base of
opment. Some infants have macrocephaly. Almost all the pons. The myelin paucity is variably described as
children die within the first few months of life. The hypomyelination or demyelination. Probably, both
third variant is milder and has been described in a are important. Perivascular accumulation of foamy
few exceptional patients who, despite episodic meta- macrophages is an argument in favor of a component
bolic derangement with ketoacidosis from infancy of active myelin breakdown. The white matter is de-
onwards, have normal or near normal motor and creased in volume in cases of longer duration result-
mental development. These patients survive at least ing in enlargement of the ventricular system. The cor-
up to the third decade. Formerly, Leigh syndrome was pus callosum is thin.
thought to represent another clinical phenotype of The condition of the gray matter is variable, but
pyruvate carboxylase deficiency, but this could not be gray matter pathology does not dominate. In some
confirmed. cases all gray matter structures are completely nor-
In the North American phenotype lactic acidemia mal. Some have noted depletion of neurons in the
is associated with a normal lactate to pyruvate ratio. cerebral cortex. Some have noted that the globus pal-
There is no hyperammonemia. Alanine and proline lidus, a nucleus rich in myelin, is involved in the
are elevated. In the French phenotype, laboratory in- process of myelin abnormality and loss. Some de-
vestigations reveal lactic acidemia with a highly ele- scribe cystic degeneration of the deep gray nuclei.
vated lactate/pyruvate ratio and a lowered 3-hydroxy-
butyrate/acetoacetate ratio. Ketoacidosis and moder-
ate hyperammonemia are present, and blood levels of 29.3 Pathogenetic Considerations
alanine, citrulline, proline, and lysine are elevated.
The level of aspartate is often decreased but may also Pyruvate carboxylase is a mitochondrial matrix en-
be normal. zyme, encoded by nuclear DNA. The gene, PC, is locat-
Diagnosis is established by demonstrating a defi- ed on chromosome 11q13.4–13.5. Pyruvate carboxy-
ciency in pyruvate carboxylase activity in fibroblasts, lase is a homotetramer consisting of four identical
white blood cells, or liver cells. The activity of propi- polypeptides, each with a covalently bound biotin
246 Chapter 29 Pyruvate Carboxylase Deficiency

molecule. There are four major biotin-dependent car- can be shown to be present with immunological tech-
boxylases: pyruvate carboxylase, propionyl-CoA car- niques, although deficient in activity. Presumably, the
boxylase, methylcrotonyl-CoA carboxylase, and French phenotype is related to absent or almost ab-
acetyl-CoA carboxylase. There are no known tissue- sent residual enzyme activity, whereas in the North
specific isoenzymes of pyruvate carboxylase. The en- American phenotype the residual activity is enough
zyme catalyzes the conversion of pyruvate, the end to ameliorate the most severe symptoms of pyruvate
product of the glycolysis, into oxaloacetate, a citric carboxylase deficiency. However, environmental fac-
acid cycle intermediate. It replenishes oxaloacetate tors, including intercurrent illnesses and other stress-
for the citric acid cycle. Oxaloacetate is also in equi- es, such as fasting, may contribute to determining
librium with aspartate. Oxaloacetate is utilized in the outcome.
synthesis of glucose, fat, some amino acids or their
derivatives, and several neurotransmitters. The roles
of pyruvate carboxylase in metabolism are tissue- 29.4 Therapy
specific. In gluconeogenetic tissue (liver, kidneys), it
catalyzes the first step in the synthesis of glucose from Treatment has proven to be quite difficult and dis-
pyruvate. In lipogenic tissue (liver, adipose tissue, lac- appointing in pyruvate carboxylase deficiency. Meta-
tating mammary gland, adrenal gland), it participates bolic acidosis must be corrected by bicarbonate ther-
in the export of acetyl groups, such as citrate, from the apy. Aspartate supplementation has been recom-
mitochondria to the cytosol. In the CNS, pyruvate mended to increase oxaloacetate concentrations and
carboxylase participates in the replenishment of neu- overcome aspartate depletion, and some patients
rotransmitter pools of glutamate, GABA, and aspar- have actually shown improvement. Glutamine, also a
tate. precursor of oxaloacetate, may also have a beneficial
Pyruvate carboxylase deficiency can occur as iso- effect. Citrate may result in reduction of the lactic
lated pyruvate carboxylase deficiency, caused by a acidosis. Thiamine has been advocated to stimulate
mutation in the pyruvate carboxylase gene, and as the pyruvate dehydrogenase complex but is ineffec-
multiple carboxylase deficiency related to biotin defi- tive. Administration of biotin to stimulate pyruvate
ciency. In isolated pyruvate carboxylase deficiency carboxylase has been beneficial in some patients, but
there is a defect in the conversion of pyruvate to ox- in many patients it is disappointing. Avoidance of
aloacetate. Availability of the latter metabolite is es- fasting with its attendant gluconeogenetic stimulus
sential for citric acid cycle activity. When oxaloac- is important, but high carbohydrate diets and high
etate synthesis from pyruvate is limited, aspartate is fat diets are not beneficial. Unfortunately, despite all
converted to oxaloacetate and depletion of aspartate therapeutic measures, the neurological outcome
occurs. Aspartate is an important component of the remains poor.
shuttle mechanism transferring reducing equivalents
across the mitochondrial membrane. Depletion of as-
partate leads to accumulation of reducing equivalents 29.5 Magnetic Resonance Imaging
(NADH) in the cytosol and mitochondrial NADH be-
comes more oxidized. This altered redox state results Reports on neuroimaging findings are scarce in pyru-
in an increase in the lactate to pyruvate ratio and a de- vate carboxylase deficiency. CT reveals diffuse hypo-
crease in the b-hydroxybutyrate to acetoacetate ratio. density of the cerebral white matter. The white matter
Depletion of aspartate also interferes with urea cycle may be mildly swollen. MRI demonstrates signal ab-
activity. Aspartate is a nitrogen donor for the urea cy- normalities in the cerebral and cerebellar white mat-
cle and depletion of aspartate leads to hyperammone- ter and in brain stem structures, in particular the base
mia, citrullinemia, and hyperlysinemia. Accumula- of the pons (Fig. 29.1). In some patients the white
tion of acetyl-CoA may also occur under these cir- matter degeneration is cystic. In the first few months
cumstances and result in overproduction of ketone of life, subependymal cysts may be seen (Fig. 29.1).
bodies. Despite the fact that pyruvate carboxylase is Probably depending on the stage of the disease, the
an important enzyme in gluconeogenesis, hypo- white matter has a swollen appearance (Fig. 29.1) or is
glycemia is not a consistent finding in pyruvate car- atrophic. The corpus callosum is thin. In patients with
boxylase deficiency. severe atrophy, there may be subdural hygromas and
The different phenotypes of pyruvate carboxylase hematomas. The signal abnormality of the cerebral
deficiency are related to the severity of enzyme defi- white matter is partly related to delayed myelination;
ciency. Many patients with the French phenotype progress of myelination is seen over time. The white
have no immunologically detectable enzyme at all, matter abnormalities tend to be diffuse in the early
but some have. In patients with the North American changes, but over time and with progress of myelina-
phenotype, the pyruvate carboxylase enzyme protein tion, they become more patchy and multifocal.
29.5 Magnetic Resonance Imaging 247

Fig. 29.1. The T2-weighted images of a female infant of 18 There are multiple subependymal cysts along the ventricles.
days with pyruvate carboxylase deficiency show that the cere- Signal changes are seen in the base of the pons and the cen-
bral and cerebellar white matter has a higher signal than nor- tral tegmental tracts. Courtesy of Dr. U.Wendel, Department of
mal for unmyelinated white matter, and is mildly swollen. Pediatrics, University Children’s Hospital, Düsseldorf, Germany
Chapter 30

Multiple Carboxylase Deficiency

30.1 Clinical Features early childhood, but patients with a partial bio-
and Laboratory Investigations tinidase deficiency may not develop symptoms until
adolescence. However, asymptomatic adults, both
Two inherited defects in biotin metabolism are with a partial and a profound biotinidase deficiency,
known: holocarboxylase synthase deficiency and bio- have been detected. When patients present, the symp-
tinidase deficiency, both autosomal recessive disor- toms are essentially the same, independent of age, al-
ders. A rare cause of biotin dependency is a genetic though older patients tend to have fewer symptoms.
defect in the transport of biotin. Alimentary biotin Most patients present between the ages of 6 weeks
deficiency is exceptional and may, for instance, be re- and 18 months. Most have progressive encephalopa-
lated to parenteral nutrition with a formula deficient thy, but some have episodes of acute deterioration.
in biotin, short bowel syndrome, or hemodialysis The most common symptoms are skin rash, partial or
without biotin supplementation. Excessive intake of complete alopecia, hypotonia, spasticity, develop-
avidin, an egg-white glycoprotein that binds specifi- mental delay, seizures including infantile spasms, and
cally and irreversibly to biotin, may also result in sensorineural hearing loss. In many patients the
biotin deficiency. All lead to deficiency of multiple seizures are difficult to control with antiepileptic
carboxylases: 3-methylcrotonyl-CoA carboxylase, medication. Less common features are keratocon-
propionyl-CoA carboxylase, pyruvate carboxylase, junctivitis, ataxia, fatigue, lethargy, tachypnea, apnea,
and the two isoenzymes of acetyl-CoA carboxylase. laryngeal stridor, scotomas, and loss of vision due to
The characteristic manifestations of multiple car- optic atrophy. Some patients develop feeding difficul-
boxylase deficiency consist of skin disease and neuro- ties, vomiting, diarrhea, hepatomegaly, splenomegaly,
logical abnormalities. The skin abnormalities are de- or coma. Immunological dysfunction may occur in
scribed as seborrheic or atopic dermatitis. They con- acutely ill patients. Older patients often present with
sist of nonspecific, patchy, maculopapular, scaly and optic atrophy, sensorineural hearing loss, and gait
erythematous eruptions; they occur especially in problems; some of them have a mental retardation,
moist and periorificial areas. In more severe cases but others are intellectually intact. Biotinidase defi-
lichenification, crusting, and open lesions may occur, ciency generally leads to progressive clinical symp-
that may become infected by Candida. The hair is of- toms ending in severe neurological damage or death
ten sparse and thin and partial or total alopecia may if untreated.
be present that can include the eyebrows and eyelash- Laboratory findings in affected children include
es. metabolic acidosis, mild hyperammonemia, and or-
Onset of clinical symptoms in patients with holo- ganic aciduria. Increased urinary excretion is usually
carboxylase synthase deficiency is between a few found of 3-methylcrotonylglycine and 3-hydroxyiso-
hours after birth and the age of 2 years. Most patients valerate reflecting methylcrotonyl-CoA carboxylase
present in the first days or weeks after birth, but oth- deficiency; 3-hydroxypropionate, propionylglycine,
ers present with acute metabolic derangement later, tiglylglycine, propionate, and methylcitrate reflecting
in the 2nd year of life. The symptoms may be precipi- propionyl-CoA carboxylase deficiency; and lactate,
tated by catabolism or increased dietary protein in- which likely reflects pyruvate carboxylase deficiency.
take. The most common symptoms include tachy- In holocarboxylase synthase deficiency the biochem-
pnea, stridor, skin rash, feeding difficulties, vomiting, ical abnormalities tend to be more severe and com-
hypotonia, developmental delay, tremor, ataxia, plete than in biotinidase deficiency. However, in some
seizures, irritability, lethargy, and coma. Alopecia oc- patients with holocarboxylase synthase deficiency
curs in some of the patients. Disorders of immune the biochemical abnormalities are only mild, rather
function have been observed with a decreased T-cell suggesting biotinidase deficiency. In some children
count. The disease generally leads to severe neurolog- with biotinidase deficiency, urinary organic acids are
ical damage or death if untreated. normal at presentation.
The age at onset in patients with a biotinidase de- The diagnosis of holocarboxylase synthase defi-
ficiency is usually later and more variable. Most ciency is confirmed by showing deficiency of 3-
patients with a profound biotinidase deficiency have methylcrotonyl-CoA carboxylase, propionyl-CoA
cutaneous and neurological symptoms in infancy or carboxylase, pyruvate carboxylase, and acetyl-CoA
30.3 Pathogenetic Considerations 249

carboxylase in lymphocytes or fibroblasts in a medi- 30.3 Pathogenetic Considerations


um with low biotin concentration. The enzyme activ-
ities remain severely deficient after in vitro preincu- Biotin is an essential water-soluble vitamin and is the
bation with biotin. The diagnosis of biotinidase defi- coenzyme for four carboxylases, propionyl-CoA car-
ciency is confirmed by showing decreased or absent boxylase, 3-methylcrotonyl-CoA carboxylase, pyru-
enzyme activity in serum, leukocytes, and fibroblasts. vate carboxylase, and acetyl carboxylase, which are
In biotinidase deficiency and acquired biotin defi- involved in the catabolism of several branched-chain
ciency, carboxylase activities in lymphocytes are usu- amino acids, gluconeogenesis, and fatty acid synthe-
ally decreased but may also be normal, depending on sis. Each of the four carboxylases is synthesized in an
the degree of biotin deficiency; the carboxylase activ- inactive form called apocarboxylase and becomes en-
ities increase significantly or normalize after in vitro zymatically active only when it is linked covalently to
preincubation with biotin. Plasma biotin concentra- biotin; it is then called holocarboxylase. Holocar-
tion is usually below normal in symptomatic patients boxylase synthase catalyzes the covalent binding of
with biotinidase deficiency and in acquired biotin biotin to a lysine residue in the four biotin-dependent
deficiency, but normal in holocarboxylase synthase enzymes. Humans cannot synthesize biotin and
deficiency. Prenatal diagnosis can be performed therefore derive the vitamin from dietary sources or
by enzyme assays in cultured chorionic villi and from recycling endogenous biotin. The carboxylases
amniotic fluid cells. are degraded proteolytically to amino acids and bio-
cytin (biotinyl-N-¥-lysine). Biocytin is then hy-
drolyzed by biotinidase to lysine and free biotin,
30.2 Pathology thereby recycling the vitamin for reutilization. Bio-
tinidase is also necessary for liberating biotin from
Reports on neuropathological findings are scarce. dietary protein-bound sources to its free, bioavailable
The external appearance of the brain is usually nor- form. Additionally, biotinidase has biotinyl trans-
mal. Ill-demarcated, partly necrotizing focal lesions ferase activity, in which biotin from biocytin is trans-
are found in the white matter of the cerebral hemi- ferred to other proteins. The gene encoding holocar-
spheres, thalamus, hypothalamus, subthalamic nucle- boxylase synthase, HLCS, is located on chromosome
us, hippocampus, mammillary bodies, substantia ni- 21q22.1. The gene encoding biotinidase, BTD, is locat-
gra, red nucleus, oculomotor nuclei, periaqueductal ed on chromosome 3p25.
gray matter in the midbrain, tegmentum of the pons, The multiple carboxylase deficiencies are geneti-
dorsomedial parts of the medulla oblongata, inferior cally determined or acquired disorders of biotin me-
olivary nucleus, deep cerebellar white matter, cerebel- tabolism and result in impaired activity of all four bi-
lar nuclei, and the posterior, lateral, and anterior otin-dependent carboxylases. There are two forms of
columns of the spinal cord. The lesions show rarefac- inherited multiple carboxylase deficiency: holocar-
tion, microcavitation, sponginess, capillary prolifera- boxylase synthase deficiency and biotinidase defi-
tion, and gliosis. Reactive astrocytes and foamy ciency. In holocarboxylase synthase deficiency, the
macrophages are present. Myelin is lost and neurons carboxylases remain in their inactive apo-form. In bi-
are relatively preserved. The lesions are similar to otinidase deficiency, endogenous biotin cannot be re-
those observed in Leigh syndrome and thiamine defi- cycled and dietary protein-bound biotin cannot be
ciency. released. Biocytin is lost in urine, leading to progres-
Another striking finding is vacuolation and edema sive biotin depletion. The clinical symptoms of holo-
of white matter tracts, including parts of the corpus carboxylase synthase deficiency and biotinidase defi-
callosum, fornix, the central tegmental tracts in the ciency overlap, but ophthalmological abnormalities
pons, the decussation of the medial lemniscus, the su- and hearing loss have not been observed in holocar-
perior cerebellar peduncles, the pyramids, and anteri- boxylase synthase deficiency. The symptomatology in
or columns of the spinal cord. Abnormalities involv- holocarboxylase synthase deficiency tends to be of
ing the lateral and posterior columns have also been earlier onset and more severe than in biotinidase de-
reported. Myelin loss is seen with relatively less severe ficiency, although there is a major overlap. In bio-
axonal loss. Defective myelination and gliosis of the tinidase deficiency, the level of the residual enzyme
cerebral and cerebellar white matter have also been activity correlates with the development of clinical
reported. The optic nerves may display severe loss of and biochemical abnormalities. Most patients with
myelinated fibers. profound biotinidase deficiency develop early signs
of the disease. Patients with partial biotinidase defi-
ciency may be healthy and develop symptoms only
under conditions of infection or starvation. A special
variant of the disease is related to decreased affinity
of biotinidase for biocytin, the so-called Km variant
250 Chapter 30 Multiple Carboxylase Deficiency

with a reduced maximum reaction velocity and an
elevated Km of biotinidase for biocytin. Fig. 30.1. A 1-year-old girl just diagnosed with biotinidase de-
ficiency. The child presented with psychomotor retardation,
spastic quadriplegia, and a severe seizure disorder. The MRI
30.4 Therapy shows a severe delay in myelination. The white matter of the
centrum semiovale looks spongy with many tiny cysts, which
Holocarboxylase synthase deficiency can often be could also be enlarged perivascular spaces. The subarachnoid
spaces are mildly enlarged, compatible with mild cerebral at-
treated effectively with pharmacological doses of bi-
rophy. Courtesy of Dr. Z. Patay, Department of Radiology, and
otin. The required dose of biotin is dependent on the
Dr. P.T. Ozand, Department of Pediatrics, King Faisal Specialist
severity of the enzyme defect and has to be assessed
Hospital and Research Center, Riyadh, Saudi Arabia
individually. With treatment the symptoms usually
either disappear or improve and further deterioration
can usually be prevented. An important problem is
that delayed diagnosis leads to permanent damage
that cannot be undone with treatment. Unfortunately,
some patients show only a partial response to biotin published, suggesting diffuse white matter vacuola-
or no response at all. Persistence and even progres- tion or edema.
sion of the encephalopathy despite very high doses of MRI shows variable abnormalities in untreated
biotin has been observed in some patients. It is likely multiple carboxylase deficiency, including delayed
that some defects in holocarboxylase synthase make myelination, diffuse cerebral white matter signal ab-
it unresponsive to biotin supplementation. normality and swelling, more patchy cerebral white
The results of biotin treatment in biotinidase defi- matter abnormalities, and cerebral atrophy (Figs. 30.1
ciency are even better. With treatment most or all and 30.2). The white matter abnormalities and cere-
symptoms disappear and further deterioration can be bral atrophy are at least partially reversible with treat-
prevented. The only problem is that damage already ment. These imaging abnormalities have a low diag-
acquired may not be entirely reversible. In particular nostic specificity. Signal abnormalities in central gray
auditory and visual deficits, but also intellectual im- matter structures, brain stem, and spinal cord have al-
pairment and motor deficits may persist in spite of so been reported, suggestive of a Leigh-like disease.
treatment. For this reason, neonatal screening for These may be entirely or partially reversible with
biotinidase deficiency has been instituted in many treatment. With Leigh-like abnormalities present on
countries. Early diagnosis and treatment prevent the MRI, the possibility of multiple carboxylase deficien-
onset of symptoms. Patients with a partial biotinidase cy should be considered.
deficiency and the biotinidase Km variant should also One paper by Wiznitzer and Bangert (2003) re-
be treated with biotin, especially as biotin has no ports signal changes in the spinal cord over its entire
known harmful effects. length in a patient with clinical ataxia and spastic
Prenatal diagnosis is possible. If holocarboxylase paraparesis. The abnormalities disappeared with
synthase deficiency is detected, antenatal treatment treatment.
can be started by treating the mother with biotin, pre-
venting acute neonatal symptoms. Intrauterine treat-
ment is not necessary for biotinidase deficiency,
because of the later onset of symptoms.

30.5 Magnetic Resonance Imaging

CT of the brain may show basal ganglia calcification 䊳

in untreated multiple carboxylase deficiency. Diffuse Fig. 30.2. A 1-year-old boy just diagnosed with biotinidase
hypodensity of the cerebral white matter within the deficiency. He presented with an acute metabolic crisis. The
neonatal period with disappearance of the hypoden- MRI shows delayed myelination and mildly enlarged sub-
sity after many months of treatment has been report- arachnoid spaces.There are more prominent signal abnormal-
ed. The problem is that the cerebral white matter is al- ities in the deep parietal white matter, where the white matter
ways hypodense in neonates due to lack of myelin, also looks spongy with many tiny cysts, possibly enlarged
and that disappearance of the hypodensity with on- perivascular spaces. Courtesy of Dr. Z. Patay, Department of
going myelination is a normal phenomenon. The Radiology, and Dr. P.T. Ozand, Department of Pediatrics, King
white matter does, however, look more hypodense Faisal Specialist Hospital and Research Center, Riyadh, Saudi
than normal and mildly swollen on the CT pictures Arabia
30.5 Magnetic Resonance Imaging 251
Chapter 31

Cerebrotendinous Xanthomatosis

31.1 Clinical Features opment of symptoms varies markedly in nature and


and Laboratory Investigations degree of progression, even within one family. In par-
ticular in the absence of the typical tendon xan-
Cerebrotendinous xanthomatosis (CTX) is a rare thomas, the diagnosis can be easily missed.
neurometabolic disorder with an autosomal recessive Laboratory investigations reveal normal or only
mode of inheritance. Prolonged neonatal cholestatic moderately elevated serum cholesterol but a marked-
jaundice is common (“hepatitis of infancy”). Most ly increased level of serum cholestanol. CSF protein
patients are of borderline or low intelligence from the may be increased. CSF cholestanol levels are elevated.
beginning and their school performance is poor. Low serum levels of 25-hydroxy vitamin D3 and 24,25-
Chronic, intractable, and unexplained diarrhea is pre- dihydroxy vitamin D3 may be found. The measure-
sent in about 50% of the affected children. The more ment of the serum cholestanol to cholesterol ratio
specific clinical manifestations usually appear in late has been advocated as a means of establishing the
childhood or early adolescence, or even later. The diagnosis, but elevated cholestanol and elevated
most commonly noted early manifestations of the cholestanol to cholesterol ratios are not very specific
disease include cataracts and xanthomas of tendons, and can also be found in patients suffering from var-
especially the Achilles tendons, but also the tendons ious liver diseases. A preferable method for establish-
of the quadriceps muscle, the triceps muscle, and the ing the diagnosis is to demonstrate the presence of
finger extensors. During the second or third decade, abnormal bile alcohols in the urine. The diagnosis
neurological problems gradually become manifest can be confirmed by demonstrating lack of 27-hy-
with signs of cerebellar ataxia, spastic paraparesis droxylase activity in cultured fibroblasts and by DNA
and tetraparesis, signs of dysfunction of the posterior techniques.
columns, and signs of peripheral polyneuropathy. X-ray examination may reveal swelling of the
Some patients have only signs of spinal cord dysfunc- Achilles tendons and, less frequently, of the tendons
tion with pyramidal tract and dorsal column involve- of the hamstrings, quadriceps, and finger extensors.
ment. Tendon reflexes are generally hyperactive. Vi- Calcification of these soft tissue masses may be seen.
bratory and position senses are diminished whereas Motor and sensory nerve conduction is often slowed.
the superficial sensory modalities remain relatively Evoked potentials, in particular SSEPs, are as a rule
intact. Foot deformity, in particular pes cavus, is often delayed. EEG shows diffuse slowing of background
noted. About 40% of the patients develop epilepsy activity with poorly organized theta and delta waves.
with generalized tonic–clonic seizures. In most cases Carriers of CTX can be identified by observing an
a decline of mental function occurs in the third abnormal increase in bile alcohols in their urine after
decade, but there is wide diversity in the rapidity of administration of cholestyramine, a drug that leads
the decline. Changes in personality and psychiatric to intestinal loss of bile acids and as a consequence to
problems may be present, varying from irritability, increased endogenous synthesis. Normal control per-
agitation, and aggressiveness to paranoid ideation, sons fail to produce the unusual bile alcohols. Carrier
hallucinations, and catatonia. Premature atheroscle- detection can also be performed by analysis of 27-hy-
rosis may lead to angina pectoris, myocardial infarc- droxylase activity in cultured fibroblasts or by DNA
tion, and cardiac failure. Less frequent complaints are techniques. Prenatal diagnosis is possible using the
pharyngeal and palatal myoclonus, tachylalia, mask- same techniques.
like facies, parkinsonism, bulbar and pseudobulbar
paresis with dysphagia and dysphonia, visual loss due
to optic atrophy, palpebral xanthelasmas, corneal 31.2 Pathology
lipoid arcus, generalized muscular wasting, bone
fractures due to osteoporosis, impaired lung function On external examination of the brain, mild atrophy is
due to pulmonary xanthomas, and signs of endocrine found, especially of the cerebellum. Sometimes xan-
dysfunction. In untreated cases death usually occurs thomas are seen in the choroid plexus.
between the fourth and sixth decades. On microscopic examination, the cerebral cortex
The problem with the diagnosis of CTX is that and hemispheric white matter usually appear normal.
there are no obligatory symptoms and that the devel- Sometimes gliosis and perivascular collections of
31.4 Pathogenetic Considerations 253

large mononuclear cells with foamy cytoplasm are Microscopic examination of Achilles tendon xan-
found. Occasionally, prominent loss of myelin and ax- thomas reveals islets of mononuclear cells with foamy
ons may be found at the level of the corona radiata, cytoplasm and clefts filled with crystalloid material
periventricular white matter, and globus pallidus. surrounded by multinucleated giant cells. The clefts
Mononuclear cells with foamy cytoplasm may be pre- are scattered in fan-shaped clusters without any rela-
sent in the basal nuclei and thalamus. The globus pal- tion to blood vessels. Under polarized light birefrin-
lidus especially may contain large mononuclear cells gence of the clefts is shown, suggesting presence of
with foamy cytoplasm, accompanied by demyelina- sterols. The cells filled with neutral fat are mainly pre-
tion of the fibers of the ansa lenticularis as they pass sent around blood vessels but also throughout the tis-
through the internal capsule. The optic nerves and sue.
tracts may exhibit loss of myelin and axons associat- Similar xanthomatous tissue can be found in the
ed with fibrillary gliosis and presence of perivascular lungs and bones. In liver tissue fatty lipofuscin-like
lipid-laden mononuclear cells. pigment granules have been reported in hepatocytes
The most conspicuous abnormalities are found in and Kupffer cells. Crystals are found in the cytoplasm
the cerebellum. Xanthomatous tissue sometimes re- of hepatocytes. Mitochondria are hypertrophied and
places most of the white matter. On microscopic ex- peroxisomes are increased in size and number. Pre-
amination extensive loss of myelin and axons is seen mature atherosclerosis of coronary arteries can be
within the cerebellar white matter, with most severe found.
involvement of the outflow tracts of the dentate nu-
cleus and the superior cerebellar peduncles. In the af-
fected areas many small and large cystic spaces and 31.3 Chemical Pathology
needle-like clefts are present. Large quantities of neu-
tral fat accumulate within large mononuclear cells In CTX patients the lipids stored in the brain and in
with foamy cytoplasm in the cysts and in perivascu- xanthomas consist of free and esterified cholestanol
lar spaces. The needle-shaped clefts contain crys- and cholesterol. Free cholestanol is found not only in
talline deposits with staining properties of sterols. the evidently affected areas of the brain, but also in
The crystalline deposits are surrounded by inflam- areas which appear normal on histological examina-
matory cells and reactive multinucleated foreign- tion. Cholestanol is found in myelin and also in all
body giant cells. These cells represent the tissue reac- other membrane structures in the brain, including
tion to deposition of sterols. There may be extensive cell membranes and membranes of subcellular struc-
loss of Purkinje cells and granule cells and destruc- tures. The concentration of unesterified cholesterol is
tion of the dentate and fastigial nuclei. Particularly in normal or only slightly increased. In demyelinated
the dentate nucleus, severe neuronal loss may be areas the concentrations of esterified cholestanol
found with presence of clefts containing crystalline and cholesterol are elevated. Concentrations of
lipid deposits, as well as reactive astrocytosis, focal cholesterol esters are nonspecifically elevated in
calcification, and deposition of hemosiderin pig- many actively demyelinating disorders, but esterified
ment. cholestanol is not present in any of these disorders.
In the brain stem the pyramidal tracts, medial lem-
niscus, transverse pontine fibers, and superior cere-
bellar peduncles are involved. At the higher levels of 31.4 Pathogenetic Considerations
the brain stem, in particular in the red nucleus and
substantia nigra, deposits of neutral lipids and crys- In CTX the basic defect is located in the mitochon-
talline sterols are present. Gray matter changes in the drial enzyme 27-hydroxylase. This enzyme catalyzes
brain stem include loss of neurons, particularly in the the initial steps in the side-chain cleavage of sterols.
inferior olives and other nuclei. In the spinal cord the The enzyme hydroxylates a spectrum of sterol sub-
pyramidal tracts and posterior columns demonstrate strates, including cholesterol and vitamin D3. The
loss of myelin sheaths and axons. sterol 27-hydroxylase gene, CYP27, is located on the
In the peripheral nerves extensive axonal degener- distal portion of the long arm of chromosome 2
ation is found. There may be additional signs of seg- (2q35). Many different mutations have been identi-
mental demyelination and remyelination with onion fied. There is no clear correlation between genotype
bulb formation. Histological examination of muscle and phenotype.
tissue discloses signs of denervation and reinnerva- The most important pathway for the metabolism
tion with type grouping. On electron microscopy, and excretion of cholesterol in humans is the forma-
large aggregates of mitochondria are seen, mainly in tion of bile acids. The two major bile acids, cholic acid
the subsarcolemmal region. The mitochondria show and chenodeoxycholic acid, are formed in the liver
mild morphological abnormalities such as increased and secreted in bile into the intestine. The enzymes
size and irregular cristae. involved in modifying the steroid nucleus of choles-
254 Chapter 31 Cerebrotendinous Xanthomatosis

terol are mainly located in the endoplasmic reticulum barrier may be up-regulated as well, in this way con-
and the cytosol. The enzymes involved in the side- tributing to the influx of sterols into the CNS.
chain degradation are mainly located in mitochon-
dria and peroxisomes. The major pathway for side-
chain cleavage is the 27-hydroxylase pathway. 31.5 Therapy
Deficient activity of 27-hydroxylase results in a
defect in bile acid biosynthesis. The formation of nor- Treatment in CTX aims at breaking the vicious circle
mal bile acids, in particular chenodeoxycholic acid, is of defective endogenous bile acid synthesis leading to
reduced. Large amounts of unusual C27 bile alcohols absence of negative feedback, which in turn leads to
are excreted in bile, feces, and urine. As bile acids are increased production of cholesterol, abnormal bile
involved in a feedback regulation of the hepatic cho- alcohols, and cholestanol. Treatment with oral bile
lesterol production, the decrease in bile acids leads to acids, in particular chenodeoxycholic acid, repairs
enhanced cholesterol production and excessive pro- the negative feedback and leads to decreased produc-
duction of bile alcohols. tion of cholesterol, cholestanol, and abnormal bile
Cholestanol is the 5a-dihydro derivative of choles- alcohols. Assessment of serum cholestanol and uri-
terol. It normally represents about 0.1–0.3% of cho- nary bile alcohols can be used to monitor treatment.
lesterol in tissues and plasma. In CTX cholestanol is Once treatment starts, the diarrhea ceases. The pro-
increased 10- to 100-fold, so that it accounts for 2% of gression of CNS damage is retarded or halted. Pa-
plasma and tissue sterols with even greater enrich- tients receiving therapy with chenodeoxycholic acid
ment in the brain (20–50%), tendon xanthomas have been reported to show reversal of their neuro-
(10%), and bile (10%). There is evidence that the logical disability, with clearing of dementia and im-
increased synthesis of cholestanol is caused by the in- proved motor function. There is also evidence of im-
creased utilization of bile intermediates as precursors proved results of paraclinical tests, such as nerve con-
for cholestanol. The bile intermediates accumulating duction velocity, evoked responses, EEG, and CT.
in CTX may be shunted into the cholestanol pathway. Combined treatment with chenodeoxycholic acid
Like cholesterol, cholestanol is transported by low- and 3-hydroxy-3-methylglutaryl coenzyme A reduc-
density lipoproteins (LDL) and high-density lipo- tase inhibitors such as simvastatin, lovastatin, or
proteins (HDL). Despite the enhanced production of pravastatin – inhibitors of cholesterol synthesis, leads
both sterols, plasma LDL concentrations are low and to a more pronounced reduction of cholesterol and
HDL cholesterol levels are also diminished. The role cholestanol than treatment with chenodeoxycholic
of LDL is to transport cholesterol from the liver to acid alone. Their long-term clinical efficacy of this
peripheral tissues. LDL turnover is exceedingly rapid combined treatment has still to be proven.
in CTX. The catabolism of LDL by the augmented Some patients experience a more marked improve-
expression of LDL receptors is sufficiently great to ment than others. As the effects of therapy depend
maintain low plasma concentrations, despite en- largely on the extent of irreversible structural damage
hanced cholesterol production. The role of HDL is to to nervous tissue, early diagnosis and early treatment
transport cholesterol from peripheral tissues to the are important. If the disease is detected in early child-
liver. HDL cholesterol levels are subnormal in many hood, treatment of the patient not yet clinically
CTX patients. This may account for the accumulation affected can prevent occurrence of complaints. It is a
of tissue sterols in atheromas and xanthomas by hin- treatment for life.
dering reverse sterol transport.
Neurophysiology and histopathology suggest that
axonal degeneration is primary in CTX, although 31.6 Magnetic Resonance Imaging
there may also be a component of primary myelin
loss. The precise pathophysiological mechanisms are CT scan of the brain has been reported to show cere-
unclear. Evidently, the deposition of cholestanol and bellar hypodensity and in some cases also moderate
cholesterol in the nervous system and the replace- hypodensity of cerebral hemispheric white matter.
ment of cholesterol by cholestanol must be important Increased density may be seen in the area of the den-
in the development of the nervous tissue damage. tate nucleus, corresponding to hemosiderin and calci-
There is evidence that there is a subtle defect in the um deposition in histopathology.
blood–brain barrier in CTX, probably caused by the The most important and earliest MRI abnormali-
accumulating toxic metabolites. The defect in the ties are noted in the cerebellum. On T2-weighted im-
blood–brain barrier may be important in the accre- ages the dentate nucleus and cerebellar hemispheric
tion of cholestanol and cholesterol within the CNS. In white matter have a high signal intensity (Figs. 31.1
CTX the fractional clearance of LDL is enhanced, and 31.2). The cerebellar foliae are prominent, indica-
most likely due to an increased activity of hepatic tive of atrophy (Fig. 31.2). In some patients, the den-
LDL receptors. LDL receptors within the blood–brain tate nucleus has a low signal intensity on T2-weighted
31.6 Magnetic Resonance Imaging 255

Fig. 31.1. The T2-weighted images in this 40-year-old male white matter. Courtesy of Dr. F. Barkhof, Department of Radiol-
CTX patient reveal signal abnormalities in the dentate nucleus ogy, VU University Medical Center, Amsterdam, and Dr. A. Ver-
and medial lemniscus in the midbrain. In addition, there are rips, Department of Pediatric Neurology, University Medical
diffuse, ill-defined slight signal abnormalities in the cerebral Center Nijmegen, The Netherlands

images (Fig. 31.2). In exceptional cases, the cerebellar ing the presence of xanthomas within the choroid
white matter lesions are surrounded by a rim of plexus (Fig. 31.4). T2-weighted images often reveal
markedly low signal intensity (Fig. 31.4). The low sig- bilateral high-signal-intensity lesions in the globus
nal on T2-weighted images reflects the presence of pallidus and the adjacent part of the internal capsule,
macroscopic xanthomas or calcium and hemosiderin in the area of the ansa lenticularis (Fig. 31.3). The
deposits. Symmetrical lesions are often present in the globus pallidus may have a low signal intensity, prob-
corticospinal tracts and medial lemniscus in the ably related to the high lipid content (Fig. 31.2).
brain stem (Figs. 31.1 and 31.2). In some patients, the Within the spinal cord, the lateral and dorsal
transverse pontine fibers contain signal abnormali- columns may have a high signal intensity on T2-
ties. Involvement of the inferior olives may occur. weighted images (Fig. 31.5). Lipid deposits in the
In the supratentorial region, ill-defined slight sig- Achilles tendon can be visualized (Fig. 31.6).
nal changes are seen in the periventricular region in The full-blown MRI pattern of CTX, consisting of
most patients, blending with the normal white matter the typical cerebellar abnormalities with high-signal-
without a sharp demarcation between the two intensity changes within the white matter and a low
(Figs. 31.1–31.3). They have a symmetrical distribu- signal intensity of the dentate nucleus on T2-weighted
tion. These abnormalities may be confluent or patchy. images, has a high diagnostic value. When only high-
U fibers and corpus callosum are spared. Slight en- signal-intensity lesions are present in the cerebellar
largement of the ventricles and subarachnoid spaces (and also cerebral) white matter, other diagnoses
may be seen. In a few patients, focal round or ovoid should be considered, including adrenomyeloneu-
masses with low signal intensity on T2-weighted im- ropathy and Refsum disease.
ages are noted within the ventricles, probably reflect-
256 Chapter 31 Cerebrotendinous Xanthomatosis

Fig. 31.2. The T2-weighted images in this 48-year-old female spheric white matter, and the hilus of the dentate nucleus dis-
CTX patient reveal diffuse, ill-defined slight signal abnormali- play an elevated signal intensity. The dentate nucleus stands
ties within the cerebral white matter, sparing the corpus callo- out as dark. Courtesy of Dr. F. Barkhof, Department of Radio-
sum, and some cerebral atrophy.The globus pallidus has a low logy, VU University Medical Center, Amsterdam, and Dr. A. Ver-
signal intensity.The pyramidal tracts in the brain stem, the me- rips, Department of Pediatric Neurology, University Medical
dial lemniscus at the level of the pons, the cerebellar hemi- Center Nijmegen, The Netherlands
31.6 Magnetic Resonance Imaging 257

Fig. 31.3. Male, 38 years of age, with CTX.The T2-weighted im- the hilus of the dentate nucleus and cerebellar hemispheric
ages show a diffuse slight signal abnormality within the cere- white matter have a high signal. Note the cerebellar atrophy.
bral white matter.There are signal abnormalities in the medial Courtesy of Dr. F. Barkhof, Department of Radiology,VU Univer-
globus pallidus and adjacent part of the posterior limb of the sity Medical Center, Amsterdam, and Dr. A.Verrips, Department
internal capsule, the pyramidal tracts, and medial lemniscus in of Pediatric Neurology, University Medical Center Nijmegen,
the midbrain. The dentate nucleus has a low signal, whereas The Netherlands

Fig. 31.4. Female, 43 years of age,


with CTX. On the T2-weighted image
on the left, the cerebellar white matter
has a high signal intensity surrounded
by a rim of very low signal intensity.
The moderately T2-weighted image on
the right shows the involvement of
the corticospinal tracts in the mid-
brain. Note the round masses with low
signal intensity in the choroid plexus
of the lateral ventricles. From Fiorelli
et al. (1990), with permission
258 Chapter 31 Cerebrotendinous Xanthomatosis

Fig. 31.5. Spinal images in a 41-year-


old woman with CTX.The sagittal
mildly T2-weighted images reveal mild
signal abnormalities in the spinal cord
over its entire length. Courtesy of
Dr. F. Barkhof, Department of Radio-
logy, VU University Medical Center,
Amsterdam, and Dr. A. Verrips, Depart-
ment of Pediatric Neurology, Uni-
versity Medical Center Nijmegen,
The Netherlands

Fig. 31.6. The T1-weighted images


show the typical swelling of the
Achilles tendon in two CTX patients
(arrows). Courtesy of Dr. F. Barkhof,
Department of Radiology, VU Uni-
versity Medical Center, Amsterdam,
and Dr. A. Verrips, Department of
Pediatric Neurology, University
Medical Center Nijmegen,
The Netherlands
Chapter 32

Cockayne Syndrome

32.1 Clinical Features usually becomes evident in the teenage years. Behav-
and Laboratory Investigations ioral problems are rare and the patients are usually
described as happy and social. In the course of the
Cockayne syndrome (CS) is a rare, inherited disorder years progressive neurological abnormalities become
characterized by cachectic dwarfism, cutaneous pho- apparent. Cerebellar signs are present with tremor,
tosensitivity, and progressive neurological dysfunc- lack of coordination, dysarthric speech, and gait atax-
tion. The mode of inheritance is autosomal recessive. ia. Pyramidal signs are usually present with hyperto-
Two types of the disease are distinguished: classical nia and hyperreflexia. The gait disorder in patients
CS or CS type I and severe CS or CS type II. who become ambulatory is striking and progressive,
In CS type I, the children seem normal at birth. due to a combination of spasticity, ataxia, and con-
Their weight, length, and head circumference are nor- tractures of the hips, knees and ankles. Sensorineural
mal. Growth failure generally begins within the first hearing loss occurs in over half the patients. My-
year of life and may be profound.Weight is affected to oclonus and involuntary choreiform and athetoid
a greater extent than length, leading to cachectic movements are rare. Overt signs of peripheral neu-
dwarfism. Subcutaneous fat is lost diffusely and al- ropathy are rare until late stages of the disease, lead-
most completely, giving the patients a starved appear- ing to a progressive, diffuse muscular atrophy, muscle
ance.A thin, prominent nose, narrow mouth and chin, weakness, and areflexia. Some patients have dimin-
sunken eyes, and loss of adipose tissue from the face ished lacrimation, decreased sweating, miotic pupils,
result in the characteristic facies of an old man or and cool and acrocyanotic limbs. This could be due to
woman. This appearance becomes more evident in autonomic dysfunction, but formal assessment of
older patients. However, percutaneous gastrostomy autonomic function has not been documented.
tube feeding allows better management of the feeding Seizures occur late and in a minority of the patients.
problems and the typical cachectic appearance may Pigmentary degeneration of the retina of the salt
be lacking or delayed. Within the first 2 years almost and pepper type is found in the majority of the pa-
all patients have become microcephalic. A thin and tients. The retina changes are progressive, and during
dry skin, fine hair, and large and sometimes mal- the first few years of life normal findings do not ex-
formed ears contribute to the characteristic facies of clude the diagnosis of CS. Other common ophthalmo-
CS patients. Dermal photosensitivity appears as sun logical abnormalities are cataract and optic atrophy.
sensitivity resulting in desquamation, scarring, and Miotic pupils, which are poorly responsive to mydri-
atrophy of exposed areas. Pigmentary abnormalities atics, corneal dystrophy, and decreased or absent
with hypopigmentation and hyperpigmentation oc- lacrimation may be found. Despite the extensive ocu-
cur occasionally. Anhidrosis may be present. Dental lar abnormalities some visual acuity usually remains,
problems are common with moderate to severe although blindness may occur.
caries. The posture of CS patients is typical. Even at an Renal problems develop in about 10% of the CS
early age there is abnormal flexion of hips and knees patients with decreased renal function, and very
while standing. In older patients the posture is rarely renal failure. Hypertension may occur.
stooped due to kyphosis combined with progressive Undescended or small testes are seen in about 30%
hip, knee, and ankle contractures. The trunk is small, of male CS patients. In female patients breasts are of-
the arms and legs are disproportionately long, and ten small and menstrual cycles irregular.
hands and feet are large. The neurological abnormalities in CS type I be-
The earliest commonly noticed neurological ab- come gradually more severe and mental failure is pro-
normality is delayed psychomotor development, gressive. Death by inanition and infection, most com-
which usually becomes apparent at the time when sit- monly respiratory infections, used to occur in the
ting, walking, and speech should develop. The delay in fourth decade of life. However, with improved nutri-
the acquisition of functions becomes progressively tion and medical care, life expectancy may be longer.
greater as the patient grows older.All patients with CS Patients with CS type II have an earlier onset of
type I are mentally retarded, but mental capacities symptoms and a more severe course than do CS type
vary from mildly to profoundly deficient. Among the I patients. They often have a low birth weight and
higher functioning patients, intellectual deterioration little postnatal increase in height, weight, and head
260 Chapter 32 Cockayne Syndrome

circumference, although some patients have a normal their upper and lower anterior margins, and osteo-
birth weight and normal weight gain during the first porosis.
few months of life. The infants used to develop an The clinical suspicion of CS can be confirmed by
emaciated appearance characteristic of CS with little UV irradiation of cultured skin fibroblasts of the pa-
subcutaneous fat, deep-set eyes, and a prominent, tients. Cell lines from CS patients are hypersensitive
beaked nose, but with tube feeding weight for height to killing by UV, and show failure of DNA and RNA
can be normal or even high, and the typical lack of synthesis to recover to normal rates after irradiation.
subcutaneous fat is not necessarily a feature of the CS cells differ in some of their cardinal laboratory
disease. In the neonatal period the children are hypo- characteristics from xeroderma pigmentosum cells.
tonic, but they develop spastic quadriplegia. Few or CS cells and most xeroderma pigmentosum cells have
no developmental milestones are reached; those that defective DNA repair of actively transcribed genes,
are reached are subsequently lost in the process of but only xeroderma pigmentosum cells have low DNA
neurological deterioration. Congenital cataract is of- repair in the genome overall, as demonstrated by de-
ten present. Other reported ocular abnormalities ficient unscheduled DNA synthesis after UV expo-
include hypoplastic optic nerves, microphthalmia, sure. Patients with a combination of CS and xeroder-
iris hypoplasia, microcornea, and reduced lacrimal ma pigmentosum also have decreased unscheduled
secretion. Kyphosis and progressive contractures of DNA synthesis after UV exposure. Prenatal diagnosis
hips and knees occur and may already be present at is possible using a test showing the defect in nu-
birth. Photosensitivity is present but may be missed cleotide excision repair or DNA analysis when the
because of lack of exposure to sunlight in small in- gene mutations responsible in a family have been
fants. Generally, dental, auditory, and cutaneous com- demonstrated.
plications are less commonly described in these pa-
tients, probably because of the severe neurological
problems and early age at death. Death usually occurs 32.2 Pathology
by the age of 6 or 7 years, but may also occur during
the first few months or years of life. The leptomeninges in classical CS appear thickened
The clinical pictures of CS type II, cerebro-oculo- and fibrotic. The brain is usually small and has an at-
facioskeletal syndrome (COFS syndrome) and rophied appearance. Cerebellar atrophy is the most
cataracts–microcephaly–failure to thrive–kyphoscol- severe. On sectioning, the lateral ventricles appear to
iosis syndrome (CAMFAK syndrome) overlap. Some be enlarged and the corpus callosum is thin. The
of the patients described with COFS or CAMFAK syn- white matter is decreased in volume and has a mot-
drome are in fact CS type II patients. tled appearance. Calcium depositions are present bi-
A few patients have the clinical features of classical laterally in the basal ganglia and dentate nuclei and
or severe CS plus the skin abnormalities of xeroder- may also be found in the white matter and cerebral
ma pigmentosum, including freckling and skin can- and cerebellar cortices. The dura may be focally calci-
cers. fied.
Laboratory investigations of blood and CSF reveal Microscopic examination of the leptomeninges re-
no consistent and no diagnostic abnormalities. An veals increased collagenous connective tissue without
elevated CSF protein content may be found. A few any inflammatory changes. The principal finding in
patients have biochemical evidence of decreased re- CS type I is lack of myelin. Numerous islands of pre-
nal function with decreased creatinine clearance. In a served myelin are present within the white matter in
few patients elevated serum cholesterol or lipoprotein a tigroid pattern. In the areas of myelin lack, axons are
levels have been noted. Nerve conduction velocities relatively preserved. There is no evident relationship
are markedly slowed, consistent with a demyelinating between blood vessels and myelin loss or presence.
polyneuropathy, in the majority of the patients. In All levels of the brain are involved, including cerebral
skeletal muscle, denervation changes can be found. hemispheric white matter, cerebellar white matter,
EEG is normal in some patients, but may show slow- and brain stem. The U fibers are not spared. In the
ing and sometimes epileptic discharges. Evoked po- spinal cord the descending tracts demonstrate a focal
tentials are often abnormally delayed. Radiological lack of myelin, whereas the ascending tracts have a
investigations of the skeleton in CS show micro- relatively higher myelin content. The myelin-deficient
cephaly, with a thick cranial vault and sometimes in- areas contain fewer oligodendrocytes than the areas
tracranial calcifications. General skeletal maturation with more myelin. Sudanophilic lipids are scanty, but
may be within normal limits or may be advanced or sometimes seen in the walls of vessels and in mi-
delayed. Less common findings include shortness and croglial cells in the brain parenchyma. No inflamma-
broadness of the metacarpals and phalanges, whose tory infiltration is found. Astrogliosis is seen in the
epiphyses sometimes have an ivory-dense appear- myelin-deficient areas. The cerebral cortex is general-
ance, and flattening of the vertebral bodies, lipping of ly intact, but sometimes there is a slight diffuse loss of
32.4 Pathogenetic Considerations 261

neurons. The cerebellar cortex is less well preserved 32.4 Pathogenetic Considerations
and the number of Purkinje cells and granule cells is
reduced. Calcium deposits are present perivascularly Three rare autosomal recessive disorders are associ-
and within the neuropil. The typical locations of cal- ated with a defect in nucleotide excision DNA repair:
cium deposits include the basal ganglia and dentate xeroderma pigmentosum, CS, and trichothiodystro-
nucleus, and may also involve cerebral and cerebellar phy with photosensitivity. Xeroderma pigmentosum
cortex and the white matter. The small deposits may is characterized by photosensitivity, abnormal pig-
coalesce into larger calculi. In some cases, nuclear ab- mentation, and a high incidence of sunlight-induced
normalities have been found in astrocytes and neu- skin cancers; signs of neurological degeneration are
rons. In these cases many of these cells have a single, rare. CS is characterized by photosensitivity, short
large, hyperchromatic, atypical nucleus. Some cells stature, and a severe neurological degenerative disor-
are multinucleated. In a few cases neurofibrillary tan- der; there is no increased risk of skin cancer. Tri-
gles have been found in the nucleus basalis, substan- chothiodystrophy, caused by a defect in nucleotide
tia nigra, locus caeruleus, and the cerebral cortex. excision repair, is characterized by brittle hair and
Their presence may be considered to be a premature nails, photosensitivity, ichthyosis, and neurological
senile change of the brain or a nonspecific finding oc- problems, but no skin cancer. Rare patients have com-
curring in chronic neurological conditions. bined symptoms of xeroderma pigmentosum and CS.
Sural nerve biopsies demonstrate chronic segmen- Complementation analysis by cell fusion has allowed
tal demyelination and remyelination with onion bulb a genetic classification of these disorders. Most CS pa-
formation. Onion bulbs are nonspecific signs of re- tients belong to one of two complementation groups,
peated demyelination and remyelination. They are CS-A and CS-B, which represent two different defec-
more pronounced in older than in younger patients. tive genes, the CSA gene located on chromosome 5
In some cases of CS, electron-dense, membrane- and the CSB gene on chromosome 10q11–21. The
bound, finely granular or lamellated inclusions have clinical phenotype of the patients (classical or severe
been reported in Schwann cells. The nature and role CS) does not correlate with these complementation
of these inclusions is unknown. groups. Patients with combined xeroderma pigmen-
Renal pathology may include thickening of the tosum and CS belong to three of the xeroderma pig-
glomerular basement membrane. mentosum complementation groups: XP-B, XP-D, and
In CS type II, neuropathological findings are simi- XP-G. The patients in complementation group XP-G
lar to those of CS type I. The leptomeninges are thick- all have severe neurodegeneration and die in early
ened. The brain is small and atrophic and the ventri- childhood, but patients belonging to complementa-
cles are enlarged. The white matter is reduced in vol- tion group XP-D may also present with severe neuro-
ume. The cerebellum is small. In the CNS severe but logical problems.
discontinuous myelin deficiency is present with a Despite the high accuracy of the DNA replication
tigroid pattern of islands with higher myelin content. process, some errors may arise spontaneously during
There is concomitant white matter gliosis. The cere- DNA replication through intrinsic instability of
brum, cerebellum, brain stem, and spinal cord are af- chemical bonds in DNA. Furthermore, chemical com-
fected. Calcium depositions, present perivascularly pounds can cause alterations in the DNA. The chemi-
and in the neuropil, may be found in basal ganglia, cal compounds that react with nucleic acids range
thalamus, dentate nucleus, and cerebral and cerebel- from alkylating agents, polycyclic hydrocarbons, and
lar cortex and meninges. The cerebral cortex is intact; aromatic amines to aflatoxin. DNA modifications can
in the cerebellar cortex loss of Purkinje cells and also arise endogenously through cellular metabolites,
granule cells is found. Bizarre astrocytes with large for instance through free radicals generated as by-
and multiple nuclei may occur. products of oxidative metabolism. The most common
physical agents causing DNA damage are UV and ion-
izing radiation. Unless repaired, DNA damage leads to
32.3 Chemical Pathology defects in subsequent replication cycles and to abnor-
malities in transcription and translation of the infor-
In CS the white matter shows a marked loss of myelin mation coded in DNA. The result may be cell death or
lipids and some increase in cholesterol esters. Besides genomic instability (mutagenesis).
these usual chemical changes related to demyelina- There are different basic mechanisms by which le-
tion, no specific abnormalities are found. sions are eliminated from DNA: direct reversal, base
excision repair, nucleotide excision repair, and DNA
double-strand repair. Nucleotide excision repair is a
complex process that detects, removes, and repairs
many types of DNA lesions, in particular bulky le-
sions. The process involves at least 30 proteins, which
262 Chapter 32 Cockayne Syndrome

act in a stepwise fashion to recognize the damage, fol- With respect to the correlation between basic de-
lowed by enzymatic incisions in the damaged strand fect and the clinical features, it is interesting to con-
on both sides of the lesion, removal of the damaged sider the differences between CS patients and patients
single-stranded segment, repair synthesis to fill in the with xeroderma pigmentosum. The former lack the
resultant gapped DNA duplex, and ligation of the re- increased risk for skin cancer despite the presence of
pair patch to the existing DNA strand. Nucleotide ex- photosensitivity and the latter usually lack signs of
cision repair has two subpathways: transcription- neurodegeneration. Xeroderma pigmentosum pa-
coupled repair and global genome repair. The tran- tients are most often deficient in both transcription-
scription repair pathway repairs lesions in the strand coupled repair and global genome repair, although
of active, RNA-polymerase-II-transcribed genes. The sometimes in global genome repair only. The photo-
global genome repair pathway removes lesions from sensitivity of CS patients is due to the failure in rapid
genes that are transcriptionally inactive. Transcrip- repair of crucial regions of DNA, which leads to hy-
tion-coupled repair is about three times faster than persensitivity of the cells to the lethal effects of UV
global genome repair. light. But considering the absence of neurodegenera-
In CS, the underlying defect affects not only the tion in most patients with xeroderma pigmentosum,
nucleotide excision repair, but also the basal tran- the defect in excision repair cannot explain the neu-
scription process and the sensitivity to apoptosis. rological problems of CS patients in a straightforward
Cultured fibroblasts from CS patients are more sensi- fashion. It is generally accepted that a defect in basal
tive than normal to killing by UV irradiation and ex- transcription plays a role in the development of the
hibit a marked, prolonged inhibition of synthesis of non-xeroderma-pigmentosum problems. Cells with a
DNA and RNA after such irradiation. Damage pro- high metabolic rate, such as cells of the nervous sys-
duced by UV light in DNA in normal cells depresses tem, which generate high levels of reactive oxygen
rates of both DNA and RNA synthesis, but these rates species, may be particularly vulnerable to the effects
soon become normal. In CS cells this rapid recovery of a combination of endogenous (oxidative) DNA
does not occur. The rapid recovery of DNA and RNA damage and transcription deficiency, induced by
synthesis in normal cells can be attributed to prefer- both DNA damage and dysfunction of CS genes. Fur-
ential rapid repair of DNA damage in regions that are thermore, an arrest of transcription provides a strong
actively transcribed, in contrast to much slower re- signal for the apoptosis pathway. CS could be a dis-
pair in the bulk of DNA. In CS cells, this preferential ease characterized by excessive cell death by apopto-
repair of transcriptionally active DNA does not take sis. The apoptosis model could also explain the prob-
place, damage in these regions being repaired at the lem of stunted growth and provide the answer to the
same (slow) rate as in the bulk of DNA. So, CS cells are question why CS patients are not prone to skin cancer,
defective in the transcription repair pathway, but pro- even in the light of sever sunlight sensitivity: the
ficient in the global genome repair of UV-induced damaged cells become apoptotic.
DNA damage. CS cells also show evidence of a defect Xeroderma pigmentosum/CS patient belong to
in basal transcription, not related to the defect in the three complementation groups: XP-B, XP-D, and
transcription-coupled repair pathway. The initial in- XP-G. XPB and XPD are DNA helicases and subunits
ability to resume transcription may be attributed to a of the DNA repair/basal transcription factor TFIIH.
failure to remove DNA lesions from the transcribed They are required for transcription initiation by RNA
strand of active genes, but defects in CSB result in fail- polymerase II under basal conditions and are in-
ure of RNA polymerase II elongation complexes to re- volved in the early steps of the nucleotide excision re-
sume transcription even after DNA damage is re- pair pathway. XPG is involved in promoting efficient
paired. An arrest of transcription provides a strong RNA polymerase II transcription, and a defect in XPG
signal for apoptosis. In addition, there is evidence causes a deficiency in transcription.As in CS, the pho-
that a defect in the ATPase domain of the CSB protein tosensitivity is attributed to the defect in nucleotide
enhances the cellular liability to apoptosis. excision repair, whereas the non-xeroderma-pigmen-
The roles of the products of the genes CSA and CSB tosum manifestations are attributed mainly to crip-
have been only partially elucidated. CSA is thought to pled basal transcription.
associate with components of the DNA repair/basal
transcription factor TFIIH and CSB. It could have a
regulatory role in transcription-coupled repair. CSB 32.5 Therapy
is a DNA-dependent ATPase and stimulates the rate of
elongation by RNA polymerase II. In addition, it has To date no effective mode of therapy has been found
been shown that integrity of the ATPase domain of in CS and the management of the disease is pure-
CSB is critical for prevention of DNA damage induced ly symptomatic. Patients should be monitored for
apoptosis. treatable complications, such as hypertension, hear-
32.6 Magnetic Resonance Imaging 263

ing loss, and dental caries. Physical therapy can be 32.6 Magnetic Resonance Imaging
helpful to avoid contractures. Emollients for dry skin,
avoidance of excessive sun exposure, and use of sun- CT scan may demonstrate bilateral calcifications of
screens are helpful in diminishing skin problems. the basal ganglia, dentate nucleus and areas of the
cerebral and cerebellar white matter and cortex
(Fig. 32.3), although in some patients the calcium de-
posits are very subtle (Figs. 32.1 and 32.2). Cerebral,

Fig. 32.1. Term born infant, now 5 weeks of age, with a clinical of subtle mineralization of the basal ganglia and thalamus.The
phenotype of CS type II and belonging to complementation axial T2-weighted images show that the brain has a somewhat
group XP-G. Note the severe hypoplasia of the cerebellum and immature gyral pattern for the age of the infant
brain stem on the sagittal images.The CT scan shows evidence
264 Chapter 32 Cockayne Syndrome

Fig. 32.2. An 18-month-old infant with a clinical phenotype sagittal images and the subtle mineralization of the basal gan-
of CS type II, belonging to complementation group CS-B. Note glia and thalamus on the CT scan. The axial T2-weighted im-
again the hypoplasia of the cerebellum and brain stem on the ages demonstrate seriously deficient myelination

cerebellar, and brain stem atrophy with prominence ing the presence of calcium depositions, but may be
of sulci and ventricular enlargement is usually seen. more accurate in delineating the locations. The
MRI confirms the presence of variable hypoplasia globus pallidus is most often involved, followed in
and atrophy, most pronounced in brain stem and frequency by the dentate nucleus. In addition, calci-
cerebellum (Figs. 32.1–32.5). There is a loss of white um depositions may be seen in the putamen and cau-
matter volume and ventricles are mildly enlarged in date nucleus, and as irregular, not necessarily sym-
most patients. MRI is less sensitive than CT in show- metrical areas in the cortex and white matter of cere-
32.6 Magnetic Resonance Imaging 265

Fig. 32.3. A 6-year-old girl with CS type I.CT scans through the The myelin deficiency is confirmed by the T1-weighted images
posterior fossa and the lateral ventricles show calcifications in (middle image, third row). The white matter has an irregular,
the dentate nucleus and the basal ganglia. The T2-weighted tigroid appearance. Courtesy of Dr. I.N. Snoeck, Juliana Chil-
MR images reveal white matter hypomyelination and atrophy. dren’s Hospital, The Hague, The Netherlands

brum and cerebellum. In addition, there are symmet- tern of myelin presence (Figs. 32.3 and 32.4). In many
rical white matter abnormalities. On T2-weighted im- of the patients the moderately high signal intensity is
ages the signal intensity of the cerebral white matter seen throughout the hemispheric white matter, in-
is abnormally high, but usually not so high as in com- cluding periventricular white matter and U fibers,
pletely unmyelinated white matter in neonates. The sometimes also the internal capsule. The corpus cal-
white matter often seems to have a finely irregular, losum has a better state of myelination. In other pa-
granular aspect, probably reflecting the tigroid pat- tients the white matter is better myelinated in the sub-
266 Chapter 32 Cockayne Syndrome

Fig. 32.4. A 7-year-old boy with CS type I. The sagittal images pect, probably reflecting the tigroid pattern of myelin pres-
reveal serious cerebellar atrophy and some atrophy of the ence. The T1-weighted gradient echo images (third row) reveal
brain stem. The cerebral white matter has a high signal on the calcium in the putamen
axial T2-weighted images with a finely irregular, granular as-

cortical areas and the white matter hyperintensity is An MRI pattern combining hypomyelination and
most marked in the periventricular area. Apparently calcium deposition is highly suggestive of CS. The im-
delayed and disturbed myelination as well as de- ages are similar to those seen in Aicardi–Goutières
myelination play a role in CS. Images in type I and syndrome, but the calcium depositions tend to be dif-
type II CS are essentially the same, but in type II CS ferent. In Aicardi–Goutières syndrome the calcium
the myelin deficiency is more profound and the cere- depositions are typically punctate, with many small
bellum is as a rule very hypoplastic. round deposits which may coalesce to larger irregular
32.6 Magnetic Resonance Imaging 267

Fig. 32.5. This 3-year-old girl with CS type I has a relatively at most subtle mineralization of the basal ganglia.The cerebral
mild phenotype. The cerebellum is both hypoplastic and white matter contains more myelin than in the other patients
atrophic. The brain stem is relatively thin. The CT scan reveals

areas. In CS the calcium depositions tend to have a


more homogeneous aspect. Apart from the calcifica-
tions, the images showing severe hypomyelination
resemble those of Pelizaeus–Merzbacher disease and
Salla disease.
Chapter 33

Trichothiodystrophy with Photosensitivity

33.1 Clinical Features pogonadism may be found. A few TTD patients have
and Laboratory Investigations been reported who lose their hair and have more se-
rious skin problems during episodes of fever. Within
Trichothiodystrophy with photosensitivity (TTD) is a a period of a few months, the scalp hair returns. Some
rare, inherited disorder with an autosomal recessive patients have increased susceptibility to infections
mode of inheritance. Multiple names and acronyms due to chronic neutropenia, lymphopenia, or im-
have been used for the same condition, including munoglobulin deficiency. Life expectancy varies. Se-
Tay syndrome, Pollitt syndrome, ONMR (onychotri- verely affected patients may die as early as at the age
chodysplasia, neutropenia, mental retardation), BIDS of 3 years, whereas other patients reach their thirties.
(brittle hair, impaired intelligence, decreased fertility, Polarization microscopy of the hair in TTD shows
short stature), IBIDS (ichthyosis, brittle hair, impaired a typical appearance of alternating dark and light
intelligence, decreased fertility, short stature), SIBIDS bands, giving the hair a tiger-tail pattern. Scanning
(osteosclerosis, ichthyosis, brittle hair, impaired intel- electron microscopy usually reveals other abnormali-
ligence, decreased fertility, short stature), and PIBIDS ties, including absent or damaged cuticle scales, irreg-
(photosensitivity, ichthyosis, brittle hair, impaired in- ular hair surface, trichoschisis, and trichorrhexis no-
telligence, decreased fertility, short stature). Presence dosa (torsions of the flattened hair shaft). The hair
of a defect in nucleotide repair has not been verified and often the nails are characterized by a reduction in
in all patients reported under these names. A few the cystine/cysteine and sulfur content, due to a de-
patients with features of both TTD and xeroderma creased amount of sulfur-rich matrix proteins. These
pigmentosum have been reported. hair findings are not diagnostic of TTD as similar ab-
One of the most striking clinical findings in TTD is normalities may be found in other disorders, such as
the abnormal hair. The scalp hair is sparse, short, thin, untreated argininosuccinic aciduria and acroder-
brittle, and dry. The other clinical features of patients matitis enteropathica.
with TTD are highly variable in expression and sever- The diagnosis can be confirmed by UV irradiation
ity. This partly explains the confusing nomenclature of cultured skin fibroblasts of the patients. Cell lines
in the literature for what is probably the same disease. from TTD patients are hypersensitive to killing by
In many cases the brittle hair is associated with neu- UV, and show failure of DNA and RNA synthesis to re-
roectodermal abnormalities. Cutaneous signs include cover to normal rates after irradiation. TTD cells have
photosensitivity, ichthyosis, keratosis, and erythema. defective DNA repair of actively transcribed genes
There is no abnormal pigmentation and no increased (transcription-coupled repair) as well as low DNA
risk of skin cancer. Some children present at birth as repair in the genome overall (global genome repair),
collodion babies. Nails are dystrophic. Dental caries is the latter demonstrated by deficient unscheduled
common. Patients have short stature and may have DNA synthesis after UV exposure. Prenatal diagnosis
microcephaly. Neurological and developmental im- is possible using a test showing the defect in nu-
pairments of patients with TTD are reminiscent of cleotide excision repair or DNA analysis when the
those found in Cockayne syndrome. The neurological responsible gene mutations in a family have been
signs may include spasticity with hyperreflexia, cere- demonstrated.
bellar ataxia, dysarthria, and neurosensory hearing
impairment. The deep tendon reflexes may also be
absent due to a concomitant polyneuropathy. Eye ab- 33.2 Pathology
normalities may include nystagmus, retinal dystro-
phy, pale optic disc, cataract, and strabismus. Intelli- At autopsy (unpublished data), micrencephaly is
gence is impaired, but to a variable degree. Patients found. The most striking finding is a lack of myelin
have a peculiar face with a beaked nose, receding throughout the cerebrum and cerebellum, with a
chin, and protruding ears. They show genital abnor- relatively higher myelin content in the brain stem.
malities including cryptorchism, hypospadias, and Loss of Purkinje cells is seen in the cerebellar cortex.
hypoplasia of the genitalia. Delayed or absent puber- Calcium deposits are less striking than in Cockayne
ty and laboratory evidence of hypergonadotropic hy- syndrome, but may be present in the basal ganglia.
33.3 Pathogenetic Considerations 269

33.3 Pathogenetic Considerations global genome repair. Damage produced by UV light


in DNA in normal cells depresses rates of both DNA
Three rare autosomal recessive disorders are associ- and RNA synthesis, but these rates soon become nor-
ated with a defect in nucleotide excision DNA repair: mal. The rapid recovery of DNA and RNA synthesis in
xeroderma pigmentosum, Cockayne syndrome, and normal cells can be attributed to preferential rapid
TTD. Xeroderma pigmentosum is characterized by repair of DNA damage in regions that are actively
photosensitivity, abnormal pigmentation, and a high transcribed, in contrast to much slower repair in the
incidence of sunlight-induced skin cancers, but rarely bulk of DNA.
by signs of neurological degeneration. Cockayne syn- The genes mutated in TTD are all related to the
drome is characterized by photosensitivity and a DNA repair/basal transcription factor II H (TFIIH).
severe neurological degenerative disorder, but no XPB and XPD are DNA helicases and subunits of
increased risk of skin cancer. TTD is characterized by TFIIH; TTDA is involved in the stabilization of TFIIH.
brittle hair and nails, photosensitivity, and neurolog- The TFIIH complex regulates initiation of transcrip-
ical problems, but no skin cancer. Complementation tion by RNA polymerase II under basal conditions,
analysis by cell fusion has allowed a genetic classifica- and in the case of DNA damage it initiates nucleotide
tion of these disorders. TTD patients belong to three excision repair. TTD cells have a defect in both com-
complementation groups, XP-B, XP-D, and TTD-A. ponents of the nucleotide excision repair, the prefer-
The XPB gene is located on chromosome 2q21and the ential repair of transcriptionally active DNA and in
XPD gene on chromosome 19q13.2. The TTDA gene the global genome repair of UV-induced DNA dam-
has not yet been characterized. The clinical pheno- age. In addition, like Cockayne syndrome, TTD cells
type of the TTD patients does not correlate with these have a defect in basal transcription. The photosensi-
complementation groups. tivity is attributed to the defect in nucleotide excision
Despite the high accuracy of the DNA replication repair, whereas the other manifestations are attrib-
process, some errors may arise spontaneously during uted mainly to impairment of basal transcription.
DNA replication through intrinsic instability of Intriguingly, in view of the marked differences in
chemical bonds in DNA. Chemical compounds can clinical phenotypes, defects in two of the genes relat-
also cause alterations in the DNA. DNA modifications ed to TTD (XPB and XPD) can also cause xeroderma
can also arise endogenously through cellular metabo- pigmentosum and the combined symptoms of xero-
lites, for instance through free radicals generated as derma pigmentosum and Cockayne syndrome. It has
by-product of oxidative metabolism. The most com- been suggested that clinical features of xeroderma
mon physical agents causing DNA damage are UV pigmentosum result from mutations that affect only
and ionizing radiation. If not repaired, DNA damage the nucleotide excision repair function of TFIIH,
leads to defects in subsequent replication cycles and while features typical of TTD and Cockayne syn-
to abnormalities in transcription and translation of drome are due to impairment of its transcription role.
the information coded in DNA. The result may be cell This notion has been supported by the spectrum of
death or genomic instability (mutagenesis). mutations observed in the different patient groups,
There are different basic mechanisms by which indicating that the site of mutation determines the
lesions are eliminated from DNA: direct reversal, base clinical phenotype. It has been demonstrated that
excision repair, nucleotide excision repair, and DNA alterations in XPB, XPD, and TTDA associated with
double-strand repair. Nucleotide excision repair is a TTD specifically reduce the cellular content of TFIIH.
complex process that detects, removes, and repairs However, the degree of reduction in the level of
many types of DNA lesions, in particular bulky le- TFIIH does not correlate with the severity of the TTD
sions. The process involves at least 30 proteins, which phenotype, suggesting that the severity of the clinical
act in a stepwise fashion to recognize the damage, fol- symptoms in TTD cannot be related solely to the
lowed by enzymatic incisions in the damaged strand effects of mutations on the stability of TFIIH. Proba-
on both sides of the lesion, removal of the damaged bly, the transcriptional activity of the residual TFIIH
single-stranded segment, repair synthesis to fill in the complexes also plays a role.
resultant gapped DNA duplex, and ligation of the In all TTD patients known so far who show loss of
repair patch to the existing DNA strand. Nucleotide hair during fever, the same missense mutation in
excision repair has two sub-pathways: transcription- the XPD gene has been demonstrated, either in the
coupled repair and global genome repair. The tran- homozygous or the compound-heterozygous state.
scription repair pathway repairs lesions in the strand There is evidence that the XPD protein with this
of active, RNA-polymerase-II-transcribed genes. The amino acid substitution is thermolabile, resulting in a
global genome repair pathway removes lesions from further reduction of TFIIH-dependent basal tran-
genes that are transcriptionally inactive. Transcrip- scription during fever.
tion-coupled repair is about three times faster than
270 Chapter 33 Trichothiodystrophy with Photosensitivity

33.4 Therapy The most striking finding on MRI is diffuse hy-


pomyelination involving the cerebral hemispheres
To date no effective mode of therapy has been found and cerebellum (Fig. 33.1). Even the brain stem may
in TTD and the management of the disease is purely be insufficiently myelinated. In addition, there may be
symptomatic. Patients should be monitored for treat- some variable atrophy due to loss of white matter vol-
able complications. Physical therapy can be helpful to ume. However, atrophy is not present in all patients.
avoid contractures. Emollients for dry skin, avoidance A neuroimaging pattern of hypomyelination and
of excessive sun exposure, and use of sun-screens are calcium depositions is highly suggestive of a DNA re-
helpful in diminishing skin problems. pair disorder, either TTD or Cockayne syndrome. The
pattern is similar to that seen in Aicardi–Goutières
syndrome, but the calcium depositions tend to be dif-
33.5 Magnetic Resonance Imaging ferent. In Aicardi–Goutières syndrome the calcium
depositions are typically punctate, with many small
CT scan may demonstrate bilateral calcifications of round deposits which may coalesce to larger irregular
the basal ganglia, but in most TTD cases no calcium areas. In DNA repair disorders, the calcium deposi-
deposits are seen. Some cerebral atrophy may be seen tions tend to have a more homogeneous aspect. In
with mildly enlarged lateral ventricles and subarach- TTD patients without evidence of calcium deposits
noid spaces. on neuroimaging, the pattern resembles that of
Pelizaeus–Merzbacher disease and Salla disease.
33.5 Magnetic Resonance Imaging 271

Fig. 33.1. The T2-weighted images in a 3-year-old boy with The coronal T1-weighted images show that there must be
TTD show insufficient myelination of the cerebral white mat- some myelin, enough to give the white matter a moderately
ter, which has a high signal. Even the corpus callosum and in- high signal.From Østergaard and Christensen (1996), with per-
ternal capsule are hypomyelinated. The cerebellar white mat- mission
ter seems to contain more myelin, but still less than normal.
Chapter 34

Pelizaeus–Merzbacher Disease
and X-linked Spastic Paraplegia Type 2

34.1 Clinical Features hypotonia, feeding problems, absent primitive reflex-


and Laboratory Investigations es, and sometimes stridor. Abnormal, nystagmoid eye
movements and extrapyramidal hyperkinesia occur
Pelizaeus–Merzbacher disease (PMD) is a rare neuro- early, followed by the development of epilepsy, spas-
logical disorder affecting the myelination of the CNS. ticity, cerebellar ataxia, and optic atrophy. Titubation
The disease has an X-linked recessive mode of inher- is often present. From birth onwards there is a com-
itance and is usually subdivided into three types: the plete failure of psychomotor development or an early
classical type (type I), the connatal type (type II), and loss of attained milestones. Microcephaly and growth
the transitional type (type III). retardation develop in the subsequent years. Progres-
The classical type has its onset in the first year of sion is rapid, with death occurring in the first decade,
life. Those affected are almost exclusively boys. The usually in early childhood.
disease manifests initially by irregular nystagmoid The transitional form between the classical and
eye movements referred to as “dancing,” “trembling,” connatal types has its onset in the neonatal or early
or “roving.” In some children stridor occurs, caused infantile period, but its course is less rapid. The dis-
by either laryngeal abductor paralysis or laryngoma- tinction between the classical, transitional, and con-
lacia. There is variable but always marked to severe natal forms of PMD is ill-defined. The age of onset
developmental delay with only very slow develop- does not appear to be the most valuable discriminat-
mental progress. Growth is retarded and the head size ing factor between the types of PMD. The rate of pro-
is small, in the low normal or microcephalic range. gression is the most useful and earliest reliable means
Seizures occur early in the course of the disease. of differentiating the different types.
Characteristically there is a tremor or bobbing, The nosology of PMD is a matter of debate. Some
nodding, or shaking movement of the head. Signs of include all patients with evidence of severe hy-
tetraspasticity, cerebellar ataxia, and extrapyramidal pomyelination on MRI and a Pelizaeus–Merzbacher-
movement disturbances with hyperkinesia, dystonia, like clinical picture under the heading of PMD, even
and choreoathetosis become manifest as the patient in sporadic cases, in female patients, or in the case of
becomes older. Most patients never walk. The sensory autosomal recessive inheritance. In particular with
system is usually well preserved. Optic atrophy with connatal onset, female patients can be found.An adult
visual failure is common. The nystagmus disappears variant of PMD has been suggested: the Löwen-
in the course of a few years. Social interactions are berg–Hill type. We prefer to include under the head-
often relatively well preserved and the intellectual ca- ing of PMD only patients in whom a proteolipid pro-
pacities are higher than the motor capacities. Skeletal tein (PLP) gene mutation has been found or in whom
abnormalities, such as osteoporosis and kyphoscolio- the family history indicates an X-linked recessive in-
sis, result from the chronic motor disease and occur heritance. Another basic defect should be sought for
after the disease has been manifest for many years. the remaining patients.
The course of the disease is chronic. Patients may im- Mothers of boys suffering from classical PMD
prove in performance up to the age of 10–12 years. sometimes develop neurological problems including
From that age onwards, a very slow progression of the spasticity, bladder dysfunction, personality changes,
neurological signs and a decline of mental level is dementia, and polyneuropathy. Incidentally female
usually noted. Death occurs in most patients in early patients in sibships with classical PMD show identical
or mid adulthood and is usually due to intercurrent clinical and postmortem neuropathological findings.
illnesses. X-linked spastic paraplegia type 2 (SPG2) and
Some patients have a milder variant. They may PMD are allelic conditions. The pure form of SPG2
achieve aided or unaided walking and demonstrate leads to spasticity only. The legs are more severely in-
slow neurological deterioration. They may have signs volved than the arms. An intention tremor may be
of polyneuropathy. present. There are no other neurological deficits and
The connatal type, also called Seitelberger type, is intelligence is normal. The disease is slowly progres-
a more rare and severe form of PMD. The disease is sive and the life span is nearly normal. The age of on-
already manifest in the neonatal or early infantile pe- set varies from a few years of life to the teenage peri-
riod. The neonatal period may be characterized by od. The complicated form of SPG2 also leads to slow-
34.2 Pathology 273

ly progressive spasticity, but there are additional relatively well preserved. Some axonal loss may be
signs in common with PMD, which may include nys- seen in completely demyelinated areas. The severity
tagmus, optic atrophy, cerebellar ataxia, sensory dis- of the concomitant fibrillary gliosis varies from slight
turbances, and dysarthria. Walking may be delayed to dense. The gray matter is also affected; the intra-
but all patients achieve unaided walking. Mild mental cortical myelin is completely or almost completely
retardation is present in some of the patients. Analy- absent, but the normal cytoarchitecture is preserved.
sis of SPG2 and PMD families shows a great overlap of The cerebellar cortex shows loss of Purkinje cells and
clinical findings, both within and between families. granular cells. Myelin is also deficient in the optic
Therefore, mutations in the PLP gene lead to a spec- nerves and chiasm.
trum of disorders ranging from connatal PMD The abnormalities in classical PMD are less pro-
through the various manifestations of the classical nounced and myelin deficiency is less severe. There is
PMD disease to pure SPG2. a patchy absence of myelin with preservation of
Routine and metabolic laboratory investigations numerous myelin islets giving the white matter a
are of no help in establishing the diagnosis. Nerve so-called tigroid pattern. Most of the myelin islets
conduction velocities and electromyography are usu- surround small blood vessels. The myelin sheaths in
ally normal, but in some patients mild to moderate these islets are thin and composed of only a few
slowing of nerve conduction velocities and neuro- myelin lamellae. Microscopically some remaining
genic changes in electromyography are found. Evoked myelin sheaths are also seen in the areas, which are
potential studies are of some help in PMD. In BAEP otherwise devoid of myelin.At most small amounts of
studies usually only wave I or waves I and II are pre- sudanophilic lipid products are found. Oligodendro-
sent and later components are absent, indicative of cytes are numerically reduced, especially in the areas
abnormalities at brain stem level. ERG is normal.VEP lacking myelin.All parts of the CNS are affected in the
and SSEP are absent or abnormal with increased la- same way, but the spinal cord, brain stem, cerebellum,
tency, abnormal shape, and decreased amplitude. diencephalic structures, and subcortical white matter
Definite diagnosis in PMD and SPG2 is DNA-based. show a relatively good state of myelin preservation. In
Prenatal diagnosis and carrier detection are possible all areas of the CNS axons are relatively well pre-
using DNA techniques. served. Where myelin is absent, the remaining white
matter is mainly composed of naked axons. Fibrillary
gliosis varies from slight to intense. The astrocytes
34.2 Pathology are sometimes hypertrophied. The gray matter is also
involved in the process; myelin sheaths are reduced in
In PMD, the brain is too light for the patient’s age and number or are absent, but the cortical cytoarchitec-
shows signs of diffuse atrophy, involving cerebral ture as well as the individual nerve cells are normal,
hemispheres and in particular brain stem and cere- although a certain loss of and damage to nerve cells
bellum. On sectioning the white matter appears re- may be seen. In the cerebellar cortex, loss of Purkinje
duced in volume to a variable extent and the corpus cells and granule cells may be evident.
callosum is markedly reduced in width. Microscopic Whereas most PMD patients do not show signs of
examination shows lack of myelin in all parts of the primary axonal degeneration, evidence of length-de-
CNS. The pathological picture is basically the same in pendent axonal degeneration is seen in patients with
all PMD patients irrespective of subtype, but with a PLP gene deletion or null mutation.
variable severity. Myelin is present in normal amounts for age in the
In connatal PMD, the pathological picture varies PNS, including spinal roots and cranial nerves with
from a marked lack of myelin to a complete absence the exception of the optic nerve in most PMD
of myelin in all parts of the brain and spinal cord. The patients. However, in some patients the peripheral
myelin present is usually found in the spinal cord and nerves are affected and myelin loss is found.
the deeper parts of the brain: the diencephalon The transitional type shows abnormalities inter-
(globus pallidus, posterior limb of the internal cap- mediate in severity between the connatal and the
sule, thalamus), the brain stem (tegmentum of pons classical types.
and mesencephalon, mesencephalic pyramidal tracts), Electron microscopy demonstrates that the few
and the central part of the cerebellum. The myelin is oligodendrocytes present contain an excess of cyto-
usually present in perivascular islets. Also, residual plasmic dense bodies and a poorly developed endo-
myelin islets are sometimes present in the subcortical plasmic reticulum. There is condensation of nuclear
white matter, especially in the pre- and postcentral chromatin, strongly suggestive of apoptosis, as has
gyri. There are no signs of active demyelination. been found in animal models.
There are no or little sudanophilic breakdown prod- Information on the neuropathology of SPG2 is lim-
ucts in the white matter. Oligodendrocytes are re- ited. A severe myelin deficiency has been reported in
duced in number or completely absent. The axons are the spinal cord, contrasting with a mild lack of myelin
274 Chapter 34 Pelizaeus–Merzbacher Disease and X-linked Spastic Paraplegia Type 2

in the cerebral hemispheric white matter with sparing very slow breakdown of myelin. Oligodendrocytes are
of the U fibers. found to be decreased in number, show morphologi-
cal abnormalities, and appear inactive. The chemical
findings of no or at most a low level of cholesterol
34.3 Chemical Pathology esters in the white matter is not in agreement with
active demyelination. The cholesterol:phospholipid:
Chemical analysis of the remaining myelin in PMD cerebroside ratio in PMD corresponds with the ratio
reveals that its lipid content is greatly reduced and its in the brain prior to myelination. The low ratio of
protein content, conversely, relatively increased. Cere- ethanolamine phosphoglycerides to choline phos-
brosides are reduced in quantity and sulfatides are phoglycerides in PMD is an indication of a poor state
greatly reduced in quantity. The remaining glyco- of maturation of the brain, as the ratio increases with
lipids contain an abnormally high proportion of maturation. The high glucose content of glycolipids is
glucose with a proportional reduction in galactose. also an indication of the immature state of the brain;
The myelin ganglioside content and cholesterol con- as maturation proceeds, glucolipids are replaced by
tent are near normal. Phospholipids are increased, galactolipids. The topography of the myelin present is
particularly sphingomyelin, choline phosphoglyc- in conformity with an arrest of myelination, which
erides, and inositol phosphoglycerides, whereas apparently occurs before birth (connatal form) or
ethanolamine phosphoglycerides and plasmalogens within the first year of life (classical and transitional
are reduced. Protein analysis shows that proteolipid form). So, the severity of the clinical phenotype seems
protein is absent, myelin basic protein is decreased, to be related to the degree of myelin deficiency.
whereas so-called Wolfgram proteins are increased. PMD caused by a defect in the gene coding for pro-
The chemical composition of whole white matter teolipid protein (PLP). The PLP gene is localized on
depends on the amount of myelin that is present. The the long arm of the X chromosome (Xq21.33–Xq22).
water content is abnormally high. Those lipids that It codes for PLP as its major gene product and addi-
are generally recognized as myelin lipids are either tionally for DM 20 due to alternate splicing of mRNA.
absent or markedly reduced. No or very little sulfatide DM 20 is identical to PLP but 35 amino acids shorter.
is present. A marked reduction is seen in cerebroside, The oligodendrocyte is the predominant cell in
cholesterol, and phospholipids. The cholesterol:phos- which the PLP gene is expressed in the CNS. PLP and
pholipid:cerebroside ratio is similar to the ratio in DM 20 are produced in the endoplasmic reticulum
brain prior to myelination. Gangliosides are found in and routed through the Golgi to the plasma mem-
normal concentrations. No or only a very small brane. PLP is a major myelin membrane protein.
amount of cholesterol esters are present. The protein Myelin basic protein and PLP normally constitute
composition of white matter is also altered with ab- more than 80% of the total CNS myelin proteins.
sence of proteolipid protein and reduction of other Myelin basic protein accounts for 30–40% of the total
myelin proteins. myelin protein, PLP for 40–50%. PLP and its less
In contrast to white matter, the chemical composi- abundant isoform DM 20 are strongly hydrophobic
tion of gray matter is much closer to normal. The con- transmembrane proteins. They are myelin-specific
centration of sulfatides and cerebrosides is decreased, and almost entirely confined to the CNS. They are,
but the concentration of phospholipids, cholesterol, however, minor constituents of the PNS and compose
and gangliosides is normal or close to normal. less than 1% of the mass of PNS myelin proteins. The
compact lamellar structure of myelin is organized
and stabilized by the two main myelin proteins,
34.4 Pathogenetic Considerations myelin basic protein as a peripheral membrane pro-
tein, and PLP as a strongly hydrophobic integral
For many decades the pathogenesis of the lack of membrane protein. Myelin basic protein contributes
myelin in the CNS in PMD has been a matter of to the compaction of the major dense lines and PLP to
debate. The original contention of Merzbacher was the tight apposition of the intraperiod lines in the
that the lack of myelin sheaths was due to faulty or myelin sheath. DM 20 is the predominant isoform in
absent myelination. Subsequently, many authors have oligodendrocyte progenitors and is expressed before
classified PMD among the leukodystrophies and PLP in the developing brain. DM 20 has biological
described the histopathological findings as tigroid functions in the maturing CNS that are distinct from
demyelination. However, several histological and the role of PLP.
chemical findings are not consistent with demyelina- Although PLP and DM 20 have been studied exten-
tion, but are consistent with a defect in myelin depo- sively, their biological functions are still not known in
sition. Histological examination fails to reveal signs detail. Mice in which PLP gene expression has been
of active demyelination. The small amounts of myelin inactivated demonstrate only subtle defects in the
degradation products are in conformity with at best a ultrastructure of CNS myelin, demonstrating that
34.4 Pathogenetic Considerations 275

neither PLP nor DM 20 is necessary for normal ments. Oligodendroglia may be particularly sensi-
myelin assembly. These mice develop widespread tive to an imbalance in the synthesis and turnover
wallerian degeneration of CNS axons, demonstrating of myelin components due to their high rate of
that PLP expression in oligodendrocytes is necessary myelin synthesis. Considering the premature death of
for the maintenance of normal axonal integrity, prob- oligodendrocytes in PLP gene duplications, it is likely
ably through oligodendrocyte–axonal interactions. that a death program is triggered early in the disease
Many different mutations in the PLP gene related course.
to PMD have been identified. Gene duplications are Gene deletions and null mutations do not lead to
the most common cause of PMD and account for over accumulation of mutated protein in the endoplasmic
60% of the mutations. Furthermore, missense muta- reticulum, do not cause increased oligodendrocyte
tions, insertions, deletions, and nonsense mutations cell death and arrest of myelination, and lead to a
have been found. Point mutations do not only occur mild phenotype. It is intriguing that in these condi-
in the coding regions of the gene, but may also affect tions the formation of compact myelin can proceed,
splice sites and noncoding regions of the gene. The while PLP and DM 20 are absent. In null mutations
severity of the clinical phenotype is related to the na- axonal abnormalities with wallerian degeneration are
ture of the mutations. The most benign course of found in the CNS. This suggests that PLP has a role in
PMD is seen in PLP gene deletions or null mutations, glial–axon communication and is somehow neces-
whereas most missense mutations lead to a serious sary for axonal maintenance. In patients with absent
disease with neonatal or prenatal onset. Gene duplica- PLP expression, a demyelinating peripheral neuro-
tions lead to a disease of intermediate severity (usual- pathy has been reported. It has been demonstrated
ly the classical phenotype). The disease severity in that PLP but not DM 20 is necessary for peripheral
duplications is proportional to the degree of overex- nerve function, suggesting that the PLP-specific
pression of the PLP gene. In some mutations only PLP domain plays an important role in this respect.
is altered whereas DM 20 is produced correctly, result- The cause of the secondary neurological deteriora-
ing in the more benign phenotype of SPG2. SPG2 may tion in PMD is insufficiently clarified. It is possible
also be seen in PLP null mutations or point mutations that the small amount of myelin present is unstable,
in some nonconserved regions of the PLP gene. leading to a very slow breakdown. In some patients
The mode of action of changes in or absence of histopathology provides evidence for axonal loss.
PLP has not yet been fully elucidated. There is exper- There seems to be an inverse relationship between
imental support for the concept that missense muta- initial clinical severity of the disease (and the severi-
tions cause conformational changes of the protein, ty of the myelin deficit) and axonal loss.Axonal loss is
“misfoldings” that prevent proper processing of PLP especially seen in patients with milder disease and
and DM 20 after biosynthesis in the endoplasmic more myelin, for instance in patients with a null
reticulum. The accumulation of the mutated proteins mutation.
in the endoplasmic reticulum of oligodendrocytes As a rule PMD affects hemizygous males. However,
leads to the so-called unfolded protein response, from the beginning the occurrence of PMD has also
which sets into motion an apoptotic cascade. The been noticed in girls and women. This phenomenon
greater the accumulation of mutated proteins, the has been ascribed to highly unfortunate X inactiva-
more intense the unfolded protein response and the tion. Some female patients may have transient neuro-
higher the likelihood of apoptosis of oligodendro- logical abnormalities of variable severity as children
cytes. Oligodendroglial cell death leads to hypomyeli- but gradually improve over a period of several years.
nation. In mutations in which PLP is mutated but nor- This functional restoration is attributed to ongoing
mal DM 20 is produced, the trafficking of mutated PLP myelination by oligodendrocytes, in which the
to the cell surface is disrupted, but the trafficking of healthy X chromosome is not inactivated. However, in
DM 20 is normal, a situation that leads to milder dis- most patients the occurrence cannot be explained by
ease. Much more myelin is produced than in classical mechanisms related to X inactivation. It has become
PMD, but the myelin is less stable. Myelin instability apparent that female patients are more common in
and loss may contribute to the clinical phenotype. families with milder forms of PMD or SPG2. Women
In gene duplications, the excessive biosynthesis of heterozygous for PLP mutations are mosaic: one pop-
PLP and DM 20 has deleterious effects on oligoden- ulation of oligodendrocytes expresses normal PLP
droglia, leading to CNS hypomyelination and sub- and synthesizes normal myelin sheaths, whereas the
sequent demyelination, but there is no evidence of other population expresses a mutant form of PLP. The
activation of the unfolded protein response. Over- lack of phenotype in most female PMD carriers can
expression of the PLP gene leads to arrested matura- be attributed to the phenomenon that oligodendro-
tion and death of oligodendrocytes. Overexpressed cytes in which the mutant X chromosome is activated
PLP is routed to late endosomes/lysosomes and caus- become apoptotic and are replaced by healthy oligo-
es sequestration of cholesterol in these compart- dendrocytes. Most of the resulting population con-
276 Chapter 34 Pelizaeus–Merzbacher Disease and X-linked Spastic Paraplegia Type 2

sists of healthy oligodendrocytes at the peak of myeli- speckled, possibly reflecting the presence of some
nation. In contrast, in female carriers of a mild muta- myelin in a tigroid pattern.Atrophy of brain stem and
tion the mutant oligodendrocytes survive and com- cerebellum may be striking. If MRI is performed dur-
pete with normal oligodendrocytes. They produce ing the first few months of life, the images are not di-
structurally flawed myelin sheaths, which are suscep- agnostic as they merely show some atrophy and delay
tible to degradation. Because the affected females of myelination or may even be near-normal in ap-
maintain a population of normal oligodendrocytes, pearance, but repeated MRI confirms the absence of
they never display as marked a phenotype as male progress of myelination. The described pattern of
members of the same family. myelin deficiency in a boy who is a few years old is
highly suggestive of PMD. Proton MRS of the cerebral
white matter may give normal results. In some pa-
34.5 Therapy tients an increased concentration of N-acetylaspar-
tate is found, probably related to denser axonal pack-
Apart from supportive care there is presently no ing in the absence of normal amounts of myelin.
effective therapy for PMD and SPG2. Transplantation Arrest of myelination may be seen in other condi-
of myelin forming cells is a promising strategy. tions, such as severe asphyxia or late congenital infec-
tions, but as a rule in these conditions additional
focal brain lesions are present which are lacking in
34.6 Magnetic Resonance Imaging PMD. Most conditions other than PMD, having an
adverse effect on myelination, lead to delayed but
CT scanning is of little help in the diagnosis of PMD, slowly progressive myelination, with advancement of
showing only atrophy. the degree of myelination on each repeat MRI if made
In contrast, the MRI pattern in PMD is usually after a sufficiently long interval. Hence, repeated MRI
highly suggestive of the disorder. In most cases, MRI is of help in establishing the diagnosis of PMD. Seri-
shows an arrest of myelination in a stage that is in ous and permanent hypomyelination is also present
itself normal (Figs. 34.1 and 34.2). There is a correla- in some DNA repair disorders and sialic acid storage
tion between the amount of myelin present and the disorders. These can be ruled out on the basis of clin-
clinical severity of the disease. In some cases of con- ical findings and appropriate biochemical tests. In ad-
natal PMD, no myelin at all is seen. The T2-weighted dition, patients with Cockayne syndrome typically
images show a high signal intensity of all unmyelinat- have calcium deposits within the basal ganglia, a phe-
ed white matter structures, whereas these structures nomenon lacking in PMD.
have a low signal intensity on T1-weighted images. In In cases of mild PMD, related to gene deletions or
fact, no high signal intensity areas are seen on T1- null mutations, more myelin is present in the cerebral
weighted images in these cases. In cases of classical hemispheres and corpus callosum (Figs. 34.3 and
PMD myelin is present in (parts of) the brain stem, 34.4). The pattern of myelination may be patchy and
(parts of) the cerebellar white matter, (parts of) the myelination may involve the subcortical areas in par-
posterior limb of the internal capsule, the thalamus, ticular. In a family with multiple males with mild
and the globus pallidus. Often the pyramidal tracts in PMD related to a null mutation (initiation codon
the brain stem lack myelin while the brain stem is mutation) we found considerable although incom-
otherwise better myelinated. In some cases addition- plete initial myelination of the cerebral hemispheres
al myelin is present in the directly periventricular (Fig. 34.3), with subsequent cerebral atrophy and
part of the corona radiata, in the subcortical white diffuse loss of myelin in a way seen in primary neuro-
matter and cortex of the pre- and postcentral gyri, degenerative disorders, suggesting underlying axonal
and in the directly periventricular part of the optic degeneration and secondary loss of myelin (Fig. 34.4).
radiation. The myelinated structures have a low signal In MRS of one of these patients a strikingly decreased
intensity on T2-weighted images and a high signal in- level of N-acetylaspartate was found within the cere-
tensity on T1-weighted images. The pattern described bral white matter, in agreement with axonal degener-
is a normal stage of myelination for a neonate or an ation. Decreases in N-acetylaspartate within the
infant in the first few months of life, but not normal cerebral white matter have also been found in other
for the age of the patient. In addition, the cerebral patients lacking PLP.
white matter is variably but often markedly reduced Some female carriers of PMD have been shown to
in volume with a mild enlargement of the ventricular have multiple foci of increased signal intensity in the
system, a thin corpus callosum, folding of the cortex cerebral white matter, but others have not. MRI is not
in thin, deep gyri, and enlargement of the subarach- suitable as a tool for carrier identification. In some
noid spaces (Fig. 34.1). The appearance of the white more seriously affected females, an MRI pattern of
matter is often not completely identical to normal profound myelin deficit may be seen, similar to the
unmyelinated white matter, but may be somewhat pattern observed in male patients.
34.6 Magnetic Resonance Imaging 277

Fig. 34.1. Boy, 29 months old, with classical PMD. The T1- presence is consistent with arrest of myelination soon after
weighted images (third row) show presence of myelin in the birth. There is some cerebral atrophy. Courtesy of Dr. J.J.M. van
central areas. On the T2-weighted images as well, some myelin Collenburg, Zwolle, The Netherlands
is seen in the cerebellum and brain stem.The pattern of myelin

In SPG2 MRI abnormalities vary. Diffuse serious signal on T1-weighted images. In other patients wide-
white matter changes may be seen as in males with spread or more limited focal lesions are seen within
classical PMD, with a high signal of the white matter otherwise well myelinated white matter. Proton MRS
on T2-weighted images and a low signal on T1-weight- of affected white matter in SPG2 patients has demon-
ed images. In some cases the MRI is suggestive of dif- strated decreased levels of N-acetylaspartate, indica-
fuse or patchy mild hypomyelination with a mildly el- tive of axonal damage or loss.
evated signal on T2-weighted images, but also a high
278 Chapter 34 Pelizaeus–Merzbacher Disease and X-linked Spastic Paraplegia Type 2

Fig. 34.2. A 6-year-old boy with classical PMD. The T1-weight- matter has a high signal intensity throughout. The brain stem
ed images reveal some myelin in the central white matter and cerebellar white matter contain more myelin,but less than
(third row), but the T2-weighted images show that the white normal. It is striking that this patient has no atrophy
34.6 Magnetic Resonance Imaging 279

Fig. 34.3. This 2.5-year-old boy has a relatively mild form of hemispheric white matter has a high signal; some deep white
PMD and an initiation codon mutation (Sistermans et al. 1996). matter in the parieto-occipital region has a low signal.The cor-
The T1-weighted images (third row) suggest an advanced pus callosum and brain stem also have a low signal intensity
stage of myelination, but the T2-weighted images demon- on the T2-weighted images
strate that myelination is far from complete. Most cerebral
280 Chapter 34 Pelizaeus–Merzbacher Disease and X-linked Spastic Paraplegia Type 2

Fig. 34.4. The same boy as in Fig. 34.3, 8 years later. He has lost between white and gray matter, indicative of diffuse myelin
myelin. The corpus callosum and deep parieto-occipital white loss. Compared to 8 years ago, there is some diffuse cerebral
matter now have a high signal on the T2-weighted images. atrophy
The T1-weighted images (third row) show a loss of contrast
Chapter 35

18q– Syndrome

35.1 Clinical Features notype or not is still controversial. The deletion in the
and Laboratory Investigations 18q– syndrome includes the locus for the myelin basic
protein gene (18q22–23). It is likely that the impair-
The 18q– syndrome is an autosomal deletion disorder ment in myelination of the CNS is at least partially
with variable phenotype. Most patients have a de no- related to haplo-insufficiency of the myelin basic pro-
vo deletion, but in some patients it is inherited. The tein gene. In patients with an interstitial deletion of
most frequent disease characteristics include mental chromosome 18, which retains the myelin basic pro-
retardation, short stature, microcephaly, midface tein region, myelination is normal (Linnankivi et al.
hypoplasia, hypertelorism, epicanthus, carp-shaped 2003). The two most important proteins of CNS
mouth, high or cleft palate, preauricular skin tags, myelin are proteolipid protein and myelin basic pro-
narrow or atretic ear canals, sensorineural or conduc- tein. Myelin basic protein accounts for 30–40% of the
tive hearing deficit, short neck, tapering fingers, clin- total myelin protein, proteolipid protein for 40–50%.
odactyly, proximally placed thumbs, prominent fin- The 18q– syndrome could be considered to be the
ger whorls, widely spaced nipples, congenital heart autosomal counterpart of X-linked Pelizaeus–Merz-
disease, genital abnormalities, and foot deformities. bacher disease, which is caused by mutations of the
Mental capacities vary from borderline to severely proteolipid protein gene. However, an important dif-
deficient. Apart from mental retardation, neurologi- ference is that one normal myelin basic protein gene
cal abnormalities include hypotonia, seizures, nystag- is present in the 18q– syndrome, whereas no normal
mus, poor coordination, tremor, and choreoathetosis. proteolipid protein gene at all is present in males
Routine and metabolic laboratory investigations suffering from Pelizaeus–Merzbacher disease. Peli-
reveal no abnormalities. IgA deficiency and abnor- zaeus–Merzbacher disease is characterized by severe
malities in growth hormone production are relatively impairment of myelination of the CNS. The extent of
frequent. Peripheral nerve conduction is normal. impairment of myelin deposition is less severe and
Study of evoked potentials may reveal prolonged cen- more variable in the 18q– syndrome. The degree to
tral conduction. Chromosomal analysis reveals a par- which myelination is affected in the 18q– syndrome
tial deletion of the long arm of chromosome 18, most has been found to correlate with the severity of the
often including the bands q22.3Æqter. Inversion of other features, whereas the myelin basic protein gene
the long arm of chromosome 18 with loss of the 18q23 is included in the deletion in all patients. It is there-
region, translocations involving 18q, ring chromo- fore unlikely that the presence of only one copy of the
some 18, and interstitial 18q23 deletions may also be myelin basic protein gene is solely responsible for
seen. producing the abnormalities in myelination.
The so-called shiverer mouse has an autosomal re-
cessive disease related to a mutation of the myelin ba-
35.2 Pathology sic protein gene. In this mouse a defect in CNS myeli-
nation is found. Clinical disease is characterized by
Reduction of cerebral white matter and delay of generalized action tremor, increasingly frequent con-
myelination are the main histopathological findings. vulsions, and premature death. Histopathological ex-
Ventricles and subarachnoid spaces may be mildly amination reveals that CNS myelin is largely absent
enlarged. and, when present, appears as abnormal whorls of cy-
toplasm-filled membranes, tightly compacted at the
intraperiod line, but uncompacted at the major dense
35.3 Pathogenetic Considerations line. The so-called myelin-deficient mouse has a du-
plication of the myelin basic protein gene and a low
18q– Syndrome is a contiguous gene syndrome. It is level of myelin basic protein mRNA. The phenotype
likely that haplo-insufficiency of genes located in the of the myelin-deficient mouse is similar to that of the
deleted region explains the clinical phenotype and shiverer mouse, but less severe.
that the variability of the phenotype depends on the Myelin basic protein is also a component of PNS
exact genes deleted. However, whether there is a cor- myelin. A curious feature is that absence or mutation
relation between the size of the deletion and the phe- of the gene has little effect on PNS myelin. PNS is on-
282 Chapter 35 18q– Syndrome

Fig. 35.1. A 2.5-year-old girl with 18q- syndrome. Note the nal intensity in the periventricular region.The corpus callosum
delay in myelination, leading to poor contrast between gray is relatively well myelinated. The gyri have a relatively thin,
and white matter. There are additional spots of abnormal sig- atrophic appearance

ly subtly altered and is functionally normal. It is sug- 35.4 Therapy


gested that some component specific to peripheral
myelin is functionally equivalent to myelin basic pro- Supportive care is the only therapeutic option.
tein and capable of substituting for this protein in its
absence.
35.5 Magnetic Resonance Imaging 283

Fig. 35.2. The transverse T2-weighted MRI series of this 3- in the temporal lobes, where the white matter still has a high-
year-old boy shows widespread patches of hypomyelination. er signal intensity than the cortex. Courtesy of Prof. Dr. U.
Myelination is delayed for the age of the child, most evidently Stephani, Kiel and Prof. Dr. B.Terwey, Bremen, Germany

35.5 Magnetic Resonance Imaging and gray matter (Fig. 35.1). The myelin deficiency in
the cerebral hemispheric white matter is often patchy
MR images show a variable myelin deficit. Initially, with focal white matter signal abnormalities (Fig.
myelination is delayed (Fig. 35.1) but further ad- 35.2). In some patients, hypomyelination of the corti-
vanced at every follow-up MRI. In older patients a sta- cospinal tracts in the brain stem and posterior limb of
ble picture of incomplete myelination is seen. The the internal capsule is present, but in most other pa-
severity of the myelin deficit is variable. In some pa- tients these structures contain a considerable amount
tients, a near-total absence of myelin in cerebral and of myelin. The corpus callosum seems to have a rela-
cerebellar white matter, internal capsule, and corti- tively normal myelin content. Measurements of white
cospinal tracts is seen with relatively normal myelina- matter T1 and T2 relaxation times show significant
tion of the corpus callosum. The cortical gyri are rel- prolongation in patients with the 18q– syndrome,
atively thin and the sulci relatively deep due to reduc- even in structures that seem well myelinated such as
tion in white matter volume. Other patients have bet- the corpus callosum, compatible with incomplete
ter myelination. The partial hypomyelination often myelination. In some patients the lateral ventricles
leads to poor differentiation on MRI between white are mildly enlarged.
Chapter 36

Phenylketonuria

36.1 Clinical Features They often have a mousy, musty odor. Seizures may
and Laboratory Investigations occur, taking the form of tonic–clonic seizures, my-
oclonic seizures, and infantile spasms. Irritability, fre-
Phenylketonuria (PKU) represents a heterogeneous quent vomiting, and insufficient growth are part of
group of disorders with autosomal recessive inheri- the clinical picture. The final mental level of PKU
tance. PKU, or hyperphenylalaninemia, is caused by children is deficient, but the children are trainable.
a deficiency of the phenylalanine hydroxylating sys- Verbal IQ is lower than performance IQ. The children
tem. The phenylalanine hydroxylating system con- are small and often microcephalic. Pyramidal signs
sists of two essential components: phenylalanine are present with hypertonia, gait disturbances, hyper-
hydroxylase and coenzyme tetrahydrobiopterin reflexia, and extensor plantar reflexes. Frequently a
(Figs. 36.1 and 36.2). PKU is in most patients caused rapid, fine, and irregular tremor of the hands is pre-
by deficiency of phenylalanine hydroxylase, and in a sent. Abnormal choreoathetoid movements may be
minority of patients by deficiency of tetrahydro- present with twisting movements, continuous repeti-
biopterin. tious finger movements, and rhythmical swinging
Classical PKU is due to severe phenylalanine hy- body movements. Psychiatric disturbances with se-
droxylase deficiency. Infants with this disease are vere hyperactivity, destructiveness, self-mutilation,
normal at birth. In the course of the first year of life and uncontrollable attacks of rage or excitement are
psychomotor retardation becomes evident. Eczema is common.
present in a considerable number of patients, usually Worldwide neonatal screening programs have led
from infancy to late childhood. The patients often, but to early detection of almost all PKU patients. Early
not always, have blond hair, fair skin, and blue eyes. dietary treatment prevents most of the described ab-

Fig. 36.1. Metabolism of phenylalanine


36.1 Clinical Features and Laboratory Investigations 285

normalities, which are now rarely seen. Intelligence is trauterine and extrauterine growth retardation,
usually within the normal range, although mean in- microcephaly, and mental handicap, occasionally
telligence in patients with classical PKU who are accompanied by malformations of the heart or other
treated early is roughly half a standard deviation low- organs. The incidence of these aberrations appears to
er than the IQ of unaffected siblings and population be correlated with the maternal concentration of
norms. Treated PKU patients are generally slower to serum phenylalanine. Reinstigation of dietary treat-
acquire language and have a higher frequency of ment prior to conception has a favorable effect on
learning difficulties and behavioral disturbances, outcome.
with hyperactivity, anxiety, and poor concentration. Mild variants of classical PKU lead to less severe
The better the dietary control in early and middle mental retardation if untreated. Patients tolerate a
childhood, the better the outcome. Despite neonatal larger intake of phenylalanine and successful treat-
screening programs, there are still a number of pa- ment requires a less rigid diet than in the case of se-
tients with PKU who are not diagnosed in the neona- vere classical PKU.
tal period and start treatment late. In general, the lat- In so-called persistent hyperphenylalaninemia
er the onset, the lower the IQ scores. Strict dietary mildly elevated serum phenylalanine levels are found,
treatment was formerly thought to be important only but there are no clinical symptoms and treatment is
during the period of major cerebral maturation and not necessary.
used to be moderated or stopped in mid to late child- Approximately 2% of newborns with PKU have a
hood. However, there is now evidence that with dis- tetrahydrobiopterin deficiency. In the most severe pa-
continuation of strict diet and a rise in phenylalanine tients microcephaly, developmental delay, and pro-
levels, some cognitive deterioration and emotional gressive neurological dysfunction occur, leading to
problems develop in many patients, together with an death in childhood, although some patients stabilize
increase in motor problems consisting of tremor and or make some developmental progress. Early in life
signs of spasticity of the legs. Reinstitution of strict patients present with microcephaly, disturbed psy-
diet may result in disappearance of these problems. chomotor development, and the other neurological
Maternal PKU appears to have detrimental effects problems associated with hyperphenylalaninemia. In
on embryogenesis and fetal development. The off- addition, signs of extrapyramidal dysfunction are
spring of females with classical PKU, who are no prominently present and consist of parkinsonism,
longer being treated, show a high frequency of in- chorea, dystonia, oculogyric spasms, or myoclonus.

Fig. 36.2. Synthesis of tetrahydrobiopterin


286 Chapter 36 Phenylketonuria

Typical signs of parkinsonism are hypokinesia, biopterin levels are extremely low. Examination of
bradykinesia, rigidity, and mask-like facies. In addi- urine pteridine profiles allows a tentative diagnosis of
tion, sweating, hyperpyrexia without infection, drool- the basic defect. In cases of tetrahydrobiopterin defi-
ing, swallowing difficulties, pinpoint pupils, truncal ciency, CSF and urine levels of homovanillic acid
hypotonia, limb hypertonia and hyperreflexia, infan- (HVA), 5-hydroxyindoleacetic acid (5HIAA), vanillyl
tile spasms, and tonic–clonic seizures often form part mandelic acid (VMA), and 3-methoxy-4-hydrox-
of the clinical picture. In milder forms, symptoms are yphenylglycol (MHPG), the major metabolites of
less marked and consist mainly of extrapyramidal dopamine, serotonin, and (nor)epinephrine in the
dysfunction or slow development. In patients with human CNS, are reduced. The levels of these metabo-
pterin-4a-carbolamine dehydratase as the cause of lites may also be reduced, although less markedly, in
the tetrahydrobiopterin deficiency, there are general- classical PKU. However, in classical PKU the levels
ly no significant clinical abnormalities other than normalize with a phenylalanine-restricted diet, which
transient alterations in tone. In tetrahydrobiopterin is not the case in disorders characterized by tetra-
deficiency caused by a defect in dihydropteridine re- hydrobiopterin deficiency. In dihydropteridine re-
ductase, rapidly progressive demyelination of the ductase deficiency, serum folate levels may be low.
CNS may occur additionally, due to defective folate The diagnosis should be confirmed by enzyme stud-
metabolism, and cause spasticity, pseudobulbar palsy, ies in erythrocytes (dihydropteridine reductase, 6-
long tract sensory loss, and cognitive deterioration. pyruvoyl tetrahydropterin synthetase), lymphocytes
Neonatal screening is performed between 6 and 14 (guanosine triphosphate cyclohydrolase), or liver
days after birth. Increased blood phenylalanine con- biopsy (6-pyruvoyl tetrahydropterin synthetase,
centration indicates a positive test. In phenylalanine guanosine triphosphate cyclohydrolase). Prenatal
hydroxylase deficiency, phenylpyruvic acid and o-hy- diagnosis can be performed by assessing pterin levels
droxyphenylacetic acid are excreted in the urine in amniotic fluid or enzyme activity in fetal erythro-
(Fig. 36.1). The blood level of tyrosine is normal or cytes, amniocytes, or chorionic villi, depending on
lower than normal, which excludes hyperphenylala- the different enzyme defects. A DNA-based prenatal
ninemia secondary to disorders of tyrosine metabo- diagnosis is possible in families with known muta-
lism. Apart from tyrosinemia, hyperphenylalanine- tions.
mia secondary to prematurity, hepatic insufficiency,
chronic renal insufficiency, and trimethoprim med-
ication should be considered. An oral phenylalanine 36.2 Pathology
loading test is performed when the child is several
months old and leads to elevated serum levels of In untreated classical PKU, the weight of the brain is
phenylalanine without elevation of tyrosine. Severe below normal. The reduction in volume is greater in
and mild variants of phenylalanine hydroxylase defi- the white than in the gray matter. The ventricles are
ciency are distinguished by the level of serum pheny- enlarged. The cortical pattern of gyri and sulci is nor-
lalanine before treatment, the level of serum pheny- mal.
lalanine and the duration of the increase after pheny- Microscopic examination of the cortex reveals evi-
lalanine loading, and the tolerance of phenylalanine dence of developmental arrest or delay of cortical
in the diet. Residual phenylalanine hydroxylase activ- neurons, with a reduction in number and size of cor-
ity can be determined in liver biopsy material. Prena- tical neurons, paucity of dendritic arborization, and
tal diagnosis and carrier detection can be performed reduced synaptic density.
with the help of DNA techniques. In untreated PKU children variable white matter
If elevated serum phenylalanine concentrations abnormalities are seen ranging from hypomyelina-
are found on neonatal screening, pterins in urine are tion and spongy change to frank demyelination.
analyzed and dihydropteridine reductase activity is Some PKU patients do not have white matter abnor-
measured to exclude dihydropteridine reductase defi- malities, but the majority of the patients have white
ciency and other disorders in the synthesis and recy- matter lesions. Hypomyelination is usually seen, af-
cling of tetrahydrobiopterin (Fig. 36.2).Assessment of fecting most severely the structures that myelinate
the ratio between total biopterin and neopterin in late. In addition, a status spongiosus of the white mat-
urine is helpful in differentiating between the various ter is usually present. The status spongiosus is most
enzyme defects. In dihydropteridine reductase defi- marked in the optic tracts, periventricular white mat-
ciency, the total biopterin level is very high, whereas ter, centrum semiovale, and in the central part of the
the neopterin level is normal or moderately in- cerebellar white matter, although at times spongy
creased; in 6-pyruvoyl tetrahydropterin synthetase lesions are also seen in the subcortical white matter.
deficiency, the neopterin level is elevated and the Apart from occasional sudanophilic fat droplets in
biopterin level is very low; and in guanosine triphos- perivascular spaces, there are no signs of active
phate cyclohydrolase deficiency both neopterin and myelin loss. Gliosis is variable. In some patients, espe-
36.4 Pathogenetic Considerations 287

cially older untreated children and adults, areas of hydropterin synthetase affect tetrahydrobiopterin
demyelination are seen in the periventricular and synthesis.
deep white matter, sparing the subcortical U fibers. In The phenylalanine hydroxylase gene, PAH, is locat-
some cases the areas are sharply defined; in others the ed on chromosome 12q22–24.1. A large number of
borders of the lesions are indistinct. In the areas of different mutations have been reported. There is evi-
demyelination intense gliosis and deposition of su- dence that the phenotypic heterogeneity of the disor-
danophilic material is seen. Oligodendrocytes are der reflects underlying genetic heterogeneity. Some
reduced in number. The axis cylinders are relatively of the mutations lead to absence of residual hydroxy-
better preserved than the myelin sheaths. The cere- lase activity, whereas others result in the presence of
bellar white matter may also be involved. Spongy some variable residual activity. A good correlation
lesions may be noted adjacent to the demyelinated has been found between the residual level of activity
areas. With advancing age white matter changes tend of mutant phenylalanine hydroxylase enzyme and the
to become more pronounced and diffuse, although clinical severity of the disease. The residual enzyme
not in all cases. activity also correlates with the pretreatment serum
In patients with dihydropteridine reductase defi- level of phenylalanine, serum phenylalanine levels
ciency, additional neuronal loss, abnormal vascular measured in standardized loading tests, and phenyl-
proliferation, and calcium deposits in the walls of alanine tolerance. Severe classical PKU is caused by
small, medium, and large arteries and veins, as well as mutations with zero residual enzyme activity. Higher
diffusely scattered pericapillary and isolated calcium enzyme activity is associated with a milder pheno-
deposits have been reported in the basal ganglia, thal- type. In persistent benign hyperphenylalaninemia
amus (Milandi et al. 1998), cerebral cortex, and white without clinical manifestations the residual enzyme
matter (Takashima et al. 1991). Diffuse myelin pallor activity is relatively high. However, the genotype is
of the cerebral and cerebellar white matter, spongy not the only factor determining the phenotype. Dif-
white matter changes, and multifocal perivascular ferent clinical outcomes despite comparable dietary
demyelination occur as well. control form evidence for additional factors. Phenyl-
alanine is transported over the blood–brain barrier,
and there is evidence that individual differences in
36.3 Chemical Pathology this transport contribute to the severity of the neuro-
logical problems. The higher the affinity of the trans-
Analysis of cerebral lipids reveals a deficiency of porter for phenylalanine, the higher the cerebral
myelin lipids, particularly cerebroside, sulfatide, and phenylalanine levels despite similar blood phenyl-
cholesterol. In the few studies performed, the defi- alanine levels, and the lower the IQ.
ciency of myelin lipids is more severe in older pa- Deficiency of phenylalanine hydroxylase results in
tients. Cholesterol esters are absent or present in only the accumulation of phenylalanine, which is transam-
small amounts. inated to phenylpyruvate, phenyllactate, phenylac-
etate, and o-hydroxyphenylacetate (Fig. 36.1). The
metabolites derived from phenylalanine contribute
36.4 Pathogenetic Considerations little to the cerebral damage in PKU. Probably the
most important effects are related to direct toxic in-
Phenylalanine is an essential amino acid, ubiquitous fluences of phenylalanine itself. In theory, deficiency
in dietary protein. It is normally transformed into of phenylalanine hydroxylase activity could lead to
tyrosine, which is in turn used for protein synthesis systemic tyrosine deficiency. However, no consistent
and is the immediate amino acid precursor for or significant reduction in plasma tyrosine has ever
melanin, dopamine, norepinephrine, and epineph- been demonstrated in untreated PKU patients, and
rine. The conversion of phenylalanine to tyrosine postnatal tyrosine supplementation does not prevent
requires the presence of phenylalanine hydroxylase occurrence of neurological abnormalities. Hyper-
and tetrahydrobiopterin as a proton donor (Figs. 36.1 phenylalaninemia has a number of adverse effects.
and 36.2). Tetrahydrobiopterin is reconverted from Owing to the competitive nature of amino acid trans-
dihydrobiopterin by dihydropteridine reductase port across the blood–brain barrier, the brain in pa-
(Fig. 36.2). In the majority of cases, PKU is caused by tients with PKU is exposed to both high phenylala-
a deficiency of the hepatic enzyme phenylalanine hy- nine levels and low concentrations of other large neu-
droxylase, and in a minority (± 2%) by tetrahydro- tral amino acids, including histidine, tyrosine, trypto-
biopterin deficiency, caused by a defect in its recy- phan, threonine, valine, methionine, isoleucine, and
cling or its synthesis (Fig. 36.2). Defects in dihy- leucine, especially methionine and tyrosine. Hyper-
dropteridine reductase and pterin-4a-carbinolamine phenylalaninemia inhibits protein synthesis, which
dehydratase affect the recycling. Defects in guanosine may be related to the inhibition of transport of amino
triphosphate cyclohydrolase I and 6-pyruvoyl tetra- acids across the blood–brain barrier. It may be that
288 Chapter 36 Phenylketonuria

the hypomyelination, almost invariably seen in un- melatonin; L-dopa is the precursor of dopamine, epi-
treated PKU patients, is related to the inhibition of nephrine, and norepinephrine. In addition, hyper-
protein synthesis. High levels of phenylalanine have a phenylalaninemia has a competitive inhibitory effect
double effect on tyrosine and tryptophan metabo- on tyrosine hydroxylase and tryptophan hydroxylase.
lism. Not only is the transport across the blood–brain So, deficiency of these enzymes has a profound effect
barrier inhibited, but high phenylalanine levels also on multiple neurotransmitter systems, which ex-
lead to competitive inhibition of the enzymes tyro- plains many of the neurological features. This effect
sine hydroxylase and tryptophan hydroxylase, en- cannot be improved by lowering phenylalanine intake
zymes involved in the synthesis of dopamine, only. A special aspect of dihydropteridine reductase
(nor)epinephrine (from tyrosine), and serotonin deficiency is related to folate metabolism. The en-
(from tryptophan). As a result of both enzyme inhibi- zyme also reduces dihydrofolate to tetrahydrofolate.
tion and deficiency of the amino acid substrates for Defective folate metabolism is important in dihy-
these enzymes, the neurotransmitters dopamine, dropteridine reductase deficiency. Megaloblastic
(nor)epinephrine, and serotonin are decreased in un- changes of blood cells are unusual, even in very low
treated patients with phenylalanine hydroxylase defi- folate concentration. However, progressive neurologi-
ciency. cal damage is frequent. Histological changes in the
It is important to distinguish permanent from re- brain are similar to those seen in congenital folate
versible effects of hyperphenylalaninemia. Reversible malabsorption and 5,10-methylene tetrahydrofolate
neuronal dysfunction, which disappears on treat- reductase deficiency, and consist of multifocal
ment, is probably related to neurotransmitter dys- perivascular demyelination.
function. If left untreated, reversible dysfunction may
become irreparable damage consisting of a reduction
in the number and growth of neuronal cells and 36.5 Therapy
synapses. The defect in protein synthesis may con-
tribute to this damage. Reversible white matter dam- Classical PKU is treated with a low-phenylalanine
age consists of delay in myelination and white matter diet. Some patients with phenylalanine hydroxylase
sponginess, probably related to a combination of deficiency are tetrahydrobiopterin-responsive. These
phenylalanine toxicity on cells and disturbed protein patients benefit from use of tetrahydrobiopterin and
synthesis. Longstanding, severely disturbed myelina- may follow a less strict diet or avoid the diet altogeth-
tion and demyelination may only be partially re- er. The low-phenylalanine diet depends on the use of
pairable. The vulnerability of the brain for hyper- synthetic, phenylalanine-low substitutes for many
phenylalaninemia is life-long. Discontinuation of diet natural foods (meat, fish, eggs, nuts, dairy products,
may again lead to neuronal dysfunction with epilep- bread), making the diet difficult to sustain over long
sy, enhanced occurrence of EEG abnormalities and periods. Treatment should be monitored regularly
cognitive decline, and to white matter damage as evi- with assessment of serum phenylalanine levels. Both
dent from MRI. early start of dietary treatment and strict compliance
The tetrahydrobiopterin deficiency states can be with the diet contribute to a favorable intellectual out-
divided into defects in synthesis and defects in recy- come. IQ clearly correlates to the quality of dietary
cling (Fig. 36.2). Defects in synthesis are related to control. It is also important to be careful that patients
deficiency of guanosine triphosphate cyclohydrolase do not develop nutritional deficiencies, in particular
or 6-pyruvoyl tetrahydropterin synthetase. The genes vitamin B12 deficiency, from failure to consume ap-
encoding these enzymes, GCH1 and PTS, are located propriate supplements. Protein insufficiency, which
on chromosomes 14q22.1–22.2 and 11q22.3–23.3, re- leads to impaired growth, can be avoided by monitor-
spectively. Dihydropteridine reductase deficiency and ing plasma prealbumin levels. Current treatment of
pterin-4a-carbinolamine dehydratase deficiency are PKU patients is not perfect, and cognitive level is, on
defects in recycling. The genes encoding these en- average, lower than expected.
zymes, QDPR and PCBD, are located on chromosome For many years it has been customary to stop
4p15.3 and 10q22, respectively. Deficiency of guano- dietary treatment in mid to late childhood or in ado-
sine triphosphate cyclohydrolase most often leads to lescence on the assumption that hyperphenylalanine-
dopa-responsive dystonia without hyperphenylala- mia is only harmful in the immature brain. However,
ninemia. Deficiency of each of the enzymes leads to discontinuation of diet may lead to some cognitive
tetrahydrobiopterin deficiency, which is a cofactor deterioration and emotional problems in many of the
(proton donor) not only in the hydroxylation of PKU patients. They show a lack of concentration, and
phenylalanine to tyrosine, but also in the hydroxyla- sometimes agitation and emotional instability. Hy-
tion of tryptophan to 5-hydroxytryptophan and in pertonia of the legs, hyperreflexia, and extensor plan-
the hydroxylation of tyrosine to L-dopa (Fig. 36.2). 5- tar responses are frequently present and may lead to
Hydroxytryptophan is the precursor of serotonin and difficulty in walking. Epilepsy, slowness of speech,
36.6 Magnetic Resonance Imaging 289

dysarthria, and tremor, in particular intention both the degree of recent exposure to high phenylala-
tremor, may occur. With high levels of phenylalanine nine levels and the number of years since the low-
EEG shows background slowing. The sensitivity for phenylalanine diet was stopped. The presence of
phenylalanine fluctuations is greater among younger white matter changes does not correlate with age at
patients and decreases with age. The complaints may the start of dieting (early versus late treatment) or
be reversible if phenylalanine restriction is resumed, with the quality of dietary control during the first
but may become permanent if longstanding. Life- years of life or subsequent years. These data suggest
long continuation of dietary treatment is advisable. that recently sustained exposure to high phenylala-
In particular, restriction of phenylalanine intake nine concentrations rather than the quality of long-
during pregnancy is important for the benefit of the term control is the major determinant of white matter
offspring. The maternal PKU syndrome, occurring in signal abnormalities as detected by MRI. A few pa-
children born to mothers with untreated PKU, con- tients who developed neurological abnormalities sev-
sists of intrauterine growth retardation, congenital eral months or years after stopping or relaxing the
heart disease, dysmorphic signs, microcephaly, and phenylalanine-restricted diet underwent sequential
mental deficiency. The syndrome can be prevented by MRI studies. Whereas initial MRI investigations
adequate treatment during pregnancy. showed white matter changes, follow-up scans after
In PKU related to tetrahydrobiopterin deficiency, resumption of a strict diet showed resolution of the
the therapeutic goal is two-fold: not only to control abnormalities within a few months in some but not all
the blood level of phenylalanine, but also to normal- patients.
ize monoamine neurotransmission. The administra- The earliest and most frequent abnormalities con-
tion of tetrahydrobiopterin is insufficient to achieve sist of high-signal-intensity lesions on T2-weighted
this latter purpose. The introduction of oral adminis- images in the parieto-occipital periventricular white
tration of a combination of tetrahydrobiopterin, matter (periatrial and peritrigonal region) (Fig. 36.3).
L-dopa, carbidopa, and 5-hydroxytryptophan, given In more severe cases, the frontal periventricular white
to bypass the impaired activity of tyrosine hydroxy- matter is also involved and the white matter abnor-
lase and tryptophan hydroxylases, is followed by malities may extend into the subcortical area, in par-
some (and sometimes a dramatic) degree of clinical ticular in the posterior region (Fig. 36.4). The corpus
and biochemical improvement. Carbidopa is a pe- callosum is relatively, but not always completely
ripheral decarboxylase inhibitor to prevent peripher- spared. The internal capsule, brain stem, and cerebel-
al conversion of L-dopa to dopamine and so prevent lar white matter are most often preserved, although in
systemic adverse effects and enhance central benefi- some patients the cerebellar white matter and brain
cial effects. The treatment should be monitored not stem tracts are involved. The white matter lesions are
only by evaluation of serum phenylalanine levels, but symmetrical and either band-like or patchy and part-
also by assessment of CSF neurotransmitter metabo- ly confluent in an irregular fashion. Additional isolat-
lites. In dihydropteridine reductase deficiency, the ed white matter spots may occasionally be present.
folate disturbance also requires treatment. Tetrahy- Frequently the frontal, less often the occipital white
drofolate should be administered in sufficient matter changes have a peculiar configuration, extend-
amounts to keep CSF concentrations in the high nor- ing like little “flames” from the ventricular border in
mal range. This level of tetrahydrofolate prevents the line with the ventricles. In few patients some general-
occurrence of demyelinating disease. ized cerebral atrophy is seen.
The nature of the white matter changes is still a
matter of conjecture as there are no direct correlative
36.6 Magnetic Resonance Imaging studies comparing MRI and histopathological find-
ings in the same patients. Considering the rapid re-
White matter abnormalities occur with high frequen- versal of MRI changes that may be produced by effec-
cy in patients with classical PKU. These white matter tive dietary intervention, white matter edema may be
changes have been reported in older children, adoles- assumed. In view of the fact that reversibility of MRI
cents, and adults. At present, there is no systematic changes was not confirmed in all cases, and consider-
MRI study involving infants and young children. ing the known histopathological sequence of events
From the studies performed it is apparent that white in untreated PKU patients (hypomyelination Æ white
matter abnormalities are less likely to be present in matter (myelin) vacuolation and edema Æ demyeli-
children under strict dietary control and in patients nation), an attractive hypothesis is that the early MRI
with milder variants of PKU. The white matter changes observed represent white matter edema with
changes are predominantly present in patients after intramyelinic vacuole formation, and that the late
they have stopped the phenylalanine-restricted diet. changes represent permanent myelin damage and
There is a positive correlation between the presence loss. The initial stage is reversible, the later stage is
and severity of the white matter abnormalities and not. Restricted diffusion has been found in the areas
290 Chapter 36 Phenylketonuria

Fig. 36.3. A 24-year-old female patient with classical PKU. eral ventricles. Courtesy of Dr. S. Blaser, Department of Diag-
Note the periventricular zone of mild signal changes. There is nostic Imaging, and Dr. A. Feigenbaum, Department of Clinical
a more prominently abnormal signal in the white matter bor- Genetics, Hospital for Sick Children, Toronto, Canada
dering the frontal and,especially,the posterior horns of the lat-

of white matter abnormalities (Philips et al. 2001; De- tetrahydropterin synthetase deficiency (Brismar et al.
zortová et al. 2001), which would be compatible with 1990; Chien et al. 2002). CT is normal up to the age of
myelin vacuolation as histopathological basis. about 1 year. In older children CT shows atrophy with
Clinically, the white matter changes correlate with enlargement of the ventricular system and subarach-
motor problems, in particular spasticity of the legs, noid spaces. In some of the patients, diffuse hemi-
processing of visual information, sustained attention, spheric white matter hypodensity is seen. MRI shows
and changes in mood and personality. No relation- diffuse hemispheric white matter hyperintensity on
ship between MRI changes and IQ has been found. T2-weighted images in these patients, but in other pa-
Proton MRS permits noninvasive measurement of tients the white matter is normal.
brain phenylalanine levels. MRS has been helpful in In dihydropteridine reductase deficiency a combi-
studying the dynamics of phenylalanine transport nation of gray and white matter changes is found
across the blood–brain barrier after phenylalanine (Figs. 36.5–36.7). CT scan shows cerebral atrophy
loading and in studying the relationship between with enlargement of the ventricular system and sub-
phenylalanine levels in the brain and neurological arachnoid spaces. In addition, calcium deposits
functions. develop and are seen bilaterally in the basal nuclei
The imaging changes observed in patients with (globus pallidus, putamen), thalamus, and cortico-
PKU due to a deficiency of tetrahydrobiopterin are subcortical junction. Variable white matter hypoden-
different from those in classical PKU. There are only a sity is seen. MRI shows some evidence of calcium de-
few reports on imaging findings, and so the incidence position in the areas mentioned, with mottled hyper-
and extent of the abnormalities are not fully known. intensity on T1-weighted images and heterogeneous
Several reports concern patients with pyruvoyl signal loss on T2-weighted images, sometimes in
36.6 Magnetic Resonance Imaging 291

Fig. 36.4. A 17-year-old female patient with classical PKU. She sum is spared.Courtesy of Dr.S.Blaser,Department of Diagnos-
has more prominent signal abnormalities in the periventricu- tic Imaging, and Dr. A. Feigenbaum, Department of Clinical Ge-
lar white matter than the patient in Fig. 36.3.The corpus callo- netics, Hospital for Sick Children, Toronto, Canada

Fig. 36.5. A girl with PKU due to dihydropteridine reductase T1-weighted images.The subcortical white matter has too low
deficiency, age 2 years and 8 months. CT scan (upper row) a signal intensity on the T1-weighted images, too high a signal
shows some cerebral atrophy with dilated lateral ventricles. intensity on the T2-weighted images. The parasagittal T1-
There are calcifications in the striatum and in the frontal and weighted MR image shows a gyriform band of high signal in-
occipital gray and white matter junction. The white matter is tensity in the cortex and corticomedullary junction, probably
hypodense in these areas. The transverse T1- and T2-weighted due to calcification. From Gudinchet et al. (1992), with permis-
MR images at the age of 4.5 years (second row) show high sion
signal intensity in the calcified spots in the basal ganglia on
292 Chapter 36 Phenylketonuria

Fig. 36.6. A 20-year-old male with dihydropteridine reduc- matter in the centrum semiovale, reaching the U fibers and in-
tase deficiency. CT (upper row, left and middle) shows some cal- volving the peritrigonal white matter and external and inter-
cification of the basal ganglia and the parietal corticosubcorti- nal capsule. The frontal white matter is less severely affected.
cal junction. The MRI shows diffuse abnormality of the white From Sugita et al. (1990), with permission
36.6 Magnetic Resonance Imaging 293

Fig. 36.7. A 26-year-old male with dihydropteridine reduc- of-the-tiger phenomenon of Hallervorden–Spatz disease. The
tase deficiency. CT (first row) shows extensive calcifications of transverse T1- (third row) and T2-weighted images, like the CT
the basal ganglia and in the frontal corticomedullary junction. scan, show cyst formation in the occipital lobes. Note the high
The transverse T2-weighted MR images (second row) show en- signal intensity on the T1-weighted image in a calcified areas.
larged ventricles. The basal nuclei have a low signal intensity From Sugita et al. (1990), with permission
with a central area of high signal intensity, similar to the eye-
Chapter 37

Glutaric Aciduria Type 1

37.1 Clinical Features within families. With early treatment the prognosis of
and Laboratory Investigations the patients improves and most or all of the neurolog-
ical damage can be prevented. Acute deterioration
Glutaric aciduria type 1 is an autosomal recessive after instigation of treatment is rare.
metabolic disorder with highly variable clinical In glutaric aciduria type 1, excessive urinary excre-
symptomatology. The disease usually presents with tion of glutaric acid, glutaconic acid, and 3-hydroxy-
an acute encephalitis-like encephalopathy in infancy glutaric acid suggests the diagnosis. The defect in me-
or childhood after a normal initial development. In tabolization of glutaryl-CoA leads to accumulation of
most cases the first episode occurs between the ages this substance in body fluids, partly esterified with
of 6 and 18 months. Frequently, the only abnormality carnitine and excreted as glutarylcarnitine. Carnitine
preceding the first episode is a developing macro- deficiency can be the result of this excessive urinary
cephaly. The macrocephaly may already be present at carnitine excretion. Lysine, hydroxylysine, and tryp-
birth. The encephalopathic episode is usually trig- tophan, precursors of glutaryl-CoA, are not elevated
gered by an infectious illness or vaccination. The in urine or body fluids. During episodes of clinical de-
episode is characterized by hypotonia, loss of head compensation, metabolic acidosis, ketosis, hyperam-
control, spasticity, dystonia, rigidity, orofacial dyski- monemia, and elevation of serum transaminases may
nesia, grimacing, seizures, opisthotonic posturing, be found. Occasionally hypoglycemia occurs in an
lowering of consciousness, and coma. Recovery is acute episode. The diagnosis of glutaric aciduria type
slow and often incomplete. There is usually a preser- 1 may be difficult to establish. Enhanced urinary ex-
vation of intellect. After the first crisis, neurological cretion of the metabolites mentioned may be inter-
signs may remain static for a long time, suggesting a mittent and only detectable during episodes of meta-
diagnosis of postencephalitic damage. More often, the bolic derangement. In some cases the urine concen-
subsequent course of disease is slowly progressive tration of glutaric acid, glutaconic acid, and 3-hydroxy-
with episodes of acute deterioration, often associated glutaric acid may even remain normal during
with an infection. Patients frequently have a tendency episodes of clinical decompensation. In most (but not
to sweat profusely. There may be episodes of un- all) of these patients a decrease in plasma carnitine
explained fever. Ocular abnormalities are frequent, levels and an increase in urinary glutarylcarnitine
including intraretinal hemorrhages, cataract, gaze levels can be found, indicative of glutaric aciduria. In
palsy, strabismus, ametropia, and pigmentary retino- some patients only elevated levels of glutaric acid can
pathy. If untreated, death usually occurs in the first be found in CSF. A definite diagnosis can be estab-
decade, in the course of an episode of acute deteriora- lished by assay of glutaryl-CoA dehydrogenase in
tion during an infection or in the course of a Reye leukocytes and fibroblasts and by means of DNA
syndrome-like episode. In other patients the course of analysis. Asymptomatic siblings of patients with glu-
disease is more chronically progressive with retarded taric aciduria type 1 should always be investigated
development during the first year of life. Subsequent- and, if diagnosed with the same disease, treated. Pre-
ly, hypotonia, dystonia, athetoid involuntary move- natal diagnosis can be performed by enzyme assess-
ments, and spasticity develop gradually over the years ment in chorionic villi or cultured amniotic cells and
with loss of motor skills. The progressive dystonia is by DNA-based tests.
disabling and leads to a loss of walking and writing
abilities. Also, articulate speech may become progres-
sively difficult and may be lost altogether. Mental 37.2 Pathology
capabilities are intact or relatively preserved. Some
patients are stable for several years, suggesting a diag- External examination of the brain may reveal cerebral
nosis of athetoid cerebral palsy. Occasional patients atrophy with an open operculum. Ventricular en-
present as adults with signs of a progressive en- largement is often noted. Most commonly noted
cephalopathy. Some patients remain asymptomatic, parenchymal abnormalities involve the putamen and
and asymptomatic adults have occasionally been head of the caudate nucleus, less often the globus pal-
found during screening of families known to be lidus. Changes tend to be more marked in patients
affected. Thus, there is a large phenotypic variation who die after several years than in infants. The lesions
37.4 Therapy 295

are characterized by loss of neurons associated with mate. Both glutaric acid and 3-hydroxyglutaric acid
gliosis. The cortex is well preserved. are toxic to neurons in culture, and this neurotoxic ef-
Marked spongiform changes are seen in the hemi- fect can be prevented by preincubation with NMDA
spheric white matter in some patients. It is the receptor blockers. In addition, glutaric acid, glutacon-
periventricular white matter that is particularly in- ic acid, and 3-hydroxyglutaric acid are competitive in-
volved, whereas the subcortical U fibers are spared. In hibitors of glutamic acid decarboxylase, the enzyme
addition, optic nerves, corpus callosum, internal cap- involved in biosynthesis of GABA, an inhibitory neu-
sule, deep cerebellar white matter, and long tracts of rotransmitter. Biochemical analysis of the brains of a
the brain stem may be involved.Vacuolation is caused few patients demonstrated elevated levels of glutaric
by myelin splitting and intramyelinic vacuole forma- acid in the frontal cortex and basal ganglia, very low
tion. In electron microscopy splitting is seen to occur glutamic acid decarboxylase activity, and very low
along the intraperiod line. Mild spongiform changes concentrations of GABA in the caudate nucleus and
due to myelin splitting may be seen in gray matter putamen. Low CSF GABA levels have also been found.
structures rich in myelin, such as the thalamus, However, the low GABA is not necessarily related to
globus pallidus, and brain stem reticular formation. inhibition of glutamic acid decarboxylase, but may
also be the result of destruction or dysfunction of
GABA-ergic neurons. Quinolinic acid, an intermedi-
37.3 Pathogenetic Considerations ate in the metabolism of tryptophan and lysine, and a
potent neurotoxin, may contribute to the neuronal
Glutaric aciduria type 1 is caused by deficiency of glu- damage in glutaric aciduria.
taryl-CoA dehydrogenase. Glutaryl-CoA, a catabolite Even less is known about the pathogenesis of
of lysine, hydroxylysine, and tryptophan is dehydro- myelin splitting and vacuolation. Myelin splitting is
genated to glutaconyl-CoA and subsequently decar- probably caused by toxic effects of accumulating sub-
boxylated to crotonyl-CoA, both steps being catalyzed stances.
by a single enzyme, glutaryl-CoA dehydrogenase. The The cause of the macrocephaly has not been ade-
enzyme is active in mitochondria as a homotetramer. quately explained. Contributing factors may be the
In glutaric aciduria type 1 this mitochondrial enzyme presence of extracerebral fluid collections, hydro-
is lacking. A single case of peroxisomal glutaryl-CoA cephalus, and myelin splitting with intramyelinic ac-
dehydrogenase deficiency has been described (Ben- cumulation of fluid. The pathogenesis of the extrac-
nett et al. 1991). Mitochondrial glutaryl-CoA dehy- erebral fluid collections is unexplained.
drogenase is a flavin adenine dinucleotide (FAD)-re-
quiring enzyme. Deficiency of this enzyme activity
has been reported in glutaric aciduria type 1 and in 37.4 Therapy
multiple acyl-CoA dehydrogenase deficiency, also
called glutaric aciduria type 2. During a metabolic crisis, it is essential to correct the
The gene encoding glutaryl-CoA dehydrogenase, metabolic decompensation as soon as possible to pre-
GCDH, is located on chromosome 19p13.2. Many dif- vent and limit the irreversible brain damage. A high-
ferent mutations have been identified in glutaric calorie glucose infusion (if necessary together with
aciduria type 1. There is no apparent correlation insulin), intravenous lipids, intravascular volume ex-
between genotype and phenotype. There is also a pansion, and correction of acidemia and hypo-
marked clinical variability among individuals who glycemia are essential. Chronic treatment consists of
are homozygous for the same mutation. This suggests a diet low in protein, in particular low in tryptophan
the existence of unrelated, genetically polymorphic and lysine, supplemented with carnitine. Carnitine is
protective mechanisms. Clinical variability may also used to stimulate the formation and excretion of glu-
be related to the flexibility of associated enzyme sys- tarylcarnitine. Riboflavin is a coenzyme for glutaryl-
tems such as those involved in g-aminobutyric acid CoA dehydrogenase, but cofactor responsiveness is
(GABA) or glutamate metabolism. Additionally, envi- rare. A trial of riboflavin can be recommended, but
ronmental factors may be important in determining the drug should be stopped if no beneficial biochem-
the severity of the phenotype. ical effect is found. After the onset of neurological
The cause of striatal dysfunction and necrosis in problems this treatment protocol leads to no or only
glutaric aciduria has only been partially elucidated. A modest improvement, but in most cases further dete-
number of factors may contribute to the damage. Ex- rioration is halted and subsequent episodes of acute
citotoxicity of accumulating substances may be an encephalopathy are prevented. Early treatment has
important factor and involve activation of N-methyl- been shown to be effective in preventing the occur-
D-aspartate (NMDA) receptors, of which glutamate is rence of most or all problems, and improvement or
the normal activator.Both glutaric acid and 3-hydroxy- resolution of neuroimaging abnormalities can be ob-
glutaric acid exhibit structural similarities to gluta- served. Some treated patients, however, still show
296 Chapter 37 Glutaric Aciduria Type 1

some deterioration, and unfortunately even a serious to cerebral atrophy, sometimes to communicating
encephalopathic episode may occur. hydrocephalus. Considering the presence of absolute
There are some additional therapeutic options. In or relative macrocephaly, mild enlargement of the
view of the decreased GABA levels, treatment with a ventricular system, and absence of significant cortical
GABA analogue, baclofen, is logical and has produced damage at autopsy, communicating hydrocephalus
some symptomatic improvement with a decrease in may be the most probable explanation. Both in the
dystonia. Gamma-vinyl GABA (vigabatrin) causes an case of subdural hygroma and in the case of enlarged
irreversible inhibition of GABA transaminase, the subarachnoid spaces, these spaces are crossed by
first step of GABA catabolism, and in this way leads to bridging veins. As a consequence, patients with glu-
increased GABA levels. Decreased dystonia has been taric aciduria type 1 are prone to developing subdur-
noted with vigabatrin therapy. Valproate has been al hemorrhages after minor head trauma. Especially
recommended because of its inhibitory action on in the presence of retinal hemorrhages, a misdiagno-
GABA transaminase. However, valproate is partly ex- sis of nonaccidental head injury and child abuse is
creted as valproylcarnitine and may cause further readily made. In many patients with glutaric aciduria
stress to the already compromised carnitine system. type 1, the sylvian fissure is wide open, forming a
Whether subdural fluid collections should be evac- large CSF space anterior to the temporal lobes. Some
uated, and whether shunt implantation should be per- authors ascribe these CSF collections to the presence
formed in cases of ventricular enlargement and pro- of bitemporal arachnoid cysts, but this is rarely in
gressive macrocephaly, is highly questionable. Reso- conformity with the configuration of the CSF collec-
lution of these abnormalities, spontaneously or with tions. Usually they are ascribed to either frontal and
metabolic treatment, has been documented. If meta- temporal atrophy or failure of opercularization. The
bolic treatment is not started, deterioration may con- aspect of the incompletely formed frontal and tempo-
tinue despite neurosurgical intervention. A prompt ral operculum is most consistent with a failure of
diagnosis of glutaric aciduria type 1 is extremely opercularization. However, in a few patients a normal
important in this context. The stress of the subdural sylvian fissure has been noted soon after birth,
hygroma or hematoma itself and the stress of the
surgery, combined with a catabolic state in a very ill
child, may provoke serious neurological injury. 䊳

Fig. 37.2. A 3-year-old girl with glutaric aciduria type 1, who


presented with an acute encephalopathic episode at the age
37.5 Magnetic Resonance Imaging of 1 year. Note the mildly enlarged subarachnoid spaces, the
open sylvian fissure, enlarged spaces anterior to the temporal
CT scan of the brain discloses enlarged CSF spaces in lobe, and mildly dilated lateral ventricles. There are extensive
most untreated patients with glutaric aciduria type 1, mild signal changes in the periventricular white matter,
even if asymptomatic. The most often noted are fluid sparing the corpus callosum and U fibers. Bilateral lesions are
collections over the convexities in the frontoparietal present in the putamen,caudate nucleus,globus pallidus,den-
and frontotemporal areas. In some cases the fron- tate nucleus, and pontine central tegmental tracts. The puta-
toparietal fluid collections have the appearance of men and caudate nucleus are atrophic. Courtesy of Dr. Z. Patay,
subdural hygromas, usually bilateral, occasionally Department of Radiology, and Dr. P.T. Ozand, Department of
unilateral. In most cases the fluid collections consist Pediatrics, King Faisal Specialist Hospital and Research Center,
of enlarged subarachnoid spaces, frequently ascribed Riyadh, Saudi Arabia

Fig. 37.1. Boy, 10 months old, with


untreated glutaric aciduria type 1.
The images show extensive bilateral
subdural hygromas and frontotempo-
ral hypoplasia with an open sylvian
fissure. Note that the arachnoid and
subdural spaces can be easily distin-
guished. Myelination is seriously
delayed.The basal ganglia have a
slightly abnormal signal intensity.
From Osaka et al. (1993), with
permission
37.5 Magnetic Resonance Imaging 297

Fig. 37.2.
298 Chapter 37 Glutaric Aciduria Type 1

Fig. 37.3. A 3-year-old girl with glutaric aciduria type 1, who have an abnormal signal and are atrophic. There are signal
was diagnosed at the age of 15 months and has been non- changes in the central tegmental tracts of the pons. Courtesy
compliant with treatment since. There is an open sylvian fis- of Dr. Z. Patay, Department of Radiology, and Dr. P.T. Ozand,
sure and free space anterior to the temporal lobe. There are Department of Pediatrics, King Faisal Specialist Hospital and
small spots of abnormal signal within the deep cerebral white Research Center, Riyadh, Saudi Arabia
matter. The putamen, globus pallidus, and caudate nucleus

whereas wide open sylvian fissures are seen several Bilateral lesions are often seen in central gray mat-
months later, forming an argument for atrophy. De- ter structures, variably including the putamen, globus
crease of enlarged CSF spaces has been noted to occur pallidus, head of the caudate nucleus, dentate nucleus,
spontaneously and with treatment. In some patients and substantia nigra (Figs. 37.1–37.4). The lesions are
hemispheric white matter hypodensity has been swollen in the acute stage and subsequently become
found on CT. atrophic. Myelination may be delayed (Fig. 37.1). In
MRI may depict transient subependymal cysts in some of the patients more prominent white matter
young and often still presymptomatic infants. MRI abnormalities are seen, most often in combination
also depicts the fluid collections (Figs. 37.1–37.3). In with striatal lesions or enlarged fluid collections as
most cases, the aspect of the wide open sylvian fis- described above (Figs. 37.2 and 37.4). The white mat-
sures is not consistent with arachnoid cysts but more ter abnormalities are extensive, symmetrical, and
probably with hypoplasia of the frontal and temporal most marked in the frontal and occipital periventric-
operculum (Figs. 37.1–37.3). The enlarged peripheral ular white matter and in the centrum semiovale. The
fluid spaces are the result of enlarged subarachnoid arcuate fibers and corpus callosum are usually spared
spaces, subdural spaces, or a combination of the two (Figs. 37.2 and 37.4). In other patients, more limited
(Figs. 37.1–37.3). Subdural hematomas may also be white matter abnormalities are seen (Fig. 37.3).
seen. In some patients the brain has a highly atrophic On the whole, the imaging findings are highly vari-
appearance with greatly enlarged extracerebral fluid able. In some patients no abnormalities are noted at
spaces (Fig. 37.1). all, whereas in others only or mainly enlarged CSF
37.5 Magnetic Resonance Imaging 299

Fig. 37.4. A 14-month-old boy with glutaric aciduria type 1. (third row). There are bilateral lesions in the putamen, globus
He was diagnosed at 8 months and responded well to treat- pallidus, dentate nucleus, and pontine central tegmental
ment.The cerebral white matter is abnormal, partly because of tracts. Courtesy of Dr. Z. Patay, Department of Radiology, and
delayed myelination, partly because of white matter damage. Dr. P.T. Ozand, Department of Pediatrics, King Faisal Specialist
It is easier to distinguish the two using T1-weighted images Hospital and Research Center, Riyadh, Saudi Arabia

spaces are seen; other cases show a predominance of ment. If already present, they tend to improve with
striatal lesions, while others have a combination of treatment, depending partly on the reversibility of the
striatal and white matter abnormalities. Most cere- abnormalities found.
bral abnormalities can be prevented by early treat-
Chapter 38

Propionic Acidemia

38.1 Clinical Features lower than that of the neonatal onset form, and the
and Laboratory Investigations prognosis with respect to intelligence is better.
During episodes of acute metabolic decompensa-
Propionic acidemia, formerly also called ketotic hy- tion, laboratory investigations reveal signs of dehy-
perglycinemia, is a disorder of organic acid metabo- dration, metabolic acidosis with ketonuria, elevated
lism with autosomal recessive inheritance. Two forms ammonia levels, and decreased carnitine levels. Lac-
of the disease can be distinguished: the severe neona- tate may be elevated and hypoglycemia may be pre-
tal onset form and the late onset form. sent. Neutropenia and thrombopenia are consistent
Many patients with propionic acidemia present in findings. Pancytopenia is rare. Specific laboratory
the neonatal period with a progressive encephalopa- findings suggesting the diagnosis are elevated plasma
thy. They are normal at birth, but after a few hours or and urine levels of propionate, glycine, hydroxypro-
days deterioration sets in with lethargy, poor feeding, pionate, propionylglycine, and methylcitrate. CSF
vomiting, tachypnea, and unexplained coma. Abnor- glycine is not elevated, as is the case in nonketotic hy-
malities in tone and movement occur. Limb hyperto- perglycinemia. The diagnosis is confirmed by assess-
nia and opisthotonic posturing or axial hypotonia ment of the enzyme propionyl-CoA carboxylase in
are often present. Abnormal pedaling movements, leukocytes or fibroblasts. Prenatal diagnosis can be
myoclonic jerks, tremors, and seizures may be pre- performed by enzyme assessment in cultured amnio-
sent. Intracranial hemorrhage may occur as a result of cytes and chorionic villi or by DNA techniques.
serious thrombocytopenia. Hepatomegaly may be
found on physical examination. In the more advanced
stage, there are signs of respiratory distress and 38.2 Pathology
bradycardia. Mortality is high, and survivors fre-
quently have severe lasting neurological impairment, Few data exist relating to histopathological findings
in particular characterized by mental retardation and in propionic acidemia. In the neonatal form, myelin
extrapyramidal movements. The disease course is splitting and myelin vacuolation have been reported,
most commonly characterized by repeated relapses. affecting the structures that contain myelin in a
In the late-onset form, the patients are older when term neonate: posterior limb of the internal capsule,
they present with an acute encephalopathy. Most pa- globus pallidus, thalamus, myelinated brain stem
tients are older than 1 year at presentation, and some tracts, cerebellar white matter, and spinal cord. The
may even present in adulthood. The acute metabolic progress of myelination is normal for a neonate. The
decompensations are precipitated by enhanced pro- cortical cytoarchitecture is normal.
tein catabolism of exogenous origin (high protein in- In older children, postmortem abnormalities
take) or endogenous origin (catabolic states, such as mainly concern the basal ganglia. Atrophy, nerve cell
in fasting, infections, or other forms of stress), but loss, and gliosis are often found in the caudate nucle-
sometimes no cause is found. The encephalopathic us and putamen, to a lesser extent in the globus pal-
episodes are characterized by ataxia, lethargy, and co- lidus and thalamus. Hypermyelination may occur in
ma, sometimes with focal neurological abnormalities the affected basal ganglia. The white matter is often
like hemiplegia. Often the children already had normal. However, atrophy and spongiosis of the cere-
chronic problems before the first episode, including bral white matter may also be found.
chronic feeding problems, persistent vomiting, failure Examination of the liver may reveal fatty degener-
to thrive, hypotonia, osteoporosis, and developmental ation.
retardation. Some patients with the later-onset form
of propionic acidemia are mildly retarded, but some
patients have normal intelligence. Some patients have 38.3 Pathogenetic Considerations
a lasting extrapyramidal movement disorder, usually
chorea. These chronic problems can be present for a The basic defect in isolated propionic acidemia is a
long time without acute metabolic decompensation deficient activity of the mitochondrial enzyme propi-
and without correct diagnosis. The mortality rate of onyl-CoA carboxylase. This enzyme has biotin as its
the encephalopathic episodes with delayed onset is cofactor and consists of two nonidentical subunits (a
38.4 Therapy 301

Fig. 38.1. A 2-month-old boy with


early presentation of propionic
academia.The cerebral white matter is
slightly higher in signal intensity than
normal for a neonate. Courtesy of
Dr. Z. Patay, Department of Radiology,
and Dr. P.T. Ozand, Department of
Pediatrics, King Faisal Specialist
Hospital and Research Center, Riyadh,
Saudi Arabia

and b). Propionyl-CoA carboxylase deficiency can be 38.4 Therapy


caused by a defect in either the a-subunit or the b-
subunit. The gene encoding the a-subunit is called In episodes of acute metabolic decompensation,
PCCA and located on chromosome 13q32. The gene treatment consists of removal of toxic products by
encoding the b-subunits is called PCCB and located dialysis or exchange transfusions. At the same time
on chromosome 3q13.3-q22. Multiple disease-causing further catabolism of proteins should be prevented
mutations have been described in both genes. and an anabolic situation should be achieved to pre-
Propionyl-CoA carboxylase converts propionyl- vent the production of more toxic products. This can
CoA into methylmalonyl-CoA, which is subsequently be achieved by continuous parenteral nutrition with a
converted into succinyl-CoA, which enters the Krebs protein-free product and continuous intravenous in-
cycle. Isoleucine, valine, threonine, and methionine fusion of insulin. Insulin has the capacity to stimulate
are precursors of propionic acid, explaining the ob- protein synthesis. Biotin can be added in an attempt
served protein and amino acid intolerance. Propi- to stimulate residual enzyme activity. Carnitine is ad-
onyl-CoA carboxylase is also involved in the degrada- ministered to stimulate excretion of propionylcarni-
tion of fatty acids with odd-numbered chain lengths tine in urine.
and cholesterol. It is presently not clear why some pa- Long-term treatment consists of dietary protein
tients have a neonatal-onset devastating disease, restriction while maintaining normal development
whereas other patients have a later onset and milder and nutrition status and preventing catabolism.
disease course. Chronic treatment with carnitine should be consid-
The myelinopathy with splitting and vacuole ered. Chronic use of metronidazole can be prescribed
formation is seen in a number of inborn errors of to reduce propionic acid production by bacteria in the
metabolism, including Canavan disease, maple syrup gut. Despite adequate treatment, the outcome of pa-
urine disease, and nonketotic hyperglycinemia, and a tients with the severe variant of propionic academia is
number of intoxications, including hexachlorophene, poor. Most of these patients have remaining morbid-
cuprizone, and triethyltin intoxication. The myelino- ity and disability and recurrent metabolic decompen-
pathy is probably a toxic phenomenon. sations may still occur. In these severe forms of propi-
302 Chapter 38 Propionic Acidemia

Fig. 38.2. This girl with propionic academia was diagnosed at gra, and dentate nucleus. The cerebral cortex has a slightly el-
the age of 3 weeks. She has had repeated crises and intensive evated signal intensity, while the cerebellar cortex and subcor-
care admissions since. The present MRI was obtained at the tical white matter have a more prominently elevated signal.
age of 2 years,when she was admitted in coma to the intensive Courtesy of Dr. Z. Patay, Department of Radiology, and Dr. P.T.
care unit. The T2-weighted images show signal abnormality Ozand, Department of Pediatrics, King Faisal Specialist Hospi-
and swelling of the caudate nucleus, putamen, substantia ni- tal and Research Center, Riyadh, Saudi Arabia

onic acidemia, liver transplantation can be consid- In older patients with propionic academia and
ered. After the procedure, a diet with normal or only acute decompensation, MRI may reveal signal abnor-
mildly restricted protein content can be resumed. malities and swelling of the basal ganglia, substantia
nigra, and dentate nucleus (Fig. 38.2). The cerebral
and cerebellar cortices and subcortical white matter
38.5 Magnetic Resonance Imaging may also display abnormal signal and mild swelling
(Fig. 38.2 and 38.3). In the chronic phase, MRI find-
In patients with neonatal presentation of propionic ings include cerebral atrophy, variably delayed and
academia and serious encephalopathy, an MRI pic- disturbed myelination, and abnormalities in the cau-
ture similar to that of maple syrup urine disease, with date nuclei, globus pallidus, and putamen (Fig. 38.4).
signal abnormality and swelling of the myelin-con- The basal ganglia may display signal changes and at-
taining structures (dorsal part of the pons, midbrain, rophy. However, none of these abnormalities is oblig-
cerebellum, posterior limb of the internal capsule, atory in patients with propionic academia. In one pa-
globus pallidus, thalamus, and central part of the tient with hemiplegia during acute metabolic decom-
corona radiata) would be expected. These have, how- pensation, MRI did not reveal a corresponding con-
ever, not been reported. Diffuse slight cerebral white tralateral cerebral lesion, apart from slight local
matter signal abnormality has been observed cortical atrophy.
(Fig. 38.1).
38.5 Magnetic Resonance Imaging 303

Fig. 38.3. A 6-year-old girl with propionic academia present- abnormalities are present in the caudate nucleus, putamen,
ed with an acute metabolic decompensation. Note the signal and dentate nucleus. Courtesy of Dr. Z. Patay, Department of
abnormalities in the cerebral and cerebellar cortex and the Radiology, King Faisal Specialist Hospital and Research Center,
subcortical white matter on the T2-weighted images. Signal Riyadh, Saudi Arabia

With treatment, improvement may occur. It is


striking that what looks like irreversible cerebral
atrophy (Fig. 38.4) improves with treatment with an
increase in volume of the cerebral mantle (Fig. 38.5).
304 Chapter 38 Propionic Acidemia

Fig. 38.4. A 10-month-old boy with untreated propionic of the lateral ventricles and subarachnoid spaces.The caudate
acidemia, who had just been diagnosed at the time. The T2- nucleus, putamen, and globus pallidus have an abnormal sig-
weighted images show delayed and irregular myelination. nal and are atrophic.The dentate nucleus also seems to have a
There is evidence of prominent cerebral atrophy with dilation slightly abnormal signal
38.5 Magnetic Resonance Imaging 305

Fig. 38.5. Follow-up of the boy shown in Fig. 38.3. Dietary area. There is a marked increase in cerebral volume as com-
treatment and medication were started soon after the age of pared to the previous MRI. The basal ganglia lesions are still
10 months and this MRI was obtained at 3 years. Myelination is visible. The dentate nucleus also still has a slightly abnormal
now much more advanced, but still incomplete in the periven- signal
tricular occipital area, the temporal lobes, and the subcortical
Chapter 39

Nonketotic Hyperglycinemia

39.1 Clinical Features glycine is essential for the diagnosis. Patients with
and Laboratory Investigations transient NKH initially have elevated CSF glycine and
an elevated CSF:plasma glycine ratio, but in the
Nonketotic hyperglycinemia (NKH) is an autosomal course of 2–8 weeks plasma and CSF glycine levels
recessive disorder of which different types are distin- return to normal.
guished: neonatal, infantile, mild-episodic, late-onset, NKH is caused by a defect in the glycine cleavage
and transient. system. This system is specifically expressed in liver,
Most NKH patients have the neonatal variant. They kidney, brain, and lymphoblasts, but not in fibroblasts
are normal at birth. Within a few days there are pro- and leukocytes. Liver biopsy used to be necessary
gressive neurological abnormalities, including lethar- for enzymatic diagnosis of NKH. Since it was shown
gy, hypotonia, myoclonic jerks, seizures, coma, and that the glycine cleavage system can be tested in
apneic spells. Most infants require assisted ventila- lymphoblasts, enzymatic diagnosis of NKH has been
tion. A few patients have been described with an in- performed using peripheral blood. However, the
creasing head circumference to above the 95th per- normal glycine cleavage activity in normal lym-
centile and hydrocephalus on neuroimaging, requir- phoblasts is low, and the reliability of this method
ing shunting. Many patients with neonatal NKH die has been questioned. Patients with neonatal NKH
within a few weeks. The patients who survive show have extremely low to undetectable glycine cleavage
signs of spasticity, opisthotonus, absence of any de- system activity, whereas in patients with atypical
velopment, severe seizures, and microcephaly. Many NKH some residual activity is found. Prenatal diag-
children who survive the neonatal period die within nosis using cultured amniotic cells is not possible,
the first year of life, but some survive into childhood because the glycine cleavage system is not manifest in
or beyond in a severely disabled condition. these cells. Prenatal diagnosis using chorionic villi
Patients with atypical NKH do not present in the tissue is feasible, although false negative and false
neonatal period. In the infantile variant, seizures have positive prenatal diagnoses have been reported. DNA
a later onset, in the first year of life, and the patients diagnosis is preferable when the mutation of the fam-
tend to survive. Mental retardation is present but is ily is known.
not as severe as in the neonatal variant of NKH.
Patients with mild-episodic NKH have mild mental
retardation and episodes of agitation, ataxia, chorea, 39.2 Pathology
and vertical supranuclear ophthalmoplegia provoked
by febrile illness. Patients with late-onset NKH have The external configuration of the brain in patients
variable neurological features, including seizures, de- with neonatal NKH is usually normal with a normal
velopmental delay, developmental regression, spastic- gyral pattern. The quantity of myelin found by micro-
ity, ataxia, and optic atrophy. Patients with transient scopic examination of the brain is normal, but all
NKH are initially indistinguishable from patients myelinated areas have a strikingly spongy appearance
with neonatal NKH. However, in the course of several imparted by the presence of numerous vacuoles. In
weeks the patients recover, most often without neuro- conformity with the neonatal state of myelination, the
logical sequelae, although some patients have severe spinal cord, brain stem (in particular brain stem
remaining neurological deficits. tegmentum), cerebellar white matter, posterior limb
In neonatal and transient NKH, a characteristic of the internal capsule, and optic nerves, tracts, and
EEG pattern is seen during the first weeks of life with chiasm are prominently involved, whereas cerebral
short bursts of high complex waves alternated with hemispheric white matter devoid of myelin is free of
hypoactivity. In neonatal NKH, this burst–suppres- vacuoles. There is usually a correlation between
sion pattern changes into hypsarrhythmia in the myelin density and density of vacuoles. In electron
course of a few weeks. microscopy, the vacuoles are found to be located
Laboratory abnormalities include elevated levels within myelin sheaths and formed by splitting of
of glycine in plasma and urine in the absence of myelin lamellae along the intraperiod lines. There are
ketoacidosis. The CSF level of glycine is elevated, as is no sudanophilic deposits and there is a paucity of
the ratio of CSF to plasma glycine. Elevation of CSF phagocytic and astroglial reactions. In children
39.3 Pathogenetic Considerations 307

beyond the neonatal period variable delay in myelina- also found in family members who never have symp-
tion has been reported. toms of NKH. Apparently carriership for NKH con-
In some patients with neonatal NKH, pachygyria tributes to the development of transient NKH, but an
and agenesis of the corpus callosum have been re- additional unknown factor or factors contribute to
ported at CT. These findings have, so far as we know, the development of symptoms. Immaturity of the
not been confirmed by more detailed MRI or autopsy glycine cleavage system in the neonatal period may be
findings. a factor. Deficiency of the L protein presents with the
Additional neuropathological changes can be symptoms of pyruvate dehydrogenase complex defi-
found related to the serious condition of the child and ciency without hyperglycinemia.
occurrence of complications: neuronal incrustation In NKH, there is an elevated concentration of
with calcium in the cortex and thalamus, more acute glycine in the brain and CSF. The glycine is held re-
anoxic neuronal changes in the cerebral cortex, and sponsible for the neurological impairment. Glycine
ischemic and/or hemorrhagic white matter changes is a major inhibitory neurotransmitter in the brain
in the periventricular area. stem and spinal cord, acting on strychnine-sensitive
Neuropathological findings in patients with a glycine receptors. The effects of high levels of glycine
neonatal presentation of NKH and survival beyond concentrations here are probably responsible for the
the first year of life include a variable diffuse decrease hypotonia, lowered consciousness, and apnea present
in white matter mass with some ventricular enlarge- in neonates. Glycine is also active in the cerebral cor-
ment and a thin corpus callosum. The white matter of tex and has excitatory properties here. It is a positive
the CNS shows diffuse vacuolation, contrasting with modulator of the N-methyl-D-aspartate (NMDA) re-
the more restricted vacuolation in neonates, in whom ceptor of glutamate, which plays an important role in
unmyelinated white matter is not vacuolated. The glutamate-induced neurotoxicity. NMDA receptors
amount of myelin may be normal or reduced without are located throughout the brain. Glycine increases
evidence of a progressive myelin disease or myelin the frequency of NMDA receptor channel opening by
loss. Cortical architecture is normal. The cerebellum accelerating recovery of the receptor following gluta-
may be of normal size or diffusely atrophic with a se- mate-induced desensitization. In this way, glycine
vere loss of Purkinje cells and granule cells. potentiates the excitotoxic action of glutamate and
may lead to intractable seizures. In animal experi-
ments it has been shown that glycine administration
39.3 Pathogenetic Considerations enhances NMDA-induced seizures in mice whose
classic glycine receptors located in spinal cord and
The basic defect in NKH involves the glycine cleavage brain stem had been blocked with strychnine. This
system. The glycine cleavage system is a mitochon- experiment indicates that glycine is a harmful en-
drial multienzyme complex that is composed of four hancer of the NMDA response. It has been shown that
proteins: P protein (a pyridoxal phosphate-depen- the developing brain has heightened susceptibility to
dent glycine decarboxylase), T protein (a tetrahydro- NMDA-mediated injury, and high levels of glycine
folate requiring aminomethyltransferase), H protein may be particularly devastating to the CNS of the
(a lipoic acid containing hydrogen carrier protein), neonate. This is in accordance with the clinical obser-
and L protein (dihydrolipoamide dehydrogenase). vation that most acute neurological problems are pre-
Three components, the P, T, and H proteins, are specif- sent in the neonatal period, and that stabilization is
ic for the glycine cleavage system. L protein is a house- seen in patients who survive this period, albeit with a
keeping enzyme, that is also used as the E3 component severe neurological handicap.
of a-keto acid dehydrogenase complexes such as the The relationships between high glycine levels and
pyruvate dehydrogenase complex. P, T, H, and L pro- myelin vacuolation and between myelin vacuolation
teins are encoded by the genes GLDC on chromosome and clinical symptomatology are unclear. A myelin
9p23–24, AMT on chromosome 3q21.1–21.2, GCSH disorder does not, as a rule, lead to severe epilepsy,
on chromosome 16q24, and GCSL on chromosome but rather to spasticity and other forms of loss of
7q31–32, respectively. The glycine cleavage system neurological function. The myelin disorder may ex-
catalyzes the conversion of glycine and tetrahydrofo- plain at least part of the clinical impairment in the
late to CO2, NH3, and methylenetetrahydrofolate. chronic stage of the disease. Myelin vacuolation is
The glycine cleavage activity is undetectable or seen in inborn errors of metabolism (Canavan dis-
extremely low in the neonatal type, whereas in the ease, maple syrup urine disease) and in intoxications
later-onset types there is still some residual activity. (hexachlorophene, triethyltin, cuprizone). The patho-
The majority of NKH patients have a defect in GLDC genetic mechanisms of myelin vacuolation are un-
and the remainder have a defect in AMT. In transient clear in all these conditions, but are probably toxic in
neonatal NKH heterozygous GLDC and GCSH muta- nature.
tions have been found. However, the same mutation is
308 Chapter 39 Nonketotic Hyperglycinemia

39.4 Therapy ty and epilepsy and some improvement of sucking


and gross motor movements.
Many therapeutic approaches have been used in the So far a combination of the two strategies, using
attempt to lower glycine concentrations in patients sodium benzoate and dextromethorphan, has had
with the neonatal-onset form of NHK. Dietary re- variable beneficial effect. Sodium benzoate is conju-
striction of glycine and serine or of all protein, gated with glycine to form hippurate, which can be
administration of drugs to stimulate excretion of effectively excreted in the urine, in this way removing
glycine in urine or bile, peritoneal dialysis, hemodial- excess glycine.A lowering to normalization of plasma
ysis, and exchange transfusions have all failed to glycine level and a lowering of CSF level can be
achieve much. achieved with sodium benzoate, accompanied by
Another strategy is aimed at blocking the effects of increased alertness of the patient and a reduction of
glycine on NMDA receptors. Strychnine blocks seizures. The effectiveness of sodium benzoate seems
glycinergic inhibitory effects, but strychnine is only to be higher in patients with atypical variants of NKH
effective on the classic glycine receptors present in the with only moderately elevated glycine levels than in
brain stem and spinal cord. Strychnine treatment is patients with neonatal NKH. Dextromethorphan, an
clinically ineffective. Diazepam is a competitor for NMDA receptor antagonist, has been used with vari-
glycine receptors. The drug has a favorable anticon- able success to reduce or stop seizures, improve alert-
vulsant effect. Treatment using NMDA receptor an- ness and social interest, and initiate some motor de-
tagonists has also been attempted. Ketamine and velopment. During temporary cessation of treatment
tryptophan, both NMDA receptor antagonists, have and during febrile infections, temporary neurological
been reported to lead to some reduction in irritabili- deterioration can occur. Imipramine, another NMDA

Fig. 39.1. Male neonate with NKH, born at term and now has a high signal throughout on these T2-weighted images.
4 days old. The brain looks diffusely abnormal and somewhat The brain stem also has an abnormal signal. The corpus callo-
swollen. In large areas the contrast between cortex and white sum is present
matter is decreased.The posterior limb of the internal capsule
39.5 Magnetic Resonance Imaging 309

receptor antagonist, has also been reported to have less prominent (Fig. 39.1). We observed diffuse brain
beneficial effects. Despite treatment, the neurological edema and swelling in a comatose neonate with NKH
handicap of the treated children remains serious and (Fig. 39.1).
there is only minor psychomotor development. On follow-up progressive cerebral and cerebellar
atrophy is observed with ventricular enlargement, en-
largement of the subarachnoid spaces, and thinning
39.5 Magnetic Resonance Imaging of the corpus callosum (Figs. 39.2 and 39.3). Myelina-
tion is seriously delayed (Figs. 39.2 and 39.3).
Limited imaging data are available in NKH. In the Long echo time proton MRS of the brain offers a
neonatal phase, MRI may show abnormalities in the noninvasive method for direct measurement of brain
signal intensity in the dorsal pons and midbrain and levels of glycine, if elevated, and can be used for diag-
posterior limb of the internal capsule. These abnor- nostic purposes and to monitor treatment. Glycine
malities are probably related to myelin vacuolation in co-resonates with myo-inositol at 3.56 ppm. For this
myelinated areas. Diffusion-weighted MRI shows a reason it is impossible to quantify glycine at short
high signal in these areas and the cerebral peduncles, echo times. However, glycine has a much longer T2
whereas ADC values in these areas are low, indicative than myo-inositol and is also visible at long echo
of restricted diffusion. The findings are reminiscent times (135–270 ms), when myo-inositol is no longer
of those in maple syrup urine disease, but tend to be seen.

Fig. 39.2. Boy, 10 months old, with


NKH.The corpus callosum is extremely
thin and hardly visible on the sagittal
images, but the presence of the cingu-
late gyrus indicates that it has devel-
oped normally.The transverse images
confirm that there is a very thin corpus
callosum.The white matter contains
very little myelin.The volume of the
cerebral white matter is reduced and
the lateral ventricles are dilated.There
is a lesion in the basal ganglia on the
left. Courtesy of Dr. Z. Patay, Depart-
ment of Radiology, and Dr. P.T. Ozand,
Department of Pediatrics, King Faisal
Specialist Hospital and Research
Center, Riyadh, Saudi Arabia
310 Chapter 39 Nonketotic Hyperglycinemia

Fig. 39.3. Patient, 13 months old, with NKH.The sagittal image cerebral white matter is mildly reduced and the lateral ventri-
demonstrates that the corpus callosum is extremely thin but cles are dilated. Courtesy of Dr. Z. Patay, Department of Radiol-
complete.The T1- and T2-weighted images show that the cere- ogy, and Dr. P.T. Ozand, Department of Pediatrics, King Faisal
bral white matter contains very little myelin.The volume of the Specialist Hospital and Research Center, Riyadh, Saudi Arabia
Chapter 40

Maple Syrup Urine Disease

40.1 Clinical Features tions. Clinical signs may be first seen between the
and Laboratory Investigations ages of 2 months and 40 years, triggered by infection,
vaccination, operation, or sudden increase in dietary
Maple syrup urine disease (MSUD) is a hetero- protein. The episodic deterioration is characterized
geneous disorder. Classification is based on clinical by maple syrup odor, cerebellar ataxia, irritability,
presentation and outcome. Five phenotypes can be and progressive lethargy. With supportive care the
distinguished: classical, intermediate, intermittent, patient recovers, but will experience repeated similar
thiamine-responsive, and dihydrolipoyl dehydroge- episodes until the correct diagnosis is established and
nase (E3)-deficient forms of MSUD. All forms have an specific dietary treatment started.
autosomal recessive mode of inheritance. Thiamine-responsive MSUD is not a well-defined
Classical MSUD is the most severe and the most subtype of MSUD. In the reported patients, the clini-
common form of the disease. Infants appear normal cal course tends to be relatively mild, even if untreat-
at birth. By the end of the first week symptoms ed, although thiamine responders may also have the
emerge, with lethargy, poor feeding (but rarely vom- classical, severe form of the disease. In general, these
iting), alternating periods of hypertonia and hypoto- patients do not have acute neonatal illness and their
nia, opisthotonus, convulsions, bulging fontanel, early clinical course is similar to that of intermediate
irregular respiration, and apnea. An odor of maple MSUD. The course of the disease is greatly ameliorat-
syrup is frequently noted, but may not initially be pre- ed by simultaneous thiamine administration and
sent. If the disease is not treated, rapidly progressive dietary treatment. Outcome is favorable.
neurological deterioration occurs, with cerebral ede- E3-deficient patients have a relatively uneventful
ma, coma, and death usually within the first month of first few months of life. Patients develop persistent
life. If an untreated patient survives the first few lactic acidosis between 2 and 6 months of life and a
weeks of life, signs of severe brain damage remain, progressive neurological deterioration sets in with
with profound psychomotor retardation, spasticity, hypotonia, developmental delay, and extrapyramidal
generalized dystonia, and cerebral blindness. Early movement abnormalities.
diagnosis and treatment may avert or reverse the Laboratory investigations reveal ketoacidosis in
neurological abnormalities, but mental and neurolog- episodes of metabolic decompensation. Concentra-
ical residua are common in treated patients. It has tions of branched-chain amino acids (leucine, valine,
been shown that the length of time after birth for and isoleucine) and related keto acids (a-ketoiso-
which the metabolic derangement is not adequately caproic acid, a-ketoisovaleric acid, and a-keto-b-
treated, and the quality of long-term metabolic con- methylvaleric acid) are elevated in blood, urine, and
trol both have important influences on eventual intel- CSF. Smaller amounts of the respective 2-hydroxy
lectual capacities. If the disease is identified and treat- acids are formed by reduction of the keto acids. An
ed within a few days after birth, IQ scores are higher unusual isomer of isoleucine, alloisoleucine, is also
and may be normal. A problem is that throughout life found. Diagnosis is confirmed by demonstration of a
intercurrent illnesses, even minor illnesses, may lead deficiency of branched-chain keto acid dehydroge-
to severe metabolic derangement, cerebral edema, nase in leukocytes or cultured fibroblasts. DNA con-
and possibly death. firmation is an option.
The intermediate variant of MSUD is milder and In the early stages of untreated classical MSUD,
patients do not have catastrophic illness in the neona- EEG shows characteristic abnormalities, variously
tal period. Many patients do not have episodes of called a “comb-like” or “picket fence” rhythm or “cen-
acute metabolic decompensation. Progressive mental tral theta spindle.” The pattern consists of bursts and
retardation is the major clinical feature, usually be- runs of 5–7 Hz, primarily monophasic, negative,
coming apparent during the first year of life. General- mu-like activity in the central and central–parasagit-
ized hypotonia and an odor of maple syrup are pre- tal regions during wakefulness and sleep with the
sent. most abundant bursts occurring during quiet, non-
Patients with the intermittent form of MSUD show REM sleep. The background pattern shows diffuse
normal early development. They are at risk for acute slowing, loss of reactivity to auditory stimuli, burst-
metabolic decompensation during stressful situa- suppression patterns, and spike and sharp wave pat-
312 Chapter 40 Maple Syrup Urine Disease

terns. The abnormalities disappear on treatment. Mo- tein, are significantly reduced. Cholesterol esters are
tor and sensory peripheral nerve conduction velocity not elevated. Free amino acids in the brain are not
is normal. altered with the exception of the branched-chain
Prenatal diagnosis can be performed by assessing amino acids, which are markedly increased. Gluta-
branched-chain keto acid dehydrogenase in cultured mine, glutamate, and GABA are significantly reduced.
amniocytes or chorionic villus cells. In families with In older, treated patients, the lipid composition of the
known mutations, DNA analysis can replace the brain is normal.
enzyme assay.

40.4 Pathogenetic Considerations


40.2 Pathology
MSUD is caused by a deficiency in activity of the
In young infants who die in the acute stage of the dis- branched-chain a-keto acid dehydrogenase complex.
ease, the brain is enlarged due to generalized edema. This is a mitochondrial multienzyme complex cat-
Brain weight is increased. Gyri may be broadened and alyzing the oxidative decarboxylation of branched-
flattened. Microscopic examination reveals a status chain a-keto acids, which are derived branched-chain
spongiosus of myelinated areas. Unmyelinated re- amino acids such as valine, leucine, and isoleucine by
gions are not affected. Thus, in neonates the areas transamination. The multienzyme complex consists
involved are the spinal cord, medulla oblongata, of three catalytic components: branched-chain a-ke-
dorsal part of pons, midbrain, cerebellar white mat- to acid decarboxylase (E1), dihydrolipoyl acyltrans-
ter, cerebellar peduncles, posterior limb of the inter- ferase (E2), and dihydrolipoyl dehydrogenase (E3).
nal capsule, and central part of the corona radiata. E1 is a heterotetramer composed of two a and two b
Sponginess may also be present in the basal ganglia, subunits. The complex also contains two specific reg-
in particular in the globus pallidus due to its density ulatory enzymes, a kinase and a phosphatase, com-
of myelinated fibers. The sponginess is caused by pounds that are responsible for regulating the catalyt-
myelin splitting at the intraperiod line and in- ic activity through phosphorylation and dephospho-
tramyelinic vacuole formation. No signs of active rylation. E1 is phosphorylated at two serine residues
myelin breakdown are seen and no sudanophilic and hence responsible for regulation of the catalytic
breakdown products. In the spongy white matter activity of the complex. Phosphorylation inactivates
marked astrocytic gliosis is present. the complex and dephosphorylation activates it.
In older, untreated infants neuropathological find- E1 binds thiamine diphosphate to create the active
ings may also be characterized by edema if the patient site for decarboxylation of the branched-chain
died during an acute metabolic decompensation. a-keto acid substrate. E2 catalyzes transfer of the
Further findings consist of a delay in myelination and acyl group from the lipoyl moiety to coenzyme A to
a status spongiosus and astrogliosis of the myelinated give rise to a branched-chain acyl-CoA. It forms the
white matter. Myelin stains reveal myelin paucity, but structural core of the enzyme complex to which E1, E3,
there is no evidence of active myelin breakdown. kinase and phosphatase are bound. E3 is identical
Oligodendrocytes are decreased in numbers. There to the dehydrogenases associated with pyruvate de-
are no phagocytic cells and no or little deposition of hydrogenase and a-ketoglutarate dehydrogenase
sudanophilic breakdown products. The myelin ab- complexes.
normalities occur in all regions; no areas are spared. Mutation in any of the four genes encoding E1, E2,
Gray matter is essentially normal. In treated infants and E3 may result in MSUD through dysfunction of
and children myelination is normal and white matter the branched-chain a-keto acid dehydrogenase com-
sponginess is minimal. plex. The gene encoding E1a, BCKDHA, is located on
The neuropathology in E3 deficiency resembles chromosome 19 at position q13.1–13.2. The gene en-
that of Leigh syndrome with lesions in the basal gan- coding E1b, BCKDHB, is located on chromosome 6 at
glia, thalami, and brain stem. position p21–22. The gene encoding E2, DBT, is locat-
ed on chromosome 1 at position p21–31. The gene
encoding E3, DLD, is located on chromosome 7 at
40.3 Chemical Pathology position q31–32. Patients with a deficiency of E3
activity have combined deficiency of branched-chain
In neonates the lipid composition of the brain is nor- a-keto acid dehydrogenase, pyruvate dehydrogenase,
mal or near-normal. In older, untreated infants the and a-ketoglutarate dehydrogenase complexes. So
findings of chemical analysis of the brain are in con- far, no clear relationship between genotype and phe-
formity with delayed myelination without active notype has emerged. There is some relationship
myelin breakdown. Major myelin components, in- between clinical phenotype and residual enzyme
cluding sulfatide, cerebroside, and proteolipid pro- activity.
40.6 Magnetic Resonance Imaging 313

Mechanisms for toxic effects of increased decompensation. Catabolic conditions after surgery
branched-chain amino acids and keto acids remain can be prevented by administration of insulin and a
largely unelucidated. The toxic effects may be related branched-chain amino-acid-free parenteral nutrition
to disturbance of neurotransmission, energy deple- regimen.
tion, and direct myelin damage. Branched-chain keto All patients with classical MSUD require life-long
acids and their hydroxy derivatives compete with dietary treatment. The long-term treatment aims at
glutamate for decarboxylation and so reduce GABA maintaining the whole-body content of branched-
production. Excess leucine may reduce cerebral sero- chain amino acids close to the minimum requirement
tonin. a-Ketoisocaproic acid inhibits pyruvate dehy- for normal body function. The amount of branched-
drogenase and a-ketoglutarate dehydrogenase, two chain amino acids necessary for adequate protein
important enzymes in mitochondrial energy pro- synthesis is given in the form of natural proteins in a
duction. The mechanism of myelin vacuolation and protein-restricted diet. A branched-chain amino-
myelin damage is unknown. Hexachlorophene and acid-free mixture of amino acids is used to supple-
triethyltin are toxins which also lead to myelin vacuo- ment the diet. The levels of branched-chain amino
lation. These substances are inhibitors of mitochon- acids in blood are measured regularly to monitor
drial oxidative phosphorylation. From experimental treatment. Deficiency of branch-chain amino acids
studies it is known that lasting effects only follow leads to growth failure, skin rash, exfoliative dermati-
chronic exposure and that early discontinuation of tis, diarrhea, and de-epithelialization of the cornea.
exposure is followed by repair. This course of events is Patients with milder forms of MSUD tolerate a high-
similar to that observed in MSUD: early treatment er protein intake. In thiamine-responsive MSUD, use
leads to resolution of abnormalities. Only in untreat- of thiamine increases protein tolerance. However, a
ed cases are lasting effects seen. The precise patho- protein-restricted diet should still be used and the
physiological mechanisms of myelin splitting are plasma levels of branched-chain amino acids moni-
unknown, either in exogenous intoxications or in tored. In order to test for thiamin-responsiveness, all
MSUD. Evidence has been found that MSUD metabo- patients should receive trial therapy with thiamin.
lites, in particular a-isocaproic acid, induce apoptosis If onset of dietary treatment leads to lowering of
in glial and neuronal cells, but the relationship with blood levels of branched-chain amino acids within a
the in vivo pathology of MSUD is unclear. few days after birth, and if adequate dietary control is
maintained over the years, prognosis is excellent and
intellectual capacities may be normal. Later onset of
40.5 Therapy treatment or poor dietary control contribute to men-
tal deficiency. With adequate dietary treatment and
In cases of acute metabolic decompensation with very careful monitoring, female patients with MSUD may
high blood and tissue levels of branched-chain amino enjoy successful pregnancies.
acids and keto acids, emergency treatment is neces- Liver transplantation has been shown to increase
sary. Exchange transfusions, peritoneal dialysis, he- whole-body branched-chain a-keto acid dehydroge-
modialysis, and hemofiltration are effective in acute- nase activity to at least the level of very mild MSUD
ly ill patients. These therapeutic measures must be variants (Wendel et al. 1999). After liver transplanta-
supplemented by high-energy intake to reverse the tion, patients no longer need protein-restricted diets
catabolic condition caused by infection or fasting. For and the risk of metabolic decompensation during
this purpose intravenous glucose, intravenous lipids, catabolic events is apparently abolished. However, liv-
or a special formula containing a mixture of complete er transplantation comes with considerable risks and
nutrients lacking only branched-chain amino acids the benefits may not be significantly different from
can be used. Concomitant administration of insulin is those of strict dietary treatment.
very effective in achieving an anabolic situation. In
acutely ill but not comatose patients, nutritional ther-
apy may be sufficient. During the course of therapy, 40.6 Magnetic Resonance Imaging
isoleucine and valine should be added to the regimen
to prevent depletion of these essential amino acids In MSUD with neonatal presentation, CT shows a very
and thus ensure effective protein synthesis. characteristic pattern with profound hypodensity
Minor illnesses may lead to catabolic conditions and swelling of the cerebellar hemispheres, dorsal
with release of amino acids from body tissues. Toxic part of the pons, midbrain, posterior limb of the
levels of branched-chain amino acids may be reached internal capsule, the globus pallidus, and often the
within a few hours with onset of as yet mild clinical thalamus. In addition, milder, generalized cerebral
symptoms. Immediate high-energy intake and tem- white matter hypodensity is seen.
porary removal of all natural protein from the diet in MRI confirms the pattern. In MSUD with neonatal
such situations may prevent a full-blown metabolic presentation, T2-weighted images show marked
314 Chapter 40 Maple Syrup Urine Disease

Fig. 40.1.
40.6 Magnetic Resonance Imaging 315

Fig. 40.1. (continued). Baby boy with MSUD. The T2-weighted cerebellar white matter, the medulla, dorsal part of the pons,
images (first three rows) show an elevated signal intensity and pyramidal tracts in the basis of the pons, midbrain, posterior
swelling of the cerebellar white matter, the medulla, dorsal limb of the internal capsule, and central part of the corona
part of the pons, pyramidal tracts in the basis of the pons, mid- radiata.The thalamus,anterior limb of the internal capsule,cor-
brain, the posterior limb of the internal capsule, and central pus callosum, and hippocampus display less severely restrict-
part of the corona radiata.The thalamus, too, has a slightly ab- ed diffusion. Courtesy of Dr. Z. Patay, Department of Radiology,
normal signal. The diffusion-weighted images (trace images, and Dr. P.T. Ozand, Department of Pediatrics, King Faisal Spe-
b = 1000, fourth and fifth rows) show restricted diffusion in the cialist Hospital and Research Center, Riyadh, Saudi Arabia

swelling and high signal intensity in the posterior ed development or signs of metabolic decompensa-
part of the pons, in the midbrain, cerebellar white tion. MRI at that time (Figs. 40.5 and 40.6) shows
matter, posterior limb of the internal capsule, thala- more diffuse abnormality of the white matter of the
mus, globus pallidus, and central part of the corona cerebral hemispheres. The cerebral white matter may
radiata (Fig. 40.1). Besides, mild diffuse cerebral have a striking stripe-like appearance, the stripes be-
white matter edema may be present. Without treat- ing partly related to enlarged perivascular spaces and
ment, the edema gradually decreases and atrophy en- partly to myelination in stripes, probably reflecting
sues. Edema is most pronounced between the third perivascular myelin deposition (Figs. 40.5 and 40.6).
week and the end of the second month. With treat- The brain stem is also involved. Both the thalamus
ment edema resolves more rapidly. and globus pallidus are affected, whereas the puta-
Remaining abnormalities in treated patients are men and caudate nucleus are normal. The cortex is
variable, depending on the severity and duration of normal. Improvement after treatment has been re-
the initial episode of metabolic derangement and the ported. In adequately treated patients with milder
subsequent metabolic control (Figs. 40.2–40.4). In variants of MSUD, CT and MRI may be normal.
some patients MRI becomes normal. In others some Diffusion-weighted imaging shows restricted dif-
signs of atrophy, delay in myelination, and white fusion in the areas of acute myelin vacuolation with
matter abnormalities are found. Many patients have low ADC values (Fig. 40.1). In neonates, this results in
remaining signal abnormalities in the midbrain, a pattern of very high signal intensity in the medulla,
thalamus, and globus pallidus. dorsal part of the pons, the midbrain, cerebellar white
If MRI is performed during a later episode of matter, posterior limb of the internal capsule, thala-
metabolic decompensation in otherwise adequately mus, globus pallidus, and central part of the corona
treated and normally developing children with classi- radiata. The restricted diffusion is probably related to
cal MSUD, it shows brain swelling and abnormally decrease of the extracellular space because of the
increased signal intensity on T2-weighted images in vacuoles within the myelin sheaths. Diffusion tensor
the deep white matter and U fibers, internal capsule, imaging reveals decreased anisotropy in the same
basal ganglia, thalamus, hypothalamus, dentate nu- regions. In the unmyelinated white matter ADC val-
cleus, and brain stem. In deep coma, all white matter ues are increased, reflecting white matter edema.
may be abnormal in signal and seriously swollen. If Proton MRS is helpful in establishing a diagnosis
adequate treatment is initiated immediately, most or in acute metabolic decompensation by revealing res-
all abnormalities disappear. onances related to branched chain amino acids and
Milder variants of MSUD usually come to medical keto acids at 0.9 ppm. Lactate may also be elevated.
attention in the second year of life because of retard- These peaks disappear with treatment.
316 Chapter 40 Maple Syrup Urine Disease

Fig. 40.2. A 4-month-old girl with neonatal-onset MSUD. She as the putamen and caudate nucleus are spared.These abnor-
was diagnosed at 3 weeks and has been receiving treatment malities are very similar to those seen in Canavan disease.
since that time.Diffuse signal abnormalities are still seen in the Courtesy of Dr. Z. Patay, Department of Radiology, and Dr. P.T.
cerebral and cerebellar white matter, and brain stem at all lev- Ozand, Department of Pediatrics, King Faisal Specialist Hospi-
els. There is, however, much less swelling than in the neonatal tal and Research Center, Riyadh, Saudi Arabia
stage.The thalamus and globus pallidus are abnormal, where-

The images of patients with classical MSUD during onset. The images of patients with milder variants of
the neonatal period are diagnostic. All areas which MSUD during the second year of life are very similar
are normally myelinated at that age have an abnormal to those of Canavan disease. However, clinical history
signal intensity and are swollen. This pattern is exclu- and laboratory findings differentiate between the
sively seen in vacuolating myelinopathies of neonatal two.
40.6 Magnetic Resonance Imaging 317

Fig. 40.3. The same girl as in Fig. 40.2, now at the age of 2 ment of Radiology, and Dr. P.T. Ozand, Department of Pedi-
years. Under treatment most abnormalities disappeared. atrics, King Faisal Specialist Hospital and Research Center,
Myelination is mildly delayed. There are slight signal abnor- Riyadh, Saudi Arabia
malities in the globus pallidus. Courtesy of Dr. Z. Patay, Depart-
318 Chapter 40 Maple Syrup Urine Disease

Fig. 40.4. Boy with neonatal presentation of MSUD. He has re- sal pons, and the dentate nucleus. Courtesy of Dr. Z. Patay,
sponded well to treatment and is now 6 years old. The MRI Department of Radiology, King Faisal Specialist Hospital and
shows signal abnormalities in the periventricular white matter, Research Center, Riyadh, Saudi Arabia
globus pallidus, midbrain (especially the dorsal part), the dor-
40.6 Magnetic Resonance Imaging 319

Fig. 40.5. An 18-month-old boy with intermittent MSUD. He there are signal abnormalities in the midbrain, pons, medulla,
experienced metabolic coma at 12 and 18 months. MRI shows and dentate nucleus. He responded well to treatment and had
extensive cerebral white matter abnormalities, in some areas a full clinical recovery. Courtesy of Dr. Z. Patay, Department of
with a stripe-like aspect.The thalamus and globus pallidus are Radiology, King Faisal Specialist Hospital and Research Center,
abnormal, while the putamen and caudate nucleus are spared. Riyadh, Saudi Arabia
The posterior limb of the internal capsule is spared.In addition,

Fig. 40.6. A 4-year-old boy recently diagnosed with MSUD. T1-weighted images (third row) demonstrate mild diffuse hyper-
The T2-weighted images (first and second rows) show extensive intensity of the white matter, consistent with diffuse hypomyeli-
cerebral white matter abnormalities, which have a stripe-like nation. Within the white matter the enlarged perivascular
aspect. The stripes are partly related to enlarged perivascular spaces are beautifully seen. The sagittal (fourth row, left and
spaces and partly to myelin deposition in a stripe-like pattern, middle) and coronal (fourth row,right) images confirm the pres-
probably perivascular myelin deposition. The thalamus and ence of the enlarged perivascular spaces and the stripe-like
globus pallidus are abnormal, while the putamen and caudate pattern of myelin deposition. Courtesy of Dr. I. Verma, Depart-
nucleus are spared.The posterior limb of the internal capsule is ment of Genetic Medicine, Sir Ganga Ram Hospital, New Delhi,
spared. There are signal abnormalities in the midbrain, pons, India. (Fig. 40.6 see next page)
medulla, dentate nucleus, and cerebellar white matter. The
320 Chapter 40 Maple Syrup Urine Disease

Fig. 40.6.
Chapter 41

3-Hydroxy-3-Methylglutaryl-CoA Lyase Deficiency

41.1 Clinical Features 41.2 Pathology


and Laboratory Investigations
Zoghbi et al. (1986) reported the findings of a brain
3-Hydroxy-3-methylglutaryl-coenzyme A lyase (HMG- biopsy during an episode of metabolic decompensa-
CoA lyase) deficiency is a rare inborn error of metab- tion. No abnormalities were found in the cerebral cor-
olism with an autosomal recessive mode of inheri- tex. In the white matter spongiosis, reactive gliosis,
tance. About 50% of the patients present in the and increased intracellular astrocytic glycogen were
neonatal period after a brief symptom-free period. found. Gibson et al. (1994) reported spongiform de-
Most of the rest become symptomatic before the age generative changes and generalized edema in a child
of 1 year. Presentation after the age of 2 years is rare. who died during a metabolic crisis. The histopatho-
Episodes of deterioration are provoked by fasting, in- logical basis of the focal and diffuse white matter sig-
fections, and high protein intake. The episodes are nal abnormalities in well-treated and clinically nor-
characterized by lowering of consciousness, hypoto- mal patients is unknown.
nia, sometimes seizures, and apnea. Many patients
have hepatomegaly. Some patients die from deep hy-
poglycemia. With appropriate treatment, most pa- 41.3 Pathogenetic Considerations
tients recover from their initial episode of metabolic
decompensation, but there may be remaining neuro- HMG-CoA lyase is a key enzyme in leucine degrada-
logical sequelae, including microcephaly, mental re- tion and in ketogenesis. The gene encoding HMG-
tardation, visual impairment, epilepsy, and central CoA lyase, HMGCL, is located on chromosome
motor deficits. 1p35.1–36.1. The enzyme HMG-CoA lyase is located
Laboratory examinations during these episodes in mitochondria and peroxisomes of all tissues.
always reveal an acidosis and almost always hypo- Leucine is sequentially converted to 3-methyl-
glycemia, which is often profound. Ketone bodies are crotonyl-CoA, 3-methylglutaconyl-CoA, and HMG-
inappropriately low. Lactate may be markedly elevat- CoA. Fatty acid degradation leads to the formation of
ed, especially in neonates. Many patients have abnor- acetyl-CoA, which joins acetoacetyl-CoA to form
mal liver function tests and hyperammonemia. Pan- HMG-CoA. HMG-CoA lyase catalyzes the conversion
creatitis is another complication that may occur. Uri- of HMG-CoA to acetoacetate. Acetoacetate and
nary organic acid analysis reveals increased excretion 3-hydroxybutyrate are the principal ketone bodies.
of 3-hydroxy-3-methylglutaric acid, 3-hydroxyisova- 3-Hydroxybutyrate is derived directly from acetoac-
leric acid, 3-methylglutaconic acid, and 3-methylglu- etate in a dehydrogenase reaction. A third ketone
taric acid. 3-Methylcrotonylglycine may also be pre- body, acetone, arises from decarboxylation of ace-
sent. The diagnosis is confirmed by enzyme assess- toacetate. Ketone bodies are formed predominantly
ment in leukocytes or cultured fibroblasts. Fasting from fatty acids and ketogenic amino acids, princi-
and leucine loading are usually unnecessary for diag- pally leucine. Ketone bodies are used as alternative
nosis. They may be useful to determine fasting and source of energy for the brain when production of
protein tolerance, but they carry the risk of invoking energy from glucose is insufficient to meet the de-
metabolic decompensation and should be performed mands. The main function of ketone bodies may be to
under close supervision. DNA confirmation is op- provide the brain with a noncarbohydrate energy
tional. Neonatal screening is possible using analysis source during fasting. The brain cannot use fatty
of 3-methylglutarylcarnitine in blood. Prenatal diag- acids as an energy source. Under normal conditions,
nosis can be performed using metabolite measure- the main energy substrates of the brain are glucose
ments, enzyme essays, and molecular techniques. and its derivatives, such as lactate, but during pro-
longed fasting ketone bodies supply almost two-
thirds of the brain energy. Ketone bodies have a glu-
cose sparing effect, turning down peripheral and cen-
tral glucose utilization. They also reduce muscular
proteolysis.
322 Chapter 41 3-Hydroxy-3-Methylglutaryl-CoA Lyase Deficiency

In HMG-CoA lyase deficiency multiple different 41.5 Magnetic Resonance Imaging


mutations have been identified in HMGCL but so far
no clear genotype–phenotype correlation has been CT scan findings during a metabolic crisis in the
found. HMG-CoA lyase deficiency causes a block in neonatal period are characterized by white matter
the production of ketone bodies, leading to exhaus- hypodensity and swelling. In the occipital region,
tion of glucose to very low levels under conditions contrast between the cortex and white matter may
when ketogenesis is required, that is, insufficient have disappeared and follow-up CT may show severe
availability of glucose to meet energy demands. The cystic degeneration of the occipital and parieto-oc-
metabolic decompensation is characterized by hy- cipital white matter. These findings are not specific
poketotic hypoglycemia. Infancy is the period of for HMG-CoA lyase deficiency, but are characteristic
highest risk for decompensation. sequelae of profound neonatal hypoglycemia. Follow-
In patients who survived a period of profound hy- up MRI shows evidence of gliotic scarring and atro-
poglycemia in the neonatal period the remaining phy of the area involved (Fig. 41.1). In addition, le-
cerebral damage may mainly involve the posterior sions in the globus pallidus, thalamus, putamen, and
part of the brain, as typically happens in neonatal hy- caudate nucleus may occur following a neonatal
poglycemia. The cause of the remaining white matter hypoglycemic crisis (Fig. 41.1).
abnormalities seen in patients with HMG-CoA lyase Hypoglycemia after the neonatal period usually
deficiency is unknown. These abnormalities must leads to more diffuse cerebral cortical damage. In
somehow be related to the specific metabolic de- some children a stroke, e.g., a unilateral anterior and
rangement, because they are not seen in other disor- middle cerebral artery infarction arising during a
ders with hypoketotic hypoglycemia. metabolic crisis, has been reported. In an untreated
infant with a moderately severe chronic metabolic de-
rangement, macrocephaly and diffuse white matter
41.4 Therapy hypodensity and swelling have been reported (Lisson
et al. 1981; Leupold et al. 1982), suggesting diffuse
The mainstay of treatment is suppression of ketogen- white matter spongiosis. Cystic degeneration of the
esis. During a metabolic crisis, immediate attention white matter was documented on follow-up.
should be focused on correction of the blood pH by MRI findings that are characteristic of HMG-CoA
sodium bicarbonate infusions and elevation of blood lyase deficiency include multiple abnormal white
glucose by glucose infusions. In addition, patients matter foci, varying in number from a few to, more
may require measures of general support including often, a multitude and varying in size from very small
assisted ventilation and repair of electrolyte and fluid – a few millimeters – to larger and more confluent le-
deficits. sions (Figs. 41.2 and 41.3). The abnormal white mat-
Long-term management of HMG-CoA-lyase-defi- ter foci may be seen on a background of slight to mild
cient patients should focus on avoidance of hypo- signal abnormalities of almost the entire cerebral
glycemia and of long fasts. A high-carbohydrate diet white matter, sparing the corpus callosum, variably
is recommended with moderate protein and fat re- sparing the U fibers (Fig. 41.2). The combination of
striction. The leucine intake should be adjusted to in- diffuse mild and multifocal more serious cerebral
sure normal growth. Patients should be under con- white matter abnormalities is unique. In addition, sig-
stant observation during periods of intercurrent ill- nal abnormalities may be seen in the posterior limb of
ness. A high carbohydrate intake must be maintained the internal capsule, hilus of the dentate nucleus, the
during any metabolic stress. If high carbohydrate in- dentate nucleus, and pontine tegmentum (Figs. 41.1–
take cannot be maintained, patients should be admit- 41.3). The cerebellar white matter is spared. These ab-
ted for intravenous glucose administration. normalities are also seen in well-treated patients who
Once the diagnosis has been established and this have never experienced a metabolic crisis and who
treatment strategy is applied, further neurological are clinically normal. The striking discrepancy be-
damage is usually prevented. tween the serious white matter abnormalities on MRI
and the lack of clinical findings in these patients is
unexplained. This observation excludes demyelina-
tion as histopathological basis.
41.5 Magnetic Resonance Imaging 323

Fig. 41.1. This 4-year-old girl with HMG-CoA lyase deficiency addition, mild signal changes are present in the basal ganglia,
experienced serious hypoglycemia in the neonatal period. She thalami, and dentate nuclei. The cerebral white matter is dif-
is severely retarded and has a visual impairment and micro- fusely abnormal. The corpus callosum is spared. From van der
cephaly. MRI shows damage of the parieto-occipital white and Knaap et al. (1998), with permission
gray matter, as may occur after neonatal hypoglycemia. In
324 Chapter 41 3-Hydroxy-3-Methylglutaryl-CoA Lyase Deficiency

Fig. 41.2. The 10-year-old sister of the patient in Fig. 41.1 also T1-weighted images only show the areas of more prominent
has HMG-CoA lyase deficiency, but never developed symp- signal abnormality; the diffuse signal changes are difficult to
toms of the disease. She was diagnosed after the diagnosis see. The corpus callosum is not involved. Signal abnormalities
was established in her sister. The T2-weighted images show are also seen in the dentate nucleus and the hilus of the den-
diffuse mild signal changes in the cerebral white matter with tate nucleus. From van der Knaap et al. (1998), with permission
additional spots of more prominent signal abnormality. The
41.5 Magnetic Resonance Imaging 325

Fig. 41.3. A 6-year-old boy with HMG-CoA lyase deficiency.He ty. These spots are also seen on the IR images (upper row, left).
had two episodes of hypoglycemia and lethargy in his third Slight signal changes are seen in the posterior limb of the in-
year of life,but his condition was never serious.His intelligence ternal capsule, thalami, pontine tegmentum, dentate nucleus,
and neurological examination are normal. MRI reveals diffuse and outflow tract of the dentate nucleus. From van der Knaap
signal changes in the cerebral white matter, sparing the U et al. (1998), with permission
fibers, with multiple foci of more prominent signal abnormali-
Chapter 42

Canavan Disease

42.1 Clinical Features diagnosis of CD was proven with biochemical or DNA


and Laboratory Investigations techniques, and the existence of a truly juvenile vari-
ant is doubtful.
Canavan disease (CD) is a rare hereditary neurologi- Laboratory investigations in CD are diagnostic. An
cal disorder affecting infants and children. The dis- increased concentration of N-acetylaspartic acid is
ease is also called spongy degeneration of the CNS of found in urine and plasma. CSF protein is sometimes
the Van Bogaert–Bertrand type. CD has an autosomal raised. EEG is diffusely slow with paroxysmal fea-
recessive mode of inheritance. It is most frequently tures. Evoked potentials are delayed or absent. Nerve
found in children of Jewish Ashkenazi origin. conduction velocity is normal. Decreased activity of
The infantile form of CD is the most common. The the enzyme aspartoacylase is found in cultured
patients appear normal at birth and the initial stages fibroblasts. Carrier detection on the basis of enzyme
of development are normal. The disease usually assessment is a problem because of large overlap be-
becomes apparent within the first 6 months of life. tween activities found in carriers and controls. Prena-
Early signs include hypotonia with poor head control, tal diagnosis by assessment of enzyme activity in am-
decreased motor activity, irritability, visual loss, poor niocytes and chorionic cells is unreliable owing to the
sucking ability, and accelerated skull growth. An ab- low levels of aspartoacylase in these cells. Assessment
normal increase in skull growth results in macro- of the level of N-acetylaspartate in amniotic fluid is
cephaly in most of the patients. Failure of develop- more reliable. Prenatal diagnosis is also possible
mental progress and regression occur. Gradually, hy- using DNA techniques.
potonia of arms and legs is replaced by spasticity and
tonic extensor spasms may occur. Axial hypotonia
persists. Blindness with coarse nystagmoid eye move- 42.2 Pathology
ments and pale optic discs at funduscopy become
evident. Occasionally sensorineural hearing loss is The brain is abnormally enlarged in CD. Otherwise
noted. Choreoathetosis, dystonia, and tonic or my- the external appearance of the cerebral gyri, cerebel-
oclonic seizures occur in some patients. Eventually a lum, and brain stem are normal. On sectioning, an ill-
chronic vegetative state with decerebrate or decorti- defined demarcation between cortex and white mat-
cate rigidity appears. Autonomic crises may occur ter is seen in all parts of the cerebral and cerebellar
with episodically increased vasomotor responses, dis- hemispheres. The deeper cortex and subcortical
turbed temperature regulation, and vomiting. The white matter are edematous and soft. Cavitation of
patients generally die before the age of 4 years. How- the affected white matter is only seen in advanced
ever, a considerable number of patient have a more stages of the disease. During the first few years, the
protracted course with a life expectancy beyond 10 ventricles are slightly narrowed. Thereafter, they
years, even into the third decade. gradually increase in size as a result of increasingly
In the uncommon congenital variant, children may severe loss of white matter.
appear normal at birth or may be in a poor condition. Histological examination shows vacuolation with
During the first few days after birth, inactivity and the presence of innumerable minor vacuoles, most
lethargy become manifest. The children cry frequent- striking in the subcortical cerebral white matter and
ly and exhibit difficulties in sucking and swallowing. in the deep layers of the cortex, giving the affected
They are hypotonic. They die within a few days or areas a spongy appearance. The periventricular white
weeks. matter is better preserved. No substances can be
A separate juvenile variant of CD has been distin- demonstrated within the vacuoles. Myelin degenera-
guished with normal development for several years. tion is present in a distribution that closely follows
Occasionally, children with CD may achieve more de- the status spongiosus. Thus, moderate to severe loss
velopmental milestones than usual and may even of myelin is seen in the subcortical areas, whereas the
achieve walking; they may have a highly protracted periventricular white matter, corpus callosum, inter-
course of disease and live longer than expected; but nal capsule, and fornix are less severely vacuolated
an entirely normal development for several years has and their myelin content is better preserved. In the
never been documented in patients in whom the areas of severe myelin loss axons and oligodendro-
42.4 Pathogenetic Considerations 327

cytes are largely intact. Myelin vacuolation and loss is In the congenital variant the sponginess is most se-
accompanied by astrogliosis. Myelin breakdown vere in the cerebellum and brain stem, which are the
products are scarce; some sudanophilic lipids may be parts with the highest myelin content at that time.
present in macrophages. Cortical neurons are normal
in number and appearance, while intracortical myelin
is lost. Within the cortex, there is a marked prolifera- 42.3 Chemical Pathology
tion of swollen astroglia with the morphologic fea-
tures of Alzheimer type II cells. Their nuclei are In CD, chemical abnormalities in the brain are limit-
abnormally large, variably shaped, with unevenly dis- ed to the white matter; the chemical composition of
persed nuclear chromatin, which tends to accumulate the gray matter is near-normal. The most prominent
near the nuclear membrane. The cytoplasm is swollen white matter abnormalities are its increased content
and watery. These cells are also seen in the subcorti- of water and N-acetylaspartate. Electrolyte analyses
cal white matter, but they do not prevail in other areas show that the composition of the excess fluid in the
of vacuolated white matter. brain is similar to that of plasma ultrafiltrate. There is
On electron microscopy, the vacuoles observed in a drastic decrease in myelin lipids and protein as
the white matter appear to be formed as a result of manifestations of the severe myelin loss. There is a
separation of myelin lamellae with intramyelinic vac- marked decrease of galactolipids and a relatively
uole formation. Splitting of the myelin lamellae smaller decrease in cholesterol and total phospho-
occurs at the intraperiod lines, where external sur- lipids. No cholesterol esters are detected in the white
faces of the original oligodendroglial membrane were matter.
closely apposed in the formation of the myelin The composition of myelin is altered. Cholesterol
sheath. The swollen astrocytes with enlarged nuclei is increased, and phospholipids are decreased, espe-
and watery cytoplasm contribute to the vacuolated cially ethanolamine phosphoglycerides. Galactolipids
aspect, in particular within the cortex. On electron are severely diminished, cerebroside more so than
microscopy these cells are shown to contain abnor- sulfatide. The total ganglioside concentration is nor-
mal, enormously elongated mitochondria. mal.
Sponginess is also present in the globus pallidus, This abnormal pattern of white matter and myelin
thalamus, and hypothalamus, whereas caudate nucle- composition is also seen in several other diseases and
us and in particular putamen tend to be better pre- is probably a result of nonspecific destruction of
served.Alzheimer type II cells are present in the basal myelin.
ganglia.
Within the cerebellum the most severe vacuolation
is seen in the Purkinje cell layer, the deeper areas of 42.4 Pathogenetic Considerations
the granule cell layer, and the subcortical white mat-
ter. Purkinje cells and granule cells are well preserved. CD is caused by a deficiency of aspartoacylase. The
Myelin loss is accompanied by astrocytosis, but axons gene for aspartoacylase, ASPA, is located on the short
are spared. The deep white matter of the cerebellum is arm of chromosome 17 (17p13-ter). Numerous differ-
better preserved.Vacuolation is present in the dentate ent mutations have been described. Homozygosity for
nucleus, but nerve cells are intact. one particular mutation (Glu285Ala) is responsible
Vacuolation, demyelination, and astrocytosis are for most Ashkenazi Jewish cases. In non-Jewish pa-
present in the midbrain, pons, medulla, and spinal tients the mutations are different and more diverse.
cord, both in descending and ascending tracts. How- There is no evident correlation between genotype
ever, the involvement of brain stem and spinal cord is and phenotype. Siblings homozygous for the same
variable in severity. Cranial nerve nuclei, other brain mutation can have a very different course of disease
stem nuclei, and spinal cord gray matter also show a and life span.
variable degree of vacuolation. The optic nerve, con- N-acetylaspartate is synthesized from acetyl-CoA
taining CNS myelin, is involved in the process. The and aspartate by L-aspartate-N-acetyl transferase.
other cranial nerves, the spinal roots, and the periph- This reaction occurs exclusively in neurons of the
eral nerves are normal. CNS. N-acetylaspartate is metabolized to aspartate
In protracted infantile patients, who survive for a and acetate by aspartoacylase. Aspartoacylase is ex-
relatively long time, the lateral ventricles are marked- clusively located in oligodendroglia. In deficiency of
ly dilated as a result of severe loss of white matter tis- aspartoacylase N-acetylaspartate concentrations in
sue. Myelin is now also lost in the deep white matter. the brain are elevated and the substance is excreted in
In addition, neuronal loss is severe, and vacuolation is large amounts in the urine.
seen in all layers of the cortex.
328 Chapter 42 Canavan Disease

The brain is the only organ in which the biosynthe- dysfunction, in contrast to earlier studies in which
sis of N-acetylaspartate has been demonstrated. Its abnormalities in mitochondrial Na+/K+-ATPase had
concentration in the brain is very high, and it is high- been reported (Adachi et al. 1966, 1972). Those who
er in gray than in white matter. N-acetylaspartate is believe that N-acetylaspartate is a molecular water
second to glutamic acid in its abundance in the hu- pump and has an important role in osmoregulation
man brain. It has been shown that N-acetylaspartate hypothesize that the myelin vacuolation and other
is mainly or exclusively localized in neurons. To date, pathological changes are the consequence of continu-
the functional role of this compound has not been ous increased hydrostatic pressure.
elucidated. Some suggest it is involved in neurotrans-
mission. The high concentration of N-acetylaspartate
in neurons may be a form of chemical compartmen- 42.5 Therapy
tation serving neurotransmission. N-acetylaspartate
is converted to aspartate and has been implicated in At present there is no specific therapy for CD patients.
the formation of glutamate. It has been found that not Seizures need to be controlled. Gene therapy has been
only N-acetylaspartate but also N-acetylaspartylglu- attempted. Clinical results have so far not been im-
tamate is elevated. Both aspartate and glutamate are pressive and at most some transient beneficial effects
excitatory neurotransmitters. N-acetylaspartylgluta- have occurred. Prolonged follow-up is not yet avail-
mate may be the storage form of glutamate. N-acety- able.
laspartylglutamate may also by itself interfere with
neurotransmitter receptors. Another suggestion is
that N-acetylaspartate may act as an osmolyte and 42.6 Magnetic Resonance Imaging
that its lack of hydrolysis leads to accumulation of
water in brain cells. N-acetylaspartate may function CT shows diffuse hypodensity of the white matter of
as a molecular water pump in myelinated neurons. It cerebral hemispheres and cerebellum. Involvement of
has also been proposed that N-acetylaspartate is an the globus pallidus with sparing of caudate nucleus
acetyl group donor in the synthesis of brain lipids, and putamen is usually seen.
including myelin lipids. In particular, the substance In infants with CD, diffuse cerebral white matter
would be important in the formation of cerebronic changes are seen (Figs. 42.1 and 42.2). In older pa-
acid, which is a precursor of ceramide, which in turn tients with CD, MRI shows that the most severe
is a constituent of sulfatide and cerebroside. Some abnormalities are present in the subcortical white
others suggest N-acetylaspartate is important in pro- matter of the cerebrum and cerebellum (Fig. 42.3).
tein synthesis. Central white matter structures, such as the periven-
The relationship between accumulation of N- tricular rim of white matter, internal capsule, and cor-
acetylaspartate and myelin vacuolation and forma- pus callosum are better preserved (Fig. 42.3). As the
tion of abnormal astrocytes has not been elucidated. disease progresses, the central white matter also be-
Those who suggest that the substance is essential for comes involved (Fig. 42.4). Thus, the spread of the
the normal synthesis of myelin lipids explain the white matter changes is centripetal. The white matter
myelin abnormality by faulty formation of one of its abnormalities are confluent and symmetrical in dis-
constituents. However, the chemical composition of tribution. The subcortical white matter has a mildly
myelin in CD shows only nonspecific abnormalities swollen aspect, broadening the gyri. Over time, white
related to the myelin breakdown process and no spe-
cific deficiencies of any of its components. Others
suggest that accumulation of N-acetylaspartate may 䊳

have toxic effects leading to myelin splitting. In this Fig. 42.1. A 4-month-old male infant with CD.Note the diffuse
respect, the comparison to hexachlorophene intoxica- signal abnormality of the cerebral and cerebellar white matter
tion and triethyltin intoxication is important, as both on the T2-weighted images (first three rows). The globus pal-
conditions are characterized by myelin splitting, vac- lidus and thalamus are involved,but the putamen and caudate
uolation, and loss, and presence of Alzheimer type II nucleus are spared. The posterior limb of the internal capsule
cells with abnormal mitochondria, as is the case in and brain stem are also abnormal. The FLAIR image (fourth
CD. Hexachlorophene and triethyltin both interfere row, left) demonstrates that the peripheral cerebral white mat-
with mitochondrial oxidative phosphorylation. N- ter has a high water content with a low signal intensity as com-
acetylaspartate is synthesized in mitochondria and if pared to the central white matter. On the trace diffusion-
this substance accumulates, its toxic effects may well weighted image (b = 1000; fourth row, middle) the central
be mainly exerted in these organelles. However, the white matter (posterior limb of the internal capsule and the
relationship between possible mitochondrial dys- optic radiation) has a high signal, whereas the same structures
function and myelin vacuolation is unclear. De Coo et have a low ADC in the range of 45–70 on the ADC map (fourth
al. (1991) could not find evidence of mitochondrial row, right), indicative of restricted diffusion in these regions
42.6 Magnetic Resonance Imaging 329

Fig. 42.1.
330 Chapter 42 Canavan Disease

Fig. 42.2.
42.6 Magnetic Resonance Imaging 331

Fig. 42.3. A 21-month-old boy with CD.These T2-weighted im- periventricular white matter, and posterior limb of the internal
ages show the involvement of the subcortical white matter, capsule, are spared. The midbrain is diffusely abnormal. From
globus pallidus, and, to a lesser extent, the thalamus. The cen- Meyding-Lamadé and Sartor (1993), with permission
tral white matter structures, including the corpus callosum,

Fig. 42.4. A 32-month-old child with CD. These T2-weighted bilaterally, whereas the putamen and caudate nucleus are
images show there is now involvement of all white matter spared.The brain stem contains signal abnormalities.There are
structures, including corpus callosum and periventricular signs of diffuse atrophy. From Kendall (1992), with permission
white matter. The globus pallidus and thalamus are involved

matter atrophy occurs with marked dilatation of the


lateral ventricles and to a lesser extent the subarach-
noid spaces (Figs. 42.4 and 42.5). Bilateral involve-
䊴 ment of the globus pallidus is seen and involvement
Fig. 42.2. The same boy with CD as in Fig. 42.1, now 12 of the thalamus often present (Figs. 42.1–42.4). The
months old. The pattern of abnormalities on the T2-weighted resulting image with sparing of putamen and caudate
images (first three rows) is essentially unchanged. The FLAIR nucleus is very typical of CD. Brain stem tracts are in-
image (fourth row, left) shows further white matter rarefaction volved as a rule (Figs. 42.1–42.5). The cerebellar white
with a lower signal of the cerebral white matter throughout. matter may also be affected. Over time, brain stem
The trace diffusion-weighted image (b = 1000; fourth row, and cerebellar atrophy ensues (Fig. 42.5).
middle) shows that large parts of the cerebral white matter Diffusion-weighted imaging shows restricted dif-
now have a low signal, whereas they have a high ADC in the fusion with low ADC values early in the course of the
range of 130–200 on the ADC map (fourth row,right),indicative disease (Fig. 42.1), whereas increased diffusion and
of increased diffusion high ACD values are found later in the course of the
332 Chapter 42 Canavan Disease

Fig. 42.5. A 19-year-old boy with CD. There is impressive cerebral and cerebellar atrophy. The images still show that the remain-
ing cerebral white matter is abnormal, but they no longer display the features characteristic of CD

disease (Fig. 42.2). The explanation is probably to be brain (Fig. 42.6) revealed signal abnormalities limited
found in the myelin vacuolation, which leads to re- to the U fibers, whereas the periventricular and deep
stricting of the extracellular space in the early stages, white matter had a normal signal. In addition, signal
whereas in the chronic stage destruction of the white abnormalities were seen in the caudate nucleus, thal-
matter matrix leads to increased size of the extracel- amus, and capsule between globus pallidus and puta-
lular space. Similar observations have been done in men. There were signal abnormalities in the external
maple syrup urine disease, a disease also character- and extreme capsules. The rest of the putamen ap-
ized by myelin vacuolation. peared to have a normal signal. The tegmentum of the
Proton MRS of the brain in CD patients reveals a pons, hilus of the dentate nucleus, and dentate nucle-
relatively high N-acetylaspartate resonance. The in- us were also involved. Thus, the pattern is very simi-
crease may only be relative in comparison with the lar to the pattern described above for classical CD, the
concentration of the other metabolites, although in- main difference being the fact that the cerebral white
crease in absolute values has also been reported. Lac- matter abnormalities were much more limited in
tate may be elevated. Choline is as a rule decreased. extent and affected the U fibers only.
The decrease in choline suggests a component of Two unusual patients with elevated urinary N-
hypomyelination rather than active myelin loss. acetylaspartate and aspartoacylase deficiency in
Zafeiriou et al. (1999) have reported MRI findings fibroblasts were reported by Toft et al. (1993). The
in two siblings with a mild and protracted course of clinical disease was much milder than in the usual
disease. The diagnosis was confirmed by finding ele- form of CD, with normocephaly, epilepsy, and mild
vated N-acetylaspartate in urine and deficiency of psychomotor retardation without regression at any
aspartoacylase in cultured fibroblasts. MRI of the time up to the age of 6 years. MR images showed sub-
42.6 Magnetic Resonance Imaging 333

Fig. 42.6. A 4-year-old female patient with a mild variant of pallidus and putamen have an abnormal signal. In addition,
CD. The U fibers are diffusely abnormal, whereas the central signal abnormalities are present in the dentate nucleus and
cerebral white matter is normal.The thalamus,head of the cau- hilus of the dentate nucleus. Subtle signal changes are seen in
date nucleus, external capsule, and capsule between globus the brain stem. From Zafeiriou et al. (1999), with permission

tle subcortical white matter changes in one patient, The differential diagnosis of diffusely swollen
whereas the white matter was normal in the other pa- white matter changes includes mainly late onset vari-
tient. Signal abnormalities were seen in globus pal- ants of maple syrup urine disease, Alexander disease,
lidus, putamen, and caudate nucleus. Proton MRS did congenital muscular dystrophy, and megalencephalic
not reveal an elevation of N-acetylaspartate, which is leukoencephalopathy with subcortical cysts. In the
highly unusual for CD. It is important to recognize presence of lesions of the globus pallidus with preser-
that the assessment of aspartoacylase activity is diffi- vation of the caudate nucleus and putamen, the im-
cult and not always reliable. Unfortunately, the diag- ages are highly suggestive of CD, but maple syrup
nosis was not confirmed by demonstration of muta- urine disease should be excluded by appropriate bio-
tions in the ASPA gene. chemical tests.
Chapter 43

L-2-Hydroxyglutaric Aciduria

43.1 Clinical Features genu of the internal capsule, optic tracts, and optic
and Laboratory Investigations radiations. Cerebral cortex, thalamus, brain stem, and
spinal cord were normal.
L-2-Hydroxyglutaric aciduria is a rare neurometabol-
ic disorder with autosomal recessive inheritance. The
children are initially normal. In the second year of life 43.3 Pathogenetic Considerations
a delay in unsupported walking, abnormal gait,
speech delay, or febrile convulsions form the present- L-2-hydroxyglutaric aciduria is caused by a deficiency
ing symptoms in most cases. In other patients no ab- of an FAD-linked, membrane-bound mitochondrial
normalities are noted until learning disabilities be- enzyme, L-2-hydroxyglutarate dehydrogenase, that
come apparent during early school years. Over the catalyzes the oxidation of L-2-hydroxyglutarate to
years slowly progressive neurological dysfunction is a-ketoglutarate. The gene encoding this enzyme,
noted, characterized predominantly by cerebellar C14orf160, also called DURANIN, is located on chro-
ataxia with nystagmus, dysarthria, head titubation, mosome 14q22.1. Elevated levels of L-2-hydroxyglu-
trunk ataxia, dysmetria, and intention tremor. Slow tarate are probably toxic for the CNS. The elevations
intellectual decline is noted in all patients. Frequently of lysine in blood and CSF may be secondary rather
noted abnormalities are extrapyramidal signs such than primary, as lysine loading appears to have no ef-
as dystonia and choreoathetosis, pyramidal signs, fect on the level of L-2-hydroxyglutaric acid in blood.
pseudobulbar signs, myoclonus, and macrocephaly. Two patients have been reported with neonatal-
Seizures occur in the majority of the patients, espe- onset encephalopathy, early death, and elevated L-2-
cially in the context of fever. In some patients stable hydroxyglutaric acid as well as lactate (Chen et al.
mental retardation is the only manifestation for years 1996; Barth et al. 1998). It is likely that these patients
and the slowly progressive decline of motor and cog- had another, unrelated metabolic disease.
nitive functions only starts in adolescence or adult-
hood. There is increasing evidence that patients with
L-2-hydroxyglutaric aciduria have an increased risk 43.4 Therapy
of CNS malignancies of various types.
Laboratory investigations reveal elevated urinary Treatment is entirely symptomatic.
excretion of L-2-hydroxyglutaric acid; plasma and
CSF levels are also increased. Lysine levels in urine,
plasma, and CSF may also be elevated. Prenatal diag- 43.5 Magnetic Resonance Imaging
nosis is possible by assessment of the L-2-hydroxyglu-
taric acid concentration in the amniotic fluid. MRI in patients with L-2-hydroxyglutaric aciduria
shows a highly characteristic and consistent pattern.
The abnormalities begin within the subcortical white
43.2 Pathology matter with multiple foci of high signal intensity on
T2-weighted images (Figs. 43.1 and 43.2). The lesions
Only a few descriptions of brain pathology are avail- have a tendency to become confluent, first in the
able. In 1994, Larnaout et al. described diffuse de- frontoparietal region (Fig. 43.1), later involving all
myelination, spongiosis, and cystic cavitation of the subcortical white matter in a confluent manner
cerebral white matter in a patient who died at the age (Figs. 43.2 and 43.3). In relatively mildly affected pa-
of 30 years. The abnormalities were most pronounced tients, the abnormalities remain multifocal and con-
in the subcortical region, in the axis of cerebral con- fined to the U fibers, whereas in patients with more
volutions. In the cerebellum, loss of granule cells and serious neurological handicap, the cerebral white
Purkinje cells and moderate pallor of the white mat- matter is more diffusely involved, although a rim of
ter were noted. The dentate nucleus and globus pal- periventricular tissue remains spared. The white mat-
lidus showed marked cell loss and severe spongiosis. ter abnormalities are mildly swollen with some
The putamen and caudate nucleus were less severely broadening of the gyri (Figs. 43.1–43.3). The aspect of
affected. Myelin was normal in the corpus callosum, swollen white matter changes is related to the spongi-
43.5 Magnetic Resonance Imaging 335

Fig. 43.1. A 12-year-old girl who has L-2-hydroxyglutaric partly confluent. Lesions are present in central gray matter
aciduria, showing the involvement of subcortical white matter structures, most prominently the globus pallidus and dentate
and the sparing of the central white matter including the cor- nucleus. The sagittal image shows that part of the inferior
pus callosum. The subcortical lesions are partly multifocal, vermis is absent

form nature of the leukoencephalopathy at histo- sule, and brain stem, are spared, although the internal
pathology. Diffusion-weighted MRI has revealed a capsule is involved in some cases. The lateral ventri-
high ADC and low signal on diffusion-weighted cles become slightly enlarged. In addition to the white
images in the abnormal white matter, consistent with matter abnormalities, changes in signal intensity are
increased freedom of water movements. Centrally typically seen in the globus pallidus, caudate nucleus,
located white matter structures, including periven- and putamen (Fig. 43.1). The globus pallidus is usual-
tricular white matter, corpus callosum, internal cap- ly most severely involved. The caudate nucleus may be
336 Chapter 43 L-2-Hydroxyglutaric Aciduria

Fig. 43.2. MR images of a 10-year-old boy with L-2-hydroxyg- white matter structures are spared, which is nicely shown by
lutaric aciduria, showing confluent involvement of the subcor- the T1-weighted image
tical white matter, which is also mildly swollen. The central

atrophic. The cerebellar vermis becomes highly The pattern of symmetrical abnormalities with
atrophic (Fig. 43.3). The cerebellar hemispheres are supratentorial subcortical white matter lesions, in-
also atrophic, but less severely. In the dentate nucleus fratentorial atrophy of the cerebellum (vermis more
a change in signal intensity is present (Figs. 43.1– than hemispheres), and lesions in both dentate nuclei
43.3). The cerebellar white matter is most often nor- is reminiscent of the patterns seen in Kearns–Sayre
mal. syndrome and Canavan disease. In both of the latter
disorders brain stem involvement is as a rule present.
43.5 Magnetic Resonance Imaging 337

Fig. 43.3. The T2-weighted images of a 25-year-old female the dentate nucleus has disappeared. What remains of the
patient with L-2-hydroxyglutaric aciduria show involvement of basal ganglia has a mildly abnormal signal. The cerebellum is
all subcortical and deep cerebral white matter with sparing of highly atrophic. Courtesy of Dr. N. Giffin, Royal United Hospital
a thin periventricular rim. The basal ganglia are atrophic and Bath, Bath, United Kingdom
Chapter 44

D-2-Hydroxyglutaric Aciduria

44.1 Clinical Features resent a separate third variant. Prenatal diagnosis


and Laboratory Investigations may not be reliable in this latter disorder.

D-2-Hydroxyglutaric aciduria is a rare neurometabol-


ic disorder with autosomal recessive inheritance. Two 44.2 Pathology
different variants are distinguished: a severe variant
and a mild variant. The two variants have never been There is a detailed histopathological description of
observed within the same family. only one patient with a severe phenotype (Eeg-Olofs-
Severe D-2-hydroxyglutaric aciduria results in neo- son et al. 2000). The boy had severe psychomotor re-
natal- or early-infantile-onset encephalopathy with tardation and epilepsy and died at the age of 14 years.
serious epilepsy, hypotonia, visual failure, and no or The middle cerebral arteries showed multiple saccu-
little development. The infants are often irritable and lar aneurysms. There was diffuse atrophy of the cere-
lethargic. Episodic vomiting may be present. Inspira- bral white matter with marked dilation of the lateral
tory stridor, dyspnea, and apnea may occur. Further ventricles. Microscopy showed loss of myelinated
signs may be spasticity, chorea, or dystonia. Both fibers and some astrogliosis within the cerebral white
macrocephaly and microcephaly may occur. Many matter. The cerebral cortex was intact. The basal gan-
patients have signs of cardiomyopathy. Hepatomegaly glia and cerebellum showed slight atrophy. Micro-
is of rare occurrence. Signs of mild facial dysmor- scopically, there was some loss of Purkinje cells and a
phism consisting of a flat face with a broad nasal reduction of cerebellar white matter. The brain stem
bridge and external ear anomalies are frequent. Early was intact.
death may occur, but the patients may also survive
with severe developmental delay and epilepsy.
Mild D-2-hydroxyglutaric aciduria is clinically 44.3 Pathogenetic Considerations
much more variable. Hypotonia, developmental
delay of variable severity, and epilepsy are the most D-2-hydroxyglutaric aciduria is caused by a deficien-
frequent signs of the disease. Macrocephaly may cy of the mitochondrial enzyme D-2-hydroxyglu-
be present. Multiple cerebral infarctions associated tarate dehydrogenase, which converts D-2-hydroxy-
with acute-onset neurological signs have been report- glutarate into 2-ketoglutarate. The enzyme is encoded
ed in one patient with D-2-hydroxyglutaric aciduria by a gene with the code name MGC25181, located on
and no identifiable known vascular risk factors chromosme 2p25.3. It is not yet known whether defi-
(van der Knaap et al. 1999b). Cardiomyopathy has ciency of this enzyme underlies the disease in all pa-
been observed in few patients. However, some pa- tients.
tients have no clinical problems that can be related to D-2-hydroxyglutaric acid is a metabolic intermedi-
the D-2-hydroxyglutaric aciduria. These patients are ate in a variety of pathways. It is striking that the uri-
detected in the course of screening for D-2-hydroxy- nary organic acid profile often shows elevated excre-
glutaric aciduria in a family with a symptomatic tion of 2-ketoglutarate. Sometimes the increased uri-
child. nary excretion of 2-ketoglutarate is accompanied by
Laboratory investigations reveal elevated urinary elevated excretion of other citric acid cycle interme-
excretion of D-2-hydroxyglutaric acid; plasma and diates. The latter abnormalities probably reflect a sec-
CSF levels are also increased. The urinary organic ondary disturbance of the citric acid cycle and mito-
acid profile often shows elevated excretion of 2-keto- chondrial respiratory chain. It has been shown that el-
glutarate, sometimes accompanied by elevated excre- evated levels of D-2-hydroxyglutaric acid in brain tis-
tion of other citric acid cycle intermediates and lac- sue lead to strong inhibition of cytochrome-c oxidase
tate. Nonketotic dicarboxylic aciduria is observed in activity (complex IV) in a dose-dependent manner in
some patients. Prenatal diagnosis is possible by as- vitro (da Silva et al. 2002). Secondary respiratory
sessment of D-2-hydroxyglutaric acid concentration chain dysfunction may be important in the patho-
in amniotic fluid. A few patients with the severe phe- physiology of the clinical symptomatology in D-2-hy-
notype have combined D- and L-2-hydroxyglutaric droxyglutaric aciduria. The nonketotic dicarboxylic
aciduria. It has been suggested that these patients rep- aciduria observed in some patients may reflect a sec-
44.5 Magnetic Resonance Imaging 339

Fig. 44.1. T1-weighted (first row) and T2-weighted (second gyration are delayed. The opercula are insufficiently devel-
row) images in a 4-month-old patient with the severe variant oped and the gyri are too coarse for the age of the infant.There
of D-2-hydroxyglutaric aciduria. The lateral ventricles are are large subependymal cysts over the body of the caudate
enlarged, most pronouncedly in the posterior region.The sub- nucleus. From van der Knaap et al. (1999a), with permission
arachnoid spaces are also mildly enlarged. Myelination and

ondary disturbance of the mitochondrial fatty acid enlarged, mainly in the posterior part. The ventricu-
b-oxidation. lar enlargement is often combined with mildly en-
larged frontal subarachnoid spaces and frontal sub-
dural effusions (Figs. 44.1 and 44.2). Secondary sub-
44.4 Therapy dural hematomas may occur. Within the first few
months of life, subependymal germinolytic cysts are
Treatment is entirely symptomatic. almost invariably present over the head and corpus of
the caudate nucleus (Fig. 44.1). They disappear with
time and are rarely seen after 6 months. Follow-up
44.5 Magnetic Resonance Imaging MRI in the second year of life shows more advanced
gyration, but myelination remains delayed and in
In the early-onset severe variant of D-2-hydroxyglu- some patients patchy white matter abnormalities are
taric aciduria, MRI performed in the first few months seen (Fig. 44.2). The lateral ventricles may become
of life shows signs of delayed cerebral maturation, in- highly enlarged due to white matter atrophy. In one
cluding delayed myelination and gyration (Fig. 44.1). patient, multiple aneurysms of the large cerebral ar-
Gyri are insufficiently branched, and in some patients teries were found (Eeg-Olofsson et al. 2000; Fig. 44.3).
focal agyria is found, particularly in the occipital re- In the mild variant of D-2-hydroxyglutaric aciduria,
gion. The sylvian fissure is wide open due to insuffi- subependymal cysts and delayed myelination may be
cient formation of the frontal and temporal opercula seen on early MRI. The lateral ventricles and the
(Fig. 44.1). The lateral ventricles are usually mildly subarachnoid spaces may be mildly dilated. Subdural
340 Chapter 44 D-2-Hydroxyglutaric Aciduria

Fig. 44.2. T2-weighted images in a patient with the severe and subarachnoid spaces are mildly enlarged. There are small
variant of D-2-hydroxyglutaric aciduria at the age of 15 subdural effusions.From van der Knaap et al.(1999b),with per-
months. Myelination is seriously delayed. In addition, there are mission
multifocal white matter abnormalities. The lateral ventricles

Fig. 44.3. T2-weighted images in a patient with the severe left. Note the aneurysms in the M2 segment of the middle
variant of D-2-hydroxyglutaric aciduria at the age of 15 years. cerebral arteries on both sides. From van der Knaap et al.
The lateral ventricles are seriously dilated. A focal area of ab- (1999b), with permission
normal signal is visible within the cerebral white matter on the

effusions may be seen. On follow-up, MRI may be the major cerebral arteries (van der Knaap et al.
normal, or it may show deficient myelination or bilat- 1999b).
eral multifocal, patchy abnormalities in the cerebral The imaging findings in patients with combined
white matter or a combination of the two. The ventri- D- and L-2-hydroxyglutaric aciduria are similar to
cles are often mildly dilated (Fig. 44.4). In one patient, those seen in the severe variant of D-2-hydroxyglutar-
multiple infarctions developed in the territories of ic aciduria.
44.5 Magnetic Resonance Imaging 341

Fig. 44.4. T2-weighted images in a patient with the mild variant of D-2-hydroxyglutaric aciduria at the age of 4 years. There are
minor abnormal white matter spots.The lateral ventricles are mildly enlarged
Chapter 45

Hyperhomocysteinemias

45.1 Introduction The most common defect of the transsulfuration


pathway and the most frequent cause of severe hyper-
Hyperhomocysteinemias are associated with a diver- homocysteinemia is a deficiency of cystathionine b-
sity of neurological problems, ranging from mental synthase. This enzyme normally catalyzes the conver-
retardation or deterioration to signs of subacute com- sion of homocysteine to cystathionine, which is con-
bined degeneration of cord and cerebral infarctions. verted to cysteine.
Homocysteine lies at an important branch point of 5,10-Methylenetetrahydrofolate reductase defi-
sulfur amino acid metabolism. It is formed from me- ciency is the most common defect of folate metabo-
thionine by demethylation. It may either be converted lism. This enzyme deficiency leads to a lack of 5-
to cysteine through the transsulfuration reaction or methyltetrahydrofolate, a cosubstrate for the methio-
remethylated to form methionine. Remethylation to nine synthase reaction, which in turn leads to an im-
methionine requires methyl donors such as betaine pairment of the remethylation of homocysteine to
or 5-methyltetrahydrofolate. The metabolism of ho- methionine.
mocysteine requires folate and cobalamin (vitamin So-called mild hyperhomocysteinemia, both in the
B12) (Fig. 45.1). Consequently, a number of defects fasting state and after methionine loading, can be the
may underlie hyperhomocysteinemia: a defect in the consequence of heterozygosity for cystathionine b-
transsulfuration pathway, a defect in cobalamin me- synthase deficiency. Heterozygosity for 5,10-methyl-
tabolism, and a defect in folate metabolism. enetetrahydrofolate reductase deficiency does not

Fig. 45.1. Homocysteine metabolism


45.2 Clinical Features and Laboratory Investigations 343

lead to hyperhomocysteinemia. However, homozy- malonyl-CoA mutase and consequently to a combina-


gosity for a mutant form of 5,10-methylenetetrahy- tion of hyperhomocysteinemia and methylmalonic
drofolate reductase, characterized by thermolability, aciduria.
can lead to mild hyperhomocysteinemia. Heterozy-
gosity for this mutation does not lead to hyperhomo-
cysteinemia. Compound heterozygosity for the classi- 45.2 Clinical Features
cal and the thermolabile mutations of 5,10-methyl- and Laboratory Investigations
enetetrahydrofolate reductase may also lead to mild
hyperhomocysteinemia. Cystathionine b-synthase deficiency leads to the clas-
Folic acid deficiency leads to a disturbance of the sical clinical picture commonly associated with the
folate cycle and, as a consequence, to hyperhomocys- term “homocystinuria.” The disease has an autosomal
teinemia. recessive inheritance. There is considerable clinical
Inborn errors affecting intracellular cobalamin variability, even within the same family. Psychomotor
metabolism lead to a deficiency of methylcobalamin retardation is often a feature of the disease and be-
and/or adenosylcobalamin. Methylcobalamin is an comes evident during the first or second year of life.
essential cofactor for methionine synthase, the en- In the course of time, the optic lenses become dislo-
zyme catalyzing the conversion of homocysteine to cated in most patients. Other ophthalmological ab-
methionine. Adenosylcobalamin is a cofactor for normalities include myopia and, less frequently, glau-
methylmalonyl-CoA mutase. The inborn errors of coma, optic atrophy, retinal abnormalities, and
cobalamin metabolism can be divided into three cataract. There are often skeletal abnormalities in-
main groups: isolated defects in methylcobalamin cluding osteoporosis, scoliosis, arachnodactyly, thin-
synthesis, isolated defects in adenosylcobalamin syn- ning and lengthening of the long bones, and other ab-
thesis, and defects in the synthesis of both methyl- normalities in the configuration and maturation of
cobalamin and adenosylcobalamin. The patients with the skeleton. The combination of ectopic lenses and
isolated defects in methylcobalamin synthesis can be tall and thin habitus may give the patient a marfanoid
divided into two complementation groups (CblE and appearance. Patients often present with signs of pre-
CblG), which are very similar clinically and biochem- mature arteriosclerosis and thromboembolism. Vas-
ically with the presence of hyperhomocysteinemia. cular occlusion can occur in any vessel at any age.
Both are characterized by a deficiency of methionine Fifty percent of the untreated patients suffer a cardio-
synthase, also known as methyltetrahydrofolate: ho- vascular incident before the age of 30 years, and cere-
mocysteine methyltransferase. Complementation bral infarction accounts for more than 60% of these
group CblG deficiency of methionine synthase is incidents. Related neurological problems include
caused by mutations in the gene encoding methion- hemiparesis and other focal neurological signs. In ad-
ine synthase. In complementation group CblE the dition, generalized dystonia, other extrapyramidal
functional deficiency of methionine synthase is movement disturbances, and epilepsy, usually with
caused by a defect in the reductive activation of me- grand mal convulsions, may occur. Behavioral distur-
thionine synthase due to a defect in methionine syn- bances and personality problems occur in a consider-
thase reductase. The patients with isolated defects in able number of patients. There are no signs of bone
adenosylcobalamin synthesis can be divided into two marrow dysfunction.
complementation groups, designated CblA and CblB. The clinical severity of 5,10-methylenetetrahydro-
Biochemically they are characterized by methyl- folate reductase deficiency varies greatly from patient
malonic aciduria without hyperhomocysteinemia. As to patient, even within the same family. The disease
hyperhomocysteinemia is not present in these dis- has an autosomal recessive mode of inheritance. On-
eases, they are not discussed in this chapter. Among set of clinical manifestations varies from early infan-
the patients with a defect in the synthesis of methyl- cy to adulthood. More than half of the patients be-
cobalamin and adenosylcobalamin, three comple- come symptomatic during the first year of life.
mentation groups can be distinguished, CblC, CblD, Among the most common early clinical manifesta-
and CblF. Biochemically they are characterized by tions are lethargy, epilepsy, hypotonia and severe de-
presence of both hyperhomocysteinemia and methyl- velopmental delay, often in combination with micro-
malonic aciduria. In CblC and CblD, there is a defect cephaly. In early-onset disease, early death is not rare
in cellular metabolism of cobalamin. In CblF there and can occur during an episode of apnea or may fol-
appears to be a defect in the process by which cobal- low respiratory problems. In older children, a distur-
amin exits from lysosomes after being taken up by re- bance of gait is a common finding. Neurological ex-
ceptor-mediated endocytosis. amination reveals a combination of spasticity and
Cobalamin cannot be synthesized by the body and sensory disturbances of the legs, consistent with com-
is dietary in origin. Deficiency leads to a disturbance bined dysfunction of the dorsal columns and pyrami-
of function of both methionine synthase and methyl- dal tracts of the spinal cord (combined degeneration
344 Chapter 45 Hyperhomocysteinemias

of the cord), variably mixed with signs of a peripher- with subacute combined degeneration of the cord,
al polyneuropathy with weakness and sensory distur- brain involvement (hypotonia, developmental delay,
bances with a glove-and-stocking distribution. The seizures, microcephaly, hydrocephaly), polyneuropa-
disturbances of postural sense lead to loss of coordi- thy, and pigmentary retinopathy. In young children
nation. Seizures, psychiatric manifestations, atheto- neurological manifestations include psychomotor re-
sis, and parkinsonism may occur. There are no signs tardation or regression, lethargy, hypokinesis, hypo-
of megaloblastic anemia. Vascular complications are tonia, brisk reflexes, ataxia, optic atrophy, and
not common in patients with 5,10-methylenetetrahy- seizures. In older children the gastrointestinal prob-
drofolate reductase deficiency, but recurrent cerebral lems are less prominent and hematological and neu-
infarction and superior sagittal sinus thrombosis rological problems dominate, with signs of myelo-
have been reported. pathy, peripheral neuropathy, tremor, dementia and
Longstanding mild hyperhomocysteinemia relat- behavioral problems, and, rarely, optic atrophy or
ed to heterozygosity for cystathionine b-synthase de- retinopathy.
ficiency and to thermolability of 5,10-methylenete- Cobalamin deficiency can be observed in a num-
trahydrofolate reductase contributes to premature ar- ber of conditions. Deficiency due to insufficient di-
teriosclerosis of small and large vessels, which may etary intake is rare, but can be observed in strict veg-
become manifest with transient ischemic attacks, la- etarians (vegans) and in breast-fed babies of mothers
cunar infarctions, cerebral infarcts in large arterial who are cobalamin-deficient. Cobalamin in the diet is
territories, and spontaneous artery dissection. The released from protein in the acid environment of the
clinical symptomatology of stroke in these patients stomach. Here it binds to R proteins of gastric and
does not differ from that in cerebrovascular disease of salivary origin. Pancreatic proteases digest the R pro-
any other origin. teins and liberate cobalamin in the upper small intes-
Folate deficiency is one of the most common vita- tine, where it forms a complex with intrinsic factor,
min deficiencies. The major causes are poor dietary synthesized by gastric parietal cells. The newly
intake; conditions leading to malabsorption, such as formed complex binds with specific receptors in the
steatorrhea and sprue; and treatment with anticon- terminal ileum and is transported into the entero-
vulsants such as phenytoin or with antifolate drugs cyte. The complex is dissociated and cobalamin is
including methotrexate, trimethoprim, and tri- transported into the portal blood bound to trans-
amterene. In an advanced stage, the deficiency can cobalamin II. Transcobalamin II is the transport pro-
cause macrocytic anemia, neutropenia, thrombope- tein for cobalamin and also facilitates cobalamin up-
nia, or pancytopenia with macrocytosis. Neurological take by tissues. Disturbances of cobalamin uptake
complications are exceptional. However, there is evi- and transport can arise at all levels. R protein defi-
dence that folate deficiency without cobalamin defi- ciency as an inherited defect has been described in a
ciency can also cause subacute combined degenera- small number of patients. Intrinsic factor deficiency
tion of the cord and cerebral abnormalities. Heredi- can be caused by deficient synthesis, synthesis of a
tary congenital folate malabsorption has been de- mutant protein with decreased activity, autoimmune
scribed in a small number of patients. These patients gastritis with antibodies against parietal cells and in-
present at the age of 2–5 months with severe mega- trinsic factor, or gastrectomy. Cobalamin malabsorp-
loblastic anemia, diarrhea, mouth ulcers, and failure tion may be due to defective receptor on the entero-
to thrive. Most patients show progressive neurologi- cyte, defective receptor internalization, surgical re-
cal deterioration, with mild to severe mental retarda- section of the terminal ileum, inflammatory diseases
tion, seizures, peripheral neuropathy, ataxia, and of the ileum (sprue, ulcerative colitis, Crohn disease)
athetosis. There is evidence that decreased serum fo- and transcobalamin II deficiency. Nitrous oxide
late is also an independent risk factor for ischemic (N2O) is a very special cause of deficient cobalamin
stroke, probably related to the elevated homocysteine activity. It oxidizes active cobalamin to an inert form.
levels. Long-term exposure to this anesthetic substance, for-
Inborn errors of cobalamin metabolism are auto- merly used in the artificial ventilation of patients
somal recessive disorders with a variable clinical pic- with tetanus, leads to megaloblastic and aplastic bone
ture. Most cases become manifest in infancy, but de- marrow changes and combined degeneration of the
lay in onset of clinical manifestations to adolescence spinal cord. Patients with latent cobalamin deficiency
or adulthood has been described. In infancy, the pa- are exceedingly sensitive to neurological deteriora-
tients present with gastrointestinal signs and symp- tion following nitrous oxide anesthesia.
toms (feeding problems, vomiting, atrophic stomati- The clinical signs and symptoms of cobalamin de-
tis, glossitis, alternating diarrhea and constipation, ficiency usually follow a chronic course and have a de-
and failure to thrive), hematological signs (macrocyt- layed onset because of the large cobalamin stores in
ic anemia, less often thrombopenia, neutropenia, and the body. In infants fed on breast milk with a low
leukopenia), and neurological problems compatible cobalamin content, symptoms are delayed by several
45.3 Pathology 345

Table 45.1. Laboratory findings in the hyperhomocysteinemias


Homocysteine Methionine Methylmalonic acid Macrocytosis

Cystathionine b-synthetase deficiency ≠ ≠ – –


5,10-Methylene-tetrahydrofolate
reductase deficiency ≠ – – –
Cbl C, D, F ≠ – ≠ +
Cbl E, G ≠ – – +
Folate deficiency ≠ – – +
Cobalamin deficiency ≠ – ≠ +

months. The signs and symptoms are hematological etary cobalamin deficiency or deficiency due to gas-
(macrocytic anemia, usually mild thrombopenia, hy- trointestinal absorption problems, serum cobalamin
persegmentation of neutrophils, aplastic anemia), levels may only be slightly or moderately low, despite
gastrointestinal (atrophic glossitis, stomatitis, consti- other clinical evidence of important deficiency. Total
pation, diarrhea), and neurological. The neurological serum cobalamin is normal in transcobalamin II de-
signs and symptoms usually follow a chronic course, ficiency, and in CblC, CblD, CblF, CblE, and CblG. Dis-
but a more or less acute spinal cross-section syn- turbances of cobalamin absorption can be investigat-
drome does occur. Neurological abnormalities con- ed with the Schilling test. This test measures urinary
sist of a progressive spastic paraparesis with ataxia excretion of radioactively labeled cobalamin after
due to impairment of postural sense. The arms are oral administration. If the urinary level is below nor-
usually affected later and to a lesser extent than the mal, the test is combined with addition of intrinsic
legs. Peripheral neuropathy occurs with progressive factor to distinguish between cobalamin deficiency
weakness, loss of reflexes (but plantar reflexes usual- caused by lack of intrinsic factor and other causes of
ly remain extensor) and distal sensory disturbances. disturbances of the vitamin absorption. Differentia-
Visual problems may occur caused by optic atrophy. tion of CblC, CblD, CblF, CblE, and CblG is possible by
Mental signs are frequent and range from disturbed biochemical and complementation studies on cul-
development in infants to regression and dementia, tured fibroblasts.
lability, depression, irritability, confusion, psychosis, Cystathionine b-synthase activity and 5,10-meth-
and lethargy in older patients. Affected infants are of- ylenetetrahydrofolate reductase activity can be as-
ten microcephalic. Epileptic seizures may occur. sessed in chorionic villi and cultured amniocytes, fa-
The most important laboratory investigations in- cilitating prenatal diagnosis. Prenatal diagnosis in
clude assessment of the levels of homocysteine, disorders of intracellular cobalamin metabolism can
methionine, and methylmalonic acid in plasma be performed on amniocytes or chorionic villus sam-
and urine and assessment of hematological abnor- ples. In many disorders, a DNA-based prenatal diag-
malities, in particular megaloblastic anemia (see nosis is also possible.
Table 45.1). It is important to note that there may be In case of subacute combined degeneration of the
no hematological abnormalities, even in the presence spinal cord, CSF protein is usually slightly elevated.
of overt neurological abnormalities. In cystathionine SSEPs show normal or moderately slowed peripheral
b-synthetase deficiency, low levels of cystathionine conduction, normal conduction or mild slowing
and cystine are additional findings. The homocys- across the cervical portion of the median SSEP, and
teine levels are highest in cystathionine b-synthase absence or severe slowing of impulse propagation
deficiency, lowest in the so-called mild hyperhomo- along the spinal cord with the peroneal SSEP. The
cysteinemias. Mild hyperhomocysteinemia can be conduction velocity of peripheral nerves is normal or
detected by performing a methionine loading test or mildly to more markedly reduced. There may be signs
by assessment of serum homocysteine levels during of denervation.
fasting. The assessment of the CSF level of S-adeno-
sylmethionine is of value as a decrease is associated
with present or imminent demyelination. 45.3 Pathology
Direct enzyme assessment confirms the diagnosis
in cystathionine b-synthase deficiency and 5,10- Two types of cerebral pathology are consistently seen
methylenetetrahydrofolate reductase deficiency. The in severe hyperhomocysteinemias: subacute com-
enzyme assessment can be performed in liver biopsy bined degeneration of the spinal cord and brain and
specimens, cultured fibroblasts, and lymphocytes. the cerebral consequences of premature vascular dis-
Serum folate is low in folate deficiency. In case of di- ease.
346 Chapter 45 Hyperhomocysteinemias

The vascular changes affect both arterial and ve- matter volume occurs with enlargement of ventricles
nous systems. Histologically, the vascular disease is and subarachnoid spaces.
characterized by intima and media smooth muscle Peripheral nerves show signs of a combination of
hyperplasia, with fibrosis and thickening of the elas- segmental demyelination and axonal degeneration.
tic lamina. In cystathionine b-synthase deficiency the main
Subacute combined degeneration of the spinal pathological findings are arteriosclerosis, arterial
cord is a demyelinating disorder. The brain is often af- thromboembolism, and venous thrombosis in chil-
fected at the same time. The form the disorder takes, dren and young adults. Resulting lesions consist of ar-
both clinically and histopathologically, is strongly in- terial and venous infarcts. Most often multiple small
fluenced by the age of the patient, or rather, the infarcts of different ages are seen, spread over the
progress of myelination at the time of onset of dis- brain. In cystathionine b-synthase deficiency de-
ease. In early infantile onset, the disease may take the myelination is rare. In one patient (Chou and Wais-
form of delayed and disturbed myelination rather man 1965), a vacuolating white matter disease was
than demyelination, whereas after completion of documented, predominantly involving the subcorti-
myelination the classical picture of subacute com- cal white cerebral and cerebellar white matter, rela-
bined degeneration of the cord and demyelinating tively sparing the corpus callosum, internal capsule,
brain disease arises. and brain stem. The spinal cord was also involved. In
Degeneration and demyelination of the spinal one patient a combination of demyelination and ex-
cord, in particular the dorsal and lateral columns, lent tensive vascular abnormalities with multiple small
the disease its name: subacute combined degenera- infarcts was found (Dunn et al. 1966).
tion of the cord. The midthoracic segments of the In 5,10-methylenetetrahydrofolate reductase defi-
cord are mainly involved where a zone of white mat- ciency vascular pathology is present but less severe
ter destruction may affect the whole cord and not on- than in cystathionine b-synthase deficiency and cere-
ly the long tracts. In the upper cervical segments the bral infarcts are rare. Demyelination of brain and
posterior columns are predominantly affected, and at spinal cord as described above is the most common
the lower lumbar levels predominantly the pyramidal type of pathology. The findings in folate deficiency
tracts. As pathological changes predominate in the are similar.
midthoracic segments, subsequent wallerian degen- In the inherited defects in cobalamin metabolism
eration may play a role in the involvement of the as- and in cobalamin deficiency, spongy demyelination
cending tracts in the cervical segments and of the de- in the patterns of subacute combined degeneration of
scending tracts in the lumbar segments. The first cord and demyelinating brain disease is the predomi-
changes consist of swelling and splitting of myelin nant finding. Additional changes are present in arte-
sheaths, followed by spongiform white matter degen- rioles and capillaries with thickening, fibrosis, and
eration and demyelination. Electron microscopy hyalination, but cerebral infarctions have not been re-
shows that the myelin splitting occurs at the intrape- ported.
riod line. A variable astrocytic reaction is present; To date, cerebral infarction is the only type of
sometimes it is considerable. pathology that has been observed in mild hyperho-
In the cerebral white matter variable demyelina- mocysteinemia, in most cases related to heterozygos-
tion is present. In many cases the demyelinating le- ity for cystathionine b-synthase deficiency. This ob-
sions are small, ill defined, and located perivascularly. servation may be biased as children and young adults
Their number is variable. They may be scanty or dis- with cerebrovascular accidents are screened for the
seminated over wide areas. Sometimes the cerebral presence of mild hyperhomocysteinemia. Systematic
white matter changes become extensive and diffuse, data on neuropathological findings in unselected pa-
but the internal capsule and brain stem remain rela- tients are not available.
tively spared. In occasional cases a predominance of
subcortical white matter involvement has been men-
tioned, with the brunt of abnormalities in the white 45.4 Pathogenetic Considerations
matter at the junction of the cortex. The histology of
the cerebral lesions is similar to that of the spinal Two types of pathology dominate in the hyperhomo-
cord. There is first a fusiform swelling of myelin cysteinemias: vascular pathology and myelinopathy.
sheaths, myelin splitting, and formation of in- The relative contribution to the clinical symptomatol-
tramyelinic vacuoles, followed by demyelination and ogy varies by disease. In cystathionine b-synthase
finally also axonal degeneration. The optic nerves are deficiency, and also in mild hyperhomocysteine-
often involved in the demyelinating process. The mia, vascular pathology dominates, whereas in 5,10-
basal nuclei including the thalamus may also become methylenetetrahydrofolate reductase deficiency, dis-
involved in the process of myelin vacuolation. In turbances of cobalamin metabolism (CblC, CblD,
more extensive white matter disease loss of white CblF, CblE, and CblG), exogenous folate deficiency,
45.4 Pathogenetic Considerations 347

and cobalamin deficiency, the myelinopathy domi- cient synthesis of choline may contribute to the mye-
nates. linopathy.
The pathogenesis of the vascular pathology is only Administration of folic acid in patients with cobal-
partially understood. There is evidence from experi- amin deficiency may precipitate or exacerbate de-
mental animal work and from in vitro research that myelination. Folic acid is not active and requires
homocysteine damages the vascular endothelium. In- conversion to active tetrahydrofolate. This reaction is
jury of arterial and venous vessel walls predisposes to very slow. Folic acid competes with the transport of
early platelet activation and thrombus formation. Ho- active tetrahydrofolate in the CNS. The resulting de-
mocysteine is rapidly auto-oxidized when added to creased availability of tetrahydrofolate for nervous
plasma, and potent reactive oxygen species, including tissue is probably the cause of enhanced demyelina-
superoxide, hydrogen peroxide, and hydroxyl radi- tion.
cals, are produced during this process. These reactive The folate cycle is not only involved in the re-
oxygen species have been implicated in the vascular methylation of homocysteine to methionine, a reac-
toxicity of hyperhomocysteinemia. Platelet abnor- tion that is crucial to the synthesis of S-adenosylme-
malities and abnormalities in soluble factors involved thionine, it is also involved in the transfer of methyl
in blood coagulation may contribute to the thrombot- groups necessary for the synthesis of purines and
ic diathesis. pyrimidines, major constituents of DNA. There is
Plasma homocysteine levels are highest in cys- general agreement that megaloblastic anemia or pan-
tathionine b-synthase deficiency, explaining the high cytopenia and the defective proliferation of rapidly
incidence of vascular accidents in this disorder. It is dividing cells with sequelae such as glossitis, intesti-
remarkable, however, that vascular abnormalities and nal problems, and hypospermia, are related to impair-
vascular accidents occur relatively frequently in the ment of DNA synthesis due to interference with folate
mild hyperhomocysteinemias, which have a lower metabolism. The main problem with folate metabo-
level of homocysteine than for instance the inherited lism in the cobalamin disorders (CblC, CblD, CblE,
cobalamin disorders, whereas vascular pathology is CblF, and CblG) and cobalamin deficiency is that
rare in the latter conditions. The chronicity of elevat- 5-methyltetrahydrofolate is not converted to tetrahy-
ed homocysteine levels before detection and treat- drofolate, whereas the conversion of 5,10-methyl-
ment, in view of the absence of other clinical prob- enetetrahydrofolate to 5-methyltetrahydrofolate is ir-
lems in heterozygosity for cystathionine b-synthase reversible. This is called the methyl–folate trap. The
deficiency, may form part of the explanation. trap leads to deficiency of folate coenzymes derived
Most current evidence points to deficiency of from tetrahydrofolate, necessary in the production of
S-adenosylmethionine being critical to the develop- purines and pyrimidines.
ment of demyelination. In the human brain S-adeno- In cases of deficiency of folate or cobalamin, or
sylmethionine is the universal methyl group donor, in inborn errors of folate or cobalamin metabolism,
acting in a wide variety of biological methylations CSF levels of 5-hydroxyindolacetic acid (5-HIAA)
that modify proteins, nucleic acids, fatty acids, and homovanillic acid (HVA), metabolites of sero-
phospholipids, and polysaccharides. S-adenosyl- tonin and dopamine, respectively, are decreased. The
methionine is necessary for the inactivation of cate- decrease is independent of the level of S-adenosylme-
cholamines and other biogenic amines. The methyl thionine. Why these metabolites are reduced is not
transfer pathway provides precursors for polyamine known. Neurotransmitter disturbances may be re-
synthesis. A relationship has been found between sponsible for the frequently observed extrapyramidal
the presence of demyelination and deficiency of movement abnormalities, epilepsy, and changes in
S-adenosylmethionine in the CSF, whereas remyelina- mood and personality.
tion under treatment is associated with a return of The gene encoding cystathionine b-synthase, CBS,
the S-adenosylmethionine level to normal. Addition- is located on chromosome 21q22.3. Different muta-
al evidence comes from animal experimental work, tions have been described. Cystathionine b-synthase
in which cycloleucine is used to elicit subacute com- requires pyridoxal phosphate (vitamin B6) as a cofac-
bined degeneration of the spinal cord. Cycloleucine tor. In the case of a mutation, there is either no resid-
causes deficiency of S-adenosylmethionine by inhibi- ual enzyme activity, reduced activity and normal
tion of methionine adenosyltransferase. The precise affinity for the cofactor, or reduced activity and re-
mechanism by which a deficiency of S-adenosyl- duced affinity for pyridoxal phosphate. The vitamin-
methionine would lead to demyelination is not B6-responsive patients do better in all aspects of the
known. The failure of methylation of arginine107- disease than nonresponsive patients, even if both
myelin basic protein may be important in this respect. groups are untreated. They have a higher IQ and all
In addition, methyl groups are necessary for synthe- other manifestations of the disease occur later.
sis of choline from ethanolamine, and these methyl The gene for 5,10-methylenetetrahydrofolate re-
groups are provided by S-adenosylmethionine. Defi- ductase, MTHFR, is located on chromosome 1p36.3.
348 Chapter 45 Hyperhomocysteinemias

Many different mutations have been described. Ther- ma homocysteine levels. The long-term effects of this
molability of the enzyme is due to a CÆT substitution treatment have not been fully assessed.
at position 677 of the gene. Homozygosity for this 5,10-Methylenetetrahydrofolate reductase defi-
mutation is associated with mild hyperhomocys- ciency has proven very resistant to treatment and on-
teinemia. The 677TÆC allele has a high prevalence ly limited success has been achieved. Therapeutic op-
(30–40%) in most populations, but varies greatly in tions include (1) use of folates such as folic acid or
different ethnic groups. About 10% of Caucasians are folinic acid in an attempt to maximize any residual
homozygous, but in populations of African descent enzyme activity; (2) use of methyltetrahydrofolate to
the prevalence is 0–2%, whereas in Asians it is about replace the missing product; (3) use of methionine to
20%. correct the cellular methionine deficiency; (4) use of
Functional deficiency of methionine synthase is pyridoxine to lower homocysteine levels because of
caused either by mutations in the gene encoding me- its role as a cofactor for cystathionine b-synthase; (5)
thionine synthase, MTR (complementation group use of cobalamin because of its role as a cofactor for
cblG), or in the gene encoding methionine synthase methionine synthase; (6) use of carnitine; and (7) use
reductase, MTRR, leading to a defect in the reductive of betaine in order to lower homocysteine levels and
activation of methionine synthase (complementation supplement methionine levels. In most patients sev-
group cblE). eral agents are used in combination.
Among the patients with a defect in the synthesis Folate deficiency can be corrected by supplemen-
of both methylcobalamin and adenosylcobalamin tation of the deficient substance.
three complementation groups are distinguished: All inborn errors of intracellular cobalamin me-
CblC, CblD, and CblF. In cblC and cblD, there is a tabolism are treated with intramuscular hydroxy-
defect in cellular metabolism of cobalamin. In cblF cobalamin. After induction, the hydroxycobalamin
there appears to be a defect in the process by which can be given orally because cobalamin absorption
cobalamin exits from lysosomes after being taken up and transport are normal. The biochemical and
by receptor-mediated endocytosis. hematological response to the therapy is usually
rapid. Plasma levels of methionine and homocysteine
can be used to monitor treatment. In CblE and CblG
45.5 Therapy the use of hydroxycobalamin is usually sufficient, al-
though supplementation with methionine has been
About half the patients with cystathionine b-synthase shown to have additional beneficial effects. In CblC of
deficiency respond to large oral doses of vitamin B6 infantile onset, hydroxycobalamin cannot always ful-
(pyridoxine). After a variable period, up to a few ly reverse the clinical and biochemical abnormalities.
weeks, homocysteine levels become normal, hyper- Betaine can be used as additive. A synergistic action
methioninemia decreases, and hypocysteinemia in- of betaine and hydroxycobalamin has been shown in
creases to normal values. In some patients the re- CblC. The long-term outcome of treatment depends
sponse is only partial and very large doses of vitamin to a great extent on the swiftness with which treatment
B6 are needed. If this is not enough to correct the bio- is started after onset of symptoms, or, in other words,
chemical abnormalities, a diet low in methionine and on the extent of irreversible neurological damage.
high in cystine should be initiated. A mildly methion- In cobalamin deficiency of all causes, hydroxy-
ine-restricted diet may be advisable for vitamin- cobalamin can be administered. In transcobalamin II
B6-responsive patients too. Extra supplementation of deficiency large doses of hydroxycobalamin are re-
betaine has been proposed to promote remethylation quired to correct cellular cobalamin deficiency. The
of homocysteine to methionine. Folate utilization effect of treatment depends on the presence and
may be increased in patients with cystathionine extent of irreversible neurological damage.
b-synthase deficiency, since it is required in methyla-
tion of homocysteine to methionine. Folate deficien-
cy can mask vitamin B6 responsiveness, and simulta- 45.6 Magnetic Resonance Imaging
neous administration of folate is necessary in some
patients to achieve biochemical normalization. As- The cerebral infarctions occurring in cystathionine
pirin has been used to reduce the tendency to devel- b-synthase deficiency, both lacunar and large infarc-
op thromboses, but has not proven to be beneficial. tions, can be visualized by MRI. The images as such
With early instigation of treatment, most of the clini- are not specific, only the age category of the patient is
cal manifestations of the disease can be prevented or unusual. Changes suggestive of single or multiple
delayed. small infarcts are most commonly observed. When
Also, in mild hyperhomocysteinemia due to het- the lesions are scattered over the brain with a pre-
erozygosity for cystathionine b-synthase deficiency, dilection for the periventricular area, the picture may
vitamin B6 combined with folate can normalize plas- resemble that of multiple sclerosis.
45.6 Magnetic Resonance Imaging 349

Fig. 45.2. A 43-year-old woman with


mild hyperhomocysteinemia who had
her first stroke with hemiparesis on
the left at the age of 22 years. At that
time a large medial cerebral artery
infarction was found. Recently she
developed signs of left hemisphere
dysfunction.The MR images show
an old, large middle cerebral artery
infarction with wallerian degeneration
of the connecting pyramidal tracts,
through the internal capsule into the
brain stem.There is a small lesion in
the paraventricular white matter on
the left. Courtesy of Dr. J.W. Snoek,
Department of Neurology, Martini
Hospital, Groningen, The Netherlands

Multiple systematic studies have documented an creased signal intensity is seen in the cerebral white
increased frequency of overt and silent infarcts of the matter, sparing the brain stem and cerebellar white
brain in patients with mild hyperhomocysteinemia matter, consistent with demyelination or insufficient
(Fig. 45.2). An increased incidence of spontaneous myelination (Figs. 45.5 and 45.7). The degree of signal
artery dissection has also been reported. One patient abnormality varies from slight to prominent. There
with mild hyperhomocysteinemia and severe cere- may be evidence of cerebral atrophy with increased
brovascular occlusive disease due to a moyamoya size of the lateral ventricles and subarachnoid spaces.
phenotype has been reported (Van Diemen-Steen- Improvement of the white matter signal and the cere-
voorde et al. 1990). bral atrophy is noted after therapy. Several patients
In subacute combined degeneration MRI reveals with 5,10-methylenetetrahydrofolate reductase defi-
spinal cord abnormalities with an increased signal in- ciency and disorders of intracellular cobalamin me-
tensity of the posterior part of the cervical spinal cord tabolism have been reported, who had internal hydro-
on T2-weighted sagittal and transverse images cephalus with marked enlargement of the lateral ven-
(Fig. 45.3). The abnormality in signal intensity is lo- tricles and macrocephaly, requiring shunt implanta-
cated in the dorsal and lateral columns. Contrast en- tion (Fig. 45.8).
hancement of the dorsal columns may be seen, but Delayed myelination and reversible cerebral atro-
does not occur in all patients. With treatment, partial phy has been documented in cobalamin-deficient in-
or complete resolution the lesion can follow. fants (Fig. 45.9). In our cobalamin-deficient patient,
MRI of cerebral white matter abnormalities has aged 19 months, a severe cerebral hemispheric white
mainly been documented in patients with 5,10-meth- matter abnormality was found, whereas the corpus
ylenetetrahydrofolate reductase deficiency (Figs. 45.4 callosum, internal capsule, and brain stem were
and 45.5) and disorders of intracellular cobalamin spared (Fig. 45.9). The white matter abnormalities
metabolism (Figs. 45.6–45.8). In untreated infants were most severe in the parietal area, extending
within the first year of life, MRI shows a delay in through the corona radiata into the arcuate fibers. In
myelination with improvement after therapy (Figs. addition, the hemispheric white matter had too high
45.4 and 45.6). In older untreated patients, an in- a signal intensity on T2-weighted images with a
350 Chapter 45 Hyperhomocysteinemias

Fig. 45.3. Spinal images of a cobalamin-deficient patient


with signs of subacute combined degeneration of the spinal
cord. Note the selective involvement of the dorsal columns
at the cervical level

patchy distribution, most pronounced in the subcor- into the U fibers (Fig. 45.11), sometimes with some
tical white matter, probably related to delayed and swelling leading to broadening of gyri. There may be
disturbed myelination. The images also showed cere- signal abnormalities in the brain stem and middle
bral atrophy in the presence of clinical microcephaly. cerebellar peduncles as well (Fig. 45.11). Small lesions
With cobalamin treatment, the white matter im- in the basal ganglia have been reported. The abnor-
proved and the atrophy resolved with return of the malities resolve partially with treatment (Figs. 45.10
head circumference to the normal centiles (Fig. 45.9). and 45.11).
In older cobalamin-deficient patients, white matter Finding evidence of combined degeneration of
abnormalities of variable extent have been described. dorsal and lateral columns is highly suggestive of one
These abnormalities are bilateral but not necessarily of the hyperhomocysteinemic disorders and should
symmetrical. The white matter changes are most of- direct the diagnostic work-up in the right direction.
ten located in the periventricular white matter as a However, both the vascular abnormalities and the
confluent rim of signal change with additionally white matter abnormalities described above are non-
smaller lesions in the deep white matter (Fig. 45.10). specific. Metabolic screening of urine is most helpful
In some patients, larger areas of white matter are in achieving the correct diagnosis.
abnormal, extending from the periventricular region
45.6 Magnetic Resonance Imaging 351

Fig. 45.4. Girl with methylenetetrahydrofolate reductase defi- The second MRI series (second row) was obtained at 22 months
ciency. The images in the first row were obtained at the age of and the third MRI was obtained at 4 years (third row). A de-
10 months.They show marked cerebral atrophy with widening crease in cerebral atrophy and a progress of myelination are
of the lateral ventricles and subarachnoid spaces. In addition, noted on follow-up, although the lateral ventricles and sub-
myelination is delayed. The frontal and parietal white matter arachnoid spaces remain dilated and myelination incomplete.
has a higher signal on T2-weighted images than expected for There remain areas in the subcortical white matter with evi-
delayed myelination only. Treatment with betaine monohy- dently elevated signal on the T2-weighted images.From Engel-
drate led to marked clinical and biochemical improvement. brecht et al. (1997), with permission
352 Chapter 45 Hyperhomocysteinemias

Fig. 45.5. Boy with methylenetetrahydrofolate reductase de- The optic radiation and cerebellar white matter have a more
ficiency at the age of 5 years.The T2-weighted MR images show normal signal. Courtesy of Dr. Z. Patay, Department of Radiolo-
that the cerebral white matter has an elevated signal through- gy, King Faisal Specialist Hospital and Research Center, Riyadh,
out. The corpus callosum also has a slightly abnormal signal. Saudi Arabia

Fig. 45.6. Girl with CblC.The first MRI was obtained at the age is still incomplete.There are more prominent white matter ab-
of 2.5 months (first and second row); the second MRI at the age normalities in the periventricular region.Courtesy of Dr.C.Fon-
of 24 months (third and fourth row). On the first MRI the cere- da, Division of Pediatric Radiology, A. Meyer Children’s Hospi-
bral white matter is abnormal and slightly swollen. Treatment tal, Florence, Italy, and Dr. A. Rossi (Rossi et al. 2001b, with per-
with hydroxycobalamin led to clinical improvement. Follow- mission)
up MRI at 2 years reveals that myelination has progressed but
45.6 Magnetic Resonance Imaging 353

Fig. 45.6.
354 Chapter 45 Hyperhomocysteinemias

Fig. 45.7. A 6-year-old untreated male patient with CblC. cerebral atrophy with mild widening of the lateral ventricles
There are prominent abnormalities in the periventricular and and subarachnoid spaces. Courtesy of Dr. C. Fonda, Division of
deep white matter. The corpus callosum is also involved, but Pediatric Radiology,A.Meyer Children’s Hospital,Florence,Italy
the internal capsule and brain stem are spared. There is some

Fig. 45.8. Girl with CblC. The first MRI was obtained at 3
months (first and second row), the second at 4 months (third
and fourth row). Note the progressive hydrocephalus. The lat-
eral ventricles have increased in size and the cerebral mantle
has become thinner.The cerebral white matter has become in-
creasingly abnormal in signal intensity.Courtesy of Dr.S.Blaser,
Department of Diagnostic Imaging, and Dr. A. Feigenbaum,
Department of Clinical Genetics, Hospital for Sick Children,
Toronto, Canada
45.6 Magnetic Resonance Imaging 355

Fig. 45.8.
356 Chapter 45 Hyperhomocysteinemias

Fig. 45.9.
45.6 Magnetic Resonance Imaging 357

Fig. 45.9. Girl with cobalamin deficiency, 19 months old at the with enlargement of the ventricular system and subarachnoid
time of the first MRI (first and second row). She was breast-fed spaces. The second MRI was obtained after 6 months of treat-
and her mother, being vegan, had a cobalamin deficiency. ment (third and fourth row) and showed marked progress of
These T2-weighted MR images suggest a combination of de- myelination and improvement of the atrophy. The third MRI
layed and disturbed myelination of the cerebral hemispheres, was obtained at the age of 4 years (fifth row). It shows that
and demyelination and gliosis in the tracts under the pericen- myelination is still incomplete in the U fibers and temporal
tral cortex. Corpus callosum, internal capsule, and brain stem lobe.There are some remaining areas of abnormal signal in the
are spared. In addition there is prominent cerebral atrophy deep parietal white matter
358 Chapter 45 Hyperhomocysteinemias

Fig. 45.10. A 51-year-old female


patient with cobalamin deficiency.
The first MRI (first row) was obtained
before treatment and shows exten-
sive, patchy cerebral white matter
abnormalities with sparing of the U
fibers. Follow-up MR images 2 months
after installment of cobalamin supple-
mentation (second row) and 44
months after initiation of treatment
(third row) show a striking improve-
ment, especially on the third MRI.
From Stojsavljević et al. (1997),
with permission
45.6 Magnetic Resonance Imaging 359

Fig. 45.11. A 48-year-old woman with cobalamin deficiency the corpus callosum, posterior limb of the internal capsule,
(first row) with follow-up 6 months after initiation of cobal- and middle cerebellar peduncles. With treatment improve-
amin supplementation (second row). Note the diffuse cerebral ment occurs. From Moritaet al. (2003), with permission
white matter abnormalities before treatment, also involving
Chapter 46

Urea Cycle Defects

46.1 Clinical Features one or two days the child becomes lethargic and hy-
and Laboratory Investigations potonic. Vomiting, seizures, hypothermia, and hyper-
ventilation occur. Lethargy increases and coma fol-
There are five well-documented urea cycle defects lows. There are signs of elevated intracranial pressure
(Fig. 46.1): with a bulging fontanel and increasing head size. In
– Carbamyl phosphate synthetase deficiency (CPSD) most cases the disease progresses rapidly to death
– Ornithine transcarbamylase deficiency (OTCD) within a few days. Survivors almost always have
– Argininosuccinate synthetase deficiency (ASSD), severe neurological sequelae.
also called citrullinemia In the case of later onset, the disease is chronic and
– Argininosuccinate lyase deficiency (ASLD), also episodic. Occurrence of symptoms may be related to
called argininosuccinic aciduria protein intake, infections, trauma, surgery, or initia-
– Arginase deficiency, also called hyperargininemia tion of valproate treatment, but not infrequently an
episode occurs without any obvious cause. The
These disorders have an autosomal recessive mode of episodes are characterized by headache, lethargy, irri-
inheritance, with the exception of OTCD, which has tability, agitation, confusion, hallucinations, vomit-
an X-linked recessive inheritance. Clinical signs of ing, hypotonia, ataxia, dysarthria, or coma. The pre-
metabolic derangement may appear at any time from sentation may also be stroke-like with signs of dys-
early infancy to adulthood, but peak periods include function of one cerebral hemisphere, in particular
the neonatal period, change to a diet with high pro- hemiplegia. In patients with later onset of clinical
tein content (replacement of milk feeding by a diet symptoms the mortality rate is still high, and highest
with higher protein content, parenteral nutrition), during the initial presenting illness. In addition to the
and episodes of infectious diseases. Valproate may al- episodic worsenings, there are often chronic clinical
so induce an episode of metabolic derangement. signs, which may include learning problems or psy-
In the case of neonatal presentation, a normal ba- chomotor retardation of variable severity, behavioral
by is born after normal pregnancy and delivery. After problems, ataxia, seizures, and hepatomegaly. Some
patients voluntarily restrict their protein intake. Nor-
mal development and neurological function up to
adulthood have been reported in rare cases. In ASLD,
coarse and friable hair (trichorrhexis nodosa) is a
special characteristic.
The clinical features and course of disease are in-
distinguishable in CPSD, OTCD, ASSD, and ASLD. It is
only in hyperargininemia that the clinical manifesta-
tions differ. In the latter disease, the clinical symp-
toms are slowly progressive and include growth fail-
ure, psychomotor retardation, progressive spastic
tetraplegia (the legs being more severely involved
than the arms), tremor, ataxia, choreoathetosis,
epilepsy, and hyperactivity. In addition, episodes of
lethargy, vomiting, and coma may occur. There is
some variability in onset and rate of progression of
the disease. Life span is usually longer than in the oth-
er urea cycle defects.
Females heterozygous for OTCD are usually free of
symptoms, but approximately 10% become sympto-
matic and have a milder and more variable course of
Fig. 46.1. The urea cycle. CPS, carbamyl phosphate syn- disease than affected males. Symptoms are episodic
thetase; OTC, ornithine transcarbamylase; ASS, argininosucci- and include headaches, vomiting, irritability, bizarre
nate synthetase; ASL, argininosuccinate lyase behavior, lethargy, ataxia, tremors, seizures, and co-
46.3 Pathogenetic Considerations 361

ma. The presentation may also be stroke-like with microscope. Alzheimer type II astrocytes are mainly
recurrent episodes of hemiparesis. A high-protein present in cerebral cortex, basal nuclei, cerebellar cor-
diet, infection, surgery, and the postpartum state may tex and nuclei, and brain stem nuclei. The presence of
precipitate attacks. Deterioration following use of val- Alzheimer type II cells depends on the presence of
proate has been described repeatedly. hyperammonemia. In adequately treated patients
Laboratory investigations reveal hyperammone- with normal ammonia levels Alzheimer type II cells
mia in all urea cycle disorders, with the exception of are absent.
hyperargininemia, in which blood ammonium levels In neonates who die in the acute phase of metabol-
may be normal. Respiratory alkalosis is often present. ic decompensation, diffuse brain swelling is found.
Urinary orotic acid is increased because of the shunt- Alzheimer type II astrocytes are present in gray
ing of nitrogen waste from the urea cycle. In CPSD matter structures. There may be signs of acute neu-
and OTCD citrulline is decreased in plasma. In ASSD ronal injury. Myelination is normal for age. In some
plasma citrulline is elevated, whereas in ASLD argini- neonates a status spongiosus of cortex and white
nosuccinate is elevated and citrulline is moderately matter is observed.
increased. In these four disorders, plasma levels of In older infants, children, and adults, atrophy of the
glutamine and alanine are frequently raised, whereas brain is often found with widening of the lateral ven-
arginine and ornithine are decreased. In hyper- tricles and spaces between the sulci. The atrophy
argininemia, arginine is elevated in plasma. In urine, varies from mild to severe. Variable focal and multi-
elevation of arginine, lysine, cystine, ornithine, cit- focal corticosubcortical necrosis may be seen with a
rulline, glutamine, and orotic acid is found. A definite tendency to microcavitation. Acute lesions are
diagnosis can be established by enzyme assessment swollen; old lesions are atrophic with presence of ule-
in liver cells. In hyperargininemia and ASLD, enzyme gyria. On microscopic examination, cortical findings
assessment is also possible in erythrocytes; in ASSD vary from normal to pseudolaminar neuronal necro-
and ASLD, enzyme assessment is also possible in sis to complete depopulation of the cortex and diffuse
fibroblasts. DNA confirmation is possible for all urea gliosis. The cortical damage may have a spongiform
cycle defects. appearance. Neuronal loss, gliosis, and spongy
All five urea cycle defects can be diagnosed antena- changes may also be seen in basal nuclei, thalamus,
tally. The techniques available for prenatal diagnosis and brain stem. White matter changes are variable.
vary from measurement of abnormal metabolites in Myelination may be normal or mildly to severely
amniotic fluid, to DNA analysis in chorionic villi or delayed. Signs of active myelin breakdown may be
amniocytes, to measurement of enzyme activity in present, but are not seen in all cases. The white matter
cultured amniocytes or in utero liver biopsy samples. may show diffuse gliosis. The white matter changes
Protein loading, alanine loading, and allopurinol may be spongiform with presence of myelin splitting
challenge to induce orotic aciduria can be used for and vacuolation. In severe cases, there is diffuse loss
carrier detection in OTCD. However, a negative test of axons and myelin, and the white matter may be
does not rule out the carrier status. DNA techniques largely replaced by gliotic tissue.
can be used for carrier detection if the mutation in In female carriers of OTCD, neuropathological
the affected patient is known. findings are apparently mainly related to chronic
hyperammonemia, which leads predominantly to
neuronal damage. Variable, sometimes extreme cere-
46.2 Pathology bral atrophy is the predominant finding. The hemi-
spheric walls are thin and the lateral ventricles are
Neuropathological findings are variable and depend dilated. The cerebral cortex shows signs of neuronal
on the age of the patient and on the relative effects of loss and gliosis. There is also loss of neurons in the
present and past acute and chronic metabolic de- basal nuclei and thalamus. Alzheimer type II astro-
rangements. Neuropathological findings are similar cytes are present in the cerebral cortex, basal nuclei,
in the different urea cycle disorders. dentate nuclei, and brain stem nuclei. The white mat-
Actual high elevations of ammonium levels lead to ter may be rarefied and gliotic with a reduced number
brain swelling. On light and electron microscopy, as- of myelinated fibers.
trocyte swelling is found. Hyperammonemia induces
the so-called Alzheimer type II change in astrocytes.
This change consists of an increase in the number and 46.3 Pathogenetic Considerations
size of astrocytic nuclei, which may become nearly
twice their normal size. These nuclei are vesicular The urea cycle (Fig. 46.1) serves two purposes: it con-
with a prominent nuclear membrane and an optical- tains, in part, the biochemical reactions required for
ly empty nucleoplasm with sparse chromatin parti- the de novo biosynthesis and degradation of arginine,
cles. The cytoplasm is not discernible with the light and it incorporates the surplus of nitrogen into urea,
362 Chapter 46 Urea Cycle Defects

which serves as a waste nitrogen product. The en- (NMDA) receptors. Moderate elevations of CSF levels
zymes involved in urea synthesis are partly located of quinolinic acid have been found in patients with a
within the mitochondria (carbamyl phosphate syn- urea cycle defect.
thetase, ornithine transcarbamylase), whereas the Unlike patients with liver failure, in whom ammo-
other enzymes are located within the cytosol (ar- nia is only one of several toxins, ammonia appears to
gininosuccinate synthetase, argininosuccinate lyase, be the only cause of the acute encephalopathy seen in
arginase). Carbamyl phosphate synthetase requires patients with urea cycle defects, except for those with
N-acetylglutamate for full activity. Most urea cycle hyperargininemia, in whom an increase in arginine
metabolic capacity resides in the liver. The gene loca- may also play a role. In hyperammonemia due to
tions of the urea cycle enzymes have been identified liver failure, MRI of the brain shows, as expected in
and the genes have been characterized. The gene for generalized disorders, a symmetrical pattern. There
carbamyl phosphate synthetase is located on chromo- are changes in signal intensity in the basal nuclei, in
some 2q35; the gene for ornithine transcarbamylase particular due to T1 shortening. In contrast, brain
is located on chromosome Xp21.1; the gene for pathology in urea cycle disorders is often character-
argininosuccinate synthetase is located on chromo- ized by focal, usually asymmetrical lesions. It remains
some 9q34; the gene for argininosuccinate lyase is to be explained why cerebral abnormalities related to
located on chromosome 7cen-q11.2; and the gene for hyperammonemia in urea cycle disorders are so dif-
arginase is located on chromosome 6q23. ferent from those observed in liver failure, and why
A urea cycle defect has two consequences: arginine the lesions tend to be asymmetrical.
becomes an essential amino acid (except in hyper- In all urea cycle disorders apart from hyper-
argininemia), and nitrogen accumulates in a variety argininemia, arginine is deficient unless externally
of molecules, in particular ammonia. Ammonia is supplied. Chronic arginine deficiency is character-
highly toxic to the brain where it interferes with ener- ized by dermatological features with erythematous
gy production and normal metabolism of neuro- scaling. A dramatic improvement of this cutaneous
transmitters. Ammonia influences the glutamine– eruption occurs with dietary arginine supplementa-
glutamate–GABA balance. Glutamate is the most im- tion. High levels of citrulline or argininosuccinate are
portant excitatory neurotransmitter, whereas GABA probably not toxic. The similarity in presentation
(g-aminobutyric acid), formed by decarboxylation among the different urea cycle defects is related to
from glutamate, is the most important inhibitory neu- hyperammonemia. The variability in clinical severity
rotransmitter. In the presynaptic neuron, glutamate is is probably related to differences in mutations (and
formed from glutamine by glutaminase. After release differences in levels of residual enzyme activity), dif-
by the presynaptic neuron, glutamate is taken up by ferences in other genetic factors and environmental
the astrocyte, in which it is processed by glutamine factors. In females carrying an OTC mutant allele on
synthetase into glutamine. Glutamine is transported one chromosome, variability in expression is related
to the presynaptic neuron, where glutaminase cat- to the proportion of hepatocytes in which the normal
alyzes the formation of glutamate available for neuro- or mutant allele is active (lyonization).
transmission. Hyperammonemia has a great impact Hyperargininemia has a clinical picture that is
on this cycle by stimulating synthesis of glutamine partially different from that of the other urea cycle
from ammonia and glutamate with consumption of defects. The progressive spasticity that dominates the
ATP. The disturbance of the balance between excitato- clinical picture in hyperargininemia is not a feature
ry and inhibitory neurotransmitters may contribute of the other urea cycle defects. It has been proposed
to the cerebral dysfunction. Glutamine synthetase is that arginine and its metabolites, the guanidine com-
mainly located in astrocytes, and high plasma levels pounds, are responsible.
of ammonia lead to accumulation of glutamine with-
in astrocytes. During hyperammonemia, the concen-
tration of glutamine in the brain becomes highly ele- 46.4 Therapy
vated. It has been proposed that the consequent
osmotic effect causes astrocytes to swell, with sub- Therapeutic strategies in urea cycle defects aim at
sequent cerebral edema. reduction of protein intake, utilization of alternative
The excitotoxin quinolinic acid has been proposed pathways of nitrogen excretion, and replacement of
to explain aspects of neuronal injury. Quinolinic acid deficient nutrients.
accumulates under hyperammonemic conditions and Emergency treatment consists of stopping all pro-
derives from tryptophan metabolism. Under such tein intake, starting high energy intake to prevent
conditions there is increased transport of tryptophan catabolic situations with breakdown of endogenous
across the blood–brain barrier. Tryptophan oxidation protein, and measures that lead to augmented nitro-
leads to the formation of quinolinic acid, which gen disposal. Sodium benzoate, sodium phenyl bu-
acts as an excitotoxin at the N-methyl-D-aspartate tyrate, or sodium phenyl acetate, supplied orally or
46.5 Magnetic Resonance Imaging 363

intravenously, can be used as substrates for an alter- 46.5 Magnetic Resonance Imaging
nate route of nitrogen disposal. Lactulose binds am-
monia in the intestinal tract and can increase ammo- In urea cycle defects, cerebral abnormalities change
nia disposal in feces. Arginine should be supplement- depending on the stage of disease. In acute episodes
ed in all urea cycle defects except hyperargininemia. of metabolic derangement, lesions appear, which may
Hemodialysis can be used in life-threatening situa- improve under treatment but leave traces. Chronic
tions. Of course, the conditions initiating the deterio- hyperammonemia also has deleterious effects. MRI
ration (infections, insufficient intake) and complica- has the advantage over neuropathological examina-
tions (seizures) have to be treated too. tions of being able to depict the dynamics of cerebral
Chronic treatment of urea cycle defects consists of lesions in urea cycle disorders.
a protein-restricted diet, sufficient caloric intake to In neonates, neuroimaging shows severe brain
avoid catabolic situations, and supplementation of swelling. On CT diffuse cerebral hypodensity with
absent substances. Protein-restricted diets have to be loss of contrast between cortex and white matter may
supplemented with essential amino acid mixtures to be seen. MRI shows diffuse cerebral edema (Fig. 46.2)
avoid deficiencies. In all urea cycle disorders apart and may demonstrate involvement of the basal gan-
from hyperargininemia, arginine cannot be synthe- glia with a high signal in the caudate nucleus, puta-
sized endogenously; it has become an essential amino men, and/or globus pallidus on T2-weighted images
acid and has to be supplied. If an essential amino acid (Fig. 46.2) and a high signal in the globus pallidus and
is lacking, protein breakdown cannot be remedied. to a lesser extent the putamen on T1-weighted images.
Long-term treatment that restricts intake of high- The deep sulci of the insular and perirolandic region
protein food, including milk and meat, leads to defi- may also display T1 shortening. MR spectroscopy may
ciency of minerals, trace elements, and vitamins, contribute by showing highly elevated glutamine lev-
which also have to be supplemented. Infections and els. If the patient survives, diffuse brain atrophy fol-
insufficient caloric intake due to anorexia carry the lows. In some cases, focal or diffuse gross cystic de-
risk of triggering an episode of severe hyperammone- generation of the cerebral hemispheres is seen on fol-
mia. Therefore, such conditions have to be treated low-up. The basal ganglia are often prominently in-
vigorously. The use of valproic acid as an antiepilep- volved as well, but the thalamus, brain stem, and
tic drug should be avoided.Valproic acid may acceler- cerebellum tend to be relatively spared.
ate the appearance of hyperammonemia. In acute metabolic derangement in older infants
The overall long-term prognosis has improved and children, small and large areas of abnormal sig-
with treatment, in particular in patients presenting nal intensity are seen in the brain, most often involv-
after the neonatal period. However, a high percentage ing both cortex and underlying white matter, giving
of treated patients are still mentally retarded. The them an infarct-like aspect (Figs. 46.4 and 46.5).
mortality rate in the group with neonatal presenta- Sometimes, the signal abnormalities involve only or
tion is still very high, and most (or all) surviving mainly the cortex (Fig. 46.3). The acute lesions are
patients are severely handicapped. moderately swollen. Often multiple lesions are pre-
Enzyme replacement therapy through liver trans- sent. The distribution of the lesions is as a rule asym-
plantation has been attempted, and it has been shown metrical or even unilateral (Figs. 46.4 and 46.5). In
to be effective in correcting the metabolic defect. some cases one hemisphere is totally involved. How-
After liver transplantation no further brain damage is ever, in other patients almost the entire brain is affect-
to be expected, but significant recovery of prior brain ed (Fig. 46.3). A combination of high plasma ammo-
damage does not occur. Therefore, liver transplanta- nia levels and an MRI picture with one or more large,
tion may be an excellent treatment for patients with- moderately swollen areas involving cortex and white
out major brain injury. However, in view of the poten- matter is highly suggestive of a urea cycle defect. This
tial morbidity and mortality associated with liver pattern can be found in all urea cycle defects when an
transplantation, the present experience is still too episode of acute metabolic derangement is present,
limited to allow a balanced opinion. Genetic correc- including those in female carriers of OTCD (Fig. 46.4)
tion of the patient’s liver cells is a promising future and patients with arginase deficiency (Fig. 46.5). In
option. the chronic stage, swelling resolves and focal areas of
atrophy and patchy signal changes of cortex and
white matter remain (Fig. 46.6). Sometimes, the le-
sions are restricted to the white matter.
In chronic hyperammonemia, defective myelina-
tion and progressive cerebral atrophy are seen. Some
patients have some nonspecific focal white matter
abnormalities.
364 Chapter 46 Urea Cycle Defects

Fig. 46.2. MRI series of a baby boy, 6 days old, with ASLD and particular, severe swelling of the brain stem. Note the signal
extreme hyperammonemia. The T1-weighted sagittal image abnormalities in the putamen and caudate nucleus. MR spec-
shows generalized edema and tonsillar herniation. The T2- tra of this patient are shown in Chap. 108
weighted transverse images show generalized edema with, in
46.5 Magnetic Resonance Imaging 365

Fig. 46.3. A 2-year-old girl, a carrier of OTCD, with an acute cerebral cortex and subcortical white matter. Courtesy of Dr. S.
episode of neurological deterioration and coma. The T2- Blaser, Department of Diagnostic Imaging, Hospital for Sick
weighted transverse images show diffuse involvement of the Children, Toronto, Canada
366 Chapter 46 Urea Cycle Defects

Fig. 46.4. A 2-year-old girl, a carrier of OTCD, with an acute smaller area of abnormal signal in the cortex and subcortical
episode of neurological deterioration and hemiplegia. The T2- white matter is seen in the right frontal region. Courtesy of Dr.
weighted images show an extensive area of high signal inten- S. Blaser, Department of Diagnostic Imaging, Hospital for Sick
sity and swelling in the left frontal and parietal white matter Children, Toronto, Canada
and cortex with blurring of the corticomedullary junction. A
46.5 Magnetic Resonance Imaging 367

Fig. 46.5. A boy,6 years of age,with hyperargininemia.The MR ally in the frontal lobes, accentuated on the left side, with in-
images were made during an episode of acute metabolic de- volvement of both gray and white matter, blurring the
compensation following protein-rich gavage feeding. The T2- gray–white matter junction. The corpus callosum is not
weighted transverse images show the signal changes bilater- involved, nor are the basal ganglia
368 Chapter 46 Urea Cycle Defects

Fig. 46.6. The same boy as in Fig. 46.5, now 2 years later. The acute abnormalities have disappeared. Note the atrophy, most
marked in the left frontal area
Chapter 47

Serine Synthesis Defect Caused


by 3-Phosphoglycerate Dehydrogenase Deficiency

47.1 Clinical Features L-serine has a central role as a precursor for sulfur
and Laboratory Investigations amino acids. It is a major source of one-carbon
groups, providing formyl groups for purine synthesis
3-Phosphoglycerate dehydrogenase deficiency is a and methyl groups for pyrimidine synthesis, the
rare neurological disorder with a prenatal onset. All remethylation of homocysteine, and many other
patients known so far have presented with congenital methylation reactions necessary in cellular metabo-
microcephaly. Some have contractures at birth. Dur- lism. It is a precursor for the synthesis of phosphoglyc-
ing the first months of life a severe psychomotor erides and complex macromolecules such as sphin-
retardation becomes apparent. Between 2 and 14 golipids and glycolipids. Serine can be converted to
months epilepsy starts, which is difficult to control glycine, which has important neurotransmitter func-
with antiepileptic medication. Different types of tions as an NMDA receptor agonist. The conversion of
seizures may be seen, including West syndrome and L-serine to glycine is accompanied by the conversion
tonic, atonic, and myoclonic seizures. The patients de- of tetrahydrofolate to 5,10-methylenetetrahydrofolate,
velop spastic tetraparesis. Other features that may be which is subsequently reduced to 5-methyltetrahydro-
seen are nystagmus, cataract, and hypogonadism. folate. The latter is a methyl donor for the remethyla-
Low levels of L-serine and glycine are found in the tion of homocysteine to methionine. L-serine can also
CSF and plasma in the fasting state. The levels are rel- be converted to D-serine, and D-serine is an even more
atively lower in CSF than in plasma. Urinary amino potent NMDA receptor agonist than glycine. L-Serine
acids are not informative. Another finding is a low is utilized in gluconeogenesis, but its role here is quan-
level of 5-methyltetrahydrofolate in CSF without an titatively of minor importance.
elevated homocysteine concentration in plasma. It is likely that, as soon as the blood–brain barrier
Megaloblastic anemia and thrombocytopenia are is established, the CNS has to rely on its own synthe-
variably present. The diagnosis is confirmed by sis of L-serine. 3-Phosphoglycerate dehydrogenase is
demonstration of deficient 3-phosphoglycerate dehy- highly expressed in fetal tissues including the CNS,
drogenase activity in cultured fibroblasts. Prenatal especially in the ventricular and subventricular zone
diagnosis is possible by means of both enzyme as- of the neural tube. This is the zone where the prolifer-
sessment and DNA analysis. ation of neural cells takes place. It is likely that im-
pairment of neuronal proliferation is responsible for
the microcephaly observed in patients at birth. The
47.2 Pathology role of L-serine in the CNS is not restricted to provid-
ing nucleotide precursors needed for cell prolifera-
At present, no autopsy reports are available. tion. Serine has also a trophic effect on neurons and
stimulates dendritogenesis and axon length.
The hypomyelination observed on MRI in patients
47.3 Pathogenetic Considerations with 3-phosphoglycerate dehydrogenase deficiency
may be secondary to low concentrations of folate
Deficiency of 3-phosphoglycerate dehydrogenase is a metabolites. Hypomyelination has been described in
disease with an autosomal recessive mode of inheri- patients suffering from other inborn errors with a low
tance. The related gene, PHGDH, is located on chromo- CSF level of 5-methyltetrahydrofolate. Low levels of
some 1q12. Different mutations have been described. S-adenosylmethionine may be important in the white
L-Serine is derived from four possible sources: di- matter abnormalities observed in folate deficiency.
etary intake, biosynthesis from 3-phosphoglycerate,
biosynthesis from glycine, and degradation of protein
and phospholipids. It is likely that the predominant 47.4 Therapy
source of L-serine is different for different tissues and
different stages of development. 3-Phosphoglycerate Treatment with high doses of L-serine is usually very
dehydrogenase catalyzes the first step in the synthesis effective in controlling the seizures. Some patients
of L-serine from the glycolytic intermediate 3-phos- need additional treatment with glycine to stop the
phoglycerate. seizures. There is some developmental progress after
370 Chapter 47 Serine Synthesis Defect Caused by 3-Phosphoglycerate Dehydrogenase Deficiency

onset of treatment, but the children remain seriously 47.5 Magnetic Resonance Imaging
handicapped. The biochemical abnormalities nor-
malize during treatment. The problem with treating MRI in untreated patients shows a reduced volume of
children diagnosed some time after birth is that irre- the cerebral white matter with enlarged subarachnoid
versible brain damage is already present, which can- spaces (Fig. 47.1). The sulci almost touch the walls of
not be reversed by treatment. There is preliminary the lateral ventricles. The corpus callosum is thin and
evidence that treatment of mothers pregnant of an short (Fig. 47.2). The cerebral white matter is defi-
affected fetus with L-serine may prevent all or most of cient in myelination, also in older children.
the clinical problems.

Fig. 47.1. Female patient with 3-phosphoglycerate dehydro- white matter. Under treatment the myelin content and the
genase deficiency at the age of 12 months before the start of volume of the cerebral white matter increase, although they
treatment (first row), and at the ages of 18 (second row) and 22 do not become normal. Courtesy of Dr. M. Pineda, Department
months (third row), both after treatment started. At 12 months of Neuropediatrics, Hospital Sant Joan de Déu, Barcelona,
there is serious myelin deficiency and atrophy of the cerebral Spain
47.5 Magnetic Resonance Imaging 371

Fig. 47.2. Midsagittal image of two


brothers with 3-phosphoglycerate
dehydrogenase deficiency, showing
the short and thin corpus callosum

Once treatment has been started, the white matter


volume increases (Fig. 47.1). In some patients myeli-
nation progresses, but not in all. In the patients in
whom myelination progresses, it still remains defi-
cient and does not seem to reach completion. Longer
follow-up is needed to document the long-term
effects of treatment.
Chapter 48

Molybdenum Cofactor Deficiency


and Isolated Sulfite Oxidase Deficiency

48.1 Clinical Features Laboratory investigations in isolated sulfite oxi-


and Laboratory Investigations dase deficiency reveal elevated urinary excretion of
sulfite, thiosulfate, and taurine. Plasma and urinary
Molybdenum cofactor deficiency and sulfite oxidase amino acid profiles show the presence of S-sulfocys-
deficiency are autosomal recessive inborn errors of teine. Plasma cystine levels are decreased. Urinary ex-
metabolism with a similar clinical presentation. In cretion of xanthine and hypoxanthine are normal.
molybdenum cofactor deficiency, not only sulfite oxi- Sulfite oxidase activity is deficient in cultured fibro-
dase but also xanthine dehydrogenase and aldehyde blasts. In molybdenum cofactor deficiency, the same
oxidase are deficient. Molybdenum cofactor deficien- laboratory findings are present, with the exception of
cy occurs more frequently than isolated sulfite oxi- xanthine and hypoxanthine, which are elevated, and
dase deficiency. plasma and urinary uric acid, which are low to virtu-
Patients with molybdenum cofactor deficiency ally absent. DNA-based diagnosis is possible for both
usually present a few days after birth with feeding dif- disorders. Elevated urinary sulfite can be detected us-
ficulties, seizures, axial hypotonia, and limb hyperto- ing dipsticks, but these are not very reliable and may
nia. The seizures are usually tonic–clonic and difficult show false negative results, because sulfite sponta-
to control. In patients who survive the neonatal peri- neously oxidizes to sulfate on standing. Prenatal diag-
od, serious psychomotor retardation is seen. Many nosis is possible by enzyme assessment in chorionic
patients develop microcephaly. Lens dislocation de- villi or cultured amniotic cells. Prenatal diagnosis by
velops in patients who survive the neonatal period. DNA analysis is also an option.
Other ocular abnormalities include spherophakia,
iris coloboma, nystagmus, and enophthalmos. Cere-
bral blindness may occur.A few patients have a milder 48.2 Pathology
disease course, with onset after the neonatal period,
usually precipitated by an infection. The clinical The neuropathology of both isolated sulfite oxidase
symptoms include hypotonia, extrapyramidal move- deficiency and molybdenum cofactor deficiency is
ment abnormalities, and sometimes lens dislocation. characterized by gross cerebral atrophy with deep
Most patients die at an early age. Exceptional patients sulci and dilated ventricles. The cerebral hemispheres
have a still milder disease with a Marfan-like habitus, are usually affected by multicystic degeneration,
dislocated lenses, learning problems, occasional which involves the white matter and inner layer of the
stroke-like episodes with unilateral motor deficits, cortex. In some areas the cortex may show complete
and survival into the third decade. degeneration. The cavities are separated by thick glial
In patients with sulfite oxidase deficiency, the clin- scar tissue. The remaining white matter contains little
ical picture is on the whole more variable than in myelin and is markedly gliotic. The basal ganglia,
molybdenum cofactor deficiency. At the severe end of thalami, and cerebellum are atrophic and show neu-
the spectrum, patients display signs of serious en- ronal loss and gliosis on microscopy. The basal gan-
cephalopathy soon after birth, with seizures and spas- glia may also be partly cystic. Remaining white and
ticity and subsequently serious psychomotor retarda- gray matter structures may show diffuse spongiosis
tion, lens dislocation, spherophakia, and early death, and may contain areas of mineralization. The brain
similar to the clinical picture of molybdenum cofac- stem is atrophic and contains little myelin, but is bet-
tor deficiency. However, patients may also have a later ter preserved.
onset, most often between the ages of 6 months and
1.5 years, and lack suggestive symptoms such as ec-
topic lenses and seizures. The onset of the disease is 48.3 Pathogenetic Considerations
often precipitated by an infection, which is followed
by developmental regression, hypotonia, and dysto- Sulfite oxidase is encoded by the gene SUOX, located
nia or choreoathetosis. Microcephaly may develop. on chromosome 12q13.2–13.3. Molybdenum cofactor
An episodic course with stepwise deterioration has consists of a unique pterin, molybdopterin, and the
also been described. metal molybdenum. The cofactor is synthesized in
48.5 Magnetic Resonance Imaging 373

humans by a complex pathway that requires the prod- precursor sulfur amino acids may lead to some im-
ucts of at least four different genes: MOCS1, MOCS2, provement. Cysteamine may be beneficial in absorb-
MOCS3, and GEPH. Disease-causing mutations have ing excess sulfite.
been identified in three of these genes: MOCS1, Treatment of seizures is important. In particular
MOCS2, and GEPH. MOCS1 is located on chromo- vigabatrin has been successful in controlling the
some 6p21.3, MOCS2 on chromosome 5q11, and seizures.
GEPH on chromosome 14q24. MOCS1 and MOCS2
have a bicistronic architecture, which means that each
gene encodes two proteins in different open reading 48.5 Magnetic Resonance Imaging
frames. The gene products, MOCS1A and B and
MOCS2A and B, are expressed either from different Neuroimaging findings in patients with isolated sul-
mRNAs generated by alternative splicing or by inde- fite oxidase deficiency and molybdenum cofactor
pendent translation of a bicistronic mRNA. The deficiency are similar. CT scan of the brain has been
gephyrin protein is required during cofactor assem- reported to show diffuse edema in the neonatal peri-
bly for insertion of the molybdenum into molyb- od. Multicystic degeneration of the cerebral hemi-
dopterin. To date, no disease-causing mutations have spheres and calcium deposits have been documented
been found in MOCS3. by follow-up CT.
Molybdenum cofactor deficiency results in simul- In early postnatal presentation, MRI shows brain
taneous loss of all cofactor-dependent enzyme activ- swelling and extensive areas of abnormal signal, con-
ities, including sulfite oxidase, xanthine dehydroge- sistent with edema and “hypoxic” changes within the
nase, and aldehyde oxidase. Sulfite oxidase catalyzes cerebral cortex (Figs. 48.1 and 48.2). Follow-up MRI
the oxidation of sulfite into sulfate. Xanthine oxidase shows extensive cystic degeneration of the cerebral
catalyzes the decomposition of xanthine into uric hemispheres, with large and smaller cysts within the
acid. Aldehyde dehydrogenase catalyzes the forma- white matter and enlargement of the ventricles and
tion of xanthine from hypoxanthine. In addition, this subarachnoid spaces (Fig. 48.2). The corpus callosum
enzyme is thought to be part of a general detoxifica- is thin. The cerebellum and brain stem are small,
tion system. Combined deficiency leads to a pheno- probably due to both hypoplasia and atrophy. The
type that is clinically quite similar to that of isolated small cerebellum with enlarged pericerebellar spaces
sulfite oxidase deficiency. Therefore, it is likely that has been described as a Dandy–Walker variant, which
the neurological disease, characterized by a severe en- is not correct, as the rest of the Dandy–Walker config-
cephalopathy, results primarily from the sulfite oxi- uration is lacking. The basal ganglia and thalami are
dase deficiency. The pathological findings resemble atrophic and may contain cysts. On more prolonged
those seen in severe perinatal asphyxia. The patho- follow-up, the large cysts in the cerebral white matter
genesis of the disease is not understood. It is not clear may collapse and a seriously atrophic brain may re-
whether the damage arises from a toxic metabolite or main. Not much myelin is deposited in the highly
from the deficit of a reaction product. The accumulat- damaged, gliotic white matter. Because of the serious
ing sulfite may be toxic. However, its mode of action is brain atrophy, subdural fluid collections develop eas-
unclear, and it is also unknown why the brain is most ily.
seriously affected. The reaction of sulfite with disul- The above pattern is indistinguishable from that
fide bonds or with sulfhydryl groups is a general seen in multicystic encephalopathy after perinatal as-
process that would be expected to occur in all organs. phyxia, neonatal presentations of a urea cycle defect,
Ocular lens subluxation may be related to a disrup- and some early-onset and severe mitochondrial dis-
tion of cystine cross-linking by excess sulfite. orders. It is essentially the pattern that is expected to
develop in neonates after profound energy failure of
any origin. It seems that serious edema preceding the
48.4 Therapy multicystic degeneration is a common factor.
Some patients with milder forms of isolated sulfite
No effective treatment is available for the severe vari- oxidase deficiency or molybdenum cofactor deficien-
ants of either isolated sulfite oxidase deficiency or cy have a much better preserved brain. Bilateral
molybdenum cofactor deficiency. The cofactor is ex- lesions in the globus pallidus with preservation of
ceedingly unstable and direct cofactor replacement is the rest of the brain have been found repeatedly
not feasible. Various therapeutic trials, including oral (Fig. 48.3). In some patients more extensive abnor-
intake of molybdenum salts and low-sulfur amino malities are seen, with signal abnormality and
acid diet, combined with oral sulfate supplementa- swelling of the caudate nucleus, putamen, globus
tion, have failed. However, some measures may bene- pallidus, and cerebral peduncles in the acute stage
fit individual patients. In particular in patients with a (Fig. 48.4) and less prominent signal abnormalities
milder clinical picture, restriction of the intake of the and atrophy of these structures in the chronic stage.
374 Chapter 48 Molybdenum Cofactor Deficiency and Isolated Sulfite Oxidase Deficiency

Fig. 48.1. Baby boy, 10 days old, with molybdenum cofactor signal in parts on T1-weighted images, as seen in necrosis.
deficiency.The T2-weighted images show serious involvement Courtesy of Dr. S. Blaser, Department of Diagnostic Imaging,
of the cortex and basal ganglia. These structures have a high Hospital for Sick Children, Toronto, Canada

Selective involvement of the globus pallidus is rare


and has been observed in creatine synthesis defects,
succinic semialdehyde dehydrogenase deficiency,
mitochondrial defects, hyperbilirubinemia, and car-
bon monoxide poisoning. Lesions in the globus pal-
lidus are also relatively frequent in neurofibromatosis
type I.
48.5 Magnetic Resonance Imaging 375

Fig. 48.2. Baby girl with isolated sulfite oxidase deficiency. obtained at the age of 31 days and demonstrate cystic degen-
The T2-weighted images in the first row were obtained at the eration of the cerebral white matter. The basal ganglia and
age of 5 days and show serious abnormalities of the cerebral cerebral cortex also contain signal abnormalities. The brain
cortex, white matter, and basal ganglia. The brain is diffusely stem and cerebellum are preserved. From Dublin et al. (2002),
swollen. The posterior fossa structures have a more normal with permission
appearance. The images in the second and third rows were
376 Chapter 48 Molybdenum Cofactor Deficiency and Isolated Sulfite Oxidase Deficiency

Fig. 48.3. Two brothers with isolated sulfite oxidase deficien- right image represents the younger brother, also at the age of
cy. Both are relatively mildly affected. The left and middle 5 years. Both have isolated globus pallidus lesions
images represent the older brother at the age of 5 years. The

Fig. 48.4. Girl, 1 year and 9 months old, with molybdenum nigra, and dentate nucleus. From Hughes et al. (1998), with
cofactor deficiency. Note the signal abnormality and swelling permission
of the putamen, globus pallidus, caudate nucleus, substantia
Chapter 49

Galactosemia

49.1 Clinical Features verbal and performance IQ slowly decline, and a sub-
and Laboratory Investigations stantial number of patients have subnormal intelli-
gence. School achievements are often worse than
Three major types of galactosemia can be distin- those of healthy sibs. At the end of the first decade of
guished, based on three different enzyme deficien- life, many children develop tremor, resting and pos-
cies. The most commonly detected type, classical tural. Other neurological signs that may develop over
galactosemia or galactosemia type 1, is caused the years include a cerebellar ataxia with clumsiness,
by a deficiency of galactose-1-phosphate uridyltrans- intention tremor and dysdiadochokinesis, hyper-
ferase. Galactosemia type 2 is caused by galactokinase reflexia, apraxia, seizures, and choreoathetosis. Some
deficiency. Galactosemia type 3 is the result of a defi- children have microcephaly. Many children have
ciency of uridine diphosphate galactose-4-epimerase. speech abnormalities, varying from dysarthria, verbal
All three types have an autosomal recessive mode of dyspraxia, and dysgrammatism to stuttering. Delayed
inheritance. growth is especially seen in girls, but final height is
Galactosemia type 1 has an incidence of 1:30,000 to usually within normal limits. A high incidence of
1:60,000. Affected infants are normal at birth. Clinical ovarian failure with hypergonadotropic hypogo-
manifestations develop a few days after the baby has nadism has been documented in female patients. It
started to have milk feeds. Symptoms include a failure may be manifest as delayed puberty, primary or sec-
to thrive, refusal to feed, vomiting, diarrhea, and ondary amenorrhea, or oligomenorrhea. Amenor-
hypotonia. Signs of deranged liver function with rhea can also occur after pregnancy. Successful preg-
jaundice and hepatomegaly usually become apparent nancies in female patients are rare, but may occur. No
after the first week of life. Hemolysis occurs in some correlation between onset and strictness of diet and
patients, contributing to the jaundice. There is an development of late complications has ever been es-
enhanced susceptibility to infections, in particular tablished. A relationship between development of
Escherichia coli infections. Sepsis often develops cataract and dietary control is present.With introduc-
within the first 2 weeks of life. Ascites may occur and tion of tighter dietary control, lens opacities usually
is a serious prognostic sign with an associated mor- gradually resolve. The patients with milder variants of
tality of about 20%. Cataracts appear within days or galactosemia type 1 have a better outlook with nor-
weeks and become irreversible within a matter of mal physical, motor and mental development when
weeks. There may be signs of elevated intracranial treated.
pressure with lethargy and diffuse cerebral edema on In patients with signs of CNS problems, EEG often
neuroimaging. If milk is not withdrawn, neonatal shows nonspecific abnormalities. SSEPs show pro-
death may follow. longed central conduction times.
There are less severe variants of the disease. Clini- Galactosemia type 2 is much rarer than type 1, and
cal presentation may be later and less life-threaten- much milder. Cataracts are the only consistent mani-
ing. Some patients are seen later in the first year of life festation of the untreated disorder. They develop in-
because of retarded psychomotor development, sidiously within weeks of birth. In exceptional cases,
cataracts, and hepatomegaly. In rare cases, a child mild hepatomegaly, mental retardation, or pseudotu-
who is several years of age is presented with psy- mor cerebri have been described.
chomotor retardation and cataracts. These children Galactosemia type 3 exists in two forms. Infants
often have a history of reduced milk intake because of with the mild form appear healthy and remain so. The
recurrent vomiting after drinking milk. severe form is similar to galactosemia type 1. Presen-
If galactose is not eliminated from the diet, tation is neonatal with jaundice, vomiting, weight
cataracts, progressive liver failure, and mental defi- loss, hypotonia, and hepatomegaly. Despite treat-
ciency develop in patients surviving the neonatal pe- ment, motor and intellectual development is retarded
riod.A galactose-free diet causes a striking regression and neural deafness is present.
of all signs and symptoms. Nausea and vomiting Whenever the diagnosis galactosemia is consid-
cease, lethargy disappears, weight gain ensues, liver ered in neonates, it is essential to stop milk feeding
problems clear, and cataracts regress. However, the immediately. In galactosemia type 1, a positive reduc-
long-term outcome is less optimistic. Over the years, tion test in urine may be the first diagnostic lead.
378 Chapter 49 Galactosemia

Apart from galactosuria, there may be evidence of a 49.2 Pathology


renal tubular defect with some proteinuria, gluco-
suria, amino aciduria, phosphaturia, and renal tubu- Few neuropathological descriptions are present and
lar acidosis. Galactitol in urine is elevated. The inter- they only concern patients with classical galacto-
mittent nature of the galactosuria makes its detection semia (type 1). External examination often reveals
difficult, in particular when milk feeds are withheld the brain to be mildly atrophic. The most prominent
from very ill infants. The diagnosis is confirmed by findings concern the cerebral white matter and cere-
the finding of high levels of galactose-1-phosphate in bellar cortex. The cerebral white matter is diffusely
red blood cells and the demonstration of a deficiency gliotic. On myelin staining, patchy pallor is found, but
of galactose-1-phosphate uridyltransferase in red or no signs of active demyelination. The white matter
white blood cells. The assay on red blood cells may, of changes are most pronounced in the periventricular
course, be falsely negative after exchange blood trans- area. The white matter may be reduced in volume
fusion. When a child has received exchange transfu- with some enlargement of the lateral ventricles and
sion, assays in blood must be postponed for 3–4 subarachnoid spaces. The cerebral cortex may either
months. Enzyme activity can also be determined in be normal or exhibit some neuronal loss. Gliosis and
cultured skin fibroblasts. Patients with some residual pigmentary degeneration of the globus pallidus and
enzyme activity appear to follow milder clinical reticular zone of the substantia nigra have been de-
courses (mild variants) than patients with no demon- scribed. There is a loss of Purkinje cells within the
strable activity. cerebellar cortex.
In galactosemia type 2, elevated galactose is found
in blood and urine. Galactose-1-phosphate is not ele-
vated. Final diagnosis is established by demonstrating 49.3 Pathogenetic Considerations
of a deficiency of galactokinase in red blood cells or
fibroblasts. Galactose is metabolized in three sequential enzy-
In galactosemia type 3, elevated galactose is pre- matic steps. The first step comprises the phosphory-
sent in blood and urine. The diagnosis may be sus- lation of galactose by galactokinase to form galac-
pected when galactose-1-phosphate in erythrocytes tose-1-phosphate. The second step is mediated by
is elevated and the activity of galactose-1-phosphate galactose-1-phosphate uridyltransferase and results
uridyltransferase is normal. The diagnosis can in an exchange of galactose-1-phosphate for the
be confirmed by demonstrating a deficiency of glucose-1-phosphate moiety of uridine diphosphate
epimerase activity in red blood cells and leukocytes. glucose to form uridine diphosphate galactose and
In the mild form of galactosemia type 3, the enzyme free glucose-1-phosphate. In the third step, uridine
deficiency is limited to blood cells and normal activi- diphosphate galactose is transformed to uridine
ty is found in fibroblasts and liver cells. In the severe diphosphate glucose by the enzyme epimerase. In this
form a generalized deficiency of epimerase is present. step galactose is converted to glucose. This step can be
In several countries, mass newborn screening is reversed, leading to endogenous synthesis of galac-
performed. Diagnostic difficulties arise in cases of tose from glucose.
partial galactose-1-phosphate uridyltransferase defi- The most common type of galactosemia, type 1,
ciency. A mutation of the transferase gene, called the is related to a deficiency of galactose-1-phosphate
Duarte variant, causes diminished red cell transferase uridyltransferase. The gene encoding this enzyme,
activity but usually no clinical disorder. The allelic GALT, is located on chromosome 9p13. A large num-
frequency of the Duarte variant is high, and com- ber of mutations have been described. Furthermore,
pound heterozygotes with one Duarte allele and one gene polymorphism has been found, leading to en-
classical galactosemia allele constitute the most com- zyme polymorphism. The most common variants are
mon biochemical phenotype detected by screening the Duarte variants (D1 and D2), but other benign
newborn infants. This condition is usually benign, variants have also been described. There is evidence
but neonates may have symptoms of galactose toxici- that the molecular heterogeneity forms the explana-
ty. Several other benign mutations of the galactose-1- tion for the variable clinical outcome.
phosphate uridyltransferase gene have been de- The human galactokinase gene, GALK, is located
scribed. on chromosome 17p24. Patients with galactokinase
Prenatal diagnosis can be performed by analysis of deficiency have mutations in this gene. A second
galactitol in amniotic fluid, by enzyme analysis in GALK cDNA has been found on chromosome 15. It is
chorionic villus cells or amniotic fluid cells, and by clear that only the first gene on chromosome 17p24
DNA techniques. Prenatal diagnosis, however, is only produces significant amounts of the enzyme; the role
rarely performed with a view to terminating the of the second gene is not known.
affected pregnancy.
49.4 Therapy 379

The epimerase gene, GALE, is located on chromo- phosphate. Aldose reductase is activated by high lev-
some 1p36. Different mutations have been detected in els of galactose, and activation of this pathway leads
epimerase deficient patients. to myo-inositol depletion. Myo-inositol is an intracel-
The pathogenetic mechanisms of tissue damage in lular osmolyte. It is essential for the synthesis of inos-
galactosemia are understood to only a limited extent. itol phospholipids, which are membrane compo-
The mechanisms may be organ-specific. nents. An important surface signal transduction sys-
Galactitol toxicity is probably responsible for the tem relies on induction of hydrolysis of plasma mem-
development of cataracts. In the presence of high brane phosphoinositides to generate intracellular
galactose levels, the enzyme aldose reductase irre- second messenger molecules, which induce the cell to
versibly reduces galactose to galactitol. Very little respond to various extracellular agonists, such as
galactitol leaves the intracellular compartment and neurotransmitters and peptide hormones. Insuffi-
no further metabolism is possible beyond the galacti- cient levels of these membrane phosphoinositides
tol step. The intracellular concentration of galactitol may lead to impaired cell function.
increases and alters the cell osmotic environment.
Water is drawn into the cell and results in lens
swelling, with denaturation and precipitation of pro- 49.4 Therapy
teins and disruption of lens architecture. Aldose re-
ductase activity is also present in the brain, where The predominant source of galactose is lactose, pre-
galactitol may accumulate intracellularly and con- sent in mammalian milk or artificial milk formulae
tribute to cellular swelling and death, especially in the and milk products. Lactose is a disaccharide. Prior to
early, untreated stage. absorption from the intestine it is hydrolyzed to its
There is evidence that galactose-1-phosphate is constituent monosaccharides, glucose and galactose.
responsible for many of the acute toxic effects in Galactose is subsequently converted to glucose. Pa-
galactosemia, including liver, kidney, and brain tients with galactosemia type 1 are treated with a
damage. Galactose-1-phosphate may contribute to galactose-free diet, causing all symptoms of acute
toxicity by reducing energy availability through inhi- galactose toxicity to disappear, including vomiting,
bition of several enzymes involved in glucose metab- diarrhea, jaundice, hepatomegaly, cerebral edema,
olism. and cataracts. Levels of galactose-1-phosphate in red
The observations that a galactose-free diet is im- blood cells and galactitol in urine drop. Treatment
possible and that galactose and galactose-1-phos- monitoring by assessment of galactose-1-phosphate
phate can also be made from endogenous sources in red blood cells or galactitol in urine is of limited
make it probable that galactose-1-phosphate and value, since even with strict adherence to diet the val-
galactitol also contribute to the occurrence of late ues remain elevated. After increased galactose intake,
complications. it takes several weeks before galactitol in urine rises
Galactose is part of many complex glycoproteins and even longer before galactose-1-phosphate rises.
and glycolipids. Defects in galactosylation of proteins On the whole, changes in galactose intake have rela-
and lipids has been proposed as pathogenetic mecha- tively little effect on metabolite levels. Perfect treat-
nisms for organ damage in galactosemia, by analogy ment is not attainable, probably because food prod-
to the congenital defects in glycosylation. In particu- ucts may still contain some galactose and, possibly
lar, the late complications of classical galactosemia more importantly, because of endogenous produc-
have been ascribed to depletion of uridine diphos- tion of galactose from glucose. There is no evidence
phate galactose (UDP-galactose), one of the products that the occurrence of late complications depends on
of galactose-1-phosphate uridyltransferase activity. strictness of compliance with the diet, which is also
UDP-galactose is the donor of the galactosyl moiety an argument in favor of endogenous galactose pro-
in the biosynthesis of glycoproteins and glycolipids, duction. In addition, an abnormal prenatal intrauter-
including gangliosides, cerebroside, and sulfatide. A ine biochemical environment may be responsible for
deficiency of UPD-galactose could limit the synthesis part of the cerebral and ovary dysfunction, which
of these macromolecules, which include major myelin cannot be reversed by postnatal treatment.
lipids. UDP-galactose is also needed for the synthesis There is no evidence that dietary treatment of
of ovarian membrane glycoproteins and glycolipids. heterozygous mothers pregnant with a galactosemic
The theory is, however, difficult to reconcile with the fetus has beneficial effects. Lactation in galactosemic
metabolic pathways, which enable synthesis of UDP- mothers may cause a significant rise in galactose-1-
galactose from glucose-1-phosphate by pyrophos- phosphate in red blood cells and galactitol in urine.
phorylase and epimerase. Considering these biochemical signs of self-intoxica-
Depletion of myo-inositol and inositol phospho- tion, breast feeding is discouraged. Hormonal re-
lipids has been found in the brain of galactosemia placement therapy is given to female patients with
patients. Myo-inositol is synthesized from glucose-6- ovary dysfunction.
380 Chapter 49 Galactosemia

Treatment of galactosemia type 2 may be limited to The first abnormality noted by MRI is that after
the elimination of milk from the diet. It is not neces- normal initial myelination the directly subcortical
sary to apply a strict galactose-restricted diet. white matter does not become as hypointense on T2-
Treatment of the mild form of galactosemia type 3 weighted images as in normal children older than
is not necessary. Treatment of the severe form is sim- 1 year, indicating hypomyelination (Figs. 49.1 and
ilar to treatment of galactosemia type 1, but more dif- 49.2). This can still be seen in teenage and adult pa-
ficult. Patients with galactosemia type 3 are unable to tients (Fig. 49.3). In addition, multiple foci of high sig-
synthesize galactose from glucose and are therefore nal intensity are seen in many patients, spread over
dependent upon exogenous sources for galactose. the hemispheric white matter (Figs. 49.1 and 49.3).
When too small amounts of galactose are ingested, The foci are bilateral and more or less symmetrical in
synthesis of galactosylated compounds, such as galac- distribution, although not perfectly symmetrical. The
toproteins and galactolipids (present in myelin white matter abnormalities are most pronounced in
lipids), is impaired. Unfortunately, there is no easily the periventricular area, round the frontal and occip-
available chemical parameter for monitoring how ital horns. In many patients the ventricular system is
much galactose should be used. slightly enlarged. Cerebellar foliae are prominently
visible in some patients. In patients with mild vari-
ants of galactosemia type 1 no abnormalities are not-
49.5 Magnetic Resonance Imaging ed on MRI.
In the early, untreated stages of the disease, proton
Neuroimaging reports all relate to galactosemia type MRS shows the presence of highly elevated peaks at
1. CT scan of the brain has been reported to show 3.67 and 3.74 ppm, representing galactitol. In treated
signs of generalized atrophy in patients with neuro- patients brain galactitol is below the level of detection
logical abnormalities. The ventricular system may be for in vivo proton MRS. Under treatment, myo-inosi-
enlarged and the cerebellum atrophic. tol levels have been found to be normal.
49.5 Magnetic Resonance Imaging 381

Fig. 49.1. A 4-year-old boy with galactosemia type 1. Note tricular region, most marked posteriorly. The FLAIR images
the seriously delayed myelination.In addition, the T2-weighted (third row) confirm the white matter abnormalities
images show foci of elevated signal intensity in the periven-
382 Chapter 49 Galactosemia

Fig. 49.2. A 6-year-old girl with galactosemia type 1. Again the process of myelination is seriously delayed. The anterior tempo-
ral white matter displays an abnormal signal

Fig. 49.3. Girl, 14 years of age, with galactosemia type 1. The T2-weighted images show a combination of hypomyelination and
patchy high signal intensity white matter abnormalities, consistent with gliosis
Chapter 50

Sjögren–Larsson Syndrome

50.1 Clinical Features 50.2 Pathology


and Laboratory Investigations
The external appearance of the brain is normal. A
Sjögren–Larsson syndrome (SLS) is a rare disorder considerable deficit in myelin is seen in the periven-
with an autosomal recessive mode of inheritance. The tricular and deep white matter, the area involved ex-
three cardinal clinical signs are mental retardation, tending from frontal to occipital, sparing the U fibers.
congenital ichthyosis, and spastic diplegia or tetra- Ballooning of myelin sheaths is a notable feature
plegia. Most patients are born prematurely. The within the areas of myelin deficit. Lipid-laden
ichthyosis is usually present at birth and initial pre- macrophages and histiocytes are present. Small lipid
sentation may be as a collodion baby. The ichthyosis droplets are contained in the cytoplasm of microglia,
tends to worsen with time, and usually presents after scattered diffusely through the white matter. In the
infancy as a brownish verrucous, lichenified hyperk- area of myelin paucity, degeneration and loss of axis
eratosis, more pronounced on the lateral aspects of cylinders and astrocytosis are found. The corpus cal-
the trunk, neck, lower abdomen, and the main flex- losum is well myelinated. Within the brain stem the
ures, with marked pruritus. The face is usually spared. pyramidal tracts show a deficit in myelin and loss of
Palms and soles may show hyperkeratosis and axis cylinders. Myelin paucity is also demonstrated in
desquamation, but abnormalities are usually mild. some of the descending tracts of the spinal cord,
Hair and nails are normal. The spasticity usually be- including the lateral corticospinal tracts, but the as-
comes manifest between 4 and 30 months of age. cending tracts are normal. Cerebellar white matter is
Many of the patients are never able to walk without normal.
assistance, and a considerable number are wheel- Microscopy of the cerebral cortex and central
chair-dependent. Those able to walk have a typical brain nuclei reveals in all areas an increase in astro-
spastic gait. Tendon reflexes are high, and bilateral cytes and accumulation of sudanophilic droplets of
Babinski signs are often present. Mental deficiency fat. Within the cerebellar cortex some loss of Purkin-
varies from moderate to severe. Speech defect is usu- je cells is seen. Some neuronal cell loss may also be
ally present and can be attributed to a combination of seen in the cerebral cortex, in particular Betz cells in
mental deficiency and pseudobulbar palsy. Occasion- the motor cortex, and in the basal ganglia, in particu-
ally epilepsy is present. SLS patients have ocular ab- lar in putamen and substantia nigra.
normalities, consisting of bilateral glistening yellow-
white dots in the macular region of the retina. They
appear during the first 2 years of life and become 50.3 Pathogenetic Considerations
more numerous over time. Photophobia and subnor-
mal vision are frequently present. Short stature and The basic defect of SLS is an impairment in fatty alco-
thoracolumbar kyphoscoliosis may be present. The hol oxidation due to deficient activity of fatty alcohol-
disease is not clearly progressive; the clinical signs are NAD+ oxidoreductase. This enzyme consists of two
more or less stable over the years; most patients sur- proteins that sequentially catalyze the oxidation of
vive into adulthood. There is no correlation between fatty alcohol to fatty aldehyde and fatty acid, reactions
the severity of the neurological symptoms and the that are catalyzed by fatty alcohol dehydrogenase and
ichthyosis. fatty aldehyde dehydrogenase, respectively. SLS pa-
The clinical diagnosis of SLS is confirmed by find- tients are specifically deficient in the fatty aldehyde
ing deficient fatty alcohol-NAD+ oxidoreductase ac- dehydrogenase component. The gene coding for the
tivity in leukocytes and cultured fibroblasts and DNA enzyme is called FALDH or, more recently, ALDH3A2
analysis. Carrier detection can be performed by en- and is located on chromosome 17p11.2. The enzyme
zyme assessment in cultured fibroblasts or DNA has a microsomal location. Fatty aldehyde dehydroge-
analysis. Prenatal diagnosis is possible with the help nase is necessary for the oxidation of fatty aldehydes
of enzyme assessment or DNA techniques in families derived from metabolism of fatty alcohols, phytanic
with known mutations. acid, ether glycerophospholipids, and leukotriene B4.
384 Chapter 50 Sjögren–Larsson Syndrome

Fatty aldehydes and fatty alcohols are aliphatic leukotriene B4 are probably responsible for the pruri-
lipids that exist as free molecules or as components of tus. There is evidence that leukotriene B4 probably
other lipids. As a consequence of the enzyme defi- plays a role during labor. Placental leukotriene B4 pro-
ciency, fatty alcohols accumulate in SLS patients (oc- duction is low during the third trimester of pregnan-
tadecanol and hexadecanol). The levels of free fatty cy and increases in spontaneous labor. Elevated levels
alcohols are increased in cultured fibroblasts and produced by the fetus may lead to preterm birth.
plasma.Although the primary enzyme deficiency is at
the aldehyde step of the fatty alcohol oxidation, free
fatty aldehydes are not increased in blood or cultured 50.4 Therapy
fibroblasts. This suggests that the defect in fatty alde-
hyde dehydrogenase results in a concomitant block- Several studies have been performed in which dietet-
age of the fatty alcohol dehydrogenase within the ic therapy was aimed at the primary biochemical
enzyme complex, which limits the production of alco- defect, trying to block the exogenous admission of
hol-derived fatty aldehyde. Another explanation toxic substances, the fatty alcohols. Unfortunately
would be that the aldehyde substrates are diverted these studies had little if any success at all. Leuko-
into other metabolites. triene B4 is synthesized from arachidonic acid via the
The symptoms of SLS patients probably arise from lipoxygenase pathway. The drug zileuton, used in
membrane alterations resulting from accumulation asthma, inhibits 5-lipoxygenase and has a beneficial
of fatty-alcohol- or fatty-aldehyde-modified lipids effect on the pruritus in patients. A favorable behav-
and proteins. The biological consequences of either ioral effect has also been observed. Some patients
long-chain fatty alcohol or fatty aldehyde accumula- have been treated with acitretin, a retinoid, which
tion are unknown. Long-chain alcohols have been leads to a marked improvement of the ichthyosis and
shown to partition into artificial lipid bilayers and pruritus. Symptomatic treatment includes care of the
synaptic vesicles. Accumulation of fatty alcohols in dermatological and neurological problems.
the skin of SLS patients probably alters the epidermal
water barrier, which is critically dependent on
the lipid composition of the stratum corneum, and 50.5 Magnetic Resonance Imaging
leads to increased transepidermal water loss and
ichthyosis. Furthermore, the catabolism of ether CT reveals diffuse or patchy cerebral white matter hy-
phospholipids (including plasmalogens) and sphin- podensities, most marked in the frontal area. No en-
golipids, both of which are major components of hancement is present after contrast administration.
myelin, generates fatty aldehydes that may be sub- MRI may show three types of white matter abnor-
strates for fatty aldehyde dehydrogenase. Accumula- malities. First of all, the process of myelination is
tion of fatty alcohols or fatty aldehydes in myelin mildly delayed and appears not to reach completion
could alter myelin stability and lead to myelin loss. in SLS patients (Figs. 50.1 and 50.2). The directly sub-
It has been shown that fatty aldehyde dehydroge- cortical U fibers and the white matter of the anterior
nase is also important in the a-oxidation of phytanic part of the temporal lobes, which are normally among
acid. The enzyme is involved in the conversion of the last to reach complete myelination, retain a slight-
pristanal into pristanic acid. Surprisingly, SLS pa- ly higher signal intensity than the adjacent cortex on
tients do not accumulate phytanic acids in their plas- T2-weighted images. Secondly, some patients have a
ma. It may be that another fatty aldehyde dehydroge- symmetrical periventricular rim of markedly in-
nase takes over. creased signal intensity, most often predominantly
Fatty aldehyde dehydrogenase is involved in the w- involving the frontal and parietal lobes (Fig. 50.2).
oxidation of leukotriene B4. Leukotriene B4 is a potent Thirdly, other patients have a symmetrical periven-
proinflammatory mediator, synthesized from arachi- tricular rim of slightly to mildly increased signal in-
donic acid. It is predominantly produced by polymor- tensity, which often has a more posterior predomi-
phonuclear leukocytes. The inactivation of leuko- nance (Fig. 50.3). The white matter changes become
triene B4 is regulated by microsomal w-oxidation. apparent during the process of myelination and seem
The urinary levels of leukotriene B4 and its metabolite to be stable. Follow-up MRI does not show an increase
w-hydroxy leukotriene B4 are elevated in SLS in the extent or severity of the white matter signal
patients. w-Hydroxy leukotriene B4 is converted to changes. In exceptional cases the white matter abnor-
w-aldehyde leukotriene B4 and subsequently w-car- malities are not confluent, but multifocal (Fig. 50.1).
boxy leukotriene B4. Urinary w-carboxy leukotriene Typically, the corpus callosum and cerebellar white
B4 is not detectable, indicating a block in the degrada- matter are spared. In exceptional cases the corpus cal-
tion of leukotriene B4 at the level of the conversion of losum shows signal abnormalities. Brain stem pyra-
w-aldehyde leukotriene B4 to w-carboxy leukotriene midal tract involvement may be seen. Mild cerebral
B4. Elevated levels of leukotriene B4 and w-hydroxy atrophy is common leading to mild but sometimes
50.5 Magnetic Resonance Imaging 385

Fig. 50.1. A 2-year-old patient with SLS. Myelination is incom- signal abnormalities in the periventricular white matter. Cour-
plete. The T2-weighted images show a high signal in the tesy of Dr. M.A.A.P. Willemsen, Department of Pediatric Neu-
directly subcortical white matter in all areas. There are focal rology, University Medical Center Nijmegen, The Netherlands

Fig. 50.2. A 10-year-old boy with SLS. Myelination is still have a sharp demarcation. The corpus callosum is largely
incomplete,which is best seen in the basotemporal white mat- spared. Courtesy of Dr. M.A.A.P. Willemsen, Department of
ter (second row, right). There are prominent signal abnormali- Pediatric Neurology, University Medical Center Nijmegen, The
ties in the periventricular and deep white matter, most pro- Netherlands
nounced in the frontoparietal region.The signal abnormalities
386 Chapter 50 Sjögren–Larsson Syndrome

Fig. 50.3. An 8-year-old patient with SLS, brother of the pa- rior region. The rim is poorly demarcated. Courtesy of Dr.
tient shown in Fig. 50.2. In this patient there is a periventricular M.A.A.P. Willemsen, Department of Pediatric Neurology, Uni-
rim of mild signal abnormality, most pronounced in the poste- versity Medical Center Nijmegen, The Netherlands

more marked dilatation of the lateral ventricles. These resonances have been assigned to methylene
There is no correlation between the severity of the and methyl groups of lipids, respectively. It is highly
white matter abnormalities and the severity of the likely that the peaks represent accumulating fatty
neurological problems or the age of the patient. alcohols or fatty aldehydes. The peaks have their
MRS consistently shows a high and narrow peak greatest height within the abnormal white matter.
at 1.3 ppm and a much smaller peak at 0.9 ppm in Neither of the peaks is seen in spectra from the cere-
spectra of the cerebral white matter of SLS patients. bral cortex.
Chapter 51

Lowe Syndrome

51.1 Clinical Features thirties. Death may occur between the first and the
and Laboratory Investigations fourth decade, secondary to inanition, pneumonia, or
uremia.
Lowe syndrome, or oculocerebrorenal syndrome of Laboratory findings show renal tubular dysfunc-
Lowe (OCRL), is a rare, X-linked hereditary disease. It tion similar to Fanconi syndrome. The tubular dys-
is present in all races, with a predominance in those of function leads to urinary loss of bicarbonate, glucose,
Caucasian and Asian ancestries. Rarely females are calcium, phosphorus, protein, and amino acids. The
affected like males. results are metabolic acidosis, hypophosphatemia,
Males with OCRL present with congenital ocular secondary hyperparathyroidism, and rickets. Urinary
manifestations including cataract, often associated excretion of carnitine is increased with decreased
with glaucoma and miotic pupils. Buphthalmos may serum carnitine. The serum muscle enzymes creatine
be present. Sight is poor and eye movements are nys- kinase, aspartate aminotransferase, and lactate dehy-
tagmoid. Additional early features include muscle hy- drogenase are sometimes elevated. Over the years
potonia, hyporeflexia, or areflexia and hypermobility progressive glomerular dysfunction develops with
of the joints. During the first year of life psychomotor uremia. A linear relation between reciprocal serum
developmental delay becomes evident. The infants creatine level and age reflects the progressive renal
have excessive crying with a high-pitched scream. failure. Abnormalities of blood chemistry values are
Asymptomatic amino aciduria and proteinuria are mitigated by replacement therapy for renal tubular
usually found during this stage. losses.
During early childhood the symptoms of renal Skeletal X-ray examinations reveal rachitic changes.
tubular dysfunction develop with metabolic acidosis EEG often shows nonspecific abnormalities, some-
and losses of calcium and phosphate. Severe deminer- times epileptic activity.
alization and rickets may develop and lead to fre- Progressive lenticular opacities are present in OCRL
quent fractures, pain, and delay of normal physical carriers, and even young carriers can be identified
development. Many patients develop scoliosis. Linear reliably in this way in known OCRL families. Lack of
growth decreases after 1 year of age, and between 1 opacities in females cannot be considered as proof of
and 3 years of age height falls below the third per- not being a carrier, but is suggestive.
centile. Head circumference is either normal or bor- The biochemical assay for deficiency of phos-
derline microcephalic. Noninflammatory arthro- phatidylinositol 4,5-biphosphate 5-phosphatase in
pathy of unknown pathogenesis with pain, joint cultured fibroblasts is the definitive laboratory test
swelling, and contractures may occur, affecting both for diagnosing patients and can be used for prenatal
large and small joints. Mental retardation is usually diagnosis. DNA techniques are available for diagnos-
moderate, but intelligence varies between normal and tic confirmation in patients, for carrier detection, and
profoundly decreased. Some patients have epilepsy. for prenatal diagnosis. De novo mutations or sporadic
Unusual stereotypic movements are often seen, espe- cases are frequent. Somatic and germline mosaicism
cially repetitive shaking of limbs. Maladaptive behav- has been described and must be taken into account
ior is frequently present and consists of temper for genetic counseling.
tantrums, high-pitched scream, and stubbornness. A
later occurring ocular complication is corneal keloid,
consisting of scar-like opacifications of the cornea 51.2 Pathology
that can develop spontaneously or following trauma.
The frequency of corneal keloid increases with age in In OCRL, data on neuropathological findings are lim-
OCRL patients. ited. The brain weight is usually below normal. Mod-
Patients with OCRL continue to grow in early erate, diffuse atrophy may be seen with cerebral vol-
adulthood. Final height, however, remains below the ume loss, some ventricular enlargement and thinning
third percentile of normal values. Severe bone disease of the corpus callosum. The meninges are diffusely
leads to orthopedic disability. Renal dysfunction is fibrotic without inflammatory changes.
slowly progressive with decreasing glomerular filtra- Variable cortical changes have been described
tion rates. Terminal renal failure occurs in the mid- ranging from normal to neuronal loss in combination
388 Chapter 51 Lowe Syndrome

with gliosis. Evidence of aberrant neuronal migration X chromosome. OCRL may also occur in XO patients.
with pachygyria or polymicrogyria has also been re- Unfortunate X inactivation is another possibility
ported. explaining exceptional female patients.
The white matter changes described are variable. Inositol containing phospholipids are collectively
The most frequent finding is gliosis. More or less referred to as phosphoinositides or phosphatidyli-
prominent myelin paucity has been described, either nositols. They are minor but ubiquitous components
focal or diffuse. White matter abnormalities are most of eukaryotic membranes. Phosphoinositides are
pronounced in the centrum semiovale and periven- important in signal transduction. The subcellular
tricular white matter. Spongy changes have been localization of PIP2 5-phosphatase is in the trans-
observed, frequently associated with perivascular Golgi network. The substrate of this enzyme, phos-
lacunes. Active myelin breakdown has never been phatidylinositol 4,5-biphosphate, serves as a sub-
reported, and the myelin paucity is usually ascribed strate for the production of second messengers, pro-
to deficient myelination. In rare cases, the white mat- motes the polymerization of actin cytoskeleton, and
ter is normal. plays a role in vesicular trafficking. In fibroblasts of
The cerebellum may be atrophic or normal. Gliosis Lowe patients a decrease in long actin stress fibers,
of the cortex and white matter have been observed. enhanced sensitivity to actin depolymerizing agents,
Variable and inconsistent abnormalities of the pe- and an increase in actin staining in a distinctly anom-
ripheral nervous system have been reported. Nerve alous distribution in the center of the cells have been
biopsy has been found to exhibit signs of axonal neu- demonstrated. Two actin-binding proteins, gelsolin
ropathy in several, but not all patients. Muscle biopsy and a-actinin, proteins regulated by both PIP2 and
has indicated selective type 1 fiber atrophy with mild Ca2+, have been found to have an abnormal distribu-
type 1 fiber predominance, considered to be consis- tion. Actin polymerization plays a key role in the for-
tent with congenital fiber type disproportion myopa- mation, maintenance, and proper function of tight
thy or, alternatively, to abnormal neuronal influence junctions and adherens junctions, which are critical
upon muscle. In other patients, muscle biopsy re- in renal proximal tubule function and the differentia-
vealed no abnormalities. tion of the lens. The relationship with the encephalo-
Renal pathology demonstrates dilated tubules with pathy is less clear.
atrophy of tubular epithelium. Many tubules contain
eosinophilic, granular protein casts. Interstitial fibro-
sis is present. Glomerular changes have been ob- 51.4 Therapy
served, in particular on ultrastructural examination.
Glomerular endothelial cells may be swollen with a The nephrologist should manage both the tubular
reduction in the number of endothelial slit pores, and glomerular complications associated with the
fusion of endothelial foot processes, and thickening renal disease in OCRL. In renal tubular acidosis and
of the glomerular basement membrane. rickets a combination of sodium-potassium citrate,
phosphorus, and large doses of vitamin D has a ben-
eficial effect. Ocular surgery is essential to prevent
51.3 Pathogenetic Considerations total blindness. The retina in patients with OCRL is
usually well-formed and capable of receiving a light
OCRL is caused by defects in the OCRL1 gene, located stimulus.
on chromosome Xq25–26. The gene product is
phosphatidylinositol 4,5-biphosphate (PIP2) 5-phos-
phatase, an inositol polyphosphate 5-phosphatase be- 51.5 Magnetic Resonance Imaging
longing to the type II 5-phosphatase family. The type
II phosphatases hydrolyze both water-soluble and MRI in OCRL shows a more consistent pattern of
lipid inositol polyphosphate substrates, whereas the abnormalities than would be suggested by the neu-
type I phosphatases only hydrolyze water-soluble ropathology. In patients with an advanced degree of
substrates. However, PIP2 5-phosphatase prefers the myelination patchy, small and larger, isolated and
lipid substrate phosphatidylinositol 4,5-biphosphate. partially confluent white matter changes are visible,
Many different mutations of the OCRL1 gene have suggestive of gliosis (Figs. 51.1 and 51.2). During the
been described in patients, most of which are truncat- first months of life these white matter abnormalities
ing mutations or missense mutations in conserved may not be noticeable within the still unmyelinated
domains of the protein. In highly exceptional cases, white matter. The changes are most pronounced in
the OCRL phenotype has been observed in female the periventricular area and centrum semiovale,
patients. In some of these female patients there is a whereas the U fibers, corpus callosum, internal cap-
mutation in the OCRL1 gene on one chromosome, sule, brain stem, and cerebellum are spared. The white
whereas there is a deletion of the region on the other matter abnormalities are highly variable in extent. In
51.5 Magnetic Resonance Imaging 389

Fig. 51.1. MR study of a 17-month-old boy with OCRL.There are multifocal small lesions in the periventricular white matter and
small cysts within the abnormal white matter

some patients only a few, small white matter lesions The MRI pattern of multifocal, irregular, patchy
are present (Fig. 51.1), whereas in other patients the white matter abnormalities with small cysts is remi-
lesions are much more extensive (Fig. 51.2). The dis- niscent of the patterns seen in hypomelanosis of Ito
tribution of the white matter changes is symmetrical and some mucopolysaccharidoses.
globally, but not in detail. Often multiple small cystic
lesions are seen within the abnormal white matter.
Mild ventriculomegaly may be present. No major cor-
tical abnormalities have been reported.
390 Chapter 51 Lowe Syndrome

Fig. 51.2.
51.5 Magnetic Resonance Imaging 391

Fig. 51.2. An 8-year-old boy with OCRL. The T2-weighted im-


ages (second and third rows) show multifocal small and larger,
partially confluent lesions in the periventricular and deep
white matter. The sagittal T1-weighted (first row) and axial
FLAIR images (fourth row) reveal a multitude of small cysts
within and outside the abnormal white matter. Courtesy of Dr.
L. Charnas, Department of Pediatric Neurology, and Dr. C Peter,
Department of Pediatric Hemato-Oncology and Bone Marrow
Transplantation, University of Minnesota, Minneapolis, USA
Chapter 52

Wilson Disease

52.1 Clinical Features neurological manifestations of WD. The rings are


and Laboratory Investigations typically 1–3 mm in diameter, green, yellow, or brown,
and located at the periphery of the cornea. They
So-called hepatolenticular degeneration was first begin as crescents in the superior quadrant and
described by the English neurologist S.A.K. Wilson in subsequently extend to the inferior, lateral, and medi-
1912 and has been named after him. Wilson disease al regions until they become circumferential and
(WD) is a genetically determined, autosomal reces- broader, spreading centrally from the limbus. Kayser–
sive disease with a prevalence of 20 to 30 per million. Fleischer rings may sometimes be seen with the
The clinical picture is extremely variable, depending unaided eye, but a slit-lamp examination often is nec-
among other things on the age at presentation. Pa- essary to detect them. Adolescents may present with a
tients present with hepatic, neurological, or psychi- deterioration of performance at school and in athlet-
atric symptomatology in roughly equal proportions, ics, difficulties with handwriting, and loss of dexteri-
with some overlap. In children, hepatic manifesta- ty. Without treatment, neurological deterioration
tions predominate, whereas in adolescents and adults continues. The full-blown neurological picture of WD
neuropsychiatric manifestations are more frequent. encompasses a variable combination of the following
Symptoms rarely occur before the age of 6, and half of signs: dysarthria, drooling, dysphagia, grimacing, ab-
the patients are symptomatic by the age of 15. Onset normal eye movements, dystonia, rigidity, bradykine-
after the age of 40 is rare, but patients presenting in sia, athetosis, wing beating, spasticity with increased
the fifth and sixth decades of life have been described. tendon reflexes and Babinski signs, cerebellar ataxia,
Hepatic manifestations in WD encompass a spec- and tremor (resting, intention, or postural), but cog-
trum of acute and chronic liver diseases. Some pa- nitive and sensory functions are typically preserved.
tients, usually younger ones, present with fulminant Seizures may occur. Because of increasing difficulty
hepatic failure, leading to jaundice, hypoalbumine- in controlling movement, the patients become unable
mia, ascites, coagulation defects, hyperammonemia, to feed themselves, and become bedridden. Flexion
and hepatic encephalopathy. Massive liver failure in contractures develop. Ultimately, the patients become
WD is often accompanied by hemolytic anemia. How- helpless, are usually alert, but unable to talk.
ever, if the liver failure is less massive, only some of Psychiatric and behavioral symptoms are variable
the features of this syndrome may be present. In old- and include reduced performance at school or at
er patients liver failure tends to be chronic and is as- work, inability to cope, depression, labile moods
sociated with portal hypertension, leading to gastric ranging from mania to depression, sexual exhibition-
or esophageal varices and/or hypersplenism with ism, and psychosis. Personality changes, particularly
thrombopenia or leukopenia. In addition, these irritability, emotionality, and increased anger are the
patients often have hypoalbuminemia, edema, and most commonly noted problems.
ascites. Some patients present with a picture of acute Ophthalmological abnormalities include, first and
hepatitis, which subsides spontaneously, and may be foremost, the Kayser–Fleischer rings, and, less fre-
attributed mistakenly to a viral infection. In some quently, sunflower cataracts consisting of green, gold-
patients cirrhosis is well compensated and the results en-brown, or gray granular deposits in the lens of the
of standard liver biochemical tests are normal. Most eye, appearing as discoid opacities in the anterior lens
patients with WD, whatever their clinical presentation capsule with petal-like fronds that radiate toward
or presymptomatic status, have some degree of liver the lens periphery like a sunflower. Neither Kayser–
disease. Fleischer rings nor sunflower cataracts affect vision
Patient with WD who present with neurological significantly. Kayser–Fleischer rings are found in al-
disease are typically in their late teens or twenties. most all patients with a neurological presentation and
The signs and symptoms are usually chronic, but in about 30% of the patients with a hepatic presenta-
occasionally acute in presentation. Frequent early tion.
symptoms include tremor, speech problems, clumsi- Hematological presentation is rare, but WD should
ness, dystonia, and personality changes. The tremor be considered in young patients with nonspherocyt-
may be unilateral or asymmetrical. Kayser–Fleischer ic, Coombs-negative intravascular hemolysis of un-
rings are almost invariably present in patients with clear etiology. Severe hemolysis frequently occurs in
52.2 Pathology 393

the setting of fulminant hepatic failure and suggests a scopic examination have a high diagnostic value. He-
diagnosis of WD. patic copper concentrations are always elevated in
Patients may have abnormalities of renal tubular untreated WD patients and also higher in patients
function with amino aciduria, proteinuria, urico- than in heterozygotes. Histological abnormalities in
suria, hypercalciuria, hyperphosphaturia, or defective WD range from steatosis to cirrhosis. Liver histology
urine acidification. Occasionally, full-blown Fanconi is normal in heterozygotes. However, in patients with
syndrome is present. Renal stones are not uncom- chronic cholestatic liver disease, elevations of liver
mon. copper concentrations may be similar to those in WD
Osteomalacia, rickets, osteoporosis, osteoarthritis, patients, and incidental WD cases with normal cop-
polyarthritis, localized bone demineralization, spon- per levels in single biopsies have been reported, re-
taneous fractures, peri- and intra-articular calcifica- lated to uneven copper distribution in the liver. In
tions, and joint hypermobility may occur in WD. selected patients a test of incorporation of a copper
However, musculoskeletal symptoms are rarely the isotope into serum ceruloplasmin is informative. Be-
presenting complaints. cause the biochemical copper profile of a normal
Hyperpigmentation of the skin may occur. Con- neonate mimics that seen in WD and clinical manifes-
gestive heart failure and cardiac dysrhythmias have tations of WD are rarely seen before the age of 6 years,
been reported. The most frequent endocrinological conventional screening of potentially affected chil-
disturbances are gynecomastia and delayed puberty dren should not be performed before the age of 2–3
due to hepatic dysfunction. Primary or secondary oli- years. DNA techniques are available for definite diag-
go- or amenorrhea is common. Successful pregnancy nosis in patients suspected of WD and for distinction
is exceptional in untreated female WD patients, and between presymptomatic patients and heterozygotes
spontaneous abortions occur frequently. in families with WD and known mutations. Prenatal
There is often an unacceptable delay in establish- diagnosis is possible using DNA techniques.
ing a diagnosis of WD.Whenever the diagnosis is sus-
pected, Kayser–Fleischer rings should be sought by
an experienced ophthalmologist using a slit-lamp. 52.2 Pathology
Kayser–Fleischer rings are almost always present in
WD patients with neurological problems, but their External examination of the brain usually reveals no
absence does not mean the diagnosis is wrong. abnormalities except for some shrinkage of the insu-
Kayser–Fleischer rings are also not pathognomonic lar cortex.
for WD, but may be seen in some other hepatobiliary Within the brain parenchyma of patients with
diseases or as a result of topical ocular application of manifestations of CNS disease, lesions are invariably
copper-containing solutions. In most patients sus- present in the corpus striatum, which appears
pected of WD, the diagnosis is confirmed by the pres- shrunken. Cavitation is often present in the putamen,
ence of both Kayser–Fleischer rings and at least one, sometimes also in the caudate nucleus, rarely in the
if not all three, of the biochemical markers: low serum globus pallidus, thalamus, and red nucleus. On micro-
level of ceruloplasmin, elevated free (nonceruloplas- scopic examination, neuronal loss is found in the
min) serum copper concentration, and increased uri- putamen and caudate nucleus. Astrocytic prolifera-
nary copper excretion (24-h urine copper). However, tion is present. Some astrocytes have the appearance
the limited value of these tests should be recognized. of Alzheimer type II cells; few resemble Alzheimer
Ceruloplasmin is low in about 80–90% of WD pa- type I cells. When cavities are present, variable num-
tients, but some WD patients have levels in the low-to- bers of lipophages and siderophages are seen in rela-
normal range due to hepatic inflammation, hepatic tion to the cavities; old and recent hemorrhages can
neoplasm, pregnancy, or use of estrogen. A low serum be found. Depositions of copper have been described
ceruloplasmin level is found in up to 20% of the het- by some, in particular in the putamen. Less constant-
erozygotes for WD, even though they will never devel- ly and less severely involved nuclei are the globus pal-
op symptoms of WD. Low serum ceruloplasmin can lidus, the subthalamic nucleus, the thalamus, dentate
also be found in other hepatic disorders, normal nucleus, substantia nigra, and other brain stem nu-
neonates and young infants, patients with malabsorp- clei. Opalski cells are typically present in the thala-
tion, malnutrition, or nephrosis, and in patients with mus, globus pallidus, and zona reticularis of the sub-
hereditary hypoceruloplasminemia. Free serum cop- stantia nigra, less often in the caudate nucleus and
per and urinary copper concentrations can be elevat- putamen. Opalski cells are large cells with volumi-
ed in other hepatobiliary diseases. In case of doubt, in nous cytoplasm and small nuclei. Like Alzheimer
particular if the ceruloplasmin level is normal and/or type I and type II cells, they are of astroglial origin.
Kayser–Fleischer rings are absent, conditions most Alzheimer type I cells and Opalski cells are character-
often seen in patients with hepatic presentation, liver istic of WD.
biopsy for quantitative copper analysis, and micro-
394 Chapter 52 Wilson Disease

In some patients destructive lesions are present in Copper is an essential trace element, being an inte-
the cerebral cortex and white matter. The distribution gral component of many important enzymes involved
of the cortical involvement corresponds with that of in cellular processes such as oxidative metabolism,
the white matter involvement. These lesions may be free radical detoxification, neurotransmitter biosyn-
very impressive and exceed the lesions in the basal thesis, and iron homeostasis. In excess, copper is also
nuclei. The lesions are typically located in the superi- a very toxic ion, because it can oxidize proteins and
or and middle frontal gyri; widespread involvement lipids in membranes, bind to proteins and nucleic
posterior to the frontal lobes is uncommon. The acids, and enhance the generation of free radicals.
changes are usually symmetrical, but may be asym- Dietary intake of copper generally far exceeds the
metrical. Cavitation of the white matter lesions may trace amounts required. Consequently, efficient and
occur. The outer layers of the cortex always retain suf- appropriate mechanisms must exist to ensure copper
ficient structural integrity to form a continuous shell. homeostasis and to transport copper to the sites
In microscopic examination of the affected cortex, the where it is required without allowing toxic accumula-
deep cortical layers appear to be affected predomi- tion of free ions. Clearance of excess copper is
nantly, whereas the more superficial cortical layers achieved via the hepatobiliary route.Various proteins
are relatively intact. The cortical abnormalities may have been recognized to be involved in copper home-
have a spongiform appearance.Alzheimer types I and ostasis and transport: albumin for copper transport
II cells and Opalski cells may be seen in the cerebral in the blood, ceruloplasmin as a copper donor to tis-
cortex. The white matter abnormalities vary from sues and enzymes, and metallothionein for intracel-
myelin paucity and spongy white matter degenera- lular copper storage. The human copper chaperone
tion to outright necrosis with presence of sudano- Atox1 is a small cytosolic protein that plays a key role
philic breakdown products. There may be prolifera- in the distribution of copper to the secretory pathway
tion of Alzheimer type I and type II astrocytes and of the cell.Atox1 facilitates copper delivery to the pro-
Opalski cells. tein ATP7B. ATP7B translocates copper in an ATP-de-
The cerebellum is relatively spared. Some loss of pendent manner into the lumen of the trans-Golgi
cortical neurons may be seen. The dentate nucleus is network for incorporation into copper-dependent
more frequently involved. Cerebellar white matter enzymes, or exports excess copper out of the cell into
pallor, gliosis, and degeneration have been described. the bile. Under steady state conditions ATP7B is local-
Brain stem nuclei may be involved. ized primarily in the trans-Golgi network, where it
In patients with fulminant liver failure, the cirrho- pumps copper from the cytoplasm into the trans-Gol-
sis is predominantly micronodular, whereas in pa- gi network lumen. Under elevated copper concentra-
tients with longstanding liver disease large areas of tions ATP7B undergoes a reversible, copper-mediated
macronodular cirrhosis are found. In the periphery of translocation from the Golgi to the apical canalicular
the nodules, copper pigments may be demonstrated membrane, where it pumps copper into the bile. Defi-
histochemically. Electron microscopy demonstrates ciency of ATP7B leads to failure to excrete copper
that hepatocytes contain giant mitochondria with from the liver into bile and to incorporate copper in-
paracrystalline inclusions. Multivesicular rounded to ceruloplasmin during ceruloplasmin biosynthesis
granules in hepatocytes are thought to be character- in the liver. This results in toxic accumulation of cop-
istic of WD. per in the liver, with subsequent overflow to the kid-
ney, brain, and cornea. WNDP is present in hepato-
cytes, kidney, placenta, brain, heart, lung, muscle, and
52.3 Pathogenetic Considerations pancreas, but reversal of dysfunction of other organs
after liver transplantation indicates that the primary
The gene responsible for WD is ATP7B, located on genetic defect leading to copper accumulation resides
chromosome 13q14.3. The gene product is a cation in the liver.
transporting P-type ATPase protein, called ATP7B or Accumulation of copper leads to liver cirrhosis,
Wilson disease protein (WNDP). Many different progressive neurological damage and renal problems.
mutations have been identified in WD patients. It is Very high free serum copper levels, in particular
reasonable to assume that the type of mutation might caused by sudden massive release of copper from the
account, at least in part, for the clinical variability liver, may result in hemolytic crises. The cause of
observed in WD patients. However, most patients are pathology in WD is copper toxicity. Organs affected
compound heterozygotes, which hampers the study by the disease contain elevated copper levels, and low-
of genotype–phenotype correlations. Considerable ering of copper leads to cessation of progression,
clinical variation is seen among patients within the some repair, and clinical improvement. Excess copper
same family, suggesting influence of other genetic causes cell injury, inflammation, and cell death. Cop-
and environmental factors. per has damaging effects on mitochondria and per-
52.4 Therapy 395

oxisomes. Effects on microtubules, on cross-linking A few highly exceptional patients have been reported
of DNA, and on plasma membranes, as well as inhibi- who show deterioration after the institution of oral
tion of a large number of enzymes, have also been zinc therapy.
shown. The state of copper is important. Copper Penicillamine has been considered the drug of first
bound to ceruloplasmin, metallothionein, or albumin choice in WD for many years. It acts by reductive
is nontoxic. It is ionic copper or copper, which is easi- chelation, reducing the copper bound to protein,
ly dissociable, that is toxic. Copper toxicity probably thereby decreasing the affinity of the protein for cop-
occurs because of the generation of oxidant radicals. per and allowing the penicillamine to bind copper.
In this respect it is important to note that the pattern Reductive chelation is much more effective than
of preferential involvement of gray matter structures chelators with a high affinity for copper, such as
in WD shows some striking similarities to the pat- EDTA. The copper mobilized by penicillamine is ex-
terns observed in respiratory chain defects and in hy- creted in urine. Penicillamine probably also induces
poxia. Caudate nucleus and putamen are preferential- hepatic metallothionein, thereby holding potentially
ly affected. Other frequently involved gray matter toxic copper in a nontoxic form. Some patients im-
structures are thalamus, globus pallidus, subthalamic prove clinically soon after therapy begins, whereas
nucleus, and periaqueductal gray matter. some may require several months of therapy before
improvement occurs. In about 10–20% of WD pa-
tients with neurological complaints, temporary exac-
52.4 Therapy erbation of symptoms is seen with the institution of
therapy. Unfortunately, some patients never recover
Therapeutic strategies in WD can focus on inhibition to their pretreatment baseline. Inadvertent discontin-
of the intestinal uptake of copper, induction of metal- uation of penicillamine therapy may lead to cata-
lothionein to detoxify copper, and chelation. When a strophic clinical deterioration and death. Adverse
diagnosis of WD is made, siblings of the affected pa- effects of penicillamine are, unfortunately, common.
tient should be screened. On average 25% of the sib- A considerable number of the patients develop an
lings will be affected but still at the “presymptomatic” early hypersensitivity reaction to penicillamine man-
phase clinically, although they always have subclinical dating its discontinuation. Treatment is restarted in
liver disease. WD is an almost completely penetrant conjunction with corticosteroids until tolerance de-
disease, and individuals having two mutations in the velops. Long-term use of penicillamine can result in
WD disease gene have an almost 100% chance of ac- serious side effects forcing discontinuation of its use.
quiring neurological, psychiatric, or hepatic disease These side effects include induction of a variety of
or a combination of these. It is therefore important to autoimmune-like disorders and induction of skin
treat these presymptomatic patients prophylactically problems due to alterations of dermal collagen and
with anti-copper medication. Patients begin to have elastin.
hepatic damage from about the age of 3 or 4 years. Triethylene tetramine dihydrochloride (trientine
Patients should be treated as soon as a diagnosis has or trien) is both a chelator, acting primarily by en-
been made, but probably not during the first 1 or 2 hancing urinary excretion of copper, and an inhibitor
years of their life. Treatment should be continued for of copper absorption in the intestine. Far less clinical
life in WD patients, whether asymptomatic or symp- experience has been gained with this drug than with
tomatic. penicillamine, but it can be used in WD patients who
Nowadays, zinc is usually preferred as the first- do not tolerate penicillamine. Tolerance of triethylene
choice drug in the treatment of WD patients, because tetramine dihydrochloride is generally excellent. Tox-
it is almost completely nontoxic. Oral zinc in the form icity during the first weeks of treatment may consist
of zinc acetate or zinc sulfate inhibits intestinal cop- of bone marrow suppression or proteinuria. Later a
per absorption and promotes fecal copper excretion. variety of autoimmune disorders may occur.
There is also evidence that zinc may induce the syn- Ammonium tetrathiomolybdate forms a tripartite
thesis of metallothionein in enterocytes and possibly complex with copper and protein that is very stable.
hepatocytes. Although zinc appears to be a more Given with food, ammonium tetrathiomolybdate
potent stimulus to metallothionein synthesis than complexes food copper with food protein, rendering
copper, copper is more avid in binding to metallo- food copper, along with copper in saliva, gastric juice,
thionein. Complexed copper may then either be and intestinal secretions, unabsorbable. This puts the
transferred into the portal circulation or remain com- patient in an immediate negative copper balance. In
plexed within the cytosolic fraction of the enterocyte addition, tetrathiomolybdate is absorbed into the
and be excreted in the feces with sloughed cells. Zinc blood and complexes free copper with blood albu-
therapy acts slowly, producing a modestly negative min. This complexed copper cannot be taken up by
copper balance. Zinc therapy is usually well tolerated, cells and is nontoxic. This drug has a faster effect than
gastric irritation being the most frequent side effect. zinc and avoids the deteriorations that may be seen
396 Chapter 52 Wilson Disease

with penicillamine.As such the drug is ideal for initial flect edema, necrosis, and gliosis. In some patients, ar-
treatment. Possible side effects are related to copper eas of low signal intensity are seen in the putamen,
deficiency and include mild bone marrow suppres- head of the caudate nucleus, globus pallidus, and thal-
sion. In addition, mild elevations of aminotransferas- amus on T2-weighted images (Figs. 52.1 and 52.2).
es may be seen. The nuclei either have a generalized low signal inten-
Because of the ubiquity of copper in our diet, a sity or have small areas of low signal intensity within
negative copper balance cannot be achieved by di- the bigger high signal intensity lesion. Some assume
etary modification alone. It seems prudent, however, that the low signal intensity on T2-weighted images is
to recommend that WD patients avoid food with a related to iron deposition, as phagocytes containing
high copper content. iron pigment after small hemorrhages are seen in
Despite the effectiveness of medication in most histological specimens. Others assume that the low
WD patients, occasional patients require liver trans- signal intensity is related to the paramagnetic effect
plantation because of hepatic failure. This procedure of copper. It is well established that copper ions in vit-
corrects the metabolic defect localized in the liver, ro have a pronounced effect on both T1 and T2. An
and transplanted patients no longer need specific increased concentration of copper ions causes a
medication for WD. decrease in T1 and T2. High signal intensity on T1-
The prognosis for WD patients has generally im- weighted images has been found in the globus pal-
proved greatly with the advent of treatment. In partic- lidus (Fig. 52.1) and, more rarely, in the putamen, cau-
ular, when treatment is started in a presymptomatic date nucleus, and around the aqueduct. This may be
stage, patients never become symptomatic. Also, the explained by a paramagnetic effect of copper, but may
vast majority of symptomatic patients improve con- also be related to hepatic dysfunction. Sometimes
siderably or recover completely. The patients with multiple small cavities are seen within the basal nu-
hepatic presentation have the least optimistic prog- clei with a signal intensity similar to that of CSF on all
nosis. Assuming they survive the initial episode, the sequences. No contrast enhancement is seen.
greatest risks are bleeding from esophageal varices Atrophy is a frequent finding. It may be focal, most
and hepatic insufficiency. The patients with neuro- frequently involving the caudate nuclei and brain
logical or psychiatric presentation have a better prog- stem. The atrophy may also be generalized involving
nosis, although their quality of life can be problemat- the cerebral hemispheres and the cerebellum, with
ic if the residual deficit is considerable. It is therefore enlargement of subarachnoid spaces and ventricular
important to start treatment as early as possible, system.
before irreparable damage has occurred. Under ade- Subcortical white matter lesions are not rare in
quate treatment, successful pregnancy is possible. WD, in contrast to what was formerly thought. The
Anti-copper therapy should be continued during white matter lesions are most often found in the
pregnancy. Zinc is safest as both penicillamine and frontal lobes, sometimes with extensions into the
triethylene tetramine dihydrochloride are potentially parietal area (Fig. 52.3). White matter lesions in other
teratogenic. areas are rare. Their location is largely subcortical
and they may not reach the ventricular wall. The over-
lying cortex may also be involved (Fig. 52.3). The
52.5 Magnetic Resonance Imaging white matter lesions are usually large and confluent,
but multiple, small, isolated lesions can also be found.
CT scan of the brain in WD may reveal a number of In exceptional cases the white matter lesions are cys-
different abnormalities including mild ventricular tic. The white matter changes are often asymmetrical
dilatation, cortical atrophy, brain stem atrophy, hypo- (Fig. 52.3), which contrasts with the symmetry of the
dense areas in the region of the putamen, globus pal- gray matter lesions. The posterior limb of the internal
lidus, and dentate nucleus, and white matter hypo- capsule and the external capsule may be affected.
densities, in particular in the frontal subcortical area. Rare localizations are the splenium of the corpus
MRI most often shows lesions in the thalamus, callosum and the white matter of the parietal, tempo-
putamen, and caudate nucleus (Figs. 52.1–52.3). ral, and occipital lobes. No contrast enhancement is
Within the putamen, the lateral rim is often most present.
prominently affected (Fig. 52.1). Other gray matter Cerebellar lesions are less frequent. Bilateral le-
structures which are also regularly affected are the sions in the dentate nuclei are most often seen. The
globus pallidus, claustrum, and subthalamic nucleus. cerebellar white matter may be abnormal, especially
The lesions are usually bilateral and symmetrical; in the area around the dentate nucleus (Fig. 52.1).
asymmetry is rare. The lesions most often have a high Brain stem lesions are frequent. The areas most
signal intensity on T2-weighted images and a low sig- frequently involved are the midbrain tectum and
nal intensity on T1-weighted images (Figs. 52.1–52.3). tegmentum, the red nucleus, substantia nigra, cen-
These changes in signal intensity are thought to re- tral part of the base of the pons, and pontine tegmen-
52.5 Magnetic Resonance Imaging 397

Fig. 52.1. A 37-year-old female with WD. Note the prominent thalamus standing out separately. Additionally, the T2-weight-
abnormalities in the basal ganglia (first row). The caudate nu- ed images (second and third row) show areas of high signal in
cleus is markedly atrophic. The caudate nucleus, the peripher- the brain stem and cerebellar white matter, whereas the den-
al rim of the putamen, and the claustrum have a high signal on tate nucleus has a low signal. The typical “panda face” is seen
proton density and T2-weighted images, whereas the central on the right in the second row. Courtesy of Dr. E. Gut, Depart-
part of the putamen has a low signal. The globus pallidus has ment of Neuroradiology, Kliniken Schmieder, Allensbach, Ger-
a high signal on T1-weighted images. The ventrolateral thala- many
mus also contains signal abnormalities, the outer rim of the
398 Chapter 52 Wilson Disease

Fig. 52.2. A 31-year-old female with WD. The T2-weighted im- signal. Within the midbrain, the typical panda face configura-
ages show high signal intensity in the ventrolateral thalamus, tion is seen (second row, left). Courtesy of Dr.T. Naidich, Depart-
its outer rim standing out separately. The putamen has an ab- ment of Radiology, The Mount Sinai School of Medicine, New
normally low signal with some tiny high-signal-intensity areas. York
The tectum and tegmentum of midbrain and pons have a high

Fig. 52.3. A 13-year-old boy with WD.Note the involvement of white matter lesions with an asymmetrical distribution. The
the putamen and thalamus, the lateral rim of the thalamus overlying cortex is affected as well. From Hedera et al. (2002),
standing out separately. Additionally, there are large focal with permission
52.5 Magnetic Resonance Imaging 399

tum. Atrophy of midbrain and pons may be seen. tory and laboratory tests differentiate between these
A T2-weighted image through the midbrain may re- disorders.
veal the so-called “face of the giant panda” appear- MRI is abnormal in almost all patients with neuro-
ance, with a low signal intensity of the superior col- logical complaints and in some neurologically nor-
liculus, loss of the normal, characteristic low signal mal patients. In exceptional cases normal MRI has
intensity of the lateral portion of the substantia nigra, been reported despite the presence of neurological
presence of the normal low signal intensity of the red abnormalities.With successful treatment, MRI abnor-
nucleus, and high signal intensity of the tegmentum malities have been shown to improve or disappear.
of the midbrain (Figs. 52.1 and 52.2). Some correlations have been found between cere-
MRI is of value in WD, both for diagnostic and bral lesions and clinical symptomatology. Dystonia
monitoring purposes. The diagnosis of WD may be is related to putamen lesions; dysarthria correlates
difficult, and MRI may suggest this possibility. The with lesions in both putamen and caudate nucleus.
most important differential diagnostic options are Both lesions in the substantia nigra and in the puta-
respiratory chain defects and extrapontine myelinol- men and caudate nucleus are associated with par-
ysis, in particular if a central lesion is present in the kinsonism. Abnormalities in efferent cerebellar path-
basis of the pons. WD and Japanese encephalitis also ways, including the superior cerebellar peduncle,
show a very similar topography of the MRI abnormal- red nucleus, and thalamus, are associated with clini-
ities. However, brain stem lesions are more frequent cal cerebellar signs. Lesions in the dentate nucleus are
in WD. WD patients more often have a mixed signal associated with a kinetic tremor and dysmetria.
intensity of the basal ganglia and a typical linear rim Lesions in the red nucleus are associated with proxi-
of signal abnormality in the putamen. Thalamic hem- mal kinetic tremor. Generalized atrophy and subcor-
orrhagic lesions and meningeal enhancement are tical white matter lesions are associated with cogni-
more often seen in Japanese encephalitis. Clinical his- tive decline.
Chapter 53

Menkes Disease

53.1 Clinical Features Sepsis and meningitis are also fairly common. Death
and Laboratory Investigations usually occurs before the age of 3 years.
Patients with unusually severe variants of MD have
Menkes disease (MD), also called kinky hair disease been reported who present soon after birth with
or trichopoliodystrophy, is a rare, genetically deter- extensive vascular disease with intracranial, gastro-
mined disease with an X-linked recessive mode of intestinal, intra-abdominal, pulmonary, and other
inheritance. About 30% of cases are related to new hemorrhages, in the absence of a coagulopathy, final-
mutations, and in these patients a positive family his- ly resulting in hemorrhagic shock and early death.
tory is lacking. Rarely, females are affected, related to Additionally, brain infarcts may occur and multiple
an accompanying chromosomal abnormality (for in- fractures may be present.
stance, XO or a translocation that disrupts the disease Milder variants of the disease occur, including
gene on the X chromosome) or carriership for a mild MD and occipital horn syndrome (OHS). Pa-
mutation and a highly unfavorable X inactivation tients with mild MD have a hypopigmented skin,
pattern. The incidence has been estimated as 1 in laxity of the skin and joints, abnormal hair, and the
100,000–250,000 live births. However, the true inci- characteristic facial appearance. The neurological
dence is unknown because patients may die in early symptoms are much milder than in classical MD. The
infancy without a diagnosis. The clinical spectrum is predominant neurological manifestations of mild
broad. MD are prominent cerebellar ataxia and dysarthria,
In classical MD, premature birth and low birth and moderate cognitive deficit.
weight are frequent. In the neonatal period, some pa- OHS was previously known as type IX Ehlers–
tients display hypothermia, hypoglycemia, poor feed- Danlos syndrome or X-linked cutis laxa. It is an X-
ing, and hyperbilirubinemia. However, these features linked recessive connective tissue disorder. The dis-
may also appear in otherwise healthy premature ba- ease derives its name from distinctive cranial exos-
bies and rarely lead to a diagnosis in the neonatal pe- toses (occipital horns) resulting from calcification of
riod. The infants are normal up to the age of 2–4 the trapezius and sternocleidomastoid muscles at
months, when loss of developmental milestones, hy- their attachment to the occipital bone. The predomi-
potonia, hypothermia, seizures, and failure to thrive nant clinical manifestations of OHS are skeletal ab-
occur. The hair abnormalities characteristic of MD normalities including the occipital horns, short and
usually become prominent at the age of 2–3 months. broad clavicles, osteoporosis, laxity of the skin and
The hair is short, sparse, coarse, twisted, and often joints, bruisable skin, and bladder diverticula. The
hypopigmented. It resembles steel wool. Eye lashes urinary tract abnormalities lead to recurrent urinary
and eyebrows can also be affected. The infants have a tract infections. Most patients have inguinal hernias.
cherubic face, with a decreased facial expression, The only apparent neurological abnormalities of pa-
prominent forehead, pudgy cheeks, sagging jowls, tients with OHS are autonomic dysfunction and slight
and scant eyebrows. The skin and joints are lax. The mental deficiency. The autonomic dysfunction may
skin is lacking in pigment. Inguinal hernias may be lead to diarrhea, orthostatic hypotension and syn-
present. Diverticula of the bladder and ureters predis- cope, and temperature instability with heat intoler-
pose to urinary tract infections and sometimes rup- ance. Life expectancy is as a rule up to at least mid-
ture. Hydronephrosis may occur. Bone changes such adulthood.
as osteoporosis are always present, and fractures of Some patients have been reported with a mixture
the long bones and ribs may occur. Pectus excavatus is of manifestations of classical MD, mild MD, and OHS.
often seen. Diarrhea is common and intestinal hem- Most patients with classical MD show moderate to
orrhage may occur. The neurological deterioration in severe EEG abnormalities including hypsarrhythmia.
classical MD is severe. Most patients never achieve Skeletal changes in classic MD are distinctive but not
head control or independent sitting. The seizures are pathognomonic. They usually become apparent by
usually intractable. Subdural hematomas and effu- the second month of life. They include wormian
sions are often present. Infections are common, in bones of the skull, generalized osteoporosis, meta-
particular respiratory and urinary tract infections. physeal spurring of long bones, and anterior flaring
of the ribs. Rib and long bone fractures, including
53.2 Pathology 401

metaphyseal fractures, are common. The clavicles are depigmentation, and/or increased copper accumula-
short with hammer-shaped distal ends. Radiographic tion in cultured fibroblasts. However, some hetero-
abnormalities in OHS include osteoporosis, occipital zygotes display no abnormalities, which can be ex-
and femoral exostoses, carpal coalescence, coxa valga, plained by the individual X inactivation pattern.
deformation of the radii, ulnae, tibiae, and fibulae, DNA-based diagnosis is very useful for carrier detec-
and short clavicles with hammer-shaped distal ends. tion. Prenatal diagnosis can be performed on the
Microscopic examination of the hair in MD reveals basis of analysis of copper levels or 64Cu uptake in cul-
pili torti (180° twisting of the hair shaft), often in tured amniotic fluid cells or chorionic villi. DNA-
combination with constrictions at regular intervals based prenatal diagnosis is possible in families with a
(trichorrhexis nodosa), varying diameters, transverse known mutation.
fractures of the hair shaft, and longitudinal splitting
of the shaft.
Serum levels of copper and ceruloplasmin are very 53.2 Pathology
low in patients with classical MD. Interpretation is
difficult in the first 2–3 weeks of life. Serum copper In many infants with classic MD, chronic subdural
and ceruloplasmin levels are normally low at birth hematomas are found. The brain shows as a rule
and rise over the first month. It may therefore be dif- severe and diffuse atrophy of the cerebral and cere-
ficult to diagnose MD by these markers in the bellar hemispheres. The cerebral arteries are thin-
neonate. An abnormal plasma or CSF catecholamine walled, tortuous, and ectatic. On sectioning, the white
pattern may be a more sensitive marker of copper matter appears shrunken and the lateral ventricles
deficiency, but a lumbar puncture is not feasible in are moderately dilated.
neonates with a hemorrhagic disease. The defect in Microscopic examination of the cerebral cortex
copper transport can be demonstrated by analysis of shows widespread neuronal loss and severe spongio-
copper accumulation in cultured fibroblasts or lym- sis. Areas of total devastation with microcystic rar-
phoblasts using radioactive copper (64Cu). Copper efaction of the cortex may be present, and in some
levels and accumulation are elevated in MD. The areas the cortex and subcortical white matter may be
activity of various copper-dependent enzymes, such cavitated. Moderate nerve cell loss is also found in the
as cytochrome-c oxidase, dopamine b-hydroxylase, basal ganglia. The thalamus and red nucleus may be
and lysyl oxidase, are reduced. CSF lactate is elevated, particularly affected and show severe degeneration.
as expected in cytochrome-c oxidase deficiency. The cerebral white matter is reduced in volume. Mi-
Catecholamine levels are abnormal in serum and CSF, croscopic examination demonstrates severe myelin
with elevated levels of the dopamine metabolite dihy- paucity and sometimes severe and diffuse gliosis. The
droxyphenylacetic acid (DOPAC) and the catechol- myelin paucity is probably explained by a combina-
amine precursor dihydroxyphenylalanine (DOPA) tion of poor myelination and myelin loss. A status
and low levels of the deaminated norepinephrine spongiosus may be seen in the white matter but frank
metabolite dihydroxyphenylglycol (DHPG), indicat- cavitation is rare.
ing reduced activity of dopamine b-hydroxylase. As a The cerebellum is markedly atrophic. On micro-
result the ratios of DOPA to DHPG and DOPAC to scopic examination, there is a striking loss of granule
DHPG are markedly elevated and valuable for a diag- cells. The Purkinje cells are also diminished in num-
nosis. Generalized amino aciduria has been observed ber and show striking dendritic abnormalities, with
in a few patients with MD, but renal tubular damage is presence of perisomatic dendrites radiating from the
rarely a problem. perikaryon. The apical dendrites may show deformi-
In patients with mild MD and OHS, serum levels of ties of the weeping willow or staghorn pattern. Den-
copper and ceruloplasmin are usually slightly to dritic swellings and axonal torpedoes may also be
moderately decreased, but may also be normal or present. On electron microscopy, a marked increase in
even lower than in patients with classical MD. The the number and size of mitochondria is found in the
increased copper levels and 64Cu uptake and retention perikaryon of Purkinje cells and to a lesser extent of
in fibroblasts or lymphoblasts from patients with neurons within the cerebral cortex and basal ganglia.
mild MD or OHS are indistinguishable from those in There are intramitochondrial electron-dense bodies.
classical MD. The activity of lysyl oxidase is signifi- In muscle biopsy of MD patients, mitochondrial
cantly decreased in skin and cultured fibroblasts from abnormalities have been found, including enlarged
patients with OHS. mitochondria with disorderly and irregular cristae.
DNA-based diagnosis is possible. Female carriers Ragged red fibers have also been reported.
are clinically almost always completely normal, with Ultrastructural studies of the skin reveal that der-
the exception that pili torti are seen in the hair of 50% mal collagen fibrils have a heterogeneous size range,
of carriers. Heterozygous females can be diagnosed with a mean diameter smaller than normal. Dermal
on the basis of these pili torti in their hair, patchy skin elastin fibers are scarce and considerably smaller
402 Chapter 53 Menkes Disease

than normal. They consist of thin strands of amor- defect in the ATP7A protein leads first of all to re-
phous elastin associated with numerous microfibrils. duced uptake of copper across the small intestine,
Cerebral and systemic arteries and arterioles are because the copper accumulates in the intestinal
tortuous with irregular lumina and frayed and split epithelial cells and is not excreted into the blood. The
intimal linings. In the walls of arteries the amount of reduced supply of dietary copper is further com-
collagen is normal, but the fibrils display a broader pounded by the defective distribution of copper with-
range of diameters than normal, with a slightly small- in the body wherever ATP7A is required for copper
er mean. Elastin fibers in the internal elastic lamina, transport. For example, ATP7A is involved in the
tunica media, and intimal layers show considerable transport of copper across the blood–brain barrier to
disruption, appearing fragmented and wider than the CNS, explaining the marked deficiency of copper
normal and displaying irregular contours. Veins are in the brain of MD patients. In contrast with the over-
less often affected. all copper deficiency in MD, copper accumulation oc-
curs in certain tissues such as the intestinal epithelial
cells, the kidney, and cultured cells, owing to the cop-
53.3 Chemical Pathology per efflux defect. It is probable that copper is normal-
ly resorbed in the proximal tubules of the kidney, as
Lipid studies in MD show decreases in myelin lipids under normal conditions very little copper is excret-
with increases in cholesterol and cholesterol esters. ed in the urine. Patients with MD cannot deliver the
The presence of cholesterol esters points to active resorbed copper from the epithelial cells of the prox-
myelin loss. In the white matter galactolipids are imal tubules into the blood.
decreased. Deficiency of copper-dependent enzymes causes
the multiple abnormalities in MD patients. Hypopig-
mented hair is caused by the deficiency of tyrosinase,
53.4 Pathogenetic Considerations an enzyme required for melanin synthesis. The
unusual hair, the pili torti, is caused by reduced
The gene for MD and its variants is MNK, also called keratin cross-linking, a process that is copper-depen-
ATP7A, located on chromosome Xq13.3. The gene dent. Connective tissue abnormalities, including
codes for a transmembrane copper-transporting arterial vessel wall abnormalities, loose skin, and
P-type ATPase. ATP7A is expressed in virtually all fragile bones, result from reduced lysyl oxidase activ-
nonhepatic tissues. The protein is located in the ity and consequent weak cross-links in collagen
trans-Golgi network, pumping copper into the Golgi and elastin. Deficiency of several enzymes, includ-
apparatus to supply copper to the copper-dependent ing cytochrome-c oxidase (oxidative respiration),
enzymes. In the presence of elevated extracellular superoxide dismutase (free radical detoxification),
copper concentrations, the protein relocates to the peptidylglycine a-amidating mono-oxygenase (mod-
plasma membrane to pump excess copper out of the ification of various peptide neurotransmitters), and
cells. When intracellular copper levels are reduced, dopamine b-hydroxylase (neurotransmitter bio-
the protein returns to the trans-Golgi network. This synthesis), may contribute to the neurological dam-
reversible relocation is very important for copper age.
homeostasis. Many different mutations have been found in
Several key metabolic processes are copper-depen- ATP7A. In classical MD, most mutations are nonsense
dent, including oxidative respiration neurotransmit- mutations, frameshifts, large deletions, and splice site
ter synthesis, connective tissue formation, free radical mutations, which lead to absence of functional pro-
detoxification, iron homeostasis, and pigmentation. tein. In mild MD, missense mutations have been
Copper is required for the catalytic activity of a num- found that block copper-induced relocation of the
ber of essential enzymes in these processes, the ma- mutated protein from the trans-Golgi network to the
jority of which catalyze oxidation–reduction reac- plasma membrane. The milder course of the disease
tions. The ability of copper, like iron, to undergo re- suggests that the mutant protein retains some copper
versible redox changes makes it a useful cofactor in transport activity. Patients with OHS have splice site
redox reactions. However, both metals also catalyze mutations that still permit the production of a small
the production of potentially damaging free radicals amounts of normal ATP7A mRNA next to abnormal-
and are toxic in excess amounts. Organisms have ly small transcripts. The milder clinical manifesta-
several mechanisms to obtain and distribute copper tions can be explained by the presence of small
safely and to excrete the excess. amounts of the normal protein. As most of the clini-
Defects in ATP7A result in a defect in the copper cal symptoms in OHS are related to lysyl oxidase
excretion of cells and an accumulation of copper in deficiency, it is assumed that this enzyme is more sen-
their cytosol. Copper efflux requires ATP7A in most sitive than the other cuproenzymes to copper defi-
cells, hepatocytes being a notable exception. The ciency.
53.6 Magnetic Resonance Imaging 403

53.5 Therapy regressed or even disappeared in some MD patients,


although an OHS-like clinical picture may persist in
In the treatment of MD patients, the first important such patients. It has been suggested that mutations
issue is early diagnosis. The second issue is to bypass with preservation of some residual ATP7A activity
the block in the intestinal absorption of copper. The may be requisite for significant clinical efficacy of
third is to deliver circulating copper to the brain. The copper replacement therapy (Kaler et al. 1996). One
fourth is that copper must be available within cells to patient has been reported in whom mislocalization of
enzymes that require copper as a cofactor. the protein in the endoplasmic reticulum due to a cer-
Oral administration of copper generally has no tain mutation could be corrected by supplementation
effect on circulating copper, but parenteral adminis- of copper (Kim et al. 2002). However, significant im-
tration of copper in any form restores circulating cop- provement has also been observed in some early-
per and ceruloplasmin to normal levels. Among the treated patients from classical MD families with se-
different forms of copper, copper histidine has been vere mutations.
reported to be taken up by the brain most efficiently. From a practical point of view, commencing par-
Copper uptake by hepatocytes and brain tissue is enteral copper histidine treatment as soon as the
physiologically mediated by histidine. Therefore, cop- diagnosis of MD has been established may be advo-
per histidine is generally used for parenteral admin- cated irrespective of the gene abnormality. Therapy
istration. While low hepatic stores are quickly replen- should be continued until it becomes evident that
ished by parenteral copper treatment, levels of copper cerebral degeneration cannot be arrested.
in the brain only increase gradually, if at all, during Symptomatic treatment including antiepileptic
treatment, consistent with trapping of copper within medication, maintenance of an adequate nutritional
the cells forming the blood–brain barrier. The activi- status, and physical therapy are important. Bisphos-
ties of copper-dependent enzymes remain subnormal phonate therapy has been used to increase bone min-
under treatment in most patients. However, it is diffi- eral density in MD patients. It is effective in the man-
cult to assess the activities of most enzymes serially, agement of osteoporosis and contributes to the pre-
as repeated skin, muscle, and brain biopsies would be vention of bone fractures.
required to do that. The darkening of the hair under
treatment indicates a higher activity of tyrosine hy-
droxylase. Ceruloplasmin is the only cuproenzyme 53.6 Magnetic Resonance Imaging
whose activity always normalizes in response to cop-
per treatment, because this enzyme is synthesized in One of the most striking findings on MRI in patients
the liver, where ATP7A is not expressed. with classical MD is excessive tortuosity of the cere-
There are two major problems with this copper bral arteries. MRA confirms tortuosity, dilatation,
replacement therapy. First of all, there is usually a and kinking of the arteries (Fig. 53.1). Tortuosity of
delay in diagnosis, and during this time brain damage the extracranial systemic arteries is also present.
continues which cannot be undone. Secondly, the Early MRI of the brain may be normal in classical
block in intestinal copper absorption may be by- MD. Soon, however, serious damage develops. Tran-
passed by parenteral administration, but the basic de- sient bilateral temporal lobe white matter abnormali-
fect affecting transport of copper into the brain and ties with prominent swelling and involvement of the
intracellular copper metabolism in tissues through- overlying cortex have been observed (also seen in
out the body cannot be fully overcome by copper Fig. 53.1). In some MD patients, brain lesions arise
replacement. that look like focal infarctions with a low ADC on dif-
Clinical outcomes of MD patients treated with cop- fusion-weighted images. The cerebral white matter
per replacement have generally been disappointing. may also be diffusely abnormal, mildly swollen, and
Some patients who are treated from an early age, have a highly abnormal signal. In that stage of the dis-
preferably <1 month, show outcomes better than ex- ease, there is no evident atrophy and there are no sub-
pected, and some even attain normal developmental dural fluid collections. The white matter swelling
milestones. However, other patients do not fare well soon evolves into atrophy and the white matter re-
despite early treatment. When treatment is com- mains poorly myelinated (Fig. 53.2). The atrophy is
menced at an older age, after onset of symptoms, progressive over time. Some white matter areas may
significant neurological recovery is not possible, become cystic. Diffuse and serious cerebral and cere-
although decreased seizure frequency, reduced irri- bellar atrophy leads to enlargement of the extracere-
tability, better sleeping, and slight advances in social bral spaces (Fig. 53.2). Subdural hygromas and
development may be seen. Copper treatment may hematomas easily develop and are almost invariably
prolong life span, but not consistently in all patients. seen in MD (Fig. 53.2). As the atrophied brain recedes
Some striking exceptions have been observed. from the dura, the bridging cortical veins may tear,
With early treatment neurological symptoms have resulting in subdural effusions. The vulnerability of
404 Chapter 53 Menkes Disease

Fig. 53.1. A 5-month-old boy with MD.The large, swollen tem- brain stem show abnormal hypertortuosity, which is not a fea-
poral lesions, which involve both white matter and overlying ture of herpes encephalitis. MRA confirms the tortuosity of the
cortex, could be considered suggestive of herpes encephalitis. cerebral arteries. Courtesy of Dr. T. Aduc, Randburg, and Dr.
However, the arteries in the sylvian fissure and around the M.M. Lippert, Pretoria, South Africa

the abnormal vessels in MD may contribute to the de- agnosis. However, the clinical picture with skin and
velopment of hematomas. In addition, there may be hair abnormalities, tortuosity of the arteries on MRI,
signal changes in the basal ganglia. and the presence of other bone abnormalities should
The most important item in the differential diag- help in the differentiation. In patients presenting with
nosis is the shaken baby syndrome. The combination predominantly white matter disease on MRI, tortuos-
of subdural hygromas and hematomas of different ity of the cerebral vessels is again an important diag-
ages and bone fractures is very suggestive of this di- nostic clue for MD.
53.6 Magnetic Resonance Imaging 405

Fig. 53.2. The same boy as in Fig. 53.1, now 6.5 months old. stage. Bilaterally low in the basal ganglia a ball of tortuous and
Note the serious atrophy with enlargement of the subarach- dilated vessels is seen, which is particularly clear in the en-
noid spaces and small subdural effusions. The process of larged view.Courtesy of Dr.T.Aduc, Randburg, and Dr.M.M.Lip-
myelination is not showing any progress beyond the neonatal pert, Pretoria, South Africa
Chapter 54

Fragile X Premutation

54.1 Clinical Features dystrophic axons. Spongiform changes are seen in the
and Laboratory Investigations cerebellar white matter and middle cerebellar pedun-
cles.
Fragile X syndrome is one of the most common in-
herited forms of mental retardation. The disorder is
caused by an expansion in excess of 200 repeats (full 54.3 Pathogenetic Considerations
mutation expansion) of a trinucleotide element,
(CGG)n, located in the 5’ untranslated region of the The fragile X syndrome is caused by an expansion in
fragile X mental retardation 1 gene (FMR1). Trinu- excess of 200 repeats of a trinucleotide element,
cleotide expansions that are in the 55–200 repeats (CGG)n, located in the 5’ untranslated region of the
range are called premutations. Normal persons have a fragile X mental retardation 1 gene (FMR1) on chro-
range of approximately 5–54 repeats. mosome Xq27.3. Trinucleotide expansions that are
Male carriers of premutations typically have nor- longer than 200 repeats are called full mutation ex-
mal intelligence, although emotional problems and pansions. Trinucleotide expansions that are in the
subtle physical features occur in about 25%. Mental 55–200 repeats range are called premutations. Nor-
retardation and autism may occur. About 20% of fe- mal persons have a range of approximately 5–54
male patients with a premutation experience prema- repeats.
ture ovarian failure and menopause. Some female The pathophysiology of the neurological syn-
premutation carriers have emotional problems, in- drome in older males with a premutation has not
cluding anxiety, obsessional thinking, or depression. been fully understood. Repeat expansions are known
Some male carriers of the fragile X premutation to influence both transcription and translation. The
over the age of 50 years experience progressive neuro- full expansion of over 200 repeats generally leads to
logical dysfunction with tremor and cerebellar ataxia, transcriptional silencing and deficiency or absence of
most frequently leading to gait difficulties and deteri- the FMR1 protein (FMRP). Lack of this protein leads
oration of fine motor skills. Additionally, progressive to the fragile X syndrome. In patients with a fragile X
cognitive decline and behavioral difficulties arise, in- premutation, FRMP levels are gradually reduced with
cluding memory loss, anxiety, mood lability, and increasing repeat number, despite elevated FMR1
reclusive behavior. In a few patients more serious de- mRNA levels, suggesting that translation is impeded
mentia occurs. More variable features include parkin- within the premutation range. The syndrome seen in
sonism, peripheral neuropathy with distal weakness older males with a premutation may be related to a
and sensory loss, lower limb proximal muscle weak- gain of function with toxic effects related to elevated
ness, and autonomic dysfunction with urinary and FMR1 mRNA levels.
bowel incontinence and impotence. Rarely, female
premutation carriers develop tremor and ataxia, but
they do not experience cognitive decline. 54.4 Therapy
The diagnosis of fragile X premutation is estab-
lished by demonstrating a CGG repeat with a length No specific treatment is available.
of 55–200 trinucleotide repeats in the FMR1 gene.

54.5 Magnetic Resonance Imaging


54.2 Pathology
In older male patients carrying the fragile X premuta-
Neuropathologic findings include cerebral and cere- tion and the clinical symptoms described above, MRI
bellar atrophy. There are eosinophilic intranuclear in- shows symmetrical signal abnormalities in the mid-
clusions in neurons and astrocytes throughout the dle cerebellar peduncles and deep cerebellar white
cortex, subcortical regions, and brain stem. The hip- matter (Figs. 54.1 and 54.2). After contrast, no en-
pocampus and the frontal cortex are the most severe- hancement occurs. The dentate nucleus is spared.
ly affected. There is evidence of cerebellar degenera- Mild to moderate atrophy of the cerebellum, mid-
tion with Purkinje cell loss, torpedo formation, and brain, and pons is frequently present. Mild to moder-
54.5 Magnetic Resonance Imaging 407

Fig. 54.1. A 61-year-old man with a fragile X premutation. A are patchy signal abnormalities in the periventricular and
CGG repeat with a length of 80 trinucleotide repeats was deep cerebral white matter. Courtesy of Dr. J. Vandevenne,
demonstrated in the FMR1 gene. Note the cerebral and cere- Department of Radiology, and Dr. A. Wibail, Department of
bellar atrophy. There are signal abnormalities in the middle Neurology, Hospitals Oost-Limburg, Genk, Belgium
cerebellar peduncles and deep cerebellar white matter. There

ate cerebral atrophy with thinning of the corpus cal- and parietal periventricular and deep white matter
losum is present in the majority of the patients (Figs. 54.1 and 54.2). MRI does not demonstrate the
(Figs. 54.1 and 54.2). In many patients additional same abnormalities in asymptomatic older male
white matter abnormalities are present in the frontal patients carrying the fragile X premutation.
408 Chapter 54 Fragile X Premutation

Fig. 54.2. A 63-year-old man with a fragile X premutation. A of elevated signal intensity. In addition, there are signal abnor-
CGG repeat with a length of 90 trinucleotide repeats was malities in the base of the pons, middle cerebellar peduncles,
demonstrated in the FMR1 gen.These sagittal T1-weighted im- and cerebellar white matter. Courtesy of Dr. J. Vandevenne,
age (first row, left) and axial FLAIR images show prominent Department of Radiology, and Dr. I. Raets, Department of Neu-
cerebral and cerebellar atrophy. There is a periventricular rim rology, Hospitals Oost-Limburg, Genk, Belgium
Chapter 55

Hypomelanosis of Ito

55.1 Clinical Features nign. CNS abnormalities are frequent and these are
and Laboratory Investigations highly variable. Neurological impairment can be
quite severe with hemimegalencephaly or migra-
Hypomelanosis of Ito (HMI), formerly also called in- tional defects, leading to intractable seizures and seri-
continentia pigmenti achromians, does not represent ous mental and motor handicap. About 80% of the
a distinct entity but is a symptom of many different patients have mental retardation, which varies from
states of mosaicism. HMI is characterized by unilater- mild to profound. Autism is seen in some patients.
al or bilateral hypopigmented whorls, streaks, and HMI has been associated rarely with vascular abnor-
patches. The abnormal pigmentation follows Blaschko malities such as moyamoya syndrome and intracra-
lines. The Blaschko lines have a bizarre fountain-like nial arteriovenous malformations. If a parent is
pattern on the back, whorls on the abdomen and lin- affected, he or she may only have cutaneous abnor-
ear stripes on the limbs. Zones of hypomelanosis are malities and no neurological problems, whereas the
easily seen in patients with a darker skin, but to see offspring has evident neurological signs.
the zones in patients with a fair skin the use of Wood’s In patients with HMI chromosome mosaicism is
light may be necessary. In most patients the zones are often restricted to the skin, while blood karyotypes
apparent at birth or within the first year of life. The may be normal. For this reason skin fibroblast chro-
mucous membranes are spared. The hypopigmented mosome analysis is recommended. Mosaicism has
whorls represent the “negative pattern” of the hyper- also been found in keratinocytes obtained from the
pigmented lesions seen in incontinentia pigmenti hypopigmented areas of the skin. The use of FISH
type II, an X-linked disorder that is lethal for males. facilitates the determination of the origin of the chro-
Another difference with incontinentia pigment type mosomes involved and the extent of the mosaicism
II is that neonatal inflammatory skin abnormalities when one is detected.
are lacking in HMI.
HMI is a nonspecific marker for genetic or chro-
mosomal mosaicism in persons with a sufficiently 55.2 Pathology
dark skin to show lighter patches. Most cases are spo-
radic, but autosomal dominant inheritance is evident Some patients with HMI may have major brain
in some. Incontinentia pigmenti type I is a subtype of abnormalities, including hemimegalencephaly and
hypomelanosis of Ito, associated with an X chromo- migrational abnormalities. The pathology of these
some/autosome translocation involving band Xp11. abnormalities is not reviewed here. The pathological
Abnormalities of the eyes, hair, teeth, muscu- correlate of the white matter abnormalities observed
loskeletal system, heart, and CNS occur in some of the relatively frequently in HMI is deficient myelination
patients with HMI. Hypopigmented areas can be ob- and gliosis.
served in the iris and the retina. Corneal opacities,
macrophthalmia, microphthalmia, strabismus, epi-
canthal folds, choroidal atrophy, and optic nerve hy- 55.3 Pathogenetic Considerations
poplasia may occur. Abnormalities in hair color,
alopecia, hypertrichosis, and trichorrhexia may be HMI is a manifestation of etiologically heterogeneous
observed. Fingernails may be abnormal. Numerous conditions, the common factor being chromosomal
types of dental dysplasia may be present. Macro- or genetic mosaicism. A mosaic is defined as an or-
cephaly is relatively common; microcephaly is less ganism composed of two (or more) genetically differ-
frequent. The face may be asymmetrical. Unilateral ent populations of cells originating from a genetical-
hypertrophy or hypotrophy of the body or part of the ly homogeneous zygote. The mosaicism either in-
body may be present. Other abnormalities observed volves a chromosomal abnormality or a gene muta-
include hypertelorism, bifid uvula, cleft palate, small tion. The chromosomal mosaicism includes an array
stature, scoliosis, clinodactyly, and syndactyly. Con- of different abnormalities: diploid/triploid mo-
genital cardiac abnormalities and unilateral kidney saicism, diploid/tetraploid mosaicism, mosaicism for
aplasia may be present. Occurrence of a tumor is a trisomies, unbalanced translocation between auto-
rare observation, and if present, it is most often be- somes or an autosome and the X chromosome, chro-
410 Chapter 55 Hypomelanosis of Ito

mosomal deletions, and ring chromosomes. In the sphere is dysplastic with a larger ventricle, a thick-
X-chromosome, band Xp11 is involved. HMI due to ened, pachygyric or polymicrogyric cortex, and fre-
Xp11 alteration is caused by functional or genetic quently heterotopic neurons in the white matter. Var-
mosaicism. Functional mosaicism results from the ious other abnormalities have been found, including
Lyon effect of X inactivation. agenesis of the corpus callosum, asymmetrical or
Mosaicism can be recognized most easily in the symmetrical ventricular dilatation, focal cerebral at-
skin, which exhibits patterns of differences in pig- rophy with porencephalic ventricular dilatation,
mentation following the Blaschko lines. The different hemiatrophy, diffuse cerebral atrophy, cerebellar hy-
degrees of pigmentation of the skin represent differ- poplasia or atrophy, vascular abnormalities, and,
ent cell populations. The abnormal cell line is con- rarely, a tumor.
fined to the skin with abnormal pigmentation; the More subtle brain abnormalities in HMI consist of
normal skin contains almost only normal cells. It has irregular, multifocal, and confluent abnormalities in
been hypothesized that the genetic and chromosomal the periventricular and deep white matter, usually
abnormalities result in abnormalities of pigmenta- with increased perivascular spaces within and, to a
tion because of altered expression of genes in the pig- lesser extent, also outside these areas (Fig. 55.1). In
mentary pathway. Such genes may be present in dif- some patients, enlarged perivascular spaces are seen
ferent regions of multiple chromosomes. without associated white matter signal abnormalities.
The enlarged spaces may also involve the corpus cal-
losum. The increased perivascular spaces are best
55.4 Therapy seen on FLAIR images (Fig. 55.1). The white matter
abnormalities are often bilateral but may be asym-
No specific treatment is available. metrical in distribution. They are static and probably
reflect white matter gliosis and focal myelination de-
fects.
55.5 Magnetic Resonance Imaging Presence of brain abnormalities in HMI appears to
be associated with an enhanced frequency of neuro-
In many patients with HMI, MRI of the brain is nor- logical problems. Major malformations are associated
mal. Some patients with HMI have major CNS abnor- with a major handicap. White matter abnormalities
malities, including migrational abnormalities and with enlarged perivascular spaces are more common-
hemimegalencephaly. The migrational abnormalities ly seen in patients with a mental handicap, although
may consist of cortical dysplasia or neuronal hetero- they have also been observed in HMI patients with a
topias. In hemimegalencephaly, the enlarged hemi- normal cognitive function.

Fig. 55.1. A 5-year-old boy with HMI, mental retardation, and ages (fourth and fifth row) allow distinction between enlarged
macrocephaly. The sagittal (first row) and axial (second and perivascular spaces with a low signal intensity and gliotic
third row) T2-weighted images show extensive, partly multifo- white matter with a high signal intensity. Courtesy of Dr. S.
cal and partly confluent abnormalities in the cerebral white Blaser, Department of Diagnostic Imaging, Hospital for Sick
matter.The posterior fossa structures are spared.The FLAIR im- Children, Toronto, Canada
55.5 Magnetic Resonance Imaging 411

Fig. 55.1. (continued).


Chapter 56

Incontinentia Pigmenti

56.1 Clinical Features include cataract, strabismus, retinal pigmentary ab-


and Laboratory Investigations normalities, retinal detachments, infarction, vitreous
hemorrhage, microphthalmia, and optic atrophy.
Incontinentia pigmenti (IP), formerly called inconti- The brain is affected in 30–50% of the patients,
nentia pigmenti type II or Bloch–Sulzberger syn- causing mental retardation, spasticity (hemiplegia or
drome, is an X-linked dominant disorder that is usu- quadriplegia), cerebellar ataxia, and seizures. The
ally fatal prenatally in males. Fifty-five percent of the head circumference is usually normal at birth, but
patients have a family history of the disease; the rest patients may develop microcephaly. Some of the
of the cases are caused by spontaneous mutations. patients have signs of serious encephalopathy in the
So-called incontinentia pigmenti type I is unrelated neonatal period, with intractable seizures suggestive
to incontinentia pigmenti type II and is in fact a sub- of viral encephalitis. Acute-onset stroke has been
type of hypomelanosis of Ito, associated with an X observed occasionally in infants and children with IP.
chromosome/autosome translocation involving band Patients with structural brain abnormalities and
Xp11. It has an X-linked dominant mode of inheri- neonatal seizures are at greater risk of motor and
tance. In contrast to incontinentia pigmenti type II, mental impairment.
the typical neonatal inflammatory skin abnormalities In IP, laboratory studies usually reveal eosinophil-
are lacking. Incontinentia pigmenti type I is dis- ia in the blood in the neonatal period. The disease is
cussed in Chap. 55. caused by mutations in the NEMO gene, located on
Affected females display skin abnormalities in four chromosome Xq28. Cells expressing the mutated X
stages: (1) perinatal inflammatory vesicles, (2) verru- chromosome are eliminated selectively around the
cous patches, (3) hyperpigmentation, and (4) dermal time of birth, so females with IP display extremely
scarring. In IP the pigmentary disturbance is a sort of skewed X inactivation, which can be demonstrated in
self-tattooing. The lesions of the first phase are pre- peripheral blood leukocytes.
sent at birth or appear within the first 2 weeks of life
in 90% of the patients, although they may also appear
later in infancy or childhood. They are erythematous, 56.2 Pathology
macular, papular, vesicular, and bullous, and suggest
an inflammatory condition of the skin. Lesions of the Pathological findings in IP are highly variable and
second stage, appearing between 2 weeks and several dependent on the stage of the disease. Fresh hemor-
months of life, are pustular, lichenoid, verrucous, ker- rhagic necrosis within the cerebral white matter with
atotic, and dyskeratotic. The third stage is character- perivenous and capillary hemorrhage, vascular con-
ized by development of pigmentation, mainly at the gestion, edema, and inflammation surrounding the
sites of the earlier lesions. In the fully developed dis- hemorrhage is seen in the early stages. The gray mat-
ease, the skin shows swirling patterns of melanin pig- ter may also be affected, but usually to a lesser extent.
mentation, especially on the trunk, with a “marble In the more chronic stages, ulegyria, cystic changes in
cake” appearance. The pigmentary abnormalities the cerebral white matter, neuronal loss, and gliosis
usually disappear by the age of 20 years. The fourth are found.
stage is characterized by atrophic scarring. Hypome-
lanotic macules, most often located over the calves,
may be the only dermatological indication of IP in 56.3 Pathogenetic Considerations
adulthood. Late recurrences of the first-stage inflam-
matory lesions are uncommon and are usually pre- Mutations in the gene NEMO, located on chromo-
ceded by an infectious episode. some Xq28, cause IP. IP is allelic with ectodermal dys-
IP may be associated with a variety of abnormali- plasia, an- or hypohidrosis, and immunodeficiency, a
ties of the eyes, teeth, and skeleton. Dystrophy of the disease caused by different mutations in the same
nails is frequent but usually mild. Any of the cuta- gene. NEMO stands for nuclear factor (NF)-k-B essen-
neous stages of IP may involve the scalp.Alopecia sec- tial modulator. The gene is also called IKBKG, which
ondary to scarring is frequent. Malformed teeth, hy- stands for inhibitor of k (kappa) light polypeptide
podontia, or adontia may occur. Ocular abnormalities gene enhancer in B cells, kinase of, gamma (g). NEMO
56.3 Pathogenetic Considerations 413

Fig. 56.1. Female neonate, 3 days old, with IP. She has signs of T1-weighted images (third row) indicates hemorrhagic necro-
a devastating encephalopathy and vesicular skin lesions. Note sis. Courtesy of Dr. N.Wolf, Department of Pediatric Neurology,
the extensive damage to large parts of the cerebral white mat- and Dr. A Seitz, Department of Neuroradiology, University Hos-
ter and cortex, more seriously on the left than on the right. pital Heidelberg, Germany
The lesions have a swollen aspect. The high signal on the

is essential for NF-k-B activation. Activated NF-k-B ing paternal meiosis, is responsible for 80% of the
normally protects against tumor necrosis factor alpha new mutations. This mutation results in a lack of
(TNF-a)-induced apoptosis. NF-k-B activation, which leads to an extreme suscep-
The most common mutation in IP is a genomic tibility to proapoptotic signals, leading to embryonic
rearrangement resulting in a large deletion within the death in males and extremely skewed X inactivation
NEMO gene. This rearrangement, which occurs dur- in females with IP. The remaining mutations are small
414 Chapter 56 Incontinentia Pigmenti

Fig. 56.2. Follow-up MRI of the girl shown in Fig. 56.1. She is involved. Courtesy of Dr. N.Wolf, Department of Pediatric Neu-
now 5 months old.There is a serious cystic degeneration of the rology, and Dr. A Seitz, Department of Neuroradiology, Univer-
deep and subcortical cerebral white matter, more prominent- sity Hospital Heidelberg, Germany
ly on the left than on the right. The overlying cortex is also

duplications, substitutions, and deletions. In surviv- their replacement by cells in which the normal X
ing males with IP, somatic mosaicism or a 47,XXY chromosome is active. This leads to extremely skewed
karyotype with skewed X inactivation has been X inactivation patterns in females, which save them
found. from the lethal effect of the mutation. Other interpre-
The evolution of the lesions in females can be in- tations suggest a vasculopathy as the major patho-
terpreted as representing death of cells that have the genetic mechanism for ophthalmological and brain
mutant-bearing X chromosome as the active one and damage.
56.5 Magnetic Resonance Imaging 415

56.4 Therapy of gliotic scar tissue, focal atrophy, and secondary


cortical dysplasia in the form of ulegyria are common
No specific treatment is available. (Fig. 56.3). The scar tissue may be unilateral or bilat-
eral, symmetrical or asymmetrical. The lesions usual-
ly involve both cortex and white matter of the cerebral
56.5 Magnetic Resonance Imaging hemispheres, but the most severe lesions are usually
seen in the deep white matter. The lesions may also be
In IP, the MRI findings are highly variable. In similar to those seen in periventricular leukomalacia
neonates with signs of active encephalopathy, MRI with mild, symmetrical or asymmetrical dilatation of
shows variably extensive areas of hemorrhagic cortex the lateral ventricles due to loss of white matter vol-
and white matter necrosis and edema suggestive ume, periventricular white matter signal abnormali-
of encephalitis, in particular herpes encephalitis ties, and disfiguration of the lateral ventricles due to
(Fig. 56.1). Cystic degeneration may follow (Fig. 56.2). the gliotic scarring (Fig. 56.3). The corpus callosum is
In some patients the lesions are small, whereas in oth- thin. Lesions may also be seen in the cerebellum. De-
er patients the abnormalities involve most of one or velopmental cerebral anomalies may also be present,
both cerebral hemispheres. In the chronic stage, signs including polymicrogyria and neuronal heterotopias.

Fig. 56.3. A 13-month-old girl with IP. Note the extensive There is retraction of the cortex on the right, which is folded
damage to the white matter of the right cerebral hemisphere. with deep narrow sulci (ulegyria). Courtesy of Dr.W.J. Feikema,
The white matter damage on the left is much milder.The later- Department of Neurology, Deventer Ziekenhuis, Deventer,The
al ventricles are dilated, more so on the right than on the left. Netherlands
Chapter 57

Alexander Disease

57.1 Clinical Features tients the clinical course is episodic and progressive,
and Laboratory Investigations as in multiple sclerosis. A chronic progressive course
with bulbar and pseudobulbar symptoms, spasticity,
Alexander disease (AD) is a rare disorder of the CNS. cerebellar ataxia, and dementia has been reported.
Almost all cases are sporadic, but there are occasion- Nystagmus and other abnormalities in eye move-
al familial cases with an apparent autosomal domi- ments may occur. Some patients only have bulbar
nant mode of inheritance. Three clinical subgroups of signs. Palatal myoclonus may present. Sometimes the
AD can be distinguished: infantile, juvenile, and disease remains asymptomatic and Rosenthal fibers
adult. are found at brain autopsy. Macrocephaly is not a sign
In infantile AD, the onset of symptoms varies from of the disease in adults.
birth to early childhood. The average age at onset is 6 Laboratory investigations are not helpful in estab-
months. There is increasing macrocephaly together lishing the diagnosis of AD. CSF is normal or shows
with feeding problems, difficulty swallowing, chok- a nonspecific increase in protein level. The CSF
ing, vomiting, failure to thrive, and signs of neurolog- aB-crystallin level may be elevated, as may be the CSF
ical deterioration. Slowing down of motor and mental level of heat shock protein 27 (HSP27), but the sensi-
development, loss of acquired developmental mile- tivity and specificity of these tests have not been as-
stones, spastic quadriparesis, and seizures are usually sessed. High-voltage slow-wave activity and focal dis-
present. Choreoathetosis and other extrapyramidal charges are recorded on the EEG in most cases, with
signs may occur. In some patients there are clinical predominance of abnormalities over the frontal area.
signs of elevated intracranial pressure with bulging A brain biopsy or autopsy revealing the characteristic
fontanel, vomiting, and papilledema at funduscopy. Rosenthal fibers used to be considered a prerequisite
Usually funduscopic findings are normal. Nystagmus for a definite diagnosis. However, DNA-based diagno-
and eye movement abnormalities may occur. The sis is now possible. Sporadic patients are hetero-
children may develop apneic attacks or chronic hy- zygous for a mutation in the GFAP gene which is de
poventilation. The average duration of the illness is novo and not found in one of the parents. In familial
2–3 years, ranging from a few months to 8 years. cases, affected family members are heterozygous for a
In juvenile AD, the age at onset of clear symptoms mutation in the gene, the disease being transmitted in
varies from 4 to 14 years, with an average age of 9 an autosomal dominant fashion.
years. However, in retrospect subtle signs of neuro-
logical problems have usually been observed since
before the age of 2 years, mainly some developmental 57.2 Pathology
delay or seizures. Many of the patients have macro-
cephaly, but this is a less consistent finding than in In infantile and juvenile AD the brain is abnormally
infantile AD. The patients suffer from progressive enlarged. External examination may reveal macro-
bulbar and pseudobulbar symptoms, with delayed gyria. Olfactory bulbs and optic nerves are some-
speech development, dysarthria, hoarseness, loss of times enlarged. In many patients the lateral ventricles
speech, increasing swallowing problems, and apneic are widened, either because of hydrocephalus or be-
attacks. Many patients have bouts of vomiting, espe- cause of atrophy and tissue loss. Hydrocephalus, if
cially during morning hours. The swallowing prob- present, is caused by narrowing of the aqueduct. Oc-
lems and vomiting often lead to insufficient gain in casional cases have been reported with a greatly ex-
weight, finally necessitating tube feeding. Spasticity, panded cavum septi pellucidi, bulging into the lateral
cerebellar ataxia, seizures, behavioral changes, and ventricles and compressing the foramina of Monro.
cognitive deterioration develop. The average duration Subependymal cysts may be seen beneath the inferi-
of illness is 8 years. or surfaces of both frontal horns. Thalami, basal gan-
In adult AD, the onset of symptoms is highly vari- glia, cerebellum, and brain stem may be atrophic on
able, occurring between the second and seventh inspection.
decades. Some of the cases are familial, with an affect- On microscopic examination the most distinctive
ed parent and one or more affected children. The clin- feature of AD is the presence of countless Rosenthal
ical features reported are highly variable. In some pa- fibers throughout the CNS. Rosenthal fibers are irreg-
57.3 Chemical Pathology 417

ularly shaped, elongated or round hyaline eosino- myelin paucity most axons are intact. The affected
philic bodies up to 50 mm in length with a diameter of white matter is markedly cellular due to abundance of
1–25 mm. They are arranged radially around blood abnormal, hypertrophied astrocytes. No inflammato-
vessels and perpendicularly to the surface of the cere- ry reaction is present. Oligodendroglia do not show
bral hemispheres, brain stem, cerebellum, and spinal any pathological changes, but may be reduced in
cord in the subependymal and subpial regions. In ad- number. Cavitation occurs relatively frequently in
dition, they are scattered throughout the white matter AD, is usually present in the deep white matter of the
in all areas of the CNS. Rosenthal fibers are most frontal lobes, and is sometimes seen in the parietal
prominent in the deep frontal white matter, the cere- lobes adjacent to the lateral ventricles and the hilus of
bral cortex, the periventricular region, the basal gan- the dentate nucleus. In the end stage, the white matter
glia, thalami, and brain stem. Deposition of Rosenthal may be severely reduced in volume.
fibers in the cerebellum is variable, being sparse in In most cases there is a lack of myelin and at the
some cases but prominent in others, involving the same time a scarcity of sudanophilic material. Some
cerebellar white matter, dentate nucleus, or, rarely, the authors suggest that the absence of typical features of
subpial layers of the cerebellar cortex. The fornix and active breakdown of myelin sheaths points to dis-
optic nerves, chiasm, and tracts may contain many turbed myelination rather than demyelination. How-
Rosenthal fibers. The peripheral or schwannian parts ever, in other cases the presence of sudanophilia and
of the cranial nerves are always free of Rosenthal macrophages accumulating neutral fat have been re-
fibers, whereas the intraparenchymal root bundles ported. Possibly, myelin paucity can be explained by a
may contain heavy deposits. The neurons of the cor- variable combination of disturbed myelination and
tex and basal ganglia are usually relatively well pre- demyelination, disturbed myelination being most
served regardless of the degree of Rosenthal fiber pronounced in the patients with early onset of dis-
deposition, but there may also be a serious loss of ease.
neurons in the frontal cortex and deep gray matter In adult cases, more rarely in juvenile cases, and in
structures. In the brain stem the subependymal accu- exceptional infantile cases, the abnormalities may be
mulation of Rosenthal fibers may lead to narrowing much more limited. Usually, brain stem, cerebellar,
of the lumen of the aqueduct, resulting in hydro- and spinal cord abnormalities dominate, with Rosen-
cephalus. thal fiber deposits and numerous hypertrophied as-
Throughout the CNS there are accumulations of trocytes in subpial, subependymal, and perivascular
hypertrophic fibrillary astrocytes, most marked in regions. Myelin paucity may be present in the affect-
the subpial, subependymal, and periventricular re- ed areas, but myelin density may also be normal.
gions. Their distribution corresponds to the greatest There may be focal, tumor-like lesions with mass ef-
concentration of Rosenthal fibers. The astrocytes are fect. There may also be a striking atrophy of the low-
often large and may contain bizarre nuclei. They have er brain stem and the upper part of the spinal cord.
large amounts of cytoplasm and, in their perikaryon, Microscopy of the lesions may demonstrate prolifer-
hyaline droplets which show the staining characteris- ated astrocytes with considerable pleomorphism of
tics of Rosenthal fibers. On electron microscopy it is the nuclei and occasionally multiple nuclei, resem-
evident that the Rosenthal fibers are abundant in as- bling an astrocytoma. The Rosenthal fiber deposition
trocytic processes and are present in smaller amounts may also be more widespread throughout the CNS.
in the astrocytic perikarya. On electron microscopy Likewise, there may be more extensive white matter
Rosenthal fibers appear as irregular, electron-dense, abnormalities, predominantly involving the frontal
osmiophilic, granular deposits closely associated and parietal white matter. The white matter involve-
with intermediate glial filaments. The granular de- ment may be patchy and multifocal or diffuse. Cavita-
posits are non-membrane-bound. tions have been reported in the frontal white matter,
Another distinctive histological feature is paucity but also in the brain stem, hilus of the dentate nucle-
of myelin. The lack of myelin sheaths is generally us, and spinal cord.
most pronounced in the frontal white matter, tempo-
ral white matter, centrum semiovale, tegmentum of
the brain stem, and ventral and lateral columns of the 57.3 Chemical Pathology
spinal cord. As a rule the frontal white matter is the
most severely involved. There is little or no sparing of Chemical analysis of brain tissue in AD reveals signs
the arcuate fibers. The internal capsule, parieto-oc- of immature myelin with a relatively high content of
cipital white matter, and cerebellum are relatively bet- glucolipids instead of galactolipids and with a rela-
ter myelinated. However, in some cases the cerebellar tively low cerebroside content.All myelin constituents
white matter is also extensively involved. The brain are present in a lower than normal concentration as a
stem involvement may be plaque-like with focal or consequence of the myelin paucity. Cholesterol esters
multifocal areas of myelin paucity. In the areas of are not elevated.
418 Chapter 57 Alexander Disease

Rosenthal fibers consist of two components: bun- conditions. They accumulate in reactive and neoplas-
dles of intermediate filaments, and aggregates of tic astrocytes, and this accumulation is associated
dense material on the filaments. Biochemically, the with translocation of the proteins from the soluble
major proteins of the aggregates are aB-crystallin fraction to the insoluble or cytoskeleton-related frac-
and HSP27.A fraction of aB-crystallin is ubiquinated. tion. In the Rosenthal fibers, aB-crystallin and HSP27
The filaments contain glial fibrillary acidic protein are present as insoluble aggregates bound to interme-
(GFAP) and vimentin. GFAP is also found in the gran- diate glial filaments. The association of aB-crystallin
ular aggregates. and HSP27 with intermediate filaments is probably
critical in the formation of Rosenthal fibers. Ubiqui-
tin is another component of the Rosenthal fibers.
57.4 Pathogenetic Considerations Rosenthal fibers contain mono- and polyubiquitinat-
ed conjugates of aB-crystallin. Conjugation with
AD is caused by mutations in the gene GFAP, which ubiquitin is the first step in a series of reactions that
encodes GFAP and is, located on chromosome 17q21. leads to intracellular nonlysosomal degradation of
The disease has an autosomal dominant inheritance proteins. However, apparently stable ubiquitin conju-
and almost all patients have a de novo mutation, i.e., gates can also be formed, suggesting that proteolysis
which is not found in one of the parents. Familial cas- is not the only function of ubiquitin conjugation.
es are mainly seen in adult AD, where patients may Ubiquitin is present in various abnormal filamentous
have offspring. So far, only missense mutations have neuronal inclusions, such as the neurofibrillary tan-
been found. It is most likely that the mutations ob- gles of Alzheimer disease, Lewy bodies in Parkinson
served in AD act in a gain-of-function fashion. GFAP- disease, and Pick bodies in Pick disease. The presence
null mice have a subtle phenotype which does not re- of ubiquitin in these inclusions may represent an
semble AD. On the other hand, transgenic mice with abortive or only partially successful attempt to de-
overexpression of human GFAP have a fatal en- grade proteins that accumulate in the abnormal states
cephalopathy that closely resembles AD.Astrocytes of mentioned.
these mice are hypertrophic and contain inclusion The formation of Rosenthal fibers is in itself a non-
bodies that are histologically and antigenically iden- specific process. Rosenthal fibers accumulate in glial
tical to Rosenthal fibers. It is not yet known whether scar tissue and glial tumors, both of which are condi-
defects in other genes may be responsible for some tions characterized by genesis of intermediate fila-
cases of AD. ments. They have been reported as a focal pheno-
GFAP is an intermediate filament protein that is menon in different types of glial tumors, multiple
expressed almost exclusively in astrocytes of the CNS. sclerosis, encephalomalacia, and syringomyelia. More
Intermediate filaments are intermediate-sized fi- widespread formation has been described in diffuse
brous cytoskeletal polymers, which together with gliomatosis, central pontine and extrapontine myeli-
smaller actin microfilaments and larger microtubules nolysis, vincristine therapy, radiation therapy, and
form the structural framework of the cytoplasm of all chronic inflammatory processes. Rosenthal fiber
eukaryotic cells. GFAP is the major intermediate fila- formation appears to reflect chronic pathological
ment protein of astrocytes. It appears to play a role in processes affecting astrocytes.
the outgrowth of processes of astrocytes. A marked There is evidence that myelin paucity is explained
increase in GFAP is part of the complex changes seen at least in part by disturbed myelination. In truly de-
in astrocytes after most types of CNS injury. Absence myelinating disorders, a macro- and microglial reac-
of GFAP leads to surprisingly few change in the un- tion of variable intensity is always found with evi-
challenged CNS, but if damage occurs, it is more se- dence of phagocytic activity and presence of prod-
vere in the absence of GFAP. For instance, experimen- ucts of myelin breakdown. In AD, no phagocytic
tal allergic encephalitis is more severe in GFAP transformation of macroglia and microglia is seen,
knock-out mice. On the other hand, accumulation of despite a conspicuous absence of myelin sheaths.
GFAP in astrocytes apparently leads to a stress re- There is a lack of histological and histochemical evi-
sponse that induces the small stress proteins aB-crys- dence for the presence of lipoid products of myelin
tallin and HSP27 and leads to the generation of breakdown. There is chemical evidence of a distur-
Rosenthal fibers. bance of myelin maturation. To stress the differences
Rosenthal fibers are inclusion bodies composed of between AD and the regular “myelinoclastic leuko-
intermediate filaments and the small stress proteins dystrophies,” the disease has been called a “dys-
aB-crystallin and HSP27. aB-Crystallin and HSP27 myelinogenic leukodystrophy.”
are both members of the so-called small heat shock The nature of the relationship between astrocytic
protein family. They are normally present in the brain abnormalities and myelin paucity, whether due to dis-
in small amounts and are water-soluble. The expres- turbed myelination, demyelination, or both, is un-
sion of these proteins is enhanced by various stress known. Astrocytes have multiple important func-
57.6 Magnetic Resonance Imaging 419

tions. They provide structural support within the ner- ally relatively late, in the stage of cystic degeneration
vous system, and they play a central role in regenera- and atrophy of the white matter.
tive repair. Astrocytic foot processes provide physical
and electrical insulation for synapses. They have an
important role in potassium distribution, preventing 57.5 Therapy
the accumulation of potassium in the extracellular
space during neuronal activity. They are involved in Treatment is entirely supportive. No causal therapy is
the metabolism of various neurotransmitters, and available.
probably have a reservoir function for nutrients. As-
trocytes and their interaction with oligodendrocytes
are a prerequisite for oligodendrocyte differentiation 57.6 Magnetic Resonance Imaging
and survival and for the deposition and maintenance
of myelin sheaths. There are gap junctions between In infantile AD, CT discloses bilateral, usually sym-
astrocytes and oligodendrocytes, which provide a metrical, moderately well-demarcated areas of re-
means of interaction.Astrocytes are the “third factor,” duced density in the frontal lobes with extensions to
allowing oligodendrocytes to myelinate axons and the temporal and parietal lobes and the external and
to maintain the myelin sheaths already deposited extreme capsules. The anterior limb of the internal
around axons. These data indicate that astrocytic dys- capsule may be involved. The subcortical arcuate
function in the immature brain of infants, in which fibers are involved in the process. Temporarily, the
myelin is still to be laid down, may have an adverse white matter abnormalities may show mass effect
effect on the process of myelination and myelin mat- with compression of the ventricles. A mild to moder-
uration, resulting in disturbed myelination and hy- ate enlargement of the lateral and third ventricles en-
pomyelination. In older patients astrocytic dysfunc- sues, caused either by atrophy or by hydrocephalus
tion may lead to disturbance of myelin maintenance, due to aqueduct stenosis. In all reported cases, the
resulting in demyelination. The reason why certain frontal white matter abnormalities are the most se-
astrocyte populations are more involved than others vere, and occipital white matter and cerebellum are
is not clear. completely or relatively spared. However, cerebellar
The megalencephaly in AD is caused by a combina- white matter may also become extensively involved.
tion of astrocytic hyperplasia and massive deposition In many cases a rim of normal or increased density is
of Rosenthal fibers. The subsequent atrophy and cyst seen in the subependymal region, including frontal
formation would be secondary to progressive astro- periventricular white matter, caudate nucleus, thala-
cytic cell death in association with loss of other ner- mus, hypothalamus, fornix, and the occipital periven-
vous tissue components. In neuroimaging the con- tricular white matter (Fig. 57.1). In some cases in-
trast enhancement is seen mainly in the frontal white creased density has also been reported in the subpial
matter, periventricular rim, basal ganglia, thalamus, cortical layers with a more patchy appearance. Con-
hypothalamus, and brain stem, which are the areas trast enhancement is seen in the areas of increased
with the highest Rosenthal fiber density. The contrast density (Fig. 57.1). Contrast enhancement has also
enhancement is probably caused by a defect in the been reported in the dorsal part of the brain stem.
blood–brain barrier related to impaired function to However, some patients show no contrast enhance-
astrocytic foot plates. The Rosenthal fibers are partic- ment, probably depending on the time of examina-
ularly present in astrocytic cell processes and foot tion, because the higher-density enhancing regions
plates, and these foot plates form an integral part of decrease as the disease progresses. The course of the
the blood–brain barrier. disease is characterized by volume loss. Cysts arise in
The topography of the pathological changes corre- the white matter, especially the frontal white matter,
lates with the clinical features. The frontal predomi- and atrophy ensues. The extent of the white matter
nance of the white matter abnormalities correlates abnormalities may increase, but the increase is usual-
with the frequently observed behavioral problems. ly not very prominent. Contrast enhancement is usu-
Most patients have epilepsy, which correlates with ally absent in late stages of the disease.
cortical involvement. Bulbar signs are prominent in In juvenile AD, CT findings include frontal white
AD patients, whereas brain stem involvement is al- matter hypodensity. Areas of increased density and
most invariably found in pathology and imaging. It is, contrast enhancement are less prominent than in in-
however, important to note that in juvenile and adult fantile AD. Over time, white matter atrophy occurs
AD the complete typical imaging picture is already and cysts may form.
present on early MRI studies obtained during the In adult AD CT abnormalities limited to the cere-
stage of minimal neurological dysfunction, and that bellar white matter and brain stem have been report-
the onset of neurological deterioration may be de- ed. The lesions may be space-occupying.
layed for many years. Spasticity and ataxia occur usu-
420 Chapter 57 Alexander Disease

Fig. 57.1. A CT scan without (first row) and with contrast (sec- trast. The same areas enhance with contrast. Courtesy of Dr. S.
ond row) in a 4-week-old baby boy with infantile AD. Note the Blaser, Department of Diagnostic Imaging, Hospital for Sick
increased density in the frontal white matter, a periventricular Children, Toronto, Canada
rim, and part of the basal ganglia on the images without con-

In infantile AD, MRI shows abnormal signal inten- 57.4). In addition, there are usually lesions in the
sity of the white matter in a symmetrical distribution brain stem, most often involving the mid brain and
with frontal predominance (Figs. 57.2–57.4). The ab- the medulla (Figs. 57.2–57.4). The hilus of the dentate
normal white matter has usually a swollen aspect, nucleus may have an abnormal signal. The fornix and
with broadening of gyri and stretching of the overly- optic nerves and chiasm may be thickened (Figs. 57.2
ing cortex (Fig. 57.4). However, in infantile cases it and 57.3). Contrast enhancement is often prominent
may be difficult or impossible to distinguish abnor- and involves the frontal white matter, ependymal lin-
mal white matter from unmyelinated white matter ing of the ventricles, the periventricular rim, the basal
and the frontal predominance may be evident only in ganglia, thalamus, dentate nucleus, brain stem le-
the degree of hyperintensity on T2-weighted images sions, fornix, and optic chiasm in variable combina-
or degree of hypointensity on T1-weighted images tions (Figs. 57.2, 57.4, 57.5 and 57.7). The frontal cor-
and the swelling (Figs. 57.2 and 57.3). In some infants tex may also show contrast enhancement (Fig. 57.4).
there is no evident frontal predominance of white Special features may be hydrocephalus due to aque-
matter abnormalities. There is a characteristic duct stenosis and major enlargements of a cavum
periventricular rim of low signal intensity on T2- septi pellucidi and cavum Vergae (Fig. 57.6). Sub-
weighted images and high signal intensity on T1- ependymal cysts may be seen at the level of the head
weighted images (Figs. 57.2–57.4). There are usually of the caudate nucleus. Over time, cavitation of the
prominent signal abnormalities and swelling of the frontal white matter may occur and may become
caudate nucleus, other basal ganglia, and thalamus prominent (Fig. 57.4). Atrophy of the affected white
(Figs. 57.2 and 57.3). These structures may have a matter, basal ganglia, thalamus, brain stem, and cere-
high signal on T1-weighted images (Figs. 57.3 and bellum occur (Fig. 57.4).
57.6 Magnetic Resonance Imaging 421

Fig. 57.2. MRI in the same boy as in Fig. 57.1, a few days later. fornix is thickened. The midbrain and medulla contain areas
The frontal white matter has at most a slightly abnormal signal of abnormal signal. After contrast (third row), the T1-weighted
intensity on the T2-weighted images; its signal intensity is images demonstrate enhancement of the periventricular rim,
close to normal for unmyelinated white matter. There is a frontal white matter, fornix, basal ganglia, and a central area in
periventricular rim of low signal intensity on the T2-weighted the thickened optic chiasm. Courtesy of Dr. S. Blaser, Depart-
images, most prominent in the frontal region. The head of the ment of Diagnostic Imaging, Hospital for Sick Children, Toron-
caudate nucleus is highly swollen and abnormal in signal. The to, Canada
422 Chapter 57 Alexander Disease

Fig. 57.3. MRI in a 7-week-old baby girl with infantile AD. The putamen have an abnormal signal intensity and are swollen.
frontal white matter has a higher signal intensity than normal The thalamus and large parts of the midbrain have an abnor-
for unmyelinated white matter on the T2-weighted images. mal signal. The fornix is thickened. From van der Knaap et al.
There is a rim of low signal on the T2-weighted images and (2001), with permission; additional images courtesy of Dr. S.
high signal on the T1-weighted images without contrast, ex- Springer, Department of Pediatrics, Ludwig Maximilian Univer-
tending into the frontal white matter.The caudate nucleus and sity, Munich, Germany

In juvenile AD, there is a frontal predominance of often seen in the midbrain (in the anterior part, the
the white matter abnormalities (Figs. 57.8–57.10). periaqueductal region, or the entire area except for
The occipital and temporal white matter may be the red nuclei and the colliculi) and the medulla
largely spared, but in some patients only a thin rim of (either in the central or the posterior part) (Fig. 57.9).
occipital arcuate fibers is spared. The abnormal The pontine tegmentum may be involved as well.
frontal white matter is usually involved throughout Contrast enhancement may involve the frontal white
and mildly swollen with some broadening of the gyri. matter, ependymal lining of the ventricles, the
However, in some patients the white matter changes periventricular rim, the basal ganglia, thalamus,
are restricted to the frontal periventricular region dentate nucleus, cerebellar cortex, brain stem lesions,
and there is no evident swelling of the abnormal and intraparenchymal trajectories of cranial nerves
white matter. There is a rim of low signal intensity on in variable combinations, but the enhancement is as a
T2-weighted images and high signal intensity on T1- rule much more subtle than in infantile AD (Figs. 57.9
weighted images (Figs. 57.8–57.11). The rim is often and 57.10). Over time cavitation of the frontal white
thin and discontinuous (Figs. 57.8–57.11). There are matter may occur (Figs. 57.10 and 57.12). The cysts
almost invariably some signal changes in the basal may become very large (Fig. 57.12), but in some pa-
ganglia and thalamus, with some swelling (Figs. 57.8, tients cysts are never seen. Invariably atrophy occurs
57.10 and 57.11). These structures may have a high of the affected white matter, the basal ganglia, and the
signal on T1-weighted images (Figs. 57.10 and 57.11). thalamus. The atrophic basal ganglia and thalamus
Very characteristic are the brain stem lesions, most have a high, normal, or low signal on T2-weighted
57.6 Magnetic Resonance Imaging 423

Fig. 57.4. The same girl as in Fig 57.3, now 3 months old. The normal signal. There is a thin periventricular rim of low signal
first and second rows show the T2-weighted images (left), T1- on T2-weighted images and high signal on T1-weighted im-
weighted images without contrast (middle) and with contrast ages, which enhances after contrast. Parts of the basal ganglia,
(right).The third row shows T2-weighted images at lower levels. frontal white matter and frontal cortex also enhance after con-
The frontal white matter now displays much more prominent trast. The fornix is thickened and enhances after contrast. The
signal abnormalities and has a swollen appearance. There are midbrain,hilus of the dentate nucleus,and medulla contain ar-
small cysts in the frontal white matter. The lateral ventricles eas of abnormal signal. From van der Knaap et al. (2001), with
have become much wider, probably due to a combination of permission; additional images courtesy of Dr. S. Springer,
white matter volume loss and hydrocephalus caused by aque- Department of Pediatrics, Ludwig Maximilian University,
duct stenosis. The basal ganglia are atrophic and have an ab- Munich, Germany
424 Chapter 57 Alexander Disease

Fig. 57.5. T1-weighted images with contrast in a 3-month-old There is cystic enlargement of the cavum septi pellucidi. The
girl (first row) and 6-month-old girl (second row) with infantile images of the second row demonstrate enhancement of a
AD.The images of the first row show contrast enhancement of periventricular rim, basal ganglia, and spots in the brain stem
spots in the frontal white matter, the basal ganglia, thickened and the colliculi
fornix, dorsal part of the midbrain, and ependymal lining.

Fig. 57.6. The T1-weighted sagittal (left) and coronal images wide; the cavum septi pellucidi is grossly dilated. Note the
(middle and right) of a 4-month-old girl with infantile AD small areas of high signal at the frontal horns, frequently seen
show large frontotemporal cystic areas. The third ventricle is in AD
57.6 Magnetic Resonance Imaging 425

Fig. 57.7. T2-weighted images (left),T1-weighted images with- high signal on T1-weighted images, which enhances after con-
out contrast (middle), and T1-weighted images with contrast trast. The rim is double, the inner rim being discontinuous.
(right) in an 18-month-old boy with late-infantile AD. The Note the small rings at the frontal horns, which enhance after
frontal white matter is abnormal in signal. The basal ganglia contrast.These small rings are often seen in AD. Courtesy of Dr.
and thalami are of mixed signal and are atrophic. There is a N. Thomas, Department of Pediatric Neurology, Southampton
periventricular rim with low signal on T2-weighted images and General Hospital, Southampton, UK

images (Fig. 57.9). The brain stem and cerebellum 3. Abnormalities of the basal ganglia and thalami,
may also become atrophic. The extent of the white either in the form of elevated signal intensity
matter abnormalities may increase over time, but the and some swelling or atrophy and elevated or de-
increase is usually not prominent. creased signal intensity on T2-weighted images
MRI is capable of suggesting the diagnosis with a 4. Brain stem abnormalities, in particular involving
high probability of accuracy, as demonstrated by a the mid brain and medulla
high correlation with de novo GFAP gene mutations. 5. Contrast enhancement involving one or more of
MRI criteria have been defined and four of the five the following structures: ventricular lining,
criteria have to be fulfilled for an MRI-based diagno- periventricular rim of tissue, white matter of the
sis. frontal lobes, optic chiasm, fornix, basal ganglia,
1. Extensive, symmetrical cerebral white matter ab- thalamus, dentate nucleus, cerebellar cortex, and
normalities with a frontal preponderance, either brain stem structures.
in the extent of the white matter abnormalities,
the degree of swelling, the degree of signal change, These criteria were designed to facilitate diagnosis in
or the degree of tissue loss (white matter atrophy typical AD patients. However, unusual MRI patterns
or cystic degeneration) have been reported in DNA-confirmed AD patients,
2. Presence of a periventricular rim of decreased sig- which do not fulfill the above criteria. In exceptional
nal intensity on T2-weighted images and elevated patients, the frontal white matter abnormalities are
signal intensity on T1-weighted images asymmetrical (Fig. 57.13). A patient with juvenile AD
has been reported, in whom cerebellar white changes
426 Chapter 57 Alexander Disease

Fig. 57.8. Series of T2-weighted images in a 5-year-old girl pearance.There is a thin periventricular rim of low signal.There
with juvenile AD.From these images the ventrodorsal gradient are small dark round rings at the frontal horns.The hilus of the
in white matter disease is evident. The basal ganglia have a dentate nucleus is affected. There are no clear brain stem ab-
slightly abnormal signal intensity and a somewhat swollen ap- normalities

and swelling were most prominent (Fig. 57.14). Over matter together with involvement of diencephalic nu-
time he also developed frontal white matter changes clei and brain stem tracts as well as contrast enhance-
(Fig. 57.15). Additionally, there are patients, as a rule ment may be seen in X-linked adrenoleukodystrophy.
with juvenile or adult AD, who exclusively or almost
exclusively have brain stem, cerebellar, or spinal cord
lesions in variable combinations (Figs. 57.16–57.19). 䊳

These lesions may be space-occupying, suggesting a Fig. 57.9. Series of T2-weighted (first and second rows) and
multifocal glioma (Figs. 57.18 and 57.19). They are contrast-enhanced T1-weighted images (third and fourth rows)
often asymmetrical. These lesions may show contrast in a 10-year-old boy with juvenile AD.There are extensive cere-
enhancement (Figs. 57.15 and 57.17–57.19). Few pa- bral white matter abnormalities with frontal predominance.
tients have been reported who only have atrophy of There is a thin, discontinuous periventricular rim of low signal
the lower part of the brain stem and upper spinal on the T2-weighted images. The basal ganglia are atrophic.
cord. None of these patients received contrast agent, There is a lesion in the midbrain and the dorsal medulla. Both
so that the pattern of enhancement cannot be as- the cerebellar hemispheric white matter and the hilus of the
sessed. dentate nucleus are abnormal, with the dentate nucleus
The typical AD MRI pattern is quite specific, dis- prominently visible in between. After contrast administration
similar from patterns observed in other white matter there is some enhancement of parts of the ependymal lining
disorders. Several leukoencephalopathies share some of the lateral ventricles, the lesions in the midbrain and medul-
of the MRI characteristics, but none shares all of la, the dentate nucleus, and the cerebellar cortex. From van der
them. Predominant involvement of the frontal white Knaap et al. (2001), with permission
57.6 Magnetic Resonance Imaging 427

Fig. 57.9.
428 Chapter 57 Alexander Disease

Fig. 57.10. An 8-year-old boy with juvenile AD. The first row and the signal abnormalities in the basal ganglia. The fornix is
contains a T2-weighted image (left) and two T1-weighted im- thickened. After contrast administration, enhancement of the
ages, one without contrast (middle) and one with contrast periventricular rim, basal ganglia, and frontal white matter is
(right), at the level of the basal ganglia. The second row con- seen. There are cysts of variable size in the frontal white mat-
tains T1-weighted images with contrast. Note the extensive ter. There is a large cavum Vergae. Courtesy of Dr. C. Leite, neu-
cerebral white matter abnormalities with frontal predomi- roradiologist, and Dr. F. Kok, pediatric neurologist, University of
nance, the periventricular rim with a low signal on the T2- São Paulo Medical School, São Paulo, Brazil
weighted image and high signal on the T1-weighted image,

However, in this disorder only or mainly the genicu- lidus are typically involved, with sparing of the cau-
late bodies are involved among the diencephalic nu- date nucleus and putamen. Contrast enhancement
clei. The brain stem lesions primarily involve the cor- does not occur. In merosin-deficient congenital mus-
ticospinal, corticobulbar, visual, and auditory tracts. cular dystrophy, extensive cerebral white matter
Contrast enhancement occurs within the outer bor- changes are present with relative sparing of the occip-
der of the white matter lesions. Similarly, some pa- ital white matter. However, the basal ganglia and brain
tients with metachromatic leukodystrophy have pre- stem are spared. In megalencephalic leukoencephalo-
dominantly frontal white matter abnormalities to- pathy with subcortical cysts, extensive cerebral white
gether with involvement of the brain stem. The brain matter changes are observed with slight swelling.
stem lesions involve the long tracts. Contrast en- However, there are invariably anterior temporal cysts,
hancement is not a feature of this disease. Canavan and often subcortical cysts in the frontoparietal area,
disease is characterized by a combination of macro- whereas the cysts in AD affect primarily the deep
cephaly, extensive cerebral white matter changes frontal white matter. Contrast enhancement is not a
(without frontal preponderance), and basal ganglia feature of megalencephalic leukoencephalopathy
abnormalities. However, the thalamus and globus pal- with subcortical cysts.
57.6 Magnetic Resonance Imaging 429

Fig. 57.11. One T2- (left) and one


T1-weighted image (right) in a 6-year-
old girl with juvenile AD, showing the
typical characteristics. Note the high
signal of the basal ganglia on the
T1-weighted image without contrast

Fig. 57.12. A 7-year-old girl with juvenile AD. Note the huge Knaap et al. (2001), with permission; additional images cour-
cysts in the frontal white matter. Note also the periventricular tesy of Dr. S. Naidu, Department of Neurogenetics, Kennedy
rim of low signal on the T2-weighted image. From van der Krieger Institute, Baltimore, USA

Fig. 57.13. This 5-year-old female with an unusual variant of her mother (father not investigated). The basal ganglia are
AD has highly asymmetrical signal abnormalities in the frontal mildly abnormal in signal and are mildly swollen,suggestive of
white matter. A brain biopsy was performed to rule out a low- AD.There are no brain stem lesions. Courtesy of Dr. N.Thomas,
grade glioma; Rosenthal fibers were found. Subsequently, a Department of Pediatric Neurology, Southampton General
mutation in the GFAP gene was demonstrated, not present in Hospital, Southampton, UK
430 Chapter 57 Alexander Disease

Fig. 57.14. A 3-year-old boy with an unusual variant of AD. His signal abnormalities in the periventricular white matter, most
cerebellum is highly enlarged, leading to obstructive hydro- prominent in the posterior region. A cerebellar biopsy re-
cephalus for which a ventriculoperitoneal shunt was placed. vealed Rosenthal fibers and a de novo mutation in the GFAP
The cerebellar white matter is abnormal in signal. He has mild gene was found
57.6 Magnetic Resonance Imaging 431

Fig. 57.15. The same boy as in Fig 57.14, now 12 years old. matter and hilus of the dentate nucleus display prominent sig-
He is neurologically remarkably stable. His cerebellum is nal abnormalities.The medulla contains lesions. After contrast
markedly reduced in size.There are periventricular white mat- administration, a lesion in the deep parietal white matter,
ter abnormalities with a frontal predominance.There are slight spots in the dentate nucleus, and the dorsal part of the medul-
signal abnormalities in the basal ganglia. The cerebellar white la show enhancement
432 Chapter 57 Alexander Disease

Fig. 57.16. A 32-year-old patient with adult AD. He had a the cerebellar white matter abnormalities and the brain stem
slowly progressive neurological course and died soon after atrophy. The lower brain stem is extremely thin. There are also
the MRI. Numerous Rosenthal fibers were found at autopsy areas of abnormal signal within the brain stem. The contrast-
and a mutation in the GFAP gene was found, not present in his enhanced T1-weighted image (third row, right) shows foci of
mother (father not investigated). Most prominent features are enhancement in the cerebellum
57.6 Magnetic Resonance Imaging 433

Fig. 57.17. An 11-year-old female patient with juvenile AD right and in the medulla. After contrast administration, en-
and a mutation in the GFAP gene.There are minimal supraten- hancement of the latter lesions is seen. The small rings at the
torial abnormalities. Some periventricular white matter abnor- frontal horns also show some enhancement. Courtesy of Dr. R.
malities are seen, most pronounced at the frontal horns. The Robinson, Department of Pediatric Neurology, Guy’s Hospital,
basal ganglia contain slight signal abnormalities. Several small London, UK
lesions are present in the middle cerebellar peduncle on the
434 Chapter 57 Alexander Disease

Fig. 57.18. A 14-year-old male patient with juvenile AD and a middle cerebellar peduncles. The hilus of the dentate nucleus
de novo GFAP mutation.There are no evident abnormalities in has an abnormal signal. After contrast administration, numer-
the supratentorial white matter.There are extensive multifocal ous foci of enhancement are seen.Courtesy of Dr.A.Reddy, De-
and confluent abnormalities in the midbrain, pons, medulla, partment of Hematology/Oncology, Children’s Health System,
and middle cerebellar peduncles with some mass effect of the Birmingham, Alabama, USA
57.6 Magnetic Resonance Imaging 435

Fig. 57.19. A 7-year-old boy with juvenile AD. He had Rosen- pons is mildly atrophic. After contrast, enhancement of the
thal fibers at histological examination and a de novo mutation fornix and the nodular lesions in the posterior fossa is seen.
in the GFAP gene. Apart from a mild dilatation of the lateral Courtesy of Dr. L. González Gutiérrez-Solana, Neuropediatrics
ventricles and a thickened fornix, the T2-weighted images do Unit, Hospital Niño Jesús, Madrid, Spain, and Dr. A. Messing,
not show supratentorial abnormalities, neither in the white Department of Pathobiological Sciences,Waisman Center and
matter nor in the basal ganglia. There are multiple nodular School of Veterinary Medicine, University of Wisconsin, Madi-
lesions in the brain stem and middle cerebellar peduncles.The son, Wisconsin, USA
Chapter 58

Giant Axonal Neuropathy

58.1 Clinical Features munostaining for glial fibrillary acidic protein. In ad-
and Laboratory Investigations dition, occasional giant axons are found, measuring
up to 100 mm or more in diameter. They are short,
Giant axonal neuropathy (GAN) is an early-onset globular to fusiform or balloon-like enlargements or
neurodegenerative disorder that affects both the PNS long cigar-like or corkscrew-shaped axonal thicken-
and CNS. The disease has an autosomal recessive ings. They stain positively with silver stains and anti-
mode of inheritance. Patients have characteristic neurofilament antibodies. The cerebral white matter
tightly curled hair, often described as frizzy or kinky. may be diffusely gliotic with a variable loss of myelin,
Acquisition of motor milestones is delayed in most, the frontal and parietal lobes being most affected and
but not all patients. The age at onset of progressive the U fibers being relatively spared. In other patients,
neurological problems is usually below 7 years. Pa- however, absence of myelin loss has been reported.
tients develop a clumsy gait and progressive weak- Rosenthal fibers are aggregated around blood vessels.
ness of the legs. The weakness spreads to involve the Low numbers of giant axons are seen within the cere-
arms as well. Neurological examination reveals signs bral white matter. The central gray matter structures
of peripheral neuropathy with absent reflexes, mus- also contain increased numbers of astrocytes, scat-
cular atrophy, weakness, and impaired sensation, tered Rosenthal fibers, and occasional giant axons.
most marked distally. There are usually also pyrami- Optic nerves and tracts show fiber loss and gliosis.
dal signs, with extensor plantar reflexes. Subsequent- The cerebellar cortex displays loss of Purkinje and
ly, dysarthria, nystagmus, ataxia, scoliosis, and intel- granule cells and hyperplasia of Bergmann astro-
lectual decline become apparent. Optic atrophy may cytes. In the cerebellar white matter, loss of nerve
occur. Patients may develop bulbar weakness, involv- fibers and increase of astrocytic processes and
ing muscles of the face, tongue, and palate. Seizures Rosenthal fibers is found. Giant axons and Rosenthal
may occur. Urinary retention and chronic constipa- fibers are scattered throughout the brain stem. In par-
tion may be vexing problems and are related to in- ticular, the pyramidal tracts are shrunken and gliotic,
volvement of peripheral autonomic nerves. Preco- display loss of nerve fibers, and contain many giant
cious puberty may occur. Most patients are of small axons. The spinal white matter contains excessive
stature. Patients usually become wheelchair-depen- numbers of astrocytes. There are subpial clusters of
dent in the first or second decade of life and die be- Rosenthal fibers. Giant axons are particularly numer-
tween the ages of 10 and 30 years. ous in the posterior columns, especially in the cer-
Neurophysiological examination reveals signs of a vical region, and the lateral corticospinal tracts,
severe axonal neuropathy. Somatosensory, visual, and especially in the lower thoracic and lumbar region.
brain stem auditory evoked potentials show pro- Electron microscopy of the axonal swellings reveals
longed conduction times or absence of reproducible enormous accumulations of neurofilament, often
responses. Sural nerve biopsy, showing axonal loss arranged in a whorl-like, interlacing pattern. Nonde-
and large axonal swellings, which consist mainly of script electron-dense granules are interspersed
tightly packed neurofilaments, supports the diagno- among filamentous masses. The myelin sheaths of the
sis. DNA confirmation is possible. enlarged axons are abnormally thin and disintegra-
tion of myelin lamellae may be seen.
Sural nerve biopsy reveals enlarged axons with
58.2 Pathology spindle-shaped focal distensions of non-myelinated
and thinly myelinated nerve fibers. Myelin sheaths
The brain is unremarkable on external examination. around the swellings are abnormally thin and the
On sectioning, some dilatation of the lateral ventri- largest swellings often lack a covering of myelin over
cles and thinning of the corpus callosum may be a part of their length. Ultrastructurally, the disten-
found. The cerebral and cerebellar white matter may sions are composed of closely packed neurofilaments,
be abnormal with sparing of the U fibers. which often form a whorl pattern. There are closely
Histological examination of the cortex reveals in- associated electron-dense granules. Microtubules,
creased numbers of astrocytes and scattered Rosen- microtubule–mitochondrial complexes, and cisterns
thal fibers. The Rosenthal fibers are positive in im- of smooth endoplasmic reticulum, instead of being
58.5 Magnetic Resonance Imaging 437

dispersed among the neurofilaments as in normal axonal degeneration and neuronal death found in
axoplasm, are frequently seen gathered together with- GAN patients point to the importance of gigaxonin
in the filament-free areas at the center or periphery of for neuronal survival.
the axonal swelling. In muscles, the typical pattern of
neurogenic atrophy is found.
In conclusion, the pathology is that of a distal 58.4 Therapy
axonopathy most severely affecting peripheral
nerves, pyramidal tracts, posterior spinal columns, No specific treatment is available. Therapy is support-
and the cerebellum. It is important to note that ive.
Rosenthal fibers have not been reported in all pa-
tients.
58.5 Magnetic Resonance Imaging

58.3 Pathogenetic Considerations The MRI findings described in GAN are variable,
probably at least partly depending on the stage of the
The gene related to giant axonal neuropathy, GAN, is disease. In some patients, no or minimal cerebral
located on chromosome 16q24.1 and encodes a ubi- white matter abnormalities are present (Fig. 58.1).
quitously expressed protein, gigaxonin. The white matter abnormalities may be diffuse and
Giant axonal neuropathy is characterized by cyto- subtle (Fig. 58.3), sometimes with multifocal spots of
skeletal abnormality. The hallmark of the disease is more prominent signal change superimposed. In oth-
the presence of giant axonal swellings, which are er patients, extensive and confluent white matter ab-
densely packed with aberrant neurofilaments, abnor- normalities are present, symmetrically involving the
mal microtubule network, and accumulation of other cerebral white matter in a diffuse fashion (Fig. 58.2)
membranous organelles. Neurofilaments belong to or with a predominance in the frontoparietal region.
the intermediate filaments. In GAN, an abnormal ac- The corpus callosum and U fibers tend to be spared.
cumulation of multiple tissue-specific intermediate The posterior limb of the internal capsule, pyramidal
filaments is found in a wider range of cells than only tracts, and medial lemniscus in the brain stem, mid-
neurons, suggesting a generalized disorganization of dle cerebellar peduncles, hilus of the dentate nucleus,
intermediate filament networks. Aggregations of and cerebellar white matter may display signal
vimentin have been reported in endothelial cells, changes (Figs. 58.2 and 58.3). The white matter ab-
Schwann cells, and cultured skin fibroblasts, and ag- normalities are progressive over time (Figs. 58.1 and
gregations of glial fibrillary acidic protein are found 58.2). The lateral ventricles become mildly dilated due
in astrocytes. Keratin intermediate filaments are to white matter volume loss and the cerebellar vermis
altered, leading to the characteristic kinky hair. becomes atrophic over time.
The cytoskeletal network is responsible for cell ar- In our experience the MR images in GAN have a
chitecture, intracellular transport, mitosis, cell mobil- certain resemblance to the images in Alexander dis-
ity, and differentiation. It is composed of micro- ease, which is interesting since both are characterized
tubules, actin microfilaments, and intermediate by Rosenthal fiber deposition. The basal ganglia
filaments, which interconnect through cross-linking may have a slightly abnormal signal and a slightly
proteins. The properties of the network formed are swollen aspect (Figs. 58.1 and 58.3). The areas with
modulated by different associated proteins. Cyto- high signal on T1-weighted images include the
skeletal organization and dynamics depend on pro- ependymal lining (Fig. 58.1), a thin periventricular
tein self-associations and interactions with a variety
of binding partners such as microtubule-associated
proteins (MAPs).
Gigaxonin binds directly to microtubule-associat- 䊳

ed protein 1B light chain (MAP1B-LC), a protein in- Fig. 58.1. A 6-year-old female patient with GAN. The T2-
volved in maintaining the integrity of cytoskeletal weighted images (upper two rows) show a diffuse slight signal
structures and promoting neuronal stability. The in- abnormality of the cerebral white matter. The basal ganglia
teraction of gigaxonin with MAP1B-LC enhances have a slightly abnormal signal and slightly swollen appear-
microtubule stability, required for axonal transport ance. The T1-weighted images (third row) show that the
over long distance. In line with this, the neurofilament ependymal lining of the lateral ventricles and the globus pal-
accumulations in GAN, leading to the segmental lidus have a slightly increased signal. After contrast adminis-
distension of axons, mainly affect distal portions tration (fourth row),subtle enhancement of the ependymal lin-
of the long tracts. Some of the mutations found in ing is seen. Courtesy of Dr. S. Blaser, Department of Diagnostic
GAN patients have been shown to lead to loss of Imaging, Hospital for Sick Children, Toronto, Canada.
gigaxonin–MAP1B-LC interaction. The devastating (Fig. 58.1 see next page)
438 Chapter 58 Giant Axonal Neuropathy

Fig. 58.1.
58.5 Magnetic Resonance Imaging 439

Fig. 58.2.
440 Chapter 58 Giant Axonal Neuropathy

rim (Fig. 58.3), and the globus pallidus (Figs. 58.1 and
58.3). The thin periventricular rim may have a low
signal on T2-weighted images (Fig. 58.3). The ependy-
mal lining shows subtle contrast enhancement
(Figs. 58.1 and 58.3). Areas in the dorsal medulla may
also show contrast enhancement (Fig. 58.2), which is
often seen in Alexander disease.

䊴 䊳

Fig. 58.2. At the age of 14 years, diffuse cerebral signal abnor- Fig. 58.3. The T2-weighted images (upper two rows) of her
malities are seen in the same girl, sparing the corpus callosum brother, who also suffers from GAN, at the age of 3 years, show
and to some extent the U fibers. The posterior limb of the in- diffuse slight signal abnormalities in the cerebral white matter
ternal capsule, pyramidal tracts and medial lemniscus in the with sparing of the U fibers and corpus callosum. The posteri-
brain stem, hilus of the dentate nucleus, and cerebellar white or limb of the internal capsule, pyramidal tracts and medial
matter are also involved. The contrast-enhanced T1-weighted lemniscus in the brain stem, hilus of the dentate nucleus, and
images reveal contrast uptake in multiple spots with the cere- cerebellar white matter are also involved.There is a thin rim of
bellum, pons, and the dorsal part of the medulla. Courtesy of low signal around the lateral ventricles on the T2-weighted
Dr. S. Blaser, Department of Diagnostic Imaging, Hospital for images, most evident in the posterior region. This rim has an
Sick Children, Toronto, Canada. (Fig. 58.2 see last page) increased signal on T1-weighted images (third row) and en-
hances after contrast (fourth row).The globus pallidus also has
a slightly increased signal on T1-weighted images (third row).
Courtesy of Dr. S. Blaser, Department of Diagnostic Imaging,
Hospital for Sick Children, Toronto, Canada.
58.5 Magnetic Resonance Imaging 441

Fig. 58.3.
Chapter 59

Megalencephalic Leukoencephalopathy
with Subcortical Cysts

59.1 Clinical Features for a few years or never achieve independent walking.
and Laboratory Investigations Some patients have a more benign clinical course. As
a teenager they may have normal mental and motor
Megalencephalic leukoencephalopathy with subcorti- function and only have borderline macrocephaly.
cal cysts (MLC) is a rare leukoencephalopathy with an Some patients still walk independently in their for-
autosomal recessive mode of inheritance. The disease ties.As the disease has been known for only a relative-
is relatively prevalent among Turkish people and in a ly short time, little information exists about average
certain Asian-Indian community, the Agarwal ethnic life span. Some patients have died in their teens or
group. Macrocephaly is present at birth or, more twenties, but others are alive in their forties.
frequently, develops during the first year of life. The In extensive laboratory investigations no abnor-
degree of macrocephaly is variable and is impressive malities are found. CSF protein is normal. Peripheral
in some of the patients. After the first year of life the motor and sensory nerve conduction velocities and
head growth rate normalizes and growth follows a electromyogram are normal. Evoked potentials are
line parallel to the 98th percentile, usually several initially normal in most patients. BAEPs remain nor-
centimeters above it. Initial mental and motor devel- mal, but VEPs and SSEPs deteriorate over the years
opment is normal in most cases, mildly delayed in with prolongation of latencies and abnormal cortical
some of the patients. Apart from progressive macro- responses. EEG is initially normal. Subsequently,
cephaly, the first clinical sign is usually a delay in background slowing occurs and abnormalities are
walking. Walking is often unstable and the child falls seen such as sharp waves, spikes, and spike-wave
frequently. After an interval of several years, a slow complexes with variable location. Abnormal photo-
deterioration of motor function is noted with devel- paroxysmal responses are seen in some of the pa-
opment of axial ataxia and ataxia of the extremities. tients but not in all.
Signs of pyramidal dysfunction are late and minor, Presently, the diagnosis is MRI-based. Analysis of
and are as a rule dominated by the signs of cerebellar the MLC1 gene can be performed, and abnormalities
ataxia. Muscle tone tends to be low, apart from some are found in 60–70% of the patients with typical MRI
ankle hypertonia. Reflexes become high and Babinski findings. Some families without mutations in MLC1
signs become apparent. Gradually the ability to walk exclude linkage to the MLC1 locus, implying that
independently is lost, and most children become there must be another gene or genes related to MLC.
completely wheelchair-dependent at the end of the Prenatal diagnosis is possible in families with known
first decade or in the second decade of life. Almost all mutations in MLC1.
patients have epilepsy from early on, usually easily
controlled with medication. Mental deterioration is
late and mild. Decreasing school performance be- 59.2 Pathology
comes evident during the later years of primary
school. In a minority of patients, intellectual capaci- Brain biopsy was performed in one patient (van der
ties are mildly decreased from the beginning. A rela- Knaap et al. 1996). The cortex was normal. A status
tively late and slow decrease in intelligence is also spongiosus of the cerebral white matter was found
noted in these patients. Speech becomes increasingly with presence of innumerable vacuoles. Myelin
dysarthric and the patients may also develop dyspha- sheaths had a normal thickness and density and there
gia. Some patients display extrapyramidal movement were only minor signs of myelin breakdown. The
abnormalities with dystonia and athetosis. Minor white matter showed intense fibrillary astrogliosis.
head trauma has been reported to induce temporary Electron microscopy revealed splitting of myelin
deterioration in some patients, most often with sheaths at the intraperiod line with intramyelinic vac-
seizures or status epilepticus, prolonged uncon- uole formation. The splitting involved only the outer
sciousness, or acute motor deterioration with gradual lamellae of the myelin sheaths; the vacuoles were cov-
improvement. ered by only one or two myelin layers. There was no
Some patients have a more severe clinical course evidence of axonal degeneration or loss. The cerebral
and maintain the ability to walk independently only cortex was normal.
59.5 Magnetic Resonance Imaging 443

59.3 Pathogenetic Considerations show white matter abnormalities on MRI. Especially


the MRI pattern of merosin-deficient congenital
There is one gene known to be related to MLC: MLC1, muscular dystrophy is very similar to that of MLC.
formerly called KIAA0027, located on chromosome Myelin vacuolation is seen in both MLC and merosin-
22qtel. Mutations in MLC1 are found in approximate- deficient congenital muscular dystrophy. Another
ly 70% of the MLC patients. Different mutations possibility is that MLC1 is involved in transporting
have been found. Within the Agarwal ethnic group, molecules that are essential for oligodendrocytes or
the patients are homozygous for the same founder myelin. Mutations in MLC1 may affect myelin sheath
mutation. Among Turkish patients, however, different formation or compaction. An argument in favor of
mutations are found and there is no evidence for a this possibility is the fact that the rapidly increasing
founder mutation among them. There is no evident macrocephaly of MLC develops during the period of
genotype–phenotype correlation. Some families most rapid myelin formation, the first year of life,
without mutations in MLC1 exclude linkage to the whereas head growth rate slows down and becomes
MLC1 locus, implying that there must be another normal in the second year of life. It is probable that
gene or other genes related to MLC. the myelin vacuoles arise during the myelin deposi-
MLC1 encodes a membrane protein of unknown tion. In agreement with this, Schmitt et al. (2003)
function with eight transmembrane domains. The show that MLC1 mRNA expression in the murine
MLC1 protein is highly conserved throughout evolu- brain peaks during a transitional period of astrocytic
tion in a variety of different vertebrate species that specialization that occurs in white matter during
produce myelin. Orthologues are not found in eu- myelination, suggesting a role for MLC1 in myelin
karyotes that do not produce myelin, suggesting an as formation.
yet undiscovered fundamental function of MLC1 re- The white matter has a very abnormal aspect on
lated to myelin. Sequence analysis of the MLC1 pro- MRI at an early age (the youngest child was 1.5 years
tein does not reveal any similarity to known proteins old when imaging was performed), when neurologi-
or protein domains. Most proteins with eight trans- cal examination is still normal or near-normal and
membrane domains have a transport function. Both evoked responses are normal. Thus, despite its highly
the C- and the N-terminus of MLC1 are located with- abnormal appearance, the white matter is functional-
in the cytoplasm. MLC1 is expressed in the brain and ly intact or largely intact during these early years. Ap-
all types of blood leukocytes. Within the brain, the parently the presence of the vacuoles in the outer part
MLC1 protein is specifically expressed in astrocytic of the myelin sheaths permits normal function. Over
endfeet in perivascular, subependymal, and subpial the years, MRI shows some decrease in white matter
regions. Astrocytic endfeet form part of the blood– swelling and some enlargement of lateral ventricles
brain barrier. This localization suggests a possible and subarachnoid spaces, a process accompanied by
role for MLC1 in a transport process across the clinical deterioration. The basis of the deterioration is
blood–brain barrier. The blood–brain barrier is a dif- not known.
fusion barrier for the exchange of molecules and is The gene KIAA0027/MLC1 has also been implicat-
formed by seamlessly joined endothelial cells of the ed in catatonic schizophrenia. However, this has so far
capillary wall, the basal lamina, and astroglial end- not been substantiated. Among patients and family
feet. The role of astrocytes in the blood–brain barrier members known to be carriers, the incidence of psy-
is not well defined. They are separated from the chiatric illness is not increased.
endothelial cells by the basal lamina and do not con-
tribute directly to the physical barrier. Perivascular
astroglial endfeet contain many transport proteins, 59.4 Therapy
including transporters of monocarboxylates, glucose,
glutamate, and water. So far all attempts at treatment have failed. Patients
Given the presence of vacuoles in the outer layer of have been treated with diuretics and acetazolamide,
the myelin sheaths in MLC, the role of astrocytic end- but neither the clinical symptoms nor the white matter
feet in water homeostasis in the brain is of special swelling improved. Treatment with creatine monohy-
interest. Different water channel proteins or aquapor- drate did not lead to objective improvement either.
ins are present in the brain. Aquaporin-4 shows a lo-
calization in the astroglial endfeet similar to MLC1.
Aquaporin-4 is associated with the dystrophin-asso- 59.5 Magnetic Resonance Imaging
ciated glycoprotein complex (DAGC). This complex
links the actin cytoskeleton to the extracellular ma- In MLC the cerebral hemispheric white matter is dif-
trix. Interestingly, some congenital muscular dystro- fusely abnormal and swollen (Figs. 59.1–59.6). The
phies, which are caused by mutations in members of swelling is most marked during the first years of life,
the dystrophin-associated glycoprotein complex, also with obliteration of peripheral CSF spaces and nar-
444 Chapter 59 Megalencephalic Leukoencephalopathy with Subcortical Cysts

Fig. 59.1.
59.5 Magnetic Resonance Imaging 445

Fig. 59.1. (continued). MRI series of a 6-year-old girl with MLC. callosum is largely spared. The posterior limb of the internal
The upper row of T1-weighted sagittal images show the capsule contains two lines of abnormal signal with a line of
swollen white matter and the cysts in the frontal and temporal normal signal in between. The midbrain, pontine tegmentum,
region. The U fibers in the posterior region of the cerebral pyramidal tracts in the basis of the pons, and cerebellar white
hemispheres, corpus callosum, brain stem, and cerebellum are matter are mildly abnormal in signal but not swollen. The
relatively normal in signal intensity. The second and third rows fourth and fifth rows contain the proton density images, which
shows these features on axial T2-weighted images. A large reveal the many subcortical cysts
cavum septi pellucidi and cavum Vergae are visible.The corpus

Fig. 59.2. Three ADC maps of the same patient as in Fig. 59.1. ADC values are normal in the basal ganglia (0.74–0.94 ¥
ADC values are highly elevated within the cerebral hemispher- 10–3 mm2/s) and mildly elevated within the cerebellar white
ic white matter, for instance 2.06–2.47 ¥ 10–3 mm2/s in the matter (1.10–1.32 ¥ 10–3 mm2/s)
frontal white matter (normal white matter ADC 0.85–0.95).

rowing of ventricles (Fig. 59.1). In older children and white matter. The external and extreme capsules are
adults the hemispheric white matter swelling is less prominently involved. The central white matter struc-
severe, peripheral CSF spaces are more prominently tures, including the corpus callosum, anterior limb of
visible, and the ventricular system becomes enlarged the internal capsule, posterior limb of the internal
(Fig. 59.3 and 59.4). On FLAIR images parts of the capsule (in part), and a periventricular rim of occipi-
cerebral white matter may have a slightly lower signal tal white matter are relatively spared. The posterior
intensity than the remainder of the abnormal white limb of the internal capsule shows either a double line
matter, related to the very high water content of the of abnormal signal intensity over its whole length
446 Chapter 59 Megalencephalic Leukoencephalopathy with Subcortical Cysts

Fig. 59.3. An 18-year-old boy with MLC. The cerebral white from the posterior limb of the internal capsule. There are mild
matter is diffusely abnormal and mildly swollen, but less so signal abnormalities in the cerebellar white matter. There are
than in the previous patient. Note again the sparing of the cor- cystic lesions in the anterior temporal and frontal regions
pus callosum and the presence of a double line of high signal

(Figs. 59.1 and 59.3) or a signal abnormality mainly in seen in the brain stem, especially the pyramidal tracts
its distal part (Fig. 59.4). Some sparing of arcuate (Fig. 59.1). There are almost invariably subcortical
fibers may be seen, most often in the occipital area cysts in the anterior temporal region, often also in the
(Figs. 59.3 and 59.4). In the majority of patients, the frontal and parietal subcortical regions (Figs. 59.1
cerebellar white matter shows only a mild abnormal- and 59.3–59.6). The cysts are bilateral. So far, the
ity in signal intensity and no swelling (Figs. 59.1, 59.3 anterior temporal cysts have only been found to be
and 59.4). In some patients minor abnormalities are lacking in some young infants and children with
59.5 Magnetic Resonance Imaging 447

Fig. 59.4. A 20-year-old woman, showing that some atrophy has now developed

MLC. The cysts tend to become larger with age and Since the gene for the disease has been found, less
may increase in number. In some patients they be- prominent white matter abnormalities have been
come very large (Fig. 59.5). The signal intensity of the documented in patients with clinically mild disease
contents of the cysts is always similar to that of CSF. In (Fig. 59.6). Early MRI may show the typical diffuse
accordance with this, T1 and T2 of cyst contents and cerebral white matter abnormalities with swelling
CSF are also similar. A patent and enlarged cavum (Fig. 59.6a), whereas follow-up MRI may show that
septi pellucidi and cavum Vergae are often present the cerebral white matter now has a normal signal
(Figs. 59.1 and 59.3). Cortical gray matter structures intensity in large areas and is less swollen (Fig. 59.6b).
and basal nuclei are always normal. It is mainly the subcortical white matter that remains
448 Chapter 59 Megalencephalic Leukoencephalopathy with Subcortical Cysts

Fig. 59.5. A 5-year-old girl with MLC.The subcortical cysts are huge. Courtesy of Dr. J. Wolf, Biochemical Genetics Clinic, Universi-
ty of Wisconsin–Madison, Madison, Wisconsin, USA

abnormal. Still, the characteristic subcortical cysts Similar white matter changes with swelling have
are present. been reported in Canavan disease, Alexander disease,
MRS has revealed a reduction of all signals per vol- L-2-hydroxyglutaric aciduria, and merosin-deficient
ume, indicative of the high water content of the brain. congenital muscular dystrophy. However, in Canavan
Relative to creatine, N-acetylaspartate is decreased disease, as a rule, MRI demonstrates additional
and myo-inositol is increased, suggesting axonal involvement of the thalamus and globus pallidus,
damage and gliosis. Diffusion tensor imaging shows not found in MLC patients. Special MRI findings in
increased ADC values in the cerebral white matter Alexander disease are a more prominent sparing of
and reduced anisotropy (Fig. 59.2). parieto-occipital white matter and often also sparing
59.5 Magnetic Resonance Imaging 449

Fig. 59.6 a. Girl with DNA-confirmed MLC. The first MRI (a) of abnormalities (a), the white matter looks much more nor-
was obtained at the age of 5 years; the second (b) at the age mal than usual for MLC on the follow-up MRI (b).The subcorti-
of 11 years. Whereas the first images show a classical pattern cal cysts, however, remain present

of the U fibers throughout. Basal ganglia and brain nuclei, and severe atrophy of the cerebellar vermis,
stem structures are typically involved. Contrast en- not observed in MLC. The MRI abnormalities ob-
hancement of certain brain structures is typically served in merosin-deficient congenital muscular dys-
present. Cavitation starts in the deep frontal white trophy are very similar to those observed in MLC.
matter. None of these features is present in MLC. In Subcortical cysts were observed in one patient with
L-2-hydroxyglutaric aciduria MRI shows additional a later-onset variant of muscular dystrophy with
involvement of caudate nuclei, putamen, dentate leukoencephalopathy and swelling.
450 Chapter 59 Megalencephalic Leukoencephalopathy with Subcortical Cysts

Fig. 59.6 b. (continued).


Chapter 60

Congenital Muscular Dystrophies

60.1 Clinical Features months following birth, the infants show profound
and Laboratory Investigations mental and motor retardation with rarely any devel-
opment beyond the newborn level. However, the clin-
Congenital muscular dystrophies (CMD) are a het- ical picture varies considerably, even within the same
erogeneous group of congenital myopathies that are sibship. Survival varies from the neonatal period to
hereditary and often progressive. They are often asso- over 5 years, but most children die within the first
ciated with abnormalities of the brain and eyes. Only year of life.
those types that are associated with significant white In Japan, FCMD is the second most frequent of the
matter abnormalities are discussed in the present muscular dystrophies, Duchenne muscular dystrophy
chapter. being the most frequent. The disease is almost exclu-
1. Fukuyama congenital muscular dystrophy (FCMD) sively reported in Japan. Inheritance is autosomal re-
2. Muscle–eye–brain disease (MEB) cessive. Onset of clinical symptoms is in the neonatal
3. Walker–Warburg syndrome (WWS) or early infantile period with marked hypotonia and
4. Merosin-deficient congenital muscular dystrophy hypokinesia. Motor development is delayed to a vari-
(MDC1A) able degree. In most patients motor functions are ac-
5. MDC1C quired gradually and the maximum motor develop-
6. MDC1D ment has been reached by the age of 2–8 years. The
majority of patients never manage to stand or walk;
WWS has many alternative names, including Walker’s the highest developmental level is usually crawling on
lissencephaly, Warburg syndrome, cerebro-oculo- hands and knees. The distribution of affected muscles
muscular syndrome, cerebro-ocular dysplasia–mus- is generalized, but proximal muscles are slightly more
cular dystrophy syndrome, and HARD±E syndrome severely affected than distal muscles. The facial mus-
(hydrocephalus, agyria, and retinal dysplasia with or cles are also affected, resulting in a hypotonic facial
without encephalocele). The disease has an autoso- expression. Muscular atrophy is prominent. Pseudo-
mal recessive mode of inheritance. Severe neurologi- hypertrophy of the calves is found in some patients.
cal dysfunction is evident from birth onwards. Hypo- After the age of about 8 years, motor functions grad-
tonia is profound and neonatal reflexes are often poor ually deteriorate. Joint contractures are not usually
or absent. In about 40% of the patients congenital found in the neonatal period, but flexion contractures
contractures are present. The affected neonates are of hips, knees, and elbow joints, limited anteflexion of
immobile and unreactive. In many patients progres- the cervical spine, and contractures of the joints of
sive hydrocephalus is evident from birth onwards the hands develop consecutively during the first few
with macrocephaly and bulging fontanel. Shunting years of life. There is invariable involvement of the
may be required. In other patients hydrocephalus is CNS, often with microcephaly, and always with severe
not evident at birth but develops soon afterwards. A mental retardation, which is not progressive. Seizures
few patients have microcephaly. About 30% of the occur in more than half the patients, usually in the
patients have an occipital meningocele or encephalo- form of generalized tonic–clonic convulsions. In a
cele. Epileptic seizures occur frequently. Ophthalmo- minority, infantile spasms are found. Ophthalmologi-
logical abnormalities are multiple and diverse and in- cal abnormalities are present in about half of the pa-
clude defects of the anterior and posterior chambers: tients and include strabismus, nystagmus, optic nerve
corneal opacities, microcornea, megalocornea, iris pallor, mild to severe myopia, cataract, and, less often,
atrophy, iris coloboma, iridolental synechiae, narrow chorioretinal degeneration and retinal vascular ab-
iridocorneal angle with or without glaucoma and normalities. The average life span in Fukuyama type
buphthalmos, cataract, persistent hyperplastic pri- CMD is estimated to be about 12 years; patients rarely
mary vitreous, chorioretinal coloboma, retinal dys- live beyond the age of 20. However, with respiratory
plasia, retinal detachment, optic disc hypoplasia, op- support patients may live beyond this age. In a few
tic disc coloboma, and unilateral or bilateral mi- cases FCMD is more severe and resembles WWS in
crophthalmia. Some patients have cleft palate and phenotype. In these cases patients have more severe
cleft lip. Genital abnormalities including small penis muscular weakness, never achieving head control or
and undescended testes are common in males. In the the ability to sit without support, they may have pro-
452 Chapter 60 Congenital Muscular Dystrophies

gressive hydrocephalus requiring shunt implantation, gression of weakness may be noted at the end of the
and they have more severe ophthalmological abnor- first decade, with death between 10 and 20 years.
malities. However, survival beyond 20 years has been de-
Most reports concerning MEB come from Finland. scribed. Some patients have a milder presentation,
The disease has an autosomal recessive mode of in- related to partial merosin deficiency. Patients have
heritance. Clinical symptoms are similar to those of later-onset slowly progressive muscle weakness,
WWS, but tend to be milder. Muscle hypotonia and mainly limb-girdle type, and achieve ambulation.
poor visual contact are noted in the neonatal or early Rarely, they may display signs of CNS dysfunction.
infantile period. The typical facial appearance is char- Some patients are adults and (still) asymptomatic.
acterized by a relatively large head with a high and A new CMD syndrome has been defined, designat-
prominent forehead, wide fontanel, flat midfacies, ed MDC1C. The patients present soon after birth with
short nose, and short philtrum. In about half of the hypotonia and severe weakness. They have weakness
patients there are some signs of hydrocephalus dur- and wasting of the shoulder girdle muscles, and
ing the first year of life, with a mildly increased head weakness and hypertrophy of the leg muscles. In
growth rate, but shunt implantation is required in some patients, cognitive development is normal, as is
only some of the patients. Motor development is vari- MRI of the brain, whereas other patients display men-
ably but generally severely retarded. Some of the pa- tal retardation and, on MRI, cerebral white matter ab-
tients show hardly any developmental progress, normalities and subcortical cerebellar cysts. MDC1C
whereas others achieve sitting without support at the is allelic with limb-girdle muscular dystrophy 21
age of 10 years and walking with support after more (LGMD21), a much milder muscular disease. MDC1C
than 10 years of life. Muscle weakness is generalized, and LGMD21 may occur in the same family.
but in the extremities weakness is slightly more Another new CMD syndrome has been designated
prominent in the proximal muscles. Facial muscles MDC1D. Apart from a congenital muscular dystro-
are usually not involved. Contractures develop gradu- phy, mental retardation is present. The phenotypic
ally and not in every patient. There is always involve- variability still has to be defined.
ment of the CNS with marked mental retardation. In addition to the above, more or less well-delin-
However, some patients do acquire the ability to eated syndromes, there are many patients remaining
speak. Most patients develop seizures, and occasion- who have an unclassified CMD. Some of these pa-
ally infantile spasms. Ophthalmological examination tients have merosin deficiency; most of them do not.
typically reveals severe visual failure with severe my- Many of these patients do not have accompanying
opia. Additional ocular signs include glaucoma, reti- brain abnormalities, but some do. Some of the pa-
nal degeneration, choroidal hypoplasia, optic nerve tients in this group of unclassified CMD cases have
pallor, and cataract. Between the ages of 5 and 25 white matter abnormalities. Some patients have white
years progress of psychomotor development ceases in matter abnormalities similar to MDC1A, whereas
many patients and deterioration sets in with loss of muscle merosin staining is normal. Other patients
mental and motor abilities and development of signs have more limited white matter abnormalities. Still
of spasticity, particularly in the legs. The age at death others have cerebellar abnormalities, brain stem ab-
is highly variable: some patients die at the age of 6 normalities, or gyral abnormalities. The classification
years, whereas others survive until their fifties. and basic defects in these patients remain to be eluci-
In Europe, merosin-deficient CMD (MDC1A) is the dated.
commonest form of CMD. Inheritance is autosomal In laboratory examinations, a moderately in-
recessive. Generalized hypotonia is noted in infancy creased CK level is consistently found in almost all
with a delay in motor development. Most children do patients. However, exceptional patients may have a
not acquire independent ambulation. Muscle weak- normal CK level, and, therefore, a normal CK value
ness and atrophy are severe and generalized and in- does not rule out the presence of a CMD. Patients with
clude facial weakness. Contractures are usually pre- a partial merosin deficiency have lower CK than clas-
sent, either at birth or later in life. Most children have sical MDC1A patients, who have markedly increased
a normal intelligence, some have a mild intellectual CK. Electromyography reveals signs of myopathy. In
impairment, but severe mental deficiency is rare. MDC1A, peripheral nerve conduction velocities are
Some of the patients have macrocephaly. Some chil- decreased. Muscle biopsy contributes to the diagnosis
dren develop seizures. Eyes and visual function are of CMD, revealing dystrophic changes. These include
normal. Additionally, there may be cardiac involve- marked variation in fiber size with presence of at-
ment that is usually subclinical, rarely clinically evi- rophic and hypertrophic fibers, fiber necrosis with
dent. There is neurophysiological evidence of periph- presence of phagocytes, increased number of fibers
eral nerve involvement with reduced motor and sen- with internal nuclei, increased interstitial fibrosis,
sory nerve conduction velocities, but clinically the and replacement by adipose tissue. No inflammatory
neuropathy is overshadowed by the myopathy. Pro- infiltration is found, with the exception of some pa-
60.2 Pathology 453

tients with MDC1A, in whom the findings may be Factors contributing to ventricular enlargement are
suggestive of myositis. There are no significant histo- aqueduct stenosis, disturbed fluid dynamics associat-
logical differences between the different CMD vari- ed with the Dandy–Walker malformation, and block-
ants. However, the use of different antibodies con- age of the arachnoid granulations by fibroglial tissue.
tributes to establishing the correct diagnosis. In The cerebral cortex is abnormally thick with absent
MDC1A there is isolated negative (usually complete- white matter interdigitations or with irregular, shal-
ly negative) immunostaining for laminin-a2 in mus- low indentations in the otherwise smooth cortical
cle. In some patients with a milder phenotype a par- ribbon. On microscopic examination the cortex is se-
tial deficiency is found. Skin biopsy is also useful to verely disorganized, with no recognizable lamination
assess the laminin-a2 status of a patient. In FCMD, and widespread disruption by gliofibrillary bundles
MEB, MDC1C, and MDC1D muscle, severely de- accompanying vessels from the pial surface.Agyria or
creased immunostaining for a-dystroglycan and a pachygyria with severe disorganization of the cortex
variable but less severe reduction for laminin-a2 are and absence of lamination is called lissencephaly type
observed. In WWS immunostaining for a-dystrogly- II. Neuronal heterotopias are present scattered in the
can is decreased, but immunostaining for laminin-a2 white matter and in the subependymal region. The
tends to be normal, although decreased immunos- white matter is reduced in volume, poorly myelinated,
taining has also been reported. It is important to real- gliotic, spongy, and often strikingly edematous with
ize that there are several antibodies against different occasionally cavitations. Myelination is virtually ab-
epitopes of laminin-a2. The results of immunostain- sent in some cases. In the brain stem corticospinal
ing may be different with different antibodies. DNA- tracts are grossly absent. The pontine nuclei and mid-
based prenatal diagnosis is possible in families with a dle cerebellar peduncles are usually absent. Within
known mutation in one of the genes related to the the smooth, afoliar cerebellar cortex microscopic
CMDs. changes are found similar to those in the cerebral cor-
tex, but the abnormalities are less severe, as some
organization into layers is usually present. Cerebellar
60.2 Pathology white matter is better myelinated.
In FCMD superficial gliomesenchymal prolifera-
In WWS the leptomeninges are thick and tend to tion is present on the surface of the brain and spinal
obliterate the subarachnoid space with fibrous and cord, leading to thickened leptomeninges adherent to
heterotopic neuroglial tissue. The brain surface may the surface of the CNS. Microscopically, glioneural
be smooth on external examination, with only an in- heterotopias are found in the leptomeninges. Exten-
terhemispheric fissure present and a markedly fore- sive cortical dysplasia of cerebrum and cerebellum is
shortened sylvian fissure due to incomplete develop- present. The pattern of the cortical dysplasia is always
ment of the opercula. The brain surface may also have symmetrical, but varies in severity from site to site
areas of pachygyria and polymicrogyria with an ir- and from case to case. As a rule, the cortical dysplasia
regular verrucous appearance. Bands of gliotic tissue is most prominent in the frontal and temporal lobes,
cross the interhemispheric fissure, especially frontal- whereas the occipital lobe is relatively spared. Occa-
ly, fusing the hemispheres. On sectioning, the lateral sionally the frontal cortex shows focal interhemi-
ventricles are markedly to severely enlarged; only in spheric fusions. The primary sulci (central, calcarine,
exceptional cases are the ventricles normal. The cere- parieto-occipital, and cingulate) are present, sec-
bral mantle is usually seriously reduced in width. The ondary sulci are shallow, and the gyral surfaces have
deep nuclei are present in their normal position but an irregular appearance. The cortical dysplasia may
are usually small. The corpus callosum and septum take the form of unlayered polymicrogyria (lissen-
pellucidum are often absent or hypoplastic. The aque- cephaly type II), or smooth, four-layered pachygyria,
duct is small and stenotic. The quadrigeminal surface or verrucose dysplasia with superficial cellular nod-
elevations are fused and the mammillary bodies do ules within a normally stratified six-layer cortex. The
not protrude from the ventral surface. The brain stem polymicrogyria is also called pachygyric polymicro-
is small and the pons markedly hypoplastic. Olfacto- gyria (or polymicrogyric pachygyria), because the
ry and optic nerves are attenuated or absent. The microgyri are fused and the external appearance of
cerebellar vermis, especially the posterior vermis, and the brain is pachygyric; polymicrogyria is only a mi-
the cerebellar hemispheres are hypoplastic, often croscopic finding. In most patients the three men-
associated with an enlarged fourth ventricle and a tioned types of cortical dysplasia are present to a vari-
retrocerebellar cyst, constituting a Dandy–Walker able extent. Many breaches are present in the glia lim-
malformation. In about 25% of the patients a posteri- itans–basal lamina complex overlying the cerebral
or encephalocele or meningocele is present, contain- cortex. In the CNS a basement membrane is observed
ing either an extension of a retrocerebellar cyst, cere- in the boundary between nervous tissue and lep-
bellar tissue, or, rarely, tissue of supratentorial origin. tomeninges forming part of the pial–glial barrier. At
454 Chapter 60 Congenital Muscular Dystrophies

the site of the breaches, neural tissue protrudes. The year-old patient. The external appearance of the brain
exposed surfaces of the extruded neuroglial elements was normal. Furthermore, no abnormalities of cere-
lack basal lamina, come into contact with adjoining bral and cerebellar cortex were found on microscopic
elements, and fuse with each other, leading to devel- examination and there were no neuronal heterotopias
opment of polymicrogyria. During this process, ves- within the white matter. Extensive myelin pallor was
sels in the subarachnoid space, which were originally found bilaterally with sparing of the arcuate fibers.
located over the cortical surface, become entrapped. Abnormalities were most severe in the frontoparietal
The border between cerebral cortex and white matter white matter, whereas the occipital white matter was
is irregular. Ectopic nerve cells are found in the sub- less severely affected. On microscopic examination
cortical white matter, near the ventricles and dissem- there was a spongy appearance of myelin. Moderate
inated within the white matter. The ventricles are of- astrocytic proliferation and vascular hyperplasia
ten mildly dilated. Overt hydrocephalus is not pre- were found. No changes were found in basal ganglia,
sent. The pyramidal tracts in the brain stem are hy- brain stem, or cerebellum, including cerebellar white
poplastic and dysplastic and have an abnormal matter. Taratuto et al. (1999) report on a 4-month-old
course. Within the cerebellar cortex, areas of polymi- patient. The patient had bilateral occipital cortical
crogyria are present. In the cerebellum mesenchymal dysplasia with irregular lamination and fusion of
tissue proliferation appears to have occurred inside adjacent gyri at microscopy. Multifocal glioneural
the cerebellar parenchyma to form numerous small leptomeningeal heterotopias were present. The white
cavities bordered by neuroglial elements. The cere- matter had a normal stage of myelination for a
bral white matter changes vary in severity. Myelin 4-month-old infant. The cerebellar vermis was hy-
paucity and gliosis are seen, but without signs of poplastic. Sural nerve biopsy in MDC1A patients
breakdown, particularly in the younger children, shows a reduction of large myelinated fibers, short
whereas normal myelination may be seen in older internodes, enlarged nodes, excessive variability of
children. In a patient in whom brain tissue was inves- myelin thickness, tomacula, and uncompacted
tigated at two different points of time, myelination myelin, but no evidence of demyelination.
was very poor and astrogliosis marked in brain biop-
sy material, whereas at autopsy 4 years later, myelina-
tion proved to be only slightly less than normal and 60.3 Pathogenetic Considerations
astrocytosis was mild.
In MEB, the cerebral gyral pattern is coarse on in- The dystrophin-associated glycoprotein (DAG) com-
spection, with an abnormal nodular surface, sugges- plex is present in several tissues including muscle,
tive of pachygyric polymicrogyria. Agyric areas have heart, nerve, and brain. This complex has a crucial
been found in the lateral convexity of the occipital role in linking the cytoskeletal proteins with the ex-
lobes. On sectioning, the cortex is abnormally thick. tracellular basal lamina. In muscle this complex func-
Microscopic examination reveals that almost the tions by linking the actin-associated cytoskeleton of
entire cortex is severely disorganized apart from the the muscle fibers to the extracellular matrix via dys-
basomedial occipital lobe and hippocampus. The ab- trophin and the laminin-a2 chain of merosin. The
normally thick cortex lacks horizontal laminae and dystrophin-associated glycoprotein complex is cru-
vertical columns. The neurons are haphazardly ori- cial for normal contraction of muscle. a-Dystrogly-
ented and form irregular clusters or islands separated can is a peripheral membrane component of the dys-
by gliovascular strands extending from the pia. In trophin-associated glycoprotein complex. Structural
places, irregular bundles of myelinated axons pene- defects in the dystrophin-associated glycoprotein
trate the cortex from the white matter. The pia–arach- complex, leading to defects in the linkage between
noid is focally thickened and adherent to the cortex. merosin in the extracellular matrix and actin in the
The pial–cortical border is irregular. The white mat- subsarcolemmal cytoskeleton, cause various muscu-
ter is reduced in volume and shows a moderate and lar dystrophies. Duchenne and Becker muscular dys-
variable degree of myelin pallor and gliosis. The ven- trophies are related to mutations in dystrophin. Muta-
tricles are dilated to a variable extent. The septum pel- tions in various sarcoglycans result in limb-girdle
lucidum may be absent. The cerebellar cortex is also muscular dystrophies. Mutations in the gene LAMA,
severely disorganized and lacks normal foliae, espe- which is located on chromosome 6q and encodes
cially on the upper surface. The vermis is severely the laminin-a2 chain of merosin, result in MDC1A.
hypoplastic. The interface between pia–arachnoid WWS, FCMD, MEB, MDC1C, and MDC1D are charac-
and cerebellar cortex is distorted. Gliovascular terized by hypoglycosylation of a-dystroglycan. Gly-
strands penetrate the cerebellar cortex. The brain- cosylation is the most common form of post-transla-
stem is thin and the pons is flat. tional protein modification, necessary for proteins to
In MDC1A only two histopathological reports have achieve their proper structure, function, and stability.
been published. Echenne et al. (1984) report on an 18- It is a complex process where sugars are added to pro-
60.3 Pathogenetic Considerations 455

teins as they pass through the endoplasmic reticulum Japanese patients with mutations in FCMD tend to
and the Golgi apparatus. Attached glycans can be di- have a very severe, WWS-like clinical phenotype.
vided into two groups according to their linkage MEB is caused by a defect in the O-mannosyl gly-
to the protein. N-Glycans are linked to asparagine; can synthesis. POMGnT1 normally adds an N-acetyl-
O-glycans are attached to serine or threonine. a-Dys- glucosamine residue to an O-linked mannose. This is
troglycan is normally a heavily glycosylated protein, the second step in O-mannose glycosylation. It has
and hypoglycosylation abolishes the binding activity been demonstrated that sialyl O-mannosyl glycan is
of the protein for laminin, neurexin, and agrin, all of the laminin-binding ligand of a-dystroglycan. The
which are components of the basement membrane. In hypothesis that defects in O-mannosylation weaken
20% of WWS patients the disease is caused by muta- the laminin-binding and consequently may be re-
tions in the gene POMT1, which is located on chromo- sponsible for muscular dystrophy led to the detection
some 9q34.1 and encodes O-mannosyltransferase 1. of a defect in POMGnT1 as the cause of MEB. Some
FCMD is related to mutations in the gene FCMD, patients with severe POMGNT1 mutations have a
which is located on chromosome 9q31 and encodes WWS-like phenotype. Patients with mutations near
the protein fukutin. The function of fukutin is un- the 5’-terminus tend to have a more severe clinical
known, but based on amino acid homology with sev- phenotype than patients with mutations near the
eral phosphoryl-ligand transferases, it is hypothe- 3’-terminus.
sized to be a glycosyltransferase. MEB is caused by Laminin is a heterotrimer composed of three
mutations in the gene POMGNT1, located on chromo- different polypeptides, called a-, b-, and g-chain,
some 1p33–34, which encodes the protein O-manno- of which different subtypes are known. These chains
syl-b1,2-N-acetylglucosaminyltransferase-1 assemble into a number of different laminin variants,
(POMGnT1). This enzyme uses an O-linked mannose but every laminin has one a-, one b-, and one g-chain.
as a substrate. Fukutin-related protein, encoded by Laminin-2, or merosin, is composed of a2-b1-g1. It is
the gene FKRP, located on chromosome 19q13.3, is the a2-chain that is mutated in MDC1A. Classical
highly homologous to fukutin and also a putative gly- MDC1A is associated with mutations that drastically
cosyltransferase. Mutations in the gene LARGE, locat- affect the expression or structure of laminin-a2. A
ed on chromosome 22q12.3-q13.1, lead to MDC1D. wide range of milder phenotypes is caused by partial
Large is a putative glycosyltransferase. laminin-a2 deficiency, produced by mutations in
In WWS, about 20% of the patients have a defect in LAMA2 that permit production of a partially func-
O-mannosyltransferase 1, the enzyme that putatively tional protein or a reduced amount of normal pro-
catalyzes the first step in O-mannosyl glycan synthe- tein. Merosin is not only present in muscle tissue, but
sis. The O-mannosyl glycans of a-dystroglycan are also in the brain. In the brain merosin is localized on
critical for binding laminin-a2. WWS is genetically the brain surface at the glia limitans and on the base-
heterogeneous and several WWS families do not link ment membrane of blood vessels. Furthermore,
to the POMT1 locus. It is very likely that defects in merosin is present in the endoneurial basement
the other proteins involved in O-mannosylglycosyla- membrane that surrounds the myelin sheath of pe-
tion underlie the remaining, unexplained WWS pa- ripheral nerve fibers and individual Schwann cells. In
tients. laminin-a2 deficiency, the basement membrane in
FCMD occurs almost exclusively in Japan. About skeletal muscle and peripheral nerves is disrupted or
95% of FCMD patients share a specific haplotype on completely absent, indicating that laminin-a2 is es-
one or both alleles in the critical region of chromo- sential for formation of basement membrane in these
some 9, supporting the hypothesis of a founder of the tissues. There is evidence that the formation of a
disease in the Japanese population. In all patients basement membrane is a prerequisite of myelination
sharing this haplotype, a 3-kb retrotransposal inser- in peripheral nerves. Under conditions that prevent
tion into the 3’-untranslated region of the gene is basement membrane formation, Schwann cells fail to
found. Various other mutations have been found. The ensheath and myelinate nerve fibers.
frequency of a severe phenotype with WWS-like Defects in the gene FKRP lead to a severe form of
manifestations such as hydrocephalus and microph- CMD, designated MDC1C, and a milder limb-girdle
thalmia is higher among patients who are compound muscular dystrophy, designated LGMD21. Most pa-
heterozygous for the Japanese founder mutation than tients with MDC1C do not have structural brain ab-
among the patients who are homozygous for the normalities. However, it has been shown that severe
founder mutation. The observed lack of Japanese pa- mutations in FKRP may lead to an MEB or even WWS
tients with two nonfounder mutations suggests that phenotype.
such cases might be fatal in utero. This may also ex- Large is a putative bifunctional glycosyltrans-
plain why few FCMD patients have been reported in ferase. One putative catalytic region has a high ho-
non-Japanese populations in which the Japanese mology to members of the bacterial WaaIJ family of
founder mutation does not occur. The rare non- putative a-glycosyltransferases involved in the syn-
456 Chapter 60 Congenital Muscular Dystrophies

Fig. 60.1. A 4-month-old boy with WWS. The third and lateral tion. The cerebral cortex is agyric on the outside, whereas the
ventricles are highly enlarged due to aqueduct stenosis. The irregular inner border indicates a disorganized,polymicrogyric
corpus callosum is thin and high-arched. Note the presence cortex, compatible with lissencephaly type II. The cerebellar
of a cingulate gyrus, indicating that there is no agenesis of the cortex is also disorganized. The cerebral hemispheric white
corpus callosum. The septum pellucidum is absent. The colli- matter has an abnormal signal throughout. Only the brain
culi are fused.The pons is extremely hypoplastic and there is a stem appears to contain some normal myelin. From Van de
pontomesencephalic kink. The cerebellum is extremely hy- Knaap et al. (1997), with permission
poplastic with characteristics of a Dandy–Walker malforma-

thesis of lipopolysaccharides or lipo-oligosaccha- their pathogenesis is unclear. Retarded but ongoing


rides. The second catalytic domain is closely related myelination may contribute in some cases, especially
to I-b-1,3-N-acetylglucosaminyl transferase, an en- in FCMD and MEB. However, in FCMD and MEB, too,
zyme required for the synthesis of the poly-N-acetyl- the cerebral white matter abnormalities have a higher
lactosamine backbone, which is attached to N-gly- signal on T2-weighted images and a lower signal on
cans, O-glycans, and glycolipids. T1-weighted images than is compatible with straight-
Whether the brain and eye symptoms in the WWS, forward hypomyelination, but reduction in the white
FCMD, MEB, MDC1C, and MDC1D are related to the matter abnormalities has been observed on follow-
defect in glycosylation of a-dystroglycan or other up. In WWS and MDC1A the white matter abnormal-
proteins is less clear. Brain-specific disruption of a- ities are more impressive and mildly swollen, suggest-
dystroglycan during embryonic development causes ing sponginess, which has been confirmed at autopsy.
neuronal overmigration and fusion of cerebral hemi- The role of the white matter abnormalities in deter-
spheres in mice. This suggests that disrupted glycosy- mining the clinical picture seems to be minor in all
lation of a-dystroglycan is also at least in part respon- CMD variants. The disease is mostly explained by the
sible for the brain abnormalities. The white matter severity of the cortical dysplasia and the muscular
abnormalities reported in the CMDs are variable and dystrophy.
60.5 Magnetic Resonance Imaging 457

Fig. 60.2. A 10-month-old girl with WWS.The lateral and third tricular heterotopias. The cerebellar cortex is also disorga-
ventricles are markedly dilated despite the shunting. Note the nized, with the presence of many small subcortical cysts. The
thin and high-arched corpus callosum,which is just visible.The cerebral hemispheric white matter has an abnormal signal
septum pellucidum is absent. The colliculi are fused. The pons throughout. Only the brain stem and cerebellar white matter
and cerebellar vermis are hypoplastic. The cerebral cortex is appear to contain myelin. Courtesy of Dr. C.E. Catsman-
pachygyric. In the anterior region the cortex is abnormally Berrevoets, Department of Child Neurology, Sophia Children’s
thick,whereas it is thinner in the parietal areas.The irregular in- Hospital and Erasmus University Medical Center, Rotterdam,
ner border indicates a disorganized, polymicrogyric cortex, The Netherlands
compatible with lissencephaly type II.There is a rim of periven-

60.4 Therapy 60.5 Magnetic Resonance Imaging

No definitive treatment is possible, only supportive In WWS, MRI almost invariably shows severe hydro-
care. Physiotherapy is of major importance. In some cephalus (Figs. 60.1–60.3). A normal ventricular size
patients a slight improvement in strength and a fall in is highly unusual. Because of the severe hydro-
CK activity have been noted on administration of cephalus and very thin cerebral mantle, the quality of
corticosteroids. However, in other patients no im- the white and gray matter may be difficult to assess. In
provement was found. Considering the major adverse patients in whom a cerebral mantle of some thickness
effects of chronic use of corticosteroids, this mode of is present, the abnormalities are easier to recognize.
therapy remains controversial. Respiratory support The cortex is either totally agyric (Fig. 60.1) or folded
may prolong life. in broad, somewhat better formed gyri (Figs. 60.2 and
60.3). The cortex is smooth on the external side but
the border with the white matter is irregular, reflect-
ing the underlying polymicrogyria and disorganiza-
tion of the cortex with frequent disruption by glio-
fibrillary bundles (Figs. 60.1–60.3). In some cases ex-
458 Chapter 60 Congenital Muscular Dystrophies

Fig. 60.3. A 6-year-old boy with WWS. The lateral and third ence of many small subcortical cysts. The cerebral hemispher-
ventricles are mildly dilated. The corpus callosum is high- ic white matter has an abnormal signal throughout and is
arched.The septum pellucidum is partially absent.The colliculi swollen.There are multiple large subcortical cysts.The cerebel-
are fused. The pons and cerebellar vermis are hypoplastic. The lar white matter also has an abnormal signal intensity.Only the
cerebral cortex is pachygyric and irregular, compatible with corpus callosum and the brain stem have a normal low signal.
lissencephaly type II.There are many small periventricular het- From Van der Knaap et al. (1997), with permission
erotopias. The cerebellar cortex is disorganized, with the pres-
60.5 Magnetic Resonance Imaging 459

Fig. 60.4.
460 Chapter 60 Congenital Muscular Dystrophies

Fig. 60.4. (continued). A male patient with FCMD.The first and dilated. The cerebral white matter is diffusely abnormal in sig-
second rows show the MRI at 5 months, the third and fourth nal at 5 months, but improvement is seen over time. At 7 years
rows contain the MRI obtained at 21 months,the fifth and sixth the posterior cerebral white matter is normally myelinated,
rows show the MRI obtained at 7 years.The frontoparietal cor- whereas the frontal periventricular and deep white matter has
tex displays the characteristics of a polymicrogyric pachygyria, an abnormal signal intensity.The corpus callosum and internal
whereas the occipital cortex is smooth and very thick. The capsule have a normal signal intensity. Courtesy of Dr. N. Aida,
cerebellar cortex is also dysplastic with distortion of the nor- Department of Radiology, Kanagawa Children’s Medical Cen-
mal horizontal foliae and presence of subcortical cerebellar ter,Yokohama, Japan
cysts. The pons is flat. The lateral ventricles are somewhat

tensive subcortical and subependymal heterotopic is also enlarged and the aqueduct is narrow. Fusion of
neuronal nodules are seen (Figs. 60.2 and 60.3). The the superior and inferior colliculi is typically seen
cerebral white matter has a high signal intensity on (Figs. 60.1–60.3). There is often a pontomesence-
T2-weighted images throughout and a low signal in- phalic kink and the pons is extremely hypoplastic
tensity on the T1-weighted images (Figs. 60.1–60.3). (Figs. 60.1–60.3). The cerebellum is highly hypoplas-
The white matter may have a mildly swollen appear- tic in all its elements, in particular in the vermis. The
ance and multiple cysts may be present within cerebellar surface is smooth, without foliae or with ir-
this highly abnormal looking white matter (Fig. 60.3). regular, small foliae. Multiple small subcortical cere-
The corpus callosum is often very thin and high- bellar cysts are seen (Figs. 60.2 and 60.3). In some pa-
arched, and may not be visible. However, the presence tients the posterior fossa is extremely small, but in
of a gyrus cinguli strongly suggests that it is not other patients the fourth ventricle is enlarged and in
an agenesis of the corpus callosum, but rather an open communication with an enlarged retrocerebel-
extreme thinning due to severe hydrocephalus lar space, forming either the full-blown Dandy–Walk-
(Figs. 60.1–60.3). The septum may be absent, proba- er malformation or the less severe Dandy–Walker
bly also related to the long-standing hydrocephalus of variant (Fig. 60.1). A posterior meningocele or ence-
antenatal origin (Figs. 60.1–60.3). The third ventricle phalocele is often present.
60.5 Magnetic Resonance Imaging 461

In FCMD, MRI reveals cerebral cortical dysplasia, consequence of white matter swelling. MRI shows
showing gyri that are too few and too broad, and in- that the cortex is not thicker than normal and does
complete opercularization (Fig. 60.4). In most areas not have the irregular inner border of pachygyric
the slightly thickened cortex has an irregular aspect polymicrogyria (Figs. 60.7 and 60.8). However, some
with little dots and an irregular inner border, reflect- MDC1A patients have occipital cortical dysplasia
ing polymicrogyria and verrucose cortical dysplasia. with an agyric outer border but irregular inner
This type of cortical dysplasia is seen in frontopari- border and evidence of subcortical ectopic neurons,
etotemporal areas (Fig. 60.4). In some areas the cortex similar to the lissencephaly type II seen in WWS
is thicker and has a smooth inner and outer surface. (Figs. 60.9 and 60.10). In patients with occipital
This type of cortical dysplasia is mainly seen in the agyria the occipital horn of the lateral ventricles may
temporo-occipital region (Fig. 60.4). The ventricular be focally dilated (Fig. 60.9). The dysplasia is bilateral
system is mildly enlarged and often has a colpo- but may be larger on one side than the other, with al-
cephalic aspect. In neonates the cerebral white matter so a more prominent enlargement of the occipital
looks normal for unmyelinated white matter. In in- horn on the most seriously affected side. In MDC1A,
fants and young children the cerebral white matter the white matter is near-normal in the first few
looks diffusely abnormal, with a higher signal on months of life (Fig. 60.9). From the second half of the
T2-weighted images and lower signal on T1-weighted first year onwards, MRI shows swollen white matter
images than is compatible with straightforward with a high signal on T2-weighted images, low on T1-
hypomyelination (Fig. 60.4a). On repeated MRI pro- weighted images. In most patients the white matter
gress of myelination is seen, the rate being variable abnormalities are extensive and confluent, but in
(Fig. 60.4b,c). In some patients there is still hardly some patients they are a bit less extensive and some-
any myelin after several years. In some older children times they are multifocal. The abnormalities have a
diffuse white matter lesions are seen. Most patients frontal predominance and the occipital white matter
have multifocal lesions in the cerebral white matter is better preserved. In some cases the subcortical
(Fig. 60.4b, c). The lesions vary in size and distribu- areas are spared throughout. The corpus callosum,
tion. In a few patients no white matter abnormalities internal capsule, optic radiation, brain stem, and cere-
are found. The corpus callosum, internal capsule, and bellar white matter are consistently spared. The white
brain stem display a normal myelin signal. MRI also matter changes appear to be nonprogressive over
shows cerebellar cortical dysplasia, with irregularly the years. In patients with occipital agyria, the
distorted folia. There are subcortical cerebellar cysts occipital white matter may be markedly abnormal
(Fig. 60.4b, c). The pons is flat (Fig. 60.4). (Fig. 60.9). In some patients hypoplasia of the pons is
In MEB, MRI shows a variable ventricular enlarge- seen (Fig. 60.7). The cerebellum may also be hy-
ment, ranging from normal to markedly dilated. The poplastic. In CMD patients with partial merosin defi-
cortical dysplasia is variable, both between different ciency, white matter abnormalities are more variable.
areas of the brain and between patients (Figs. 60.5 Some patients have extensive white matter abnormal-
and 60.6). The frontal, temporal, and parietal areas are ities similar to those seen in classical MDC1A. In oth-
the most abnormal, whereas the configuration of the ers the white matter abnormalities are more limited
occipital cortex is close to normal. The cortex is in extent (Fig. 60.10). Occipital agyria may also occur
slightly too thick and folded in broad gyri with an in patients with partial merosin deficiency (Fig.
irregular inner border, compatible with pachygyric 60.10).
polymicrogyria (Figs. 60.5 and 60.6). Operculariza- In the two patients with adult-onset signs of mus-
tion is incomplete. Neuronal heterotopias may be cular dystrophy and cerebral dysfunction reported by
seen in the white matter. The cerebral white matter is van Engelen et al. (1992), the white matter abnormal-
either normal (Fig. 60.6) or contains multifocal white ities are identical to those of MDC1A patients. A spe-
matter abnormalities (Fig. 60.5). The corpus callosum cial finding is that the oldest patient, 29 years of age,
may be dysplastic and thin and the septum pellu- has cysts in the subcortical area in the tips of the tem-
cidum may be incomplete. The superior and inferior poral lobes and the parietal area. With these cysts the
colliculi are fused. The pons is hypoplastic. The cere- MR pattern becomes indistinguishable from the pat-
bellum, in particular the vermis, may be small. The tern of megalencephalic leukoencephalopathy with
cerebellar cortex is dysplastic and there are multiple subcortical cysts, described in Chap. 59.
small subcortical cerebellar cysts (Figs. 60.5 and 60.6). In the MDC1C patients with mutations in the
In classical MDC1A, MRI shows prominent white FKRP gene and cerebral abnormalities, variable cere-
matter changes (Figs. 60.7 and 60.8). The white matter bral white matter abnormalities and the presence of
appears mildly swollen, leading to broadening of the subcortical cerebellar cysts have been reported
gyri with stretching of the overlying cortex. On CT it (Figs. 60.11 and 60.12). However, most patients with
is difficult to distinguish between primary cortical FKRP mutations have normal MRI.
dysplasia and secondary broadening of the gyri as a
462 Chapter 60 Congenital Muscular Dystrophies

Fig. 60.5. A 2.5-year-old girl with MEB. The frontal, temporal, white matter contains extensive signal abnormalities, whereas
and parietal cortex is the most abnormal, whereas the config- the occipital white matter is normal. The corpus callosum,
uration of the occipital cortex is close to normal.The abnormal internal capsule, brain stem, and cerebellar white matter have
cortex is slightly too thick and folded in broad gyri with an a normal signal.The inferior vermis is hypoplastic.The superior
irregular inner border, indicative of pachygyric polymicro- and inferior colliculi are fused. The pons is hypoplastic. There
gyria. Neuronal heterotopias are seen in the white matter are multiple small subcortical cerebellar cysts
along the lateral ventricles. The frontal, parietal, and temporal
60.5 Magnetic Resonance Imaging 463

Fig. 60.6. A 9-year-old boy with MEB.


The frontal cortex is too thick and has
an irregular inner border, indicative of
underlying polymicrogyria.The inferi-
or vermis is hypoplastic.The superior
and inferior colliculi are fused.The
pons is hypoplastic.There are multiple
small subcortical cerebellar cysts.
The lateral ventricles are mildly dilat-
ed, more so on the right than the left.
The white matter is normal. Courtesy
of Dr. P.G. Barth, Department of Child
Neurology, Academic Medical Center,
Amsterdam, The Netherlands

In the single MDC1D patient reported (Long- It is clear that MRI plays an important role in the
man et al. 2003), MRI revealed diffuse cerebral white classification of CMD patients during life. The corti-
matter abnormalities with mild swelling of the ab- cal dysplasia, pons hypoplasia, subcortical cerebellar
normal white matter leading to broadening of cysts, and different types of white matter involvement
gyri (Fig. 60.13). The cerebral cortex showed signs are highly suggestive of the diagnosis CMD. MDC1A
of diffuse polymicrogyria. The pons was hypoplas- may be confused with megalencephalic leuko-
tic. encephalopathy with subcortical cysts, but the sub-
cortical cysts are as a rule lacking.
464 Chapter 60 Congenital Muscular Dystrophies

Fig. 60.7. A 6-year-old girl with classical MDC1A. The MRI whereas the occipital white matter is better preserved. The
shows prominent white matter changes. The abnormal white corpus callosum, internal capsule, optic radiation, brain stem,
matter is mildly swollen with broadening of the gyri. MRI and cerebellar white matter are spared. The pons is mildly
shows that the cortex is of normal thickness and does not have hypoplastic
an irregular border.The frontal white matter is most abnormal
60.5 Magnetic Resonance Imaging 465

Fig. 60.8. A 9-year-old girl with MDC1A.The abnormalities are very similar to those seen in Fig. 60.7

Fig. 60.9. A 2-month-old girl with MDC1A. Note the occipital agyria.The white matter under the area of agyria has an abnormally
high signal.The remainder of the white matter looks normal for unmyelinated white matter
466 Chapter 60 Congenital Muscular Dystrophies

Fig. 60.10. A 7-year-old boy with CMD and partial merosin tesy of Dr. H. Stroink and Dr. C.E. Catsman-Berrevoets, Depart-
deficiency. Note the agyria in the occipital and basotemporal ment of Child Neurology, Sophia Children’s Hospital and Eras-
region. There are multifocal white matter abnormalities. Cour- mus University Medical Center, Rotterdam, The Netherlands

Fig. 60.11. A girl with MDC1C. The first MRI (first row) was ob- cortex is dysplastic and there are subcortical cerebellar cysts.
tained at the age of 3 years; the follow-up MRI (second row) at The pons is hypoplastic. From Louhichi et al. (2004), with per-
the age of 6 years.The initial MRI shows multifocal large white mission, and courtesy of Dr. F. Fakhfakh, Laboratoire de Géné-
matter lesions, mainly involving the deep white matter. On fol- tique Moleculaire Humaine, Faculté de Médecine de Sfax, and
low-up, most white matter abnormalities have disappeared Dr. C. Triki, Service de Neurologie, CHU Habib Bourguiba, Sfax,
and only a few foci of abnormal signal remain. The cerebellar Tunisia
467

Fig. 60.12. A 7-year-old boy with MDC1C. The MRI shows ex- bellar cysts. From Louhichi et al. (2004), with permission, and
tensive white matter abnormalities,which partially spare the U courtesy of Dr. F. Fakhfakh, Laboratoire de Génétique Molecu-
fibers. The pons and cerebellar vermis are hypoplastic. The laire Humaine, Faculté de Médecine de Sfax, and Dr.C.Triki, Ser-
cerebellar cortex is dysplastic and there are subcortical cere- vice de Neurologie, CHU Habib Bourguiba, Sfax, Tunisia
468 Chapter 60 Congenital Muscular Dystrophies

Fig. 60.13. A 14-year-old girl with MDC1D.The sagittal images sum, internal capsule, brain stem, and cerebellar white matter
show hypoplasia of the pons and fusion of the superior and in- have a normal signal. The U fibers in the occipitotemporal re-
ferior colliculi. There is no cerebellar hypoplasia and there are gion are spared. The cerebral cortex diffusely has an irregular
no evident subcortical cerebellar cysts. The cerebral white border,suggesting underlying polymicrogyria.From Longman
matter is diffusely highly abnormal in signal. In the frontal and et al. (2003), with permission, and courtesy of Dr. F. Muntoni,
particularly the anterior temporal region the abnormal white Dubowitz Neuromuscular Centre, Hammersmith Campus,
matter is swollen with broadening of gyri. The corpus callo- London, UK
Chapter 61

Myotonic Dystrophy Type 1

61.1 Clinical Features genital DM 1 is seen. The disease is transmitted as an


and Laboratory Investigations autosomal dominant trait, but the mother is the af-
fected parent in over 90% of the cases of congenital
Myotonic dystrophy type 1 (DM 1) or Curschmann– DM 1.
Steinert disease is a dominantly inherited progressive Diagnosis is established by clinical examination
myopathy. It is one of the most frequent muscle dis- and EMG. On EMG prolonged trains of high-frequen-
orders, with a prevalence of approximately 1:8,000. cy discharges arising from single fibers or groups of
Symptoms of the disease usually become apparent muscle fibers occur in response to electrode insertion
between the ages of 15 and 40 years, but may be found or movement. EMG can be negative in young patients.
in childhood. Major symptoms include myotonia, Serum creatine kinase levels may be mildly elevated.
progressive muscle weakness, and atrophy. In partic- DNA techniques are available to confirm the diagno-
ular the face, jaw, neck, and distal muscles of the arms sis and for prenatal diagnosis.
and legs are affected. Involvement of the bulbar mus-
cles causes dysphagia and dysarthria. Myotonia leads
to a prolonged after-contraction of the affected mus- 61.2 Pathology
cles, which persists after the voluntary muscle con-
traction has ceased. Myotonia is increased by fatigue, Neuropathological data on DM 1 are scarce. Some ex-
emotion, and cold. Other features of the disease in- ternal atrophy of the cerebral hemispheres may be
clude cataract, early frontal baldness (more conspicu- present. Microscopic examination may reveal lesions
ous in males than in females), hearing impairment, within the cerebral white matter, especially in the sub-
and testicular atrophy leading to impotence and in- cortical areas. Microscopically, they are characterized
fertility. The heart is often affected. Cardiac conduc- by myelin paucity and preservation of myelin sheaths
tion defects and dilated cardiomyopathy may occur in perivascular regions in a tigroid pattern. Axons are
and may contribute to a shortened life span. Smooth relatively preserved. Macrophages containing fatty
muscles, particularly those of the pharynx, esopha- material and fibrillary astrocytes are scanty. It has
gus, and gastrointestinal tract, are also involved. Hy- been demonstrated that the areas of myelin paucity
perinsulinism occurs with enhanced frequency. Mild- correspond with areas of signal abnormality on MRI.
ly impaired intelligence is frequently present. Hyper- Additional findings that have been reported are
somnia may occur. variable neuronal loss and astrocytosis in the fron-
DM 1 has a congenital variant, with variable hypo- toparietal and occipital cortices, neurofibrillary tan-
tonia and weakness present at birth. This variant is gles in the limbic cortex, intracytoplasmic inclusion
present in 10–15% of the patients. In congenital DM 1 bodies within neurons of the thalamus and substan-
respiratory problems and feeding difficulties are tia nigra, disordered cortical architecture, and neu-
common. Joint deformities may be present. Myotonia ronal heterotopias. The neuronal heterotopias are fre-
is not usually present in neonates and seldom devel- quently found adjacent to the areas of myelin paucity.
ops until later in childhood. Cataracts and endocrine The absence of gliosis and lipid-laden macro-
abnormalities usually develop even later. Mental re- phages within the areas of abnormal white matter as
tardation is present in nearly all patients with con- well as their association with neuronal heterotopias
genital DM 1 and is more severe than the cognitive suggest that the white matter lesions are developmen-
problems seen in patients with adolescent and adult- tal abnormalities and not caused by demyelination.
onset disease. Some patients do not survive the
neonatal period.
A characteristic feature of DM 1 is the tendency of 61.3 Pathogenetic Considerations
the symptomatology to have an earlier onset and in-
creasing severity with transmission to subsequent DM 1 is caused by an expansion of an unstable cyto-
generations, a phenomenon called anticipation. In sine-thymine-guanine (CTG) trinucleotide repeat in
early generations, cataract is often the only abnor- the 3’ untranslated region of a protein kinase gene,
mality; in a subsequent generation adult myotonia DMPK, located on chromosome 19q13.3. In the nor-
and weakness occur, while in the next generation con- mal population, the repeat length ranges between
470 Chapter 61 Myotonic Dystrophy Type 1

4 and 37 copies, whereas alleles in DM 1 patients vary 61.4 Therapy


from 50 to several thousands of CTG repeats. The
longer the CTG repeat sequence, the earlier the onset First and most important is awareness of the potential
and more severe the clinical symptomatology. Alleles problems associated with DM 1. Anesthesia and
with 50–80 repeats are classified as protomutations, surgery come with particular hazards due to the com-
with which the DM 1 phenotype is mild and often bination of respiratory muscle weakness and in-
does not cause subjective symptoms. Alleles with creased sensitivity to anesthetics and muscle relax-
more than 80 repeats usually cause the classic DM 1 ants. Cardiac problems such as atrial fibrillation and
phenotype. The phenomenon of anticipation is asso- flutter and heart block are important causes of death
ciated with an increase in the length of the CTG repeat and require careful attention and treatment. Aware-
sequence with transmission to subsequent genera- ness of smooth muscle dysfunction and the associat-
tions. Rare cases of reverse mutations have been re- ed potential gastrointestinal complaints including
ported, in which the prolonged CTG repeat sequence abdominal pains and pseudo-obstruction may pre-
in the parent is decreased in size to a normal length in vent unnecessary surgery. Symptomatic relief of the
the offspring. myotonia can, if necessary, be obtained by drugs such
The expanded repeat is located in the 3’ untranslat- as procainamide, diphenylhydantoin (phenytoin),
ed part of the gene. Three models have been proposed and mexiletine. The effect of these drugs lies in the
to explain how an expansion of this untranslated CTG stabilization of the muscle membrane; they have no
repeat may cause the dominantly inherited DM 1 phe- effect on muscle weakness. Physical therapy may be
notype. Haploinsufficiency of the DMPK gene may necessary. Cataract extraction can be important in or-
produce cellular dysfunction. Alternatively, the ex- der to preserve visual acuity.
panded repeat may lead to a dominant gain of func-
tion. Finally, the CTG repeat expansion may change
the chromatin structure, which interferes with the ex- 61.5 Magnetic Resonance Imaging
pression of adjacent genes.
The in vivo biological substrates of the enzyme It was already known from CT that mild ventricular
DMPK are unknown. Since DMPK exhibits kinase ac- enlargement and white matter hypodensity are not
tivity, any perturbation of its expression or activity rare in DM 1.
will potentially affect other molecules. Because DM 1 MRI confirms the frequent presence of mild ven-
is characterized by muscle dysfunction and myoto- triculomegaly and enlargement of subarachnoid
nia, ion channels could be substrates for DMPK. How- spaces (Figs. 61.1–61.4). In the majority of the DM 1
ever, other proteins of as yet unknown function have patients MRI also shows white matter abnormalities
also been found associated with DMPK. Dysfunction in the cerebral hemispheres (Figs. 61.1–61.4). The dis-
of such proteins may explain the extramuscular tribution of the white matter lesions is variable, rang-
symptoms, including the mental deficit. ing in location from predominantly in the periven-

Fig. 61.1. These T2-weighted MR images show the white mat- involved as well. There is some atrophy with enlargement of
ter changes in a 60-year-old female patient with DM 1. The the subarachnoid spaces. From Damain et al. (1992), with per-
white matter abnormalities are patchy and predominantly mission
located in the subcortical areas. The temporal white matter is
61.5 Magnetic Resonance Imaging 471

tricular region to predominantly in the deep white in both conditions. Patients with congenital DM 1
matter or predominantly in the subcortical areas. In tend more often to have enlargement of the lateral
particular the anterior part of the temporal lobes is ventricles and a small corpus callosum.
frequently involved (Figs. 61.1–61.4). The white mat- Magnetization transfer ratios have found to be
ter changes are often patchy with in part isolated and decreased not only in the MRI-visible white matter
in part irregularly confluent lesions of variable size. lesions, but also in the normal-appearing white mat-
The lesions tend to be bilateral but are often not sym- ter, suggesting presence of more widespread structur-
metrical in detail. The extent of the white matter al abnormalities. T2 relaxation measurements simi-
changes varies from minor, with some small white larly revealed increased values within the white mat-
matter lesions (Figs. 61.4 and 61.5), to more extensive ter lesions, but again also in the normal-appearing
and more confluent lesions (Figs. 61.1–61.3). In ex- white matter.
ceptional patients, the cerebral white matter abnor- The pattern of MRI abnormalities with multifocal
malities are diffuse. The white matter abnormalities and often not entirely symmetrical lesions would
may show an increase over time. The white matter rather suggest acquired disorders, especially inflam-
changes occur both in congenital MD1 and later-on- matory and infectious disorders. The pattern is
set forms and have the same imaging characteristics particularly similar to that seen in congenital cyto-

Fig. 61.2. A 63-year old female patient with DM 1.The patchy, are mildly dilated. Note the temporal white matter abnormali-
multifocal white matter abnormalities mainly involve the deep ties. Courtesy of Dr. G. Bachmann, Department of Diagnostic
cerebral white matter. The abnormalities are asymmetrical in Radiology, Kerckhoff-Klinik, Bad Nauheim, Germany
distribution. The lateral ventricles and subarachnoid spaces

Fig. 61.3. The T2-weighted images in a 61-year-old male pa- zo, Department of Neurological Sciences, Second University of
tient with DM 1 reveal similar abnormalities, again with a Naples, Italy
slightly asymmetrical distribution. Courtesy of Dr. A. Di Costan-
472 Chapter 61 Myotonic Dystrophy Type 1

Fig. 61.4. This 54-year-old female patient with DM 1 has only ment of Diagnostic Radiology, Kerckhoff-Klinik, Bad Nauheim,
mild white matter abnormalities. The temporal abnormalities Germany
are most pronounced. Courtesy of Dr. G. Bachmann, Depart-

Fig. 61.5. This 34-year-old female patient with DM 1 has min- Courtesy of Dr.A.Di Costanzo,Department of Neurological Sci-
imal white matter abnormalities. There is an indication of ences, Second University of Naples, Italy
slight abnormalities in the anterior temporal white matter.

megalovirus infection. In the latter condition, multi-


focal lesions are seen, mainly in the deep cerebral
white matter, and the anterior temporal lobes are
frequently involved. However, in congenital cyto-
megalovirus infection, the largest lesions are as a rule
present in the parietal region, which is not the case in
DM 1. Of course, in the presence of myotonia, there is
little reason for confusion.
Chapter 62

Myotonic Dystrophy Type 2

62.1 Clinical Features In a large overview study on DM 2 (Day et al. 2003),


and Laboratory Investigations 90% of the affected individuals had electrical myoto-
nia, 82% had weakness, 61% cataracts, 23% diabetes
Myotonic dystrophy type 2 (DM 2) is a dominantly in- mellitus, and 19% cardiac involvement. First symp-
herited progressive myopathy. DM 2 closely resem- toms of the disease occur between the ages of 13 and
bles adult-onset myotonic dystrophy type 1 (DM 1). 67, with a median of 48 years. Muscle symptoms,
Initially, three different disorders were delineated on including pain, stiffness, myotonia, and weakness, are
the basis of clinical findings: proximal myotonic the most common symptoms in DM 2 patients. Fluc-
myopathy (PROMM), proximal myotonic dystrophy tuating or episodic burning muscle pain is frequently
(PMD), and myotonic dystrophy type 2 (“DM 2”). present, mainly during the night. Cataracts are a
Since all three have been shown to be part of one prominent and early feature, sometimes already
genetic spectrum, DM 2 is used for all three and has seen before the age of 20. Cataract extraction may be
replaced the terms PROMM and PMD. necessary from as early as 30 years to as late as in the
Patients with PROMM have been reported as being seventies. Cardiac complaints include palpitations,
completely asymptomatic, mildly affected, or consid- intermittent tachycardia, and episodic syncope. These
erably disabled. Clinical onset is between 20 and 60 symptoms increase in frequency with age. Cardiac
years of age. Most patients have cataract before the conduction abnormalities and cardiomyopathy may
age of 50. The muscle weakness is predominantly underlie these complaints. DM 2 patients can develop
proximal. Additional signs include muscle pain, fluc- fatal arrhythmias unexpectedly.
tuating weakness and stiffness, muscle cramps, fasci- On EMG a broad spectrum of spontaneous activi-
culations, calf hypertrophy, tremor, seizures, sen- ty, such as fibrillations, positive sharp waves, fascicu-
sorineural deafness, frontal baldness, male hypogo- lations, myotonic and pseudomyotonic discharges,
nadism, insulin-resistant diabetes mellitus, hypothy- neuromyotonia-like discharges, and high-frequency
roidism, hyperhidrosis, dysphagia, constipation, bizarre discharges, can be found. Motor unit action
intermittent episodes of chest pain, and cardiac con- potentials may show reduced mean duration, in-
duction defects. Cognitive impairment, hypersomnia, creased polyphasia, and increased satellite potentials.
and apathy are rare and at most mild. Serum creatine kinase levels are usually elevated, typ-
Patients with PMD have been reported to present ically less than five times the upper limit of normal,
with cataract, late-onset sensorineural hearing loss, but may also be normal. g-Glutamyl transferase is
late-onset prominent proximal weakness, wasting, normal or mildly elevated. Primary male hypogo-
myotonia and male hypogonadism. nadism is found in the majority of the affected men,
“DM 2” as defined initially includes patients with with elevated LH and FSH, low testosterone, and
myotonia, diffuse (proximal and distal) or exclusively oligospermia. Hypogammaglobulinemia may be
distal weakness, frontal balding, cataracts, infertility, found. Thyroid function tests may indicate hypothy-
and cardiac conduction defects. Additional features roidism. In the case of diabetes mellitus, insulin in-
include calf hypertrophy and hyperhidrosis. sensitivity is found with elevated basal insulin levels
So, like DM 1, DM2 is a clinically heterogeneous or prolonged insulin elevations. ECG shows impulse
multisystem disorder. Some affected DM 2 patients generation and conduction defects. Echocardiogra-
exhibit remarkable clinical similarity to those with phy may reveal a cardiomyopathy. DNA techniques
classic DM 1, with myotonia, proximal and distal are available to confirm the diagnosis and for prena-
muscle weakness, frontal balding, cataracts, and car- tal diagnosis.
diac conduction defects, whereas other patients can
be more easily distinguished. One clear distinction is
the absence of a congenital form of DM 2. Other dif- 62.2 Pathology
ferences include a lack of mental retardation in juve-
nile patients; less evident central hypersomnia; less Neuropathological data on DM 2 are lacking.
distal, facial, and bulbar weakness; and less pro-
nounced muscle atrophy.
474 Chapter 62 Myotonic Dystrophy Type 2

Fig. 62.1. The T2-weighted images in this 66-year-old male external capsule are also affected. The lateral ventricles and
patient with DM 2 show diffuse cerebral white matter abnor- subarachnoid spaces are mildly dilated. Courtesy of Dr. E.
malities with sparing of the corpus callosum and relative spar- Hund, Department of Neurology, and Dr.Sartor, Department of
ing of the U fibers. Spots of abnormal signal are present in the Neuroradiology, University of Heidelberg Medical School, Hei-
basal ganglia.The anterior limb of the internal capsule and the delberg, Germany

62.3 Pathogenetic Considerations Not all families with a DM 2 phenotype display


linkage with chromosome 3q21.3. So, there must be
DM 2 is caused by an unstable expansion of a cyto- still other genes related to this phenotype.
sine-cytosine-thymine-guanine (CCTG) repeat in in-
tron 1 of the zinc finger protein 9 gene (ZNF9), a gene
located on chromosome 3q21.3. The repeat length 62.4 Therapy
varies between 75–11,000, with a mean of approxi-
mately 5,000. Expansion sizes in the blood of affected So far, there is no specific treatment for DM 2. Physi-
children are usually shorter than in their parents. No cal therapy may be of some benefit. In patients in
significant correlation is found between the age of on- whom myotonia is a prominent problem, mexiletine,
set and expansion size. phenytoin, or carbamazepine may be beneficial.
The gene has two products, a-ZNF9 and b-ZNF9, When cardiac conduction defects are present, antiar-
produced by alternative splicing. ZNF9, also called rhythmic drugs should be avoided. Patients should
cellular nucleic acid binding protein, contains seven undergo cardiological examination and follow-up as
zinc finger domains. It is though to be an RNA-bind- soon as the diagnosis is established. Cataracts may
ing protein. It is widely expressed, with the highest ex- require surgical intervention at some point. Cardiac
pression in heart and skeletal muscle. The pathophys- therapy, including pacemaker implantation, may be
iology of the disease is presently unclear. necessary. Patients should be checked for hypo-
62.5 Magnetic Resonance Imaging 475

Fig. 62.2. The MRI of this 64-year-old female patient with ventricles and subarachnoid spaces are dilated due to reduced
DM 2 reveals extensive, patchy cerebral white matter abnor- volume of the cerebral white matter. Courtesy of Dr. E. Hund,
malities, sparing the corpus callosum and partially sparing the Department of Neurology, and Dr. Sartor, Department of
U fibers. The internal and external capsules are affected. There Neuroradiology, University of Heidelberg Medical School, Hei-
are spots of abnormal signal in the basal ganglia. The lateral delberg, Germany

thyroidism regularly. Whether DM 2 patients ex- 62.5 Magnetic Resonance Imaging


perience respiratory depression in response to
benzodiazepines, opiates, or barbiturates, whether MRI of the brain is often normal, but may show rather
myotonia is aggravated by depolarizing muscle relax- extensive abnormalities in the cerebral white matter
ants, and whether DM 2 patients may develop malig- (Figs. 62.1 and 62.2). The U fibers tend to be spared.
nant hyperthermia during or after anesthesia is The white matter abnormalities may be diffuse
unknown. (Fig. 62.1) or, more often, patchy (Fig. 62.2). These ab-
normalities may be accompanied by neurological
problems including clinical episodes of stroke,
parkinsonian features, seizures, and mental changes.
Chapter 63

X-linked Charcot–Marie–Tooth Disease

63.1 Clinical Features weakness, bulbar dysarthria, dysphagia, and loss of


and Laboratory Investigations the gag reflex. Nausea and vertigo may be present. The
patients are usually alert during the episode, but som-
Charcot–Marie–Tooth disease (CMT) is a clinically nolence, behavior abnormalities, and disorientation
and genetically heterogeneous group of peripheral may also occur. They may complain of headache.
nerve disorders characterized by distal weakness, After the episode, patients recover fully to their previ-
atrophy, sensory loss, and decreased tendon reflexes. ous condition. The episodes seem to be provoked by
CMT has been classified according to the pattern of minor infections, surgery, sports, or staying at high
inheritance and whether the abnormalities primarily altitudes.
affect myelin (CMT1) or axons (CMT2). The X-linked Nerve conduction velocities are reduced in CMTX,
dominant form of CMT (CMTX) shows both demyeli- but the reductions are less severe than in the other
nating and axonal aspects, causing problems in cate- forms of demyelinating CMT, consistent with moder-
gorizing CMTX as a subtype of CMT1 or CMT2. ate demyelination of the investigated nerves. The re-
CMTX affects both males and females, but males ductions in conduction velocities may be variable
are affected more severely than females, with onset of from one nerve segment to the other. Distal latencies
symptoms at an earlier age. Asymptomatic female are prolonged. Compound muscle action potentials
carriers have been reported. The clinical manifesta- and sensory nerve action potentials have reduced
tions of CMTX vary. Onset may be congenital or de- amplitudes. The electromyogram is usually normal,
layed until the third decade. Symptoms develop in a but sometimes fibrillation potentials are found, sug-
length-dependent distribution, meaning that the gesting axonal involvement. Nerve conduction veloc-
longest nerves are involved earliest. Thus, the weak- ities are typically slower in affected males than in
ness always starts in the lower leg muscles with stum- their affected female relatives. Abnormalities in brain
bling and steppage gait. With progression of the dis- stem auditory evoked responses have been reported
ease, the weakness spreads to the hands. The weak- repeatedly with slowed central conduction. Sensory
ness varies widely, from mild, causing no functional evoked responses may also show a slowing of central
impairment, to severe, necessitating the use of a conduction.
wheelchair. Foot deformities, including pes cavus and
hammer toes, are common. Muscle atrophy and loss
of tendon reflexes occur. Distal sensory loss with 63.2 Pathology
glove and stocking distribution occurs, involving
touch, pin prick, temperature, vibration, and position Findings in sural nerve biopsy specimens are variably
sense. Many patients have balance difficulties with a described as primary axonal degeneration and pri-
positive Romberg sign. Scoliosis may occur. Breathing mary demyelinating. Findings include loss of myeli-
difficulties due to phrenic nerve involvement occur in nated fibers and the presence of groups of small, thin-
some patients. ly myelinated and unmyelinated regenerating fibers.
Most CMTX patients do not have overt clinical Axons with disproportionately thin myelin sheaths in
CNS manifestations, but subclinical evidence of CNS relation to the axon caliber and small onion bulb for-
dysfunction is common. Hyperreflexia, extensor mations, indicative of demyelination and consecutive
plantar reflexes, some spasticity, and cerebellar dys- remyelination, may be present.
function have been observed in addition to the pe- No brain autopsy studies are available
ripheral neuropathy. Some patients have sensorineur-
al hearing loss. Patients may experience episodes of
transient neurological problems of central origin, 63.3 Pathogenetic Considerations
lasting hours to weeks. The problems may fluctuate in
severity during that time. Signs reported include CMTX is caused by mutations in the gene, called
dysarthria, cerebellar ataxia, gait ataxia, pyramidal CX32 or GJB1, coding for connexin 32, also called gap
weakness of the extremities (hemiparesis, monopare- junction protein b1. The gene is located on chromo-
sis, paraparesis, or tetraparesis), and cranial nerve some Xq13.1. Connexin 32 is a gap junction protein
palsies including third cranial nerve palsy, facial that is found in both the PNS and the CNS. It is incor-
63.5 Magnetic Resonance Imaging 477

Fig. 63.1. MRI of a 14-year-old male patient with CMTX during um of the corpus callosum is affected. There is no evident in-
an episode of CNS symptoms shows mild signal changes in the volvement of the brain stem and cerebellar peduncles. From
parieto-occipital white matter, sparing the U fibers.The spleni- Schelhaas et al. (2002), with permission

porated into cell membranes as a subunit of a cellular, transmembrane, and intracellular domains
hemichannel called the connexon. One connexon of the protein. By analogy to channelopathies, altered
comprises six subunits and two connexons form a gap gating properties of connexons due to mutated con-
junction between two cells. Gap junctions are inter- nexin 32 could give rise to transient CNS symptoms.
cellular channels that span the space between two Situations of metabolic stress may further alter the
neighboring cells and result from the association of function of gap junctions and cause transient symp-
two half-channels, connexons, contributed separately toms.
by each of the participating cells. These gap junctions
allow rapid exchange of ions, small metabolites, and
second messengers between cells or parts of cells. In 63.4 Therapy
the PNS, connexin 32 is expressed in Schwann cells at
the Schmidt–Lantermann incisures and the paran- At present, no specific therapy exists for patients with
odal regions, the noncompacted parts of myelin. In CMTX.
the CNS it is expressed in oligodendrocytes and neu-
rons. Connexin is also expressed in nonnervous tis-
sues including liver, pancreas, and secretory epitheli- 63.5 Magnetic Resonance Imaging
um.
Mutations either lead to loss of function or to al- Outside the episodes of CNS dysfunction, MRI of the
tered gating properties of the gap junction channels. brain is most often described as normal. During the
There is not yet a hot spot for connexin 32 mutations episodes of CNS symptoms, transient white matter
associated with CNS symptoms. The mutations asso- abnormalities occur (Fig. 63.1). They are located in
ciated with CNS disease have been found in the extra- the central or posterior part of the centrum semio-
478 Chapter 63 X-linked Charcot–Marie–Tooth Disease

vale, the splenium of the corpus callosum, posterior ter abnormalities are reversible within a few months,
limb of the internal capsule, and middle cerebellar the restricted diffusion cannot be caused by cytotox-
peduncles. The U fibers are spared. The signal ic edema. Myelin splitting and intramyelinic edema,
changes are milder than those seen in demyelination with compression of the extracellular space, is a more
or white matter inflammatory disease. They are sym- likely explanation.
metrical and confluent. After contrast administra- No systematic study of CMTX patients has been
tion, no enhancement is seen. Diffusion-weighted performed to document the MRI findings of the brain
imaging shows restricted diffusion. As the white mat- outside the episodes of CNS dysfunction.
Chapter 64

Oculodentodigital Dysplasia

64.1 Clinical Features 64.3 Pathogenetic Considerations


and Laboratory Investigations
ODDD is related to dominant mutations in the con-
Oculodentodigital dysplasia (ODDD) is an inherited nexin 43 gene, GJA1, at chromosome 6q22–23. A high
disorder that affects the development of the face, eyes, rate of de novo mutations is observed.
limbs, and teeth. The disorder has an autosomal dom- Connexin 43, like other connexin proteins, is a
inant mode of inheritance. It displays a high pene- transmembrane protein with an intracellular N-ter-
trance but variable expression. There is also intrafa- minus, four transmembrane domains, two extracellu-
milial variation of the clinical characteristics. Isolated lar loops, one cytoplasmic loop, and an intracellular
patients, related to de novo mutations, are frequently C-terminus. Six connexins can form a hemichannel or
observed. connexon, a specialized intracellular structure sur-
Affected patients have a narrow nose with thin, an- rounding a pore. Two connexons in apposing cell
teverted nostrils and a prominent columnella. Char- membranes can align to form an intercellular gap
acteristic eye manifestations include short palpebral junction. These channels provide a direct low-resis-
fissures, epicanthal folds, hyper- or hypotelorism, mi- tance intracellular pathway for the passage of ions
crophthalmia, and bilateral microcornea, often with and small molecules. Gaps junctions have been found
abnormalities of the iris. Some patients develop glau- in most tissues. Most tissues express more than one
coma or optic atrophy. Bilateral complete syndactyly type of connexin. Multiple types of connexins can
of the fourth and fifth fingers is the characteristic dig- assemble to form gap junctions between cells, with
ital malformation (type III syndactyly). The third fin- the diversity of combinations influencing the nature
ger may also be involved. Syndactyly of the third and of the cell-to-cell communication.
fourth toes may be present. Midphalangeal hypopla- Almost all GJA1 mutations in ODDD are missense
sia, distal phalangeal hypoplasia, or aplasia of one or mutations. The lack of mutations resulting in the in-
more digits or toes may be present. Associated camp- troduction of a stop codon into the protein suggests
todactyly or clinodactyly of the fifth fingers is a com- that the mechanism underlying ODDD is not a loss of
mon finding. The teeth are small and there is general- connexin 43 function. This hypothesis is reinforced
ized hypoplasia of the enamel. Partial anodontia may by the finding that the mouse knock-out does not
be present. Caries and premature loss of teeth may oc- have an ODDD-like phenotype. There is a strong cor-
cur. Less common features include dry, thin, slow- relation between the sites of GJA1 expression during
growing, and sparse hair (hypotrichosis), cleft lip and mouse embryonic development and the ODDD phe-
palate, conductive hearing loss, cranial and mandibu- notype, suggesting that connexin 43 plays a key role
lar hyperostosis, and microcephaly. in normal facial and limb development. The patho-
Neurological symptoms are frequent in ODDD. physiology of the disease in ODDD has still to be elu-
Slowly progressive spastic paraparesis of the legs has cidated.
been reported most often. Other reported problems ODDD and Hallermann–Streiff syndrome share
include ataxia, dysarthria, ptosis, nystagmus, gaze several clinical characteristics. Some patients with
palsy, strabismus, visual impairment, neurogenic Hallermann–Streiff syndrome have two mutations
bladder dysfunction, bowel disturbance, seizures, and in the GJA1 gene, whereas their parents are carri-
mild mental retardation. However, the clinical expres- ers. However, in other typical Hallermann–Streiff
sion of neurological features often varies widely syndrome patients no mutation in GJA1 can be
among affected individuals within and between found.
ODDD kindreds. The onset of the neurological prob-
lems is variable. It is usually evident by the second
decade of life, but may be much later. 64.4 Therapy

At present, only symptomatic therapy is available for


64.2 Pathology patients with ODDD.

No autopsy studies of ODDD are available.


480 Chapter 64 Oculodentodigital Dysplasia

Fig. 64.1. A 5.5-year-old girl with ODDD. The T2-weighted im- row, left) and FLAIR images (third row, middle and right) show
ages (first and second row) show ill-defined, mild periventricu- that the subcortical white matter is better preserved. Courtesy
lar signal abnormalities, extending downwards into the poste- of Dr. B. Tinselboer and Dr. F. Beemer, Department of Clinical
rior limb of the internal capsule. The inversion recovery (third Genetics, University Medical Center Utrecht, The Netherlands

64.5 Magnetic Resonance Imaging tively sparing the U fibers. The signal abnormalities
extend downwards into the posterior limb of the in-
In some patients with ODDD, MRI does not reveal ab- ternal capsule and the corticospinal tracts in the
normalities. Patients with ODDD and progressive brain stem. Low signal intensity on T2-weighted im-
neurological problems have white matter abnormali- ages has been reported in the pericentral cortex,
ties on MRI (Fig. 64.1). The cerebral white matter is globus pallidus, and substantia nigra. The imaging
somewhat reduced in volume. A zone of mildly ab- abnormalities in ODDD may not be very specific, but
normal signal surrounds the lateral ventricles, rela- they are consistent among patients.
Chapter 65

Leukoencephalopathy with Vanishing White Matter

65.1 Clinical Features months. In these infants evidence may be found of


and Laboratory Investigations involvement of other organs, including cataract (oil
droplet cataract), hepatosplenomegaly, kidney hy-
Leukoencephalopathy with vanishing white matter poplasia, pancreas involvement, and ovarian dysgen-
(VWM) has also been called childhood ataxia with esis.
central nervous system hypomyelination (CACH) At the other end of the clinical spectrum are pa-
and myelinopathia periaxialis diffusa.“Cree leukoen- tients who are completely normal until onset of the
cephalopathy” and “ovarioleukodystrophy” have both disease in adulthood. In some patients occasional
been found to be variants of VWM. seizures are the first sign of the disease. In other pa-
VWM is a disease with an autosomal recessive tients, the disease starts with psychiatric symptoms.
mode of inheritance. It has been shown to be one of Some patients slowly develop dementia. In other pa-
the more prevalent leukoencephalopathies in chil- tients motor deterioration dominates the clinical pic-
dren. Its incidence is similar to that of metachromat- ture. The episodes of major deterioration are usually
ic leukodystrophy. The classical and most frequent less marked in patients with a later onset. Later onset
variant has its onset in childhood, most often between most often implies a slower and more protracted dis-
the ages of 2 and 6 years. The disease is characterized ease course, although unexpected rapid progression
by chronic progressive neurological deterioration and death within a few months may also occur. In
with cerebellar ataxia, usually less prominent spastic- adult females with the disease, primary or secondary
ity, and relatively mild mental decline. Optic atrophy ovarian failure may occur.
with loss of vision may occur, but not in all patients. Laboratory tests are unrevealing in VWM. The on-
Epilepsy is present in most patients, but is rarely a ly consistent abnormality ever demonstrated is mod-
prominent feature. Characteristically, there are addi- erately elevated CSF glycine. The CSF:serum glycine
tional episodes of major and rapid deterioration fol- ratio is also elevated and may even be higher than
lowing minor head trauma and especially febrile in- 0.08. A ratio higher than 0.08 is thought to be indica-
fections. Occasionally, these episodes are provoked by tive of nonketotic hyperglycinemia. It is not known
fright. During these episodes, patients rapidly lose whether CSF glycine is elevated in all patients with
motor faculties and become very hypotonic. Irritabil- VWM and whether a normal level excludes the
ity, vomiting, and seizures usually progress to somno- diagnosis. The glycine elevation is probably a non-
lence and loss of consciousness. These episodes may specific finding related to ongoing excitatory brain
end in coma. Death usually follows an episode of damage. DNA testing is available for diagnostic con-
coma. If recovery occurs, it is usually incomplete. firmation, carrier testing, and prenatal diagnosis.
Some patients have a more severe variant of the
disease with onset within the first year of life and ear-
ly demise. An example is found in so-called “Cree 65.2 Pathology
leukoencephalopathy,” a disease that was described
among the Cree Indians. The onset of Cree leukoen- In VWM, the brain is generally of normal size and
cephalopathy is between the ages of 3 and 9 months, weight, but in a few cases it is slightly heavier or
and death occurs before the age of 2 years. Some lighter than normal. The gyri are usually normal or
(non-Cree) patients have an even more severe pheno- atrophic. When the brain is removed from the skull,
type with antenatal onset. In the third trimester of the rarefied and cystic white matter usually collapses.
pregnancy, decreased fetal movements, oligohydram- On macroscopic examination, gray matter structures
nios, growth failure, and development of micro- appear unaffected. The lesions are found primarily in
cephaly occur. At birth, contractures may be seen. cerebral white matter, with the cerebellum and brain
From birth onwards or from soon after birth, there stem much less severely involved. The cerebral white
are increasing problems, with a rapid downhill course matter varies from gelatinous to cystic to frankly
characterized by feeding problems, vomiting, failure cavitary. The frontoparietal white matter, particularly
to thrive, irritability, apathy, axial hypotonia, limb deep and periventricular, appears to be more com-
hypertonia or hypotonia, seizures, apneic episodes, monly involved, with relative sparing of the temporal
coma, respiratory failure, and death within a few lobe. White matter areas that are characteristically
482 Chapter 65 Leukoencephalopathy with Vanishing White Matter

relatively spared include the optic system, corpus cal- markers (bcl2, survivin), and proapoptotic markers
losum, anterior commissure, and internal capsule. (bac, bax, TUNEL, and activated caspase-3) has been
The subcortical arcuate fibers also tend to be spared, found in oligodendrocytes. There is a trend that in
but not consistently. In the brain stem the central early-onset devastating encephalopathy, loss of oligo-
tegmental tract at the level of the pons is often in- dendrocytes dominates with enhanced staining for
volved in a bilateral and symmetrical distribution. apoptosis markers, whereas in older patients with
Microscopically, the grossly affected white matter longstanding disease, proliferation of oligodendro-
shows myelin pallor, thin myelin sheaths, vacuolation, cytes dominates with striking increases in their num-
myelin loss, cystic change, and rarely active demyeli- bers. There is a meager to moderate response by as-
nation. Lipophages containing myelin breakdown trocytes and microglial cells in this disease, even in
products are rare. There is no significant inflammato- areas near the cavitation. Only scant lipophages are
ry response (lymphocytes, plasma cells, neutrophils, seen, and the astrocytes appear to be dysmorphic
eosinophils). The gray matter is generally spared or with blunt, broad processes rather than their typical
greatly preserved in comparison to white matter. delicate arborizations.
However, astrocytosis and microgliosis of the overly-
ing cortex may be found. Subtle mineralization of the
basal ganglia has been observed. While axonal loss is 65.3 Pathogenetic Considerations
complete in areas of cavitation, the less involved areas
demonstrate a more variable loss of axons. Some- VWM is related to defects in translation initiation
times the axonal loss is as severe as the myelin loss, factor eIF2B. eIF2B consists of five nonidentical sub-
but in other cases or areas axons are relatively spared. units (eIF2Ba, eIF2Bb, eIF2Bg, eIF2Bd, and eIF2Be),
Myelin sheaths are abnormal and vary from pale to all of which are encoded by different genes (EIF2B1,
thin to vacuolated. Vacuolated myelin is noted in pre- EIF2B2, EIF2B3, EIF2B4, and EIF2B5, respectively)
served areas, often near the cystic to cavitated lesions. located on different chromosomes (12q24.3, 14q24,
The vacuoles are bordered by material staining posi- 1p34.1, 2p23.3, and 3q27, respectively). Mutations in
tive with Luxol fast blue or with myelin basic protein, any of these genes can independently cause the dis-
suggesting that these vacuoles reflect intramyelinic ease.
edema. Ultrastructural studies have revealed some Translation of mRNA into polypeptides is one of
myelin vacuoles suggestive of intramyelinic edema. the major energy-consuming processes in the cell and
However, a re-evaluation of the vacuolated white mat- is therefore, not surprisingly, a tightly regulated
ter following the recognition of “foamy” oligodendro- process. The initiation phase, in which ribosomes are
cytes has raised questions about how much vacuola- assembled on mRNA, is controlled via several differ-
tion of white matter is related to intramyelinic edema ent signaling pathways. Multiple so-called eukaryotic
and how much is due to the presence of these vacuo- initiation factors (eIFs) are involved in translation
lated oligodendrocytes. The radiating stripes within initiation, and among them the guanine nucleotide
the rarefied white matter, seen on MRI, seem to corre- exchange factor eIF2B plays a key regulatory role. A
late with blood vessels accompanied by reactive as- crucial step in translation initiation is the delivery
trocytes. by eIF2 of the initiator methionyl-transfer RNA (Met-
Different types of glia are involved in the disease tRNAi) to the small ribosomal subunit. Upon recogni-
process. In and around the areas that are cavitated, tion of the start codon and binding of methionine to
oligodendrocytes demonstrate marked losses, al- it, the eIF2-bound guanosine triphosphate (GTP) is
though they are still relatively high in number as hydrolyzed and eIF2 is released in its inactive guano-
compared to other types of cells. In areas that are bet- sine diphosphate (GDP)-bound form. In order to
ter preserved an increase in apparently mature oligo- bind another Met-tRNAi and initiate the production
dendrocytes is seen, most consistently in the arcuate of another protein, active eIF2 must be regenerated by
fibers, other regions bordering the cystic to cavitated exchange of GDP for GTP. This step is catalyzed by
lesions, anterior commissure, the internal capsule, eIF2B. The exchange of GDP for GTP by eIF2B is re-
and the corpus callosum. Oligodendrocytes may have quired for each round of translation initiation. Thus,
an abnormal appearance and look vacuolated or eIF2B is necessary for the production of all proteins
“foamy.” At the ultrastructural level the vacuoles are in the body. Regulation of this step can control global
membranous structures associated with mitochondr- rates of protein synthesis under diverse conditions.
ial membranes, and, in places, contiguous with myelin Protein synthesis is markedly inhibited under a
lamellae. These oligodendrocytes also combine many variety of stress conditions and in the recovery phase
mitochondria, proteolipid protein mRNA, and finger- that follows. This response is part of a protective
print structures – the latter two attributes also noted mechanism of cells elicited by various stimuli, includ-
in nonvacuolated oligodendrocytes. Positive staining ing thermal, chemical, oxidative, or physical trauma,
for both proliferative markers (Ki-67), antiapoptotic called the cellular stress response or heat shock re-
65.4 Therapy 483

sponse. Stress may lead to misfolding and denatura- unfolded protein response in VWM. However, many
tion of proteins, contributing to cell dysfunction and aspects of the disease are poorly understood. eIF2B is
death. The inhibition of normal RNA translation dur- ubiquitously expressed in all cells of the body. It is
ing stress is thought to enhance cell survival by limit- unclear why the brain is preferentially or selectively
ing the accumulation of denatured proteins and sav- affected, whereas other organs are only involved in
ing cellular energy. the most serious variants of the disease.
Inhibition of mRNA translation can be achieved It is becoming increasingly clear that there is some
through the modification of several initiation factors. genotype–phenotype correlation. Some mutations
Most stress conditions, including heat stress, lead to are consistently, although not invariably, associated
activation of specific kinases that phosphorylate eIF2 with a mild phenotype, whereas other mutations are
on its a-subunit. In this phosphorylated form, eIF2 consistently associated with a severe phenotype. One
binds tightly with eIF2B and in this way is a competi- example is the Arg113His mutation in eIF2Be, which
tive inhibitor of eIF2B, preventing the recycling of in the homozygous state is almost always associated
eIF2. The concentration of eIF2 usually exceeds that with a late onset and slow progress. It is remarkable
of eIF2B. Therefore, even modest levels of eIF2a that histidine is normal at position 113 in mouse and
phosphorylation can potentially lead to complete in- rat. However, childhood-onset patients homozygous
hibition of translation initiation and protein synthe- for the Arg113His mutation have been observed too.
sis. In certain cell types, inactivation of eIF2B at The second example is the Arg195His in eIF2Be, the
40–41 °C can be achieved without changes in eIF2a Cree leukoencephalopathy mutation, which in the
phosphorylation. eIF2B activity can also be regulated homozygous state is invariably associated with an
through other pathways, such as phosphorylation at early onset and early death. There is also evidence for
different sites, which can enhance or suppress eIF2B a correlation between decrease in guanine-nucleotide
activity.Whether these latter pathways are involved in exchange factor (GEF) activity of eIF2B and the age at
the regulation of eIF2B activity under stress condi- onset of the disease. Mutations with a more serious
tions is unclear. impact on eIF2B function seem to be associated with
The essential role of eIF2B, both in normal protein a more serious disease course. However, there is also
production and in its regulation under different con- major variation between patients carrying the same
ditions, including elevated temperature, is reflected mutations and between affected siblings in the same
by the evolutionary conservation of the complex and family. So, environmental and other genetic factors
the nonviability of yeast null mutants for each of the influence the phenotype as well.
subunits except eIF2Ba. In VWM patients, most mu-
tations are missense mutations. So far, major muta-
tions, which prevent the expression of full-length 65.4 Therapy
eIF2B subunits, have only been observed in the com-
pound heterozygous state with a missense mutation There is presently no causal treatment for VWM, but
as second mutation. certain preventive measures seem advisable. Particu-
At a biochemical level, evidence has been provided lar stress conditions should be avoided in patients
that mutations reduce eIF2B activity. Certain muta- with VWM: infections, high temperatures, and head
tions impair the ability to form the five-subunit eIF2B trauma. Vaccinations to prevent infectious diseases
holocomplexes, leading to diminished eIF2B activity. are important. In the case of fever, it is essential to
Point mutations in the catalytic domain, located at keep the temperature down with antipyretics, if nec-
eIF2Be, impair its ability to bind eIF2. Some other mu- essary with cooling. It is important to be liberal with
tations in eIF2Bb actually enhance eIF2 binding, also antibiotics. In children with frequent upper respirato-
impairing eIF2B function. ry tract infections, daily low-dose antibiotics may be
The pathophysiology of VWM is still difficult to considered. Patients have reported deterioration after
explain. Serious deteriorations often follow febrile in- sun bathing. Playing for a long time in the full sun
fections and other forms of cellular stress. Inhibition during hot weather may, therefore, be something to
of eIF2B via the phosphorylation of eIF2a is an im- avoid. It is impossible to avoid the minor head trau-
portant mechanism for slowing down protein synthe- mas of daily life, but it is better to avoid certain types
sis in response to, for example, the accumulation of of physical contact sports. During episodes of major
unfolded proteins in the endoplasmic reticulum. This deterioration, corticosteroids have led to temporary
regulation of eIF2B is likely to be of particular impor- improvements. However, this effect is not consistent
tance in preventing denatured proteins from accumu- and lasting beneficial effects have never been ob-
lating during cellular stress and could provide a clue served. Considering the potential adverse effects of
as to why VWM is exacerbated by episodes of infec- steroids, in particular worsening of infections, their
tion and trauma. There is evidence of activation of the use during episodes of deterioration is not advocated.
484 Chapter 65 Leukoencephalopathy with Vanishing White Matter

65.5 Magnetic Resonance Imaging missure is spared. The cerebellar white matter may
have a normal (Fig. 65.4) or mildly abnormal signal
MRI of the brain shows extensive cerebral white mat- (Figs. 65.1, 65.2 and 65.5). However, the process of rar-
ter changes in VWM (Figs. 65.1–65.5). Over time, MRI efaction and cystic degeneration appears to be re-
shows evidence of disappearance of the affected white stricted to the cerebral hemispheric white matter.
matter, which is replaced by fluid (Figs. 65.2 and 65.3). Over time, atrophy of the cerebellum and sometimes
On T2-weighted images, abnormal white matter and also the brain stem ensues. There are often signal ab-
cystic white matter both have a high signal intensity normalities in the midbrain and pons, and sometimes
and cannot be distinguished. Proton-density or in the medulla (Figs. 65.2 and 65.5). Especially, the
FLAIR images are necessary to demonstrate the white central tegmental tracts in the pontine tegmentum
matter rarefaction and cystic degeneration. Abnor- are often involved. Unlike the cerebral and cerebellar
mal white matter has a high signal on proton-density white matter abnormalities, the brain stem signal ab-
and FLAIR images, whereas cystic white matter has a normalities, when present, may improve again and
low signal intensity, similar to the signal of CSF. Rar- even disappear. They seem to be particularly promi-
efied white matter has an intermediate signal intensi- nent during episodes of lowered consciousness
ty, not as low as CSF. Within the rarefied and cystic (Figs. 65.2 and 65.5). MRI-visible spinal cord lesions
white matter a stripe-like pattern is visible, suggest- are rare but may occur.
ing remaining tissue strands (Fig. 65.6). Heavily T2- The cerebral and cerebellar cortex have a normal
weighted images are as a rule not suitable to show the appearance. Most often, the basal ganglia and thala-
stripes, because these images do not allow distinction mus also have a normal signal intensity, but particu-
between the stripes of abnormal white matter and larly thalamus and globus pallidus lesions may occur
rarefied or cystic white matter, both appearing bright. (Figs. 65.2 and 65.5). Thalamus lesions may also im-
In exceptional cases, the stripes are dark on T2- prove and disappear all together. The globus pallidus
weighted images (Fig. 65.6). The U fibers are spared may have a low signal intensity on T2-weighted im-
to a variable extent. In some patients a rim of subcor- ages, suggesting mineralization (Figs. 65.4 and 65.5).
tical white matter is spared, whereas in other patients The above MRI description applies to the late-in-
the U fibers are abnormal (Fig. 65.7). The sparing of fantile, childhood, juvenile, and adult-onset disease
the U fibers is seen best on T1-weighted images (Figs. 65.1–65.10). The MRI picture may be much
(Fig. 65.7). MRI has been performed in a few more difficult to diagnose in early-infantile VWM. In
presymptomatic and oligosymptomatic individuals evidently affected neonates, the brain may only look
and in all persons diffuse cerebral white matter ab- immature with a gyral pattern that is too coarse for
normalities have been found, although initially not the gestational age of the child and with immature
necessarily with evidence of rarefaction or cystic de- white matter having a high water content and little
generation (Figs. 65.8 and 65.9). In the end stage of myelin, but little or no rarefaction (Figs. 65.11–65.13).
VWM all cerebral hemispheric white matter may have Over time, the cerebral white matter may look in-
vanished, leaving a ventricular wall and cortex with creasingly abnormal, rarefied, and cystic, as seen in
little or nothing in between (Figs. 65.2 and 65.3). It is the later-onset variants of the disease, but the cerebral
striking that although the white matter may be high- white matter may also become highly atrophic, the
ly cystic, the brain does not collapse and rarely shows ependymal lining (almost) touching the depth of the
evidence of external atrophy. On the contrary, the gyri (Fig. 65.13), very much unlike what is seen in the
cerebral white matter may have a (mildly) swollen later-onset variants of the disease. In these severely
appearance with broadening of gyri (Fig. 65.3). Even affected infants, the brain as a whole is highly atroph-
when there seems to be hardly any cerebral white ic, including also the corpus callosum, cerebellum,
matter left, there is a distance between ependymal lin- and brain stem. In early-onset VWM, the cerebellar
ing and the cortex, apparently filled with fluid. The white matter may also become cystic.
lateral ventricles may be mildly to moderately dilated, Proton MRS shows stage-dependent abnormali-
but not seriously so.White matter swelling is predom- ties. In the initial stages, when there is little white mat-
inantly seen in young children, whereas more serious ter rarefaction, the white matter spectrum is relative-
atrophy is seen in teenagers and adults with VWM ly preserved. With ongoing rarefaction and cystic de-
(Fig. 65.4). generation, the signals decrease, finally to disappear
Often a dilated cavum septi pellucidi and cavum altogether. Finally, the spectrum is a CSF spectrum
Vergae are present. The inner rim of the corpus callo- with some lactate and glucose and no or minor “nor-
sum is usually involved (Figs. 65.8 and 65.9), and in mal” peaks. The cortex spectrum remains well pre-
later stages the corpus callosum becomes thin served throughout.
(Fig. 65.10). The posterior limb of the internal capsule A study using phosphorus MRS of the brain
is often involved, whereas the anterior limb of the in- (Blüml et al. 2003) revealed evidence for an altered
ternal capsule is rarely involved. The anterior com- energy state of the residual cells in the cerebral white
65.5 Magnetic Resonance Imaging 485

Fig. 65.1. The cerebral white matter is diffusely abnormal in signal intensity of CSF, consistent with white matter rarefac-
this 3.5-year-old girl with VWM.The posterior limb of the inter- tion and cystic degeneration. Within these areas dots and
nal capsule is affected. The brain stem is intact. The cerebellar stripes with a high signal intensity are seen, consistent with re-
white matter has a mildly abnormal signal intensity.The FLAIR maining strands of abnormal tissue. The sagittal T1-weighted
images (third row, left and middle) demonstrate that within the image (third row, right) shows a pattern of radiating stripes,
white matter that is abnormal on the T2-weighted images compatible with better preserved tissue strands
there are areas with a low signal intensity, close or equal to the

matter. Nucleoside triphosphate and inorganic phos- tion, the nucleoside triphosphate to inorganic phos-
phate were reduced, whereas phosphocreatine was phate ratio normally being lower in gray matter than
elevated. The relative preservation of gray matter over in white matter. Of the metabolites involved in
white matter may have contributed to this observa- biosynthesis and catabolism of membrane phospho-
486 Chapter 65 Leukoencephalopathy with Vanishing White Matter

Fig. 65.2.
65.5 Magnetic Resonance Imaging 487

Fig. 65.3. The T2-weighted (first row) and FLAIR images (sec- images (third row) show that all cerebral white matter has van-
ond row) of a severely affected 2-year-old boy with VWM show ished. The lateral ventricles are mildly dilated, but the cerebral
the characteristic abnormalities. Note the large cavum septi cortex has not collapsed over the central structures. On the
pellucidi. Ten months later he has lost almost all functions. He contrary, the vanished white matter look swollen with broad-
is blind and has no intentional movements. The MRI was per- ening of gyri
formed because he had papilledema at funduscopy.The FLAIR

Fig. 65.2. The T2-weighted images of this 3-year-old girl with tion and cystic degeneration than in the previous patient. Five
VWM (first two rows) show diffuse involvement of the cerebral months later (third and fourth rows), she is in coma. The FLAIR
white matter, posterior limb of the internal capsule, and cere- images (third row, left) show that the cerebral white matter is
bellar white matter. Within the pons the central tegmental now largely cystic. The T2-weighted images show that the
tracts and the pyramidal tracts are affected. The FLAIR image globus pallidus, midbrain, and medulla now have an abnormal
(first row, left) shows much more serious white matter rarefac- signal.The girl died soon after the MRI
488 Chapter 65 Leukoencephalopathy with Vanishing White Matter

Fig. 65.4. This 18-year-old girl has a milder form of VWM. Al- the cerebral white matter is rarefied but not cystic. The FLAIR
though she has been symptomatic since the age of 4 years,she image (third row, right) shows the characteristic radiating
can still walk without support. The T2-weighted images show stripes of better preserved tissue strands. The globus pallidus
diffuse white matter involvement with some atrophy, but the has a low signal intensity, probably related to mineralization.
proton-density images (third row, left and middle) show that The brain stem and cerebellar white matter are intact

lipids, glycerophosphoethanolamine was reduced changed with serious rarefaction and altered ratios of
and phosphotidyl ethanolamine was increased, composing cells. Why a defect in the regulation of
whereas choline-containing phosphorylated metabo- protein synthesis would selectively affect ethanol-
lites were unchanged. It is difficult to interpret these amine phospholipid metabolism and leave choline
findings, most of all because the composition of the phospholipid metabolism unaffected is presently un-
remaining brain tissue in VWM is dramatically clear.
65.5 Magnetic Resonance Imaging 489

Fig. 65.5. The girl illustrated in this figure presented for the and FLAIR (third row,right) images show evidence of some bet-
first time at the age of 16, lapsed into coma and died after 4 ter preserved tissue strands.The T2-weighted images show sig-
months. The T2-weighted images show the diffuse white mat- nal abnormalities in the internal capsule, thalamus, midbrain,
ter involvement, but the proton-density images (third row, left basis of the pons, central tegmental tracts, middle cerebellar
and middle) show that the white matter is rarefied but not cys- peduncles, cerebellar white matter, and medulla
tic. This was confirmed at autopsy. Both the proton-density
490 Chapter 65 Leukoencephalopathy with Vanishing White Matter

Fig. 65.6. The first row of images was obtained in a 5-year-old dots with a high signal intensity are seen, consistent with
boy with VWM; the second row in a 6-year-old girl with VWM. In transections of strands of abnormal white matter. In the girl an
the boy the typical MRI features of VWM are seen with a pat- unusual pattern is seen. The sagittal (left) and axial (middle)
tern of radiating stripes on the sagittal T1-weighted image T2-weighted images show the diffuse abnormality of the cere-
(left), diffuse white matter abnormalities without evidence of bral white matter.Within the abnormal white matter a pattern
stripes and dots on the axial T2-weighted image (middle), of dark radiating stripes is seen, apparently representing
whereas on the FLAIR image (right) the abnormal white matter strands of normal tissue. The FLAIR image (right) shows some
has a high signal in some parts and a low signal in others, the white matter rarefaction in the parietal area.Images of the sec-
latter compatible with rarefaction and cystic degeneration. ond patient courtesy of Dr. T. Polster, Gilead Pediatric Center,
Within the rarefied white matter of the centrum semiovale, Bielefeld, Germany
65.5 Magnetic Resonance Imaging 491

Fig. 65.7. The first row of images originates from a 9-year-old (right) images reveal the characteristic features of VWM.The IR
girl with VWM, the second row from a 16-year-old girl with image (left) shows that the U fibers are spared in the first
VWM. The T2-weighted (middle) and FLAIR or proton-density patient, whereas they are affected in the second patient
492 Chapter 65 Leukoencephalopathy with Vanishing White Matter

Fig. 65.8. This 18-year-old university student presented with cerebral white matter and the inner rim of the corpus callo-
a single seizure and had no abnormalities at neurological ex- sum. The FLAIR images (second row) do not show evidence of
amination. She has a DNA-confirmed diagnosis of VWM. The white matter rarefaction. Courtesy of Dr. E Storey, Neurogen-
T2-weighted images (first row) show diffuse involvement of the etics Clinic, Royal Melbourne Hospital, Parkville, Australia

Fig. 65.9. A 13-year-old girl has a history of complicated mi- matter and inner rim of the corpus callosum with sparing of all
graines,but no abnormalities at neurological examination.She subcortical white matter.There is no evidence of white matter
has a DNA-confirmed diagnosis of VWM. These FLAIR images rarefaction. Courtesy of Dr. M. D’Hooghe, Department of Neu-
show abnormalities in the periventricular and deep white rology, Sint-Jan General Hospital, Brugge, Belgium
65.5 Magnetic Resonance Imaging 493

Fig. 65.10. A 49-year-old man with a history of presenile dementia and motor problems. His MRI is typical for VWM and the
diagnosis was DNA-confirmed. Note the thin corpus callosum. From Prass et al. (2001), with permission

Fig. 65.11. The VWM patient in this figure had an antenatal ages were obtained at the age of 3 months and revealed most
onset of symptoms with decreased fetal movements and of all an immature brain with failure of myelination and insuf-
oligohydramnios. At birth she had dislocated hips and was hy- ficient gyral development.The FLAIR image shows some white
potonic. She followed a rapidly downhill course with in- matter rarefaction. The right image was obtained at 6 months
tractable seizures, feeding difficulties, hypotonia, apathy, and and revealed more prominently abnormal cerebral white mat-
finally coma and respiratory failure.At physical examination oil ter with additional involvement of the globus pallidus and the
droplet cataracts and hepatosplenomegaly were found. She right thalamus. From van der Knaap et al. (2003), with permis-
died at 8 months.The left (T2-weighted) and middle (FLAIR) im- sion
494 Chapter 65 Leukoencephalopathy with Vanishing White Matter

Fig. 65.12. This figure shows the images of two siblings with tained at 3 and 4 months revealed more prominent abnormal-
antenatal onset of VWM and death at the ages of 3.5 and 4 ities of the cerebral white matter. Myelination and gyration
months, respectively.The first row of images was obtained at 1 have not progressed. There is white matter volume loss with
month in a girl;the second row shows the follow-up images ob- enlargement of the lateral ventricles. Note the swollen and
tained at 4 months. The third row shows the images of her af- cystic anterior temporal white matter. The cerebellum is small
fected brother, obtained at 3 months. The images obtained at and the brain stem looks atrophic.Courtesy of Dr.R.van Coster,
1 month reveal an immature brain with failure of myelination Department of Pediatric Neurology, C. Hooft University Hospi-
and insufficient gyration. Note the swelling of the anterior tal, Gent, Belgium
temporal white matter, which contains a cyst. The images ob-

Fig. 65.13. A patient with antenatal onset of VWM underwent ing white matter, the ependyma touches the cortex.The later-
MRI at the ages of 5 days (first and second rows) and 5 months al ventricles are now highly dilated. There is a cyst in the ante-
(third and fourth rows). Initially, the brain has an immature rior temporal region. The remaining cerebral white matter
appearance with coarse gyri and cerebral white matter with a looks highly abnormal and swollen. The cerebellum has be-
high water content. The inferior horns of the lateral ventricles come highly atrophic. The brain stem is also thin. From
are dilated. On follow-up, most of the cerebral white matter Boltshauser et al. (2002) and van der Knaap et al. (2003), with
has disappeared, but, unlike the typical appearance of vanish- permission
65.5 Magnetic Resonance Imaging 495

Fig. 65.13.
Chapter 66

Aicardi–Goutières Syndrome

66.1 Clinical Features the same family, one affected child may achieve no de-
and Laboratory Investigations velopmental milestones at all and be severely spastic,
whereas the affected sibling is walking with support.
Aicardi–Goutières syndrome is a rare leukoen- Laboratory investigations have revealed no abnor-
cephalopathy with an autosomal recessive mode of malities apart from mild chronic lymphocytic pleo-
inheritance. The disease presents at birth or within cytosis in the CSF, with a cell count usually varying
the first few months of life. In some patients intrauter- between 10 and 300 cells/mm3. The CSF lymphocyto-
ine growth retardation and a deceleration of head sis is more or less obligatory in the early stages of the
growth have been reported. Early postnatal signs are disease, but may disappear afterwards. The duration
irritability, jitteriness, feeding problems, and failure of the persistence is probably quite variable. Protein
to thrive. Often a high-pitched cry is noted. Recurrent level varies from normal to more than 1 g/l. There is
febrile episodes of unknown cause may occur in the no evidence of intrathecal immunoglobulin synthe-
presence of normal laboratory investigations. Gener- sis: IgG index is normal and there are no oligoclonal
alized hypertonia and spasticity are present from the bands. Interferon-a is elevated in blood and CSF, but
beginning, or develop in the course of the first much higher in CSF than in blood, indicative of in-
months. Dystonic posturing may occur. Truncal hy- trathecal synthesis. The elevation of interferon-a is
potonia, poor head control, and opisthotonic postur- probably consistently present, at least in the early
ing are often noted. Head growth is usually poor with stages of the disease and probably also for a long time
progressive microcephaly, but some patients have a thereafter. Electron microscopy of CSF lymphocytes
normal head circumference. Gain in height and may reveal tubuloreticular inclusions, indicative of
weight is usually normal or just below normal. Some interferon deposits. Similar deposits may also be pre-
but not all patients have seizures.Vision is often poor. sent in endothelial cells on skin biopsy. There is no
Some patients have pale optic discs, whereas others evidence of congenital infections; TORCH (toxoplas-
have normal fundi. Pupillary reactions are normal. mosis, rubella, cytomegalovirus, and herpes simplex)
Nystagmus may be present. There is a variably severe screening is negative. Extensive screening for inborn
developmental delay. In many patients no, or at most errors of metabolism has never revealed abnormali-
few, developmental milestones are reached. However, ties. In some patients increased CSF pterins and low-
some patients achieve walking with or without sup- ered CSF folate levels have been found.
port. Some patients learn to talk. Multiple patients have been reported with atypical
Some patients have extraneurological features. The findings. At present it is not clear whether these cases
most prominent of these are cutaneous lesions, which are variant forms of Aicardi–Goutières syndrome or
mainly affect the fingers and toes, and sometimes should be considered separate, distinct disease enti-
also the ear lobes, in the form of scaly erythematous ties. A few patients reported had atypical manifesta-
lesions and an acrocyanotic appearance reminiscent tions including microcytic anemia (Kumar et al.
of chilblains. Patients may have puffy edema and 1998) or growth hormone deficiency and retinal de-
swelling of the proximal parts of fingers and toes generation (Bönnemann et al. 1991). Another condi-
tapering towards the end. tion to be considered is the microcephaly–intracra-
The disease is usually considered to be progressive, nial calcifications syndrome (MICS), also called pseu-
although in patients with severe and early-onset dis- do-TORCH syndrome, which has a neonatal-onset
ease it may be difficult to determine whether this is encephalopathy and shares essential features with
the case. Patients with milder disease may show ongo- Aicardi–Goutières syndrome, especially calcium de-
ing development for many years, although in others a posits within the cerebral white matter and basal gan-
clear regression is noted. Many patients die early, glia, but also has features suggestive of a congenital
within either months or a few years of birth. Most infection, such as neonatal thrombocytopenia, he-
children die in a vegetative state from pneumonia. patosplenomegaly, congenital microcephaly, and
However, survival beyond the teenage period has also cataracts in the presence of negative TORCH antibod-
been observed. When multiple affected patients are ies (Burn et al. 1986; Reardon et al. 1994; Wieczorek et
present within the same family, striking differences in al. 1995, Monastiri et al. 1997; Slee et al. 1999; Vivarel-
the severity of the disease may be observed. Within li et al. 2001). Cree encephalitis, reported among the
66.3 Pathogenetic Considerations 497

Cree Indians, leads to microcephaly, intracerebral cal- tion is minor and limited to the areas of necrosis and
cifications, hepatosplenomegaly, and thrombocy- to the leptomeninges.
topenia, similar to the pseudo-TORCH syndrome
(Black et al. 1988). Lupus erythematosus has been de-
scribed in two siblings with a pseudo-TORCH syn-
drome born to consanguineous parents (Dale et al. 66.3 Pathogenetic Considerations
2000). The patients presented with early-onset en-
cephalopathy and typical signs of pseudo-TORCH The disease is has an autosomal recessive mode of in-
syndrome. Within a year they developed hypocom- heritance. The basic defect is unknown. A genome-
plementemia and a systemic lupus erythematosus- wide linkage study has revealed a locus for a disease
like rash. The diagnosis of systemic lupus erythe- gene on chromosome 3p21, but no gene associated
matosus was made on the basis of a characteristic au- with Aicardi–Goutières syndrome has yet been iden-
toantibody profile. Finally, there are reports on pa- tified. Evidence for locus heterogeneity has been pro-
tients who share similarities with Aicardi–Goutières vided, suggesting the existence of at least one addi-
syndrome patients, including the cerebral white mat- tional disease gene. The problem presently is the
ter abnormalities and punctate calcium deposits in clinical definition of the disease. There is a striking
the white matter and basal ganglia, but also have clinical heterogeneity and it is not known whether
symptoms not reported in other patients, such as hy- Aicardi–Goutières syndrome, variant forms, MICS/
pothyroidism and pericardial effusions, and lack CSF pseudo-TORCH syndrome, and Cree encephalitis
lymphocytosis and elevated CSF interferon-a levels represent the same disorder or not. They may repre-
(Schwarz et al. 2002). sent closely related but different disorders with de-
fects in different single genes, probably all involved in
similar or the same processes, or they may represent
one disorder, associated with not a single but a small
66.2 Pathology number of genes that are involved in the same
process.
Postmortem investigations demonstrate gross atro- The chronic CSF lymphocytosis and elevated in-
phy of the cerebral hemispheres, corpus callosum, terferon-a levels are central and striking features in
brain stem, and cerebellum. On sectioning, reduction Aicardi–Goutières syndrome. Interferons constitute a
in white matter volume and enlargement of the ven- family of signal proteins. Upon binding to specific re-
tricles are noted. Calcium deposits are consistently ceptors they induce activation of a signal transduc-
present. Most dense calcium deposits are seen in the tion pathway that activates a broad range of genes,
globus pallidus, putamen, caudate nucleus, thalamus, which are involved in antiviral, immunomodulatory,
and dentate nucleus. Less dense calcium deposits are and antiproliferative activities. Interferon-a is in-
variably noted in the cerebral cortex, cerebral white creased to variable levels in viral infections, depend-
matter, cerebellar white matter, and cerebellar cortex. ing on the stage of the disease. Interferon-a does not
The calcium deposits are usually punctate, but large cross the blood–brain barrier, so if levels are higher in
concrements may also be seen. On microscopy the the CSF than in the blood, it is a result of intrathecal
calcium deposits are shown to be vascular and synthesis. It is elevated in the CSF during viral menin-
perivascular. Many small vessels, both veins and ar- gitis and encephalitis, but elevation is not a sign of
teries, exhibit thickened media and adventitial wall neurodegenerative disorders in general. It is probable
with circular and focal calcium deposits within the that interferon-a is important in the pathophysiology
media, adventitia, and perivascular spaces. In addi- of Aicardi–Goutières syndrome. Transgenic mice
tion, some lamellar calcium deposits not associated with astrocyte-targeted chronic overproduction of
with blood vessels are seen in the cerebral and cere- interferon-a develop progressive inflammatory en-
bellar white matter. The cerebral and cerebellar white cephalopathy with angiopathy and calcifications in
matter and the brain stem display a diffuse inhomo- the basal ganglia. These neuropathological features
geneous myelin deficiency associated with astrocyto- are very similar to those found in Aicardi–Goutières
sis. Areas of complete myelin deficiency may show syndrome, suggesting a causal relationship between
cavitation. Signs of active myelin breakdown are lack- the elevated levels of interferon-a and the disease.
ing; no sudanophilic material is present. Some ascribe Histopathological findings suggest that a calcify-
the myelin deficiency to hypomyelination and others ing microangiopathy may underlie Aicardi–Goutières
to myelin loss. In the cerebral and cerebellar cortex a syndrome. Small vessels have a highly abnormal ap-
multitude of wedge-shaped microinfarctions has pearance with thickened walls and intramural and
been reported with abnormal small vessel prolifera- perivascular calcium deposits. The wedge-shaped mi-
tion. These infarctions are probably related to the croinfarctions in the cerebral and cerebellar cortices
widespread calcifying microangiopathy. Inflamma- are highly suggestive of involvement of small arteri-
498 Chapter 66 Aicardi–Goutières Syndrome

oles. The inhomogeneous, irregular aspect of the syndrome, CT did not reveal any calcifications, or on-
white matter abnormalities, with patches of pre- ly in the cerebral white matter along the ventricles.
served myelin and patches of myelin loss and cavita- Apparently the calcifications are a characteristic, but
tion alternating in an apparently haphazard way, is not obligatory part of the disease. Over time the calci-
compatible with an underlying microangiopathy. A um deposits tend to increase in extent and number.
microangiopathy in the context of an inherited dis- On CT, variable white matter hypodensity and atro-
ease is not unique. The combination with microan- phy has been reported.
giopathy and leukoencephalopathy is also seen in Calcifications are less easily seen on MRI, but MRI
Labrune syndrome and CADASIL, although in the lat- more easily shows the abnormality of the white mat-
ter condition there are no calcium deposits. An open ter. With few exceptions, the cerebral and cerebellar
question is how an “interferonopathy” could be relat- white matter is diffusely abnormal (Figs. 66.2 and
ed to the calcifying microangiopathy seen in Aicar- 66.6). In some patients the signal abnormality is mild
di–Goutières syndrome and the white matter looks hypomyelinated rather
than “diseased.” In these patients, MRI demonstrates
the presence of myelin in some structures that nor-
66.4 Therapy mally myelinate early, suggesting arrest of myelina-
tion at an early stage. In other patients the white mat-
At present, there is no therapy apart from supportive ter signal abnormalities are much more pronounced,
care. Prenatal diagnosis is not possible as yet. suggesting that the white matter signal abnormality
cannot simply be explained by insufficient and arrest-
ed myelination, but that there must be additional
66.5 Magnetic Resonance Imaging white matter damage. So, MRI suggests that the white
matter abnormalities seen in Aicardi–Goutières syn-
CT is very important in the diagnosis of this disease, drome are the result of a variable combination of hy-
readily demonstrating the presence of calcium de- pomyelination, demyelination, gliosis, and white mat-
posits (Figs. 66.1, 66.3, 66.5, and 66.8). The calcium ter microinfarctions. In some patients, the cerebral
deposits are most often seen in the globus pallidus, white matter looks much better, with more myelin
putamen, caudate nucleus, thalamus, and dentate nu- and only some focal white matter abnormalities
clei. Additionally, calcium deposits may be seen in the (Fig. 66.4). Over time there is variable atrophy of the
cerebral white matter, in particular in the periventric- brain, in particular due to loss of cerebral white mat-
ular area alongside the ventricle wall, and in the cere- ter volume (compare Figs. 66.6 and 66.7). The atrophy
bellar white matter. The deposits are typically punc- becomes severe in some patients, whereas in other
tate, and may coalesce to form large concrements. patients there is no evidence of progression of the
However, in patients in whom the basal ganglia calci- atrophy. Some patients have subcortical cysts in the
fications have become diffuse, there are usually still anterior temporal area (Figs. 66.2 and 66.7). These
punctate calcifications elsewhere in the brain. In sev- cysts may become large. Occasionally exceptional pa-
eral affected sibs of typical cases of Aicardi–Goutières tients are seen who are the sibling of a more typically

Fig. 66.1. CT of a 3-year-old severely affected patient with deposits in the white matter have the typical punctate
Aicardi–Goutières syndrome.There are calcium deposits in the appearance, whereas the basal ganglia calcium deposits are
basal ganglia and periventricular white matter. The calcium more diffuse
66.5 Magnetic Resonance Imaging 499

Fig. 66.2. MRI of the same patient as shown in Fig. 66.1 at the cerebral white matter abnormalities.The corpus callosum and
same age. The T1-weighted images show cysts in the anterior internal capsule are spared
temporal region. The T2-weighted images show extensive

affected patient and in whom the cerebral white mat- Goutières syndrome. Congenital infections, in partic-
ter looks much closer to normal than is usual for ular congenital cytomegalovirus infection and toxo-
Aicardi–Goutières syndrome. plasmosis can lead to a similar pattern of calcium
The MRI pattern shows similarities to the leukoen- depositions and in particular congenital cytomegalo-
cephalopathy and calcium deposits seen in Cockayne virus infection may be associated with extensive
syndrome. However, in the latter disease the calcium white matter abnormalities and anterior temporal
deposits tend to be larger and more confluent, while subcortical cysts.
the punctate pattern is very typical of Aicardi–
500 Chapter 66 Aicardi–Goutières Syndrome

Fig. 66.3. CT scan of the affected brother of the patient punctate calcium deposits in the basal ganglia and cerebral
shown in Figs. 66.1 and 66.2. He is 7 years old. He has a much white matter
milder phenotype and can walk with support. Note the subtle

Fig. 66.4. MRI of the same patient as shown in Fig. 66.3 at the tricular white matter. Myelination is delayed, as most evident
same age. The cerebral white matter looks much better. There in the temporal lobe
are multifocal,partially confluent abnormalities in the periven-
66.5 Magnetic Resonance Imaging 501

Fig. 66.5. CT scan of a 4-month-old patient with Aicardi– um deposits in the periventricular and deep white matter,
Goutières syndrome, who is extremely severely affected. He basal ganglia, thalami, and dentate nucleus
has a sister with the same diagnosis. There are punctate calci-

Fig. 66.6. MRI of the same patient as shown in Fig. 66.5, also at
the age of 4 months.Note the atrophy of pons and cerebellum.
The anterior temporal white matter is swollen and highly
rarefied. The cerebral white matter is reduced in volume and
diffusely abnormal in signal intensity. There are small spots of
low signal intensity in the periventricular white matter on the
T2-weighted image, reflecting calcium deposits. The coronal
FLAIR images reveal subependymal cysts and highly rarefied
or cystic anterior temporal white matter.
(Fig. 66.6 see page 502)

Fig. 66.7. MRI of the same patient as shown in Figs. 66.5 and
66.6, now at the age of 16 months. Note the serious atrophy of
the cerebral white matter. There are anterior temporal cysts.
The subependymal cysts have disappeared
(Fig. 66.7 see page 503)
502 Chapter 66 Aicardi–Goutières Syndrome

Fig. 66.6.
66.5 Magnetic Resonance Imaging 503

Fig. 66.7.
504 Chapter 66 Aicardi–Goutières Syndrome

Fig. 66.8. CT of a 4-month-old patient with Aicardi–Goutières syndrome, demonstrating how extensive the calcium deposits
may be. Courtesy of P.G. Barth, Department of Child Neurology, Academic Medical Center, Amsterdam, The Netherlands
Chapter 67

Leukoencephalopathy with Calcifications and Cysts

67.1 Clinical Features iron deposits. The white matter shows myelin loss,
and Laboratory Investigations microcystic degeneration, and gliosis.

Leukoencephalopathy with calcifications and cysts


(LCC) is a very rare leukoencephalopathy. Affected 67.3 Pathogenetic Considerations
siblings have been reported, suggesting an autosomal
recessive mode of inheritance. Onset varies between The disease probably has an autosomal recessive
early infancy and adolescence. Some children display mode of inheritance. The basic defect is unknown. A
intrauterine growth retardation. The disease is slowly genome-wide linkage study has been started.
progressive with signs of spasticity, cerebellar ataxia, The pathophysiology of the disease has not been
extrapyramidal movement abnormalities, epilepsy, elucidated, but it is clearly a proliferative angiopathy
and mental deterioration. In addition, the patients involving the vessels of the central nervous system
may develop focal neurological deficits and signs of and the retina. The genesis of the cysts is unclear.
increased intracranial pressure related to growing in-
tracranial cysts which require neurosurgical treat-
ment. Optic atrophy with deterioration of vision and 67.4 Therapy
blindness may occur. Some of the children have bilat-
eral Coats retinopathy. Coats retinopathy is a congen- Growing cysts may require neurosurgical interven-
ital retinal telangiectatic disease, characterized by ab- tion to alleviate the elevated intracranial pressure.
normal retinal vascular permeability and telangiecta- Other than that, only supportive care is available. Pre-
sia, leading to a progressive exudative retinal detach- natal diagnosis is as yet not possible.
ment and blindness.
As with all newly described syndromes for which
the basic defect is not yet known, it is difficult to de- 67.5 Magnetic Resonance Imaging
fine the phenotype. Several patients have been re-
ported who lack some of the features described above CT is very important in the diagnosis of LCC, readily
and have additional features not mentioned above. A demonstrating the presence of calcium depositions
few patients had additionally dyskeratosis congenita- (Figs. 67.3 and 67.6). The calcium deposits are most
like symptoms (sparse hair, dysplastic nails, pigmen- often seen in the thalamus, basal ganglia, the deep
tation abnormalities of the skin) and Fanconi ane- white matter, the white matter–cortex border, cerebel-
mia-like features (thrombopenia and aplastic ane- lar white matter, dentate nucleus, brain stem, and lin-
mia). Other features reported include microcephaly, ing of the cysts. They are not necessarily symmetrical.
osteopenia, osteosclerosis with a tendency to frac- They are progressive over time. The calcium deposits
tures, and short stature. are different from those seen in Aicardi–Goutières
Laboratory investigations are unrevealing. Blood syndrome. In the latter disease the calcium deposits
calcium, phosphorus, and alkaline phosphatase are are small and punctate, and even in patients with
normal. Parathormone levels are normal. CSF is nor- more confluent calcifications, smaller punctate calci-
mal. fications can still be seen. In LCC the calcium deposits
are larger and bulky.
Calcifications are less easily seen on MRI. Gradi-
67.2 Pathology ent-echo images can be used for that purpose
(Fig. 67.1). MRI is, however, superior in depicting the
The most prominent finding in tissue obtained in abnormality of the white matter (Figs. 67.1, 67.2, 67.4,
neurosurgical interventions consists of angiomatous and 67.5). The cerebral white matter shows large areas
changes. Numerous small, tortuous blood vessels are of signal abnormality. The abnormal areas may have
found with normal endothelial lamina but an irregu- a mildly swollen aspect with broadening of gyri and
lar, often calcified wall. These vessels are surrounded stretching of the overlying cortex. The white matter
by many whirled and irregular Rosenthal fibers, abnormalities may be asymmetrical. They increase in
eosinophilic bodies, microcalcifications, and ferric extent over time.
506 Chapter 67 Leukoencephalopathy with Calcifications and Cysts

Fig. 67.1. A boy with LCC syndrome started to have increas- The cerebral white matter abnormalities are more serious on
ing walking difficulties at the age of 9–10 years. The first MRI the left; the cerebellar white matter abnormalities are more
was obtained at the age of 10 years. Note the calcium deposits serious on the right.The abnormal white matter has a swollen
in the basal ganglia, thalami, dentate nuclei, and lining of the aspect. We thank Dr. P.G. Barth, Department of Child Neu-
cysts in the deep frontal white matter on the left and the cere- rology, Academic Medical Center, Amsterdam, The Nether-
bellum on the right, indicated by a low signal intensity on lands, for referral of the patient
the gradient-echo (first row, left) and T2-weighted images.

In addition, growing cysts may be seen, most often white matter abnormalities and calcium depositions.
located in the region of the basal ganglia and third However, the cysts are unique. In Aicardi–Goutières
and fourth ventricle (Figs. 67.1–67.6). They may be- syndrome the calcium deposits are typically punc-
come large and lead to compression of brain tissue or tate. In Cockayne syndrome the calcium deposits
obstruct the normal CSF flow, leading to secondary tend to be larger and more confluent, but the white
hydrocephalus. Some of the cysts seem entirely intra- matter disease is symmetrical and there are no cysts.
ventricular, whereas other cysts are evidently primar- CNS infections, in particular congenital cytomegalo-
ily intraparenchymal. It is important to stress that the virus infection, may lead to calcium depositions and
cysts are not present in all patients, or not yet. Patients extensive white matter abnormalities, but the pattern
may be known for many years with calcium deposits of white matter abnormalities is typical and there are
and white matter abnormalities before they develop often anterior temporal subcortical cysts. In disor-
cysts. ders involving parathormone, similar calcium de-
The imaging pattern showing a combination of posits may be seen but the white matter abnormali-
leukoencephalopathy and calcium deposits is remi- ties and cysts are lacking.
niscent of that seen in other disorders leading to
67.5 Magnetic Resonance Imaging 507

Fig. 67.2. The same patient 1 year later. With surgical intervention, the cysts have become smaller

Fig. 67.3. The same patient, now 13 years old.There is a grow- nuclei. There are multiple calcium deposits in the cortico-sub-
ing cyst in the region of the thalamus on the right.The CT scan cortical junction in the cerebral hemispheres
shows the calcium in the basal ganglia, thalami, and dentate

Fig. 67.4. The same boy, now 19 years old.There are large cysts in the region of the thalamus on both sides
508 Chapter 67 Leukoencephalopathy with Calcifications and Cysts

Fig. 67.5.
67.5 Magnetic Resonance Imaging 509

Fig. 67.6. CT scan at the age of 22 reveals the increase in calcium as compared to the first CT (Fig. 67.3)

Fig. 67.5. The same boy, now 22 years of age. The cysts in the
region of the thalami have regressed without surgical inter-
vention. The cysts in the left hemisphere have become very
large and have led to a marked midline shift. The T1-weighted
images without (third row) and with contrast (fourth row) re-
veal enhancement in the areas of calcium deposits: the lining
of cysts and the basal ganglia and thalami.
Chapter 68

Leukoencephalopathy with Brain Stem and Spinal Cord


Involvement and Elevated White Matter Lactate

68.1 Clinical Features 68.4 Therapy


and Laboratory Investigations
No specific treatment is available. Supportive care is
Leukoencephalopathy with brain stem and spinal important.
cord involvement and elevated white matter lactate
(LBSL) is a rare disorder with an autosomal recessive
mode of inheritance. The initial development is nor- 68.5 Magnetic Resonance Imaging
mal. In some of the patients, independent walking is
unstable from the beginning. Motor deterioration LBSL has a distinct MRI pattern. The cerebral white
starts at a variable age in childhood or adolescence matter is involved to a variable extent. In some pa-
with signs of spasticity and ataxia involving the legs tients the abnormalities are extensive (Figs. 68.1 and
more than the arms. The disease is slowly progressive. 68.2), in others they are more limited (Figs. 68.6 and
Patients may become wheelchair-dependent in their 68.9). The white matter abnormalities are progressive
teens or twenties, but some still walk in their forties. over time, with new structures being involved and
Manual dexterity also becomes decreased to a vari- some white matter atrophy (Fig. 68.2). In all patients,
able degree. At neurological examination, most pa- even in the oldest, the U fibers are spared (Fig. 68.1).
tients have a distal decrease in position and vibration In some patients, the cerebral white matter abnormal-
sense. Some patients develop epilepsy with infrequent ities are homogeneous (Fig. 68.10), but in most pa-
seizures. Some of the patients have learning problems tients the abnormal cerebral white matter has an in-
from early on, but cognitive decline is of late occur- homogeneous, spotty aspect (Figs. 68.1, 68.6, and
rence. Some patients have more serious disease than 68.9). On FLAIR images the white matter abnormali-
others.A striking feature is that some patients experi- ties are also inhomogeneous with spots of lower
ence an episode of more rapid and partially reversible signal intensity suggestive of focal rarefaction
neurological deterioration accompanied by fever fol- (Fig. 68.5). In the mildest form, there may be a few
lowing minor head trauma. focal lesions in the cerebral white matter on a back-
Somatosensory evoked potentials with stimulation ground of limited white matter abnormalities, both in
of the tibial and median nerves are delayed or nega- extent and degree of signal change (Figs. 68.6 and
tive. Sensory and motor nerve conduction velocities 68.9). The corpus callosum is involved, almost always
are normal.
Extensive laboratory investigations have been un-
revealing. Only in some of the patients have mild ele- 䊳

vations of serum or CSF lactate been found on sever- Fig. 68.1. A 34-year-old lady with LBSL, presently wheelchair-
al occasions. However, extensive mitochondrial work- bound. The T2-weighted images (first, second, and third rows)
up in fresh muscle tissue has not revealed abnormal- show extensive inhomogeneous signal abnormalities in the
ities in any of the patients. No mitochondrial DNA cerebral white matter with sparing of the U fibers.The posteri-
mutations have been found. or part of the corpus callosum is affected, whereas the anteri-
Presently, the diagnosis is MRI-based. Prenatal di- or part is spared. Signal changes are also seen in the posterior
agnosis is unfortunately not possible. limb of the internal capsule.Within the midbrain, signal abnor-
malities are seen in the pyramidal tracts. At the level of the
pons, the entire basis, the medial lemniscus, mesencephalic
68.2 Pathology trigeminal tracts, intraparenchymal parts of the trigeminal
nerves, and superior cerebellar peduncles are affected bilater-
No pathology information is available. ally. At the level of the medulla, the inferior cerebellar pedun-
cles, the anterior spinocerebellar tracts, the decussation of the
medial lemniscus and the pyramids are involved. The cerebel-
68.3 Pathogenetic Considerations lar white matter is diffusely affected. The T1-weighted images
(fourth row) show the inhomogeneities in signal intensity of
The disease has an autosomal recessive mode of in- the abnormal white matter with foci of more prominent signal
heritance, but the genetic defect is unknown. change
68.5 Magnetic Resonance Imaging 511

Fig. 68.1.
512 Chapter 68 Leukoencephalopathy with Brain Stem and Spinal Cord Involvement and Elevated White Matter Lactate

Fig. 68.2.
68.5 Magnetic Resonance Imaging 513

Fig. 68.3. Diffusion-weighted images of the same patient as and third rows) show that some areas, in particular rims of
shown in Fig. 68.2 at the age of 19 years. The Trace diffusion- lesions, have a low ADC, whereas the remainder of the white
weighted images (first row, b value = 1000) show a high signal matter has a high ADC
intensity of the abnormal white matter.The ADC maps (second

Fig. 68.2. These T2-weighted images of a female patient with al lemniscus. At the level of the pons, bilateral signal changes
LBSL were obtained at the ages of 10 years (first and second were seen in the medial lemniscus, mesencephalic trigeminal
rows) and almost 20 years (third and fourth rows). The typical tract, intraparenchymal part of the trigeminal nerve, and the
abnormalities are present. Over time the supratentorial white superior cerebellar peduncle, whereas on follow-up pyramidal
matter becomes more atrophic and the abnormalities become tract abnormalities are seen as well. The cerebellar white mat-
less homogeneous. At the level of the midbrain signal abnor- ter abnormalities were initially subcortical and become diffuse
malities become visible in the pyramidal tracts and the medi-
514 Chapter 68 Leukoencephalopathy with Brain Stem and Spinal Cord Involvement and Elevated White Matter Lactate

Fig. 68.4. Spinal images obtained in the patient


shown in Figs. 68.2 and 68.3. The patient under-
went multiple MRI studies in 10 years and the
spinal imaging findings were always the same, with
signal abnormalities in the dorsal columns and lat-
eral corticospinal tracts over the entire length of
the spinal cord

Fig. 68.5. FLAIR images of the patient shown in Figs. 68.2–68.4. These images show that the abnormal white matter is
inhomogeneous with areas of lower signal, suggesting focal white matter rarefaction

more seriously in the posterior part than the anterior Within the brain stem and spinal cord, the disease in-
part (Figs. 68.1, 68.2, and 68.6), but sometimes the volves certain tracts selectively. The pyramidal tracts
corpus callosum is homogeneously affected through- are affected over their entire length extending down-
out. The posterior limb of the internal capsule is also wards through the posterior limb of the internal cap-
affected (Figs. 68.1, 68.2, 68.5, 68.6, 68.9, and 68.10). sule and the brain stem into the lateral corticospinal
68.5 Magnetic Resonance Imaging 515

Fig. 68.6. Male LBSL patient at the age of 17 years.He has min- um of the corpus callosum and posterior limb of the internal
imal neurological problems, mainly consisting of minor spas- capsule are abnormal. At the level of the pons signal abnor-
ticity of the legs and some bladder dysfunction.The T2-weight- malities are seen in the intraparenchymal part of the left
ed images show diffuse mild signal changes in the periventric- trigeminal nerve.There is a small lesion in the superior cerebel-
ular and deep cerebral white matter. There are superimposed lar peduncle on the left. At the level of the medulla lesions are
focal lesions with more prominent signal change. The spleni- seen in the pyramids

tracts of the spinal cord (Figs. 68.1, 68.2, 68.4, radiata above the level of the thalamus (Figs. 68.1,
68.8–68.10). Sensory tracts are also affected over their 68.2, 68.9, and 68.10). The transverse pontine fibers
entire length, involving the dorsal columns in the become involved in later stages of the disease
spinal cord, the medial lemniscus through the brain (Fig. 68.1). Cerebellar connections are selectively in-
stem up to the level of the thalamus, and the corona volved, first the superior and inferior cerebellar pe-
516 Chapter 68 Leukoencephalopathy with Brain Stem and Spinal Cord Involvement and Elevated White Matter Lactate

Fig. 68.7. Diffusion-weighted images in the same patient sity. On the ADC map (middle and right), the focal lesions have
as in Fig. 68.6.The Trace diffusion-weighted image (left, b value a low ADC, whereas the areas of subtle signal change on the
= 1000) shows that the focal lesions have a high signal inten- conventional images have high ADC values

Fig. 68.8. Spinal images obtained in the patient


shown in Figs. 68.6 and 68.7. There are signal abnor-
malities in the dorsal columns and lateral corti-
cospinal tracts over the entire length of the spinal
cord

duncles (Figs. 68.1, 68.2, 68.9, and 68.10), and only at a parenchymal trajectories of the trigeminal nerve and
late stage the middle cerebellar peduncles (Fig. 68.1). the mesencephalic trigeminal tracts (Figs. 68.1, 68.2,
The anterior spinocerebellar tracts at the level of the 68.9, and 68.10). The cerebellar white matter may also
medulla also become abnormal (Fig. 68.1). A remark- develop signal abnormalities, first in the subcortical
able finding is the consistent involvement of the intra- regions (Fig. 68.2) and subsequently spreading in-
68.5 Magnetic Resonance Imaging 517

Fig. 68.9. A female 44-year-old LBSL patient with minimal enchymal trajectory of the trigeminal nerve. At the level of
neurological problems. In the periventricular and deep cere- the medulla, abnormalities are seen in the inferior cerebellar
bral white matter subtle signal abnormalities are seen with peduncles and pyramids.The lowest section is through the up-
small foci of more prominent signal change. The posterior per part of the cervical spinal cord and shows the involvement
limb of the internal capsule is affected. At the level of the of the posterior columns and the lateral corticospinal tracts.
pons, bilateral abnormalities are seen in the pyramidal tracts, Courtesy of Dr. M. Heitbrink and Dr. B. Wiarda, Department of
medial lemniscus, superior cerebellar peduncles, and intrapar- Radiology, Medical Center Alkmaar, The Netherlands

wards to involve all cerebellar white matter through- and gliosis, respectively. White matter choline is
out (Figs. 68.1 and 68.2). The cerebellum becomes at- increased, too, but the increase is minor, suggesting
rophic over time. mildly enhanced membrane turnover, possibly
The white matter abnormalities are progressive myelin loss. White matter lactate is elevated in almost
over time. Cerebral white matter abnormalities may all patients, but to a variable degree. Cortex spectra
be present without any abnormality in the brain stem are normal. A minor increase in lactate in the cortex
or cerebellum, but at that time the spinal cord already voxel is probably explained by admixture of both
contains signal abnormalities in the lateral and dorsal white matter and CSF in the voxel, lactate levels of up
tracts. Without spinal imaging, an MRI-based diag- to 1.2 mmol/l being normal in the CSF.
nosis is not possible in the early or mildest stages of Diffusion-weighted imaging in LBSL patients
the disease. Over time, the brain stem abnormalities consistently shows high signal in the affected white
develop, and finally so do cerebellar white matter ab- matter on the Trace diffusion-weighted images (b val-
normalities. ue = 1000) (Figs. 68.3 and 68.7), most prominently in
A similar MRI pattern has not been described in the focal lesions, with more severe signal change on
any other condition; if present, it is diagnostic. the conventional images (Fig. 68.7). The ADC maps
MRS shows a significant decrease in N-acetylas- consistently show a complicated pattern with a mix-
partate and increase in myo-inositol in the abnormal ture of high and low ADC values in areas with high
white matter, suggesting axonal damage or loss signal on the Trace diffusion-weighted images. The
518 Chapter 68 Leukoencephalopathy with Brain Stem and Spinal Cord Involvement and Elevated White Matter Lactate

Fig. 68.10. A 10-year-old boy with LBSL. In this patient the cerebral white matter abnormalities are homogeneous. Courtesy of
Dr. S. Blaser, Department of Diagnostic Imaging, Hospital for Sick Children, Toronto

low ADCs are mainly seen at the border of the white this is at present unknown, but it is clear that concepts
matter abnormalities (cerebral U fibers, peripheral such as vasogenic edema, cytotoxic edema, and T2
rim of the cerebellar white matter abnormalities, the shine-through effects are insufficient to explain these
small focal lesions). The remainder of the abnormal observations.
white matter has elevated ADC values. The reason for
Chapter 69

Hypomyelination with Atrophy


of the Basal Ganglia and Cerebellum

69.1 Clinical Features sponses show a normal latency for waves I and II,
and Laboratory Investigations whereas the later waves are delayed or not recordable.
Motor and sensory nerve conduction velocities are
A distinct leukoencephalopathy has been described normal. Sural nerve biopsy is unrevealing.
in a few patients, characterized by hypomyelination
and atrophy of the basal ganglia and cerebellum
(HABC), giving the disease its name. Both males and 69.2 Pathology
females are affected. It is highly likely that the disease
is inherited. However, since all patients so far have No autopsy studies are available.
been isolated cases, it is unclear whether the mode of
inheritance is autosomal recessive, or whether it is
autosomal dominant with all cases representing de 69.3 Pathogenetic Considerations
novo mutations.
The disease has its onset in infancy or early child- The disease is most likely inherited, but the mode of
hood. The severity of the disease is highly variable. inheritance is at present unclear. The disease is char-
The most severely affected patients present soon after acterized by a disturbance of normal development
birth with poor eye contact and absence of any motor and degeneration. From the beginning there is a pic-
development. Ophthalmological examination reveals ture of myelin deficiency. In some patients MRI sug-
pale optic discs, consistent with hypomyelination of gests that there is some additional loss of myelin.
the optic nerves. Over the years there are signs of There is progressive atrophy of the putamen, caudate
slowly progressive spasticity and extrapyramidal nucleus, and cerebellum. The cerebral white matter
movement abnormalities including rigidity, dystonia, also shows loss of volume over time. The putamen
and choreoathetosis. The patients seem to have a bet- and caudate nucleus disappear without evidence of
ter mental function than motor function and appear remaining scar tissue on MRI, suggesting atrophy by
to have a social awareness. They may have some means of apoptosis rather than necrosis. The atrophy
seizures. The severely affected patients tend to have a is more severe in the clinically more severely affected
small stature and have a head circumference below patients.
the third percentile. Patients with intermediate sever-
ity of disease have delayed early development, but
achieve grasping and unsupported sitting. In the 69.4 Therapy
mildest cases the patients may have normal initial de-
velopment and they achieve unsupported walking. In At present, no specific therapy exists for patients with
these patients slow deterioration becomes evident in HABC. Seizures may require initiation of antiepilep-
early childhood with increasing spasticity, ataxia, and tic treatment. The severe extrapyramidal symptoms
often prominent extrapyramidal movement abnor- may require use of drugs also used for parkinsonism,
malities consisting of dystonia, choreoathetosis, and including L-dopa. However, the extrapyramidal
rigidity. Some patients are predominantly spastic. movement abnormalities tend to be drug-resistant.
The patients typically have learning problems, but Some patients require intrathecal baclofen to reduce
further cognitive decline is at most minor. Occasion- the serious hypertonia.
al epileptic seizures may occur. Vision is normal and
ophthalmological examination reveals no abnormali-
ties. Height and head circumference are normal. 69.5 Magnetic Resonance Imaging
Laboratory examinations, including extensive
metabolic studies, are unrevealing. CSF neurotrans- The diagnosis in HABC is MRI-based. Early MRI is
mitters and neurotransmitter metabolites have been characterized by the presence of very little myelin. In
studied in several patients and found to be within the some patients, the putamen is already absent within
normal range. Electroretinogram is normal. Visual the first year of life and the caudate nucleus is small,
evoked responses and somatosensory evoked re- making the diagnosis of HABC possible (Fig. 69.1).
sponses are delayed. Brain stem auditory evoked re- However, in other patients the MRI within the first
520 Chapter 69 Hypomyelination with Atrophy of the Basal Ganglia and Cerebellum

Fig. 69.1. Girl with HABC and a severe


phenotype. At the age of 11 months
(first row) the signal intensity of all
cerebral white matter is high on the
T2-weighted image (right), consistent
with hypomyelination.The same
image shows that the thalamus is of
normal size and the globus pallidus is
visible, whereas there is no putamen.
The caudate nucleus is very small and
lacks the normal intermediate signal
intensity.The follow-up images at
the age of 10 years (second row) show
the atrophy of the cerebellum.The
T2-weighted image (right) shows that
the white matter still has a high signal
and has become seriously atrophic.
The thalamus and globus pallidus are
of normal size but the putamen and
caudate nucleus are not visible.
From Van der Knaap et al. (2002),
with permission

Fig. 69.2. Boy with HABC and a milder


phenotype at the age of 18 months
(first row) and 6 years (second row).The
sagittal images (left) show progressive
cerebellar atrophy over time.The axial
T2-weighted images (right) show per-
sistent hypomyelination of the cere-
bral white matter. Initially, the basal
ganglia have a normal appearance,
but on follow-up the putamen has
disappeared. Note the normal
thalamus and globus pallidus.
From Van der Knaap et al. (2002),
with permission
69.5 Magnetic Resonance Imaging 521

year of life shows nothing other than myelin deficien- other patients the white matter has a moderately high
cy; the putamen and caudate nucleus are still present signal intensity on both T2- and T1-weighted images
(Fig. 69.2). The severity of the myelin deficit is vari- (Fig. 69.4), indicative of diffuse deposition of some
able. In some patients the cerebral hemispheric white myelin (moderate hypomyelination). It is strik-
matter has a high signal intensity on T2-weighted im- ing that the pyramidal tracts in the brain stem are
ages and a low signal intensity on T1-weighted images also hypomyelinated. Over time, the putamen disap-
(Fig. 69.3), indicative of serious hypomyelination. In pears (Figs. 69.1–69.5). The caudate nucleus becomes

Fig. 69.3. A 9-year-old boy with HABC. The sagittal T1-weight- fifth rows), consistent with a serious lack of myelin. On the
ed image (first row, left) shows the prominent cerebellar atro- T2-weighted images the thalamus and globus pallidus are nor-
phy. The cerebral white matter has a high signal intensity on mal, whereas there is no putamen and the caudate nucleus is
the axial T2-weighted images (first, second, and third rows) and small
a low signal intensity on T1-weighted images (fourth and
522 Chapter 69 Hypomyelination with Atrophy of the Basal Ganglia and Cerebellum

Fig. 69.3. (continued).

smaller, disappearing in some patients (Figs. 69.1 and The full-blown picture of hypomyelination and
69.5). The thalamus and globus pallidus remain of missing putamen is diagnostic of HABC and does
normal size (Figs. 69.1–69.5). The cerebellum be- not suggest any other disorder. However, for as long
comes progressively atrophic (Figs. 69.1 and 69.3). as the putamen is still visible, other disorders leading
The atrophy affects the vermis more seriously than to hypomyelination should be considered, includ-
the hemispheres. Over the years, variable atrophy of ing Pelizaeus–Merzbacher disease, Salla disease, and
the cerebral white matter occurs, associated with DNA repair disorders.
variable dilatation of the lateral ventricles. In some of Proton MRS reveals that within the white matter
the patients, there appears to be some additional loss total N-acetylaspartate and choline are normal, argu-
of the little myelin present (Figs. 69.4 and 69.5). The ing against significant neuronal/axonal loss and ac-
atrophy of cerebral white matter and the basal ganglia tive demyelination. Myo-inositol and total creatine are
is more serious in the clinically severely affected pa- elevated, suggesting significant white matter gliosis.
tients.
69.5 Magnetic Resonance Imaging 523

Fig. 69.4.
524 Chapter 69 Hypomyelination with Atrophy of the Basal Ganglia and Cerebellum

Fig. 69.4. (continued). A 6-year-old girl with HABC. The cere- bellar white matter are also insufficiently myelinated. The
bral white matter has a high signal intensity on the axial (first T2-weighted images show a normal thalamus and globus pal-
and second rows) and coronal (third row) T2-weighted images lidus, whereas there is no putamen and the caudate nucleus is
and an intermediate to high signal intensity on T1-weighted very small. Courtesy of Dr. S. Blaser, Department of Diagnostic
images (fourth and fifth rows), consistent with moderate hy- Imaging, Hospital for Sick Children, Toronto, Canada
pomyelination.The middle cerebellar peduncles and the cere-
69.5 Magnetic Resonance Imaging 525

Fig. 69.5. The same girl as in Fig. 69.4, now 8 years old. She is nucleus is no longer visible. Courtesy of Dr. S. Blaser, Depart-
clinically deteriorating. The T1-weighted images (first and sec- ment of Diagnostic Imaging, Hospital for Sick Children, Toron-
ond rows) show white matter atrophy and loss of myelin. The to, Canada
coronal T2-weighted images (third row) show that the caudate
Chapter 70

Hereditary Diffuse Leukoencephalopathy


with Neuroaxonal Spheroids

70.1 Clinical Features the affected white matter. Neuroaxonal spheroids are
and Laboratory Investigations round to sausage-shaped axonal swellings, which are
easily identified with Bielschowsky, Bodian, and anti-
Hereditary diffuse leukoencephalopathy with spher- neurofilament immunostains. The leukoencephalo-
oids (HDLS) is an autosomal dominant progressive pathy is most severe in the frontal, frontoparietal, and
disease. The disease was described for the first time in temporal areas and may be mildly asymmetrical. The
multiple members of a large Swedish pedigree in 1984 U fibers are relatively spared. The abnormalities tend
(Axelsson et al. 1984). In this family, 17 of 71 subjects to be most pronounced in the white matter below the
from 4 generations were affected. The age at onset pre- and postcentral gyri and extend through the pos-
varied from 8 to 60 years, with a mean of 36 years. terior limb of the internal capsule into the pyramidal
The age at death was 39 to 89 years, with a mean of tracts of the brain stem. The corpus callosum is vari-
57 years. The time between onset and death varied ably affected. The abnormal white matter may show
from 3 months to over 30 years. Some patients rapid- vacuolization. Reactive astrocytes and macrophages
ly developed severe dementia and died within a are present, but no inflammatory cells. The cerebral
few months of the onset of symptoms, whereas in oth- cortex and basal ganglia are normal and contain no or
ers the course was prolonged with dementia develop- only a few spheroids.Within the cerebellum, a marked
ing over decades. Sporadic cases have also been re- loss of Purkinje cells is seen, but the cerebellar white
ported. matter is normal. Electron microscopy of the spher-
The predominant clinical manifestations are oids reveals neurofilaments scattered among elec-
psychiatric and include depression, anxiety, alcohol tron-dense material and mitochondria.
abuse, irritability, and aggressiveness. Psychotic
symptoms may occur with confusion, delusions,
and hallucinations. The most frequent neurological 70.3 Pathogenetic Considerations
symptoms are dementia, seizures, impaired balance,
retropulsion, gait apraxia, spasticity, ataxia, and uri- The homogeneity of the clinical picture and histo-
nary incontinence. Extrapyramidal symptoms may pathological findings strongly suggests that HDLS is
occur with hyperkinesia, chorea, tremor, and oral a distinct disease entity. The disease has an autosomal
dyskinesia. dominant mode of inheritance. Isolated cases are
EEG usually shows nonspecific abnormalities with probably the result of new mutations. The genetic de-
slowing of the background pattern and sometimes fect and the pathophysiology of HDSL are as yet un-
paroxysmal changes. The abnormalities often have a resolved. Considering the more or less commensurate
frontotemporal predominance. They may be asym- loss of axons and myelin sheaths, the preferential in-
metrical. Routine and metabolic laboratory investiga- volvement of long tracts, and the presence of axonal
tions reveal no abnormalities. The diagnosis is at pre- swellings, it is likely that axons are the primary target
sent based on histopathological findings. of the disease.
Axonal spheroids are pathological findings char-
acteristic of the neuroaxonal dystrophies. They occur
70.2 Pathology most often in the context of neuronal degenerative
disorders, such as infantile neuroaxonal dystrophy
External examination of the brain shows mild atro- (Seitelberger disease) and Hallervorden–Spatz dis-
phy of the frontoparietal regions. The thalamus and ease. The combination of leukoencephalopathy and
the rostral part of the caudate nucleus may be mildly neuroaxonal spheroids in the abnormal white matter
reduced in size. The lateral ventricles are moderately is rare. Apart from HDSL, this combination is ob-
enlarged. The corticospinal tracts and the basis of served in dermatoleukodystrophy with neuroaxonal
the pons are atrophic. On microscopy, a widespread spheroids (Matsuyama et al. 1978) and polycystic
leukoencephalopathy is found, characterized by a lipomembranous osteodysplasia with sclerosing
commensurate loss of myelin sheaths and axons and leukoencephalopathy (Nasu–Hakola disease). Both
the presence of numerous neuroaxonal spheroids in disorders are clinically different from HDLS.
70.5 Magnetic Resonance Imaging 527

70.4 Therapy the caudate nucleus may be flattened. There may be


cerebellar atrophy.
Supportive care is the only therapeutic option. The above MRI findings may confirm the diagno-
sis within a pedigree with known HDLS. However, the
MRI findings in themselves are not specific and do
70.5 Magnetic Resonance Imaging not allow a definite diagnosis. The diagnosis needs to
be confirmed by histopathology.
MR images demonstrate signal abnormalities bilater- The differential diagnosis of HDLS includes disor-
ally within the cerebral white matter, most pro- ders with frontal cortical degeneration, such as fron-
nounced either within the white matter under the totemporal dementia and Pick disease. In these disor-
pre- and postcentral gyri (Figs. 70.1–70.3) or within ders MRI shows atrophy mainly of the frontotempo-
the frontal white matter. The signal abnormalities ral areas. Sometimes there are additional white mat-
may be patchy or more confluent, and may be sym- ter changes, which are ill-defined and usually mild. If
metrical or asymmetrical. They are ill-defined. The present, they make differentiation from HDLS diffi-
corpus callosum is thin and may contain areas of ab- cult.
normal signal. The signal abnormalities extend The differential diagnosis also includes disorders
downwards through the posterior limb of the internal with frontal lobe dysfunction caused by white matter
capsule into the pyramidal tracts of the brain stem degeneration, such as metachromatic leukodystro-
(Fig. 70.1). The affected cerebral white matter is phy, X-linked adrenoleukodystrophy with frontal pre-
atrophic with widening of the lateral ventricles and dominance, Nasu–Hakola disease, Binswanger dis-
subarachnoid spaces (Figs. 70.1–70.3). The head of ease, CADASIL, orthochromatic pigmentary leukody-

Fig. 70.1. A 26-year-old female patient with HDSL. This extend downwards into the posterior limb of the internal cap-
diagnosis was confirmed histopathologically in her father sule and the pyramidal tracts in the midbrain. There is a mild
(Fig. 70.2). Note the patchy and asymmetrical abnormalities in cerebral atrophy.From van der Knaap et al.(2000),with permis-
the white matter under the central sulcus. The abnormalities sion
528 Chapter 70 Hereditary Diffuse Leukoencephalopathy with Neuroaxonal Spheroids

Fig. 70.2. A 55-year-old man with HDSL, father of the patient They are partially confluent,partially multifocal.The abnormal-
in Fig. 70.1. The diagnosis was confirmed at autopsy. There is ities extend downwards into the posterior limb of the internal
serious cerebral atrophy. The white matter abnormalities are capsule. From van der Knaap et al. (2000), with permission
most prominent in the tracts under the pericentral cortex.

strophy, and adult-onset autosomal dominant leuko-


dystrophies. Most disorders can be ruled out by typi-
cal clinical, physical, and laboratory findings and
neuroimaging differences. Some disorders require
histopathological confirmation.
70.5 Magnetic Resonance Imaging 529

Fig. 70.3. A 38-year-old woman with HDLS.The diagnosis was white matter, most prominently in the central tracts. From van
confirmed at autopsy. There is prominent cerebral atrophy. der Knaap et al. (2000), with permission
There are patchy, multifocal abnormalities in the cerebral
Chapter 71

Dentatorubropallidoluysian Atrophy

71.1 Clinical Features tosis in the globus pallidus, subthalamic nucleus, red
and Laboratory Investigations nucleus, and dentate nucleus. Mild changes occur in
the caudate nucleus, putamen, thalamus, and inferior
Dentatorubropallidoluysian atrophy (DRPLA) is a olives. There is only subtle involvement of the sub-
rare disease with an autosomal dominant mode of stantia nigra. The superior cerebellar peduncles, con-
inheritance. The disease has a pronounced ethnic taining the outflow tracts of the dentate nuclei, are at-
predilection and occurs most frequently in Japan. rophic. The base of the pons and the tegmentum of
However, an increasing number of American and Eu- the midbrain and pons display variable atrophy.
ropean patients have been described. Although spo- Unlike findings in other dementias, there is no signif-
radic cases have been reported, nearly all cases are fa- icant cell loss in the nucleus basalis of Meynert.
milial with two or more successive generations affect- Immunohistochemistry demonstrates the presence
ed. There is evidence of anticipation: in the next gen- of ubiquin-positive intranuclear aggregates in neu-
eration the disease tends to have an earlier onset and rons and also astroglia. In addition, filamentous
be more severe. ubiquinated intracytoplasmic inclusions may be
The presenting symptoms are a variable mixture of found in neurons of affected structures.
cerebellar ataxia, myoclonus, epilepsy, choreoatheto- In younger patients patchy periventricular white
sis, dementia, and psychiatric abnormalities. The age matter abnormalities are found, whereas in older pa-
at onset ranges from the first to the sixth decade, with tients the white matter abnormalities are more con-
an average age of 32 years. In the early-onset forms fluent and symmetrical, and affect the deep white
progressive myoclonic epilepsy and dementia are the matter of the centrum semiovale. Microscopy reveals
predominant symptoms, whereas in the patients with gliosis and demyelination in the affected areas. Occa-
later onset progressive cerebellar ataxia and choreo- sionally the cerebellar white matter is involved. In-
athetosis are more prominent. Among the older pa- tranuclear aggregates have been demonstrated in
tients two clinical presentations can be distinguished: oligodendrocytes within the affected white matter.
a pseudo-Huntington form with prominent choreoa- Spinal cord involvement has been reported, with
thetosis, cognitive dysfunction, and psychiatric syn- degeneration of the posterior columns, spinocerebel-
dromes, and a form with progressive ataxia with less lar tracts, corticospinal tracts, and anterior horn cells.
severe cognitive impairment. In the Haw River variant of the disease, marked
In the younger group of patients, DRPLA has to be neuronal loss of the dentate nuclei, microcalcification
distinguished from other diseases with progressive of the globus pallidus, neuroaxonal dystrophy of the
myoclonic epilepsy, such as neuronal ceroid lipofusci- nucleus gracilis, and demyelination of the centrum
nosis, sialidosis, myoclonus epilepsy with red ragged semiovale have been found.
fibers (MERRF), and Unverricht disease. In the older
patients DRPLA has to be distinguished from Hunt-
ington disease, which may have a very similar clinical 71.3 Pathogenetic Considerations
presentation. Laboratory investigations are unreveal-
ing in DRPLA, but DNA confirmation is possible. DRPLA is caused by an unstable expansion of a cyto-
sine-adenine-guanine (CAG) repeat in the gene
DRPLA, located on chromosome 12p13.31. The prod-
71.2 Pathology uct of DRPLA is atrophin-1, a protein that is widely
expressed in many tissues including the brain. It
There are some variations in the histopathological shares no homology with other known proteins. Ex-
findings between different families. Most data on pansion of the CAG repeat in the DRPLA gene in nor-
histopathology in DPRLA come from Japanese pa- mal subjects ranges from 10 to 21, in patients from
tients, in whom histopathological findings are the 58 to 82. No overlap has been reported between nor-
most uniform. On gross inspection there may be mal alleles and mutant alleles.
some atrophy of the base of the pons and the cerebel- In the brain atrophin-1 is located in the cytoplasm
lar vermis. The principal histopathological alter- of neurons. In DPRLA subjects both expanded and
ations consist of severe neuronal cell loss and astrocy- normal alleles are detectable in the brain, with re-
71.5 Magnetic Resonance Imaging 531

gional variation. Atrophin-1 has been immunocyto- eases, but recently histopathological studies have
chemically localized to somatic and somatodendritic shown nuclear inclusions in oligodendrocytes of
compartments of cortical and cerebellar neurons. In DRPLA patients as well. In transgenic DRPLA mice,
the cerebellum the protein is especially abundant in too, oligodendrocytes demonstrated the same inclu-
Purkinje cells and neurons of the dentate nucleus. sions. Apparently not only neurons but also glial cells
The unstable CAG repeat encodes for a polygluta- are affected in DRPLA. It is probable that oligoden-
mine repeat. Polyglutamine repeats also occur in my- droglial involvement is responsible for the white
otonic dystrophy, fragile X syndromes, spinobulbar matter abnormalities that may be observed in DRPLA
muscular atrophy (Kennedy disease), Huntington dis- patients.
ease, spinocerebellar ataxia type 1 (SCA1), spinocere-
bellar ataxia type 2 (SCA2), Machado–Joseph disease
(SCA3) and spinocerebellar ataxia type 7 (SCA7). The 71.4 Therapy
number of repeats clearly influences the phenotypic
expression. As in other trinucleotide repeat syn- No causal treatment is available, but symptomatic care
dromes, the number of repeats increases with succes- is important. Anticonvulsants should be used when
sive generations. This leads to an earlier onset and seizures are present. Psychiatric problems can be
worsening of the phenotype in every following gener- treated with appropriate psychotropic medications.
ation, a phenomenon referred to as “anticipation.” In
the patients presenting with progressive myoclonic
epilepsy end dementia (early presentation) the num- 71.5 Magnetic Resonance Imaging
ber of repeats is much higher than in the patients
without the progressive myoclonic epilepsy (late pre- Standard sequences usually suffice to make an inven-
sentation). tory of the abnormalities in DPRLA. Sophisticated
The molecular basis for the selective neuronal techniques as diffusion tensor imaging may be useful
damage in dentatorubropallidoluysian atrophy is not to identify involved tracts, as may be fiber tracking.
yet fully understood. Misfolding and conformational Gradient echo techniques may be useful in the detec-
alterations of the mutant protein due to polygluta- tion of calcifications.
mine expansion may lead to aberrant protein–protein The characteristic findings on conventional MR
interactions and targeting of the misfolded proteins include atrophy of the tegmentum of the midbrain,
for degradation. Ubiquination of proteins in the ag- pons, dentate nucleus, superior cerebellar peduncles,
gregates indicates targeting for degradation. Protein and cerebellum (Figs. 71.1 and 71.2). The lateral
aggregation is probably a critical molecular compo- ventricles show moderate to severe enlargement and
nent of polyglutamine diseases. These aggregates are commonly there is moderate but clear cortical atro-
present as nuclear inclusions in the neurons selective- phy. The globus pallidus and subthalamic nucleus are
ly affected by the disease. Transglutaminases appear usually either normal or slightly atrophic on MRI.
to play a role in the formation of the protein aggre- High signal intensity of the globus pallidus on pro-
gates. Transglutaminases are a large family of pro- ton-density and T2-weighted images is often present
teins with the common capacity to catalyze cross- from early on (Figs. 71.1–71.3). Bilateral symmetric
linking of protein substrates, resulting in the forma- signal changes in the thalamus and abnormal signal
tion of large protein aggregates that are insoluble to intensity in the midbrain and pons are characteristic
all known protein detergents. These aggregates may (Figs. 71.1–71.3). The red nucleus characteristically
contribute to neuronal dysfunction and degenera- has normal signal (Fig. 71.2). In older patients there is
tion. It may also be that mutant proteins are most tox- an increase of lesions in the midbrain and thalamus.
ic when roaming freely and that the aggregates repre- Although not invariable, white matter abnormali-
sent the cell’s effort to convert the toxic proteins into ties are characteristically present in patients with
more innocuous clumps. The differences in clinical DRPLA. In younger patients patchy periventricular
expression and cell specificity of the affected neurons areas with high signal intensity may be found on T2-
in the different polyglutamine disorders could be re- weighted, proton-density, and FLAIR images. Conflu-
lated to different factors, including the cellular distri- ent white matter abnormalities are found in older pa-
bution of the polyglutamine protein, protein interac- tients with a diffuse increase in signal in the periven-
tors, and cell specificity of certain modifying pro- tricular and deep white matter, only sparing most of
teins. The latter group may include cell-specific trans- the U fibers (Figs. 71.1–71.3). The pattern of cerebral
glutaminases and cysteine proteases. white matter abnormalities in combination with sig-
Nuclear inclusions were previously considered to nal changes in the globus pallidus, thalamus, and
be a feature specific to neuronal degenerative dis- brain stem is typical of an advanced stage of DRPLA.
532 Chapter 71 Dentatorubropallidoluysian Atrophy

Fig. 71.1. A 73-year-old female patient with DRPLA. The mid- U fibers and the corpus callosum. Note the signal abnormali-
brain, pons, and cerebellum are atrophic. There is also some ties in the globus pallidus, thalamus, midbrain, and pons. From
cerebral atrophy. There are diffuse signal abnormalities in the Uyama et al. (1995), with permission
cerebral hemispheric white matter with partial sparing of the
71.5 Magnetic Resonance Imaging 533

Fig. 71.2. A 60-year-old female patient with DRPLA.The sagit- caudate nucleus, globus pallidus, putamen, thalamus, mid-
tal and axial FLAIR images reveal extensive signal abnormali- brain, tegmentum of the pons, and dorsal part of the medulla.
ties in the cerebral white matter with partial sparing of the U The red nuclei are spared. Note the atrophy of the cerebellum,
fibers. The inner rim of the corpus callosum shows an abnor- pons and superior cerebellar peduncles. From Yoshii et al.
mal signal. There are additional signal abnormalities in the (1998), with permission
534 Chapter 71 Dentatorubropallidoluysian Atrophy

Fig. 71.3. A 68-year-old female patient with DRPLA. Note the sum and some of the U fibers. There are also signal abnormal-
evident pontine and cerebellar atrophy. There are extensive ities in the globus pallidus,thalamus,midbrain,and pons.From
cerebral white matter abnormalities, sparing the corpus callo- Uyama et al. (1995), with permission
Chapter 72

Cerebral Amyloid Angiopathy

72.1 Clinical and Laboratory Findings leptomeningeal amyloidosis. They vary in presenta-
tion but dementia, hemiplegic migraine, spasticity,
Cerebral amyloid angiopathy (CAA) was recognized blindness, and deafness are usually present. These
as a disease entity at the beginning of the twentieth patients have amyloid deposition in the vitreous and
century. The first reports appeared in the German lit- retinal vessels, leptomeningeal vessels, and other
erature in 1907 and 1909. In 1935 a familial form of organs.
this disease was reported from Iceland, later followed The incidence of sporadic CAA increases with age.
by the description of a Dutch family (1964), a Flemish The differentiation from other conditions with small
variant (1992), and a British type (1996). In the latter vessel disease may be difficult. There is a relationship
variant hemorrhage is not a prominent feature, as it is with sporadic Alzheimer disease. Amyloid deposition
in the Icelandic and Dutch type.Apart from the famil- in vessel walls is also a factor of importance in spo-
ial forms, there are also sporadic forms. radic and familial forms of Alzheimer disease. CAA is
The classic presentation of CAA is lobar hemor- found in 92% of postmortem studies of brains of pa-
rhage, often multiple, associated with an acute neuro- tients with Alzheimer disease. Amyloid deposition is
logical presentation. CAA is responsible for 5–20% of accepted as one of the hallmarks of Alzheimer dis-
nontraumatic cerebral hemorrhages and 30% of lo- ease. CAA, however, can occur without any other sign
bar hemorrhages. The most frequent locations are: of Alzheimer disease, and Alzheimer disease can
frontal (35%), parietal (26%), temporal (14%), and occur without CAA. If CAA is present there is risk of
occipital (19%). Cerebellar and basal ganglia hemor- lobar hemorrhage. There is no obvious relation with
rhages are infrequent; brain stem hemorrhages are the occurrence, location, and severity of neuritic
extremely rare. Sporadic CAA may present with a va- plaques or neurofibrillary tangles.
riety of symptoms other than lobar hemorrhage. Apart from a relationship with Alzheimer disease,
There may be petechial hemorrhages in cortical and CAA is reported to occur in transmissible spongiform
subcortical areas and these may produce either recur- encephalopathies (prion diseases), especially in Ger-
rent transient neurological symptoms or rapidly pro- stmann–Sträussler–Scheinker disease, and, less com-
gressive dementia. Dementia is estimated to occur in monly, in Creutzfeldt–Jakob disease; in malignant
about 40% of patients with CAA. The progression of neoplasms treated with irradiation, parkinsonism–
dementia is as a rule much faster than in Alzheimer dementia complex of Guam, and dementia pugilisti-
disease. ca. A few cases of primary angiitis of the CNS have
The familial forms of CAA have an autosomal been reported in combination with CAA.
dominant inheritance. There are differences in pre- Routine laboratory tests are unrevealing. The diag-
sentation between the affected families. The Icelandic nosis of CAA should be suspected on the basis of typ-
form presents in the third decade with multiple in- ical clinical and neuroimaging findings. The diagno-
tracerebral hemorrhages and infarctions. The disease sis may be confirmed by a leptomeningeal biopsy, but
progresses rapidly to dementia, paralysis, and early this is only done in exceptional cases. In familial CAA
death. The Dutch form has a later onset, in the fourth histological confirmation in one patient is necessary,
to fifth decade of life. Chronic migrainous headache, whereas in other affected family members suggestive
as in CADASIL, may precede the onset by years. The clinical and neuroimaging findings suffice for a diag-
presenting symptom is most often intracerebral nosis. The Boston group for research of CAA has
hemorrhage, sometimes ischemic strokes. Neuro- come up with criteria for the diagnosis of CAA-relat-
psychiatric symptoms and dementia are constant ed hemorrhage (see Knudsen et al. 2001). Patients
features. The Flemish variant presents with intracere- fulfilling these criteria, however, have a hemorrhage,
bral hemorrhage and dementia. The British variant so that patients without hemorrhage are excluded.
shows progressive dementia, spasticity, and ataxia The MR criteria mentioned in this set of criteria also
(Worster–Drought syndrome). In this form the entire do not specify the mandatory inclusion of gradient
CNS is involved, including cerebral white matter, cere- echo techniques, which would help to identify pa-
bellum, brain stem, and spinal cord. Hemorrhage is tients with and without hemorrhages with CAA,
not prominent. Other families from Japan, Italy, and in addition to being helpful to demonstrate other
North America have been reported with oculo- possible causes.
536 Chapter 72 Cerebral Amyloid Angiopathy

72.2 Pathology 72.3 Pathogenetic Considerations

Gross inspection of brain sections reveals a combina- The central biochemical event in the pathophysiology
tion of larger and smaller hemorrhages, most often in of CAA is the polymerization of soluble subunit pro-
the cortex and subcortical regions; small infarctions, teins into insoluble amyloid fibrils. Several different
mainly affecting the cerebral cortex; and extensive forms of amyloid subunit proteins are deposited in
white matter abnormalities, mainly involving the the vasculature leading to amyloid angiopathy,
deep and periventricular white matter, usually spar- including Ab protein, cystatin C, transthyretin, and
ing the U fibers. Lacunae may also be present in the gelsolin.
basal ganglia. Ventricular dilatation is usually also Ab protein is a hydrophobic, nonglycosylated pep-
present. tide of 39–43 amino acids. Ab protein is derived from
On microscopic examination the lesions in the the amyloid precursor protein (APP) encoded by the
white matter consist of patchy gliosis and myelin loss, gene APP located on chromosome 21q22.1. APP is one
the latter in particular in the parieto-occipital areas. of the genes for familial Alzheimer disease, AD1. In
U fibers, corpus callosum, internal capsule, and optic sporadic CAA, CAA with sporadic Alzheimer disease,
radiation are usually spared. Perivascular spaces are familial Alzheimer disease, Down syndrome, and the
enlarged and contain mononuclear cells and hemo- Dutch type of CAA, the vascular amyloid is composed
siderin-laden macrophages. Around the ventricles of Ab protein subunits. Subtle differences exist be-
Rosenthal fibers may be found. tween the vascular amyloid in CAA and amyloid
The most striking finding in CAA is that smooth parenchymal deposits, such as in amyloid plaques in
muscle in the vessel wall of small blood vessels is Alzheimer disease. Vascular amyloid is composed
replaced by a hyaline, eosinophilic material. The de- mainly of the 39-amino-acid Ab species, plaque amy-
posits stain with Congo red, hence the name “con- loid predominantly of a 42-amino-acid Ab species.
gophilic angiopathy.” The term “amyloid” is applied to Different point mutations in APP, causing an amino
a number of proteins sharing the property of Congo acid substitution in the Ab protein, have been found
red staining, which then exhibits green birefringence in the Dutch type of hereditary cerebral hemorrhage
under polarized light. This latter property is depen- with amyloidosis (HCHWA-D) and in Italian families
dent upon the configuration of a twisted b-pleated with similar clinicopathological findings to those in
sheet. This is a feature of amyloid laid down under HCHWA-D.
different circumstances and in different organs of the Additionally, the e4 allele (apoEe4) of apolipopro-
body, which, despite identical staining properties, tein E (apoE) is a risk factor for CAA and Alzheimer
may have different amino acid sequences in the con- disease and the e2 allele has been linked to greater
stituent polypeptide chains. Specific antibodies risk of CAA and cerebral hemorrhage. The mecha-
against amyloid b peptide (Ab), cystatin C, and thyre- nism underlying this increased risk is not completely
ostatin can be used for immunohistochemical char- clear, but there is increasing evidence that the ability
acterization. It is of interest that in patients with a of apoE to interact with Ab and influence its confor-
combination of CAA and Alzheimer disease the de- mation and clearance plays a major role. ApoE ap-
posits in the vessel wall are immunoreactive with pears critically involved in the conversion of Ab into
both anti-Ab and anti-cystatin C. Fibrinoid necrosis forms which have a high b-sheet content with associ-
and hyaline changes in vessel walls are usually also ated cellular toxicity.
present in sporadic cases of CAA and larger cortical In the Icelandic form of CAA, HCHWA-I, the vas-
vessels are often thrombosed. Ultrastructurally, amy- cular amyloid is composed of a mutated form of cys-
loid fibrils in blood vessels appear as interwoven bun- tatin C, a cysteine proteinase inhibitor, playing a role
dles of 10-nm filaments, short and disarranged. In in intracellular catabolism of peptides and proteins.
early CAA only the outer part of the vessel wall is Cystatin C is encoded by the gene CST3, located on
involved, but when the deposits become larger, they chromosome 20p11.2.
occupy the abluminal part of the basement mem- In the familial oculo-leptomeningeal amyloidoses
brane and adjacent smooth muscle cells show degen- the vascular amyloid is transthyretin, a prealbumin
erative features. When severely affected, the walls of that is also present in hereditary peripheral amyloi-
blood vessels may be replaced entirely by bundles of doses. Transthyretin is an amino acid residue carrier
fibrils with loss of smooth muscle cells and with radi- protein for thyroid hormone and retinol binding
ation of fibrils into the surrounding neuropil. protein in plasma and CSF. The gene encoding
transthyretin is TTR, located on chromosome
18q12.1. In CAA related to transthyretin most of the
families (Japan, Portugal) have an association with
the same point mutation in TTR, but other mutations
have also been reported.
72.5 Magnetic Resonance Imaging 537

In the British form of CAA a novel 4-kDa protein ing to loss of smooth muscle cells of the media, fol-
fragment called amyloid-bri (ABri) was identified in lowed by vascular dilatation, spindle-shaped microan-
isolated amyloid fibrils. ABri is a fragment of a pre- eurysms, intimal thickening with thinning and dis-
cursor protein (BriPP), a putative type II single-span- ruption of media and adventitia, changes in vascular
ning transmembrane precursor that is encoded by the permeability, invasion of plasma components into the
gene BRI, located on chromosome 13. A mutation in vascular wall (fibrinoid necrosis), possible additional
the stop codon of this gene generates a longer open factors (e.g., hypertension, Ab-induced production
reading frame, resulting in a larger, 277-amino-acid of superoxide free radicals from endothelial cells),
protein, whereas the wild-type protein is 266 amino activation of fibrinolytic systems by Ab, infiltration of
acids. Cleavage of 34 amino acids at the C-terminal inflammatory cells with production of cytokines, and
end of the extended protein generates the amyloido- the ApoEe2 allele), ending in hemorrhage.
genic fragment, ABri. The frequency of sporadic congophilic angiopathy
The Finnish, gelsolin-related type of CAA is noted in the normal aging subject is variable. In patients
worldwide in relatives carrying the same mutation in over the age of 60 years presenting with lobar hemor-
the gelsolin gene on chromosome 9. rhage, the incidence is high. In stroke patients, too,
Amyloidosis is a phenomenon in which native with the proper techniques, MRI detects microhem-
globular proteins form long fibrils with a characteris- orrhages in more than 20% of patients. In individuals
tic three-dimensional structure. Although emerging without hemorrhage or other clinical symptoms, the
from different proteins, amyloid fibrils show a com- percentages in population studies differ. There is little
mon X-ray diffraction pattern, indicative of a cross-b evidence of the disorder under the age of 60. In the
structure, where the b-sheets run parallel to the fibril population over 60 years of age there is evidence of
axis and the b-strands forming the sheets are perpen- congophilic angiopathy in about 30% of individuals.
dicular to this axis. Well studied amyloidogenic pro- In normal aging the changes are usually mild and
teins include the Ab protein. The tendency to self-ag- limited to the cerebral cortex, most often in the pari-
gregate into insoluble fibrils is stronger in Ab 42 frag- eto-occipital region. The presence of microhemor-
ments of APP than in Ab 39 or Ab 40 fragments. Fac- rhages in a high percentage of normal aging people
tors promoting in vivo Ab-amyloid formation are: can induce false positives when brain biopsies are
∑ Overproduction, increased levels of APP (in gene considered to obtain a histological diagnosis.
overdose and trisomy 21)
∑ A highly amyloidogenic sequence (as in the codon
693 mutation of the Dutch variant of CAA) 72.4 Therapy
∑ Altered proteolysis of APP (as in familial
Alzheimer disease due to codon 692 mutation) There is no effective treatment for the underlying dis-
∑ A seeding nucleation event, analogous to seeding ease process of CAA. Once a patient is diagnosed with
in crystal formation CAA, measures can be taken to prevent brain hemor-
∑ Time/patient age rhage as much as possible. High blood pressure
should be treated. Blood thinners such as Coumadin
Many theories describe the detailed process of amy- (warfarin), antiplatelet agents such as aspirin, or
loid deposition in the parenchyma or the vessel wall. medications designed to dissolve blood clots may
In Alzheimer disease the amyloid cascade is a good cause hemorrhage in patients with CAA, and should
example. It describes the stepwise progression in the be avoided if possible. If these medications are re-
deposition of amyloid in Alzheimer disease, starting quired for other conditions, such as heart disease, the
with either the genetic influence of the APP or prese- potential benefits must be carefully weighed against
nilin 1 and 2 genes, or the failure of clearance of Ab the increased risks. Seizures should be treated with
because of the apoEe4 allele, increasing the Ab42 lev- antiepileptic drugs, although some drugs should be
els, followed by oligomerization of Ab42 in limbic and avoided because of their antiplatelet effect. Once a he-
association cortices, with gradual deposition of Ab42 morrhage has occurred, supportive and symptomatic
oligomers, activation of microglia, complement, as- medical care is important. Sometimes neurosurgical
trocytes, altering neuronal ionic homeostasis, leading intervention is necessary to reduce the pressure with-
to oxidative injury, production of changes in tau pro- in the brain.
teins, and formation of tangles and plaques, leading
to neuronal dysfunction, dementia, and death. Other
theories try to explain why vascular amyloidosis in 72.5 Magnetic Resonance Imaging
the absence of other risk factors leads to hemorrhage,
an important feature in CAA. One of these theories In patients presenting with an intracerebral hemor-
suggests a cascade starting with amyloid deposition rhage, both CT and MRI will show the hemor-
in the media and adventitia of cortical arteries, lead- rhage(s), together with its secondary effects, such as
538 Chapter 72 Cerebral Amyloid Angiopathy

Fig. 72.1. A 54-year-old female with a familial history of amy- of very low signal intensity, dispersed throughout the brain.
loid angiopathy. The first row demonstrates T1-weighted The low signal intensity is caused by hemosiderin residues of
parasagittal images of the left hemisphere with a parietal microhemorrhages. The combination of lobar hemorrhage
lobar hemorrhage. The second and third rows represent with multiple spot-like microhemorrhages is characteristic
T2*- (GE-)weighted images revealing smaller and larger spots of CAA

Fig. 72.2. Transverse FLAIR images of a 72-year-old man with cause of the white matter changes is not evident from these
slowly progressive cognitive deterioration (first and second images.The images made with a T2*-weighted sequence (third
rows). The images show slightly asymmetrical confluent areas and fourth rows) reveal multiple spots with very low signal
of hyperintensity in the deep and periventricular white matter, intensity, representing microhemorrhages. These images sug-
in some places extending into the U fibers. The underlying gest that the patient is suffering from a sporadic form of CAA
72.5 Magnetic Resonance Imaging 539

Fig. 72.2.
540 Chapter 72 Cerebral Amyloid Angiopathy

mass effect worsened by edema, imminent tentorial times there is involvement of the midbrain and pons,
herniation, hydrocephalus, and breakthrough of and occasionally there is a lesion in the cerebellum.
blood into the ventricles and subarachnoid space. The The usual work-up for hemorrhages in the brain
location of the hemorrhage(s) is important: CAA he- should be performed, including MR angiography,
morrhages are mainly lobar and predominantly in preferably phase contrast angiography when blood
the parieto-occipital region (Fig. 72.1), in contrast to is present, and contrast administration to exclude
hypertension-related and aneurysm-related hemor- metastases or arteriovenous malformations. The
rhages. In CAA hemorrhages in the brain stem and most important and mandatory MR sequence in cas-
posterior fossa are rare. Especially cases with multiple es of CAA is a gradient echo pulse sequence, because
hemorrhages are suspect for CAA, more so in the of its sensitivity to changes in magnetic susceptibility
absence of vascular risk factors. In addition to hemor- (Figs. 72.1 and 72.2). These T2*-weighted images may
rhages MRI will show infarctions, when present, in show multiple residues of petechial hemorrhages
more detail than CT. Lacunar infarctions will be seen all through the brain (Figs. 72.1 and 72.2). This fea-
best on FLAIR images, where they appear as black ture may be seen, however, in numerous disorders
dots with a bright rim. In nearly all cases MRI will involving small vessels, such as hypertensive en-
show extensive white matter abnormalities in the cephalopathy, vasculitides, atherosclerosis, CADASIL,
periventricular and deep white matter, but sparing or Rendu–Osler disease, and may be mimicked by
the U fibers, with confluent and isolated lesions which multiple melanoma metastases. They are also present
are not necessarily symmetrical in distribution. in a considerable percentage of normal aging individ-
There are often lesions in the basal ganglia; some- uals.
Chapter 73

Cerebral Autosomal Dominant Arteriopathy


with Subcortical Infarcts and Leukoencephalopathy

73.1 Clinical Features Table 73.1. Diagnostic criteria for probable CADASIL (Davous
1998)
and Laboratory Findings
1. Young age at onset (<50 years)
The acronym CADASIL stands for cerebral autosomal 2. At least two of the following clinical findings:
dominant arteriopathy with subcortical infarcts and – Stroke-like episodes with permanent
leukoencephalopathy. This disease was formerly neurological signs
known as familial Binswanger disease or hereditary – Migrainous headaches
– Major mood disturbances
multi-infarct dementia. Although it was initially – Subcortical dementia
thought to be a rare disease, many families with 3. No vascular risk factors related to deficit
CADASIL have now been reported worldwide.
4. Evidence of inheritance with autosomal dominant
CADASIL is the most common hereditary form of transmission
stroke leading to progressive dementia.
5. On MRI, white matter abnormalities without cortical
The clinical picture is dominated by recurrent sub- infarcts
cortical ischemic infarctions, beginning somewhere
between 40 and 60 years, or occasionally at a younger
age. There is usually a striking absence of vascular
risk factors: there is no history of hypertension, no Diagnosis based on the electron microscopic demon-
hypercholesterolemia, and no hyperhomocysteine- stration of granular osmiophilic material (GOM) ad-
mia. Migrainous headaches and psychiatric symp- jacent to the basement membrane of smooth muscle
toms are frequent. They are often the initial symp- cells of arterioles has a specificity of 100%, but the
toms and may precede other symptoms of the disease sensitivity is rather low (~50%). Immunostaining
for many years. The neurological symptoms progress with antibodies against the Notch3 protein may in-
over months to years, most often in a step-like fash- crease the sensitivity of skin biopsy techniques sub-
ion, and may worsen in the second decade after onset. stantially; in patients with CADASIL, there is an ab-
Hemiparesis, hemianopia, dysarthria, and cerebellar normal accumulation of the Notch3 protein in the
ataxia may characterize these events. Other symp- vessel walls. Diagnosis by mutation detection is also
toms include seizures, pseudobulbar palsy, urinary possible. In the absence of such evidence criteria for
incontinence, and unexplained “CADASIL coma.” probable CADASIL have been proposed, which are
This latter event is a subacute encephalopathy, in summarized in Table 73.1.
some cases accompanied by fever, with focal neuro- Discovery of de novo mutations in isolated pa-
logical symptoms and seizures, eventually progress- tients emphasizes that a possible diagnosis of
ing to coma. The condition is reversible in most cases CADASIL should not be rejected in the absence of a
but may recur. In the history of nearly all the report- family history. Neuroimaging plays an important role
ed patients migraines with visual or sensory auras are in the initial diagnosis. The pattern of white matter
present. In other patients the diagnosis “relapsing–re- abnormalities is highly suggestive (see below) and the
mitting multiple sclerosis” was maintained for years. MRI-based diagnosis of CADASIL can be substantiat-
Psychiatric symptoms, including confusion, severe ed with many more details than suggested under
forms of depression, and signs of frontal dysfunction, point 5 of the above criteria. Especially involvement
are manifest in more than 30% of the patients. A sub- of the anterior temporal pole has a high sensitivity
cortical dementia, similar to that seen in subcortical and specificity (both 85–90%). CSF analysis shows
arteriosclerotic encephalopathy, evolves. There is oligoclonal bands in quite a number of cases, al-
considerable variability in clinical symptoms and though these are considered to be nonspecific.
course of disease among the patients, also within fam-
ilies. The median survival is 64 years for males and 69
years for females. 73.2 Pathology
A definite diagnosis of CADASIL can be made on
the basis of histopathological findings in skin, mus- The primary disease process is a vasculopathy, most
cle, or brain biopsy or when the typical clinical find- prominently involving the small perforating arteries
ings can be linked to a mutation in the gene NOTCH3. of the cerebral white matter. Within the media of the
542 Chapter 73 Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy

small arterial vessels, deposits of GOM are seen on CADASIL, a dramatic and selective accumulation of
electron microscopy, which displace degenerating the extracellular domain of the receptor protein
smooth muscle cells of the arterial media in small occurs at the cytoplasmic membrane of vascular
vessels, a feature now considered the pathological smooth muscle cells, in close vicinity to but not with-
hallmark of CADASIL. GOM has an extracellular in GOM. This suggests that CADASIL mutations
location and is not membrane-bound. The chemical specifically impair the clearance of the notch3
nature of GOM is unknown. The material usually ectodomain, but not the cytosolic domain, from the
stains with PAS, consistent with acid polysaccharide. cell surface.
Other histochemical stains have shown that the mate- It is evident that the clinical manifestations, di-
rial does not contain amyloid, elastin, chromatin, cal- verse as they may be, are the result of the vascular
cium, or iron. Immunoenzymatic and immunofluo- changes leading to hypoperfusion, demyelination,
rescence studies have also shown absence of im- gliosis, and lacunar infarctions. How the genetic
munoglobulins or complement proteins, cystatin C, changes translate into the vascular changes and the
transthyretin, gelsolin, fibrinogen, cathepsin D and phenotypic variability is still incompletely under-
a1-antichymotrypsin. Although the disease manifests stood.
itself only through neurological and neuropsychiatric
symptoms, it is a generalized disorder and the ultra-
structural lesions of the arterial wall are also found in 73.4 Therapy
other organs, including the spinal cord, spleen, heart,
muscle, skin, and peripheral nerves. Biopsy of these As in other disorders with cerebrovascular disease,
tissues can confirm the diagnosis. Skin biopsy is most strategies are directed at improving cerebral perfu-
often used for that purpose. sion.Anticoagulants and thrombolytic agents, such as
The concentric thickening of the wall leads to is- warfarin and heparin, are not advocated because of
chemia, infarctions, and myelin loss, which are shown the risk of hemorrhage. Aspirin, despite its low effica-
by microscopic examination of the brain. Multiple cy in some patients, is considered the best choice
small infarcts and lacunae are seen in the basal gan- with the smallest chance of complications. Neuro-
glia, thalamus, internal capsule, periventricular white protective agents, antioxidants, N-methyl-D-aspartate
matter, and brain stem. The cerebellum is less often (NMDA) antagonists, and voltage-sensitive calcium
affected. The periventricular white matter may be se- channel antagonists, for example nimodipine and
verely demyelinated. The U fibers and the cortex are flunarizine, also have been suggested. Other com-
usually spared. Cortical infarcts are rare and of small pounds, like propentofylline, a xanthine derivative
size. Fresh and old hemorrhages may also be found, with multiple actions, have been tested positively in
most often in advanced stages of the disease. Alzheimer disease, and might also be beneficial in
vascular dementias.

73.3 Pathogenetic Considerations


73.5 Magnetic Resonance Imaging
CADASIL is related to dominant mutations in
NOTCH3 on chromosome 19q13.1. The gene codes for Although there are many similarities between
a large transmembrane receptor protein. The extra- CADASIL, subcortical arteriosclerotic encephalopa-
cellular portion of the Notch3 protein contains 34 thy, and cerebral amyloid angiopathy, the MR pattern
tandem repeats of an epidermal growth factor (EGF) of fully developed CADASIL has several features that
motif, each of which contains six cysteine residues can make the MR diagnosis highly suggestive and, in
binding within the domains as three cysteine–cys- combination with the clinical features and family his-
teine disulfide bonds. Mutations that have been tory, even diagnostic.
demonstrated in CADASIL occur within these EGF MRI shows extensive white matter abnormalities
repeats and always involve either loss or gain of a cys- in the absence of cortical infarcts (Figs. 73.1–73.3).
teine residue. The mutations result in altered disulfide There is a tendency to symmetry. The centrum semi-
binding within the repeats by changing the number of ovale is involved, with relative sparing of the U fibers.
cysteines from six to an odd number. There is evi- The abnormalities extend downwards into the exter-
dence that mutations may lead to a gain of function. nal and extreme capsules. In the frontal lobes the
Other evidence demonstrates that some CADASIL white matter abnormalities extend into the U fibers.
mutations reduce the activity of the Notch3 receptor. The white matter of the temporal lobes is involved in
There is considerable phenotypic variability, which is typical cases (Figs. 73.1–73.5). Moderate to severe in-
thus far unexplained. volvement of the temporal lobes has a sensitivity of
In normal tissue, gene expression for NOTCH3 is 89% and a specificity of 83%. Involvement of the ex-
highly restricted to vascular smooth muscle cells. In treme and external capsule has a sensitivity of 93%,
73.5 Magnetic Resonance Imaging 543

Fig. 73.1. Images of a 45-year-old woman with CADASIL, glia, thalamus, pons, and cerebellum is also more or less sym-
showing most of the typical MR features. The T2-weighted im- metrical. The CT scan (first row, left) and the T1-weighted im-
ages (first row, middle and right; second row, left and middle) ages (third row) show multiple small hemorrhages in the cen-
show nearly symmetrical involvement of the white matter in trum semiovale and the posterior temporal lobe on the right.
the centrum semiovale,with extensions into the arcuate fibers, The T1-weighted images, including the sagittal image (second
the external capsules, and the temporal lobes. The corpus cal- row, right), show the widened perivascular spaces in the cen-
losum is relatively spared. The involvement of the basal gan- trum semiovale and the basal ganglia.

but a low specificity of 45%. To differentiate between the white matter changes with high signal intensity,
small lacunar infarctions and other white matter ab- whereas cystic infarctions have very low signal inten-
normalities, such as demyelination and gliosis, FLAIR sity. Diffusion-weighted imaging may demonstrate
images are very useful (Figs. 73.2–73.5). They show the relatively fresh infarctions.
544 Chapter 73 Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy

Fig. 73.2. The MR pattern in a 61-year-old woman with bellar atrophy. Courtesy of Dr. K. Demuth, Department of Neu-
CADASIL is similar to the pattern seen in Fig. 73.1,although the rology, Marien Hospital, Stuttgart, Germany
white matter abnormalities are more confluent. There is cere-

Good quality images show that, apart from the sig- the often multiple small or lacunar infarctions in the
nal changes in the white matter, the Virchow–Robin basal ganglia, one may see a more generalized low sig-
spaces are widened (Fig. 73.1). This leads to an état nal intensity on T2-weighted images, in particular in
criblé of the basal ganglia. The widening of the the globus pallidus (Figs. 73.3 and 73.4). CT does not
perivascular spaces may also involve the centrum show calcification in the basal ganglia, so that the ex-
semiovale and the temporal lobes. planation must be deposition of other substances, for
In longer-standing cases of CADASIL signal example iron.
changes may occur in the basal ganglia. Apart from
73.5 Magnetic Resonance Imaging 545

Fig. 73.3.
546 Chapter 73 Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy

Fig. 73.3. (continued). Series of FLAIR images in a 59-year-old signal intensity.The sagittal (fourth row) and coronal (fifth row)
woman, a representative of a known CADASIL family. In this FLAIR images further illustrate and complete this pattern. The
series many MR characteristics of CADASIL are present: more frontal cortico-subcortical involvement is depicted, as are the
or less symmetrical involvement of the centrum semiovale, multiple lacunar infarctions and the extension of the lesions
external capsules, temporal lobes, and basal ganglia; multiple into the external capsules and the temporal lobes. There is al-
lacunar infarctions; some involvement of the cerebellum. Even so involvement of the corpus callosum and the fornix (mid-
on the FLAIR images the basal ganglia have a relatively low sagittal image, fourth row, right)

Abnormalities in the mesencephalon, pons, and of the above-mentioned findings may already be pre-
cerebellum are often less symmetrical than lesions in sent in clinically unaffected members of patients’
the cerebral hemispheres. One may find an occasion- families.
al infarction in the mesencephalon, pons, and cerebel- In the early stages of the disease, the pattern of
lum, and a high signal in the transverse fibers of the MRI abnormalities is less characteristic and more dif-
pons (Figs. 73.1, 73.2, and 73.4). ficult to differentiate from other vascular disorders
MR angiography usually does not show the (Fig. 73.5). Abnormalities of the anterior temporal
changes in the small vessels that cause the problems white matter and the external capsule are early find-
in CADASIL. At the same time intra-arterial DSA ings and, in the presence of multiple small infarc-
seems contraindicated because of the unexplained tions, suggestive of CADASIL.
extremely high complication rate of up to 50% in in- The differential diagnosis of CADASIL includes
tra-arterial DSA in CADASIL patients. other familial disorders presenting with multiple
After contrast injection there is no enhancement recurrent incidents and white matter lesions. Other
seen in any of the lesions, unlike, for example, Bin- familial disorders, in which recurrent incidents or
swanger disease, where new infarcts will display en- step-like progression is a prominent symptom, in-
hancement in the subacute phase, or multiple sclero- clude multiple sclerosis, the dyslipoproteinemias
sis, where acute plaques show enhancement. (e.g., familial hypercholesterolemia), disorders of
Gradient refocused T2*-weighted images show in connective tissue (Ehlers–Danlos syndrome, Marfan
many cases spots of low signal intensity, representing syndrome), hyperhomocysteinemia, amyloid an-
microhemorrhages. This feature is also seen in other giopathy, and mitochondrial encephalopathies. Re-
small vessel diseases. Small hemorrhages may also be current strokes in young adults may also be due to
seen on CT and T1-weighted images (Fig. 73.1). cerebral vasculitis, systemic lupus erythematosus,
MR diffusion-weighted imaging is capable of drug abuse (especially cocaine), cardiac diseases,
quantitative estimation of the structural damage of sickle cell disease, etc. Most of these diseases can be
the brain, which in its turn may relate to the clinical ruled out by the mode of inheritance, the clinical ap-
severity of the disorder. Changes in diffusion-weight- pearance and course, biological tests, and laboratory
ed imaging consist of an up to 60% increase in diffu- findings. MRI of these diseases and disorders shows a
sivity in lesions with high signal intensity on FLAIR pattern of irregular cortical and subcortical infarc-
or T2-weighted images, with loss of diffusion aniso- tions, lacking the tendency towards symmetry seen in
tropy in those areas. In normal-appearing white mat- CADASIL. In the disorders mentioned, the infarcts
ter, however, changes in diffusivity were also found. may involve both cortex and white matter, whereas
These findings do not explain one of the most in- CADASIL involves white matter and basal ganglia,
triguing MRI aspects of CADASIL, namely that many without cortical infarctions. Temporal lobe involve-
73.5 Magnetic Resonance Imaging 547

Fig. 73.4. In this 48-year-old man, a representative of another external capsules and evident involvement of the temporal
large family with CADASIL, the MR pattern is difficult to distin- lobes.There are many lacunar infarctions, which, together with
guish from that of other vascular disorders.The involvement of the dark appearance of the globus pallidus, often seen in pa-
the centrum semiovale is patchy and less symmetrical than in tients with CADASIL, indicate the correct diagnosis
more advanced CADASIL.There is incipient involvement of the

ment also argues strongly for CADASIL. The differen- hemiplegic migraine can be confused with the begin-
tial diagnosis of white matter disorders with cogni- ning of CADASIL, but does not show white matter
tive impairment and possible severe temporal lobe abnormalities comparable to those of CADASIL.
involvement is from myotonic dystrophy. Familial
548 Chapter 73 Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy

Fig. 73.5. A 31-year-old male member of the same family CADASIL. The images show multiple lacunar infarctions. In
as the patient in Fig. 73.4. Without knowing the diagnosis in addition, there is incipient involvement of the external cap-
the family, it would be difficult to diagnose this patient with sules and the white matter of the anterior temporal lobes
Chapter 74

Cerebral Autosomal Recessive Arteriopathy


with Subcortical Infarcts and Leukoencephalopathy

74.1 Clinical and Laboratory Findings elastic membrane. The thickened vessel walls do not
stain with PAS stain or Congo red, there is no ultra-
This disease was first described in 1976 by Maeda et structural granular osmiophilic material in the vessel
al. as a “familial unusual encephalopathy of Bin- walls, and there is no b-amyloid immunoreactivity in
swanger’s type without hypertension” in four pa- vascular walls. The presence of PAS-positive material
tients, two of whom were brothers. In nearly all cases and the ultrastructural finding of granular osmio-
reported after this first publication, the parents were philic material in the vessel walls are considered to be
consanguineous, often first cousins, providing evi- the hallmarks of CADASIL, making CARASIL distinct
dence for an autosomal recessive mode of inheri- from CADASIL. There is also no increase of amyloid
tance. Over the years the syndrome has crystallized plaques or neurofibrillary tangles. Large arteries at
sufficiently to allow definition of the clinical picture. the base of the brain display no or mild atheromatous
The disease occurs predominantly in males, with a changes; severe changes are rare. In visceral organs
male:female ratio of 7.5:1. So far, all patients de- arterial changes are less severe. A muscle biopsy,
scribed are Japanese. The disease has a relatively ear- however, may show the described vessel changes. The
ly age of onset, most often between 25 and 30 years. superficial temporal arteries are often involved, pos-
The course of the disease is relatively rapid and is sibly explaining the alopecia.
characterized by step-like or chronic-progressive
pyramidal and extrapyramidal symptoms, pseudo-
bulbar palsy, and cognitive deterioration. The neuro- 74.3 Pathogenetic Considerations
logical symptoms are associated with alopecia from
youth. There is diffuse hair-thinning with similarities In CARASIL the brain damage is caused by narrowing
to what is seen in radiation injury and systemic lupus of the lumina of arterioles, especially of long perforat-
erythematosus. Patients experience acute lumbago. ing arteries supplying the periventricular and deep
The lumbago is caused by kyphotic and spondylotic white matter. There is an obvious similarity to other
changes of the vertebrae, often in an unusual location, diseases in which systemic narrowing of penetrating
the lower thoracic and higher lumbar region. Patients small medullary vessels is at the core of the patho-
lack vascular risk factors, especially hypertension. physiology. The resulting decrease in cerebral perfu-
Diffuse white matter disease is seen on MRI or at sion leads to white matter pallor, demyelination, glio-
autopsy. The disease is often compared to Binswanger sis, preinfarct status, and lacunar infarctions. The dis-
disease, but the MR images more closely mimic the ease is hereditary, but not linked to the NOTCH 3
MRI characteristics of CADASIL (see below). Life ex- gene, which is responsible for CADASIL. The genetic
pectancy in these relatively young patients is short- basis of CARASIL has not yet been elucidated.
ened considerably. Most patients die within 10 years
after onset of the disease in a vegetative state.
Laboratory examinations are unrevealing. There 74.4 Therapy
are no biochemical changes in blood or urine samples.
So far no therapy has been applied aimed at improve-
ment of cerebral blood flow and counteracting is-
74.2 Pathology chemia, as described in the chapter on CADASIL.
Treatment is mainly symptomatic. Disc herniation
The cortex is intact. There is diffuse demyelination of and lumbar stenosis may demand surgical interven-
the cerebral white matter, with relatively well pre- tion.
served U fibers. Within the affected white matter and
within the basal ganglia, there are small softened and
cystic foci. The corpus callosum is affected in about 74.5 Magnetic Resonance Imaging
half of the patients. In the abnormal white matter
arteriosclerosis is seen involving arteries of 100- to MRI shows a pattern of white matter involvement
400-mm caliber, with fibrous intimal proliferation, similar to that of CADASIL. The white matter lesions
severe hyalinosis, and splitting of the intima and the are confluent and more or less symmetrical (Figs. 74.1
550 Chapter 74 Cerebral Autosomal Recessive Arteriopathy with Subcortical Infarcts and Leukoencephalopathy

Fig. 74.1.
74.5 Magnetic Resonance Imaging 551

Fig. 74.2. A 42-year-old male patient with CARASIL. The MRI callosum is thin but seems to have a normal signal. There are
was obtained 16 years after the onset of the disease. Note the many small lesions in the basal ganglia and brain stem. The
extensive cerebral white matter abnormalities, which do not T1-weighted images do not show evidence of small hemor-
spare the U fibers. The internal and external capsules are af- rhages. From Fukutake and Hirayama (1995), with permission
fected as well.The cerebral white matter is atrophic; the corpus

and 74.2). They involve the cerebral hemispheric certainly not completely spared (Fig. 74.1), and be-
white matter and both internal and external capsules. come extensively affected in later stages (Fig. 74.2).
The temporal lobes are affected as well, including the The corpus callosum tends to be spared. There are
temporal poles. The U fibers are relatively spared, but small focal lesions in the basal ganglia, thalami, and
brain stem (Figs. 74.1 and 74.2). FLAIR images show
small lacunae (Fig. 74.1), whereas diffusion-weighted
䊴 images may reveal small fresh infarctions (Fig. 74.1).
Fig. 74.1. A 39-year-old woman with CARASIL. The MRI was Over time, the white matter becomes increasingly
obtained 17 months after the onset of neurological symp- atrophic (Fig. 74.2).
toms. There are symmetrical abnormalities in the cerebral The pattern of confluent periventricular and deep
hemispheric white matter and both internal and external cap- white matter involvement in combination with multi-
sules. The U fibers are partially spared. The corpus callosum is focal small lesions of the basal ganglia, thalami, and
intact. There are small additional lesions in the basal ganglia brain stem is typically the pattern seen in vascular
and brain stem.The diffusion-weighted image (first row,middle disorders. However, the white matter abnormalities
image) shows a small fresh infarction.The FLAIR images (fourth are more homogeneously confluent than in Bins-
row) reveal many small lacunae. From Yanagawa et al. (2002), wanger disease. Unlike in CADASIL, there are no
with permission; additional images courtesy of Dr. S. Yana- petechial hemorrhages.
gawa, Department of Neurology, and Dr. S. Ikeda,Third Depart-
ment of Medicine, Shinsu University School of Medicine, Mat-
sumoto, Japan
Chapter 75

Polycystic Lipomembranous Osteodysplasia


with Sclerosing Leukoencephalopathy
(Nasu–Hakola Disease)

75.1 Clinical Features 75.2 Pathology


and Laboratory Findings
On gross inspection, the brain shows atrophy of vari-
Polycystic lipomembranous osteodysplasia with able severity, especially in the frontal and temporal
sclerosing leukoencephalopathy (PLOSL), also called regions. The large cerebral arteries at the base of the
Nasu–Hakola disease, was described in the early brain appear normal. On coronal sectioning, the ven-
1970s by Nasu et al. (Japan) and Hakola (Finland). tricles are enlarged, again predominantly in the
The disorder was recognized as a combination of frontal and temporal lobes. There is a decrease in
abnormalities in the nervous system and adipose tis- white matter volume. The white matter appears gray-
sue in the skeleton. Initially most cases were reported ish and is of rubbery consistency. The basal ganglia
from Japan and Finland, but over the years the disease and thalami are smaller than normal. The globus pal-
has been reported on a more global scale, with papers lidus shows a brownish discoloration and contains
from other Nordic countries, Italy, and Belgium to deposits of sandy material.
North America and South Africa. There is no doubt Microscopy shows a diffuse, symmetrical myelin
that other countries will follow. The disease has an pallor and marked astrofibrillary gliosis of the cere-
autosomal recessive mode of inheritance. Patients are bral white matter, predominantly in the frontal and
often born of consanguineous parents. temporal region, but also in the corpus callosum, in-
The initial development of patients is normal. The ternal capsule, and sometimes cerebellar white mat-
first symptoms develop in the second or third decade ter. The arcuate fibers are better spared in the tempo-
of life and are mainly related to the skeletal abnor- ral than in the frontal regions. There is little su-
malities. The disorder affects especially the distal danophilic material in perivascular macrophages, al-
parts of the long cancellous bones, metacarpals, though sometimes sudanophilic material is more
metatarsals, and phalanges. The first symptoms are prominent and diffusely scattered in the affected
painful wrists and ankles, distortions and fractures, white matter. No metachromatic material is found.
either occurring spontaneously or induced by minor There are no signs of inflammation. Signs of axonal
trauma. In the third decade a slowly progressive de- degeneration may be seen within the cerebral hemi-
mentia starts to develop, with the characteristics of a spheric white matter, basal ganglia, cerebellum, and
prefrontal psychological syndrome, including per- brain stem, with axonal loss, fragmentation, and pres-
sonality changes, euphoria, loss of social inhibitions, ence of spheroid bodies. Electron microscopy of these
memory loss, and confabulations. Gradually other axonal spheroids reveals a compact collection of cell
symptoms appear. Seizures enter the clinical picture, organelles, including degenerated mitochondria,
in addition to gait disturbances, paraplegia, chor- neurofilaments, and vesicles with patchy dense mate-
eiform movements, myoclonia, and urinary inconti- rial. The presence of these axonal spheroids may sug-
nence. In the last phase of the disorder, cortical in- gest a link to leukoencephalopathies with axonal
volvement becomes manifest with manifestations spheroids. The cerebral cortex is usually intact. The
such as aphasia, agraphia, acalculia, and alexia. Bul- thalamus is rarely seriously affected. In the globus
bar symptoms may develop as well. The prognosis is pallidus and putamen, basophilic calcospherites and
poor; the average life expectancy is 40 years. Howev- moderate neuronal loss are seen. There are changes in
er, patients with a slower and more benign disease the blood vessels (small arterioles and capillaries),
course have also been reported. most prominently in the white matter. The vessels
Laboratory findings may include an increased have plump endothelium. The basement membranes
alkaline phosphatase level. Skeletal X-rays reveal of these blood vessels are thickened and often multi-
multiple cystic lesions symmetrically located in the plied, related to concentric deposition of collagen IV.
metatarsal areas of long bones, the phalanges, Immunohistochemically extravasation of plasma
metacarpals, and metatarsals. Biopsy of the bone constituents can be demonstrated.
shows the characteristic histological membranocys- Peripheral nerves may be affected and axonal de-
tic changes – lesions that can also be found in subcu- generation has been reported.
taneous adipose layers. DNA confirmation of the In addition to the brain, adipose tissue and bone
diagnosis is possible. marrow are of interest. In adipose tissue and fatty
75.5 Magnetic Resonance Imaging 553

bone marrow, membranocystic lesions are found. The TYROBP-mediated signaling pathway plays an
Convoluted membranous structures lie scattered or important role in human brain and bone tissue. The
conglomerated among mature fat cells. The mem- pathogenesis of the disease has still to be clarified.
branes, which are mainly eosinophilic but partly ba- The precise biochemical and structural steps leading
sophilic, form irregular cystic cavities containing to the histopathology of the disease are not yet under-
pale, homogeneous material. Ultrastructurally, the stood. Patients with PLOSL have no defects in cell-
membranes consist of undulating bands composed of mediated immunity, suggesting a remarkable capaci-
numerous minute tubular structures, arranged per- ty of the human immune system to compensate for
pendicularly to the inner surface of the cavity. The the inactive TYROBP-mediated activation pathway.
cavity is filled with a lightly osmiophilic amorphous The characteristic bone cysts may reflect chronic dys-
substance. The outer zone of the bands shows tubular function of osteoclasts.
crypts with microvesicles. The tubulovesicular struc-
ture of the membranocystic lesions arises at the
lipid–cytoplasmic interface of degenerated fat cells 75.5 Magnetic Resonance Imaging
and enters the interstitium to form the lesion, concur-
rent with the collapse of fat cells. There are also mem- The combined findings of X-rays of the skeleton and
branocystic structures with thinner membranes CT and MRI of the brain are diagnostic. X-rays of
without tubular structures. Membranocystic struc- the long bones, phalanges, and metacarpal and
tures are not specific for PLOSL. They can be induced metatarsal bones show the typical cystic changes, in
by several forms of chronic circulatory insufficiency many cases with evidence of earlier fractures. CT
and trauma. shows calcification of the basal ganglia, especially in
the putamen and caudate nucleus (Figs. 75.1 and
75.2), sometimes also in the globus pallidus, rarely in
75.3 Chemical Pathology the dentate nucleus (Fig. 75.2). The abnormalities in
the white matter are better demonstrated on MRI.
Lipid analysis of brains of patients with PLOSL has re- MRI shows bilateral, symmetric, cerebral white
vealed large amounts of free fatty acids, but no cho- matter abnormalities with a high signal intensity on
lesterol esters. Within the white matter, total lipid, T2-weighted and FLAIR images (Figs. 75.2–75.4). The
cholesterol, and cerebroside content are mildly to se-
riously decreased.

75.4 Pathogenetic Considerations

One gene for PLOSL is TYROBP, also called DAP12,


located on chromosome 19q13.1. The gene encodes
TYRO protein tyrosine kinase binding protein
(TYROBP), also called DAP12. TYROBP is an adaptor
molecule that couples a variety of cell surface recep-
tors expressed by myeloid cells, including natural
killer cells, and plays a major role in the transduction
of cell activation signals. It is expressed by a wide va-
riety of myeloid cells and also by microglia and osteo-
clasts.On the plasma membrane of these cells,TYROBP
associates with activating receptors recognizing major
histocompatibility complex class I molecules.
More recently, TREM2 has been identified as the
second PLOLS gene. The gene is located on chromo-
some 6p21.2. The protein TREM2 belongs to the im-
munoglobulin superfamily and forms a receptor sig-
naling complex with TYROBP. Its pattern of expres-
sion closely follows the pattern of TYROBP. It triggers
activation of immune responses in macrophages and Fig. 75.1. Female PLOSL patient, 38 years old. The CT scan
monocyte-derived dendritic cells. PLOSL forms an demonstrates the calcium deposits in the basal ganglia. Cour-
example of a disease in which mutations in two differ- tesy of Dr.Y. Ueki, Dr. N. Kohara, Dr. H. Fukuyama, and Dr.Y. Miki,
ent subunits of a multisubunit receptor complex re- Departments of Neurology and Brain Pathophysiology, Facul-
sult in an identical phenotypic appearance. ty of Medicine, Kyoto University, Kyoto, Japan
554 Chapter 75 Polycystic Lipomembranous Osteodysplasia with Sclerosing Leukoencephalopathy (Nasu–Hakola Disease)

Fig. 75.2. Male PLOSL patient, 29


years old.The CT scan demonstrates
the calcium deposits in the caudate
nucleus and dentate nucleus.The MRI
reveals abnormalities in the periven-
tricular and deep white matter,
sparing the U fibers.The hilus of the
dentate nucleus and deep cerebellar
white matter is affected as well.
From Malandrini et al. (1996),
with permission

signal change is often mild and ill demarcated. The 75.4). There is progressive cerebral atrophy with
temporal lobe tends to be better preserved (Figs. 75.3 widening of the ventricles and subarachnoid spaces
and 75.4). The U fibers also tend to be spared (Figs. 75.3 and 75.4). The atrophy may dominate in
(Figs. 75.2–75.4). On T2-weighted, but more conspicu- the frontal area. Perfusion studies have been per-
ously on gradient-recalled-echo images, the basal formed with SPECT and PET with evidence of hypo-
ganglia appear too dark in comparison with average perfusion in the frontal and temporal lobes.
normal findings in this age group (Figs. 75.3 and
75.5 Magnetic Resonance Imaging 555

Fig. 75.3. A 32-year-old man with PLOSL.The T2-weighted im- intensity of the putamen and globus pallidus, related to calci-
ages reveal diffuse, ill-defined signal abnormalities in the cere- um deposits, as evident on the CT scan (not shown). Courtesy
bral white matter, relatively sparing the U fibers and temporal of Dr. T. Autti, Department of Radiology, Helsinki University
white matter. There is some cerebral and cerebellar atrophy Central Hospital, Helsinki, Finland
with moderate widening of CSF spaces. Note the low signal
556 Chapter 75 Polycystic Lipomembranous Osteodysplasia with Sclerosing Leukoencephalopathy (Nasu–Hakola Disease)

Fig. 75.4. Brother of the patient shown in Fig. 75.3, also suffer- sity of the basal ganglia on these T2-weighted images. Cour-
ing from PLOSL, 32 years old. Despite being the same age at tesy of Dr. T. Autti, Department of Radiology, Helsinki Universi-
MRI, this patient has much more severe cerebral atrophy and ty Central Hospital, Helsinki, Finland
also some cerebellar atrophy. Note again the low signal inten-
Chapter 76

Pigmentary Orthochromatic Leukodystrophy

76.1 Clinical Features with relative sparing of the U fibers. The frontal white
and Laboratory Investigations matter is most prominently affected. Axons are rela-
tively spared, but axonal swellings may be seen in the
A limited number of families with pigmentary affected white matter. The brain stem and cerebellar
orthochromatic leukodystrophy (POLD) have been white matter are inconstantly and mildly affected.
described. In several families affected siblings were Wallerian degeneration may be seen in brain stem
observed, whereas the parents did not display signs of tracts. There are scanty macrophages containing
dementia, providing evidence for an autosomal reces- sudanophilic material. Astrocytes, microglia, and
sive mode of inheritance. However, occurrence of pa- macrophages contain a brown-yellow, autofluores-
tients in successive generations has also been report- cent pigment in the cytoplasm that stains positive
ed, suggesting an autosomal dominant inheritance. with PAS. The inclusions have the staining properties
Autosomal dominant inheritance with incomplete of ceroid lipofuscin. Macrophages and astrocytes are
penetrance would explain the two observations. The inconstantly positive in iron stains. The demyelinated
mean age at onset of symptoms is 42 years ± 11.5 white matter is diffusely gliotic. Oligodendrocytes are
(range 25–70 years).Women account for two-thirds of markedly decreased in areas of demyelination, but
the patients. Childhood onset of POLD has been re- preserved or relatively increased in numbers in the
ported occasionally (Seiser et al. 1990; Harding et al. less severely affected areas. There are no inflammato-
1990). ry infiltrates.
Most patients present with dementia and behav- Electron microscopy of the white matter shows
ioral disturbances. The disease is chronic-progressive that most macrophages and many astrocytes and
and is characterized by progressive cognitive decline, oligodendrocytes are filled with membrane-bound
spastic hemiplegia or tetraplegia, cerebellar ataxia, intracytoplasmic inclusions containing electron-
and pseudobulbar features with dysphagia, dys- dense multilamellar material in a curved or straight
arthria, and finally loss of speech. Myoclonia and parallel arrangement or with fingerprint profiles,
epilepsy are also relatively frequent findings. Optic accompanied by various amounts of lipofuscin and
atrophy and visual loss may occur. The duration of lipid droplets. Curvilinear profiles may also be ob-
the illness is 7 ± 5.5 years, with a range of 1–18 years, served.
indicating that the rate of progression is highly vari-
able. Two sisters with POLD had primary infertility
with streak ovaries at autopsy. 76.3 Pathogenetic Considerations
The diagnosis is at present based on histopatho-
logical findings. When the diagnosis has been estab- At present, the basic genetic defect and the pathogen-
lished by means of histopathology in one patient, esis of the disease are unknown. One paper (Marotti
MRI of the brain revealing a leukoencephalopathy is et al. 2004) reports a father and two daughters with
sufficient to confirm the diagnosis in family members adult-onset leukoencephalopathy and at autopsy
who become clinically symptomatic. widespread destruction of cerebral white matter with
presence of neuroaxonal spheroids as well as pig-
mented glial cells and macrophages. So in this family
76.2 Pathology histology revealed characteristics of both POLD and
hereditary diffuse leukoencephalopathy with spher-
External examination reveals mild to marked atrophy oids (HDLS, Chap. 70), suggesting perhaps an etio-
of the cerebrum and cerebellum. On sectioning, logical relationship between the two.
dilatation of the lateral ventricles is found, in addition
to gray-brown discoloration of the cerebral white
matter, most marked in the frontal and periventricu- 76.4 Therapy
lar regions.
Microscopic examination reveals prominent, sym- Only symptomatic therapy is available for patients
metrical demyelination of the cerebral white matter with POLD.
558 Chapter 76 Pigmentary Orthochromatic Leukodystrophy

76.5 Magnetic Resonance Imaging matter volume loss. The corpus callosum was also
atrophic. The white matter abnormalities were
MRI has been reported in one patient (Dousset et al. bilateral but not entirely symmetrical. The MRI find-
1998) and revealed extensive signal abnormalities ings are not distinct and do not allow a specific
in the cerebral white matter, accompanied by white diagnosis.
Chapter 77

Adult-Onset Autosomal Dominant


Leukoencephalopathies

Multiple families have been described with an adult ly stages. The abnormalities are patchy and inhomo-
onset leukoencephalopathy and an autosomal domi- geneous in signal intensity. The deep white matter is
nant mode of inheritance. When there is no specific most prominently affected. The cerebral white matter
disease marker, it is hard to tell whether families have abnormalities are progressive and extend posteriorly.
the same disease or different disorders. In hereditary They finally involve all cerebral white matter with rel-
diffuse leukoencephalopathy with spheroids (see ative preservation of the U fibers. Signal abnormali-
Chap. 70), the white matter disease is histopathologi- ties in the middle cerebellar peduncles are early find-
cally accompanied by numerous neuroaxonal spher- ings. In the later stages, the cerebellar white matter is
oids within the affected white matter, which is the also affected. Brain stem abnormalities are frequent,
basis for the diagnosis. In pigmentary orthochromat- including involvement of the pyramidal tracts, medi-
ic leukodystrophy (see Chap. 76), histopathological al lemniscus, and cerebellar peduncles.
examination reveals demyelination accompanied by Autopsy reveals widespread myelin loss in isolated
astrocytes and phagocytic cells that contain in their and confluent patches in the cerebral white matter
cytoplasm membrane-bound inclusions of brown- and relative preservation of axons within the affected
yellow, autofluorescent pigment, which has the ap- areas. Irregular islands of relatively normal white
pearance of multilamellar material in a curved or matter are seen within and at the margins of the af-
straight parallel arrangement or with fingerprint pro- fected regions. In the areas of myelin loss, oligoden-
files on electron microscopy. However, in other fami- drocytes are abundant. The abnormal white matter is
lies with adult-onset autosomal dominant leukoen- vacuolated. Despite the severity of the white matter
cephalopathy such characteristic histopathological abnormalities, macrophages, activated microglia,
findings are lacking. In the present chapter we focus lipid accumulation, astrocytic proliferation, and fib-
on those families in which MRI findings have been re- rillary gliosis are scarce. Inflammatory infiltrates are
ported. absent. The U fibers are relatively spared. Gray matter
Adult-onset autosomal dominant leukoencephalo- structures are intact. The cerebellar white matter and
pathy has been described in multiple members cerebellar peduncles are severely affected, similar to
of an American-Irish family (Eldridge et al. 1984; the cerebral white matter. Brain stem tracts are also
Schwankhaus et al. 1988, 1994; Coffeen et al. 2000). In affected, but the changes are milder than in the cere-
this family, the onset of clinical symptoms is in the bral and cerebellar white matter.
fourth or fifth decade of life. The first symptoms con- Calandriello et al. (1992) describe a family with an
sist of autonomic abnormalities, including bowel and autosomal dominant leukoencephalopathy and a
bladder dysfunction, impotence, orthostatic hypoten- highly variable age at onset, ranging between 9 and 66
sion, and decreased sweating. After several years, years, making evaluation of this family difficult.
other neurological symptoms appear, such as loss of Details concerning MRI findings are lacking.
fine motor skills. Subsequently, overt cerebellar and Abe et al. (1993) describe a family with adult-onset
pyramidal dysfunction develops, eventually leading autosomal dominant leukoencephalopathy and clini-
to complete loss of voluntary movements. Signs of cally progressive tetraparesis, dysarthria, dysphagia,
posterior column dysfunction are frequent. Behav- and urinary incontinence. Mental capacities seem to
ioral problems, cognitive deficits, and abnormalities be relatively better preserved. MRI shows diffuse sig-
of the central visual pathways are mild and of later nal abnormalities of the cerebral white matter, brain
onset. Sensorineural hearing loss is common. The stem tracts, and cerebellar white matter. A striking
peripheral nervous system is spared. The disease is finding is the preservation of the optic radiation, con-
slowly progressive, and survival of 20 years is com- sidered to be a distinct finding.
mon. Laboratory tests are unrevealing. Evoked poten- Quattrocolo et al. (1997) and Bergui et al. (1997)
tials show central conduction delays. Peripheral nerve describe a large Italian family with adult-onset auto-
conduction velocities are normal. A gene locus on somal dominant leukoencephalopathy and clinically
chromosome 5q31 has been identified, but the gene progressive spasticity, pseudobulbar dysfunction,
itself has not yet been found. urinary incontinence, and sometimes action tremor.
Neuroimaging shows cerebral white matter Apart from slight memory impairment, no cognitive
changes with frontoparietal predominance in the ear- decline is noted and there are no behavioral changes.
560 Chapter 77 Adult-Onset Autosomal Dominant Leukoencephalopathies

The peripheral nervous system is spared. The mean pathy. The disease is characterized by cerebellar
age at onset is 45 years and the duration of the disease ataxia as the initial symptom and later dementia and
10 years. MRI of the brain shows diffuse signal abnor- signs of pyramidal dysfunction. MRI shows a diffuse
malities within the cerebral white matter, most leukoencephalopathy involving all cerebral and cere-
prominently involving the deep cerebral white matter. bellar white matter and the brain stem. In addition,
The U fibers and corpus callosum are initially spared some cerebral atrophy develops. Histopathological
but become involved in the later stages of the disease. examination reveals vacuolar degeneration of the
Some cerebral hemispheric atrophy develops over white matter, sparing the U fibers.
time. The posterior limb of the internal capsule and In 2003, another family with adult-onset auto-
brain stem tracts become affected. The cerebellum somal dominant leukoencephalopathy was reported
and the optic radiation remain spared, even in the late by Letournel et al. The patients have a variable combi-
stages of the disease. There are no data on histo- nation of dementia, motor signs, and epilepsy.
pathology. MRI findings have not been reported in detail and
Fukazawa et al. (1997) describe a family with an histopathological finding are nonspecific, showing an
autosomal dominant leukoencephalopathy. The onset orthochromatic leukoencephalopathy involving the
of clinical symptoms in this family is in childhood, cerebral hemispheres, but sparing the cerebellum and
but the progression is extremely slow, over decades. brain stem.
The first signs include cerebellar dysfunction and It is hard to tell whether consistent differences in
some mental deterioration. The subsequently devel- clinical and MRI findings are sufficient to distinguish
oping spasticity leads to serious disability in the third different disease entities. For instance, in some fami-
or fourth decade of life. The MRI shows diffuse white lies signs of autonomic dysfunction are consistently
matter signal abnormalities, involving all cerebral present, whereas they are absent in other families.
white matter from cortex to ventricular lining and Likewise, the cerebellar white matter is consistently
also the internal capsule and corticospinal tracts in involved in some families, whereas in other families
the brain stem. The cerebellum and remainder of the cerebellum is consistently spared. It is presently
the brain stem have a normal signal, although the important to document these families in every detail
cerebellum shows some atrophy. The findings may from clinical, MRI, MRS, and histopathological per-
be compatible with hypomyelination, but data on spectives. It is to be expected that the underlying
histopathology are lacking. gene defect(s) will soon be found in the larger fami-
In 2001 Tagawa et al. described another family with lies, facilitating further analysis of the remaining
an adult-onset autosomal dominant leukoencephalo- families.
Chapter 78

Inflammatory and Infectious Disorders

Although apparently different with respect to the un- eign cells. Two ways of activating complement are dis-
derlying cause, inflammatory and infectious diseases tinguished. One is the so-called alternative pathway,
have much in common. In both conditions the human which is activated by a microbial polysaccharide. This
defense system plays an active role, counteracting the pathway belongs to the innate, nonspecific immune
cause of the disturbance, killing the intruder, and system. The other, the classical pathway, is a specific
repairing the tissue damage. response triggered by antibody–antigen complexes.
Inflammation has principally a protective role. Both systems work closely together.
Paradoxically, in a large number of human diseases Acute phase proteins are plasma proteins which
inflammatory reactions are part of the pathogenetic undergo a dramatic increase in concentration in
mechanisms causing tissue damage. Inflammatory response to infection or injury. A number of factors
reactions can be not only protective but also histotox- belong to this group, including C-reactive protein
ic. The inflammatory process is characterized by a (CRP) and fibrinogen. C-reactive protein has the abil-
complex interplay between blood cells, blood vessels, ity to bind to a number of micro-organisms which
and tissue. Although the process is complex, it follows contain phosphorylcholine on their membranes. The
well-orchestrated patterns. complex formed is able to activate complement by the
Many components of the defense system have to classical pathway, resulting in opsonization of the
come into action to destroy the invading micro-or- microbe for adherence to phagocytes.
ganism or counteract adverse interactions. The de- Interferons are broad-spectrum antiviral agents.
fense system depends largely on the immune system, Different molecular forms of interferon have been
which can be subdivided into an innate (nonadap- identified. There are at least 14 different interferons-
tive) and an acquired (adaptive) part. Repeated infec- a (IFN-a), produced by leukocytes, while fibroblasts
tions do not improve the innate system, whereas the and other cells produce interferon-b (IFN-b), inter-
adaptive immune system has a “memory bank” with feron-g (IFN-g), and macrophage activating factors,
improved resistance after repeated attacks. In both which switch on the microbicidal mechanisms of the
systems a molecular (solvable, humoral) component macrophage. A subpopulation of T lymphocytes, T
can be distinguished from a cellular component. helper cells, will produce lymphokines, amongst them
In the innate system the soluble factors are lyso- IFN-g, if bound to an antigen in association with a
zyme, complement, acute phase proteins, and interfer- major histocompatibility complex (MHC) class II
on. molecule on a macrophage surface. IFN-g is also pro-
The enzyme lysozyme is a soluble bactericidal sub- duced by cytotoxic T lymphocytes which recognize
stance, abundantly present in body fluids. It is capable antigen in association with MHC class I molecules.
of splitting the exposed peptidoglycan wall of suscep- The major histocompatibility complex (MHC) is a
tible bacteria. complex of proteins which is of predominant impor-
Complement consists of a complex series of many tance in the transplant rejection process. In man the
proteins, and forms one of the enzyme systems in MHC is the human leukocyte antigen (HLA) cluster
plasma triggered during inflammation. Once activat- encoded for by chromosome 6. It has been recognized
ed, the complement system produces a rapid, highly that proteins encoded in this particular region of
amplified response to a trigger stimulus, mediated by chromosome 6 are involved in many aspects of im-
a cascade reaction, in which the product of one reac- munological recognition, including both interaction
tion is the enzyme catalyst of the next reaction. Com- between lymphoid cells and interaction between lym-
ponents of complement are designated by the letter phocytes and antigen presenting cells. Three classes
“C” followed by a number related to the chronology of of MHC molecules are distinguished. Class I mole-
its discovery. Unfortunately, the sequence in which cules associate with antigen on the surface of virally
the complement components come into action is not infected cells to signal cytotoxic T lymphocytes. Class
identical to the sequence of their discovery. The ac- II molecules signal T helper cells to activate B cells
tions of activated complement range from increasing and macrophages. T helper cells are only activated
vascular permeability to mast cell degranulation, op- when both antigen and MHC class II molecules are
sonization and phagocytosis of bacteria, neutrophil presented on the antigen-presenting cell. Class III
activation, chemotaxis, and lysis of bacteria and for- genes encode components of the complement.
562 Chapter 78 Inflammatory and Infectious Disorders

The cellular part of the innate immune system is on triggering. This triggering occurs by components
formed by phagocytes, natural killer cells, and mast of the complement system. Mast cell activation fol-
cells. lows two major pathways. The first pathway involves
The phagocytes can be divided into monocytes and the release of the preformed mediators from the
polymorphonuclear granulocytes. The latter can be granules, such as histamine, causing vasodilatation,
further divided into neutrophils, eosinophils, and increased capillary permeability, and chemokinesis;
basophils, according to the histological staining of neutral proteases, which activate complement;
their granules. Granulocytes do not show any speci- eosinophil and neutrophil chemotactic factor; and
ficity for antigens, but together with antibodies and platelet activating factor, which induces mediator re-
complement, they play an important role in protec- lease, including interleukins and tumor necrosis fac-
tion against micro-organisms. Their predominant tor with multiple actions such as macrophage activa-
role is phagocytosis. In the process of phagocytosis tion and triggering of acute phase proteins. The sec-
both oxygen-dependent and oxygen-independent ond pathway results in the release of newly synthe-
mechanisms play a role. The oxygen burst is the sized mediators via the phospholipase A2 pathway.
process in phagocytosis by which oxygen is convert- This enzyme initiates arachidonic acid degradation.
ed to free radicals. Breakdown of arachidonic acid is achieved by differ-
Oxygen radicals and their metabolites constitute ent enzymes. Lipoxygenase leads to the promotion of
an important class of inflammatory mediators. Free leukotrienes, which influence the microcirculation
radicals are atoms or molecules containing an un- and enhance chemotaxis. Cyclo-oxygenase leads to
paired electron in the outer orbit. Phagocytic cells, the formation of prostaglandins and thromboxanes,
when activated, exhibit a sharp increase in molecular which affect bronchial muscle, platelet aggregation,
oxygen consumption, the so-called respiratory burst, and vasodilatation.
and produce a battery of biologically active oxygen Complex and efficient as the innate immune sys-
radicals and derived metabolites. The stimuli for tem appears, many micro-organisms find ways to cir-
phagocyte-derived oxygen radical production are cumvent these responses. Here the adaptive immune
multiple, including endotoxins, g-immunoglobulins, system is important, which also has humoral and cel-
and cytokines. Reduction of molecular oxygen by lular components. The humoral adaptive immune sys-
addition of a single electron results in formation of tem leads to the formation of antibodies, which form
the superoxide anion (O2•). In aqueous environments, specific responses to specific antigens. Antibodies
O2• exists in equilibrium with its protonated form, form a highly specific answer to those micro-organ-
H2O• (perhydroxyl radical). In the presence of super- isms which escape the innate immune system. Anti-
oxide dismutase O2• can undergo dismutation, result- bodies are capable of binding specifically to the at-
ing in H2O2 formation. Hydrogen peroxide has the ca- tacking microbe, activating the complement system,
pacity to oxidize directly a wide spectrum of biologi- and stimulating phagocytic cells. The antibody mole-
cal molecules. Phagocyte-derived oxygen radicals cule therefore has three main regions, two regions
and their metabolites are important mediators of tis- concerned in communicating with complement and
sue damage in inflammatory disorders. The concur- phagocytes, and one region for binding to an individ-
rent rise in pH due to free radical formation allows ual micro-organism. The latter region carries the ex-
cationic proteins to function optimally, damaging the ternal recognition function. The first two functions
bacterial membrane. The process of phagocytosis is are constant. The recognition function, however, de-
adequately supported by the complement system, mands numerous adaptations of recognition sites.
which can prepare micro-organisms for phagocytosis Antibodies, when bound to a microbe, will link to the
by opsonization. first molecule in the classical complement sequence.
Natural killer cells are large granular lymphocytes The cells involved in the adaptive immune system
with a characteristic morphology, capable of recog- are mainly lymphocytes. All the cells of the immune
nizing structures on high-molecular-weight glyco- system are derived from pluripotent stem cells
proteins which appear on the surface of virally infect- through two main lines of differentiation: the lym-
ed cells and allow them to be differentiated from un- phoid lineage, producing lymphocytes, and the
infected cells. At the binding site, pores are forced in myeloid lineage, producing phagocytes and mast
the infected cell and the cell is then killed by nuclear cells. There are three kinds of lymphocytes with dif-
fragmentation through calcium-dependent endonu- ferent functions: T cells and B cells and the so-called
clease. The various interferons produced by virally in- third population cells. T cells differentiate initially in
fected cells augment natural killer cytotoxicity and the thymus; B cells in fetal liver, spleen, and in adult
form an integrated feedback defense system. bone marrow. Morphologically, T and B cells are iden-
Mast cells have a central role in the acute inflam- tical; functionally they can be distinguished. B cells
mation process. They are loaded with granules con- are classically defined by the production of im-
taining preformed mediators, which are released up- munoglobulins (antibodies), which are presented on
Inflammatory and Infectious Disorders 563

the cell surface. The T cells can be subdivided into T start to proliferate. The antigen-presenting cells in the
helper cells, T suppressor cells, and T killer cells. Their CNS may be perivascular macrophages, the gatekeep-
function is to recognize foreign intruders, to activate ers of the CNS. Microglial cells may also act as anti-
B cells, and to kill invading micro-organisms. The gen-presenting cells within the CNS. Sensitized or
third population cells do not consistently carry mark- activated T cells start to release substances such as
ers of either T or B cells. They possess specific recep- interferons (interferon-g), which activate local cells
tors for g-immunoglobulins and form the greater part such as microglia, astrocytes, and perivascular
of natural killer and antibody-dependent cellular macrophages. This local activation results in upregu-
cytotoxic effectors. In another classification system, lation of adhesion molecules on endothelial cells,
dependent on the surface proteins reacting to clusters enhanced expression of major histocompatibility
of antigens (CD = cluster determinants), lympho- complex, and release of chemoattractive substances,
cytes can be subdivided in terms of different cell which attract and facilitate the entrance of more T
types. The T helper cells are classified as CD4+ cells cells, macrophages, and B lymphocytes. B cells are
(formerly T4 cells); the T suppressor/killer cells as triggered to become plasma cells and produce anti-
CD8+ cells (formerly T8 cells). bodies. Macrophages, microcytes, astrocytes, and
To orchestrate the actions of all the cells involved mast cells start to produce inflammatory mediators
in the inflammatory process, a great variety of solv- such as tumor necrosis factors, reactive oxygen
able mediators are used for communication between species, interleukins, vasoactive amines, leukotrienes,
the cells. On the effector side, too, many solvable complement, and hydrolytic enzymes. The result is,
factors play an important role in the inflammatory eventually, vascular dilatation, increased capillary
reaction. An important group of such mediators has permeability, exudation of fluid in tissue, and ex-
been given the name cytokines. Cytokines can be di- travasation of numerous cells, leading subsequently
vided into monokines, produced by monocytes and to disruption of the blood–brain barrier, perivascular
macrophages, and lymphokines, produced by lym- inflammation, formation of edema and demyelina-
phocytes. Cytokines are hormone-like substances tion. Phagocytosis of myelin by macrophages may be
and are the most important mediators of the action of initiated by antimyelin antibodies, or by opsonization
T lymphocytes. The immune functions of T lympho- via their receptor for complement component.
cytes are reflected by a specific set of cytokines, the Knowledge of the factors mediating inflammation
lymphokines, produced by these cells after activation. has stimulated attempts to undo undesired reactions
Subpopulations of T cells, such as T1 and T2 helper by counteracting or blocking the involved pathways.
cells, can be distinguished by the specific lym- Interferon-g, for example, proved to have a negative
phokines they produce. Important lymphokines, influence on the course of secondary progressive
some of which have been mentioned before, are inter- multiple sclerosis. Interferon-b, known to counteract
leukins, interferons, lymphotoxin, growth factors, and the action of interferon-g, seems to have a beneficial
tumor necrosis factor. CD4+ cell has the cytokine effect on the course of this disease.
profile of the T1 helper cell. The T1 helper cell acti- In infectious disorders several pathogenetic factors
vates the B lymphocytes to produce antibodies and it play a role in evoking the host response. Exotoxins, for
causes eosinophilia and mast cell hyperplasia. The example, are produced by micro-organisms during
CD8+ lymphocyte has T2 cell characteristics and is their growth. They have a proteinaceous nature and
responsible for cytotoxicity, complement fixation, therefore act as antigens. They evoke an antibody
and macrophage activation. The CD8+ lymphocyte response and in this way give rise to neutralizing
profile, therefore, protects against viruses, tumors, antitoxins. Endotoxins are membrane molecules,
and infections. It is also responsible for transplant re- lipopolysaccharides, present in the outer cell mem-
jection, inflammation, and autoimmune reaction. Cy- brane of gram-negative bacteria. The polysaccharide
tokines form a functional network. The various fac- chain is variable and determines the antigenic struc-
tors, messengers, and effectors act in a subtle interac- ture of the bacteria. The lipid fraction anchors the
tion. These interactions may be synergistic, additive, lipopolysaccharide in the outer bacterial membrane,
or antagonistic, and function in order to maintain a is less variable, and is responsible for the toxic effects.
balance of optimal efficacy in the protection of the After liberation of the lipopolysaccharides from the
host. bacterial membrane they adsorb on the lipid mem-
The cooperative cellular and mediator activity in brane of many cells, especially neutrophils. This trig-
inflammatory (either infectious or noninfectious) dis- gers a whole cascade of immune reactions. Another
orders of the CNS can be briefly summarized as fol- way to evoke immune reactions is by presentation of
lows: antigens by specialized cells, also called antigen-pre-
Circulating T cells recognize the antigen con- senting cells, such as the Langerhans and dendritic
cerned in combination with the major histocompati- cells. These cells recognize the foreign intruders,
bility complex class II on antigen-presenting cells and probably by lack of identifying cell surface membrane
564 Chapter 78 Inflammatory and Infectious Disorders

MHC class I compounds. Viruses use the replication For many viruses a necessary condition for the
system of host cells and bring their antigens to ex- entry into cells is the availability of specific receptor
pression on the surface of the host cells, activating molecules at the surface of such cells. The distribu-
natural killer cells. tion of these receptor molecules will largely deter-
The presence and multiplication of the original mine the specific regions of the brain preferentially
micro-organism sustains the disease or expands it. attacked by the virus and the population of cells in the
The micro-organisms produce a gamut of products affected regions that are injured or destroyed. Some
to assist in their spread, including streptokinase, viruses like JC papovavirus, responsible for progres-
hyaluronidase, and neuraminidase. In many cases it is sive multifocal leukoencephalopathy, infect oligoden-
difficult to distinguish the direct influence of the mi- drocytes with sparing of the neurons. This explains
cro-organisms on the tissue from the immunological the affinity of this virus for white matter. Rabies virus
reaction the attack provokes. A demonstration of this has great affinity for the cerebellum and limbic sys-
is found in subacute sclerosing panencephalitis, tem, whereas poliomyelitis affects the motor nuclei in
caused by the measles virus. In subacute sclerosing the cortex, brain stem, and anterior horns of the
panencephalitis, as in other persistent viral infec- spinal cord. Creutzfeldt–Jacob disease, caused by pri-
tions, deposits of immune complexes can be demon- ons, has been shown by MRI to affect in particular the
strated in the wall of small cerebral blood vessels. The basal ganglia. These differences in topographical dis-
presence of immune complexes and the histological tribution are considered to be the result of specific
findings of perivascular edema, inflammation, and receptor interactions.
demyelination provide strong evidence that, in addi- Viruses enter the cells of the host either by fusing
tion to the actual invasion by the virus, there is a con- with the plasma membrane and discharging their
temporaneous immune-mediated response to this contents directly in the cytosol, or by being internal-
virus which is responsible for much of the tissue dam- ized through the endocytotic pathway. The first way is
age. The same could be true for subacute AIDS en- used by herpes simplex and corona viruses. Internal-
cephalitis and tropical spastic paraparesis due to ization does not have special requirements in these
HTLV-1 infection. cases. Other viruses do not have this option and have
Selective neuronal and/or glial damage in infec- to follow the other path, involving binding to a recep-
tious disorders and the predilection for particular ar- tor and collection of receptor–ligand complexes at
eas (topistic areas) is in most cases difficult to under- the cell membrane. The viruses are then transported
stand. Experience has taught that herpes simplex within endocytotic vesicles. After penetration of the
virus has an affinity for the frontal and temporal cell membrane, the replicative machinery of the virus
lobes; fungal infections attack primarily gray matter itself comes into play. Using the host cell system of
structures; Haemophilus influenzae has a preference nucleic acid (DNA/RNA) replication and transcrip-
for cortico-subcortical areas; cytomegalovirus (CMV) tion, viruses can replicate and transcribe their DNA
encephalopathy and AIDS encephalopathy in adults or RNA and in so doing multiply themselves; the new
are predominantly located in the cerebral white mat- virus particles leave the cell in search of new host
ter and spare the U fibers. There is often a consider- cells. Ultimately the balance between the viral ma-
able difference between the course of a disease and chinery and the host immune system will determine
the predilection sites between congenital infections the outcome of the infection.
and infections acquired later, after birth. A good ex- In the noninfectious inflammatory disorders other
ample is the difference between congenital, connatal, mechanisms play a role in invoking the host response,
and adult CMV infection, and between congenital and not all of which are well understood. The inflamma-
postnatal toxoplasmosis. tory reactions may be triggered by “primary” autoim-
In some infections the preferential location is sim- mune reactions, in which autoantibodies are primar-
ply related to the porte d’entrée. For example, infec- ily directed against autoantigens, as is the case in vas-
tions that involve the leptomeninges will enter the culitis of the CNS, in systemic lupus erythematosus,
brain via the Virchow–Robin spaces and are located Behçet disease, giant cell arteritis, and rheumatoid
predominantly in the cortex (Haemophilus influen- vasculitis. The hypothesis is that these primary auto-
zae) and in the basal ganglia (Cryptococcus neofor- immune disorders are the result of a dysfunction of T
mans) or both. In herpes simplex virus, a primary cell regulation. The inflammatory response in the
infection in the throat or mouth is followed by the CNS may also be secondary to an infectious disease
persistent presence of latent virus in sensory ganglia elsewhere in the body or a vaccination. Mycoplasma
and possibly also in the brain, after entry via the infections of the lung, for example, may lead to an
olfactory route. Following activation of the latent acute disseminated encephalomyelitis, caused by the
virus, direct spread may explain the frontotemporal formation of antibodies that cross-react with CNS
predominance. Within the affected area, herpes sim- antigens on myelin membranes or surface molecules
plex virus infects all types of cells. of CNS cells. This reaction resembles in many re-
Inflammatory and Infectious Disorders 565

spects the experimental allergic encephalomyelitis ∑ Specific infection of oligodendrocytes leading to


provoked by immunization with components of the cell death and subsequent loss of the myelin
myelin membrane (basic myelin protein, for exam- sheath extension of the oligodendrocytes. This is
ple). Such a secondary autoimmune response proba- the case in progressive multifocal leukoen-
bly also forms the basis of several paraneoplastic syn- cephalopathy.
dromes of the CNS, such as limbic encephalitis, and ∑ Myelin can also be an innocent bystander and be-
Purkinje cell degeneration. Here, too, the hypothesis come a victim in a process that hits all white mat-
is that antibodies formed against tumor antigens ter components and possibly gray matter as well.
cross-react with structural components of the CNS. This occurs in a multitude of infections, such as
As far as the involvement of myelin in inflammato- those caused by Haemophilus influenzae and her-
ry and infectious disorders is concerned, several bio- pes simplex virus, and in toxoplasmosis.
mechanisms can be distinguished: ∑ Finally, dysmyelination may be the consequence of
∑ Immune reaction against one of the components infectious or inflammatory changes to unmyeli-
of myelin. This occurs in acute disseminated en- nated white matter, damaging the white matter
cephalomyelitis. This condition can be simulated matrix. Most of the congenital infections can have
in an animal model, experimental allergic en- this effect, often in addition to other visible struc-
cephalomyelitis. In this experimental condition tural lesions.
the animal has been sensitized against basic
myelin protein.
Chapter 79

Multiple Sclerosis
J. Valk, F. Barkhof

79.1 Clinical Features main types. In most patients the disease starts with a
and Laboratory Investigations relapsing-remitting course (RR), defined by the oc-
currence of relapses clearly separated in time, with
Multiple sclerosis (MS) is the most common demyeli- stable intervals in between. Disability in RR patients
nating disorder of the CNS. The peak incidence is at develops because recovery between relapses is in-
30 years of age. MS rarely commences in childhood or complete; in two-thirds of the patients the relapsing-
after the age of 50 years. Females are affected twice as remitting phase is followed by a secondary progressive
often as males. MS shows a characteristic geographi- course (SP) with increasing disability as measured on
cal distribution. It is rare in tropical areas and in- a disability scale over a period of 12 months. Finally
creases in frequency at higher latitudes. It has been there is a primary progressive form in 10–15% of the
estimated, for example, that the prevalence in the patients, with progressive disability from the onset. In
United States varies from 6–14 per 100,000 inhabi- this group the disease usually starts later and the fe-
tants in the southern states to about 40–60 per male preponderance is less evident (Lublin and Rein-
100,000 inhabitants in the northern states. No definite gold 1996). The term benign form (BF) is used when
relationship has been established with the climatic after initial relapses and remissions no further pro-
characteristics of the latitude. Within this overall lat- gression occurs and the patient is neurologically fully
itudinal distribution a rather large range of inci- functional 15 years after the first symptoms. Special
dences of MS has been observed at the same latitudes. attention has been given in the last decade to patients
Occasionally clusters of MS have been reported. In with a clinically isolated symptom (CIS) because of
these instances a remarkable number of patients the potential to delay or prevent the possible progres-
acquire MS within a short period of time (a few sion to MS with early treatment. Finally, tumefactive
months). Etiological factors for these MS pockets are forms of MS present with symptoms of a space-occu-
as yet unknown. Sometimes such clusters of MS as- pying lesion. Medical history, the presence of other
sume the proportions of an epidemic, the incidence of clinical symptoms or lesions on neuroimaging, and
MS rising over a period of several years in a large the therapeutic effect of corticosteroids may help in
area. In these cases the nature of the introduced envi- establishing the correct diagnosis.
ronmental factor or factors still remains to be eluci- In classical MS, clinical signs and symptoms are re-
dated. The importance of environmental factors is lated to lesions present in various tracts of the CNS.
stressed by the findings of migrational studies. A de- Most last for days up to months or are permanent.
crease in the risk of developing MS has been noted in Sometimes symptoms of extremely short duration
young individuals migrating from high-risk to low- occur, lasting for seconds to hours. Common features
risk areas and an increase in risk after migration from are fatigue, impairment of vision due to optic neuri-
low-risk to high-risk areas. Such changes in risk have tis, motor disturbances caused by pyramidal tract in-
not been found in older individuals, and the data sug- volvement, sensory disturbances including Lher-
gest that the risk of acquiring MS is largely estab- mitte’s symptom, cerebellar ataxia and dysarthria,
lished around the age of 15. There is also strong evi- diplopia, micturition problems, sexual disturbances,
dence of a genetically influenced susceptibility to MS. hearing loss, vertigo, and balance abnormalities. Less
In general, first-degree relatives of probands have a common are epileptic seizures, signs of peripheral
risk that is 30–50 times greater than the risk for the neuropathy, trigeminal neuralgia, hemifacial spasms,
general population. and dementia, although some degree of cognitive im-
A number of variants of MS can be distinguished: pairment is present in up to half of MS patients. There
classical MS (also called Charcot type), neuromyelitis is almost always a tendency for the frequency of
optica (NMO, or Devic disease), concentric sclerosis episodes to decrease as time passes, or for the pro-
(CS, or Baló disease), and diffuse sclerosis (DS, or gression in the progressive form to slow down.
Schilder disease). The extent of the resulting disability is extremely
The clinical presentation of classical MS is ex- variable. In general, it appears that the average life ex-
tremely variable. The extreme acute progressive form pectancy in young patients after the onset of MS is
(Marburg type) is rare. For the more slowly develop- about 35 years.About 60–70% of patients remain am-
ing forms it has become usual to distinguish four bulant. Long-term prognosis is not influenced by
79.1 Clinical Features and Laboratory Investigations 567

pregnancy, illness, or anesthesia. Patients with many Compared to classical MS, CS runs a more rapidly
relapses in the early phase more rapidly acquire a progressive, usually monophasic course. The initial
severe state of disability than those with less frequent symptoms are often suggestive of a stroke; less fre-
relapses (Confavreux et al. 2000). A large number of quently psychiatric symptoms predominate. The neu-
hyperintense lesions on T2-weighted images is also rological symptoms are sometimes associated with
indicative of more rapid deterioration. When more fever and headache and then resemble the clinical
than 10 lesions are present, the EDSS score within 4 picture of an infection or tumor. The disease is pro-
years will be higher than 6, which means inability to gressive and can be fatal, usually as a consequence of
walk without support. Abnormal evoked responses in respiratory problems and infection. A more pro-
the early phase of the disorder are also indicative of a longed survival for several years has also been report-
poor prognosis. It should, however, be noted that ed.
these predictors are rather weak. DS (encephalitis periaxialis diffusa or Schilder dis-
NMO (neuromyelitis optica or Devic disease) is a ease) is a demyelinating disease related to MS, which
clinical syndrome consisting of optic neuritis, often primarily affects children. Clinical features are intel-
bilateral with total blindness, in combination with lectual impairment, epileptic seizures, signs of pyra-
transverse myelitis, which usually has a thoracic lo- midal tract involvement (occasionally unilaterally
calization. The optic neuritis and transverse myelitis with hemiplegia), cerebellar ataxia, visual impair-
either occur simultaneously or are separated by a ment (caused by retrobulbar neuritis or demyelina-
brief interval of several days to several weeks. Men tion of the occipital lobes), pseudobulbar palsy (lead-
and women are affected more or less equally. The age ing to problems with speech and swallowing), deaf-
of patients ranges from 5 to 65 years, but patients are ness, diplopia, extrapyramidal movement abnormali-
rarely older than 50 years. The group most common- ties, and incontinence. A predominantly psychiatric
ly affected is young adults. This disease occurs most symptomatology is relatively frequent. In most cases
frequently in the Asian population, where the overall there is rather rapid progression of neurological signs
incidence of MS is low. The prognosis is rather poor. over the course of 1–2 years. In a minority, the de-
Until recently, about 15–20% of patients died in the myelinating process is fulminant and accompanied
acute stage due to an ascending spinal disorder with by cerebral edema. Rarely, the course of disease is
respiratory paralysis; another 30% died with compli- characterized by exacerbations. In exceptional cases
cations after many months. A poor neurological out- significant and prolonged improvement occurs; an
come with severe disability is reported in another arrest of the disease is observed in exceptional cases.
15% of patients. Complete or nearly complete recov- Definite diagnosis has always been a problem in
ery is found in about 35%. Improved supportive care MS. The clinical features may mimic many other neu-
has reduced mortality and residual disability. About rological disorders, including ischemic disorders,
half of the surviving patients experience no recur- neoplastic diseases, vasculitides, granulomatous dis-
rence of neurological disease; about one-third of the eases, and infections. Definitive diagnostic tests are
remainder suffer a relapse of optic neuritis, one-third lacking. This diagnostic uncertainty has led to the de-
a relapse of optic neuritis and transverse myelitis, and finition of diagnostic criteria. In 1965, Schumacher
one-third develop a multifocal white matter disease. was the first to draw up clinical criteria for the diag-
Wingerchuck et al. (1999) formulated absolute cri- nosis of definite MS. The basic idea behind these cri-
teria and supportive criteria for the diagnosis NMO. teria is that there must be symptoms and objective
The absolute criteria are: optic neuritis, acute myelitis, signs of multifocal white matter disease with dissem-
and no evidence of clinical disease outside the optic ination in space and time, for which there is no better
nerve and spinal cord. Supportive criteria are subdi- neurological explanation. These criteria have been re-
vided into major and minor support criteria. Major peatedly modified, and clinical criteria for possible
support criteria are: negative brain MRI at onset; MRI and probable MS have been added (Rose et al. 1976).
lesions in the spinal cord extending over more than In 1983, Poser et al. were the first to draw up diagnos-
three vertebral segments; and CSF pleocytosis >50 tic criteria that were not completely clinical but incor-
white blood cells/mm3. Minor supportive criteria are: porated supportive laboratory data (CSF abnormali-
bilateral optic neuritis; severe optic neuritis with ties) and paraclinical evidence of multifocal white
fixed visual acuity of less than 20/200 in at least one matter lesions (CT and evoked responses). Since that
eye; and severe, fixed, attack-related weakness in one time, MRI has become more generally used and has
or more limbs. acquired a prominent role as a paraclinical test. MR
CS (encephalitis periaxialis concentrica or Baló has the possibility to prove dissemination in time (by
disease) is a very rare MS variant, which usually af- new enhancing lesions or new T2 lesions) and dissem-
fects young adults. For unknown demographic rea- ination in space (by multiple brain and spinal cord lo-
sons there is a much higher incidence in the Philip- calizations). In the most recently proposed diagnostic
pines. Both sexes are affected more or less equally. criteria for multiple sclerosis by McDonald et al.
568 Chapter 79 Multiple Sclerosis

Table 79.1. Criteria for diagnosis of MS (McDonald et al. 2001)


Clinical presentation Additional data needed for MS diagnosis

Two or more attacks; objective clinical evidence None


of 2 or more lesions
Two or more attacks; objective clinical evidence of 1 lesion Dissemination in space, demonstrated by MRI
or
Two or more MRI-detected lesions consistent
with MS plus positive CSF
or
Await further clinical attack implicating a different site
One attack; objective clinical evidence of 2 or more lesions Dissemination in time, demonstrated by MRI
or
Second clinical attack
One attack; objective clinical evidence of 1 lesion Dissemination in space, demonstrated by MRI
(monosymptomatic presentation; clinically isolated syndrome) or
Two or more MRI-detected lesions consistent with MS
plus positive CSF
and
Dissemination in time, demonstrated by MRI
or
Second clinical attack
Insidious neurological progression suggestive of MS Positive CSF
and
Dissemination in space, demonstrated by:
(1) Nine or more T2 lesions in brain
or
(2) 2 or more lesions in spinal cord
or
(3) 4–8 brain lesions plus 1 spinal cord lesion
or
Abnormal VEP associated with 4–8 brain lesions,
or with fewer than 4 brain lesions plus 1 spinal
cord lesion demonstrated by MRI
and
Dissemination in time, demonstrated by MRI
or
(4) Continued progression for 1 year

If criteria indicated are fulfilled the diagnosis is “multiple sclerosis” (MS); if the criteria are not completely met, the diagnosis is
“possible MS.” If the criteria are fully explored and not met, the diagnosis is “not MS.”
No additional tests are required; however, if tests (MRI, CSF) are undertaken and are negative, extreme caution should be taken
before making a diagnosis of MS. Alternative diagnoses must be considered.There must be no better explanation for the clinical
picture.
MRI demonstration of space dissemination must fulfill the criteria derived from Barkhof et al. (1997) and Tintoré et al. (2000).
“Positive CSF” is determined by oligoclonal bands detected by established methods (preferably isoelectric focusing) different
from any such bands in serum, or by a raised IgG index.

(2001), these MRI criteria have been formalized. The that of oligoclonal banding about 90%. The specifici-
MRI criteria for dissemination in space are based on ty and predictive value of these CSF investigations are
the Barkhof criteria (Barkhof et al. 1997) with a mod- highly dependent on the so-called pretest probability
ification as proposed by Tintoré et al. (2000). These of MS. The problem is that a number of diseases mim-
new MRI criteria have been validated and make an icking MS, such as infections, acute disseminated en-
earlier diagnosis of MS feasible without sacrificing cephalomyelitis, and vasculitis, are apt to lead to an
accuracy (Barkhof et al. 2003; Dalton et al. 2003). increased production of IgG in the CSF with oligo-
Table 79.1 summarizes the diagnostic criteria as rec- clonal banding. It should also be noted that some pa-
ommended by the International Panel on the Diagno- tients with clinically definite MS have a normal IgG
sis of Multiple Sclerosis. index and lack CSF oligoclonal bands. Levels of CSF
Valuable laboratory findings supporting the diag- IgM and IgA may also be elevated in MS. The CSF pro-
nosis of MS are oligoclonal bands in the CSF and an tein content may be slightly raised, but very rarely
increased IgG index as a sign of increased synthesis of exceeds the level of 1 g/l. The white cell count may be
IgG within the blood–brain barrier. The sensitivity of increased, but only in exceptional cases is it higher
assessment of the IgG index in MS is about 80% and than 20 cells/ml. Another CSF abnormality may be an
79.2 Pathology 569

elevation of myelin basic protein in active MS. Its lev- in cases of clinically indefinite MS. VEPs and SSEPs
el is normal in stable MS. The finding of increased appear to have a higher diagnostic yield than BAEPs.
amounts of myelin basic protein is indicative of active However, abnormalities are nonspecific and must be
demyelination and as such not specific for MS. Re- interpreted with care in the context of the clinical pic-
cently analysis of antibodies against myelin oligoden- ture.
drocyte glycoprotein and myelin basic protein in pa- In NMO, CSF abnormalities are similar to those in
tients with clinically isolated symptoms predict for MS, with the exception of a more common occur-
early conversion to clinically definite MS (Berger et al. rence of lymphocytic pleocytosis.
2003). Free light chains of immunoglobulins and the Laboratory examinations in CS rarely yield much
k:l light chain ratio may be increased in the CSF. In- information. CSF usually reveals no pleocytosis,
creased CSF free k chains appear to be relatively spe- sometimes an increased amount of red blood cells.
cific for MS. Abnormally high levels have been found Total protein is only occasionally elevated.
in 85% of patients with clinically definite MS, in 20% In DS the CSF is often normal, but slight lympho-
of patients with CNS infections, and only exceptional- cytosis is occasionally found. The protein level is not
ly in noninfectious controls. Concentrations of solu- infrequently elevated, as is the IgG index. Oligoclonal
ble adhesion molecules, sVCAM-1 and sICAM-1, in bands have been reported.
serum and CSF correlate with activity of MS, as indi-
cated by gadolinium-enhancing lesions on MRI, and
may possibly be used as a surrogate marker for dis- 79.2 Pathology
ease activity.
A number of changes in the subset distribution of Usually the external appearance of the brain is rela-
T lymphocytes has been reported in the peripheral tively normal in MS. In chronic cases slight atrophy
blood of MS patients. CD (cluster of differentiation may be present with widening of sulci and slight en-
markers on hematopoietic cells) 4+ T cells (T helper- largement of the ventricular system. Occasionally
inducer cells) can be subdivided in two mutually ex- firm, depressed lesions are seen on the surface of the
clusive subsets: “naive” cells that have not yet been brain stem, spinal cord, and in the optic nerves. On
stimulated, and “memory” cells that have been stimu- sectioning, numerous lesions of varying size become
lated before. These subsets can be recognized by dif- apparent in the white matter of CNS. Even more are
ferences in CD antigens. Memory cells can produce revealed by microscopic examination, especially if
large amounts of cytokines after activation and show MRI-guided. The distribution of plaques varies great-
increased expression of a set of adhesion molecules, ly among MS patients, but the following are recog-
sVCAM-1 and sICAM-1. In the peripheral blood of nized as preferential localizations: the periventricular
patients with active MS, naive cells are decreased in white matter, in particular the lateral angles of the lat-
number; in inactive MS this fraction is normal. In the eral ventricles, floor of the fourth ventricle, cerebellar
peripheral blood of patients with active MS, lympho- peduncles, cervical part of the spinal cord, and the
cytes have been found with a higher expression of the optic nerves. In severe, long-standing cases, numer-
activation marker CD26 than in patients with inactive ous lesions are found in most parts of the CNS. Al-
MS and healthy controls. In CSF of active MS patients though the distribution of lesions is not precisely
CD4+ T cells are relatively over-represented as com- symmetrical, predominant involvement of one hemi-
pared to CD4+ T cells in peripheral blood.Among the sphere is rare. A significant proportion of the plaques
CD4+ T cells in the CSF, memory cells are increased are found in the border zone between gray and white
whereas naive cells are almost absent. Subset changes, matter. The extent of cortical involvement has been
therefore, may reflect disease activity and can be used underestimated for a long time. They are found with
for monitoring purposes. Increased numbers of TNF- rigorous myelin stains (e.g., using antibodies against
producing T cells are associated with an enhanced myelin basic protein or proteolipid protein), because
rate of lesion development on MRI (Killestein et al. the normal amount of myelin in the cortex is minimal
2001). More recently, the role of CD8+ T cells (sup- and classical inflammatory changes are lacking
pressor-cytotoxic cells) has been reinforced, and (Peterson et al. 2001). In many cases a diffuse subpial
again relationships with MRI lesion development demyelination can be found (Bö et al. 2003).
have been found (Killestein et al. 2003). Further ex- Microscopically, the earliest stage of the MS plaque
aminations are required to reveal the significance of consists of perivenular lymphocytic infiltration. Such
these findings. lesions are now found more easily using postmortem
Other tests often used in the diagnostic assessment MRI as a guide (De Groot et al. 2001). The subsequent
of MS are the visual, sensory and auditory evoked po- stage is characterized by more diffuse tissue infiltra-
tentials (VEP, SSEP, and BAEP). These tests are helpful tion by inflammatory cells and macrophages associ-
in detecting silent white matter lesions, thus provid- ated with edema, demyelination, proliferation, and
ing evidence of multifocal white matter involvement hyperplasia of astrocytes, and appearance of in-
570 Chapter 79 Multiple Sclerosis

creased numbers of lipid-laden macrophages and toxic) T lymphocytes. Initially these T lymphocytes
demyelinated axons. Axonal damage already occurs are predominantly present in the center of the plaque,
during the phase of acute demyelination. Loss of but as the lesion enlarges, T cells move to the periph-
myelin and oligodendrocytes eventually becomes eral part of the lesion. The CD4+ cells invade the nor-
complete. As plaques enlarge and coalesce, the mal white matter. The majority of the inflammatory T
perivenular distribution becomes less apparent. Al- cells in MS are memory cells. The margins of the
though variable between patients, axonal damage can plaque contain predominantly CD8+ cells and in-
be extensive from early on. In the course of time, creased numbers of oligodendrocytes and astrocytes.
axonal loss can become very substantial (Bjartmar et With increasing age of the plaque, myelin and
al. 2003). In the gray matter plaques, too, the myelin is macrophages disappear from the central part; the
predominantly affected and neuronal cell bodies are plaque margins contain lymphocytes, oligodendro-
largely preserved. In lesions of several months’ dura- cytes, lipid-laden macrophages, and astrocytes, sug-
tion, inflammation is far less pronounced, fewer lipid- gestive of low-grade activity at these margins. In
laden macrophages are seen, and fibrillary gliosis be- chronic-active MS, small numbers of inflammatory
comes increasingly prominent. Chronic-inactive MS cells are scattered throughout the normal-appearing
plaques have a sharply demarcated border and are white matter, suggestive of a diffuse, slow demyelinat-
hypocellular, demyelinated, and gliotic with almost ing process. Chronic-inactive MS lesions contain few
total oligodendrocyte loss. Inflammatory cells and inflammatory cells. In chronically affected tissue an
lipid-laden macrophages are no longer present. The interesting recent finding is the presence of T cells
remaining elements are axons and astrocytic process- and their association with heat shock proteins ex-
es. Axonal damage has led to wallerian degeneration, pressed on oligodendrocytes. These cells have previ-
which is most evident in the long tracts. Rarely is the ously been implicated in the pathogenesis of rheuma-
damage sufficiently severe to produce a cyst. In the toid arthritis, but the presence of these cells with still
same patient lesions of different ages are present. unclear function now seems to be a more general
In MS, plaque-like areas are observed, in which finding in autoimmunity. They may play a role either
myelin has not completely disappeared, and which do in tissue repair or in perpetuating the inflammatory
not have the appearance of the typical plaque. These process.
lesions are called shadow plaques. The myelin sheaths In the review of Lassman et al. (2001) four different
in these plaques are abnormally thin and are of rela- types of histopathological reactions were distin-
tively uniform thickness. The internodes are short. guished, possibly reflecting four different types of MS
The number of oligodendrocytes is increased in the lesions: (1) macrophage-mediated, with T-cell-medi-
lesion. These features are characteristic of remyelina- ated inflammation as a putative mechanism; (2) anti-
tion, and so the shadow plaques probably represent body-mediated, with T-cell-mediated inflammation
areas of remyelination. with complement-mediated lysis of antibody-target-
The description as given fits the relapsing-remit- ed myelin as a putative mechanism; (3) distal oligo-
ting form of MS. Histological examination is usually dendrogliopathy, with T-cell-mediated small vessel
performed in patients who have suffered from the vasculitis with secondary ischemic damage of the
disease for a long time.An exception is the acute form white matter as a putative mechanism; and (4) prima-
of MS, in which the lesions are days to weeks old and ry oligodendrocyte damage and secondary demyeli-
show acute inflammatory and demyelinating changes. nation, with T-cell-mediated inflammation and de-
As a consequence of the lack of early histological in- myelination induced by macrophage toxins on the
formation, there is no parallel description of histol- background of metabolically impaired oligodendro-
ogy in some clinical MS subtypes, such as benign MS. cytes as a putative mechanism. It is clear that such a
In primary progressive MS more diffuse demyelina- postmortem classification does not easily translate
tion with more diffuse and less intense inflammation into clinical practice, but it may help a better under-
is found.A mixture of the multiple lesions and the dif- standing of the differences between subtypes of MS.
fuse pattern is seen in patients in whom the disease In NMO the spinal cord, optic nerves, and chiasm
was initially relapsing-remitting, but secondarily be- appear swollen and congested externally if the patient
came progressive in its course. has died relatively early in the course of disease.
Immunocytochemical studies have demonstrated Demyelination and inflammation with perivascular
that the inflammatory cells of acute MS plaques are lymphocytic infiltration and fat granules are seen at
mainly macrophages and lymphocytes, with few plas- microscopy. In severe cases necrosis of gray and white
ma cells. Macrophages stain positive for the major matter occurs, leading to cavitation. However, at the
histocompatibility complex (MHC) class II, which periphery of such lesions the relative sparing of axons
implies a role for these cells in local antigen presenta- is evident. Acute lesions may be hemorrhagic. The
tion to T cells. The T cells present are a mixture of spinal cord lesion is often large and extends over
CD4+ (helper-inducer) and CD8+ (suppressor-cyto- many segments, usually in the low cervical and high
79.3 Chemical Pathology 571

thoracic areas. Lesions in the conus may also occur. In generation is common. There are not only naked ax-
the optic nerves and chiasm extensive loss of myelin, ons but also axons partially covered with thin layers
gliosis, and some loss of axons occur. As a rule, of myelin as signs of abortive remyelination.
additional areas of demyelination are found in the
predilectional regions of classical MS, such as peri-
ventricular areas and brain stem. 79.3 Chemical Pathology
The characteristic lesions in CS are areas of alter-
nating zones of myelinated and demyelinated tissue, Chemical analysis of the composition of MS plaques
either with a concentric pattern or with a more irreg- reveals a number of alterations: increase in water, de-
ular arrangement. The size of lesions varies from tiny crease in total lipid, in particular in cholesterol, cere-
to about 4–5 cm in diameter. The location and num- brosides, sulfatides, ethanolamine plasmalogens, and
ber of lesions vary widely. Sometimes large areas are serine phosphoglycerides. Cholesterol esters are in-
almost completely involved. The lesions may occur creased. In the lesion the major myelin proteins –
anywhere in the CNS; only the spinal cord is rarely myelin basic protein, proteolipid protein, and myelin-
affected. The rings of the lesion terminate abruptly associated glycoprotein – are reduced. Myelin basic
where they contact gray matter. The central core is the protein is in fact virtually absent from the center of
starting point of the lesion and consists of a venule most plaques, and the decrease in concentration of
with a cuff of inflammatory cells. In the course of myelin-associated glycoprotein extends into the nor-
time, the central core becomes intensely gliotic. The mal-appearing white matter around the plaque, sug-
core is surrounded by zones of demyelination, in gesting that this protein disappears or is altered be-
which axons are preserved and myelin is replaced by fore myelin breakdown starts. The protein losses in
gliosis. With increasing distance from the core, the the plaque are accompanied by an increase in pro-
stage of myelin breakdown in the affected zones teins of a lower molecular weight, which may be pro-
becomes less advanced, and the gliosis is less severe. teolytic breakdown products. In and around the MS
Acute lesions are surrounded by edema. In chronic lesion proteinases and other hydrolytic enzymes are
lesions the involved area becomes scarred and at- increased. The observed chemical changes in the MS
rophic. The concentric lesion may become so disinte- plaques are variable and depend on the extent of
grated that it is difficult to recognize. Ultrastructural demyelination. An early MS lesion contains more
examination of the myelinated zones reveals that they myelin and more sudanophilic material, which is bio-
are largely composed of thinly myelinated fibers. A chemically defined as cholesterol esters. Old plaques
few normally myelinated and some demyelinated have no or little myelin left and lack sudanophilic
axons are also present. These bands contain many material.
cells, including oligodendrocytes, lymphocytes, and Myelin isolated from the plaque has a composition
astrocytes. These changes are reminiscent of those typical of abnormal myelin during aspecific degrada-
seen at the edge of a chronic-active MS plaque and are tion. No myelin abnormalities specific for MS have
interpreted by some as zones of remyelination. been demonstrated.
Throughout the white matter, numerous venules Much effort has been devoted to the study of the
show cuffs of inflammatory cells. Very often there are chemical composition of normal-appearing white
also lesions characteristic of classical MS. matter in MS. In the first place, the myelin yield of the
In DS widespread demyelination is found, with normal-appearing white matter is strikingly low.
variable axonal damage in the centrum semiovale of There is a decrease in total lipid, in phospholipids
both cerebral hemispheres, most often involving the (particularly ethanolamine plasmalogens), in both
occipital lobes. Usually the corpus callosum is also af- galactolipids (cerebroside, and sulfatide), and in
fected and interconnects the lesions of the two sides. myelin proteins. Frequently, cholesterol esters are
The lesions are not completely symmetrical. They found. Several investigators found an elevation of
have a sharp edge. A rim of subcortical white matter hydrolytic enzymes in normal-appearing white mat-
is commonly preserved, but a lesion may also spread ter. These findings are qualitatively similar to those
into the gray matter. In the acute stage demyelination observed in MS plaques, but are considerably smaller
is associated with dense perivascular infiltrates of in magnitude. All these data together provide strong
lymphocytes, plasma cells, and lipid-filled macro- evidence for the presence of minor microscopic ab-
phages. The myelin disintegration leads to the forma- normalities of the MS type in the white matter that
tion of sudanophilic material. Areas may be become appears macroscopically normal. This suggestion has
frankly necrotic, and cavitation may occur. Glial reac- been confirmed by microscopic examination of sam-
tion is present, with giant multinucleated and hyper- ples of normal-appearing white matter. It is clear that
trophied astrocytes. In cases of long duration, inflam- the disease process is widespread and not simply re-
matory cells and macrophages containing sudano- stricted to plaques. There is a general myelin deficit
philic material disappear. Evidence of wallerian de- throughout the white matter.
572 Chapter 79 Multiple Sclerosis

79.4 Pathogenetic Considerations indicative of hyperactive B lymphocytes, and the de-


ficiency of suppressor T lymphocyte function may be
The search for the cause of MS has engaged many in- signs of a fundamental defect in immunoregulation.
vestigators for many years. Three main lines can be The role of CD8+ suppressor-cytotoxic cells versus
distinguished in theories about the etiology of MS: CD4+ helper cells has been a point of discussion.
one line stressing the importance of immunological These subsets interact with specific MHC molecules
reactions, the second line pointing to the evidence for to regulate the immune response. MHC class II
genetic factors, the third one advocating environmen- restricted CD4+ T cells are the major producers of
tal factors. These lines are complementary and not cytokines and are associated with delayed-type hy-
mutually exclusive. persensitivity and antibody response. MHC class I re-
A major theory proposes that MS results from al- stricted CD8+ T cells are associated with cytotoxicity.
terations in the immune system. A suggestion in this There is evidence that CD4+ cells are the key initia-
direction came from the observation of some similar- tors of tissue destruction in MS. CD8+ T cells have re-
ity between MS and experimental allergic encephalo- ceived less attention, but there is growing evidence
myelitis (EAE) in animals. EAE is induced by immu- that MHC class I restricted CD8+ T cell responses
nizing animals with antigens normally present in the may have a critical role in the pathogenesis of MS. In
CNS myelin, such as myelin basic protein (MBP), pro- addition to CD4+ cells, CD8+ lymphocytes are pre-
teolipid protein (PLP), and myelin oligodendrocyte sent in active demyelinating lesions, and in fact pre-
glycoprotein (MOG), with an adjuvant containing dominate in many lesions. CD8+ T cells recognize
heat-killed mycobacterium to stimulate the innate peptides presented by MHC class I molecules. Adult
immune system. In about 1–2 weeks the animals de- oligodendrocytes express class I MHC molecules and
velop encephalomyelitis with perivascular infiltrates do not express class II MHC molecules, even when
composed of lymphocytes and macrophages in the stimulated by interferon-g. This implies that when
white matter, followed by demyelination. In mice EAE adult oligodendrocytes are targets for T cell reactivi-
is caused by activated CD4+ T cell lymphocytes spe- ty, they will probably be recognized by MHC class I
cific for MBP, PLP or MOG, as proven in vitro and in restricted CD8+ T cells and not by class II restricted
cloning experiments. The lesions of EAE and of MS CD4+ T cells.
have the character of delayed hypersensitivity reac- The cause of the immunological alterations in MS
tions. The sequence of events in EAE and possibly by and their role in the pathogenesis of MS have not yet
analogy MS could be as follows: autoreactive T cells been elucidated. The antibody response in the CSF
specific for myelin proteins may be present in the cir- may be an expression of an autoimmune process
culation of normal individuals. Immunization with a against normal or altered brain constituents. This
myelin antigen together with an adjuvant leads to T autoimmune process may be idiopathic, or triggered
cell activation against epitopes of autologous myelin by a viral infection or other exogenous or endo-
proteins. Activated T cells stick to the endothelial lin- genous antigens that cross-react with brain con-
ing and cross into the cerebral parenchyma. In the stituents. Low concentrations of antibodies have been
CNS the activated T cells encounter myelin proteins demonstrated in the CSF reacting with myelin pro-
and release cytokines that recruit and activate teins, oligodendroglia, glycolipids, and nuclear anti-
macrophages and other T cells that lead to myelin de- gens, but no single MS-specific antigen that reacts
struction. EAE has been used to analyze the specifici- with most of the IgG has ever been identified. It is not
ty of myelin-reactive T cells and help to define im- excluded that the observed antibodies are epiphe-
munodominant regions of myelin proteins that may nomena without pathogenetic importance. Another
be relevant to MS in humans. explanation may be that T helper-inducer lympho-
Abnormalities of immunoregulation and of hu- cytes in MS are activated and autoreactive. Many
moral and cellular immunity appear to be an impor- other changes in immune-related factors have been
tant part of the disease process in MS. Inflammatory observed, such as circulating immune complexes,
cells – lymphocytes, plasma cells, and macrophages – altered levels of cytokines and complement compo-
are present in perivascular areas in the CNS in active nents, and increased prostaglandin synthesis, but the
disease and take part in the disease process. In nearly significance of these findings is not known. A con-
all MS patients, there is evidence for an increased syn- vincing observation about the role of immune re-
thesis of immunoglobulins within the blood–brain sponses in MS has been the decrease in MS-activated
barrier. A low CD8+ T suppressor cell activity and a lesions in pregnant women. There is a sharp decrease
high ratio of CD4+ T helper to CD8+ T suppressor in lesions in all cases, with a return to the prepregnant
cells is present in the blood during exacerbations of status in the months after delivery. In another study,
MS, and also chronically progressive MS patients have the beneficial effect of pregnancy was indicated by
similar abnormalities of peripheral blood T lympho- the finding that the mean disease duration before be-
cyte subsets. The high levels of antiviral antibodies, coming wheelchair-dependent was 50% longer in pa-
79.4 Pathogenetic Considerations 573

tients who became pregnant after the first symptoms concordance rate (a concordance rate of 100% would
of MS. be expected among monozygotic twins if MS were ex-
Lymphocytes become activated by an unidentified clusively genetically determined), and the increased
cause and by a multistep process penetrate the prevalence of MS in dizygotic twins as compared to
blood–brain barrier. The capillary endothelial cells in siblings (1–6% of the siblings of MS patients are also
the CNS are not fenestrated and are connected affected) strongly suggests the involvement of envi-
through tight junctions. The capillary endothelial ronmental as well as genetic factors.
cells express cellular adhesion molecules (V-CAM) Further evidence for a genetic component in the
and class II molecules of the MHC.Activated lympho- etiology of MS comes from the observation of associ-
cytes can pass the endothelial barrier assisted by ations between MS and specific human leukocyte
adhesion molecules such as integrins, in particular a4 antigen (HLA) alleles. The HLA genes encoding for
integrin, which binds to V-CAM. Once the activated these antigens, which are expressed on cell surfaces of
lymphocytes have extravasated, they still need help in lymphocytes, are found on the short arm of chromo-
passing through a barrier of extracellular matrix con- some 6. The HLA system consists of five loci – A, B, C,
sisting of type IV collagen. T cells are then targeted to D, and DR – and each of the HLA loci has a large num-
proteins of the myelin sheath, myelin basic protein, ber of alleles. The HLA region can be used as an excel-
myelin oligodendroglial glycoprotein, and prote- lent genetic marker with known chromosomal loca-
olipid protein, as well as stress proteins like aB crys- tion. There is a highly significant association between
tallin, present in the myelin sheath after activation by HLA-DR2 and MS and a less strong association be-
the inflammatory response. T cells produce cyto- tween HLA-A3 and B7 and MS in Caucasians. How-
kines, notably TNF-b and TNF-a, and then influence ever, there are great differences in observed HLA as-
macrophages, microglial cells and astrocytes to pro- sociations in populations of different racial back-
duce nitric oxide, a major mediator in inflammatory ground, and it is clear that the mentioned HLA alleles
reactions, and osteopontin, a multifunctional protein in themselves are neither necessary nor sufficient to
abundantly expressed during inflammation. Macro- lead to the development of MS. The meaning of the
phages and microglia are induced to phagocytose HLA associations is not entirely understood. A likely
large pieces of the myelin sheath. explanation is that the MS-related gene or genes lie on
This short and incomplete summary of the initia- the same chromosome as the HLA genes and are in
tion of the inflammatory process is intended to give linkage disequilibrium with specific HLA alleles. This
an impression of the complexity of the process, in means that certain combinations of alleles occur sig-
which the role of the many players only gradually be- nificantly more frequently than would be expected by
comes clear. Knowledge of this process in detail has chance. It is also possible that specific HLA alleles are
already opened therapeutic windows and will open directly involved in the etiology of MS. The HLA sys-
more in the future. It helps us to understand why tem is part of the immune response system. Alleles of
many therapeutic interventions only yield partial re- certain class II genes, HLA DR and HLA DQ, confer
sults. Much still depends on the unraveling of the pri- the strongest risk of MS. In addition to the well-estab-
mary cause of the inflammatory reaction. lished MHC association, genome-wide screens of
The evidence for genetic factors comes from re- families with multiple cases of MS also suggest a role
ports of familial cases and unusually high-risk fami- for several additional unidentified genes, each with a
lies, and from studies which consistently show higher modest effect. Transcriptional profiling using gene
concordance rates for MS in monozygotic twins than microarrays and large-scale sequencing of tran-
in dizygotic twins. The concordance rate in monozy- scripts from MS lesion material reveal expression of
gotic twins reported in the literature varies from 10% genes involved in the pathogenesis of acute disease,
to 70%, and that in dizygotic twins from 2.3% to 20%. like immunoglobulins, interleukin 6, and osteopon-
Selection bias probably leads to overestimation of the tin.
rate of concordance among twins. On the other hand, It is clear that MS is not a genetically determined
however, a twin sample collected at one point in time disease with a Mendelian mode of inheritance. It is
probably underestimates the concordance rate, as more probable that multiple susceptibility genes for
more individuals will develop MS in the course of MS exist, and that the expression of subtle changes in
time, and the concordance rate will increase with in- these genes (polymorphisms) depends on environ-
creased duration of follow-up. The prevalence of MS mental factors. The chromosomal localization of
among relatives of MS patients is increased, and the some of the genes determining susceptibility to MS is
increase becomes more pronounced the closer the de- probably in or near the HLA region, or their expres-
gree of kinship to the propositus. This observation is sion depends on the action of certain HLA alleles. A
consistent with a genetic hypothesis, but common en- method consisting of pooling DNA from MS individ-
vironmental experiences with relatives, and especial- uals and typing these pools for around 60,000 mi-
ly twins, may also play a role. The low overall twin crosatellite markers, suggested by Barcellos et al.
574 Chapter 79 Multiple Sclerosis

(1997), was realized in an extensive study known as In CS, some consider the concentric lesion to be a
Genetic Analysis of Multiple Sclerosis in EuropeanS variant of an MS plaque in which the center of the le-
(GAMES), described by Sawcer and Compston (2003). sion represents the initial small focus of acute de-
GAMES is essentially an indirect screen for associa- myelination, and in which the concentric rings are
tion and is therefore dependent on the assumption formed by a centrifugal progression of episodes of
that at least some of the markers tested will have alle- demyelination and remyelination. The zones of pre-
les in linkage disequilibrium with gene variants that served myelin within the concentric lesions are sup-
influence the susceptibility to MS. All previous ge- posed to be formed by episodic remyelination at the
nomic screens in MS families only suggested linkage borders of demyelinating foci, which is followed by
with 6p21, the area containing the genes for the major further centrifugal spread of the demyelination. A
histocompatibility complex. Although its approach remarkable difference with MS is that the concentric
has limits and flaws, GAMES may have the potential lesion never invades gray matter structures, unlike
of identifying one or more novel associations outside MS plaques. There is, as yet, no explanation for the
the MHC region at 6p21. higher incidence of CS in the Philippines.
Epidemiological studies of migrants suggest that Loose and indiscriminate use of the term “diffuse
environmental factors, particularly when present be- sclerosis” (DS) has led to a great deal of confusion in
fore the age of 15 years, are involved in the etiology of nomenclature. Schilder was the first to describe the
MS. One of the most important theories about the na- disease in three cases of what he called “encephalitis
ture of the environmental factors speculates on a viral periaxialis diffusa.” However, on closer inspection of
etiology. However, a causative virus has never been clinical data and neuropathological findings, it was
reproducibly isolated from the CNS of MS patients, concluded that one of these patients probably had X-
nor has viral antigen been demonstrated in a consis- linked adrenoleukodystrophy and another acute dis-
tent fashion. Ultrastructural examination of brain tis- seminated encephalomyelitis. Only one patient is now
sue has never unequivocally revealed virus particles. considered to be an example of DS. After Schilder
Antibodies to multiple viruses are elevated in the CSF many authors used the name “diffuse sclerosis” for a
of MS patients. It is probable that these antibodies re- wide range of unrelated demyelinating disorders. It is
sult from a nonspecific immunostimulation. It is true that a number of diseases, especially X-linked
commonly accepted that relapses in MS are often trig- adrenoleukodystrophy, may be difficult to differenti-
gered by infection with viruses. Viruses, such as her- ate from DS on clinical and pathological grounds
pes virus-6, influenza, measles, papilloma virus, and alone. In these cases, assessment of various enzyme
Epstein–Barr virus, have genes encoding peptides activities and ultrastructural examination are indis-
containing amino acid sequences similar to those pensable in establishing the correct diagnosis. In the
found in the major structural proteins of myelin. An- course of time, an increasing number of diseases
tibodies reacting with protein sequences from these could be distinguished from DS, and some investiga-
microbes may cross-react with components of the tors have suggested abandoning the term “Schilder
myelin sheath. T cells also recognize peptide se- DS.” However, we are of the opinion that the term
quences in the myelin sheath that are shared with mi- “Schilder DS” should be reserved for myelinoclastic
crobial sequences. Once an immune cell is activated, diffuse sclerosis as a variant of MS. The pathology of
either by a foreign microbe or a self-protein, it may DS does not differ substantially in its light or electron
penetrate the blood–brain barrier and the inflamma- microscopic appearances from the classical dissemi-
tory cascade as described above will follow (Buljevac nated form of MS. The only difference involves the di-
et al. 2002). mension of the demyelinating lesions and the rapid
The precise nature of the relationship between MS, progression of the process. In 1985, Poser offered the
CS, NMO, and DS is not known. The frequent occur- following definition of DS: the disease is a subacute or
rence of histopathological typical MS lesions in CS, chronic myelinoclastic disorder resulting in the for-
NMO, and DS provides evidence for essential similar- mation of one or more, commonly two, roughly sym-
ities in etiology and pathogenesis. In NMO, it is im- metrical plaques measuring at least 2 ¥ 3 cm in two of
portant to distinguish the MS-related disease from the three dimensions, involving the centrum semio-
acute disseminated encephalomyelitis and a vas- vale of the cerebral hemispheres. Other diseases that
culitic process, especially lupus erythematosus, both can lead to a similar picture should be excluded. Ac-
of which may produce an identical clinical picture cording to this view, the pathogenesis of DS is largely
and a rather similar pathological picture. As NMO is identical with that of MS, and the question is which
relatively frequent in Asian populations, it has been factor is responsible for the difference. It has been
suggested that racial-genetic factors lead to a modi- suggested that the large areas of demyelination may
fied appearance of MS. be due to the fact that the child’s nervous system, be-
79.5 Therapy 575

ing still immature, is more susceptible to an injurious MRI. However, IFNB is only partially effective at the
agent. It is improbable, however, that the immaturity tested doses. Patients in the high-dose group contin-
of the brain alone accounts for the difference com- ued to have exacerbations, although at a reduced rate
pared to classical MS, as classical MS can also occur and of milder clinical severity. The beneficial effect of
during childhood. IFNB is probably due to its ability to inhibit interfer-
The correlation between neuropathological lesions on-g (IFNG) synthesis, to improve defective suppres-
and clinical signs and symptoms is rather poor in MS. sor activity in MS patients, and to inhibit MHC class
There are many silent lesions. Clinically silent lesions II antigen expression induced by IFNG on the sur-
probably occur when demyelination affects some but faces of antigen-presenting cells. Patients in the
not all fibers of a pathway, or when remyelination chronic progressive phase still with relapses also
occurs. A conduction block occurs in demyelinated benefit from continuous IFNB treatment. The indica-
fibers, but conduction remains intact in unaffected tions for INFB are relapsing-remitting forms of MS
fibers. The very transient symptoms in MS are proba- and clinically isolated syndromes. Interferons are less
bly related to a reduction of the functional reserve of effective in primary progressive MS and secondary
a fiber tract for the conduction of nerve impulses be- progressive MS.
cause of demyelination. Slight alterations of conduc- Mitoxantrone (Novantrone in the US and Canada),
tion capacities, e.g., those due to a rise in body tem- which belongs to the group of tumor antibiotics, has
perature, may result in the appearance of symptoms been evaluated in a number of studies. Patients were
from a fiber tract in which a plaque has reduced the treated with intravenous administration every 3
functional reserve but not to less than the minimum months for 24 months. Mitoxantrone was found to de-
number of fibers necessary for normal function. Im- lay the time to first treated relapse and time to disabil-
provement occurs as soon as conduction of electrical ity progression in patients with secondary progres-
impulses is restored. Recovery after a relapse is prob- sive disease or progressive relapsing-remitting dis-
ably largely related to remyelination, which leads to ease. Monitoring of white blood cells and liver func-
abolition of the conduction block. tion remained necessary because of an increased risk
of infection.
Copolymer 1, known as glatimer acetate, is a mix-
79.5 Therapy ture of polypeptides (L-alanine, L-lysine, L-glutamine,
and L-tyrosine) that acts as an immunological agent
The interest in an immunological basis for MS has led and appears to simulate myelin basic protein. It has
to the development of a variety of treatments de- been reported to reduce the number of relapses with
signed to alter or suppress the immune response. One possible improvement of functional abilities. Its
of the first of these studies advocated the use of greatest advantage is its low toxicity. Glatimer acetate
adrenocorticotropic hormone (ACTH) in acute exac- stimulates antigen-specific suppressor cells. T cells
erbations. It was demonstrated that ACTH shortens activated by glatimer acetate protect against inflam-
the recovery time but does not alter the eventual lev- matory damage by the production of anti-inflamma-
el of recovery. More recent studies indicate that high- tory cytokines. It reduces the number of relapses. It is
dose intravenous methylprednisolone also hastens effective in MS patients under the age of 16 years.
recovery, but that the effect on suppression of Long-term studies indicate that the EDSS score in pa-
gadolinium-enhancing lesions, used as surrogate tients treated with glatimer acetate hardly increases
marker of MS activity, is short-lived. Intrathecal cor- over the years and that outcome parameters such as
ticosteroids did not prove to be any more helpful than brain atrophy and black holes show favorable results.
orally administered corticosteroids. In MRS studies the N-acetylaspartate (NAA) increas-
More aggressive immunosuppressive agents have es with treatment. Probably this effect is due to the in-
also been used, such as azathioprine, cyclophos- crease of brain-derived neurotrophic factor under
phamide, cyclosporin A, and total lymphoid irradia- glatimer acetate treatment.
tion. Plasma exchange has been used in limited stud- The glycoprotein a4 b1 integrin, also known as very
ies. However, the limited beneficial effects of these late antigen 4 (VLA-4) is expressed on the surface of
treatment modalities usually do not outweigh the of- lymphocytes and monocytes and is an important me-
ten serious side effects. Recently the intravenous ad- diator of cell adhesion and transendothelial migra-
ministration of immunoglobulins has been the sub- tion. Treatment with an antibody against a4 integrin
ject of interest. reduced signs of disease activity and inflammation in
Success has been reported with interferon beta-1b mice and gave positive results in a small, placebo-
(IFNB) in relapsing-remitting MS, with significant fa- controlled clinical trial with a humanized monoclon-
vorable effects on exacerbation rates, times between al antibody (natalizumab) (Miller et al. 2003).
first and second exacerbations, severity of exacerba- New treatment modalities are based upon the un-
tions, MS activity, and lesion load as determined by derstanding of T cell activation and cytokine produc-
576 Chapter 79 Multiple Sclerosis

tion in autoimmune diseases. From a therapeutic 79.6 Magnetic Resonance Imaging


point of view there are several ways in which exces-
sive effector functions of activated T cells can be MR has obtained a prominent role in diagnosis, fol-
counteracted, for instance by injection of monoclon- low-up, therapy monitoring, and research in MS. MRI
al antibodies against T cell membrane molecules. The protocols for patients with or suspected of having MS
presence of excessive intrathecal immunoglobulins in depend on the clinical symptoms and the information
MS may be related to the activation of CD4+ T lym- required from the MRI. Different techniques yield
phocytes, also inducing B lymphocyte production. A different information (Table 79.2).
large multicenter anti-CD4+ antibody trial was start- The most common MRI appearance of the relaps-
ed, but the lack of efficacy has ended this trial in an ing-remitting and secondary progressive forms of MS
early phase. As monoclonal antibodies act more se- consists of multiple lesions with intermediate or low
lectively, they may be more effective in suppressing signal intensity on T1-weighted images, high signal
disease activity. intensity on T2-weighted images, isolated or confluent
Far fewer therapeutic trials have been performed or both, with a bilateral but usually not entirely sym-
in CD, NMO, and DS. In these groups no reports on metrical distribution, and preferentially located along
the effects of INFB or glatimer acetate are available. In the lateral angles of the ventricles in the efferent and
CD a beneficial effect of prednisone therapy has been afferent tracts of the corpus callosum (Figs. 79.1–
described, but not consistently. In NMO remarkable 79.5). Some of these lesions have a typical ovoid form,
improvement has been reported under treatment pointing towards the convexity of the brain, known as
with corticosteroids, immunosuppressants, and lym- Dawson’s fingers (Figs. 79.1, 79.5). This is the result of
phocyte plasmapheresis. However, due to the very low the perivenular distribution of MS lesions. This
incidence of NMO, no controlled trial has provided is known from histopathology, but can now also
proof of such favorable effects. In DS a good clinical be illustrated by high-resolution MR venography
reaction to corticosteroids, ACTH, and immunosup- (Fig. 79.1). The corpus callosum is thinner than usual
pressants is generally observed. and the inner rim has an irregular border related to
Many other attempts have been made to control the presence of lesions. Parasagittal and sagittal T2-
the progress of MS. In this section we discuss some weighted and FLAIR images show this to best advan-
prominent endeavors – certainly not all of them – to tage (Fig. 79.5). Other lesions are spread over the
demonstrate the many possible entries to the disease frontal, parietal, and occipital white matter and, less
process. frequently, the temporal lobes. There are no or few le-
Finally, and also importantly, there are many pal- sions in the basal ganglia. Lesions may occur in the
liative measures to ease the problems of MS patients: midbrain (8%), pons (12%), cerebellar hemispheres
physiotherapy is helpful in keeping the patient mobile (4%), and medulla oblongata (1–2%). Specific struc-
as long as possible; spasmolytic drugs have a place in tures may be involved, such as the medial longitudi-
combating spasticity and bladder dysfunction; and nal fasciculus (clinically presenting as internuclear
antibiotics are necessary in intercurrent infections. ophthalmoplegia), the trigeminal nucleus or emerg-

Table 79.2. MR sequences that may be included in a protocol with an indication of the MS features in which they are effective.
This list serves as a general orientation and is not intended to be a protocol applicable to all MS patients.
Conventional:
T1-weighted, transverse “Black holes”,“gray” holes, prognosis
T2-weighted and proton density, transverse T2 lesions, distribution, lesion load
FLAIR (2D, 3D) Most sensitive for T2 lesions
T1-weighted with gadolinium Active lesions, blood-brain barrier disruption
FLAIR sagittal Corpus callosum lesions
STIR Visual pathways, optic neuritis
Proton density, T1-weighted, cardiac gated Spinal cord lesions

Special:
MTR or histograms Structural integrity, follow-up, research, study of normal appearing white matter
DWI, DTI Structural changes, ADC, FA, fiber tracking
MRS Chemical composition

FLAIR, fluid-attenuated inversion recovery; STIR, short tau inversion recovery; DWI, diffusion-weighted imaging; DTI, diffusion
tensor imaging
79.6 Magnetic Resonance Imaging 577

Fig. 79.1. The upper row of images


shows left a proton density image
with a typical ovoid lesion and right
a T1-weighted image with intravenous
contrast, with enhancement of the
lesion.The lower row of images shows
a special high-resolution technique
that allows submillimeter venography
of the CNS.The image on the right
shows the lesion extending from the
frontal horn towards the convexity in
its relation to the local perforating
vein. From Tan et al. (2000), with
permission

ing nerve (presenting as trigeminal neuralgia), and longer enhancing lesions together with new or reacti-
the vestibular nucleus (presenting with vertigo) vated lesions that enhance. The most important role
(Figs. 79.6 and 79.7). Cortical lesions are often present of gadolinium-enhanced MRI has emerged as moni-
but are difficult to appreciate on MRI. Cortical lesions toring the efficacy of drug treatment (Figs. 79.10 and
extending into the white matter or lesions located in 79.11). Long-term follow-up studies have shown that
the U fibers are well detectable (Fig. 79.8). Such juxta- newly appearing gadolinium-enhancing lesions are
cortical lesions form an important hallmark of MS on far more frequent than clinical exacerbations in re-
MRI and have been incorporated into new diagnostic lapsing-remitting and secondary progressive forms
criteria (McDonald et al. 2001, see Table 79.1). of MS. If one accepts gadolinium enhancement as a
Depending on the clinical request, contrast may be sign of new disease activity, this can serve as a means
given. Active lesions may enhance. Enhancement in- of monitoring the therapeutic response of the disease
dicates disruption of the blood–brain barrier, proba- to new drugs. It has been shown that changes in the
bly the first local change of a developing MS plaque. number of gadolinium-enhancing lesions during the
Inflammation and edema will follow. The enhance- course of the disease correspond better to changes in
ment lasts between 2 and 6 weeks. MRI provides the the EDSS than changes in lesion load on T2-weighted
possibility to identify acute, new or reactivated, images. The apparent advantage for therapeutic trials
lesions (Fig. 79.9). Enhancing lesions are often used in is the gain in time and the smaller study population
clinical trials as surrogate markers for disease activi- one has to examine to obtain statistical significance of
ty. Contrast-enhanced MRI can help, not only by results. The eventual clinical trial based on clinical re-
demonstrating “dissemination in space” with the sults can in this way be limited to the most promising
presence of more than one lesion in the CNS at differ- therapeutic compounds.
ent locations, but by also demonstrating “dissemina- Two manifestations of MS outside the brain de-
tion in time” in showing older nonenhancing or no serve special mention: optic neuritis and spinal cord
578 Chapter 79 Multiple Sclerosis

Fig. 79.2. The classical MR pattern of MS. The transverse T2- the ventricles, in the corpus callosum, in the mesencephalon,
weighted series in this 42-year-old woman shows lesions in the pons, around the fourth ventricle, and in the medulla ob-
the centrum semiovale, around the lateral upper borders of longata.There are no lesions in the basal ganglia

lesions, because they both require special MRI tech- The majority of MS lesions of the spinal cord occur
niques. in the cervical spinal cord and the conus medullaris.
The STIR sequence in particular has been effective In some patients lesions are solely present in the
in demonstrating optic neuritis. Other fat saturation spinal cord. They appear most often as high-signal-
techniques, using a saturation pulse to suppress the intensity lesions on proton density and T2-weighted
fat signal, can be combined with gadolinium en- images and may enhance with contrast (Fig. 79.13).
hancement (Fig. 79.12). There is, as a rule, no or very little mass effect. Mass
effect can, however, occasionally be considerable and
79.6 Magnetic Resonance Imaging 579

Fig. 79.3. Same patient as


in Fig. 79.2. It is useful in
patients suspected of having
MS to include the spinal cord
in the examination.The
disability of the patient is
often also or mainly related
to the spinal cord lesions

Table 79.3. Spinal lesions in patients with MS and patients Indications, techniques, and the role of asympto-
with other neurological disorders. The difference is statistical- matic spinal cord lesions in the differential diagnosis
ly significant (Bot et al. 2002)
from MS versus other diseases such as vascular dis-
Spinal cord Normal Abnormal orders have recently been summarized (Lycklama á
Nijeholt et al, 2003).
Other neurological disorders 39 4
(n = 43)
Many patients with definite MS do not fit the “typ-
ical” description given above. Nearly anything goes
MS (n = 25) 2 23
with MS. Lesions may show mass effect or become
cystic. The distribution may not be typical. Criteria as
suggested by Barkhof et al. (1997), incorporated in the
McDonald criteria (see Table 79.1), are often helpful
the lesion may then mimic an intramedullary tumor. in establishing the diagnosis.
When patients present with symptoms indicative of a Sometimes MS presents with clinical signs of
spinal cord lesion, MRI of the spinal cord may be the raised intracranial pressure and a tumefactive lesion
first requested examination. If a lesion indicative of on MRI, often enhancing. In quite a few patients this
MS is found, an MRI of the brain should follow to look observation has prompted a brain biopsy. The pres-
for corroborative evidence, in the form of lesions in ence of other white matter lesions and the “open ring”
the brain, to support the diagnosis of MS. The great- sign may lead to the correct diagnosis and manage-
est technical problems in detecting spinal cord MS ment of the patient (Fig. 79.18). The open ring sign is
lesions are encountered in the thoracic region. Phased often present in large, contrast-enhancing demyeli-
array coils have increased the signal-to-noise ratio nating lesions and consists of a partly enhancing ring
and detail in studies of the spinal cord. Conventional around the lesion (Masden et al. 2000).
dual-echo spin echo sequences with cardiac gating Much attention has also been given to clinical iso-
yield the best results (Fig. 79.14) (Lycklama á Nijeholt lated symptoms (CIS) that may or may not progress
et al. 1996). In more than 90% of patients with MS, to MS. This isolated symptom is often optic neuritis,
spinal lesions can thus be identified (Figs. 79.15– but other signs are possible. MRI is a powerful predic-
79.17). The detection of spinal lesions also helps in tor of the later diagnosis of definite MS, better
the differentiation of MS from vascular lesions than the presence of HLA-DR2 or CSF oligoclonal
(Table 79.3; Bot et al. 2002). bands.
580 Chapter 79 Multiple Sclerosis

Fig. 79.4. A 38-year-old woman suspected of having MS. The of the ventricles and the corpus callosum.There are no lesions
MR picture is compatible with this diagnosis. The transverse elsewhere in the brain
T2-weighted images show lesions around the lateral borders

Normal-appearing white matter (NAWM) has be- ues of normal-appearing white matter are increased
come an important issue in MRI of MS. MR tech- and FA values lower as compared to normal white
niques have confirmed the histopathological and matter in non-MS patients. Changes in metabolite
chemical observations that white matter that appears concentrations on MR spectra lead to the same con-
normal on T1-weighted, T2-weighted, and FLAIR im- clusion.
ages show diminished structural integrity when mag- The relationship between the lesions demonstrat-
netization transfer ratios (MTRs) are estimated ed on MRI and clinical findings is generally speaking
(Fig. 79.19). With diffusion tensor imaging, ADC val- poor. In patients with advanced disease of the sec-
79.6 Magnetic Resonance Imaging 581

Fig. 79.5. Same patient as in Fig. 79.4. The upper two rows dis- around the lateral ventricular borders. In this patient, too, mul-
play sagittal and parasagittal proton density images, revealing tiple lesions in the spinal cord confirm the diagnosis
the shape and extent of the lesions in the corpus callosum and

ondary progressive type, the correspondence be- hancement of lesions (Van Walderveen et al, 1995,
tween the developing atrophy and the number of 1998). Histopathologically, they correspond to areas
white matter lesions and the EDSS disability score is with severe matrix destruction and axonal loss (Van
better. Waesberghe et al. 1999).
In estimating the prognosis of MS, so-called black There is a growing interest in remyelination, part
holes have been proven to have a special place.“Black of the healing process of MS. Some lesions disappear
holes” are MS lesions that appear very dark on T1- from MRI in MS patients with follow-up MRI, where-
weighted images and bright on T2-weighted images as others do not. The question is whether the disap-
(Fig. 79.20). The presence of black holes correlates pearing lesions remyelinate and therefore become in-
better with a poor prognosis than the presence of le- visible, or shrink and turn into little gliotic scars. New
sions on T2-weighted images and gadolinium en- MR methods may be helpful in answering this ques-
582 Chapter 79 Multiple Sclerosis

Fig. 79.6. Transverse FLAIR series of a 40-year-old woman pre- mas. The surprise finding in this patient consisted of lesions
senting with a 6-week history of vertigo attacks. She had pre- supratentorially suggestive of MS and a distinct lesion on the
viously undergone operations on both ears for cholesteato- left side in the vestibular nucleus and nerve

tion, for example by demonstrating changes in the conclusion of this study was that T1-weighted images
MTR or ADC. Not all lesions that remyelinate neces- and MTRs have additional value in distinguishing le-
sarily disappear from the MR image; many maintain sions with and without remyelination (Barkhof et al.
a high signal on T2-weighted images. In a post- 2003). In addition, ADC values and contrast-en-
mortem study, MRI of the 1-cm-thick slices was used hanced T1-weighted images may also hold informa-
to detect lesions (Barkhof et al. 2003). In this study re- tion in patients with remitting MS (Fig. 79.21). MR
myelination was found in 42% of the lesions, the re- data obtained in demyelinating lesions induced by
myelination being partial in 19% and full in 23%. The lysophosphatidyl choline in animals showed high
79.6 Magnetic Resonance Imaging 583

Fig. 79.7. T1-weighted contrast enhanced images in the same patient as in Fig. 79.6.The two enlarged images on the right show
the enhancement of the lesions in the vestibular nucleus and tract (ring and arrow)

ADC values in the peripheral edema and moderately in fiber tracts when remyelination occurs. Other tech-
low values in the center of the lesions in the early niques may also be considered, such as functional
phase of the lesions. Substantial reduction of ADC MRI in combination with fiber tracking.
values occurs in both parts of the lesions with re- Magnetization transfer ratios (MTRs) are often
myelination (Degaonkar et al. 2002). used in follow-up studies. Changes in MTR reflect
Several special MR techniques have been shown to changes in structural integrity and correlate with
be effective in extracting more information on the na- axonal damage. MTR histograms of the whole brain
ture and degree of structural damage of brain tissue. or segments (white matter, gray matter) are used as a
They are mainly applied in research settings and in tool to measure progression or regression of the dis-
protocols of clinical trials. ease.
Diffusion-weighted imaging usually reveals a high Proton MR spectroscopy (1H-MRS) has added con-
to very high signal in hyperacute lesions and some- siderably to the understanding of the pathophysiolo-
times low ADC values (Fig. 79.22). It is difficult to gy of MS. 1H-MRS has been performed as a single
draw conclusions from that, because the underlying voxel technique or as chemical shift imaging (CSI) in
mechanism of the signal change in these cases is not acute, subacute, and chronic lesions, and also in nor-
completely understood. Low ADC values do not cor- mal-appearing white and gray matter, in particular in
respond entirely with enhancement, probably be- patients with relapsing-remitting MS and secondary
cause enhancement depends on leakage through the progressive MS. MRS can analyze the biochemical
blood–brain barrier, whereas low ADC values in MS changes and their time course. In acute lesions
lesions more likely reflect the initial inflammatory choline and lactate increase early in the demyelinat-
reaction. Low ADC values in MS are not indicative of ing process, reflecting inflammation and demyelina-
cytotoxic edema. Diffusion tensor imaging and fiber tion, followed in most lesions by a decrease in NAA,
tracking may be helpful in demonstrating the repair reflecting axonal loss. The amount of NAA loss (be-
584 Chapter 79 Multiple Sclerosis

Fig. 79.8. Juxtacortical lesions are important in a definite on the proton density (left) and T2-weighted images (middle).
diagnosis of MS, but can be difficult to identify on MR images. After contrast injection there is enhancement of one of the
In this series of three transverse images tiny lesions are noted lesions (arrows)

Fig. 79.9. On the left a proton density


image shows multiple lesions, sugges-
tive of MS.The T1-weighted contrast
enhanced image on the right shows
enhancement of some, but not of
all lesions, confirming dissemination
in time and dissemination in space

tween 28% and 66%) reflects the severity of axonal well as moderate NAA decrease and choline increase
damage, and correlates in a later phase with disability in normal-appearing white matter.
scores. Especially in patients with secondary progres- Special variants of MS may have a different MR ap-
sive MS, the loss of NAA is more pronounced than in pearance. MS can occur in very young children. As in
the more benign forms of MS. In 1H-MRS with short adults, the diagnosis has to be considered when mul-
echo times an increase in myo-inositol is seen, reflect- tifocal white matter abnormalities are seen, some of
ing gliosis. The presence of “lipids” may also be not- which may take up contrast after injection. The le-
ed. 1H-MRS with metabolite nulling allows further sions in young children tend to be larger, more often
identification of these “lipids” at 0.9 and 1.3 ppm and involving the basal ganglia and less often involving
identifies them rather as proteins and polypeptides, the lateral borders of the ventricles. Although the
probably products of myelin protein breakdown. numbers of childhood MS patients reported are
When confirmed, these resonances can be used as small, it seems that the incidence of brain stem le-
markers of myelin fragments. Over time, partial or sions is somewhat higher in children than in adults.
full recovery of NAA losses may occur. Full recovery As in adult MS, the contrast uptake disappears after
may be seen in patients with mild disease and mild to several weeks and the lesions become less conspicu-
moderate loss of NAA. Choline values decrease from ous on T2-weighted images (Figs. 79.23–79.25).
high in the initial phase to normal in a period of 2 In NMO, one does not necessarily expect lesions in
years. 1H-MRS has shown gray matter involvement as the cerebral white matter as the clinical condition
79.6 Magnetic Resonance Imaging 585

Fig. 79.10. Series of contrast-enhanced T1-weighted images at the ventricular level, obtained in successive months. Enhancing
lesions come and go.This type of serial study can be used as surrogate marker for disease activity in MS

mainly consists of problems of vision (optic neuritis) atypical cases of X-linked adrenoleukodystrophy
and paraplegia (transverse myelitis). In some cases, with frontal or parietal, asymmetrical lesions that
however, we have found multiple white matter lesions confusion with DS may exist. However, with laborato-
in the cerebral hemispheres. These MRI findings are ry investigations it is easy to distinguish X-linked
in accordance with histology (Fig. 79.26). adrenoleukodystrophy from DS. DS is very rare, but it
In DS the lesions are large, at least 3 ¥ 2 cm in size, should be borne in mind when diagnosing cases of
mostly localized in the parietal region on both sides severe white matter affection occurring in the first
and, in accordance with the original drawings two decades of life.
of Schilder, connected via the corpus callosum. From CS is easily recognizable on MRI and shows alter-
the drawings of Schilder’s original publication (Fig. nating layers of myelinated and unmyelinated fibers,
79.27), one can also conclude that the lesions will ap- leading to a striking appearance (Fig. 79.29). In CS the
pear symmetrical in some transverse sections but not MR images show the ring-like lesions, often already
in all. These lesions spare the arcuate fibers, unless the visible on the proton density and T2-weighted images,
disease is very advanced. After injection of gadolini- with an enhancing rim after contrast injection.
um there is enhancement of the peripheral active rim In the differential diagnosis of MS, it is important
(Fig. 79.28). It may be difficult to distinguish DS from to realize that many other conditions can lead to mul-
X-linked adrenoleukodystrophy on the basis of imag- tifocal lesions located chiefly in the white matter with
ing findings. Bilateral, confluent white matter lesions prolonged T1 and T2. They are found in arteriosclerot-
connected via the corpus callosum and showing rim ic disorders, head trauma, migrainous headache,
enhancement are also seen as a rule in X-linked vasculitis (including systemic lupus erythematosus,
adrenoleukodystrophy. However, in the latter disease other rheumatoid disorders, and Behçet disease), ear-
the location of the lesions is preferentially occipital ly metastatic lesions, leukemic deposits in the brain,
and the lesions are usually symmetrical. It is in the Lyme disease, neurosarcoidosis, toxoplasmosis, and
586 Chapter 79 Multiple Sclerosis

Fig. 79.11. A comparable series


to that in Fig. 79.10 at the level of
the brain stem, showing the same
phenomenon

as age-related white matter changes in the elderly. “MS-like” white matter lesions are often seen in the
Without further information, MRI cannot always reli- fifth and sixth decades of life, and that the incidence
ably differentiate between conditions mimicking the of MS is low in this age group, argues for the reser-
MR pattern of MS. However, MRI is not the only tool vation of MRI as a tool in the classification of MS for
which can be used to reach the diagnosis. Clinical research mainly in the under-50 age group. Spinal
evidence and the patient’s history, age, and laboratory imaging may be of considerable help in the differen-
findings may suggest a diagnosis other than MS. The tial diagnosis of vascular disorders versus MS (Figs.
fact that some of the above-mentioned causes of 79.30, 79.31) (Lycklama á Nijeholt et al. 2003).
79.6 Magnetic Resonance Imaging 587

Fig. 79.12. Optic neuritis of the left eye. The coronal STIR im-
age shows on the right side a normal optic nerve, with signal
intensity identical to the white matter in the brain. On the left
side the optic nerve has a bright signal, compatible with an
active neuritis of the optic nerve

Fig. 79.13. Spinal imaging is


extremely important in MS.
On the T2-weighted sagittal im-
age on the left an intramedullary
lesion is seen, enhancing on the
T1-weighted contrast enhanced
image on the right
588 Chapter 79 Multiple Sclerosis

Fig. 79.14. To assure high-quality images


of the spinal cord special techniques have to
be employed. In these two images (proton
density on the left, T2-weighted on the right)
cardiac gating was used to eliminate flow
artifacts. On the proton density image the
CSF is isointense with the spinal cord and
the lesions stand out

Fig. 79.15. The same technique is applied


in this case, demonstrating that even in a
case with severe atrophy and relatively
wide CSF spaces, the influence of flow
artifacts can be minimized.The lesions in
the atrophic cord are well visible
79.6 Magnetic Resonance Imaging 589

Fig. 79.16. The technique employed


in the previous images is also success-
ful in the thoracic area.The lesions
are well demonstrated.The transverse
magnetization transfer gradient echo
image (MT-FLASH) depicts the lesion
in the axial direction

Fig. 79.17. Transverse images of the spinal cord lesions are T2-weighted images combined with a series of transverse
important as well.There are several possible ways of obtaining T2-weighted images,demonstrating the position of the lesions
good transverse images. This figure shows a series of sagittal within the spinal cord structures
590 Chapter 79 Multiple Sclerosis

Fig. 79.18. Transverse T2-weighted


image and contrast-enhanced
T1-weighted image to demonstrate
the “open ring” sign.The lesion shown
on the T2-weighted image is difficult
to differentiate from a low-grade
astrocytoma or abscess. After contrast
the lesion shows ring enhancement,
but the ring is not fully closed.This
feature is rarely seen in tumors or
abscesses, more often in tumefactive
MS lesions

Fig. 79.19. These series of images


represents magnetization transfer
imaging.The upper row shows
two transverse images, respectively
T2-weighted (left) and T1-weighted
(right).The lower row shows a
T2-weighted image after a magnetiza-
tion transfer prepulse (left) and the
resulting magnetization transfer
map (right).This map allows direct
measurements of the MTR, either
pixel by pixel or by ROIs
79.6 Magnetic Resonance Imaging 591

Fig. 79.20. Three sagittal FLAIR


images show typical MS lesions.The
transverse T1-weighted image shows
“black holes.”These images relate to
a 43-year-old patient with a secondary
progressive form of MS, now with
severe disability and cognitive deteri-
oration.The presence of black holes
predicts a poor outcome
592 Chapter 79 Multiple Sclerosis

Fig. 79.21. This series of images


probably illustrates one of the
MR features of remyelination in a
remitting patient. Presented are
two FLAIR images (first row), two
T2-weighted images (second row),
two Trace diffusion-weighted images
with a b value of 1000 mm2/s (third
row) and two ADC maps (fourth row).
The FLAIR image shows a target
lesion, with a higher signal centrally
than the surrounding ring (arrow).This
lesion has a high signal on the Trace
diffusion-weighted image and low
signal intensity on the ADC map
(0.52–0.64 ¥ 10–3 mm2/s), and no
enhancement (not shown).The low
ADC values in this area probably
represent remyelination
79.6 Magnetic Resonance Imaging 593

Fig. 79.22. The upper row of transverse T2-weighted images row of T1-weighted contrast-enhanced images shows contrast
shows a number of lesions in a case of acute MS. The second uptake in some of the lesions.The ADC values in the lesions are
row shows three Trace diffusion-weighted images (b = 1000), just below normal
in which most of the lesions have a very high signal. The third
594 Chapter 79 Multiple Sclerosis

Fig. 79.23. MS in a girl of 6 years. The FLAIR images show a geniculate bodies, and medially involving the splenium of the
large lesion on the left side in the centrum semiovale, extend- corpus callosum. There is also a lesion on the right side of the
ing into the U fibers. A second lesion involves a large area at midbrain and pons. Courtesy of Dr. M.A.A.P Willemsen, Depart-
the right side near the trigonum, extending laterally into the ment of Pediatric Neurology, University Medical Center Nij-
basal ganglia, the internal capsule, the medial and lateral megen, The Netherlands
79.6 Magnetic Resonance Imaging 595

Fig. 79.24. Same patient as in Fig. 79.23. In children with MS, the transverse images at the indicated levels give further infor-
too, imaging of the spinal cord is important. The lesions in the mation about the position of the lesions in the cord
spinal cord are well seen on the T2-weighted sagittal images;
596 Chapter 79 Multiple Sclerosis

Fig. 79.25. Sagittal FLAIR images in a boy of 13 years shows the classic pattern of adult MS
79.6 Magnetic Resonance Imaging 597

Fig. 79.26. A set of images illustrating neuromyelitis optica. side. In this case the images through the head (proton density,
The sagittal T1-weighted contrast-enhanced images of the T2-weighted,T1-weighted with contrast) show a clinically silent
spinal cord show severe atrophy of the cord and multiple en- lesion with enhancement. In other cases no abnormalities in
hancing lesions.The coronal STIR image of the head shows an the brain are found
elevated signal of both optic nerves with atrophy on the left
598 Chapter 79 Multiple Sclerosis

Fig. 79.27. Illustration from the original paper of Schilder


(1912) on DS. The drawings show the extensive, asymmetrical
involvement of the frontal and parietal lobes, connected via a
lesion in the corpus callosum
79.6 Magnetic Resonance Imaging 599

Fig. 79.28. A 27-year-old man with a recent acute exacerba- Large lesions are seen on both sides in the frontal and parietal
tion of neurological signs and symptoms, including hemiple- regions, with an asymmetrical distribution, preponderance on
gia, aphasia, headache, and drowsiness. The MR tableau is a the left, and involvement of the corpus callosum connecting
collage of T2-weighted images (upper left) and T1-weighted im- the lesions on both sides. There is ring-like enhancement in
ages without and with contrast in three planes.Compare espe- the zone of acute inflammation. There was considerable im-
cially the coronal image with Schilder’s diagram (Fig. 79.27). provement after treatment with corticosteroids
600 Chapter 79 Multiple Sclerosis

Fig. 79.29. A 25-year-old woman


with CS. MRI shows a large lesion with
a concentric pattern of rings in the
white matter of the centrum semio-
vale on the right side.There is an
abrupt termination of the concentric
rings where the lesion abuts on the
gray matter. After contrast injection
only the outer rings enhance. From
Korte et al. (1994), with permission
79.6 Magnetic Resonance Imaging 601

Fig. 79.30. In patients over 50 years of age, differentiation be- sis MS.The lesions in the spinal cord make this diagnosis more
tween MS and vascular lesions may be difficult. In this patient certain. (Fig. 79.30 continued see next page)
the FLAIR images of the brain are compatible with the diagno-
602 Chapter 79 Multiple Sclerosis

Fig. 79.30. (continued).


79.6 Magnetic Resonance Imaging 603

Fig. 79.31. This 50-year-old patient has a few scattered lesions in the brain.The spinal cord images do not reveal any abnormal-
ity.This argues strongly against MS (see Table 79.3)
Chapter 80

Acute Disseminated Encephalomyelitis


and Acute Hemorrhagic Encephalomyelitis

80.1 Clinical Symptoms apparently independent of the nature and severity of


and Laboratory Investigations the antecedent infection or immunization.
There is a close relationship with the Guillain–
Acute disseminated encephalomyelitis (ADEM) is Barré syndrome, and in fact a combination of ADEM
an uncommon inflammatory disorder within the and Guillain–Barré syndrome has been described.
CNS, predominantly within the white matter of the Clinically, ADEM can usually be distinguished from
brain and spinal cord. “Postinfectious/postvaccina- multiple sclerosis, because it presents with signs of
tion encephalopathy” is a synonym. Poser used the severe multifocal involvement of the CNS at the same
term “disseminated vasculomyelinopathy,” indicating time. If initially monosymptomatic with optic neuri-
the probably causative small vessel vasculopathy tis, the optic neuritis tends to be bilateral. No clinical
(Poser 1969). Many other synonyms exist. feature, however, is exclusive to one or the other dis-
ADEM and acute hemorrhagic encephalomyelitis order.
(AHEM) represent two clinical variants of a single Laboratory investigations reveal a peripheral
pathological process. They follow a viral infection or leukocytosis. The CSF shows pleocytosis. The cellular
vaccination or occur without recognized antecedent. exudate is mainly lymphocytic and rarely exceeds
Implicated viral infections are measles, chickenpox, 100 cells/ml in ADEM. In AHEM the initial cells are
rubella, smallpox, infectious mononucleosis, herpes granulocytic, and erythrocytes are also often seen.
simplex, herpes zoster, mumps, influenza, and Myco- Glucose concentration is normal. There is usually a
plasma pneumoniae infection. They rarely occur fol- mild elevation of the CSF proteins, with an oligoclon-
lowing a bacterial infection. A nonspecific upper res- al banding pattern and a heightened IgG index as
piratory tract infection is the most common an- signs of intrathecal IgG synthesis. An elevated level of
tecedent. The latent period varies from several days to myelin basic protein is shown, indicating myelin de-
several weeks, the mean being 4–6 days. Onset of the struction. CSF abnormalities may persist for a long
neurological symptoms is often abrupt, with convul- time after clinical recovery. Rarely, the CSF is com-
sions and progression to somnolence and coma. Ill- pletely normal throughout the course of the illness.
ness may also commence subacutely with symptoms The EEG often shows bilateral slow activity. These ab-
of headache, fever, irritability, drowsiness, and vomit- normalities are nonspecific and add little to the diag-
ing. Nuchal rigidity may be present. The neurological nosis.
signs are quite polymorphic and consist of hemiple-
gia, diplegia or tetraplegia, cerebellar ataxia, cranial
nerve palsies, optic neuritis, nystagmus, sensory loss, 80.2 Pathology
and bladder dysfunction. Subcortical blindness is
rare. Extrapyramidal movement abnormalities such Edema is often the most conspicuous gross finding in
as chorea, athetosis, and ballismus occur. The pro- ADEM, but the external appearance of the brain may
gression of the disease is variable. Patients with AHEM also be completely normal. Microscopic examination
have a more severe clinical course and progress more demonstrates a diffuse inflammatory process in the
rapidly into delirium and coma. The highest mortali- brain, brain stem, and spinal cord, with inflammatory
ty is seen during the first week of the illness, and in cells around veins and venules. Sometimes the
fact most patients who survive the first week eventu- perivascular infiltrates are associated with signs of
ally recover, with varying degrees of disability. Pro- vasculitis, showing inflammatory cells in vessel walls
longed disturbances in level of consciousness entail a with or without frank necrosis. The cerebral hemi-
poor prognosis both for morbidity and mortality. spheres are usually more or less symmetrically in-
Possible neurological sequelae include epilepsy, spas- volved. Both the white matter and gray matter are
tic paresis, ataxia, decreased vision, and cognitive and affected, but the white matter shows more severe
psychiatric disturbances, but many patients have no changes. The most distinctive histological change is
remaining deficits. Most of the neurological syn- perivascular demyelination. Within the lesions
dromes have a monophasic course lasting several myelin sheaths are lost, but the axons are relatively
weeks. Recurrent attacks of ADEM/AHEM may occur. unaffected. Occasionally the demyelinating lesions
The occurrence of the neurological abnormalities is are confluent and form large demyelinated areas. At
80.3 Pathogenetic Considerations 605

later stages of the disease, the inflammatory exudate to nervous tissue antigen and especially to myelin ba-
is replaced by fibrous gliosis. sic protein has been shown in a variety of postinfec-
In AHEM, necropsy reveals the brain to be swollen tious neurological disorders, including ADEM and
and soft in consistency. On sectioning, numerous AHEM. Experimental allergic encephalomyelitis re-
small hemorrhages are seen, mainly in the white mat- sults from T cell sensitization to myelin basic protein.
ter. The cerebral cortex and basal ganglia are often The delayed hypersensitivity to myelin basic protein
spared. The hemorrhagic lesions are often but not al- leads to an attack on myelin sheaths with subsequent
ways symmetrical. Confluence of many small lesions demyelination. The demyelination is predominantly
leads to large hemorrhagic lesions. On microscopic perivascular as the responsible T cells originate from
examination the abnormalities are seen to be related the blood. ADEM and AHEM may represent the
to blood vessels, predominantly small veins but also human counterparts of experimental allergic ence-
small arteries. There is necrosis of vessel walls, with phalomyelitis with breakdown of tolerance to myelin
fibrinous exudation, perivascular edema, hemor- antigens. It is possible that the viral proteins serve as
rhages, neutrophilic granulocytes in the vessel walls, an antigen that cross-reacts with myelin antigens. It is
perivascular spaces, and adjacent brain tissue, and also possible that during the initial phase of viral in-
perivascular demyelination. The demyelination is vasion there is subclinical involvement of the CNS,
usually associated with necrosis and at least some with release or exposure of sequestered neural anti-
loss of axons. In severely necrotic areas all axons may gens and subsequent sensitization to them. These the-
disappear. Perivascular demyelination is also seen ories, however, do not explain the rarity of ADEM and
surrounding apparently normal vessels. AHEM following a viral infection of the CNS, such as
Both in ADEM and in AHEM the severity, type, and herpes virus or cytomegalovirus, nor its low inci-
localization of the pathological changes are unrelated dence in general. How a wide variety of infections and
to the type of preceding disease. In both syndromes vaccinations can induce similar sensitization to
the parenchymal pathology is often associated with myelin antigens has also not been explained satisfac-
meningeal lymphocytic infiltrates. torily.
An alternative theory stresses the importance of
the vascular changes, which are almost invariably
80.3 Pathogenetic Considerations present, and has coined the term “disseminated vas-
culo-myelinopathy.” The detection of circulating anti-
The early theories on ADEM and AHEM speculated gen–antibody complexes in the serum of patients
that these syndromes might represent a delayed but with a variety of postinfectious neurological disor-
direct invasion of the CNS by a virus or reactivation ders led to the assumption of a vascular lesion due to
of a latent virus. There has, however, been a consider- the entrapment of immune complexes in vessel walls
able amount of evidence against these theories. The and the subsequent inflammatory response. Because
pathological changes are fairly uniform and quite un- perivascular demyelination can result from vascular
like those of the viral encephalitides, the demonstra- injury alone, the participation of delayed hypersensi-
tion of viral antigens or viral particles in the brain or tivity would not be necessary. The presence or ab-
the CSF being an exception. sence of immune complexes, the size and the number
Some observations led to the theory that ADEM of the complexes, and not the antecedent illness
and AHEM represent an autoimmune response to would thus be the major factor in the development of
myelin constituents. First of all, the same neurological ADEM and AHEM, and the development of delayed
syndrome was found in patients vaccinated with ra- hypersensitivity to myelin antigens would be merely
bies vaccine grown in rabbit spinal cord. Secondly, a an epiphenomenon resulting from nervous tissue
comparable neurological syndrome was reported in damage. However, it is not clear why the nervous sys-
animals after the repeated injection of CNS ho- tem should be preferentially involved in immune-
mogenate in combination with Freund’s adjuvant. complex-mediated vasculitis. Only some indirect evi-
This syndrome is known as experimental allergic en- dence supports this theory: the detection of circulat-
cephalomyelitis. Pathologically, the lesions of ADEM ing complexes in some patients with various post-
and experimental allergic encephalomyelitis have infectious neurological disorders, the presence of
marked similarities. The white matter shows perivas- systemic features compatible with immune complex
cular cuffing by inflammatory cells and demyelina- disease in occasional patients, and the occurrence of
tion. The cerebral hemispheres, cerebellum, brain similar nervous system abnormalities in other hu-
stem, and spinal cord are involved.A hyperacute form man disorders caused by immune complexes. Im-
of allergic encephalomyelitis has been induced in an- mune complexes have, however, not been found at
imals with alteration of the immunization regimen. sites of vessel injury, and it is not impossible that the
Pathologically, more hemorrhagic features are pre- complexes themselves only represent an epiphenom-
sent, similar to AHEM. Sensitization of lymphocytes enon.
606 Chapter 80 Acute Disseminated Encephalomyelitis and Acute Hemorrhagic Encephalomyelitis

The two hypotheses presented here are not mutu- is exceptional. Smaller, multiple sclerosis-like lesions
ally exclusive. Immune complex vasculitis results in occur in a small number of patients. The lesions have
increased vascular permeability. Changes in vascular a preference for the occipital and parietal area. The
permeability and perivascular inflammation can ei- white matter lesion may “spill into” the cortex with
ther alter the antigenicity of myelin or release an anti- some focal cortical involvement (Fig. 80.3). Mass ef-
gen previously sequestered by a competent blood– fect is rare but tumefactive lesions in the frontal and
brain barrier. The cell-mediated immune response parietal lobes have been described. It should be noted
could then perpetuate the damage and lead to de- that less frequently ADEM presents with involvement
myelination. In some patients perivascular demyeli- of the basal ganglia and brain stem (Fig. 80.4). Spinal
nation is seen, whereas in others the pathological pic- MR is mandatory for a comprehensive inventory of
ture is dominated by perivascular necrosis. It is hy- lesions (Fig. 80.4). In nearly all cases spinal lesions
pothesized that pure demyelination may be caused by can be demonstrated. Incidentally spinal lesions are
delayed hypersensitivity to myelin antigens alone and the sole MR presentation of the disease. Some pa-
that necrosis associated with inflammatory infiltra- tients have optic neuritis (Fig. 80.4).After contrast in-
tion occurs when there is production of antibodies jection the white and/or gray matter lesions may en-
directed against several components of the brain hance (Fig. 80.3), but usually not all lesions enhance,
parenchyma. and contrary to what is usually stated, our experience
is that in many cases enhancement is at most subtle
(Fig. 80.2), or is not present at all (Fig. 80.1). The
80.4 Therapy enhancement is often patchy, while sometimes the
whole lesion enhances. Ring enhancement may occur.
Corticosteroids are central in the management of The presence of both enhancing and nonenhancing
ADEM and AHEM. Dramatic clinical improvement lesions argues in favor of the lesions’ being in differ-
may be seen. Relapses may occur when steroids are ent stages of development, despite the supposed
withdrawn, and improvement may then recur when monophasic character of the disease. Repeated MRI
steroids are reinstituted. The fact that the improve- during the course of the disease may show disappear-
ment is sometimes absent or only slight may be ex- ance of some lesions and concurrent appearance of
plained by the presence of irreversible structural others. These observations modify the view that
damage prior to institution of the steroid therapy. It is ADEM is always simply monophasic. Disappearance
recommended that corticosteroid therapy be initiat- of white matter lesions is sometimes rapid (Fig. 80.5),
ed as soon as possible in the treatment of these syn- but may also take a long time, as much as 18 months,
dromes. and part of the white matter damage may be perma-
In desperate cases, alternative therapeutic strate- nent (Fig. 80.6). New lesions may appear despite clin-
gies may include more drastic immunosuppression ical improvement.
and plasmapheresis. Diffusion-weighted imaging in ADEM reveals sim-
ilar results as in multiple sclerosis. In the peracute
phase of the lesion ADC values may be low; after 2–3
80.5 Magnetic Resonance Imaging weeks, the ADC values are higher than average in the
core of the lesion and mixed in the borders; in the late
In ADEM, CT scan of the brain shows multifocal or phase the abnormal findings tend to disappear.
diffuse white matter damage, but it may be normal in Magnetization transfer ratios (MTRs) have been
the acute stage, or may remain normal throughout. estimated in ADEM. Histogram MTR analysis of nor-
Not all lesions show enhancement after contrast in- mal-appearing white matter in patients with multiple
jection. CT can seldom explain the full extent of clin- sclerosis and ADEM show significantly lower MTRs
ical disability, and, conversely, several lesions may be and peak positions and significantly higher diffusivi-
seen for which no correlative clinical signs are ob- ty in multiple sclerosis than in ADEM, suggesting that
served. Clinical improvement is accompanied by dis-
appearance of contrast enhancement and complete or
partial resolution of low-density lesions.
In AHEM the white matter lesions are character- 䊳

ized on CT by extensive edema with prominent mass Fig. 80.1. A 2-year-old girl with ADEM.The T2-weighted trans-
effect and the presence of hemorrhages. verse images (first and second rows) show large, bulky lesions
MRI in ADEM shows multiple, usually large white in the centrum semiovale, predominantly in the frontal lobe.
matter lesions with an asymmetrical distribution There are also lesions in the midbrain and pons. This is the
(Figs. 80.1–80.3). In very extensive cases, in which “common” pattern of ADEM. On the T1-weighted contrast-
almost all white matter is involved, the asymmetrical enhanced images (third and fourth rows) no enhancement of
distribution becomes less clear. Symmetry of lesions lesions is seen, despite the acute clinical presentation
80.5 Magnetic Resonance Imaging 607

Fig. 80.1.
608 Chapter 80 Acute Disseminated Encephalomyelitis and Acute Hemorrhagic Encephalomyelitis

Fig. 80.2. A 5-year-old girl with a rapidly progressive presen- left middle cerebellar peduncle.T1-weighted coronal contrast-
tation of ADEM. The transverse FLAIR images (upper two rows) enhanced images (third row) show only a few tiny spots of en-
show many lesions dispersed throughout the brain, with ex- hancement
ception of the temporal lobes.There is also a large lesion in the

in ADEM the normal-appearing white matter is less tion or vaccination, or when the disease presents with
involved than in multiple sclerosis. These measure- a single symptom such as optic neuritis, it is impossi-
ments have been made outside of the acute phase. ble to differentiate between acute multiple sclerosis
Apart from the difference in preferential involve- and ADEM on basis of MR images. In a later phase,
ment of brain structures, it may be difficult to distin- comparison as described of MTRs and MRS in nor-
guish ADEM from multiple sclerosis in the initial mal-appearing white matter can help in the decision.
phase. Without a clinical history of a preceding infec- Repeated MRI examinations over a long period of
80.5 Magnetic Resonance Imaging 609

Fig. 80.3. In this 3-year-old girl with ADEM the T2-weighted tex. After contrast injection the T1-weighted images (third row)
transverse images (upper two rows) show large lesions that in- show enhancement of many lesions
volve the subcortical white matter,with extension into the cor-

time may also be helpful, in particular in combination when the patient is asymptomatic. New lesions on
with follow-up of the clinical course. Stationary last- MRI repeated 3 months after the attack is considered
ing lesions are indicative of ADEM, while newly ap- to be predictive of multiple sclerosis. However, some
pearing lesions argue against this diagnosis, but this ADEM patients have multiple episodes in the course
is not an absolute rule. New lesions may appear in of several years.
ADEM despite ongoing clinical improvement, but In exceptional cases, one may find a clinical picture
new lesions do, as a rule, not appear after the attack suggestive of ADEM while MRI shows predominantly
610 Chapter 80 Acute Disseminated Encephalomyelitis and Acute Hemorrhagic Encephalomyelitis

Fig. 80.4.
80.5 Magnetic Resonance Imaging 611

or exclusively gray matter lesions (Fig. 80.7). These In AHEM, the hemorrhagic component can be
gray matter lesions may disappear. Probably the “gray identified on CT, but also with confidence by MRI, ei-
matter ADEM” is the counterpart of the “regular ther by FLAIR or by gradient echo techniques. The
ADEM,” just as Guillain–Barré syndrome is usually a hemorrhagic aspect, which is highly unusual in mul-
demyelinating polyneuropathy, but is sometimes ax- tiple sclerosis and its variants, may help to establish
onal. the correct diagnosis (Fig. 80.8).

䊴 䊳

Fig. 80.4. MRI in a 6-year-old girl presenting with lowered Fig. 80.5. An 11-year-old boy with ADEM. In the acute stage
consciousness, spastic paraparesis, and diminished vision of (first two rows), the T2-weighted transverse images show ex-
the right eye. The FLAIR images (first row) show a gray matter tensive, patchy involvement of the white matter in the cen-
lesion, a large lesion in the right thalamus with some mass ef- trum semiovale, bilateral and symmetrical involvement of the
fect.A coronal STIR image (second row,left) shows swelling and internal capsule, involvement of the pulvinar, and also sym-
too high a signal in the right optic nerve, representing optic metrical involvement of structures in the pons. At follow-up,
neuritis. The transverse T1-weighted images after contrast in- 2 weeks later (third and fourth rows), the lesions have disap-
jection (second row, middle and right) show enhancement of peared completely, reflecting full clinical recovery.
the right optic nerve.The third row shows the spinal images. A (Fig. 80.5 see next page)
T2-weighted sagittal image of the cervical and upper thoracic
cord (left) shows a large intramedullary lesion in the upper cer-
vical cord. The proton density image (middle) reveals multiple
additional smaller lesions in the thoracic cord. A T1-weighted
contrast-enhanced sagittal image of the spinal cord (right)
shows enhancement of multiple lesions
612 Chapter 80 Acute Disseminated Encephalomyelitis and Acute Hemorrhagic Encephalomyelitis

Fig. 80.5.
80.5 Magnetic Resonance Imaging 613

Fig. 80.6.
614 Chapter 80 Acute Disseminated Encephalomyelitis and Acute Hemorrhagic Encephalomyelitis

Fig. 80.6. (continued). In a 6-year-old girl with ADEM and a spicuity.The ADC values were low (–30 %). Follow-up MRI after
serious presentation, the T2-weighted images (upper two rows) 6 months (fourth and fifth row) shows cortical and central atro-
show multifocal involvement of subcortical and deep white phy and some ill-defined areas of signal abnormality in the
matter. Diffusion-weighted–Trace (b = 1000) images (third cerebral white matter
row) show high signal in the affected regions with greater con-

Fig. 80.7. In this 2.5-year-old boy with ADEM, the lesions are predominantly located in the gray matter. Exceptions are lesions
in the middle cerebellar peduncle on the left side
80.5 Magnetic Resonance Imaging 615

Fig. 80.8. An 8-year-old boy, acutely ill following a minor in- rolandic area shows too low a signal intensity, caused by the
fection a few weeks before admission.The T2-weighted images presence of breakdown products of blood, including hemo-
show involvement of the pons and mesencephalon, especially siderin. The presence of considerable hemorrhage classifies
on the right side, and large, confluent areas of abnormal white this patient as a case of AHEM. Courtesy of Dr. A. Goulao, Lis-
matter in the cerebral hemispheres. The gyral pattern in the bon, Portugal
Chapter 81

Acquired Immunodeficiency Syndrome

81.1 Clinical Features herpes simplex encephalitis, herpes zoster encephali-


and Laboratory Investigations tis and radiculitis, cytomegalovirus encephalitis,
papovavirus infection with progressive multifocal
The acquired immunodeficiency syndrome (AIDS), leukoencephalopathy, infection with Aspergillus fu-
as defined according to the criteria of the Centers of migatus, Candida albicans, and Cryptococcus neofor-
Disease Control, is a state characterized by one or mans, nocardiosis, coccidioidomycosis, Mycobacteri-
more opportunistic diseases indicative of underlying um tuberculosis infection, atypical mycobacterial in-
cellular immunodeficiency, in the absence of underly- fections, toxoplasmosis, and neurosyphilis. However,
ing causes of cellular immunodeficiency other than only a minority (about 30%) of the CNS affections
human immunodeficiency virus type 1 infection and can be attributed to opportunistic infections. A pri-
in the absence of all other causes of reduced resis- mary HIV-1 infection is the most common cause of
tance reported to be associated with opportunistic neurological dysfunction. Acute diffuse leukoen-
diseases. The opportunistic diseases in AIDS include cephalitis, subacute encephalitis, aseptic meningitis,
opportunistic infections and neoplasms that can re- vacuolar myelopathy, and inflammatory demyelinat-
sult from immunodeficiency. AIDS is caused by a ing peripheral neuropathy are generally assumed to
retrovirus named human immunodeficiency virus be caused by direct HIV-1 infection. It should be not-
type 1 (HIV-1). This virus has previously been desig- ed that the following clinical descriptions refer to
nated human T cell lymphotropic virus type III untreated patients. With treatment the disease course
(HTLV-III), lymphadenopathy-associated virus (LAV), will be halted or modified.
and AIDS-related virus (ARV). High-risk groups in Acute diffuse leukoencephalitis has been de-
non-Third-World countries have been defined as scribed in a limited number of cases. Clinically this
homosexual or bisexual males, intravenous drug disease is characterized by rapid mental deteriora-
abusers, immigrants from Haiti and Central Africa, tion, progressive tetraparesis, and in some cases death
blood transfusion recipients, heterosexual partners within a few days.A relapsing-remitting course of dis-
of HIV-infected individuals, children of mothers in- ease has also been described, indistinguishable from
fected with HIV, and hemophiliacs who have received multiple sclerosis.
factor VIII concentrate. About 72% of AIDS patients Subacute HIV-1 encephalitis, also called AIDS en-
originate from the first of these risk groups and 17% cephalopathy or AIDS dementia complex, is the most
from the second group. The other risk groups consti- frequent neurological manifestation of AIDS and
tute only minor percentages of the total AIDS patient eventually afflicts many AIDS patients. Subacute HIV-
population. 1 encephalitis usually develops after other complica-
AIDS can manifest itself in many different ways. tions of AIDS have appeared, but it may also be the
Most patients present with malignant tumors and/or first major or even the sole clinical manifestation of
infections that are infrequently seen in immunocom- HIV-1 infection. In subacute HIV encephalitis cogni-
petent individuals. The most common tumors in tive, motor, and behavioral abnormalities are usually
AIDS are Kaposi sarcoma, primary CNS lymphoma, early features. Early cognitive abnormalities are im-
systemic non-Hodgkin lymphoma, and plasmocy- paired memory, loss of concentration, confusion, and
toma. The opportunistic infections most frequently slowing of mentation and movement. The onset of de-
include Pneumocystis carinii pneumonia, toxoplas- mentia is usually insidious, but a rapid onset over a
mosis, cryptococcal meningitis, candidiasis of the up- period of a few days and accelerations are not rare.
per gastrointestinal tract, and many infections with Behavioral abnormalities include apathy, social with-
other viruses, fungi, mycobacteria, and parasites. drawal, dysphoric mood, organic psychosis, and re-
Nonspecific signs and symptoms of the disease are gressive behavior. Early motor symptoms are lack of
weight loss, fatigue, malaise, night sweats, fever, cysts balance, weakness of the legs, tremor, and loss of co-
in the parotid glands and generalized lymphadeno- ordination. Neurological examination often reveals
pathy. Neurological signs and symptoms are frequent ataxia and pyramidal tract signs. The majority of pa-
and are reported in 30–75% of patients. Malignant tients develop a severe and global dementia within 2
lymphoma and metastatic Kaposi sarcoma may affect months after the onset of symptoms, but sometimes
the CNS. Opportunistic infections of the CNS include the initial course is more protracted, and a mild or
81.1 Clinical Features and Laboratory Investigations 617

moderate impairment of intellectual functions is pre- of grades I, II, and III can be distinguished. A steadily
sent for several months prior to the subsequent onset progressive spastic–ataxic paraparesis, which usually
of a more severe global dementia. The dementia of evolves over several weeks to months, is most charac-
subacute HIV-1 encephalitis has been described as teristic of patients with grade III, severe myelopathy.
subcortical because of the relative absence in many Signs and symptoms are similar but less severe in pa-
patients of seizures and other signs of focal cortical tients with grade II myelopathy. Clear myelopathic
involvement. The affected patients usually remain signs are infrequent in patients with grade I myelopa-
alert. Neurological symptoms in the end stages are thy.
variable and include aphasia, ataxia, hypertonia, Patients with AIDS are susceptible to a number of
motor weakness with paraparesis, quadriparesis or PNS complications. Peripheral nerve affections occur
hemiparesis, pseudobulbar palsy with dysphagia and in about half of the patients with subacute encephali-
dysarthria, tremor, blindness, extrapyramidal signs tis. Cranial nerve signs are less prominent.Apart from
with rigidity, incontinence, myoclonus, epileptic herpes zoster radiculitis, a distal symmetrical periph-
seizures, and sometimes organic psychosis. Retinopa- eral polyneuropathy, chronic inflammatory demyeli-
thy is found in about one-third of the patients with nating polyradiculoneuropathy, and mononeuritis
subacute HIV-1 encephalitis. Cotton wool spots are multiplex have been described. Distal symmetrical
the most frequently observed retinal abnormalities; peripheral neuropathy manifests itself by distal sen-
less frequently a hemorrhagic retinitis is seen. Optic sory disturbances and a mild degree of weakness in
neuritis is a rare finding. Death usually occurs within the distal muscles of the lower extremities. The symp-
1 year after the first signs of encephalopathy are noted. toms are predominantly sensory with burning,
Other neurological complications include cerebral painful paresthesias and numbness. In more ad-
hemorrhage and cerebral infarction. Thrombocy- vanced stages the muscular weakness may become
topenia predisposes AIDS patients to cerebral hemor- more marked and spread to the arms. Chronic in-
rhage, while nonbacterial thrombotic endocarditis flammatory demyelinating polyradiculoneuropathy
may lead to cerebral infarction. resembles the Guillain–Barré syndrome but the
The majority of children with AIDS are born to course is subacute or, more often, chronic. Weakness
women who have AIDS or pre-AIDS themselves and is most severe in distal muscles but also affects prox-
who are intravenous drug abusers, sexual partners of imal muscles. Sensory abnormalities are usually less
male members of high-risk groups, or of Haitian ori- marked. In severe cases weakness becomes general-
gin. In a smaller percentage of the affected children, ized, and facial diplegia, bulbar weakness, and respi-
transfusion-associated AIDS has been implicated. ratory difficulties result. In mononeuritis multiplex,
Clinical characteristics of AIDS in children include nerve dysfunction is relatively abrupt in onset. In the
failure to thrive, generalized lymphadenopathy, he- course of time, multiple nerves become involved, re-
patosplenomegaly, recurrent bacterial infections, and sulting in patchy weakness and sensory loss.
infections with opportunistic agents. Neurological Diagnosis of HIV-1 infection cannot be made with
abnormalities are variable. The most consistent find- standard serological tests in the early phase of the dis-
ings are acquired microcephaly and moderate to se- ease. Recombinant enzyme-linked immunosorbent
vere mental retardation. Motor deficits ranging in assays (ELISAs) are commonly negative in the early
severity include pyramidal tract signs with spastic phase, as are tests for antibodies. Serological tests be-
paraparesis or tetraparesis and pseudobulbar palsy. come positive after 3–4 weeks. Early diagnosis is pos-
Hypotonia with hyperreflexia is frequent in affected sible with the serum or plasma viral p24 antigen test,
children, especially early in the course of disease. In also used in blood donors to detect viral infection. Es-
some children, psychomotor retardation is stationary. timation of plasma viral RNA levels is even more sen-
In others progressive mental and motor deterioration sitive than the p24 antigen test. Counts of CD4+ lym-
occurs with a remitting course. Subacute HIV-1 en- phocytes give important information of the immune
cephalitis can be found in this latter group of patients. status of the patient. Studies of T lymphocyte subpop-
Vacuolar myelopathy is a common neurological ulations in AIDS reveal a depletion of CD4+ T helper
complication in AIDS. It is frequently difficult to cor- cells with an inversion of the CD4+ helper to CD8+
relate spinal cord abnormalities with clinical symp- suppressor cell ratio. In active HIV-1 infections the
toms and signs in individual patients since many pa- count is low. The most specific diagnosis of HIV in-
tients have coexistent encephalopathy or peripheral fection is by direct identification of the virus in tissue
neuropathy. Clinical features may be a spastic mono- or blood of suspected patients or indirectly by
paresis, paraparesis, or tetraparesis with hyperreflex- demonstrating the presence of antibodies to the
ia and extensor plantar reflexes, but sometimes re- virus. Longitudinal studies have shown that in serum,
flexes are absent. Incontinence and sexual distur- HIV-1 antigens appear early and transiently after pri-
bances are frequent, as are sensory abnormalities mary HIV-1 infection. Antibody production follows,
with sensory ataxia. Neuropathologically, myelopathy after which HIV antigens may disappear.
618 Chapter 81 Acquired Immunodeficiency Syndrome

Immunological investigations may include tests Conduction block is a common finding. EMG is either
demonstrating cutaneous anergy to recall antigens, normal or shows signs of denervation and reinnerva-
depressed response in lymphocyte transformation tion.
assays, and usually a decreased natural killer cell ac-
tivity. Lymphopenia may be present. These immuno-
logical abnormalities may be absent early in the dis- 81.2 Pathology
ease. Abnormalities in B cell function and humoral
immunity have been reported. Immunoglobulin lev- It has been estimated that approximately 40% of pa-
els may be high due to nonspecific polyclonal B cell tients with AIDS have clinically apparent CNS dys-
activation. Circulating immune complexes may be function. At autopsy, neuropathological abnormali-
present. Serological studies for a number of infections ties are found in about 80% of patients, and 20%
are frequently positive, such as hepatitis A and B, her- of patients have multiple coexisting CNS disease
pes group viruses, and toxoplasmosis. If indicated, processes. Neuropathological signs of subacute HIV-1
cultures of body fluids for bacteria, mycobacteria, encephalitis are found in about 50% of patients with
viruses, and fungi may be performed. AIDS, and it is accordingly the most common CNS
The CSF of patients with HIV infection of the CNS illness associated with AIDS.
usually has slightly raised protein content and less of- The brains of patients with subacute HIV-1 en-
ten a mononuclear pleocytosis with cell counts of up cephalitis, also called giant cell encephalitis or mi-
to 50 cells/mm3. The CSF of children with AIDS en- croglial nodule encephalitis, usually show some de-
cephalopathy shows these abnormalities less often. gree of atrophy at postmortem examination. Mild to
The CSF of patients with AIDS encephalopathy often moderate ventricular dilatation is associated with an
contains oligoclonal immunoglobulin bands, which apparent reduction of cerebral white matter volume
have anti-HIV activity. CSF anti-HIV antibodies are and shrunken cortical gyri. The most prominent
present in most AIDS patients, irrespective of the microscopic abnormalities in the brain involve the
presence of neurological complications. These anti- white matter and deep gray matter. Subacute HIV-1
bodies occur early in the course of the disease, indica- encephalitis is characterized by intraparenchymal
tive of an early seeding of HIV to the CNS. A longitu- and perivascular infiltrations of lymphocytes and
dinal study showed that HIV-1 antigen is present macrophages, located in both gray and white matter.
transiently in CSF before the occurrence of antibod- The perivascular infiltrations occur typically around
ies. HIV-1 antigen continues to be present in children capillaries and venules and are most frequently de-
and adults with progressive AIDS encephalopathy. As tected in the centrum semiovale, basal ganglia, and
a rule, it is not found in the CSF of asymptomatic pons. In mild cases the infiltrates are lymphocytic
seropositive controls, some of whom have anti-HIV and scanty. In more severe cases, inflammatory infil-
antibodies in the CSF. Persistence of HIV-1 antigens trates are composed mainly of foamy macrophages, in
in the CSF most probably reflects ongoing CNS in- some instances intermixed with multinucleated giant
volvement. HIV-1 antigens in the CSF are not a result cells. These multinucleated cells are thought to be de-
of leakage of viral proteins through the blood–brain rived from macrophages as they display macrophage
barrier, as in some patients the quantity of HIV-1 markers, and are considered to be a histopathological
antigens is larger in CSF than in serum, whereas oth- hallmark of HIV-1 encephalitis.
er patients have extremely high serum levels and no The most common white matter abnormality is
HIV-1 antigens in the CSF. The same arguments apply diffuse pallor, demonstrated by myelin staining. The
to anti-HIV antibodies, proving that the synthesis of borders of these large areas of pallor are ill defined.
these CSF antibodies occurs within the blood–brain The subcortical white matter is relatively spared.
barrier. There is some variability in the regional intensity of
EEG often shows moderate, generalized slowing of the pallor, which is most prominent in the centrum
the background pattern in subacute HIV-1 encephali- semiovale. The myelin loss is accompanied by a loss of
tis, occasionally with non-periodic high-voltage spik- axons, but the axons are relatively better preserved.
ing. Even in patients with seizures or myoclonic jerks, Diffuse reactive astrocytosis occurs and generally
paroxysmal discharges are not usually seen. In pa- parallels the inflammatory and parenchymal changes.
tients with distal symmetrical peripheral neuropathy In addition, most patients have small, poorly defined
the nerve conduction velocity is normal or slightly re- foci of more complete demyelination. These usually
duced. EMG shows some signs of denervation in dis- have a perivascular distribution. The lesions are
tal muscles. In chronic inflammatory demyelinating associated with inflammatory infiltrates, which are
polyneuropathy a marked slowing of nerve conduc- composed of lymphocytes, lipid-laden foamy macro-
tion is the rule. The abnormalities are frequently phages, reactive astrocytes, microglia, and sometimes
patchy. F responses are prolonged or absent, which is multinucleated cells. Another common finding is
in conformity with a component of radiculopathy. vacuolation of the white matter, most frequently ob-
81.2 Pathology 619

served in the centrum semiovale and less commonly In multiple sclerosis-like leukoencephalopathy,
in the internal capsule, brain stem, and cerebellum. multiple, large, well-defined areas of demyelination
Electron microscopic examination shows intramye- can be shown, which may involve hemispheric white
linic vacuoles. matter, internal capsule, basal ganglia, optic tracts,
The cerebral cortex is relatively normal.Astrocyto- corpus callosum, brain stem, and cerebellum. These
sis is usually found only in the deep cortical layers. lesions are characterized by myelin loss with presence
Considerable neuronal damage and loss is seen only of lipid-laden macrophages and preservation of ax-
in cases of severe white matter changes. Inflammato- ons and nerve cell bodies, associated with marked
ry infiltrations with presence of macrophages, lym- gliosis and presence of perivascular inflammatory
phocytes, multinucleated cells, and reactive astro- infiltrates. Multinucleated giant cells and microglial
cytes are present in the basal ganglia and brain stem. nodules are reported as absent. Direct evidence of
Sometimes focal areas of coagulation necrosis with HIV-1 infection has not (yet) been provided, although
cavitation are seen, principally in the basal ganglia. increase in CSF HIV-1 antigen during exacerbations
Microglial nodules with multinucleated giant cells has been demonstrated in some cases.
are found in both gray and white matter, in the latter In children with progressive HIV-1 encephalopathy,
case often accompanied by demyelinating lesions. brain weight is below normal for age, and sometimes
HIV-1 virus has been identified in these nodules. In a there is obvious atrophy. Most of the brains show
number of cases, however, the microglial nodules evidence of white matter disease, including pallor,
contain cells with the intranuclear and intracytoplas- diffuse lack of myelin, and astrocytosis. Probably
mic inclusion material typical of cytomegalovirus in- both hypomyelination and demyelination are respon-
fection. Microglial nodules may be the consequence sible for the diffuse lack of myelin. The axons are rel-
of either HIV-1 infection or cytomegalovirus infec- atively better preserved than in adults. Calcification
tion. Cytomegalovirus-associated nodules are usually often occurs within or adjacent to the walls of small
distinguished by their predominant localization vessels in the basal ganglia and central white matter
within the cortical gray matter and by the presence of of the cerebral hemispheres, especially in the frontal
characteristic intranuclear inclusion bodies within or lobes. Inflammatory cell infiltrates which resemble
near the nodules. classic microglial nodules but which are larger are
Ultrastructural studies of the brain in subacute commonly observed. They consist of microglia, astro-
HIV-1 encephalitis reveal many virus-like particles in cytes, lymphocytes, a few plasma cells, and often
the cytoplasm of most macrophages and multinucle- multinucleated cells, some of which have the propor-
ated giant cells and, less often, in astrocytes, but not in tions of multinucleated giant cells. The inflammatory
neurons and oligodendrocytes. These particles are infiltrates are most often located in basal ganglia and
double-membraned structures, which contain cylin- pons, but are also found in other gray and white mat-
drical nucleoids, characteristic of the Lentivirus sub- ter structures of brain and spinal cord. In addition,
family of the retrovirus group. These structures have vascular or perivascular inflammation may be pre-
been identified as HIV-1. In addition to complete in- sent, involving small or medium-sized arteries or
fectious HIV-1 virions, HIV-1 RNA, HIV-1 DNA, and veins. The inflammation of intraparenchymal vessels
HIV-1 core proteins have been identified in the brains is accompanied by intimal fibrosis of medium-sized
and also in the CSF of patients with subacute HIV-1 vessels and endarteritis obliterans with focal throm-
encephalitis. Macrophages and multinucleated cells bosis of small vessels.
of macrophage origin appear to be infected, but there Spinal cord disease afflicts approximately 25% of
is evidence that some other cells are also infected to a AIDS patients. Pathological changes are most promi-
lesser extent. nent in the lateral and posterior columns at the tho-
Histological examination of acute diffuse leukoen- racic level, less severe in the anterolateral and anteri-
cephalitis reveals demyelination, most pronounced in or columns. White matter changes are often asym-
the centrum semiovale. There is also axonal loss, but metrical and are not confined to specific anatomic
axons are relatively better preserved than myelin. tracts. Microscopically, the disease is characterized by
Microglial nodules and multinucleated giant cells intramyelinic vacuoles, similar to those seen in cere-
may be present but have also been reported as absent. bral and cerebellar white matter. Axons are intact ex-
Inflammatory reaction is variable. The demyelinating cept in the most severely affected areas. The white
lesion may extend into the frontal, occipital, and tem- matter vacuolation is associated with few lipid-laden
poral area. The basal ganglia, thalamus, and brain macrophages, usually located within the vacuoles. Re-
stem may also be involved in the process. The lesions active astrocytes are rare, and inflammation is not
may be large and multifocal or highly confluent, sym- present as a rule.
metrical or asymmetrical. Presence of HIV-1 genome
in the brain has been shown. Tests for all other infec-
tions, in particular cytomegalovirus, are negative.
620 Chapter 81 Acquired Immunodeficiency Syndrome

81.3 Pathogenetic Considerations cell) immune system essentially ceases to function,


but there are also marked abnormalities in B cell acti-
Human immunodeficiency virus type 1 (HIV-1) has vation (humoral immune system) and immunoregu-
been identified as the primary cause of AIDS. HIV-1 lation. There is a state of polyclonal B cell activation
is the prototypical member of the Lentivirinae sub- with high immunoglobulin levels and a poor anti-
family of retroviruses affecting humans. Lentiviruses body response to new antigens. The deficiency of cel-
characteristically cause indolent infections in their lular immunity leads to an extreme susceptibility to
host. These infections are notable for involvement of viruses, fungi, mycobacteria, and parasites – agents
the nervous system, long periods of clinical latency, that require cell-mediated immunity for contain-
and weak humoral immune responses complicated by ment. Neoplasms arising in a cellular immunodefi-
persistent virus presence. One feature that distin- ciency state include primarily lymphomas and Ka-
guishes the lentiviruses from other retroviruses is the posi sarcoma. The B cell dysregulation may explain
remarkable complexity of their viral genomes. Most the frequent occurrence of severe pyogenic infec-
retroviruses capable of replication contain only three tions, in particular with Streptococcus pneumoniae
genes. HIV-1, however, contains in its RNA genome at and Haemophilus influenzae. The elevated produc-
least six additional genes. It is probable that the dis- tion of nonspecific immunoglobulins may lead to
tinct but concerted actions of these additional genes autoimmune processes such as immune thrombo-
underlie the profound pathogenicity of HIV-1. From cytopenia. In addition, CD4+ T antigens are also pre-
a therapeutic standpoint, this same genomic com- sent on the cell surface of certain types of macro-
plexity may also be the Achilles heel of the virus. phages and monocytes, which may therefore also be
A broad-based search for antagonists specific for infected by HIV-1. The infection of these cells results
these HIV-1 gene products has started. High-resolu- in additional immunological deficits, especially in
tion electron microscopy has revealed the HIV-1 viri- chemotaxis.
on as an icosahedral structure containing 72 external HIV-1 leads to a persisting infection. The viral ge-
spikes. These spikes are formed by the two major vi- netic information is integrated in the DNA of the host
ral envelope proteins, gp120 and gp41. The HIV-1 cell and the virus can only be destroyed by killing the
lipid bilayer is also studded with various host pro- infected cells. It has been demonstrated that a small
teins, including class I and class II major histocom- percentage of infected cells may survive and con-
patibility (MHC) antigens, acquired during virion tribute to virus persistence. Infected monocytes and
budding. HIV-1 has a selective tropism of CD4+ macrophages may contribute to the persistence of
T helper-inducer lymphocytes. The human CD4+ HIV-1 as they appear to be relatively resistant to the
T lymphocyte and monocyte are the major cellular cytolytic effect of HIV-1.
targets for HIV-1 infection, because the CD4+ mem- HIV-1 appears to be transmitted predominantly by
brane antigen represents the principal, if not sole, contact with infected blood or semen. However, the
high-affinity receptor for this retrovirus. After entry virus has also been isolated from cervicovaginal se-
of the CD4+ T lymphocyte, the viral RNA is tran- cretions, tears, urine, saliva, breast milk, and CSF.
scribed into DNA and subsequently integrated into These fluids could possibly also be involved in the
the CD4+ T lymphocyte DNA during cell division. In transmission of AIDS. HIV may be transmitted to a
inactive CD4+ T cells much of the viral DNA remains child in utero, during birth, or postnatally through in-
unintegrated in the cytoplasm. The HIV replication fected breast milk.
cycle is restricted until the CD4+ T cell is activated by The clinical picture of patients with HIV-1 infec-
other pathogens (for instance, hepatitis B virus, her- tions can range from asymptomatic (carrier with
pes simplex virus, cytomegalovirus) or by allogeneic viremia or antibody or both) through chronic gener-
stimulation (for instance, exposure to allogeneic se- alized lymphadenopathy to a variable degree of clini-
men, blood, or allografts). After activation, transcrip- cally manifest immunodeficiency. Estimation of viral
tion occurs in RNA, followed by protein synthesis. load and CD4+ lymphocytes may help to predict
Viral proteins and viral RNA assemble. Mature virus- which patients are likely to have the full-blown dis-
es are formed by budding from the cytoplasmic mem- ease. The latency period between infection and full-
brane. In the process of HIV replication the CD4+ blown AIDS ranges from several months to several
T cell is killed. The mechanism of cell death is unclear. years.
The killing of CD4+ T lymphocytes leads to depletion Substantial evidence supports a direct etiological
of this type of cells. Faster depletion of CD4+ T cells role for HIV-1 in subacute encephalitis, aseptic
may result from enhanced susceptibility to superin- meningitis, vacuolar myelopathy, and peripheral neu-
fection by other pathogens, for example, by cyto- ropathy. The intra-blood-brain-barrier synthesis of
megalovirus. CD4+ T lymphocytes play a central role HIV-1-specific antibodies has been demonstrated in
in the immune response, and their depletion leads to the majority of AIDS patients with neurological
many immunological abnormalities. The cellular (T symptoms but also in patients without these. The
81.4 Therapy 621

oligoclonal IgG bands in the CSF of AIDS patients tive is that the release of HIV-1 envelope glycopro-
have been demonstrated to contain anti-HIV activity. teins from infected macrophages may interfere di-
The presence of HIV-1 antigens in CSF appears to be rectly with neuronal function as these HIV-1 proteins
strongly associated with CNS involvement. HIV-1 has may directly suppress neuronal responses to neu-
been isolated from CSF in patients with subacute en- rotropic factors and cause shortened neuronal sur-
cephalitis and aseptic meningitis. The fact that virus vival.
has been isolated from CSF cannot be attributed sole- The frequent involvement of the CNS in AIDS is
ly to the presence of infected lymphocytes since many probably related to a number of factors. The brain is
samples are free of cells. HIV DNA and RNA se- an immunologically privileged organ. HIV-1 within
quences have been demonstrated in CNS tissue of pa- brain tissue may be hidden and protected from im-
tients with subacute encephalitis. HIV-1 has been iso- mune surveillance. In addition, the damage to the im-
lated from brain in subacute encephalitis, from the mune system caused by HIV infection promotes the
spinal cord in vacuolar myelopathy, and from the sur- persistence of viral infection. In infants the known
al nerve in a patient with peripheral neuropathy. The susceptibility of the immature CNS to viral invasion
multinucleated giant cells seen in subacute encephali- plays a role and may explain the high incidence of
tis show a striking similarity to the multinucleated AIDS encephalopathy in young children infected with
cells that develop from the fusion of T lymphocytes HIV.
infected by HIV-1 in vitro. Ultrastructural examina-
tion has identified intact retroviral particles within
the multinucleated giant cells in some cases. The 81.4 Therapy
multinucleated cells are probably of macrophage
origin, as they share morphological features and Therapy for HIV-1 infection has been increasingly
macrophage markers. Very similar multinucleated successful in considerably reducing morbidity and
giant cells have been reported in lymph nodes in mortality. This has been achieved by a logical attack
AIDS patients. There is evidence that the multinucle- on targets provided by the life cycle of the HIV-1 virus
ated giant cells are of hematogenous origin. It is pos- and by using a combination of therapies in the
sible that the virus penetrates the CNS by migration so-called highly active anti-retroviral treatment
of infected mononuclear cells from the blood, but a (HAART). To understand the different approaches it
direct viral invasion of the CNS is not excluded. Im- is most illustrative to follow this life cycle of HIV-1
munohistochemical identification of HIV antigen has and indicate where interference with the cycle is pos-
shown that the most frequently infected cells include sible.
macrophages, microglia, and multinucleated giant The HIV-1 virus consists of an outer envelope of
cells; less frequently capillary endothelial cells, astro- protein, fat, and sugars wrapped around an inner
cytes, and oligodendroglia are infected, and also neu- core, the capsid, which contains genetic information
rons, but only rarely. The virus is concentrated in, in the form of two strands of RNA, and a number of
although not limited to, the white matter. special enzymes. Proteins of HIV-1 are strongly
The precise mechanisms underlying the structural attracted to CD4+ surface receptors presented on
cerebral damage remain to be determined. The low- dendritic cells, macrophages, monocytes, and, most
level infection, sometimes seen in a few neurons, abundantly, on T4 lymphocytes (helper cells). After
astrocytes, and oligodendrocytes, cannot be held binding to the CD4+ surface receptor, other proteins,
responsible for the severe CNS damage that is often such as glycoproteins gp120 and gp41, are activated,
present. The functional and structural damage of the allowing the HIV cell to fuse with the outside of the
CNS is therefore probably related indirectly to the cell. The therapeutic intervention at this point is ad-
HIV-1 infection, which primarily affects monocytes ministration of entry blockers. Attempts have been
and macrophages and to a lesser degree endothelial made to use CD4+ protein decoys, which have so far
cells. Data indicate that the extent of endothelial and not been very successful. More promising as an entry
macrophage infection is more commensurate with inhibitor seems enfuvirtide (T20). Enfuvirtide is a
the clinical findings than the extent of neuronal and complex peptide consisting of 36 amino acids. The
glial infection. This observation led to the suggestion compound binds to a subunit, gp41, of the glycopro-
that the CNS dysfunction reflects an infection of the tein gp120, located on the viral envelope, blocking the
endothelial cells that impairs the blood–brain barrier entry of the virus in the cell. Data about side effects
and leads to fluctuations in fluid and electrolyte lev- and long-term results are still scarce.
els and to structural damage. Another possibility is Once the viral capsid is inside the cell, the two
that activated macrophages secrete a variety of mate- strands of RNA are transcribed into DNA, the “provi-
rials, such as tumor necrosis factor, interleukin, and ral” DNA, by the enzyme reverse transcriptase. Inter-
proteolytic enzymes, that cause brain tissue damage ventions at this step have been successful and several
and impairment of neurological function.An alterna- types of reverse transcriptase inhibitors have been
622 Chapter 81 Acquired Immunodeficiency Syndrome

developed and play an important role in combination patients. Patients with progressive multifocal ence-
therapies. The main groups are nucleoside reverse phalopathy have shown temporary improvement. The
transcriptase inhibitors, nonnucleoside reverse tran- improved CD4+ cell count is another sign of improve-
scriptase inhibitors, and nucleotide reverse transcrip- ment.
tase inhibitors. Side effects of these multidrug regimens are com-
Approved reverse transcriptase inhibitors include: mon, myopathy, neuropathy, gastrointestinal prob-
∑ Nucleoside analogues: zidovudine, didanosine, lems, pancreatitis, bone marrow and hepatic toxicity,
zalcitabine, stavudine, lamuvidine, abacavir and fat redistribution being the most common. Both
∑ Nucleotide analogues: tenofovir disoproxil fu- lipoatrophy and lipohypertrophy are possible, lipohy-
marate pertrophy probably caused by the protease inhibitors
∑ Non-nucleoside reverse transcriptase inhibitors: and lipoatrophy caused by the reverse transcriptase
delavirdine, efavirenz, nevirapine inhibitors. Lipohypertrophy causes a protease paunch
and buffalo hump. Lipoatrophy can disfigure the face
The next step in the development of new virions is the and often requires cosmetic surgery. Bone marrow
entry of the proviral DNA into the cell’s nucleus, suppression is another possible side effect, leading to
where the enzyme integrase connects the viral DNA anemia and thrombopenia. Children born from
with the host cell’s DNA. Integrase inhibitors are still mothers using antiretroviral drugs during pregnancy
under research, but may eventually open a therapeu- may present with severe mitochondrial disorders re-
tic window at this level. lated to depletion of mitochondrial DNA. The effects
When the T lymphocyte is activated, special en- of HIV nucleoside analogue reverse transcriptase in-
zymes transcribe DNA into messenger RNA. In this hibitors (NRTIs) on mitochondrial function have
phase, transcription inhibitors could be expected to been known for some time. NRTIs are phosphory-
have therapeutic action, but so far they do not play an lated intracellularly to dideoxynucleoside triphos-
important role in HIV therapy. phates. These compounds compete with the natural
Messenger RNA is now translated into new viral substrates (deoxythymidine triphosphates) for HIV
proteins, which are kept inside the cell. In the final reverse transcriptase, but, since they lack a 3’ hydrox-
part of the process these strings of proteins are cut up yl group, the effect is termination rather than length-
by the enzyme protease to form proteins that will ening of the DNA chain. They are also substrates for
handle the different functions of the new HIV virus. DNA polymerase g, the enzyme required for replica-
The logical answer to this phase was the development tion of mitochondrial DNA. Mitochondrial DNA en-
of protease inhibitors, which has been successful. Pro- codes crucial mitochondrial proteins and is present
tease inhibitors now play a prominent role in HIV in multiple copies per cell. Decreased concentrations
therapy.Approved protease inhibitors are: saquinavir, of mitochondrial DNA have been shown in muscles of
ritonavir, indinavir, nelfinavir, amprenavir. patients with zidovudine-induced myopathy. NRTIs
This is not the place to give recommendations on are widely used to diminish the rate of transmission
combination therapy, which would be very soon of HIV across the placenta from infected mothers to
outdated. Federal guidelines and recommendations children. In a series of 1784 children (Blanche et al.
are available on the Internet and updated regularly 1999), 8 children with probable mitochondrial dys-
(http://www.projinf.org/fs/antiVirSrat.html). On this function were observed, without HIV infection, a
site generic names and brand names of the antiviral much higher frequency than could be expected in the
drugs are given, together with the names of the phar- normal population (1 in 5,000–20,000). Quite a few
maceutical companies. Federal guidelines are given uncertainties remain (Morris and Carr 1999). At this
for the circumstances in which therapy should start, moment continuation of the policy of the treatment
how best to combine preparations, changes of thera- of HIV positive pregnant women with antiretroviral
py, when indicated, and advice for treatment of preg- therapy is still recommended.
nant women. It is common policy now to prescribe a A more definite approach is the development of an
combination of a protease inhibitor with two nucleo- HIV-1 vaccine. The development of such a vaccine has
side analogues. Initially the combination therapy had been complicated by the remarkable sequence het-
the disadvantage that many pills had to be taken every erogeneity of the HIV-1 envelope proteins. Hyperim-
day, some before meals, some after meals, three or munization of chimpanzees with HIV-1 envelope
four times a day. This hardly promoted therapy com- protein did not protect them against HIV-1 infection
pliance, seriously contributing to the risk of drug re- after injection of small quantities of the same strain
sistance. There are attempts to replace these regimens of bHIV-1. More recent vaccination studies in Asian
by a once-a-day intake of drugs, and several drugs monkeys with a simian virus (SIV-1) infection, which
have already been approved for once-a-day use. The causes an AIDS-like disease, have generated more
newer therapeutic regimens have been successful in positive results and suggest that an effective HIV-1
restoring at least some immunocompetence in the vaccine may be an achievable goal.
81.5 Magnetic Resonance Imaging 623

Preventive measures still prevail in the battle quence of opportunistic infections, tumors, and vas-
against AIDS, the universally propagated “safe sex” cular lesions of the brain. The presence of more than
being the major public prevention factor. Other pre- one type of lesion within one patient frequently com-
ventive measures are directed towards the avoidance plicates the interpretation of CT findings.
of contact with infected blood, screening of blood MRI has greater sensitivity than CT in identifying
donors for AIDS, and inactivation of virus in blood CNS lesions in this patient population, with the ex-
products such as factor VIII concentrate. Specific ception of calcifications. HIV-1 infection of the brain
measures have also been advocated in the manage- leads to a number of patterns identifiable on MRI.
ment of HIV-1-positive pregnant women, in order to The most common pattern seen in AIDS dementia
reduce the risk of neonates acquiring the disease in complex is generalized atrophy with widened ventri-
the peri- or postnatal period. cles and enlarged cortical sulci. Probably, however, at-
In opportunistic infections and CNS tumors ap- rophy is a relatively late finding. MRS is more sensi-
propriate therapeutic measures have to be taken. tive, already showing a decrease in the N-acetylaspar-
Identifying the causative agent in infections or the tate (considered to be a neuronal marker) while im-
nature of the tumor is, therefore, important and the ages are still normal.
major role of MRI. The second pattern represents the subacute HIV-1
encephalitis and reflects the histological findings of
diffuse, mild myelin pallor or demyelination with
81.5 Magnetic Resonance Imaging preference for the frontal and parietal lobes. MRI
shows symmetrical periventricular areas of mildly in-
Neuroimaging studies are essential in the evaluation creased signal intensity of the white matter, not in-
of AIDS patients with neurological symptoms. A high volving the U fibers, with preferential involvement of
percentage of the patients infected with HIV-1 have the frontal and parietal lobes (Figs. 81.1 and 81.2).
CNS complications, including the AIDS dementia There is no enhancement after contrast injection. The
complex, opportunistic infections, and brain tumors. brain stem and cerebellum are not involved. In the
The primary purpose of brain imaging is to detect course of the disease the white matter abnormalities
potentially treatable opportunistic lesions. progress, resulting in severe hemispheric involve-
Most patients with advanced subacute HIV-1 en- ment (Fig. 81.2).
cephalitis have CT evidence of cerebral atrophy with In acute diffuse leukoencephalitis, large multifocal
enlargement of subarachnoid spaces and ventricles. or more diffuse and extensive white matter abnor-
CT evidence of atrophy may precede clinical signs of malities are present. In the case of multifocal lesions,
encephalitis, whereas in other cases initial CT scans these are not necessarily symmetrical (Fig. 81.3).
may be normal and the atrophy becomes evident on Hemispheric white matter, internal capsule, thala-
later scans. Marked hypodensities in the white matter mus, and brain stem may be involved. In cases in
of both cerebral hemispheres are sometimes noted. which the disease course resembles multiple sclerosis,
An additional abnormality shown by CT scan in some multifocal white matter involvement is seen (Fig.
children with HIV-1 infection of the CNS, and less 81.3). Contrast enhancement of the lesions has not
frequently in adults, is calcification of basal ganglia been reported.
and, less often, of the periventricular white matter The frequency of superimposed infections is rela-
and the frontal subcortical white matter. CT scans tively low, but double and multiple infections may oc-
may also reveal abnormalities which are a conse- cur. Of the opportunistic infections, cerebral toxo-

Fig. 81.1. T2-weighted transverse


images of 40-year-old male AIDS
patient with an advanced stage of
subacute HIV encephalitis.The MR
images show widening of the ventri-
cles and cortical atrophy.There are
symmetrical signal abnormalities in
the cerebral white matter, most promi-
nently involving the frontal lobes.
In the posterior regions, the signal
abnormalities are more subtle and
the U fibers are spared
624 Chapter 81 Acquired Immunodeficiency Syndrome

Fig. 81.2. A 48-year-old man presenting with AIDS presented corticospinal tracts of the midbrain and the white matter of
with headaches, loss of concentration, mild cognitive impair- the temporal lobes. There is slight cerebral atrophy. This pat-
ment, and changes in behavior. The transverse FLAIR images tern is characteristic of subacute HIV infection. Courtesy of
show symmetrical involvement of the periventricular and Dr. M. Heitbrink and Dr. B. Wiarda, Department of Radiology,
deep cerebral white matter, sparing the U fibers.The posterior Medical Center Alkmaar, The Netherlands
limbs of the internal capsules are involved, continued in the

plasmosis is the most common. It is the presenting anti-toxoplasmosis treatment when multiple focal le-
opportunistic infection in at least 5% of the AIDS pa- sions with ring enhancement are seen. If toxoplasmo-
tient population. The lesions of toxoplasmosis are sis is present, the lesions will improve within a few
multifocal and unequal in size (Fig. 81.4). Sometimes weeks of therapy. Differentiation of larger single le-
the lesions show the so-called “target” sign, with a rim sions is from lymphoma. Progressive multifocal
of high signal intensity on T2-weighted images and a leukoencephalitis is discussed in Chap. 82. In this dis-
core that is isointense with normal white matter. Le- ease, the lesions are confined to the white matter and
sions occur in all areas including the subcortical re- extend into the arcuate fibers. There is a typically
gion, basal ganglia, brain stem, and, less frequently, sharp border between gray matter and demyelinated
the cerebellar hemispheres (Fig. 81.4). The lesions can white matter, leading to a characteristic pattern. Usu-
be very large, have contact with the ventricles, and ally the lesions do not enhance, but there are excep-
mimic a lymphoma. After contrast injection the le- tions. Cryptococcus neoformans infection is the most
sions usually enhance. In about 5% of the cases, how- frequent intracranial fungal infection (2–7.5% of
ever, no or only slight enhancement is seen. This may AIDS patients). The infection is preferentially located
be due to the absence of inflammation as a conse- in the basal ganglia spreading along the Virchow–
quence of the defective immune system of the patient, Robin spaces. This pattern of spread may produce a
resulting in little or no capsule formation around the typical appearance, suggestive of this type of infec-
toxoplasmosis abscess. Possibly, this finding predicts tion. Cytomegalovirus encephalitis resembles in
a poor prognosis. Pragmatically one could consider many aspects the subacute AIDS encephalitis and
81.5 Magnetic Resonance Imaging 625

Fig. 81.3. A 24-year-old man, suffering from AIDS, presented ond and third rows) show lesions consistent with acute diffuse
with a rapidly progressive neurological syndrome and mental leukoencephalitis. There is mildly increased signal intensity in
and emotional deterioration. The T1-weighted sagittal image the centrum semiovale and around the ventricles, also involv-
(first row, left) shows a high-signal-intensity lesion in the basal ing the posterior limb of the internal capsule and the white
ganglia on the left side, probably a hemorrhage. The trans- matter tracts in the pons. The lesions are asymmetrical at the
verse proton density (first row) and T2-weighted images (sec- level of pons, midbrain, and periventricular area

may present with the same clinical symptoms. Pa- cases a ventriculitis will develop. After contrast injec-
tients may also be asymptomatic. On MRI, diffuse, tion this appears as an enhanced subependymal rim
symmetrical, periventricular high-signal-intensity around the ventricles.
changes are seen on T2-weighted images, indistin- Of the CNS tumors complicating AIDS, primary
guishable from subacute HIV encephalitis. In some lymphoma is the most frequently observed (about
626 Chapter 81 Acquired Immunodeficiency Syndrome

Fig. 81.4. A 48-year-old male AIDS patient was admitted to cerebellum, pons, and basal ganglia.The second type of lesion
the hospital with general malaise and left-sided hemiparesis, consists of a more symmetrical involvement of the periventric-
cerebellar ataxia, and diplopia. He also showed intellectual ular and deep cerebral white matter. The latter type of lesion
deterioration. The MR images show two types of lesions. The represents subacute HIV encephalitis; the dispersed, smaller
first type consists of multiple smaller, isolated lesions in the lesions were demonstrated to be toxoplasmosis

5% of AIDS patients). In AIDS, lymphomas may be connected with the ventricular wall and enhance after
multifocal, which is rare in other patients. Also, the contrast administration. Thallium-201 SPECT may
enhancement of lymphoma in AIDS patients may be offer a solution in distinguishing between infection
less homogeneous. Differentiation from toxoplasmo- and lymphoma. Leptomeningeal lymphoma can be
sis on MRI may be difficult. Both can present as single diagnosed by the presence of pericerebral tissue that
or multifocal lesions with mass effect; both can be enhances with contrast. Metastases of Kaposi sarco-
81.5 Magnetic Resonance Imaging 627

ma have been observed in the CNS and in the skull of HIV-1 encephalitis in children, although white
base, but are extremely rare. matter changes may be present. Opportunistic infec-
In HIV-1 encephalitis in children, apart from the tions are less frequent in children than in adults. Cen-
ventriculomegaly and atrophy also found in adults, tral mass lesions are usually caused by primary lym-
there is often symmetrical bilateral calcification of phoma.
the basal ganglia and the white matter adjacent to the In the spinal cord lesions have been described in
frontal horns. Contrast enhancement of the basal AIDS patients. On MRI these lesions are not distin-
ganglia may also occur, sometimes starting unilater- guishable from lesions as seen in transverse myelitis
ally, becoming bilateral later on. Calcifications may in multiple sclerosis. Swelling of the cord is seen ini-
also be related to a concurrent cytomegalovirus infec- tially with moderate enhancement, eventually fol-
tion. White matter disease is not a prominent feature lowed by atrophy.
Chapter 82

Progressive Multifocal Leukoencephalopathy

82.1 Clinical Features brief, lasting only a few days. At the other end of the
and Laboratory Investigations scale, the disease can last for a year or more. In AIDS
patients with secondary PML some survival time can
Progressive multifocal leukoencephalopathy (PML) is be gained by treatment with highly aggressive anti-
a rare demyelinating infection of the CNS caused by a retroviral therapy (HAART). Patients showing spon-
polyomavirus, the JC virus. It usually occurs in im- taneous improvement have also been described.
munodeficient patients. Its association with such un- It is clear that a wide range of clinical neurological
derlying diseases as lymphoma, multiple myeloma, abnormalities may be encountered in PML and no
leukemia, sarcoidosis, tuberculosis, Whipple disease, syndrome is especially distinctive of the disease. The
systemic lupus erythematosus, systemic carcinomas, combination of a coexisting chronic systemic disease
renal transplantation, bone marrow transplantation, with fairly rapidly progressive multifocal or diffuse
AIDS, and immunosuppressed states is well docu- cerebral disease should suggest the presence of PML.
mented, although the disease has also been reported As a great proportion of the general population,
in patients without evidence of an immunological probably 80–90%, has been exposed to JC virus in the
deficit. During the last few years, PML has most often form of a banal childhood upper respiratory tract
occurred in the context of AIDS. The prevalence of infection and thus has developed IgG antibodies
PML in AIDS patients is estimated to be in the range against the JC virus, serological tests are not helpful in
of 4–7%, which is probably an underestimation. In establishing the diagnosis. Routine cytochemical CSF
most cases, PML is a late complication of a pre-exist- examinations, such as cell count and protein concen-
ing chronic systemic disease, which has typically been tration, are also of little value, because abnormalities
present for a long time before the neurological abnor- may be due to the underlying pre-existent disease.
malities appear. This underlying disease does not dif- The JC virus has never been cultured from blood or
fer appreciably either in its clinical or pathological as- CSF, nor have viral antigens been identified in the
pects from the same disease in other patients in CSF. In patients who are not suffering from HIV infec-
whom PML does not occur. There is no way of pre- tion, detection of intrathecal antibodies against the JC
dicting the occurrence of PML. PML occurs predom- virus has a sensitivity of about 70% and a specificity
inantly in adults. There is a male preponderance in of 99%. As in other microbial diseases, the PCR tech-
the proportion of about 5:3. nique of amplifying DNA has had a major impact on
The clinical signs and symptoms of PML are those the diagnosis of PML. Detection of JC viral DNA by
expected from the presence of multiple CNS lesions of PCR in the CSF has a sensitivity of about 75% and
varying size, mainly affecting the white matter of the specificity between 90% and 99%. The high specifici-
cerebral hemispheres but indiscriminate in their lo- ty obviates in many cases the need for a brain biopsy.
calization. In the early stages the most frequently ob- However, the problem of false negative results in 25%
served signs are monoparesis, hemiparesis, personal- of the cases remains, which may prompt a second CSF
ity change, cognitive impairment, ataxia, dysarthria, examination or still make a brain biopsy necessary.
dysphasia, and cerebral visual impairment. Quadri- Semiquantitative measurements of JC viral DNA load
paresis, severe dementia, and coma characterize the in CSF shows correlation with survival and could be
more advanced stages. Headaches and seizures are used as a method of monitoring therapeutic efficacy,
unusual, and there is no evidence of increased in- in addition to clinical and neuroimaging findings.
tracranial pressure. Rarely, involuntary movements
indicative of an extrapyramidal disorder are ob-
served, such as choreiform movements, dystonia, 82.2 Pathology
athetosis, or parkinsonism. Once the disease appears,
it generally progresses until the patient dies. Usually, The brain displays no external abnormalities in PML.
as with many incapacitating cerebral diseases, death On sectioning, multiple grayish granular-appearing
results from terminal bronchopneumonia. In most lesions are seen in the white matter, often associated
patients a period of about 2–6 months elapses be- with a loss of the distinct border between the cortex
tween the first appearance of neurological symptoms and subcortical white matter. Asymmetrical involve-
and death. In a few patients the illness is extremely ment is the rule. Although the lesions are often found
82.3 Pathogenetic Considerations 629

bilaterally, they are usually more extensive in one group of papovaviruses. This observation is con-
cerebral hemisphere than in the other. firmed by immunofluorescent studies with the use of
Microscopically the lesions vary from small foci of specific antibodies against polyomavirus. Using the
demyelination to extensive areas of myelin loss, occu- in situ hybridization techniques with a polyomavirus
pying a major portion of a cerebral lobe or hemi- DNA probe, the infection of oligodendrocytes and
sphere. The small foci tend to be round or oval, but astrocytes with this virus can also be shown.
the larger areas of demyelination are irregularly out-
lined, probably resulting from coalescence of smaller
lesions. The lesions may occur anywhere in the white 82.3 Pathogenetic Considerations
matter of the CNS, no part of the CNS being entirely
spared. However, the lesions tend to be less frequent PML is a demyelinating disease associated with a
in the brain stem and the cerebellum, and involve- polyomavirus, which belongs to the Papovaviridae.
ment of the spinal cord and peripheral nerves is The family Papovaviridae consists of two genera,
uncommon. Localization of lesions in the corti- Papillomavirus and Polyomavirus. These are small
comedullary junction and subcortical white matter is DNA-containing viruses.A human polyomavirus, des-
very typical. The deep cortical layers often also show ignated JC virus after the individual from whom it was
loss of myelin sheaths, but the cortical cytoarchitec- first isolated, has been implicated as the etiological
ture remains intact. agent in nearly all cases of PML. In a minority of the
The lesions are characterized by loss of myelin cases a related polyomavirus, simian virus 40 (SV40),
with relative sparing of the axons. Sometimes the ax- has been held responsible for the disease. Although
ons are destroyed along with the myelin, leaving the disease PML is rare, infection with the JC virus is
nothing but a meshwork of astrocytic processes and quite common. Antibodies to JC virus have been
glial fibrils or actual cavities. In the center of the le- found in up to 90% of the population, and the preva-
sion, where myelin sheaths have been destroyed, lence of antibodies rises with increasing age. Infection
oligodendrocytes are absent. At the periphery of the is usually acquired early in life: by the age of 15, 65%
lesions the oligodendrocytes are markedly altered, of children have anti-JC virus antibodies. In any given
with enlargement of their nuclei and effacement of PML patient there is no information about whether
the normal chromatin pattern. These abnormal nu- these antibodies were present before the onset of the
clei often contain basophilic or eosinophilic inclusion disease. The virus is distributed throughout the world.
bodies. Another cellular change is a conspicuous al- The question of how JC virus can be ubiquitous yet
teration of astrocytes. Some of the astrocytes in and not cause more widespread clinical illness is not sim-
around the lesion are enlarged into gigantic forms, ple to answer. Recent research has provided informa-
and their nuclear structure is profoundly altered. Hy- tion about the pathways the JC virus utilizes to enter
perchromatism, occasionally bizarre lobulation of the the human host and ultimately the brain. Traffic of JC
nucleus, multinucleation, and mitotic figures are virus through the body is complex, involving cellular
characteristic abnormalities. The change in individ- receptors, DNA-binding proteins, and a variety of
ual cells is at times so severe that the cells become in- viral regulatory regions in multiple target cells. JC
distinguishable from the neoplastic cells that charac- virus DNA has been detected in human tonsil tissue,
terize malignant astrocytomas. The relative absence including stromal cells and lymphocytes. Therefore,
of a cellular inflammatory reaction is striking. In tonsils may be the initial site of infection. For this in-
many cases inflammatory cells are absent altogether, fection to occur virions have to bind to special recep-
and in other cases they are few. Numerous lipid-laden tors, enter cells, and have their DNA integrated into
macrophages can be identified within the area of host cell DNA. Viral DNA is replicated and translated
demyelination. into proteins, and via a number of steps JC virions are
PML does little damage to nerve cell bodies. Le- assembled and passed to circulating tonsillar lym-
sions may be situated in gray matter structures, espe- phocytes. As with other human viral pathogens, sus-
cially the deep cortical layers.Within these lesions the ceptibility to infection is determined at least partly by
myelin sheaths are destroyed, and the astrocytes and viral attachment to cellular receptors. This specificity
oligodendrocytes show the characteristic changes, of receptors plays a role in the types of cells that are
whereas most of the nerve cells appear to be intact. most vulnerable to infection. Different JC virus geno-
Electron microscopic studies show virus-like par- types with different regulatory regions have consis-
ticles within the abnormal oligodendrocytic nuclei tently been found in different tissues, probably con-
and, to some extent, also in altered astrocytes. These tributing to differential vulnerability of different tis-
particles are seen in isolation, in irregular groups, in sues to infection. Viral reactivation occurs by im-
a crystal-like pattern, or in the form of filaments. The munosuppression of the host. Reactivated JC virus
size, shape, surface details, and arrangement of these will infect B lymphocytes. These cells can cross the
particles are characteristic of the polyomavirus sub- blood–brain barrier and infect oligodendrocytes.
630 Chapter 82 Progressive Multifocal Leukoencephalopathy

Oligodendrocytes have a receptor profile that makes Cytarabine (arabinosylcytosine) administered intra-
them extremely vulnerable to JC virus infection. venously and intrathecally has been demonstrated to
Viremia would be another explanation of the trans- be more promising in PML. The rationale for the use
port of JC virus from infected organs to the brain, but of this agent is that polyomaviruses are DNA viruses,
this has never been observed. T cells cannot be carri- and that cytarabine impairs DNA synthesis. Long-
ers either, because JC virus cannot infect T lympho- term survival of PML patients treated with cytarabine
cytes or bind to T cell membranes. Infected B lym- has been reported. The improvement of the neurolog-
phocytes have been found in multiple PML brain tis- ical condition varies from moderate to marked in
sue samples. these patients. In some patients there is very rapid
Various papovaviruses are known to induce cell improvement, occurring within 48 h of initiation of
transformation in vitro and are oncogenic in ham- therapy. In others there is a delay of several weeks.
sters. Papovaviruses, as mentioned before, are subdi- Sometimes cytarabine is only transiently beneficial,
vided into two subgroups: the papillomaviruses, and sometimes it has no beneficial effect at all.
known to cause warts and cervical carcinoma, and Neither type and duration of underlying disease nor
polyomaviruses, such as the JC virus, causing PML duration and course of neurological symptoms prior
and of which the oncogenicity is a suspicion of more to treatment appear to yield a clue as to the different
recent date. Papovaviruses, their antigens, or viral responses to therapy. Some studies, however, have
DNA sequences have been detected in some human been unable to find the same positive results using
tumor cells. Pathologically, two cell types in the brain cytarabine. Treatment of PML with a combination of
are infected by the polyomavirus in PML. First, oligo- cytarabine and interferon has occasionally been re-
dendrocytes are lytically infected, and virus particles ported as successful. Treatment of patients with HIV-
fill the nucleus to give it a characteristic appearance. 1 infection and PML with highly aggressive antiretro-
Oligodendroglial cell death causes demyelination. It is viral therapy has led to a better immune status in pa-
postulated that there is also an infection of the astro- tients, and sometimes also to a positive effect on the
cytes, in which the viral genome is integrated into the coexisting PML.
cellular DNA, leading to cell transformation. The fact Therapy should be directed at eradication of the
that astrocytic cell transformation in PML is virally viral infection not only by specific antiviral agents but
induced is indirectly supported by the known onco- also by enhancing host immune defenses. Lowering of
genic potential of papovaviruses in hamsters, the re- the immunosuppressive medication may be appro-
ported simultaneous occurrence of PML and gliomas, priate. Early diagnosis before actual destruction of
and the demonstration of shared internal capsid anti- nervous tissue has occurred may be necessary for
gens of a polyomavirus in the nucleus of giant astro- treatment to be successful.
cytes in PML.
A defect in cell-mediated immune defenses ap-
pears to be a central factor in the reactivation process 82.5 Magnetic Resonance Imaging
of JC virus and the development of PML. The lack of
cellular inflammatory reaction in white matter le- CT scan shows low-density white matter lesions with
sions also supports the notion of a deficit in cellular characteristically scalloped lateral borders following
immunity. Yet this deficit alone does not provide an the contours of the gray–white matter junction,
adequate explanation for the occurrence of PML whereas the medial border is smoother in outline.
since diseases associated with such immunological Enhancement of some of the lesions may be seen.
deficiency and treatment with immunosuppressive Small and early lesions are not detected, and CT often
drugs are relatively frequent, while PML is rare. The shows no abnormalities in the early stages of the dis-
problem is made even more difficult by the fact that ease.
PML may even occur in the presence of apparently MRI shows that PML lesions can be found any-
intact immune responses. where in the white matter of the cerebral hemi-
Simian virus 40 is of minor importance in PML. In spheres, the brain stem, and the cerebellum, but most
general, infection with this virus is rare in humans, often involve the cerebral subcortical area (Figs. 82.1–
only 5% showing antibodies. The source of the virus 82.3). The lesions have an intermediate to low signal
is unknown. intensity on T1-weighted images, and a high or very
high signal intensity on T2-weighted images. The af-
fected white matter appears somewhat swollen, which
82.4 Therapy becomes more evident in those areas where the U
fibers are involved. The subcortical white matter le-
At present the prognosis in PML is generally poor. sions then have a scalloped appearance, outlining the
Treatment with idoxuridine and vidarabine, both an- overlying cortex and stretching it (Figs. 82.1, 82.2 and
tiviral agents, has been shown to be nonbeneficial. 82.4). The sharp edge between affected white matter
82.5 Magnetic Resonance Imaging 631

Fig. 82.1. The characteristic image of


PML: a large lesion in the right parietal
lobe, involvement of the corpus callo-
sum, and a smaller lesion in the
left hemisphere.These T2-weighted
images show a sharp border between
the hyperintense white matter lesion
and the normal cortex.The cortex is
stretched out over the lesion. In this
case there was no enhancement

Fig. 82.2. A 63-year-old male with AIDS and PML.These images show a large frontal lesion on the right side with partial enhance-
ment after contrast injection, as is seen on the transverse and coronal T1-weighted contrast-enhanced images

and cortical gray matter is due to the anatomical bar- to the appearance of a widespread white matter disor-
rier formed by the noninfected neuronal layer with its der (Fig. 82.4). Enhancement after the injection of
relative myelin paucity as opposed to the myelin-rich gadolinium is seen in a minority of patients, but is not
white matter and infected oligodendrocytes. In ex- exceptional (Figs. 82.2 and 82.4). If enhancement is
ceptional cases MRI shows some extension of a white seen, it is usually only present in some of the lesions;
matter lesion into the cortex, probably where intra- it is faint and peripheral. More solid enhancement is
cortical myelin has been attacked (Fig. 82.3). Lesions seen in rare cases. There is evidence that mass effect
of the external capsule have been found in 38% of and (temporary) enhancement on MR images, as ex-
PML patients, and posterior fossa lesions in 32% pressing the presence of an immune reaction of the
(Fig. 82.4). Isolated posterior fossa lesions have been patient, represent positive predictive factors for more
reported occasionally. Basal ganglia may be involved, prolonged survival. PML lesions have different ap-
particularly in the myelin-containing laminae sepa- pearances on diffusion-weighted images depending
rating the various nuclei and subnuclei (Figs. 82.3 and on the stage of the disease. Newer lesions and the ad-
82.4). The myelin content of the thalamus and globus vancing edge of large lesions have a high signal on
pallidus is higher than of the putamen and caudate Trace diffusion-weighted images and a normal to low
nucleus, and lesions would be expected to occur more ADC. Older lesions and the center of large lesions
often in these areas. Involvement of the corpus callo- have a low signal on Trace diffusion-weighted images
sum is often seen (Figs. 82.1 and 82.4). Although the and increased ADC values. High signal on Trace diffu-
epithet “multifocal” suggests more than one lesion in sion-weighted images and low ADC mark the regions
all cases, this is not necessarily true. Single lesions of active infection with cell swelling, distinguishing
may occur. Confluence of several lesions may give rise them from burnt-out areas of gliosis.
632 Chapter 82 Progressive Multifocal Leukoencephalopathy

Fig. 82.3. One parasagittal T1-weighted and a series of T2- volved: putamen, globus pallidus, and caudate nucleus on the
weighted MR images obtained in a 48-year-old man with AIDS left are affected, and some lesions extend into the cortical gray
and PML are shown. The images show multiple lesions in the matter
white matter. Note that gray matter structures are also in-

PML has to be differentiated from other multifocal encephalopathy syndrome, and noninfectious in-
white matter disorders, especially from those occur- flammatory disorders, white matter lesions are as a
ring in AIDS patients, such as cytomegalovirus infec- rule also asymmetrical, and differentiation from PML
tions, and from acute and subacute HIV encephalitis may be impossible on the basis of MRI features alone.
itself. These latter infections, however, do not usually Clinical and laboratory information is necessary. In
involve the U fibers, do have different signal intensity several inborn errors of metabolism and in several
characteristics on T1- and T2-weighted images, and toxic encephalopathies, spongiform white matter
tend to be symmetrical. Lesions in systemic lupus changes occur, resulting in some white matter
erythematosus may sometimes mimic PML, while swelling and stretching of the overlying cortex, which
PML may also occur in the context of systemic lupus is otherwise intact. The appearance of the white mat-
erythematosus. Multifocal white matter involvement ter changes shows similarities to those observed in
with some swelling of the subcortical lesions and PML, but the white matter abnormalities are usually
stretching of the overlying cortex may also be seen in symmetrical and usually generalized rather than
noninfectious inflammatory disorders, in particular multifocal. Some cases of PML resemble gliomatosis
multiple sclerosis and acute disseminated en- cerebri on MRI. However, in gliomatosis cerebri the
cephalomyelitis. Similar lesions have been reported in border between white and gray matter is as a rule
the context of the reversible posterior encephalo- blurred, contrasting with the sharp border seen in
pathy syndrome. In vasculitis, posterior reversible PML.
82.5 Magnetic Resonance Imaging 633

Fig. 82.4.
634 Chapter 82 Progressive Multifocal Leukoencephalopathy

Fig. 82.4. (continued). A 43-year-old man with rapidly pro- are the internal capsule and the laminae separating the differ-
gressive neurological deterioration caused by PML. This series ent nuclei, resulting in a pattern of concentric partial circles.
of T2-weighted images (first, second, and third rows) show ex- The brain stem and middle cerebellar peduncle on the right
tensive involvement of white matter structures. The cerebral are affected. The affected white matter is mildly hypointense
white matter is affected, more seriously on the right side, with on T1-weighted images (fourth row).There is one small focus of
involvement of the corpus callosum.The abnormal white mat- contrast enhancement. Diffusion-weighted Trace images
ter is mildly swollen with thickening of the corpus callosum. (b = 1000, fifth row) show large areas of hyperintensity, which
The images could be considered suggestive of gliomatosis correspond to low ADC values and restricted diffusion. These
cerebri, but there is a sharp demarcation between the affected findings indicate active infection of large areas, consistent
cerebral white matter and the unaffected overlying cortex.The with the dramatic clinical picture in the patient
basal ganglia are involved, but the structures mainly affected
Chapter 83

Brucellosis

83.1 Clinical Features 83.2 Pathology


and Laboratory Findings
There are few reports on the neuropathology of neu-
Brucellosis is a zoonosis endemic in the eastern part robrucellosis. They describe various degrees of vas-
of the Mediterranean, the Arabian peninsulas, and the cular inflammation, ranging from chronic infiltration
Indian subcontinent. Infection of humans occurs by with lymphocytes, plasma cells, and macrophages to
ingestion of raw meat, milk products, or direct con- acute polymorphic infiltration that may cause ab-
tact with infected animals. Four species of Brucella scesses, necrosis, and aneurysm formation. Diffuse
can infect humans: Brucella melitensis, Brucella abor- meningeal inflammation may occur as well as inflam-
tus, Brucella suis, and Brucella canis. After infection, matory cell infiltration of the perineurium of nerve
bacteremia follows and the disease spreads to many roots. Little has been published about the nature of
organs and organ systems. Brucella species diffusely the involvement of the cranial nerves and the inner
colonize the lymphoreticular system, with prolifera- ear. Inflammation of the inner ear seems a more prob-
tion of lymphocytes and macrophages. Formation of able cause of the progressive hearing loss than affec-
granulomas follows. tion of the eighth cranial nerve.
The clinical symptoms are protean. The disease Although demyelination is presumed by several
usually starts with an episode of malaise, fever, and authors to be present in patients with white matter
arthralgia, and progresses slowly to more severe con- abnormalities, especially those presenting with the
stitutional symptoms, including headache, nausea, clinical picture of ADEM, no histopathological evi-
vomiting, otalgia, hearing loss, ophthalmoplegia, and dence of demyelination has been provided. Occasion-
cardiac and neurological symptoms. Involvement of ally data have been obtained because a brain biopsy
the CNS occurs in three major forms: leptomeningeal was performed. In a patient with diffuse white matter
involvement in the bacteremic phase of the disease, involvement and progressive encephalopathy, biopsy
peripheral nervous system involvement with poly- revealed abundant inflammatory cells in the white
radiculoneuropathy, and slowly progressive central matter, with lymphocytic infiltrates in the lepto-
nervous system involvement with mild confusion, meninges surrounding the cortical vessels (Seidel et
focal neurological symptoms such as dysarthria, and al. 2003). No significant demyelination was found in
cognitive impairment, but occasionally also with the affected area. Reactive astrogliosis was found in
multifocal serious symptoms due to multiple brain the cortex. Marked activation of microglia was
abscesses. In children and occasionally in adults the demonstrated by immune staining of MHC complex
clinical and MRI features may resemble those of acute class II antigens. The infiltrates in the white matter
disseminated encephalomyelitis (ADEM). In some consisted mainly of CD8+ T-cell lymphocytes.
cases diabetes insipidus is present as the result of
granulomatous involvement of the sellar contents.
Spinal symptoms may result either from direct in- 83.3 Pathogenetic Considerations
volvement of the spinal cord or from compression
due to involvement of the vertebral column with for- Brucellae are intracellular, nonmobile, gram-negative
mation of granulomas. coccobacilli capable of replication within mononu-
Laboratory confirmation of brucellosis may be dif- clear phagocytes. Ingestion of infectious products,
ficult. The CSF shows usually only mild pleocytosis most often unpasteurized milk, is followed by
and normal protein and glucose concentrations. IgG hematogenous dissemination, residence of the bru-
antibodies against Brucella may be found in the CSF, cellae in the reticuloendothelial system, and subse-
even with negative serum titers. Enzyme-linked im- quent involvement of multiple organs or organ sys-
munosorbent assay (ELISA) of the CSF may further tems, resulting in a variety of clinical symptoms. In
assist in the diagnosis. In rare cases brain biopsy may 4–15% of patients the CNS is involved. The presence
lead to a correct diagnosis. of the brucellae leads to an inflammatory–granulo-
matous response and, in the CNS, sometimes to mul-
tiple brain abscesses. Involvement of venous struc-
tures may lead to sinus thrombosis. The abundance of
636 Chapter 83 Brucellosis

Fig. 83.1. Brucellosis in a 70-year-old man. The FLAIR images The T1-weighted images after contrast (third row) show en-
(first and second rows) show frontoparietal involvement of the hancement of the leptomeninges. From Al-Sous et al. (2004),
arcuate fibers and a hyperintense rim around the ventricles. with permission

CD8+ T-cell lymphocytes in white matter lesions sug- 83.4 Therapy


gests that the white matter lesions are induced by T-
cell-mediated toxic injury. This would imply that the The correct diagnosis is essential because brucellosis
pathogenesis of the lesions is different from lesions is a treatable condition. Once diagnosed, neurobru-
seen in autoimmune-related disorders such as acute cellosis can be treated with a combination of antibi-
disseminated encephalomyelitis. It is unclear why in otics. Most often this is done by an intravenous course
some cases the sellar region is involved or the spinal for several weeks, followed by an oral course for many
cord in others. months.
83.5 Magnetic Resonance Imaging 637

Fig. 83.2. Brucellosis in a 50-year-old man. The FLAIR images extension into the deep frontal white matter, than seen in
show a more irregular pattern of involvement of the arcuate Fig. 83.1. In this case there are also lesions in the midbrain and
fibers and a more prominent rim around the ventricles, with pons. From Al-Sous et al. (2004), with permission

83.5 Magnetic Resonance Imaging Leptomeningeal involvement occurs usually in the


early phase of the disease.
An MR inventory of suspected inflammatory–granu- White matter involvement may occur in a number
lomatous disorders should aim at the depiction of the of different patterns. First of all, a multiple sclerosis or
various disease manifestations of these disorders, ADEM-like type of involvement may occur with
guided by the clinical symptoms. The structures that asymmetrical large patchy hyperintense lesions on
should obtain special attention are: T2-weighted and FLAIR images (Figs. 83.1 and 83.2).
∑ The leptomeninges, especially in the early phase of Secondly, a symmetrical pattern of diffuse involve-
the disease (T2-weighted images, T1-weighted im- ment of subcortical white matter is sometimes seen
ages without and with contrast, in transverse and without involvement of the corpus callosum. Some-
coronal planes) (Fig. 83.1) times the abnormalities are more extensive and dif-
∑ The white matter (proton density, T2-weighted, fusely involve the subcortical and periventricular re-
and FLAIR images, contrast-enhanced T1-weight- gions, again without participation of the corpus cal-
ed images) (Figs. 83.1–83.3) losum (Fig. 83.3). Occasionally there are (asymmet-
∑ The sellar region (sagittal and coronal T1-weight- ric) lesions in the basal ganglia (Fig. 83.3). Some of
ed images with fat-suppression, without and with these basal ganglia lesions may be hemorrhagic and
contrast enhancement (Fig. 83.4) some may enhance after contrast administration. In
∑ The spinal cord (sagittal T1- and T2-weighted im- later phases lacunar infarctions may be seen, mainly
ages of the entire spinal canal, transverse images in the basal ganglia. In all cases and especially in the
when lesions are present (Figs. 83.3 and 83.5) early phase of the disease, leptomeningeal enhance-
ment may occur (Fig. 83.1).
638 Chapter 83 Brucellosis

Fig. 83.3. Brucellosis in a 65-year-old patient with both cere- and a more solid component in the deep frontal white matter.
bral and spinal abnormalities.The two upper images on the left, The abscess is surrounded by an enhancing rim; the solid part
FLAIR and T2-weighted respectively, show symmetrical in- of the lesions enhances in toto. The sagittal and transverse
volvement of most of the cerebral white matter, most promi- T2-weighted images of the thoracic spinal cord show a lesion
nently of the arcuate fibers and the external capsule. The low- in the center of the thoracic cord (arrow). From Seidel et al.
er left image, T1-weighted with contrast, shows meningeal in- (2003), with permission
volvement and abscess formation with a cyst-like component

Lesions in the perisellar, suprasellar, and intrasel- cludes inflammatory conditions like multiple sclero-
lar region may be the only intracranial manifestation sis, acute demyelinating encephalomyelitis, neu-
of brucellosis (Fig. 83.4). They may also occur in com- roborreliosis, and systemic lupus erythematosus.
bination with white matter lesions. When more diffuse and symmetrical white matter ab-
Spinal cord abnormalities may show enhancement normalities are present, the differential diagnosis
in the acute phase. Nerve roots may also show en- may be more difficult and may includes many inher-
hancement (Fig. 83.5). In the later phases spinal cord ited conditions, depending on the exact pattern of ab-
atrophy is usually found. normalities present. Leptomeningeal enhancement
Unusual manifestations are superior sagittal sinus after contrast injection may support the diagnosis of
thrombosis and single or multiple brain abscesses neurobrucellosis. The peri-, intra-, and parasellar
(Fig. 83.3). manifestations have to be differentiated from neu-
When multiple sclerosis-like white matter abnor- rosarcoidosis, pituitary tumors, Langerhans cell histi-
malities are present, the differential diagnosis in- ocytosis, and Whipple disease.
83.5 Magnetic Resonance Imaging 639

Fig. 83.4. Brucellosis in a 30-year-old


woman. Coronal T1-weighted images
with contrast show a suprasellar gran-
uloma as the sole manifestation of
the disease.The differential diagnosis
of this phenomenon includes Langer-
hans cell histiocytosis, Whipple
disease, neurosarcoidosis, and Tolosa–
Hunt syndrome. From Al-Sous et al.
(2004), with permission

Fig. 83.5. T1-weighted images in the


transverse and sagittal plane in a
32-year-old man with brucellosis.The
images on the left are T1-weighted
images without contrast, those on the
right with contrast. After contrast
administration, caudal roots show
enhancement. From Al-Sous et al.
(2004), with permission
Chapter 84

Subacute Sclerosing Panencephalitis

84.1 Clinical Features generalized, and psychomotor convulsions. They may


and Laboratory Investigations even precede the myoclonic jerking. Mental deterio-
ration becomes progressively more obvious. At this
Subacute sclerosing panencephalitis (SSPE) is one of stage, choreoathetosis, ataxia, tremor, and spasticity
the slow virus infections of the CNS. It is a rare disor- are common. The duration of stage II varies from 1
der, but it is the commonest of the chronic virus infec- month to 1 year or more. In some patients the disease
tions to affect children. It is caused by measles virus is arrested in stage II and remains in this stage for
and occurs in approximately 4:100,000 cases of years. In stage III there is severe dementia. The spas-
measles. SSPE is a disease of childhood and adoles- ticity increases, and the child becomes bedridden. Ex-
cence, with an age of onset range of 4–25 years and a trapyramidal dysfunction of the parkinsonian type is
peak incidence at 9–17 years. SSPE is rare in adults. frequent. Nasogastric tube feeding is often required at
The disease occurs on average 9 years after the initial this time because of progressive bulbar palsy. Hyper-
infection. There is a racial difference in incidence of thermia may be found without evidence of infection.
SSPE: in the United States the incidence is four times It is the rule that the myoclonia diminishes in stage
higher in whites than in blacks. Boys outnumber girls III. Stage IV is the final stage in which the child is in a
by 3 or 2 to 1. Children who acquire measles infection vegetative state, characterized by mutism and decor-
under the age of 1 year are more likely to develop ticate or decerebrate postures. There is no bladder or
SSPE. The mean age at infection is 12–14 months. In bowel control. Ophthalmological abnormalities occur
exceptional cases SSPE can also occur after live in 50% of patients and include optic atrophy and
measles immunization, but the incidence is highly re- chorioretinitis. The chorioretinitis may be most
duced after immunization. Since the introduction of marked at the maculae, where scars are seen with
measles vaccination, the incidence of SSPE has be- characteristic pigmentation. There are signs of auto-
come very low in countries where a high immuniza- nomic dysfunction, such as hyperthermia, severe per-
tion coverage (~ 95%) is achieved. spiration, and changes in heart rate and blood pres-
The clinical syndrome of SSPE is rather variable. sure. The duration of stage IV varies from 1 to 6 years.
The duration of the disease ranges from 3 months to Most patients die during stage III or IV from car-
more than 7 years. Although there is a marked varia- diorespiratory complications related to impaired cen-
tion in presenting signs and in the sequence of events, tral mechanisms controlling temperature, cardiac,
four clinical phases can usually be discerned. Stage I and respiratory functions. Another common cause of
is characterized by an insidious deterioration of be- death is infection. Spontaneous long-term remissions
havior and intellectual performance. The duration of are exceedingly rare, but may occur.
this stage is usually several months, but the exact time The clinical diagnosis of SSPE is confirmed by the
of onset of the disease is often difficult to determine. finding of increased levels of anti-measles virus anti-
Personality changes occur with withdrawn, timid, or body in serum and CSF with an elevated CSF-to-
aggressive behavior. These changes are followed by serum ratio in antibody level. The CSF is clear, and the
lethargy, drooling, slurred speech, and paucity of pressure and glucose concentration are normal. Mod-
speech. Soon after the onset of intellectual deteriora- erate pleocytosis (5–20 mononuclear cells per milli-
tion, visual disturbances can often be detected. These liter) is often present. The protein level is usually nor-
are either related to progressive chorioretinitis or to mal or mildly elevated; a level that exceeds 0.90 g/l is
lesions of the central visual pathways. In stage II, mas- uncommon. The level of IgG as a percentage of total
sive, repetitive, and frequent myoclonic jerking oc- protein is highly increased, and oligoclonal bands are
curs. The myoclonia develops slowly and irregularly, present. Measles-specific IgG antibodies represent
but gradually affects all somatic muscle groups, espe- nearly 10–20% of total serum IgG and about 75% of
cially the axial muscles, in a reasonably symmetric the total CSF IgG. There is always an elevated CSF-to-
fashion and at a regularly repetitive rate. The my- serum ratio of IgG, especially of measles virus anti-
oclonus may be sufficiently severe to throw the child bodies, indicating local production of measles virus
to the floor. The jerking interferes with intentional antibodies within the CNS.
movements, giving the impression of clumsiness. The Another confirmatory finding is a characteristic
jerking is absent during sleep. Seizures of a more con- EEG pattern of periodic complexes consisting of bi-
ventional type may also occur, such as focal motor, lateral synchronous, symmetric, 2- to 4-Hz high-am-
84.3 Pathogenetic Considerations 641

plitude sharp and slow wave bursts, which may occur 84.3 Pathogenetic Considerations
every 5–7 s. These periodic complexes occur simulta-
neously with the involuntary myoclonic movements. Measles virus is a member of the Morbillivirus genus
In stage I the EEG may be normal or show only mild in the Paramyxoviridae family. SSPE is a rare compli-
to moderate, nonspecific slowing. Stage II is charac- cation of measles infection and differs from the acute
terized by the occurrence of the periodic complexes; measles encephalitis that may occur during or imme-
the background pattern is still relatively normal. In diately after acute measles infection. The main risk
stage III, the background rhythm slows, and fewer factor is acquiring the infection at or before the age of
bursts of periodic complexes occur. Stage IV has slow 1 year. The elevated serum and CSF anti-measles
delta activity and rare SSPE complexes. These com- virus antibody levels in patients suspected of having
plexes in association with myoclonic seizures provide SSPE are indicative of an active measles virus infec-
strong corroborative evidence of SSPE. However, tion of the brain. Inclusions with paramyxovirus par-
periodic complexes are also seen in other diffuse ticles have been demonstrated in neuronal and glial
cerebral disorders, such as some lysosomal storage cells in SSPE. The cellular inclusions react with anti-
disorders, mitochondrial encephalopathy (MERRF), measles virus antibodies. For a long time, however,
cerebral anoxia, and widespread infections of the the virus could not be recovered from brain tissue by
brain. conventional methods. Successful isolation of the
measles virus was finally accomplished by cocultivat-
ing brain cells of SSPE patients with cells known to
84.2 Pathology support measles virus replication. After the isolation
of the virus, the agent was studied extensively to de-
The pathology of SSPE is restricted to the CNS. The termine its structural and biological characteristics.
leptomeninges are thickened. The brain may show at- Its ultrastructural features are similar to those of
rophy during the terminal stages of disease but is oth- measles virus. Further studies of the immune re-
erwise normal on external examination. On slicing sponse in patients demonstrated the absence of
the brain, discolored areas are seen. Microscopically, serum antibody to the matrix (M) protein of measles
the disease appears to be multifocal in character and virus. The M protein is associated with the inner sur-
may involve all portions of the CNS, with the excep- face of the viral membrane. It is important in the as-
tion of the cerebellum, which is rarely affected. The sembly of the virus particle, which occurs by a bud-
frontal lobes are involved first, followed by the pari- ding process from the surface membrane of the in-
etal, temporal, and occipital lobes, basal ganglia, brain fected cell. Subsequent studies demonstrated the ab-
stem, and spinal cord. The typical pathological find- sence of measles virus M protein in the brain tissue of
ings include perivascular infiltration by mononuclear patients with SSPE. More recent studies have demon-
cells in gray and white matter and proliferation of strated that the measles viruses isolated from the CNS
both macroglia and microglia.Astrocyte proliferation of SSPE patients have major mutations in the M pro-
can be particularly pronounced in white matter. Neu- tein gene. As a consequence, the budding process is
ronal degeneration and neuronal loss may be severe. defective.Without budding, the replicating intracellu-
Neurofibrillary tangles are seen. Demyelination is fo- lar virus may go into a dormant phase. However, M
cal and not always conspicuous. The subcortical white protein gene mutations thought to be characteristic
matter is mainly affected, first with inflammation and of SSPE viruses have also been found in measles virus
then with destruction. In the later stages of the dis- isolated in the acute stage of measles infection and it
ease both cortical atrophy and demyelination are is therefore unclear whether these mutations are crit-
more pronounced. Intranuclear and intracytoplasmic ical for the development of SSPE. Regardless of the
inclusions are present in neurons, astrocytes, and precise explanation, the absence of M protein ex-
oligodendrocytes. Immunohistochemistry has re- plains many of the virological features of SSPE. Lack
vealed measles virus antigen in neurons, oligoden- of M protein results in a persistent, abortive infection,
drocytes, and inflammatory cells. CD4+ T lympho- in which no mature infectious virus is produced in
cytes have been demonstrated in the perivascular ar- the extracellular space. The internal components of
eas and CD8+ T lymphocytes in the parenchyma. the virus accumulate in the cells. It is thought that in
Ultrastructural examination shows the presence of the time between primary measles virus infection
paramyxovirus particles. A spectrum of viral inclu- and the onset of SSPE symptoms, viral nucleocapsids,
sions and particles has been reported. Some inclu- RNA, and probably other gene products accumulate
sions fill nuclei, whereas smaller particles, nucleocap- in cells of the nervous system and spread from cell to
sids, and virions of different configurations are found cell through the nervous system. The onset of SSPE
in either the nucleus or the cytoplasm of oligoden- symptoms is usually ascribed to altered cell function
droglial cells and neurons. Nuclear bodies and gran- and cell death, resulting from excessive accumulation
ulofilamentous inclusions occur in astrocytes. of viral products. The intracellular virus is never in
642 Chapter 84 Subacute Sclerosing Panencephalitis

contact with the extracellular space, as a consequence avirin stopped the progression of brain atrophy and
of the absence of M protein and the resulting inabili- improved the clinical condition.
ty of the virus to assemble and bud. Only when cells The problem in trials concerning SSPE is the very
begin to deteriorate and die does the virus make con- small number of patients that can be included in the
tact with the immune system, causing a rise in anti- Western hemisphere, so that multicenter, internation-
measles antibodies, except antibodies to M protein. al collaboration is necessary.
The antibodies produced are ineffective in eradicat-
ing the intracellular virus.
Other mutations in the measles virus genome may 84.5 Magnetic Resonance Imaging
contribute to the risk of SSPE. Fusion protein (F pro-
tein) and hemagglutinin (H protein) are two surface CT is normal during the initial stage of the disease,
glycoproteins. Truncations in the cytoplasmic do- shows multiple low-density areas in the white matter
main of the F protein impede efficient virus assembly and basal nuclei in subsequent stages, and shows se-
and budding. Loss of glycosylation in the H protein vere atrophy in the end stage. Low-density areas may
leads to inefficient membrane transport. These fea- show contrast enhancement.
tures lead to reduced amounts of envelope protein on In the earliest stage, MRI may be normal, but it
the cell surface and may prevent association with the shows abnormalities before CT does. Early changes
M protein, which leads to hampered assembly and may involve the basal ganglia (Fig. 84.1), lateral genic-
budding of mature measles virus particles. Mutations ulate bodies, or the disease may present with multifo-
in H and F proteins may also lead to enhanced cell- cal white matter lesions, which have a preference for
cell fusion and spread of the virus from cell to cell, hemispheric subcortical white matter (Fig. 84.2). In
making it possible for the defective virus to accumu- more extensive lesions, the deep and periventricular
late readily and spread without the need at any stage white matter are involved additionally (Fig. 84.3). The
for viral maturation and budding. abnormalities are often asymmetrical (Fig. 84.2), but
Host factors seem also to be important in predis- in many cases they are extensive and symmetrical
posing individuals to SSPE. In human beings the pri- (Fig. 84.3). The overlying cortex is often also affected
mary infection seems to occur during a critical peri- (Fig. 84.2). Large focal lesions involving white and
od in early life when passive maternal immunity has gray matter, resembling infarctions, may occur. These
faded, in particular before the age of 1 year. Immatu- lesions may enhance initially and subsequently be-
rity of the host immune system and CNS has been come nonenhancing. The white matter changes may
suggested to contribute to the increased risk for SSPE decrease or resolve, even in the presence of clinical
development among infants. Other genetic factors deterioration. Bilateral striatal lesions (Fig. 84.1),
(male versus female; racial differences) seem to con- bilateral lesions in the middle cerebellar peduncles,
tribute and may determine components of the im- brain stem lesions, and cerebellar abnormalities may
mune system. occur. In end-stage disease, severe atrophy of cere-
brum, basal ganglia, brain stem, and cerebellum is
found, often with global and generalized white matter
84.4 Therapy signal abnormality.
MR spectroscopy, either as chemical shift imaging
There is no effective treatment for SSPE. The disease or as single voxel measurements, may help to estimate
leads to death in most cases, but there may be sponta- the stage of the disease. In stage I, MRS findings do not
neous temporary improvement or an arrest of further differ significantly from those in normal age-matched
progression for a number of years. Attempts to alter volunteers. In stage II, there is still a normal N-acetyl-
the disease course with antiviral agents, such as aspartate/creatine ratio, but an increased choline/
amantadine (Symmetrel), 5-bromodeoxyuridine, or creatine ratio. The myo-inositol/creatine ratio is in-
inosiplex (Isoprinosine,Viruxan) have failed, which is creased in this and the subsequent stages. This would
not surprising in view of the marked cell-associated correspond with an early inflammatory reaction. In
nature of the virus. Intraventricular interferon-alfa- stage III, MRS shows decreased N-acetylaspartate/
2b, usually applied in combination with oral inosi- creatine, increased choline/creatine, increased myo-
plex, seems to have a higher remission and survival inositol/creatine, and elevated lactate and lipid peaks,
rate. Some patients have been treated with a combina- suggesting demyelination, gliosis, neuronal loss, and
tion of intraventricular interferon-alfa-2b and rib- macrophage activation, macrophages being highly
avirin. Subcutaneous interferon-beta has been ap- dependent on anaerobe glycolysis.
plied in patients who could not receive interferon-al- The differential diagnosis includes other causes of
fa and has also demonstrated beneficial effects. Both multifocal abnormalities of gray and white matter, in-
medications are broad-spectrum antiviral agents cluding acute disseminated encephalomyelitis, pro-
with activity against both DNA and RNA viruses. In gressive multifocal leukoencephalopathy, multiple
the case reports of a few patients, the addition of rib- sclerosis, and vasculitides. In some cases of SSPE,
84.5 Magnetic Resonance Imaging 643

Fig. 84.1. In this 9-year-old boy with SSPE, MRI initially showed much more severe, and shows lesions of the body of the cau-
only a few moderately abnormal white matter lesions in the date nucleus on the left and in the putamen on both sides.
centrum semiovale. The MRI in this figure was performed 7 There is still a somewhat higher signal intensity of the parietal
months later, when signs of neurological dysfunction were deep white matter

Fig. 84.2. In this 3-year-old boy with SSPE, T2-weighted trans- The overlying cortex is also abnormal. Courtesy of Dr. M. Pine-
verse and coronal images show asymmetrical involvement of da, Department of Neuropediatrics, Clinic-Hospital Sant Joan
the subcortical and deep white matter, most severe on the left. de Déu, Barcelona, Spain
644 Chapter 84 Subacute Sclerosing Panencephalitis

Fig. 84.3. This 20-year-old man with SSPE is in a far advanced also extensive signal abnormalities in the brain stem, middle
stage of the disease. There are extensive abnormalities in the cerebellar peduncles, internal capsule. The basal ganglia dis-
cerebral white matter, most severe in the posterior region, also play subtle signal abnormalities. There is some atrophy. Cour-
involving the splenium of the corpus callosum, reminiscent of tesy of Dr. Z. Patay, Department of Radiology, King Faisal Spe-
the cerebral form of X-linked adrenoleukodystrophy.However, cialist Hospital and Research Center, Riyadh, Saudi Arabia
there is no enhancement after contrast (not shown).There are

MRI shows symmetrical white matter lesions in the chondrial disorders, in particular Leigh syndrome,
occipital area and splenium of the corpus callosum, and other neurodegenerative disorders. EEG and lab-
simulating X-linked adrenoleukodystrophy. In cases oratory findings help to establish the correct diagno-
with involvement of the basal nuclei, the differential sis.
diagnosis includes Creutzfeldt–Jakob disease, mito-
Chapter 85

Congenital and Perinatal Cytomegalovirus Infection

85.1 Clinical Features tion, jaundice, hepatosplenomegaly, petechiae or pur-


and Laboratory Investigations pura, and pneumonia. Hepatomegaly, splenomegaly,
and petechiae are the most common. The liver is usu-
Cytomegalovirus (CMV) is a member of the Her- ally smooth and nontender and commonly measures
pesviridae family of large DNA viruses, along with 5 cm or more below the right costal margin. Ascites
herpes simplex virus types 1 and 2, Epstein–Barr may be present prenatally and persist postnatally for
virus, varicella-zoster virus, and human herpes virus- 1–2 weeks. The hepatomegaly usually resolves by 3
es 6 and 7. These viruses share the biological proper- months of age, and persistence beyond 1 year is high-
ties of latency and potential reactivation. Infection ly unusual. Mild hepatitis is usually present. Hyper-
with one member of the family does not confer im- bilirubinemia, on the other hand, may be quite strik-
munity against infection or disease with the other ing, with very high conjugated (direct) bilirubin lev-
members of the herpes family. Naturally acquired els. The abnormal results of liver function tests grad-
CMV infection induces cross-reactive immunity to ually resolve during the first few weeks of life. Chronic
infection with new strains of CMV, but this protection hepatitis due to congenital infection with CMV is un-
is not complete, because reinfection with a second usual but may occur. Enlargement of the spleen is
strain of CMV has been documented. very common in congenital CMV infection, and in
Infection with CMV occurs in several ways: first, by some cases it may be the only abnormality detectable
close or intimate contact of either a sexual or nonsex- at birth. Petechiae in congenital CMV disease are usu-
ual nature with another person who is shedding the ally pinpoint and generalized over the infant’s trunk
virus in bodily secretions; secondly, vertically from and extremities. If present at birth, they can be tran-
mother to infant by transplacental infection; thirdly, sient and resolve within 48–72 h. They can be the
by blood product transmission from a CMV-seropos- only apparent manifestation of CMV infection. How-
itive donor. In congenital and neonatal CMV infec- ever, more commonly, the triad of hepatomegaly,
tion, the mother plays a dominant role. Infection of splenomegaly, and petechiae is seen. Petechiae are
the mother with CMV can be either a primary infec- usually, but not always, accompanied by thrombo-
tion or a reactivation of an earlier infection. The risk cytopenia, and platelet counts in the first few weeks of
of congenital CMV infection in the infant is much life range from 2,000/mm3 to 125,000/mm3. Hemolyt-
higher in the case of a primary infection in the moth- ic anemia may also be present. Pneumonitis is unusu-
er than in the case of reactivation of an old infection. al but, if present, it is usually a severe, interstitial
The transmission rate in a primary infection is pneumonitis occurring in the context of a diffuse,
40–50%, whereas it is approximately 1–2% in recur- multisystem infection. Post-transfusion CMV infec-
rent infection. Infants may also be infected during de- tion in neonates, especially premature infants, may
livery from CMV-infected maternal vaginal secre- cause a syndrome of shock, lymphocytosis, and pneu-
tions or, postnatally, by infected breast milk. Children monitis. The mortality rate of neonatally sympto-
not congenitally or perinatally infected may acquire matic CMV infection is 10–30%.
the disease during the toddler or preschool years by CNS manifestations are common in neonates with
contact with family members or other children. Blood a symptomatic CMV infection and include lethargy,
transfusions can also be a source of CMV infection; poor feeding, seizures, hypertonia or hypotonia,
donor-to-recipient transmission of CMV has been microcephaly, chorioretinitis, and sensorineural
documented. CMV may also be transmitted and pro- deafness. Ocular involvement with CMV occurs in
duce a congenital infection if a pregnant woman or 10–20% of symptomatic infants. Most commonly it
her fetus receives a blood product transfusion from a produces a chorioretinitis that is usually old and inac-
CMV-seropositive donor. tive at birth. The retinitis is usually unilateral but can
Approximately 1% of all newborns are congenital- produce blindness if the macula is involved, as well as
ly infected with CMV, making CMV the most com- strabismus and optic atrophy. Congenital CMV and
mon congenital infection. Of the neonates infected congenital toxoplasmosis produce similar lesions;
with CMV, approximately 10% will have symptoms at however, congenital CMV characteristically does not
birth that are commonly associated with congenital produce microphthalmia or cataracts, and alternative
CMV disease, including intrauterine growth retarda- diagnoses, such as congenital rubella or toxoplasmo-
646 Chapter 85 Congenital and Perinatal Cytomegalovirus Infection

sis or metabolic disorders, should be considered if congenital CMV infection, but this approach has sev-
these eye findings are present. Microcephaly may be eral drawbacks.Although the absence of IgG antibod-
present at birth. It may be part of the overall small size ies to CMV in cord or infant blood probably rules out
of a growth-retarded infant or may be disproportion- congenital CMV infection in an immune-competent
ate and accompanied by normal weight, length, and mother–infant pair, the presence of IgG antibodies
chest circumference. Infants may also have septal de- has limited value, because 50–80% of women of
fects, biliary atresia, inguinal hernias, hip dislocation, childbearing age will have anti-CMV IgG antibodies
and other musculoskeletal abnormalities. In addition, that will be transplacentally transferred to their in-
infants with toxoplasmosis, herpes simplex, syphilis, fant. A significantly higher titer of IgG antibodies to
and HIV infections may be coinfected with CMV, and CMV in the infant than in the mother may imply an
infants with metabolic disorders may have congenital active congenital infection, but in practice this differ-
CMV infection as well. ence is usually difficult to ascertain. Serological sam-
About 90% of the infants with an intrauterine CMV ples obtained serially at 1, 3, and 6 months may rule
infection are asymptomatic at birth. Ten to fifteen per- out congenital infection if the level of CMV antibod-
cent of the infants with a congenital CMV infection ies gradually declines, but if the levels persist, serolog-
that is clinically silent in the neonatal period, and al- ical test results alone cannot determine whether the
most all neonates with a neonatally symptomatic in- infection was of congenital or postnatal origin. The
fection, develop persistent problems, most commonly presence of IgM antibodies at birth, however, is high-
neurological impairment, sensorineural hearing ly suggestive of a congenital CMV infection, provided
deficits, and decreased vision related to chorioretinitis the test was performed properly, but confirmatory
or optic atrophy. Approximately half of the infants urine culture for CMV is recommended for a defini-
with symptomatic and 15% of infants with asympto- tive diagnosis. It is important to realize that a negative
matic congenital CMV infection will have or develop CMV IgM antibody test does not exclude the diagno-
an associated hearing loss. The hearing loss may still sis of congenital infection. Anti-CMV IgM antibodies
develop many years after the initial infection and may are found in only 70–80% of the congenitally infected
fluctuate in severity. The vision loss, too, may be pro- infants. More recently, the PCR technique has been in-
gressive or of late onset. The severity of the neurolog- troduced to demonstrate the presence of the virus in
ical impairment is highly variable.At the severe end of tissue or body fluids. CMV DNA has been detected by
the spectrum are profound mental deficiency and mo- PCR in urine, saliva, and CSF.
tor handicap, often with serious spasticity; at the mild The biggest problem with establishing the diagno-
end are learning, behavioral, and motor coordination sis of congenital CMV has been that it cannot be con-
problems. Epilepsy may occur, in particular in patients firmed after the neonatal period. In the absence of
with more severe handicap. The encephalopathy has overt neonatal signs, infants with congenital or peri-
never been observed to be progressive. natal CMV infection are not tested for the presence of
These numbers indicate that congenital CMV is the virus, viral DNA, or antibodies within the appro-
one of the leading causes of mental deficiency. The priate time frame of 3 weeks. The infants with a
numbers also indicate that most infants with a con- neonatally asymptomatic congenital CMV infection
genital CMV infection have no sequelae at all. who develop neurological sequelae usually come to
The diagnostic time frame for congenital or peri- medical attention after the age of 6–9 months because
natal CMV infection is only the first 3 weeks after of developmental delay or hearing loss, and at that
birth. CMV infection is very common and a postnatal time it is no longer possible to confirm the diagnosis
infection may readily occur. It is therefore important with conventional techniques. However, the PCR
that the test is performed soon after birth. Positive technique allows a delayed diagnosis if neonatal body
tests after the age of 3 weeks can indicate either a con- fluids are still available. The Guthrie card is usually
genital, perinatal, or postnatal infection. the only source of neonatal blood kept beyond the
An important test for the diagnosis of congenital neonatal period. The Guthrie card is the filter paper
CMV infection in neonates is the isolation of the virus used to collect blood spots in the neonatal period for
from urine, saliva, or tissue obtained during the first 3 neonatal screening tests. These cards are stored for a
weeks of life. All infants in whom the diagnosis is sus- variable number of months to years. The introduction
pected should have a viral culture performed. The of PCR on Guthrie cards for the diagnosis of congen-
virus is usually present in a very high titer, and cul- ital CMV creates the opportunity for correct diagno-
tures are commonly positive within 2–3 days of incu- sis beyond the neonatal period. The sensitivity of the
bation. Excretion of the virus in the urine may persist PCR for CMV DNA on Guthrie cards is 100% and the
for years. specificity is 99%. The only disadvantage of the test is
Standard serological tests, such as detection of that the PCR is negative if the neonate no longer had
CMV IgG and IgM antibodies, alone or as part of a viremia. The chance that a neonate with a congenital
TORCH titer panel, are commonly used to diagnose CMV infection no longer has viremia is very small but
85.3 Pathogenetic Considerations 647

may exist, especially if the CMV infection occurred family, with a double-stranded DNA genome of more
long before birth. than 240 kilobase pairs, capable of encoding more
In the neonate with congenital infection, the differ- than 200 proteins. The function of most of these pro-
ential diagnosis includes any of the TORCH (toxo- teins remains unclear. As with the other herpes virus-
plasmosis, rubella, CMV, herpes simplex) agents. Con- es, the structure of the viral particle is that of an
genital toxoplasmosis may mimic congenital CMV in- icosahedral capsid, surrounded by a lipid bilayer out-
fection. In parts of Europe, particularly France and er envelope.
Belgium, congenital toxoplasmosis is a common and Better understanding of the process of viral repli-
significant problem. It is less common in the USA. cation provides insights into molecular mechanisms
Other congenital infections to be considered include of immunity and opens therapeutic windows. CMV
lymphocytic choriomeningitis virus (LCMV) infec- replicates very slowly in cell culture, reflecting its very
tion, syphilis, enteroviral disease, and human immun- slow pattern of growth in vivo (in contrast to herpes
odeficiency virus (HIV) infection. simplex virus infection, which progresses very rapid-
ly). The replication cycle of CMV is divided temporal-
ly into three regulated classes: immediate early, early,
85.2 Pathology and late.
Immediate early gene transcription occurs in the
Pathological data in congenital and neonatal CMV in- first 4 h following viral infection, and key regulatory
fections stem from lethal infections and are therefore proteins are made which allow the virus to take con-
highly biased towards the severe end of the clinical trol of cellular machinery. The major immediate ear-
spectrum. Some of these infants were stillborn; others ly promoter of the CMV genome involved in this part
survived for several days or weeks. Microcephaly was of the process is one of the most powerful eukaryotic
present in most of the patients. Other findings includ- promoters described in nature, and has been exploit-
ed meningoencephalitis, periventricular necrosis ed in modern biotechnology as a useful promoter for
with associated calcifications, more diffuse calcifica- driving gene expression in gene therapy and vaccina-
tions, extensive cortical necrosis with calcifications tion studies.
frequently involving the convexity of the gyri, distur- Following synthesis of immediate early genes, the
bance of neuronal migration (ranging from lissence- early gene products are transcribed. Early gene prod-
phaly to polymicrogyria), ventriculomegaly, cerebel- ucts include DNA replication proteins and some
lar hypoplasia, and marked disruption of the cerebel- structural proteins.
lar architecture. In about half of the cases intranu- Finally, the late gene products are made approxi-
clear inclusion bodies are found, while systemic mately 24 h after infection, and these proteins are
inclusion bodies are found in nearly all cases, in par- chiefly structural proteins that are involved in virion
ticular in the kidney, lungs and liver. assembly and egress. Synthesis of late proteins is high-
In clinical specimens, one of the classic hallmarks ly dependent on viral DNA replication and can be
of CMV infection is the cytomegalic inclusion cell. blocked by inhibitors of viral DNA polymerase, such
These massively enlarged cells (the property of cyto- as ganciclovir. The lipid bilayer outer envelope con-
megaly gave CMV its name) contain intranuclear tains the virally encoded glycoproteins, which are the
inclusions, which histopathologically have the ap- major targets of host neutralizing antibody responses.
pearance of owl’s eyes. The presence of these cells These glycoproteins are candidates for human vaccine
indicates productive infection, although they may be design. The proteinaceous layer between the envelope
lacking even in actively infected tissues. In most cell and the inner capsid, the viral tegument, contains pro-
lines, CMV is difficult to culture in the laboratory, teins that are major targets of host cell-mediated im-
but in vivo infection seems to involve chiefly epithe- mune responses. Of these tegument proteins, the most
lial cells, and, with severe disseminated CMV disease, important is the so-called major tegument protein,
involvement can be observed in nearly all organ sys- UL83 (phosphoprotein 65 [pp65]). Another clinically
tems. important protein, the UL97 gene product, is a phos-
There are no pathology data on the nature of the photransferase. Although the function of this protein
white matter abnormalities observed in children with in the viral life cycle is unknown, this protein is clini-
a mild neurological handicap. cally important because a substrate of the kinase is the
antiviral drug, ganciclovir, which, once phosphorylat-
ed, becomes an effective anti-CMV drug.
85.3 Pathogenetic Considerations Immunity to CMV is complex and involves hu-
moral and cell-mediated responses. Several CMV
CMV is a member of the family of eight human her- gene products are of particular importance in CMV
pes viruses, and is designated human herpes virus 5 immunity. The outer envelope of the virus, which is
(HHV-5). It is the largest member of the herpes virus derived from the host cell nuclear membrane, con-
648 Chapter 85 Congenital and Perinatal Cytomegalovirus Infection

tains multiple virally encoded glycoproteins. Glyco- developing brain. Others have postulated a direct
protein B (gB) and glycoprotein H (gH) appear to be teratogenic effect of CMV on the developing fetus.
the major determinants of protective humoral immu- Observation of CMV-induced alternations in the cell
nity. Antibodies to these proteins are capable of neu- cycle and CMV-induced damage to chromosomes
tralizing the virus, and gB and gH are targets of inves- supports this speculation; however, this hypothesis
tigational CMV subunit vaccines. However, although has been difficult to verify experimentally.
humoral responses are important in control of severe It has been hypothesized that the nature and extent
disease, they clearly are inadequate in preventing of the cerebral abnormalities is related to the gesta-
transplacental infection, which can occur even in tional age at the time of the infection. Infection of the
women who have anti-CMV antibodies. fetus early in pregnancy would lead to abnormalities
The generation of cytotoxic T cell responses in migration with cortical dysplasia, whereas infec-
against CMV may be a more important host immune tions late in pregnancy would lead to white matter ab-
response in the control of infection. In general, these normalities only. This hypothesis is probably general-
cytotoxic T cells involve major histocompatibility ly correct. However, we have seen patients in whom
complex (MHC) class I restricted CD8+ responses. a primary CMV infection was documented in the
Although many viral gene products are important in mother shortly before conception, and in whom only
generating these responses, most CMV-specific cyto- a mild encephalopathy with multifocal white matter
toxic T cells target pp65, an abundant phosphoprotein lesions without gyral abnormalities was found. It is, of
in the viral tegument, the product of the CMV UL83 course, possible that the transplacental transfer of the
gene. virus occurred late in pregnancy.
Recent investigations into the molecular biology of
CMV have revealed the presence of many viral gene
products which appear to modulate host inflammato- 85.4 Therapy
ry and immune responses. Several CMV genes inter-
fere with normal antigen processing and generation Four antiviral chemotherapeutic agents – ganciclovir,
of cell-mediated immune responses. To date, three vi- foscarnet, cidofovir, and formivirsen – are licensed
ral gene products have been identified that inhibit specifically for treatment of serious, life-threatening
MHC class I antigen presentation. One is the US11 or sight-threatening CMV in immunocompromised
gene product, which exports the class I heavy chain patients. Currently a randomized, controlled multi-
from the endoplasmic reticulum to the cytosol, thus center clinical trial evaluating the use of ganciclovir
rendering it nonfunctional. Another is the US3 gene for the treatment of infants with symptomatic con-
product, which retains MHC molecules in the endo- genital CMV infection and evidence of CNS involve-
plasmic reticulum, preventing them from traveling to
the plasma membrane. Finally, the US6 protein in-
hibits peptide translocation by transporters associat-
ed with antigen processing. Other viral gene prod- 䊳

ucts, encoded by the UL33, US27, and US28 genes, are Fig. 85.1. Boy with congenital CMV infection (diagnosis con-
functional homologues of cellular G-protein-coupled firmed by a positive PCR for CMV DNA on the Guthrie card),
receptors. They may, via molecular mimicry, subvert who presented in the course of the first year of life because of
normal inflammatory responses, and in the process serious developmental delay, spasticity, and microcephaly. A
promote tissue dissemination of virus and interfere CT scan was performed at 9 months of age (first row) and
with host immune response. The CMV genome also showed many small calcifications, especially at the cortico-
encodes a homologue of the cellular MHC class I subcortical junction. Some small calcium deposits are seen in
gene, which appears to contribute to the ability of the basal ganglia and deep white matter. In addition, the CT
CMV to evade host defense. The UL144 gene encodes suggests a diffuse cortical dysplasia. MRI was performed at the
a structural homologue of the tumor necrosis factor age of 16 months. The T2-weighted images (second and third
receptor superfamily, which may contribute to the rows) reveal extensive cortical dysplasia, most seriously affect-
ability of CMV to escape immune clearance. ing the lateral sides of the brain.The cortex is too thick,the out-
Little is known about the molecular mechanisms side folding is too coarse, whereas the inner border of the cor-
responsible for the pathogenesis of tissue damage tex is irregular, compatible with polymicrogyric pachygyria. In
caused by CMV, particularly for congenital CMV in- addition, the images show ventriculomegaly, dilated inferior
fection. Although the CNS is the major target organ horns, and multifocal white matter abnormalities, the largest
for tissue damage in the developing fetus, culturing lesions being present in the deep parietal white matter. The
CMV from the CSF of symptomatic congenitally T1-weighted images (fourth row) confirm the cortical dysplasia
infected infants is difficult. Because CMV can infect (left), the dilated inferior horn (middle), and tiny high-signal-
endothelial cells, it has been postulated that a viral intensity spots in the white matter (right), probably related to
angiitis may be responsible for perfusion failure of calcium deposits
85.4 Therapy 649

Fig. 85.1.
650 Chapter 85 Congenital and Perinatal Cytomegalovirus Infection

ment is in progress. It is unknown whether this early ly. White matter abnormalities are reported in 14% of
and intensive administration of ganciclovir will has- the children, whereas no abnormalities were demon-
ten resolution of acute disease, beneficially influence strated in 86% of the children. Few children have cal-
growth and development, decrease auditory and visu- cifications or mild ventriculomegaly (Fig. 85.1).
al impairments, or improve intellectual outcome in Characteristically the calcifications in congenital
these infants. There is evidence that with ganciclovir CMV are distributed in a linear, periventricular pat-
treatment the incidence of delayed hearing loss is tern ranging from tiny, punctate lesions to large de-
lower. However, because of the potential bone marrow posits of calcium that appear to line the entire ven-
suppression, the possibility of as yet unforeseen long- tricular system. Calcifications also may involve the
term effects, and the as yet unproven benefit on long- cortical and subcortical regions or involve the basal
term neurodevelopmental outcome, it is recommend- ganglia (Fig. 85.1). Infants with intracranial calcifica-
ed that ganciclovir should not be routinely used to tions are more likely to display cognitive and audio-
treat infants with congenital CMV disease until the logical deficits later in life than those infants who do
results of ongoing clinical trials establish its safety not have detectable abnormalities.
and efficacy. Anecdotal evidence suggests that criti- With respect to MRI, conventional MR sequences,
cally ill newborns, especially those who are premature including T1- and T2-weighted images, are useful to
and have CMV pneumonia, may benefit from ganci- show morphological and structural details. Gradient
clovir treatment. Because of the side effects of thera- echo sequences should be added to better show calci-
py with currently available antiviral agents, treatment fications. Either an inversion recovery technique or a
of newborns with an asymptomatic congenital CMV T1-weighted 3D gradient echo sequence can help to
infection is presently not advocated, even though assess the presence and extent of the cortical gyral
these infants are particularly at some risk for devel- abnormalities.
oping hearing loss. Studies reporting MRI findings in patients with
The antiviral drugs that are now in use for the neonatal symptomatic congenital CMV infection are
treatment of CMV either directly or indirectly inhibit restricted to small numbers. The findings include di-
viral polymerase or are able to reduce viral prolifera- lated ventricles, enlarged subarachnoid spaces, cere-
tion in patients with signs of CMV disease. These bral gyral abnormalities, cerebellar hypoplasia, cere-
drugs, however, do not clear the virus completely and bellar cortical dysplasia, delayed myelination, and
have serious side effects. Strains of CMV with re- white matter lesions. The ventricular dilatation may
duced susceptibility to these antiviral drugs have be extreme with serious thinning of the cerebral
been reported. There are, fortunately, newer therapies mantle. In the early stages, subependymal germi-
on their way, based upon better knowledge of the nolytic cysts and intraparenchymal, intraventricular,
physiology and life cycle of the virus. Attempts are and subdural hemorrhages may be present.
being made to improve the efficacy of orally taken In patients with proven but neonatally asympto-
drugs. Research is also going on in purine and pyrim- matic congenital CMV, white matter abnormalities
idine nucleoside analogues, as well as in nonnucleo- are frequently observed. The occurrence of anterior
side CMV inhibitors and CMV protease inhibitors. temporal cysts, often in combination with dilated in-
The ultimate goal is the development of a vaccine that ferior horns has been described repeatedly.
prevents congenital CMV disease. The gyral abnormalities in congenital CMV main-
ly involve the lateral aspects of the cerebrum (Figs.
85.1 and 85.2), although they may also be diffuse
85.5 Magnetic Resonance Imaging (Fig. 85.3). In patients with diffuse gyral abnormali-
ties, the cortex may be thin and almost agyric. In ad-
Most published neuroimaging reports concern CT dition, the lateral ventricles are often dilated in these
scans obtained during the follow-up of patients with patients. If the abnormalities are more limited, they
a neonatal symptomatic congenital CMV infection. most often consist of polymicrogyria (polymicro-
Frequent findings are intracranial calcifications (33– gyric pachygyria). The cortex is slightly thickened
54%), unilateral or bilateral ventriculomegaly (10– and folded in abnormally broad and shallow gyri
37%), white matter abnormalities (0–22%), neuronal (Figs. 85.1 and 85.2). The inner border of the cortex is
migration abnormalities with cortical dysplasia irregular, indicative of underlying polymicrogyria.
(0–10%), and an extensive, destructive encephalo- The gyral abnormalities may be unilateral or asym-
pathy (5–13%). Occasionally, subdural effusions or metrical (Figs. 85.4 and 85.5). They may also present
hemorrhage are seen. In 20–30% of the children no in the form of schizencephaly.
abnormalities are found. The pattern of white matter abnormalities ob-
In patients with a neonatal confirmed but asymp- served in congenital CMV infection is distinct. Most
tomatic congenital CMV infection, CT scans show often, the white matter abnormalities consist of mul-
milder abnormalities, and shows them less frequent- tifocal lesions with the largest lesions in the parietal
85.5 Magnetic Resonance Imaging 651

Fig. 85.2. Images of a 3-year-old girl with congenital CMV (di- The inferior horns of the lateral ventricles are dilated. Along
agnosis confirmed by a positive PCR for CMV DNA on the the border of the left ventricle, in the parietal region, white
Guthrie card). The T2-weighted images (first and second rows) matter lesions are seen.The FLAIR images (third row) show the
show polymicrogyric pachygyria, more severe on the right white matter lesions more clearly
than on the left, and dilated ventricles, larger on the right side.

area and with predominant involvement of deep lateral but not always symmetrical. In patients with
white matter, relatively sparing the immediately gyral abnormalities, both diffuse (Fig. 85.3) and mul-
periventricular and subcortical white matter (Figs. tifocal (Figs. 85.1, 85.2, 85.4 and 85.5) white matter ab-
85.6–85.8). Numerous small additional lesions are normalities may occur. The white matter abnormali-
usually seen in the frontal white matter (Figs. 85.6 and ties may be limited in extent (Figs. 85.2 and 85.4), but
85.8). The white matter abnormalities are as a rule bi- may also be extensive (Figs. 85.3 and 85.5). In partic-
652 Chapter 85 Congenital and Perinatal Cytomegalovirus Infection

Fig. 85.3. A 11-month-old boy with CMV (diagnosis con- of low signal intensity around the ventricles, probably a rim of
firmed by a positive PCR for CMV DNA on the Guthrie card). ectopic neurons. The lateral ventricles are dilated. The inferior
The T2-weighted images show polymicrogyria of both hemi- horns are markedly dilated and the hippocampus has an ab-
spheres, but also of the cerebellar cortex. Only the posterior normal shape. Note the diffusely abnormal cerebral white
part of the brain has better gyral development. There is a rim matter

ular when the abnormalities are extensive, they are syndrome, muscle–eye–brain disease, and the Fuku-
often mistaken for a genetic leukoencephalopathy yama type of congenital muscular dystrophy. Howev-
and extensive laboratory tests are performed in that er, in addition to muscle weakness, patients with these
direction. latter conditions have other MRI abnormalities, such
In addition, anterior temporal abnormalities, in- as pontine hypoplasia and subcortical cerebellar
cluding abnormal and swollen white matter, subcorti- cysts. None of these are seen in congenital CMV. A
cal cysts, and focal enlargement of the anterior part of combination of multifocal white matter abnormali-
the inferior horn, either alone or more often in com- ties and anterior temporal cysts has been reported
bination, have been demonstrated to be particularly previously in a few children with a clinically static en-
suggestive of congenital CMV (Figs. 85.1–85.8). The cephalopathy (Olivier et al. 1998). It has been suggest-
enlarged inferior horns are related to an abnormal ed that this might be a novel, genetically determined
configuration of the hippocampi, which have a verti- leukoencephalopathy. Considering our findings, con-
cal orientation instead of the normal horizontal ori- genital CMV should also be considered.
entation, and are abnormally small (Fig. 85.7). A CT scan showing calcification could raise the
The MRI pattern suggestive of congenital CMV suspicion of congenital toxoplasmosis. In contrast to
does not resemble the pattern of any of the known congenital CMV, the intracranial calcifications ob-
leukoencephalopathies with the exception of some of served in congenital toxoplasmosis are usually scat-
the congenital muscular dystrophies. A combination tered diffusely throughout the brain and not in the
of cortical dysgyria and diffuse or multifocal white classic periventricular distribution of CMV, which
matter abnormalities is also seen in Walker–Warburg may be an important clue.
85.5 Magnetic Resonance Imaging 653

Fig. 85.4. A 6-year-old boy with con-


genital CMV infection, diagnosed at
birth. Note the unilateral left-sided
gyral abnormalities.The abnormalities
in the deep parietal white matter are
most pronounced on the right.The
inferior horn is dilated on the left

Fig. 85.5.
654 Chapter 85 Congenital and Perinatal Cytomegalovirus Infection

Fig. 85.5. (continued). A 17-month-old boy with congenital T1-weighted axial image (third row, left) shows the left-sided
CMV (diagnosis confirmed by a positive PCR for CMV DNA on gyral abnormalities. The sagittal T1-weighted images (third
the Guthrie card). The T2-weighted images (first and second row, middle and right) demonstrate the presence of multiple
rows) depict extensive, partly confluent, partly multifocal intraparenchymal and intraventricular cysts. Note the cysts in
white matter abnormalities. On the left side of the brain, corti- the anterior temporal white matter and the enlarged inferior
cal dysplasia is present, best visible on the higher sections.The horn. Courtesy of Dr. A. Clarke, Department of Pediatric Neurol-
inferior horns of the lateral ventricles are dilated and the ante- ogy, St. George’s Hospital, London, UK
rior temporal white matter is abnormal and swollen. The

Fig. 85.6. The boy presented with neonatal signs of a congen-


ital CMV infection and was diagnosed in the neonatal period.
The first MRI was obtained at the age of 1 month (first and sec-
ond rows). The T2-weighted images show that the cerebral
white matter has an abnormally high signal intensity and is
slightly swollen. The abnormalities are most pronounced in
the posterior region. It is difficult to identify lesions within
the high signal intensity of unmyelinated white matter. The
sagittal T1-weighted images (first row, left and middle) show
swelling of the anterior temporal white matter, a small cyst in
the inferior horn, and a subependymal cyst in the thalamocau-
date notch. The follow-up MRI was obtained at the age of
1.5 years (third and fourth rows).The T2-weighted images show
the typical multifocal white matter abnormalities with the
largest lesions in the deep parietal white matter. Note the
signal abnormality in the anterior temporal white matter.
85.5 Magnetic Resonance Imaging 655

Fig. 85.6.
656 Chapter 85 Congenital and Perinatal Cytomegalovirus Infection

Fig. 85.7.
85.5 Magnetic Resonance Imaging 657

Fig. 85.8. A 1.5-year-old boy, diagnosed with congenital CMV white matter is highly abnormal in signal and has a somewhat
in the neonatal period. Note the multifocal white matter ab- swollen appearance. The FLAIR images (third row, middle and
normalities with the largest lesions in the deep parietal white right) suggest that the anterior temporal white matter is
matter. The inferior horn is dilated and the anterior temporal almost cystic

Fig. 85.7. A baby girl presented soon after birth with an en- mainder of the cerebral white matter.The subependymal cysts
cephalopathy. She was diagnosed with a congenital CMV in- have disappeared. MRI obtained at the age of 2.5 years (third
fection on the basis of a positive PCR for CMV DNA on the and fourth rows) shows that the cerebral white matter looks
Guthrie card. The first MRI was obtained at 6 weeks (first row), much better myelinated.There are some remaining multifocal
which showed diffusely mildly abnormal cerebral white mat- white matter abnormalities, most prominent in the deep pari-
ter but no well-delineated lesions. There are subependymal etal region. The inferior horns are highly dilated. The coronal
cysts in the left thalamocaudate notch and in the left inferior FLAIR image (fourth row, middle) shows the abnormal shape of
horn. The hippocampus has an abnormal shape. Follow-up the hippocampus. The sagittal T1-weighted image (fourth row,
MRI at the age of 4 months (second row) shows that the pari- right) shows a cystic lesion anterior to the highly dilated inferi-
etal white matter is now evidently more abnormal than the re- or horn.
Chapter 86

Whipple Disease

86.1 Clinical Features serum iron, low serum carotene, proteinuria, hema-
and Laboratory Investigations turia, and steatorrhea. In Whipple disease with CNS
involvement, CSF oligoclonal banding and an in-
Whipple disease is a multisystem granulomatous dis- creased IgG level have been reported. In the initial
order. There is a male:female ratio of 6:1. The peak age phase an inflammatory reaction with CSF pleocytosis
of occurrence is between 50 and 55 years, but Whip- and elevated protein are usually seen. Increased IgA
ple disease may also occur in childhood and in senes- intrathecal synthesis, continuing even under therapy,
cence. Many organs may be involved, but predomi- has also been reported, probably indicating a still
nantly the gastrointestinal tract is affected. Migratory, active infection.
nonerosive arthritis of the large joints may precede The diagnosis is confirmed by either PCR of the
the intestinal symptoms by 2–10 years. The chief CSF or light and electron microscopy of a biopsy of
complaints of patients with systemic Whipple disease the jejunum, a retroperitoneal lymph node, or of
are diarrhea, weight loss, fever, arthritis, and abdom- affected brain tissue, demonstrating the presence of
inal swelling. Hypothalamic–pituitary involvement is the bacillus Tropheryma whippelii.
common in patients with systemic Whipple disease
and may lead to symptoms such as asomnia or hyper-
somnia, hyperphagia, polydipsia and weight gain. 86.2 Pathology
Physical findings in the multiorgan form of the dis-
ease may include lymphadenopathy, hypotension, The gross pathological features of Whipple disease
hyperpigmentation, hepatomegaly, splenomegaly, as- include generalized cerebral atrophy and small
cites, edema, cardiac murmur, pericarditis, pneumo- chalky nodules or granulomas up to 2 mm in diame-
nia, and occasionally ophthalmoplegia. ter, scattered diffusely over the cerebral and cerebel-
The CNS is involved in conjunction with other or- lar cortex and the subcortical white matter. The
gans in 10–40% of patients. CNS complaints may in- changes are focal, so that one area of the brain may be
clude headache, diplopia, depression, personality normal whereas an adjacent area may show florid ab-
changes, and cognitive decline. In a few patients the normalities. Microscopically the granulomas contain
CNS is the sole site of infection, without any sign of strongly PAS-positive-staining macrophages, sur-
involvement of other organs. The neurological symp- rounded by large reactive astrocytes. With more
toms in these cases are protean and include slowly widespread disease, PAS-positive cells infiltrate the
progressive dementia, headache, hypothalamic dys- white matter and may also extend into the subarach-
function, meningitis, seizures, supranuclear ophthal- noid spaces and be associated with death of neurons,
moplegia, myoclonus, ataxia, nystagmus, uveitis, pa- formation of vacuoles, and demyelination. Bacilli and
pilledema, hemiparesis, peripheral neuropathy, and debris of bacilli may be found in the PAS-positive ma-
myopathy. A movement disorder referred to as oculo- terial. Microinfarctions have been reported and are
masticatory myorhythmia, characterized by rhyth- probably caused by microthrombi related to vegeta-
mic opening and closure of the mouth by action of the tions on the heart valves, as found in a substantial
masticatory muscles, in combination with a slow number of patients at necropsy.
(1-Hz) convergent–divergent pendular nystagmus, is Epon-embedded toluidine-blue-stained semithin
considered virtually pathognomonic of Whipple dis- sections prepared for electron microscopy demon-
ease of the CNS. The triad of progressive dementia, strate gliotic gray matter with perivascular accumula-
myoclonus, and external ophthalmoplegia is also tion of macrophages containing multiple intracyto-
highly suggestive of the disease. plasmic osmiophilic dark granules. Ultrastructurally
Laboratory findings in systemic Whipple disease these dark granules consist of lipofuscin and unique
include elevated erythrocyte sedimentation rate, ele- membrane-bound inclusions containing numerous
vated liver enzymes, low serum potassium and calci- membranous profiles. Transitional forms from these
um, low serum protein and albumen, anemia, low unique inclusions to lipofuscin are also observed.
86.5 Magnetic Resonance Imaging 659

Fig. 86.1. Lesions in the hypothalamus often occur in Whip- with hypopituitarism. Similar lesions may be seen in Langer-
ple disease and are demonstrated here in T1-weighted, con- hans cell histiocytosis, neurosarcoidosis, brucellosis, and hypo-
trast-enhanced images in three planes in a 44-year-old man thalamic tumors. From Brändle et al. (1999), with permission

86.3 Pathogenetic Considerations unclear how the infection breaches the blood–brain
barrier. During active disease bacilli are found in
Whipple disease is caused by a bacillus with unique macrophages and free in the affected area.
characteristics. With electron microscopy it has been
consistently recognized in affected tissue, either free
or degraded to varying degrees within macrophages. 86.4 Therapy
It is a weakly gram-positive rod-shaped bacillus
which is not acid-fast. It is 1–2 mm in length and has a Therapy consists of a combination of antibiotics,
thick wall, the inner layer of which stains with PAS including penicillin, streptomycin, tetracycline, and
dyes. Its presence accounts for the brightly staining trimethoprim–sulfamethoxazole. Treatment is con-
PAS-positive macrophages which are seen in biopsy tinued for 1–2 years and relapses may occur even
material and contain bacillary debris. Culturing of the after many years.
bacillus has proven to be difficult.
Molecular genetic techniques led to the identifica-
tion of a 1321-base 16S rRNA gene sequence in tissue 86.5 Magnetic Resonance Imaging
derived from small bowel biopsy of patients assumed
to have Whipple disease. Using PCR, positive reac- As variable as the protean neurological manifesta-
tions have been obtained from other tissues, includ- tions of Whipple disease are the findings with MRI.
ing heart, vitreous fluid, peripheral blood cells, and Lesions may involve gray and white matter without a
pleural effusions. It has been difficult to identify se- distinct pattern. To illustrate this we cite a number of
quences from brain tissue; recently, however, a high findings in patients with confirmed Whipple disease:
yield has been obtained with PCR of the CSF. The ∑ Lesions in the mammillary bodies and hypothala-
bacillus was eventually characterized by 16S rRNA mus (Figs. 86.1 and 86.2)
gene analysis to be a member of the actinomycetes. ∑ Lesions in the hippocampal gyrus, uncus, and me-
The bacillus has been given the name Tropheryma dial or mediobasal temporal lobes (Fig. 86.2)
whippelii. It is still unclear whether all forms of Whip- ∑ Lesions in the hypothalamus, thalamus, and basal
ple disease are caused by this bacillus. ganglia
The occurrence in nature of this bacillus is still un- ∑ Lesions in the optic chiasm and tracts (Fig. 86.2)
clear. This is also the case with the transmission of the ∑ Diffuse cerebral atrophy and ventriculomegaly
disease to humans and the pathophysiology once the ∑ Lesions in the brain stem (Fig. 86.2)
infection is established. Humans are the only host for ∑ Spinal involvement (Fig. 86.3)
the disease. There is no evidence for human-to-hu- ∑ Multiple lesions throughout white and gray mat-
man transmission and the disease does not occur in ter, varying in size (Fig. 86.4)
clusters. Because the main site of the disease is in
the gastrointestinal tract, it has been assumed that A constant feature is enhancement after contrast. The
infection follows ingestion, subsequently spreading enhancing lesions are very similar to those seen in
hematogenously or via lymphatic channels. It is also other inflammatory and infectious disorders. From
660 Chapter 86 Whipple Disease

Fig. 86.2.
86.5 Magnetic Resonance Imaging 661

Fig. 86.3. The patient shown in Fig. 86.2 also has a lesion in Unit,Centre Hospitalier et Universitaire de Grenoble,Grenoble,
the spinal cord (arrows).From Kremer et al.(2001),with permis- France
sion and courtesy of Dr.S.Grand,Magnetic Resonance Imaging


the description of these lesions there seems to be
Fig. 86.2. A 68-year-old man with Whipple disease. The axial some preference for the hypothalamus and medial
FLAIR (first and second rows) and T2-weighted images (third temporal lobe. A clear message from these findings is
row) show a periventricular hyperintense rim and small focal that contrast must be given in patients with an unusu-
lesions in the deep white matter.The lower slices show lesions al pattern of lesions and an unexplained neurological
in the posterior limb of the internal capsule, hypothalamus, disorder.
thalamus, the mediobasal parts of the temporal lobes, the an- The differential diagnosis of multiple enhancing
terior commissure, mammillary bodies, optic chiasm, mesen- lesions of variable size involving both gray and white
cephalon,pons,middle cerebellar peduncles,and medulla.The matter is from multiple metastases, infectious and in-
coronal images (fourth row), FLAIR (left) and T1-weighted after flammatory disorders, including multiple sclerosis,
contrast injection (middle and right), show enhancement of acute disseminated encephalomyelitis, systemic lupus
the involved structures in the hypothalamus and mediobasal erythematosus, and Behçet disease. In patients with
temporal area, with an asymmetrical distribution. From Kre- predominant lesions in the hypothalamus and
mer et al. (2001), with permission and courtesy of Dr. S. Grand, suprasellar region, the differentiation is from Langer-
Magnetic Resonance Imaging Unit, Centre Hospitalier et Uni- hans cell histiocytosis, sarcoidosis, brucellosis, lym-
versitaire de Grenoble, Grenoble, France phoma, and other tumorous processes.
662 Chapter 86 Whipple Disease

Fig. 86.4.
86.5 Magnetic Resonance Imaging 663

Fig. 86.4. (continued). A 4-year-old boy with Whipple disease. isolated, of different sizes, involving both white and gray mat-
T2-weighted images are in the left column,T1-weighted images ter, randomly spread over the brain in an asymmetrical fash-
are at the same level in the middle column, and T1-weighted ion. Note that only few lesions enhance and that the enhance-
images with contrast in the right column. The images give an ment is partial. Some show a vague ring-like enhancement.
overview of the lesions. The multifocal lesions are rounded, From Duprez et al. (1996), with permission
Chapter 87

Toxic Encephalopathies

There is growing awareness that chronic intoxications disturbances; extrapyramidal movement abnormali-
by industrial, agricultural, iatrogenic, and environ- ties; disturbance of specific senses; disturbance of
mental pollution may have teratogenic or oncogenic coordination; and behavioral and psychological
effects or may cause neurological or psychiatric syn- changes.
dromes. A toxic encephalopathy (TE) is the result of In many forms of TE, clinical manifestations are
the interaction between a chemical compound and acute and severe, requiring immediate treatment.
the brain. Disturbance of normal brain function may Imaging of the brain is then only of secondary inter-
be caused by multiple factors, such as depletion of ox- est, although in unsuspected cases MRI may play a
idative energy, mitochondrial dysfunction, interfer- leading role in diagnosis. The role of imaging is as a
ence with biochemical pathways, alteration of mem- rule more important in cases of subacute or chronic
brane function and stability, enzymatic dysfunction, TE with slowly progressive neurological damage. In
derangement of neurotransmission, and altered ion such cases, MRI may demonstrate striking abnormal-
balance. It has often taken some time before the true ities and can be of importance for further diagnosis
cause of the encephalopathy was recognized, reflect- and therapy monitoring. However, even in cases with
ing the difficulty in recognizing that slow deteriora- histologically proven cerebral damage, MRI is not
tion of neurological functions may indicate poison- always positive. For example, tardive dyskinesia is a
ing by a toxin. In many cases, religious, superstitious, severe movement disorder due to the chronic use of
or racial “explanations” have been believed for a long neuroleptic drugs. Histology shows a decrease in the
time. The list of examples of TEs is long. Well-known number of ganglion cells in the substantia nigra. MRI
examples include: shows no abnormalities. In other cases, too, when the
1941 Spastic paraparesis, caused by Lathyrus encephalopathy is the result of interference with neu-
sativus peas; the toxic agent was identified as rotransmitters, for example in the malignant neu-
b-N-methylamino-L-alanine (BMAA) roleptic syndrome, MRI fails to show abnormalities.
1953 Guamanian type of parkinsonism, caused by Knowledge of the biomechanisms whereby toxins
the seeds of Cycas circinalis; the toxic agent of endogenous or exogenous origin cause encephalo-
was originally identified as b-N-oxalylmethy- pathy helps us to understand the patterns of lesions
lamino-L-alanine (BOMAA); later this theory seen in imaging studies. MRI abnormalities are, as a
was withdrawn, because blood concentrations rule, symmetrical as toxins have no left/right prefer-
were too low to explain the disease ence. Characteristic MRI patterns showing involve-
1948 Hexachlorophene encephalopathy, especially ment of particular brain structures are the result of
in neonates differences in regional vulnerability of brain tissue to
1950 Encephalopathy caused by monosodium glu- environmental perturbations and to biochemical
tamate in baby food changes. Here the concept of selective vulnerability
1953 Minamata disease (mercury encephalopathy) comes in (see also Chap. 3).
1960 House painter’s dementia, caused by organic As a general rule, specific groups of toxins tend to
solvents and carbon disulfide affect specific brain structures more than others. That
1983 Methylphenyltetrahydropyridine (MPTP),“syn- is, certain regions and systems within the brain have
thetic heroin”, causing striatal dopamine defi- greater affinity for and greater sensitivity to specific
ciency and parkinsonism types of toxins. These regions of identical affinity and
vulnerability were recognized by German neu-
In the past few decades drug-related encephalo- ropathologists in the first part of the twentieth centu-
pathies due to heroin, cocaine, or ecstasy, and iatro- ry, who designated them the topistische Bezirke or
genic intoxications due to more aggressive therapies topistic areas. Topistic areas often involve more than
against cancer and graft-versus-host disease, have be- one structure; they often encompass a whole func-
come more prominent and widespread. tional chain of neurons and tracts.
TE presents with one or more of the following The principle of functionally related systems is well
neurological or psychiatric symptoms (Bonhoeffer established in neurology. Topistic areas related to
types): decreased concentration and consciousness; functional systems can be readily identified during
excitability and convulsions; motor and sensory normal physiological development of the brain and
Toxic Encephalopathies 665

Fig. 87.1. T2-weighted series of transverse images in a 63- structures are initially spared. In the subsequent days a de-
year-old man with carbon monoxide intoxication. The globus layed leukoencephalopathy may follow
pallidus shows high signal intensity, whereas the other brain

in systemic degenerative disorders. As early as 1920, structures to energy depletion is also reflected in the
Flechsig recognized that functionally related systems preferential affliction of gray matter structures in car-
myelinate at the same time. Similarly, functionally re- bon monoxide intoxication, affecting especially the
lated and interdependent nuclei appear to degenerate globus pallidus (Fig. 87.1), and Leigh disease, where
at the same time in multiple system atrophy and pro- the putamen, caudate nucleus, globus pallidus, peri-
gressive supranuclear palsy. aqueductal gray matter, tectum and tegmentum of the
Other mechanisms of selective vulnerability in TE midbrain, and dentate nuclei may be involved. Wer-
are related to the similarity in particular physico- nicke encephalopathy, a toxic encephalopathy caused
chemical characteristics that make different geo- by thiamine deficiency in alcoholics, shows a similar-
graphic areas equally vulnerable to a particular nox- ity in pattern of involvement to Leigh disease, pre-
ious agent. Apparently diverse areas may prove to sumably because thiamine deficiency also influences
have similar oxygen requirements, chemical compo- energy metabolism. A difference is that in Wernicke
sition, and/or neurotransmitter dependency and den- encephalopathy the mammillary bodies are nearly
sity. always involved and putamen and caudate nucleus
Gray matter structures have higher cellular activi- are nearly always preserved (Fig. 87.2). The reason for
ty and a higher oxygen requirement than white mat- this difference is unknown.
ter structures and, therefore, are more vulnerable to An example of selective vulnerability resulting
oxygen deprivation. The damage that results from from specific chemical composition is found in
oxygen deprivation is actually mediated by toxic myelin. Myelin has high lipid content and a slow
products, such as excitatory amino acids and free rad- turnover. As a result, all the myelinated tracts are par-
icals inducing irreversible neuronal damage and ticularly vulnerable to the accumulation of lipophilic
death. The selective vulnerability of gray matter substances and to lipid peroxidation. Lipophilic sub-
666 Chapter 87 Toxic Encephalopathies

Fig. 87.2. In the acute phase of


Wernicke encephalopathy the
mammillary bodies are nearly always
involved.The axial proton density
image shows the lesions. On the coro-
nal T1-weighted contrast-enhanced
image the mammillary bodies show
uptake of contrast

Fig. 87.3. T1-weighted images of a


18-year-old girl with severe behavioral
disturbances after having taken a love
potion containing ecstasy. Cyst-like
lesions are seen in the globus pallidus
and the posterior parts of the puta-
men on both sides

stances easily cross the blood–brain barrier. One in- ties do not usually show abnormalities. However, in
stance of such intoxication has become notorious in some cases MRI successfully depicts the topistic
medical literature: hexachlorophene encephalopathy, areas, as shown in a case of ecstasy intoxication
a vacuolating myelinopathy, which was found in in- (Fig. 87.3).
fants who were washed with antiseptic hexachloro- Selective vulnerability is also related to the level of
phene solutions for dermal problems. The skin of activity during development. This concept was partic-
preterm neonates proved to be more permeable to ularly stressed by Dobbing (1968) and has broadened
these agents than the more mature skin, resulting in the insight into the origin of congenital malforma-
increased absorption and toxicity. In adults, vacuolat- tions of the CNS. The greatest impact of noxious
ing myelinopathy has been described after the use of agents is on those structures that grow and develop at
hexachlorophene solutions in vaginal tampons and as the highest rate at the time of insult. Thus, migra-
an antiseptic agent on burned areas. Intoxication with tional disorders result when toxic insults occur in the
triethyltin has identical effects. third to fifth month of gestation, the period in which
Topistic areas related to the distribution of a partic- neuronal migration occurs. Similarly, disorders of
ular neurotransmitter are best visualized by positron myelination are observed when toxic insults occur
emission tomography (PET). PET, for example, shows during the last trimester of pregnancy and the first
the distribution of 18fluorodopa in the basal ganglia. year of life, because myelination of the CNS occurs at
Methylphenyltetrahydropyridine (MPTP) interferes a high rate in these periods. Since normal myelination
selectively with the dopamine neurotransmitters and depends upon complex interactions between axons,
leads to severe parkinsonism. Tardive dyskinesia and myelin-forming oligodendrocytes, and the provision
malignant neuroleptic syndrome are other examples of substrates by the environment, the delicate interac-
of TEs with involvement of topistic areas related to tive process is easily disturbed by adverse factors such
specific neurotransmitters. In this kind of involve- as nutritional deficiencies, inborn errors of metabo-
ment of a neurotransmitter system, imaging modali- lism, and intoxications.
Toxic Encephalopathies 667

Toxic Encephalopathies

Exogenous Endogenous
Inborn errors of metabolism:
Globoid cell leukodystrophy
Glutaric aciduria type I
Methylmalonic acidemia

Canavan disease
External Internal
Classification Parainfectious agents
Sources Paraneoplastic agents
Industrial Hepatocerebral syndromes
Agricultural Endotoxins
Pharmacological (drugs) Ion balance disorders
Uremia
Effects
Oncogens
Teratogens
(Para)sympathicomimetics
(Para)sympathicolytics

Chemical composition
Lipophilic substances (organic solvents)
Heavy metals
Ethanol

Fig. 87.4. Classification of toxic encephalopathies

In this respect, it should be mentioned that age can cation is imperfect because it depends, in part, on the
be a prominent pathoplastic factor in TE. Many fetal point of view of the observer. A bacterial endotoxin
intoxications, such as those caused by maternal use of can be classified either as exogenous-external, be-
antiepileptic or antidepressive drugs, maternal alco- cause the infection by gram-negative bacteria stems
hol abuse, and maternal drug abuse, lead to serious from outside the body, or as exogenous-internal be-
syndromes of which the fetal alcohol syndrome and cause the endotoxin causing the related encephalopa-
fetal hydantoin syndrome are well-known examples. thy is produced by the bacteria within the body. We
Fetal intoxications may result in cerebral maldevelop- prefer exogenous-internal, because the toxin itself is
ment, cerebral disruptions, and arrest or delay of produced within – and in interaction with – the body.
cerebral maturation. The effects of intoxications may The largest group of toxins is exogenous-external.
also be different in different age groups. Lead intoxi- Toxins within this group are usually categorized by
cation in adults differs in presentation from that in chemical composition, source, or effect. Dietary and
childhood. This is also true of salt intoxication, where metabolic deficiencies may lead to abnormal bio-
the progress of myelination is an important factor in chemical processes that produce effects comparable
the distribution of the toxic damage. to intoxications.
To facilitate the analysis of TEs, we have classified The group of exogenous-external TEs includes all
them according to origin of the toxins within or out- intoxications from iatrogenic, agricultural, industrial,
side the blood–brain barrier (endogenous versus ex- environmental, and social (drugs, alcohol, nicotine)
ogenous). Exogenous TEs are then further subdivid- sources that affect the CNS. Belonging to this group
ed depending on whether the toxic substance origi- are intoxications with the Lathyrus sativus peas, or-
nates inside or outside the body (exogenous-internal ganic mercury poisoning, organic lead poisoning,
versus exogenous-external) (Fig. 87.4). This classifi- and toluene exposure, as well as other well-known en-
668 Chapter 87 Toxic Encephalopathies

Fig. 87.5. Mercury encephalopathy


in a 50-year-old man with a 2-year
course of progressive cerebellar and
focal cerebral signs following use of
a mercury-containing compound to
prevent tulip bulbs from cropping.
In particular on the gradient echo
images, which are more sensitive to
magnetic susceptibility changes,
the abnormal mineral depositions are
clearly shown in cerebellar tissue and,
to a less severe extent, in the occipital
area

cephalopathies associated with ethanol abuse, such as in particular the calcarine cortex. MR images show
the Marchiafava–Bignami syndrome, Wernicke en- the deposition of mercury in cerebellar structures
cephalopathy, and Korsakoff syndrome. Iatrogenic and under the occipital cortex (Fig. 87.5).
exogenous-external TEs include all TEs caused by the
ingestion of prescribed drugs, such as chemothera- Lead Encephalopathy. Children may have a lower
peutic agents, anticonvulsants, tranquilizers, and tolerance to lead than adults. Acute lead intoxication
anesthetic gases, or caused by medical treatment, is associated with convulsions, delirium, meningism,
such as progressive aluminum encephalopathy in pa- and papilledema. Chronic intoxication is associated
tients undergoing renal dialysis. Some fetal intoxica- with dementia, peripheral neuropathy, anemia, and a
tion syndromes can also be considered in this catego- variety of visceral features. Neuropathologically the
ry. A few examples are mentioned here. mildest chronic form of lead intoxication is the selec-
tive, segmental demyelination of peripheral nerves. In
Organic Mercury Poisoning. Ingestion of fish caught acute cases there may be demyelination and necrosis
in the poisoned bay of Minamata led to a neurologi- of central and cerebellar white matter. Neuronal dam-
cal disorder that was eventually identified as being age is pronounced.
caused by organic mercury. Mercury intoxication has
also been reported following accidental ingestion of Aluminum Dementia. Aluminum has been indicated
wheat that had been treated with organic mercury as a toxic factor in dialysis dementia. The excessive
compounds to prevent cropping. Neuropathology aluminum was probably derived from the high alu-
shows degeneration of the granular layer of the cere- minum concentration in the dialysate and from the
bellum and patchy loss of cells in the cerebral cortex, phosphate binding gels, which contain aluminum.
Toxic Encephalopathies 669

Fig. 87.6. Two men, aged 34 (first row) and 26 years (second ternal capsule (note the swelling), and the cerebellar white
row), with severe neurological disability after sniffing toxic matter. In all patients investigated in Amsterdam, an identical
heroin. There are lesions in the parieto-occipital white matter, MRI pattern has been observed
splenium of the corpus callosum, the posterior limb of the in-

Wernicke Encephalopathy. Wernicke encephalopa- Immunosuppressive Agents. In graft-versus-host


thy results from a deficiency of vitamin B1 (thiamine), disease, immunosuppressive agents such as cyclo-
and, as such, is not confined to chronic alcoholics. Be- sporine and tacrolimus are used to prevent the re-
cause vitamin B1 is a cofactor of transketolase, thi- pelling of transplanted organs. They may cause what
amine deficiency causes decreased activity of this en- is usually referred to as posterior reversible en-
zyme. The precise relationship of reduced enzyme ac- cephalopathy syndrome. This reaction will be dis-
tivity to damage in the characteristic “topistic area” cussed separately in Chap. 92.
represented by Wernicke encephalopathy is conjec-
tural. Korsakoff disease is now generally seen as a Heroin Pyrolysite. During the 1980s, it was discov-
chronic stage of Wernicke encephalopathy, with con- ered that a group of chronic heroin addicts in Amster-
sistent atrophy of the mammillary bodies and vari- dam manifested progressive neurological symptoms
able involvement of the dorsomedial nucleus of the after sniffing heroin (“chasing the dragon”). On neu-
thalamus. ropathological examination, a vacuolating myelino-
pathy was discovered in these cases. MR studies re-
Cytostatic Agents. Cytostatics, such as methotrexate vealed extensive involvement of the white matter of
and 5-fluorouracil, especially in combination with the cerebral hemispheres and the cerebellum and
levimasole, can lead to severe changes in the white clearly depicted the affected tracts from the parietal
matter when they are used to treat extracranial malig- cortex to the brain stem (Fig. 87.6). The toxic sub-
nancies or intracranial tumors (often in combination stance in the heroin has not been identified. One as-
with radiotherapy). These reactions will be dealt with sumes that it must be a lipophilic substance. Identical
in Chap. 88. neurological findings and MRI abnormalities have
670 Chapter 87 Toxic Encephalopathies

Fig. 87.7. Series of T2-weighted MR images in transverse and ing myelinopathy also exists in this case.The images beautiful-
coronal planes of a 22-year-old male cocaine sniffer. The ab- ly demonstrate all involved structures. Courtesy of Dr. J. Tam-
normalities are similar to those found in patients sniffing hero- raz, Department of Magnetic Resonance and Neuroimaging,
in. The swelling of the white matter suggests that a vacuolat- Hôtel-Dieu de France, Université Saint-Joseph, Beirut, Lebanon

been found among cocaine addicts (Fig. 87.7). Abuse tion, and level of exposure. The authors (Burg and
of cocaine more often leads to vascular complica- Gist 1999) compared the reporting rates for 25 health
tions. outcomes across subgroups. Statistically significant
increases in reporting rates were seen with, increased
Organic Solvents. Individuals who have been profes- maximum trichloroethylene exposure for the out-
sionally exposed over a long time to the inhalation of come stroke, increased cumulative chemical exposure
organic solvents may develop an organic psychologi- for respiratory allergies, and duration of exposure for
cal syndrome with loss of concentration, irritability, hearing impairment. Another registry study among a
emotional instability, and memory disorders. Al- population of demented individuals concluded that
though in most cases no structural abnormality can there was no support for the hypothesis that occupa-
be proven by MRI, in a number of cases changes in the tional exposure to organic solvents is a cause of de-
white matter, sometimes also the gray matter, varying mentia (Palmer et al. 1998). More recently, 74 patients
in degree from mild to severe, may be found in indi- were examined who had a long history of exposure to
viduals who have been exposed many years to organ- carbon disulfide. They were assessed with respect to
ic solvents or pesticides. In the United States of Amer- neuropsychological performance and MRI abnor-
ica a National Exposure Registry has published data malities (Cho et al. 2002). MRI findings revealed a sig-
on a longitudinal surveillance of populations exposed nificantly higher number of lacunae in the high expo-
to low concentrations of specific substances in the en- sure group than in a group with lower exposure.
vironment. The trichloroethylene subregistry con- Periventricular hyperintensities were most often lo-
tained 4041 individuals, who were divided into four cated in frontal and occipital areas (Fig. 87.8). Neu-
subgroups by types of exposure (chemicals), dura- ropsychological differences between those groups
Toxic Encephalopathies 671

Fig. 87.8. FLAIR series in a patient with carbon disulfide intox- what less symmetrical.The central part of the pons is affected.
ication. The images show a nearly symmetrical hyperintensity Courtesy of Dr C.C. Huang, Department of Neurology, Chang
of the deep and periventricular white matter and the corpus Gung Memorial Hospital and University, Taipei, Taiwan
callosum.There are multiple small basal ganglia lesions, some-

Fig. 87.9. A 68-year-old man with progressive memory loss, vale, both isolated and more confluent.The ventricles and sub-
who worked in the paint industry for many years and was pro- arachnoid spaces are moderately enlarged, related to cerebral
fessionally exposed to organic solvents. The T2-weighted MR atrophy.There are some isolated lesions in the basal ganglia
images show lesions scattered through the centrum semio-
672 Chapter 87 Toxic Encephalopathies

Fig. 87.10. T2-weighted series of a 64-year-old female patient over 30 years. There are widespread, focal, in some areas con-
with progressive subcortical dementia and gait disturbances. fluent, signal abnormalities in the white matter of the hemi-
This patient had been handling insecticides and pesticides for spheres and middle cerebellar peduncles

were not significant. In our own experience with MR matter changes started in the deep white matter and
and EEG studies of 50 persons with long-lasting ex- progressed to the periphery. Other reports state di-
posure to organic solvents MR showed only minor minished distinction between gray and white matter.
white matter abnormalities in two persons. Whether Another highly lipophilic substance, is fentanyl, a
this was related to the exposure to organic solvents re- synthetic opioid many times stronger than morphine.
mained unclear. On the other hand, we have seen two It is used in anesthesia and in intensive care units. We
patients with severe exposure to organic waste prod- observed a patient who had attempted suicide by
ucts and pesticides for more than 30 years who had transdermal application of fentanyl. MRI in this
severe white matter abnormalities dispersed over the comatose patient showed extensive white matter ab-
entire brain (Figs. 87.9 and 87.10). normalities, with ADC characteristics of increased
Toluene is one example of a typical lipophilic sub- diffusivity (Fig. 87.11). The leukoencephalopathy was
stance that persists in the myelin for a long time, lead- most prominent in the parieto-occipital region. In
ing to myelin and gray matter damage. Toluene is a this respect the pattern resembles the pattern seen in
volatile liquid that is used as an industrial solvent. It posterior reversible encephalopathy syndrome. In
is inhaled by some individuals as a form of substance this case the abnormalities were not transient; the pa-
abuse. Long-term abuse leads to cognitive deteriora- tient did not survive.
tion, cerebellar ataxia, tremor, and anosmia. Chronic Common to disorders caused by exogenous-inter-
use of toluene leads to white matter lesions in 46% of nal toxins is a focal or generalized process in the body
the patients, cortical and central atrophy in 26%, and outside the blood–brain barrier that produces a toxin
thalamic hypointensity in 20% (Aydin et al. 2002). that crosses the blood–brain barrier, enters the brain,
The white matter changes in 41 patients were focal in and gives rise to encephalopathy. Encephalopathies
53% and diffuse in 47% (Aydin et al. 2002). White related to toxins released by bacteria elsewhere in
Toxic Encephalopathies 673

Fig. 87.11.
674 Chapter 87 Toxic Encephalopathies

Fig. 87.11. (continued). A 39-year-old nurse committed sui- The lesions have a high signal on the Trace diffusion-weighted
cide using fentanyl dermal patches. The T2-weighted images images (fifth row, b = 1000); the ADC values are, however, high
(first and second rows) show predominant involvement of the (sixth row). This MR pattern is comparable to what is seen in
parieto-occipital areas, but also involvement of the basal gan- posterior reversible encephalopathy syndrome. Courtesy of
glia (especially the globus pallidus) and the posterior limb of Dr. M. Heitbrink and Dr. B. Wiarda, Department of Radiology,
the internal capsule. The FLAIR images (third and fourth rows) Medical Center Alkmaar, The Netherlands
more clearly show that there is also cerebellar involvement.

the body, and also paraneoplastic syndromes and Parainfectious encephalopathy has been reported
parainfectious degeneration can be included in this with Escherichia coli, Mycoplasma pneumoniae, and
category. Tumors may contain antigens that lead to diphtheria infections. The reaction may be toxic,
the formation of antibodies which cannot distinguish allergic, or both.
between tumor cells and own-body cells, for example Changes in the ion balance are held responsible for
Purkinje cells. Malignant disease anywhere in the central pontine and extrapontine myelinolysis. Hy-
body may have a remote effect on the peripheral ponatremia is considered to be an important initial
nerves, the spinal cord, and on the brain, causing factor in these disorders, subsequently precipitated
peripheral neuropathy, subacute necrotizing myelo- by (too) fast restoration of the blood sodium concen-
pathy, and encephalomyeloradiculitis. The ence- tration. These conditions are discussed in more detail
phalomyeloradiculitis group includes two special in Chap. 89.
forms: brain stem encephalitis, and so-called limbic Inborn errors of metabolism may lead to accumu-
encephalitis in which the changes are restricted to lation of toxic substances produced outside the
the limbic system (Fig. 87.12). The different mani- blood–brain barrier and their then crossing the
festations presumably reflect the different antigenic blood–brain barrier, leading to toxic brain damage.
content of these brain structures. It is possible that a Many of the disorders described in the first part of
more general toxic effect plays an additional role. this book may be considered an exogenous-internal
TE. Examples are many of the amino acidopathies
Toxic Encephalopathies 675

Fig. 87.12. Patient with limbic encephalitis. The images show ment of the thalamus, in particular the pulvinar. In this case
the typical symmetrical involvement of the medial parts of there are also symmetrical lesions in the posterolateral parts of
both temporal lobes, including the hippocampus, involve- the midbrain

and organic acidopathies, including the urea cycle myelin. Hypocortisolism, hypercortisolism, and a de-
disorders. In Wilson disease, the encephalopathy is re- ficiency of growth hormone also result in hypomyeli-
lated to excessive deposition of copper in the brain, nation in infants.
especially in the globus pallidus. Failure of internal organs may lead to exogenous-
Hormonal abnormalities in infants may disturb internal TEs. Well-known examples are renal failure
the normal process of myelination. Thyroid deficien- and hepatic failure. Hepatocerebral syndromes mani-
cy in the neonatal period causes disturbed develop- fest by cerebral atrophy, by changes in signal intensi-
ment of the CNS with a decreased formation of ty in the basal ganglia, and by T1-shortening of the
676 Chapter 87 Toxic Encephalopathies

Fig. 87.13. T1-weighted (SE) sagittal


and transverse (IR) images in 3-year-
old boy with hepatic failure and
hepatic encephalopathy.The images
show the typical T1 shortening in the
basal ganglia but also of the white
matter. Due to T1 shortening the basal
ganglia can hardly be distinguished
from the surrounding white matter.
The pons shows T1 shortening in the
posterior part, leading to a pattern on
the T1-weighted sagittal image as seen
in neonates

white matter (particularly in infants and children). ganglia can make the basal ganglia indistinguishable
1H-MR spectroscopy shows an increase in glutamine
from white matter on T1-weighted images
and a depletion of myo-inositol.Ammonia plays a key (Fig. 87.13).At the same time, there is T1 shortening in
role in hepatic encephalopathy. It appears to cause the white matter, typically sparing the pons. In older
neurotoxicity by interacting with the glutamate/glut- patients, this spread of T1 shortening over the white
amine/GABA balance. Apart from its other roles in matter is less clear. The T1 shortening of gray and
metabolism, glutamate is the most important excita- white matter may be observed with a number of tox-
tory neurotransmitter. g-Aminobutyric acid (GABA), ic-metabolic conditions, not just hepatic en-
formed by decarboxylation from glutamate, is the cephalopathy. For instance, we have seen this phe-
most important inhibitory neurotransmitter. In the nomenon in a child with hemorrhagic shock and en-
presynaptic neuron glutamate is formed from gluta- cephalopathy syndrome (Fig. 87.14). It has also been
mine by glutaminase. After neurotransmission gluta- reported in patients who received parenteral nutri-
mate is taken up by astrocytes where it is processed tion over a long period.
by glutamine synthetase into glutamine. Glutamine is GABA is considered to be an inhibitory neuro-
transported to the presynaptic neuron where glutam- transmitter and the counterpart of glutamate. It has
inase catalyzes the formation of glutamate available been used in anesthesia for good reasons, but has ob-
for neurotransmission. Hyperammonemia has a great tained a bad reputation as a “rape” drug. We have ob-
impact on this cycle by stimulating glutamine synthe- served cortical infarctions and neurological deficits
sis via glutamine synthetase, by possible inhibition after the use of this drug, probably at excessive dosage
of glutaminase, and by inhibition of glutamate reup- (Fig. 87.15).
take by the astrocyte. However, the relation between The endogenous forms of TE are the smallest
the neurotransmitters’ changes and the T1 shortening group. A number of inborn errors of metabolism
as seen on MR is not clear. Other explanations for the should be included here, because their effects are me-
T1 shortening of the basal ganglia have been suggest- diated by a toxic substance produced locally. In par-
ed, such as accumulation of manganese or of lipid ticular, this is the case in lysosomal storage disorders,
particles. In MR images, T1 shortening in the basal such as Krabbe disease or globoid cell leukodystro-
Toxic Encephalopathies 677

Fig. 87.14. A 7-month-old girl with


hemorrhagic shock and encephalo-
pathy syndrome. Hemorrhagic laminar
cortical lesions are seen in the occipi-
tal lobe.There is also a striking
T1 shortening in the globus pallidus
on the T1-weighted images.The
T2-weighted transverse image (second
row, right) shows generalized atrophy,
presence of lesions in the occipital
area, but no clear signal changes
in the basal ganglia

phy. The primary defect is galactocerebroside b- child has reached, as well as upon the nature of the
galactosidase deficiency. This enzyme has two func- toxin, its concentration, and the duration of the expo-
tions: it degrades cerebroside into galactose and ce- sure. Toxic or teratogenic substances have a great im-
ramide and it hydrolyzes psychosine (galactosyl pact on the developing fetus. The effects are often
sphingosine). Psychosine is a toxic metabolite that is widespread and involve multiple parts of the body or
essentially nonexistent in normal brain. Presence of the whole body in addition to the brain. The gamut of
elevated psychosine in Krabbe disease causes early, possible dysgenesis is extensive and ranges from fatal
very rapid, almost complete death of the oligoden- malformation with early spontaneous abortion to
droglia with extensive loss of myelin sheaths main- mild alteration in morphology and function. The
tained by these cells. In addition, inability to degrade sources of the toxins include:
cerebroside leads to an abnormally high concentra- 1. Medical treatment. Syndromes can result from the
tion of this substance within the myelin membrane, use of established drugs prescribed to the mother,
eventually resulting in myelin instability and break- but taken during pregnancy. Most such cases are
down. In Krabbe disease, the disturbance of both accidental. A few arise when drugs are given
functions of galactocerebroside b-galactosidase con- knowingly in desperate cases. Antiepileptic drugs
tributes to the myelin loss. In metachromatic leuko- are known to cause developmental damage, result-
dystrophy it is the local storage of sulfatide that caus- ing, for example, in the fetal hydantoin syndrome
es myelin damage and loss. Other inborn errors of and the fetal valproate syndrome.
metabolism, glutaric aciduria type 1 and methyl- 2. Alcohol and drug abuse. An important cause of fe-
malonic acidemia, produce compounds with a direct tal dysgenesis is the use of drugs or alcohol during
toxic effect on the basal ganglia (see also Chap. 3). pregnancy. The fetal alcohol syndrome has been
The toxic interactions interfering with fetal devel- reported extensively in the literature. These chil-
opment can be considered as exogenous-external in- dren are growth retarded, have short palpebral fis-
toxication, reaching the unborn child via the mother. sures, a low nasal bridge, epicanthus, a long convex
The fetal syndromes are unique, because the effect of upper lip, and mental retardation. The numerous
the toxin depends upon the stage of development the CNS anomalies of the fetal alcohol syndrome in-
678 Chapter 87 Toxic Encephalopathies

Fig. 87.15. Transverse FLAIR (first row) and sagittal contrast raped. GABA was used to subdue the victim.The images show
enhanced T1-weighted images (second row) of a 17-year-old multiple cortical infarctions
girl who had been administered GABA and subsequently

clude: derangement of neuronal and glial migra- anal atresia, sacral dysgenesis or agenesis, high
tion, microcephaly, hydrocephaly, porencephaly, position of the conus medullaris, and diverse uro-
agenesis of the corpus callosum, meningomyelo- logical, neurological, and orthopedic disorders.
cele, and Dandy–Walker malformation. Microcephaly can be caused by maternal phenyl-
3. Metabolic disorders of the mother. Fetal develop- ketonuria.
ment may suffer if the mother has a metabolic dis- 4. Teratogenic chemical substances. Compounds
order. Diabetes mellitus may be one causative fac- with teratogenic action may be encountered in the
tor in the caudal regression syndrome, a complex direct environment (pollution, professional con-
abnormality of the caudal end of the embryo with tact with chemicals).
Chapter 88

Iatrogenic Toxic Encephalopathies

In the last decades more aggressive treatment meth- In this chapter we will discuss the side effects of
ods have been used to control otherwise fatal dis- drugs used in modern therapeutic strategies. Fortu-
eases. This kind of treatment has a considerable im- nately most of these side effects are reversible or, de-
pact on the temporary improvement, cure, and sur- spite their visibility on MRI, do not lead to permanent
vival rate of patients with life-threatening conditions. major neurological dysfunction. An early clinical and
It has been known for a long time that radiation ther- radiological diagnosis is important. The therapeutic
apy of the brain, either for prophylaxis as in acute regimen should be modified or aborted and, if neces-
lymphoblastic leukemia (ALL) of childhood, or for sary and possible, replaced by a therapy with similar
therapeutic reasons as in tumors of the brain, can lead effect. This will in most cases lead to disappearance of
to radiation damage of the brain, either as radiation the clinical symptoms and MR manifestations, but
necrosis or leukoencephalopathy, focal or diffuse (see not in all.
Chap. 102). When radiation therapy is used in combi- The reported reactions to the drugs cited in the
nation with intravenous or intrathecal methotrexate, Table 88.1 may be divided in two groups of abnormal-
the lesions can be even more pronounced. The intro- ities:
duction of bone marrow and organ transplantation in
the treatment of a number of disorders has increased
the numbers both of reversible and of permanent le- 88.1 Multifocal Inflammatory
sions in the central and/or peripheral nervous system Leukoencephalopathy
related to the medical treatment before and after the
intervention. This means that radiologists are in- In the patients with multifocal inflammatory leuko-
creasingly being confronted with lesions of the brain encephalopathy (MIL) the lesions may appear multi-
that are therapy-related, either during the period of ple sclerosis-like (Fig. 88.1). There are multiple le-
intervention, during the months following it, or even sions in the centrum semiovale, some with the ap-
after an intervention that took place many years pre- pearance of Dawson’s fingers. Many lesions may en-
viously hance after contrast injection. The enhancement
The list of compounds that can be responsible for may be ring-like. In other cases the lesions have
brain lesions is long, and for many of them the nature the appearance of progressive multifocal leukoen-
of the damage they may cause is known. Table 88.1 cephalopathy, with larger confluent lesions extending
shows where the pharmaceutical compound has its into the arcuate fibers. Sometimes the multiple en-
place in present treatment plans. Notice that for some hancing lesions are taken for cerebral metastases and
interventions an aggressive pretreatment is neces- brain biopsy may be performed. The latter is also
sary. This pretreatment may cause lesions of the prompted by the clinical presentation of a multifocal
brain, but it has also been shown that the choice of neurological syndrome with subacute confusion,
pretreatment medication can influence the later out- ataxia, dysarthria, diplopia, seizures, and other neuro-
come of drugs used after the intervention. logical signs. Neuropathology of MIL shows marked

Table 88.1. Schematic overview of


Pretreatment Treatment Post-treatment
the most common drugs used before
and after the central intervention.
Immunosuppression Radiotherapy Anti-GVHD
The list of drugs is incomplete
Chemotherapy Bone marrow transplant Cyclosporine
Cyclophosphamide Organ transplant Tacrolimus (FK506)
Doxorubicin Cytostatics Corticosteroids
Vincristine Chemotherapy
Prednisone 5-Fluorouracil
L-Asparaginase Levamisole
Cytosine arabinoside Pegaspargase
Cisplatin Progenitor stem cell support
BCNU

GVHD, graft-versus-host disease; BCNU, 1,3-bis-(2-chloroethyl)-1-nitrosurea


680 Chapter 88 Iatrogenic Toxic Encephalopathies

Fig. 88.1. Multifocal inflammatory leukoencephalopathy in a age on the right). After discontinuation of levamisole and un-
57-year-old woman during adjuvant therapy with levamisole der treatment with steroids, the clinical picture improved dra-
for malignant melanoma. Over a period of 3 weeks the patient matically. The MRI improved too. From Kimmel et al. (1995),
became progressively confused and ataxic. The proton densi- with permission, and courtesy of Dr. E.F.M. Wijdicks, Depart-
ty image (left) shows multifocal white matter lesions, which ment of Neurology, Mayo Clinic, Rochester Minnesota, USA
enhance after contrast (contrast-enhanced T1-weighted im-

Fig. 88.2. The T2-weighted images show the classical pattern terior cerebral artery and the middle and anterior cerebral
of the posterior reversible encephalopathy syndrome with in- artery
volvement of the border zones between the middle and pos-
88.2 Posterior Reversible Encephalopathy Syndrome 681

focal cellularity of the white matter, with mononu- tion is only one of the possible neurological side ef-
clear cells scattered throughout the white matter and fects of these drugs, and effects on other organs are
around the blood vessels. The mononuclear cells are also known. In PRES there is, as in hypertensive en-
mainly macrophages. Knowledge of the drugs the pa- cephalopathy, preferential involvement of the occipi-
tient is taking will, of course, lead to the correct diag- tal lobes, but other locations – the frontal lobes, bor-
nosis and treatment. MIL is mostly seen in patients der zones, mesencephalon, and pons and cerebellum
with colon cancer treated with a combination of 5- – have also been reported. The preference for areas in
fluorouracil (5-FU) and levamisole, the latter proba- the vertebrobasilar arterial territory is considered to
bly playing the dominant role and 5-fluorouracil act- be due to the lack of sympathetic–parasympathetic
ing as potentiator of the immunostimulatory actions fibers around these vessels, with poorer autoregula-
of levamisole (Fig. 88.1). A patient has been reported tion as a result. There are many other suggestions for
who developed MIL after treatment with 5-fluo- how to explain the selective damage: for example, lo-
rouracil alone. This patient, however, had a deficiency cal endothelial damage with the release of vasoactive
of dihydropyrimidine dehydrogenase, an enzyme peptides leading to labile blood pressure and va-
necessary for 5-fluorouracil catabolism. MIL has also sospasm, and thrombotic microangiopathy leading to
been described in patients treated with 5-fluorouracil microvascular damage. None of these theories is com-
derivatives, such as tegafur [1-(2-tetrahydrofuryl)-5- pletely satisfactory. In some cases only the subcortical
fluoro-uracil] and carmofur (1-hexylcarbamoyl-5- white matter is involved, the gray matter being
fluoro-uracil).After discontinuing the therapy the en- spared. In other cases the cortex is also involved.
cephalopathy usually still progresses for 1 or 2 There is evidence for a relation between the condi-
months, and then gradually clinical and radiological tioning regimens used before allogeneic bone mar-
improvement occurs, although often incomplete. row transplant and the type of lesions observed under
post-treatment medication. Patients under post-
treatment medication who were pretreated with cy-
88.2 Posterior Reversible clophosphamide and a chemotherapeutic agent, such
Encephalopathy Syndrome as busulfan or thiotepa, develop cortical abnormali-
ties with various degrees of subcortical white matter
Drugs used in the prevention of graft-versus-host involvement. Nontransplant and transplant patients
disease (GVHD) may lead to a reversible encephalo- treated with cyclophosphamide and total body irradi-
pathy syndrome, posterior reversible encephalopathy ation have lesions in the subcortical and deep white
syndrome (PRES), that bears resemblance to what is matter, without cortical involvement. This exempli-
seen in acute hypertensive encephalopathy (Figs. fies how the final result is the last stage of a complex
88.2–88.4; see also Chap. 92). In fact, the reaction may process. Cyclosporine-related toxicity is more often
be mediated by hypertension, but there are excep- seen in liver transplantation than in cardiac or renal
tions. Clinically the presentation of the syndrome transplantation. The elimination of cyclosporine is
consists of altered mental status, headaches, seizures, through hepatic metabolism via the P-450 cyto-
and cerebral blindness. Drugs most often used in chrome oxidase system, and the concentration of bio-
GVHD are cyclosporine and tacrolimus. Cyclosporine logically active cyclosporine depends on the blood
is of great importance in organ and bone marrow cholesterol level. For this reason, cyclosporine toxici-
transplantation. The activity of cyclosporine is T-cell- ty occurs especially in the period shortly after liver
mediated and inhibits calcineurin, which plays a role transplantation, when the new liver has not regained
in calcium-mediated cell death. Cyclosporine is also its full functional capability. It is also clear that the
used in autoimmune disorders, insulin-dependent di- pre-existing condition of the patient, which necessi-
abetes mellitus, inflammatory bowel disease, chronic tated the liver transplant and may have included a he-
asthma, rheumatoid arthritis, aplastic anemia, and patocerebral syndrome, alcoholic liver cirrhosis, and
psoriasis. Tacrolimus (FK 506) suppresses both the a severe degree of alcohol-related neurological dis-
cell-mediated and humoral immune responses by se- ease, may obscure the symptoms of cyclosporine or
lectively inhibiting the expression of T lymphocytes tacrolimus toxicity. In these potentially reversible tox-
of a subset of early-phase activation genes, including ic disorders diffusion-weighted images and ADC
interleukin (IL)-2, IL-3, IL-4, interferon-g, tumor maps are important. They help to differentiate be-
necrosis factor, and granulocyte colony-stimulating tween cytotoxic edema and vacuolating myelinopa-
factor. Tacrolimus binds, as cyclosporine, to specific thy on one hand, and vasogenic edema on the other.
intracellular protein ligands, which play a role in As a rule, increased diffusivity is seen in reversible
maintaining the stability of intracellular structures. conditions and is helpful in the initial diagnosis,
Tacrolimus and cyclosporine inhibit the activity of whereas decreased diffusivity usually predicts a
calcineurin, but it is unclear how this relates to the de- poorer prognosis.
velopment of leukoencephalopathy. The PRES reac-
682 Chapter 88 Iatrogenic Toxic Encephalopathies

Fig. 88.3. Images of a 14-year-old girl with Henoch–Schönlein FLAIR images show some minor lesions in the typical spots in
purpura treated with cyclosporine. T2-weighted images did the posterior parts of the hemispheres
not show abnormalities, despite neurological symptoms.

In the last years it has become clear that toxic ef- after bone marrow transplant, which involves the use
fects may appear a long time, even many years, after of total body irradiation and immunosuppressive and
the intervention, and the link between the interven- chemotherapy, severe, fatal leukoencephalopathy may
tion and the brain abnormalities may not be obvious. occur.
We have just started to learn that even many years
88.2 Posterior Reversible Encephalopathy Syndrome 683

Fig. 88.4. A 45-year-old woman


underwent lung transplantation and
treatment with cyclosporine to pre-
vent graft-versus-host disease.The
sagittal T2-weighted images (first row)
show a lesion selectively located in
the mesencephalon and upper pons.
The FLAIR images (second row) show
this lesion even more clearly.The
contrast-enhanced T1-weighted
images (third row) demonstrate that
the core of the lesion enhances. After
the medication was changed, the
lesion disappeared within 2 weeks
Chapter 89

Central Pontine and Extrapontine Myelinolysis

89.1 Clinical Features tion of neurological findings is not specific for


and Laboratory Findings CPM/EPM and may also be seen in other types of
brain dysfunction, particularly in pontine infarction
Central pontine myelinolysis (CPM) is a demyelinat- or hemorrhage. Clinically, CPM/EPM can only be sus-
ing disorder affecting mainly the central part of the pected, especially if the neurological condition of a
pons. In some cases, CPM is accompanied by ex- patient deteriorates after correction of hyponatremia
trapontine myelinolysis (EPM, together CPM/EPM). (serum sodium less than 120 mEq/l). Symptoms of
CPM/EPM is not a primary disease but develops the disease appear within hours or up to a week after
against a background of other, usually severe condi- correction of hyponatremia. Many patients with neu-
tions, apparently as a complication of the main dis- rological manifestations of CPM/EPM die of compli-
ease. It occurs most often in alcoholics but has also cations or underlying disease. However, other pa-
been reported in association with diabetic ketoacido- tients survive with partial or complete recovery.
sis, psychogenic excessive water drinking, inappro- In CPM the EEG is rarely abnormal, because even
priate antidiuretic hormone secretion, malnutrition, large lesions of the base of the pons are compatible
and chronic debilitating diseases. It is probable that with a normal EEG. The most frequently observed
the underlying cause of myelinolysis in all these dis- EEG abnormality is a generalized slowing, correlating
orders is a derangement of serum sodium concentra- with the level of consciousness. In EPM, the EEG may
tion, particularly a rapid correction of hyponatremia show focal abnormalities. BAEPs and SSEPs often
and rapid changes in osmotic conditions. show abnormalities consistent with a pontine lesion,
Most of the patients are severely ill as a result of the but may also be completely normal. The abnormali-
primary disorder. It takes a period of a few days for ties are nonspecific.
patients to develop the clinical symptoms caused by
CPM/EPM. Mental confusion is present in many but
not all cases. Sometimes patients suddenly lapse into 89.2 Pathology
coma for no apparent reason. No particular type of
mental change is specific for CPM and EPM, as there In CPM there is usually a single symmetrical lesion,
are a variety of associated clinical conditions, some of often with a typical “w” form, located in the central
which have their own mental counterparts, such as part of the base of the pons. It can be large, occupying
Wernicke encephalopathy, delirium tremens and var- almost the entire base of the pons, or very small. Gen-
ious metabolic encephalopathies. Frequently a bulbar erally the lesion is continuous and sharply defined, al-
or pseudobulbar syndrome is present, manifesting it- though an island of tissue is occasionally preserved.
self in dysarthria or anarthria, dysphagia, and some- In exceptional cases the lesion develops unilaterally
times weakness of the neck muscles. Involvement of or appears to be multifocal. Rarely, the lesion spreads
the facial nerve is uncommon; when present, it is usu- into the tegmentum pontis or into the middle cerebel-
ally of the central type and part of a hemispheric syn- lar peduncles. It does not usually extend to the surface
drome. Occasionally a limitation of conjugate move- of the pons.
ment of the eyes is found. Oculomotor or abducens Histologically, there is severe and sometimes com-
nerve dysfunction is rarely observed. Pupillary dis- plete loss of myelin in the lesion, with a concomitant
turbances may be present. The weakness of the limbs loss of oligodendrocytes. Demyelination affects both
is often severe, resulting in tetraparesis or tetraplegia. the long tracts (pontocerebellar transverse fibers and
The usual type of motor weakness is flaccid with hy- the long descending tracts) and the pontine nuclei.
poreflexia. Extensor plantar reflexes and abnormal The axons are usually well preserved, although a con-
bulbar reflexes are often present. Because of the siderable loss of axons may occur. The most severely
weakness the testing of cerebellar function is often affected area may be cystic in nature due to almost to-
impossible. Extrapyramidal features may occur. Sen- tal disappearance of myelin and axons. Axonal loss is
sory findings are rare. associated with degenerative changes in nerve cells.
The clinical manifestations of CPM/EPM vary, ac- However, on the whole, neurons of the pontine nuclei
cording to the extent of the lesions, from minimal to a are relatively spared even in areas of severe demyeli-
complete locked-in syndrome or coma. The constella- nation. Reactive astrocytosis and macrophage re-
89.3 Pathogenetic Considerations 685

sponse are usually present. Macrophages contain blocking effect. Alcohol withdrawal leads to rapid re-
remnants of disintegrated myelin and lipid material. turn of antidiuretic hormone function, thus causing
Neither vascular disease nor inflammation is seen. hyponatremia. With intravenous tubing, aggressive
In EPM, which accompanies CPM in 10% of pa- treatment of hyponatremia by infusion of hypertonic
tients, similar demyelinating lesions occur outside solutions became possible in any of the implicated
the pons, with a predilection for the white matter of diseases. The clinical syndrome and the characteristic
the cerebellum, especially the folia. Also affected are histological lesions of CPM/EPM can be reproduced
the mammillary bodies, the tegmentum of the mid- in experimental work with dogs and rats by rapid
brain, the lateral geniculate bodies, the thalamus, correction of hyponatremia. In these experiments,
striatum, the internal, external, and extreme capsules, hyponatremia in itself does not appear sufficient to
the anterior commissure, the fornix, the deep layers of induce CPM/EPM, and neither self-correction of the
the cerebral cortex and subjacent white matter of the serum sodium nor a rapid rise from normonatremia
crowns and sides of the cerebral gyri. Not all these to hypernatremia results in CPM/EPM.
structures are necessarily affected in any one patient; However, there are other, conflicting data. Some in-
the precise localization of demyelinated lesions varies vestigators found that the rate of increase in serum
from patient to patient. In exceptional cases, histolog- sodium in patients with CPM/EPM had been more
ical abnormalities consistent with the diagnosis EPM rapid than in patients with hyponatremia but without
occur without any pontine lesion. CPM/EPM. Others found no difference in the rate of
increase of serum sodium in the two groups. Some-
times patients with a slow rise in serum sodium still
89.3 Pathogenetic Considerations develop CPM/EPM. Some authors stress that most of
the patients developing CPM/EPM became hyperna-
CPM and EPM have been reported in quite a number tremic after correction and state that it is the eleva-
of conditions: alcoholism, psychogenic water drink- tion of serum sodium to hypernatremic levels rather
ing, diabetic ketoacidosis, subdural abscess, brain than the rapid correction that predisposes to CPM/
tumor, cerebral trauma, meningitis, encephalitis, EPM. Others have found that CPM/EPM can develop
renal dialysis, malnutrition, lung infections, adreno- without overcorrection, while a mild hyponatremia
cortical insufficiency, postoperative hyponatremia, still exists. In a recent discussion, attention was paid
inappropriate antidiuretic hormone secretion, extra- to the combined role of hyponatremia and hypoxia.
cerebral malignancy, and other chronic debilitating The new point of view was that independent of the
diseases. The etiology is not completely understood. rate of correction of the hyponatremia, brain stem
Abnormalities in serum sodium levels have been the lesions develop when hypoxia is present.
subject of considerable interest in this respect. There The exact pathogenesis of demyelination in CPM/
is evidence that rapid correction of hyponatremia EPM is not clear. It is known that generalized cellular
may cause CPM/EPM. Clinical observations strongly edema is the hallmark of acute hyponatremia. In cas-
suggest this causal relationship as the clinical symp- es of chronic hyponatremia, brain cells have adapted
tomatology in CPM/EPM is usually preceded by a to this condition by extruding sodium, potassium,
rapid rise in serum sodium after a period of sustained chloride, and water. The loss of more solute than wa-
hyponatremia. Usually, no neurological injury is ter renders cellular and extracellular fluid equally
seen when the chronic hyponatremia is corrected hypotonic, but reduces cell swelling. Restoration of
slowly. In all diseases inferred to be able to induce normal serum osmolality removes water from the hy-
CPM/EPM, hyponatremia is a common complication, potonic tissue and causes the adapted, normovolemic
for which rigorous treatment is often instituted after brain cells to shrink to volumes less than normal. As
hospitalization. brain cells replenish lost electrolytes, the normal cel-
There are strong indications that CPM/EPM is in- lular volume is re-established. Complications may re-
deed an iatrogenic disease. The first description of sult if normonatremia is achieved too rapidly before
this syndrome dates from the 1950s. It is inconceiv- cellular restoration of lost solute. The rapid rise in
able that it could have been missed by neurologists sodium level results in an osmotic endothelial injury
and pathologists of the nineteenth and early twenti- and in opening of the blood–brain barrier. The pro-
eth centuries with their strong reliance on post- posed mechanisms for opening the blood–brain bar-
mortem examination. In the 1950s more liberal use rier are endothelial cell shrinkage allowing fluid into
was made of intravenous tubing, and there was inter- the intercapillary and pericapillary spaces. Others
est in treating alcoholic withdrawal states by massive suggest that the barrier opening is in fact due to en-
administration of intravenous fluid. Thiazide diuret- hanced vesicular transport. In any case, the penetra-
ics were introduced for clinical use at this time. These tion of the blood–brain barrier is proven by the pres-
factors probably contributed to the genesis of hy- ence of biliary pigment in the myelinolytic lesion of
ponatremia. Alcohol has an antidiuretic-hormone- some highly jaundiced patients. It is hypothesized
686 Chapter 89 Central Pontine and Extrapontine Myelinolysis

that the endothelial injury leads to a release of ously ill patients are in general vulnerable to the dis-
myelinotoxic factors from the damaged cells, in turn ease. Again, this fact may simply indicate that very
leading to demyelination. Alternatively, and not nec- sick patients face an increased risk of hyponatremia
essarily mutually exclusively, the osmotic opening of or a greater likelihood of undergoing vigorous cor-
the blood–brain barrier results in vasogenic edema. rection of hyponatremia. The other possibility is that
Indeed, edema has been observed in CPM lesions. It is the brains of debilitated patients are less resistant to a
known that edema may itself be myelinotoxic. If the rapid rise in serum sodium. Here, too, thiamine defi-
presumption of this mechanism is correct, it may ex- ciency may play a role.
plain the localization of lesions, which preferentially
develop at sites characterized by an extensive admix-
ture of gray and white matter. Of all regions in the 89.4 Therapy
brain, the pons has the greatest degree of gray–white
apposition. Areas such as the basal ganglia, thalamus, No therapy is available for CPM/EPM. As soon as the
geniculate bodies, and cortex–white matter junctions neurological symptomatology has developed, only
also have extensive apposition of gray and white mat- symptomatic measures can be taken. Prevention must
ter. Edema or related myelinotoxic factors are princi- therefore be the focus of clinical efforts. The preven-
pally derived from the highly vascular gray matter tion of CPM/EPM relates to the medical approach to
and are thus able to affect the adjacent heavily myeli- hyponatremia, especially chronic hyponatremia. As
nated white matter. However, the mechanism where- there are different opinions about the role of hypona-
by edema may cause demyelination remains to be de- tremia and the rise of serum sodium level in the gen-
termined.An alternative hypothesis states that edema esis of CPM/EPM, there are likewise many guidelines
of the pons would result in strangulation of tracts and for the correction of hyponatremia. In severe hypona-
nuclear masses. In the base of the pons, longitudinal tremia with a serum sodium concentration below
and transverse fibers interlace. Because of this grid- 120 mmol/l, correction is necessary. The recommend-
like anatomy, the pons is particularly susceptible to ed rate of correction varies between 0.5 to 2 mmol/l
damage when edema occurs, as the edema liter- per hour, or 12 to 48 mmol/l per day. The fact that hy-
ally strangles the myelin sheaths and small blood ves- ponatremia itself may lead to brain damage forms an
sels. If mild, this would result in a reversible physio- argument against very slow correction.After reaching
logical block of neurotransmission. If edema persist- a concentration of 120–130 mmol/l, water restriction
ed and became more severe, irreversible demyelina- in combination with a diet with normal salt content is
tion would occur. recommended. In chronic asymptomatic hypona-
Although the summarized views seem reasonable, tremia, simple water restriction and a diet with nor-
there are some objections. This grid pattern is not mal salt content may suffice from the beginning. Oth-
present in extrapontine localizations of myelinolysis. ers prefer a combination of normal saline with fluid
There are regions in the brain in which myelinated restriction. Of course, diuretics, which cause salt loss,
fibers are interspersed with layers of gray matter that are discontinued. It is advisable to avoid hyperna-
do not show myelinolysis. And, finally, the factor of tremia. In the light of recent discussion, one should
edema, invoked in several theories of myelinolysis, is also be aware of the influence of hypoxic–ischemic
little documented. conditions and avoid or correct those as soon as pos-
Another variable to be considered is the role of sible.
the underlying condition. The over-representation of In addition to the rate of sodium correction, the
alcoholics in the myelinolysis population requires total osmotic load from contrast material in neu-
explanation. It may simply be related to the fact that roimaging must be considered. Intravenous contrast
alcoholics admitted to hospital are in withdrawal and material should not be used in the immediate post-
at the same time vigorously treated with intravenous correctional period of hyponatremia.
fluids.Another possibility is that the alcoholic state or
the withdrawal of alcohol somehow makes the brain
more sensitive to the rapid rise of serum sodium from 89.5 Magnetic Resonance Imaging
hyponatremia. This is conceivable, since alcohol re-
portedly affects brain hydration. Thiamine deficiency CT scanning may show fairly characteristic abnor-
may be another factor. Thiamine deficiency may malities in CPM, consisting of a roundish or “w-” or
cause depletion of ATP, which may affect the trans- “batwing-shaped” hypodense area in the midline of
port of electrolytes in the cell membranes, producing the pons, approximately equidistant from the floor of
endothelial damage and making cells more vulnera- the fourth ventricle and the ventral surface of the
ble to osmotic change. The fact that CPM/EPM has pons, with, in the acute phase, peripheral enhance-
been observed in the setting of a variety of medical ment after contrast injection. The fourth ventricle
disorders other than alcoholism suggests that seri- shows no displacement, and the brain stem is of nor-
89.5 Magnetic Resonance Imaging 687

Fig. 89.1. A 56-year-old man with an


episode of general malaise, vomiting,
and desiccation. Hyponatremia of
111.5 mmol/l was corrected in 2 days
to 134 mmol/l, followed by severe
neurological symptoms. Midsagittal
T1 and transverse T2-weighted MR
images demonstrate the characteristic
features of CPM in the subacute
phase, typically sparing the outer
rim of the pons

Fig. 89.2. A 44-year-old man with


chronic alcohol abuse and hypona-
tremia.The MRI was obtained in the
acute phase of CPM.The characteristic
w-shaped lesion central in the pons
in the acute phase is especially well
seen on the transverse T2-weighted
image. Courtesy of Dr. B.G. Ziedses
des Plantes Jr., Deventer Hospital,
Deventer, The Netherlands

mal size. The CT scan can also detect abnormalities trolateral longitudinal fibers are spared. In severe cas-
consistent with EPM. Sometimes the CT scan is found es, necrosis and cavitation may develop. The lesions
to be normal in CPM/EPM. In some patients the hy- may spread into the pontine tegmentum and mesen-
podensities of the CT scan disappear as the patient cephalon. Most reports reject a correlation between
improves, but persistence of hypodensities has also the severity of the MRI findings and the clinical con-
been observed despite clinical cure. CT findings ap- dition. On the other hand, in the follow-up of patients
pear to correlate rather poorly with the clinical syn- a good correlation between clinical improvement and
drome. Furthermore, the specificity of CT findings in the fading of lesions on MRI is observed. Even with
CPM is not high, as differentiation between CPM and complete clinical restoration, however, abnormalities
other pontine lesions, especially pontine infarction, is on MRI remain visible for a long time. Ring enhance-
often difficult or impossible. It is not an infrequent ment of the lesions in the pons after injection of intra-
occurrence that posterior fossa artifacts, characteris- venous contrast has been observed in the acute phase.
tic on CT, prevent optimal assessment of the pontine Although in MRI the quantities of injected contrast
area. material are far less than in CT, administration of
MRI is capable of showing the characteristics of contrast should be avoided, if possible, in order not to
CPM and EPM in more detail. MRI is normal or may aggravate the patient’s ionic imbalance. Moreover,
even show low signal intensity in the pons on T2- enhancement of the lesions in the pons does not con-
weighted images at onset of clinical symptoms. In tribute to the diagnosis. Diffusion-weighted imaging
classic cases of CPM, repeat MRI after a few days may add helpful information. In the acute phase Trace
shows symmetric high signal intensity on T2-weight- diffusion-weighted images show a high signal in the
ed images, representing myelinolysis of the base of central pontine lesion with corresponding low ADC
the pons, spreading centrifugally from the median values, in conformity with initial cellular edema. In a
raphe, characteristically leaving the outer rim of the few days ADC values pseudonormalize.
pons unaffected (Figs. 89.1 and 89.2). Hence, the ven-
688 Chapter 89 Central Pontine and Extrapontine Myelinolysis

Fig. 89.3. A 64-year-old man became critically ill after surgery tive edema in the right temporal lobe.Furthermore,there is ex-
for removal of a meningioma. Surgery and postoperative tensive myelinolysis in the cerebellar white matter, the middle
course were complicated by severe blood loss and hypona- cerebellar peduncles, the internal capsule, and the subcortical
tremia, both of which were corrected. After initial good recov- white matter, with preponderance on the right side. The pon-
ery, deterioration of the clinical condition occurred. The T2- tine white matter is slightly affected. Gradual return to normal
weighted coronal and transverse images show the postopera- occurred in the weeks that followed

The findings as described are highly characteristic Sometimes lesions in the basal ganglia are seen to-
of CPM. With the typical antecedents there is no great gether with the typical lesions in the pons. These cas-
difficulty in establishing a diagnosis. Differentiation es can be considered transitional between CPM and
in less clear cases is mainly from pontine glioma and CPM/EPM. In EPM, lesions are most often seen in the
pontine infarctions. putamen and caudate nucleus. In addition, lesions
89.5 Magnetic Resonance Imaging 689

may occur in the globus pallidus, thalamus, genicu- lishing the diagnosis EPM is not difficult in the pres-
late bodies, mammillary bodies, hippocampus, inter- ence of the characteristic pons lesion. The differential
nal and external capsule, corpus callosum, anterior diagnosis in EPM without a pontine lesion is more
commissure, and cerebellar and cerebral gyral convo- difficult and includes acute disseminated encephalo-
lutions (Fig. 89.3). Lesions may also be confined to the myelitis, encephalitis, and toxic-metabolic ence-
middle cerebellar peduncles (Fig. 89.3). EPM may phalopathies.
occur in isolation or be combined with CPM. Estab-
Chapter 90

Hypernatremia

90.1 Clinical Features Laboratory findings include a serum sodium level


and Laboratory Findings higher than 150 mEq/l by definition. Sodium excre-
tion is usually found at maximum physiological rates
It has been known for a long time that hypernatrem- of 270–290 mEq/l in urine, but is sometimes in excess
ia is a highly toxic condition which, if not adequately of that, as high as 400 mEq/l, the latter at the expense
and immediately treated, may end with severe neuro- of fluid homeostasis and thus leading to volume de-
logical deficits, coma, and death. In most patients hy- pletion.
pernatremia is a result in most patients of dehydra-
tion due to either insufficient intake or excessive loss
of water, as in diarrhea, vomiting, adrenal dysfunc- 90.2 Pathology
tion, or hypothalamic dysfunction caused by head
trauma, brain surgery, pituitary tumors, cranio- In the fatal incidents in neonates and infants patho-
pharyngiomas, other brain tumors, abscesses, or he- logical findings are uniform. Most of the abnormali-
morrhages. Hypernatremia may also follow the dis- ties are found in the lungs and brain. In the lungs he-
continuation of intake of lithium after long-lasting morrhages are found, sometimes very extensive. In
treatment. This results in a defect in concentrating the brain, subdural and focal subarachnoid hemor-
urine, leading to hypernatremia. rhages are seen. On sectioning, petechial and larger,
Another, less frequent, cause of hypernatremia is a more confluent hemorrhages are seen, in particular
too high intake of salt, either accidentally or nonacci- in the basal ganglia and the white matter. The cerebral
dentally, in the latter case often as form of abuse or and cerebellar cortex is relatively spared. In the basal
punishment. Attempts at abortion by intra-amniotic ganglia discolored spots are seen. The substance of
injection of hypertonic salt solution may lead to seri- the white matter in the centrum semiovale is macro-
ous hypernatremia. Accidental disasters of intake of scopically changed, with areas of gray-brownish dis-
high doses of salt have been reported, when in the coloration.
nursery sugar is inadvertently replaced by salt in Microscopic examination confirms the multiple
preparing one of the formulas for neonates or when a petechial hemorrhages, together with thrombosis in
10% salt solution is used for infusion in neonates in- many small vessels. Around these thrombotic vessels
stead of a 10% glucose solution. Use of the wrong flu- ring-like hemorrhages are present, with perivascular
id for emetics, gastric lavage, and other medical inter- edema. The discoloration seen in the white matter
ventions has been reported as an iatrogenic cause of appears to be related to the thrombotic vessels and
hypernatremia in other cases. A separate chapter in focal hemorrhages. Hemosiderin is present in these
salt poisoning is the nonaccidental, forced intake of lesions, making it possible to date these hemorrhages
too large quantities of salt as form of punishment, pu- to the onset of the clinical symptoms. In some pa-
rification for religious motives, or child abuse. In tients thrombosis of venous sinuses is seen. The over-
many cases in children nonaccidental salt poisoning all picture is that of a hemorrhagic encephalopathy,
is part of the “Munchausen by proxy” syndrome. with thrombosis, derangement of microcirculation,
The initial clinical presentation of hypernatremia and necrosis.
may vary from drowsiness, inactivity, and irritability
to altered consciousness, confusion, seizures, and ev-
idence of dehydration, including sunken eyes, dry 90.3 Pathogenetic Considerations
mouth, dry mucosa, doughy skin turgor, raised blood
pressure, and irregular pulse. Coma may ensue. Fur- The fluid balance of the body is maintained by multi-
ther deterioration may occur after the initial diagno- ple hormonal and neurogenic mechanisms. These
sis and installation of treatment, and severe neurolog- keep the total quantity of fluid, the concentration of
ical deficits may develop and remain even after suc- solutes, and thus the osmolality of body fluids within
cessful regulation of the osmotic balance. The death narrow limits. Disruption of these homeostatic mech-
rate among patients with serious hypernatremia is anisms may lead to either overhydration with hypo-
high. osmolality, or dehydration with hyperosmolality. Ex-
90.5 Magnetic Resonance Imaging 691

cessive intake of fluid on the one hand, or excessive normalization of serum sodium levels should be
intake of salt on the other, may cause the regulatory achieved in 48–72 h. Dialysis has also been advocated.
capacity of the homeostatic system to be exceeded. The damage done to brain, lungs, and kidney devel-
Damage of the regulatory systems in the hypothala- ops rapidly, and hypernatremia, especially in chil-
mus, by trauma, brain surgery, tumor, or other ex- dren, is fatal in over 50% of cases. Treatment should
panding lesions, may also lead to deregulation of the therefore start as soon as possible. Special attention
osmotic sensors or deficiency of antidiuretic hor- is, of course, also necessary for renal, pulmonary, and
mone. cardiac function. Even when the patient survives,
The effects of hypernatremia on the brain appear severe neurological deficit may remain.
with some delay. It takes time before the blood–brain
barrier is disrupted. In experiments, animals with hy-
pernatremia show similar clinical signs as humans: 90.5 Magnetic Resonance Imaging
lethargy, irritability, tremor, ataxia, seizures, and
eventually coma and death. On autopsy shrinkage of Very few reports are available on neuroimaging find-
the brain is obvious with hemorrhagic fluid present ings in patients with hypernatremia. On CT, extensive
around the brain. In some instances there are massive cerebral and cerebellar hemorrhages have been de-
subdural hematomas. These are probably caused by scribed. CT may also show bilateral transient thalam-
tearing of veins which bridge the space between the ic hypodensity. One report describes the MR findings
brain and the calvarium. Smaller hemorrhages at the in a neonate with hypernatremic dehydration. This
brain surface and in the parenchyma are the result of was a 15-day-old girl who received insufficient quan-
direct damage to the endothelium of the smaller ves- tities of breast milk for several days. MR showed a
sels and capillaries. Increased osmolality of the blood superior sagittal sinus thrombosis with a venous he-
causes water to move from the brain toward the morrhagic infarction, and some smaller hemorrhages
blood. The brain reacts to the osmotic insult with the with surrounding edema.
generation of intracellular osmolytes, so-called idio- We have seen a 2.5-month-old child, a victim of
genic osmolytes. These osmolytes serve to regulate child abuse, who was given a small amount of sodium.
volume and can reach high concentrations within On admission the blood sodium level was 200 mEq/l.
cells. The intracellular osmolality is thereby increased The first MRI showed extensive abnormalities con-
and the loss of cellular fluid counteracted. Several fined to the nonmyelinated frontotemporal areas,
substances have been identified as putative organic whereas the myelinated areas seemed to be preserved
osmolytes, among them myo-inositol, N-acetylaspar- (Fig. 90.1). The lesions showed irregular enhance-
tate, glycerophosphoryl choline, and taurine. How- ment, indicating a defective blood–brain barrier
ever, rapid correction and normalization of blood (Fig. 90.1). After the initial phase of swelling, shrink-
osmolality may subsequently lead to relative hyperos- age of the affected parts followed with evidence of he-
molality of the brain, resulting in shift of fluid from morrhagic necrosis (Fig. 90.2). Finally, serious atro-
the intravascular compartment to the brain par- phy of the frontal and temporal lobes and gliosis de-
enchyma, brain edema, and possibly pontine and ex- veloped (Fig. 90.3). The child remained severely neu-
trapontine myelinolysis. rologically damaged.
When MRI is required in patients suspected of
having hypernatremia, the protocol should include
90.4 Therapy gradient echo images to identify hemorrhage, patho-
logically a constant finding. Proton MRS could be of
Therapy consists mainly of restoring the osmotic bal- interest because it will be able to monitor osmolytes
ance and bringing the sodium levels back to normal such as myo-inositol. The presence of hemorrhages,
values. Too rapid correction of the sodium levels may however, would probably disturb magnetic field ho-
be followed by pontine and extrapontine myelinoly- mogeneity.
sis, a complication more often seen in too rapid When nonaccidental salt poisoning is suspected,
correction of hyponatremic states. For this reason one should be alert for other possible signs of child
it has been advised that the decrease in plasma sodi- abuse.
um should not exceed 0.5 mEq/l per hour and that
692 Chapter 90 Hypernatremia

Fig. 90.1.
90.5 Magnetic Resonance Imaging 693

Fig. 90.2. After 1.5 weeks atrophy of the affected areas evolves.The T1-weighted axial (second row) and sagittal images (third row)
demonstrate the hemorrhagic component of the necrosis

Fig. 90.1. A 2.5-month-old girl was the victim of nonacciden-


tal salt intoxication. The T2-weighted images (first and second
row) show involvement of the, at this stage of development,
unmyelinated parts of the brain: the frontal and temporal
lobes. The involved parts are partly swollen. The myelinated
internal capsule, brain stem, and cerebellar white matter are
intact. The T1-weighted images (third row) show that myelina-
tion is normal and that the unmyelinated cerebral white matter
has a low signal intensity. After contrast injection (fourth row),
there are some areas of contrast enhancement as evidence of
disruption of the blood–brain barrier
694 Chapter 90 Hypernatremia

Fig. 90.3. Another month later, severe atrophy of frontal and temporal lobes and ventriculomegaly are seen.Bilateral frontal sub-
dural effusions are present
Chapter 91

Marchiafava–Bignami Syndrome

91.1 Clinical Features 91.2 Pathology


and Laboratory Investigations
The principal pathological change is necrosis of the
Marchiafava–Bignami syndrome (MBS) is a disorder corpus callosum. The necrosis may involve the entire
characterized by primary degeneration of the corpus length of the corpus callosum or only a part of it. The
callosum in chronic alcoholics. Although a high pro- middle layers of the splenium and genu are mainly af-
portion of reported cases have been Italians, drinking fected, an upper and lower rim of the callosal fibers
Italian wine, the condition has also been described in usually being preserved. The lesion extends laterally
many non-Italians drinking non-Italian wine. Most to the edges of the corpus callosum. When necrosis is
affected cases were middle-aged men whose daily incomplete, demyelination is found, with relative
wine consumption had been 2 l or more for many preservation of axons and a moderate glial reaction.
years. In cases of total necrosis, both myelin and axons have
The disease has three major clinical forms: acute, disappeared and are replaced by an accumulation of
subacute, and chronic. The acute form is character- macrophages and perivascular cells together with a
ized by sudden onset with severe disturbances of con- variable glial reaction. The lesion is sharply delineat-
sciousness, sometimes heralded by convulsions. The ed. In acute cases the lesion appears in the form of a
initial phase is followed by the development of persis- band of edematous necrosis with fresh coagulation.
tent coma or stupor with pyramidal signs and hyper- An upper and a lower band of normal callosal tissue
tonia. Patients are generally mute, but if they say a few remains. In cases of long duration, the corpus callo-
words, they appear to be severely dysarthric. They sum is thinned and atrophic with a slit or band of de-
may die within a few days. The subacute condition is myelination in the middle, most conspicuous in genu
characterized by rapidly progressive dementia, some- and splenium. Macrophages accumulate in perivascu-
times following an initial acute state with temporary lar areas.
coma or convulsions. The dementia has characteris- Other regions of white matter are sometimes in-
tics of a split-brain syndrome. The patients are usual- volved, including the centrum semiovale in the
ly dysarthric. Hypertonia of the limbs is quite com- frontal, parietal, and occipital region as an extension
mon and marked by strong opposition to any move- of the lesion of the corpus callosum. Sometimes the
ment, either flexion or extension. There is spastic flex- lesion includes all the white matter up to the U fibers.
ion of the arms and extension of the legs with Furthermore, the cerebellar peduncles and the anteri-
hyperreflexia and extensor plantar reflexes. A facial or commissure may be affected. The fornix is thought
grimace and trismus may be present, as well as never to be affected. Also, these lesions are primarily
opisthotonus. In the end stage the patients are unable characterized by demyelination with accumulation of
to walk or stand. Severe dementia usually progresses macrophages. In complete necrosis axons are also
to a vegetative state and death within a few months. In lost. The ventricles are moderately enlarged, and in
the chronic form, which is much less common, de- most cases there is some cortical atrophy. There is oc-
mentia progresses slowly over a number of years. casionally cortical laminar necrosis and, exceptional-
Neurological examination reveals diffuse rigidity, ly, necrosis of the basal ganglia.
dysarthria, and inability to stand or walk, as in the
subacute form. This condition progresses steadily to-
wards death several years later. It should be noted, 91.3 Pathogenetic Considerations
however, that in a few cases improvement of the pa-
tient’s condition and disappearance of image abnor- The pathogenesis of MBS is still unknown. A toxic
malities have been reported. factor present in cheap red wine seems to be the clos-
Laboratory investigations may reveal vitamin defi- est constant association, but the factor responsible
ciencies due to malnutrition but they are probably not has never been identified. It has been shown that vit-
directly related to MBS. Combinations of Wernicke amin deficiencies are not important in the pathogen-
encephalopathy with MBS have been described. There esis, as a supply of vitamins does not prevent MBS in
is no laboratory test for MBS. animals. There are, however, a few descriptions of the
696 Chapter 91 Marchiafava–Bignami Syndrome

Fig. 91.1. A 53-year-old man with MBS with acute onset. MRI of the middle layer of the corpus callosum,in particular in sple-
on the ninth day (left image) shows contrast-enhancing lesions nium and genu, but no contrast enhancement. The splenium
in the genu and the splenium of the corpus callosum. At 3 and genu show cystic degeneration. From Caparros-Lefebvre
weeks the images (middle and right images) show involvement et al. (1994), with permission

combination of MBS with Wernicke encephalopathy. has been described in the early stages of the disease.
Lesions of the white matter affecting the corpus callo- In the later stages there is atrophy of the corpus
sum and anterior commissure have been described callosum, progressive and marked widening of
following chronic cyanide intoxication and chronic the cerebral sulci, and dilatation of the lateral ventri-
methyl alcohol intoxication. However, the patho- cles.
genetic similarities on the molecular level between The corpus callosum lesions are much more easily
MBS and these intoxications are unclear. Why the visualized by MRI (Figs. 91.1 and 91.2). The sagittal
central layers of genu and splenium of the corpus cal- MR images are diagnostic: they show the typical pat-
losum are preferentially affected in MBS remains ob- tern of the corpus callosum splitting into three layers,
scure. The cause of the additional gray matter lesions, with relative sparing of the upper and lower layers. In
usually in the form of laminar necrosis of the cerebral the initial stages of the disease the corpus callosum
cortex, has not been explained satisfactorily. The appears swollen and contrast enhancement may be
most attractive hypothesis would seem to be that the present, in particular in the genu and splenium. Cal-
cortical necrosis is secondary to the callosal lesion, losal hemorrhage may occur in the subacute stage.
which leads to loss of neurons due to secondary de- The middle layer may become cystic in the later
generation. stages of the disease and the corpus callosum be-
comes atrophic. Cerebral hemispheric white matter
may be involved, with a less characteristic appear-
91.4 Therapy

There is no effective treatment for MBS; only sympto-


matic treatment is possible. However, it is important
not to overlook other complications of long-standing
alcoholism, particularly vitamin deficiencies and dis- 䊳

turbances of electrolytes. Fig. 91.2. A 56-year-old man with MBS. The first and second
rows of images were obtained in the acute phase. The sagittal
T2-weighted images (first row) show that especially the middle
91.5 Magnetic Resonance Imaging layer of the corpus callosum is involved.The axial FLAIR images
(second row) show the predominant involvement of the
CT scan may show abnormality of the corpus callo- corpus callosum with some additional lesions in the deep
sum with some swelling and edema in the acute form. periventricular white matter. A follow-up MRI was obtained
The corpus callosum lesions in the subacute or chron- 50 days later (third and fourth rows). The sagittal T1-weighted
ic forms of the disease, which are not associated with images (third row) show cavitation of the middle layer of the
swelling, may be more difficult to detect on CT. Ex- genu and splenium of the corpus callosum. The hyperintense
tensive hypodense areas may be found in the cerebral lesions on the axial T2-weighted images (fourth row) are less
hemispheric white matter. Contrast enhancement conspicuous. From Yamamoto et al. (2000), with permission
91.5 Magnetic Resonance Imaging 697

Fig. 91.2.
698 Chapter 91 Marchiafava–Bignami Syndrome

ance. Apart from the corpus callosum, other inter- There are many diseases that produce lesions
hemispheric commissural fibers may be involved, in the corpus callosum, the most prominent being
such as the anterior commissure. Cortical laminar multiple sclerosis. Others are acute disseminated
necrosis and basal nuclei involvement, when present, encephalomyelitis, progressive multifocal leukoen-
can be visualized by MRI. Involvement especially of cephalopathy, adrenoleukodystrophy, and mitochon-
the putamen, in some cases even reversible, has been drial disorders. These disorders rarely produce the
described. split corpus callosum pattern seen in MBS. Cyanide
Because in nearly all patients excessive alcohol intoxication is said to present occasionally with split-
abuse plays a dominant role, the MRI study should ting of the corpus callosum. The context is then obvi-
also look for signs of other alcohol and thiamine ously different. In most patients with MBS, the mid-
deficiency-related abnormalities, such as changes in sagittal MR image, showing the splitting of the spleni-
mammillary bodies, walls of the third ventricle and um and genu of the corpus callosum into three layers,
basal ganglia, tectum and tegmentum of the midbrain together with the medical history and clinical find-
and pons, dentate nucleus and cerebellar cortex. ings of MBS, lead to the correct diagnosis.
Chapter 92

Posterior Reversible Encephalopathy Syndrome

92.1 Clinical Features 92.3 Pathogenetic Considerations


and Laboratory Investigations
There are many theories trying to explain the nature
Hypertensive encephalopathy may lead to reversible and location of the lesions seen in PRES. The under-
clinical symptoms including headache, nausea, vom- lying problems are complex, and it is likely that mul-
iting, altered mental functioning, cerebral blindness, tiple factors play a role in the pathogenesis. This point
and epileptic seizures. can be illustrated by a discussion of a few different
The syndrome can be present in a number of con- underlying disorders.
ditions characterized by hypertension: eclampsia, Transient symptoms of hypertensive encephalopa-
renal insufficiency, hemolytic–uremic syndrome, thy were recognized clinically even in the first half of
thrombotic thrombocytopenic purpura, and acute in- the twentieth century in pre-eclampsia and eclamp-
termittent porphyria. In most cases the symptoms sia. The clinical picture of pre-eclampsia consists of
and MR abnormalities will disappear with antihyper- hypertension, confusion, proteinuria, and edema oc-
tensive treatment. With or without hypertension, curring in the second half of pregnancy. Symptoms
the clinical and MR pattern may also be seen in pa- vary in severity and multiorgan involvement is com-
tients undergoing anticancer therapy, bone marrow, mon. Neurological symptoms include headache, con-
or organ transplantation, using either anticancer fusion, hyperreflexia, visual hallucinations, and cere-
medication or immunosuppressive therapy (see also bral blindness. Eclampsia is characterized by the oc-
Chap. 88). currence of an epileptic seizure or coma in a patient
Because of the first reported findings on MRI and with a pre-eclampsia syndrome. This syndrome and
the transient nature of the clinical symptoms, the the neurological implications have been studied ex-
name reversible posterior leukoencephalopathy syn- tensively. Many factors have been identified that pre-
drome (RPLS) was suggested. When it became clear dispose to eclampsia, including an immunological
that often gray matter was also involved, the name maternal reaction (probably triggered by placental
was changed into posterior reversible encephalopa- debris), dietary factors (such as deficiency of magne-
thy syndrome (PRES). This name has stuck so far, but sium, calcium, and zinc, and excess of sodium) and
does not cover the entire spectrum of manifestations genetic factors (such as abnormal alleles for the genes
of transient clinical symptoms and MRI abnormali- of tumor necrosis factor-a, angiotensinogen, factor V,
ties seen in relation to hypertensive and drug-related and nitric oxide synthase). The maternal response
encephalopathies. leads to widespread cellular dysfunction with in-
creased capillary permeability, raised fibronectin lev-
els, and damaged endothelium. Subsequently, coagu-
92.2 Pathology lation abnormalities develop with increased activa-
tion and consumption of platelets, low antithrombin
Pathological descriptions are rare, as expected in a III levels, abnormal prostaglandin levels, and in some
usually reversible disorder. Occasionally the results of cases diffuse intravascular coagulation. Endocrine
a brain biopsy are mentioned, usually with vasogenic abnormalities include activation of the renin–an-
edema as the only finding. The best data have been giotensin–aldosterone axis, abnormal catecholamine
obtained in eclampsia, because in the first half of the levels, and abnormalities of progesterone metabo-
twentieth century the condition could be lethal. The lism. This constellation leads to hypertension, edema,
descriptions give a varied picture of cerebral pathol- proteinuria, bleeding tendencies, renal dysfunction,
ogy, with white matter edema, most pronounced in liver damage, and neurological abnormalities. The
the watershed areas, sometimes with pronounced hypertension exceeds the regulatory capacity of the
mass effect, potentially resulting in tentorial hernia- vessel walls (muscular regulation), so that the local
tion. In addition, there may be microinfarctions, cor- autoregulation (regulating size of the vessels and the
tical petechiae, and pericapillary hemorrhages. Larg- admitted blood flow) depends solely on the sympa-
er hemorrhages may occur in subcortical regions, thetic innervation of the vessels, which is relatively
deep white matter, and basal ganglia. poor in the vertebrobasilar system. Hypertension and
700 Chapter 92 Posterior Reversible Encephalopathy Syndrome

immune-mediated endothelial dysfunction lead to porphyria present with subacute episodes of abdom-
extravasation of red cells and plasma proteins, result- inal pain, vomiting, nausea, bowel distension or ileus,
ing in cerebral edema. Vasospasm, prostaglandin de- and neurovisceral and circulatory disturbances. The
ficiency, defects in the e-NOS gene coding for nitric neurological problems are extremely variable and in-
oxide synthase, and endothelial damage play a role clude autonomic neuropathy, peripheral axonal neu-
in the eventual progression of the lesion to ischemia ropathy, and CNS dysfunction. In subacute attacks,
and infarction. Thus, in eclampsia a varied picture central and peripheral neurological symptoms may
of cerebral pathology, with cerebral edema, micro- become prominent. Patients may have seizures, visual
infarctions, cortical petechiae, and pericapillary hallucinations, and cerebral blindness. Bilateral le-
hemorrhages, results in the clinical manifestations sions in the parieto-occipital and posterior frontal
of headache, confusion, visual disturbances, and lobes have been found in these patients. The lesions
seizures. disappear after treatment. The similarity of the clini-
Reversible leukoencephalopathies predominantly cal and imaging pattern to other cases of PRES is evi-
in the posterior cerebral areas also have been de- dent. Some of the patients have hypertension during
scribed in the hemolytic–uremic syndrome (HUS) these subacute episodes, but not all. It is likely that
and thrombotic thrombocytopenic purpura (TTP). other factors play a role in the development of CNS
In these disorders PRES is only one of many possible dysfunction
manifestations. Both disorders are microangiopathic Hypertension has not been found in all patients
hemolytic anemias characterized by hemolytic ane- with PRES. Nocturnal registration of blood pressure
mia, thrombocytopenia, CNS dysfunction, fever, in patients with PRES without hypertension has
and/or renal dysfunction. HUS is usually seen in in- shown transient rises in blood pressure, a “riser” pat-
fants and children, whereas TTP more often presents tern, in some patients. It remains to be proven
in adults. Clinical findings include acute renal insuffi- whether that is the case in a significant number of pa-
ciency and encephalopathy, often with seizures, hem- tients with PRES.
orrhagic colitis, and upper respiratory infection. MRI In eclampsia, HUS, and TTP, as well as in acute in-
and CT abnormalities develop over days, showing termittent porphyria and other disorders in which
patchy white matter areas in the frontal, parietal, and PRES develops, hypertension is only one of the
occipital gray and white matter. In patients who died, factors that play a role in the development of the
occlusive platelet thrombi were found, primarily con- encephalopathy. This explains why not all patients
fined to the cortical gray matter. There was mild en- develop the syndrome under the same conditions.
dothelial proliferation in the larger vessels. Capillar- Autoregulation of blood flow (velocity) and flux (vol-
ies showed proliferation, or “wickerwork” formation. ume) is dependent on local vascular wall (muscular)
Many causes have been described for HUS and TTP. response and neuronal, sympathetic regulation. The
In children hemorrhagic colitis is the most frequent predominant involvement of the parieto-occipital
cause; in adults, immune-mediated diseases, for ex- lobes, but also the involvement of brain stem and
ample, systemic lupus erythematosus. HUS and TTP cerebellum, may be explained by the relative poverty
are classified as “uremic encephalopathies.” The of the sympathetic innervation of the vertebrobasilar
imaging findings may be similar to those described in vascular system. Other factors mentioned as con-
hypertensive encephalopathies of other origins. Most tributing to PRES are direct toxic influences on the
probably the encephalopathy is the result of more endothelium, vasoconstriction due to the liberation
than one factor. In patients with severe hypertension of endothelin, and the formation of microthrombi.
it is assumed that acute increase of blood pressure ex- What is not explained in any of these theories is the
ceeds the limits of local autoregulation. Distal vessels location of lesions in border zones, both parieto-
and capillaries are exposed to too high a pressure and occipital and frontal, suggesting temporary hypoper-
to hyperperfusion. This causes damage to the en- fusion as responsible factor.
dothelium so that blood plasma and, in some cases,
erythrocytes can pass the blood–brain barrier.
Reversible encephalopathic clinical symptoms and 92.4 Therapy
lesions on MRI resembling those of PRES may also be
seen in acute intermittent porphyria. Acute intermit- When hypertension is present, treatment of the hy-
tent porphyria is a disorder with an autosomal domi- pertension and any underlying disease may lead to
nant mode of inheritance, caused by a deficiency of disappearance of clinical symptoms and MR abnor-
porphobilinogen deaminase. More than 80% of carri- malities, usually within 14 days. This, of course, also
ers never develop symptoms. Symptoms are often depends on the underlying cause. In patients with
precipitated by the influence of drugs, hormones, and eclampsia, delivery of the child when possible and,
nutritional factors that change the rate of heme syn- where necessary, treatment of hematological abnor-
thesis in the liver. Patients with acute intermittent malities, renal disorders, and hypertension should be
92.5 Magnetic Resonance Imaging 701

instituted. The positive influence of magnesium sul- nolysis. Isolated thalamic involvement also occurs in
fate on the eclamptic condition has been recognized deep venous thrombosis. With a protracted state of
for many years, as has the role of pyridoxine in the the lesions low-grade gliomas have to be ruled out.
past few decades. In patients in whom PRES is caused When only the midbrain, pons, or cerebellum is af-
by immunosuppressive and/or chemotherapeutic fected, a correct diagnosis may be extremely difficult
drugs, the drug regimen has to be abandoned or and depends on careful interpretation of all MR data
changed. Once the treatment is stopped, further dete- in relationship to all the available clinical data.
rioration may occur for 1 or 2 weeks before improve- Most important are the addition of diffusion-
ment sets in. Complete disappearance of clinical and weighted imaging and the use of ADC maps (Fig.
imaging symptoms and signs may occur, but residual 92.6). These techniques allow differentiation of areas
damage is not rare. with low diffusivity, which may progress to infarction,
and areas with high diffusivity, probably representing
vasogenic edema, which may revert to normal. Unfor-
92.5 Magnetic Resonance Imaging tunately, results reported in the literature are contra-
dictory. Some authors report decreased diffusivity;
In many cases of PRES and its variants CT shows hy- others show increased ADC values, or “pseudo”-nor-
podensity in the affected areas, predominantly the malized ADC values, the latter, according to the
parieto-occipital regions. In some cases hemorrhages authors, the result of intravoxel presence of both va-
are seen, related to extravasation of erythrocytes. sogenic and cytotoxic edema. However, most evi-
MRI is more sensitive than CT and allows better dence so far suggests that lesions with a high ADC,
delineation of the abnormalities, such as an invento- probably representing vasogenic edema, are re-
ry of all involved areas, their location in border zones, versible, whereas lesions with a low ADC, represent-
and the involvement of gray as well as white matter. ing cytotoxic edema, are irreversible.
Standard MR techniques, such as T1-weighted, proton Perfusion studies with SPECT and MRI have also
density, T2-weighted, and FLAIR images demonstrate shown conflicting results. Both hypo- and hyperper-
the lesions. FLAIR usually depicts hemorrhages fairly fusion have been reported in the involved areas, as
well. Gradient refocused images should be added to has diffuse hypoperfusion over the entire brain. Be-
pick up microhemorrhages. Contrast enhancement cause in many cases border zones are involved, one
may be seen in some of the lesions. might expect hypoperfusion to occur at some point
The lesions are predominantly located in the pari- during the development of the disorder. Perhaps the
eto-occipital region. In that location they tend to- differences in results of perfusion studies may be ex-
wards symmetry, but asymmetrical presentations are plained by differences in the time elapsed between
not rare. The splenium of the corpus callosum may be onset of the disorder and the time of the MR exami-
involved. Usually the deep white matter is involved, nation. The severity and rate of development of the
but the overlying cortex is usually affected as well. In process and underlying factors may be also of impor-
other cases the lesions are located in the parieto-oc- tance.
cipital and frontal border zones (Fig. 92.1), suggesting MRS has revealed diffuse metabolic abnormalities
that an episode of hypoperfusion has occurred. One in PRES, even in areas that have a normal signal on
must be aware of other presentations: PRES may af- imaging. Reduced ratios of N-acetylaspartate over
fect solely the thalamic nuclei, the frontal border choline and N-acetylaspartate over creatine, with
zones, the midbrain, the pons, or the cerebellum high choline concentrations in the normal-appearing
(Figs. 92.2 and 92.3). In the initial period some mass white matter have been reported. Quantification of
effect may be present, with secondary effects such as the metabolites confirmed these findings: low con-
hydrocephalus. In some patients the lesions are ex- centration of N-acetylaspartate and high concentra-
tensive (Fig. 92.1); in others they are much more lim- tion of choline. After treatment MRS findings return
ited (Fig. 92.4). With treatment MRI abnormalities to normal. Unfortunately there are also MRS studies
disappear completely or nearly completely in most reporting either no biochemical abnormalities in the
patients (Figs. 92.2 and 92.5). affected areas during the acute stages of the disease,
Differentiation from other disorders depends on or no improvement of the MR spectra despite disap-
the location. When the border zones alone are affect- pearance of clinical symptoms. Because most studies
ed, other disorders leading to hypoperfusion must al- comprise a single observation or data on a small
so be considered. Isolated involvement of the pons number of patients, it remains difficult to draw gener-
has to be differentiated from central pontine myeli- al conclusions.
702 Chapter 92 Posterior Reversible Encephalopathy Syndrome

Fig. 92.1. An 11-year-old girl with hypertensive encephalopa- gray and white matter. No abnormalities are seen in midbrain,
thy.T2-weighted images (first and second row) show the typical pons, or cerebellum. On FLAIR images (third row) the lesions
pattern of the posterior reversible encephalopathy syndrome are more conspicuous, but FLAIR may be less good in depict-
(PRES): nearly symmetrical lesions in the parieto-occipital ing lesions in the posterior fossa
lobes and the frontal border zones. The lesions involve both
92.5 Magnetic Resonance Imaging 703

Fig. 92.2. T2-weighted transverse images in a 58-year-old in the midbrain and pons. These latter lesions show some
man with hypertensive encephalopathy (first three rows).There swelling. A repeat MRI was obtained a few weeks later (fourth
are diffuse mild white matter abnormalities in the cerebral and fifth rows) and show that with treatment all lesions have
hemispheres, but the more important abnormalities are seen disappeared
704 Chapter 92 Posterior Reversible Encephalopathy Syndrome

Fig. 92.2. (continued).

Fig. 92.3. Two coronal T2-weighted


images of a 32-year-old pregnant
woman with eclampsia.The left image
shows extensive involvement of
the parieto-occipital lobe; the right
image shows involvement of the basal
ganglia, predominantly on the right
side, and of the mesial temporal struc-
tures. After delivery and therapy an
almost complete recovery followed
92.5 Magnetic Resonance Imaging 705

Fig. 92.4. In this 7-year-old girl with reflux nephropathy and hypertensive encephalopathy the T2-weighted images show only a
few lesions with an asymmetrical distribution
706 Chapter 92 Posterior Reversible Encephalopathy Syndrome

Fig. 92.5. A 63-year-old woman was admitted to the intensive the lesions on follow-up. The MRI findings are highly sugges-
care unit with septic shock, respiratory infection, and diarrhea. tive of PRES, although the causative factors in this patient are
After recovery she complained of memory problems. The up- not known. Courtesy of Dr. E.P.J. Arnoldus, Department of Neu-
per row of FLAIR images shows lesions in this phase. The lower rology, TweeSteden Hospital, Tilburg, The Netherlands
row of T2-weighted images shows complete disappearance of
92.5 Magnetic Resonance Imaging 707

Fig. 92.6. The first two rows contain the FLAIR images of a the right side, and some tiny hyperintense spots in the frontal
52-year-old man with hypertensive encephalopathy, showing border zones. The fifth and sixth rows show ADC maps with
the common MR pattern of PRES lesions. The third and fourth a mixture of higher and lower than average values in the
rows show Trace diffusion-weighted images (b = 1000), with involved areas
bright spots in the parieto-occipital area, more extensive on
708 Chapter 92 Posterior Reversible Encephalopathy Syndrome

Fig. 92.6. (continued)


Chapter 93

Langerhans Cell Histiocytosis

93.1 Clinical and Laboratory Findings loma. The symptoms and signs of pituitary–hypo-
thalamic involvement vary widely and may include
Histiocytosis-X was the name initially given to a changes in social behavior, appetite, changes of sleep
group of disorders with seemingly unrelated clinical pattern, polyuria and polydipsia, indicating posterior
features, but characterized by the same pathological pituitary dysfunction, and growth failure, precocious
finding: infiltration of the involved tissue by large or delayed puberty, amenorrhea, hypothyroidism, or
numbers of histiocytes, often organized as granulo- hypocortisolism, indicating anterior pituitary dys-
mas. The disease was later renamed Langerhans cell function.
histiocytosis (LCH), after the cell (X) was identified as Intra- and extra-axial granulomas cause symp-
the Langerhans cell. LCH includes eosinophilic gran- toms dependent on their location. The intra-axial le-
uloma, Letterer–Siwe disease and Hand–Schüller– sions can occur in many places, supra- and infraten-
Christian disease. The eosinophilic granuloma in- torial, and behave as space-occupying lesions. Extra-
volves the bone. Letterer–Siwe disease is a multiorgan axial lesions may exist as a continuous extension from
disease in children under the age of 2 years. Hand– bone lesions, or they may develop as lesions from the
Schüller–Christian disease includes the triad of ex- leptomeninges. They may exert pressure on the brain.
ophthalmos, diabetes insipidus, and bone lesions. LCH may also occur in the choroid plexus.
The second type of histiocytic disorders comprises Involvement of structures in the posterior fossa,
non-Langerhans histiocytosis, which includes Erd- in particular the cerebellum, are, after diabetes in-
heim–Chester disease, isohemophagocytic lympho- sipidus, the second most common manifestation of
histiocytosis, and histiocytosis with massive lymph- CNS involvement in LCH, and may occur many years
adenopathy. Erdheim–Chester disease is mentioned after the initial diagnosis of LCH. Lesions in the pos-
explicitly here, because it can mimic many of the terior fossa may consist of local granulomas, or of
features of brain involvement in LCH. symmetrical abnormalities in the white matter of the
The third type of histiocytic disorders comprises hilus of the dentate nucleus and corpus medullare of
malignant histiocytosis. the cerebellum. Related clinical features are ataxia, re-
LCH can now be defined as abnormal proliferation flex abnormalities, tremor, dysarthria, and dysphagia.
of clonal, dendritic Langerhans cells, which can be Combinations of the symptoms and progression of
found in lungs, bone, skin, liver, spleen, lymph nodes, the disease are not rare and may lead to severe CNS
thymus, bone marrow, brain, pituitary, and other en- deterioration.
docrine organs. In adults usually only two organs are Combinations of diabetes insipidus and cerebellar
involved, whereas in younger children LCH is most symptoms also occur in Erdheim–Chester disease.
commonly a multiorgan disease. Other manifestations of this disease can also be simi-
Primary involvement of the brain is rare in LCH. lar to LCH.
Patients who develop CNS disease are likely to have Diagnosis of LCH in nearly all cases demands a
multiorgan disease, with lesions in the calvaria and biopsy and histological confirmation of the patholog-
the temporal and orbital bones. Involvement of the ical Langerhans cell.
brain and related structures can be subdivided into
four different forms: (1) involvement of the pitu-
itary–hypothalamic structures; (2) intra- or extra-ax- 93.2 Pathology
ial granulomatous lesions; (3) abnormalities in the
posterior fossa, in particular the cerebellopontine Histologically, the granulomatous lesions consist of a
pathways; and (4) combinations of these three mani- proliferation of clonal Langerhans cells, combined
festations, with overlapping symptoms. with an inflammatory reaction. Four stages are usual-
Involvement of the pituitary–hypothalamic struc- ly distinguished: a hyperplastic–proliferative stage, a
tures leads to the most common symptom of cerebral granulomatous stage, a xanthomatous stage, and a
involvement, diabetes insipidus. Diabetes insipidus stage of fibrosis. Lesions in the first stage are most
can precede other CNS lesions by more than 3 years. likely to show the characteristics of LCH. Electron mi-
When diabetes insipidus occurs as isolated symptom, croscopy reveals Birbeck granules, characteristic of
it is often referred to as Ayala disease or Gagel granu- LCH. Immunostaining can be performed for the
710 Chapter 93 Langerhans Cell Histiocytosis

S-100 protein and for CD1a (OKT6) antigenic reactiv- neously with transformation to a fibroxanthomatous
ity, but these tests have a lower level of confidence. state. Such lesions lose the characteristics of LCH. The
Histochemistry demonstrates that LCH and adjuvant involvement of other organs, which is usually present,
T cell lymphocytes in lymph nodes produce high also needs consideration in the treatment planning.
levels of several cytokines, including tumor necrosis Surgical removal may be an option for intracere-
factor-a (TNF-a), granulocyte–macrophage colony- bral granulomas. When the resection is incomplete
stimulating factor (GM-CSF), interferon-g, and inter- radiotherapy is usually added. Extra-axial locations at
leukins (IL)-1, -2, -3, -4, -7, and -10. In pulmonary the convexity, subdural or arachnoidal, are resected
lesions LCH expresses the costimulatory molecules when possible. Lesions of the skull base are less suit-
B7-1 and B7-2. ed to radical surgical removal. Radiotherapy and
In the lesions in the cerebellar hemispheres no chemotherapy are then the remaining options. In pa-
Langerhans cells can be demonstrated. The lesions tients with multiple lesions radiotherapy combined
are characterized by demyelination, gliosis, and loss with chemotherapy may control the disease for many
of Purkinje and granular cells. years. In patients with diabetes insipidus primary
treatment depends on the neuroimaging findings. In
patients with a nearly empty sella and lack of high
93.3 Pathogenetic Considerations signal on T1-weighted MRI of the posterior part of the
pituitary, substitution therapy is indicated. In pa-
LCH is a proliferative disorder of mesenchymal cells, tients with granulomatous involvement of the pitu-
and can therefore only develop where mesenchymal itary–hypothalamic system, surgery, irradiation and
tissue is present: within the CNS in the meninges, the chemotherapy in addition to hormonal substitution
adventitia of blood vessels, and among free microglial therapy have to be considered. Substances with favor-
cells. It is unknown what triggers the transformation able effects on LCH are steroids, vinblastine, and
of normal antigen-presenting dendritic Langerhans etoposide (VP16). Etoposide is a semisynthetic epi-
cells into the clonal Langerhans cells that are defec- podophyllotoxin used in the treatment of malignan-
tive in this ability. Clonal proliferation leads to func- cies of the macrophage and monocyte lineage, but al-
tional deficiency. One theory assumes that LCH cells so effective in LCH. It is noteworthy that etoposide
arise from the adventitia of blood vessels, causing may cause a high signal intensity of the basal ganglia
perivascular histiocytic aggregates that develop into on T1-weighted MR images.
larger granulomatous masses with variable portions Lesions in the cerebellar white matter not contain-
of foamy histiocytes, eosinophils, microglia, lympho- ing Langerhans cells cause a treatment dilemma. If an
cytes, and plasma cells. In later stages fibrosis and as- autoimmune cause is supposed, one could expect
trocytic gliosis become dominant. Why granulomas benefits from corticosteroids or immunosuppressive
have a special affinity to the pituitary and hypothala- therapy. If another explanation is supposed, for in-
mic structures remains so far an unanswered ques- stance cytokine toxicity, therapy is unclear. So far no
tion. definite guidelines have been suggested for the treat-
The symmetrical lesions located in the cerebellar ment of the cerebellar white matter abnormalities.
white matter, also involving Purkinje and granular
cells, but without Langerhans cells on histological ex-
amination, require a separate explanation. One of the 93.5 Magnetic Resonance Imaging
theories compares the cerebellar and pontine lesions
in LCH to the cerebellar degeneration seen in parane- MR is aimed at depicting the different possible mani-
oplastic syndromes. In this paraneoplastic syndrome festations and the different stages of LCH of both the
antineuronal (anti-Yu) antibodies can be demon- skull and the intracranial contents. To visualize le-
strated. The search for antibodies against cerebellar sions of the skull, the cranial vault as well as the skull
tissue has, however, been negative in LCH patients. base, one can effectively use T1-weighted images, with
Another theory proposes a toxic role for the high con- and without fat suppression, with and without con-
centrations of cytokines produced by LCH cells, but trast. This can be combined with using the same T1-
again definitive proof is lacking. weighted sequences for visualizing the intracranial
contents, both the extra- and intra-axial structures, as
these sequences are also effective in depicting sub-
93.4 Therapy dural and leptomeningeal disease manifestations.
Granulomatous LCH lesions in the skull base and
Therapy in LCH with CNS involvement is dependent brain will enhance after contrast injection. In addi-
on the nature and extent of the lesion and the sec- tion T2-weighted and FLAIR images are indicated to
ondary implications. It is important to realize that in survey the intracranial contents for other manifesta-
some patients the lesions tend to regress sponta- tions.
93.5 Magnetic Resonance Imaging 711

lobes, show up on the sequences indicated above. In


all cases one can expect enhancement of the granulo-
matous lesions.
There are also two possible types of lesions in the
posterior fossa. First of all, granulomatous manifesta-
tions of LCH can be observed, space-occupying, en-
hancing after contrast, and usually asymmetrical.
Secondly, bilateral, symmetrical, confluent signal ab-
normalities can be seen in the white matter of the
hilus of the dentate nucleus, the corpus medullare of
the cerebellum, and the pons (Figs. 93.3 and 93.4). The
bilateral cerebellar lesions do not represent LCH le-
sions, do not show mass effect, and do not enhance
(Figs. 93.3 and 93.4). In these patients, the dentate nu-
cleus often has a high signal on T1-weighted images.
The globus pallidus and, less often, the caudate nucle-
us may also have a high signal intensity on T1-weight-
ed images (Figs. 93.3 and 93.4). Within the supraten-
torial white matter, enlarged perivascular spaces are
frequently present. Contrast enhancement following
a vascular pattern may be present. More prominent,
Fig. 93.1. A 65-year-old female patient with histologically
patchy white matter abnormalities not following a
confirmed LCH. The midsagittal, T1-weighted, contrast-en-
hanced, fat-saturated image depicts a lesion mainly involving
vascular pattern may also be present.
the hypothalamus
In Erdheim–Chester disease, a non-Langerhans
histiocytosis, similar lesions as described for LCH
may be seen (Figs. 93.2 and 93.5). Granulomatous le-
sions may be found in the sellar and suprasellar re-
In patients with involvement of the pituitary–hy- gion as well as in other areas (Fig. 93.2). Progressive
pothalamus axis, sagittal and coronal T1-weighted intra-axial lesions have been reported involving the
images with fat suppression, without and with con- pons, superior part of the medulla oblongata, the
trast injection, will show in some patients a partially cerebellar peduncles, and areas around the fourth
empty sella and lack of the bright signal of the poste- ventricles, as well as an extra-axial mass with encase-
rior pituitary, and in other patients a granulomatous ment of the vertebral artery. Autopsy in one of these
lesion in the suprasellar region, around the pituitary cases revealed histiocytic infiltration of the cerebellar
stalk, with or without extension into the skull base pathways, in particular the base of the pons and the
and meninges (Fig. 93.1). Infundibular thickening middle cerebellar peduncles. The nonenhancing sym-
and infundibular atrophy are frequent findings. Oth- metrical bilateral lesions in the cerebellar hemi-
er isolated or multifocal intracerebral lesions, for ex- spheres with clinically a progressive spastic–ataxic
ample LCH of the choroid plexus, or space-occupying syndrome may also occur in Erdheim–Chester dis-
lesions in the frontal, temporal, parietal, or occipital ease (Fig. 93.5).

Fig. 93.2. A 55-year-old woman


with Erdheim–Chester disease, as
confirmed at autopsy.The sagittal
T1-weighted image with contrast
shows an intra- and suprasellar
process, enlarging the sella turcica
and reaching upward unto the optic
chiasm. A second, extra-axial lesion is
located anteriorly in the cranioverte-
bral junction. Both lesions enhance
homogeneously.The axial FLAIR
image reveals the lesion with high
signal intensity. From Thorns et al.
(2003), with permission
712 Chapter 93 Langerhans Cell Histiocytosis

Fig. 93.3. A 16-year-old male patient


with LCH.The T1-weighted images
(upper row) reveal high signal intensity
in the globus pallidus.The coronal
T2-weighted images (second row) of
the posterior fossa show hyperinten-
sity of the cerebellar white matter
and the brain stem
93.5 Magnetic Resonance Imaging 713

Fig. 93.4. A 17-year-old boy with LCH. The upper row shows row shows the T2-weighted images at the level of the brain
T2-weighted (left), proton density (middle), and T1-weighted stem and cerebellum. The tracts in the midbrain and base of
(right) images at the level of the basal ganglia.The globus pal- the pons as well as the cerebellar white matter display a high
lidus has an increased signal on all these images. The second signal. From Saatci et al. (1999), with permission

Fig. 93.5. A 44-year-old man with


Erdheim–Chester disease.The
T2-weighted images show hyperin-
tensity of the cerebellar white matter.
From Weidauer et al. (2003), with
permission
Chapter 94

Post-Hypoxic–Ischemic Damage

94.1 Pathogenetic Factors such as the endoplasmic reticulum and mitochon-


dria, and it is not until these are flooded that the cyto-
In hypoxic and ischemic conditions the supply of sol concentration of Ca2+ will increase. Ca2+ is a cofac-
oxygen and nutrients to brain tissue is compromised. tor for certain enzymes, including proteases and
If this condition lasts for some time, changes occur in phospholipases, which will attack functional compo-
cells and tissues, eventually leading to cell death. nents of the cell such as lipids and proteins. High Ca2+
The first change to occur at a molecular level is the levels in the cytosol are at the core of many detrimen-
replacement of the highly efficient aerobic glycolysis, tal biochemical cascades.
which delivers 38 molecules of adenosine triphos- The oxygen paradox is based on the observation
phate (ATP) for one molecule of glucose, by ineffi- that oxygen supplied after a hypoxic episode causes
cient anaerobic glycolysis with only 2 molecules of additional damage. This is explained by the action of
ATP for one molecule of glucose. Under aerobic con- oxygen radicals. Free radicals are compounds with a
ditions the major product of glycolysis is pyruvate, lone electron in an outer orbital, which are, therefore,
which is metabolized to acetyl-CoA (activated ac- highly reactive. They are normally present in the mi-
etate), which enters the citric acid cycle. Under anaer- tochondrial electron transport chain of all cells and
obic conditions, pyruvate is not converted into acetyl- are kept under control by physical and chemical cou-
CoA, but into lactate, a potentially harmful com- pling. The defense mechanisms against the toxic ef-
pound. fect of oxygen radicals include a low oxygen pressure
Energy depletion leads to membrane depolariza- at the cellular level, and enzymatic decomposition of
tion and disturbed transport of ions across mem- oxygen radicals by enzymes and scavengers, such as
branes of organelles and cells. Concentration and superoxide dismutase and catalase, as well as antioxi-
voltage gradients are no longer maintained; water is dants, such as a-tocopherol (vitamin E) and ascorbic
osmotically drawn into the organelles and cells, caus- acid (vitamin C). The latter compounds are fat-solu-
ing them to swell. These changes may be reversible, ble and water-soluble, respectively. Superoxide dis-
but if the hypoxic condition lasts too long, irreparable mutase snatches away the superoxide ion; catalase
damage with cell death will occur. Morphologically counteracts the effects of hydrogen peroxide. Howev-
this point of no return is indicated by the appearance er, under conditions of hypoxia–ischemia, and in par-
of cloudy precipitations within the mitochondria.Ap- ticular after restoration of oxygen supply, these de-
parent changes in the inner membrane of the mito- fense mechanisms may be overwhelmed. One of the
chondrion, where oxidative phosphorylation takes cascades that then become active is the xanthine oxi-
place, occur much later. dase reaction. Under anaerobic conditions, ATP is
broken down in larger than usual quantities via ADP
to AMP, to inosine and hypoxanthine, and finally to
94.2 Reperfusion Damage uric acid under the enzymatic influence of xanthine
dehydrogenase. The raised Ca2+ concentration in the
Repair of the condition that has caused the hypoxic– cytosol activates proteases, which convert xanthine
ischemic changes, with restoration of blood circula- dehydrogenase into xanthine oxidase, thereby pro-
tion and oxygen supply, can introduce additional ducing a superoxide anion (O•). Superoxide anions,
damage in ischemic cells. This means that cells that once formed from this xanthine oxidase system, react
are reversibly damaged are again threatened, now by with iron salts to form perhydroxyl radicals (OH•).
the harmful effects of reperfusion. Two factors are as- This chemical process is described as the superoxide-
sumed to play a critical role as causative agent in this driven, iron-promoted Haber–Weiss reaction. This
additional damage: the calcium factor and the oxygen reaction occurs when free Fe2+ is present: under hem-
paradox. orrhagic conditions or in structures whose natural
The concentration of Ca2+ in blood is much higher iron content is high. Targets for the perhydroxyl radi-
than in the cell. A Ca2+ gradient is maintained across cals are polyunsaturated fatty acids, compounds that
the cell membrane with ATP as energy donor. Lower- are highly vulnerable to peroxidation. Polyunsaturat-
ing of the ATP concentration leads to influx of Ca2+ ed fatty acids reside in the midzone of biomem-
and Na+ into the cell. Ca2+ is first stored in organelles, branes, spatially separated from the site of generation
94.2 Reperfusion Damage 715

of active oxygen. Under hypoxic–ischemic condi- CNS and there is evidence that it may contribute to
tions, with activation of Ca2+-dependent enzymes in excitotoxicity. It is a simple molecule which is synthe-
the cells, in particular proteases and phospholipases, sized from L-arginine by the enzyme nitric oxide syn-
this is no longer the case. The polyunsaturated fatty thase. Nitric oxide synthase is present in endothelial
acids in the cell membrane come under attack and be- cells. There are several isoforms and the neuronal iso-
come new sources of oxygen radicals. The superoxide form is activated by high cytosol Ca2+. Once formed,
anion withdraws a proton from a polyunsaturated fat- nitric oxide diffuses to neighboring cells and reacts
ty acid, creating a lipid with an unpaired electron. This with guanylate cyclase, inducing the formation of
newly created lipid radical (L•) reacts with O2 (reper- cyclic guanosine monophosphate from guanosine
fusion) and forms a lipid peroxyl radical (LOO•). This triphosphate. Cyclic guanosine monophosphate in-
compound will withdraw a proton from a nearby fluences the relaxation of smooth muscles, including
polyunsaturated fatty acid, creating a new radical and blood vessels. Nitric oxide preferentially combines
initiating a chain reaction. Iron plays a very important with superoxide anions when present, leading to the
catalytic role in lipid peroxidation. In a normal adult formation of highly reactive peroxynitrite, which
about 4 g iron is present in hemoglobin. Myoglobin causes cell damage. The resulting drop in cyclic
contains 10% of total body iron and a small part is guanosine monophosphate leads to vasoconstriction.
present in enzymes and in the transport protein trans- On the other hand, nitric oxide can be changed to a
ferrin. The remaining iron is found in storage proteins chemical state that has the opposite effect, i.e., a pro-
such as ferritin. Free iron is not usually present in pe- tective effect. In the presence of electron donors such
ripheral blood. Iron from ferritin is liberated at pH as ascorbate or cysteine, nitric oxide becomes ni-
values lower than 6, which may occur under condi- trosonium ion (NO+), which binds to a regulatory site
tions of severe hypoxia. Superoxide anions are also ca- on the most prominent receptor of excitatory amino
pable of dissociating the iron–ferritin complex. Lipid acids, resulting in decreased activity of this receptor.
peroxyl radicals can even dissociate iron from hemo- The deleterious action of free radicals, as mentioned
globin molecules. Hemorrhagic conditions, such as before, is dependent on the presence of oxygen.
hemorrhagic infarctions, infections, contusions, and Therefore, most of the damage occurs during reperfu-
subarachnoid hemorrhages, create the optimal envi- sion after ischemia. The capacity of tissue to neutral-
ronment for these deleterious reactions. ize free radicals is called the antioxidance capacity.
In the formation of free radicals the polyunsaturat- During ischemia and reperfusion, this capacity de-
ed fatty acid arachidonic acid, abundant in cell mem- creases rapidly. The decrease is greater in experi-
branes, has a special place.Arachidonic acid is metab- ments in which some blood is still perfusing the is-
olized by at least three enzymes: cyclooxygenase, chemic area than when the oxygen supply is totally
lipoxygenase, and cytochrome P-450 oxygenase. All arrested. This is understandable because the presence
three pathways are capable of producing superoxide of oxygen is needed for free radical damage to occur.
radicals. The cyclooxygenase pathway leads to the The recognition of the harmful effects of free rad-
formation of prostaglandins and thromboxanes, icals has induced a worldwide search for counteract-
which influence, amongst others, the microcircula- ing compounds. Several pharmaceuticals are under
tion. The lipoxygenase pathway leads to the forma- consideration as cerebroprotective agents. Free radi-
tion of leukotrienes, which may induce edema and cal scavengers are used to diminish negative effects of
necrosis and further inhibit microcirculation. Cy- cranial trauma or cerebral hypoxia. Substances such
tochrome P-450 oxygenase leads directly to the for- as vitamin E, N-acetylcysteine, and dimethylsulfoxide
mation of oxygen radicals. There is interaction at are used for this purpose.Vitamin E works by offering
each level of the components of this cascade, leading a competitive binding site to superoxide anions. Al-
to either stimulation or inhibition of effects of the lopurinol counteracts the effects of xanthine oxidase,
other components. and is effective in diminishing the influence of one of
Free radicals are formed by many other mecha- the cascades. The 21-amino steroids (lazaroids) have
nisms. They can be formed by membrane-bound been proven to be potent inhibitors of lipid peroxida-
enzymes on the surface of neutrophils and phago- tion. Calcium entry blockers are considered to pre-
cytes, which are activated by endotoxins from bacte- vent high Ca2+ concentrations in the cytosol and in-
ria, or by cytokines, inducing the so-called oxygen hibit the formation of Ca2+-dependent enzymes. Not
burst of phagocyte membrane compounds in the all newly conceived drugs have met the theoretical ex-
acute phase of infections. pectations. Often, of course, in clinical situations the
Nitric oxide (NO•) is a reactive free radical, which therapy starts when most of the damage has already
can inhibit mitochondrial respiration. It is one of the been done. Moreover, the processes behind post-hy-
major defense molecules against microbes and malig- poxic–ischemic encephalopathy are complicated.And
nant cells. It has recently been reported to act as a also: free radicals are not the only factors that have to
highly unorthodox messenger molecule within the be taken into account.
716 Chapter 94 Post-Hypoxic–Ischemic Damage

94.3 Excitatory Amino Acids concentration in the cell. These non-NMDA receptors
open channels to allow Na+ and Cl– into the cell under
Overstimulation of neurons by excitatory amino anoxic conditions, passively causing the entrance of
acids (EAAs), in particular glutamate and aspartate, H2O and cell edema.
can lead to cell dysfunction and, eventually, neuronal The action of these EAA receptors is fundamental
death. EEAs serve as excitatory neurotransmitters, as in the concept of excitotoxicity. Effects of overstimu-
opposed to inhibitory neurotransmitters, of which lation by EAAs have been considered to play a role in
g-aminobutyric acid (GABA) is the most prominent. many different neurological disorders, for example, in
These amino acids play a role in the normal messen- epilepsy, hypoglycemia, trauma, AIDS dementia com-
ger system of the CNS. Under normal circumstances plex, Huntington chorea, amyotrophic lateral sclero-
the potentially harmful accumulation of EAAs is pre- sis, toxic encephalopathies, and perhaps Alzheimer
vented from happening. After conversion from gluta- disease, making the described mechanism a “final
mine, glutamate is released in the synapse, making common pathway” of neuronal injury.
contact with glutamate-responding receptors. On the Evidence has been provided to support the concept
distal side of the synapse, glutamate is broken down that in vivo high levels of endogenous EAAs, such as
to glutamine, which re-enters the presynaptic side, or glutamate and aspartate, also play a critical role in
is taken up by a glutamate transporter into a nearby brain damage caused by hypoxic–ischemic insults. As
astrocyte for further transformation. The ensuing has already been stressed, glutamate reclaim after the
neuronal depolarization is very short-lived. This glu- synaptic release is highly energy-dependent. In low-
tamate reuptake chain is energy-dependent. In ener- energy states glutamate accumulates in the extracel-
gy-deprived conditions glutamate accumulates in the lular spaces, leading to neuronal overstimulation with
extracellular space and causes neuronal overstimula- excessive anionic and cationic fluxes and, in the acute
tion. There are several mechanisms leading to abnor- phase, to intracellular edema and osmotic lysis, which
mal glutamate accumulation. For example, the role of may be the cause of instant cell death. Resulting high
the glutamate transporter in the astrocyte may be concentrations of Ca2+ in the cytosol contribute to de-
compromised, because of the collapse of the gradients layed cell death. In vitro tests have shown that these
for Na+ and K+ across the cell membrane. Its function two processes, the osmotic instant lysis and the de-
may even be reversed and the transporter may be- layed Ca2+-dependent neuronal death, can be separat-
come a source of extracellular glutamate. Or, in case ed by manipulating the conditions. The possibility
of cell injury, glutamate may leak out of damaged of preventing each type of damage by different bio-
cells. Every cell contains enough glutamate to cause chemical environments also confirms this difference.
damage to many neighboring cells. This knowledge is potentially of use in therapeutic in-
Further analysis of the excitatory synapses reveals terventions.
that at least three types of glutamate receptor can be Antagonists of EAAs have been tested in cell cul-
distinguished. The EAA receptors are classified ac- tures, animal experiments, and clinical trials of their
cording to their typical agonists: N-methyl-D-aspar- “cerebroprotective” properties. In the clinical tests, es-
tate (NMDA), quisqualate (QA), kainate (KA), and pecially, the results so far have been disappointing.
2-amino-4-phosphonobutyrate (AP4). Each class of This may be because the whole process is extremely
receptor has distinct pharmacological properties and complicated and an antagonist interferes somewhere
anatomical distribution. The NMDA receptor is the in this complex process with unexpected reactions
best understood. It contains an NMDA recognition and effects, or because there is insufficient passage
site and a cationic ionophore, allowing Na+ and Ca2+ through the blood–brain barrier, or because the ad-
to enter the cytosol. Other sites are an Mg2+ binding ministration of drugs in humans usually occurs when
site and a Zn2+ binding site, probably for noncompet- the damage has already been done.
itive inhibition of the ionophore. A glycine site has al-
so been recognized for positive allosteric modulation
at the receptor site. Agonists and antagonists of each 94.4 Patterns of Morphological Changes
of these sites are known. For example, agonists for of White Matter in Hypoxic–Ischemic
the NMDA site are L-glutamate and L-aspartate; an Encephalopathy
antagonist is carboxypiperazine propyl 1-phospho-
nate. At the ionophore site antagonists are phencycli- Hypoxemia, a reduced oxygen content of the arterial
dine, imine maleate (MK 801), ketamine, and dex- blood, occurs clinically in many conditions, including
tromethorphan. At the glycine site, glycine and D-ser- asphyxia, severe pulmonary failure, respiratory insuf-
ine are agonists; kynurenate is an antagonist. ficiency, arrest, and incorrect anesthesia. Ischemia or
Agonists and antagonists for the quisqualate and oligemia is a reduction in blood flow. This condition
kainate receptors have also been identified. They are exists where arteries are occluded, in exsanguination,
directly involved in the regulation of Ca2+ and Na+ and in cardiac arrest. Hypoxia, ischemia, and hypo-
94.4 Patterns of Morphological Changes of White Matter in Hypoxic–Ischemic Encephalopathy 717

glycemia have the same effect on the brain. Neurons sis), striatum necrosis, and Purkinje cell necro-
are usually most susceptible, but there is also a hier- sis. At least two factors are involved in this se-
archy of vulnerability within the group of neurons, as lective involvement of particular brain struc-
is discussed in the chapter on selective vulnerability tures. First the distribution of excitatory amino
(Chap. 3). Under certain conditions, however, for ex- acid receptors is important. The differences in
ample an early stage of development, white matter concentration of EAA receptors can lead to
may be more vulnerable than gray matter. This is par- focal damage in areas with higher receptor
ticularly the case in preterm infants. density. These areas are to a large extent the ar-
Several patterns of morphological post-hypoxic– eas involved as mentioned above. Secondly, a
ischemic damage can be distinguished: role is also played by reperfusion damage and
1. Focal lesions, the distribution of which depends at free radical action. As has been explained,
least in part on vascular patterns: the Haber–Weiss reaction occurs especially in
(a) Arterial territorial pattern. If the perfusion areas with a high iron concentration, including
through one major artery fails, the territory the thalamus, globus pallidus, substantia nigra,
supplied by this artery is compromised. The and several cortical layers.
initial cytotoxic edema affects all structures, (b) Specific involvement of zones of active myelina-
gray and white. tion. Following asphyxia in term-born neo-
(b) The arterial border zone pattern, due to dimin- nates, lesions are frequently seen in the dorsal
ished perfusion, mostly leads to changes in the part of the putamen, the ventrolateral nucleus
parietal region, or along the frontal parasagit- of the thalamus, and in the hippocampus. They
tal border between middle and anterior cere- are often combined with damage in the white
bral arteries. Other border zones may, howev- and gray matter bordering the central sulcus,
er, also be involved. leading to local atrophy and sclerotic ulegyria.
(c) Focal symmetrical abnormalities occur prefer- The explanation for the vulnerability of these
entially in the basal ganglia in patients with affected areas at this time is the active myelina-
chronic hypertension and concurrent vascular tion taking place at term in these areas, with
changes. Etat lacunaire is the ancient denota- high chemical turnover and high regional
tion of a condition in which multiple small, blood flow.
old, cystic infarcts are seeded throughout the (c) Diffuse white matter injury. In delayed post-
basal ganglia, leading to a highly characteristic hypoxic–ischemic demyelination there is usu-
appearance. This condition is difficult to sepa- ally diffuse involvement of all the white matter.
rate from the état criblé (cribriform atrophy) This is seen in rare cases after cardiac arrest,
caused by the widening of the Virchow–Robin errors in anesthesia, and in toxic encephalo-
spaces around tortuous vessels. pathies, in particular after carbon monoxide
(d) Periventricular leukomalacia occurs in older and cyanide poisoning. The same pattern is
patients in subcortical arteriosclerotic en- seen in diffuse white matter injury after irradi-
cephalopathy, also referred to as Binswanger ation and chemotherapy, in which diffuse vas-
disease. Periventricular leukomalacia also oc- cular changes play an intermediary role.
curs in prematurely born infants. 3. Miscellaneous lesions affecting both gray and
2. Lesions due to generalized hypoxia–ischemia: white matter:
(a) Cortical and subcortical gray matter damage. A number of congenital conditions such as en-
Typical involvement of the “topistic” areas as cephaloclastic schizencephaly, hydranencephaly,
described by Vogt and Vogt (1937) is the result and porencephaly are probably the result of hy-
of generalized hypoxia–ischemia. Morpho- poxic–ischemic conditions during pregnancy.
logically changes occur in the most vulnerable The conditions that primarily lead to white matter
areas, resulting in cortical laminar necrosis, changes will be discussed in separate chapters.
Sommer’s sector necrosis (hippocampus necro-
Chapter 95

Post-Hypoxic–Ischemic Encephalopathy of Neonates


J. Valk, R.J. Vermeulen, M.S. van der Knaap

95.1 Clinical Features often have epilepsy. Delayed visual maturation and
and Laboratory Investigations cerebral visual failure are usually present.
A third group of neonates develop a multicystic
Periventricular leukomalacia (PVL) is a post-hypox- encephalopathy (MCE). Patients with MCE are more
ic–ischemic leukoencephalopathy resulting from pre- often born at term than preterm. In the majority of
or perinatal hypoxic–ischemic insults. Little (1861) the children the clinical signs of a devastating en-
was the first to describe the clinical picture of the con- cephalopathy are unexpected. The typical clinical
dition. It occurs in particular in preterm neonates course usually includes initially not very alarming
with a gestational age of 32–36 weeks. Major risk fac- signs of fetal distress, such as prolonged bradycardia
tors are prematurity and intrauterine infection. Be- or meconium-stained amniotic fluid, followed by
fore that time, at a gestational age of 28–32 weeks, ger- moderate starting problems after delivery and rapid
minal layer, intraventricular, and intraparenchymal partial recovery. Several hours later a devastating
hemorrhages predominate. PVL is rarely seen in term encephalopathy develops, characterized by lowered
neonates; if present, it is often in combination with consciousness, seizures and, on ultrasound, general-
cardiac disorders, intrauterine growth retardation, or ized cerebral edema. These patients have a very poor
any other sign of intrauterine incidents. There is a outcome, with microcephaly, severe developmental
distinct relationship between PVL and the clinical retardation, spastic tetraplegia, visual handicap, and
concept of “cerebral palsy,” although it should be real- seizures.
ized that the concept of cerebral palsy comprises all A special clinical picture is seen as the result of
infants with a stable handicap which occurred before acute profound hypoxia–ischemia in term or post-
the age of one year. The increasing technology in term babies. In the course of the first year of life an ex-
neonatal medicine has made it possible to keep more trapyramidal movement disorder becomes apparent
premature infants alive. As a consequence, PVL as a with choreoathetosis, dystonia, and orobuccolingual
cause of cerebral palsy has increased in relative fre- dyskinesia in combination with variable spasticity
quency. and variable mental retardation, although mental ca-
The typical location of PVL in the periventricular pacities are often better preserved. The motor handi-
region, interfering with the corticospinal tracts of the cap is usually very severe, the combination of ex-
legs more than of the arms, is responsible for the re- trapyramidal and pyramidal abnormalities leading to
sulting spastic diplegia, tetraplegia (legs more severe- serious impairment of intentional movements. This
ly affected than arms), or hemiplegia (leg more se- clinical picture is associated with lesions of the basal
verely affected than arm). Patients with motor dis- nuclei and the central, perirolandic cortical and sub-
ability and PVL have a relatively high incidence of cortical area. A similar clinical condition may occur
seizures. Epilepsy in patients with PVL is associated in preterm neonates, when they undergo a similar
with multiple seizure types. Mental development is episode of acute profound hypoxia–ischemia, but the
relatively better preserved than motor development, central cortical and subcortical areas are not similar-
although there is increasing evidence that there are ly involved, probably due to the state of myelination in
also cognitive deficits in PVL. Extension of the leuko- the preterm born infants.
malacia into the optic radiation may lead to cortical It is estimated that post-hypoxic–ischemic en-
blindness or, more often, delayed visual maturation. cephalopathy is the cause of 90% of cases of cerebral
The CNS manifestations of PVL take 1 or 2 years to palsy. For decisions on how to proceed with the treat-
show the full clinical impact. ment of a very sick patient in the neonatal intensive
In older preterm infants the white matter damage care unit, it is extremely useful to be able to predict
tends to have a more peripheral, subcortical location. the outcome of damage at an early stage. For the pre-
This condition is known as subcortical leukomalacia diction of the outcome of pre- and perinatal injury
(SCL). In fact, PVL and SCL form a continuum, and one has to rely in the early phase on clinical sympto-
SCL as a rule includes periventricular white matter matology and paraclinical test results available at
damage. Clinically, so-called SCL leads to a more se- the time. A classification of the severity of post-
vere handicap than PVL. The children have spastic hypoxic–ischemic encephalopathy was proposed by
tetraplegia, are mentally more severely retarded, and Sarnat and Sarnat (1976) for neonates with a gesta-
95.2 Pathology 719

tional age of over 36 weeks. This classification, based nique. A lowered pulsatility index is associated with
on clinical and EEG findings, has been shown to have more severe post-hypoxic–ischemic encephalopathy,
prognostic significance and is the standard of clinical although the overlap with normal is rather large.
scoring in neonatal intensive care unit in infants with Near-infrared spectroscopy is a method with po-
post-hypoxic–ischemic encephalopathy. Stage I usu- tential to measure changes in the oxy–deoxyhemo-
ally occurs during the first 24 h of life and is charac- globin ratio in the brain and to estimate cerebral
terized by jitteriness, a state of irritability, hyperalert- blood flow. The method has the advantage of being
ness, sympathetic nervous system preponderance, bedside and providing continuous information.
uninhibited reflexes, and a normal EEG. Stage II There is, however, only a limited window available,
consists clinically of hypotonia, lethargy, or obtun- because of interference with the skull. The method is
dation for at least 12 h after birth, strong distal flex- in clinical use in some intensive care units and in re-
ion, parasympathetic predominance, and multifocal search settings.
seizures. EEG displays periodicity, sometimes preced- PET studies have occasionally been used to deter-
ed by continuous delta wave activity. The period be- mine the metabolic rate and the cerebral blood flow
tween 48 and 72 h is critical, during which the en- of the various brain regions. This is rather a research
cephalopathy either worsens or improves. Stage III is than a clinical tool for post-hypoxic–ischemic en-
characterized by suppression of brain stem and auto- cephalopathy in neonates.
nomic functions, stupor or coma, and generalized Clinical signs, EEG, US, and evoked potentials are
flaccidity. Mechanical ventilation is necessary. The of the highest importance as predictors of the out-
EEG is isoelectric or shows a burst–suppression pat- come of perinatal asphyxia in the acute stage, espe-
tern. Persistence of stage II for less than 5 days and cially in those newborns that require intensive treat-
absence of entry in stage III are associated with favor- ment to survive. In early stages of post-hypoxic–is-
able outcome. Entry into stage III, failure of the EEG chemic damage special MR techniques, including dif-
to return to normal, or persistence of stage II for fusion-weighted imaging and MRS, have proved to be
longer than 7 days predicts a poor neurological out- of great importance in the prediction of outcome. In a
come. later phase the extent of lesions on MRI, in particular
In a large cohort, including 1807 preterm (32–36 the presence of subcortical damage, glial retraction,
weeks gestational age) and very preterm (25–32 and ulegyria, the presence of secondary phenomena,
weeks gestational age) infants it was shown that there such as atrophy and hydrocephalus, and the distur-
was a significant inverse relationship between umbil- bance of myelination are helpful in predicting the de-
ical cord pH and base excess values and subsequent gree of future disability.
adverse outcome for infants delivered preterm (Vic-
tory et al. 2003). Determination of CSF lactate levels in
150 nonasphyxiated and 46 asphyxiated neonates 95.2 Pathology
showed a significant relationship between elevated
lactate levels, fetal distress, and APGAR scores and PVL was described histological for the first time in
seems to be an objective way of assessing the severity 1867 by Virchow and in 1868 and 1873 by Parrot. They
of cerebral hypoxia (Mathew et al. 1980). described “pale” infarcts in the periventricular white
Abnormal VEP and SSEP results carry some nega- matter as yellowish or chalky plaques, 1–6 mm in di-
tive prognostic value. ameter, 1–15 mm from the ependymal surface of the
Ultrasound (US), being versatile and a bedside ex- lateral ventricles. Softening of the plaques forms cav-
amination, is the imaging modality of first choice in ities filled with a milky fluid. The most common lo-
the neonatal intensive care unit. It can help to identi- calizations are the area anterior to the frontal horn,
fy areas of hyperechogenicity located around the the superolateral angles of the lateral ventricles, the
trigonum or the frontal horns with onset 3–7 days peritrigonal area, and the area around the occipital
after the hypoxic–ischemic incident. This hypere- horns.
chogenicity is either transient or progresses to cysts Microscopically the lesions initially show coagula-
in about 7–14 days. This time delay is also reported in tion necrosis, with nuclear pyknosis and sponginess
histological studies. US can be helpful in demonstrat- of the tissue. Astrocytic proliferation at the borders
ing SCL, depending on the type of equipment. US is sets in after a few days and varicose axon swellings
also helpful in demonstrating germinal layer hemor- develop. Microglia proliferate and lipid-laden macro-
rhage, intraventricular hemorrhage, ventricular di- phages accumulate. The organizing lesions are delin-
latation, intraparenchymal hemorrhage, focal infarc- eated by active gliosis. Cavitation forms within a few
tions, porencephalic cysts, and lesions in the basal weeks. The cavities are lined by fibrillary glial scar tis-
ganglia. An important acquisition is the ability to sue. Swollen axons mineralize quickly and persist for
measure flow velocity and pulsatility index in major many months. Most of the infarcts are ischemic and
cerebral vessels with the Doppler ultrasound tech- lack hemorrhage.Vascular-occlusive changes are usu-
720 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

ally absent. Occasionally hemorrhages are found with markably thinned. Softening of the putamen, globus
a distribution similar to that of the leukomalacia. pallidus, and lateral thalamus may be present. There
They are due to secondary hemorrhages into periven- are foci of necrosis involving different levels of the
tricular infarcts. Under certain circumstances these brain stem down to the lower medulla. The spinal
hemorrhages can become massive. cord is not involved. Microscopically intensive gliosis
The severity of the gliotic reaction and its extent and the presence of fat-laden macrophages are seen
depend on age. Before 24–28 weeks gestational age, in the walls of the cysts. In the basal ganglia symmet-
the immature brain cannot fully respond to insults rical necrotic changes are apparent: the neuropil is
with astrogliosis. In premature neonates of 28–32 destroyed and few neurons remain. Scattered calcifi-
weeks gestational age, gliosis is usually limited to cations and encrusted neurons are seen in the cere-
the periventricular area (PVL). In older premature bral cortex and basal ganglia. In the cerebellum there
neonates gliosis extends more often into the subcorti- is loss of Purkinje cells, many of which are swollen,
cal area (SCL). This subcortical extension of the gliot- with cactus-like dendrites in the molecular layer.
ic lesion is probably related to both gestational age
and the severity of the hypoxic–ischemic insult. If
subcortical cysts develop and the child dies during 95.3 Pathogenetic Considerations
this stage, multicystic subcortical degeneration is
found at autopsy. If the child survives, the cysts usual- PVL and germinal-layer-related hemorrhages are
ly disappear and gliotic scarring occurs with retrac- considered to be due to hypoxic–ischemic insults of
tion of white matter deforming ventricular system the preterm neonate, occurring in the pre-, peri-, or
and cortex. The distribution of these lesions follows a postnatal period. Age seems to be the pathoplastic
pattern parallel to the periventricular area, closer to factor. The incidence of germinal-layer-related hem-
the cortical lining, extending from the frontal to the orrhage is highest before the 32nd week of gestation-
occipital region. The scars of PVL and SCL remain al age; the highest incidence of PVL lies between the
visible throughout life. The residual lesions of PVL 32nd and 36th week of gestational age, but there cer-
are characterized by irregular borders of the ventri- tainly is an overlap, and both conditions may be pre-
cles, where periventricular cysts have made contact sent at the same time. Both conditions are rare in
with the lumen of the ventricles, with glial retraction neonates born at term. The high incidence of germi-
often at the level of the trigonum, focal loss of white nal-layer hemorrhage before the age of 32 weeks’ ges-
matter, and deep sulci abutting the walls of the ventri- tation can be explained by the presence of the germi-
cles. In the case of SCL, glial retraction disfigures the nal layer up until that time. After the 32nd week of
centrum semiovale and causes crowding of gyri, gestation the germinal layer rapidly disappears. This
eventually leading to parietal and occipital ulegyria germinal layer or matrix layer is considered to be the
(= sclerotic polymicrogyria). nurturing bed for the neuronal and glial cells of the
In term neonates the pattern of damage is differ- developing brain. It is extremely well vascularized,
ent. The post-hypoxic–ischemic lesions occur prefer- whereas the vascular channels have thin endothelial
entially in a triangular area in the parasagittal, corti- walls without supportive tissue. The germinal layer
cal, and subcortical region bordering the sulcus cen- therefore bleeds easily. A hemodynamic contribution
tralis. The resulting gliotic scar has a typical triangu- comes from the pressure-passive cerebral blood flow
lar shape with retraction of the parietal cortex, in stressed premature children, causing a direct de-
leading to local ulegyria. This is often combined with pendency of the cerebral blood flow on the systemic
lesions in the dorsal part of the putamen and the ven- blood pressure and blood flow. The existence of a
trolateral part of the thalamus. The hippocampus patent ductus arteriosus (ductus Botalli) and its
may also be involved. The nuclei of the brain stem and drug-induced (indomethacin) closure can have a
the dentate nucleus may also be affected in this con- profound influence on the cerebral circulation, as it
dition, and pontosubicular necrosis, a histological en- lacks the regulatory mechanism to buffer these
tity, could also fit into this gamut of post-hypoxic–is- changes. Another important factor is the immaturity
chemic damage of the term neonate. of the lung, which requires mechanical ventilation
Histopathology in MCE reveals multiple cystic with high pressures leading to indirect changes in the
cavities in the white matter, extending from the ven- blood pressure. All these factors explain the relative
tricular wall to the inner cortex, affecting frontal, high frequency of development of germinal-layer he-
parietal, and occipital lobes. Septa bridge the cyst morrhage in early preterm babies. The hemorrhages
walls. The medial and lateral parts of the temporal potentially break through into the ventricles, which
lobes are usually spared. Some convolutions are may or may not expand. Under certain conditions
shrunken, and ulegyric regions are present. The later- there is also a breakthrough of hemorrhage into the
al ventricles are dilated, but do not communicate with parenchyma. The intraparenchymal hemorrhages
the cysts. The corpus callosum and fornix may be re- have a poor prognosis. There is growing evidence that
95.3 Pathogenetic Considerations 721

intraparenchymal extension of the hemorrhage oc- vironment, monitoring, and life support measures.
curs in tissue that has already suffered from hypoxia. These include artificial ventilation, administration of
In other words, the hemorrhage occurs in infarcted surfactant, gastric tube feeding, and drug-induced
tissue, usually a venous infarction. closure of the ductus arteriosus, to mention but a few.
In explaining the selective vulnerability of white Often there were already prenatal problems, which
matter in PVL and SCL, multiple risk factors probably after preterm birth continue into postnatal problems.
play a role. There are three major factors in the selec- It is often not possible to pinpoint one precise hypox-
tive vulnerability of the periventricular white matter ic–ischemic incident as the cause of brain damage. In
in premature infants: the incomplete state of develop- term-born neonates, on the other hand, most cases of
ment of the vascular supply to the cerebral white mat- hypoxia–ischemia are due to a single, severe, perina-
ter; the maturation-dependent impairment in regula- tal incident. The cerebral damage resulting from an
tion of cerebral blood flow; and the maturation-de- acute episode of severe asphyxia is completely differ-
pendent vulnerability of the oligodendroglial precur- ent from the cerebral damage in premature neonates.
sor cells. The first two factors are the underlying Selective vulnerability of the brain in this type of in-
cause of susceptibility to ischemic factors. Arterial cident is apparently dictated by the biochemically
end zones and border zones around the ventricles are most active parts of the brain, which demand the
defined by branches of arteries penetrating the cere- most glucose and oxygen: the zones of active myelina-
bral wall from the pial surface and arteries from the tion. The resulting damage in the term-born neonate
choroid plexus penetrating the ventricular wall. Pres- has the form of the so-called central cortico-subcor-
sure-passive cerebral blood flow in stressed prema- tical pattern, with leukomalacia in the myelination
ture children causes a direct dependency of the cere- zone, extending band-shaped from the basal ganglia
bral blood flow on the systemic blood pressure and into the pre- and postcentral gyri. There are also le-
blood flow, hypotension leading to decreased cerebral sions in the dorsal part of the putamen, the ventrolat-
perfusion. It has been shown that oligodendroglial eral part of the thalamus, and the hippocampus, also
precursor cells are highly vulnerable to attacks by free actively myelinating at that time. In the premature
radicals, which are generated by the cascade of events neonate with a single episode of acute profound as-
in ischemia–reperfusion events. Mature oligoden- phyxia, there are as a rule only lesions in the basal
droglia are more resistant to this type of insult. The ganglia and thalami. This can be explained by the fact
presence of intraventricular and intraparenchymal that the central cortico-subcortical area is not yet
blood is a risk factor. In free radical attacks the pres- myelinating in premature infants. As far as lesions in
ence of oxygen and free iron is an important catalyst the basal ganglia and hippocampus are concerned,
for the Haber–Weiss reaction and the formation of excitotoxicity and high local density of glutamate re-
the perhydroxyl radicals, and the subsequent process ceptors also play a role.
of lipid peroxidation. Iron is present in hemorrhagic The pathophysiology of MCE is less clear. MCE is
tissue. Fe2+ can be liberated from ferritin when the pH usually considered a rare condition in neonates, but
value is lower than 6. Iron is also actively acquired still accounts for about 10% of the patients with post-
during the differentiation of oligodendroglia. In the hypoxic–ischemic injury (Sie et al. 2000a). The risk
ischemia–reperfusion cascade elevation of extra- factors for this group show a striking difference as
cellular glutamate, contributing to excitotoxicity, is a compared to patients developing PVL and those de-
factor contributing to tissue damage. Cytokines re- veloping the cortico-subcortical pattern. PVL is
leased in the presence of maternal (antenatal) and mainly found in preterm infants who suffer from pro-
fetal or neonatal infection contribute to the tissue longed and repeated partial hypoxia–ischemia; the
damage. Hypocarbia leading to vasoconstriction has cortico-subcortical pattern mainly in term-born in-
been shown to be a contributing factor to the white fants, following a single episode of acute profound hy-
matter damage in premature neonates. This finding is poxia–ischemia; MCE is mainly seen in term-born in-
of clinical importance, since premature children with fants, initially with signs of mild hypoxia–ischemia,
respiratory distress supported by artificial ventilation not requiring vigorous resuscitation at delivery, fol-
are at risk of developing hypocarbia. Detailed knowl- lowed by a delayed-onset devastating encephalopathy
edge of all the pathogenic factors active in this field is with generalized brain edema. This sequence of de-
essential to pave the way for interventions and treat- layed-onset neuronal death resembles the sequence of
ment regimes. ischemia–reperfusion damage and secondary energy
The pathophysiology of encephalopathy in prema- failure as substantiated in animal experiments.
turely born neonates differs in many aspects from Without being linked directly to the concept of
that of term-born neonates. In the postnatal period, MCE, it has been noted that neonates, after an initial
the preterm neonate with a low birth weight and im- period of suboptimal responses, improve to a better
mature organs is going through an extremely stress- functional level within the first day, only to present
ful period, in which the baby depends on a special en- with a catastrophic reaction about 24 h after the first
722 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.1. A CT scan of a girl with PVL at the age of 2 years (first with dilatation of the lateral ventricles, deformation of the
row) shows asymmetrical ventriculomegaly and calcifications ventricular walls (due to scarring), white matter volume loss,
around the ventricles. The T2-weighted MR images obtained and periventricular gliosis.In addition,pulvinar lesions are pre-
at the age of 3 years (second and third rows) show severe PVL sent

episode. The explanation for the first episode of sub- oxidation, and accumulation of excitatory amino
optimal function is sought in the acute reaction of acids to delayed cell death. This sequence of events
neurons to energy depletion and cell membrane does not differ in essence from the general cascades
paralysis with edema, swelling, and impaired func- triggered whenever energy failure occurs and can be
tion. When the cell survives, a second, calcium-de- seen as a final common path of secondary neuronal
pendent mechanism is triggered, leading via activa- death.
tion of enzymes, formation of free radicals, lipid per-
95.5 Magnetic Resonance Imaging 723

95.4 Therapy beneficial results have not yet been reported. Energy
depletion leads to the accumulation of excitatory
Because of the serious consequences of post-hypox- amino acids, glutamate in particular. Glutamate ex-
ic–ischemic damage, many attempts have been di- erts its stimulating action via at least three different
rected at preventing PVL and other forms of post-hy- receptors, which have unequal topographical distrib-
poxic–ischemic damage from happening. Intensive ution. Of the glutamate receptors, the pharmacologi-
check-ups during pregnancy, early in utero diagnosis cal properties of the NMDA receptor are best under-
of congenital abnormalities, electronic fetal monitor- stood. An attempt has been made to counteract exci-
ing during delivery, and technically advanced neona- totoxicity by the administration of antagonists for
tal intensive care units have not, however, led to a specific sites of the NMDA receptor. Although neuro-
reduction in the incidence of cerebral palsy, which protective effects have been obtained in vitro, clinical
stands at 1–2 per 10,000 newborns. This is at least trials in preterm neonates have been disappointing.
partially due to a population shift. Neonates of very This is probably because glutamate is extremely im-
low birth weight can now be kept alive and carry high portant for many reactions, and nonselective inhibi-
risk factors for the development of post-hypoxic–is- tion may lead to tampering with the other functions
chemic damage. Preventive measures applied in the of this neurotransmitter. For instance, maturing neu-
past to reduce brain metabolism and thus to increase roreceptors in developing neurosynapses also play a
the resistance against hypoxia, such as the use of phe- role in the maturation of brain parts and brain func-
nobarbital and low body temperature, have failed to tions; in the fetal period they are intermediates of cell
improve the outcome. Better understanding of the plasticity and essential in processes of memory and
pathophysiological mechanisms underlying post-hy- learning.
poxic–ischemic encephalopathy has triggered new Once a static encephalopathy has developed,
efforts to develop cerebroprotective drugs. It has the child should be given physical therapy, special ed-
become clear that disrupted calcium homeostasis ucation if necessary, and neurological treatment to
within ischemic cells is an essential factor in delayed limit secondary consequences such as spasticity and
cell death. Energy depletion leads to high calcium epilepsy.
concentration within the cytosol, triggering many
other cascades that can damage the cell. Calcium en-
try blockers would seem to be a logical answer to this 95.5 Magnetic Resonance Imaging
problem. Results of trials, however, have not been
satisfactory. One of the cascades triggered by high Because of its versatility and ability to be used at the
cytosol calcium is the transformation of xanthine de- bedside, neurosonography (ultrasound, US) will re-
hydroxygenase into xanthine oxidase, both of them main the first-line imaging modality in intensive care
enzymes of the adenosine-nucleotide biochemical units for neonates. Detection and staging of germi-
pathway. The formation of xanthine oxidase leads to nal-layer-related hemorrhage can be done satisfacto-
formation of free radicals and to cell damage by lipid rily with US. This is also true for the evolution of PVL,
peroxidation after reperfusion. It was hoped that which can be followed day by day. Many other brain
blocking of this cascade would improve the outcome. conditions can be correctly diagnosed by US: for
Free radical scavengers have also been tested for their example, cerebral infarctions, hydrocephalus, poren-
ability to prevent reperfusion damage. Substantial cephaly, hydranencephaly, and congenital anomalies,

Table 95.1. MRI sequences for the evaluation of neonates


Sequence Purpose/contribution

Sagittal T1-weighted spin echo Small hemorrhages, cysts, congenital anomalies


Proton density and T2-weighted spin echo Signal changes of white matter and cortical rim, ventricular size
FLAIR Differential diagnosis: asphyxia versus infection
Gradient echo Macro- and microhemorrhages, calcifications
IR Progress of myelination, gray/white matter differentiation
T1-weighted + contrast Cortical laminar necrosis, infections
DWI–anisotropy–Trace Infarcted and preinfarcted tissue, pattern of abnormalities
DTI-FA; fiber tracking Quantitative estimation of brain maturation
MRS Biochemical developmental age; presence of lactate
MRA Vascular anomalies, abnormal vein of Galen

FLAIR, fluid-attenuated inversion recovery; IR, inversion recovery; DWI, diffusion-weighted imaging; DTI-FA, diffusion tensor
imaging–fractional anisotropy; MRS, MR spectroscopy; MRA, MR angiography
724 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.2. a Boy born at 38 weeks of gestation.The pregnancy (third row) images show an intraparenchymal hemorrhage
was complicated by hypertension and perinatal asphyxia oc- on the left. This type of hemorrhage usually has its origin
curred. MRI at the age of 1 week showed mildly dilated lateral in a venous infarction and suggests a previous condition of
ventricles.T2-weighted (first and second rows) and T1-weighted venous congestion

such as schizencephaly, corpus callosum agenesis, calcifications in the damaged periventricular region
vein of Galen malformations, and Dandy–Walker (Fig. 95.1).
malformations. When uncertainty remains after US, or discrepan-
The role of CT is limited to the diagnosis of hem- cy exists between US findings and clinical condition,
orrhages and of calcifications, an important feature of MRI is indicated. The role of MRI in the diagnosis of
some disorders, for example bilateral thalamic necro- post-hypoxic–ischemic conditions in neonates has
sis. In later cases of PVL, CT may be helpful to show become increasingly prominent. This is the result of
95.5 Magnetic Resonance Imaging 725

Fig. 95.2. b Follow-up studies after 3 months (first row, T2- there is loss of white matter in the left hemisphere. Myelina-
weighted; second row T1-weighted) and 18 months (T2-weight- tion is delayed in the left hemisphere
ed,third row) show that the left lateral ventricle is enlarged and

improvement of MR techniques and, not least, to bet- development of faster and new MR sequences. When
ter adaptation of the MR procedure to the needs of the requirements for MRI of neonates are fulfilled, a
preterm or term neonates. These improvements con- protocol for MRI of neonates could include quite a
sist of improvement of monitoring vital signs with gamut of sequences (Table 95.1). With the sequences
MR-compatible equipment, the development of MR- listed in Table 95.1 it is possible to survey the condi-
compatible incubators containing the usual life sup- tion of the neonatal brain tissue and in many cases to
port systems, adapted head coil for neonates, and the predict outcome, which is often of major importance
726 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.3. a A boy born at 34 weeks’ gestation with respirato- images (third row) show the row as bright dots along the ven-
ry insufficiency.The T2-weighted images (first and second rows) tricular borders, representing small hemorrhagic lesions (ar-
show a row of dark dots along the lateral upper borders of the row)
ventricles (arrow) and narrow ventricles. Sagittal T1-weighted

in patient management. In daily routine not all these ence of hemorrhages or calcifications. MRS may con-
sequences will be applied; the first six, however, can tribute to the prediction of outcome of post-hypox-
considerably help in the initial diagnosis. Fast (turbo) ic–ischemic damage in the neonatal period. An ele-
spin echo sequences decrease acquisition time but vated level of lactate, particularly when present in the
have the disadvantage of canceling inhomogeneities basal nuclei of a term neonate, predicts a poor out-
in the local magnetic field and thus masking the pres- come.
95.5 Magnetic Resonance Imaging 727

Fig. 95.3. b Follow-up at the age of 18 months.The T2-weight- volume loss, also predominantly in the peritrigonal area with
ed images show the typical triad of PVL: irregular borders of cortical sulci abutting the ventricles; and periventricular gliosis
the lateral ventricles, especially the trigonum; white matter

MR patterns in post-hypoxic–ischemic encephalo- also has high signal intensity due to the high water
pathy can be subdivided into early and late, residual, content of the premature brain. In this early phase dif-
patterns. fusion-weighted images (Trace images, ADC maps)
Post-hypoxic–ischemic encephalopathy in preterm are helpful. In acute PVL, the affected area has lower
neonates encompasses several conditions. signal intensity on ADC maps and decreased ADC
Venous infarctions, often in the wake of germinal- values. However, it should be noted that the hemor-
layer- (or matrix-)related hemorrhage, are often still rhagic components also influence the ADC due to
referred to as grade 4 hemorrhages. Venous infarc- susceptibility effects. Diffusion tensor imaging and
tions, frequently located along the border of the later- quantification of fractional anisotropy values may
al ventricles, may occur isolated or together with assist in the diagnosis. In the second week of life the
more extensive PVL (Fig. 95.2). changes become more obvious, also on conventional
The most important post-hypoxic–ischemic en- images. ADC values become higher; subependymal
cephalopathy in preterm neonates is PVL. In the first cystic changes appear around the ventricles and will
postnatal week the diagnosis may be difficult. Small often make contact with the ventricles. The most
bright foci around the ventricles on T1-weighted common late pattern of PVL consists of slightly
sagittal or hypointense foci on T2*-weighted trans- widened lateral ventricles with irregular borders; loss
verse images (Figs. 95.3a and 95.4a) are not always of white matter volume, with cortical sulci nearly
followed by cystic changes. More extensive lesions touching the ventricular walls; gliosis in the periven-
usually do (Figs. 95.3b and 95.4b). It is difficult in the tricular area; and focal or diffuse thinning of the cor-
first week to differentiate periventricular white mat- pus callosum (Figs. 95.1, 95.3b, 95.4b, 95.5, and 95.6).
ter changes, with high signal on proton density and The presence of gliosis depends on the gestational age
T2-weighted images, from normal white matter, which at the time of the insult; immaturity of astrocytes be-
728 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.4. a Boy born at 36 weeks’ gestation. In the last rows) show small hemorrhages in the germinal matrix along
trimester there were diminished fetal movements. During de- the ventricular border. The sagittal T1-weighted images (third
livery worsening of the condition occurred with cardiac decel- row) show a number of bright spots along the border of the
erations, necessitating emergency cesarean section. He had lateral ventricles, consistent with hemorrhages
low APGAR scores. The T2-weighted images (first and second

fore 28 weeks’ gestation may lead to less prominent or PVL is not the only manifestation of post-hypox-
absent gliosis. DTI fiber tracking may show the de- ic–ischemic encephalopathy in preterm neonates.
fects in the ascending and descending cortical tracts. Findings at autopsy are enlightening. In 97 autopsies
Severe cases of PVL are often associated with lesions of preterm neonates (Skullerud and Westre 1986), 48
in the thalamus, especially in the pulvinar (Fig. 95.1). had germinal-layer-related hemorrhage, 23 had PVL,
95.5 Magnetic Resonance Imaging 729

Fig. 95.4. b T2-weighted images at 18 months show the triad of PVL. Myelination is delayed

and 57 had pontosubicular necrosis. Of course these praise in the acute phase; the images suggest most
findings are not representative of the surviving group. often diffuse edema. Diffusion-weighted Trace images
The diagnosis pontosubicular necrosis is rarely con- bring out the pattern with high signal intensity, with
sidered in MR reports of preterm neonates and may low ADC values in the affected areas. This pattern
demand more attention. Sagittal proton density and typically involves the perirolandic cortex, the corti-
T2-weighted images may help to demonstrate abnor- cospinal tracts, the dorsal part of the putamen, the
malities in the subiculum. ventrolateral part of the thalamus, the hippocampus,
PVL may occur in term-born neonates, but less and, in the most severe cases, dentate nucleus and
frequently than in preterm neonates. Acute profound brain stem nuclei. The typical late manifestation of
hypoxia–ischemia occurs more frequently in term severe post-hypoxic–ischemic damage in the term-
neonates. Conventional MR images, including con- born neonate consists of gliosis and or atrophy in the
trast-enhanced images, may show diffuse cerebral above-mentioned brain areas (Figs. 95.9 and 95.10).
edema. Because the cerebellum is less susceptible to In MCE the initial pattern seen on MR is general-
hypoxia, it often is not involved. This results in what ized brain edema of the hemispheres, with sparing of
may be called a “white cerebrum” on ADC maps or the cerebellum. This is followed by the development
“black cerebellum” sign on Trace diffusion-weighted of large subcortical cysts that may be spread through
images, the reverse of what was already known on CT all cerebral lobes or confined to the frontal and pari-
as the “white cerebellum” sign (Figs. 95.7 and 95.8). eto-occipital lobes. The cerebellum is least affected
This appearance is even more obvious on coronal dif- (Figs. 95.11 and 95.12).
fusion-weighted Trace images. When the insult is less Aside from the described patterns, other presenta-
severe, another typical pattern develops with lesions tions of post-hypoxic–ischemic encephalopathy are
in the basal ganglia and the central cortico-subcorti- possible. Cortical laminar necrosis (Fig. 95.13) usual-
cal area (Figs. 95.9 and 95.10). On conventional im- ly shows on T1-weighted and FLAIR images as a thin
ages this pattern of damage may be difficult to ap- rim of higher signal intensity within the cortical
730 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.5. A girl was born at a gestational age of 31 weeks with show white matter loss and gliosis restricted to the frontal
perinatal asphyxia and respiratory distress at birth demanding lobes. Frontal localization of PVL is unusual
prolonged artificial ventilation. At 2 years T2-weighted images

layer. On proton density and T2-weighted images the The infarction most often involves the entire middle
cortical ribbon in the affected areas is usually less cerebral territory.A polycystic degeneration of the af-
clearly seen. After contrast injection there may be fected area follows.
enhancement of this rim. On follow-up this will result In our experience border zone infarctions are rela-
in gliosis and atrophy of the affected areas. tively rare, but do occur (Fig. 95.15).
In neonates territorial infarctions are relatively Other lesions caused by intrauterine hypoxic con-
rare, but will be seen occasionally. Often they involve ditions of the neonate include hydranencephaly,
the territory of the middle cerebral artery (Fig. 95.14). porencephaly, and acquired schizencephaly.
95.5 Magnetic Resonance Imaging 731

Fig. 95.6. Images of a 32-year-old mentally retarded and visu- weighted (first and second rows) and FLAIR (third row) images
ally handicapped man with a history of perinatal asphyxia. It is show severe white matter loss and gliosis in the parieto-occip-
unclear whether he also suffered from hypoglycemia. The T2- ital area and crowding of the occipital gyri (ulegyria)
732 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.7. a Images of a female neonate, born at a gestational tion between white and gray matter.There is generalized ede-
age of 41.5 weeks, who suffered from extremely severe perina- ma.The brain stem and cerebellum are the only structures that
tal asphyxia. The MRI was obtained at obtained at 4 days. The seem to have preserved their normal gray–white differentia-
proton density images (first row) show an almost complete tion. The contrast-enhanced T1-weighted images (third row)
loss of white–gray matter demarcation. The basal ganglia show diffuse faint enhancement of the basal ganglia and per-
stand out as white. The T2-weighted images (second row) con- haps the cortex
firm the loss of the cortical ribbon with decreased demarca-
95.5 Magnetic Resonance Imaging 733

Fig. 95.7. b Transverse diffusion-weighted Trace images (first coronal images (third row,Trace diffusion-weighted image left,
row,b = 1000) and ADC maps (second row) show diffuse abnor- and ADC map in the middle) are much easier to interpret. Only
malities in the supratentorial regions, which makes them diffi- the cerebellum has a normal aspect.This “dark” cerebellum on
cult to read: because all structures are abnormal, they do not Trace images is the counterpart of the “white” cerebellum as
stand out as abnormal as compared to normal structures. The seen on CT in severe hypoxia (third row, right)
lower slices show the normal brain stem and cerebellum. The
734 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.8. a
95.5 Magnetic Resonance Imaging 735

Fig. 95.8. (continued). a Images of a term-born male neonate, generalized edema. The FLAIR images (third and fourth row)
victim of a very difficult delivery and repeated attempts at show fuzziness of brain structures. The contrast-enhanced
vacuum extraction.The MRI was obtained at the age of 4 days. T1-weighted images (fifth and sixth rows) show a remarkable
The T2-weighted images (first and second rows) show loss of pattern of enhancement in the rolandic area. It remains diffi-
the cortical ribbon with decreased demarcation between cult to assess the condition of the neonate’s brain
white and gray matter over the large parts of the brain and

Fig. 95.8. b Diffusion-weighted Trace images (first and second


rows, b = 1000) and ADC maps (third and fourth rows) show
involvement of the cortical and subcortical structures of both
hemispheres. Only the cerebellum has a normal aspect.
(Fig. 95.8. b see next page)
736 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.8. b
95.5 Magnetic Resonance Imaging 737

Fig. 95.9. a
738 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.9. b
95.5 Magnetic Resonance Imaging 739

Fig. 95.9. c

Fig. 95.9. a A boy was born at 42 weeks gestational age by and help to establish the prognosis of the child.The ADC maps
emergency cesarean section. There was a severe hypoxic–is- (third and fourth rows) confirm the low ADC values in the
chemic insult. The T2-weighted images (first and second rows) bright areas of the Trace images. c Follow-up MRI at 2 years
show perhaps a less clear cortical ribbon in the parietal region shows the pattern also on conventional images. The T2-
and ill-defined basal ganglia, but the abnormalities are not weighted images (first and second rows) show a gliotic lesion
prominent and the images are difficult to interpret. The con- involving the cortex and subcortical white matter in the
trast-enhanced T1-weighted images (third and fourth rows) perirolandic area, and lesions in the dorsal part of the puta-
confirm the fuzziness together with some enhancement of the men, the ventrolateral part of the thalamus, the substantia
basal ganglia. b Diffusion-weighted Trace images (first and nigra, and the hippocampus on both sides.The pattern is even
second rows, b = 1000) reveal the cortico-subcortical pattern more conspicuous on FLAIR images (third row)
740 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.10. The same pattern of perirolandic cortico-subcortical gliosis and lesions in thalamus, putamen, and dentate nucleus
in a 3-year-old girl who had a history of severe perinatal asphyxia at term birth
95.5 Magnetic Resonance Imaging 741

Fig. 95.11. a A boy was born at 32 weeks gestational age after hemorrhages and ventriculomegaly.There is also blood in the
fetal distress. He had a severe anemia and rhesus antagonism. ventricles. The cerebral white matter is swollen and has an
T1-weighted (first and second rows) and T2-weighted (third row) abnormal signal intensity. (Fig. 95.11. b see next page)
images 1 month after birth show extensive intraparenchymal
742 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.11. b T2-weighted series at the age of 2 months show a multicystic encephalopathy
95.5 Magnetic Resonance Imaging 743

Fig. 95.12. A boy was born at 42 weeks’ gestation. An initially shows a multicystic encephalopathy and ventriculomegaly.
mild hypoxic–ischemic encephalopathy was soon followed by The basal ganglia are also cystic. The brain stem and cerebel-
a catastrophic encephalopathy. This MRI at the age of 2 years lum are spared
744 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.13. a A boy was born at 39 weeks’ gestation with seri- gions and fuzziness of the basal ganglia. Contrast-enhanced
ous perinatal asphyxia and a poor start. MRI was obtained at T1-weighted images (third row) show enhancement of the
3 days. The T2-weighted images (first and second rows) show perirolandic cortex on the right, but no clear other abnormali-
a poorly defined cortical ribbon in the parieto-occipital re- ties
95.5 Magnetic Resonance Imaging 745

Fig. 95.13. b Diffusion-weighted Trace images (first and sec- involved, whereas the parieto-occipital and perirolandic cor-
ond rows, b = 1000) and ADC maps (third row) show a different tex is severely involved. The ADC values in the affected areas
pattern than in previous patients. The basal ganglia are not are low, as is indicated for some spots
746 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.13. c Follow-up MRI was obtained at the age of 14 tracts in the perirolandic area, as is seen in the cortico-subcor-
months.The FLAIR (first and second rows) and T2-weighted im- tical pattern. There are no abnormalities in the basal ganglia.
ages (third row) show gliosis in the cortex and subcortical Additional gliosis is present in the parieto-occipital region
95.5 Magnetic Resonance Imaging 747

Fig. 95.14. A term-born neonate was born by vacuum extrac- Follow-up images after a few years (third row) show the typical
tion. The upper row of conventional T2-weighted and T1- pattern of remains of a perinatal middle cerebral artery infarc-
weighted images without and with contrast show an infarc- tion. The entire right hemisphere is smaller than the left. The
tion in left middle cerebral artery territory, most clear on the middle cerebral artery territory has undergone polycystic
T2-weighted images.The second row shows a diffusion-weight- degeneration, with now only some membranous remains in
ed Trace image (left, b = 1000) and ADC map (right) at the same the area
level, both indicating restricted diffusion in the infarcted area.
748 Chapter 95 Post-Hypoxic–Ischemic Encephalopathy of Neonates

Fig. 95.15. A female infant was born at a gestational age of 35 tic tetraparesis, left more than right. The MRI at 7 years shows
weeks. Soon after birth she started to have epileptic seizures the pattern of extensive border zone infarctions. The gliosis is
and developed microcephaly, developmental delay, and spas- best seen on the FLAIR images (third row)
Chapter 96

Neonatal Hypoglycemia
R.J. Vermeulen, J. Valk

96.1 Clinical and Laboratory Findings ronal damage and histochemical differences. Hypo-
glycemia leads to reduced concentrations of pyruvate
Neonatal hypoglycemia is a condition that frequently and lactate and diminishes the production of protons
occurs in sick newborns. The symptomatology in the by the Krebs cycle, resulting in tissue alkalosis, while
acute stage can range from agitation, somnolence, hypoxia–ischemia is characterized by elevated lactate
and epileptic seizures to status epilepticus and coma. and acidosis. In contrast to hypoxia–ischemia, hypo-
Several groups of neonates are at risk of hypo- glycemia does not lead to pannecrosis, but to selective
glycemia because of a lack of glucose reserves (dys- neuronal necrosis. This neuronal necrosis involves
maturity and prematurity) or an increased need for the cerebral cortex, the hippocampus, the caudate
glucose related to high weight and maternal diabetes. nucleus, and sometimes the spinal cord. In contrast to
The causes of severe neonatal hypoglycemia are hypoxia–ischemia, the brain stem and the cerebellum
diverse and can be separated in three large groups: are spared in hypoglycemia. In addition, the cortical
hyperinsulinism (Beckwith–Wiedemann syndrome, neuronal necrosis is more superficial in hypo-
nesidioblastosis–adenoma spectrum, hyperplasia of glycemia, whereas the middle cortical layers are pref-
the pancreas, leucine sensitivity), endocrine deficien- erentially targeted by hypoxia–ischemia. Axon-spar-
cies (cortisol deficiency, growth hormone deficiency, ing parenchymal lesions with selective dendritic
glucagon deficiency, hypothyroidism, panhypopitu- swellings are often considered a hallmark of hypo-
itarism), and hereditary metabolic defects (defects in glycemic damage. These swollen dendrites contain
carbohydrate, amino acid, organic acid, and fatty acid swollen mitochondria. In the next phase swollen mi-
metabolism). tochondria are seen in the cell body and cell mem-
Long-term outcome of infants with severe neona- brane irregularities appear. Finally, the neurons be-
tal hypoglycemia varies from fatal, through poor with come necrotic, as indicated by mitochondrial floccu-
severe mental retardation, epilepsy, and visual im- lent densities and frank membrane rupture. There is
pairment, to absence of evident neurological conse- a free admixture of amorphous cytoplasm within the
quences. Visual impairment is an important clinical extracellular space, indicating cellular dissolution.
manifestation of neonatal hypoglycemia and results The cytoplasm of the affected cells stains acidophilic
from the preferential damage of the parieto-occipital in light microscopy. The dendritic death of neurons is
white and gray matter. In addition, optic atrophy has characteristic of excitotoxicity.
been described, most probably secondary to the le- Neuropathological observations of damage to the
sions in the parieto-occipital region through the neonatal brain in pure hypoglycemia are limited. In
mechanism of transsynaptic degeneration. the few studies of untreated or inadequately treated
It should be noted that there is no consensus about hypoglycemic neonates, involvement of all parts of
the exact definition of hypoglycemia. It has been the brain and the anterior horns of the spinal cord has
shown that even moderate hypoglycemia is a poten- been observed, with particularly severe lesions in the
tial hazard for the neonatal brain. For instance, glu- posterior parts of the cerebrum, the parieto-occipital
cose levels below 2.6 mmol/l can be associated with lobes. Involvement of arterial border zones, as may be
motor and cognitive impairment. Deterioration of seen in hypoxic–ischemic encephalopathy, is not
motor and cognitive skills is not only related to the seen. As in adults, cortical involvement includes the
depth of the hypoglycemia, but also to the number of superficial cortical layers and not the deeper layers as
days with hypoglycemic episodes. in hypoxia–ischemia. Microscopically the findings in-
clude acute degeneration of nerve cells and glial cells.
In most infants the nerve cells in the cortex are small
96.2 Pathology with abnormal nuclei. There is a regional distribution
of neuronal damage, with the most severe signs of
Most data on hypoglycemic brain damage have been acute degeneration in the occipital cortex and the
obtained in adults. In human adults it is often difficult least signs of involvement in the temporal cortex.
to distinguish hypoglycemic from hypoxic–ischemic Fragmented cells may be demonstrated in the caudate
brain damage on morphological grounds. There nucleus and putamen, the claustrum, and the granu-
are, however, differences in distribution of the neu- lar layer of the cerebellum. Another type of damage is
750 Chapter 96 Neonatal Hypoglycemia

found in large neurons (thalamus, hypothalamus, rate of the brain does not change, in contrast to under
globus pallidus, and Purkinje cells) showing chroma- conditions of hypoxia–ischemia, where the metabolic
tolysis with swelling and granularity of the cyto- rate decreases. A difference between the posterior
plasm. Large vacuoles may be observed in the motor part and the rest of the brain has, however, not been
nuclei of the brain stem. demonstrated.
Low glucose levels lead to a reduction of energy
supply and to protein and lipid catabolism. Because of
96.3 Pathogenetic Considerations the lower glucose levels, levels of lactate and pyruvate
are also reduced. Consequently proton production by
Hypoxia–ischemia and hypoglycemia both lead to the Krebs cycle is reduced, leading to tissue alkalosis,
energy failure, and one would expect similar patterns in contrast to ischemia, which is characterized by ele-
of brain damage. There are, however, major differ- vated levels of pyruvate and lactate and acidosis. In
ences, especially well known from the patterns of hypoglycemia decarboxylation of pyruvate is de-
brain damage following perinatal asphyxia and creased, leading to a shortage of CoA, a major inter-
neonatal hypoglycemia. Only in neonatal hypo- mediate in the pathway to oxaloacetate. Oxaloacetate
glycemia is preferential damage of the parieto-occip- is the a-keto acid in a transamination reaction with
ital region seen. The differences require an explana- glutamate, yielding aspartate and a-ketoglutarate.
tion. Shortage of oxaloacetate subsequently leads to a re-
The immature brains of neonates have the facility duction of tissue glutamate and an increase in aspar-
to use alternative energy donors (for instance lactate tate. This increase in the aspartate/glutamate ratio is
and ketone bodies), an ability that gradually disap- the reverse of what is seen in hypoxic–ischemic con-
pears with age. This and the lesser energy demand of ditions.Aspartate is more selectively active on NMDA
the neonatal brain explain why the immature brain is receptors than glutamate. NMDA antagonists are, at
more resistant to hypoglycemia than more mature least in experimental situations, more effective in hy-
brains. This is probably the reason why hypoglycemic poglycemia than in hypoxia–ischemia. However, no
brain damage rarely occurs in neonates without a pattern of distribution of NMDA receptors is known
predisposing factor. Glucose utilization in newborns that would explain the vulnerability of the posterior
as measured with 2-deoxy-2-[17F]fluoro-D-glucose part of the cerebral hemispheres in neonates.
PET does not demonstrate a higher glucose turnover Delivery of glucose to the brain requires so-called
in the occipital cortical area than in other cortical ar- glucose transporter proteins. Endothelial cells play a
eas. crucial role in the transport of glucose over the blood
Neuronal death by hypoxic–ischemic energy de- -brain barrier since they are equipped with GLUT1
pletion includes instant cell death and delayed cell glucose transporters. In addition, GLUT3, a neuronal
damage and death, the latter mediated by a cascade of glucose transporter, shows a developmental regula-
events including membrane depolarization, calcium tion of expression, at least in the newborn rat. How-
influx into the cytosol, release of proteases and lipas- ever, it has not been reported that this transporter has
es, formation of free radicals, lipid fragmentation and a regional distribution that would explain the selec-
formation of arachidonic acid, and lipid peroxida- tive vulnerability of the parieto-occipital region.
tion. Brain damage in hypoglycemia is not the direct The GLUT5 transporter is selectively expressed in mi-
and immediate result of the lack of glucose, but fol- croglia, whereas the other types of glucose trans-
lows the steps of delayed cell death.
It has been suggested that one of the important
differences between hypoxic–ischemic and hypo-
glycemic conditions concerns the cerebral blood flow. 䊳

The two- to threefold increase in cerebral blood flow Fig. 96.1. A male neonate suffered from severe and repeated
that occurs in patients with hypoglycemia is then as- hypoglycemia. This first MRI was obtained at the age of 16
sumed not to occur in hypoxia–ischemia. This, how- days. The T2-weighted images (first and second rows) show
ever, is not true in all cases of hypoxia–ischemia, blurring of the cortical ribbon in the parieto-occipital and tem-
where an initial rise in cerebral blood flow of the poral areas and swelling of these areas. The signal intensity of
same order may occur, followed by a decrease when the white matter is too high for normal unmyelinated white
generalized hypoxia–ischemia also affects the cardiac matter in these regions, suggesting edema.There is also a high
muscle. The cardiac muscle seems more resistant to signal intensity in the peripheral parts of the cerebellar hemi-
low glucose levels than to hypoxia–ischemia. This dif- spheres, which is extremely unusual in post-hypoxic–ischemic
ference could be important. Correction of the hypo- encephalopathy. The T1-weighted images (third and fourth
glycemia leads to a decrease in cerebral blood flow to rows) show areas of cortical laminar hyperintensity in the
values about 30% below normal. PET studies reveal frontal and parietal areas and loss of gray–white matter dis-
that under hypoglycemic conditions the metabolic tinction in the posterior parieto-occipital and temporal area
96.3 Pathogenetic Considerations 751

Fig. 96.1.
752 Chapter 96 Neonatal Hypoglycemia

Fig. 96.2. Diffusion-weighted imaging was performed in the minent in the parieto-occipital area, but also in both frontal
same boy at 16 days. The Trace diffusion-weighted (b = 1000) and temporal lobes. The ADC maps (second row) confirm the
images (first row) show extensive hyperintensities, most pro- severe restriction in water diffusion in the affected areas

porters (2, 4, and 7) are only expressed at very low glycemia should be avoided because of the risk of
levels in the brain. neurological complications. After correction of the
So none of the factors mentioned above would ex- hypoglycemia, tapering of intravenous glucose infu-
plain either the selective involvement of the parieto- sion should be slow in order to avoid secondary hypo-
occipital gray and white matter in neonates or the glycemia. In addition, treatment of any specific caus-
sparing of the cerebellum and brain stem. es underlying the hypoglycemia, including endocrine
dysfunction and inborn errors of metabolism, should
be instituted as appropriate.
96.4 Therapy

Prevention is the most important form of treatment. 96.5 Magnetic Resonance Imaging
It is essential to identify infants at risk of hypo-
glycemia and to detect the first signs of its occur- The standard protocol used for preterm and term
rence. Special risk factors have been discussed above. neonates, described in Chap. 95, is also appropriate
In the postnatal period maintenance of body temper- for imaging hypoglycemic brain lesions. This proto-
ature, oral feeding within 3–4 h after birth and moni- col includes T1-weighted, proton density, and T2-
toring the clinical signs of hypoglycemia (jitteriness weighted images for morphological and pathomor-
and seizures) are important in the prevention and phological information. Gradient echo refocused im-
early detection of hypoglycemia. Even in neonates ages are used to detect hemorrhagic components and
with only mildly low glucose levels, treatment should calcifications. In addition diffusion-weighted Trace
be initiated, starting with a bolus infusion followed by images and ADC maps should be obtained, to reveal
a continuous glucose infusion. Importantly, hyper- the extent of damage and to discover abnormal
96.5 Magnetic Resonance Imaging 753

Fig. 96.3. This series of FLAIR images, obtained at 21 days in The underlying white matter has become dark, suggesting
the same boy, confirm the extensive cortical involvement. On- white matter degeneration and rarefaction
ly the most anterior part of the frontal cortex seems spared.

areas with a normal appearance on conventional se- dark (Fig. 96.3), compatible with ongoing cortical
quences. Where available, MRS should be part of the necrosis and white matter degeneration and rarefac-
examination. tion. Soon macrocysts develop, the cysts extending
The most constant finding on MRI in early hypo- from cortex to ventricular wall (Fig. 96.4). The cortex
glycemic encephalopathy consists of hyperintense covering these cysts is extremely thin. However, the
and swollen areas on proton density, T2-weighted, and initial hypoglycemic brain lesions are reversible in
FLAIR images, predominantly located in the parieto- some cases or are reversible in part of the brain (com-
occipital lobes (Fig. 96.1). The lesions tend to be more pare Figs. 96.2 and 96.4). In the final stages MRI
or less symmetrical and may involve the splenium of shows tissue loss, especially of the white matter, with
the corpus callosum. T1-weighted images may show crowding of the overlying gyri (ulegyria) in the pari-
linear high-signal changes in the depth of the cortical eto-occipital region. Usually the trigonum and occip-
sulci, in particular in the parieto-occipital areas ital horns of the lateral ventricles are dilated.
(Fig. 96.1). After intravenous gadolinium the latter MRS should show low concentrations of lactate in
abnormalities may become more conspicuous. In the the initial lesions, in contrast to what is seen in peri-
initial phase the affected areas show high signal in- natal asphyxia. MR phosphorus spectroscopy is able
tensity on diffusion-weighted Trace images and ADC to confirm the alkalosis in hypoglycemia versus the
values are decreased by about 25–40% compared to acidosis in hypoxia–ischemia.
normal (Fig. 96.2).Very low ADC values (0.50–0.70) of The differential diagnosis in the early phase of hy-
the affected parieto-occipital lobes suggest irre- poglycemia includes only a few other disease states.
versible cytotoxic edema leading to permanent loss of The most prominent is bilateral occlusion of both
brain tissue. Follow-up FLAIR images show cortical posterior cerebral arteries, as may occur in patients
hyperintensity, whereas the white matter becomes with preceding severe brain edema. The P3 segment
754 Chapter 96 Neonatal Hypoglycemia

Fig. 96.4. This series of T1-weighted images was obtained rhagic. Note that not all areas with severe diffusion restriction
in the same boy at 6 weeks. There is multicystic degeneration underwent the same degeneration
of both parieto-occipital lobes. The lesions are partly hemor-

of the posterior cerebral artery may be compressed ischemic damage is often present in hypoglycemia,
against the tentorial ridge and bilateral posterior ter- which may make the hypoglycemic damage pattern
ritorial infarctions may result. Concurrent hypoxic– less clear.
Chapter 97

Delayed Posthypoxic Leukoencephalopathy

97.1 Clinical Features clinical history, physical findings, and imaging tech-
and Laboratory Investigations niques. The EEG contains diffuse bilateral slow wave
activity with low voltage.
The usual pathological sequels of hypoxia in the CNS
consist of damage to the neurons of the cortex and the
subcortical gray matter structures. Selective injury to 97.2 Pathology
the cerebral white matter as a consequence of hypox-
ia–ischemia occurring in the neonatal period is far The neuropathological findings are highly variable,
less common after hypoxia–ischemia occurring later depending on the severity and nature of the insult
in life. However, as early as 1925 the German patholo- and the time between the event and the pathological
gist Grinker noticed a diffuse symmetrical leukoen- examination of the brain. Here only the neuropatho-
cephalopathy occurring days after carbon monoxide logical findings of DPHL in the more chronic stage of
intoxication. The condition was named after him: the disease are described.
Grinker myelinopathy. As in the original description, The external appearance of the brain is normal or
posthypoxic leukoencephalopathy may occur imme- mildly atrophic. On sectioning, confluent white mat-
diately subsequent to a hypoxic–ischemic insult, but ter lesions are found bilaterally, with a fairly symmet-
usually there is an early phase of improvement after rical distribution. Microscopically, the central white
the initial stage of cerebral injury, followed, several matter of both hemispheres contains areas of diffuse
days or weeks later, by recurrence of impaired con- demyelination with loss of oligodendroglial cells and
sciousness and other neurological signs. In these cas- proliferation of astrocytes. The axons are relatively
es CT and MR show more or less severe leukoen- spared, but areas of extensive necrosis may occur
cephalopathy, and therefore this condition is referred with loss of both myelin and axons. Such necrosis
to as delayed posthypoxic leukoencephalopathy is seen predominantly in arterial end and border
(DPHL). The neurological abnormalities in DPHL zones of the deep white matter, while in the less dis-
vary from patient to patient and include spastic pare- tant arterial end fields of the white matter only
sis of the extremities, a parkinsonian syndrome, demyelination is observed. The arcuate fibers and
choreiform movements, visual failure, myoclonus, white matter underneath the ependyma are better
seizures, psychosis, and mental deterioration. The preserved. Patches of myelin persist around numer-
condition results in a chronic state of global demen- ous vessels.
tia, a vegetative state, or death, but recovery may also The cortex is usually spared, but concomitant areas
occur. of necrosis may be present in the cerebral cortex,
The most common cause of DPHL is intoxication especially in an arterial border zone distribution.
with carbon monoxide or cyanide. This may happen Necrotic areas are often present in the basal ganglia.
accidentally, but also occurs in homicidal or suicidal The brain stem and cerebellum are usually, but not
cases. In the USA not less than 6.4% of suicide at- always, unaffected.
tempts involve carbon monoxide. This figure is prob-
able not far from that in other countries in the west-
ern hemisphere. 97.3 Pathogenetic Considerations
Laboratory tests in the acute phase include, first of
all, determination of blood gases to assess oxygena- The susceptibility of tissues to anoxic–ischemic dam-
tion and acid–base status, since acidosis frequently age depends on the extent of vascular supply, the
accompanies the hypoxia. In cases of suspected car- presence and quality of collateral circulation, the
bon monoxide poisoning, the level of carboxyhemo- metabolic activity, and thus the energy demands of
globin is determined. In carbon monoxide poisoning, the particular tissues. In intoxications the specific
the ECG often shows signs of ischemia with inverted chemical affinity and vulnerability of certain brain
T waves and ST wave depression. If other intoxica- structures also play a prominent role, as in the initial
tions are suspected, specific laboratory estimations of phase of carbon monoxide poisoning. In the brain
the level of toxic agents are necessary. The diagnosis anoxic–ischemic processes most commonly affect the
of DPHL is established at a later stage with the help of cerebral cortex, while the white matter is completely
756 Chapter 97 Delayed Posthypoxic Leukoencephalopathy

or relatively spared. This observation can be ex- oxyhemoglobin dissociation curve to the left, and tis-
plained by the fact that the white matter is metaboli- sue oxygen tension must therefore fall to much lower
cally less active than the cortex. Other explanations levels before the remaining oxyhemoglobin can give
are found in the distribution of excitatory amino acid up its oxygen, a factor aggravating the tissue hypoxia.
synapses and local physicochemical factors at the cel- Moreover, carbon monoxide inhibits cellular respira-
lular level. tion by binding to cytochrome oxidase. In addition to
However, a diffuse injury of the white matter is hypoxia, carbon monoxide often causes general hy-
seen in DPHL. DPHL occurs under circumstances of potension by the formation of carboxymyoglobin in
prolonged hypoxia, hypotension, and metabolic im- the myocardium, which in turn leads to myocardial
balance. The underlying causes comprise respiratory dysfunction. This combination of hypoxia and gener-
failure, cardiac arrest, and systematic hypotension. al circulatory collapse probably explains why DPHL is
Precipitating events are carbon monoxide poisoning, so often seen in carbon monoxide poisoning.
cyanide poisoning, carbon disulfide poisoning, hero- Cyanide may also lead to DPHL. Cyanides have
in overdose, morphine intoxication, anesthetic acci- specific inhibitory effects on the cytochrome oxidase
dent, postoperative shock, and many other events. respiratory enzyme system of cells due to a strong
The white matter lesions of DPHL are located in affinity of cyanides for the iron core of the cyto-
arterial end and border zones. For their arterial blood chromes. In this way cyanides lead to tissue hypoxia
supply, the cerebral cortex and arcuate fibers depend despite the presence of sufficient amounts of oxygen.
on cortical branches of the major cerebral arteries
and their leptomeningeal anastomoses. For their
supply the periventricular and deep white matter 97.4 Therapy
are dependent on penetrating arteries which traverse
the brain from the cortex towards the ventricular Prevention of cerebral hypoxia and ischemia and the
ependyma. These arteries have few collateral branch- prompt restitution of normal oxygenation, blood
es; one artery nourishes one white matter unit. The pressure, and acid–base balance after any hypoxic–is-
basal ganglia receive their supply from end arteries. chemic insult are the only possible measures in the
The deep white matter lesions of DPHL are frequent- prevention and treatment of DPHL. The treatment of
ly accompanied by lesions in the basal ganglia, as well choice for patients with carbon monoxide poisoning
as in border zones of the cerebral cortex. However, is exposure to hyperbaric oxygen in order to wash out
there is no clear correlation between gray and white the carbon monoxide as soon as possible. Cyanide
matter damage, and white matter damage does not poisoning can be treated with hydroxycobalamin and
appear to depend directly on the degree of anoxia. sodium thiosulfate in the acute stage. Adequate treat-
Although some consider the white matter lesions to ment of the acute poisoning may prevent the occur-
be merely a border zone effect, it is probable that rence of DPHL. Once DPHL has developed, the only
something other than hypoxia alone is required for option is to provide supportive care.
lesions of this kind to be produced. There are several
reasons for this assumption. Cerebral DPHL occurs
only rarely, in contrast to the much more frequent 97.5 Magnetic Resonance Imaging
anoxic–ischemic gray matter damage. In addition,
gray matter structures are relatively spared in DPHL, In DPHL, the involvement of the white matter is gen-
which suggests that the hypoxic–ischemic process in erally symmetrical and confluent and located in arte-
itself is not profound as these structures are rather rial border and end zones, due to the underlying sys-
sensitive to lack of oxygen. This leads to the impres- temic cause. In carbon monoxide poisoning, exten-
sion that white matter damage is particularly likely to sive, confluent deep white matter involvement with
occur under conditions of prolonged depression of late occurrences has been reported, but more focal
both oxygenation and circulation. Acidosis may be and asymmetrical white matter involvement has also
another adverse factor in this context. Drug overdose, been observed.
for instance morphine intoxication, leading both to a White matter lesions may be accompanied by le-
depression of respiration and to hypotension, is par- sions in gray matter structures in arterial terminal
ticularly apt to lead to DPHL, much more often than, and border zones of the cortex and in the basal
for instance, a cardiac arrest without antecedent im- ganglia as the remains of the initial event. In carbon
pairment of respiration. monoxide intoxication, the globus pallidus is pre-
Carbon monoxide intoxication relatively frequent- ferentially affected. In cases where the original hy-
ly leads to DPHL. It causes tissue hypoxia by re- poxic–ischemic encephalopathy had another origin,
versibly binding to hemoglobin in red blood cells, other gray and white matter structures may also be
thereby reducing the oxygen-carrying capacity of the involved. There are remarkably few reports on imag-
blood. The presence of carboxyhemoglobin shifts the ing findings in DPHL. The contribution of MRS is
97.5 Magnetic Resonance Imaging 757

Fig. 97.1. A 56-year-old man with


chronic exposure to carbon disulfide.
The T2-weighted images show nearly
symmetrical involvement of the deep
white matter.There is also a moderate
cerebral atrophy.The basal ganglia
and the pons show lesions. From Ku
et al. (2003), with permission and cour-
tesy of Dr C.C. Huang, Department
of Neurology, Chang Gung Memorial
Hospital and University, Taipei, Taiwan

described in few articles. Diffusion-weighted Trace


images and ADC maps have the possible contribution
of distinguishing more recent from older lesions.
758 Chapter 97 Delayed Posthypoxic Leukoencephalopathy

Fig. 97.2. A 6-year-old boy underwent surgery for a congeni- showed signs of spastic tetraplegia. MRI shows extensive
tal cardiac defect. Postoperatively he did well. Four weeks later changes in the white matter, largely sparing the arcuate fibers.
he showed behavioral changes. He suffered two cardiac ar- There are also lesions in the internal capsule,basal ganglia,and
rests,necessitating resuscitation,after which he remained sub- mesencephalon.The cerebellum is not affected
comatose. Neurological recovery was slow and only partial. He
Chapter 98

White Matter Lesions of the Elderly

98.1 Clinical Features and change of water content, with consequently loss
and Laboratory Findings of fractional anisotropy; and that there are decreasing
magnetization transfer ratios, indicating loss of
Age-related changes of the brain have been docu- structural integrity of the brain tissue. MRS reveals a
mented in numerous MRI and histopathological decrease of N-acetylaspartate, reflecting loss of neu-
studies. Changes that are found at histopathological rons and axons. Reduced cerebral perfusion is report-
examination in elderly individuals without neurolog- ed as measured with MR techniques and PET, with
ical or neuropsychological deficits – in other words, unchanged oxygen extraction on PET. Impressive as
in normal aging – include enlargement of the ventri- these findings may seem, they have not enabled defi-
cles and the cerebral sulci reflecting gray and white nition of clear cut-off points between successful ag-
matter loss, decrease in neurons and synapses, and in- ing, mild cognitive impairment, and progression to-
crease in lipofuscin and mineral deposits in brain wards multi-infarct dementia in the individual case.
structures. On MRI white matter hyperintensities
may appear on T2-weighted and FLAIR images, in-
creasing with age, varying from multifocal spots in 98.2 Pathology
the deep white matter or a pencil-stripe-like rim
around the ventricles to a confluent, bilateral, more or Reports on the pathological substrate of leukoaraio-
less symmetrical, periventricular leukoencephalopa- sis on MRI, either in normal aging or in patients with
thy. The more or less extensive signal changes of the different degrees of cognitive decline, show a wide
periventricular and deep white matter have been giv- variety of findings. This is not surprising, given the
en the descriptive name “leukoaraiosis” (white matter bias in the examined populations, which most often
rarefaction) by Hachinski et al. (1987), and have been involve only a small number of patients. In nearly all
the subject of many studies since then. Attempts have cases gross anatomy shows that the cerebral cortex
been made to document these changes more precise- appears normal or shows only minor changes. The
ly with a variety of MR techniques, to search for neu- white matter is either macroscopically normal or
ropathological correlates, and to find out which white shows a grayish discoloration, with a rubbery consis-
matter signal alterations represent apparently inno- tency. White matter volume may be reduced. White
cent changes by clinical standards, and which signal matter abnormalities may be focal, isolated, or more
alterations indicate future clinical progress towards confluent. The basal ganglia do not, as a rule, show
cognitive impairment and eventually vascular de- changes. At microscopy, there are variable degrees of
mentia. Similar attempts have been made from the ependymal denudation and frontal and peritrigonal
neuropsychological point of view, where the search is white matter changes, consisting of loosely struc-
still going on to define “age-related memory disor- tured tissue with widened Virchow–Robin spaces.
der,” “age-associated mental impairment,” “benign The periventricular changes vary from white matter
senescent forgetfulness,” and “mild cognitive impair- pallor (meaning less intensive myelin staining, the
ment” (with decliners and nondecliners) in such a earliest change in the periventricular white matter,
way that homogeneous subgroups can be identified without overt demyelination, axonal loss, presence of
for further studies. Again an important aim is predic- foamy macrophages, or gliosis) to periventricular de-
tion of outcome. The combined efforts of all disci- myelination (describing overt myelin loss with astro-
plines involved so far have not come up with a final cytic proliferation and in some cases some inflamma-
answer. tory reaction). The lesions may be patchy or more
Much was expected of quantitative MR techniques. confluent. In more severe cases the periventricular
These techniques can more precisely indicate that and deep white matter changes are continuous. The
with age more white matter than gray matter is lost; cause of these changes is to be found in vessel wall
that increases in relaxation times are more pro- abnormalities. These abnormalities range from non-
nounced in white matter than gray matter; that there specific intima thickening without narrowing of the
is an increase of apparent diffusion coefficients in lumen, to changes in the smooth muscle walls of the
both white and gray matter, but more so in white than small vessels leading to gradual narrowing or occlu-
in gray matter, indicating more free water movement sion of the small vessels.
760 Chapter 98 White Matter Lesions of the Elderly

There is no sharp transition between these rela- ogy and the influence of lesions on neuropsychologi-
tively mild changes and the pattern seen in vascular cal functioning.
dementia patients. In patients with vascular demen- The distribution of the lesions may also be ex-
tia, there are also lacunar infarctions, involvement of plained by the unique blood supply to the periven-
the corpus callosum, involvement of the basal ganglia tricular and deep white mater. Arteries originating in
(the latter often showing an état criblé or état lacu- the arachnoid spaces perforate the brain tissue, run-
naire or both), abnormalities of the transverse fibers ning from the cortical surface towards the periven-
of the pons, and coagulation necrosis and cavitation tricular white matter. Although De Reuck’s hypothe-
of the deep white matter. sis (1971) of subependymal border zones has been
challenged, it remains true that there is a scarcity of
anastomoses of the large perforating arteries in these
98.3 Pathogenetic Considerations areas, so that in fact one vessel irrigates only one
metabolic unit. Recurrent transient drops in cerebral
There are large differences in the estimation of the blood flow can lead to ischemia in these units, often
presumed clinical significance of white matter abnor- referred to as incomplete infarction. These changes
malities in the elderly, and there is a great variability may progress in some patients, explaining the whole
in reported histopathological correlates. The most range of abnormalities found in histopathology, from
prevalent morphological substrate is perivascular white matter sponginess, pallor, patchy demyelina-
tissue change. Reasons for the perivascular tissue tion, astrocytic proliferation, to more serious changes
changes are supposed to be the thickening of vessel such as coagulative necrosis and cavitation. In this
walls (by a number of different causes), damage to view leukoaraiosis is the expression of a diffuse cere-
the surrounding tissue by a waterhammer effect of bral hypoperfusion of variable severity, in its mildest
pulsating arterioles with diminished elasticity, and form showing changes in the white matter on MRI
perivascular edema resulting from leakage of the without pertinent pathological findings other than
blood–brain barrier. In a high percentage of cases possibly death of oligodendrocytes, and without neu-
hypertension plays a prominent role, and it is cer- rological and/or neuropsychological deficits, and in
tainly a major factor in changing the vessel walls by its most severe form bearing the characteristics of
lipohyalinosis and consequently narrowing of the subcortical arteriosclerotic encephalopathy.
lumina of small brain vessels. Several other disorders Focusing on white matter abnormalities in the
lead to the same result, despite the different material elderly, population-based studies have tried to identi-
deposited in the vessel walls, with amyloid angio- fy risk factors for both the occurrence of white matter
pathy, arteriosclerosis, and CADASIL as examples. hyperintensities on T2-weighted images, and factors
Some factors must be responsible for the selective predicting the long-term outcome of members of
involvement of the periventricular and deep white these populations. Not surprisingly, these factors
matter as a common finding. Several factors have include higher age, higher ischemic score on the
been suggested. Hachinski scale (0–18), history of stroke, lacunar in-
Chronic hypoperfusion may have a selective influ- farction on MRI, hypertension (systolic >140 mmHg;
ence on oligodendrocytes and myelinated axons, as diastolic >95 mmHg), male gender, atrial fibrillation,
was supported by animal experimentation. Experi- coronary artery infarcts, diabetes mellitus, smoking,
mental studies of brain ischemia on rat and gerbil alcohol abuse, drug abuse, hyperhomocysteinemia,
brains show that both oligodendrocytes and myeli- antiphospholipid antibodies, several coagulation dis-
nated axons are highly vulnerable to ischemia and orders, the presence of the apolipoprotein E e4 allele,
that chronic cerebral hypoperfusion leads to progres- and probably other genetic factors. A study involving
sive rarefaction and glial activation in the white mat- psychiatric patients showed periventricular and deep
ter. Three hours after carotid occlusion in rats, oligo- white matter abnormalities in a high percentage of
dendrocytes display characteristics of irreversible patients with major depression. Important as these
injury, such as pyknosis and plasma membrane rup-
ture. Three factors, in these experiments, led to vac-
uolation of the white matter: intramyelinic fluid accu-
mulation, enlarged extracellular spaces, and swelling 䊳

of astrocytic processes. These changes precede irre- Fig. 98.1. a FLAIR (upper two rows) and T2-weighted (lower
versible neuronal injury, indicating a time-indepen- two rows) images of a 64-year-old woman with “benign senes-
dency of these processes and partly explaining the cent memory impairment.” The images show hyperintense
selective vulnerability of white matter in chronic changes in the deep and periventricular white matter and
hypoperfusion. Animal models, however, cannot easi- basal ganglia. The FLAIR images show the abnormalities with
ly be designed to mimic the age-related changes in the greater conspicuity. Clinical and MRI follow-up over the course
human brain and explain the various kinds of pathol- of 4 years did not show any progression (nondecliner)
98.3 Pathogenetic Considerations 761

Fig. 98.1. a
762 Chapter 98 White Matter Lesions of the Elderly

Fig. 98.1. b The gradient echo images


of the same 64-year-old woman show
hemosiderin deposits, residues of
microhemorrhages, in the basal
ganglia, more prominent on the left
side, and around the anterior commis-
sure. CT scan showed no calcifications

studies are to establish rules for population-based present. The lesions seen on MRI in older individuals
prevention, in individual cases they offer only guide- can be graded as follows:
lines. ∑ Frontal and occipital caps: white matter hyperin-
The overall impression one may develop is that tensity around the frontal horns and the triangu-
leukoaraiosis is too general a concept, so that exam- lar area of the ventricles
ined populations and selected cases do not form a ho- ∑ A periventricular 1- to 3-mm-thick rim of high
mogeneous entity. Certainly some general rules for signal intensity, best seen on proton density or
prevention have been drawn from the research results FLAIR images
obtained so far, but future research should aim at the (These two findings are considered to be without
definition of more homogeneous subgroups to obtain clinical significance and represent areas of looser
better insight in the pathophysiological mechanisms tissue and widened Virchow–Robin spaces)
underlying the final common visual product leuko- ∑ Patchy deep white matter lesions, partly isolated,
araiosis. partly confluent
∑ Confluent deep white matter hyperintensity, con-
tinuous with periventricular white matter changes
98.4 Therapy ∑ One or a few lacunar infarctions within the affect-
ed deep white matter
The development of leukoaraiosis is strongly related
to age, and secondarily to risk factors. As they get MR has been used extensively to study the process of
older, nearly 100% of people will have white matter aging of brain structures in vivo. Pathological studies
lesions. At the moment of detection of white matter of the brain depict the terminal phases of disease on-
lesions, whether related to neurological or neuropsy- ly and are limited by the relatively small number of
chological complaints or as an incidental finding, risk samples that can be examined per patient, as a rule in
factors should be searched for, where possible treated, the order of 350–450 at most. In contrast, MR data
and habits such as smoking, excessive drinking, and about normal aging are abundant and can be used
lack of exercise should be changed. The most impor- as reference data for MR studies in older patients,
tant way to take action is by promoting preventive for example to provide normal values per age group
measures that would improve the lifestyle and health of ventricular and sulcal width, hippocampal and
of the general population. temporal lobe volume, magnetization transfer ratios
and histograms, fractional anisotropy, apparent dif-
fusion coefficients, T1 and T2 relaxation times, region-
98.5 Magnetic Resonance Imaging al cerebral blood volume, and metabolite concentra-
tions.
With the introduction of CT it became clear that there General experience is that over the age of 50 years
were age-related changes of the brain, amongst them one may expect to see white matter abnormalities in
more or less extensive periventricular areas of hypo- patients and controls, increasing exponentially with
density. MRI showed these changes to better advan- age (Figs. 98.1 and 98.2). Population-based studies
tage as hyperintense changes on T2-weighted images, have linked these white matter abnormalities to vari-
and later even better on FLAIR images. Many scales ous risk factors, and also tried to find predictive fac-
were developed to grade these signal abnormalities tors that would indicate risks for future strokes and
and to relate them to neurofunctional deficits, when cognitive decline. The problem with these studies,
98.5 Magnetic Resonance Imaging 763

Fig. 98.2. FLAIR images of a 71-year-old man presenting with infarctions are noted.The Trace diffusion-weighted image and
mild apraxia of the left arm and no cognitive impairment. In ADC map (third row, middle and right) show a small recent
the periventricular white matter hyperintensities a few lacunar infarction with low ADC values in the right periventricular area

especially those that include large populations, is hemorrhages (Fig. 98.1) and thus have identified a
that only basic MR techniques have been used – T1- group with possibly a different pathogenesis, other
and T2-weighted images – without employment of risk factors, and different prognosis. Other MR se-
techniques that allow a better definition of tissue quences would possibly have led to further differenti-
characteristics. The inclusion, for example, of gradi- ation, for example, on the basis of the estimation of
ent echo refocusing pulse sequences would have tissue integrity with magnetization transfer ratios, or
shown the members of these populations with micro- on the basis of the change of brain metabolites in
764 Chapter 98 White Matter Lesions of the Elderly

Fig. 98.3.
98.5 Magnetic Resonance Imaging 765

Fig. 98.3. (continued). A 62-year-old man without cognitive area, and a single spot in the right parietal area.The fourth row
complaints, presenting with left-sided hemianopia. The FLAIR shows Trace diffusion-weighted images with b = 1000; the fifth
images (upper two rows) show hyperintensities consistent with row shows ADC maps. The left parietal lesion is bright on the
normal aging, except for the left parietal lesion, involving cor- Trace images and has ADC values as low as 0.52 ¥ 10–3 mm2/s,
tex and subcortical white matter. The gradient echo images confirming the presence of a fresh infarction
(third row) show a few low-intensity spots in the left parietal

MRS, including the presence of lactate in the lesions. information about neuronal and axonal loss, and
Probably the most important MR technique to relate about the presence of lactate, indicating anaerobic
the white matter abnormalities to clinical findings is glycolysis.
perfusion imaging. This allows not only the estima- In many patients follow-up studies will be re-
tion of the degree of hypoperfusion in the involved quired. Postprocessing of data is then also very im-
areas, but also the reserve capacity of the tissue, giv- portant, to obtain adequate comparable information.
ing a quantitative measure of the severity of the white This will give a clue with respect to the rate of pro-
matter damage. gression, and information about the efficacy of thera-
In daily routine one will be confronted mainly with peutic measures and changes in lifestyle (Fig. 98.4).
patients referred for memory disorders and cognitive Differential diagnosis is important, because many
decline. Apart from ruling out rare causes such as other disorders may present with deep white matter
brain tumors, chronic subdural hematomas, and so abnormalities. Disorders with multifocal and partial-
on, a protocol should be used that will enable assess- ly confluent white matter abnormalities, often accom-
ment of most of the factors responsible for cognitive panied by cognitive impairment, are amyloid an-
decline according to present knowledge. This means giopathy, CADASIL, multiple sclerosis, cerebral vas-
an inventory of cerebral and cerebellar cortical and culitis, systemic lupus erythematosus, chronic expo-
deep gray matter structures, including the hippocam- sure to organic solvents (housepainter’s dementia),
pus and temporal lobe structures, of white matter ab- and several infections including HIV encephalopathy
normalities, and of the presence of microhemor- and progressive multifocal leukoencephalopathy. In
rhages (Fig. 98.1). In patients with rapidly progressive Alzheimer disease, in particular in the late-onset
cognitive decline diffusion-weighted imaging with forms, white matter abnormalities are common.
ADC maps should be added to the program (Figs. 98.2 Disproportional hippocampal atrophy suggests Alz-
and 98.3). In centers dedicated to the research of mild heimer disease. It is important to realize that more
cognitive impairment, or where vascular factors seem and more “mixed” dementias are being recognized,
to be important, MR angiography should be included. caused by a combination of vascular and neurode-
MRS and chemical shift imaging may give additional generative factors.
766 Chapter 98 White Matter Lesions of the Elderly

Fig. 98.4. FLAIR images of a 67-year-old woman with mild Also clinically there was clear progression of the cognitive
cognitive impairment (first row).There are white matter hyper- disorder. ADC values in this patient in the centrum semiovale
intensities in the deep and periventricular white matter and had gone up from 0.85–0.95 ¥ 10–3 mm2/s to 1.20–1.25 ¥
corpus callosum, and scattered small lesions in the basal 10–3 mm2/s; fractional anisotropy values changed from 0.450
ganglia. Images of the same patient 4 years later (second to 0.224, indicating loss of structural integrity. MRS showed
row) show progression of the abnormalities, with lacunar in- lactate in the lesions in the last examination. All evidence is
farctions in both hemispheres and the brain stem (not shown). that this patient is a decliner
Chapter 99

Subcortical Arteriosclerotic Encephalopathy

99.1 Clinical Features 99.2 Pathology


and Laboratory Investigations
Externally the brain in patients with SAE appears
This disease was first described by Binswanger in normal. The brain weight is average. In most cases the
1894 as encephalitis subcorticalis chronica progressi- arteries at the base of the brain show moderate to se-
va, later renamed by Olzewski (1962) as subcortical vere arteriosclerosis. The lateral ventricles are mod-
arteriosclerotic encephalopathy (SAE). It was be- erately to highly enlarged. In advanced cases the cere-
lieved for a long time that the disease was very rare. bral hemispheric white matter appears wrinkled,
Only a few documented cases appeared in the litera- firm, rubbery, and discolored gray or yellow, especial-
ture before the advent of CT and MRI. CT and, even ly in the periventricular, frontal, and parietal regions.
more, MRI have made it clear that white matter ab- Histopathologically, the white matter lesions consist
normalities are frequent in the brains of older pa- of partial loss of myelin sheaths, oligodendroglial
tients. This has opened a discussion that is still going cells, and axons, producing a decrease of the mesh-
on about the diagnostic criteria for the disease. These work density of white matter tissue, along with mild
criteria comprise: reactive fibrillary gliosis and sparse macrophages.Ar-
∑ A vascular risk factor or evidence of systemic vas- teriosclerosis is invariably present in these areas of
cular disease incomplete infarction, and there is severe stenosis of
∑ Evidence of focal cerebral disease the smallest vessels by fibrohyalinose material. The U
∑ Evidence of subcortical dysfunction fibers are usually spared. The temporal lobes, too, in
∑ Bilateral deep white matter abnormalities on CT contrast to the findings in CADASIL, are uninvolved.
or MRI État criblé, widening of the Virchow–Robin spaces, is
∑ Absence of multiple or bilateral cortical lesions on an almost constant feature. État criblé results from
CT or MRI spiraled elongation of penetrating arteries and arteri-
∑ Absence of severe dementia (mini mental scale oles in white matter and central gray matter nuclei.
>10) Blood vessels in areas of état criblé are thickened,
ectatic, and have sclerotic walls. The perivascular tis-
The age of onset is between 40 and 60 years and most sue shows reactive astrocytosis and isomorphic glio-
patients have a history of chronic hypertension, often sis with glial fibers extending along degenerated ax-
poorly controlled, and often one or more vascular in- ons. There is perivascular leakage of serum proteins.
cidents. The patients usually present with a slowly de- Multiple lacunar infarctions, état lacunaire, are also a
veloping subcortical frontal dysfunction with loss of frequent finding. Lacunes are small cavitary lesions
interest, lack of drive, alternations in mood and per- that result from ischemic strokes due to occlusion of
sonality, loss of appropriate social conduct and lack of penetrating cerebral arterioles. Lacunes predominate
judgment, parkinsonian gait disturbances, urinary in the basal ganglia, internal capsule, pons, and cen-
incontinence, and pseudobulbar palsy. Minor strokes trum semiovale. Fresh lacunes show necrosis and liq-
may occur with a variable degree of improvement of uefaction, followed by absorption of necrotic materi-
the neurological deficits. Gradually the dementia pro- al by fatty macrophages. In the chronic stage an irreg-
gresses and more neurological symptoms become ular cavity remains, whose walls show dense fibrillary
manifest, including dysarthria, clumsiness, distur- connective tissue and gliosis. Reabsorption of minute
bances of gait, ataxia and apraxia, and finally pro- hemorrhages may also result in lacunes leaving he-
found global dementia. It may be difficult to distin- mosiderin-filled macrophages in the walls and in the
guish SAE on clinical grounds from Alzheimer dis- vicinity of the lesions.
ease and other vascular dementias.
There are no specific laboratory markers for the
disease. Discussion has been going on for a long time 99.3 Pathogenetic Considerations
whether or not SAE is a disease entity, but after the re-
views of Fisher (1989) and Caplan (1995) there seem The main characteristic of SAE is arteriosclerosis
to be sufficient arguments to do so. with narrowing and occlusion of the deep perforating
cerebral arteries and their branches. These arteries
768 Chapter 99 Subcortical Arteriosclerotic Encephalopathy

and arterioles are end vessels without collateral circu- ication. The robust evidence for the effectiveness of
lation. They form an arterial end and border zone in cholinergic treatments in Alzheimer disease, and the
the periventricular region. The cortex and subcortical frequent occurrence of mixed dementias, has led to
white matter are within the territory of supply of the the testing of galantamine (Reminyl) in a group of pa-
cortical vessels and their leptomeningeal anasto- tients with SAE. Galantamine has a dual cholinergic
moses. mode of action, and reduces behavioral symptoms in
SAE is characterized by microinfarctions, focal or Alzheimer disease. This is the result of its potential to
diffuse demyelination, and gliosis of the periventric- modulate systems involving other neurotransmitters
ular and deep white matter, in combination with lacu- such as serotonin and dopamine, which affect mood
nar infarctions in the basal ganglia and brain stem. It and emotional balance.Application of galantamine in
is commonly accepted that SAE type encephalopathy SAE patients led to significant cognitive improve-
is caused by ischemia in the distal watershed territo- ment over 6 months, long-term maintenance of cog-
ries described above. The ischemia is the combined nition for at least 12 months, and improvement of
effect of arteriosclerosis and decreased brain perfu- both behavioral and functional symptoms. These da-
sion from hypotension or low cardiac output. Elonga- ta suggest a result that is, so far, unsurpassed by other
tion of the medullary arteries with dilatation of the drugs or treatment regimens.
perivascular spaces leads to an état criblé.
There are numerous risk factors underlying these
changes, such as age, hypertension, hypotension, 99.5 Magnetic Resonance Imaging
smoking, inadequate diet, and diabetes mellitus.
A number of genetic factors may also play a role. In patients with presenile dementia and a history of
Hyperhomocysteinemia and hyperlipidemia have hypertension the diagnosis SAE depends on two pa-
been identified as important risk factors. Effects of rameters: the clinical establishment of a subcortical
sustained daytime hypertension, hypertensive crises, type of dementia and the establishment of diffuse
and the absence of a normal nocturnal dip in blood damage to periventricular and deep white matter by
pressure are considered to be particularly damaging. means of an imaging modality, CT or MRI. In uncom-
Another factor that may play a role in the develop- plicated cases of SAE, CT and MRI identify a relative-
ment and progression of SAE is coagulation activa- ly well preserved cortex, moderately to more serious-
tion, leading to a hypercoagulable state. In a selected ly enlarged ventricles, and a broad area of reduced
group of SAE patients, fibrinogen, thrombin–an- density around the ventricles on CT or, on MRI, a
tithrombin complex, prothrombin fragment1+2, and zone of high signal intensity on T2-weighted and
cross-linked D-dimer were found to be significantly FLAIR images, involving the periventricular and deep
increased. Obstructive sleep apnea, which produces white matter (Figs. 99.1–99.3). The more central le-
increased platelet activation, higher epinephrine sions, closest to the ventricles, are usually confluent;
levels with vasoconstrictive effect, and higher blood farther away from the center there are often many iso-
pressure, also plays a role. lated lesions of different size (Figs. 99.1 and 99.3).
There is little doubt that the changes caused by Within the confluent areas there may be spots with
these factors are responsible for the abnormalities still higher signal intensity on T2-weighted images,
seen on MRI. That these MRI changes do not neces- possibly representing infarctions. In some cases small
sarily have the same histopathological background is cavities are present with low signal intensity on
illustrated by the observation that some patients with FLAIR images, often with a bright rim, as residues of
severe periventricular abnormalities on MRI have a lacunar infarctions (Fig. 99.3). In the early phases of
mild clinical presentation, whereas other patients the disease the U fibers are usually spared (Fig. 99.1).
with much less severe MRI abnormalities have ad- In cases of longer standing white matter disorder the
vanced clinical symptoms. subcortical fibers may also be partially involved
(Figs. 99.2 and 99.3). In some cases cortical infarc-
tions are also seen. Apart from the periventricular
99.4 Therapy and deep white matter lesions there are often isolated
hyperintense lesions scattered throughout the basal
There is no cure for SAE. Whenever hypertension is ganglia, the pons, and midbrain, representing small
present, treating it may slow down the progress of dis- infarctions (Figs. 99.1 and 99.2). Widened Virchow–
ease. Other risk factors such as hyperlipidemia and Robin spaces and lacunar infarctions are best seen on
hyperhomocysteinemia should be treated, and bad FLAIR images (Fig. 99.3). The corpus callosum is usu-
habits such as smoking and excessive drinking ally less affected than in multiple sclerosis, but this is
should be given up. There is some evidence that in an a rule with many exceptions. Sagittal FLAIR images
early stage the disease can still be influenced by these will often show involvement of the under layer and
measures. Some success has been booked with med- thinning of the corpus callosum (Fig. 99.3).
99.5 Magnetic Resonance Imaging 769

Fig. 99.1. FLAIR images of a 56-year-old woman. The patient ment of the external capsule and the dark appearance of the
had the antecedents of SAE with longstanding hypertension globus pallidus, but the temporal lobes are not affected. Fami-
and multiple transient ischemic attacks. Her images show ly history and tests for CADASIL were negative
some of the characteristics seen in CADASIL, with the involve-

Standard MRI series are fully capable of displaying in N-acetylaspartate and, in some cases, presence of
most of the cerebral abnormalities. Fast imaging se- some lactate.
quences can often be used to advantage. FLAIR is very The differential diagnosis includes in the first
useful to show older lacunar infarctions and enlarged place periventricular and deep white matter changes
perivascular spaces within the affected white matter which occur in normal elderly people with varying
and the basal ganglia as spots with low signal intensi- severity. The differentiation from normal pressure
ty, and to show the demarcation of the ventricles from hydrocephalus may be difficult when the ventricles
the periventricular abnormalities. FLAIR is also use- are greatly enlarged. Additionally, the combination of
ful when estimation of lesion load is required in re- SAE and normal pressure hydrocephalus seems very
search programs. Gradient refocused images should possible. Stiffening of the walls of the ventricles in
be standard in a MR protocol for dementing illnesses SAE may even play a role in the pathogenesis of nor-
in the older age group, because they will show spots mal pressure hydrocephalus. Other vascular diseases
with high magnetic susceptibility, representing he- such as amyloid angiopathy and CADASIL have to be
mosiderin deposits in microhemorrhages. They are considered. Unlike CADASIL, patients with SAE do
found in a high percentage of patients with SAE and not have prominent abnormalities in the external and
in many other vascular disorders. Diffusion-weighted extreme capsules and the anterior temporal lobes,
imaging may be helpful in identifying fresh infarc- although the images may show some similarities
tions, which will otherwise be lost in the brightness of (Fig. 99.1). Rarer are vasculitides, Fabry disease, Lyme
the lesions that probably represent old infarctions, disease, HIV infection, leukoencephalopathy after
myelin loss, and gliosis. MRS usually shows decrease chemotherapy or radiotherapy, and toxic encephalo-
770 Chapter 99 Subcortical Arteriosclerotic Encephalopathy

Fig. 99.2. A 58-year-old man with progressive subcortical de- fibers. Small lesions are seen in the left cerebellar hemisphere.
mentia and transient ischemic attacks. The T2-weighted series Diffuse hyperintensities are present in the pons. There are
shows a periventricular leukoencephalopathy extending into small, punctate lesions in the basal ganglia and in the corpus
the centrum semiovale, in some areas also involving the U callosum

Fig. 99.3. A 64-year-old man with more advanced dementia visualized. The sagittal FLAIR images (third and fourth rows)
and a history of hypertension and transient ischemic attacks. show more clearly the involvement of the corpus callosum
The axial FLAIR images (first and second rows) show ventricu- and the fornix, both thinned and partly hyperintense.The état
lomegaly, periventricular leukoencephalopathy, and promi- criblé of the basal ganglia is beautifully seen
nent white matter atrophy. État criblé of the basal ganglia is
99.5 Magnetic Resonance Imaging 771

Fig. 99.3.
772 Chapter 99 Subcortical Arteriosclerotic Encephalopathy

pathies. In general, vascular diseases share a pattern and often also brain stem lesions. The presence of
of irregularly confluent periventricular white matter small lacunes adds to the suspicion of a vascular dis-
abnormalities with often multiple isolated lesions at order.
the periphery, multifocal lesions in the basal ganglia,
Chapter 100

Vasculitis

100.1 Introduction To organize the discussion we suggest the follow-


ing classification:
The term “vasculitis” refers to inflammation of the A subclassification that may be helpful in reaching
vessel walls. There are many causes of vasculitis, but a diagnosis is division according to the size of vessels
they result in only a few histological patterns of vas- affected by the disorder, even though there is a con-
cular inflammation, of which necrotizing vasculitis siderable overlap. Takayasu disease and giant cell ar-
and granulomatous reaction are the most prominent. teritis affect large vessels; primary angiitis of the CNS
Because vessels of any type in any organ can be affect- and polyarteritis nodosa affect medium-sized vessels;
ed, there are a wide variety of clinical manifestations. Wegener granulomatosis, Churg–Strauss syndrome,
This, combined with etiological nonspecificity of the rheumatoid disorders, SLE, microscopic polyarteritis,
histological lesions, complicates the diagnosis of scleroderma, and many other vasculitic disorders in-
vasculitis. The past few decades have seen important volve small vessels: arterioles, capillaries, and
progress in this field, enabling a laboratory diagnosis venules. Behçet disease involves all types and sizes of
in some cases. A good example is the discovery vessels, arterial or venous (Fig. 100.1).
in 1982 of the anti-neutrophil cytoplasm antibodies
(ANCAs), subdivided into a perinuclear form
(pANCA), related to Churg–Strauss syndrome and 100.2 Clinical Presentation
microscopic polyarteritis, and a cytoplasmic form and Laboratory Findings
(cANCA), related to Wegener disease. The blood tests
for these antibodies are now widely available, but they As in all relapsing multifocal disorders that may affect
need careful application and interpretation. Other both gray and white matter, the clinical manifesta-
findings, such as the detection of anti-neuronal anti- tions of the disease depend on the localization of the
bodies, anti-centromere antibodies, and anti-phos- abnormalities, progression over time, and the nature
pholipid antibodies (APLA), have shed some light on of the disease. Symptoms may be neurological or psy-
the pathophysiology of a number of disorders and chiatric or both, and cognitive deterioration may be
have demonstrated a link between systemic lupus the leading feature. When the cerebral manifestations
erythematosus (SLE) and Sneddon disease. Many are part of a systemic disorder the diagnosis may be
more autoantibodies have been identified and they easier, unless the cerebral symptoms are the first or
may help in better understanding the complex only manifestation.
pathology underlying these disorders. Despite these Clinically there are many factors to consider: age,
discoveries the diagnosis of vasculitis remains often gender, presence of skin lesions, involvement of other
difficult, in particular in the primary vasculitides of organs (in particular kidneys, lungs, and paranasal si-
the CNS. nuses), medication, drug abuse, and neurological

Fig. 100.1. Different vasculitic disorders may


preferentially attack blood vessels of a particular
size. AS, arteriosclerosis; cAA, cerebral amyloid
angiopathy; CADASIL, cerebral autosomal
dominant arteriopathy with subcortical infarcts
and leukoencephalopathy; PACNS, primary
angiitis of the CNS; PAN, polyarteritis nodosa;
MPAN, micropolyarteritis nodosa; SLE, systemic
lupus erythematosus
774 Chapter 100 Vasculitis

Table 100.1. Classification of CNS vasculitis (including nonvas-


culitic disorders that may present with similar pathology to
some of the vasculitides)
Primary vasculitis of the CNS
Granulomatous angiitis of the CNS
(primary angiitis of the CNS)
Giant cell arteritis (arteritis temporalis)
(Benign primary angiitis of the CNS)
Secondary vasculitis of the CNS
Systemic vessel wall disease
Takayasu arteritis
Polyarteritis nodosa
Moyamoya syndrome
Autoimmune-mediated disorders
Rheumatoid arteritis
Systemic lupus erythematosusa (SLE)
Sneddon diseasea
Anti-phospholipid antibody (APLA) syndrome
Sjögren disease
Scleroderma
Neurosarcoidosis
Wegener granulomatosisb Fig. 100.2. Intra-arterial DSA of the right internal carotid
Churg–Strauss syndromeb artery in a 37-year-old woman with biopsy-proven granulo-
Microscopic polyarteritisb
matous angiitis of the CNS, showing the irregular size of the lu-
Behçet disease
Infectious vasculitis men of the posterior temporal artery. Other vessels also show
Borreliosis (Lyme disease) irregular borders
Lues
Tuberculosis
Varicella-zoster
Herpes zoster ophthalmicus (delayed hemiplegia)
Drug-related vasculitis
Aminopenicillins Ergot alkaloids es, for example, in moyamoya syndrome, Takayasu
Allopurinol Interleukin-2 disease, and infectious vasculitis. In quite a number of
Retinoids Methylphenidate cases a leptomeningeal or brain biopsy will still be re-
Quinolones Penicillin
Cocaine, heroin, amphetamines, ecstasy quired, especially in primary angiitis of the CNS, to
ascertain the diagnosis. However, this is not full proof
Disorders primarily obstructing the vascular lumenc
Disorders of coagulation either.
Intravascular lymphomatosis The most important features and data of the differ-
Sickle cell disease ent forms of vasculitis will be briefly reviewed.
a
APLA-related disorder.
b
ANCA-related disorder.
c
Disorders that may present with similar pathology to some 100.2.1 Primary Vasculitis of the CNS
of the vasculitides, but have an endoluminal cause and often
require different treatment. Granulomatous angiitis (also primary angiitis of the
CNS, PACNS) is the most important representative of
primary vasculitides of the CNS. The presenting clin-
signs, including cognitive deterioration, focal cortical ical signs of granulomatous angiitis are nonspecific,
symptoms, stroke-like episodes, etc. often suggesting global dysfunction of the CNS.Acute
Relevant laboratory tests include erythrocyte sedi- or subacute onset confusion, headache, change of per-
mentation rate, complement activation, immune sta- sonality, paresis, cranial neuropathy, or loss of con-
tus and antibodies in blood and CSF, protein and cell sciousness are often presenting signs but as such non-
content of the CSF, and assessment of coagulation fac- specific. The most frequent presenting symptoms of
tors. All these items may be unrevealing, even in his- granulomatous angiitis are headache (68%), paresis
tologically confirmed cases of vasculitis. (56%), and confusion (55%). In almost 25% of the
MRI may play an important role, even though the patients fever and elevated blood pressure are noted.
abnormalities found on MRI are usually not diagnos- Dermatological abnormalities are rare. Funduscopy
tic. It is rare, however, that cerebral vasculitis exists reveals vascular changes in 25% of patients.
with a negative MRI. A normal MRI should even lead The erythrocyte sedimentation rate is in most cas-
to reconsideration of the diagnosis. MRA may be es elevated. CSF pressure is usually increased, and
helpful in showing the vascular abnormalities, but CSF is nearly always abnormal with increased total
special contrast-enhanced techniques should be protein, lymphocytic pleocytosis, and in 30% of the
used. Intra-arterial DSA may be helpful in some cas- patients decreased glucose levels.
100.2 Clinical Presentation and Laboratory Findings 775

Fig. 100.3. T1-weighted images with


contrast (first row) of a 46-year-old
woman with granulomatous angiitis
of the CNS, showing an enhancing
infarction in the right parietal lobe.
MRA (second row) shows rapid and
irregular tapering of the more distal
branches of the middle and posterior
cerebral arteries

Postmortem examination of brain tissue and ex- Giant cell arteritis or arteritis temporalis usually
amination of brain biopsy material show an inflam- occurs in patients, male and female, who are over the
matory process of small arteries and arterioles, pref- age of 55 years. It often involves the superficial tem-
erentially involving deep white matter and lepto- poral arteries, which become swollen, tortuous, ten-
meningeal vessels. Intima proliferation and fibrosis der, and nodular. Pulsations are usually diminished.
are frequent, with multinuclear giant cells of the Lang- Eventually the vessels become hardened and shrink.
hans type and foreign body type. In comparison with Clinical problems most often consist of acute visual
the other vessel layers, the media is relatively spared. failure of one eye. Signs of CNS dysfunction are rela-
Granulomata with macrophages and lymphocytes are tively rare. However, all larger and medium-sized ves-
present. The cause of the disease is unknown. sels of the head and neck may become involved. Of
MRI and angiography are helpful in revealing the particular interest are the carotid, vertebral, and oph-
CNS lesions, consisting of multiple white and gray thalmic arteries, including the ciliary arteries and the
matter lesions and changes of caliber in the cerebral central artery of the retina. The cervical portions of
vessels (Figs. 100.2 and 100.3). MRA is usually not the carotid and vertebral arteries are usually in-
informative, but may show vascular irregularities volved, the intracranial arteries to a lesser extent, and
(Fig. 100.3). The MRI pattern is not specific, but may the spinal arteries least of all.
suggest a vasculitis by a combination of multiple The erythrocyte sedimentation rate is in most cas-
white and gray matter lesions and infarctions of dif- es elevated. Histologically segmental, multifocal pa-
ferent sizes (Fig. 100.4). Diffusion-weighted imaging narteritis is found. The intima of the vessels is thick-
may show lesions with high signal intensity and low ened by subendothelial fibrosis, narrowing or occlud-
ADC values, representative of infarctions. ing the lumen. The internal elastic lamina is severely
Without treatment the disease is usually fatal. but irregularly fragmented. The media is infiltrated
However, granulomatous angiitis responds to high- by small and large mononuclear cells, some of the ep-
dose steroids and cytotoxic agents such as cyclophos- ithelioid type. Giant cells of either Langhans or for-
phamide and azathioprine. eign body type are almost invariably present, either in
the media close to the damaged internal elastic lami-
776 Chapter 100 Vasculitis

Fig. 100.4. A 57-year-old woman with multiple stroke-like vascular territory, similar to what is seen in MELAS. A mito-
episodes over the years. FLAIR images show a large temporo- chondrial disorder was ruled out and the diagnosis granulo-
parieto-occipital infarct on the right side and several lesions in matous angiitis of the CNS was established. Courtesy of Dr. M.
the left hemisphere, with some focal cortical atrophy. Note Heitbrink and Dr. B. Wiarda, Department of Radiology, Medical
that the distribution of the infarction does not reflect a single Center Alkmaar, The Netherlands

na or in the adjacent intima. With healing there is 100.2.2 Secondary Vasculitis of the CNS
scarring and occasionally aneurysm formation. In
some cases the vessels are occluded by organizing 100.2.2.1 Systemic Vessel Wall Disease
thrombus. Takayasu arteritis affects the aortic arch and its
MRI will show intracerebral lesions, but there is branches, in particular in young women of Asiatic
no specific pattern. MRA usually lacks resolution to origin. The smaller intracranial vessels are usually
visualize the abnormal vessels. Intra-arterial DSA not involved. The neurological symptoms are variable
may be more helpful in showing the diseased vessels. because they depend on the extent of the blood vessel
Diagnosis, however, is usually established by biopsy abnormalities, in particular the involvement of the
of the temporal vessels. A negative biopsy does not carotid and vertebral arteries. Visual problems are
exclude the diagnosis. relatively frequent, most often as one-sided amauro-
Therapy includes the use of corticosteroids and sis fugax. Many other neurological symptoms are
immunosuppressives. possible, including hemiparesis, aphasia, cranial
Benign primary angiitis of the CNS is best charac- nerve palsies, coordination problems, and vertigo.
terized as a self-limiting form of granulomatous angi- Laboratory investigations are unrevealing. Histo-
itis of the CNS. This, of course, is a diagnosis a poste- logically there are lesions in the media and adventitia
riori, because there are no criteria by which to distin- of the vessels, but they are predominant in the adven-
guish progressive forms from nonprogressive. titia. The lesions demonstrate collagenous prolifera-
tion and perivascular lymphocyte infiltration. The
media shows an inflammatory granulomatous re-
100.2 Clinical Presentation and Laboratory Findings 777

Fig. 100.5. Two intra-arterial DSA images (left) of Takayasu artery (middle) shows irregular narrowing of the arteries. The
disease. On the left-hand image the narrowing of the neck ves- MRA image on the right shows narrowing and wall irregulari-
sels is clearly visible. The selective injection of the subclavian ties of the subclavian arteries and the neck arteries

action, narrowing the lumen. The cerebral lesions small aneurysms. The parenchymal lesions are small
are secondary to either diminished perfusion or em- infarctions, multiple and disseminated, sometimes
boli. hemorrhagic. Part of the periarteritis nodosa group is
MRA, especially contrast-enhanced MRA of the abuse-associated vasculitis, best described in intra-
aortic arch and branches, is now fully capable of mak- venous methamphetamine users, and hepatitis B
ing an inventory of all involved vessels (Fig. 100.5). virus-associated vasculitis. In these cases, too, the dis-
Treatment consists of corticosteroids, immuno- order is systemic.
suppressives, and, where necessary, interventional ra- MRI shows the cerebral involvement as multiple
diology or vascular surgery for bypasses, stent place- white and gray matter lesions, both lacunar and terri-
ment, and transposition of vessels. torial infarctions. High-resolution MRA may also
In polyarteritis nodosa CNS manifestations usually show vascular abnormalities.
develop in patients who have had systemic disease for Moyamoya syndrome, (moyamoya meaning “puff
several years, in particular with involvement of the of smoke,”) was long considered an ethnic disease
kidneys and the lungs. Clinically two main groups of limited to patients of Japanese ancestry. Today, how-
CNS involvement can be distinguished: one with gen- ever, it is clear that the disease occurs in Europe and
eral signs of CNS involvement, including changes of America as well. The disease is a progressive arteritis
consciousness and epileptic seizures; the other with of the supraclinoid portion of the carotid arteries,
focal or multifocal signs, including ataxia, aphasia, eventually leading to complete occlusion. The disease
hemiparesis, sensory disorders, ophthalmoplegia, usually starts at a young age, rarely under the age of
and visual disorders. The most common neurological 10 years. The fact that the disease is limited to the
presentation is polyneuropathy. carotid arteries has led to the suggestion that the dis-
The erythrocyte sedimentation rate is often but ease is related to a defect in embryogenesis, which is
not always increased. CSF is usually normal. Histolog- different for the carotid arteries from what it is for the
ically the small arteries and arterioles are most often vertebrobasilar system. In the later phases an exten-
affected, especially in the leptomeninges, the deep sive collateral circulation characteristically develops,
white and gray matter, and the choroid plexus. The af- partly via external carotid arteries, partly via the pos-
fected vessels show fibrinoid or hyaline necrosis of terior cerebral artery circulation.
the media and destruction of the internal elastic lam- Laboratory investigations are unrevealing in moya-
ina. Inflammatory granuloma affects the entire thick- moya syndrome. Histopathological examination in
ness of the vessel wall, with secondary intima lesions, moyamoya syndrome reveals that the principal alter-
eventually obstructing the vessel lumen. Segmental ations are stenosis and occlusion of the distal por-
vessel wall necrosis may lead to the formation of tions of the internal carotid arteries and the proximal
778 Chapter 100 Vasculitis

Fig. 100.6. FLAIR images of a 9-year-old boy with moyamoya most intensive in the basal ganglia. The intra-arterial DSA of
syndrome and multiple cortical infarctions (first two rows).The the left internal carotid artery (fourth row, next page) shows
image pattern is similar to what is seen in MELAS, but the im- the typical changes of moyamoya syndrome with a “puff” of
ages through the basal ganglia suggest the presence of many abnormal vessels in the center. The later phase (fifth row)
abnormal vessels. The third row shows the source images of shows the remaining circulation via the posterior cerebral
the MRA, revealing the abundance of small and very small ves- artery, with the collateral “puff of smoke” in the basal ganglia,
sels all through the brain, reflecting the collateral circulation, and the leptomeningeal collaterals in the parietal lobe

parts of the anterior and middle cerebral arteries. intima shows massive fibrous thickening, without
This is combined with numerous dilated, thin-walled atheromatous features. The internal elastic lamina is
collateral arteries branching from the posterior parts usually preserved but extremely wavy and often du-
of the circle of Willis. In the stenotic arteries the plicated or triplicated. The media is atrophic. There is
100.2 Clinical Presentation and Laboratory Findings 779

Fig. 100.6. (continued).

no inflammatory infiltration, but thrombosis, re- There is no real cure for the disease, but microvas-
canalization, and aneurysm formation may occur. cular surgery may help to restore some of the perfu-
The diagnosis can be established with radiological sion by vascular anastomoses
techniques. The anastomoses via lenticulostriate
arteries in the basal ganglia give rise to the typical 100.2.2.2 Autoimmune-Related Disorders
appearance of a cloud (“puff of smoke”) on intra-ar- The rheumatological syndromes associated with CNS
terial DSA (Fig. 100.6), whereas on MRI the usual ap- disease due to vasculitis include a broad spectrum of
pearance is of multiple infarctions, both territorial autoimmune diseases. At one end of this spectrum
and hypoperfusion-related border zone infarctions. one finds organ-specific diseases with organ-specific
Either the cortex or the cerebral white matter is prin- antibodies, for example Hashimoto disease of the thy-
cipally affected (Figs. 100.6 and 100.7). On high-reso- roid. In the middle of the spectrum the lesions tend to
lution images multiple dilated small vessels may be be localized in one organ but the antibodies are not
visible in the basal ganglia (Figs. 100.6 and 100.7). In organ-specific. A typical example is primary biliary
advanced cases there may be dilated small vessels cirrhosis, where the small bile ductules are the main
scattered throughout the brain. target of inflammatory cell infiltration but the serum
780 Chapter 100 Vasculitis

Fig. 100.7.
100.2 Clinical Presentation and Laboratory Findings 781

antibodies, mainly antimitochondrial, are not organ-
Fig. 100.7. A series of T2-weighted images (first and second specific.At the other end of the spectrum the disorder
rows) of a 9-year-old boy with moyamoya syndrome.There are is not organ-specific; lesions and antibodies are not
extensive white matter abnormalities in the deep white mat- confined to a single organ. In SLE, and many other
ter of the cerebral hemispheres suggesting a leukodystrophy. autoimmune disorders, lesions are seen in the skin,
Note that within the abnormal white matter many punctate renal glomeruli, joints, serous membranes, and blood
lesions are present, with high signal intensity in parts and low vessels.
signal intensity in others. The blown-up image of the basal
Rheumatoid disease with CNS vasculitis is a rela-
ganglia shows this to better advantage (third row, left).The ax-
tively rare occurrence. In all patients joint disease
ial and sagittal T1-weighted images (third row, middle and
is apparent. Patients may have had demonstrable
right) show that there is an abundance of small abnormal ves-
rheumatoid arthritis for between 1 and 30 years prior
sels in the brain parenchyma. The source images of the MRA
to the onset of their neurological problems. CNS dis-
and the MRA itself (fourth row) confirm this. The MRA demon-
ease manifests itself nonspecifically by a multitude of
strates that the large cerebral arteries are occluded. Note the
possible neurological signs and symptoms, including
contribution of external vessels to perfusion of the cerebral
seizures, dementia, hemiparesis, cranial nerve palsies,
parenchyma on the left side of the image. The intra-arterial
blindness, cerebellar ataxia, and dysphasia.
DSA confirmed the diagnosis of moyamoya syndrome

Fig. 100.8. The transverse and coronal FLAIR images of a patible with a diagnosis of MELAS. In this case the clinical and
9-year-old boy presenting with progressive dementia show laboratory diagnosis was SLE
multiple, mainly cortical infarctions. The MRI would be com-

Fig. 100.9. The FLAIR images of a 25-year-old woman with a nosed with SLE. Courtesy of Dr. M. Driessen-Kletter, Depart-
multiple sclerosis-like presentation show an MRI pattern com- ment of Neurology, Twenteborg Hospital, Almelo, The Nether-
patible with this diagnosis. Shortly after the MRI she was diag- lands
782 Chapter 100 Vasculitis

Fig. 100.10.
100.2 Clinical Presentation and Laboratory Findings 783

Fig. 100.10. (continued). Images of a 50-year-old female pa- very dark, suggesting calcium deposits. Gradient echo images
tient with SLE.The upper row of T1-weighted images show high (fourth row) show hypointensity in the same areas. The CT im-
signal intensity in the basal ganglia, the pulvinar, and the den- ages (fifth row) confirm that there are massive calcifications in
tate nucleus. The T2-weighted images (second and third rows) these areas. Note that the corpus medullare of the cerebellum
demonstrate a broad rim of hyperintensity around the lateral is also calcified. Courtesy of Dr. E. Gut, Department of Neuro-
ventricles. The basal ganglia, pulvinar, dentate nucleus, and radiology, Kliniken Schmieder, Allensbach, Germany
multiple areas of the cortico-subcortical junction appear

Serum rheumatoid factors are present and the ery- sis, lymphopenia, elevated anti-DNA antibodies, false
throcyte sedimentation rate is elevated. Histological positive test to syphilis (VDRL), complement activa-
examination reveals in a number of cases amyloid de- tion, and an abnormal titer of autoantibodies, such as
posits together with signs of vasculitis. lupus anticoagulant, anti-cardiolipin and other
MRI is nonspecific and in most cases shows scat- APLA. Immune complexes and diminished levels or
tered white matter lesions, sometimes mimicking altered metabolism of the fourth component of the
multiple sclerosis. The presence of systemic abnor- complement cascade have been found in the CSF of
malities will in most cases suggest the diagnosis. patients with CNS disease. Antibodies to neuronal
Therapy of rheumatoid disorders is often complex antigens, often cross-reacting with lymphocytic anti-
and may include antiphlogistic medication, corticos- gens, are preferentially seen in the serum and CSF of
teroids, immunosuppressives, cytokines and many patients with neurological manifestations. The prin-
supportive measures. cipal diagnostic test for the disease is the presence of
Systemic lupus erythematosus (SLE) is a chronic, antinuclear antibodies; antibodies against double-
relapsing-remitting disease, with variable involve- stranded native DNA are specific for SLE.
ment of different organ systems. The disease occurs Histological true vasculitis of the CNS is rare, but
preferentially in adolescent and young adult women. vasculopathic changes are common. Microscopic
Frequent findings include malar rash (butterfly rash), changes are most marked in small vessels and consist
nonerosive arthritis involving multiple joints leading of acute fibrinoid necrosis and marked thickening of
to tenderness, swelling, and effusion, serositis of pleu- the vessel wall with minimal inflammatory cell infil-
ra or pericardium, renal abnormalities, Raynaud tration. Some vessels are occluded by thrombi with
phenomenon, lymphadenopathy, and gastrointestinal corresponding microinfarcts. Autoimmune factors
complaints. Cerebral involvement often presents with that play a role in the cerebral vascular changes in SLE
neuropsychiatric symptoms. Neurological disease is have been identified as lupus anticoagulant, APLA,
the second or third leading cause of death after renal and anti-cardiolipin antibodies. They link SLE with
disease. Neurological manifestations usually follow the pure anti-phospholipid syndrome and with Sned-
systemic manifestations by more than a year. The don disease, which is probably a variation of the anti-
symptomatology is related to the site of CNS involve- phospholipid syndrome.
ment.Also, there may be spinal cord involvement with Many different MRI patterns can be encountered in
transverse myelitis. SLE. Sometimes the MRI pattern mimics MELAS,
There are criteria for the diagnosis of SLE (Ameri- showing mainly cortical infarctions (Fig. 100.8).
can College of Rheumatology), which contain both Sometimes it mimics multiple sclerosis (Fig. 100.9). In
clinical symptoms and more or less specific laborato- other cases there are more widespread white matter
ry findings, such as hemolytic anemia with reticulo- lesions (Figs. 100.10 and 100.11). In some patients se-
784 Chapter 100 Vasculitis

Fig. 100.11. A 56-year-old woman with a long history of com- duncles, the hilus of the dentate nucleus, and the peridentate
plicated SLE.There is symmetrical involvement of the cerebral white matter. Courtesy of Dr. R. Schiffmann, Developmental
hemispheric white matter, the anterior and posterior limbs of and Metabolic Neurology Branch,National Institutes of Health,
the internal capsule, the external capsule, the thalamus (espe- Bethesda, Maryland, USA
cially the pulvinar), the midbrain, pons, middle cerebellar pe-

vere involvement of the basal ganglia is seen, a pattern toid arthritis, thrombocytopenic purpura, and poly-
that has to be differentiated from central venous cythemia. At an early age the patients develop cere-
thrombosis. In still other patients there is extensive brovascular manifestations, including larger and
calcification of the basal ganglia, dentate nucleus, cen- smaller arterial territorial infarctions, with severe
trum semiovale, and cortico-subcortical junction neurological consequences and often cognitive dete-
(Fig. 100.10). Enhancement may occur in active le- rioration.
sions. In some cases, despite neuropsychiatric symp- Sneddon syndrome is linked with the presence of
toms, conventional MRI is normal. In those cases esti- anti-cardiolipin antibodies (one of the APLA), and
mation of magnetization transfer histograms may de- therefore Sneddon syndrome may be regarded as
tect abnormalities, without otherwise visible lesions. belonging to the APLA-related disorders. However,
Therapy is usually with corticosteroids and im- the presence of APLA is highly variable in Sneddon
munosuppressives. disease (41% positive). In APLA-positive patients,
Sneddon syndrome is characterized by a combina- audible mitral regurgitation is observed more fre-
tion of generalized livedo racemosa (or reticularis), a quently. There is a close relationship between Sneddon
cutaneous condition featured by a reddish-purple syndrome, SLE, and “pure” APLA syndrome. It has
reticulated pattern predominantly present on trunk been suggested that Sneddon syndrome might cover
and extremities, and diffuse intravascular coagula- a continuum of various clinical and biological enti-
tion leading to cerebrovascular manifestations. The ties, ranging from APLA-related SLE patients, to pri-
skin abnormality is not specific, but may be also be mary APLA-positive patients, and Sneddon syn-
associated with polyarteritis nodosa, SLE, rheuma- drome in the middle.
100.2 Clinical Presentation and Laboratory Findings 785

Fig. 100.12. Gradient echo images of


two patients with Sneddon syndrome.
The upper two images are of a 53-year-
old woman who has suffered multiple
infarctions in the past. She is now
wheelchair-bound and suffering from
progressive cognitive impairment.The
lower two images are of a 42- year-old
woman who has also suffered multi-
ple transient ischemic attacks and
permanent infarctions. She also has
impairment of cognitive functions.
In both women there is ventricu-
lomegaly.The gradient echo images
show the preferential occurrence of
the hemorrhages at the surface of the
brain and the ependymal lining.This is
not often seen in other vasculitides.
The superficial hemorrhages result in
focal superficial hemosiderosis.The
black rim around the brain stem in the
upper images is also caused by hemo-
siderin. In the lower images punctate
black dots are seen nearly everywhere
in the brain, reflecting microhemor-
rhages

Nonspecific laboratory findings include an elevat- unless they also have thrombocytopenia. On the con-
ed erythrocyte sedimentation rate. The finding of trary, patients present with a hypercoagulative state
APLA in addition to typical skin abnormalities con- which leads to thrombotic complications, stroke, my-
firms the diagnosis. ocardial infarctions, dementia, and fetal loss.
Histopathologically Sneddon syndrome particu- APLA syndrome is found in up to 30–50% of pa-
larly attacks small cortical arteries. Infarctions are, tients with SLE. Patients without SLE or other sys-
therefore, most often located in cortical areas. They temic disease can also develop APLA syndrome, re-
are often hemorrhagic, and hemosiderin deposits are ferred to either secondary or primary or pure APLA
also found in the leptomeninges. syndrome. Children may develop secondary APLA
MRI shows a variable number of smaller and larg- syndrome during viral infections. Up to 30% of pa-
er infarctions, in which cortical involvement is most tients with HIV-infection develop secondary APLA
prominent. With gradient echo techniques multiple syndrome.“Pure” APLA syndrome occurs in patients
hemosiderin deposits with extensive superficial he- without any of these antecedents.
mosiderosis is the most common finding. In our ex- Venous thrombosis is a relatively frequent occur-
perience this finding is more constant in Sneddon rence in APLA syndrome. About 30% of patients with
syndrome than in other vasculitides (Fig. 100.12). APLA syndrome acquire deep venous thrombosis. In
Treatment according to the guidelines for SLE and patients with APLA syndrome and SLE the figure is
APLA syndrome is the rule. even 40%. In drug-induced or parainfectious APLA
Anti-phospholipid antibody syndrome. Anti-phos- syndrome it is less than 5%. Patients with APLA syn-
pholipid antibodies (APLA) are antibodies against drome may prove difficult to treat for venous throm-
phospholipids and are found in a variety of clinical bosis and they have a high rate of recurrence. In
disorders. The discovery of APLA followed the obser- young adults with APLA syndrome there is an in-
vation that in a patient with SLE the activated partial creased risk of arterial disease, leading to myocardial
thromboplastin time (aPTT) was prolonged (lupus infarctions, stroke, and a higher incidence of periph-
anticoagulant). Despite this, patients with high levels eral vascular occlusions. In addition to brain damage
of APLA do not develop hemorrhagic complications by territorial infarctions, patients often present with
786 Chapter 100 Vasculitis

early-onset dementia, the average age at onset being culopathy with a perivascular mononuclear reaction
52 years. In these patients there is usually no history infiltrating in the cerebral parenchyma, usually in-
of major strokes. Associated findings include livedo volving the white matter.
racemosa, Raynaud phenomenon, superficial throm- On MRI more or less extensive white and gray mat-
bophlebitis, cardiac valve vegetations, and mitral re- ter lesions have been reported, including partial terri-
gurgitation. torial infarctions. Microbleeds, present in histopatho-
Laboratory testing for APLA syndrome is complex logical studies, can be seen on gradient echo se-
and several subtests are involved. There is no simple quences. Lesions in the posterior fossa are rare. The
screening test for the condition. Testing should be MRI pattern is not specific and the differential diag-
done when one or more of the major clinical manifes- nosis includes multiple sclerosis, SLE, Behçet disease,
tations occur: arterial or venous thrombosis, throm- and microscopic polyarteritis nodosa.
bocytopenia, or frequent miscarriages. Primary Scleroderma (scleros=hard, derma=skin) is a pro-
APLA syndrome patients are more often male, and gressive disease that leads to hardening of the skin
have a low titer of anti-nuclear antibodies. and connective tissue and is part of the group of
MRI can, with the appropriate techniques, demon- arthritic conditions called connective tissue diseases.
strate the involvement of the CNS. MRI shows white There are two main forms of scleroderma, a localized
matter abnormalities, suggestive of small vessel dis- and a systemic form. The localized form is subdivid-
ease. Apart from the white matter involvement, le- ed into two forms, morphea and linear scleroderma.
sions usually extend into the cortical layers. In fresh Morphea is a cutaneous lesion with indurated, slight-
infarctions diffusion-weighted imaging is helpful. ly depressed plaques of thickened dermal fibrous tis-
Gradient echo sequences will show microhemor- sue, white or yellowish, with pinkish-purplish halo.
rhages when present. The linear form of scleroderma is a line of thickened
Therapeutically one has to be aware that, although tissue, which affects the skin, but can also affect the
APLA syndrome is considered to be an autoimmune muscles and bone underneath, limiting movements
disease, immunosuppressive therapy does not pre- of limbs when affected. The underlying cortex can al-
vent recurrent thrombosis, fetal loss, or neurological so be affected when the abnormality occurs on the
syndromes and should therefore not be considered. A forehead. Linear scleroderma is usually present on
possible exception to this rule is the treatment of cat- arms, legs, or on the forehead. It may appear as a long
astrophic cases, in which also plasmapheresis may be streak resembling a deep saber wound, often referred
attempted. In thrombotic forms anticoagulant thera- to as “en coup de sabre.” The systemic form of sclero-
py is usually the treatment of choice. derma can affect any part of the body, skin, blood ves-
In Sjögren syndrome, an autoimmune disorder, sels, and internal organs. The systemic form is also re-
neurological complications are not uncommon, most ferred to as diffuse scleroderma or CREST (calcinosis,
often affecting the PNS. CNS involvement is reported Raynaud syndrome, esophageal problems, sclero-
in 25–30% of the patients, presenting as trigeminal dactylia, and telangiectasia) syndrome. The presence
neuropathy, recurrent aseptic meningoencephalitis, of two of the five symptoms mentioned is enough to
or unifocal or multifocal cerebral parenchymatous le- make the diagnosis CREST. When CREST is present
sions.Also lesions of the spinal cord may occur result- together with other symptoms of scleroderma, it is re-
ing from a necrotizing vasculitis and leading to trans- ferred to as “limited scleroderma plus CREST.”
verse myelitis, chronic progressive myelitis, a Brown– In scleroderma many organs and organ systems
Séquard syndrome, or a neurogenic bladder. Patients may be involved, including skin; blood vessels; respi-
may also present with neuropsychiatric symptoms. ratory system; musculoskeletal system; cardiovascu-
The involvement of the salivary and lacrimal glands lar system; genitourinary system; ears, nose, and
(the “sicca” manifestations) is usually manifest before throat (sicca syndrome); renal system; endocrine sys-
CNS or PNS symptoms, but may also appear later. tem; PNS; and CNS. Focusing on neurological in-
More general constitutional symptoms may also be volvement: peripheral nerve manifestations may lead
present, such as fatigue, malaise, low-grade fever, Ray- to pain and paresthesias from nerve entrapment, for
naud syndrome, lymphadenopathy, arthralgia, myal- instance trigeminal neuralgia and carpal tunnel syn-
gia, as well as involvement of lungs, kidneys, muscles, drome. Involvement of the CNS may lead to (partial)
and joints. Lymphoproliferative disorders are poten- territorial strokes, epileptic seizures, with as patho-
tial complications. This disease has much in common logical substrate focal narrowing of middle-sized
with rheumatoid arthritis and SLE. arteries, and more or less extensive intracerebral cal-
Pathological studies of peripheral nerves and mus- cifications, probably related to an underlying cere-
cle show acute or chronic vasculitis or perivascular brovascular pathology. Some patients present with re-
inflammation. In a few cases in which biopsy of the current loss of consciousness and multiple sponta-
CNS was performed, either an unambiguous vasculi- neous intracerebral hemorrhages.
tis was found or, more typically, a mononuclear vas-
100.2 Clinical Presentation and Laboratory Findings 787

Anti-centromere antibodies have been found in but findings may also mimic tuberculosis, multiple
patients with scleroderma and Raynaud syndrome sclerosis, lymphomas, and fungal infections of the
and in CREST patients and may facilitate the diagno- brain. Extra-axial manifestations may show features
sis. Extractable nuclear antibodies and other autoan- comparable to convexity meningiomas. Spread along
tibodies may also be found. When peripheral skin le- the perivascular spaces, leptomeningeal involvement,
sions are present, the diagnosis can be confirmed by involvement of the optic nerve, the pituitary gland,
skin biopsy. The skin is thickened as a result of over- the floor of the third ventricle, and the hypothalamus
production of collagenous tissue, overgrowing hair have all been reported. Spinal manifestations are not
follicles and sweat glands. rare.
Histologically there are two major findings: fibrot- Although the cause of sarcoidosis still is unknown,
ic changes, resulting from endothelial damage of it is generally accepted that sarcoidosis results from
small vessels and subsequent collagen deposition, exposure of genetically predisposed individuals to
and vasculopathy, consisting of fibrinoid necrosis of specific environmental agents. The finding of associ-
arterioles, lymphoid hyperplasia, and thickening of ations between the human leukocyte antigens of the
the basement membrane. major histocompatibility complex and sarcoidosis
The cause of scleroderma is unknown. Recently it supports this hypothesis. So does the difference in
has been suggested that cellular microchimerism prevalence rates between ethnic groups.Another pos-
with a lifelong status could form an immunological sible association is between sarcoidosis and bacterial
basis for amplification of autoimmune reactions lead- DNA, especially DNA of Propionibacterium acnes or
ing to clinical manifestations of systemic sclerosis. granulosum. Further studies will have to confirm the
Microchimerism has been defined by the presence of value of these findings.
a small number of circulating cells transferred from Laboratory tests are not very helpful in establish-
another individual. This transfer may occur during ing the diagnosis. A positive angiotensin converting
pregnancy between mother and fetus or, in multiges- enzyme (ACE) test in the CSF may support the diag-
tational pregnancies, between fetuses. Other causes nosis, but the test may also be negative or neutral. The
are blood transfusion, bone marrow transplantation, results of this test should therefore be interpreted
and organ transplants. Microchimerism has been im- with care. The cell count in the CSF is usually slightly
plicated not only in systemic sclerosis, but in the elevated, as is the protein content.
pathogenesis of autoimmune diseases more general- Histologically the manifestations can be divided
ly. An increased number of microchimeric cells has into those with proliferation of granulomatous tissue,
been found in peripheral blood and tissues of pa- leading to more solid lesions in the suprasellar re-
tients with systemic sclerosis, and they have been gion, the brain, and spinal cord, and those with lep-
proven to be specifically activated and capable of rec- tomeningeal and perivascular involvement with fea-
ognizing human leukocyte antigens (HLA) from pa- tures of an inflammatory vasculitis.
tients. MRI can portray the major manifestations: the
At this moment there is no specific treatment for suprasellar–leptomeningeal involvement (Fig. 100.13)
scleroderma. Attempts are under way to build upon and the spread along the perivascular spaces
the remarkably successful use of TNF-a neutralizing (Fig. 100.14), in both cases with enhancement after
treatments for rheumatoid arthritis, which may have contrast injection. In other cases the lesions mimic
paved the way for similar approaches in rheumatoid- multiple sclerosis patterns. In several patients an ex-
like disorders tra-axial process very similar to a meningioma has
MRI is nonspecific and presentations vary widely. been described. Intramedullary lesions are also easily
Infarctions may occur in middle-sized artery territo- demonstrated on spinal MR.
ries. In addition, macro- and microhemorrhages and Therapeutically corticosteroids may be beneficial.
extensive calcifications may be present. In other cases In more chronic cases cyclophosphamide and
lesions mimic patterns found in multiple sclerosis. methotrexate are treatment options.
Where frontal linear scleroderma is present one may Wegener granulomatosis (Wegener disease) may
expect underlying cortical abnormalities, calcifica- lead to cerebral lesions, either as lesions primarily lo-
tions, and atrophy. cated in the nasal cavities and sinuses and extending
Neurosarcoidosis occurs in approximately 5% of intracranially, or as necrotizing cerebral vasculitis,
patients with systemic sarcoidosis, also called Besnier– nearly always in the presence of active sinusitis, otitis,
Boeck–Schaumann disease. Sarcoidosis is a systemic or lung disease. A limited form of Wegener disease,
granulomatosis of unknown cause, especially affect- without upper and lower respiratory tract disease, has
ing the lungs with lung fibrosis, but also found in hi- been repeatedly described and has a better prognosis.
lar lymph nodes, skin, liver, spleen, eyes, phalangeal The clinical features are protean, but ocular manifes-
bones, parotid glands, and the CNS. In the CNS facial tations are common.
nerve paralysis is the most common manifestation,
788 Chapter 100 Vasculitis

Fig. 100.13. Sagittal T1-weighted, contrast-enhanced, fat-sup- mus, pituitary stalk, the leptomeninges around the brain stem
pressed images of a patient with neurosarcoidosis, showing and cerebellum, in the sulcus cinguli, and in the foramen of
the characteristic images of involvement of the hypothala- Magendie

Fig. 100.14. In this patient with neurosarcoidosis, the involve- both cortex and white matter. After contrast injection (middle
ment concerns the perivascular spaces. The two FLAIR images and right columns) enhancement occurs, in part with a stripe-
(left column) show multiple infarct-like lesions involving in like pattern following the course of the perivascular spaces
100.2 Clinical Presentation and Laboratory Findings 789

The diagnosis may be sustained by the presence of Churg–Strauss syndrome belongs to the group of
cytoplasmic anti-neutrophil antibodies (ANCAs). necrotizing vasculitides. It is characterized by an
The optimal evaluation of serum ANCA levels re- eosinophil-rich and granulomatous inflammation
quires assessment of the presence of two principal and necrotizing vasculitis involving the small- to
ANCA targets: myeloperoxidase and proteinase 3. medium-sized vessels, affecting particularly the respi-
The cytoplasmic (c-ANCA) staining pattern of neu- ratory tract. The disease is associated with asthma and
trophils stems largely from antibodies to proteinase 3; peripheral eosinophilia, often combined with necro-
the perinuclear (p-ANCA) staining pattern is derived tizing glomerulonephritis. Cerebral manifestations
from antibodies to myeloperoxidase and several oth- occur in about 10% of the patients. About 60% of pa-
er antigens, including lactoferrin, cathepsin G, and tients are p-ANCA-positive. MRI findings are variable
elastase. ANCAs are detected in patients with differ- and the cerebral lesions are similar to those seen in
ent disorders, such as infections, inflammatory bowel many other vasculitides. They consist of macro- or mi-
disease, autoimmune hepatitis, and malignancies. croinfarctions and micro- or macrohemorrhages.
With the presence of c-ANCAs, high titers of pro- Microscopic polyarteritis (polyangiopathy) is also
teinase 3, and the absence of antinuclear antibodies, characterized by necrotizing vasculitis of the small
the diagnosis of Wegener disease becomes more de- vessels, arteriolae, capillaries, or veins, without gran-
finitive. ulomas. The disease is nearly always associated with
Histologically Wegener disease is characterized by necrotizing glomerulonephritis and often with pul-
necrotizing panarteritis of the middle great vessels. monary capillaritis and hemorrhage. Peripheral neu-
Multiple foci of arteritis develop in the nasal sinuses, ropathy occurs less frequently than in polyarteritis
the respiratory tract, the eye, and the kidneys. There is nodosa. Sinusitis, which is rare in polyarteritis no-
much similarity between Wegener disease and in- dosa, occurs in 9% of patients with microscopic pol-
volvement of the CNS by other granulomatous and yarteritis. CNS involvement is rare, but when present
arteritic diseases, including Churg–Strauss syndrome will be caused by necrotizing vasculitis.
and microscopic polyarteritis nodosa. The p-ANCA reaction is found in 50–80% of the
There is a close relationship between Wegener dis- patients with microscopic polyarteritis and in less
ease, Churg–Strauss syndrome, and microscopic pol- than 20% of patients with polyarteritis nodosa. His-
yarteritis (as distinct from polyarteritis nodosa). The tologically there is necrotizing vasculitis of small ves-
latter involves predominantly if not exclusively arter- sels, with microinfarctions and microhemorrhages.
ies, whereas the other disorders virtually always affect MRI may show any degree of small vessel disease,
vessels smaller than arteries. with involvement of both white and gray matter. Ad-
In limited Wegener disease the histopathological ditionally, MRI may show small and larger infarc-
characteristics are similar to those of the typical dis- tions. MRA is usually negative.
ease, but without the upper and lower respiratory Behçet disease is a multisystem recurrent vasculitis
tract and kidney involvement. The ANCA test is less of supposedly autoimmune origin, involving many
likely to be positive. In limited Wegener disease organs. The original clinical triad of symptoms in-
50–60% of the patients have sinusitis, and 6% have cludes oral ulcers, genital ulcers, and anterior or pos-
hearing loss due to formation of granulomatous tis- terior uveitis. The disease is frequently encountered
sue between eustachian tube and tympanum. De- in a specific geographic distribution extending from
struction of the petrous part of the temporal bone Japan to the eastern Mediterranean countries, pass-
and inflammatory masses at the base of the skull may ing through China and Iran: the area that was suppos-
also be present. Erosion of the lamina papyracea of edly the ancient silk route. The disease mainly affects
the orbital walls may occur. Involvement of single or young adults. The female:male ratio is 11:1 in Turkey,
multiple cranial nerves may occur, especially of the less pronounced in other countries. In Turkey the
eighth, ninth, tenth, and twelfth cranial nerves. The prevalence is 80–300 per million inhabitants, whereas
cranial neuropathies are caused either by necrotizing the prevalence is 1 per 100,000 in the USA.
small vessel vasculitis or by a granulomatous process Behçet disease is known to cause a variety of other
in the meninges. manifestations, such as arthritis, arterial and venous
Leptomeningeal enhancement on contrast-en- thrombosis, pulmonary angiitis, cardiac disorders,
hanced MRI has been described in Wegener granulo- cutaneous lesions, rectocolitis, and lesions of the
matosis. Isolated intracranial or spinal lesions may al- CNS. CNS involvement is estimated to occur in
so be seen (Fig. 100.15). The lesions will enhance after 5–50% of the cases. Neurological manifestations oc-
contrast injection (Fig. 100.15). MR and CT examina- cur in a wide variety of forms, due to the involvement
tions of the nasal sinuses and mastoids are important of either the leptomeninges, the brain parenchyma, or
in the evaluation of cerebral involvement by continu- the blood vessels. All sizes of blood vessels are in-
ous extension. volved, and arteries as well as veins. Clinically the
CNS involvement becomes apparent as meningoen-
790 Chapter 100 Vasculitis

Fig. 100.15. A 9-year-old boy with


Wegener disease.The FLAIR images
through the brain show subcortical
and cortical spots with hyperintensity.
In the thoracic spinal cord there is
an intramedullary lesion with ring
enhancement and lower signal
intensity in the core of the lesion.
In the lungs there are multiple
granulomas

cephalitis, cranial nerve palsies, cerebellar ataxia, in its ability to attack blood vessels of any kind and
spastic para- or tetraparesis, raised intracranial pres- any size, veins as well as arteries, with preference for
sure, and dementia. Parenchymal CNS involvement certain organs. In the brain Behçet disease involves
occurs in the majority of the patients, predominantly predominantly the white matter of the cerebral hemi-
in the basal ganglia and brain stem and spinal cord. spheres, the basal ganglia, and the brain stem. Ob-
CSF showed pleocytosis and elevated protein or an el- struction of arteries may lead to territorial infarcts;
evated CSF pressure. involvement of smaller vessels leads to small vessel
The cause of Behçet disease is unknown. Viral and disease and lacunar infarctions; thrombosis of ve-
bacterial infections have been suggested, until now nous structures may lead to venous infarctions or
without sufficient evidence. It is widely assumed that raised intracranial pressure.
Behçet disease is an autoimmune disorder, even MRI plays an important role in the diagnosis and
though not all the facts agree with that view. Argu- follow-up of neuro-Behçet, and should aim at depict-
ments against the autoimmune hypothesis are the ing the various forms of CNS involvement:
male predominance, the lack of any specific antigen ∑ White matter abnormalities often involve the
or antibody, and the lack of a relationship with HLA cerebral hemispheres, with isolated and confluent
class II antigens. Genetic susceptibility, however, ex- lesions in the centrum semiovale and somewhat
ists in some populations. In Turkey 84% of patients less prominent involvement of periventricular ar-
with Behçet disease are positive for HLA B51, which is eas than in multiple sclerosis. However, they are
not found in other populations. often difficult to distinguish from multiple sclero-
Histologically the vasculitis in the CNS is charac- sis.
terized by perivascular cell infiltration, infarctions ∑ Lesions are often present in the basal ganglia, mid-
with small necrotic areas surrounding the blood ves- brain, and pons; the brain stem lesions are more
sels, microhemorrhages, loss of myelin, and gliosis. frequent, larger, and more extensive than in multi-
Behçet disease is unparalleled among the vasculitides ple sclerosis (Figs. 100.16 and 100.17).
100.2 Clinical Presentation and Laboratory Findings 791

Fig. 100.16. Behçet disease may cause lesions anywhere in enhancement of the lesions on the T1-weighted images after
the brain, but most often involves the brain stem and basal contrast (third row). Courtesy of Dr. G. Akman-Demir, Depart-
ganglia in an asymmetrical fashion.The FLAIR images (first and ment of Neurology, Medical Faculty, Istanbul University, Istan-
second rows) of a 53-year-old woman depict the lesions in the bul, Turkey
midbrain, pons and around the fourth ventricle. Note the

∑ Small, and occasionally large, infarctions may be ∑ Leptomeningeal involvement is especially to be


present; diffusion-weighted images may be added expected in patients presenting with cranial nerve
to the usual protocol, which includes proton den- palsies and in those with meningoencephalitis;
sity, T2-weighted, and FLAIR images, in order to contrast administration and T1-weighted images
catch fresh infarctions. with fat suppression are required for adequate de-
piction.
792 Chapter 100 Vasculitis

Fig. 100.17. A 28-year-old man with Behçet disease. Note the of Neurology, Medical Faculty, Istanbul University, Istanbul,
involvement of the internal capsule on the right, the midbrain Turkey
and the medulla. Courtesy of Dr. G. Akman-Demir, Department

∑ Visualization of microhemorrhages, as often seen 100.2.2.3 Infectious Vasculitis


in pathological specimens, demands a gradient A number of infectious disorders may give rise to vas-
echo technique on MRI. culitis and related cerebral lesions. Lyme disease
∑ In patients with raised intracranial pressure, (neuroborreliosis) is an infectious disease caused by
thrombosis of the large venous sinuses has to be Borrelia burgdorferi, which may affect many organs,
ruled out; on a T1-weighted sagittal image this such as skin (90% of all patients), joints, heart, CNS,
may already be obvious when the superior sagittal and PNS. The first manifestation is often a skin lesion,
sinus is involved. Thrombosis of other intracra- known as erythema (chronicum) migrans. This may
nial veins can be made visible with MR phlebogra- be followed by bacterial dissemination, presenting
phy. initially as a flu-like disorder, with fever, malaise, and
∑ In patients presenting with large territorial infarc- diffuse pain. About 15% of patients subsequently de-
tions MRA may show the occluded vessels. velop one or more features of CNS involvement: lym-
∑ Spinal cord lesions will be visible on proton densi- phocytic meningitis (in the USA) or meningoen-
ty and T2-weighted images, and the spinal lesions cephalitis (in Europe), uni- or bilateral facial paresis
will usually enhance after contrast injection, as (to be differentiated from neurosarcoidosis and Guil-
will many of the intracranial lesions in Behçet dis- lain–Barré syndrome), and polyradiculitis (which
ease (Fig. 100.16). may mimic Guillain–Barré syndrome). In a minority
of patients focal encephalomyelitis will develop with
Therapy in Behçet disease may be topical when oral prominent white matter involvement on MRI.
aphthae and genital ulcers are concerned. More ex- In untreated or unsuccessfully treated patients, late
tensive systemic involvement usually requires corti- neurological symptoms may develop, which include
costeroid and immunosuppressive therapy. chronic encephalomyelitis, with focal neurological
100.2 Clinical Presentation and Laboratory Findings 793

Fig. 100.18. Images of a 10-year-old girl with neuroborrelio- lesions enhance. These images do not allow differentiation
sis. The T2-weighted images show multiple lesions of different from acute disseminated encephalomyelitis (ADEM) or multi-
sizes, asymmetrically spread through the centra semiovalia of ple sclerosis. From Demaerel et al. (1995), with permission and
both hemispheres. The T1-weighted, contrast-enhanced im- by courtesy of Dr. P. Demaerel, Department of Radiology, Uni-
ages in the coronal and sagittal planes show that several versity Hospitals Gasthuisberg, Leuven, Belgium

abnormalities and also focal abnormalities on MRI. A bite, erythema migrans, and one or more of the man-
chronic infection may lead to confusional states, with ifestations within the scope of Lyme disease.
memory loss and cognitive impairment. Due to limi- The cause of Lyme disease is an infection by a
tations in diagnostic technology, Lyme disease is still member of a group of spirochetes, originally named
primarily a clinical diagnosis, usually accepted only Borrelia burgdorferi, now referred to as Borrelia
when the antecedents of the patient include a tick- burgdorferi sensu lato, subdivided into at least three
794 Chapter 100 Vasculitis

Fig. 100.19. A 10-year-old boy with Lyme disease presented transverse plane (second row, right) the enhancement and
with low back pain radiating to his legs. The T1-weighted, con- thickening of nerve roots is confirmed. From Demaerel et al.
trast-enhanced sagittal images (first row) of the lower thoracic (1998), with permission and by courtesy of Dr. P. Demaerel,
and lumbar spine show the enhancing and thickened caudal Department of Radiology, University Hospitals Gasthuisberg,
roots. In the coronal plane (second row, left and middle) and Leuven, Belgium
100.2 Clinical Presentation and Laboratory Findings 795

species: Borrelia burgdorferi sensu stricto, Borrelia infection is less common, but arachnoiditis and focal
burgdorferi garinii, and Borrelia burgdorferi afzelii. intramedullary tuberculomas may occur. Tubercu-
The first group is responsible for the North American lous spondylitis may lead to secondary involvement
Lyme infections, the latter two for the European of spinal cord and roots. Because of migration of in-
forms, which have some differences in clinical fea- dividuals from regions in the world where tuberculo-
tures. The disease is a zoonosis, with humans as inad- sis is still endemic and the higher incidence of cases
vertent host. The disease is transmitted to humans by in immunocompromised individuals, one should be
a bite from the hard-shelled ixodid ticks. These ticks aware of the possibility of a tuberculous infection.
are especially abundant in areas where the ecological The demonstration of Mycobacterium tuberculosis
circumstances, climate, food, and the different vectors or bacterial DNA in the body fluids or biopsy materi-
for their procreation are present. The life cycle of the al makes a certain diagnosis. Brain involvement is of-
ticks runs a three-phase course over a 2-year cycle. ten secondary to a pulmonary infection. Indirect
Even in areas with a high proportion of infected ticks markers are pleocytosis in the CSF, high protein and
only 1–2% of humans with a tick bite will become low glucose levels and presence of indicators of in-
infected. flammation in the plasma. Often CSF has trans-
Diagnosis of Lyme disease is not straightforward. formed into a thick, yellowish fluid, which may be the
Serological tests take a long time and may be unreli- cause of a communicating hydrocephalus.
able because of crossover reactions, which can lead to Histologically, the different forms of tuberculosis
false positives. CSF examination may be positive for of the brain and meninges have different aspects. Tu-
inflammatory markers, pleocytosis, slight rise in pro- berculomas occur in the brain parenchyma and in the
tein, and usually normal glucose. A test can be per- leptomeninges and choroid plexus. The larger tuber-
formed to show anti-Borrelia antibodies with a Lyme- culomas have a granulomatous border, often with
specific ELISA. PCR may be applied to detect bacteri- Langhans giant cells, encompassing a central caseat-
al DNA. ing necrosis. They are usually small and multiple, but
There is no specific MR pattern of Lyme disease. incidentally may become larger and have mass effect.
There may be many small lesions dispersed through Leptomeningeal tuberculosis can consist of isolated
the hemispheres, sometimes very similar to the pat- tuberculomas of varying size, often associated with
tern of multiple sclerosis (Fig. 100.18). Lesions in the tuberculomas in the brain tissue. A second form ap-
corpus callosum are less common. After injection of pears as generalized meningitis, presenting as a gray-
contrast, active lesions may enhance (Fig. 100.18). ish, gelatinous, thick exudate, predominantly at the
Special techniques should be used to image the cra- base of the brain. The exudate consists of polymor-
nial nerves when involved; in the active phase these phonuclear cell infiltrations, fibrin exudation, endar-
will also enhance. Leptomeningeal enhancement may teritis, hemorrhages and caseous necrosis. Perivascu-
occur in patients with meningoencephalitis, but in lar intraparenchymal inflammatory extensions are
our experience this is rare in the chronic form. Le- frequent and hemorrhagic infarctions may ensue.
sions in the spinal cord and involvement of nerve MR images, standard morphological and T1-
roots may also occur (Fig. 100.19). weighted images with and without contrast, where
Antibiotic therapy is effective when given appro- available with fat suppression, depict the described
priately and cures up to 90–95% of patients. In pa- manifestations and secondary effects (Fig. 110.20).
tients with chronic disease the percentage is lower, MRA demonstrates the affected vessels. Diffusion-
80–85%. weighted imaging will show infarction or abscesses.
Tuberculosis is caused by Mycobacterium tubercu- Therapy consists of tuberculostatic medication
losis. Involvement of the brain is usually due to and necessary supplements.
hematogenous spread from a primary focus, in most Syphilitic angiitis is caused by a spirochete, Tre-
cases the lungs. Intracranial manifestations include ponema pallidum, and presents clinically as a multi-
extension of a subpial or subependymal focus, result- system infection. Neurosyphilis is one of its manifes-
ing in involvement of the basal leptomeninges, which tations. Neurosyphilis can be classified into distinct
causes basal leptomeningitis. Occlusion of the CSF syndromes that span all stages of dissemination of
pathways leads to hydrocephalus. The infection may the disease.
also induce vasculitis of the smaller and middle-sized In the first phase the local genital infection and
cerebral arteries, often the lenticulostriate arteries or skin abnormalities prevail, but a meningeal syphilitic
the posterior cerebral artery branches, the thalamo- infection at the onset may lead to cranial nerve palsies
perforate arteries, leading to small infarctions in the and ocular changes. Four to 7 years after the primary
basal ganglia and deep white matter. Multiple miliary infection, a meningovascular syphilis may develop,
abscesses may also occur, as well as tuberculous intra- with focal nervous system ischemia, secondary to
parenchymal granulomas (tuberculomas), cerebritis, thrombosis. Parenchymatous manifestations of neu-
larger abscesses, and pachymeningitis. Spinal cord rosyphilis occur 10–30 years after the primary infec-
796 Chapter 100 Vasculitis

Fig. 100.20. Tuberculous meningoencephalitis. The first row leukomalacia. Focal lesions are seen in the basal ganglia
shows one T1-weighted and two T2-weighted images; the sec- and mesencephalon.The contrast-enhanced images show the
ond row shows T1-weighted images with contrast. The images typical basal leptomeningitis
in the first row show ventriculomegaly and periventricular

tion with general paresis or tabes dorsalis. In this late rapid plasma reagin (RPR) test, are more sensitive
phase other parenchymatous lesions, gummata, or than specific. False positive results can be obtained in
cranial nerve involvement, hearing loss, or optic neu- SLE, herpes simplex infections, pregnancy, and Lyme
ritis may occur. In this late phase cognitive decline, disease.
delusions, and paresis are part of the clinical picture, Pathophysiologically the infection leads to an
although nowadays, because of proper treatment, obliterative endarteritis of terminal arterioles, result-
rarely observed. In combination with HIV infection, ing in inflammatory and necrotic lesions.
the neurological manifestations of neurosyphilis oc- MRI is not specific in cases of neurosyphilis. Con-
cur at an earlier date, accelerate faster, and are more trast-enhanced T1-weighted images show the lep-
severe. tomeningitis, infarctions, and gummata when pre-
The gold standard for the diagnosis is the demon- sent. In HIV-positive patients the diagnosis may be
stration of spirochetes by dark-field examination, even more difficult.
which is easy when skin or genital lesions are present. When treated in time the infection can be cured.
In neurosyphilis this is more difficult and the diagno- Various regimes of antibiotics are now available. In
sis is suggested by clinical findings, medical history, patients with HIV, however, treatment failure is fre-
and CSF analysis. Serological studies can confirm the quent.
diagnosis. Treponema tests include the fluorescent Varicella-zoster complications stem from infection
treponemal antibody absorption double staining with the varicella-zoster virus, a virus that occurs ex-
hemagglutination test, and the Treponema immobi- clusively in humans and causes chickenpox (varicel-
lization test. Nontreponemal tests, such as the Venere- la). It then becomes latent in cranial nerve and dorsal
al Disease Research Laboratory (VDRL) test and the root ganglia, and frequently reactivates decades later
100.2 Clinical Presentation and Laboratory Findings 797

Fig. 100.21. Left-sided infarction of


the brain following left-sided herpes
zoster ophthalmicus

to produce shingles (herpes zoster) and postherpetic (mean age 55 years, range 7–96 years). The hemiple-
neuralgia. Latency depends on age of the virus and gia occurs one week to 2 years after the onset of the
immune status of the patient. The reactivated virus herpes zoster ophthalmicus. Delayed hemiplegia fol-
may cause symptoms in elderly immunocompetent lowing herpes zoster ophthalmicus has often been
individuals or in immunocompromised patients and considered to be the equivalent of granulomatous
may produce disease of the brain and spinal cord. Re- angiitis. Sufficient clinical differences, however, exist
cently varicella-zoster virus has been detected in to justify the assumption of two distinct entities.
blood vessels and other tissues by PCR, and this has Symptoms tend to be milder in this disorder than in
widened the clinical spectrum of acute and chronic primary granulomatous angiitis of the nervous sys-
disorders ascribed to varicella-zoster virus, including tem. Most patients with this disorder survive, even
large vessel granulomatous arteritis, myelitis, and without steroids.
small vessel encephalitis, all of which may occur with- Microscopic tissue examination reveals the same
out the rash typical of varicella-zoster. histological abnormalities in both disorders.
Varicella-zoster becomes latent in ganglia along MRA is nonspecific, but will show the unilateral in-
the entire neuraxis. Unlike herpes virus, it cannot be volvement of the brain (Fig. 100.21).
cultivated from human ganglia. With special tech-
niques, such as in situ hybridization, Southern blot, 100.2.2.4 Drug-Related Vasculitis
and PCR analysis, viral DNA has been found in There is a long list of drugs that may cause drug-in-
trigeminal and thoracic ganglia. Early reactivation of duced vasculitis, including therapeutic and diagnos-
the virus after chickenpox with an interval of only tic pharmaceuticals and substances used in drug
several months, associated with granulomatous ar- abuse. To the first category belong, among many oth-
teritis of the medium-sized vessels, may be seen in ers, allopurinol, amphetamine, ergot alkaloids,
children presenting with an infarction. These infarc- ephedrine, ginseng, interleukin-2, methylphenidate,
tions may be located in the posterior fossa, with basi- oxymethazoline, penicillin, and hepatitis B vaccine.
lar artery arteritis, or, more often, in the carotid artery To the second category belong cocaine, heroin,
territories, causing either larger territorial infarctions methylphenidate, and methamphetamines. In many
or infarctions in the territories of the lenticulostriate cases of the first group the vasculitis is restricted to
arteries. Myelitis in immunocompetent individuals is the skin, but extension to other organs is very well
usually less severe than in immunocompromised pa- possible. Extension to the brain is rare.
tients. In the latter group small vessel encephalitis Cocaine may lead to vasculitis, vasospasm, and in-
may develop, with poor prognosis. creased platelet aggregation resulting in infarctions
MRI is not specific in varicella-zoster vasculitis, (strokes), leukoencephalopathy, and hemorrhages.
but may show resulting infarctions. MRA may indi- Chronic cocaine dependency has also been linked to
cate the affected middle-sized vessels. a moyamoya-like vasculopathy, with obstructed ves-
There is no curative therapy for the vasculitis, sels and extensive collateral circulation, in particular
which is in most cases self-limiting. Supportive mea- the basal ganglia. The risk of myocardial infarction is
sures depending on the neurological symptoms are increased in young individuals using cocaine.
indicated. In heroin abuse strokes and hemorrhages are less
Delayed contralateral hemiplegia following herpes frequent than in cocaine abuse. A spongiform
zoster ophthalmicus tends to occur in middle age leukoencephalopathy is far more frequent in heroin
798 Chapter 100 Vasculitis

Fig. 100.22. A 43-year-old woman presenting with a series of matter and deep white matter. This pattern can be seen in
transient ischemic attacks, all related to different brain local- intravascular coagulation, for example in sepsis, but also in
izations. T2-weighted images of the brain show multiple le- intravascular metastases.In this case the cause was intravascu-
sions of different sizes involving cortex and subcortical white lar lymphomatosis

abuse (see Chap. 87), but has occasionally been de- IVL have been described in the age range of 30–80
scribed in cocaine abuse. years. The clinical signs and symptoms of IVL may
MRI patterns in drug-related vasculitides are in- mimic those of vasculitis, and the cerebral form has a
consistent and depend on the vessels involved. Le- variable presentation. Often there are stroke-like
sions will usually have the features of infarctions. Dif- episodes or repetitive transient ischemic attacks. Fo-
fusion-weighted imaging, therefore, should be part of cal cortical symptoms, confusion, disorientation, de-
the MR protocol. mentia, and seizures may follow. Meningoencephali-
tis-like presentation has also been described.
Proper diagnosis is important, because IVL is a
100.2.3 Disorders Primarily Obstructing treatable condition and early diagnosis may prevent
the Vessel Lumen irreparable damage. Manifestations in other organs –
skin or lungs, for example – anemia, and high ery-
Hypercoagulative states. There are a number of inher- throcyte sedimentation rate may help to arrive at the
ited and acquired disorders that lead to a change in correct diagnosis. Usually the diagnosis can be made
the complex process of coagulation, which may in- by examination of blood or bone marrow samples,
voke hemorrhages when hemostasis is insufficiently biopsies of skin lesions, or bronchial lavage in pul-
controlled or thrombosis with obliteration of vessel monary manifestations.
lumina when coagulation surpasses its necessary bio- Cerebral manifestations as seen on MRI include
logical function and causes unsolicited thrombosis. infarct-like lesions dispersed throughout the brain
Such inherited factors are: elevated von Willebrand (Fig. 100.22), venous occlusion of the large sinus or
factor, factor V Leiden, fibrinogen polymorphisms, cortical veins with venous infarctions, and enhance-
protein C and protein S deficiencies, and many others. ment of leptomeninges after contrast injection. Diffu-
In various conditions hypercoagulation may be in- sion-weighted imaging may help to indicate the na-
duced by changes in the blood composition, as may ture of the cerebral lesions and identify them as in-
happen in cancer patients, and in postoperative and farctions. MRA may show abnormalities of arteries
post-traumatic conditions with extended immobi- but is not diagnostic. In cases where there is venous
lization. The resulting lesions will occur in unpre- involvement, MR phlebography is useful to show the
dictable places and therefore neurological signs and obstructed veins.
MR findings are not specific. Treatment consists of polychemotherapy.
Intravascular lymphomatosis (IVL) is a very rare The name of sickle cell disease stems from the ab-
non-Hodgkin lymphoma characterized by prolifera- normal form of the red blood cells, which look like
tion of lymphoma cells in the vascular lumina, both crescents. It is an autosomal recessive disease which
arterial and venous, without involvement of adjacent has its highest incidence in Africa. It is caused by a de-
parenchymal tissue. IVL with cerebral involvement is fect in the b-hemoglobin gene. Sickle cell anemia will
predominantly of B cell lineage. Patients with cerebral develop in homozygously affected individuals. Het-
100.3 Magnetic Resonance Imaging 799

erozygous carriers will have some abnormal erythro- hemoglobin causes the red blood cells to stop sick-
cytes, but symptoms will be only precipitated under ling. It has been proven effective in preventing crisis.
conditions with low oxygen tension, such as in high- The long-term side effects of this medication are,
lying geographical regions. Sickle cells are less prone however, still unknown. Animal experiments with
to parasitic (malarial) infection than normal blood gene replacement of the defective gene have given
cells. It is hypothesized that in former centuries carri- new hope.
ers of sickle cell disease were more likely to survive
malaria and were responsible for the recurrence of
sickle cell carriership in subsequent generations. To- 100.3 Magnetic Resonance Imaging
day the disease affects 1 in 500 newborns with Afro-
American parents and 1 in 1000 newborns with His- Vasculitis can lead to a wide variety of MRI patterns.
pano-American parents in the USA. The disease is The lesions can be focal, asymmetrically distributed
found everywhere in Africa, throughout Middle and throughout the brain parenchyma, sometimes mim-
South America and Cuba, and in Mediterranean icking multiple sclerosis, or lesions can be more con-
countries such as Italy, Greece, and Turkey. fluent. Infarctions may occur in vascular territories or
The clinical picture is highly variable, as many dif- border zones, or they may present as small lacunar in-
ferent organs may suffer ischemic insults. The nature farctions. Involvement of the basal ganglia, thalamus,
of the disease, however, is such that it often causes midbrain, pons, medulla oblongata, and leptomenin-
cerebrovascular accidents. Sickle cell disease is one of ges is often seen. Lesions may become hemorrhagic
the many sources of watershed and arterial territori- and macro- and microhemorrhages may occur. With
al infarctions in children and relatively young adults. cortical and leptomeningeal localizations superficial
Neurological symptoms depend on the location of the hemosiderosis may follow. In later phases of the dis-
infarctions. ease atrophy may become apparent.
At the molecular level sickle cell anemia is caused It is evident that the MRI pattern of vasculitis is not
by a single base mutation leading to the substitution specific. Disorders which may share this gamut of
of valine for glutamic acid at position 6 of the b-he- MRI patterns include multiple sclerosis, acute dis-
moglobin molecule. Homozygously affected individ- seminated encephalomyelitis, extrapontine myelino-
uals have abnormal hemoglobin, hemoglobin S. Un- lysis, progressive multifocal leukoencephalopathy,
der circumstances with low oxygen tension and radiation vasculopathy, subcortical arteriosclerotic
acidosis the abnormal hemoglobin molecules poly- encephalopathy (Binswanger disease), eclampsia,
merize, resulting in increased cell rigidity and the neoplasms and emboli. Clinical and laboratory data
characteristic sickle shape. This leads to sludging are helpful in further differentiation of these disor-
in smaller vessels and to hemolysis. The majority of ders.
strokes occur in younger patients. Many factors can Some of the vasculitic disorders may present with
precipitate stroke, such as dehydration, pregnancy, a more characteristic MRI pattern. Vasculitis in SLE
low hematocrit value, cardiomegaly, and abnormal can lead to extensive calcifications, which may be
liver functions. prominent in the basal ganglia, the geniculate bodies,
Sickled cells are less flexible, which slows their pas- and the dentate nucleus. On T1-weighted images this
sage through the vessels. The red blood cells contain- may lead to high signal intensity in the involved areas,
ing the abnormal hemoglobin S seem also to be ab- sometimes blotting out the basal ganglia. This phe-
normally adherent to the endothelium. Because of the nomenon may also be seen in such disorders as
stasis, platelets will also become more adherent, and hypoparathyroidism, pseudo-hypoparathyroidism,
fibrin deposition follows, with subsequent occlusion liver failure, and parenteral nutrition. The signal
of the lumen. Ischemic infarctions account for 70% of intensity on T2-weighted images is decreased. These
cerebrovascular episodes, and these infarctions are calcifications are largely symmetrical, but there seem
most often caused by occlusion of large arteries at the to be exceptions to this rule.
base of the brain. Occlusion of smaller vessels most An MRI protocol covering the features of vasculitis
often involves smaller cortical branches. should include T1- and T2-weighted, FLAIR images,
On MRI lesions may include small, disseminated and diffusion-weighted imaging (Trace images and
infarctions, border zone or territorial infarctions, and ADC maps). In addition gradient echo images are
in rare cases thrombosis of the large venous sinuses. mandatory to note the hemosiderin deposits, which
MRA may show obstructed vessels. are ubiquitous in most cases of vasculitis. MRA can
The anticancer drug hydroxyurea has given pa- be helpful, but a contrast-enhanced high-resolution
tients with sickle cell disease some hope. It was found (512 ¥ 512) sequence should preferably be used. We
that taking this drug caused an increase in the pro- prefer slow machine injection of 15–20 ml contrast,
duction of fetal hemoglobin, which is normally pre- about 1 ml per second, followed by 20 ml of saline,
sent only in newborns. The presence of sufficient fetal during the MRA acquisition, with a presaturation slab
800 Chapter 100 Vasculitis

blocking the larger venous sinuses. With this proto- may lead to the correct diagnosis. It should be noted
col, MR images will show the central cerebral vessels that in most of the vasculitic disorders, but in partic-
as well as their second and third ramifications. After ular in SLE and Behçet disease, mild to extensive
this procedure a contrast-enhanced, fat-suppressed, white matter disease can be found in absence of clin-
T1-weighted image is obtained in at least one direc- ical neurological abnormalities. The reverse is also
tion to identify enhancement of lesions, a frequent true: in SLE, despite evident neurological manifesta-
observation in vasculitis. tion, the findings at MRI and autopsy may be disap-
When unusual MR patterns are present, vasculitis pointing. Quantitative estimation of CNS involve-
should be always be considered a possible cause, espe- ment may be reached by magnetization transfer ratio
cially with clinical suspicion. The combination of histogram analysis, diffusion tensor imaging, ADC
MRI findings with the clinical and laboratory data mapping, and MRS.
Chapter 101

Leukoencephalopathy and Dural Arteriovenous Fistulas


R. van den Berg, G.J. Lycklama à Nijeholt, and J.M.C. van Dijk

101.1 Clinical Features 101.2 Pathology


and Laboratory Investigations
Histopathologically, DAVFs are located within the
Cranial dural arteriovenous fistulas (DAVFs) repre- wall of the sinus. The fistula itself has no intervening
sent 10–15% of all intracranial arteriovenous lesions. capillary bed or nidus and consists of small venules.
The exact etiology of cranial DAVFs is still unknown. Intimal hyperplasia of both dural arteries and veins is
Development of DAVFs has been described after noticed. The arterioles show hypertrophied walls,
surgery, head trauma, and in relation to dural sinus mainly characterized by media hyperplasia. Orga-
thrombosis. In adults, it is generally accepted that nized thrombi have been demonstrated in the dural
DAVFs are acquired conditions. Pediatric cases are sinuses in up to 100% of cases.
rare; DAVFs have infrequently been demonstrated in Immunohistochemically, DAVFs show strong
utero and may be present in neonates, associated with staining for basic fibroblast growth factor (bFGF) in
a dural sinus malformation. the subendothelial layer and hypertrophied media of
Clinical symptoms of DAVFs are related to the the arteries in the sinus wall and in the fibrous
fistula itself, e.g., pulsatile tinnitus, or to the venous connective tissues around the sinuses, sparing the
hypertension in the involved venous territory. The endothelium. Vascular endothelial growth factor
clinical symptomatology and the risk of aggressive (VEGF) stains positively in the endothelium of the
complications, such as intracranial hemorrhage, cor- dural sinus, small arteries, and veins in the sinus
relate directly with the venous drainage pattern of walls. In addition, VEGF stains positively in the en-
DAVFs. Depending on the venous drainage and the dothelium of many capillaries in the sinuses that are
flow characteristics, DAVFs may cause orbital symp- obstructed by an organized thrombus. No such find-
toms including exophthalmia and cranial nerve ings are encountered in dural sinuses of control spec-
deficits. A focal area of cortical venous reflux – retro- imens. The factors stimulating bFGF and VEGF are
grade drainage in a cortical leptomeningeal vein – not known, but it is postulated that tissue hypoxia
may cause focal neurological deficits, such as aphasia and or intraluminal shear stress resulting from ve-
or motor weakness. DAVFs can also cause remote nous hypertension stimulates the expression of these
symptoms. Lesions located in the cavernous sinus, angiogenic growth factors.
tentorium, and foramen magnum can cause venous The acute and chronic parenchymal abnormalities
congestion of the brain stem or spinal cord with relat- in the case of cortical venous reflux (aggressive type
ed symptomatology. The retrograde transmission of of fistulas) are related to venous hypertension. Acute
the arterial pressure in a more extensive venous net- changes include diffuse cerebral edema and petechial
work, in combination with impaired venous outflow hemorrhages within the gray and white matter.
through the sinuses, may lead to venous hyperten- Chronic changes include markedly dilated and thick-
sion, resulting in CSF absorption abnormalities and ened, hyalinized walls of the parenchymal veins. Glio-
papilledema. These venous pressure disturbances sis may occur within the white matter.
may lead to symptoms of parkinsonism, such as
rigidity, bradykinesia, and gait disturbances, and
global cognitive dysfunction with dementia as the 101.3 Pathogenetic Considerations
most severe presentation.
The presence of cortical venous reflux in cranial The association of DAVFs with venous thrombosis
DAVFs carries an annual mortality rate of 10.4%. The has been described frequently. Another important
annual risk of hemorrhage is estimated to be 8.1% factor contributing to the development of DAVFs is
per year. The risk of nonhemorrhagic neurological venous or sinus hypertension. Sinus thrombosis does
deficit is 6.9% per year. The annual event rate for not always lead to the development of DAVFs, and it
patients with aggressive DAVFs is therefore 15%. has been stated that venous hypertension is a pre-
requisite for formation of a DAVF, even in the absence
of sinus thrombosis. DAVFs have also been reported
to develop following intracranial surgery, trauma,
surgery in remote areas of the body, and in the post-
802 Chapter 101 Leukoencephalopathy and Dural Arteriovenous Fistulas

Table 101.1. Classification of cranial dural arteriovenous fis- drainage. This causes chronic (venous) ischemia. In
tulas those cases where extensive reflux is seen in combina-
Borden classification tion with impaired venous outflow through the sinus-
es, venous pressures can rise to very high levels.
1. Venous drainage directly into dural venous sinus
or meningeal vein
2. Venous drainage into dural venous sinus 101.4 Therapy
with cortical venous reflux
3. Venous drainage directly into subarachnoid veins
(only cortical venous reflux)
The natural history of the disease is related to the ve-
nous drainage pattern of the DAVF. DAVFs with ante-
Cognard classification grade drainage only present with focal symptoms due
to the fistula itself, and morbidity and mortality are
I. Venous drainage into dural venous sinus limited. In these cases treatment should aim only at
with antegrade flow diminution of the focal symptomatology. DAVFs with
IIa. Venous drainage into dural venous sinus cortical venous reflux, however, may lead to severe
with retrograde flow complications and require aggressive treatment. Dis-
IIa-b. Venous drainage into dural venous sinus connection of the cortical venous reflux is obligatory
with retrograde flow and cortical venous reflux
to protect the patient from sequelae such as intracra-
IIb. Venous drainage into dural venous sinus nial hemorrhage and nonhemorrhagic neurological
with antegrade flow and cortical venous reflux
deficits. Partial treatment will not reduce the risk of
III. Venous drainage directly into subarachnoid veins occurrence of these complications.
(only cortical venous reflux)
Both endovascular embolization and surgery are
IV. Type III with venous ectasias of the draining
subarachnoid veins available to treat DAVFs. Surgical disconnection of
the fistulous vein(s) used to be the gold standard for
treatment, but with the introduction of liquid adhe-
sive embolics, which have been shown to produce a
partum period. The wide variety of etiological factors durable result without recanalization, these tech-
should not be regarded as direct causes of DAVFs; niques should be regarded as equal. The choice of
rather, they represent environments that may be con- treatment mode should be decided by the interven-
ducive to the development of a DAVF in particular tional neuroradiologist and the neurosurgeon.
patients. If endovascular treatment is chosen, the arterial
Two principal theories on the pathogenesis of route is used preferentially. The goal of arterial em-
DAVFs have been proposed. The first claims that bolization, in which a liquid embolic agent such as n-
DAVFs are caused by enlargement of preexisting mi- butyl cyanoacrylate (NBCA) should be regarded as
crofistulas in the dura. These microfistulas enlarge superior to polyvinyl alcohol (PVA) particles, is to
because of increased venous pressure, associated ei- penetrate the fistulous point and to occlude the prox-
ther with sinus thrombosis or with sinus outflow ob- imal part of the refluxing vein. If this cannot be ac-
struction. The second theory points to the develop- complished, occlusion of the fistulous vein through
ment of angiogenic factors, such as the above men- an alternative route is indicated. The least aggressive
tioned bFGF and VEGF, either directly from the orga- is selective disconnection of the refluxing vein(s).
nization of a sinus thrombosis or indirectly from This will leave the DAVF itself untreated. If the fistu-
local tissue hypoxia due to an increased venous pres- lous zone is more extensive and the cortical venous
sure. reflux is found on multiple sites of the dural sinus, or
Many classifications have been proposed for if venous hypertension is the main problem, oblitera-
DAVFs, of which the Borden classification and the tion of the sinus may be the only treatment option
Cognard classification are most commonly used. The left. However, first a thorough angiographic examina-
common concept of both classifications is to differen- tion of the venous drainage of the normally draining
tiate between DAVFs with antegrade drainage in the veins, including the veins of the posterior fossa, is
dural sinus (benign type: Borden 1, Cognard types I necessary. Sacrifice of the dural sinus can only be per-
and IIa), and DAVFs with cortical venous reflux (ag- formed if adequate venous drainage of the brain is
gressive type: Borden 2 and 3, Cognard type IIb and guaranteed. Only if the sinus is no longer used for the
higher) (Table 101.1). The importance of the venous drainage of the brain parenchyma, and the veins of
drainage pattern lies in the correlation with clinical the posterior fossa do not enter the involved segment,
symptomatology and the risks of hemorrhage. The the sinus can be sacrificed.
retrograde transmission of the arterial pressure in Selective disconnection of the cortical venous re-
the venous network leads to venous hypertension flux or sacrifice of (a part of) a dural sinus can also be
and congestion and impairs parenchymal venous performed by an open surgical approach or by a com-
101.5 Magnetic Resonance Imaging 803

bination of endovascular and surgical approaches. In


patients with occlusion of the affected sinus at both
proximal and distal sites (isolated sinus), a direct ap-
proach to the diseased sinus can be obtained through
a small burr hole that allows direct puncture of the
dural sinus. Subsequently the fistulous zone of the
DAVF can be closed using coils or other thrombo-
genic material.

101.5 Magnetic Resonance Imaging

In the presence of a DAVF with cortical venous reflux,


unenhanced CT images may show hypodensities, rep-
resenting areas of gliosis, edema, or venous ischemia.
Abnormally enlarged pial veins can be depicted due
to their increased density compared to the brain
parenchyma (Fig. 101.1). Contrast-enhanced CT will
show extensive enhancement of the enlarged pial
venous network.
In the absence of cortical venous reflux, a DAVF
might be occult on MRI. In such cases, the location of
the DAVF within the dura and the lack of a mass effect
on the brain parenchyma make it very difficult to see
the nidus of the fistula on MRI. MRA is more sensitive Fig. 101.1. A 57-year-old man presented with rapid cognitive
in depicting the nidus, although the lack of flow infor- decline related to a DAVF located in the torcular region. On the
mation in time is one of the drawbacks. MRA after plain CT scan dilated pial veins are demonstrated in the right
injection of a bolus of intravenous contrast may solve temporal region
this limitation.
In DAVFs with cortical venous reflux MRI shows
prominent flow voids on the surface of the brain
corresponding to dilated cortical vessels, or more
subtle serpiginous or dot-like vascular structures
(Fig. 101.2). These are highly suggestive of the correct
diagnosis. Hydrocephalus secondary to the venous
hypertension in the superior sagittal sinus can be pre- Angiography is obligatory to confirm the diagno-
sent. The brain parenchyma, particularly the white sis DAVF and for treatment planning. Selective con-
matter, may show T2 hyperintensity (Fig. 101.2). This trast injections into the different branch arteries of
is related to venous hypertension and congestion of the external carotid artery will reveal rapid arteriove-
the brain in the earlier stages and gliosis in later nous shunting through the fistula into the cerebral ve-
stages. The cerebral involvement may be extensive nous system, thereby arterializing the venous system.
(Fig. 101.2) or focal (Fig. 101.4). Dependent on the lo- Important concomitant findings are outflow obstruc-
calization of the shunt, the cerebellum, deep gray nu- tion due to venous sinus occlusion, which can result
clei, or brain stem may be affected. DAVFs have been in extracranial drainage via collateral routes, includ-
described as the cause of bilateral thalamic hyperin- ing the orbital system, and which augments the risk of
tensities on T2-weighted images. retrograde flow into the cortical and cerebellar veins.
The differential diagnosis in bilateral thalamic T2 The transit time of contrast when injected selectively
hyperintensities should include basilar artery infarc- into the internal carotid artery is delayed, compatible
tion, tumor infiltration, and deep venous occlusion. with venous congestion. In the normal situation
The differential diagnosis of more diffuse T2 hyperin- venous drainage is seen 4–6 s after the beginning of
tensities would include superior sagittal sinus throm- the arterial phase. In addition to the analysis of the
bosis with venous congestion, diffuse glioma, and venous reflux, the venous drainage of the brain
other leukoencephalopathies. However, the combina- should be examined with the same diligence, not on-
tion of an abundance of dilated pial vessels, contrast ly to detect focal areas of delayed venous drainage,
enhancement of these vessels, and deep white matter but also to determine whether sacrifice of a dural
T2 hyperintensity is highly suggestive of a DAVF and sinus or a refluxing vein is a potential treatment
mandates further angiographic analysis. option.
804 Chapter 101 Leukoencephalopathy and Dural Arteriovenous Fistulas

Fig. 101.2.
101.5 Magnetic Resonance Imaging 805

Fig. 101.3. Angiography of the pa-


tient presented in Fig. 101.2. Selective
injection of the external carotid artery
(first row, left) shows hypertrophy of
the superficial temporal artery and
middle meningeal artery. Immediate
enhancement of the superior sagittal
sinus is seen, suggesting the presence
of a DAVF. Selective internal carotid
artery injection (first row, right; middle
row, left) shows filling of the artery of
the falx cerebri through a (hyper-
trophied) ophthalmic artery (first row,
right).There is early enhancement of
the superior sagittal sinus, consistent
with a DAVF.There is slow passage of
contrast through the brain parenchy-
ma because of venous congestion,
resulting in a late parenchymal and
venous phase.This is also illustrated
by the poor visibility of peripheral
arteries in the early phase (middle row,
left) and by the “pseudo-phlebitic”
aspect of the brain parenchyma in a
later phase of the angiogram (middle
row, right). After treatment by both an
endovascular approach (selective glue
injections in external carotid branches
feeding the arteriovenous fistula) and
by direct placement of coils in the
superior sagittal sinus (third row, left;
middle row, right), there is a marked
reduction in the size of external carotid
artery branches.The internal carotid
artery injection after treatment shows
abnormal drainage of the brain due to
occlusion of the superior sagittal sinus
(third row, right).The brain is no longer
using the superior sagittal sinus for its
drainage. However the transit time of
contrast is much shorter.The cognitive
symptoms of the patient have dis-
appeared almost completely

Fig. 101.2. A 67-year-old woman had in her medical history


an operation for an acoustic neurinoma on the right. She pre-
sented with a rapid cognitive decline.The T2-weighted images
(first two rows) show a diffuse signal increase in the deep white
matter of both cerebral hemispheres. Abnormal flow voids are
visible on the cerebral surface and in the brain parenchyma,
especially in the temporal lobes and in the posterior fossa.The
T1-weighted images after contrast (third and fourth rows) show
extensive enhancement of cortical and parenchymal veins
806 Chapter 101 Leukoencephalopathy and Dural Arteriovenous Fistulas

Fig. 101.4. FLAIR images of a 51-year-old patient who had a single epileptic attack with visual signs.The images show an area of
increased signal in the left occipital lobe (second row)
101.5 Magnetic Resonance Imaging 807

Fig. 101.5. Angiography of the pa-


tient presented in Fig. 101.4. Selective
injection of the left vertebral artery
shows no definite abnormalities
(first row), except for nonenhancement
of the left transverse and sigmoid
sinus. Selective external carotid artery
injection shows early filling of a dural
tentorial sinus, located in the tentori-
um cerebelli, due to a DAVF located
there.The fistula is fed by a trans-
mastoid branch of the occipital artery
(second row, right).There is reflux from
the dural sinus into cortical veins
in the area (third row), leading to
local venous congestion
Chapter 102

Leukoencephalopathy After Radiotherapy


and Chemotherapy

102.1 Clinical Features Late delayed reactions are generally irreversible.


and Laboratory Investigations The process begins insidiously with personality
changes, gradually progressing over several months.
In the treatment of malignancies three modalities Initially there is excessive drowsiness and loss of
play a major role: surgery, radiotherapy, and chemo- initiative and interest. In the course of time there is a
therapy. Other treatment modalities such as hyper- decrease of cognitive functioning, confusion, irri-
thermia and laser coagulation, often image-guided, tability, and memory loss, eventually leading to glob-
are of some, but lesser importance. Radiotherapy and al dementia.
chemotherapy are not only applied in the treatment This classical description is used to describe time-
of primary brain tumors and metastases, but in- linked reactions after radio- and chemotherapy. In
trathecal and intravenous administration of chemo- addition, the “focal radiation injury” and “focal white
therapeutic drugs, together with cranial or total neu- matter injury” resulting from stereotactic radio-
raxis irradiation, are also widely used in the prophy- surgery, gamma knife surgery, or intensity-modulat-
laxis of cerebral involvement in extracerebral malig- ed radiotherapy may be seen as fitting within the
nancies. classical concept. Clinically some of these injuries
For many years the brain was considered to be rel- have important consequences. Focal radiation injury
atively resistant to therapeutic doses of irradiation caused by irradiation of extracranial structures, e.g.
and chemotherapy, because neurons do not multiply of nasopharyngeal squamous cell tumors or pituitary
and the turnover of glial cells is relatively slow. Also, tumors, may lead to damage of the brain – in these
the blood–brain barrier may prevent the penetration cases, damage to both temporal lobes, resulting in a
of chemotherapeutics into the brain. These concepts complex behavioral change, the Klüver–Bucy syn-
have to be modified, because it has become clear that drome.
adverse effects are not exceptional. There are a number of irradiation- and chemo-
In the classical description of the effects of radio- therapy-related patterns that need special considera-
therapy three types of damage are distinguished ac- tion because of their consequences for the prognosis
cording to their time of occurrence: acute reactions, and sometimes the need for therapeutic manage-
which occur during the time of treatment and may ment. This holds especially for multifocal inflamma-
change the treatment schedule; early delayed reac- tory leukoencephalopathy (MIL), and the posterior
tions, which are usually transient and appear from a reversible encephalopathy syndrome (PRES). MIL
few weeks to a few months after treatment; and late and PRES are addressed separately in Chaps. 88 and
delayed reactions, with onset from several months to 92.
several years after treatment.
Acute reactions are usually mild and of little con-
sequence, but severe reactions may occur. Clinically 102.2 Pathology
they may present as mild signs of increased intracra-
nial pressure. The patient may become confused, in- Neuropathological changes in acute reactions after
coherent, and disoriented. In more severe cases the radiotherapy are primarily characterized by cerebral
patient suffers from headaches, nausea, vomiting, and edema, with flattening of gyri, obliteration of sulci,
sometimes elevation of body temperature. Seizures and signs of tentorial herniation. The lateral ventri-
occasionally occur, and the patient may lapse into co- cles are narrowed. Vascular changes are present and
ma. Discontinuation of the treatment and corticos- consist of fibrinoid necrosis and thickening of the en-
teroid administration may be necessary and life-sav- dothelium with extravasation of fibrinous material
ing. and perivascular lymphocytic infiltration.
Early delayed reactions are usually transient and Because the early delayed reaction is usually tran-
disappear without treatment. Various clinical symp- sient and nonlethal in nature, the amount of informa-
toms have been reported: somnolence, nausea, vomit- tion available on the histopathological features is lim-
ing, dysarthria, dysphagia, cerebellar ataxia, and nys- ited. Foci of demyelination with central necrosis and
tagmus. petechial hemorrhages have been described. Lym-
phocytes and plasma cells are found in the perivascu-
102.3 Pathogenetic Considerations 809

lar spaces, and there is pronounced microglial and as- chloroethyl-nitrosourea (BCNU), methotrexate, and
trocytic proliferation in the affected areas. Vascular cisplatin. They enhance the effect of radiation-in-
changes are not prominent in the affected areas, but duced changes. On the other hand, radiation may in-
occasionally the lesions are more marked and show duce changes in the permeability of the blood–brain
fibrinoid necrosis, fibroproliferative vessel thicken- barrier and thus affect the delivery of potentially tox-
ing, enlargement of endothelial cells, and capillary ic agents. Other factors in the patient, such as nutri-
proliferation. The cytoarchitecture of the gray matter tional status, type of primary malignancy, or pre-
is usually intact. transplant status, and the presence of a paraneoplas-
The neuropathological findings of the late delayed tic syndrome, may well contribute to the development
leukoencephalopathy are rather specific. The changes and severity of cerebral lesions.
consist of demyelination, astrogliosis, multifocal co- The acute post-therapy syndrome is thought to be
agulative necrosis, and cavitation. The periventricular due to vasogenic edema resulting from damage to the
white matter and the centrum semiovale are involved capillary endothelium.
bilaterally, whereas the subcortical arcuate fibers, the Early delayed reactions are believed to be due to
cerebral cortex, and deep gray matter structures are demyelination and may be reversible to some degree.
usually spared. Within and around the necrotizing There are suggestions that they may be the result of
lesions conspicuous swelling of axons occurs. An in- an autoimmune reaction following sensitization for
flammatory response is usually absent. There are some myelin antigen that has become exposed by
marked vascular lesions, characterized by hyaliniza- therapy-induced tissue necrosis. Antigens are re-
tion, fibrosis, and necrosis of vessel walls and vascu- leased in the intracellular spaces by damaged myelin
lar thrombosis. Areas of endothelial proliferation and and glial cells, and may evoke hypersensitivity reac-
various degrees of adventitial fibroblast proliferation tions. This hypothesis has never been proven, but the
are found. Obliteration of the lumen may result. De- perivascular inflammatory reaction may be an argu-
position of iron salts and calcium in the vessel walls ment. Another mechanism which may account for
may occur. Calcifications may be extensive and be the marked demyelination in the absence of vascular
present in the subcortical areas, the basal ganglia, changes in early delayed post-treatment leukoen-
and, less commonly, in the pons. It is noteworthy that cephalopathy is primary damage to glial cells, in par-
mineralizing angiopathy is more often seen in chil- ticular oligodendrocytes. Sometimes striking glial
dren than in adults. proliferations are noticed, associated with bizarre
The installment of prophylactic irradiation and cells and giant multinuclear astrocytes, supporting
radiotherapy for acute lymphatic leukemia and the this hypothesis.
treatment of genetic disorders with bone marrow In the late delayed reactions vascular changes with
transplantation have focused attention on the side secondary ischemic changes form the most likely ex-
effects of these therapies. In most patients these reac- planation for the tissue damage. The endothelium of
tions are transient and have the nature of temporary blood vessels is one of the most sensitive tissues of the
white matter edema, as has been confirmed by an brain. Damage to the endothelium leads to endothe-
occasional biopsy, and more recently by quantitative lial proliferation and changes in the vessel wall, with
MR data. Multifocal white matter necrosis has the subsequent obliteration of the lumen and ischemia.
same neuropathological features as the radiation Small vessels are usually most affected, but larger ar-
necrosis described above for late delayed post-irradi- teries may also suffer, and may be partially or totally
ation reactions. occluded, possibly with formation of moyamoya-like
collaterals.
It is important to realize that the adverse conse-
102.3 Pathogenetic Considerations quences of irradiation of the brain, in particular in
combination with chemotherapy, may be more severe
Cranial irradiation and systemic, intracarotid, intra- in infants and children than in adults. The conse-
venous, and intrathecal chemotherapy, alone or in quences for the immature brain may differ from those
combination, may result in lesions of the CNS, of for the more mature brain. From the management of
either the white matter, the gray matter, or both. In medulloblastomas in young children it has become
cases of combined therapy it is impossible to delin- clear that aggressive treatment approaches, especially
eate the relative contributions of radiation and craniospinal irradiation, can harm the developing
chemotherapy to the development of cerebral lesions. brain. It is hard to predict what dose of radiotherapy
The total radiation dose and possible overdosage on will be harmful in each individual child. It is well
specific targets, as well as the dose of systemic intra- known that very young children will have significant
venously or intrathecally administered chemothera- learning problems after full-dose radiotherapy, and
peutics all influence the outcome. Some chemothera- that even older children may develop difficulties in
peutic agents are radiosensitizers, for example bis- school. However, a reduction in dosage may also re-
810 Chapter 102 Leukoencephalopathy After Radiotherapy and Chemotherapy

duce efficacy on the tumor. Hence, approaches using changes on T2-weighted images and subcortical hy-
reduced-dose craniospinal irradiation and chemo- perdensities on CT, consistent with calcifications. The
therapy, in order to reduce cognitive, endocrine, and authors attribute these changes to a mineralizing an-
psychological deficits, may decrease late effects, but giopathy, commonly thought to be the result of cra-
carry with them the risk of having more treatment nial irradiation in combination with chemotherapy.
failures. From this and other reports it has become clear that
The combination of radiotherapy and chemother- chemotherapy without irradiation may also lead to
apy leads to synergistic inhibition of the synthesis of leukoencephalopathy and mineralizing angiopathy.
macromolecules and DNA repair, possibly further
contributing to the damage. In a postmortem study of
children with childhood leukemia treated with differ- 102.4 Therapy
ent modes of application of this combined therapy,
comparing those who showed leukoencephalopathy In acute reactions corticosteroids are useful in allevi-
and those who did not show leukoencephalopathy, it ating cerebral vasogenic edema and may be life-sav-
became clear that the development of white matter ing in patients with imminent tentorial herniation.
damage did not correlate with age, despite the differ- The importance of recognizing early delayed reac-
ent stages of myelination and neuronal differentia- tions is the fact that they are usually transient and do
tion in the pediatric age group. Nor was there a rela- not necessarily require intervention or indicate a fail-
tionship with intercurrent infections, nutrition, or the ure of therapy. In the late delayed reaction corticos-
presence of CNS leukemia. There was a clear relation- teroids play a minor role. In cases with focal radiation
ship with the radiation dose the child had received in necrosis, surgery is an option. In patients with neuro-
combination with intrathecal methotrexate. The total logical syndromes caused by toxic effects of cytostat-
amount of intrathecally administered methotrexate ic or immunosuppressive drugs that may be re-
seemed less important. The dose of methotrexate was versible, abortion of the therapy or a switch to other
important when given intravenously: the incidence of drugs may have a beneficial effect. Psychiatric syn-
leukoencephalopathy increased with the total dose of dromes, especially when caused by more permanent
intravenously administered methotrexate. damage of both temporal lobes, as happens in the ir-
To add substance to this discussion, the multicen- radiation of nasopharyngeal and pituitary tumors,
ter study of the German Late Effects (of acute lym- are difficult to treat and may require special mea-
phatic leukemia treatment) Working Group, summa- sures.
rizes findings in a large population treated with stan-
dard protocols (Hertzberg et al. 1997). In this study
118 former patients with acute lymphatic leukemia in 102.5 Magnetic Resonance Imaging
first continuous remission underwent CT and/or
MRI. The group was subdivided into: group A (39 pa- MRI is generally the first choice of imaging modali-
tients), receiving intrathecal methotrexate and sys- ties in the follow-up of patients treated with cranial
temic medium–high dose methotrexate; group B (41 irradiation and/or chemotherapy, because its sensi-
patients), receiving cranial irradiation (16.8 Gy) and tivity is much better than that of CT. Only in the
intrathecal methotrexate or systemic methotrexate; detection of microcalcifications, such as occur in
and group C (38 patients), receiving irradiation mineralizing angiopathy, does CT have an advantage.
(17.1 Gy) and intrathecal methotrexate. Abnormal All stages of radiation and chemotherapy injuries
MRI and CT scans were found in 61 of the 118 pa-
tients, consisting of white matter changes (diffuse or
focal), brain atrophy, and calcifications. Of these 61 䊳

patients, 15 were from group A (38.5%), 23 from Fig. 102.1. A 62-year-old woman treated with chemotherapy
group B (56.1%), and 23 from group C (60.5%). Pa- and irradiation for a right frontal glioma. The initial tumor is
tients with definite CNS changes showed impaired barely visible on the FLAIR images (first three rows) within a
neuropsychological function. It is clear from this re- now much larger area of high signal involving the periventric-
port that methotrexate without irradiation can also ular and deep frontal white matter, right more than left, and
lead to serious CNS changes, both of gray and white the corpus callosum.T1-weighted images after contrast (fourth
matter. This has been reported in more detail by row) show a rim of a few millimeters’ thickness along the wall
Lövblad et al. (1998). They describe the cases of four of the left lateral ventricle as well as an enhancing dot in the
children treated with high-dose intravenous and in- right frontal area.The white matter changes can be attributed
trathecal methotrexate without irradiation. In all to the irradiation and chemotherapy.The enhancement,which
these cases the cure was prolonged, lasting for more did not change during further follow-up, probably represents
than 1 year. These children developed serious CNS either inactivated tumor tissue,or,more probably,radiation-in-
abnormalities with diffuse hyperintense white matter duced changes
102.5 Magnetic Resonance Imaging 811

Fig. 102.1.
812 Chapter 102 Leukoencephalopathy After Radiotherapy and Chemotherapy

Fig. 102.2. A 52-year-old male patient presented with a par- the irradiation field. Coronal T2-weighted images show the im-
tial seizure. He had been treated previously for a right-sided pact of the radiation on the right temporal lobe, involving
nasopharyngeal squamous cell carcinoma with partial exci- both white and gray matter
sion and radiotherapy.The right temporal lobe was included in

have in common an increase in free tissue and, some- ferentiate radiation necrosis from tumor recurrence.
times, intracellular water. The consequence of this is a Perfusion imaging and MRS have a prominent role
higher signal intensity on T2-weighted and lower sig- in distinguishing tumor recurrence from radiation
nal intensity on T1-weighted images. This signal be- necrosis. In necrotizing tissue perfusion is low,
havior may, however, reflect many forms of underly- whereas in tumor recurrence it is usually high. MRS –
ing pathology, such as impairment of the blood–brain if possible, chemical shift imaging to cover the whole
barrier due to endothelial damage and vasogenic area – shows in tumor recurrence high choline, lac-
edema, demyelination, gliosis, ischemia, and tissue tate, and often the presence of some residual brain
necrosis. Newer techniques, such as diffusion-weight- metabolites, for instance N-acetylaspartate in re-
ed imaging, perfusion imaging, and proton MRS, have duced concentration. In necrotic tissue N-acetylas-
been of considerable help in the determination of the partate is usually absent, as is choline, whereas lactate
structural tissue changes. Diffusion-weighted imag- is present. It is, however, not rare that both tissue
ing, in combination with calculated ADC maps, or necrosis and tumor recurrence are present at the
diffusion tensor imaging with the calculation of frac- same time. Microbleeds can be made visible on MR
tional anisotropy, have made it possible to differenti- images with gradient echo or hybrid spin-echo–gra-
ate between vasogenic edema and cytotoxic edema, dient-echo techniques.
and permits a better estimation of the prognosis of In the acute reaction MR findings are nonspecific.
the abnormalities found. Contrast administration The images may be completely normal or subtly ab-
plays an important role, even though it does not dif- normal with poorly defined multifocal areas of hy-
102.5 Magnetic Resonance Imaging 813

Fig. 102.3. A 4-year-old boy was treated prophylactically with cerebral peduncles in the midbrain. The T1-weighted images
irradiation and intrathecal methotrexate because of acute (second row) reveal cysts in the basal ganglia, indicating that
lymphocytic leukemia. The T2-weighted images (upper row) cystic necrosis has developed. With contrast, an enhancing
show involvement of the basal ganglia, internal capsule, and recurrent tumor is seen in the third ventricle

perintensity on T2-weighted images, most often in not reversible. Enhancing lesions within the white
both hemispheres. The abnormalities usually disap- matter correlate with tissue necrosis at autopsy.
pear spontaneously, given an uneventful clinical Transient white matter abnormalities are often
course. found in patients treated for acute lymphocytic
In early delayed reactions, occurring a few weeks leukemia with prophylactic cranial irradiation,
to months after treatment, the white matter changes chemotherapy, and bone marrow transplantation.
are also usually transient. Changes on MRI include The lesions are located in the periventricular area and
high signal intensity on T2-weighted or FLAIR images may be more or less extensive. There is no clear rela-
in the basal ganglia, the cerebral peduncles, and the tionship between the severity of the lesions on MRI
deep white matter. Diffusion-weighted imaging also and the clinical condition and outcome. Diffusion-
shows high signal intensity in these areas, with also weighted imaging and ADC maps may be helpful by
a high ADC value. The term “T2 shine-through” is showing the nature of the lesions. The lesions may
sometimes used for this phenomenon, but seems not remain visible for a long time after the clinical disap-
quite correct, because the combination of high signal pearance of symptoms.
on diffusion-weighted imaging and high diffusivity Late delayed reactions occur months to years after
may reflect an underlying condition, most probably, the initial treatment. Depending on the field of irradi-
but not only, vasogenic edema. It is in general an indi- ation the lesions are more focal (Figs. 102.1–102.3) or
cation of a benign nature of the lesion. In some pa- more generalized (Figs. 102.4 and 102.5). So-called
tients who develop a diffuse leukoencephalopathy a “diffuse radiation injury” can be caused by irradia-
few weeks to a few months after treatment, the course tion, by combined irradiation and chemotherapy, and
is not benign and the white matter abnormalities are by chemotherapy alone. The white matter lesions,
814 Chapter 102 Leukoencephalopathy After Radiotherapy and Chemotherapy

Fig. 102.4. A 29-year-old woman with disseminated breast also involving the external capsule and the temporal lobes. In
carcinoma. Metastases in the brain were treated with radio- the cerebellum there are multiple, asymmetrical lesions and
therapy and systemic cytostatic treatment. The T2-weighted linear high-signal bands in the cerebellar foliae. T1-weighted
transverse series 6 months after radiotherapy (upper two rows) images after contrast (third row) show multiple metastases
shows diffuse symmetrical deep white matter hyperintensity, and leptomeningeal carcinomatosis

when irradiation has covered the whole brain, are lo- ed, and FLAIR images. ADC values are usually only
cated in the white matter of both hemispheres and are slightly above those of normal cerebral tissue. Despite
symmetrical and confluent. Combinations of diffuse the sometimes extensive white matter lesions, pa-
radiation injury and recurrent tumor may occur tients may be asymptomatic. In more severe cases
(Fig. 102.3). The lesions of late delayed reactions have there may be slowing down of mental activity and
a high signal intensity on proton density, T2-weight- cognitive impairment. The most severe form of late
102.5 Magnetic Resonance Imaging 815

Fig. 102.5. A 24-year-old man was treated for acute lympho- arcuate fibers in parietal, frontal, and temporal lobes, as well as
cytic leukemia with chemotherapy and intrathecal methotrex- bilateral involvement of the posterior limb of the internal cap-
ate.Two months later he developed progressive encephalopa- sule, thalamus, corticospinal tracts in the midbrain, and both
thy. The T2-weighted images show extensive involvement of middle cerebellar peduncles

delayed reaction is necrotizing encephalopathy with A disseminated necrotizing leukoencephalopathy


areas of focal necrosis (Fig. 102.3). The borders of with characteristic contrast enhancement of the white
these lesions may enhance. To verify this diagnosis, matter has been reported in patients with acute
perfusion studies may be performed, showing re- lymphoblastic leukemia after intense chemotherapy
duced perfusion, or MRS, showing loss of metabolites with methotrexate and prophylactic cranial irradiation,
without a rise in choline concentration and with var- with either a fulminant or a less fulminant course. In
ious amounts of lactate. these cases methotrexate was delivered both intra-
816 Chapter 102 Leukoencephalopathy After Radiotherapy and Chemotherapy

Fig. 102.6.
102.5 Magnetic Resonance Imaging 817

Fig. 102.7. The T1-weighted images


of the same patient as illustrated in
Fig. 102.6, without (first row) and with
contrast (second row), reveal multiple
small punctate areas of contrast
uptake, suggesting perivascular
enhancement.These findings suggest
an underlying angiitis or vasculopathy

venously and intrathecally. MRI initially shows exten- enhancement disappears but atrophy sets in, leading
sive white matter abnormalities, with focal but sym- to death within a few years. The MR pattern differs
metrical enhancement. On follow-up studies the from that seen in autoimmune suppressive therapy-
related MIL.
Severe late delayed reactions may also develop
䊴 after treatment with a combination of irradiation,
Fig. 102.6. An 18-year-old girl was treated 7 years ago with chemotherapy, and bone marrow transplantation.
chemotherapy, irradiation, and autologous bone marrow Years after treatment, and following an initially good
transplantation for a non-Hodgkin lymphoma. She has been response, a progressive encephalopathy may develop
tumor-free since the treatment. However, since 1.5 years with loss of mental faculties and an array of neurolog-
after the treatment she has developed slowly progressive ical symptoms and ending in death. MRI shows white
encephalopathy with recently more rapid decline. The ence- matter abnormalities predominantly involving the
phalopathy was characterized by concentration and memory arcuate fibers and progressive atrophy (Figs. 102.6
problems, personality changes, subsequent global cognitive and 102.7).
impairment, and finally increasing ataxia and spasticity. The Mineralizing angiopathy is more often seen in chil-
T2-weighted images (first three rows) show extensive white dren than in adults. In children it is the most common
matter changes,especially involving the arcuate fibers.There is abnormality seen on MRI and/or CT. Calcifications
some cerebral atrophy with ventriculomegaly, including the are found in the subcortical white matter and some-
temporal horns, and widening of the subarachnoid spaces. times in the basal ganglia, in particular in the puta-
There are bilateral lesions in the thalamus. Diffusion-weighted men.
images (fourth row) at different b values and an ADC map Lesions after gamma-knife therapy, stereotactic
show increased ADC values in the abnormal white matter radiosurgery, and localized overdosage are not funda-
areas (ADC values in the affected region 1.24–1.55). These mentally different from the classic description. Here,
values, unfortunately, only reflect the final phase of the too, the whole gamut of reactions is possible: from va-
process. Because of the insidious start and progress of the sogenic transient edema, to severe white and/or gray
encephalopathy, initial ADC values are not available matter lesions, to a cavitating (leuko)encephalopathy.
Chapter 103

Gliomatosis Cerebri

103.1 Clinical Features 103.3 Pathogenetic Considerations


and Laboratory Investigations
No familial cases of gliomatosis cerebri have been
Several synonyms are used for this condition: diffuse reported. One study looking at the chromosomes
glioma of the brain, gliomatosis diffusa, gliomatous of cells of gliomatosis cerebri revealed that the
hypertrophy, blastomatous type of diffuse sclerosis, majority of the abnormal cells had the karyotype
and central diffuse schwannosis. 44 XY, (del(6)(q25), del(14)(q21), del(15;21)(q10;q10),
The clinical presentation is variable. Headache is add(18)(q22), del(19)(p12), add(20)(p13), -21.A small-
present in most cases, and may be accompanied by fo- er proportion of cells had 88 chromosomes with a
cal seizures, changes in mental state, psychiatric syn- doubling of the normal karyotype.With the exception
dromes, ataxia, dysphagia, dysphasia, and memory of the chromosome 6 deletion, these chromosomal
loss. The clinical picture suggests in most cases a mul- changes do not resemble those found in astrocy-
tifocal progressive disorder. Initially, however, symp- tomas, suggesting that gliomatosis cerebri is a sepa-
toms may be difficult to interpret, with only loss of rate entity (Hecht et al. 1995). Herrlinger et al. (2002)
concentration, behavioral problems, or psychiatric summarize the molecular genetic findings and find
symptoms. Some patients present with diminished that genetic alterations in diffuse gliomatosis are not
vision only, and papilledema is found at fundoscopy. different from those found in infiltrating astro-
The clinical symptoms are often discrepantly mild in cytomas. They conclude on these grounds that glio-
view of the extensive abnormalities found on MRI, matosis cerebri should be considered a particularly
but they are progressive and lead to death. The medi- invasive subform of glioma, rather than a distinct
an survival time is 14 months. tumor entity that is entirely different from other
Over the last 5–10 years there seems to have been cerebral gliomas. An extra argument for this is found
an increase in the number of cases of gliomatosis in the dedifferentiation of gliomatosis cerebri into
cerebri, and younger patients are affected than in for- anaplastic astrocytomas and glioblastoma multi-
mer years. The reason for this is unclear. forme.
In one report, gliomatosis cerebri was seen follow-
ing radiation and chemotherapy for a extraneural
103.2 Pathology metastasis of primary nongerminomatous germ cell
tumor in the pineal region, providing evidence for
Macroscopically, there is swelling of the involved exogenous induction of the tumor.
structures. Although all parts of the brain may be in-
volved, including the brain stem and cerebellum, 䊳

there is a preference for the central, periventricular Fig. 103.1. Series of transverse T2-weighted images in a 56-
areas and mesolimbic parts of the temporal lobe. The year-old woman (first two rows). She has a history of changes in
microscopic hallmark of gliomatosis cerebri is the personality over the past 2 years, leading to suspicion of fron-
presence of many moderately pleomorphic glial cells, totemporal dementia. The images show bilateral, nearly sym-
infiltrating pre-existing structures, without signifi- metrical signal abnormalities of the frontal lobes, connected via
cant destruction. Natural borders between structures the genu of the corpus callosum and spreading toward the
are not respected. Usually, these cells are astrocytic in deep insular structures on both sides.The affected corpus callo-
type and react positively with glial fibrillary acidic sum is markedly swollen.There is diminished gray–white matter
protein (GFAP). In other cases, the infiltrating cells distinction in the involved areas. The third and fourth rows con-
have oligodendroglia-like elements and only a few tain diffusion-weighted images (Trace diffusion-weighted im-
cells are GFAP-positive astrocytes. Locally further ages with b = 1000 in the third row; ADC maps in the fourth row).
dedifferentiation may take place, and some parts of The Trace diffusion-weighted images show a moderate increase
the lesion may progress to anaplastic astrocytoma or in signal intensity in the involved area, whereas the ADC values
glioblastoma multiforme. Finally, the entire neuraxis are too high, indicating increased mobility of water. Note that
may be involved. this combination of moderately high signal increase on Trace
diffusion-weighted images together with high ADC values in
the affected area does not reflect vasogenic edema
103.3 Pathogenetic Considerations 819

Fig. 103.1.
820 Chapter 103 Gliomatosis Cerebri

Fig. 103.2. Series of FLAIR images in a 25-year-old man with involvement of the temporal lobes. In many places where
biopsy-proven gliomatosis cerebri.The biopsy was taken from the tumor touches upon the cortico-subcortical junction, the
the left frontal lobe. In this case the abnormalities are mildly gray–white matter differentiation has disappeared
asymmetrical and have a prominent mass effect.There is clear

103.4 Therapy 103.5 Magnetic Resonance Imaging

Because neurosurgery is not an option in most cases, Most useful for the diagnosis of gliomatosis cerebri
radiotherapy and chemotherapy are the only remain- are proton density, T2-weighted, and FLAIR sequences.
ing therapies available. The results have been disap- They show increased signal intensity of the involved
pointing for many years, but with the drug temozolo- areas, with poor demarcation from noninvolved tis-
mide longer survival has been achieved. sue, and with mass effect. When the cortico-subcorti-
103.5 Magnetic Resonance Imaging 821

Fig. 103.3. This 17-year-old girl presented with bilateral pa- trast administration, showing some contrast enhancement in
pilledema and no other neurological signs. The FLAIR images the right frontal lobe. The two images on the right were ob-
(upper two rows) reveal an asymmetrical presentation of tained 1 h after the injection and show much more prominent
gliomatosis cerebri with most prominent abnormalities on the contrast enhancement. The latter images make clear that the
right. The third row shows coronal T1-weighted images with damage to the blood–brain barrier is far more extensive than
contrast. The left-hand image was obtained directly after con- the first postcontrast image would suggest

cal junction is involved, there may be a striking loss of will be locally narrowed, with distortion of its normal
demarcation of gray and white matter and the sugges- border (Figs. 103.2 and 103.3). In these centrally
tion of some swelling (Figs. 103.1–103.3). When the located cases, the corpus callosum is nearly always
periventricular structures are involved, the ventricle involved and may be severely swollen (Fig. 103.2).
822 Chapter 103 Gliomatosis Cerebri

Fig. 103.4. Follow-up study of the same girl shown in Fig. 103.3, 6 months after radiotherapy and chemotherapy. It is clear that
the process has advanced and the brain stem is seriously involved

There may be a striking symmetry of the abnormali- Diffusion-weighted imaging with ADC maps is
ties (Fig. 103.1), although this may disappear when useful, in combination with chemical shift imaging
the disease progresses. The disease may also be high- and perfusion imaging, to find the places with the
ly asymmetrical in the initial stage (Fig. 103.4). In highest malignancy to direct a brain biopsy, when
some patients the spread of the disease clearly follows considered necessary, or to direct a local boost or in-
white matter tracts, with less severe involvement of tensity-modulated radiotherapy. In the affected areas
gray matter structures. Focal calcification has been MRS shows decreased N-acetylaspartate, normal or
reported. increased choline, and, in some patients, strikingly
In most cases, there is no or only partial enhance- increased myo-inositol. Myo-inositol is exclusively
ment of the involved structures after contrast injec- present in glial cells.
tion. This, however, depends on the technique used. The MR pattern of the disease is nearly always di-
With triple-dose gadolinium one may see some agnostic. In cases with central location and symmet-
enhancement; a delayed scan (e.g., 1 h delay) may rical involvement of the thalamus, central venous
show enhancement in an unexpectedly large area thrombosis has to be excluded, for which purpose
(Fig. 103.3). Leptomeningeal dissemination may be MRA can be used. Postictal changes may mimic the
observed, the disseminated lesions showing contrast MR appearance of gliomatosis cerebri. In these cases
enhancement. the abnormalities disappear within 14 days.
Chapter 104

Diffuse Axonal Injury

104.1 Clinical Features spect. EEG will reflect the seriousness of the function-
and Laboratory Findings al disturbance of the brain, and will provide impor-
tant information. MRI, in particular with more recent
Head trauma, especially in motor vehicle accidents, MR techniques, such as diffusion-weighted imaging,
can lead to a wide spectrum of cerebral and intracra- perfusion imaging, and MRS, may be helpful in pre-
nial lesions, with usually different clinical presenta- dicting outcome.
tions (epidural and subdural hematomas, subarach-
noid and intracerebral hemorrhages, cerebral contu-
sions, generalized cerebral edema, and secondary 104.2 Pathology
phenomena, such as hydrocephalus, raised intracra-
nial pressure, and tentorial herniation), and different Pathological findings depend heavily on the time be-
findings on CT and MRI. tween the initial accident and the postmortem analy-
About 50% of patients with traumatic accelera- sis. Because of the many secondary reactions that
tion–deceleration injuries are diagnosed with diffuse occur after the initial trauma, histological findings
axonal injury (DAI), also called shearing injury. Clin- may be different in different stages. Unfortunately,
ically, DAI is characterized by loss of consciousness very often the time between the accident and death is
following the accident, usually without a lucid inter- not reported.
val, a very low score on the Glasgow Coma Scale, and In general, macroscopic findings may be normal or
discrepantly subtle abnormalities on CT scans. When reveal focal atrophy, either cortical or in the rostral
lesions are seen on CT, they consist mainly of pe- part of the brain stem. In early cases brain edema may
techial hemorrhages at the cortico-subcortical junc- be severe and tentorial herniation may be present.
tion, in the body or splenium of the corpus callosum, Microscopic examination may demonstrate that
and/or in the basal ganglia and brain stem. axons are torn completely, but more often the damage
Intracranial pressure measurements in patients is incomplete. Focal alterations in the axoplasmic
with DAI are, at least in the beginning, normal, in con- membrane may result in impairment of the axoplas-
trast to the case with most of the other brain injuries. mic transport. Swelling ensues and the axon is dis-
DAI may result in death. It is unclear how often sected. Early damage to the axons is shown by the
death occurs in DAI, but one may assume that it oc- presence of large numbers of eosinophilic and argy-
curs in a high percentage of the patients. When it rophilic bulbs on nerve fibers, forming the so-called
comes to a correct estimation of the frequency of retraction balls, the pathological hallmark of shear-
death, the problem is that numbers provided by dif- ing injury. Macroscopically, lesions in DAI are usually
ferent specialists – neurologists, traumatologists, and ovoid or elliptical, following the long axis of the
neuropathologists – are not consistent. In some re- injured axons. Their distribution is not uniform or
ports (by neurologists) it is stated that DAI rarely re- symmetrical, but they occur particularly at the junc-
sults in death, whereas others (neuropathologists) tion of gray and white matter, in the corpus callosum,
claim that DAI is an important cause of death in accel- septum, fornix, internal capsule, deep gray matter,
eration–deceleration trauma. This difference is at tegmentum, and cerebellar foliae dorsal to the dentate
least partly due to the different populations they en- nuclei. The lesions are often hemorrhagic; the hemor-
counter.About 90% of patients with a clinical diagno- rhages occur in a linear pattern, following the distor-
sis of DAI, however, will remain in a vegetative condi- tion of layers. In later stages, atrophy dominates the
tion. picture. Of the basal ganglia, the lateral and ventral
In the first episode patients with DAI stay in the in- nuclei of the thalamus are most atrophic, usually with
tensive care unit, in many cases artificially ventilated. sparing of the anterior and dorsomedial nuclei, the
During this period it would be important for the pulvinar, the centromedian nuclei, and lateral genicu-
management of these patients to have a reliable pre- late bodies. Cholinergic neurons have been found to
diction of outcome, most importantly to maintain the be more susceptible than neurons belonging to other
option to discontinue treatment that will not further categories of neurotransmitters. Immunocytochemi-
improve the condition of the patient. Clinical and cal staining for b-amyloid precursor protein (b-APP)
neurophysiological data are important in that re- detects with great sensitivity axons that have im-
824 Chapter 104 Diffuse Axonal Injury

paired fast axonal transport. In normally functioning brain injury. Homovanillic acid, a breakdown product
axons b-APP is transported with fast axonal trans- of the adrenergic neurotransmitter systems, is signif-
port and never builds up to a concentration that al- icantly decreased soon after brain injury, and the
lows its detection in tissue samples. When this fast level correlates with the depth of coma.
axonal transport system is damaged, b-APP rapidly
accumulates in the disrupted segment. This accumu-
lation occurs before morphological methods detect 104.4 Therapy
the axonal damage. It is not known whether axonal
damage thus detected is potentially reversible. The Dopamine, one of the catecholamines, is an important
b-APP staining technique has demonstrated that neurotransmitter in the CNS. In DAI a reduction of
even in apparently minor head trauma damage may dopamine turnover has been found. This observation
occur to axons. In animal experiments there is a good has prompted the introduction of amantadine (Sym-
correlation between the amount of axonal damage metrel) therapy in DAI. Amantadine is a drug known
and the clinical outcome. from treatment of Parkinson disease. Amantadine
causes release of dopamine from central neurons,
facilitates dopamine release by nerve impulses, and
104.3 Pathogenetic Considerations delays the uptake of dopamine by neural cells. It may
also have a profound N-methyl-D-aspartate receptor
DAI is a shearing injury. It has been found experi- antagonist effect, which may contribute to the neuro-
mentally that shearing injury is not induced by linear protective effects after injury, by decreasing gluta-
or translational forces but rather by rotational forces. mate concentrations and thus excitotoxicity. In a
A sudden acceleration–deceleration impact can pro- randomized crossover design study in DAI patients,
duce rotational forces. Where the lesions occur de- there was a consistent trend toward more rapid func-
pends on the distance to the rotational center, the arc tional improvement with amantadine treatment,
of rotation, and the duration and intensity of the regardless of when during the first three months
force. Because of the relative fixation of certain parts after injury the amantadine treatment was started
of the brain to the rigid skull, the deep and superficial (Meythaler et al. 2002). From these findings it is also
portions may not move at the same rate and can even clear that medication that results in dopaminergic
move in different directions. This will result in shear blockade is contraindicated in the early stage of
strain that manifests across the axons and results in recovery from DAI.
axonal injury and rupture. Different brain parts have
different consistencies depending on cell composi-
tion and cell density. Injuries to the brain will be most 104.5 Magnetic Resonance Imaging
prominent at their junction, where differences in tis-
sue densities are greatest. One such vulnerable site is In emergency departments CT is usually the first
the gray–white matter junction, involved in 60–70% imaging modality used in cases of head trauma. In
of patients with DAI. Other vulnerable sites are the most presentations of head trauma CT is capable of
corpus callosum, corticospinal tracts, basal ganglia, producing a correct diagnosis. CT has the advantage
and the brain stem. The initial damage to the brain is over MRI of more clearly showing skull fractures. In
followed by secondary reactions related to hemor- DAI there is usually a discrepancy between the depth
rhage, edema, changes in local perfusion, and trigger- of the coma as expressed in the Glasgow Coma Score
ing of biochemical cascades. Swelling of the brain and the lack of or subtle findings on CT (Fig. 104.1).
may lead to tentorial herniation; swelling of the brain White matter abnormalities develop over time
stem may lead to hydrocephalus. Disruption of neu- (Fig. 104.1).
ronal and axonal connections leads to wallerian de- MRI is the best imaging modality by which to con-
generation and atrophy. firm the diagnosis and to classify the lesions, usually
It is probably the extent of the lesions and the in- employing the grading scale of Gennarelli. This scale
volvement of the rostral brain stem that leads to the divides the findings into three groups: lesions with
vegetative state in many of the patients. It is thought and without hemorrhage at the gray–white matter
that damage to the rostral part of the brain is the junction, especially in the temporal and frontal areas
cause of a reduction in dopamine turnover. In the first (type 1), combined with lesions in and around the
few hours after the traumatic brain injury, cate- corpus callosum (type 2), and with lesions in the basal
cholamines in the CSF are raised. Soon the cate- ganglia and the rostral brain stem (type 3). The scale
cholamine production is chronically decreased and roughly correlates with outcome.
the levels in the CSF drop. Plasma norepinephrine Conventional MRI with T1-weighted, T2-weighted,
levels have been shown to correlate with the Glasgow T2*-weighted, and FLAIR images is more sensitive in
Coma Score and may correlate with the outcome of depicting tissue changes than is CT (Figs. 104.2–
104.5 Magnetic Resonance Imaging 825

Fig. 104.1. A 36-year-old man with severe head trauma. The a developing leukoencephalopathy in the frontal region. The
first row of images, obtained on admission, shows evidence of third row shows the images obtained 6 weeks after the acci-
white matter shearing in the frontal subcortical region, corpus dent. The hemorrhages have now disappeared. The frontal
callosum, basal ganglia, and thalami with presence of small cortex seems intact, whereas the frontal white matter is hypo-
hemorrhages. The second row of images, obtained 1 week dense, related to wallerian degeneration
later, shows the hemorrhages to be more pronounced.There is

104.5). The role of T2*-weighted images in establish- niques, such as diffusion-weighted images with Trace
ing the diagnosis is considerable. Even early on he- and ADC maps, tensor diffusion imaging with frac-
mosiderin deposits depict the linear nature and mul- tional anisotropy and fiber tracking, perfusion imag-
tiplicity of lesions (Figs. 104.2 and 104.5). The role of ing, and magnetization transfer ratio maps are added
MR is even more pronounced when newer tech- to the protocol. These techniques allow assessment of
826 Chapter 104 Diffuse Axonal Injury

Fig. 104.2.
104.5 Magnetic Resonance Imaging 827

Fig. 104.3. A 14-year-old girl was the victim of a traffic acci- the lesions with greater conspicuity, especially those in the
dent. The FLAIR images on the left (first column) show some basal ganglia, thalami, and the medial parts of the occipital
subdural blood in the left occipital region and a lesion in the lobes. ADC values (third column) are low (<40–50 % of normal
left frontal lobe.There are lesions in the basal ganglia and thal- values) in the affected areas
ami. Diffusion-weighted Trace images (second column) show

the presence and extent of lesions at an early stage, of ADC maps, is extremely sensitive in depicting post-
the loss of structural integrity, and of changes in traumatic changes at a very early stage. Most of the
brain perfusion. lesions show a high signal on the Trace diffusion-
Diffusion-weighted imaging, commonly using a weighted images. Many of these lesions show a lower
combination of Trace diffusion-weighted images and than normal ADC value (Figs. 104.3 and 104.6). It re-
mains to be seen whether, as suggested, lower ADC
values correspond with poorer outcome.
䊴 It is too early to describe the potential role of diffu-
Fig. 104.2. A 43-year-old female, hit by a car, remained co- sion tensor imaging in combination with fiber track-
matose with low Glasgow Coma Scale values. The FLAIR im- ing. One can imagine that fiber tracking may illus-
ages (first and second rows) show small lesions at the cortico- trate the disrupted connections in the brain and pre-
subcortical junction in both frontal lobes, a small hemorrhag- dict the loss of local functionality.
ic lesion in the posterior corpus callosum, contusions in the MRS has been repeatedly mentioned as capable of
right frontal operculum and the left putamen, blood around better predicting the functional outcome of brain in-
the parieto-occipital lobes on the right,a contusion in the right juries. The MR parameter used in these studies is the
temporal lobe, and bilateral lesions in the midbrain, pons, and N-acetylaspartate concentration. The N-acetylaspar-
left superior cerebellar peduncle. Gradient echo images (third tate concentration is thought to reflect the quantity
and fourth rows) show hemosiderin deposits in some of the le- and integrity of axons and neurons in the voxel. Since
sions, especially in the cortico-subcortical lesions in the frontal axonal damage is a good predictor of outcome, there
lobes is obviously a role for MRS. The drawback here is that
828 Chapter 104 Diffuse Axonal Injury

Fig. 104.4. Series of transverse proton density images of a terior limb of the internal capsule,and in the rostral brain stem.
32-year-old man who was involved in a frontal collision show All three types of the Gennarelli scale are present in this
lesions at the cortico-subcortical junction, in the truncus and patient (see also Fig. 104.7)
splenium of the corpus callosum, basal ganglia, thalamus, pos-

in most studies MRS is postponed until the patient is The most important patient management decisions
in a stable condition, which means a considerable should ideally be made in the first 2 weeks, when pa-
time interval between the accident and the per- tients are still in the intensive care unit and ventilator-
formance of MRS, varying between 3 and 35 days. dependent.
104.5 Magnetic Resonance Imaging 829

Fig. 104.5. Gradient-echo (FLASH) images of the same patient as in Fig. 104.4 show the hemorrhagic component in each of the
lesions. Blood has also leaked into the ventricles
830 Chapter 104 Diffuse Axonal Injury

Fig. 104.6.
104.5 Magnetic Resonance Imaging 831

Fig. 104.7. The three types according to Gennarelli are all pre- (second column) reveal the corpus callosum lesions. The im-
sent in this patient and shown in this figure.The images on the ages on the right (third column) demonstrate the brain stem
left (first column) show the cortical lesions. The middle images lesions

Fig. 104.6. Diffusion-weighted Trace images of the same pa-


tient as in Figs. 104.4 and 104.5 (first and second rows,b = 1000)
show high signal intensity in nearly all lesions. ADC maps
(third and fourth rows) show very low values (as low as
0.53 ¥ 10–3 mm2/s, normal 0.85–0.95 ¥ 10-3 mm2/s) in some of
the lesions
Chapter 105

Wallerian Degeneration and Myelin Loss Secondary


to Neuronal and Axonal Degeneration

105.1 Introduction 105.2 Pathology

There are basically two causes of wallerian degenera- Characteristic of wallerian degeneration is the fact
tion in our definition: neuronal cell death and axonal that the sequence and timing of its changes are high-
lesions. It should be noted that our definition is wider ly stereotyped. The cascade of events is similar in the
than usual and includes not only acute axonal lesions, PNS and CNS, but much slower in the CNS. Most ex-
but neuronal and axonal lesions of any kind. Degen- perimental studies of wallerian degeneration in the
eration of the entire arborization of a neuron with its CNS have been carried out in the optic nerve, degen-
axon and axonal branches inevitably follows necrosis erating as a result of enucleation of the eye. Tract de-
of the neuronal cell body. Examples are degenerative generation in the dorsal column of animals has also
diseases affecting neuronal cell bodies and axons, been studied after myelotomy. Human studies mainly
such as amyotrophic lateral sclerosis, Friedreich atax- concern wallerian degeneration after hemorrhage,
ia, olivopontocerebellar atrophy, and neuronal ceroid stroke, tumors, and traumatic lesions.
lipofuscinosis.A lesion of the axon that leads to an in- Abnormalities in axons severed from their neu-
terruption of its continuity gives rise to degeneration ronal perikarya precede changes in the myelin sheath.
of the distal part, whereas the proximal portion as a There is no spatiotemporal gradient of degradation:
rule survives. The myelin in the distal portion under- abnormalities appear over the whole length of the
goes dissolution as a consequence of the axonal de- axons simultaneously. About 8–15 days after the
generation, as the integrity of the myelin sheaths de- event, the axons have disintegrated, whereas the
pends on continued contact with a viable axon. The myelin still appears normal. Most axonal debris dis-
changes in the distal part of the interrupted nerve are appears from degenerating tracts within about 1
called wallerian degeneration in a narrower sense, month. In the process of wallerian degeneration,
following Waller’s original description of the changes degradation of myelin sheaths becomes apparent
that he observed after cutting the glossopharyngeal after 30–90 days and the myelin sheaths disintegrate.
and hypoglossal nerves in the frog in 1850. The breakdown of myelin into simpler lipids does not
Any lesion of axons that leads to an interruption start until after about 100 days and takes place in the
and any lesion of nerve cell bodies that leads to cell cytoplasm of phagocytic cells.
death are followed by wallerian degeneration. In all The pivotal process of wallerian degeneration is
white matter disorders, wallerian degeneration even- granular disintegration of the cytoskeleton, in which
tually plays a role, as myelin loss is followed by sec- the normal cytoskeletal elements of the axon, the
ondary axonal degeneration, which in turn is fol- neurofilaments and microtubules, are abruptly
lowed by wallerian degeneration of the distal parts of depredated to granular and amorphous debris. This
the axons and their myelin sheaths. Wallerian degen- change occurs throughout the distal stump in the first
eration as such cannot be considered a condition that 48 h after dissection. Granular disintegration of the
primarily affects myelin. Nevertheless, wallerian de- cytoskeleton is the result of the activation of ion-sen-
generation with myelin loss secondary to neuronal sitive proteases within the distal stump. A rise in in-
and axonal degeneration are discussed here as a com- tra-axonal calcium to critical levels initiates the pro-
ponent of all disorders and because the MRI appear- teolysis of the axoplasm. The reason for the rise in cal-
ance may be mistaken for primary white matter affec- cium is still unclear. Some evidence suggests that
tion. This is especially the case when the cortex and macrophages may also participate in the granular
the neuronal cell bodies are unaffected.An example is disintegration of the cytoskeleton: anti-macrophagic
the traumatic shearing of nerve fibers, which may antibodies appear to delay axonal breakdown. The
lead to bilateral extensive white matter degeneration, proteases, however, appear to be intrinsic to the axon.
whereas the cortex is remarkably normal. Granular disintegration of the cytoskeleton is associ-
ated with loss of the axolemma as well as the axo-
plasm, so that the myelin sheath surrounds an appar-
ently empty cylinder formerly occupied by the axon.
The granular debris still contains some axonal resid-
ua, as shown by the ability to stain the degenerating
105.4 Pathogenetic Considerations 833

fibers by silver impregnation for long periods after myelin by macrophages and astrocytes and results in
axotomy, and by the staining with some anti-neurofil- transformation of myelin sheaths into uniformly lay-
ament antibodies, even in the absence of surviving fil- ered lipid structures. Cholesterol esters are the major
amentous organelles. Following granular disintegra- constituents of the lipid droplets and the crystal in-
tion of the cytoskeleton, the myelin sheaths lose their clusions of phagocytic cells. The change in biochemi-
regular smooth outline and undergo segmentation cal composition of whole white matter depends on
into short portions with closed ends, termed “ovoids,” the extent of myelin loss. Cholesterol esters are rela-
which break down into smaller globules. The myelin tively elevated.
collapses radially upon itself, the lamellae become
loose, and the periodicity of myelin is gradually lost.
Clearing of the debris is performed by macrophages. 105.4 Pathogenetic Considerations
Without macrophages there is no efficient clearing. In
the PNS this is achieved by circulating macrophages; Wallerian degeneration entails both breakdown of
in the CNS the process is much slower and not com- the axon and clearance of myelin from the distal
pletely understood. stump of the transected nerve fibers as described in
Electron microscopy reveals the degenerating the previous paragraph. Wallerian degeneration is
axons and axonal debris to be surrounded by myelin one of the most prevalent types of cellular pathology
sheaths, occasionally irregularly folded. The myelin in nervous system disease.
sheaths initially show a regular structure with major The pathogenesis of wallerian degeneration de-
and minor dense lines and a periodicity of 105 Å. pends largely upon the type of initial injury. The time
Splitting of myelin lamellae is noted. The structure of schedule differs according to whether the lesion oc-
myelin lamellae continues to change with time, devel- curs acutely (as in stroke, intracerebral hemorrhage,
oping into uniformly layered structures with a peri- and traumatic diffuse axonal injury), more slowly (as
odicity of 40–50 Å. At this stage, the myelin degrada- in progressive neurodegenerative disorders such as
tion products become smaller and are phagocytosed. amyotrophic lateral sclerosis, Friedreich ataxia, neu-
Unstructured lipid droplets as well as complex lamel- ronal ceroid lipofuscinosis, and Alzheimer disease),
lar inclusions are typically found in phagocytic cells or is accompanied by an inflammatory reaction (in
during the later stages of degeneration. Inclusions of infectious or inflammatory disease, such as multiple
unstructured lipid crystals are sometimes seen. In the sclerosis and HIV encephalopathy).
final stages, numerous different inclusions are seen. The isolation of myelin retaining an almost normal
The role of oligodendrocytes in wallerian degener- protein and lipid composition despite wallerian de-
ation in the CNS is not clear. It is probable but not cer- generation as late as 90 days after the injury is consis-
tain that oligodendrocytes do not participate, or par- tent with the morphological evidence that in waller-
ticipate only to a minor extent. Macrophages, astro- ian degeneration in the CNS the breakdown of myelin
cytes, and multipotent glial cells have been observed occurs mainly within the cytoplasm of phagocytic
to contain myelin debris. cells. It would seem that the myelin sheath is degrad-
Histological evidence indicates that wallerian de- ed as a unit, with nonselective digestion of myelin
generation takes place at different rates depending on constituents, cholesterol esters being the only resid-
a number of variables. The most important factor in ual constituents.
determining the course of the wallerian degeneration A model of four stages of myelin degradation in
is whether the axonal or neuronal lesion is acute and wallerian degeneration has been proposed (Lassman
monophasic or chronically progressive. et al. 1978a, b):
1. The stage of mechanical deterioration of myelin
sheaths, showing normal myelin periodicity un-
105.3 Chemical Pathology der the electron microscope.
2. The stage of degradation of digestible proteins, re-
Biochemical analysis of CNS myelin undergoing wal- sulting in the transformation of myelin sheaths in-
lerian degeneration shows a gradual decrease in all to uniformly layered structures.
myelin constituents but otherwise a remarkable nor- 3. The stage of lipid degradation, accompanied by
mality in the composition of the remaining myelin. the occurrence of unstructured lipid droplets and
Only an increase in cholesterol esters and a slight de- crystals in phagocytic cells.
crease of ethanolamine phosphoglycerides have been 4. The stage of deposition of poorly digestible lipids
demonstrated. Neither are any major abnormalities or lipoproteins, resulting in numerous different
found in the protein constituents. Basic protein is lost electron microscopic inclusion types in phagocyt-
from the myelin sheath relatively early in the process ic cells. Resolution of tissue debris and atrophy are
of myelin degradation, due to an increase in pro- the final stage in the process.
teinase activity. This occurs before phagocytosis of These four stages are reflected in MRI.
834 Chapter 105 Wallerian Degeneration and Myelin Loss Secondary to Neuronal and Axonal Degeneration

Fig. 105.1. A patient with subacute


meningoencephalitis involving the
leptomeninges over the left hemi-
sphere and the cortex in the occipital
region. On the T2-weighted images
(first row) the white matter under-
neath the affected cortex has a lower
signal intensity than the white matter
elsewhere.This represents the second
stage of wallerian degeneration.The
second row shows T1-weighted images
after contrast with occipital cortical
enhancement

105.5 Magnetic Resonance Imaging to be hydrophobic, causing low signal intensity


on proton density and T2-weighted images. This
In the CNS, wallerian degeneration of long tracts can occurs between 4 and 14 weeks after the incident
usually be well depicted. As a multiplanar modality, (Fig. 105.1).
the MR plane can be chosen to depict the signal 3. Next, myelin lipid breakdown starts to take place,
changes over their full length. Depending on the loca- with gliosis and increased water content, leading
tion of the primary lesion or lesions, wallerian tract to a higher signal intensity of the involved tracts
degeneration can be unilateral or bilateral, and, if bi- on proton density and T2-weighted images
lateral, symmetrical or asymmetrical. Normally the (Figs. 105.2 and 105.3).
long tracts of densely packed white matter have 4. Finally, after a number of months to years, the ab-
somewhat lower signal intensity on proton density normal tissue constituents are removed and atro-
and T2-weighted images than the remainder of the phy results (Fig. 105.4).
white matter. In wallerian degeneration the signal in
the tract is lower or higher than usual compared to Conventional MRI is able to depict stages 2–4. Al-
the normal white matter tracts, depending on the though there is considerable overlap in time, each
stage of degeneration. In fact MRI can help to distin- phase has been recognized. There is a crossover of
guish the four stages of wallerian degeneration, each contrast between the degenerating tract and the sur-
resulting from a rather well-defined phase in the rounding tissue between stages 2 and 3,“fogging” the
process of degeneration: MR depiction of wallerian degeneration.
1. Physical degeneration of the axon occurs first, Most attention has been paid to local wallerian de-
with only mild biochemical changes. This happens generation following acute focal lesions and involving
in the first 2–4 weeks after the incident. This stage single tracts, most often sensorimotor tracts. Acute
is not visible on conventional MR images. diffuse wallerian degeneration occurs in cases of
2. The first stage is followed by myelin protein break- diffuse axonal injury, with disruption of corti-
down without myelin lipid breakdown. The result- comedullary connections. The wallerian degenera-
ing material with high lipid content is considered tion leads to diffuse white matter degeneration in the
105.5 Magnetic Resonance Imaging 835

Fig. 105.2. The third phase of wallerian degeneration is clear- generation all the way down into the brain stem. From Wara-
ly displayed on these T2-weighted images, showing the initial gai et al. (1994), with permission
lesion and the corticospinal tracts undergoing wallerian de-

with loss of neurons, such as in amyotrophic lateral


sclerosis, neuronal ceroid lipofuscinosis, Niemann–
Pick disease type C, Friedreich ataxia, Alzheimer dis-
ease, and in slowly developing inflammatory disor-
ders. Neurons and axons disappear gradually and not
all at the same time. That means that stages of waller-
ian degeneration overlap and that there are no stages
to be visualized separately. In these disorders, MRI
shows cortical atrophy and often, in addition, a
slight to moderate increase of signal intensity in the
cerebral hemispheric white matter (Fig. 105.5). The
changes in normal-appearing white matter in relaps-
ing–remitting and secondary progressive forms of
multiple sclerosis are probably also, at least in part,
the result of wallerian degeneration.
Wallerian degeneration has also been reported
in the spinal cord, in particular in the posterior
columns. This occurs after trauma of the spinal cord
or in the course of a myelopathy, either caused by
Fig. 105.3. Prototype of wallerian degeneration late in the degenerative changes of the vertebral column, or by
third phase. The T2-weighted image shows a hemorrhagic infectious or inflammatory lesions. From the lesion in
lesion in the corona radiata on the left.Wallerian degeneration the spinal cord wallerian degeneration occurs in two
is seen in the corticospinal tract with high signal intensity. directions: downwards in the motor tracts, and up-
Atrophy is setting in wards in the sensory tracts. In some degenerative cas-
es there is a gap between the location of the original
myelopathic lesions and the wallerian degeneration,
course of weeks (see Chap. 104). The subcortical and the latter becoming visible several segments higher in
deep white matter have a low density on CT images the afferent tracts. Given time, the parts in between
and remarkably low signal intensity on proton densi- also show signal intensity changes in the affected
ty and T2-weighted MR images. This occurs in a phase tracts.
where protein degradation is most prominent and To improve the detection of wallerian degenera-
lipid degradation still absent. In the next phase the tion on MR, special techniques, such as diffusion-
involved area shows higher signal intensity on T2- weighted imaging and magnetization transfer ratio
weighted and FLAIR images. Finally, atrophy with (MTR) measurements can be used. The most impor-
focal gliosis results. tant advantage is that with these techniques changes
In addition, focal or diffuse, low-grade wallerian can be detected in an earlier phase than with conven-
degeneration occurs in neurodegenerative disorders tional MRI. In the first stage of wallerian degenera-
836 Chapter 105 Wallerian Degeneration and Myelin Loss Secondary to Neuronal and Axonal Degeneration

Fig. 105.4. T2-weighted transverse series show a left opercular infarction. In the fourth stage of wallerian degeneration, atrophy
results, as is shown in the brain stem (arrows)

tion following an acute lesion, diffusion-weighted ues, and a decreased MTR in the tracts involved. The
imaging reveals a decreased apparent diffusion co- MTR shows changes in the affected tracts before there
efficient in the tracts involved in the degenerative are abnormalities in T2 that lead to signal changes on
process (Figs. 105.6 and 105.7), whereas the MTR in conventional images (including FLAIR). Follow-up
these tracts is increased, and anisotropic diffusion- studies show increasingly abnormal values over time.
weighted imaging or diffusion tensor imaging reveals In contrast with wallerian degeneration following an
loss of anisotropy in the affected tracts. The subse- acute incident, there is no transition from one phase
quent changes are more gradual and are progressive of degeneration to the other.
over time. From the second stage of wallerian degen- MRS allows detection of changes in concentration
eration onwards and continuing in the subsequent of N-acetylaspartate in regions with loss of neurons
stages, the ADC is increased, MTR is decreased, and or axons. Follow-up studies with MRS in patients
further loss of anisotropy occurs. with amyotrophic lateral sclerosis demonstrate that a
Long-term follow-up studies with diffusion tensor marked decrease in the N-acetylaspartate/choline
imaging in patients with more slowly progressive ratio is associated with the development of signs of
tract degeneration, such as in amyotrophic lateral upper motor neuron disease, and may even precede
sclerosis, show loss of anisotropy, decreased ADC val- the appearance of pyramidal signs.
105.5 Magnetic Resonance Imaging 837

Fig. 105.5. In this 10-year-old boy with Niemann–Pick disease Note the cerebral atrophy. A combination of atrophy and sub-
type C, wallerian degeneration is seen in the white matter, sec- tle white matter signal abnormalities is suggestive of primary
ondary to cortical neuronal loss. There is a diffuse increase in neuronal or axonal degeneration rather than a primary
signal intensity of the cerebral white matter on T2-weighted myelinopathy
images and loss of distinction between gray and white matter.

Fig. 105.6. A 5-week-old female infant with onset of seizures Trace diffusion-weighted image (middle) reveals a high signal
3 days before the MRI. Neurological examination revealed in the left cerebral peduncle, whereas the ADC map (right)
right arm impairment. MRI reveals a left middle cerebral artery reveals a decreased ADC in that area. From Mazumdar et al.
infarction. The images at the level of the midbrain are shown. (2003), with permission
The T2-weighted image (left) reveals no abnormalities. The
838 Chapter 105 Wallerian Degeneration and Myelin Loss Secondary to Neuronal and Axonal Degeneration

Fig. 105.7. A 4-day-old neonate with


right middle cerebral artery infarction.
The two images in the first row are
T2-weighted fast spin echo images
(left transverse, right coronal).
Increased signal intensity is present
in the white matter of the right
temporal lobe and insular region;
the overlying cortex has a high signal
in some parts, a low signal in others.
A focus of hemorrhage is seen in the
corona radiata and putamen.The
images show abnormally high signal
in the right corticospinal tract (arrow-
heads). Diffusion-weighted images
(second row left transverse, right
coronal) show increased signal inten-
sity in the territorial infarct and the
right cerebral peduncle (arrowheads).
The corresponding ADC maps (third
row left transverse, right coronal) show
reduced ADC values in the infarcted
region and the right cerebral pedun-
cle (arrowheads). Images of the
fourth row containing the transverse
diffusion-weighted image (left) and
corresponding ADC map (right) show
involvement of the splenium of the
corpus callosum with decreased
diffusion. From Mazumdar et al.
(2003), with permission
Chapter 106

Diffusion-Weighted Imaging

106.1 Introduction movement at the molecular level. MRI is based upon


the resonance condition of protons brought about by
A drop of ink in a glass of water will spread through- a strong static magnetic field. Protons in this condi-
out the water by a process known as diffusion. This tion can absorb and subsequently release energy of a
phenomenon was first observed by Brown in 1827 special frequency (resonance frequency). The re-
and bears his name, brownian movement. His experi- sponse or released energy of the protons can be col-
ments, executed with Clarkia pollen spread on a wa- lected by coils placed around the body or a body part
ter surface, led to wide speculations about the nature as the MR signal. The MR signal is used to reconstruct
of the movements observed. “He suspended some of images and depends on many factors, such as pulse
the pollen grains in water and examined them close- sequence, slice thickness, matrix, and number of ac-
ly, only to see them ‘filled with particles’ that were quisitions. The strength of the signal is ultimately the
‘very evidently in motion’. He was soon satisfied that sum of protons in and out of phase in a pixel (picture
the movement ‘arose neither from currents in the flu- element) or, more correctly, in a voxel (volume ele-
id, nor from its gradual evaporation, but belonged to ment). With all other parameters identical, the signal
the particle itself ’. In due course he was to carry out is at its maximum when all protons in a voxel are in
careful experiments to disprove these alternative ex- phase. The signal decreases when a number of pro-
planations, and it has been shown that Brown was tons are out of phase (or dephased). Diffusion of wa-
able to anticipate the later objections of those who ter molecules leads to dephasing of protons and
would doubt his capacity to have observed what he therefore loss of signal. This means that in MR images
claimed. It must have been tempting for Robert unrestricted diffusion leads to loss of signal and re-
Brown to assume, as had other workers before him, stricted diffusion leads to a smaller loss of signal. On
that here was the very essence of life.” (Ford, 1992) a relative scale this means that tissue with restricted
Much later it became clear that the movement of diffusion will appear brighter in the image, while tis-
particles was the result of random movement of wa- sues with less restricted diffusion will appear darker.
ter molecules. Temperature proved to be an impor- The contribution to dephasing of protons caused
tant factor in determining the rate of the movement of by diffusion of water molecules in the order of cell
molecules, and therefore the process is also referred dimensions, 5–25 mm2/s, is not detectable on images
to as thermodynamic movement. The human body made with standard MR pulse sequences. Two devel-
consists to 60–80% of water. Molecular movements in opments were necessary to realize the visualization of
the body are modified by complex tissue compart- diffusion effects with MR equipment. The first was
ments. The brain, for example, consists of many dif- the improvement of the temporal resolution by ultra-
ferent structures, restricting to a greater or lesser ex- fast imaging techniques (now usually single shot echo
tent the free movements of water molecules. This planar pulse sequences). These techniques have the
leads to differences in diffusion of water molecules advantage of “freezing” motion. Motion in the order
between different brain structures, which makes dif- of cell dimensions is rapidly overshadowed by move-
fusion a potential source of image contrast.When wa- ments of the patient, voluntary and involuntary,
ter movement is relatively unrestricted in all direc- by respiration, and pulsation of vessels and cere-
tions, diffusion is isotropic; when restrictions are pre- brospinal fluid. The second requirement was the
sent in one or more directions, water movement is introduction of a set of strong, opposed “diffusion”
anisotropic. Diffusion in gray matter in adults is near- gradients enhancing the diffusion process. Applica-
ly isotropic; on the other hand, diffusion in white tion of these techniques has made it possible to intro-
matter, with its compact and organized structure of duce diffusion-weighted imaging (DWI) into clinical
myelinated axons and fiber tracts, is anisotropic. Dis- practice. The MR application of DWI is practiced on
ease states may also lead to changes in the freedom of two levels: a relatively simple level with rapid acquisi-
water diffusion. tion of clinically useful information, and a more
It has become possible, by using MR techniques complex level allowing more precise estimation and
with a spatial resolution in the order of square mil- quantization of anisotropy and 3D analysis of the
limeters, to measure changes in the freedom of water data.
840 Chapter 106 Diffusion-Weighted Imaging

106.2 DWI Techniques

For DWI it is necessary to use ultrafast sequences


to “freeze” motion, as mentioned above. Even then,
however, the small contribution of diffusion to the
dephasing of protons is not visible on single shot
echo planar images and is easily overshadowed by T2
effects. It is necessary to enhance the diffusion effects
by applying strong, opposed diffusion gradients Fig. 106.1. Stejskal–Tanner pulse sequence, in which G is the
(Stejskal–Tanner sequence) (Stejskal and Tanner diffusion gradient strength, D the distance between diffusion
1965; Fig. 106.1) that cancel all “coherent” motion gradients, and d the duration of the diffusion gradient. The
within the voxel and enhance the effect of diffusion of diffusion sensitivity (gradient moment or b value) of this se-
protons. quence increases when (1) the gradient strength (G) increases,
The diffusion sensitivity is expressed as the b value (2) the duration of the gradient (d) is longer), and (3) the time
of the sequence and usually varies between 500 between the opposed gradients (D) is longer. These relations
and 4000 s/mm2. On commercial scanners the maxi- are brought together in the following equation: b = g2 G2 d2
mum value often does not exceed 2000; usually (D–d/3), in which g is the gyromagnetic ratio
b = 1000 s/mm2. Quantitative information about dif-
fusion can be obtained in several ways. The simplest
way is to measure the apparent diffusion coefficient describe the effective averaged diffusion per voxel.
(ADC) of the tissue. There is an exponential decrease The diffusion coefficient in free water at 25 °C is about
in signal intensity with increasing b values. The ADC 25 ¥ 10–3 mm2/s. In brain tissue ADC values are in the
is measured by estimating the slope of the exponen- order of 0.70–0.90 ¥ 10–3 mm2/s. In acute lesions, such
tial curve describing the signal intensities at different as in acute ischemia, the ADC values decrease by
b values. Measurements at two or more different about 50% to reach values of 0.40–0.60 ¥ 10–3 mm2/s.
b values (usually 500 and 1000) are used to calculate In chronic ischemic lesions, with liquefaction and
the ADC. The word “apparent” refers to the fact that in necrosis, these values may go up to 1.80–2.40 ¥
vivo it is not possible to measure the “pure” diffusion 10–3 mm2/s.
coefficient D. The diffusion coefficient measured – the In routine DWI, diffusion gradients are applied in
apparent diffusion coefficient – averages different three orthogonal directions. Images can be obtained
tissues with different diffusion coefficients present in on the basis of the results of each gradient separately,
one voxel; it is also influenced by other parameters, in respectively the slice (s), read (r), and phase (p)
such as the presence of microparticles of iron. ADCs direction (Fig. 106.2). Where fiber tracts are parallel

Fig. 106.2. Diffusion-weighted images in a patient with a diffusion gradient. Tracts perpendicular to the gradient show
right-sided homonymous hemianopia, made with three differ- high signal intensity, tracts parallel to the gradient show lower
ent diffusion gradients in x (read), y (phase), and z (slice) direc- signal intensity. Note that the small lesion in the left occipital
tions, showing anisotropy dependent on the direction of the pole demonstrates restricted diffusion in all three directions
106.2 DWI Techniques 841

Fig. 106.3. The images on the left are


made with a single diffusion gradient.
In the left upper image one sees, in
addition to the bright spot in the
thalamus, high signal intensity in fiber
tracts perpendicular to the gradient
direction.The lower left image shows
anisotropy of the splenium of the
corpus callosum with a gradient in the
slice direction.The images on the right
are Trace diffusion-weighted images,
which average diffusivity of three
(or more) gradient settings to mini-
mize the influence of anisotropy in
the image.The upper right image now
shows only the lesion in the right
thalamus.The lower right image does
not show any areas of restricted
diffusion

with the direction of the applied gradient, signal loss extremely important for the demonstration of re-
due to diffusion will be at its maximum; where fiber stricted diffusion to look at the ADC values and veri-
tracts are perpendicular to the gradient direction, sig- fy the presence of a decreased ADC (Fig. 106.4).
nal loss due to diffusion will be at its minimum and Diffusion can be viewed as the product of the de-
the signal will remain relatively high. The opposite is grees of freedom of movement of water in three di-
true for ADC maps: the greater the restriction in dif- mensions (x, y, and z). With equal movement oppor-
fusion, the lower the ADC value, and the lower the sig- tunity in all directions (isotropy), the product is a
nal intensity of that area. There is an important disad- rounded sphere and the diffusion vectors are equally
vantage to using images with the application of only long.Where diffusion is anisotropic, the product is an
one diffusion gradient in one direction. Fiber tracts ellipsoid, the vectors unequal in different directions.
perpendicular to the gradient direction have a rela- The main vector, its size and direction can be estimat-
tively high signal, which may obscure hyperintense ed. With gradients applied in three directions, the D
lesions with restricted diffusion in or in the neighbor- value can be estimated voxel by voxel by averaging the
hood of these structures or simulate a hyperintense D values obtained in three (or more) different gradi-
lesion. This pitfall can be omitted by averaging the ent directions: D = (Dxx + Dyy + Dzz) / 3. This aver-
ADC values of the measurements with (at least) three aging procedure will also cancel anisotropy in the
diffusion gradient directions (Fig. 106.3). image. The resulting image is called a Trace image
Diffusion is a process in time. That means that the (Figs. 106.3 and 106.5).
MR sequence has to cover a minimum time slot to ob-
serve the diffusion process. Therefore, a relatively Diffusion Tensor Imaging In isotropic substances,
long echo time has to be applied, resulting in a T2- diffusion can be described by a single parameter, the
weighted EPI sequence. This leads to a high signal in- diffusion coefficient, or rather, the apparent diffusion
tensity of structures with a long T2. This high signal coefficient, ADC. The attenuation of the MR signal
area may retain its brightness on diffusion-weighted depends on D and the b value, characterizing the gra-
images, simulating restricted diffusion. This phe- dient pulse:
nomenon is referred to as “T2 shine-through” effect.
ADC maps do not share this problem. It is therefore A = exp(–bD)
842 Chapter 106 Diffusion-Weighted Imaging

Fig. 106.4. The next series shows the application of DWI techniques in a clinical
situation, the effect of different b values of the diffusion gradients and the added
value of ADC maps.The upper row represents the conventional T2-weighted image
and T1-weighted images without and with contrast. The lesion in the right middle
cerebral artery territory is visible. The middle row shows Trace diffusion-weighted
images with different b value settings; from left to right: b = 50 s/mm2, 500 s/mm2,
1000 s/mm2. With higher b values more and more structural information gets lost
in the image, whereas the information about the abnormal area increases. The
Trace diffusion-weighted images must be checked against the corresponding ADC
maps (lower row). The combination of high signal intensity on Trace diffusion-
weighted images and low ADC values (<50 %) argues for the presence of cytotoxic
edema or other causes of restricted diffusion

Fig. 106.5. The image on the left


was obtained with a single gradient
in the slice direction. In the Trace
diffusion-weighted image on the
right the effect of anisotropy in
the splenium of the corpus callosum,
as seen on the left, has disappeared
106.2 DWI Techniques 843

Fig. 106.6. Diffusion tensor (matrix). In isotropic conditions


Dxx, Dyy, and Dzz suffice to determine the diffusion vectors. In
anisotropic conditions Dxy, Dxz, and Dyz are also important to
define the strength and directions of vectors in a voxel

in which A is attenuation, b is diffusion sensitivity,


and D is the diffusion coefficient. With anisotropy
present, a single parameter is no longer sufficient.
With the mathematical description of the diffusion
tensor, effects can be fully extracted, characterized,
and exploited. The tensor Deff is necessary to describe
the molecular movements in each possible direction.
In this model isotropic diffusion can be depicted as a
rounded sphere, anisotropic diffusion as an ellipsoid
tilted in the direction of the main orientation of the Fig. 106.7. Ellipsoid representing the diffusion tensor. The
diffusion (Fig. 106.6). ellipsoid is characterized by the maximum diffusion in each
The anisotropic diffusion process in each voxel can direction and represents the relationship between the princi-
be characterized by Deff, consisting of a 3 ¥ 3 tensor pal coordinate axes (x’, y’, and z’) and the laboratory reference
matrix consisting of nine combinations, of which frame (x, y, z). The radii (bold black arrows) represent the mag-
three are similar and six are independent. By sam- nitude and direction of the diffusion after unit time.The l1, l2,
pling signal attenuation after applying diffusion-sen- and l3 represent the so-called eigenvalues of diffusivity in all
sitizing gradients in at least six different noncollinear directions, shaping the ellipsoid
directions, these six (or more) elements can be deter-
mined. They represent diffusion along the three main
coordinate axes of the ellipsoid, thus defining the
shape of the ellipsoid (Fig. 106.7).
The data so obtained are rotationally independent:
the head can be placed in an arbitrary position in the
main magnet and this will not influence the results.
The data can be used to calculate either the “diffusiv-
ity”, <D> (the directionally averaged ADC), or the de-
gree of anisotropy, most often expressed as relative
anisotropy (RA), or as fractional anisotropy (FA). In
2D representations of diffusion tensor imaging (DTI),
FA is the most frequently used of many possible para-
meters describing anisotropy. RA is the ratio of vari-
ance of the eigenvalues to their mean. FA is the ratio
of the anisotropic component of the diffusion tensor
over the whole diffusion tensor. FA indicates how
elongated the ellipsoid is (0 = isotropy, 1 = maximum
anisotropy, often multiplied by 1000). FA is usually
displayed as a map in gray shades (higher FA values
reflecting more pronounced anisotropy show a high-
er signal intensity on gray scale images, low values
appear darker) (Fig. 106.8) or in color codes. FA im-
ages show more contrast between gray and white
matter than do T1- and T2-weighted images. The rea-
son for this difference is still not fully clear, but may Fig. 106.8. A fractional anisotropy (FA) map provides a mea-
have to do with the major difference in density of sure of anisotropy of different structures. Differences in FA
axon fibers. In the literature ratios are used to quanti- between gray and white matter structures are greater than
tate FA values, because values are not the same on all differences in T1, T2, or ADC values. In a FA map the distinction
systems. The ratio is calculated as: between gray and white matter structures is superb
844 Chapter 106 Diffusion-Weighted Imaging

Fig. 106.9. Diffusion tensor imaging


allows fiber tracking as is demonstrat-
ed in this image. DTI was done with
12 diffusion gradient directions. Fibers
were tracked connecting left and right
hemisphere via the corpus callosum,
showing the potential of the method.
The upper image shows a comparable
anatomical specimen

FA lesion tinguish afferent from efferent tracts; the spatial res-


rFa = × 100 % olution within acceptable acquisition times is still
FA reference point
crude; and crossing fibers may lead to confusing
or, somewhat more complex: results. Nevertheless, the potential of the method is,
FA patients – (FA controls) especially in combination with other techniques such
∆FA = × 100 % as functional MRI, impressive.
(FA controls)
in which equation “(FA controls)” stands for the mean
value from the regions of interest in volunteers. In 106.3 DWI and Pathophysiological
particular in multicenter studies and trials ratios will Backgrounds
have to be used to overcome inequalities in equip-
ment performance. DWI, usually in its simplest form with three diffusion
A map can also be produced displaying the princi- gradients, anisotropy images, Trace images, and ADC
pal vector orientation per voxel. This is most often maps, is now widely used in clinical practice. It has
displayed as a color map in which the vectors (small become a valuable tool in the detection and diagnosis
arrows) are presented in color codes per direction. of a growing number of neurological disorders. The
DTI may be used to create a 3D map by connecting pathophysiological background of the changes in the
adjacent vectors with nearly the same orientation. acute phase of diseases and the changes over time
Software programs usually allow the setting of a into subacute and chronic conditions are only partly
threshold that indicates the degree of accepted understood. Diffusion is a parameter that is indepen-
anisotropy (FA value), and also the setting of an arbi- dent of the relaxation properties (T1 and T2) of tissue.
trary angle to decide whether a fiber connection will Brain tissue consists of cell structures, with mem-
be accepted. The result is fiber tracking: white matter branes dividing tissue fluid into extracellular and in-
tracts can be visualized (Fig. 106.9). DTI cannot dis- tracellular compartments, organelle compartments,
106.3 DWI and Pathophysiological Backgrounds 845

Fig. 106.10. DWI and ADC maps, often in combination with bright signal in these lesions. The lower row of ADC images
perfusion imaging, play an important role in stroke units, to shows very low ADC values in the lesions, confirming the pres-
identify salvageable tissue. This set of images shows such a ence of cytotoxic edema.The added value to the FLAIR images
practical application in a 66-year-old stroke patient.The upper is the time dependency of the DWI–ADC techniques, allowing
row of FLAIR images shows lesions in the right middle cerebral an estimate of the age of the infarction
artery territory. The second row of DWI–Trace images shows

blood compartments consisting of arterioles, venules, The best understood changes are those in acute
and capillaries sustaining the microcirculation, and ischemic lesions (Fig. 106.10). Hypoxemia and is-
fiber tract compartments with myelin and axons, all chemia lead to membrane depolarization, changes in
having their impact on the complex process of diffu- membrane permeability, changes in ion exchange,
sion. and influx of water into the cell. Swelling of cells, both
846 Chapter 106 Diffusion-Weighted Imaging

Fig. 106.11. The practical use of DWI


and ADC maps is also demonstrated
in this 38-year-old female, in whom
the conventional images suggested
the diagnosis glioblastoma multi-
forme.The patient showed no sign
of infectious disease either clinically or
biochemically.The upper two images,
T2-weighted and T1-weighted with
contrast, show a multicystic lesion in
the right frontal lobe, with edema and
ring enhancement.The Trace images
and ADC map on the lower row show
that within the cysts water diffusion is
severely restricted, indicating a possi-
ble abscess, which was confirmed
at surgery

glia and neurons, lead to compression of the extracel- Many other factors may influence the degree of
lular space and restriction of movement of extracellu- freedom of movement of water molecules in brain tis-
lar water, and probably also to restriction of intracel- sue. High cellular density and high nucleus/cyto-
lular water movements due to changes in organelles. plasm ratio of packed cells in tumors may cause low
This results in a high signal on DWI–Trace images ADC values. In brain abscesses, the restriction of wa-
and low ADC values. Subsequent lysis and shrinking ter movement is based upon cellular density, forma-
of cells and rarefaction of tissue leads to increase of tion of septa, and gelatinous or caseous contents of
extracellular space and water content, with conse- the abscess cyst (Fig. 106.11). Epidermoids also show
quently a decrease of signal intensity on Trace images restricted diffusion, caused by the gelatinous condi-
and increase of ADC values. The time frame for these tion of the fluid and the presence of septa.
changes in arterial territorial infarctions is in the or- In focal epilepsy there is a reduction of 20% of
der of 2–4 weeks. It should be noted that this time ADC values of the tissue involved in the ictal phase,
frame is different in border zone infarctions, where while MRA, MR perfusion studies, and SPECT show
high signal on Trace images and low ADC values per- evidence of hyperperfusion in that area. In the inter-
sist for much longer, even months. This is the result of ictal phase, the ADC values return to normal in days
a more chronic and persistent perfusion deficit. to weeks. In addition to swelling of involved neurons,
In acute encephalitis and cerebritis, for example, in the increase in regional blood volume may influence
herpes simplex encephalitis,ADC values can be as low the measurements by an increase of magnetically ac-
as in ischemic lesions. In viral infections there is tive blood products, for example deoxyhemoglobin.
swelling of cells and decrease of extracellular space in Blood present in post-traumatic or postoperative
the initial phase, comparable to what happens in is- cysts may lead to a pseudo-low ADC and in that way
chemia. It remains to be seen whether ADC and FA suggest an abscess. T1-weighted images usually will
provide a means of estimating prognosis or of esti- solve this problem, when it arises.
mating disease activity in subacute lesions or recur- In white matter disorders, inherited or acquired,
rent infection. multiple factors resulting in changes in diffusivity
106.5 Hypoxic–Ischemic Conditions in Neonates 847

have to be considered, such as cellular density and not quite comparable. Measures such as “apparent”
swelling in acute inflammation or infection (multiple anisotropy are less well defined than FA and RA and
sclerosis, acute disseminating encephalomyelitis, vi- less reproducible. ADC values may also differ accord-
ral encephalitis), restriction of water movement in ing to the machine and pulse sequences used, for ex-
acute intramyelinic edema (posterior reversible en- ample due to the b value setting and the number of
cephalopathy syndrome) and vacuolating myelinopa- applied diffusion gradients. Roughly, the ADC values
thy in toxic and metabolic conditions (maple syrup are high in neonates, different in different anatomic
urine disease, Canavan disease, megalencephalic regions, varying from 1.05 to 1.64 ¥ 10–3 mm2/s.At the
leukoencephalopathy with subcortical cysts, heroin age of about 10 months this range is in the order of
intoxication, hexachlorophene intoxication, CO in- 0.75 to 0.92 ¥ 10–3 mm2/s, close to values seen in
toxication), and axonal swelling in neurodegenerative young adults (Table 106.1).
and traumatic disorders (amyotrophic lateral sclero- A difficulty with reporting on quantitative diffu-
sis, wallerian degeneration, diffuse axonal injury). sion data in neonates and children is that few of the
data are obtained by methods that fully account for
the effects of diffusion anisotropy. ADC values are of-
106.4 Brain Maturation ten obtained by measurements in one direction only.
and DWI–DTI Changes When anisotropy is present, these estimations are not
accurate. Even when measurements are made in three
DWI and DTI have the unique capability to show orthogonal directions, it is not possible to accurately
changes in anisotropy.Anisotropy is largely due to the and quantitatively determine anisotropy. The full dif-
presence of myelinated fiber tracts. It has been fusion tensor must be sampled in at least six different
shown, however, that changes in anisotropy (FA) oc- directions. Another concern is partial volume averag-
cur before myelination becomes visible, confirming ing in structures close to CSF spaces, because this will
that anisotropy is partly the result of precursors of artificially increase the measured ADC values. Nu-
myelin and, probably more importantly, axonal or merical data available at this moment should be
fiber density. The changes in FA in different brain applied with great care in clinical situations.
areas have been extensively documented. FA shows an The development of anisotropy is reflected in the
increase over time, while ADC values drop. Unfortu- FA values in Table 106.2.
nately the methods used to establish timetables are

106.5 Hypoxic–Ischemic Conditions


Table 106.1. ADC values in full-term neonates (adapted from in Neonates
Neil et al. 1998)
Region ADC values DWI and DTI have a great impact on the diagnosis
(¥ 10–3 mm2/s) of post-hypoxic–ischemic changes in neonates. On
conventional MR sequences patterns of hypoxic–is-
White matter centrum semiovale 1.30–1.60
chemic encephalopathy in preterm and term neo-
Head of caudate nucleus 1.18–1.31 nates are often difficult to visualize in the immediate
Insular gray matter 1.12–1.24 postnatal period. This is due to the only partly myeli-
Lentiform nucleus 1.15–1.22 nated state of the neonate’s brain, with a high signal
Thalamus 1.01–1.15 on T2-weighted images of the unmyelinated parts,
Anterior limb internal capsule 1.14–1.24 obscuring the lesion. DWI often reveals the abnormal
Posterior limb internal capsule 1.03–1.09
areas with high sensitivity. In particular the extent
of lesions in early periventricular leukomalacia, in
Cerebellar hemisphere 1.07–1.14
the cortico-subcortical pattern, and in basal ganglia

Table 106.2. Regional FA values at different ages (adapted from Schneider et al. 2004)
2 Months 12 Months 24 Months 144 Months

Genu corpus callosum 0.41 0.52 0.63 0.72


Splenium corpus callosum 0.45 0.61 0.68 0.76
Frontal white matter 0.16 0.29 0.34 0.39
Centrum semiovale 0.28 0.38 0.42 0.53
Posterior internal capsule 0.52 0.63 0.68 0.76
848 Chapter 106 Diffusion-Weighted Imaging

Fig. 106.12. Images of a term-born neonate with acute pro- images demonstrate the typical cortico-subcortical pattern,
found hypoxia–ischemia. The upper row of conventional T2- allowing an early diagnosis with a poor prognosis. It may take
weighted images show generalized edema, including swollen much longer before this pattern emerges on conventional im-
and abnormal basal ganglia. The lower two rows of DWI–Trace ages

lesions can be visualized within hours after the insult Table 106.3. ADC values in early periventricular leukomalacia
(Fig. 106.12). When interpreted with care, DWI and (¥ 10–3 mm2/s) (adapted from Bozzao et al. 2003)
DTI data are extremely helpful. Region Non-PVL PVL
As an example, the values in periventricular leuko-
malacia are instructive, because they seem to allow Posterior limb internal capsule 1.06 0.96
early confirmation of the presence of the lesions Corona radiata 1.23 0.97
(Table 106.3). Frontal white matter 1.35 1.19
Parietal white matter 1.52 1.15

PLV, periventricular leukomalacia


106.6 Leukoencephalopathies 849

106.6 Leukoencephalopathies gory of patients. Some findings of DWI and DTI have
direct implications for the management of patients.
DWI and DTI have been applied extensively in multi- An important example is found in X-linked adreno-
ple sclerosis. In most studies estimation of FA and leukodystrophy. DTI can play a role in the detection of
ADC has been used to compare different forms of early abnormalities in patients, not yet seen on con-
multiple sclerosis and different tissue components, ventional images. Early changes are important in esti-
such as plaques, white matter around plaques, nor- mating when to offer the patient hematopoietic stem
mal-appearing white matter, and enhancing versus cell transplantation. DWI and DTI should be com-
nonenhancing lesions. Some of the results are sum- pared in this respect with MRS and magnetization
marized in Table 106.4. transfer imaging.
From Table 106.4 it is clear that the discriminating In progressive inherited disorders the time factor
power of FA is considerably greater than that of ADC. plays an important role. In early metachromatic
As expected, an increase in ADC is linked to a de- leukodystrophy ADC values are low and DWI shows a
crease of FA. high signal. This effect is not completely understood,
The findings in normal-appearing white matter of but is thought to be related to the accumulation of
a high ADC and low FA correspond with findings in abnormal metabolites (sulfatides). In later phases of
MTR and MRS studies. DWI and MTR findings corre- the disease,ADC increases and FA decreases as sign of
late better with the disability scores of multiple scle- disintegration of affected structures. Similar findings
rosis patients than do conventional MR estimates and are reported in globoid cell leukodystrophy.
even lesion load estimations. In primary progressive The type of underlying pathology is important. In
multiple sclerosis conventional MR abnormalities are maple syrup urine disease with neonatal onset,
minimal. DTI reveals abnormal values of FA and ADC myelin vacuolation occurs in all myelinated parts
in the corpus callosum and internal capsule of these of the brain in the newborn. These parts show low
patients. Hypointense lesions on T1-weighted images, ADC values with about 80% decrease, and very high
known as black holes and indicating a poor progno- signal intensity on DWI (Fig. 106.13). In other disor-
sis, have the lowest diffusivity of all multiple sclerosis ders, such as nonketotic hyperglycinemia, Kearns–
lesions. T1 isointense lesions have the highest FA of all Sayre syndrome, Canavan disease, L-2-hydroxyglutar-
lesions studied and apparently have maintained tis- ic aciduria, and heroin encephalopathy, intramyelinic
sue integrity. edema is also present and supposed to be the cause of
Normal-appearing gray matter in multiple sclero- restricted water movement and low ADC values in the
sis has also been the subject of a number of studies. In early phase of the disease. In chronic myelin vacuola-
histogram analysis of diffusivity of gray matter in pa- tion, as seen in megalencephalic leukoencephalo-
tients with primary progressive multiple sclerosis, pathy with subcortical cysts and the end stages of
patients with secondary progressive multiple sclero- Canavan disease, no evidence of restriction of water
sis, and controls, statistically significant differences diffusion is found, but increased diffusion is found
were found. The ADC and FA values in normal-ap- (Fig. 106.14).
pearing gray matter are respectively higher and lower Among the white matter disorders in children, hy-
than in controls, although the differences are less pro- pomyelination is a frequent finding. It is striking that
nounced than in white matter. The differences be- whereas the signal intensity of cerebral white matter
tween patients with primary progressive multiple is high on T2-weighted images in both hypomyelina-
sclerosis and controls were clearly less than between tion and demyelination, diffusion anisotropy can
secondary progressive multiple sclerosis patients and distinguish the two. Whereas diffusion anisotropy
controls, but still significant. decreases in diseases characterized by myelin loss,
DWI and DTI have a place in research on patients marked diffusion anisotropy remains present in hy-
with inherited leukoencephalopathies, but have also pomyelinating conditions. This is an argument sug-
obtained a place in the clinical work-up of this cate- gesting that it is not just myelination of fiber tracts
that is responsible for the anisotropy, but rather that
the tracts themselves are responsible (Fig. 106.15).
Table 106.4. FA and ADC in different multiple sclerosis lesions
(from Guo et al. 2002) The combination of the nature of white matter
changes and the changes over time in the lesion dic-
Lesions FA ADC tates the findings on DWI and DTI, usually expressed
(¥ 10–3 mm2/s)
as ADC and FA values. This largely explains the con-
Plaque 0.280 1.03 troversies in reports on ADC and FA findings in sev-
Periplaque white matter 0.383 0.79 eral disorders, in particular posterior reversible en-
Normal-appearing
cephalopathy syndrome and some inborn errors of
white matter 0.493 0.74 metabolism.An example is found in the reports in the
Control white matter 0.537 0.73 literature on ADC values in mitochondrial en-
cephalopathy with lactate acidosis and stroke-like
850 Chapter 106 Diffusion-Weighted Imaging

Fig. 106.13.
106.6 Leukoencephalopathies 851

Fig. 106.14. Images of an infant with Canavan disease at the vacuolating myelinopathy, as is maple syrup urine disease, ini-
ages of 4 months (upper row) and 12 months (lower row). Ini- tially with compression of the extracellular space and restrict-
tially the ADC values in the affected white matter areas are low. ed diffusion. The increase in ADC values probably indicates
In the following months they become much higher than the that the tissue disintegrates and becomes rarefied over time
values of normal tissue. Canavan disease is characterized by a

episodes (MELAS). Both low and high ADC values infarctions have low ADC values and follow the time
have been found in fresh lesions (Fig. 106.16). This course and structural changes seen in common in-
could depend on the delay between the insult and the farctions of vascular origin, whereas the “other” le-
measurement. It could also mean that MELAS pro- sions with high ADC values shortly after the insult
duces two different types of lesions, of which the represent another type of pathology, yet to be eluci-
dated.
In ischemic white matter lesions, for example sub-
cortical arteriosclerotic encephalopathy, loss of struc-
tural integrity is expressed by a relatively high ADC
䊴 (1.12 ± 15 ¥ 10–3 mm2/s) and moderately low FA val-
Fig. 106.13. A neonate with maple syrup urine disease. The ues (0.480–0.530). Figures in the same order have
pathological substrate is a vacuolating myelinopathy, which been obtained in patients with CADASIL. In these
only affects myelinated areas. On the T2-weighted images (first vascular disorders there is a strong correlation be-
and second rows) the myelinated parts of the brain show tween the measure of loss of integrity and results of
swelling and higher signal intensity than the surrounding un- cognitive tests.
myelinated structures. The differentiation of abnormal from
normal tissue can be optimized by Trace diffusion-weighted
imaging (third and fourth rows). Note that the optic tract and
chiasm are also myelinated at this stage
852 Chapter 106 Diffusion-Weighted Imaging

Fig. 106.15.
106.6 Leukoencephalopathies 853

Fig. 106.16. A 12-year-old boy with MELAS develops a stroke- temporal infarction and a low ADC in the occipital infarction,
like episode, initially with dysphasia, followed by hemianopia. suggesting the influence of time on the ADC
The ADC map is most informative, showing a high ADC in the

Fig. 106.15. A series of T2- (first row) and T1-weighted (second


row) images of a 6-year-old boy with Pelizaeus–Merzbacher
disease, showing severe hypomyelination. Unlike what one
would expect, Trace diffusion-weighted images (b value 1000,
third row) do not add much information and the ADC values
(fourth row) are close to normal: ADC of frontal white matter is
0.76–0.84, that of the basal ganglia 0.54–0.84, and that of the
cortex 0.84–0.89 ¥ 103 mm2/s
Chapter 107

Magnetization Transfer Imaging

107.1 Techniques weak MT effect is always present, because each slice-


selective pulse will serve as an off-resonance pulse for
Conventional MRI depends on the contribution to the the adjacent slices. This effect is stronger when fast
MR signal of the freely mobile protons of water mol- MR sequences are used with a high number of refo-
ecules because of their great abundance and slow re- cusing pulses.
laxation, leading to a high and sharp resonance with a Myelin forms a major part of the white matter. It is
bandwidth of approximately 20 Hz. Protons bound to a lipid bilayer, in which proteins are embedded. The
macromolecules form a second pool and do not di- major lipids in myelin are cholesterol and glyc-
rectly contribute to the MR signal because of their rel- erophospholipids. The lipid bilayer is wrapped in the
atively low concentration and rapid relaxation, result- form of large lamellae around the axons, with a small
ing in a low resonance with much broader bandwidth quantity of extracellular fluid between the layers. This
of several thousand hertz. The broad peak of these creates a huge surface for interaction between the
frequencies is symmetrically arranged around the bound and free water molecules, which facilitates MT.
resonance of mobile protons. Indirectly these bound Compartmentalization of the extracellular fluid fur-
water molecules can influence the MR signal because ther contributes to the MT effect. The MT effect of the
there is a physicochemical exchange and cross-relax- myelin bilipid–water interface is responsible for the
ation between the two pools of protons. Dipole–di- bright appearance of myelin on T1-weighted images.
pole coupling results in an exchange of magnetiza- Both cholesterol and cerebrosides are particularly
tion, called magnetization transfer, where each pool important in determining the myelin bilipid–water
has influence on the relaxation rate of the other. Wolff interface interactions, which facilitate T1 relaxation
and Balaban (1989) demonstrated in vitro and vivo due to MT. The signal loss of normal white matter due
that it is possible to selectively excite the bound pool to MT effects ranges from 30% to 50%. It depends on
of protons by using a radiofrequency (RF) pulse that the RF power applied and scan parameters. Gray mat-
is off-resonance for the free water protons. The exci- ter shows a somewhat smaller MT effect than white
tation of the macromolecular bound protons leads to matter. CSF, containing practically no macromole-
transfer of magnetization to the mobile water protons cules, shows a 0–2% signal reduction due to MT.
in the magnetization transfer (MT) process. This re- Magnetization transfer imaging (MTI) is used for
sults in decrease of the MR signal after on-resonance two reasons. First, MT effects lead to a general signal
excitation. reduction of the MR image, often referred to as back-
The rates of MT between macromolecular bound ground suppression. The diminution of signal inten-
protons and mobile water protons differ between tis- sity of brain tissue can be applied to increase the con-
sues, resulting in different degrees of signal decrease trast between nonenhancing and enhancing tissue in
for different tissues, generating a new form of MR studies with contrast injection. Background suppres-
contrast. The rate of MT is dependent on a number of sion with MT is also used in MR angiography.
factors: the relative proportion of the two pools of Secondly, MTI can be used as a separate imaging
protons; the magnetic field strength; the power of the technique to characterize lesions. In tissue character-
preirradiation with an off-resonance RF prepulse; ization, the magnetization transfer ratio (MTR) is
and the imaging parameters. The contrast on T1- commonly used. The formula is simple: MTR =
weighted images is normally already partially deter- [(M0 – Ms) / M0] ¥ 100%, in which M0 is the signal
mined by magnetization transfer, so the more T1- intensity obtained without the MT prepulse, Ms or
weighted the image, the smaller the extra MT effect. Msat the signal intensity with the prepulse applied.
The highest MT effect is obtained when an MT pre- The stronger the MT effect, the higher the MTR. MTR
pulse is applied to a proton density spin echo or gra- is lower when there is less effect of the macromolecu-
dient echo sequence. The initially used continuous ir- lar proton pool. Thus, MTR is a measure of the degree
radiation with an off-resonance frequency, adminis- of the structural integrity of brain tissue.
tered with a separate RF unit, is currently replaced by MT can be applied in different ways. MTR mea-
a sinc-shaped pulse wave of short duration, applied surements can be displayed as a MTR map in which
between each on-resonance excitation using the main each voxel value represents the percentage of signal
RF transmitter. In conventional multislice MRI a loss caused by MT (Fig. 107.1). The MTR map allows
107.2 Normal Age-Dependent Changes 855

Fig. 107.1. The upper row shows a


T2-weighted image without MT pulse
and a T1-weighted image in a patient
with multiple sclerosis.The second row
shows on the left side the T2-weighted
image with MT pulse. From the images
without and with MT pulse, the MTR
map can be calculated, which is
displayed on the right. From this
map MTR values (percentage) can be
obtained per voxel or per ROI

voxel-by-voxel measurement or estimation of the phy. A measure of atrophy can be obtained by divid-
average MTR for a specific region of interest. Another ing the number of voxels representing CSF by the
approach of MTR data that has attracted much atten- total number of segmented voxels.
tion is the creation of MTR histograms, either of the A drawback of both 2D and 3D MT is that MT val-
whole brain parenchyma or of segmented areas, such ues vary considerably between scanners, even with
as the basal ganglia or the frontal lobe.White and gray similar implementations. This is especially important
matter can also be separated. In a number of postpro- in multicenter trials, and in follow-up studies when a
cessing steps the intracranial contents are separated different scanner is used for the same patient.
from the skull and orbital contents by a semiautomat-
ed software program. In the next step MTRs for every
intracranial voxel are calculated. Implementation of a 107.2 Normal Age-Dependent Changes
threshold value of 10–20% separates the CSF from the
brain parenchyma. Voxels with a value higher than Important changes in MT have been reported related
the 10–20% threshold are defined as brain parenchy- to brain maturation. With increasing density and
ma. The frequency distribution of MTR values can complexity of brain tissue, MT changes, mainly ex-
now be displayed as an MTR histogram of the whole pressed as an increase in MTRs. The changes in the
brain (Fig. 107.2). The peak height is defined by the MT of white matter run parallel with the progress of
highest number of voxels with a certain MTR value. myelination. Fiber density or axonal density, however,
This measure is influenced by changes in brain tissue plays an important role as well. Generally speaking,
related to demyelination, gliosis, and axonal loss, but with increasing age of the infant, there is an increase
also by atrophy. The peak value, therefore, has to be in MT effect and a higher MTR. This effect is greater
normalized by dividing the peak height by the total in white matter than in gray matter structures. In the
number of segmented voxels. The relative peak height neonatal brain MTR values are fairly homogeneously
thus obtained is a measure of the amount of remain- distributed, related to the fact that at that time there is
ing brain tissue, independent of brain size and atro- little structural difference between gray and white
856 Chapter 107 Magnetization Transfer Imaging

Fig. 107.2. Normalized MTR, ADC and FA histograms from the


brain parenchyma of a 38-year-old healthy person that were
obtained after removal of cerebrospinal fluid and extracere-
bral tissue pixels. Normalized MTR histograms have proven to
be very useful in follow-up studies in many disease conditions.
It remains to be seen whether combining histogram analysis
of ADC and FA values with MTR histograms will yield valuable
information. From Rovaris et al. (2003), with permission

matter. MTR values in the unmyelinated neonatal to improve the results of MR angiography, where the
white matter are in the order of 13–19%, followed by interest lies in the depiction of arteries and veins and
a two- to threefold rise to 34–38% with myelination. less in the visualization of background structures. For
Local differences in fiber density and myelin density this reason, MT prepulses are a standard part of many
lead to increasing regional variation in MTR. The MR angiography pulse sequences. Background re-
highest MTR values after myelination are found in the duction can also be applied to improve the effect of
corpus callosum and posterior limb of the internal contrast enhancement. This can be used to reduce the
capsule, the lowest values in the frontal and occipital contrast dose or to have more contrast effect with the
white matter; this is probably related to fiber density. same dose, for instance in the search for the number
Overall the most rapid changes in MTR in white mat- of enhancing multiple sclerosis (MS) lesions or mul-
ter occur during the first year of life. The changes in tiplicity of metastases.
MTR of gray matter are less conspicuous and level off The second application is based upon the possibil-
at the age of 10 months. The peak height of the MTR ity offered by MT to give a quantitative impression of
histogram, however, continues to change over a the structural integrity of brain tissue. In order to as-
longer period of time, up to the age of approximately sess the local structural integrity two types of MT
4 years. postprocessing are currently in use: a 2D estimation
of the MT effect, preferably as a MTR map, or mea-
surement of the MT effect in regions of interest. By
107.3 Magnetization Transfer using whole brain histograms, the lesion load of the
in Disease States entire brain can be assessed. These techniques are ap-
plied to disorders characterized by focal or diffuse
MT prepulses, MTI, and MTR can be used for a num- disintegration of cerebral tissue. They can be used to
ber of clinical applications. First of all, background quantitatively assess the degree of tissue damage in
reduction, as produced by MT prepulses, can be used areas that are also abnormal on conventional MR
107.3 Magnetization Transfer in Disease States 857

Table 107.1. MTR and axonal density in normal appearing white matter and multiple sclerosis lesions versus degree of
T1-hypointensity
Degree of T1-hypointensity MTR (range) % axonal density (range)

normal appearing white matter (n=24) 0.32 (0.26–0.36) 90 (60–100)


isointense (n=18) 0.24 (0.21–0.33) 80 (20–100)
mildly hypointense (n=38) 0.24 (0.16–0.31) 50 (10–100)
severely hypointense (n=53) 0.15 (0.10–0.28) 30 (0–70)

images, but they are also used to assess structures Table 107.2. MTR in different zones of cerebral X-linked
that are normal-appearing on conventional images. adrenoleukodystrophy
Changes in MT effect may occur in regions where Tissue MTR (%)
conventional MR techniques do not show abnormali-
ties, for instance in normal-appearing white and gray Unaffected white matter 46
matter in MS, wallerian degeneration, amyotrophic Zone of inflammation and partial myelin loss 35
lateral sclerosis, systemic lupus erythematosus, Zone of severe myelin loss and gliosis 20
AIDS-related dementia, Alzheimer disease, X-linked
adrenoleukodystrophy, and in many other disorders. (Melhem et al. 1996)
In all these disorders, lower MTR values in normal-
appearing structures signify a diminution of struc-
tural integrity and the presence of a disease state. A correlative MTR–histopathology study in multi-
MT has been applied most extensively in MS, ple sclerosis has shown that myelin is not the only fac-
adding to our understanding of the disease. First of tor involved in the MT effect.A significant correlation
all, lesions visible on conventional images can be could be demonstrated between MTR and axonal loss
studied. MTR values for lesions visible on T2-weight- (Van Waesberghe et al. 1999), indicating that the MTR
ed MR images are significantly lower than for nor- reflects not only myelin density but also axonal den-
mal-appearing white matter, although with a wide sity.
range of MTR values. Average MTR for MS lesions Table 107.1 shows the relationship between axonal
was found to be 26.3%, with a lower mean value in density and MTR. It also demonstrates the relation-
chronic progressive (23.3%) than in relapsing-remit- ship between hypointensity of MS lesions on T1-
ting MS (27.6%) (Filippi 1999). Follow-up studies weighted images, MTR, and axonal density (Van
show a further reduction of MTR values over time, Waesberghe et al. 1999).
more pronounced in patients with secondary pro- MTR has been applied to compare MS clinical sub-
gressive MS than in those with relapsing-remitting types (relapsing-remitting MS, secondary progressive
MS. Decreased MTR values were also found in nor- MS, and primary progressive MS) and to correlate the
mal-appearing white matter in the neighborhood of data with disability scores. While the MTR of white
focal T2 lesions. Using threshold techniques, lesion matter is not significantly different for the different
segmentation can be obtained and the MTR lesion MS subtypes, MTR histogram analysis reveals a sig-
load can be estimated. MTR histogram analysis re- nificant distinction between relapsing-remitting and
veals that there is a loss of high MTR values and a gain progressive MS and between primary and secondary
of voxels with low MTR values in MS patients as com- progressive MS. Findings in primary progressive MS
pared to normal persons. The increase in voxels with are interesting. Primary progressive MS shows few le-
a low MTR value, however, makes up for only a low sions on conventional MR. MTR histograms show a
percentage (in the order of 15%) of the total decrease lower peak height in primary progressive MS than in
in voxels with a high MTR value, suggesting that a all the other MS subgroups, with an about normal av-
higher percentage of “lost” voxels can be attributed to erage brain MTR and normal peak position, suggest-
white matter atrophy. ing that there is a markedly reduced amount of nor-
MT has been one of the techniques confirming that mal brain tissue (Filippi 1999). The correlation be-
normal-appearing white matter in MS is not normal. tween MTR results and disability is better than the
With MT, subtle but consistent abnormalities have correlation between estimations of lesion load and
been found, more pronounced with increasing dis- disability. Cognition tests showed correlations with
ability and in progressive MS. MT has also been used MT parameters of brain atrophy and remaining nor-
to study gray matter involvement. MTR of gray mat- mal brain tissue (Kalkers et al. 2001). Correlation be-
ter is significantly lower in patients with relapsing-re- tween gray matter involvement and EDSS is seen as
mitting MS than in controls, confirming that MS is a an indication that estimation of gray matter abnor-
diffuse disease affecting the whole brain. mality may be useful in assessing clinical disability.
858 Chapter 107 Magnetization Transfer Imaging

MTR can be used in monitoring of the disease in morbidity and mortality of the procedure, and the
MS. In view of all the therapeutic trials going on in fact that one cannot predict which boy carrying the
this disease, objective and quantitative parameters biochemical and genetic defect will develop the dev-
are essential to monitoring the disease course and the astating cerebral demyelinating disease and which
efficacy of the treatments applied. For instance, MTR boy will develop adrenomyeloneuropathy, the later-
has been used to estimate the effect of interferon be- onset and milder form of the disease, hematopoietic
ta-1a (Kita et al. 2000) and interferon beta-1b (Richert stem cell transplantation cannot be performed at an
et al. 1998) in MS patients. In these studies MTR was early age as a preventive measure. Careful monitoring
found to be comparable with the more common esti- at regular intervals is presently the best solution. Con-
mation of the number of new enhancing lesions over ventional MRI is important but it has been shown
time. repeatedly that proton MRS, diffusion tensor imag-
Another important application of MT is in X- ing, and MT are more sensitive and are able to
linked adrenoleukodystrophy. Monitoring of the onset demonstrate the onset of the disease and quantify
of cerebral demyelination is extremely important in disease progression where conventional MRI does
young boys. As soon as the first significant signs of not (yet) show changes. It has been shown in X-linked
progressive cerebral demyelination are found, hemato- adrenoleukodystrophy that MTRs correlate closely
poietic stem cell transplantation is performed in an with the degree of loss of tissue integrity (Melhem et
attempt to halt the disease. Because of the significant al. 1996).
Chapter 108

Magnetic Resonance Spectroscopy: Basic Principles


and Application in White Matter Disorders
M.S. van der Knaap, P.J.W. Pouwels

108.1 Basic Principles moments have a random direction and there is no net
magnetic vector. The intrinsic magnetic phenomena
Pauli was the first, in 1924, to suggest that electrons can be detected by applying an external magnetic
spin at high speed. Because the spinning electrons field. The nuclear magnetic moments tend to align
have mass, they have a certain angular moment and, themselves parallel or antiparallel to the external
thus, as spinning electric charges, a magnetic mo- magnetic field. On the other hand, however, nuclei
ment. Later, it was observed that certain nuclei also tend to be in constant random motion due to thermal
have magnetic moments. What these nuclei have in effects. The percentage of nuclei that align with the
common is the fact that they are isotopes with an odd external magnetic field depends on the strength of
atomic mass and/or odd atomic number, whereas the magnetic field relative to the random thermal
nuclei with both an even atomic number and an even effects. Slightly more nuclei align with the external
atomic mass do not have a magnetic moment. Subse- magnetic field than against it, because the first posi-
quently it was noted that when these nuclei are placed tion is more stable than the second. Stability is relat-
in a powerful magnetic field, they show a precession- ed to the amount of energy that a nucleus possesses.
al motion about the axis of the field. In 1946 Bloch Stability is greater at low energy levels. In fact, there is
and Purcell simultaneously discovered the possibility a continuous transition of nuclei between high- and
of resonant energy absorption and emission of pre- low-energy positions, but there is a slight net surplus
cessing nuclei, the basis of magnetic resonance imag- in the low energy position (1:105 at 1.5 T).
ing (MRI) and spectroscopy (MRS). For this work A spinning nucleus precesses about the axis of the
they were awarded the Nobel Prize in 1952. MRS has magnetic field. This motion is called Larmor preces-
developed into a very important tool in molecular sion, and its frequency the Larmor frequency. The
chemistry and physics to reveal molecular structure, Larmor frequency (w) depends on the magnetic field
chemical reaction rates, and diffusion processes. The strength H0 and characteristics of the particular nu-
first spectroscopy experiments on living systems were cleus, as expressed in the gyromagnetic ratio g. This
performed on small-bore systems with tiny objects, dependency is represented in the Larmor equation
such as red blood cells and excised tissue. Wider w = g H0. Resonance is a property of physical systems
bores have since been developed, allowing the study to oscillate at a preferred frequency which is charac-
of muscle disorders and experimental work with teristic of the system. The characteristic frequency is
small animals. Subsequently, in vivo MRS of human referred to as the resonance frequency. The most effi-
brain and other organs has also become possible. cient energy transfer to atomic particles precessing in
Atomic nuclei are composed of protons and neu- magnetic fields occurs in their resonance frequency,
trons. These nuclear particles exhibit spin.A proton is the Larmor frequency. For magnetic field strengths
positively charged, and this spinning charge gener- used in MRS these resonance frequencies are within
ates a small magnetic field. Although a neutron is the radiofrequency (RF) band of the electromagnetic
electrically neutral, its component electrical charges spectrum. A short burst of RF energy is known as an
are not uniformly distributed within its volume, and RF pulse. When an RF pulse is administered, this en-
thus the neutron also generates a magnetic field when ergy is absorbed, and more nuclei move into the high-
spinning, but smaller than that produced by a spin- energy position.When the RF pulse is terminated, the
ning proton. The magnetic moments of these parti- nuclei return to equilibrium and energy is released
cles are directed randomly, and they cancel each oth- at the same frequency. The RF signal emitted can be
er out in nuclei with an even number of protons and detected.
neurons. When the nucleus has an odd number of The basis of MRS is the chemical shift phenome-
neutrons and/or protons, the nucleus has a net spin, non. The atomic nucleus is surrounded by an electron
and this spinning charge has a magnetic moment. cloud and other atomic nuclei. If an external magnet-
These nuclei exhibit the magnetic resonance phe- ic field is applied, precession is also induced in this
nomenon. Nuclei of interest for in vivo spectroscopy electron cloud, resulting in a small magnetic field.
are 1H, 31P, 23Na, and also 13C, 15N, 17O, and 19F as mark- This small magnetic field has an influence on the
ers of biochemically interesting compounds. If there atomic nucleus and modifies the effect of the external
is no external magnetic field, the nuclear magnetic magnetic field at the site of the nucleus. Because of
860 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

this slight change in the local magnetic field, the nu- Table 108.1. Resonance frequencies and relative MR sensitiv-
cleus resonates at a slightly different frequency. This ities at 1.5 T
shift in frequency is called chemical shift. The chemi- Nucleus Larmor frequency Relative
cal shift is determined by the electrochemical envi- in MHz MR sensitivity
ronment of the resonating nucleus, and therefore the 1H 63.86 1
precise shift is characteristic of a particular atomic 31
nucleus in a particular compound. The bandwidth of P 25.85 0.066
19F 60.08 0.83
the RF pulse, of course, must contain all the frequen-
cies that are necessary to excite the particular atomic 23
Na 16.89 0.093
nucleus in compounds of interest. The RF signal ob- 13C 16.06 0.016
tained in an MRS experiment contains many slightly 15N 6.47 0.001
different frequencies. Fourier transformation of the
signal transforms a complex time-domain signal into
a complex frequency-domain signal. The resulting
spectrum depicts all the nuclear resonances as a func-
tion of their frequency. Each peak in the spectrum MRS methods are relatively insensitive. Weak MR
represents the atomic nucleus in a particular com- signals are measured from relatively low-concentra-
pound. The area under each peak is proportional to tion compounds. For this reason spectra are obtained
the number of nuclei producing that peak. MR spec- from relatively large volumes of interest (so-called
tra can be calibrated to yield absolute concentrations voxels). The voxel size necessary to produce a spec-
of the metabolites represented. The chemical shift de- trum with a reasonable signal-to-noise ratio is larger
pends on the magnetic field strength. However, in for nuclei with low abundance and low inherent sen-
MRS the resonance frequencies are not expressed in sitivity. For 1H-MRS voxels typically have a size of
absolute units (hertz) but in relative units (parts per 4–12 ml, whereas for 31P typical voxel sizes are in the
million, ppm) related to the resonance frequency of a range of 24–63 ml. A further improvement of the
given reference compound. The relative units, ppm, spectral signal-to-noise ratio is obtained by signal av-
are independent of magnetic field strength. In this eraging. For 1H-MRS 64–128 acquisitions are com-
way the results obtained in experiments performed at mon, whereas 128–256 acquisitions are generally
different magnetic field strengths become directly used for 31P-MRS. It is also essential to use MR instru-
comparable with respect to chemical shift. Another ments with optimal technical equipment to ascertain
phenomenon that determines the appearance of a optimal primary sensitivity. The magnetic field
spectrum is the influence of other nuclei in the same should be extremely homogeneous over the volume
molecule on the nucleus of interest, which is called analyzed. Any significant inhomogeneity in the mag-
J-coupling. If there is no coupling, the resonance re- netic field spreads out and blurs chemical shift spec-
mains a single peak (a so-called singlet). If a certain tral lines due to the spread of Larmor frequencies
nucleus in particular compound – in the case of across the volume. The resulting spectral line broad-
1H-MRS a proton – is influenced by other protons
ening is undesirable because it reduces the signal-to-
only a few chemical bonds apart, coupling will induce noise ratio and hampers the ability to distinguish two
a splitting of the resonance. Depending on the num- closely neighboring resonance lines. The frequencies
ber of neighboring protons, the resonance may be and frequency separations increase linearly with field
split into a doublet, a triplet, or more complex pat- strength. High magnetic field strengths are therefore
terns. Due to J-coupling spectra will look different at necessary to optimize spectral resolution. High spec-
different field strengths. tral resolution is particularly important for proton
The nuclei that one would like to observe can be se- spectroscopy, where most interest is focused on
lected by using the appropriate RF pulse. MRI most metabolites within a narrow chemical shift range, and
commonly makes use of the magnetic properties of where the peaks of the metabolites often overlap. For
protons and is in fact 1H MRI. The protons that con- MRS in human beings, a magnetic field strength of
tribute to the signal intensity on the images are main- 1.5–4 T is used. In experimental work with animals,
ly present in water and fat. MRS, however, focuses not magnetic field strengths of 4–8 T or even higher are
only on protons (1H-MRS), but also on phosphorus preferred.
(31P-MRS) and, less frequently, on other nuclei such as For the purpose of cleaning the final spectrum or
13C, 15N, 19F, and 23Na. In human tissues, these nuclei
obviating unwanted resonances, the spectroscopist
are present in much lower concentrations than are has access to many suppression and editing tech-
protons, and the overall sensitivity of the nuclei is niques. The largest peak in the 1H spectrum re-
much lower than that of protons. Table 108.1 lists the presents water. The concentration of water is about
resonant frequencies and relative MR sensitivities of 104–105 times higher than the concentration of the
various nuclei at a magnetic field strength of 1.5 T. other metabolites. Consequently, the water peak dom-
108.2 Metabolites 861

inates the spectrum, while the other resonances


are barely visible. For this reason, water suppression
techniques are invariably applied to remove the water
peak from the spectrum and make the other peaks
better visible and quantifiable. Lipids present in
the skull and subcutaneous tissue are another poten-
tial problem, as their concentration is also high
and their resonances overlap with those of interest-
ing brain metabolites. Several techniques are avail-
able to overcome the contamination of a spectrum
by fat: location of the volume of interest far from
the skull, fat suppression techniques, and outer vol-
ume suppression when an entire brain slice is investi-
gated. Fig. 108.1. Localized 31P spectrum of the human brain. The
Even with all these precautions, MRS remains a rel- spectrum is obtained at 1.5 T with the ISIS technique, TR
atively insensitive technique requiring concentra- 3750 ms, 256 measurements, voxel size 63 ml
tions at least in the millimolar range for metabolites
to be visualized. Many important but too diluted
compounds have so far remained undetectable by in 108.2 Metabolites
vivo MRS. In addition, only molecules sufficiently
small and mobile to tumble freely render MR signals In a spectrum from the human brain, 31P-MRS reveals
useful for in vivo work. No useful signals can be seven major peaks (Fig. 108.1): phosphomonoesters
obtained from large molecules such as proteins, even (PME), inorganic phosphate (Pi, including H3PO4,
soluble ones, nor from most membrane compounds H2PO4–, HPO42–, and PO43–), phosphodiesters (PDE),
or small molecules bound to large ones. The broaden- phosphocreatine (PCr), and the g-, a-, and b-phos-
ing of spectral lines makes differentiation of these phate groups of adenosine triphosphate (ATP), res-
molecules impossible. However, under certain condi- onating at 6.5, 4.9, 2.6, 0, –2.6, –8.0, and –16.5 ppm
tions they will contribute to the spectrum as a respectively. ADP contributes to the g- and a-ATP
“macromolecular baseline.” peaks. NAD and NADH contribute to the a-ATP peak.
For in vivo MRS it is important that a volume of in- The main component of the PME peak is phospho-
terest can be selected from which the spectrum is de- ethanolamine. Other components are phospho-
rived. A simple technique is the placement of a sur- choline, glycerol 3-phosphate and sugar phosphates.
face coil over the volume of interest. The spatial defi- Minor components are phosphoserine and phospho-
nition of such coils is rather poor since the intensity inositol. Phosphoethanolamine is a precursor in the
of signal reception decreases with distance. Several anabolism of phosphatidylethanolamine and a prod-
techniques have been devised to improve the spatial uct in the catabolism of sphingomyelin. Phospho-
response of surface coils and to make positioning of choline is a precursor of phosphatidylcholine and
the volume of interest more accurate. One major dis- sphingomyelin and a product in the catabolism of
advantage remains, which is that surface coils are on- sphingomyelin. Glycerol 3-phosphate is a precursor
ly suitable for volumes of interest near the surface. of phosphatidylethanolamine, phosphatidylcholine,
Image-localized single voxel spectroscopy by gradi- and plasmalogens and a product in the catabolism of
ent control allows direct and precise definition of a the same compounds. The PME peak is elevated in all
volume of interest on a proton image. Major advan- rapidly growing tissues with rapid membrane synthe-
tages are the ability to select volumes located in the sis, such as tumors and the growing brain. It is proba-
deeper parts of the brain and the ability to define a ble that the elevation is caused by the enhanced pres-
volume of interest guided by the abnormalities seen ence of compounds meant for the production of
on the image, avoiding contamination of the spec- membrane phospholipids. Generally, the PME peak is
trum by signals from undesired regions. In chemical considered to reflect phospholipid anabolic activity.
shift imaging (CSI), also called spectroscopic imag- This notion has been confirmed by the finding that
ing, spectra are obtained from many contiguous vox- the level of PME correlates linearly with the rate of
els at the same time, covering more or less an entire phospholipid synthesis.
brain slice. 1H CSI is usually applied to investigate one The 31P spectrum of the brain contains a large,
of multiple brain slices of 1–2 cm thickness with an broad peak centered in the PDE region, underlying
in-plane resolution of 1 ¥ 1 cm2. The information thus the remainder of the spectrum (Fig. 108.2). This large,
obtained can be used to construct maps for separate broad peak originates from large, relatively immobile
metabolites, which display the distribution of a par- membrane phospholipids. This broad component is
ticular metabolite over the brain slice. commonly removed from the spectrum by filtering
862 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

known to be present in brain tissue in significant


amounts, is used as a reference.
The chemical shift of Pi relative to PCr is a function
of pH. The pH determines the relative amounts of
H2PO4–,and HPO42– in the equilibrium H2PO4–AHPO42–
+ H+, and the precise resonance frequency of Pi as
a whole. Using the equation of Petroff et al. (1985), the
pH can be deduced from the chemical shift of Pi rela-
tive to PCr. This pH holds for the compartment in
which these phosphates are present, which in the
brain is the intracellular compartment.
Peak assignment in the 1H spectrum is problemat-
ic because of overlapping resonances. Straightfor-
ward assignment is possible for the singlet represent-
ing the N-acetyl methyl resonance of N-acetylaspar-
tate (NAA) at 2.02 ppm, the methyl and the methylene
resonance of total creatine (Cr), including free crea-
tine and phosphocreatine, at 3.02 ppm and 3.93 ppm,
respectively, and the methyl resonances of choline-
containing compounds (Cho) at 3.22 ppm (Fig. 108.3).
The methyl resonance of N-acetylaspartyl glutamate
(NAAG) is a shoulder on the methyl resonance of
NAA with only a slight difference in chemical shift
(2.05 as opposed to 2.02 ppm). It is responsible for
Fig. 108.2. A An unfiltered 31P spectrum of the brain. B The about 10–25% of the NAA peak. Since the singlet res-
same spectrum after filtering.The spectra are obtained at 1.5 T onances of NAA, Cr, and Cho exhibit relatively long T2
with the ISIS technique, TR 3750 ms, 256 measurements, voxel relaxation times, they may be specified in spectra at
size 63 ml long echo times (135 or 270 ms) (Fig. 108.3a). When
shorter echo times (15–30 ms) are used, a consider-
ably increased number of resonances with short T2
relaxation times can be visualized (Fig. 108.3b). Most
techniques as a standard processing procedure. The obvious are the appearance of a strong signal from
remaining PDE peak contains mainly contributions multiple collapsed resonances of myo-inositol (mIns)
from more mobile phospholipids and relatively at 3.56 ppm.A complex pattern of coupled resonances
minor contributions from phospholipid breakdown between 2.1 and 2.5 ppm together with a further
products, including glycerophosphocholine and glyc- group of resonances around 3.8 ppm are assigned to
erophosphoethanolamine, freely soluble cytosolic glutamine and glutamate. Resonances of g-aminobu-
molecules. These latter compounds are catabolic tyric acid (GABA) are overlapped by the larger reso-
products of phosphatidylcholine, phosphatidyletha- nances of Cr, glutamate, and NAA (GABA resonances
nolamine, and plasmalogens. at 1.90, 2.30, and 3.03 ppm). Additional resonances
The 31P spectrum contains multiple peaks repre- are seen originating from the aspartyl group of
senting high-energy phosphorus compounds (ATP NAA (2.48, 2.60, and 2.66 ppm), glucose (3.43 and
and PCr) and Pi. Intracellular PCr acts via the creatine 3.80 ppm) and scyllo-inositol (3.35 ppm). The singlet
kinase reaction as a buffer to maintain a constant ATP of glycine (3.55 ppm) overlaps with the mIns peak.
level in the face of variable energy demands. In condi- The quartet of taurine (centered at 3.35 ppm) partly
tions of decreasing ATP, PCr reacts with ADP to pro- overlaps with scyllo-inositol. Lactate is not usually
duce ATP and creatine with creatine kinase as catalyz- visible under normal conditions, but can be visual-
ing enzyme. The reaction is reversible and PCr can be ized as a doublet centered at 1.33 ppm if elevated.Ala-
re-synthesized from creatine and ATP. This so-called nine can only be seen when increased in concentra-
creatine phosphate shuttle facilitates energy distribu- tion, then giving rise to a doublet at 1.47 ppm. Pyru-
tion and responds to energy demands. The PCr/Pi vate is below the level of detection under normal cir-
ratio is often used as a measure of the energy status of cumstances, but when elevated it gives rise to a single
the brain. The ATP level only drops when PCr is ex- peak at 2.36 ppm. In case of elevated tissue levels of
hausted. In view of the metabolic stability of ATP, it free lipids, for instance as a result of myelin break-
has often been used as an internal reference for quan- down or spectral contamination by fat from the skull,
titation. In particular the b-ATP peak, which does broad resonances are seen at 0.9 and 1.3 ppm origi-
not contain any contributions of other compounds nating from the methyl and methylene groups of the
108.2 Metabolites 863

NAA and its derivative NAAG are synthesized in neu-


ronal mitochondria. However, the catabolic enzyme
for NAA hydrolysis, aspartoacylase, is expressed only
in oligodendrocytes, and the catabolic enzyme for
NAAG is expressed only in astrocytes. Little is known
about the role of NAA in normal function or disease
states. Possible functions include a role as precursor
of the putative neurotransmitter NAAG, as storage
form of the neurotransmitter aspartate, as acetyl
donor in lipid synthesis (important in myelin synthe-
sis), as stabilizer of the concentration of acetyl-CoA,
as molecular water pump, and as having a role in os-
motic regulation, protein synthesis, and cell signal-
ing. NAA is considered to be a neuron- and axon-spe-
cific marker. In disease conditions NAA is often low,
related to neuronal or axonal dysfunction or loss. Par-
tial recovery of NAA level has been described in im-
proving cerebral disorders. It is, therefore, not correct
to interpret a decrease in NAA as a straightforward
indication of irreversible neuronal loss. However, it is
difficult to separate the effects of recovery of NAA
concentrations in recovering neurons from the effects
of tissue contraction after removal of damaged tissue
elements. Temporary reductions in NAA have been
ascribed to temporary metabolic depression and de-
creased mitochondrial energy production.
The Cr peak represents the total amount of crea-
tine and phosphocreatine present in the creatine ki-
nase shuttle. The total creatine pool remains fairly
constant under a variety of conditions. For this reason
Cr has often been used as internal reference for quan-
titation. However, since Cr is not constant under all
Fig. 108.3. Localized 1H spectra of parietal cortex (12 ml) of a conditions, other means of referencing are preferable.
4-year-old healthy girl, obtained at 1.5T. A At a long echo time The Cr concentration of glia is relatively high and
of 135 ms (PRESS, TR/TE = 3000/135 ms) only singlet reso- consequently increased Cr is seen in conditions of
nances of NAA, Cr, and Cho are visible. B At a short echo time gliosis. Cr is only present intracellularly, and Cr has
of 20 ms (STEAM, TR/TE = 6000/20 ms) additional resonances also been considered a marker for cellular density.
of mIns, Glx (glutamate and glutamine), and other metabolites The Cho peak contains contributions from various
are present in the spectrum compounds. Water-soluble choline-containing com-
pounds in the brain, including choline, glycerophos-
phocholine, and phosphocholine, are responsible for
lipids. Because of severe overlap of resonances in the largest part of the peak. A smaller contribution to
spectra recorded with short echo times, it is difficult the peak comes from choline-containing phospho-
to quantify metabolite concentrations. Special tech- lipids, including sphingomyelin and phosphatidyl-
niques such as spectral editing have been developed choline. Acetylcholine is also a water-soluble com-
to quantify glutamine, glutamate, and GABA sepa- pound, but has a low concentration. Elevated Cho is
rately. Alternatively, a spectrum can be quantified by seen in conditions of high cell density and enhanced
describing it as a linear combination of model spectra membrane turnover, such as brain growth, myelina-
of all individual metabolites included in a database tion, demyelination, inflammation, and tumor growth.
using prior knowledge. Cho is also an osmolyte and its level may reflect com-
NAA is a compound found at high concentration in pensation for osmotic changes.
the CNS, whereas only traces are found in other tis- Lactate occupies a special position in energy me-
sues. In the mature brain NAA is almost entirely con- tabolism. Being the end product of glycolysis, it rises
fined to neurons and their axons: 40% of the NAA is in concentration whenever the glycolytic rate in a vol-
in the nerve-ending mitochondrial fraction and 60% ume of tissue exceeds the tissue’s capacity to catabo-
is in the cytosol. In cell cultures NAA has also been lize lactate or export it to the blood stream. Lactate
demonstrated in oligodendroglial precursor cells. levels are increased under conditions of anaerobic
864 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

glycolysis, for example in failure of energy supply or active tissue damage, such as hypoxia–ischemia, as
in respiratory chain defects. Macrophages are highly one of the so-called excitatory amino acids. Apart
dependent on anaerobic glycolysis for their energy from its role as neurotransmitter and precursor of
production. Elevated lactate is also seen in conditions GABA, glutamate is important as a Krebs cycle com-
characterized by the presence of increased numbers ponent via a-ketoglutarate. Considering its predomi-
of macrophages, such as active demyelination and nantly glial localization, glutamine may be considered
tissue necrosis. Increased lactate secondarily leads to a glial marker.
increased alanine, which may become visible in the Taurine is a key component of the cytosol buffer
spectrum. and serves to optimize cytosol buffering capacity at
The mIns peak is also a composite peak, containing physiological pH. It is involved in neurotransmission,
contributions from myo-inositol as main component, osmoregulation, and brain growth. It controls magne-
and, additionally, inositol monophosphate, phos- sium homeostasis and calcium homeostasis.
phatidyl inositol (a membrane phospholipid), and
inositol diphosphate as minor components. Inositol
diphosphate is, however, a very important compo- 108.3 Normal Values: Age Dependency
nent, functioning as a second or third messenger for and Regional Variability
various hormone actions. Inositol diphosphate is ac-
tive in releasing calcium from the endoplasmic retic- The major biochemical changes related to processes
ulum and mitochondria. Many enzymes are depen- of brain maturation are reflected in spectroscopic
dent upon the inositol-induced calcium release. The changes (Figs. 108.4 and 108.5).
function of myo-inositol itself is still largely un- The most striking finding in 31P-MRS (Fig. 108.4)
known, but it may be the storage form of the inositol- of neonatal brain is a very high PME peak, in particu-
dependent messenger system. Another important lar in fetuses and preterm babies, in whom the PME
function of myo-inositol is that of osmoregulator. In peak is higher than all other peaks in the spectrum.
diabetes mellitus, inositol phosphate has been con- Relative to b-ATP, PME decreases with ongoing brain
sidered as a mediator of some of the secondary com- maturation, to reach final values at about 2 years of
plications, in particular polyneuropathy. In galac- age. In terms of absolute metabolite concentrations
tosemia some of the late complications have been at- also, PME decreases with increasing age. The eleva-
tributed to depletion of myo-inositol. Also in hepatic tion of the PME peak in early development is mainly
encephalopathy a depletion of myo-inositol is seen. related to elevated phosphoethanolamine. The high
myo-Inositol is only present in glia and mIns can PME peak can be attributed to active membrane
therefore be used as a glial marker. In conditions of phospholipid synthesis, in particular active myelina-
gliosis, the mIns level is increased. scyllo-Inositol is a tion. Myelination is almost complete at the age of 2
stereoisomer of myo-inositol and usually the varia- years.
tion in myo-inositol level and scyllo-inositol level PDE is very low in fetal brain. PDE increases with
change simultaneously. However, in some individuals ongoing brain maturation, but is still relatively low in
high scyllo-inositol levels are found in the presence of term neonates. The PDE/b-ATP ratio and absolute
normal myo-inositol levels. concentrations of PDE increase with age. Final values
Glutamate is the most important excitatory neuro- are reached at the age of about 2 years. PDE originates
transmitter, whereas GABA, formed by decarboxyla- to a large extent from phospholipids and to a smaller
tion from glutamate, is the most important inhibitory extent from low-molecular-weight soluble metabo-
neurotransmitter. In the presynaptic neuron gluta- lites in phospholipid breakdown. The increase in PDE
mine is converted to glutamate and ammonia by glu- with increasing brain maturation is primarily related
taminase. Glutamate can either be converted to GABA to the increasing membrane density of the brain,
by glutamic acid decarboxylase or be released in the which is largely caused by progressing myelination.
synaptic cleft. After release by the presynaptic neu- With increasing membrane content of the brain,
ron, glutamate is taken up by the astrocyte in which it membrane turnover also increases, and, accordingly,
is processed by glutamine synthetase into glutamine, so does the concentration of intermediary products
which is transported back to the presynaptic neuron. of phospholipid breakdown, contributing to the
This cycle operates between the presynaptic neuron, height of the PDE peak.
the neuronal cleft, and the astrocyte. Hyperammone- The PME/PDE ratio can be used as a maturation
mia has a great impact on this cycle by stimulating index for brain development.
glutamine synthesis via glutamine synthetase, by pos- There are major changes in energy metabolism re-
sible inhibition of glutaminase, and by inhibition of lated to brain maturation. On the basis of findings in
glutamate reuptake by the astrocyte. In hyperam- animal experimental research it was assumed that the
monemia glutamate decreases and glutamine in- cerebral concentration of ATP, reflected in b-ATP,
creases. Glutamate may be elevated in conditions of does not change with age and is constant irrespective
108.3 Normal Values: Age Dependency and Regional Variability 865

Fig. 108.4. Four 31P spectra of the brain obtained at different and PCr relative to b-ATP with increasing age. The spectra are
ages after birth at term: 1 month (A), 4 months (B), 2 years (C), obtained at 1.5 T with the ISIS technique, TR 3750 ms and 256
and 15 years (D).Note the decrease in PME and increase in PDE measurements, voxel size 54–63 ml

of maturational stage. However, evidence has been NAA is low at birth. NAA/Cr ratio and absolute
provided by Buchli et al. (1994) that the ATP concen- NAA values have been shown to increase after birth.
tration of the brain increases significantly with age The steepest increase occurs during the first year of
during early development. Relative to b-ATP, PCr life, with very gradual increases during childhood,
increases rapidly after birth and reaches final values and final values are reached around the age of 10
after a few months. Pi/b-ATP does not undergo signif- years. The increase in NAA can be attributed to
icant changes. Buchli et al. (1994), using absolute processes of neuronal maturation, including increase
quantitative data, found some postnatal increase in Pi in number of axons, dendrites, and synaptic connec-
and a more marked increase in PCr and ATP. So, there tions.
is a maturational increase in the creatine kinase reac- There is a rapid increase in the cerebral Cr concen-
tion rate with age. PCr/Pi is considered a measure of tration before and around term, and final values are
the phosphorylation potential and energy status of reached after a few months. The difference from adult
the examined tissue; the increase may, therefore, im- values is small but significant. Therefore, Cr is an in-
ply an increase in energy reserve in infant brain tis- ternal reference of limited value, especially in early
sue. This increase would be compatible with the in- development.
crease in rate of energy metabolism observed in post- Cho is high in neonates and decreases thereafter.
natal cerebral maturation. Cho/Cr ratio and absolute Cho values decrease with
Human cerebral pH shows some decrease with on- increasing cerebral maturation, and final values are
going cerebral maturation. reached after 3–5 years. The high Cho in early devel-
1H spectra also undergo major developmental
opment can be attributed to a high rate of membrane
changes. In neonates, in particular in preterms, synthesis and turnover, in particular related to the
the spectrum is dominated by Cho and mIns peaks, process of myelination.
whereas in the adult the NAA peak is largest (Fig. The mIns concentration is high at birth and de-
108.5). creases rapidly. Final values are reached within the
866 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

Fig. 108.5. Four 1H spectra of parietal white matter obtained (B), at 4 years (C), and at 9 years (D). Note the increase in NAA
at 1.5 T (STEAM, TR/TE = 2500/20 ms) and at different ages: and decrease in Cho and mIns relative to Cr with increasing
32 weeks postconceptional age (A), 3 months after term birth age

first year of life. The significance of the high mIns scribed above are valid for brain structures in gener-
concentration in early development is not known. al, but they may differ in detail for different brain
The resonances originating from taurine are high structures. These regional and age-dependent spec-
at birth, and gradually decrease. tral changes make it necessary to obtain region-spe-
The peaks originating from glutamine and gluta- cific normal values for different ages.
mate do not change significantly.
Metabolite concentrations show regional variabili-
ty, determined by the structures included in the study 108.4 MRS in Cerebral Disorders:
(Fig. 108.6). Depending on the size of the individual Process-Specific Abnormalities
voxels, partial volume effects are important. Due to
the cortical foldings it is not possible to obtain spec- Two types of spectroscopic abnormalities are seen in
troscopic data from a voxel containing cortex only, white matter disorders:
and it is difficult to choose a voxel containing white 1. Process-specific spectroscopic abnormalities, re-
matter only. The impact of the problem is more severe lated to delayed maturation and tissue damage.
on 31P-MRS than on 1H-MRS, because larger voxels 2. Disease-specific spectroscopic changes, directly
are used. In 31P-MRS, PME, PDE, and Pi are higher in related to the particular disorder under investiga-
white matter than in gray matter, whereas PCr is high- tion.
er in gray matter. In 1H-MRS it is possible to obtain
spectra from voxels containing mainly cortex or In this section, the process-specific abnormalities are
white matter or central nuclei. It has been shown that discussed.
NAA, Cr, mIns, glutamate, and glutamine are higher In retarded brain maturation, MRI only shows
in gray matter spectra than in white matter spectra, delayed myelination, because, apart from gyration,
whereas Cho is higher in white matter spectra processes of neuronal maturation cannot be visual-
(Fig. 108.6). The normal spectroscopic findings in the ized. In MRS one expects metabolite levels appropri-
intact brain stem, cerebellum, and basal ganglia are ate for a younger age than the age of the patient. The
again different. The maturational changes as de- use of MRS to provide quantitative measures for
108.4 MRS in Cerebral Disorders: Process-Specific Abnormalities 867

Fig. 108.6. 1H spectra of a 4-year-old healthy girl, obtained (C) basal ganglia (5 ml), and (D) cerebellar vermis (8 ml). The
with STEAM (TR/TE = 6000/20 ms) at 1.5 T. Spectra are from spectra are plotted on the same vertical scale to allow a quali-
(A) parietal cortex (10 ml), (B) parietal white matter (4 ml), tative comparison

processes of cerebral maturation is limited by the


rather wide normal variation.
In demyelinating disorders, it is primarily the
myelin sheath that is lost; secondarily, axonal damage
and loss occurs. Histologically, demyelinating disor-
ders are characterized by rarefaction of the white
matter with respect to its normal constituents. Gliot-
ic scar tissue fills in the free spaces that arise as a con-
sequence of the loss of myelin and axons. Conse-
quently, demyelinating disorders do not lead to sig-
nificant cerebral atrophy in the early stages. Only
when the demyelination is severe and long-standing
does atrophy ensue. The rarefaction of white matter
implies that the total amount of membrane phospho- Fig. 108.7. 31P spectrum of a patient with severe demyelina-
lipids per volume of brain tissue decreases, resulting tion. Note the very low PDE. Compare this spectrum to that
in a decrease in PDE in the 31P spectrum (Fig. 108.7). of Fig. 108.1, which was obtained from a normal child of the
The decrease in the ratio of PDE to b-ATP has been same age
shown to be proportional to the extent of the demyeli-
nation. The PME peak, which is proportional to the
rate of membrane phospholipid synthesis, remains panying demyelination are reflected in a decrease in
normal until demyelination is very severe. In severe NAA in the 1H spectrum. The decrease in NAA occurs
demyelination a variable decrease in PME/b-ATP ra- early in the process. The extent of NAA loss can vary
tio is observed. The neuronal damage and loss accom- considerably, depending on the stage of the disease
868 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

Fig. 108.8. 1H spectra obtained with STEAM (TR/TE = 6000/ Fig. 108.9. 1H spectra of white matter affected by demyelina-
20 ms) at 1.5 T. A The white matter spectrum (5 ml) of a tion (4–5 ml, STEAM, TR/TE = 6000/20 ms, 1.5 T) show variable
9-month-old girl with advanced-stage infantile Krabbe dis- extents of metabolic abnormalities. A In a 4-year-old X-linked
ease shows almost complete absence of NAA, increased adrenoleukodystrophy patient with nonprogressive lesions,
Cho and mIns, as well as a clear elevation of lactate, represent- metabolic abnormalities in the affected white matter are lim-
ed by the doublet at 1.33 ppm. B The white matter spectrum ited to a small decrease of NAA and increases of Cho and mIns.
(5 ml) of a 4-year-old girl with the much slower disease course B In a 10-year-old X-linked adrenoleukodystrophy patient with
of late-infantile/early-juvenile Krabbe disease is characterized highly active demyelination, a complete absence of NAA,
by a strong elevation of mIns and a less severe decrease of a strong increase of Cho, and lipid resonances at 0.9 and
NAA 1.3 ppm, overlapping with a doublet of lactate, are found

and destructiveness of the process, as illustrated in 108.10a). In the end-stage phase of demyelination,
Fig. 108.8 for patients with infantile and juvenile Cho decreases again, lactate and lipids disappear, and
Krabbe disease. In active demyelination Cho is elevat- the spectrum becomes “empty,” dominated by a high
ed, related to enhanced membrane lipid turnover mIns peak. The cortex spectrum remains relatively
(Fig. 108.9). In addition, elevated protein and lipid intact (Fig. 108.10b).
peaks at 0.9 and 1.3 ppm are occasionally seen, relat- In primary neuronal degenerative disorders, neu-
ed to the enhanced presence of myelin breakdown rons undergo degenerative changes, die, and disap-
products (Fig. 108.9b). A variable increase in lactate pear, together with their axons and myelin sheaths.
occurs in active demyelination, presumably related to White matter rarefaction does not occur and gliosis is
the infiltration of the tissue by macrophages (Fig. usually inconspicuous. Tissue loss is primarily evi-
108.9b). Increased levels of glutamate/glutamine have dent as atrophy.As expected, the PDE/b-ATP ratio has
been observed, which may be related to processes been found to remain within the normal range, even
of active tissue degeneration. The concentration of in far advanced disease. The neuronal damage is
mIns increases as a consequence of gliosis (Fig. reflected in a decrease in NAA, but the decrease in
108.4 MRS in Cerebral Disorders: Process-Specific Abnormalities 869

Fig. 108.10. The 1H spectrum of affected white matter (A; Fig. 108.11. The 1H spectrum of white matter (A; 5 ml, STEAM,
5 ml, STEAM, TR/TE = 6000/20 ms, 1.5T) of a 10-year-old girl TR/TE = 6000/20 ms, 1.5 T) of an 8-year-old boy with the Turk-
with an advanced stage of metachromatic leukodystrophy ish variant of late-infantile neuronal ceroid lipofuscinosis
only contains signals of mIns, Cho, and Cr, indicating the pure shows a marked decrease of NAA and an increase of lactate.
gliotic contents of the tissue. B In contrast, the spectrum of B The loss of NAA is also pronounced in the cortex spectrum
the cortex (8 ml) still contains a relatively high signal of NAA (8 ml). In the cortex spectrum all signals are relatively low due
to atrophy and admixture of more CSF than usual

NAA underestimates the neuronal/axonal loss as


compared to primary demyelinating disorders be-
cause of tissue contraction (Fig. 108.11a). However,
in neuronal degenerative disorders the decrease in
NAA in the cortex spectrum is more marked
(Fig. 108.11b) than in demyelinating disorders (Fig.
108.10b). In neuronal degenerative disorders the
NAA decrease occurs before evident atrophy on MRI.
So, in this respect, MRS is more sensitive than MRI. In
neuronal degenerative disorders, Cho may be elevat-
ed related to enhanced membrane turnover. Some
elevation of mIns is sometimes seen, related to some
gliosis.
In hypomyelination, the level of Cho tends to be
low, indicative of decreased (myelin) membrane syn-
thesis and turnover (Fig. 108.12). In the case of con-
Fig. 108.12. The 1H spectrum of white matter (5 ml, STEAM,
comitant white matter gliosis, mIns and Cr are elevat-
TR/TE = 6000/20 ms, 1.5 T) of a 25-year-old male with hypo-
ed, as is seen in H-ABC (hypomyelination with atro-
myelination shows a decrease of Cho whereas all other
phy of the basal ganglia and cerebellum) (Fig. 108.13).
metabolite concentrations are within normal ranges
In the case of concomitant axonal damage and loss,
870 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

Fig. 108.13. The 1H spectrum of white matter (5 ml, STEAM, Fig. 108.15. The 1H spectrum (6 ml, STEAM, TR/TE = 6000/
TR/TE = 6000/20 ms, 1.5 T) of an 8-year-old boy with H-ABC at 20 ms, 1.5 T) of a white matter lesion of a 44-year-old female
1.5 T, shows a slight decrease of Cho, as well as an increase of with relapsing-remitting multiple sclerosis shows a decrease
Cr and especially of mIns of NAA and an increase of mIns

In several disorders, white matter pathology is


dominated by white matter rarefaction and cystic
degeneration. Examples are vanishing white matter
disease and severe variants of sulfite oxidase deficien-
cy/molybdenum cofactor deficiency. In particular
in vanishing white matter, the process of progres-
sive white matter rarefaction can be followed with
1
H-MRS. The peaks representing metabolites normal-
ly present in the spectrum gradually become lower
and disappear (Fig. 108.14). The peaks representing
lactate and glucose become visible (Fig. 108.14). Lac-
tate and glucose are normally below the level of detec-
tion in brain tissue, but they have a higher concentra-
tion in the CSF, which makes them detectable in high-
ly rarefied and cystic white matter, where most of the
white matter has been replaced by tissue water or CSF.
Acute inflammatory white matter lesions, related
to encephalitis, acute disseminated encephalomye-
litis, and multiple sclerosis, are characterized by a de-
Fig. 108.14. 1H spectra of white matter (6 ml, STEAM, TR/TE crease in NAA and variable increases in Cho, lactate,
= 6000/20 ms, 1.5 T) of patients with vanishing white matter. lipids, and mIns (Fig. 108.15). The findings are indis-
A In the spectrum of a 10-year-old girl with mild disease all tinguishable from those seen in active demyelinating
metabolites are only slightly decreased, indicating a rarefac- metabolic disorders and related to the same basic
tion of the white matter. B The spectrum of a 3-year-old girl pathological processes, including axonal damage and
with totally cystic white matter on MRI only contains lactate loss, enhanced membrane turnover, macrophage in-
(doublet at 1.33 ppm) and glucose (signals at 3.4 and 3.8 ppm), filtration, enhanced presence of membrane break-
indicating the replacement of white matter by CSF down products, and gliosis.
As MRS allows in vivo assessment of cerebral ener-
gy metabolism, it can be applied to evaluate cerebral
consequences of cellular energy failure in mitochon-
white matter NAA is decreased. Some have found an drial defects. Inborn errors involving mitochondrial
increased NAA in the white matter of patients with oxidative phosphorylation and electron transport
Pelizaeus–Merzbacher disease, which can be ex- may lead to failure to synthesize sufficient ATP, accu-
plained by the higher axonal density per tissue vol- mulation of ADP, and failure of PCr synthesis from Cr.
ume in the absence of myelin sheaths. In addition, mitochondrial dysfunction may lead to
108.4 MRS in Cerebral Disorders: Process-Specific Abnormalities 871

Fig. 108.16. The 1H spectrum of white matter (5 ml, STEAM,


TR/TE = 6000/20 ms, 1.5 T) of a 15-year-old boy with Kearns–
Sayre syndrome shows a decrease of NAA and mIns, as well as
an increase of lactate (doublet at 1.33 ppm) and glucose (res-
onances at 3.43 and 3.8 ppm)

Fig. 108.18. A The 1H spectrum of white matter (5 ml, STEAM,


TR/TE = 6000/20 ms, 1.5 T) of a 2-year-old boy with Leigh syn-
drome (Leigh/NARP mutation) is completely inconspicuous.
B A spectrum of the lesion in the basal ganglia (4 ml) in the
same patient contains a small lactate doublet. It should,
however, be noted that lactate is not necessarily detected in
patients with mitochondrial disorders, not even in areas which
are abnormal on MRI

disorders is, in most respects, indistinguishable from


that of hypoxia. In 1H-MRS in a variety of mitochon-
Fig. 108.17. A The 1H spectrum of an acute lesion in the drial encephalopathies, including Leigh syndrome,
occipital cortex (8 ml, STEAM, TR/TE = 6000/20 ms, 1.5 T) of a NARP syndrome, Kearns–Sayre syndrome (Fig.
12-year-old boy with MELAS shows a prominent doublet of 108.16), MELAS (Fig. 108.17), MERRF, and Leber
lactate and complete loss of NAA. B The spectrum of an hereditary optic atrophy, variable elevations of cere-
adjoining older lesion in the parietal cortex (6 ml) contains bral lactate are found, but lactate is not unequivocally
only some small signals from remaining lactate, Cho, and Cr elevated in all patients and/or in all brain structures
(Fig. 108.18). Cross et al. (1993) found a good correla-
tion between CSF levels of lactate: when the CSF lac-
accumulation of unoxidized reducing equivalents in tate was below 2.5 mmol/l, no lactate was seen in 1H
the form of NADH, NADPH, and lactate. Alanine may spectra of the brain, whereas in all cases with a CSF
be elevated secondary to the lactate elevation. The lactate above 4.0 mmol/l, elevated lactate was found in
pattern of metabolic disturbances in mitochondrial MRS. However, patients have been described repeat-
872 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

bral hypoxia–ischemia, acute reciprocal changes oc-


cur in the concentrations of PCr and Pi, so that the
PCr/Pi ratio falls. Only when PCr/Pi reaches a low val-
ue does the concentration of ATP also decline. Intra-
cellular pH is reduced. These changes are rapidly re-
versed when cerebral perfusion and oxygenation are
restored. Over the subsequent few days, spectral ab-
normalities develop again in spite of the absence of
any ongoing hypoxia–ischemia. Again there are
roughly reciprocal changes in PCr and Pi and, in se-
verely affected infants, a fall in ATP. However, intracel-
lular pH tends to rise rather than to fall. Later Pi in-
creases, well out of proportion to the fall in PCr, and
in severely damaged brains PCr and ATP are absent,
Fig. 108.19. The 1H spectrum of frontal cortex (5 ml, STEAM, leaving only a large residual Pi peak in the spectrum.
TR/TE = 6000/20 ms, 1.5 T) of a 7-month-old girl with pyruvate In less severely affected infants who recover, the 31P
dehydrogenase complex deficiency shows not only a promi- metabolite ratios return to normal over the course of
nent lactate doublet (1.33 ppm), but also a clear singlet at about 2 weeks, but sometimes the total phosphorus
2.36 ppm, which can be assigned to pyruvate.The spectrum is signal is reduced, indicating permanent loss of cells.
further characterized by decreased signals of NAA and mIns The explanation of the presence of two stages is that
the initial acute hypoxic–ischemic episode initiates a
series of reactions, which later cause progressive dis-
edly with absence of lactate in 1H spectra of the brain, ruption of oxidative phosphorylation in brain tissue.
while CSF lactate was high, as well as patients with el- This disruption is termed “secondary energy failure.”
evated lactate in 1H spectra of the brain while CSF lac- This course of events explains why 31P-MRS findings
tate was normal. There is a regional variability in ele- are often normal on the first day of life, to become ab-
vations of cerebral lactate, lactate being most pro- normal over the subsequent few days. The 31P-MRS
nounced in regions where MRI shows fresh structur- findings appear to have prognostic value. Of the chil-
al abnormalities, as illustrated in Fig. 108.17. The dren whose PCr/Pi falls below the 95% confidence
lactate decreases again when the lesion turns into in- limits for normal infants, two-thirds die and almost
active scar tissue. In some patients the elevated ala- all survivors have a serious neurological handicap.
nine can be visualized. Elevated cerebral glucose lev- Nearly all infants with a decrease in ATP die. With re-
els may be seen (Fig. 108.16), ascribed to a high de- spect to 1H spectroscopic findings, it has been shown
gree of nonoxidative glycolysis. In pyruvate dehydro- that lactate is elevated during the hypoxia–ischemia,
genase complex deficiency, elevated cerebral pyruvate to recover partially soon after restoration of normal
may be demonstrable in addition to the elevated lac- oxygenation of the brain and rise again during the
tate (Fig. 108.19). 31P-MRS in mitochondrial disorders first 2 days, mirroring the phenomenon of secondary
may reveal a decrease in PCr and increase in Pi in the energy failure seen in 31P-MRS. NAA begins to de-
presence of a normal pH, while calculations yield an crease several hours after the hypoxic–ischemic
increase in ADP, a decrease in phosphorylation po- episode. mIns may also increase within the first few
tential, and an increased percentage of the maximal days. Glutamate may be relatively elevated for a while
rate at which ATP is being synthesized, the latter re- after the acute hypoxic–ischemic incident, suggesting
flecting the increased demand on the mitochondria excitotoxic effects. In contrast to the normalization of
able to participate in oxidative phosphorylation. the 31P spectra, 1H spectra remain abnormal. NAA
These abnormalities are, however, again not invari- may recover partially, but does not return to normal.
ably present. The above 1H and 31P spectroscopic fea- The increased lactate may persist for weeks. 1H spec-
tures are a measure of the derangement of cerebral troscopic findings also have prognostic value. The
energy metabolism and can be used to monitor the lowest NAA levels are found in the children with the
course of disease in mitochondrial disorders, in par- poorest outcome. The presence of elevated lactate and
ticular to evaluate the effects of treatment. In addi- also of mIns carries a poor prognosis with respect to
tion, nonspecific spectroscopic changes can be pre- survival and neurological handicap.
sent in mitochondrial disorders, in particular a de- In conditions of chronic hypoxia–ischemia, such
crease in NAA, glutamate, and Cr in lesions, related to as subacute arteriosclerotic encephalopathy and
neuronal dysfunction and loss. CADASIL, damage-specific spectroscopic abnormali-
Similar spectroscopic changes related to cellular ties are found, including decreased NAA and in-
energy failure are present in hypoxic–ischemic en- creased Cho and mIns, whereas lactate may be nor-
cephalopathy of neonates. In the acute phase of cere- mal or mildly elevated.
108.5 MRS in Cerebral Disorders: Disease-Specific Abnormalities 873

In cases of hyperammonemia of any origin, in-


cluding urea cycle defects, propionic acidemia, Reye
syndrome, Wilson disease, and other causes of hepat-
ic failure, 1H spectra show changes directly related to
the high levels of ammonium. Hyperammonemia has
a great impact on the equilibrium between glutamate,
glutamine, and GABA by stimulating glutamine syn-
thesis from glutamate, resulting in an accumulation
of glutamine and a depletion of glutamate. Elevated
levels of glutamine can be visualized easily by 1H-
MRS (Fig. 108.20). Other consistent and striking
spectroscopic changes in hyperammonemia concern
a decrease in mIns and Cho (Fig. 108.20). As mIns is
an osmolyte, depletion of mIns is probably an osmot-
ic response to glutamine accumulation. In addition,
mIns is a precursor of glucuronic acid, which helps to
detoxify xenobiotics by conjugation. Depletion of
mIns could partially be the result of excessive detoxi-
fication. The decrease in Cho is probably also an os-
motic response. In cases of neuronal damage, NAA is
decreased, but in many patients with hyperammone-
mia, NAA is normal. The spectroscopic changes di-
rectly related to hyperammonemia are reversible af-
ter normalization of ammonia levels, implying that
1H-MRS is a very useful tool for monitoring cerebral

abnormalities in hyperammonemias.
Fig. 108.20. A The 1H spectrum of basal ganglia (8 ml, STEAM,
TR/TE = 6000/20 ms, 1.5 T) of a 6-year-old boy with a porto-
caval shunt and serum ammonia levels of 150–200 mmol/l 108.5 MRS in Cerebral Disorders:
shows the effects of hyperammonemia in the brain:apart from Disease-Specific Abnormalities
an increase in glutamine, the spectrum is characterized by low
signals from mIns and Cho, compensating the osmotic pres- Specific changes, related to the specific disorder, can
sure. B The resonances from glutamine dominate the spec- be found in particular in some of the inborn and ac-
trum obtained from basal ganglia and white matter (27 ml, quired errors of metabolism.
STEAM, TR/TE = 2500/20 ms, 1.5 T) in a 1-week-old neonate The relative increase in NAA in Canavan disease is
suffering from argininosuccinate lyase deficiency (urea cycle well known (Fig. 108.21). It is specifically the metabo-
defect) and serum ammonia levels of more than 1000 mmol/l. lism of NAA that is disturbed in this disorder. When
The signals of the osmolytes mIns and Cho are small in com- absolute quantitation is used and metabolites are ex-
parison to the usually much higher levels in neonates pressed in millimoles per liter of brain tissue, Cho
and Cr appear to be low and NAA can vary between
the normal range and up to a two-fold elevation. In
direct biochemical measurements the increase in the
The 1H spectrum contains resonances of several level of cerebral NAA expressed in micromoles per
metabolites that have a function as idiogenic os- milligram protein is much more pronounced. The
molytes. mIns is probably the most important among apparent discrepancy between MRS and biochemical
them, but Cho, Cr, NAA, glutamine and glutamate al- findings can be explained by the different way of ex-
so have a role in maintaining iso-osmosis. It has been pressing the concentration: per volume (MRS) or per
shown that in hypernatremia mIns, scyllo-inositol, dry weight (biochemical analyses). In Canavan dis-
Cho, Cr, and glutamine/glutamate are elevated, re- ease, the white matter has a reduced myelin content
turning to normal when the serum sodium level re- and cellularity, and the white matter is converted into
turns to normal. In hyponatremia, mIns, Cho, Cr, and a loose, vacuolated meshwork, with glia and bare
NAA are decreased and return to normal with nor- axons left as tissue elements. This serious white mat-
malization of the serum sodium level. In our patient ter rarefaction implies that a large part of the selected
with extremely elevated levels of D-arabitol and volume of interest for MRS consists of water. The low
ribitol in the brain (see below), NAA, mIns, Cr, and Cho with low Cho/Cr ratio in Canavan disease is
Cho are decreased, probably a compensatory reaction probably related to the very low membrane content of
to the osmotic effects of the accumulating polyols. the white matter. In itself the elevation of NAA is
874 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

Fig. 108.22. The 1H spectrum of white matter (6 ml, PRESS,


TR/TE = 6000/30 ms, 2 T) of a 3-year-old with an NAA deficien-
cy does not contain signals from NAA or NAAG. From Martin et
al. (2001), with permission

Fig. 108.21. A The 1H spectrum of white matter (4 ml, STEAM, Fig. 108.23. The 1H spectrum of predominantly white matter
TR/TE = 6000/20 ms, 1.5 T) of a 4-month-old boy with Canavan (27 ml, STEAM, TR/TE = 1500/270 ms, 1.5 T) of a 3-year-old boy
disease shows elevated signals of NAA and mIns, whereas Cr with maple syrup urine disease in the acute state shows next
and Cho are decreased. B Only NAA and mIns are visible in the to the doublet of lactate a doublet around 0.9 ppm,assigned to
white matter spectrum (3.5 ml, STEAM, TR/TE = 6000/20 ms, the methyl group of branched-chain amino acids (probably
1.5 T) of the same patient at the age of 1 year. leucine) or keto acids. From Felber et al. (1993), with permission

specific for Canavan disease, as in all other conditions In maple syrup urine disease, 1H-MRS during
NAA is normal or decreased, but not increased. In ad- acute metabolic decompensation shows elevated lac-
dition, mIns is increased and in some of the Canavan tate and resonances around 0.9 ppm corresponding to
patients lactate is elevated. cerebral accumulation of branched-chain amino
One patient has been described who had no de- acids and keto acids (Fig. 108.23). NAA is decreased.
tectable NAA in the brain (Martin et al. 2001), proba- With treatment and clinical recovery the 1H spectra
bly related to an as yet unidentified defect in NAA normalize.
synthesis (Fig. 108.22). Surprisingly, the clinical pic- In nonketotic hyperglycinemia, 1H spectra show an
ture was characterized by psychomotor retardation elevated glycine signal at 3.55 ppm at both short and
only, whereas MRI revealed delayed myelination at long echo times. Using a short echo time, the glycine
the age of 3 years. peak co-resonates with the mIns peak, but using echo
Salla disease and severe infantile sialic acid storage times of 135 or 270 ms, the peak at 3.55 ppm repre-
disease are characterized by accumulation of N-acetyl- sents glycine only, glycine having a much longer
neuraminic acid (sialic acid). N-acetylneuraminic acid T2 than mIns (Fig. 108.24). There is evidence that
resonates close to the methyl resonance of NAA, lead- the time course of glycine content in brain tissue,
ing to a pseudo-elevated NAA peak in the 1H white as shown by MRS, correlates more closely with the
matter spectrum. In addition, the Cho peak is abnor- clinical course than do plasma and CSF glycine val-
mally low, related to hypomyelination, and the Cr peak ues.
is elevated, probably related to white matter gliosis.
108.5 MRS in Cerebral Disorders: Disease-Specific Abnormalities 875

Fig. 108.25. The 1H spectrum of white matter (4 ml, STEAM,


TR/TE = 6000/20 ms, 2 T) of a 17-month-old girl with succinate
dehydrogenase deficiency is dominated by the singlet from
succinate at 2.40 ppm.In addition,NAA is severely reduced,and
Fig. 108.24. A The 1H spectrum of cortex and white matter lactate is elevated.From Brockmann et al.(2002),with permission
(12 ml, STEAM, TR/TE = 2500/20 ms, 1.5 T) of a 1-week-old
neonate with nonketotic hyperglycemia shows a very high
signal at 3.55 ppm. B A spectrum obtained with a long echo
time (PRESS, TR/TE = 2500/270 ms) shows that this signal is
largely due to glycine, since mIns does not contribute to the
spectrum at such long echo times

Succinate dehydrogenase deficiency is a mitochon-


drial disorder with a defect in complex II. Elevated
succinate, represented by a resonance at 2.40 ppm, can
be found in some patients (Fig. 108.25), providing
evidence for the diagnosis (Brockmann et al. 2002).
A severe reduction of the Cr peak in 1H MR spec-
tra of the brain is a central feature of the so-called cre-
atine deficiency syndromes (Fig. 108.26). The lack of
creatine in the brain can be caused by a defect in the
synthesis of creatine due to either guanidinoacetate

Fig. 108.26. A The 1H spectrum of white matter (4 ml, STEAM,


TR/TE = 6000/20 ms, 1.5 T) of a 2-year-old boy with guanidi-
noacetate methyltransferase deficiency does not contain sig-
nals from Cr. B The presence of Cr 3 months later shows the
positive effect of treatment with oral creatine supplementa-
tion in patients with a creatine synthesis defect
876 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

Fig. 108.27. The 1H spectrum of white matter (8 ml, STEAM, Fig. 108.28. The 1H spectrum of basal ganglia (3.5 ml, STEAM,
TR/TE = 3000/20 ms, 1.5 T) of a 16-year-old boy with Sjögren– TR/TE = 1600/10 ms, 1.5 T) of a 9-day-old patient with galac-
Larsson syndrome shows a prominent signal at 1.3 ppm, prob- tosemia due to a deficiency of galactose-1-phosphate uridyl-
ably representing long-chain fatty alcohols. Courtesy of Dr. transferase. Resonances at 3.67 and 3.74 ppm correspond to
M.A.A.P. Willemsen, Department of Pediatric Neurology, Uni- galactitol. From Wang et al. (2001), with permission.
versity Medical Center St Radboud,Nijmegen,The Netherlands

methyltransferase (GAMT) deficiency or glycine 1H-MRS allows monitoring of the cerebral pheny-

amidinotransferase (AGAT) deficiency, or by a defect lalanine levels in phenylketonuria (PKU). The a and b
in the transport of creatine across the blood–brain protons of phenylalanine give rise to complex multi-
barrier (X-linked creatine transporter defect). In plets of low intensity at the region between 3 and
GAMT deficiency, the spectrum may also reveal a res- 4 ppm, which cannot be distinguished from overlying
onance at 3.78 ppm, representing guanidinoacetate, strong signals of abundant metabolites under in vivo
which is not seen in the other creatine deficiency syn- conditions. In contrast, all signals of the phenyl pro-
dromes. In the defects in creatine synthesis, oral crea- tons collapse into a single peak at 7.36 ppm of suffi-
tine substitution leads to striking improvement of the cient intensity for quantitation. However, this region
symptoms of the patients and a return of the Cr peak of the spectrum, too, normally contains several over-
into the spectrum, although the Cr concentration re- lapping peaks between 6.5 and 8.5 ppm. The most
mains below normal. Oral creatine substitution does efficient way of removing these background signals is
not influence the cerebral creatine level in patients to use the spectra of normal controls for subtraction.
with a transporter defect. As the cerebral phenylalanine level in classical
In Sjögren–Larsson syndrome a prominent singlet phenylketonuria patients under free nutrition is typ-
is found at 1.3 ppm, the lipid region of the spectrum ically below 1 mmol/l, selection of a large volume of
(Fig. 108.27). The peak is seen at both short and long interest for MRS, of the order of 25 ml or larger, is re-
echo times. In addition, in most patients increased quired to obtain a sufficient signal-to-noise ratio for
resonances at 0.8–0.9 ppm are seen. These findings reliable quantitation. PRESS is preferable to STEAM
are compatible with the presence of an abnormal for better signal to noise. In addition, short echo
amount of lipids, the methylene protons resonating at times of 20 ms or less are necessary to minimize sig-
1.3 ppm and the methyl protons resonating at 0.8– nal loss due to T2-weighting of the spectrum.
0.9 ppm. The peaks are only found in the cerebral Blood–brain barrier kinetics for phenylalanine entry
white matter with the highest levels in the periven- into the brain can be studied using 1H-MRS. Brain
tricular region, where T2-weighted images show sig- phenylalanine levels do not correlate closely with
nal abnormalities, and not in the cerebral gray matter blood phenylalanine levels. The individual variation
or cerebellum (Willemsen et al. 2004). The identity of in brain-to-blood phenylalanine ratio is large, even
the lipids responsible for the peaks has not yet been among patients with similar blood levels. This indi-
elucidated, but most likely the resonances represent vidual variability may contribute to the different neu-
long-chain fatty alcohols. rological outcomes in PKU patients.
108.5 MRS in Cerebral Disorders: Disease-Specific Abnormalities 877

Fig. 108.29. 1H spectra of white matter (5 ml) of a 14-year-old to J-coupling in the long echo time spectrum. Comparison
boy with deficiency of ribose-5-phosphate isomerase leading with model spectra of D-arabitol (C, D) and ribitol (E, F),
to polyol accumulation, obtained with (A) STEAM (TR/TE = acquired with corresponding short echo times (C, E) and long
6000/20 ms) and (B) PRESS (TR/TE = 3000/135 ms) at 1.5 T. The echo times (D, F) illustrates the contribution of the two polyols
large signals between 3.6 and 3.8 ppm are partly inverted due to the spectra in A and B

Untreated galactosemia is characterized by elevat- One patient has been described with highly elevat-
ed galactitol concentrations in the brain, which disap- ed levels of the polyols ribitol and D-arabitol in the
pear with treatment. Galactitol gives rise to reso- brain (van der Knaap et al. 1999). The patient suffered
nances at 3.67 and 3.74 ppm (Fig. 108.28). from a slowly progressive encephalopathy and pe-
ripheral neuropathy. MRI of the brain revealed exten-
878 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

sive white matter abnormalities, most pronounced in cerebral involvement at the time of the transplanta-
the subcortical area, with some swelling of the abnor- tion. If applied early in the course of the disease,
mal white matter. 1H-MRS of the brain revealed high- hematopoietic stem cell transplants may prevent fur-
ly elevated peaks between 3.6 and 3.8 ppm, which ther deterioration and may even lead to improvement
could be identified as representing D-arabitol and or disappearance of cerebral lesions on MRI. The
ribitol (white matter concentrations 8.9 mmol/l and therapy is not recommended for patients with rapid-
2.9 mmol/l, respectively) (Fig. 108.29). The presence ly advancing or severe cerebral involvement, because
of these polyols could be confirmed in all body fluids in these patients the procedure may lead to accelera-
with a brain:CSF:plasma gradient of 70:40:1 for D-ara- tion of the disease or survival in a very poor neuro-
bitol and 125:55:1 for ribitol, suggesting a neurometa- logical condition or vegetative state. Hematopoietic
bolic defect. The basic defect has been demonstrated stem cell transplantation is not recommended for
to be a deficiency of ribose 5-phosphate isomerase, a asymptomatic patients, because of the more than
defect in the pentose phosphate pathway (Huck et al. 50% chance that these patients will not develop the
2004). serious cerebral form of the disease, the absence of a
In diabetes mellitus, 1H-MRS is able to detect the way of predicting whether they will, and the high
brain levels of glucose (resonances at 3.43 and mortality associated with hematopoietic stem cell
3.80 ppm) and the ketone bodies acetone (resonance transplantation. Therefore, monitoring of the disease
at 2.22 ppm), acetoacetate (resonance at 2.26 ppm) on MRI, MRS, and neuropsychological tests from the
and b-hydroxybutyrate (doublet at 1.2 ppm), allowing presymptomatic stage onwards is extremely impor-
monitoring of glucose metabolism at a brain level. In tant. As soon as there is evidence of beginning cere-
cystic leukoencephalopathies, 1H-MRS of cystic areas bral disease, hematopoietic stem cell transplantation
reveals the resonances of glucose and lactate, present should be attempted. MRI provides important infor-
in the CSF. An important example is vanishing white mation for monitoring purposes: the presence of
matter disease, in which the white matter is typically lesions, progression of lesion size, and presence or
rarefied and cystic. 1H MR spectra of the white matter absence of contrast enhancement. The presence of
reveal that the peaks representing metabolites nor- contrast enhancement is predictive of progressive
mally present in the spectrum are very low or absent disease. MRS also provides important information. In
and that peaks of lactate and glucose are visible. an area of active demyelination, not only a decrease in
NAA is found, but also an increase in Cho, Cr, mIns,
and lactate, related to enhanced membrane turnover,
108.6 Monitoring increased cellular density, inflammation, macrophage
infiltration, and gliosis. In the inner area of inactive,
Both the process-specific and disease-specific spec- burnt-out demyelination, NAA is very low, but Cho
troscopic abnormalities provide excellent possibili- and lactate are no longer elevated. The spectrum in
ties for disease monitoring. With respect to monitor- the burnt-out area has a pattern reflecting gliosis
ing of disease-specific abnormalities, a good example dominated by Cr and mIns. The spectroscopic find-
is found in the creatine synthesis defects. Oral crea- ings in the area just outside the MRI-visible lesion are
tine supplementation leads to clinical improvement. highly predictive of disease progression. Elevated lev-
Unfortunately, assessment of metabolites in body els of Cho, decreased NAA/Cr ratios, and elevated
fluids is of little help in monitoring the treatment of mIns in this area are highly predictive of progressive
the disease. 1H-MRS gives the most direct measure of disease, whereas normal Cho and mIns levels and
the efficacy of the treatment in the cerebral creatine NAA/Cr ratios are predictive of stable disease. Thus,
level. MRS is more sensitive than MRI in detecting incipi-
Process-specific spectroscopic findings can also be ent demyelination. It is, however, important to realize
very useful in monitoring disease.Among the inherit- that even in normal-appearing white matter remote
ed white matter disorders, an example of the impor- from the lesion and in the white matter of patients in
tant role of MRS in monitoring disease is found in X- whom the disease has not yet started, NAA is often
linked adrenoleukodystrophy (XALD). Central issues below normal in XALD males (Fig. 108.30) In addi-
in the treatment of males carrying the biochemical tion, MRS has a predictive value for the outcome of
defect of XALD are the questions of in which patient patients treated with a hematopoietic stem cell trans-
and when to perform hematopoietic stem cell trans- plantation. A good outcome is highly correlated with
plantation. This form of treatment is only applied for the NAA concentration in the affected white matter,
the rapidly progressive cerebral disease, which usual- and thus the preservation of neuronal tissue, before
ly has its onset in childhood, and not for the later-on- the transplantation.
set variant adrenomyeloneuropathy (AMN). The out- MRS is also a very important tool for purposes of
come of hematopoietic stem cell transplantation in monitoring and prediction of the course of the dis-
cerebral XALD is highly dependent on the degree of ease in acute brain injury, including diffuse axonal in-
108.6 Monitoring 879

Fig. 108.30. Average 1H spectra of controls and patients with matter in patients without neurological symptoms (n = 21) are
X-linked adrenoleukodystrophy, obtained with STEAM (TR/TE characterized by decreased NAA and increased concentra-
= 6000/20 ms) at 2.0 T. In comparison to (A) white matter spec- tions of Cho, Cr, mIns, and on average an increase of lactate.
tra of an age-matched control group (n = 19), the spectra of (B) (D) Ongoing demyelination in patients with advanced cere-
normal-appearing white matter of patients without neurolog- bral X-linked adrenoleukodystrophy (n = 9) leads to severe
ical symptoms (n = 47) show on average slightly lower NAA neuronal loss (almost complete absence of NAA and decrease
and higher Cho. (C) Spectra of actively demyelinating white of Cr)

jury after head trauma, near-drowning, and menin- slices allows monitoring of spectroscopic changes of
goencephalitis. There is a good correlation between large areas of the brain with a spatial resolution that
early NAA measurements and outcome: the lower the is adequate for the purpose. Single voxel spectroscopy
NAA levels, the poorer the outcome. The presence of has the advantage of better spectral quality and use of
elevated lactate and lipid peaks adds to the prediction shorter echo times. The spectroscopic abnormalities
of poor outcome. Elevated Cho may be found, but this found depend on the stage of the lesion. Cho is
finding does not seem to have predictive value. This already increased in a prelesional area up to 12
information can be used in making important deci- months before the lesion becomes visible on conven-
sions in the continuation of treatment of such pa- tional MRI. Acute lesions, as identified by gadolinium
tients. enhancement on MRI, show a reduction of NAA, and
Multiple sclerosis (MS) is probably the most im- an increase in Cho, mIns, lipids, and, often, lactate.
portant indication for monitoring by MRS. Therapeu- The earliest spectroscopic changes consist of in-
tic trials require objective and preferably quantitative creased Cho and lactate, whereas a decrease in NAA
measures of changes in disease. Spectroscopic imag- occurs a few days later. Lactate disappears in a matter
ing with acquisition of data in multiple transverse of weeks. Enhanced Cho decreases within a couple of
880 Chapter 108 Magnetic Resonance Spectroscopy: Basic Principles and Application in White Matter Disorders

tides and neutral fat, reflected in elevated protein and


lipid signals in MRS. From early on axons are dam-
aged, with decreasing NAA. The decrease in NAA re-
flects not only axonal transections in acute lesions,
but also a significant component of metabolic dys-
function of injured axons. The first component con-
cerns the permanent damage, the second may be
transient. Large destructive lesions can additionally
show transient decreases in Cr during the hyperacute
phase, probably reflecting metabolic dysfunction.
The decrease in PDE reflects myelin loss. Chronic and
inactive lesions are hypocellular, demyelinated, and
gliotic. Inflammation is no longer present. Chronic
lesions that are very hypointense on T1-weighted
images – the so-called “black holes” – have the lowest
NAA and Cr, correlating with the profound tissue
destruction, axonal loss, and hypocellularity of these
lesions (Fig. 108.31). An additional important finding
has been that normal-appearing white matter in MRI
is not completely normal in MRS (Fig. 109.31). This is
true for all MS variants, although observations vary.A
decreased or normal NAA, normal or increased Cr,
and increased mIns in 1H-MRS and a decrease in
total 31P peak integrals in 31P-MRS are seen, as well as
a reduction in the broad component of the 31P spec-
trum, consistent with a low-grade demyelination,
gliosis, and neuronal damage too subtle to be detect-
ed by MRI. In the cerebral cortex of MS patients, too,
Fig. 108.31. 1H spectra (8 ml, TR/TE = 2500/135 ms, 1.5 T) of decreased NAA and Cr have been found, ascribed to
two patients with secondary progressive MS. A The normal- neuronal metabolic dysfunction, damage, and loss.
appearing white matter of a 45-year-old male with MS is com- Several studies have indicated a correlation between
parable to the white matter of age-matched controls, apart the extent of decrease in NAA and disability, which is
from a slight decrease of NAA, whereas (B) a lesion that stronger than the correlation between T2 lesion load
is hypointense on T1-weighted images (a “black hole”) of a and disability. The total brain content of NAA has
43-year-old male is characterized by a severe decrease of NAA, been used as an indicator of total neuronal and axon-
accompanied by a decrease of Cr al damage, which is the irreparable part of the pathol-
ogy in MS.

months and eventually returns to normal. A partial 108.7 Conclusion


recovery of NAA is often seen over a period of months
after acute onset of the lesion – probably the result of Reviewing the information provided by MRS about
a combination of actual recovery of axonal integrity brain metabolism in normal cerebral maturation, in
and tissue contraction due to disappearance of edema disturbed maturation, and in disease conditions,
and removal of disintegrated tissue components. The shows that MRS has proven itself to be a tool of signif-
lipid resonances remain elevated for several months. icant usefulness in clinical medicine and of great
The rise in mIns may be temporary or permanent. potential for research purposes. For clinical applica-
31P-MRS reveals decreased PDE in acute lesions. Spec-
tions, the greatest power of MRS is found in monitor-
troscopic findings reflect the changes at the histolog- ing disease processes, in particular with a view to
ical level. Correlation with pathological findings in evaluating therapeutic interventions and establishing
biopsy specimens confirms an association between prognosis. In particular in the early stages of disease
acute inflammation, hypercellularity due to a mixed processes, MRS often appears to be more sensitive
macrophage–astrocyte response, and demyelination than MRI. For research applications, the greatest
with increased levels of Cho and lactate. In acute le- potential of MRS may be in contributing to the eluci-
sions myelin sheaths show signs of disintegration and dation of pathophysiological processes in cerebral
breakdown accompanied by liberation of polypep- disorders.
Chapter 109

Pattern Recognition in White Matter Disorders

109.1 Introduction trated in Fig. 109.2. In the early phase of the disease,
the MRI pattern is diagnostic. In the end phase al-
MRI is highly sensitive in the detection of white mat- most all cerebral white matter is affected and distin-
ter lesions. A close association has been demonstrat- guishing characteristics have been lost. A second
ed between the occurrence of white matter abnormal- problem is illustrated in Fig. 109.3, where the pattern,
ities observed with MRI and those found at autopsy. It for reasons unknown, is the inverse of the pattern
has been generally assumed that the specificity of commonly observed in this disease. The computer
MRI is much lower than its sensitivity. The specificity program should allow for these exceptions to the rule
of MRI, however, depends not only on the potential when they are known to occur. The third problem is
and limitations of the method, but also on the capa- illustrated in Fig. 109.4. Sometimes superficial read-
bilities of the person interpreting the MR images ing of the images strongly suggests a certain diagno-
(Fig. 109.1). Hence, optimization of the diagnostic sis, whereas on closer examination only some but not
specificity of MRI in white matter disorders is all of the main MRI features of a disease are present.
achieved by optimizing the quality of both the imag- And sometimes the resemblance between patterns is
ing and its interpretation. so strong that it is difficult or impossible to discrimi-
Aids to the perceptual and decision processes nate disorders on the basis of the MRI pattern recog-
can be constructed to support the interpretative nition program alone.
process. One such aid is systematic and detailed The benefits of MRI pattern recognition are di-
image analysis. A checklist or score list, which verse. First of all, it facilitates the diagnostic process
prompts the image reader to assess and record a scale and reduces the list of necessary laboratory tests.
value for each feature, is helpful in this respect. A sec- Sometimes the activity of only one enzyme has to be
ond aid is a computer program that integrates these assessed or only one gene has to be analyzed after
scale values into a pattern, compares the pattern ob- reading the MRI. In this way, MRI pattern recognition
tained with the known patterns in a database, and reduces the burden for patients and families and is a
then reaches a differential diagnosis. The reader of money-saving strategy. Secondly, the recognition of
the images can use the computer estimates of the like- patterns provides important scientific information.
lihood of various disorders as a guide in the diagnos- Selective vulnerability of brain structures for differ-
tic process. ent noxious influences underlies the development of
It is important is to realize that MRI pattern recog- different patterns of involvement of brain structures
nition has its limitations. In the first place, MRI pat- in different disorders. So far, our understanding of the
terns have characteristic features only during a cer- reasons for the selective vulnerability has remained
tain phase of progressive disorders. This is well illus- highly limited.

Fig. 109.1. T1- and T2-weighted images


of a 10-month-old male with glutaric
aciduria type I.The images show diffuse
bilateral subdural hygromas and fronto-
temporal hypoplasia. Myelination is
severely retarded. Note that the arach-
noid and subdural spaces can be clearly
distinguished.The presence of bilateral
subdural hygromas could be wrongly
interpreted as evidence of child batter-
ing. Knowledge of the specific features
of glutaric aciduria type I with presence
of frontotemporal opercular hypoplasia
precludes this mistake. Courtesy of
Osaka et al. (1993), with permission
882 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.2. The upper row of T1-weight-


ed (IR) images in this 3-year-old boy
show the pattern that is typical for the
childhood cerebral form of X-linked
adrenoleukodystrophy: peritrigonal
and occipital leukoencephalopathy,
sparing the U fibers, involving the
geniculate bodies and the splenium
of the corpus callosum, with typical
involvement of corticospinal tracts in
pons and mesencephalon.The lower
row of one T1- and one T2-weighted
image are of the same child, 3 years
later. No pattern is recognizable as all
white matter structures are involved
and all characteristic features of the
disease are lost

Fig. 109.3. T2-weighted images in a


6-year-old boy show bilateral, sym-
metrical involvement of the frontal
white matter and the frontospinal
tracts in the anterior limb of the inter-
nal capsule, which can be followed in
the brain stem.The spread of the
disease is evidently in a ventrodorsal
direction.The diagnosis is X-linked
adrenoleukodystrophy with reversed
pattern. Courtesy of Dr. P. Hoogland
and Dr. W.F.M. Arts, Juliana Children’s
Hospital, The Hague, The Netherlands

109.2 Noncomputerized reader of the image can regard large parts of an MR


Pattern Recognition system and the imaging process as a “black box,” he or
she needs to have a more than general knowledge of
Pattern recognition in the daily practice of medical the imaging process, the parameters involved (repeti-
imaging involves three levels of action which are nec- tion time, echo time, inversion time, number of exci-
essary to permit optimal interpretation of the image. tations, slice thickness, gradient strength and perfor-
These levels can be described as image formation, im- mance, diffusion sensitivity), the influence of para-
age analysis, and image interpretation. meter settings on the image, the possible artifacts,
The first level is the technological level and has to and the ways improving quality when special answers
do with the formation of the image. Although the are required.
109.2 Noncomputerized Pattern Recognition 883

Fig. 109.4. A 23-year-old man had experienced moderate phy.There was no laboratory evidence for a peroxisomal disor-
psychomotor retardation from birth onwards and progressive der. Although the changes in the peritrigonal area and occipi-
disturbances of gait for the last 2 years.The T2-weighted sagit- tal lobe could be the remnants of periventricular leukomala-
tal and transverse images show bilateral peritrigonal and oc- cia, the involvement of the internal capsule and the involve-
cipital involvement of the white matter; the splenium of the ment of the splenium of the corpus callosum make this diag-
corpus callosum and the posterior limb of the internal capsule nosis highly improbable. Despite the highly characteristic
are also affected. There is no enhancement after gadolinium image and perfect symmetry this remains an unsolved case,
injection, which argues against X-linked adrenoleukodystro- which needs to be followed up

The second level is the level of image analysis, in still other sources, in an assessment that aims at an-
which the structural elements of the image are ana- swering the clinical question.
lyzed, weighed as to their normality or abnormality, The first level, image formation, will not be dis-
and described as such. This is an important step in the cussed in this chapter. Some details of special tech-
process of teaching and learning, because the analysis niques are discussed in Chaps. 106, 107, and 108.
of structural elements depends very much on experi- The second level is the analytical level, which in-
ence, knowledge of anatomy, knowledge of normal cludes a systematic analysis and classification of
brain maturation, and knowledge of pathology. structural elements of the image (see Tables 109.1–
The first and second levels lay the foundation for 109.3). Many separate gray and white matter struc-
the third, the interpretation of the image. In this tures should be scored as normal or abnormal.Which
process the image as such is transcended. Interpret- structural elements of an image need to be evaluated
ing the image requires a combination of a certain un- has been determined by experience of their discrimi-
derstanding of image formation, analysis of the struc- nating value. For example, if one were not aware from
tural elements, experience, and knowledge of disease previous experience and histopathological studies
entities, histopathology, pathophysiology, biochem- that the arcuate fibers are spared in many white mat-
istry, and toxicology, and possibly knowledge from ter disorders, but that, for example, L-2-hydroxyglu-
884 Chapter 109 Pattern Recognition in White Matter Disorders

Table 109.1. List of structural elements to be analyzed for MRI pattern recognition
Cerebral cortex Occipital/frontal/parietal/temporal
Arcuate fibers Occipital/frontal/parietal/temporal
Lobar (deep) white matter Occipital/frontal/parietal/temporal
Periventricular white matter Occipital/frontal/parietal/temporal
Internal capsule Anterior limb/posterior limb
External capsule + extreme capsule
Caudate nucleus
Putamen
Globus pallidus
Thalamus
Corpus callosum Rostrum/genu/corpus/splenium
Cerebellar cortex
Cerebellar white matter
Hilus of dentate nucleus
Cerebellar pedunculi
Dentate nucleus
Midbrain Central part/peripheral rim/tectum and tegmentum/specific tracts
Pons Central part/peripheral rim/tegmentum/specific tracts
Medulla Dorsal part/specific tracts

Scoring: no abnormality/slight to mild abnormality/severe abnormality

Table 109.2. List of general characteristics to be analyzed for taric aciduria starts in the arcuate fibers, it would be
MRI pattern recognition senseless to make involvement of the arcuate fibers a
Predominance Frontal/occipital/parietal/temporal point of discrimination in the diagnostic process. The
Periventricular/lobar (deep)/ structural elements that need to be scored may also
arcuate fibers have to be adapted to the diseases under investiga-
Supratentorial/posterior fossa tion. Some diseases affect very special brain stem
structures, as for example in leukoencephalopathy
Symmetry Perfectly symmetrical/slightly with brain stem and spinal cord abnormalities and el-
asymmetrical/asymmetrical
evated lactate (LBSL). In this disease brain stem
Extension Small isolated lesions/large isolated structures need to be scored in detail, which is not
lesions/irregularly confluent necessary for most other disorders. Then there are so-
lesions/highly confluent
lesions/combination of these called “general characteristics” that need to be scored,
including symmetry, confluent versus isolated/multi-
Appearance Swelling/atrophy/rarefaction/cystic focal involvement of the white matter, predominant
degeneration
localization of the abnormalities, white matter
Signal intensity Slightly to mildly abnormal/severely swelling, atrophy, rarefaction, cystic degeneration,
abnormal/mixed and the evolution over time. Finally, analysis of “extra
Homogeneity Homogeneous/inhomogeneous/ characteristics” is important, including contrast en-
two zones hancement, calcium deposits, hemorrhages, and the
Demarcation Sharp/vague/mixed presence of localized cysts.
Structural elements that have been identified by us
as having in general the highest discriminating value
are: symmetry versus asymmetry, confluent involve-
Table 109.3. List of extra characteristics to be analyzed for ment of the white matter versus multifocal isolated le-
MRI pattern recognition sions, the predominant localization of lesions in the
Calcium deposition Absent/present
brain, the additional involvement of gray matter
structures, contrast enhancement, and the presence
Hemorrhage Absent/present of calcium deposits.
Contrast enhancement Absent/present In many cases, symmetry is a striking feature of
inherited white matter disorders and toxic en-
Ventricular enlargement No/slight to mild/severe
cephalopathies, although not without exceptions,
Enlargement of pericerebral No/slight to mild/severe whereas asymmetry is most often seen in acquired
subarachnoid spaces white matter disorders, particularly inflammatory
Cerebellar atrophy No/slight to mild/severe disorders and infections. The appearance of the le-
sions is important: isolated, or confluent, or both. In
Myelination Normal/ delayed/no
or hardly any myelin
most inherited white matter disorders, in toxic en-
cephalopathies, and in diffuse white matter injury af-
109.2 Noncomputerized Pattern Recognition 885

ter irradiation and chemotherapy, the lesions are con- Table 109.4. Specificity of MRI patterns
fluent; as a rule no isolated lesions are seen. Multifo- Diagnostic
cal and isolated lesions are more commonly seen in Cerebral form of X-linked adrenoleukodystrophy
acquired conditions. In multiple sclerosis a mixture of
Zellweger syndrome
isolated and confluent lesions is the rule. The same is
Cerebrotendinous xanthomatosis
true of Binswanger disease.A global survey of the dis- (if fat deposits are present)
tribution of the lesion may give an indication of the Periventricular leukomalacia
type of disorder we are dealing with. Sparing of the U Megalencephalic leukoencephalopathy
fibers is seen in many inherited white matter disor- with subcortical cysts
ders, in vascular disorders, and in subacute HIV en- Canavan disease
cephalitis. Several organic and amino acidopathies, Maple syrup urine disease
contrariwise, preferentially involve the U fibers. L-2-Hydroxyglutaric aciduria
Extra characteristics such as calcifications occur in Alexander disease
Cockayne syndrome, in some patients with X-linked Some toxic encephalopathies
adrenoleukodystrophy, in malignant phenylketon- Kearns–Sayre syndrome
uria, in Aicardi–Goutières syndrome, in some pa- Multiple sclerosis, when McDonald criteria are met
tients with systemic lupus erythematosus, in children
with AIDS encephalopathy, in congenital cytomegalo- Highly suggestive
virus infection, in some patients with a mitochondri- Metachromatic leukodystrophy
al leukoencephalopathy, and in some children after Globoid cell leukodystrophy
cranial irradiation and chemotherapy. In some white Mucopolysaccharidoses
matter disorders enhancement with contrast is seen Leigh syndrome
in part of the lesion, often in the active border. This MELAS
observation can be very helpful in the diagnostic Cockayne syndrome
process. In X-linked adrenoleukodystrophy the en- Phenylketonuria
hancing border separates the area of complete de- Glutaric aciduria type I
myelination from the area with ongoing demyelina- Acute disseminated encephalomyelitis
tion. Enhancement is also a prominent feature of mul- Wilson disease
tiple sclerosis and Alexander disease. Central pontine myelinolysis
Perhaps other criteria could also be used in the
recognition of patterns of white matter disorders on Suggestive
the images, such as measurements of the absolute sig- Lowe syndrome
nal intensities and measurements of T1 and T2, ADC, Pelizaeus–Merzbacher disease
fractional anisotropy, and MTR. For instance, in sub- Multiple sclerosis
acute HIV encephalitis, the signal intensity of the Subacute HIV encephalitis
white matter lesion, at least in the beginning of the Binswanger disease
disease, is not as high on T2-weighted images as it is in Wallerian degeneration
most other white matter disorders. Neuropathologi- Toxic encephalopathies
cal examinations confirm that demyelination in sub-
acute HIV encephalitis is only partial and mild. On Possible
the MR images the involved white matter has a coarse, Multiple sclerosis, less advanced cases
granular texture, which corresponds well with the Extrapontine myelinolysis
histological finding of numerous small foci of more
Atypical
complete demyelination.
Unusual appearance with established diagnosis,
In our definition, specificity exists in degrees. The e.g., asymmetric cerebral involvement in X-linked
pattern emerging from the image can be expressed as adrenoleukodystrophy; Alexander disease starting
being diagnostic (pathognomonic), highly sugges- in the cerebellum
tive, suggestive, possible, atypical, and impossible. It is Impossible
only in the first category that the MRI pattern is
The pattern excludes the suspected diagnosis
pathognomonic for one specific disorder. In the other
categories, clinical and laboratory evidence is neces-
sary to complete the diagnosis. The lists given should
be considered as examples; they are by no means the diagnostic process in these disorders. Very often,
complete (Table 109.4). by adding clinical information or laboratory data, a
Quite a few white matter disorders can be listed higher diagnostic category can be achieved.Addition-
under the categories “diagnostic” or “highly sugges- al facts that will help to reach a diagnosis are many
tive,” implying that MRI makes a firm contribution to and include facts in the clinical history and findings
886 Chapter 109 Pattern Recognition in White Matter Disorders

at physical examination. The latter may include ab- vere/mild/not involved) indicating the severity of
normalities of eyes, face, hair, internal organs, skin, involvement of the structures. Also, disease groups
skeleton, and peripheral nerves. Examples are facial had to be clustered to some extent. This would seem
dysmorphia in Cockayne syndrome and mucopoly- to be sensible, because it is practically impossible to
saccharidoses, ectopic lenses in hyperhomocystein- differentiate all individual conditions on MR criteria
uria, and multiple organ involvement in Zellweger alone. Another limitation of such a computer pro-
syndrome and in mitochondrial disorders. gram is that its quality is highly dependent on the
quantity and quality of the data it contains. To im-
prove the quality of the program in this respect a mul-
109.3 Computer-Assisted ticenter database with well-defined criteria for inclu-
Pattern Recognition sion of cases would be extremely helpful in narrowing
the confidence intervals in rare disorders.
With the increasing complexity of diagnostic pro- Of course, such a computer system cannot compete
cesses and with the increase in number of recognized, with the flexibility and speed of the human brain in
rare disorders, the need to implement expert systems pattern recognition. Computer systems are, therefore,
is felt. To develop these, it is essential to collect data not to be regarded as competing, but as complemen-
systematically and quantitatively and to set up large tary: a support to the experienced and a learning tool
data banks. We created a database that could be used for the inexperienced.
to develop a computer-assisted pattern recognition
program in white matter disorders, with the excep-
tion of ischemic white matter disease related to arte- 109.4 Practical Application
riosclerosis (van der Knaap et al. 1991). Over a certain of Pattern Recognition
period of time, the MR images of all patients under
the age of 50 years or, when risk factors for vascular Usually MR images are interpreted without the help
disease were present, under the age of 40 years, with of a computer program. However, in daily practice,
lesions exclusively or predominantly involving the our approach should also be systematic and we
white matter, were scored with the help of a detailed should progress logically through the diagnostic
scoring list (see Tables 109.1–109.3). The total study process. We will analyze the logical steps involved in
population numbered 1483. The diagnosis in the pa- systematic reading of the MR image, well aware that
tients was known. The patients were grouped into di- several steps may occur synchronously and that the
agnostic categories according to the classification we sequence is variable.
proposed, which stresses etiological and histological The first step identifies the nature of the cerebral
similarities between disorders of the same category. abnormalities. In the global analysis of MR images of
The data obtained were analyzed. The frequency the brain, the first consideration relates to the struc-
of occurrence of image abnormalities and features tures involved: gray matter, white matter, or both.
per disease category or subcategory were counted Next, the examiner tries to identify the nature of the
and the results presented as histograms (examples disorder with which he or she is confronted. If gray
in Figs. 109.5–109.8). The outlines of the histograms – matter is involved, are there signs of a congenital
the “skylines” – can be considered to be characteristic anomaly with ectopic gray matter? Is there gray mat-
of the disease or disease category. ter atrophy or are there parenchymatous lesions? If
In addition, a computer program was developed to there is a white matter disorder, of what nature is it? Is
estimate post-MRI probabilities of possible diag- it demyelination, delayed myelination, or hypomyeli-
noses in new patients with white matter abnormali- nation? Is there white matter swelling? Is there white
ties. The computer program was based on Bayes’ the- matter rarefaction or cystic degeneration? Are there
orem. Prevalences of the different disease categories white matter cysts? Is there a loss of white matter vol-
and subcategories and frequencies of image abnor- ume or gliotic retraction?
malities per disease were estimated from the data of The second step considers the symmetry or asym-
this study. When the imaging findings of a new pa- metry of the abnormalities. Symmetrical white mat-
tient were entered, the computer program would pro- ter involvement occurs in most of the inherited white
vide a differential diagnosis. For each possible diag- matter disorders, toxic encephalopathies, and some
nosis the positive predictive value and a two-sided of the other acquired white matter disorders. Many
95% confidence interval could be computed. of the acquired disorders lead to asymmetrical le-
Of course, there are several limitations to the prac- sions, with some exceptions. A tendency towards
tical use of a computer-assisted diagnostic system. symmetry is present in periventricular leukomalacia,
The large amounts of data involved make clustering subcortical arteriosclerotic encephalopathy, subacute
necessary; for example, a two-point scale (yes/no) HIV encephalitis, and congenital CMV encephalopa-
had to be used instead of a three-point scale (se- thy.
109.4 Practical Application of Pattern Recognition 887

Fig. 109.5. Histogram of the frequency of involvement of the tion; s, small isolated lesions; l, large isolated lesions; i, irregular-
structural elements in the cerebral form of X-linked adreno- ly confluent lesions; c, highly confluent lesions; sd, sharp de-
leukodystrophy. co, cortex; uf, arcuate fibers; lo, lobar (deep) marcation; md, mixed demarcation; vd, vague demarcation;
white matter; pv, periventricular white matter; f, frontal; p, pari- has, highly abnormal signal intensity; mas, mildly abnormal
etal; t, temporal; o, occipital; cc, corpus callosum; ci, internal signal intensity; ms, mixed signal intensity; h, homogeneous
capsule; ce, external and extreme capsules; bn, basal nuclei signal intensity; ih, inhomogeneous signal intensity; 2z, two
(globus pallidus, putamen, caudate nucleus); th, thalamus; bs, zones discernible in the lesion; ca, calcification; nmy, normal
brain stem; cw, cerebellar white matter; cg, cerebellar cortical myelination; dmy, delayed myelination; nomy, no or hardly any
gray matter; cp, middle cerebellar pedunculi; nd, dentate nu- myelin present; dv, deformation of the ventricular system
cleus; sy, symmetrical distribution; asy, asymmetrical distribu-

The third step defines the aspect of the lesions. Are are usually asymmetrical, partly confluent, but main-
they confluent, isolated, or both; diffuse or multiple? ly isolated, and located in rather specific areas of the
In the inherited white matter disorders lesions are of- brain.
ten confluent and diffuse. Multiple sclerosis, of The fourth step establishes the predominant loca-
course, is the prototype of a disease with lesions that tion of the lesions. Confluent, symmetrical lesions in
888 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.6. Histogram of the frequency of involvement of the structural elements in Pelizaeus–Merzbacher disease. Abbrevia-
tions, see Fig. 109.5

the periventricular area narrow the number of diag- The fifth step defines the pattern of spread of the
nostic possibilities considerably. Sparing of the arcu- disease. Adding the pattern of spread to the analysis
ate fibers is characteristic of the sphingolipidoses, further helps to distinguish certain entities. The cere-
whereas the reverse is true of some of the organic and bral form of X-linked adrenoleukodystrophy usually
amino acidopathies, which start in the U fibers. Le- spreads in a dorsoventral direction; the direction of
sions confined to the cerebellar white matter are rare. spread of Alexander disease is ventrodorsal, that of
Cerebrotendinous xanthomatosis, adrenomyeloneu- Canavan disease centripetal, and that of metachro-
ropathy, Refsum disease, and Langerhans cell histio- matic leukodystrophy centrifugal.
cytosis can be examples. Multiple sclerosis has a pref- The sixth step weighs the contribution of gray
erence for the upper edges of the lateral ventricles and matter involvement relative to white matter involve-
the centrum semiovale. The cerebral form of X-linked ment. Is the gray matter involvement a major or a
adrenoleukodystrophy usually presents in the occipi- minor part of the disease? Are cortical or central
tal lobes. Herpes simplex virus infections favor the gray matter structures involved? Deep gray matter
frontal and temporal lobes. is often involved in mitochondrial disorders, in Bin-
109.5 Pattern Recognition in Unclassified Leukoencephalopathies 889

Fig. 109.7. Histogram of the frequency of involvement of the structural elements in multiple sclerosis. Abbreviations, see
Fig. 109.5

swanger disease, in vasculitides, in glutaric aciduria progressive multifocal leukoencephalitis enhance-


type I and many other organic acidurias, in toxic ment occurs occasionally.
encephalopathies such as carbon monoxide intoxi-
cation, cyanide intoxication, amphetamine intoxica-
tion, and in post-hypoxic–ischemic conditions. In- 109.5 Pattern Recognition in Unclassified
volvement of the basal ganglia is rare in multiple Leukoencephalopathies
sclerosis.
The seventh step evaluates whether the lesions en- MRI pattern recognition has not changed the fact that
hance after contrast injection. Sometimes enhance- in about 50% of the children with significant white
ment of the lesions leads to a characteristic pattern, as matter abnormalities on MRI, no specific diagnosis
in cerebral X-linked adrenoleukodystrophy and can be established despite an extensive laboratory
Alexander disease. Active lesions in multiple sclerosis work-up; the disease remains unclassified. The per-
enhance. In acute disseminated encephalomyelitis centage is lower in adults. Pattern recognition works
not all lesions enhance or no lesions enhance at all; in well in patients with a diagnosis that can be con-
890 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.8. Histogram of the frequency of involvement of the structural elements in central pontine myelinolysis. Abbreviations,
see Fig. 109.5

firmed by laboratory tests because patients with the contributed to the identification of several hitherto
same or a similar disorder have been shown to share unidentified white matter disorders: megalencephal-
the MRI pattern. We decided to apply MRI pattern ic leukoencephalopathy with subcortical cysts (MLC);
recognition to the large group of patients with an vanishing white matter disease (VWM); hypomyeli-
unclassified leukoencephalopathy, assuming that nation with atrophy of the basal ganglia and cerebel-
patients with the same disease entity would again lum (HABC); and leukoencephalopathy with brain
share MRI characteristics. Application of pattern stem and spinal cord involvement and elevated white
recognition to the MR images of large numbers of pa- matter lactate (LBSL).
tients with an unclassified leukoencephalopathy led The general MRI-oriented approach to unclassi-
to the identification of several distinct patterns seen fied leukoencephalopathies has been described else-
in multiple patients. MRI pattern recognition has where (van der Knaap et al. 1999). Seven major cate-
109.7 Examples 891

gories can be identified using simple and robust MRI 109.6 Typical and Atypical MRI Patterns
criteria, mainly based on the predominant location of
the white matter abnormalities. Our aim was to divide The steps leading to the identification of so far un-
the unclassified leukoencephalopathies into more classified white matter disorders are based upon a
workable groups. These categories are distinct, as has strict adherence to MRI criteria. The resulting homo-
been validated statistically. geneous group allows further exploration of the bio-
Group A is characterized by severe hypomyelina- chemical or genetic background. Once the biochemi-
tion. This is the largest single category among the un- cal or genetic background is established, less typical
classified leukoencephalopathies. In all patients cur- cases can be analyzed, so that information can be ob-
rently known disorders leading to hypomyelination tained about the phenotypic variations of the disease,
were ruled out, including Pelizaeus–Merzbacher dis- including variations in MRI patterns. Important ex-
ease, Salla disease, and DNA repair disorders. amples can be found in the chapter on Alexander dis-
Group B is characterized by global involvement of ease (Chap. 57) and the chapter on vanishing white
the cerebral white matter. In this group two separate matter (Chap. 65).
disease entities were identified: B1, megalencephalic
leukoencephalopathy with subcortical cysts, and B2,
leukoencephalopathy with vanishing white matter. 109.7 Examples
These diseases, initially identified by their MRI pat-
tern as separate entities, are now also confirmed to be The following examples serve as illustrations of the
genetic entities. described approach.
Group C is characterized by extensive, predomi-
nantly frontal white matter abnormalities, relatively
sparing of the occipital lobes, with basal ganglia ab- 109.7.1 Example I
normalities and also in many cases brain stem abnor-
malities. Most of the patients in this category had A 4-year-old girl presented with bilaterally dimin-
Alexander disease, a diagnosis that can now also be ished vision. The initial MRI showed optic neuritis,
genetically confirmed. more severe on the right side. There was one lesion in
Group D is characterized by predominantly peri- the cerebral parenchyma on the right side around and
ventricular white matter abnormalities. This is a het- under the anterior commissure. She responded favor-
erogeneous group of disorders, often genetic, with a ably to a short course of methylprednisolone, but
progressive clinical disease course. within a week after discontinuation she returned with
Group E is characterized by predominant involve- severe symptoms of myelopathy in addition to de-
ment of the deep (lobar) white matter. Deep or lobar creased vision. Figure 109.9 shows the MRI findings
white matter is located in between the periventricular at that time. The lower row of FLAIR images shows
white matter and the U fibers. In most patients the involvement of the caudate nucleus and putamen
white matter abnormalities consisted of multifocal on the right side and bilateral lesions in the pulvinar.
isolated lesions. In most patients the encephalopathy The lesion around the right anterior commissure is
was static. A considerable proportion of the patients conspicuous. The signal intensity of the external/
probably had congenital cytomegalovirus infection, a extreme capsules on both sides is too high and there
diagnosis that can now be confirmed using PCR for is a distinct lesion in the parietal operculum on the
cytomegalovirus DNA on the filter paper containing left. There is also involvement of the dorsal part of
neonatal blood spots (the Guthrie card). the pons. At upper left, a sagittal T2-weighted image
Group F is characterized by predominant involve- of the cervicothoracic spine shows swelling of the
ment of the U fibers. cord with central high signal intensity (white arrows).
Group G is characterized by abnormalities pre- At upper right, a STIR image at the level of the chiasm
dominantly in the white matter of the posterior demonstrates swelling of the right part of the optic
fossa. chiasm with a central lesion (arrow). Although the
Subdividing patients with an unclassified leukoen- combination of optic neuritis followed by myelopathy
cephalopathy using these categories will facilitate fu- is reminiscent of Devic neuromyelitis optica, the
ture research on homogeneous subgroups of patients extent of cerebral lesions in this case argues in favor of
and allow pooling of data across multiple centers. the diagnosis acute disseminated encephalomyelitis.
892 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.9. Example I


109.7 Examples 893

Fig. 109.10. Example II

109.7.2 Example II posterior white matter, but also affecting the spleni-
um of the corpus callosum, extending towards the
The images shown in Fig. 109.10 reveal a diagnostic basal ganglia, and including the corticospinal tracts.
pattern. They relate to a 32-year-old man with pro- After contrast (third row), enhancement of a rim
gressive neurological disability. The FLAIR images within the lesion is seen.Although unusual at this age,
(first and second rows) show perfectly symmetrical, the pattern is diagnostic of the cerebral form of
confluent white matter lesions, mainly involving the X-linked adrenoleukodystrophy.
894 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.11. Example III


109.7 Examples 895

109.7.3 Example III per part of the left hemisphere, involving both cen-
trum semiovale and periventricular region. The cor-
A 7-year-old boy presented with intractable seizures tex in contact with this lesion appears too thick.
on the right. An MRI was obtained, which showed Parasagittal T1-weighted images of the left hemi-
extensive white matter abnormalities for which no sphere (second row) show the abnormalities in both
metabolic cause could be found (Fig. 109.11). The boy the gray and white matter. The IR images (third row)
was sent for a second opinion. Clinical history and also show the cortical and subcortical abnormalities.
neurological examination revealed evidence of an The coronal FLAIR images (fourth row) show the tri-
old, mild, right-sided hemiparesis. Sequential MRIs angular shape of the signal abnormalities, tapering as
taken during the following 2 years showed static they approach the lateral ventricle. The pattern is that
white matter abnormalities. The T2-weighted images of a balloon-type focal cortical dysplasia (Taylor
(first row) show white matter abnormalities in the up- type), simulating a white matter disease.

Fig. 109.12. Example IV


896 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.13. Example V

109.7.4 Example IV 109.7.5 Example V

The images of this 1-year-old girl were sent for an The T1-weighted parasagittal images (second row of
opinion because of an unclassified white matter dis- Fig. 109.13) show abnormalities in the temporal lobe
ease (Fig. 109.12). The upper two rows of T2-weighted of three different patients: cysts in the temporal pole
images show subcortical white matter lesions with in all three patients and widened temporal horns in
broadening of the overlying cortex. The sagittal T1- the first two. The diagnosis in each case can be made
weighted images (third row) show small indentations in conjunction with the axial T2-weighted images
in the ventricular lining (arrows), representing (first row). The images on the left showing multifocal
subependymal nodules. These nodules are dark on lesions with the largest lesions in the deep parietal
the T2-weighted images, while they are white on T1- white matter, together with the temporal abnormali-
weighted images. The pattern of cortico-subcortical ties, in a microcephalic child are highly suggestive of
tubers and subependymal nodules is indicative of congenital cytomegalovirus infection. The images in
tuberous sclerosis. the middle showing diffuse cerebral white matter ab-
normalities, diffuse cortical dysplasia, and pontine
hypoplasia (small sagittal image) demonstrate the
109.7 Examples 897

Fig. 109.14. Example VI

pattern of Walker–Warburg congenital muscular dys- 109.7.6 Example VI


trophy. The images on the right showing, in addition
to the cysts in the anterior temporal lobe, diffuse cere- In this patient with hypomelanosis of Ito (Fig.
bral white matter abnormalities and frontoparietal 109.14), the FLAIR images (first row) show confluent
subcortical cysts (small image) in a megalencephalic white matter abnormalities in the centrum semiovale
child are diagnostic of megalencephalic leukoen- and periventricular region. In addition, multiple
cephalopathy with subcortical cysts. black dots are seen, some arranged in rows. The sagit-
In the next series of examples (VI–X) unusual MR tal T2-weighted images (second row) show that the
findings are presented: black and white dots and dots are related to radially arranged widened perivas-
stripes. cular spaces.
898 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.15. Example VII

109.7.7 Example VII ond row of FLAIR images shows a multitude of black
dots but no stripes. They represent focal cysts, not
In this case of Lowe syndrome (Fig. 109.15), the first clearly following the perivascular spaces. The third
row of T2-weighted images show symmetrical white row of T1-weighted sagittal images confirms that the
matter abnormalities in the deep and periventricular black holes do not or do not only represent widened
white matter. Horizontal stripes in the corpus callo- perivascular spaces.
sum indicate widened perivascular spaces. The sec-
109.7 Examples 899

Fig. 109.16. Example VIII

109.7.8 Example VIII

In some subtypes of mucopolysaccharidosis, widened


perivascular spaces are the hallmark of the MR pat-
tern. This is demonstrated in the T1- and T2-weighted
transverse images and T1-weighted sagittal images
(Fig. 109.16) of a patient with Hurler syndrome. Often
the corpus callosum is involved.
900 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.17. Example IX

109.7.9 Example IX dentate nucleus, there is a striking widening of the


perivascular spaces, especially well depicted in the T1-
Figure 109.17 contains the T2-weighted transverse, weighted image (first row, right) and the sagittal and
sagittal, and coronal images and one T1-weighted im- coronal T2-weighted images (third row). Myelin depo-
age of a 4-year-old child with a mild variant of maple sition also seems to have happened mainly in perivas-
syrup urine disease. In addition to the abnormalities cular areas, leading to dark stripes on T2-weighted
in the globus pallidus, thalamus, midbrain, pons, and images.
109.7 Examples 901

Fig. 109.18. Example X

109.7.10 Example X

A 10-year-old boy presented with multiple transient


episodes of neurological dysfunction. In Fig. 109.18,
the T2-weighted images on the left show a multitude
of tiny lesions in the white matter and basal ganglia
on a background of more confluent cerebral white
matter lesions. The T1-weighted images in the middle
show a multitude of dark stripes, which were con-
firmed to be abnormal vessels on the MRA (source
image shown upper right), whereas the conventional
angiography shows the pattern of moyamoya syn-
drome.
The next three examples show leukoencephalo-
pathies linked to chromosomal abnormalities.
902 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.19. Example XI

109.7.11 Example XI

A 2-year-old boy presented with borderline macro-


cephaly, retarded development, and dysmorphic fea-
tures. Chromosomal analysis revealed an inversion-
duplication of chromosome 8p. The T2-weighted MR
images (Fig. 109.19) show dilated CSF spaces and
multiple white matter abnormalities. They were
found to be stable on follow-up.
109.7 Examples 903

Fig. 109.20. Example XII

109.7.12 Example XII

A 5-year-old girl was known to have psychomotor re-


tardation, dysmorphic features, eye abnormalities,
and hearing difficulties. Chromosomal analysis re-
vealed an unbalanced translocation between chromo-
some 6p25 and chromosome 20q13. The T2-weighted
images (Fig. 109.20) show spotty white matter abnor-
malities in the deep and periventricular white matter
and several dot-like lesions in the basal ganglia. The
coronal FLAIR image (lower right) confirms that
some of the abnormalities are related to enlarged
perivascular spaces.
904 Chapter 109 Pattern Recognition in White Matter Disorders

Fig. 109.21. Example XIII

109.7.13 Example XIII

A 2-year-old girl had a similar clinical picture and


MRI findings (Fig. 109.21; compare with Fig. 109.20).
Chromosomal analysis was normal. Because of the
striking clinical and MRI similarities, a submicro-
scopic deletion of chromosome 6p25 was suspected.
FISH analysis confirmed the chromosome 6p25 dele-
tion, illustrating the power of pattern recognition in
the reading of MR images.
References and Further Reading

1 Myelin and White Matter Dambska M,Laure-Kaminowska M.Myelination as a parameter


of normal and retarded brain maturation. Brain Dev 1990;
Asotra K, Macklin WB. Protein kinase C activity modulates 12: 214–220
myelin gene expression in enriched oligodendrocytes. Davison AN, Dobbing J. Myelination as a vulnerable period in
J Neurosci Res 1993; 34: 571–588 brain development. Br Med Bull 1966; 20: 40–44
Baumann N, Pham-Dinh D. Biology of oligodendrocyte and Deshmukh DS,Vorbrodt AW, Lee PK, Bear WD, Kuizon S. Studies
myelin in the mammalian central nervous system. Physiol on the submicrosomal fractions of bovine oligodendroglia:
Rev 2001; 81: 871–927 lipid composition and glycolipid biosynthesis. Neurochem
Benjamins JA, Iwata R, Hazlett J. Kinetics of entry of proteins Res 1988; 13: 571–582
into the myelin membrane. J Neurochem 1978; 31: 1077– De Vries GH, Norton WT. The fatty acid composition of sphin-
1085 golipids from bovine CNS axons and myelin. J Neurochem
Benveniste EN, Merrill JE. Stimulation of oligodendroglial pro- 1974; 22: 251–257
liferation and maturation by interleukin-2. Nature 1986; Dietrich RB, Bradley WG, Zaragoza IV, Otto RJ, Taira RK, Wilson
321: 610–613 GH, Kangerloo H. MR evaluation of early myelination pat-
Berlet HH,Volk B. Studies of human myelin proteins during old terns in normal and developmentally delayed infants. AJNR
age. Mech Ageing Dev 1980; 14: 211–222 Am J Neuroradiol 1988; 9: 69–76
Berndt JA, Kim JG, Hudson LD. Identification of cis-regulatory Dobbing J.Vulnerable periods in developing brain. In: Davison
elements in the myelin proteolipid protein (PLP) gene. AN, Dobbing J, eds. Applied neurochemistry. Oxford: Black-
J Biol Chem 1992; 267: 14730–14737 well, 1968: 287–316
Boiron F, Spivack WD, Deshmukh DS, Gould RM. Basis for phos- Dobbing J, Sands J. Quantitative growth and development of
pholipid incorporation into peripheral nerve myelin. J Neu- human brain. Arch Dis Child 1973; 48: 757–767
rochem 1993; 60: 320–329 Duhamel-Clerin E, Villarroya H, Mehtali M, Lapie P, Besnard F,
Bologa L. Oligodendrocytes, key cells in myelination and tar- Gumpel M, Lachapelle F. Cellular expression of an HMGCR
get in demyelinating diseases.J Neurosci Res 1985; 14:1–20 promoter-cat fusion gene in transgenic mouse brain: evi-
Bongarzone ER, Howard SG, Schonmann SG, Schonmann V, dence for a developmental regulation in oligodendrocytes.
Campagnoni AT. Identification of the dopamine D3 recep- Glia 1994; 11: 35–46
tor in oligodendrocyte precursors: potential role in regula- Dziewulska D, Jamrozik Z, Podlecka A, Rafalowska J. Do astro-
tion differentiation and myelin formation. J Neurosci 1998; cytes participate in rat spinal cord myelination? Folia
18: 5344–5353 Neuropathol 1999; 37: 81–86
Brody BA, Kinney HC, Kloman AS, Gilles FH. Sequence of central Farrer RG, Benjamins JA. Entry of newly synthesized ganglio-
nervous system myelination in human infancy. I. An autop- sides into myelin. J Neurochem 1992; 58: 1477–1484
sy study of myelination. J Neuropathol Exp Neurol 1987; 46: Fishman MA, Agrawal HC, Alexander A, Golterman J, Marten-
283–301 son RE, Mitchell RF. Biochemical maturation of human cen-
Brown MC, Moreno MB, Bongarzone ER, Cohen PD, Soto EF, tral nervous system myelin.J Neurochem 1975; 24: 689–694
Pasquini JM. Vesicular transport of myelin proteolipid and Flechsig P. Developmental (myelogenetic) localisation of the
cerebroside sulfates to the myelin membrane. J Neurosci cerebral cortex in the human subject. Lancet 1901; II: 1027–
Res 1993; 35: 402–408 1029
Burger D, Steck AJ, Bernard CCA, Kerlero de Rosbo N. Human Flechsig P. Anatomie des menschlichen Gehirns und Rücken-
myelin/oligodendrocyte glycoprotein: a new member of marks. Leipzig: Georg Thieme, 1920, 7–119
the L2/HNK-1 family. J Neurochem 1993; 61: 1822–1827 Fors L,Hood L,Saavedra RA.Sequence similarities of myelin ba-
Butt AM, Berry M. Oligodendrocytes and the control of myeli- sic protein promoters from mouse and shark: implications
nation in vivo: new insights from the rat anterior medullary for the control of gene expression in myelinating cells.
velum. J Neurosci Res 2000; 59: 477–488 J Neurochem 1993; 60: 513–521
Campagnoni AT. Molecular biology of myelin proteins from Futerman AH, Stieger B, Hubbard AL, Pagano RE. Sphin-
the central nervous system. J Neurochem 1988; 51: 1–14 gomyelin synthesis in rat liver occurs predominantly at the
Campagnoni AT, Verdi JM, Verity AN, Amur-Umarjee S. Post- cis and medial cisternae of the Golgi apparatus.J Biol Chem
transcriptional events in the expression of myelin protein 1990; 265: 8650–8657
genes. Ann NY Acad Sci 1990; 605: 270–279 Gilles FH. Myelination in the neonatal brain. Hum Pathol 1976;
Carson MJ, Behringer RR, Brinster RL, McMorris FA. Insulin-like 7: 244–248
growth factor I increases brain growth and central nervous Gilles FH, Shankle W, Dooling EC. Myelinated tracts: growth
system myelination in transgenic mice. Neuron 1993; 10: patterns.In:Gilles FH,Leviton A,Dooling EC.The developing
729–740 human brain. Boston: Wright, 1983, 117–192
Compston A, Zajicek J, Sussman J,Webb A, Hall G, Muir D, Shaw Goddard DR, Berry M, Butt AM. In vivo actions of fibroblasts
C,Wood A, Scolding N. Glial lineages and myelination in the growth factor-2 and insuline-like growth factor-I on oligo-
central nervous system. J Anat 1997; 190: 161–200 dendrocyte development and myelination in the central
nervous system. J Neurosci Res 1999; 57: 74–85
906 References and Further Reading

Goodrum JF, Earnhardt T, Goines N, Bouldin TW. Fate of myelin Matthieu JM. An introduction to the molecular basis of inherit-
lipids during degeneration and regeneration of peripheral ed myelin diseases. J Inherit Metab Dis 1993; 16: 724–732
nerve: an autoradiographic study. J Neurosci 1994; 14: Matthieu JM, Comte V, Tosic M, Honegger P. Myelin gene ex-
357–367 pression during demyelination and remyelination in ag-
Gould RM, Spivack W, Cataneo R, Holshek J, Konat G. Lipids and gregating brain cell cultures. J Neuroimmunol 1992; 40:
myelination. In: Crescenzi S, ed. A multidisciplinary ap- 231–234
proach to myelin diseases. New York: Plenum, 1987: 87–102 McLaurin J, Ackerley CA, Moscarello MA. Localization of basic
Gould RM, Freund CM, Palmer F, Feinstein DL. Messenger RNAs proteins in human myelin. J Neurosci Res 1993; 35: 618–628
located in myelin sheath assembly sites. J Neurochem Menkes JH. The leukodystrophies. N Engl J Med 1990; 322:
2000; 75: 1834–1844 54–55
Gupta SK, Pringle J, Poduslo JF, Mezei C. Induction of myelin Meyer-Franke A, Shen S, Barres BA. Astrocytes induce oligo-
genes during peripheral nerve remyelination requires a dendrocyte processes to align with and adhere to axons.
continuous signal from the ingrowing axon. J Neurosci Res Mol Cell Neurosci 1999; 14: 385–397
1993; 34: 14–23 Mickel HS, Gilles FH. Changes in glial cells during human telen-
Hasegawa M, Houdou S, Mito T,Takashima S, Asanuma K, Ohno cephalic myelinogenesis. Brain 1970; 93: 337–346
T. Development of myelination in the human fetal and in- Mikol DD, Rongnoparut P, Allwardt BA, Marton LS, Stefansson
fant cerebrum: a myelin basic protein immunohistochemi- K. The oligodendrocyte-myelin glycoprotein of mouse: pri-
cal study. Brain Dev 1992; 14: 1–6 mary structure and gene structure. Genomics 1993; 17:
Jacoby CG,Yuh WTC, Afifi AK, Bell WE, Schelper RL, Sato Y. Accel- 604–610
erated myelination in early Sturge-Weber syndrome Mitchell LS, Gillespie SC, McAllister F, Fanarraga ML, Kirkham D,
demonstrated by MR imaging. J Comput Assist Tomogr Kelly B, Brophy PJ, Grittiths IR, Montague P, Kennedy PGE.
1987; 11: 226–231 Developmental expression of major myelin protein genes
Jeckel D, Karrenbauer A, Birk R, Schmidt RR, Wieland F. Sphin- in the CNS of X-linked hypomyelinating mutant rumpshak-
gomyelin is synthesized in the cis Golgi. FEBS Lett 1990; er. J Neurosci Res 1992; 33; 205–217
261: 155–157 Morell P, ed. Myelin, 2nd ed. New York: Plenum Press, 1984
Kamholz J, Toffenetti, Lazzarini RA. Organization and expres- Morell P, Wiesmann U. A correlative synopsis of the leukodys-
sion of the human myelin basic protein gene. J Neurosci trophies. Neuropediatrics 1984; 15 (suppl): 62–65
Res 1988; 21: 62–70 Morell P, Quarles RH, Norton WT. Formation, structure, and bio-
Keene LMF, Hewer EE. Some observations on myelination in chemistry of myelin. In: Siegel GJ, Agranoff BW, Albers RW,
the human central nervous system. J Anat 1931; 66: 1–13 eds. Basic neurochemistry: molecular, cellular and medical
Kinney HC, Brody BA, Kloman AS, Gilles FH. Sequence of central aspects, 4th ed. New York: Raven Press, 1989, 109–136
nervous system myelination in human infancy. II. Patterns Norton WT. Recent advances in myelin biochemistry. Ann NY
of myelination in autopsied infants. J Neuropathol Exp Acad Sci 1984; 436: 5–10
Neurol 1988; 47: 217–234 Norton WT, Autilio LA.The lipid composition of purified bovine
Kinney HC, Karthigasan J, Borenshteyn NI, Flax JD, Kirschner brain myelin. J Neurochem 1966; 13: 213–222
DA. Myelination in the developing human brain: biochemi- Norton WT, Cammer W. Isolation and characterization of
cal correlates. Neurochem Res 1994; 19: 983–996 myelin. In: Morell P, ed. Myelin. Plenum, New York, 1984,
Konola JT, Yamamura T, Tyler B, Lees MB. Orientation of the pp 147–195
myelin proteolipid protein C-terminus in oligodendroglial Notterpek LM, Rome LH. Functional evidence for the role of ax-
membranes. Glia 1992; 5: 112–121 olemma in CNS myelination. Neuron 1994; 13: 473–485
Langworthy OR. Development of behavior patterns and Pagano RE. The Golgi apparatus: insights from lipid biochem-
myelinization of the nervous system in human fetus and in- istry. Biochem Soc Trans 1990; 18: 361–366
fant. (Contributions to embryology, vol XXIV) Washington Patsalos PN, Wiggins RC. Brain maturation following adminis-
DC: Carnegic Institute of Washington, 1933: 1–57 tration of phenobarbital, phenytoin, and sodium valproate
Lappe-Siefke C, Goebbels S, Gravel M, Nicksch E, Lee J, Braun to developing rats or to their dams: effects on synthesis of
PE, Griffiths IR, Vane K-A. Disruption of Cnp1 uncouples brain myelin and other subcellular membrane proteins. J
oligodendroglial functions in axonal support and myelina- Neurochem 1982; 39: 915–923
tion. Nat Genet 2003; 33: 366–374 Percy AK, McKhann GM. The biochemistry of myelin and the
Larocca JN, Rodriguez-Gabin AG. Myelin biogenesis: vesicle leukodystrophies. In: Vinken PJ, Bruyn GW, eds. Handbook
transport in oligodendrocytes. Neurochem Res 2002; 27: of clinical neurology, vol 10, Amsterdam: North Holland
1313–1329 Publishing Company, 1970, 134–149
Lemke G. Unwrapping the genes of myelin. Neuron 1988; Poduslo SE, Jang Y. Myelin development in infant brain. Neu-
1: 535–543 rochem Res 1984; 9: 1615–1626
Lemke G. The molecular genetics of myelination: an update. Pope A. Neuroglia: quantitative aspects. In: Schoffeniels E,
Glia 1993; 7: 263–271 Franck G, Hertz L,Tower DB, eds. Dynamic properties of glia
Ludwin SK. Remyelination in the central nervous system and cells. New York: Pergamon Press, 1977, 13–20
the peripheral nervous system. In: Waxman SG, ed. Ad- Poser CM. Discussion des rapports sur les maladies démyélin-
vances in neurology: functional recovery in neurological isantes. Proc Third Intern Congr Neuropathol. Brussels:
disease, vol 47, New York: Raven Press, 1988: 215–254 Editions Acta Medica Belgica, 1957, 106–111
Martin DW. Membranes. In: Martin DW, Mayes PA, Rochwel VW, Poser CM.Leukodystrophy and the concept of dysmyelination.
Granner DK, eds. Harper’s review of biochemistry, 20th ed. Arch Neurol 1961; 4: 323–332
Los Altos: Lange Medical Publications, 1985, 448–463 Poser CM. Dysmyelination revisited.Arch Neurol 1978; 35: 401–
Martin E,Boesch C,Zuerrer M,Kikinis R,Molinari L,Kaelin P,Bolt- 407
shauser E,Duc G.MR imaging of brain maturation in normal
and developmentally handicapped children. J Comput
Assist Tomogr 1990; 14: 685–692
References and Further Reading 907

Probstmeier R, Fahrig T, Spiess E, Schachner M. Interactions of Vinores SA, Herman MM. Phagocytosis of myelin by astrocytes
the neural cell adhesion molecule and the myelin-associat- in explants of adult rabbit cerebral white matter main-
ed glycoprotein with collagen type I: involvement in fibril- tained on Gelfoam matrix. J Neuroimmunol 1993; 43: 169–
logenesis. J Cell Biol 1992; 116: 1063–1070 176
Richardson EP. Myelination in the human central nervous Vogel US, Thompson RJ. Molecular structure, localization, and
system. In: Haymaker W, Adams RD, eds. Histology and possible functions of the myelin-associated enzyme
histopathology of the nervous system. Springfield: Charles 2’,3’-cyclic nucleotide 3’-phosphodiesterase. J Neurochem
C Thomas, 1982, 146–173 1988; 50: 1667–1677
Rodriguez M. Central nervous system demyelination and re- Vogt O. Quelques considérations genérales sur la myélo-archi-
myelination in multiple sclerosis and viral models of dis- tecture du lobe frontal. Rev Neurol 1910; 20: 405–420
ease. J Neuroimmunol 1992; 40: 255–264 Vourc’h P, Andres C. Oligodendrocyte myelin glycoprotein
Rodriguez M, Prayoonwiwat N, Howe C, Sanborn K. Proteolipid (OMgp): evolution, structure and function. Brain Res Rev
protein gene expression in demyelination and remyelina- 2004; 45: 115–124
tion of the central nervous system: a model for multiple Waxman SG, Ritchie JM. Molecular dissection of the myelinat-
sclerosis. J Neuropathol Exp Neurol 1994; 53: 136–143 ed axon. Ann Neurol 1993; 33: 121–136
Rorke LB, Riggs HE, Showers MJC, Cabrera CV, Cohn M. Myelina- Waxman SG, Sims TJ. Specificity in central myelination: evi-
tion of the brain in the newborn. Philadelphia: Lippincott, dence for local regulation of myelin thickness. Brain Res
1969: 1–105 1984; 292: 179–185
Royland J,Konat GW,Kanoh M,Wiggins RC.Down regulation of Waxman AG, Black JA, Sontheimer H, Kocsis JD. Glial cells and
myelin-specific mRNAs in the mechanism of hypomyelina- axo-glial interactions: implications for demyelinating disor-
tion in the undernourished developing brain.Dev Brain Res ders. Clin Neurosci 1994; 2: 202–210
1992; 65: 223–226 Weimbs T, Stoffel W. Proteolipid protein (PLP) of CNS myelin:
Royland JE, Konat G, Wiggins RC. Abnormal upregulation of positions of free, disulfide-bonded and fatty acid thioester-
myelin genes underlies the critical period of myelination in linked cysteine residues and implications for the mem-
undernourished developing rat brain. Brain Res 1993; 607: brane topology of PLP.Biochemistry 1992;31:12289–12296
113–116 Wiggings RC. Myelination: a critical stage in development.
Royland JE, Konat GW, Wiggins RC Myelin gene activation: Neurotoxicology 1986; 7: 103–120
a glucose sensitive critical period in development. J Neuro- Williams KA, Deber CM. The structure and function of central
sci Res 1993; 36: 399–404 nervous system myelin. Crit Rev Clin Lab Sci 1993; 30: 29–64
Russell JW, Chen H-L, Golovoy D. Insuline-like growth factor-1 Wisniewski KE, Schmidt-Sidor B. Postnatal delay of myelin for-
promotes myelination of peripheral sensory axons. J Neu- mation in brains from Down syndrome infants and chil-
ropathol Exp Neurol 2000;59:575–584 dren. Clin Neuropathol 1989; 8: 55–62
Saito M,Yu RK. Role of myelin-associated neuraminidase in the Wood PM, Bunge RP. The origin of remyelinating cells in the
ganglioside metabolism of rat brain myelin. J Neurochem adult central nervous system: the role of the mature oligo-
1992; 58: 83–87 dendrocyte. Glia 1991; 4: 225–232
Schousboe A, Westergaard N, Waagepetersen HS, Larsson OM, Yakovlev PI, Lecours AR. The myelogenetic cycles of regional
Bakken IJ, Sonnewald U. Trafficking between glia and neu- maturation of the brain.In:Minkowski A,ed.Regional devel-
rons of TCA cycle intermediates and related metabolites. opment of the brain in early life. Oxford: Blackwell, 1967:
Glia 1997; 21: 99–105 3–70
Seitelberger F. Structural manifestations of leukodystrophies. Yamamoto Y, Yoshikawa H, Nagano S, Kondoh G, Sadahiro S,
Neuropediatrics 1984; 15 (suppl): 53–61 Gotow T, Yanagihara T, Sakoda S. Myelin-associated oligo-
Shine HD, Readhead C, Popko B, Hood L, Sidman RL. Morpho- dendrocytis basic protein is essential for normal arrange-
metric analysis of normal, mutant, and transgenic CNS: cor- ment of the radial component in central nervous system
relation of myelin basic protein expression to myelinogen- myelin. Eur J Neurosci 1999; 11: 847–855
esis. J Neurochem 1992; 58: 342–349 Ye P, Carlson J, D’Ercole AJ. Insuline-lilke growth factor-I influ-
Sinoway MP, Kitagawa K, Timsit S, Hashim GA, Colman DR. Pro- ences the initiation of myelination: studies of the anterior
teolipid protein interactions in transfectants: implications commissure of transgenenic mice. Neurosci Lett 1995; 201:
for myelin assembly. J Neurosci Res 1994; 37: 551–562 235–238
Skoff RP. Neuroglia: a reevaluation of their origin and develop- Zurbriggen A,Vandevelde M, Steck A, Angst B. Myelin-associat-
ment. Pathol Res Pract 1980; 168: 279–300 ed glycoprotein is produced before myelin basic protein in
Smith R. The basic protein of CNS myelin: its structure and lig- cultured oligodendrocytes. J Neuroimmunol 1984; 6: 41–49
and binding. J Neurochem 1992; 59: 1589–1608
Stevens B, Porta S,Haak LL, Gallo V, Fields RD. Adenosine: a neu-
ron-glial transmitter promoting myelination in the CNS in
response to action potentials. Neuron 2002; 36: 855–868
2 Classification of Myelin Disorders
Stryer L. Introduction to biological membranes. In: Stryer L, ed.
Biochemistry, 2nd ed. New York: Freeman, 1981, 205–230 Adams RD, Kubik CS. The morbid anatomy of the demyelina-
Svennerholm L. Some aspects of the biochemical changes in tive diseases. Am J Med 1952; 12: 510–546
leukodystrophy. In: Folch PJ, Bauer H, eds. Brain lipids and Adams RD, Richardson EP. The demyelinative diseases of the
lipoproteins and the leukodystrophies. Amsterdam: Else- human nervous system. In: Folch P, Folch J, eds. Chemical
vier, 1963, 104–119 pathology of the nervous system. Oxford: Pergamon Press,
van der Knaap MS,Valk J, Bakker CJ, Schooneveld M, Faber JAJ, 1961: 162–194
Willemse J, Gooskens PHJM. Myelination as expression of Alzheimer A. Beiträge zur Kenntnis der pathologischen Neu-
the functional maturity of the brain. Dev Med Child Neurol roglia und ihrer Beziehungen zu den Abbauvorgängen im
1991; 33: 849–857 Nervengewebe. Nissl-Alzheimer Arbeiten 1910; 3: 401–562
908 References and Further Reading

Austin J, McAfee D, Armstrong D, O’Rourke M, Shearer L, Bach- Lassmann H, Ammerer HP, Kulnig W. Ultrastructural sequence
hawat B. Low sulfatase activities in metachromatic leuko- of myelin degradation. Wallerian degeneration in the rat
dystrophy.Trans Am Neurol Assoc J 1964:147–150 optic nerve. Acta Neuropathol (Berl) 1978; 44: 91–102
Bérard-Badier M, Paillas JE, Gastaut H, Edgar GWF. Essai sur la Matthieu JM. An introduction to the molecular basis of inherit-
significance des démyelinisations dans l’idiotie amauro- ed myelin diseases. J Inherit Metab Dis 1993; 16: 724–732
tique infantile. Psychiatr Neurol 1958; 132: 50–93 Menkes JH. The leukodystrophies. N Engl J Med 1990; 322:
Bielschowsky M,Henneberg R.Ueber familiäre diffuse Sklerose 54–55
(leukodystrophia cerebri progressiva hereditaria) J Psychol Merzbacher L. Eine eigenartige familiär-hereditäre Erkran-
Neurol 1928; 36: 131–181 kungsform (aplasia axialis extracorticalis congenita). Z
Blackwood W.The histological classification of diffuse demyeli- Gesamte Neurol Psychiatrie 1910; 3: 1–138
nating cerebral sclerosis. In: Cerebral lipidosis, a sympo- Morell P, Wiesmann U. A correlative synopsis of the leukodys-
sium. Oxford: Blackwell Scientific Publications, 1957: 1–10 trophies. Neuropediatrics 1984; 15 (suppl): 62–65
Carswell R. Pathological anatomy: illustrations of the elemen- Neubürger K. Histologisches zur Frage der diffusen Hirn-
tary forms of disease. London: Longmans, Green , 1938 sklerose. Z Gesamte Neurol Psychiatrie 1921; 73: 336–352
Challa VR. White matter lesions in MR imaging of elderly sub- Peiffer J. Differentiation of various types of leukodystrophy.
jects. Radiology 1987; 164: 874–875 World Neurology 1962; 3: 580–597
Charcot JM. Lectures on the diseases of the nervous system, Pelizaeus F. Ueber eine eigenartige familiäre Entwicklungs-
vol. 3. London: New Sydenham Society, 1868 (Lectures de- hemmung vornehmlich auf motorischem Gebiet. Arch
livered in 1868, English translation published in 1877) Psychiatrie Nervenkr 1899; 31: 100–104
Cruveilhier J. Anatomie pathologique du corps humain, vol II, Poser CM. Discussion des rapports sur les maladies demyelin-
fasc XXXII. Paris: Baillière, 1835–1842 isantes. Proc Third Intern Congr Neuropathol. Brussels. Edi-
Davison AN, Dobbing J. Myelination as vulnerable period in tions Acta Medica Belgica 1957, 106–111
brain development. Br Med Bull 1966; 20: 40–44 Poser CM.Leukodystrophy and the concept of dysmyelination.
Diezel PB. Histochemische Untersuchungen an den Globoid- Arch Neurol 1961; 4: 323–332
zellen der familiaeren infantilen diffusen Sklerose vom Ty- Poser CM. Dysmyelination revisited. Arch Neurol 1978; 35:
pus Krabbe. Virchows Arch Pathol Anat 1955; 327: 206–228 401–407
Edgar GWF. Approche biochimique des lipidoses et des leu- Poser CM. The dysmyelinating diseases. In: Baker AB, Joynt RJ,
codystrophies. Rev Neurol 1955; 92: 277–284 eds. Clinical neurology, vol 3. Philadelphia: Harper & Row,
Fardeau M, Lapresle J. Maladie de Tay-Sachs avec atteinte im- 1987: chap 34
portante de la substance blanche. A propos de deux obser- Raine CS. The neuropathology of myelin diseases. In: Morell P,
vations anatomo-cliniques. Rev Neurol 1963; 109: 157–175 ed. Myelin, 2nd ed. New York: Plenum Press, 1984, 259–310
Foelling A. Ueber Ausscheiding von Phenylbrenztraubensäure Ranvier L-A (1878) Leçons sur l’histologie due système
in den Harn als Stoffwechselanomalie in Verbindung mit nerveux (2 vol.). Weber E. ed. Paris: Savy
Imbezilität. Z Physiol Chem 1934; 227: 169–176 Schilder P. Zur Kenntnis der sogenannten diffusen Sklerose.
George AE, de Leon MJ, Gentes CI, Miller J, London E, Budzilo- Ueber Encephalitis periaxialis diffusa. Z Gesamte Neurol
vich GN, Ferris S, Chase N. Leukoencephalpathy in normal Psychiatrie 1912; 10, 1–60
and pathologic aging: CT of brain lucencies. AJNR Am J Scholz W. Klinische, pathologisch-anatomische und erbbiolo-
Neuroradiol 1986; 7: 561–566 gische Untersuchungen bei familiärer,diffuser Hirnsklerose
Gupta SR, Naheedy MH, Young JC, Ghobrial M, Rubino FA, Hin- im Kindesalter. Z Gesamte Neurol Psychiatrie 1925; 99:
do W. Periventricular white matter changes and dementia. 651–717
Clinical, neuropsychological, radiological and pathological Seitelberger F. Structural manifestations of leukodystrophies.
correlation. Arch Neurol 1988; 45: 637–641 Neuropediatrics 1984; 15 (suppl): 53–61
Hallervorden J. Die zentralen Entmarkungskrankheiten. Dtsch Stam FC, Heslinga JM, Deierkauf FA, Booij HL. Leukodystrophy
Z Nervenheilk 1940; 150: 201–239 of the Norman-Greenfield and Krabbe type. Psychiatry
Hauw JJ, Delaère P, Seilhean D, Cornu P. Morphology of de- Neurol Neurosurg 1962; 65: 254–265
myelination in the human central nervous system. J Neu- Stam FC. Concept, classification and nosology of the leuko-
roimmunol 1992; 40: 139–152 dystrophies. In:Vinken PJ, Bruyn GW, eds. Handbook of clin-
Herschkowitz N, Schulte FJ. The lipidoses: from detect to dys- ical neurology, vol 10. Amsterdam: North Holland Publish-
function. Neuropediatrics 1984; 15: 110–111 ing Company, 1970, 1–42
Heubner O. Ueber diffuse Hirnsklerose. Charité-Ann 1897; 22: Suzuki K, Suzuki Y. Globoid cell leukodystrophy (Krabbe’s dis-
298–310 ease): deficiency of galactocerebroside beta-galactosidase.
Huk WJ, Bydder GM, Curati WL. Degenerative disorders of the Proc Natl Acad Sci 1970; 66: 302–309
brain and white matter diseases. In: Huk WJ, Gademann G, Thieffry S, Bertrand I, Bargeton E, Edgar GWF, Arthuis M. Idiotie
Friedmann G, eds. Magnetic resonance imaging of central amaurotique infantile avec alterations graves de la sub-
nervous system diseases. Berlin: Springer, 1990: 197–224 stance blanche. Rev Neurol 1960; 102: 130–152
Jervis GA.Studies on phenylpyruvic oligofrenia: the position of Virchow R. Ueber das ausgebreitete Vorkommen einer
the metabolic error. J Biol Chem 1947; 169: 651–656 dem Nervenmark analogen Substanz in den tierischen Ge-
Johnson MA, Pennock JM, Bydder GM, Steiner RE, Thomas DJ, weben. Virchows Arch Pathol Anat: 1854; 562–572
Hayward R, Bryant DRT, Payne JA, Levene MI, Whitelaw A, Von Hirsch T, Peiffer J. Ueber histologische Methoden in
Dubowitz LMS, Dubowitz V. Clinical NMR imaging of the der Differentialdiagnose von Leukodystrophien und Lipi-
brain in children: normal and neurologic disease. AJR Am J dosen. Arch Psychiatrie Z Neurol 1955; 194: 88–104
Roentgenol 1983; 141: 1005–1018 Von Hirsch T, Peiffer J. A histochemical study of the pre-lipid
Krabbe K. A new familial infantile form of diffuse brain-sclero- and metachromatic degenerative products in leucodystro-
sis. Brain 1916; 39: 74–114 phy. In: Cerebral lipidoses, a symposium. Oxford: Blackwell
Scientific Publications, 1957, 68–76
References and Further Reading 909

3 Selective Vulnerability Okun JG, Horster F, Farkas LM, feyh P, Hinz A, Sauer S, Hoffmann
GF, Unsicker K, Mayatepek E, Kolker S. Neurodegeneration
Albin RL. Basal ganglia neurotoxins. Neurol Clin 2000; 18: 665– in methylmalonic aciduria involves inhibition of complex II
680 and the tricarboxylic acid cycle, and synergistically acting
Barboriak DP, Provenzale JM, Boyko OB. MR diagnosis of excitotoxicity. J Biol Chem 2002; 277: 14674–14680
Creutzfeldt-Jakob disease:significance of high signal inten- Pasternak JF, Predey TA, Mikhael MA. Neonatal asphyxia: vul-
sity of the basal ganglia. AJR Am J Roentgenol 1994; 162: nerability of basal ganglia, thalamus, and brainstem. Pedi-
137–140 atr Neurol 1991; 7: 147–149
Davison AN, Dobbing J. Myelination as a vulnerable period in Pulsinelli WA. Selective neuronal vulnerability: morphological
brain development. Br Med Bull 1966; 22: 40–44 and molecular characteristics. Prog Brain Res 1985; 63:
Dubeau F, De Stefano N, Zifkin BG, Arnold DL, Shoubridge EA 29–37
Oxidative phosphorylation defect in the brains of carriers Reneman L, Majoie CB, Habrakeb JB, Den Heeten GJ. Effects of
of the tRNAleu(UUR) A3243G mutation in a MELAS pedi- ecstasy (MDMA) on the brain in abstinent users; initial ob-
gree. Ann Neurol 2000; 47: 179–185 servations with diffusion and perfusion MR imaging. Radi-
Gosztonyi G, Koprowski H. The concept of neurotropism and ology 2001; 220: 611–617
selective vulnerability (“pathoclisis”) in virus infections of Reneman L, Majoie CBLM, Flick H, den Heeten GJ. Reduced N-
the nervous system- a historical overview. Curr Top Micro- acetylasparate levels in the frontal cortex of 3,4-methyl-
biol Immunol 2001; 253: 1–13 enedioxymetamphetamine (ecstasy) users: preliminary re-
Gosztonyi G,Koprowski H.The concept of neuropotism and se- sults. AJNR Am J Neuroradiol 2002; 23: 231–237
lective vulnerability (“pathoclisis”) in virus infections of the Ricaurte GA, Yuan J, Hatzidimitriou G, Cord BJ, McCann UD. Se-
nervous system – a historical overview. Curr Top Microbiol vere dopaminergic neurotoxicity in primates after a com-
Immunol 2001; 253: 1–13 mon recreational dose regimen of MDMA (“ecstasy”). Sci-
Govaert P, Lequin M, Swarte R, Robben S, de Coo R, Weisglas- ence 2002; 297: 2260–2263
Kuperus N, de Rijke Y, Sinaasappel M, Barkovich J. Changes Schmidt R, Offenbacher H, Fazekas F, Payer F, Kleinert R, Porsch
in globus pallidus with (pre)term kernincterus. Pediatrics G. Magnetic resonance imaging, computed tomography,
2003; 112: 1256–1263 and autopsy findings after cardiorespiratory arrest. J Neu-
Guentchev M, Wanschitz J, Voigtlander T, Flicker H, Badka H. roimaging 1991; 1: 197–199
Selective neuronal vulnerability in human prion diseases. Scholz W. Selective neuronal necrosis and its topistic patterns
Fatal familial insomnia differs from other types of prion dis- in hypoxemia and oligemia. J Neuropathol Exp Neurol
eases. Am J Pathol 1999; 155: 1453–1457 1953; 12: 249–261
Hawker K, Lang AE. Hypoxic–ischemic damage of the basal Scholz W. Topistic lesions. In: Schadé JP, McMenemey WH, eds.
ganglia. Mov Disord 1990; 5: 219–224 Selective vulnerability of the brain in hypoxaemia. Oxford:
Johnston MV, Goldstein GW. Selective vulnerability of the de- Blackwell ScientificPublications, 1963: 257–267
veloping brain to lead. Curr Opin Neurol 1998; 11:689–693 Sims NR. Energy metabolism and selective neuronal vulnera-
Johnston MV, Hoon AH. Possible mechanisms in infants for bility following global cerebral ischemia. Neurochem Res
selective basal ganglia damage from asphyxia, kernicterus 1992; 17: 923–931
or mitochondrial encephalopathies. J Child Neurol 2000; Spielmeyer W. Zur Pathogenese örtlich elektiver Gehirnverän-
15: 588–591 derungen. Z Neurol Psychiatrie 1925; 99: 756–777
Kodama T, Numaguchi Y, Gellad FE, Dwyer BA, Kristt DA. Mag- Suzuki H, Takanashi J, Saeki N, Kohno Y. Temporal parental nu-
netic resonance imaging of limbic encephalitis. Neuroradi- trition in children causing T1 shortening in the anterior pi-
ology 1991; 33: 520–523 tuitary gland and globus pallidus. Neuropediatrics 2003;
Kölker S, Kohr G, Ahlemeyer B, Okun JG, Pawlak V, Horster F, 34: 200–204
Mayatepek E, Krieglstein J, Hoffmann GF. Ca(2+) and Na(+) Tarasów E, Panasiuk A, Siergiejczyk L, Orzechowska-Bobkie-
dependence of 3-hydroxyglutarate-indiced excitotoxicity wicz A, Lewszuk A, Waleck J, Porkopowicz D. MR and 1H MR
in primary neuronal cultures from chick embryo telen- spectroscopy of the brain in patients with lever cirrhosis
cephalons. Pediatr Res 2002; 52: 199–206 and early stages of hepatic encephalopathy. Hepatogas-
Kölker S, Mayatepek E, Hoffmann GF. White matter disease in troenterology 2003; 50: 2149–2153
cerebral organic acid disorders: clinical implications and Valk J. Selective involvement of CNS structures in pediatric
suggested pathomechanisms. Neuropediatrics 2002; 33: neuroradiology. Riv Neuroradiol 1993; 6: 3–10
225–231 Valk J, van der Knaap MS. Selective vulnerability in toxic en-
Lipton SA, Rosenberg PA.Excitatory amino acids as a final com- cephalopathies and metabolic disorders. Riv Neuroradiol
mon pathway for neurologic disorders. N Engl J Med 1994; 1996; 9: 749–760
330: 613–622 Vogt C, Vogt O. Sitz und Wesen der Krankheiten im Licht der
Ludolph AC, Riepe M, Ullrich K. Excitotoxicity, energy metabo- topistischen Hirnforschung und des Varierens der Tiere.
lism and neurodegeneration. J Inherit Metab Dis 1993; 16: J Psychol Neurol 1937; 47: 237–457
716–723 Yamada M, Inaba A,Yamawaki M, Ishida K,Yokota T, Uchihara T,
McCann UD, Szabo Z, Scheffel U, Dannals RF, Ricaurte GA. Eishi Y, Okeda R. Paraneoplastic encephalo-myelo-gan-
Positron emission tomography evidence of toxic effect of glionitis: cellular binding sites of the antineuronal anti-
MDMA (ecstasy) on brain serotonin neurons in human be- body. Acta Neuropathol (Berl) 1994; 88: 85–92
ings. Lancet 1998; 352: 1433–1437
Meyer A. Section of psychiatry. The selective regional vulnera-
bility of the brain and its relation to psychiatric problems.
Proc R Soc Med 1936; 29: 1175–1181
910 References and Further Reading

4 Myelination and Retarded Myelination Engelbrecht V, Malms J, Kahn T, Grünewald S, Mödder U. Fast
spin-echo MR imaging of the pediatric brain. Pediatr Radiol
Ajayi-Obe M, Saeed N, Cowan FM, Rutherford MA, Edwards AD. 1996; 26: 259–264
Reduced development of cerebral cortex in extremely Engelbrecht V, Rassek M, Preiss S, Wald C, Mödder U. Age-de-
preterm infants. Lancet 2000; 356: 1162–1163 pendent changes in magnetization transfer contrast of
Autti T, Raininko R, Vanhanen SL, Kallio M, Santavuori P. MRI of white matter in the pediatric brain. AJNR Am J Neuroradiol
the normal brain from early childhood to middle age.II.Age 1998; 19: 1923–1929
dependence of signal intensity of T2-weighted images. Engelbrecht V, Scherer A, Rassek M, Witsack HJ, Mödder U. Dif-
Neuroradiology 1994; 36: 649–651 fusion-weighted MR imaging in the brain in children: find-
Ball WS Jr. Imaging of the brain in children. Curr Opin Radiol ings in the normal brain and in the brain with white matter
1991; 3: 895–905 diseases. Radiology 2002; 222: 410–418
Barkovich AJ, Kjos BO, Jackson DE, Norman D. Normal matura- Ferrie JC, Barantin L, Saliba E, Akoka S, Tranquart F, Sirinelli D,
tion of the neonatal and infant brain: MR imaging at 1.5T1. Pourcelot L. MR assessment of the brain maturation during
Radiology 1988; 166: 173–180 the perinatal period: quantitative t2 MR study in premature
Barkovich AJ, Gressens P, Evrard P. Formation, maturation, and newborns. Magn Reson Imaging 1999; 17: 1275–1288
disorders of brain neocortex. AJNR Am J Neuroradiol 1992; Finelli DA, Hurst GC, Amantia P Jr, Gullapali RP, Apicella A. Cere-
13: 423–446 bral white matter: technical development and clinical ap-
Barkovich AJ. Concepts of myelin and myelination in neuro- plications of effective magnetization transfer (MT) power
radiology 2000; 21: 1099–1109 concepts for high-power transfer, thin-section, quantitative
Battin MR, Maalouf EF, Counsell SJ, Herlihy AH, Rutherford MA, MT examinations. Radiology 1996; 199: 219–226
Azzopardi D, Edwards AD. Magnetic resonance imaging of Flechsig P. Anatomie des menschlichen Gehirns und Rücken-
brain in very preterm infants: visualization of the germinal marks auf myelogenetischer Grundlage. Leipzig: Georg
matrix, early myelination, and cortical folding. Pediatrics Thieme, 1920
1998; 101: 957–962 Forbes KPN, Pipe JG, Bird CR. Changes in brain water diffusion
Benes FM, Turtle M, Khan Y, Farol P. Myelination of a key relay during the 1st year of life. Radiology 2002; 222: 405–409
zone in the hippocampal formation occurs in the human Garel C, Chantrel E, Brisse H, Elmaleh M, Luton D, Oury JF, Sebag
brain during childhood, adolescence, and adulthood. Arch G, Hassan M. Fetal cerebral cortex: normal gestational land-
Gen Psychiatry 1994; 51: 477–484 marks identified using prenatal MR imaging. AJNR Am J
Blaser S, Harwood-Nash DCF. Radiology of the developing cen- Neuroradiol 2001; 22: 184–189
tral nervous system. Curr Opin Neurol Neurosurg 1992; 5: Ge Y, Grossman RI, Babb JS, Rabin ML, Mannon LJ, Kolson DL.
843–848 Age-related total gray matter and white matter changes in
Breger RK,Yetkin FZ, Fisher ME, Papke RA, Haughton VM, Rimm normal adult brain. Part I: volumetric MR imaging analysis.
AA.T1 and T2 in the cerebrum: correlation with age, gender, AJNR Am J Neuroradiol 2002; 23: 1327–1333
and demographic factors. Radiology 1991; 181: 545–547 Ge Y, Grossmann RI, Babb JS, Rabin ML, Mannon LJ, Kolson DL.
Chi JG, Dooling EC, Gilles FH. Gyral development of the human Age-related total gray matter and white matter changes is
brain. Ann Neurol 1977; 1: 86–93 normal adult brain. Apart II: Quantitative magnetization
Childs AM, Remenghi LA, Evans DJ, Ridgeway J, Saysell M, Mar- transfer ratio histogram analysis. AJNR Am J Neuroradiol
tinez D, Arthur R,Tanner S, Levene MI. MR features of devel- 2002; 23: 1334–1341
oping periventricular white matter in preterm infants: evi- Girard N, Raybaud C, du Lac P. Étude de la myélinisation
dence of glial cell migration. AJNR Am J Neuroradiol 1998; cérébrale en IRM. MRI study of brain myelination. J Neuro-
19: 971–976 radiol 1991; 18: 291–307
Childs AM, Ramenghi LA, Cornette L,Tanner SF, Arthur RJ, Mar- Hansen PE, Ballesteros MC, Soila K, Garcia L, Howard JM. MR
tinez D, Levene MI. Cerebral maturation in premature in- imaging of the developing human brain. Part 1: prenatal
fants: quantitative assessment using MR imaging. AJNR Am development. Radiographics 1993; 13: 21–36
J Neuroradiol 2001; 22: 1577–1582 Hittmair K, Wimberger D, Rand T, Prayer L, Bernert G, Kramer J,
Counsell SJ, Maalouf EF, Fletcher AM, Duggan P, Battin M, Lewis Imhof H. MR assessment of brain maturation: comparison
HJ, Herlily AH, Edwards D, Bydder GM, Rutherford MA. MR of sequences. AJNR Am J Neuroradiol 1994; 15: 425–433
imaging assessment of myelination in the very preterm Hittmair K, Kramer J, Rand T, Bernert G, Wimberger D. Infraten-
brain. AJNR Am J Neuroradiol 2002; 23: 872–881 torial brain maturation: a comparison of MRI at 0.5 and 1.5
Counsell SJ, Kennea NL, Herlihy AH, Allsop JM, Harrison MC, T. Neuroradiology 1996; 38: 360–366
Cowan FM, Hajnal JV, Edwards B, Edwards AD, Rutherford Holland BA, Haas DK, Norman D, Brant-Zawadski M, Newton
MA. T2 relaxation values in the developing preterm brain. TH. MRI of normal brain maturation. AJNR Am J Neuroradi-
AJNR Am J Neuroradiol 2003; 24: 1654–1660 ol 1986; 7: 201–208
Dietrich RB, Badley WG, Zaragoza EJ IV, Otto RJ,Taira RK,Wilson Hosoya T, Adachi M, Yamaguchi K, Haku T. MRI anatomy of
GH, Kangarloo H. MR evaluation of early myelination pat- white matter layers around the trigone of the lateral ventri-
terns in normal and developmentally delayed infants. AJNR cle. Neuroradiology 1998; 40: 477–482
Am J Neuroradiol 1988; 9: 69–76 Huisman TAGM, Wisser J, Martin E, Kubik-Huch R, Marineck B.
Dooling EC, Chi JG, Gilles FH. Telencephalic development: Fetal magnetic resonance imaging of the central nervous
changing gyral patterns. In: Gilles FH, Leviton A, Dooling EC. system: a pictorial essay. Eur Radiol 2002; 12: 1952–1261
The deverloping human brain. Growth and epidemiologic Hüppi PS, Maier SE, Peled S, Zientara GP, Barnes PD, Jolenz FA,
neuropathology. Boston: John Wright, 1983: 94–104 Volpe JJ. Microstructural development of human in as-
Duprez T, Ghaniani S, Smith AM, Gadisseux JF, Evrard P. Focal sessed in vivo by diffusion tensor magnetic resonance
seizure-induced premature myelination: speculation from imaging. Pediatr Res 1998; 44: 584–590
serial MRI. Neuroradiology 1998; 40: 580–582
References and Further Reading 911

Hüppi PS, Warfield S, Kikinis R, Barnes PD, Zientara GP, Jolensz Naidich TP, Grant JL, Altman N, Zimmerman RA, Birchansky SB,
FA, Tsuij MK, Volpe JJ. Quantitative magnetic resonance Braffman BM, Daniel JL. The developing cerebral surface.
imaging of brain development in premature and mature Neuroimaging Clin N Am 1994; 4: 201–240
newborns. Ann Neurol 1998; 43: 224–235 Neill JJ, Shiran SI, McKinstry RC, Schefft GL, Snyder AZ, Almli CR,
Johnson MA, Pennock JM, Bydder GM, Steiner RE, Thomas DJ, Akbubak E, Aronovitz JA, Miller JP, Lee BCP, Conturo TE. Nor-
Hayward R, Bryant DRT, Payne JA, Levene MI, Whitelay A, mal brain in human newborns: apparent diffusion coeffi-
Dubowitz LMS, Dubowitz V. Clinical NMR imaging of the cient and diffusion anisotrophy measured by using diffu-
brain in children: normal and neurologic disease. AJNR Am sion tensor MR imaging. Radiology 1998; 209: 57–66
J Neuroradiol 1983; 4: 1013–1025 Parazzini C, Baldoli C, Scotti G,Triulzi F.Terminal zones of myeli-
Jolensz FA, Polak JF, Adams DF, Ruenzel PW. Myelinated and nation: MR evaluation of children aged 20–40 months.
nonmyelinated nerves: comparison of proton MR proper- AJNR Am J Neuroradiol 2002; 23: 1669–1673
ties. Radiology 1987; 164: 89–91 Pennock JM, Bydder GM, Dubowitz LMS, Johnson MA. Magnet-
Jones RA, Palasis S, Grattan-Smith JD. The evolution of the ap- ic resonance imaging of the brain in children. Magn Reson
parent diffusion coefficient in the pediatric brain at low Imaging 1986; 4: 1–9
and high diffusion weightings. J Magn Reson Imaging Rademacher J, Engelbrecht V, Bürgel U, Freund H-J, Zilles K.
2003; 18: 665–674 Measuring in vivo myelination of human white matter fiber
Keene MFL, Hewer EE. Some observations on myelination in tracts with magnetization transfer MR.Neuroimage 1999; 9:
the human nervous system. J Anat 1931; 6: 1–13 393–406
Korogi Y, Takahashi M, Sumi M, Hirai T, Sakamoto Y, Ikushima I, Robertson RL, Robson CD. Diffusion imaging in neonates. Neu-
Miyayama H. MR signal intensity of the perirolandic cortex roimaging Clin N Am 2002; 12: 55–70
in the neonate and infant. Neuroradiology 1996; 38: Ruoss K, Lövblad K, Schroth G, Moessinger AC, Fusch C. Brain
578–584 development (sulci and gyri) as assessed by early postnatal
Krier EL, Truwit CL. The normal and abnormal genu of the cor- MR imaging in preterm and term newborn infants. Neuro-
pus callosum: an evolutionary, embryologic, anatomic, and pediatrics 2001; 32: 69–74
MR analysis. AJNR Am J Neuroradiol 1996; 17: 1631–1641 Schmithorst VJ, Wilke M, Dardzinki BJ, Holland SK. Correlation
Lövblad KO, Schneider J, Ruoss K, Steinlin M, Fusch C, Schroth of white matter diffusivity and anisotrophy with age during
G.Isotropic apparent diffusion coefficient mapping of post- childhood and adolescence: a cross-sectional diffusion-
natal cerebral development. Neuroradiology 2003; 45: tensor MR imaging study. Radiology 2002; 222: 212–218
400–403 Schneider JFL, Il’yasov KA, Hennig J, Martin E. Fast quantitative
Martin E, Kikinis R, Zuerrer M, Boesch C, Briner J, Krewitz G, diffusion-tensor imaging of cerebral white matter from the
Kaelin P. Developmental stages of human brain: an MR neonatal period to adolescence. Neuroradiology 2004; 46:
study. J Comput Assist Tomogr 1988; 12: 917–922 258–266
Martin E,Boesch C,Zuerrer M,Kikinis R,Molinari L,Kealin P,Bolt- Shaw DWW, Weinberger E, Astley SJ, Tsuruda JS. Quantitative
shauser E,Duc G.MR imaging of brain maturation in normal comparison of conventional spin echo and fast spin echo
and developmentally handicapped children. J Comput As- during brain myelination.J Comput Assist Tomogr 1997; 21:
sist Tomogr 1990; 14: 685–692 867–871
Martin E, Krassnitzer S, Kealin P, Boesch Ch. MR imaging of the Sie LT, van der Knaap MS, van Wezel-Meijler G, Valk J. MRI as-
brainstem: normal postnatal development. Neuroradiolo- sessment of myelination of motor and sensory pathways in
gy 1991; 33: 391–395 the brain of preterm and term-born infants. Neuropedi-
McArdle CB, Richardson CJ, Nicholas DA, Mirfakhraee M, Hay- atrics 1997; 28: 97–105
den CK, Amparo EG. Developmental features of the neona- Sie LT, Barkhof F, Lafeber HN, Valk J, van der Knaap MS. Value of
tal brain: MR imaging. 1. Gray-white matter differentiation fluid-attenuated inversion recovery sequences in early MRI
and myelination. Radiology 1987; 162: 223–229 of the brain in neonates with a perinatal hypoxic-ischemic
Miller SP,Vigneron DB, Henry RG, Bohland MA, Ceppi-Cozzio C, encephalopathy. Eur Radiol 2000; 10: 1594–1601
Hoffman C, Newton N, Partridge JC, Ferriero DM, Barkovich Stricker T, Martin E, Boesch C. Development of the human cere-
AJ. Serial quantitative diffusion tensor MRI of the prema- bellum observed with high-field-strength MR imaging.
ture brain: development in newborns with and without in- Radiology 1990; 177: 431–435
jury. J Magn Reson Imaging 2002; 16: 621–632 Takeda K, Nomura Y, Sakuma H,Tagami T, Okuda Y, Nakagawa T.
Mukherjee P, Miller JH, Lee BCP, Almli CR, McKinstry RC. Normal MR assessment of normal brain development in neonates
brain maturation during childhood: developmental trends and infants: comparative study of T1-and diffusion-weight-
characterized with diffusion tensor MR imaging. Radiology ed images. J Comput Assist Tomogr 1997; 21: 1–7
2001; 221: 349–358 Tanner SF, Ramenghi LA, Ridgway JP, Berry E, Saysell MA, Mar-
Mukherjee P, Miller JH, Shimony JS, Philip JV, Nehra D, Snyder tinez D, Arthur RJ, Smith MA, Levene MI. Quantitative com-
AZ, Conturo TE, Neil JJ, McKinstry RC. Diffusion-tensor MR parison of intrabrain diffusion in adults and preterm and
imaging of gray and white matter development during term neonates and infants. AJR A J Roentgenol 2000; 174:
normal brain maturation. AJNR Am J Neuroradiol 2002; 23: 1643–1649
1445–1456 Toft PB, Leth H, Peitersen B, Lou HC, Thomson C. The apparent
Murakami JW, Weinberger E, Shaw DWW. Normal myelination diffusion coefficient of water in gray and white matter of
of the pediatric brain imaged with fluid-attenuated inver- the infant brain. J Comput Assist Tomogr 1996; 20: 1006–
sion-recovery (FLAIR) MR imaging. AJNR Am J Neuroradiol 1011
1999; 20: 1406–1411 Van Buchem MA, Steens SCA, Vrooman HA, Zwinderman AH,
Naidich TP, Grant JL, Altman N, Zimmerman RA, Birchansky SB, McGowan JC, Rassek M, Engelbrecht V. Global estimation of
Braffman B, Daniel JL.The developing cerebral surface. Pre- myelination in the developing brain on the basis of magne-
liminary report on the patterns of sulcal and gyral matura- tization transfer imaging: a preliminary study. AJNR Am J
tion – anatomy, ultrasound, and magnetic resonance imag- Neuroradiol 2001; 22: 762–766
ing. Pediatr Neuroradiol 1994; 4 201–239
912 References and Further Reading

van der Knaap MS, Valk J. MR imaging of the various stages of Bass NH,Witmer EJ, Dreifuss FE. A pedigree study of metachro-
normal myelination during the first year of life. Neuro- matic leukodystrophy. Neurology 1970; 20: 52–62
radiology 1990; 31: 459–470 Baumann N, Masson M, Carreau V, Lefevre M, Herschkowitz N,
van der Knaap MS, van Wezel-Meijler G, Barth PG, Barkhof F, Turpin JC. Adult forms of metachromatic leukodystrophy:
Ader H, Valk J. Normal gyration and sulcation in preterm clinical and biochemical approach. Dev Neurosci 1991; 13:
and term neonates: appearance on MR images. Radiology 211–215
1996; 200: 389–396 Baumann N, Turpin JC, Lefevre M, Colsch B. Motor and psycho-
Wimberger DM, Roberts TP, Barkovich AJ, Prayer LM, Moseley cognitive types in adult metachromatic leukodystrophy:
ME, Kucharczyk J. Identification of ‘premyelination’ by genotype-phenotype relationship? J Physiol 2002; 96: 301–
diffusion-weighted MRI. J Comput Assist Tomogr 1995; 19: 306
28–33 Berger J, Löschl B, Bernheimer H, Lugowska A,Tylki-Szymanska
Yakovlev PI, Lecours AR. The myelogenetic cycles of regional A, Gieselmann V, Molzer B. Occurrence, distribution, and
maturation of the brain.In:Minkowski A,ed.Regional devel- phenotype of arylsulfatase A mutations in patients with
opment of the brain in early life. Oxford: Blackwell, 1967: metachromatic leukodystrophy. Am J Med Genet 1997; 69:
pp 3–70 335–340
Zhai G, Lin W,Wilber KP, Gerig G, Gilmore JH. Comparison of re- Berger J, Gmach M, Mayr U, Molzer B, Bernheimer H. Coinci-
gional white matter diffusion in healthy neonates and dence of two novel arylsulfatase A alleles and mutation
adults performed with a 3.0T head-only MR imaging unit. 459+1G>A within a family with metachromatic leukodys-
Radiology 2003; 229: 673–681 trophy: molecular basis of phenotypic heterogeneity. Hum
Zilles K, Schleicher A, Langemann C, Amunts K, Morosan P, Mutat 1999; 13: 61–68
Palomero-Gallagher N, Schormann T, Mohlberg H, Bürgel U, Cengiz N, Özbenli T, Onar M, Yildiz L, Erta OB. Adult metachro-
Steinmetz H, Schlaug G, Roland PE. Quantitative analysis of matic leukodystrophy: three cases with normal nerve con-
sulci in the human cerebral cortex: development, regional duction velocities in a family. Acta Neurol Scand 2002; 105:
heterogeneity, gender difference, asymmetry, intersubject 454–457
variability and cortical architecture. Hum Brain Map 1997; Clarke JTR, Skomorowski MA, Chang PL. Marked clinical differ-
5: 218–221 ence between two sibs affected with juvenile metachro-
matic leukodystrophy. Am J Med Genet 1989; 33: 10–13
Comabella M,Waye JS, Raguer N, Eng B, Domínguez C, Navarro
C, Borras C, Krivit W, Montalban X. Late-onset metachro-
5 Lysosomes and Lysosomal Disorders matic leukodystrophy clinically presenting as isolated pe-
ripheral neuropathy: compound heterozygosity for the
Cavalli V, Corti M, Gruenberg J. Endocytosis and signaling cas- IVS2+1G A mutation and a newly identified missense mu-
cades: a close encounter. Febs Lett 2001; 498: 190–196 tation (Thr408Ile) in a Spanish family. Ann Neurol 2001; 50:
Dierks T, Schmidt B, Borissenko LV, Peng J, Preusser A, Mariap- 108–112
pan M, von Figura K. Multiple sulfatase deficiency is caused Consiglio A, Quattrini A, Martino S, Bensadoun JC, Dolcetta D,
by mutations in the gene encoding the human Ca-formyl- Trojani A, Benaglia G, Marchesini S, Cestari V, Oliverio A, Bor-
glycine generating enzyme. Cell 2003; 113: 435–444. dignon C, Naldini L. In vivo gene therapy of metachromatic
Gerasimenko JV, Gerasimenko OV, Petersen OH. Membrane re- leukodystrophy by lentiviral vectors: correction of neu-
pair: Ca2+-elicited lysosomal exocytosis. Curr Biol 2001 ;11: ropathology and protection against learning impairments
R971-R974 in affected mice. Nat Med 2001; 7: 310–316
Ghosh P, Dahms NM, Kornfeld S. Mannose 6-phosphate recep- Coulter-Mackie MB, Gagnier L.Two novel mutations in the aryl-
tors: new twists in the tale. Nat Rev 2003; 4: 202–212 sufatase A gene associated with juvenile (R39OQ) and
Haltia M. The Neuronal Ceroid-Lipofuscinoses. J Neuropathol adult onset (H397Y) metachromatic leukodystrophy. Hum
Exp Neurol 2003; 62: 1–13 Mutat 1998; suppl 1: S254–S256
Mullins C, Bonifacino JS. The molecular machinery for lyso- Dayan AD. Peripheral neuropathy of metachromatic leucodys-
some biogenesis. Bioessays 2001; 23: 333–343 trophy: observations on segmental demyelination and re-
Scriver CR, Beaudet AL, Sly WS,Valle D (eds).The metabolic and myelination and the intracellular distribution of sulphatide.
molecular basis of inherited disease. Lysosomal disorders. J Neurol Neurosurg Psychiatry 1967; 30: 311–318
McGraw-Hill, New York, 8th ed., 2001: pp 3371- 3896. Faerber EN, Melvin JJ, Smergel EM. MRI appearances of
metachromatic leukodystrophy. Pediatr Radiol 1999; 29:
669–672
6 Metachromatic Leukodystrophy Felice KJ, Gomez Lira M, Natowicz M, Grunnet ML,Tsongalis GJ,
Sima AAF, Kaplan RF. Adult-onset MLD: a gene mutation
Arbour LT, Silver K, Hechtman P, Treacy EP, Coulter-Mackie MB. with isolated polyneuropathy. Neurology 2000; 55: 1036–
Variable onset of metachromatic leukodystrophy in a Viet- 1039
namese family. Pediatr Neurol 2000; 23: 173–176 Francis GS, Bonni A, Shen N, Hechtman P,Yamut B, Carpenter S,
Aurebeck G,Osterberg K,Blaw M,Chou S,Nelson E.Electron mi- Karpati G, Chang PL. Metachromatic leukodystrophy: multi-
croscopic observations on metachromatic leukodystrophy. ple nonfunctional and pseudodeficiency alleles in a pedi-
Arch Neurol 1964; 11: 273–288 gree: problems with diagnosis and counseling. Ann Neurol
Austin JH. Metachromatic form of diffuse cerebral sclerosis; 1993; 34: 212–218
diagnosis during life by urine sediment examination. Neu- Gieselmann V, von Figura K. Advances in the molecular genet-
rology 1957; 7: 415–426 ics of metachromatic leukodystrophy. J Inherit Metab Dis
Barth ML, Fensom A, Harris A. The arylsulphatase A gene and 1990; 13: 560–571
molecular genetics of metachromatic leukodystrophy. Gieselmann V, Polten A, Kreysing J, von Figura K. Arylsulfatase
J Med Genet 1994; 31: 663–666 A pseudodeficiency: loss of a polyadenylation signal and
N-glycosylation site. Proc Natl Acad Sci 1989; 86: 9436–9440
References and Further Reading 913

Gieselmann V, Fluharty AL,Tonnesen T, von Figura K. Mutations Kapaun P, Dittmann RW, Granitzny B, Eickhoff W, Wulbrand H,
in the arylsulfatase A pseudodeficiency allele causing Neumaier-Probst E, Zander A, Kohlschüetter A. Slow pro-
metachromatic leukodystrophy. Am J Hum Genet 1991; 49: gression of juvenile metachromatic leukodystrophy 6 years
407–413 after bone marrow transplantation. J Child Neurol 1999;
Gieselmann V, Zlotogora J, Harris A,Wenger DA, Morris CP. Mol- 222–228
ecular genetics of metachromatic leukodystrophy. Hum Kappler J, von Figura K, Gieselmann V. Late-onset metachro-
Mutat 1994; 4: 233–242 matic leukodystrophy: molecular pathology in two sib-
Gieselmann V, Polten A, Kreysing J, von Figura K. Molecular lings. Ann Neurol 1992; 31: 256–261
genetics of metachromatic leukodystrophy. J Inherit Metab Kidd D, Nelson J, Jones F, Dusoir H, Wallace I, McKinstry S, Pat-
Dis 1994; 17: 500–509 terson V. Long-term stabilization after bone marrow trans-
Gieselmann V, Polten A, Kreysing J, von Figura K. Molecular ge- plantation in juvenile metachromatic leukodystrophy. Arch
netics of metachromatic leukodystrophy. J Inherit Metab Neurol 1998; 55: 98–99
Dis 1994; 17: 500–509 Kim TS, Kim IO, Kim WS, Choi YS, Lee JY, Kim OW, Yeon KM, Kim
Gieselmann V, Matzner U, Hess B, Lüllmann-Rauch R, Coenen R, KJ, Hwang YS. MR of childhood metachromatic leukodys-
Hartmann D, D’Hooge R, Dedeyn P, Nagels G. Metachromat- trophy. AJNR Am J Neuroradiol 1997; 18: 733–738
ic leukodystrophy: molecular genetics and an animal mod- Kreysing J, von Figura K, Gieselmann V. Structure of the arylsul-
el. J Inherit Metab Dis 1998; 21: 564–574 fatase A gene. Eur J Biochem 1990; 191: 627–631
Gieselmann V.Metachromatic leukodystrophy:recent research Krivit W, Shapiro E, Kennedy W, Lipton M, Lockman L, Smith S,
developments. J Child Neurol 2003; 18: 591–594 Gail Summers C, Wenger DA, Tsai MY, Ramsay NKC, Kersey
Gieselmann V, Franken S, Klein D, Mansson JE, Sandhoff R, Lüll- JH,Yao JK, Kaye E.Treatment of late infantile metachromat-
mann Rauch R, Hartmann D, Saravanan VPM, de Deyn PP, ic leukodystrophy by bone marrow transplantation. N Engl
D’Hooge R, van der Linden AM, Schaeren-Wiemers N. J Med 1990; 322: 28–32
Metachromatic leukodystrophy: consequences of sul- Krivit W, Shapiro E, Hoogerbrugge PM, Moser HW. State of the
phatide accumulation. Acta Paediatr Suppl 2003; 443: art review: bone marrow transplantation treatment for
74–79 storage diseases. Bone Marrow Transplant 1992; 10 (suppl
Ginsberg L, Gershfeld NL. Membrane bilayer instability and the 1): 87–96
pathogenesis of disorders of myelin. Neurosci Lett 1991; Krivit W, Lockman LA, Watkins PA, Hirsch J, Shapiro EG. The fu-
130: 133–136. ture for treatment by bone marrow transplantation for
Gort L, Coll MJ, Chabas A. Identification of 12 novel mutations adrenoleukodystrophy, metachromatic leukodystrophy,
and two new polymorphisms in the arylsulfatase A gene: globoid cell leukodystrophy and Hurler syndrome. J Inherit
haplotype and genotype-phenotype correlation studies in Metab Dis 1995; 18: 398–412
Spanish metachromatic leukodystrophy patients.Hum Mu- Landrieu, P, Blanche S, Vanier MT, Metral S, Husson B, Sandhoff
tat 1999; 14: 240–248 K, Fischer A. Bone marrow transplantation in metachromat-
Grégoire A, Périer O, Dustin P. Metachromatic leukodystrophy, ic leukodystrophy caused by saposin-B deficiency: a case
an electron microscopic study. J Neuropathol Exp Neurol report with a 3-year follow-up period. J Pediatr 1998; 133:
1966; 25: 617–636 129–132
Guffon N, Souillet G, Maire I, Dorche C, Mathieu M, Guibaud P. Leistner S,Young E, Meaney C,Winchester B. Pseudodeficiency
Juvenile metachromatic leukodystrophy: neurological out- of arylsulphatase A: strategy for clarification of genotype in
come two years after bone marrow transplantation.J Inher- families of subjects with low ASA activity and neurological
it Metab Dis 1995; 18: 159–161 symptoms. J Inherit Metab Dis 1995; 18: 710–716
Haltia T, Palo J, Haltia M, Icen A. Juvenile metachromatic McKhann GM Metachromatic leukodystrophy: clinical and
leukodystrophy: clinical, biochemical, and neuropathologic enzymatic parameters. Neuropediatrics 1984; 15: 4–10
studies in nine new cases. Arch Neurol 1980; 37: 42–46 Malm G, Ringdén O, Winiarski J, Gröndahl E, Uvebrant P, Eriks-
Harvey JS, Carey WF, Morris CP. Importance of the glycosylation son U, Håkansson H, Skjeldal O, Månsson JE. Clinical out-
and polyadenylation variants in metachromatic leukodys- come in four children with metachromatic leukodystrophy
trophy pseudodeficiency phenotype. Hum Mol Genet treated by bone marrow transplantation. Bone Marrow
1998; 7: 1215–1219 Transplant 1996; 17: 1003–1008
Holtschmidt H, Sandhoff K, Kwon HY, Harzer K, Nakano T, Suzu- Matzner U, Hartmann D, Lüllmmann-Rauch R, Coenen R,
ki K. Sulfatide activator protein. J Biol Chem 1991; 266: Rothert F, Månsson JE, Fredman P, D’Hooge R, De Deyn PP,
7556–7560 Gieselmann V. Bone marrow stem cell-based gene transfer
Inui K, Furukawa M, Nishimoto J, Okada S,Yabuuchi H. Metabo- in a mouse model for metachromatic leukodystrophy:
lism of cerebroside sulphate and subcellular distribution of effects on visceral and nervous system disease manifesta-
its metabolites in cultured skin fibroblasts derived from tions. Gene Ther 2002; 9: 53–63
controls, metachromatic leukodystrophy, globoid cell Mei Liu H. Ultrastructure of central nervous system lesions
leukodystrophy and Farber disease. J Inherit Metab Dis in metachromatic leukodystrophy with special reference
1987; 10: 293–296 to morphogenesis. J Neuropathol Exp Neurol 1968; 27:
Inui K, Furukawa M, Okada S, Yabuuchi H. Metabolism of cere- 624–644
broside sulfate and subcellular distribution of its metabo- Minamikawa-Tachino R, Maeda Y, Fujishiro I, Itoh K, Satake A,
lites in cultured skin fibroblasts from controls, metachro- Aoki S, Yamada H, Suzuki Y, Sakuraba H. Three-dimensional
matic leukodystrophy, and globoic cell leukodystrophy. brain visualization for metachromatic leukodystrophy.
J Clin Invest 1988; 81: 310–317 Brain Dev 1996; 18: 394–399
Jayakumar PN, Aroor SR, Jha RK, Arya BYT. Computed tomogra- Navarro C, Fernandez JM, Domínguez C, Fachal C, Albarez M.
phy (CT) in late infantile metachromatic leucodystrophy. Late juvenile metachromatic leukodystrophy treated with
Acta Neurol Scand 1989; 79: 23–26 bone marrow transplantation: a 4-year follow-up study.
Jervis GA. Infantile metachromatic leukodystrophy. J Neu- Neurology 1996; 46: 254–256
ropathol Exp Neurol 1960; 19: 323–340
914 References and Further Reading

Norman RM, Urich H, Tingey AH. Metachromatic leuco-en- Von Hirsch T,Pfeiffer J.Über histologische Methoden in der Dif-
cephalopathy: a form of lipidosis. Brain 1960; 83: 369–380 ferentialdiagnose von Leukodystrophien und Lipoidosen.
Penzien JM, Kappler J, Herschkowitz N, Schuknecht B, Arch Psychiatrie Z Neurol 1955; 194: 88–104
Keinekugel P, Propping P, Tonnesen T, Lou H, Moser H, Zierz Wrobe D, Henseler M. Huettler S, Pascual Pascual SI, Chabas A,
S, Conzelmann E, Gieselmann V. Compound heterozygosity Sandhoff K. A non-glycosylated and functionally deficient
for metachromatic leukodystrophy and arylsulfatase A mutant (N215H) of the sphingolidpid activator protein B
pseudodeficiency alleles is not associated with progressive (SAP-B) in a novel case of metachromatic leukodystrophy
neurological disease. Am J Hum Genet 1993; 52: 557–564 (MLD). J Inherit Metab Dis 2000; 23: 63–76
Polten A, Fluharty AL, Fluharty CB, Kappler J, von Figura K, Zafeiriou DI, Kontopoulos EE, Michelakakis HM, Anastasiou AL,
Gieselmann V. Molecular basis of different forms of Gombakis NP. Neurophysiology and MRI in late-infantile
metachromatic leukodystrophy. N Engl J Med 1991; 324: metachromatic leukodystrophy. Pediatr Neurol 1999; 21:
18–22 843–846
Pridjian G, Humbert J, Willis J, Shapira E. Presymptomatic late- Zhang XL, Rafi MA, DeGala G, Wenger DA. Insertion in the mR-
infantile metachromatic leukodystrophy treated with bone NA of a metachromatic leukodystrophy patient with sphin-
marrow transplantation. J Pediatr 1994; 125: 755–758 golipid activator protein-1 deficiency. Proc Natl Acad Sci
Qu Y, Shapira E, Desnick RJ. Metachromatic leukodystrophy: 1990; 87: 1426–1430
subtype genotype/phenotype correlations and identifica-
tion of novel missense mutations (P148L and P191T) caus-
ing the junvenile-onset disease. Mol Genet Metab 1999; 67:
206–212
7 Multiple Sulfatase Deficiency
Regis S,Filocamo M,Corsolini F,Caroli F,Keulemans JLM,Digge-
len van OP, Gatti R. An Asn >Lys substitution in saposin B in- Al-Moutaery KR, Choudhury AR, Hassanen MO. Cervical cord
volving a conserved amino acidic residue and leading to compression and severe hydrocephalus in a child with
the loss of the sinle N-glycosylation site in a patient with Saudi variant of multiple sulfatase deficiency. Acta Neu-
metachromatic leukodystrophy and normal arylsulphatase rochir (Wien) 1994; 131: 160–163
A activity. Eur J Hum Genet 1999; 7: 125–130 Aqeel AA, Ozand PT, Brismar J, Gascon GG, Brismar G, Nester M,
Regis S, Corsolini F, Stroppiano M, Cusano R, Filocamo M. Con- Sakati N. Saudi variant of multiple sulfatase deficiency.
tribution of arylsulfatase A mutations located on the same J Child Neurol 1992; 7 (suppl): S12–S21
allele to enzyme activity reduction and metachromatic Austin JH. Studies in metachromatic leukodystrophy. Arch
leukodystrophy severity. Hum Genet 2002; 110: 351–355 Neurol 1973; 28: 258–264
Reider-Grosswasser I, Bornstein N. CT and MRI in late-onset Basner R, von Figura K, Glössl J, Klein U, Kresse H, Mlekusch W.
metachromatic leukodystrophy. Acta Neurol Scand 1987; Multiple deficiency of mucopolysaccharide sulfatases in
75: 64–69 mucosulfatidosis. Pediatr Res 1979; 13: 1316–1318
Sangalli A, Taveggia C, Salviati A, Wrabetz L, Bordignon C, Sev- Bateman BJ, Philippart M, Isenberg SJ. Ocular features of multi-
erini GM. Transduced fibroblasts and metachromatic ple sulfatase deficiency and a new variant of metachromat-
leukodystrophy lymphocytes transfer arylsulfatase A to ic leukodystrophy. J Pediatr Ophthalmol Strabismus 1984;
myelinating Glia and deficient cells in vitro. Hum Gene Ther 21: 133–139
1998; 9: 2111–2119 Bharucha BA,Nalk G,Savliwala AS,Joshi RM,Kumta NB.Siblings
Schlote W, Harzer K, Christomanou H, Paton BC, Kustermann B, with the Austin variant of metachromatic leukodystrophy
Schmid B, Seeger J, Beudt U, Schuster I, Langenbeck U. multiple sulfatidosis. Indian J Pediatr 1984; 51: 477–480
Sphingolipid activator protein 1 deficiency in metachro- Burch M, Fesnom AH, Jackson M, Pitts-Tucker T, Congdon PJ.
matic leucodystrophy with normal arylsulphatase A activi- Multiple sulphatase deficiency presenting at birth. Clin
ty. A clinical, morphological, biochemical, and immunolog- Genet 1986; 30: 409–415
ical study. Eur J Pediatr 1991; 150: 584–591 Burk RD,Valle D,Thomas GH, Miller C, Moser A, Moser H, Rosen-
Scholz W. Klinische, pathologisch-anatomische und erbbiologi- baum KN. Early manifestations of multiple sulfatase defi-
sche Untersuchungen.Z Neurol Psychiatrie 1925;99:651–717 ciency. J Pediatr 1984; 104: 574–578
Shapiro EG, Lockman LA, Knopman D, Krivit W. Characteristics Conary JT, Hasilik A, von Figura K. Synthesis and stability of
of the dementia in late-onset metachromatic leukodystro- steroid sulfatase in fibroblasts from multiple sulfatase defi-
phy. Neurology 1994; 44: 662–665 ciency. Biol Chem Hoppe-Seyler 1988; 369: 297–302
Solders G, Celsing G, Hagenfeldt L, Ljungman P, Isberg B, Constantopoulos G. Multiple sulfatase deficiency with a novel
Ringdén O. Improved peripheral nerve conduction, EEG biochemical presentation. Eur J Pediatr 1988; 147: 634–638
and verbal IQ after bone marrow transplantation for adult Cosmo MP, Pepe S, Annuziata I, Newbold RF, Grompe M, Paren-
metachromatic leukodystrophy. Bone Marrow Transplant ti G, Ballabio A. The multiple sulfatase deficiency gene
1998; 22: 1119–1122 encodes and essential and limiting factor for the activity of
Stillman AE, Krivit W, Shapiro E, Lockman L, Latchaw RE. Serial sulfatases. Cell 2003; 113: 445–456
MR after bone marrow transplantation in two patients with Fedde K, Horwitz AL. Complementation of multiple sulfatase
metachromatic leukodystrophy. AJNR Am J Neuroradiol deficiency in somatic cell hybrids. Am J Hum Genet 1984;
1994; 15: 1929–1932 36: 623–633
Tylki-Szymañska A, Czartoryska B, Lugowska A. Practical sug- Guerra WF, Verity A, Fluharty AL, Nguyen HT, Philippart M. Mul-
gestions in diagnosing metachromatic leukodystrophy in tiple sulfatase deficiency: clinical, neuropathological, ultra-
probands and in testing family members. Eur Neurol 1998; structural and biochemical studies.J Neuropathol Exp Neu-
40: 67–70 rol 1990; 49: 406–423
Van Bogaert L, Dewulf A. Diffuse progressive leukodystrophy Harbord M, Buncic JR, Chuang SA, Skomorowski MA, Clarke
in the adult with production of metachromatic degenera- JTR. Multiple sulfatase deficiency with early severe retinal
tive products (Alzheimer-Baroncini). Arch Neurol Psychia- degeneration. J Child Neurol 1991; 6: 229–235
try 1939; 42: 1083–1097
References and Further Reading 915

Horwitz AL, Warshawsky L, King J, Burns G (1986) Rapid degra- Austin J, Suzuki K, Armstrong D, Brady R, Bachhawat BK,
dation of steroid sulfatase in multiple sulfatase deficiency. Schlenker J, Stumpf D. Studies in globoid (Krabbe) leuko-
Biochem Biophys Res Commun 1986; 135: 389–396 dystrophy (GLD). Arch Neurol 1970; 23: 502–512
Kepes JJ, Berry A, Zacharias DL. Multiple sulfatase deficiency: Bajaj NPS, Waldman A, Orrell R, Wood NW, Bhatia KP. Familial
bridge between neuronal storage diseases and leukodys- adult onset of Krabbe’s disease resembling hereditary
trophies. Pathology 1988; 20: 285–291 spastic paraplegia with normal neuroimaging. J Neurol
Landgrebe J, Dierks T, Schmidt B, von Figura K. The human Neurosurg Psychiatry 2002; 72: 635–638
SUMF1 gene, required for posttranslational sulfatase modi- Baker RH,Trautmann JC,Younge BR, Nelson KD, Zimmerman R.
fication, defines a new gene family, which is conserved Late juvenile-onset Krabbe’s disease. Ophthalmology 1990;
from pro- to eukaryotes. Gene 2003; 316: 47–56 97: 1176–1180
Loffeld A, Gray RGF, Green SH, Roper HP, Moss C. Mild ichthyosis Baram TZ, Goldman AM, Percy AK. Krabbe disease: specific MRI
in a 4-year-old boy with multiple sulphatase deficiency. Br J and CT findings. Neurology 1986; 36: 111–115
Dermatol 2002; 147: 353–355 Barone R, Brühl K, Stoeter P, Fiumara A, Pavone L, Beck M. Clini-
Macaulay RJB, Lowry NJ, Casey RE. Pathologic findings of mul- cal and neuroradiological findings in classic infantile and
tiple sulfatase deficiency reflect the pattern of enzyme de- late-onset globoid-cell leukodystrophy (Krabbe disease).
ficiencies. Pediatr Neurol 1998; 19: 372–376 Am J Med Genet 1996; 63: 209–217
Mancini GMS, Diggelen van OP, Huijmans JG, Stroink H, Coo de Bernal OG, Lenn N. Multiple cranial nerve enhancement in
RFM. Pitfalls in the diagnosis of multiple sulfatase deficien- early infantile Krabbe’s disease. Neurology 2000; 54: 2348–
cy. Neuropediatrics 2001; 32: 38–40 2349
Nevsimalova S, Elleder M, Smid F, Zemankova M. Multiple sul- Bernardi B, Fonda C, Franzoni E, Marchiani V, Della Guistina E,
phatase deficiency in homozygotic twins. J Inherit Metab Zimmerman RA. MRI and CT in Krabbe’s disease: case
Dis 1984; 7: 38–40 report. Neuroradiology 1994; 36: 477–479
Perlmutter-Cremer N, Libert J,Vamos E, Sphel M, Kiebaers I. Un- Bernardi GL, Herrera DG, Carson D, DeGasperi R, Gama Sosa
usual early manifestation of multiple sulfatase deficiency. MA, Kolodny EH, Trifiletti R. Adult-onset Krabbe’s disease in
Ann Radiol 1981; 24: 43–48 siblings with novel mutations in the galactocerebrosidase
Raynaud EJ, Escourolle R, Baumann N, Turpin JC, Dubois G, gene. Ann Neurol 1997; 41: 111–114
Malpuech G, Lagarde R. Metachromatic leukodystrophy. Bischoff A, Ulrich J. Peripheral neuropathy in globoid cell
Arch Neurol 1975; 32: 834–838 leukodystrophy (Krabbe’s disease). Ultrastructural and his-
Rommerskirch W, von Figura K. Multiple sulfatase deficiency: tochemical findings. Brain 1969; 92: 861–870
catalytically inactive sulfatases are expressd from retrovi- Biswas S, Levine SM. Substrate-reduction therapy enhances
rally introduced sulfatase cDNAs. Proc Natl Acad Sci 1992; the benefits of bone marrow transplantation in young
89: 2561–2565 mice with globoid cell leukodystrophy. Pediatr Res 2002;
Schmidt B, Selmer T, Ingendoh A, von Figura K. A novel amino 51: 40–47
acid modification in sulfatases that is defective in multiple Cavanagh N, Kendall B. High density on computed tomogra-
sulfatase deficiency. Cell 1995; 82: 271–278 phy in infantile Krabbe’s disease: a case report. Dev Med
Soong BW, Casamassima AC, Fink JK, Constantopoulos G, Hor- Child Neurol 1986; 28: 799–802
witz AL. Multiple sulfatase deficiency. Neurology 1988; 38: Choi S, Enzmann DR. Infantile Krabbe disease: complementary
1273–1275 CT and MR findings. AJNR Am J Neuroradiol 1993; 14:
Tanaka A, Hirabayashi M, Ishii M,Yamaoka S. Complementation 1164–1166
studies with clinical and biochemical characterizations of a Demaerel Ph,Wilms G,Verdru P, Carton H, Baert AL.MR findings
new variant of multiple sulphatase deficiency. J Inherit in globoid cell leukodystrophy. Neuroradiology 1990; 32:
Metab Dis 1987; 10: 103–110 520–522
Vamos E, Lubairs I, Bousard N, Liberi J, Pirimutter N. Multiple De Stefano N, Dotti MT,Mortilla M, Pappagallo E,Luzi P,Rafi MA,
sulphatase deficiency with early onset. J Inherit Metab Formichi P, Inzitari D, Wenger DA, Federico A. Evidence of
1981; 4: 103–104 diffuse brain pathology and unspecific genetic characteri-
Waheed A, Hasilik A, von Figura K. Enhanced breakdown of zation in a patient with an atypical form of adult-onset
arylsulfatase A in multiple sulfatase deficiency. Eur J Krabbe disease. J Neurol 2000; 247: 226–228
Biochem 1982; 123: 317–321 Ellis WG, Schneider EL, McCulloch JR, Suzuki K, Epstein CJ. Fetal
globoid cell leukodystrophy (Krabbe disease). Pathological
and biochemical examination. Arch Neurol 1973; 29:
253–257
8 Globoid Cell Leukodystrophy Epstein MA, Zimmerman RA, Rorke LB, Sladky JT. Late-onset
globoid cell leukodystrophy mimicking an infiltrating
Andrews JM, Cancilla PA, Grippo J, Menkes JH. Globoid cell glioma. Pediatr Radiol 1991; 21: 131–132
leukodystrophy (Krabbe’s disease): morphological and bio- Eto Y, Suzuki K. Brain spingoglycolipids in Krabbe’s globoid cell
chemical studies. Neurology 1971; 21: 337–352 leucodystrophy. J Neurochem 1971; 18: 503–511
Arvidsson J, Hagberg B, Mansson J-E, Svennerholm L. Late Eto Y, Suzuki K, Suzuki K. Globoid cell leukodystrophy (Krabbe’s
onset globoid cell leukodystrophy (Krabbe’s disease) – a disease): isolation of myelin with normal glycolipid compo-
Swedish case with 15 years of follow-up. Acta Paediatr sition. J Lipid Res 1970; 11: 473–479
1995; 84: 218–221 Farina L, Bizzi A, Fiocchiaro G, Pareyson D, Shghirlanzoni A,
Austin J. Studies in globoid (Krabbe) leukodystrophy. Arch Bertagnolio B, Savoiardo M, Naidu S, Wenger DA. MR imag-
Neurol 1963; 9: 207–231 ing and proton MR spectroscopy in adult Krabbe disease.
Austin J. Studies in globoid (Krabbe) leukodystrophy. J Neu- AJNR Am J Neuroradiol 2000; 21: 1478–1482
rochem 1963; 10: 921–930 Farley TJ, Ketonen LM, Bodensteiner JB, Wang DD. Serial MRI
and CT findings in infantile Krabbe disease. Pediatr Neurol
1992; 8: 455–458
916 References and Further Reading

Feanny SJ, Chuang SH, Becker LE, Clarke JTR. Intracerebral Jones BV, Barron TF, Towfighi J. Optic nerve enlargement in
paraventricular hyperdensities: a new CT sign in Krabbe Krabbe’s disease. AJNR Am J Neuroradiol 1999 20: 1228–
globoid cell leukodystrophy. J Inherit Metab Dis 1987; 10: 1231
24–27 Kapoor R, McDonald WI, Crockard A, Moseley IF. Clinical onset
Finelli DA, Tarr RW, Sawyer RN, Horwitz SJ. Deceptively normal and MRI features of Krabbe’s disease in adolescence. J Neu-
MR in early infantile Krabbe disease. AJNR Am J Neuroradi- rol Neurosurg Psychiatry 1992; 55: 331–332
ol 1994; 15: 167–171 Kobayashi T, Shinoda H, Goto I, Yamanaka T, Suzuki Y. Globoid
Fu L, Inui K, Nishigaki T,Tatsumi N,Tsukamoto H, Kokubu C, Mu- cell leukodystrophy is a generalized galactosylsphingosine
ramatsu T, Okada S. Molecular heterogeneity of Krabbe dis- (psychosine) storage disease. Biochem Biophys Res Com-
ease. J Inherit Metab Dis 1999; 22: 155–162 mun 1987; 144: 41–46
Giri S, Jatana M, Rattan R, Won J-S, Singh I, Sing AK. Galactosyl- Kobayashi T,Goto I,Yamanaka T,Suzuki Y,Nakano T,Suzuki K.In-
sphingosine (psychosine) –induced expression of cyto- fantile and fetal globoid cell leukodystrophy: analysis of
kine-mediated inducible nitric oxide synthases via AP-1 galactosylceramide and galactosylsphingosine. Ann Neu-
and C/EBP: implications for Krabbe disease. FASEB J 2002; rol 1988; 24: 517–522
16: 661–672 Kolodny EH, Raghavan S, Krivit W. Late-onset Krabbe disease
Given CA 2nd, Santos CC, Durden DD. Intracranial and spinal (globoid cell leukodystrophy): clinical and biochemical fea-
MR imaging findings associated with Krabbe’s disease : tures of 15 cases. Dev Neurosci 1991; 13: 232–239
case report. AJNR Am J Neuroradiol 2001; 22: 1782–1875 Krabbe K. A new familial, infantile form of diffuse brain sclero-
Goebel HH, Harzer K, Ernst JP, Bohl J, Klein H. Late-onset sis. Brain 1916; 39: 74–114
globoid cell leukodystrophy:unusual ultrastructural patho- Krivit W, Lockman LA, Watkins PA, Hirsch J, Shapiro EG. The fu-
logy and subtotal b-galactocerebrosidase deficiency. ture for treatment by bone marrow transplantation for
J Child Neurol 1990; 5: 299–307 adrenoleukodystrophy, metachromatic leukodystrophy,
Goebel HH, Kimura S, Harzer K, Klein H. Ultrastructural pathol- globoid cell leukodystrophy and Hurler syndrome. J Inherit
ogy of eccrine sweat gland epithelial cells in globoid cell Metab Dis 1995; 18: 398–412
leukodystrophy. J Child Neurol 1993; 8: 171–174 Krivit W, Shapiro EG, Peters C, Wagner JE, Cornu G, Kurtzberg J,
Grewal RP, Petronas N, Barton NW. Late onset globoid cell Wenger DA, Kolodny EH, Vanier MT, Loes DJ, Dusenbery K,
leukodystrophy. J Neurol Neurosurg Psychiatry 1991; 54: Locman LA. Hematopoietic stem-cell transplantation in
1011–1012 globoid-cell leukodystrophy. N Engl J Med 1998; 338:
Hagberg B. Krabbe’s disease: clinical presentation of neuro- 1119–1126
logical variants. Neuropediatrics 1984; 15: 11–15 Krivit W, Peters C, Shapiro EG. Bone marrow transplantation as
Hagberg B, Kollberg H, Sourander P, Akesson HO. Infantile effective treatment of central nervous system disease in
globoid cell leucodystrophy. Neuropediatrics 1969; 1: 74– globoid cell leukodystrophy, metachromatic leukodystro-
88 phy, adrenoleukodystrophy, mannosidosis, fucosidosis, as-
Harzer K, Knoblich R, Rolfs A, Bauer P, Eggers J. Residual galac- partylglucosaminuria,Hurler,Maroteaux-Lamy,and Sly syn-
tosylsphingosine (psychosine) b-galactosidase activities dromes, and Gaucher disease type III. Curr Opin Neurol
and associated GALC mutations in late and very late onset 1999; 12: 167–176
Krabbe disease. Clin Chim Acta 2002; 317: 77–84 Kurokawa T, Chen YJ, Nagata M. Late infantile Krabbe leukody-
Henderson RD, MacMillan JC, Bradfield JM. Adult onset Krabbe strophy: MRI and evoked potentials in a Japanese girl. Neu-
disease may mimic motor neurone disease. J Clin Neurosci ropediatrics 1987; 18: 182–183
2003; 10: 638–639 Kwan E, Drace J, Enzmann D. Specific CT findings in Krabbe dis-
Hittmair K,Wimberger D,Wiesbauer P, Zehetmayer M, Budka H. ease. AJNR Am J Neuroradiol 1984; 5: 453–458
Early infantile form of Krabbe disease with optic hypertro- LeVine SM, Pedchenko TV, Bronshteyn IG, Pinson DM. L-cy-
phy: serial MR examinations and autopsy correlation. AJNR closerine slows the clinical and pathological course in mice
Am J Neuroradiol 1994; 15: 1454–1458 with globoid cell leukodystrophy (twitcher mice). J Neu-
Ida H, Rennert OM, Watabe K, Eto Y, Maekawa K. Pathological rosci Res 2000; 60: 231–236
and biochemical studies of fetal Krabbe disease. Brain Dev Liu HM. Ultrastructure of globoid leukodystrophy (Krabbe’s
1994; 16: 480–4 disease) with reference to the origin of globoid cells. J Neu-
Ieshima A, Eda I, Matsui A, Yoshino K, Takashima S, Takeshita K. ropathol Exp Neurol 1970; 29: 441–462
Computed tomography in Krabbe’s disease: comparison Loes DJ, Peters C, Krivit W. Globoid cell leukodystrophy: distin-
with neuropathology. Neuroradiology 1983; 25: 323–327 guishing early-onset from late-onset disease using a brain
Igisu H, Nakamura M. Inhibition of cytochrome C oxidase by MR imaging scoring method. AJNR Am J Neuroradiol 1999;
psychosine (galactosylsphingosine). Biochem Biophys Res 20: 316–323
Commun1986; 137: 323–327 Loonen MCB, van Diggelen OP, Janse HC, Kleijer WJ, Arts WFM.
Itoh M, Hayashi M, Fujioka Y, Nagashima K, Morimatsu Y, Mat- Late-onset globoid cell leucodystrophy (Krabbe’s disease):
suyama H. Immunohistological study of globoid cell clinical and genetic delineation of two forms and their rela-
leukodystrophy. Brain Dev 2002; 24: 284–290 tion to the early-infantile form. Neuropediatrics 1985; 16:
Jardim LB, Giugliani R, Fensom AH.Thalamic and basal ganglia 137–142
hyperdensities – a CT marker for globoid cell leukodystro- Lyon G, Hagberg B, Evrard Ph, Allaire C, Pavone L, Vanier M.
phy? Neuropediatrics 1992; 23: 30–31 Symptomatology of late onset Krabbe’s leukodystrophy:
Jardim LB, Giugliani R, Pires RF, Haussen S, Burin MG, .Rafi MA, the European experience. Dev Neurosci 1991; 13: 240–244
Wenger DA.Protracted course of Krabbe disease in an adult Malone MJ, Szöke MC, Looney GL. Globoid leukodystrophy: I.
patient bearing a novel mutation. Arch Neurol 1999; 56: Clinical and enzymatic studies. Arch Neurol 1975; 32:
1014–1017 606–612
Jatana M, Giri S, Singh AK. Apoptotic positive cells in Krabbe Marks HG, Scavina MT, Kolodny EH, Palmieri M, Childs J. Krabbe
brain and induction of apoptosis in rat C6 glial cells by psy- disease presenting as a pheripheral neuropathy. Muscle
chosine. Neurosci Lett 2002; 330: 183–187 Nerve 1997; 20: 1024–1028
References and Further Reading 917

Matsuda J, Vanier MT, Saito Y, Tohyama J, Suzuki K, Suzuki K. A Svennerholm L,Vanier MT,Mansson JE.Krabbe disease:a galac-
mutation in the saposin A domain of the sphingolipid acti- tosylsphingosine (psychosine) lipidosis. J Lipid Res 1980;
vator protein (prosaposin) gene results in a late-onset, 21: 53–64
chronic form of globoid cell leukodystrophy in the mouse. Tada K,Taniike M,Tsukamoto H, Inui K, Okada S. Serial magnet-
Hum Mol Genet 2001; 10: 1191–1199 ic resonance imaging studies in a case of late onset globoid
Matsumoto R, Oka N, Nagahama Y, Akiguchi I, Kimura J. Periph- cell leukodystrophy. Neuropediatrics 1992; 23: 306–309
eral neuropathy in late-onset Krabbe’s disease: histochem- Taniike M, Mohri I, Eguchi N, Irikura D, Urade Y, Okada S, Suzuki
ical and ultrastructural findings. Acta Neuropathol (Berl) K. An opoptotic depletion of oligodendrocytes in the
1996; 92: 635–639 twitcher, a murine model of globoid cell leukodystrophy.
McKelvie P, Vine P, Hopkins I, Poulos A. A case of Krabbe’s J Neuropathol Exp Neurol 1999; 58: 644–653
leukodystrophy without globoid cells. Pathology 1990; 22: Tullu MS, Muranjan MN, Kondurkar PP, Bharucha BA. Krabbe
235–238 disease – clinical profile. Indian Pediatr 2000; 37: 939–946
Menkes JH, Duncan C, Moossy J. Molecular composition of the Turazzini M, Beltramello A, Bassi R, Del Colle R, Silvestri M. Adult
major glycolipids in globoid cell leukodystrophy. Neurolo- onset Krabbe’s leukodystrophy: a report of 2 cases. Acta
gy 1966; 16: 381–393 Neurol Scand 1997; 96: 413–415
Mitsuo K, Kobayashi T, Shinnoh N, Goto I. Biosynthesis of galac- Vanhanen L-L, Raininko R, Santavuori P. Early differential diag-
tosylsphingosine (psychosine) in the Twitcher mouse. Neu- nosis of infantile neuronal ceroid lipofuscinosis, Rett Syn-
rochem Res 1989; 14: 899–903 drome, and Krabbe disease by CT and MR. AJNR Am J Neu-
Olsson Y, Sourander P, Svennerholm L. Experimental studies on roradiol 1994; 15: 1443–1453
the pathogenesis of leucodystrophies.I.The effect of intrac- Vasconcelles E, Smith M. MRI nerve root enhancement in
erebrally injected sphingolipids in the rat’s brain. Acta Neu- Krabbe disease. Pediatr Neurol 1998; 19: 151–152
ropathol (Berl) 1966; 6: 153–163 Verdru P, Lammens M, Dom R, Van Elsen A. Globoid cell
Percy AK, Odrezin GT, Knowles PD, Rouah E, Armstrong DD. leukodystrophy: a family with both late-infantile and adult
Globoid cell leukodystrophy: comparison of neuropatholo- type. Neurology 1991; 41: 1382–1384
gy with magnetic resonance imaging. Acta Neuropathol Wenger DA, Louie E. Pseudodeficiencies of arylsulfatase A and
(Berl) 1994; 88: 26–32 galactocerebrosidase activities. Dev Neurosci 1991; 13:
Phelps M, Aicardi J,Vanier MT. Late onset Krabbe’s leukodystro- 216–221
phy: a report of four cases. J Neurol Neurosurg Psychiatry Wenger DA, Rafi MA, Luzi P. Molecular genetics of Krabbe dis-
1991; 54: 293–296 ease (globoid cell leukodystrophy): diagnosis and clinical
Sabatelli M,Quaranta L,Madia F,Lippi G,Conte A,Lo Monaco M, implications. Hum Mutat 1997; 10: 268–279
Di Trapani G, Rafi MA, Wenger DA, Vaccaro AM, Tonali P. Pe- Wenger DA, Rafi MA, Luzi P, Datto P, Constantino-Ceccarini E.
ripheral neuropathy with hypomyelinating features in Krabbe disease: genetic aspects and progress toward ther-
adult-onset Krabbe’s disease. Neuromusc Disord 2002; 12: apy. Mol Genet Metab 2000; 70: 1–9
386–391 Zafeiriou DI, Michelakaki EM, Anastisiou AL, Gombakis NP,
Sasaki M, Sakuragawa N,Takashima S, Hanaoka S, Arima M. MRI Kontopoulos EE. Serial MRI and neuropsychological studies
and CT findings in Krabbe disease. Pediatr Neurol 1991; 7: in late-infantile Krabbe disease. Pediatr Neurol 1996; 15:
283–288 240–244
Satoh J-I, Tokumoto H, Kurohara K,Yukitaka M, Matsui M, Kuro- Zafeiriou DI, Anastasiou AL, Michelakaki EM, Augoustidou-
da Y, Yamamoto T, Ruruya H, Shinnoh N, Kobayashi T, Kukita Savvopoulou PA, Katzos GS, Kontopoulos EE. Early infantile
Y,Haysashi K.Adult-onset Krabbe disease with the homozy- Krabbe disease: deceptively normal magnetic resonance
gous T1853C mutation in the galacotocerebosidase gene. imaging and serial neurophysiological studies. Brain Dev
Neurology 1997; 49: 1392–1399 1997; 19: 488–491
Shen JS,Watabe K, Oshashi T, Eto Y. Intraventricular administra- Zarifi MK,Tzika AA, Astrakas LG, Poussaint TY, Anthony DC, Dar-
tion of recombinant adenovirus to neonatal twitcher ras BT. Magnetic resonance spectroscopy and magnetic
mouse leads to clinicopathological improvements. Gene resonance imaging findings in Krabbe’s disease. J Child
Ther 2001; 8: 1081–1087 Neurol 2001; 16: 522–526
Sourander P,Hansson HA,Olsson Y,Svennerholm L.Experimen-
tal studies on the pathogenesis of leucodystrophies. II. The
effect of sphingolipids on various cell types in cultures
from the nervous system. Acta Neuropathol (Berl) 1966; 6:
9 GM1 Gangliosidosis
231–242
Suzuki K. Twenty five years of the “psychosine” hypothesis: a Al-Essa M, Bakheet SM, Patay ZJ, Nounou RM, Ozand TP. Cere-
personal perspective of its history and present status. Neu- bral fluorine-18 labeled 2-fluoro-2-deoxyglucose positron
rochem Res 1998; 23: 251–259 emission tomography (FDG PET), MRI, and clinical observa-
Suzuki K. Globoid cell leukodystrophy (Krabbe’s disease): up- tions in a patient with infantile Gm1 gangliosidosis. Brain
date. J Child Neurol 2003; 18: 595–603 Dev 1999; 21: 559–562
Suzuki K, Grover WD. Krabbe’s leukodystrophy (globoid cell Beratis NG, Varvarigou-Frimas A, Beratis S, Sklower SL. Angio-
leukodystrophy). Arch Neurol 1970; 22: 385–396 keratoma corporis diffusum in GM1 gangliosidosis, type 1.
Suzuki K, Suzuki Y. Globoid cell leucodystrophy (Krabbe’s dis- Clin Genet 1989; 36: 59–64
ease): deficiency of galactocerebroside b-galactosidase. Cabral A, Portela R, Tasso T, Eusebio F, Moreira A, Marques dos
Proc Natl Acad Sci 1970; 66: 302–309 Santos H, Soares J, Moura-Nunes JF. A case of GM1 gan-
Suzuki Y, Suzuki K.The twitcher mouse: a model for Krabbe dis- gliosidosis type I. Ophthalmic Paediatr Genet 1989; 10:
ease and for experimental therapies. Brain Pathol 1995; 5: 63–67
249–258
918 References and Further Reading

Callahan JW. Molecular basis of GM1 gangliosidosis and Shen W-C, Tsai FJ, Tsai C-H. Myelination arrest demonstrated
Morquio disease, type B. Structure-function studies of lyso- using magnetic resonance imaging in a child with type
somal b-galactosidase and the non-lysosomal b-galactosi- GM1 gangliosidosis. J Formos Med Assoc 1998; 97: 296–299
dase-like protein. Biochim Biophys Acta 1999; 1455; 85–103 Silva CMD, Severini MH, Sopelsa A, Coelho JC, Zaha A, d’Azzo A,
Chen CY, Zimmerman RA, Lee CC, Chen FH, Yuh YS, Hsiao HS. Giugliani R. Six novel b-galactosidase gene mutations in
Neuroimaging findings in late infantile GM1 gangliosido- Brazilian patients with GM1-gangliosidosis. Hum Mutat
sis. AJNR Am J Neuroradiol 1998; 19: 1628–1630 1999; 13: 401–409
Folkerth RD, Alroy J, Bhan I, Kaye EM. Infantile Gm1 gangliosido- Suzuki K, Suzuki K, Chen GC. Morphological, histochemical and
sis: complete morphology and histochemistry of two au- biochemical studies on a case of systemic late infantile lipi-
topsied cases, with particular reference to delayed central dosis (generalized gangliosidosis). J Neuropathol Exp Neu-
nervous system myelination. Pediatr Dev Pathol 2000; 3: rol 1968; 27: 15–38
73–86 Suzuki K, Suzuki K, Kamoshita S. Chemical pathology of GM1
Gascon GG, Ozand PT, Erwin RE. GM1 Gangliosidosis type 2 in gangliosidosis (generalized gangliosidosis). J Neuropathol
two siblings. J Child Neurol 1992; 7: S41–S50 Exp Neurol 1969; 28: 25–73
Goebel HH. Morphology of the gangliosidoses. Neuropedi- Suzuki Y, Sakuraba H, Oshima A, Yoshida K, Shimmoto M,
atrics 1984; 15: 97–106 Takano T, Fukuhara Y. Clinical and molecular heterogeneity
Guazzi GC, D’Amore I, van Hoof F, Fruschelli C, Alessandrini C, in hereditary b-galactosidase deficiency. Dev Neurosci
Palmeri S, Federico A. Type 3 (chronic) GM1 gangliosidosis 1991; 13: 299–303
presenting as infanto-choreo-athetotic dementia, without Tanaka R, Momoi T,Yoshida A, Okumura M,Yamakura S,Takasa-
epilepsy, in three sisters. Neurology 1988; 38: 1124–1127 ki Y, Kiyomasu T, Yamanaka C. Type 3 GM1 gangliosidosis:
Hinek A, Zhang S, Smith AC, Callahan JW. Impaired elastic-fiber clinical and neurological findings in an 11-year-old girl.
assembly by fibroblasts from patients with either Morquio J Neurol 1995; 242: 299–303
B disease or infantile GM1-gangliosidosis is linked to defi- Tominaga L, Ogawa Y,Taniguchi M, Ohno K, Matsuda J, Oshima
ciency in the 67-kD spliced variant of b-galactosidase. Am J A, Suzuki Y, Nanba E. Galactonojirimycin derivatives restore
Hum Genet 2000; 67: 23–36 mutant human b-galactosidase activities expressed in fi-
Inui K, Namba R, Ihara Y, Nobukuni K, Taniike M, Midorikawa M, broblasts from enzyme-deficient knockout mouse. Brain
Tsukamoto H. A case of chronic GM1 gangliosidosis pre- Dev 2001; 23: 284–287
senting as dystonia: clinical and biochemical studies. J Neu- Urban Z, Boyd CD. Elastic-fiber pathologies: primary defects in
rol 1990; 237: 491–493 assembly – and secondary disorders in transport and deliv-
Kasama T, Taketomi T. Abnormalities of cerebral lipids in GM1- ery. Am J Hum Genet 2000; 67: 4–7
gangliosidoses, infantile, juvenile, and chronic type. Jpn J Uyama E,Terasaki T,Watanabe S,Naito M,Owada M,Araki S,An-
Exp Med 1986; 56: 1–11 do M.Type 3 GM1 gangliosidosis:characteristic MRI findings
Kaye EM, Alroy J, Raghavan SS, Schwarting GA, Adelman LS, correlated with dystonia. Acta Neurol Scand 1992; 86:
Runge V,Gelblum D,Johann G,Thalhammer DVM,Zuniga G. 609–615
Dysmyelinogenesis in animal model of GM1 gangliosidosis. van der Voorn JP, Kamphorst W, van der Knaap MS, Powers JM.
Pediatr Neurol 1992; 8: 255–261 The leukoencephalopathy of infantile GM1 gangliosidosis:
Kobayashi T, Suzuki K. Chronic GM1 gangliosidosis presenting oligodendrocytic loss and axonal dysfunction. Acta Neu-
as dystonia: II. Biochemical studies. Ann Neurol 1981; 9: ropathol (Berl) 2004; 107: 539–545
476–483 Walkley SU, Baker HJ, Rattazzi MC, Haskins ME,Wu JY. Neuroax-
Kobayashi O, Takashima S. Thalamic hyperdensity on CT in the onal dystrophy in neuronal storage disorders: evidence for
infantile GM1-gangliosidosis. Brain Dev 1994; 16: 472–474 major GABAergic neuron involvement. J Neurol Sci 1991;
Kohlschütter A. Clinical course of GM1 gangliosidoses. Neuro- 104: 1–8
pediatrics 1984; 15: 71–73 Wood PA, McBride MR, Baker HJ, Christian ST. Fluorescence po-
Lin H-C, Tsai F-J, Shen C-H, Peng C-T. Infantile form Gm1 gan- larization analysis, lipid composition, and Na+, K+-ATPase ki-
gliosidosis with dilated cardiomyopathy: a case report. netics of synaptosomal membranes in feline GM1 and GM2
Acta Paediatr 2000; 89: 880–883 gangliosidosis. J Neurochem 1985; 44: 947–956
Morrone A, Bardelli T, Donati MA, Giorgi M, di Rocco M, Gatti R, Yoshida K, Oshima A, Sakuraba H, Nakano T,Yanagisawa N, Inui
Parini R, Ricci R, Tadeucchi G, D’Azzo A, Zammarchi E. K, Okada S, Uyama E, Namba R, Kondo K, Iwasaki S,Takamiya
b-Galactosidase gene mutations affecting the lysosomal K, Suzuki Y. GM1 gangliosidosis in adults: clinical and mole-
enzyme and the elasin-binding protein in GM1-gangliosi- cular analysis of 16 Japanese patients. Ann Neurol 1992; 31:
dosis patients with cardiac involvement. Hum Mutat 2000; 328–332
15: 354–366
Nardocci N, Bertagnolio B, Rumi V, Combi M, Bardelli P, Angelini
L. Chronic GM1 gangliosidosis presenting as dystonia: clini-
cal and biochemical studies in a new case. Neuropediatrics
10 GM2 Gangliosidosis
1993; 24: 164–166
O’Brien JS, Storb R, Raff RF, Harding J, Appelbaum F, Morimoto Argov Z, Navon R. Clinical and genetic variations in the syn-
S, Kishimoto Y, Graham T, Ahern-Rindell A, O’Brien SL. Bone drome of adult GM2 gangliosidosis resulting from hex-
marrow transplantation in canine GM1 gangliosidosis. Clin osaminidase A deficiency. Ann Neurol 1984; 16: 14–20
Genet 1990; 38: 274–280 Bach G,Tomczak J, Risch N, Ekstein J.Tay-Sachs screening in the
Pampiglione G,Harden A.Neurophysiological investigations in Jewish Ashkenazi population: DNA testing is the preferred
GM1 and GM2 gangliosidoses. Neuropedatrics 1984; 15: procedure. Am J Med Genet 2001; 99: 70–75
74–84 Barnes D, Misra VP, Young EP, Thomas PK, Harding AE. An adult
Sandhoff K, Conzelmann E.The biochemical basis of gangliosi- onset hexosaminidase A deficiency syndrome with senso-
doses. Neuropediatrics 1984; 15: 85–92 ry neuropathy and internuclear ophthalmoplegia. J Neurol
Neurosurg Psychiatry 1991; 54: 1112–1113
References and Further Reading 919

Barness LA, Henry K, Kling P, Laxova R, Kaback M, Gilbert-Bar- Hund E, Grau A, Fogel W, Fosting M, Cantz M, Kunstermann-
ness E. A 7-year old white-male boy with progressive neu- Kuhn B, Harzer K, Navon R, Goebel HH, Meinck H-M.Progres-
rological deterioration. Am J Med Genet 1991; 40: 271–279 sive cerebellar ataxia, proximal neurogenic weakness and
Beck M, Sieber N, Goebel HH. Progressive cerebellar ataxia in ocular motor disturbances: hexosaminidase A deficiency
juvenile GM2-gangliosidosis type Sandhoff. Eur J Pediatr with late onset in four siblings.J Neurol Sci 1997; 145: 25–31
1998; 157: 866–867 Jeyakumar M, Butters TD, Cortina-Borja M, Hunnam V, Proia RL,
Brett EM, Ellis RB, Haas L, Ikonne JU, Lake BD, Patrick AD, Pery VH,Dwek RA,Platt FM.Delayed symptom onset and in-
Stephens R. Late onset GM2 gangliosidosis: clinical, patho- creased life expectancy in Sandhoff disease mice treated
logical, and biochemical studies on 8 patients. Arch Dis with N-butyldeoxynojirimycin. Proc Natl Acad Sci 1999; 96:
Child 1973; 48: 775–785 6388–6393
Brismar J, Brismar G, Coates R, Gascon G, Ozand P. Increased Jeyakumar M, Norflus F, Tifft CJ, Cortina-Borja M, Butters TD,
density of the thalamus on CT scans in patients with GM2 Proia RL, Perry VH, Dwek RA, Platt FM. Enhanced survival in
gangliosidosis. Am J Neurol 1990; 11: 125–130 Sandhoff disease mice receiving a combination of sub-
Çaliskan M, Özmen M, Beck M, Apak S.Thalamic hyperdensity – strate deprivation therapy and bone marrow transplanta-
is it a diagnostic marker for Sandhoff disease? Brain Dev tion. Blood 2001; 97: 327–329
1993; 15: 387–388 Johnson WG, Chutorian A, Miranda A. A new juvenile hex-
Chavany C, Jendoubi M. Biology and potential strategies for osaminidase deficiency disease presenting as cerebellar
the treatment of GM2 gangliosidosis. Mol Med Today 1998; ataxia: clinical and biochemical studies. Neurology 1977;
4: 158–165 27: 1012–1018
Chen B, Rigat B, Curry C, Mahuran DJ. Structure of the GM2A Karni A, Navon R, Sadeh M. Hexosaminidase A deficiency man-
gene: identification of an exon 2 nonsense mutation and a ifesting as spinal muscular atrophy of late onset. Ann Neu-
naturally occurring transcript with an in-frame deletion. rol 1988; 24: 451–453
Am J Hum Genet 1999; 65: 77–87 Koelfen W, Freund M, Jaschke W, Koenig S, Schultze C. GM2 gan-
Conzelmann E, Kytzia HJ, Navon R, Sandhoff K. Ganglioside gliosidosis (Sandhoff’s disease): two year follow-up by MRI.
GM2 N-acetyl-b-D-galactosaminidase activity in cultured Neuroradiology 1994; 36: 152–154
fibroblasts of late-infantile and adult GM2 gangliosidosis Kotagal S,Wenger DA, Alcala H, Gomez C, Horenstein S. AB vari-
patients and of healthy probands with low hexosaminidase ant GM2 gangliosidosis: cerebrospinal fluid and neu-
level. Am J Hum Genet 1983; 35: 900–913 ropathologic characteristics. Neurology 1986; 36: 438–440
Cordeiro P,Hechtman P,Kaplan F.The GM2 gangliosidoses data- Kroll RA, Pagel MA, Roman-Goldstein S, Barkovich AJ, D’Agosti-
bases: allelic variation at the HEXA, HEXB, and GM2A. Genet no AN, Neuwelt EA. White matter changes associated with
Med 2000; 2: 319–327 Feline GM2 gangliosidosis (Sandhoff disease): correlation of
Di Gregorio F, Ferrari G, Marini P, Siliprandi R, Gorio A.The influ- MR findings with pathologic and ultrastructural abnormal-
ence of gangiosidosis on neurite growth regeneration. ities. AJNR Am J Neuroradiol 1995; 16: 1912–1226
Neuropediatrics 1984; 15: 93–96 Lui Y, Wada R, Kawai H, Sango K, Deng C, Tai T, McDonald MP,
D’Souza G, McCann CL, Hedrick J, Fairley C, Nagel HL, Kushner Araujo K, Crawley JN, Bierfreund U, Sandhoff K, Suzuki K,
JD, Kessel R. Tay-Sachs disease carrier screening: a 21-year Prioa RL. A genetic model of substrate deprivation therapy
experience. Genet Test 2000; 4: 257–263 for a glycosphingolipid storage disorder. J Clin Invest 1999;
Federico A, Palmeri S, Malandrini A, Fabrizi G, Mondelli M, 103: 497–505
Guazzi GC.The clinical aspects of adult hexosaminidase de- Meek D, Wolfe LS, Andermann E, Andermann F. Juvenile pro-
ficiencies. Dev Neurosci 1991; 13: 280–287 gressive dystonia: a new phenotype of GM2 gangliosidosis.
Fukumizu M,Yoshikawa H,Takashima N, Kurokawa T.Tay-Sachs Ann Neurol 1984; 15: 348–352
disease: progression of changes on neuroimaging in four Mugikura S, Takahashi S, Higano S, Kurihara N, Kon K, Sakamo-
cases. Neuroradiology 1992; 34: 483–486 to K. MR findings in Tay-Sachs disease. J Comput Assist
Goebel HH,Stolte G,Kustermann-Kuhn B,Harzer K.B1 Variant of Tomogr 1996; 20: 551–555
GM2 gangliosidosis in a 12-year-old patient. Pediatr Res Nassogne MC, Commare MC, Lellouch-Tubiana A, Emond S,
1989; 25: 89–93 Zerah M, Caillaud C, Hertz-Pannier L, Saudubray JM. Unusu-
Gordon BA, Gordon KE, Hinton GG, Cadera W, Feleki V, Bayleran al presentation of GM2 gangliosidosis mimicking a brain
J, Hechtman P. Tay-Sachs disease: B1 variant. Pediatr Neurol stem tumor in a 3-year-old girl. AJNR Am J Neuroradiol
1988; 4: 54–57 2003; 24: 840–842
Gray RGF, Green A, Rabb L, Broadhead DM, Besley GTN. A case Navon R. Molecular and clinical heterogeneity of adult GM2
of the B1 variant of GM2-gangiosidosis. J Inherit Metab Dis gangliosidosis. Dev Neurosci 1991; 13: 295–298
1990; 13: 280–282 Navon R, Argov Z, Brand N, Sandbank U. Adult GM2 gangliosi-
Grosso S, Farnetani MA, Berardi R, Margollicci M, Galluzzi P, dosis in association with Tay-Sachs disease: a new pheno-
Vivarelli R, Morgese G, Balestri P. GM2 gangliosidosis variant type. Neurology 1981; 31: 1397–1401
B1. Neuroradiologic features. J Neurol 2003; 250: 17–21 Navon R, Argov Z, Frisch A. Hexosaminidase A deficiency in
Guidotti JE, Mignon A, Haase G, Caillaud C, McDonnell N, Kahn adults. Am J Med Genet 1986; 24: 179–196
A, Poenaru L. Adenoviral gene therapy of the Tay-Sachs dis- Neote K, Mahuran DJ, Gravel RA. Molecular genetics of b-hex-
ease in hexosaminedase A-deficient knock-out mice. Hum osaminidase deficiencies. Adv Neurol 1991; 56: 189–207
Mol Genet 1999; 8: 831–838 Neufeld EF. Natural history and inherited disorders of a lysoso-
Hittmair K,Wimberger D, Bernert G, Mallek R, Schindler EG. MRI mal enzyme a-hexosaminidase. J Biol Chem 1989; 264:
in a case of Sandhoff’s disease. Neuroradiology 1996; 38: 10927–10930
S178-S180 Norflus F, Tifft CJ, McDonald MP, Goldstein G, Crawley JN, Hoff-
mann A, Sandhoff K, Suzuki K, Proia RL. Bone marrow trans-
plantation prolongs life span and ameliorates neurologic
manifestations in Sandhoff disease mice. J Clin Invest 1998;
101: 1881–1888
920 References and Further Reading

Oya Y, Proia RL, Norflus F, Tifft CJ, Langaman C, Suzuki K. Distri- Walkley SU, Siegel DA, Dobrenis K. GM2 Gangliosidosis and
bution of enzyme-bearing cells in GM2 gangliosidosis mice: pyramidal neuron dendritogenesis. Neurochem Res 1995;
regionally specific pattern of cellular infiltration following 20: 1287–1299
bone marrow transplantation. Acta Neuropathol (Berl) Walkley SU, Siegel DA, Dobrenis K, Zervas M. GM2 gangliosido-
2000; 99: 161–168 sis as a regulator of pyramidal neuron dendritogenesis.Ann
Paw BH, Moskowitz SM, Uhrhammer N, Wright N, Kaback MM, NY Acad Sci 1998; 845: 188–199
Neufeld EF. Juvenile GM2 gangliosidosis caused by substi- Walkley SU, Zervas M, Wiseman S. Gangliosidosis as modula-
tution of histidine for arginine at position 499 or 504 of the tors of dendritogenesis in normal and storage disease-
a-subunit of b-hexosaminidase. J Biol Chem 1990; 265: affected pyramidal neurons. Cereb Cortex 2000; 10: 1028–
9452–9457 1037
Platt FM, Butters TD. New therapeutic prospects for the gly- Ward CP, Fensom AH, Green PM. Biallelic discrimination assays
cosphingolipid lysosomal storage diseases. Biochem Phar- for the three common Ashkenazi Jewish mutations and a
macol 1998; 56: 421–430 common non-Jewish mutation, in Tay-Sachs disease, using
Platt FM, Neises GR, Reinkensmeier G, Towsend MJ, Perry VH, fluorogenic taqman probes. Genet Test 2000; 4: 351–358
Proia RL, Winchester B, Dwek RA, Buters TD. Prevention of Willner JP, Grabowski GA, Gordon RE, Bender AN, Desnick RJ.
lysosomal storage in Tay-Sachs mice treated with N- Chronic GM2 gangliosidosis masquerading as atypical
butyldeoxynojirimycin. Science 1997; 276: 428–431 Friedreich ataxia: clinical, morphologic, and biochemical
Platt FM, Jeyakumar M, Andersson U, Priestman DA, Dewk RA, studies of nine cases. Neurology 1981; 31: 787–798
Butters TD.Inhibition of substrate synthesis as a strategy for Yoshikawa H, Yamada K, Sakuragawa N. MRI in the early stage
glycolipid lysosomal storage disease therapy. J Inherit of Tay-Sachs disease. Neuroradiology 1992; 34: 394–395
Metab Dis 2001; 24: 275–290 Yüksel A,Yalçinkaya C, I0lak C, Gündüz E, Seven M. Neuroimag-
Pullarkat RK, Reha H, Beratis NG. Accumulation of ganglioside ing findings in four patients with Sandhoff disease. Pediatr
GM2 in cerebrospinal fluid of a patient with the variant Neurol 1999; 21: 562–565
AB of infantile GM2 gangliosidosis. Pediatrics 1981; 68:
106–108
Rapin I, Suzuki K, Suzuki K, Valsamis MP. Adult (chronic) GM2
gangliosidosis: atypical spinocerebellar degeneration in a
11 Fabry Disease
Jewish sibship. Arch Neurol 1976; 33: 120–130
Rattazzi MC, Dobrenis K.Treatment of GM2 gangliosidosis: past Abe A, Gregory S, Lee L, Killen PD, Brady RO, Kulkarni A, Shay-
experiences, implications, and future prospects. Adv Genet man JA. Reduction of globotriasylceramide in Fabry dis-
2001; 44: 317–339 ease mice by substrate deprevation. J Clin Invest 2000; 105:
Rubin M, Karpati G,Wolfe LS, Carpenter S, Klavins MH, Mahuran 1563–1571
DJ. Adult onset motor neuropathy in the juvenile type of Altarescu G, Moore DF, Pursley R, Campia U, Goldstein S, Bryant
hexosaminidase A and B deficiency. J Neurol Sci 1988; 87: M, Panza JA, Schiffmann R. Enhanced endothelium-depen-
103–119 dent vasodilation in Fabry disease. Stroke 2001; 32: 1559–
Salaman MS, Clarcke JTR, Midroni G, Waxman MB. Peripheral 1562
and autonomic nervous system involvement in chronic Asano N, Ishii S, Kizu H, Ikeda H, Ikeda K, Kato A, Martin OR, Fan
GM2-gangliocidosis. J Inherit Metab Dis 2001; 24: 65–71 JQ. In vitro inhibition and intracellular enhancement of
Sandhoff K, Harzer K, Wässle W, Jatzkewitz H. Enzyme alter- lysosomal a-galactosidase A activity in Fabry lymphoblasts
ations and lipid storage in three variants of Tay-Sachs dis- by 1-deoxygalactonojirimycin and its derivatives. Eur J
ease. J Neurochem 1971; 8: 2469–2489 Biochem 2000 267: 4179–4186
Schepers U, Glombitza G, Lemm T, Hoffmann A, Chabas A, Ashley GA; Shabbeer J, Yasuda M, Eng CM, Desnick RJ. Fabry
Ozand P, Sandhoff K. Molecular analysis of a GM2-activator disease: twenty novel a-galactosidase A mutations causing
deficiency in two patients with GM2-gangliosidosis AB the classical phenotype. J Hum Genet 2001; 46: 192–196
variant. Am J Hum Genet 1996; 59: 1048–105 Baehner F, Kampmann C, Whybra C, Miebach E, Wiethoff CM,
Schnorf H, Gitzelmann R, Bosshard NU, Spycher M, Waespe W. Beck M. Enzyme replacement therapy in heterozygous
Early and severe sensory loss in three adult siblings with females with Fabry disease: results of a phase IIIB study.
hexosaminidase A and B deficiency (Sandhoff disease). J J Inherit Metab Dis 2003; 26: 617–627
Neurol Neurosurg Psychiatry 1995; 59: 520–523 Brantom MH, Schiffmann R, Sabnis SG, Murray GJ, Quirk JM,
Specola N,Vanier MT, Goutières F, Mikol J, Aicardi J.The juvenile Altarescu G, Goldfarb L, Brady RO,Balow JE,Austin HA, Kopp
and chronic forms of GM2 gangliosidosis: clinical and enzy- JB.Natural history of Fabry renal disease.Medicine 2002; 81:
matic heterogeneity. Neurology 1990; 40: 145–150 122–138
Streifler JY, Gornish M, Hadar H, Gadoth N. Brain imaging in DeVeber GA,Schwarting GA,Kolodny EH,Kowall NW.Fabry dis-
late-onset GM2 gangliosidosis. Neurology 1993; 43: 2055– ease: immunocytochemical characterization of neuronal
2058 involvement. Ann Neurol 1992; 31: 409–415
Suzuki K,Vanier MT.Biochemical and molecular aspects of late- Dütsch M,Marthol H,Stemper B,Brys M,Haendl T,Hilz MJ.Small
onset GM-2 gangiosidosis: B1 variant as a phenotype. Dev fiber dysfunction predominates in Fabry neuropathy. J Clin
Neurosci 1991; 13: 288–294 Neurophysiol 2002; 19: 575–586
Thomas PK, Young E, King RHM. Sandhoff disease mimicking Eng CM, Guffon N, Wilcox WR, Germain DP, Lee P, Waldek S,
adult-onset bulbospinal neuropathy. J Neurol Neurosurg Caplan L, Linthorst GE, Desnick RJ. Safety and efficacy of
Psychiatry 1989; 52: 1103–1106 recombinant human a-galactosidase a replacement thera-
Wada R, Tifft CJ, Proia RL. Microglial activation precedes acute py in Fabry’s disease. N Engl J Med 2001; 345: 9–16
neurodegenration in Sandhoff disease and is suppressed Feldt-Rasmussen U, Rasmussen AK, Mersebach H, Rosenberg
by bone marrow transplantation. Proc Nat Acad Sci 2000; KM, Hasholt L, Sorensen S. Fabry disease: a new challange in
97:10954–10959 endocrinology and metabolism? Eur J Endocrinol 2002;
146: 741–742
References and Further Reading 921

Filling-Katz MR, Merrick HF, Fink JK, Miles RB, Sokol J, Barton Nakao S, Takenaka T, Maeda M, Kodama C, Tanaka A, Tahara M,
NW. Carbamazepine in Fabry’s disease: effective analgesia Yoshida A, Kuriyama M, Hayashibe H, Sakuraba H,Tanaka H.
with dose-dependent exacerbation of autonomic dysfunc- An atypical variant of Fabry’s disease in men with left ven-
tion. Neurology 1989; 39: 598–600 tricular hypertrophy. N Engl J Med 1995; 333: 288–293
Frustazi A, Chimenti C, Ricci R, Natale L, Russo MA, Pieroni M, Nelis GF,Jacobs GJA.Anorexia,weight loss,and diarrhea as pre-
Eng CM, Desnick RJ. Improvement in cardiac function in the senting symptoms of angiokeratoma corporis diffusum
cardiac variant of Fabry’s disease with galactose-infusion (Fabry-Anderson’s disease). Dig Dis Sci 1989; 34: 1798–1800
therapy. N Engl J Med 2001; 345: 25–32 Pastores GM,Thadhani R.Enzyme-replacement therapy for An-
Gahl WA. New therapies for Fabry’s disease. N Engl J Med 2001; derson-Fabry disease. Lancet 2001; 358: 601–603
345: 55–57 Rahman AN, Lindenberg R. The neuropathology of hereditary
Grewal RP. Stroke in Fabry’s disease. J Neurol 1994; 241: dystopic lipidosis. Arch Neurol 1963; 9: 373–385
153–156 Schiffmann R, Kopp JB, Austin HA, Sabnis S, Moore DF,Weibel T,
Grewal RP, McLatchey SK. Cerebrovascular manifestations in a Balow J, Brady RO. Enzyme replacement therapy in Fabry
female carrier of Fabry’s disease. Acta Neurol Belg 1992; 92: disease. A randomized controlled trail. JAMA 2001; 285:
36–40 2743–2749
Hajioff D, Enever Y, Quiney R, Zuckerman J, Macdermot K, Takanashi J, Barkocich JA, Dillon WP, Sherr EH, Hart KA, Pack-
Mehta A. Hearing loss in Fabry patients: the effect of agalsi- man S. T1 hyperintensity in the pulvinar: key imaging fea-
dase alpha replacement therapy. J Inherit Metab Dis 2002; ture for diagnosis of Fabry disease. AJNR Am J Neuroradiol
26: 787–794 2003; 24: 916–921
Kaye EM, Kolodny EH, Logigian EL, Ullman MD. Nervous system Takenaka T, Murray GH, Qin G, Quirk JM, Oshima T, Qasba P,
involvement in Fabry’s disease: clinicopathological and Clark K,Kulkarni AB,Brady RO,Medin JA.Long-term enzyme
biochemical correlation. Ann Neurol 1988; 23: 505–509 correction and lipid reduction in mulitple organs of prima-
Kleijer WJ, Hussaarts-Odijk LM, Sachs ES, Jahoda MGJ, Niermei- ry and secondary transplanted Fabry mice receiving trans-
jer MF. Prenatal diagnosis of Fabry’s disease by direct analy- duced bone marrow cells. Proc Natl Acad Sci USA 2000; 97:
sis of chorionic villi. Prenat Diagn 1987; 7: 283–287 7515–7520
Kornreich R, Bishop DF, Desnick RJ. a-galactosidase A gene re- Whybra C, Kampmann C, Willers I, Davies J, Winchester B,
arrangements causing Fabry disease. J Biol Chem 1990; Kriegsmann J, Brühl K, Gal A, Bunge S, Beck M. Anderson-
265: 9319–9326 Fabry disease: clinical manifestations of disease in female
MacDermot KD,Holmes A,Miners AH.Anderson-Fabry disease: hetrozygotes. J Inherit Metab Dis 2001; 24: 715–724
clinical manifestations and impact of disease in a cohort of Wiedemann F, Breuning F, Beer M, Sandstede J, Turschner O,
60 obligate carrier females. J Med Genet 2001; 38: 769–775 Voelker W,Ertl G,Knoll A,Wanner C,Strotmann JM.Improve-
MacDermot KD. Anderson-Fabry disease: clinical manifesta- ment of cardiac function during enzyme replacement ther-
tions and impact of disease in a cohort of 98 hemizygous apy in patients with Fabry disease, a prospective strain rate
males. J Med Genet 2001; 37: 750–760 imaging study. Circulation 2003; 108: 1299–1301
Menzies DG, Campbell IW. Magnetic resonance in Fabry’s dis-
ease. J Neurol Neurosurg Psychiatry 1988; 51: 1240–1241
Mitsias P, Levine SR. Cerebrovascular complications of Fabry’s
disease. Ann Neurol 1996; 40: 8–17
12 Fucosidosis
Moore DF, Scott LTC, Gladwin MT, Altarescu G, Kaneski C, Suzu-
ki K, Pease-Fye M, Ferri R, Brady RO, Herscovitch P, Schiff- Darby JK, Willems PJ, Nakashima P, Johnsen J, Ferrell RE, Wijs-
mann R. Regional cerebral hyperperfusion and nitric oxide man EM, Gerhard DS, Dracopoli NC, Housman D, Henke J,
pathway dysregulation in Fabry disease. Reversal by en- Fowler ML, Shows TB, O’Brien JS, Cavalli-Sforza LL. Restric-
zyme replacement therapy. Circulation 2001; 104: 1506– tion analysis of the structural a-L-fucosidase gene and its
1512 linkage to fucosidosis. Am J Hum Genet 1988; 43: 749–755
Moore DF, Altarescu G, Ling GSF, Jeffries N, Frei KP, Weibel T, Galluzzi P, Rufa A, Balestri P, Cerase A, Federico A. MR brain
Charria-Ortiz G, Ferri R, Arai AE, Brady RO, Schiffmann R. Ele- imaging of fucosidosis type I. AJNR Am J Neuroradiol 2001;
vated cerebral blood flow velocities in Fabry disease with 22: 777–780
reversal after enzyme replacement. Stroke 2002; 33: Gordon BA, Gordon KE, Seo JC, Yang M, DiCioccio RA, O’Brien
525–431 JS. Fucosidosis with dystonia. Neuropediatrics 1995; 26:
Moore DF, Altarescu G, Barker WC, Patronas NJ, Herscovitch P, 325–327
Schiffmann R. White matter lesions in Fabry disease occur Inui K, Akagi M, Nishigaki T, Muramatsu T, Tsukamoto H, Okada
in ‘prior’ selectively hypometabolic and hyperperfused S. A case of chronic infantile type of fucosidosis: clinical and
brain regions. Brain Res Bull 2003; 62: 231–240 magnetic resonance imaging. Brain Dev 2000; 22: 47–49
Moore DF, Ye F, Schiffmann R, Butman JA. Increased signal in- Ismail EAR, Rudwan M, Shafik MK. Fucosidosis: immunoloigal
tensity in teh pulvinar on T1-weighted images:a pathogno- studies and chronological neuroradiological changes. Acta
monic MR imaging sign of Fabry disease. AJNR Am J Neuro- Paediatr 1999; 88: 224–227
radiol 2003; 24: 1096–1101 Kretz KA, Cripe D, Carson GS, Fukushima H, O’Brien JS.Structure
Morgan SH, Rudge P, Smith SJM, Bronstein AM, Kendall BE, Hol- and sequence of the human a-L-fucosidase gene and
ly E, Young EP, Crawfurd MA, Bannister R. The neurological pseudogene. Genomics 1992; 12: 276–280
complications of Anderson-Fabry disease (a-galactosidase Miano M, Lanino E, Gatti R, Morreale G, Fondelli P, Celle ME,
A deficiency): investigation of symptomatic and presymp- Stroppiano M, Crescenzi F, Dini G. Four-year follow-up of a
tomatic patients. QJM [N Ser] 1990; 75: 491–504 case of fucosidosis treated with unrelated donor bone
Moumdjian R,Tampieri D, Melanson D, Ethier R. Anderson-Fab- marrow transplantation. Bone Marrow Transplant 2001; 27:
ry disease: a case report with MR, CT and cerebral angiogra- 747–751
phy. AJNR Am J Neuroradiol 1989; 10: S69-S70
922 References and Further Reading

Michalski J-C, Klein A. Glycoprotein lysosomal storage disor- Kachur E, del Maestro R. Mucopolysaccharidoses and spinal
ders: a- and b-mannosidosis, fucosis and a-N-acetylgalac- cord compression: case report and review of the literature
tosaminidase deficiency. Biochim Biophys Acta 1999; 1455: with implications of bone marrow transplantation. Neuro-
69–84 surgery 2000; 47: 223–229
Ng Ying Kin NMK. Composition of the urinary glycoconjugates Kulkarni MV, Williams JC, Yeakley JW, Andrews JL, McArdle CB,
excreted by patient with type I and type II fucosidosis. Clin Narayana PA, Howell RR, Jonas AJ. Magnetic resonance
Chem 1987; 33: 44–47 imaging in the diagnosis of the cranio-cervical manifesta-
Provenzale JM, Barboriak DP, Sims K. Neuroradiologic findings tions of the mucopolysaccharidoses. Magn Reson Imaging
in fucosidosis, a rare lysosomal storage disease. AJNR Am J 1987; 5: 317–323
Neuroradiol 1995; 16: 809–813 Lee C, Dineen TE, Brack M, Kirsch JE, Runge VM. The mu-
Taylor RM, Farrow BRH, Stewart GJ, Healy PJ,Tiver K. Lysosomal copolysaccharidoses: characterization by cranial MR imag-
enzyme replacement in neural tissue by allogeneic bone ing. AJNR Am J Neuroradiol 1993; 14: 1285–1292
marrow transplantation following total lymphoid irradia- Levin TL, Berdon WE, Lachman RS, Anyane-Yeboa K. Ruzal-
tion in canine fucosidosis. Transplant Proc 1987; 9: 2730– Shapiro C, Roye DP. Lumbar gibbus in storage diseases and
2734 bone dysplasias. Pediatr Radiol 1997; 27: 289–294
Taylor RM, Farrow BRH, Stewart GJ. Amelioration of clinical dis- Martin JJ, Ceuterick C. The contribution of pathology to the
ease following bone marrow transplantation in fucosidase- study of storage disorders. Pathol Res Pract 1988; 183:
deficient dogs. Am J Med Genet 1992; 42: 628–632 375–385
Terespolsky D, Clarke JTR, Blaser SI. Evolution of the neu- Murata R, Nakajima S,Tanaka A, Miyagi N, Matsuoka O, Kogame
roimaging changes in fucosidosis type II. J Inherit Metab S, Inoue Y. MR imaging of the brain in patients with mu-
Dis 1996; 19: 775–781 copolysaccharidosis. AJNR Am J Neuroradiol 1989; 10:
Tiberio G. Filocamo M, Gatti R, Durand P. Mutations in fucosido- 1165–1170
sis gene: a review. Acta Genet Med Gemellol 1995; 44: Nelson J, Shields D, Mulholland HC. Cardiovascular studies in
223–232 the mucopolysaccharidoses. J Med Genet 1990; 27: 94–100
Vellodi A, Cragg H, Winchester B, Young E, Downie CJC, Hoare Perretti A, Petrillo A, Pelosi L, Balbi P, Parenti G, Riemma A,
RD, Stockes R, Banerjee GK. Allogenic bone marrow trans- Strisciuglio P. Detection of early abnormalities in the mu-
plantation for fucosidosis. Bone Marrow Transplant 1995; copolysaccharidoses by the use of visual and brainstem au-
15: 153–158 ditory evoked potentials. Neuropediatrics 1989; 21: 83–86
Willems PJ, Gatti R, Darby JK, Romeo G, Durand P, Dumon JE, Purpura DP, Suzuki K. Distortion of neuronal geometry and for-
O’Brien JS. Fucosidosis revisited: a review of 77 patients. Am mation of aberrant synapses in neuronal storage disease.
J Hum Genet 1991; 38: 111–131 Brain Res 1976; 116: 1–21
Willems PJ, Seo H-C, Coucke P, Tonorenzi R, O’Brien JS. Spec- Schiffmann R, Brady RO. New prospects for the treatment of
trum of mutations in fucosidosis. Eur J Hum Genet 1999; 7: lysosomal storage diseases. Drugs 2002; 62: 733–742
60–67 Scott HS, Bunge S, Gal A, Clarke LA, Morris CP, Hopwood JJ. Mu-
tation update. Molecular genetics of mucopolysaccharido-
sis type I: diagnostic, clinical, and biological implications.
Hum Mutat 1995; 6: 288–302
13 Mucoplysaccharidoses Seto T, Kono K, Morimoto K, Inoue Y, Shintaku H, Hattori H, Mat-
General suoka O, Yamano T, Tanaka A. Brain magnetic resonance
imaging in 23 patients with mucopolysaccharidoses and
Albano LMJ, Sugayama SSMM, Bertola DR, Andrade CEF, Uta- the effect of bone marrow transplantation. Ann Neurol
gawa CY, Puppi F, Nader HB, Toma L, Coelho J, Leistner S, 2001; 50: 79–92
Burin M, Giugliani R, Kim CA. Clinical and laboratorial study Shih S-L, Lee Y-J, Lin S-P, Sheu C-Y, Blickman JG. Airway changes
of 19 cases of mucopolysaccharidoses. Rev Hosp Clin Fac in children with mucopolysaccharidoses. CT evaluation.
Med S Paulo 2000; 55: 213–218 Acta Radiol 2002; 43: 40–43
Barone R, Parano E,Trifiletti RR, Fiumara A, Pavone P.White mat- Sly WS, Vogler C. Brain-directed gene therapy for lysosomal
ter changes mimicking a leukodystrophy in a patient with storage disease: going well beyond the blood-brain barrier.
mucopolysaccharidosis: characterization by MRI. J Neurol Proc Natl Acad Sci 2002; 9: 5760–5762
Sci 2002; 195: 171–175 Stone JE. Urine analysis in the diagnosis of mucopolysaccha-
Dekaban AS, Constantopoulos G. Mucopolysaccharidosis ride disorders. Ann Clin Biochem 1998; 35: 207–225
types I, II, IIIA and V. Pathological and biochemical abnor- Wraith JE. The mucopolysaccharidoses: a clinical review and
malities in the neural and mesenchymal elements of the guide to management. Arch Dis Child 1995; 72: 263–267
brain. Acta Neuropathol (Berl) 1977; 39: 1–7
Ero Y, Ohashi T. Gene therapy / cell therapy for lysosomal stor-
age disease. J Inherit Metab Dis 2000; 23: 293–298 Hurler Disease
Fensom AH, Benson PF. Recent advances in the prenatal diag-
nosis of the mucopolysaccharidoses. Prenat Diagn 1994; Afifi AK, Sato Y,Waziri MH, Bell WE. Computed tomography and
14: 1–12 magnetic resonance imaging of the brain in Hurler’s dis-
Gieselmann V. Lysosomal storage diseases. Biochim Biophys ease. J Child Neurol 1990; 5: 235–241
Acta 1995; 1270: 103–136 Barone R, Parano E,Trifiletti RR, Fiumara A, Pavone P.White mat-
Herrick IA, Rhine EJ. The mucopolysaccharidoses and anaes- ter changes mimicking a leukodystrophy in a patient with
thesia: a report of clinical experience. Can J Anaesth 1988; mucopolysaccharidosis: characterization by MRI. J Neurol
35: 67–73 Sci 2002; 195: 171–175
References and Further Reading 923

Baxter MA, Wynn RF, Deakin JA, Bellantuono I, Edington KG, Watis RWE, Spellacy E, Adams JH. Neuropathological and clini-
Cooper A, Besley GTN, Church HJ, Wraith JE, Carr TF, Fair- cal correlations in Hurler disease. J Inherit Metab Dis 1986;
bairn LJ. Retrovirally mediated correction of bone marrow- 9: 261–272
derived mesenchymal stem cells from patients with mu- Winters PR, Harrod MJ, Molenich-Heetred SA, Kirkpatrick J,
copolysaccharidosis type I. Blood 2002; 99: 1857–1859 Rosenberg RN. a-L-Iduronidase deficiency and possible
Beesley CE, Meaney CA, Greenland G, Adams V,Vellodi A,Young Hurler-Scheie genetic compound. Clinical, pathologic and
EP, Winchester BG. Mutational analysis of 85 mucopolysac- biochemical findings. Neurology 1976; 26: 1003–1007
charidosis type I families: frequency of known mutations, Wraith JE. Enzyme replacement therapy in mucopolysacchari-
identification of 17 novel mutations and in vitro expression dosis type I: progress and emerging difficulties. J Inherit
of missense mutations. Hum Genet 2001; 109: 503–511 Metab Dis 2001; 24: 245–250
Braunlin EA, Rose AG, Hopwood JJ, Candel RD, Krivit W. Coro-
nary artery patency following long-term successful en-
graftment 14 years after bone marrow transplantation in Hunter Disease
the Hurler syndrome. Am J Cardiol 2001; 88: 1075–1077
Di Natale P, di Domenico C,Villani GRD, Lombardo A, Follenzi A, Adinolfi M. Hunter syndrome: cloning of the gene, mutations
Naldini L. In vitro gene therapy of mucopolysaccharidosis and carrier detection. Dev Med Child Neurol 1993; 35:
type I by lentiviral vectors. Eur J Biochem 2002; 269: 79–85
2764–2771 Bergstrom SK, Quinn JJ, Greenstein R, Ascensao J. Long-term
Gabrielli O, Salvolini U, Maricotti M. Cerebral MRI in two broth- follow-up of a patient transplanted for Hunter’s disease
ers with mucopolysaccharidosis type I and different clinical type IIB: a case report and literature review. Bone Marrow
phenotypes. Neuroradiology 1992; 34: 313–315 Transplant 1994; 14: 653–658
Grewal SS, Krivit W, Defor TE, Shapiro EG, Orchard PJ, Abel SL, Clarke JTR, Willard HF, Teshima I, Chang PL, Skomorowski MA.
Lockman LA, Ziegler RS, Dusenbery KE, Peters C. Hurler syn- Hunter disease (mucopolysaccharidosis type II) in a karyo-
drome.Outcome of second hematopoietic cell transplanta- typically normal girl. Clin Genet 1990; 37: 355–362
tion in Hurler syndrome. Bone Marrow Transplant 2002; 29: Clarke JTR, Greer WL, Strasberg PM, Pearce RD, Skomorowski
491–496 MA, Ray PN.Hunter disease (mucopolysaccharidosis type II)
Hite SH, Peters C, Krivit W. Correction of odontoid dysplasia fol- associated with unbalanced inactivation of the X chromo-
lowing bone-marrow transplantation and engraftment (in somes in a karyotypically normal girl. Am J Hum Genet
Hurler syndrome MPS 1H). Pediatr Radiol 2000; 30: 464–470 1991; 49: 289–297
Johnson MA, Desai S, Hugh-Jones K, Starer F. Magnetic reso- Davitt SM,Hatrick A,Sabharwal T,Pearce A,Gleeson M,Adam A.
nance imaging of the brain in Hurler syndrome. AJNR Am J Tracheobronchial stent insertions in the management of
Neuroradiol 1984; 5: 816–819 major airway obstruction in a patient with Hunter syn-
Kakkis ED, Muenzer J, Tiller GE, Waber L, Belmont J, Passage M, drome (type II mucopolysaccharidosis). Eur Radiol 2002; 12:
Izykowski B, Phillips J, Doroshow R, Walot I, Hoft R, Neufeld 458–462
EF. Enzyme-replacement therapy in mucopolysaccharido- Flomen RH, Green PM, Bentley DR, Giannelli F, Green EP. Detec-
sis I. N Engl J Med 2001; 344: 182–188 tion of point mutations and a gross deletion in six Hunter
Keeling KM, Brooks DA, Hopwood JJ, Li P, Thompson JN, syndrome patients. Genomics 1992; 13: 543–550
Bedwell DM. Gentamicin-mediated suppression of Hurler O’Brien DP, Cowie RA, Wraith JE. Cervical decompression in
syndrome stop mutations restores a low level of a-L- mild mucopolysaccharidosis type II (Hunter syndrome).
iduronidase activity and reduces lysosomal glycosamino- Child Nerv Syst 1997; 13: 87–90
glycan accumulation. Hum Mol Genet 2001; 10: 291–299 Parsons VJ, Hughes DG, Wraith JE. Magnetic resonance imag-
Krivit W, Lockman LA, Watkins PA, Hirsch J, Shapiro EG. The fu- ing of the brain, neck and cervical spine in mild Hunter’s
ture for treatment by bone marrow transplantation for syndrome (mucopolysaccharidoses type II). Clin Radiol
adrenoleukodystrophy, metachromatic leukodystrophy, 1996; 51: 719–723
globoid cell leukodystrophy and Hurler syndrome. J Inherit Shimoda-Matsubayashi S, Kuru Y, Sumie H, Ito T, Hattori N, Oku-
Metab Dis 1995; 18: 398–412 ma Y, Mizuno Y. MRI findings in the mild type of mu-
Loeb H, Jonniaux G, Resibois A, Cremer N, Dodion J,Tondeur M, copolysaccharidosis II (Hunter’s syndrome). Neuroradiolo-
Gregoire PE, Richard J, Cieters P. Biochemical and ultrastruc- gy 1990; 32: 328–330
tural studies in Hurler’s syndrome. J Pediatr 1968; 73: 860– Shinomiya N,Nagayama T,Fujioka Y,Aoki T.MRI in the mild type
874 of mucopolysaccharidosis II (Hunter’s syndrome). Neurora-
Rauch RA, Friloux LA III, Lott IT. MR Imaging of cavitary lesions diology 1996; 38: 483–485
in the brain with Hurler/Scheie. AJNR Am J Neuroradiol Vellodi A, Young E, Cooper A, Lidchi V, Winchester B, Wraith JE.
1989; 10: S1–S3 Long-term follow-up following bone marrow transplanta-
Schmidt H, Ullrich K, von Lengerke H-J, Kleine M, Brämswig J. tion for Hunter disease. J Inherit Metab Dis 1999; 22:
Radiological findings in patients with mucopolysacchari- 638–648
dosis I H/S (Hurler-Scheie syndrome). Pediatr Radiol 1987; Vinchon M, Cotten A, Clarisse J, Chiki R, Christiaens J-L. Cervical
17: 409–414 myelopathy secondary to Hunter syndrome in an adult.
Vellodi A,Young EP, Cooper A, Wraith JE, Winchester B, Meaney AJNR Am J Neuroradiol 1995; 16: 1402–1403
C, Ramaswami U, Will A. Bone marrow transplantation for Wehnert M, Hopwood JJ, Schröder W, Herrmann FH. Structural
mucopolysaccharidosis type I: experience of two British gene aberrations in mucopolysaccharidosis II (Hunter).
centres. Arch Dis Child 1997; 76: 92–99 Hum Genet 1992; 89: 430–432
Walkley SU, Haskins ME, Shull RM. Alterations in neuron mor-
phology in mucopolysaccharidosis type I*: a Golgi study.
Acta Neuropathol (Berl) 1988; 75: 611–620
924 References and Further Reading

Wilson PJ, Morris CP, Anson DS, Occhiodoro T, Bielicki J, Zafeiriou DI, Savvopoulou-Augoustidou PA, Sewell A, Pa-
Clements PR,Hopwood JJ.Hunter syndrome: isolation of an padopoulou F, Badouraki M, Vargiami E, Gombakis NP, Kat-
iduronate-2-sulfatase cDNA clone and analysis of patient zos GS. Serial magnetic resonance imaging findings in mu-
DNA. (mucopolysaccharidosis type II / lysosomal storage copolysaccharidosis IIIB (Sanfilippo’s syndrome B). Brain
disorder / X chromosome-linked mutations / sulfatase se- Dev 2001; 23: 385–389
quence homology) Proc Natl Acad Sci 1990; 87: 8531–8535
Young ID, Harper PS, Newcombe RG, Archer IM. A clinical and
genetic study of Hunter’s syndrome. 2. Differences be- Morquio Disease
tween the mild and severe forms. J Med Genet 1982; 19:
408–411 Giugliani R, Jackson M, Skinner SJ, Vimal CM, Fensom AH, Fah-
Zafeiriou DI, Augoustidou-Savvopoulou PA, Papadopoulou FA, my N, Sjövall A, Benson PF. Progressive mental regression in
Gombakis NP, Katzos GS, Kontopoulos EE, van Diggelen OP. siblings with Morquio disease type B (mucopolysacchari-
Magnetic resonance imaging findings in mild mucopoly- dosis IVB). Clin Genet 1987; 32: 313–325
saccharidosis II (Hunter’s syndrome). Eur J Paediatr Neurol Hughes DG, Chadderton RD, Cowie RA, Wraith JE, Jenkins JPR.
1998; 2: 153–156 MRI of the brain and craniocervical junction in Morquio’s
disease. Neuroradiology 1997; 39: 381–385
Mikles M, Stanton RP. A review of Morquio syndrome. Am J
Sanfilippo Disease Orthopod 1997; 26: 533–540
Nelson J, Grebbell FS. The value of computed tomography in
Andria G, di Natale P, del Giudice E, Strisciuglio P, Murino P. San- patients with mucopolysaccharidosis. Neuroradiology
filippo B syndrome (MPS III B): mild and severe forms with- 1987; 29: 544–549
in the same sibship. Clin Genet 1979; 15: 500–504 Nelson J, Kinirons M. Clinical findings in 12 patients with MPS
Barone R, Nigro F, Triulzi F, Musumeci, S, Fiumara A, Pavone L. IVA (Morquio’s disease). Clin Genet 1988; 33: 121–125
Clinical and neuroradiological follow-up in mucopolysac- Nelson J, Thomas PS. Clinical findings in 12 patients with MPS
charidosis type III (Sanfilippo syndrome). Neuropediatrics IVA (Morquio’s disease). Further evidence for heterogene-
1999; 30: 270–274 ity. III. Odontoid dysplasia. Clin Genet 1988; 33: 126–130
Fu H, Samulski RJ, McCown TJ, Picornell YJ, Fletcher D, Muenzer Nelson J, Broadhead D, Mossman J. Clinical findings in 12 pa-
J. Neurological correction of lysosomal storage in a mu- tients with MPS IVA (Morquio’s disease). Further evidence
copolysaccharidosis IIIB mouse model by adeno-associat- for heterogeneity. I. Clinical and biochemical findings. Clin
ed virus-mediated gene delivery. Mol Ther 2002; 5: 42–49 Genet 1988; 33: 111–120
Gïngŏör N,Tunçbilek E. Sanfilippo disease type B. A case report Northover H, Cowie RA,Wraith JE. Mucopolysaccharidosis type
and review of the literature on recent advances in bone IVA (Morquio syndrome): a clinical review. J Inherit Metab
marrow transplantation.Turk J Pediatr 1995; 37: 157–163 Dis 1996; 19: 357–365
Haust MD, Gordon BA, Hong R, Choi JH, Langer LO, Spranger J, Oshima A, Yoshida K, Shimmoto M, Fukuhara Y, Sakuraba H,
Opitz JM. Clinicopathological conference: an adolescent Suzuki Y. Human b-glycosidase gene mutations in Morquio
girl with severe mental impairment and mucopolysaccha- B disease. Am J Hum Genet 1991; 49: 1091–1093
riduria. Am J Med Genet 1985; 22: 1–27 Rigante D, Antuzzi R, Ricci R, Segni G. Cervical myelopathy in
Jones MZ, Alroy J, Rutledge JC, Taylor JW, Alvord EC Jr, Toone J, mucopolysaccharidosis type IV. Clin Neuropathol 1999; 18:
Applegarth D, Hopwood JJ, Skutelsky E, Ianelli C, Thorley- 84–86
Lawson D, Mitchell-Herpolsheimer C, Arias A, Sharp P, Evans Stevens JM, Kendall BE, Crockard HA, Ransford A.The odontoid
W, Sillence D, Cavanagh KT. Human mucopolysaccharidosis process in Morquio-Brailsford’s disease; the effects of
IIID: clinical, biochemical, morphological and immunohis- occipitocervical fusion. J Bone Joint Surg [Br] 1991; 73:
tochemical characteristics. J Neuropathol Exp Neurol 1997; 851–858
56: 1158–1167
Kurihara M, Kumagai K, Yagishita S. Case reports. Sanfilippo
Maroteaux-Lamy Disease
syndrome type C: a clinicopathological autopsy study of a
long-term survivor. Pediatr Neurol 1996; 14: 317–321
Ozand PT,Thompson JN, Gascon GG, Sarvepalli SB, Rahbeeni Z, Herskhovitz E,Young E, Rainer J, Hall CM, Lidchi V, Chong K,Vel-
Nester MJ, Brismar J. Sanfilippo type D presenting with ac- lodi A. Bone marrow transplantation for Maroteaux-Lamy
quired language disorder but without features of mu- syndrome (MPS VI):long-term follow-up.J Inherit Metab Dis
copolysaccharidosis. J Child Neurol 1994; 9: 408–411 1999; 22: 50–62
Petitti N, Holder CA, Williams DW III. Mucopolysaccharidosis III Hite SH, Krivit W, Haines SJ, Whitley CB. Syringomyelia in mu-
(Sanfilippo syndrome) type B. Cranial imaging in two cases. copolysaccharidosis type VI (Maroteaux-Lamy syndrome):
J Comput Assist Tomogr 1997; 21: 897–899 imaging findings following bone marrow transplantation.
Vallani GRD, Follenzi A,Vanacore B, di Domenico C, Naldini L, di Pediatr Radiol 1997; 27: 736–738
Natale P. Correction of mucopolysaccharidosis type IIIb Tamaki N, Kojima N, Tanimoto M, Suyama T, Matsumoto S.
fibroblasts by lentiviral vector-medicated gene transfer. Myelopathy due to diffuse thickening of the cervical dura
Biochem J 2002; 364: 747–753 mater in Maroteaux-Lamy syndrome: report of a case. Neu-
van de Kamp JJP, Niermeijer MF, von Figura K, Giesberts MAH. rosurgery 1987; 21: 416–419
Genetic heterogeneity and clinical variability in the Sanfil- Uçakhan OO, Brodie SE, Desnick R, Willner J, Asbell PA. Long-
ippo syndrome (types A, B and C). Clin Genet 1981: 20: term follow-up of corneal graft survival following bone
152–160 marrow transplantation in the Maroteaux-Lamy syndrome.
van Schrojenstein-de Valk HMJ, van de Kamp JJP. Follow-up on CLAO J 2001; 27: 234–237
seven adult patients with mild Sanfilippo B-disease. Am J
Med Genet 1987; 28: 125–129
References and Further Reading 925

Vestermark S, Tønnesen T, Schultz Andersen M, Güttler F. Men- Chigorno V, Tettamanti G, Sonnino S. Metabolic processing of
tal retardation in a patient with Maroteaux-Lamy. Clin gangliocidosis by normal and Salla human fibroblasts in
Genet 1987; 31: 114–117 culture. J Biol Chem 1996; 36: 21738–21744
Vougioukas VI,Berlis A,Kopp MV,Korinthenberg R,Spreer J,van Daneman A, Stringer D, Reilly BJ. Neonatal ascitis due to lyso-
Velthoven V. Neurosurgical interventions in children with somal storage disease. Radiology 1983; 149: 463–467
Maroteaux-Lamy syndrome. Case report and review of the Echenne B, Vidal M, Maire I, Michalski JC, Baldet P, Astruc J. Sal-
literature. Pediatr Neurosurg 2001; 35: 35–38 la disease in one non-Finnish patient. Eur J Pediatr 1986;
Wicker G, Prill V, Brooks D, Gibson G, Hopwood J, von Figura K, 145: 320–322
Peters C. Mucopolysaccharidosis VI (Maroteaux-Lamy syn- Fois A, Balestri P, Mancini GMS, Borgogni P, Margollicci MA,
drome). J Biol Chem 1991; 266: 21386–21391 Molinelli M, Alessandri C, Gerli R. Free sialic acid storage dis-
ease. Eur J Pediatr 1987; 146: 195–198
Gillan JE, Lowden JA, Gaskin K, Cutz E. Congenital ascitis as a
Sly Disease presenting sign of lysosomal storage disease. J Pediatr
1984; 104: 225
Bernsen PLJA, Wevers RA, Gabreëls FJM, Lamers KJB, Sonnen Grosso S, Berardi R, Farnetani MA, Margollicci M, Mancini GMS,
AEH, Schuurmans Stekhoven JH. Phenotypic expression in Morgese G, Balestri P. Multiple neuroendocrine disorder in
mucopolysaccharidosis VII. J Neurol Neurosurg Psychiatry Salla disease. J Child Neurol 2001; 16: 775–777
1987; 50: 699–703 Haataja L, Parkkola R, Sonninen P, Verhanen S-L, Scheutker J,
Fukuda S,Tomatsu S,Sukegawa K,Sasaki T,Yamada Y,Kuwahara Äärimaa T,Turpeinen U, Renland M, Aula P. Phenotypic vari-
T, Okamoto H, Ikedo Y, Yamaguchi S, Orii T. JSSIEM Meeting. ation and magnetic resonance imaging (MRI) in Salla dis-
Molecular analysis of mucopolysaccharidosis type VII. J ease, a free sialic acid storage disorder. Neuropediatrics
Inherit Metab Dis 1991; 14: 800–804 1994; 25: 238–244
Kyle JW, Birkenmeier EH, Gwynn B, Vogler C, Hoppe PC, Hoff- Havelaar AC, Mancini GMS, Beerens CEMT, Souren RMA,Verhei-
mann JW, Sly WS. Correction of murine mucopolysacchari- jen FW. Purification of the lysosomal sialic acid transporter.
dosis VII by a human b-glucuronidase transgene. Proc Natl Functional characteristics of a monocarboxylate trans-
Acad Sci 1990; 87: 3914–3918 porter. J Biol Chem 1998; 273: 34568–34574
Ross CJD, Bastedo L, Maier SA, Sands MS, Chang PL. Treatment Havelaar AC, Beerens CEMT, Mancini GMS,Verheijen FW.Trans-
of a lysosomal storage disease, mucopolysaccharidosis VII, port of organic anions by the lysosomal sialic acid trans-
with microencapsulated recombinant cells. Hum Gene porter: a functional approach towards the gene for sialic
Ther 2000; 11: 2117–2127 acid storage disease. FEBS Lett 1999; 446: 65–68
Sewell AC, Gehler J, Mittermaier G, Meyer E. Mucopolysacchari- Jonas AJ. Studies of lysosomal sialic acid metabolism: reten-
dosis type VII (b-glucuronidase deficiency): a report of a tion of sialic acid by Salla disease lysosomes. Biochem Bio-
new case and a survey of those in the literature. Clin Genet phys Res Commun 1986; 137: 175–181
1982; 21: 366–373 Kirchner L, Kircher S, Salzer-Muhar U, Paschke E, Birnbacher R,
Tomatsu S, Fukuda S, Sukegawa K, Ikedo Y,Yamada S,Yamada Y, Stöckler-Ipsiroglu S. Infantile sialic acid storage disease and
Sasaki T, Okamoto H, Kuwahara T, Yamaguchi S, Kiman T, protein-losing gastroenteropathy. Pediatr Neurol 2003; 28:
Shintaku H, Isshiki G, Orii T. Mucopolysaccharidosis type VII: 313–317
characterization of mutations and molecular heterogene- Lemyre E, Russo P, Melançon SB, Gagné R, Potier M, Lambert M.
ity. Am J Hum Genet 1991; 48: 89–96 Clinical spectrum of infantile free sialic acid storage dis-
ease. Am J Med Genet 1999; 82: 385–391
Linnankivi T, Lönnqvist T, Autti T. A case of Salla disease with in-
14 Free Sialic Acid Storage Disorder volvement of the cerebellar white matter. Neuroradiology
2003; 45: 107–109
Mancini GMS, Verheijen FW, Glajaard H. Free N-acetylneu-
Aula N, Salomäki P,Timonen R,Verheijen F, Mancini G, Månsson
raminic acid (NANA) storage disorders: evidence for defec-
J-E, Aula P, Peltonen L. The spectrum of SLC17A5 gene mu-
tive NANA transport across the lysosomal membrane. Hum
tations resulting in free sialic acid-storage diseases indi-
Genet 1986; 73: 214–217
cates some genotype-phenotype correlation. Am J Hum
Mancini GMS, Verheijen FW, Beerens CEMT, Renlund M, Aula P.
Genet 2000; 67: 832–840
Sialic acid storage disorders: observations on clinical and
Autio-Harmainen H, Oldfors A, Sourander P, Renlund M,
biochemical variation. Dev Neurosci 1991; 13: 327–330
Dammert K, Similiä S. Neuropathology of Salla disease. Ac-
Mancini GMS, Beerens CEMT, Aula PP, Verheijen FW. Sialic acid
ta Neuropathol (Berl) 1988; 75: 481–490
storage diseases. A multiple lysosomal transport defect for
Baumkötter J, Cantz M, Mendla K, Baumann W, Friebolin H,
acidic monosaccharides. J Clin Invest 1991; 87: 1329–1335
Gehler J, Spranger J. N-Acetylneuraminic acid storage dis-
Mancini GMS, Hu P, Verheijen FW, van Diggelen OP, Janse HC,
ease. Hum Genet 1985; 71: 155–159
Kleijer WJ, Beemer FA, Jennekens FGI. Salla disease variant
Biancheri R,Verbeek E, Rossi A, Gaggero R, Roccatagliata L, Gat-
in a Dutch patient. Potential Value of polymorphonuclear
ti R, van Diggelen OP,Verheijen FW, Mancini GMS. An Italian
leucocytes for hetrozygote detection. Eur J Pediatr 1992;
severe Salla disease variant associated with a SLC71A5 mu-
151: 590–595
tation earlier described in infantile sialic acid storage dis-
Mancini GMS, Havelaar AC, Verheijen FW. Lysosomal transport
ease. Clin Genet 2002; 61: 443–447
disorders. J Inherit Metab Dis 2000; 23: 278–292
Blom HJ, Andersson HC, Seppala R,Tietze F, Gahl WA. Defective
Nakano C, Hirabayashi Y, Ohno K,Yano K,Yano T, Mito T, Sakurai
glucuronic acid transport from lysosomes of infantile free
M. A Japanese case of infantile sialic acid storage disease.
sialic acid storage disease fibroblasts. Biochem J 1990; 268:
Brain Dev 1996; 18: 153–156
621–625
926 References and Further Reading

Parazzini C, Arena S, Marchetti L, Menni F, Filocamo M, Verhei- Thomas GH, Scocca J, Libert J, Vamos E, Miller CS, Reynolds LW.
jen FW, Mancini GMS, Parini R. Infantile sialic acid storage Alterations in cultured fibroblasts of sibs with an infantile
disease: serial ultrasound and magnetic resonance imag- form of free (unbound) sialic acid storage disorder. Pediatr
ing features. AJNR Am J Neuroradiol 2003; 24: 398–400 Res 1983; 17: 307–312
Paschke E, Höfler G, Roscher A. Infantile sialic acid storage dis- Tietze F,Seppala R,Renlund M,Hopwood JJ,Harper GS,Thomas
ease: the fate of biosynthetically labelled N-acetyl-(3H)- GH, Gahl WA. Defective lysosomal egress of free sialic acid
neuraminic acid in cultured human fibroblasts. Pediatr Res (N-acetylneuraminic acid) in fibroblasts of patients with
1986; 8: 773–777 free sialic acid storage disease. J Biol Chem 1989; 26:
Paschke E,Trinkl G, Erwa W, Pavelka M, Mutz I, Roscher A. Infan- 15316–15322
tile type of sialic acid storage disease with sialuria. Clin Tondeur M, Libert J, Vamos E, Van Hoof F, Thomas GH, Strecker
Genet 1986; 29: 417–424 G. Infantile form of sialic acid storage disorder: clinical, ul-
Pueshel SM, O’Shea PA, Alroy J, Ambler MW, Dangond F, Daniel trastructural, and biochemical studies in two siblings. Eur J
PF, Kolodny EH. Infantile sialic acid storage disease associat- Pediatr 1982; 139: 142–147
ed with renal disease. Pediatr Neurol 1988; 4: 207–212 Varho T, Komu M, Sonninen P, Holopainen I, Nyman S, Manner
Renlund M.Clinical and laboratory diagnosis of Salla disease in T, Sallinpää M, Aula P, Lundbom N. A new metabolite con-
infancy and childhood. J Pediatr 1984; 104: 232–236 tributing to N-acetyl signal in 1H MRS of the brain in Salla
Renlund M, Aula P, Raivio KA, Autio S, Sainio K, Rapola J, Koskela disease. Neurology 1999; 52: 1668–1672
S-L. Salla disease: a new lysosomal storage disorder with Varho T, Jääskeläinen S, Tolonen U, Sonninen P, Vainionpää L,
disturbed sialic acid metabolism. Neurology 1983; 33: Aula P, Sillanpää M. Central and peripheral nervous system
57–66 dysfunction in the clinical variation of Salla disease. Neurol-
Renlund M,Chester MA,Lundblad A,Parkkinen J,Krusius T.Free ogy 2000; 55: 99–103
N-acetylneuraminic acid in tissues in Salla disease and the Varho TT, Alajoki LE, Posti KM, Korhonen TT, Renlund GG, Ny-
enzymes involved in its metbolism. Eur J Biochem 1983; man SRG, Sillanpää, Aula PP. Phenotypic spectrum of Salla
130: 39–45 disease, a free sialic acid storage disorder. Pediatr Neurol
Renlund M, Kovanen PT, Raaivio K, Aula P, Gamhberg CG, Ehn- 2002; 26: 267–273
holm C. Studies of the defect underlying the lysosomal Verheijen FW, Verbeel E, Aula N, Beerens CEMT, Havelaar AC,
storage of sialic acid in Salla disease. J Clin Invest 1986; 77: Joosse M, Peltonen L, Aula P, Galjaard H, van der Spek PJ,
568–574 Mancini GMS. A new gene, encoding an anion transporter,
Renlund M, Tietze F, Gahl WA. Defective sialic acid egress from is mutated in sialic acid storage diseases. Nat Genet 1999;
isolated fibroblast lysosomes of patients with salla disease. 23: 462–465
Science 1986; 232: 579–763 Wolburg-Buchholz K,Schlote W.Familial lysosomal storage dis-
Robinson RO, Fenson AH, Lake BD. Salla disease–rare or under- ease with generalized vacuolisation and sialic aciduria.
diagnosed? Dev Med Child Neurol 1997; 39: 153–157 Sporadic Salla disease. Neuropediatrics 1985; 16: 67–75
Salomäki P, Aula N, Juvonen V, Renlund M, Aula P. Prenatal de-
tection of free sialic acid storage disease: genetic and bio-
chemical studies in nine families. Prenat Diagn 2001; 21:
354–358
15 Neuronal Ceroid Lipofuscinoses
Schleutker J, Leppänen P, Månson J-E, Erikson A, Weissenbach General
J, Peltonen L, Aula P.Lysosomal free sialic acid storage disor-
ders with different phenotypc presentations – infantile- Autti T, Raininko R, Vanhanen SL, Santavuori P. Magnetic reso-
form sialic acid storage disease and Salla disease – repre- nance techniques in neuronal ceroid lipofuscinoses and
sent allelic disorders. Am J Hum Genet 1995; 57: 893–901 some other lysosomal diseases affecting the brain. Curr
Schmid JA, Mach L, Paschke E, Glössl. Accumulation of sialic Opin Neurol 1997; 10: 519–524
acid in endocytic compartments interferes with the forma- Autti R, Raininko R, Santavuori P, Vanhanen SL, Poutanen VP,
tion of mature lysosomes. J Biol Chem 1999; 27: 19063– Haltia M. MRI of neuronal ceroid lipofuscinosis. II. Post-
19071 mortem MRI and histopathological study of the brain in 16
Seppala R,Renlund M,Bernardini I,Tietze F,Gahl WA.Renal han- cases of neuronal ceroid lipofuscinosis of juvenile or late in-
dling of free sialic acid in normal humans and patients with fantile type. Neuroradiology 1997; 29: 371–377
Salla disease or renal disease. Lab Invest 1990; 63: 197–203 Boldrini R, Biselli R, Santorelli FM, Bosman C. Neuronal ceroid
Sewell AC, Poets CF, Degen I, Stöß, Pontz BF. The spectrum of lipofuscinosis: an ultrastructural, genetic, and clinical study
free neuraminic acid storage disease in childhood: clinical, report. Ultrastruct Pathol 2001; 25: 51–58
morphological and biochemical observations in three non- Boustany RMN, Kolodny EH. Neurological progress. The neu-
Finnish patients. Am J Med Genet 1996; 63: 203–208 ronal ceroid lipofuscinoses: a review. Rev Neurol 1989; 145:
Sewell AC, Murphy HC, Illes R. Proton nuclear magnetic reso- 105–110
nance spectroscopic detection of sialic acid storage dis- Brooks SS. Genetic counseling in the neuronal ceroid lipofusci-
ease. Clin Chem 2002; 48: 357–359 noses. Adv Genet 2001; 45: 141–158
Sonninen P, Autti T, Varho T, Hämälläinen, Raininko R. Brain in- Dawson G, Cho S. Batten’s disease: clues to neuronal protein
volvement in Salla disease.AJNR Am J Neuroradiol 1999;20: metabolism in lysosomes. J Neurosci Res 2000; 60: 133–140
433–443 Dunn DW. CT in ceroid lipofuscinosis. Neurology 1987; 37:
Sperl W,Gruber W,Quatacker J,Monnens L,Thoenes W,Fink FM, 1025–1026
Paschke E. Nephrosis in two siblings with infantile sialic Elleder M, Franc K, Kraus J, Nevšímalová S, Sixtová K, Zeman J.
acid storage disease. Eur J Pediatr 11991; 49: 447–482 Neuronal ceroid lipofuscinosis in the Czech Republic:
Stevenson RE, Lubinsky M, Taylor HA, Wenger DA, Schoer RJ, analysis of 57 cases. Report of the ‘Prague NLC group’. Eur J
Olmstead PM. Sialic acid storage disease with sialuria: clini- Paediatr Neurol 1997; 4: 109–114
cal and biochemical features in the severe infantile type.
Pediatr 1983; 72: 441–449
References and Further Reading 927

Gardiner RM. The molecular genetic basis of the neuronal Wisniewski KE, Kida E, Golabek AA, Kaczmarski W, Connell F,
ceroid lipofuscinoses. Neurol Sci 2000; 21: S15-S19 Zhong N. Neuronal ceroid lipofuscinoses: classification and
Goebel HH. Prenatal ultrastructural diagnosis in the neuronal diagnosis. Adv Genet 2001; 45: 1–34
ceroid lipofuscinoses. Pathol Res Pract 1994; 190: 728–733 Zhong N. Neuronal ceroid lipofuscinoses and possible patho-
Goebel HH. The neuronal ceroid-lipofuscinoses. J Child Neurol genic mechanism. Mol Genet Metab 2000; 71: 195–206
1995; 10: 424–437 Zhong N. Molecular genetic testing for neuronal ceroid lipo-
Goebel HH. Morphological diagnosis in neuronal ceroid lipo- fuscinoses. Adv Genet 2001; 45: 141–158
fuscinosis. Neuropediatrics 1997; 28: 67–69
Goebel HH. The new nosography of the neuronal ceroid-lipo-
fusinoses. Ann Pathol 2000; 20: 479–491 Neuronal Ceroid Lipofuscinosis: CLN1
Goebel HH.Morphological aspects of the neuronal ceroid lipo-
fuscinoses. Neurol Sci 2000; 21: S27-S33
Åberg K,Heiskala H,Vanhanen S-L,Himberg JJ,Hosking G,Yuen
Goebel HH, Mole SE, Lake BD, eds.The neuronal ceroid lipofus-
A, Santavori P. Lamotrigine therapy in infantile neuronal
cinoses (Batten disease). Amsterdam: IOS Press, 1999
ceroid lipofuscinosis (INCL). Neuropediatrics 1997; 28:
Goebel HH, Schochet SS, Jaynes M, Brück, W, Kohlschütter A,
77–79
Hentati F. Progress in neuropathology of the neuronal
Barohn RJ, Dowd DC, Kagen-Hallet KS. Congenital ceroid lipo-
ceroid lipofuscinoses. Mol Genet Metab 1999; 66: 367–372
fuscinosis. Pediatr Neurol 1992; 8: 54–59
Haltia M. The neuronal ceroid-lipofuscinoses. J Neuropathol
Bizzozero GA.The mechanisms and functional roles of protein
Exp Neurol 2003; 62: 1–13
palmitoyalation in the nervous system. Neuropediatrics
Jolly RD. Dalefield RR, Palmer DN. Ceroid, lipofuscin and the
1997; 28: 23–26
ceroid-lipofuscinoses (Batten disease). J Inherit Metab Dis
Confort-Gouny S,Chabrol B,Vion-Dury J,Mancini J,Cozzone PJ.
1993; 16: 280–283
MRI and localized proton MRS in early infantile form of neu-
Junaid NA, Pullarkat RK. Biochemistry of neuronal ceroid lipo-
ronal ceroid-lipofuscinosis. Pediatr Neurol 1993; 9: 57–60
fuscinoses. Adv Genet 2001; 45: 93–106
Haltia M, Rapola J, Santavuori P, Keränen A. Infantile type of so-
Kida E, Golabek AA, Wisniewski KE. Cellular pathology and
called neuronal ceroid-lipofuscinosis. 2. Morphological and
pathogenic aspects of neuronal ceroid lipofuscinoses. Adv
biochemical studies. J Neurol Sci 1973; 18: 269–285
Genet 2001; 45: 35–68
Haltia M, Rapola J, Santavuori P. Infantile type of so-called neu-
Kleijer WJ, van Diggelen OP. Prenatal diagnosis of neuronal
ronal ceroid-lipofuscinosis. Histological and electron mi-
ceroid lipofuscinoses. Prenat Diagn 2000; 20: 819–821
croscopic studies. Acta Neuropathol (Berl) 1973; 26: 157–
Machen BC, Williams JP, Lum GB, Dyken P, Joslyn JN, Harpen
170
MD, Dotson P. Magnetic resonance imaging in neuronal
Hofmann SL, Das AD, Lu JY, Wisniewski KE, Gupta P. Infantile
ceroid lipofuscinosis. J Comput Tomogr 1987; 11: 160–166
neuronal ceroid lipofuscinosis: no longer just a ‘Finnish’ dis-
Mitchison GM, Mole SE. Neurodegenerative disease: the neu-
ease. Eur J Paediatr Neurol 2001; 5: 47–51
ronal ceriod lipofuscinoses (Batten disease).Curr Opin Neu-
Jongen PJH, Gabreëls FJM, Schuurmans Stekhoven JH, Renier
rol 2001; 14: 795–803
WO, Le Coultre R, Begeer JH. Early infantile form of neuronal
Mole SE, Mitchison HM, Munroe PB. Molecular basis of the neu-
ceroid lipofuscinosis. Clin Neurol Neurosurg 1987; 89:
ronal ceroid lipofuscinoses: mutations in CLN1, CLN2, CLN3
161–167
and CLN5. Hum Mutat 1999; 14: 199–215
Lake BD, Brett EM, Boyd SG. A form of juvenile Batten disease
Peltonen L, Savukoski M, Vesa J. Genetics of the neuronal
with granular osmiophilic deposits. Neuropediatrics 1996;
ceroid lipofuscinoses. Curr Opin Genet Dev 2000; 10: 299–
27: 265–269
305
Lönnqvist T, Vanhanen SL, Vettenranta K, Autti T, Rapola J, San-
Raininko R, Santavuori P, Heiskala H, Sainio K, Palo J. CT findings
tavuori P, Saarinen-Pihkala UM. Hematopoietic stem cell
in neuronal ceroid lipofuscinoses. Neuropediatrics 1990;
transplantation in infantile neuronal ceroid lipofuscinosis.
21: 95–101
Neurology 2001; 57: 1411–1416
Santavuori P. Neuronal ceroid-lipofuscinoses in childhood.
Lu J-Y, Verkruyse KA, Hofmann SL. The effects of lysomotropic
Brain Dev 1988; 10: 80–83
agents on normal and INCL cells provide further evidence
Santavuori P, Linnankivi T, Jaeken J, Vanhanen S-L, Telakivi T,
for the lysosomal nature of palmitoyl-protein thioesterase
Heiskala H. Psychological symptoms and sleep distur-
function. Biochim Biophys Acta 2002; 1583: 35–44
bances in neuronal ceroid-lipofuscinoses (NCL). J Inherit
O’Rawe A, Mitchison HM, Williams R, Wheeler R, Andermann F,
Metab Dis 1993; 16: 245–248
Andermann E, Hart YM, Martin JJ, Philippart M, Stephanson
Santavuori P, Lauronen L, Kirveskari E, Åberg L, Sainio K, Autti T.
JBP, Gardiner RM, Mole SE. Genetic linkage analysis of a vari-
Neuronal ceroid lipofuscinoses in childhood. Neurol Sci
ant of juvenile onset neuronal ceroid lipofuscinosis with
2000; 21: S35–S41
granular osmiophilic deposits. Neuropediatrics 1997; 28:
Santavuori P,Vanhanen S-L, Autti T. Clinical and neuroradiolog-
21–22
ical diagnostic aspects of neuronal ceroid lipofuscinoses
Philippart M, Chugani HT, Bateman JB. New Spielmeyer-Vogt
disorders. Eur J Paediatr Neurol 2001; 5: 157–161
variant with granular inclusions and early brain atrophy.
Wisniewski KE, Kida E, Patxot OF, Connell F. Variability in the
Am J Med Genet 1995; 57: 160–164
clinical and pathological findings in the neuronal ceroid
Salonen T, Heinonen-Kopra O, Vesa J, Jalanko A. Neuronal traf-
lipofuscinoses: review of date and observations. Am J Med
ficking of palmitoyl protein thioesterase provides an excel-
Genet 1992; 42: 525–532
lent model to study the effects of different mutations,
Wisniewski KE,Kida E,Connell F,Zhong N.Neuronal ceroid lipo-
which cause infantile neuronal ceroid lipofuscinosis. Mol
fuscinoses: research update. Neurol Sci 2000; 21: S49–S56
Cell Neurosci 2001; 18: 131–140
Wisniewski KE,Zhong N,Philippart M.Pheno-/genotypic corre-
Santauvori P, Haltia M, Rapola J. Infantile type of so-called neu-
lations of neuronal ceroid lipofuscinoses. Neurology 2001;
ronal ceroid-lipofuscinosis. Dev Med Child Neurol 1974; 16:
57: 576–581
644–653
928 References and Further Reading

Santavuori P,Raininko R,Vanhanen SL,Launes J,Sainio K.MRI of Ezaki J, Tadeka-Ezaki M, Kominami E. Tripeptidyl peptidase I,
the brain, EEG sleep spindles and SPECT in the early diag- the late infantile neuronal ceroid lipofuscinosis gene prod-
nosis of infantile neuronal ceroid lipofuscinosis. Dev Med uct, initiates the lysosomal degeneration of subunit c of
Child Neurol 1992; 34: 61–79 ATP synthase. J Biochem 2000; 128: 506–516
Santavuori P, Vanhanen S-L, Sainio K, Nieminen M, Wallden T, Fueki N, Linuma K, Kojima A, Yanai K, Haginoya K, Tada K, Ido T,
Launes J, Raininko R. Infantile neuronal ceroid-lipofusci- Ito M. Reduced regional cerebral metabolic rate for glucose
nosis (INCL):diagnostic criteria.J Inherit Metab Dis 1993; 16: at the terminal stage in a case of late infantile neuronal
227–229 ceroid lipofuscinosis. J Child Neurol 1990; 5: 98–100
Svennerholm L, Fredman P, Jungbjer B, Mansson JE, Rynmark Kurata K, Hayashi M, Satoh J, Kojima H, Nagata J, Tamagada K,
BM, Boström K, Hagberg B, Noren L, Santavuori P. Large al- Shinohara T, Morimatsu Y. Kominami E. Pathological study
terations in ganglioside and neutral glycosphingolipid pat- on sibling autopsy cases of the late infantile form of neu-
terns in brains form cases with infantile neuronal ceroid ronal ceroid lipofuscinosis. Brain Dev 1999; 21: 63–67
lipofuscinosis/polyunsaturated fatty acid lipidosis. J Neu- Petersen B,Handwerker M, Huppertz H-I.Neurological findings
rochem 1987; 49: 1772–1783 in classical late onset infantile neuronal ceroid-lipofusci-
Tyynelä J, Palmer DN, Baumann M, Haltia M. Storage of nosis. Pediatr Neurol 1996; 15: 344–347
saposins A and D in infantile neuronal ceroid-lipofusci- Seitz D, Grodd W, Schwab A, Seeger U, Klose U, Nägele T. MR
nosis. FEBS Lett 1991; 330: 8–12 imaging and localized proton MR spectroscopy in late in-
Van Diggelen OP, Thobois S, Tilikete C, Zabot M-T, Keulemans fantile neuronal ceroid lipofuscinosis. AJNR Am J Neuro-
JLM, van Bunderen PA, Taschner PEM, Losekoot M, Voznyi radiol 1998: 19: 1373–1377
YV. Adult neuronal ceroid lipofuscinosis with palmitoyl- Sondhi D, Hackett NR, Alpblett RL, Kaminsky SM, Pergolizzi RG,
protein thioesterase deficiency:first adult-onset patients of Crystal RG. Feasibility of gene therapy for late neuronal
a childhood disease. Ann Neurol 2001; 50: 269–272 ceroid lipofuscinosis. Arch Neurol 2001; 58: 1793–1798
Vanhanen S-L, Raininko R, Santavuori P. Early differential diag- Umehara F, Higuchi I,Tanaka HK, Niiyama T, Ezaki J, Kominani E,
nosis of infantile neuronal ceroid lipofuscinosis, Rett syn- Osame M. Accumulation of mitochondrial ATP synthase
drome, and Krabbe disease by CT and MR. AJNR Am J Neu- subunit c in muscle in a patient with ceroid lipofuscinosis
roradiol 1994; 15: 1443–1543 (late infantile form). Acta Neuropathol (Berl) 1997; 93:
Vanhanen S-L, Liewendahl K, Raininko R, Nikkinen P, Autti T, 628–632
Sainio K, Santavuori P. Brain perfusion SPECT in infantile Warburton MJ, Bernardini F.Tripeptildyl-peptidase I deficiency
neuronal ceroid-lipofuscinosis (INCL) comparison with in classical late-infantile neuronal ceroid lipofuscinosis
clinical manifestations and MRI findings. Neuropediatrics brain tissue. Evidence for defective peptidase rather than
1995; 27: 76–83 proteinase activity. J Inherit Metab Dis 2000; 23: 145–154
Vanhanen S-L, Raininko R, Santavuori O, Autti T, Haltia M. MRI
Evaluation of the brain in infantile neuronal ceroid-lipofus- Neuronal Ceroid Lipofuscinosis: CLN 3
cinosis. 1. Postmortem MRI with histopathological correla-
tion. J Child Neurol 1995; 10: 438–443
Autti T, Raininko R, Vanhanen SL, Santavuori P. MRI of neuronal
Vanhanen S-L, Raininko R, Autti T, Santavuori P. MRI Evaluation
ceroid lipofuscinosis. I. Cranial MRI of 30 patients with juve-
of the brain in infantile neuronal ceroid-lipofuscinosis. 2.
nile neuronal ceroid lipofuscinosis. Neuroradiology 1996;
MRI findings in 21 patients. J Child Neurol 1995; 10:
38: 476–482
444–450
Bennett MJ, Gayton AR, Rittey CDR, Hosking GP. Juvenile neu-
Vanhanen S-L, Sainio J, Lappi M, Santavuori P. EEG and evoked
ronal ceroid-lipofuscinosis: developmental progress after
potentials in infantile neuronal ceroid-lipofuscinosis. Dev
supplementation with polyunsaturated fatty acids. Dev
Med Child Neurol 1997; 39: 456–463
Med Child Neurol 1994; 36: 630–638
Vanhanen SL, Puranen J, Autti T, Raininko R, Liewendahl K,
Boustany RMN, Filipek P. Seizures, depression and dementia in
Nikkinen P, Santavuori P, Suominen P,Vuari K, Häkkinen AM.
teenagers with Batten disease. J Inherit Metab Dis 1993; 16:
Neuroradiological findings (MRS, MRI, SPECT) in infantile
252–255
neuronal ceroid-lipofuscinosis (infantile CLN1) at different
Brod RD, Packer AJ, Van Dyk HJL. Diagnosis of neuronal ceroid
stages of the disease. Neuropediatrics 2004; 35: 27–35
lipofuscinosis by ultrastructural examination of peripheral
Vesa J, Hellsten E, Verkruyse LA, Camp LA, Rapola J, Santavuori
blood lymphocytes. Arch Ophthalmol 1987; 105: 1388–
P, Hofmann SL, Peltonen L. Mutations in the palmitoyl pro-
1393
tein thioesterase gene causing infantile neuronal ceroid
Bruun I, Reske-Nielsen E, Oster S. Juvenile ceroid-lipofuscinosis
lipofuscinosis. Nature 1995; 376: 584–587
and calcifications of the CSF. Acta Neurol Scand 1991; 83:
1–8
Hofman IL. Observations in institutionalized neuronal ceroid-
Neuronal Ceroid Lipofuscinosis: CLN2 lipofuscinosis patients, with special reference to involun-
tary movements. J Inherit Metab Dis 1993; 16: 249–251
Carlesimo M,Giustini S,Rossodivita A,Cardona F,Calvieri S.Late Järvelä I, Autti T, Lamminranta S, Aberg L, Raininko R, San-
infantile ceroid-lipofuscinoses. An ultrastructural study. Am tavuori P. Clinical and magnetic resonance imaging find-
J Dermatopathol 1993; 15: 456–460 ings in Batten disease: analysis of the major mutation
D’Incerti L. MRI in neuronal ceroid lipofuscinosis. Neurol Sci (1.02-kb deletion). Ann Neurol 1997; 42: 799–802
2000; 21: S71–S73 Kimura S, Goebel HH. Light and electron microscopic study of
Ezaki J, Wolfe LS, Kominami E. Decreased lysosomal subunit c- juvenile neuronal cerroid-lipofuscinosis lymphocytes.Pedi-
degrading activity in fibroblasts from patients with late in- atr Neurol 1988; 4: 148–152
fantile neuronal ceroid lipofuscinosis. Neuropediatrics
1997; 28: 53–55
References and Further Reading 929

Lauronen L, Munroe PB, Järvelä I, Autti T, Mitchison HM, O’Rawe Holmberg V, Lauronen K, Autti T, Santavuori P, Savukoski M,
AM, Gardiner RM, Mole SE, Puranen J, Häkkinen AAM, Kire- Uvebrand P, Hofman I, Peltonen L, Järvelä I. Phenotype-
verski E, Santavuori P. Delayed classical and protracted phe- genotype correlation in eight patients with Finnish variant
notypes of compound heterozygous juvenile neuronal late infantile NCL (CLN5). Neurology 2000; 55: 579–581
ceroid lipofuscinosis. Neurology 1999; 52: 360–365 Isosomppi J, Vesa J, Jalanko A, Peltonen L. Lysosomal localiza-
Manca V, Kanitakis J, Zambruno G, Thiovilet J, Gonnaund P. Ul- tion of the neuronal ceroid lipofuscinosis CLN5. Hum Mol
trastructural study of the skin in a case of juvenile ceroid- Genet 2002; 11: 885–891
lipofuscinosis. Am J Dermatopathol 1990; 12: 412–416 Lauronen L, Huttunen J, Kirveskari E,Wikström H, Sainio K, Aut-
Mitchison HM, Taschner PEM, Kremmidiotis G, Callen DF, Dog- ti T, Santavuori P. Enlarged Si and SII somatosensory evoked
gert NA, Lerner TJ, Janes RB,Wallace BA, Munroe PB, O’Rawe responses in the CLN5 form of neuronal ceroid lipofusci-
AM, Gardiner RM, Mole SE. Structure of the CLN3 gene and nosis. Clin Neurophysiol 2002; 113: 1491–1500
predicted structure, location and function of CLN3 protein. Rapola J, Lake BD. Lymphocyte inclusions in Finnish-variant
Neuropediatrics 1997; 28: 12–14 late infantile neuronal ceroid lipofuscinosis (CLN5). Neuro-
Piattella L, Cardinali C, Zamponi N, Papa O. Spielmeyer-Vogt pediatrics 2000; 31: 33–34
disease: clinical and neurophysiological aspects. Child Nerv Rapola J, Lähdetie J, Isosomppi J, Helminen P, Penttinen M,
Syst 1991; 7: 226–230 Järvelä I. Prenatal diagnosis of variant late infantile neu-
Pullarkat RK, Morris GN, Pullerkat PS, LaBadie GU, Zawitosky SE. ronal ceroid lipofuscinosis (vLINCLfinnish; CLN5). Prenat
Biochemical studies of the juvenile form of ceroid-lipofus- Diagn 1999; 19: 685–688
cinosis. Am J Med Genet 1992; 42: 575–579 Santavuori P, Rapola J, Nuutila A, Raininko R, Lappi M, Launes J,
Wisniewski KE, Zhong N Kaczmarski W, Karzmarski A, Sklower- Herva R, Sainio K.The spectrum of Jansky-Bielschowsky dis-
Brooks S, Brown WT. Studies of atypical JNCL suggest over- ease. Neuropediatrics 1991; 22: 92–96
lapping with other NCL forms. Pediatr Neurol 1998; 18: Santavuori P, Rapola J, Raininko R, Autti T, Lappi M, Nuutila A,
36–40 Launes J, Sainio K. Early juvenile neuronal ceroid-lipofusci-
Wisniewski KE, Zhong N, Kaczmarski W, Kaczmarski A, Kida E, nosis or variant Jansky-Bielschowsky disease: diagnostic
Brown WT, Schwarz KO, Lazzirini AM, Rubin AJ, Stenroos ES, criteria and nomenclature. J Inherit Metab Dis 1993; 16:
Johnson WG, Wisniewski TM. Compound heterozygous 230–232
genotype is associated with protracted neuronal ceroid Savukoski M, Klockars T, Holmberg V, Santavuori P, Lander ES,
lipofuscinosis. Ann Neurol 1998; 43: 106–110 Peltonen L. CLN5, a novel gene encoding a putative trans-
membrane protein mutated in Finnish variant late infantile
neuronal ceroid lipofuscinosis. Nat Genet 1998; 19:
Neuronal Ceroid Lipofuscinosis: CLN4 286–288
Tyynelä J, Suopanki J, Santavuori P, Baumann M, Haltia M. Vari-
Augustine A, Fricchione G, Woznicki R, Broberg D, Holt J. Adult ant late infantile neuronal ceroid-lipofuscinosis: pathology
neuronal ceroid lipofuscinosis presenting with psychiatric an biochemistry. J Neuropathol Exp Neurol 1997; 56: 369–
symptoms (case report). Int J Psychiatry Med 1993; 19: 375
315–322 Uvebrant P, Hagberg B. Neuronal ceroid lipofuscinosis in Scan-
Berkovic S, Carpenter S, Andermann F, Andermann E, Wolfe LS. dinavia. Epidemiology and clinical pictures. Neuropedi-
Kufs’ disease: a critical reappraisal. Brain 1988; 111: 27–62 atrics 1997; 28: 6–8
Constantinidis J,Wisniewski KE,Wisniewski TM.The adult and a Varilo T, Savukoski M, Norio R, Santavuori P, Peltonen L, Järvelä
new late adult forms of neuronal ceroid lipofuscinosis. Acta J. The age of human mutation: genealogical and linkage
Neuropathol (Berl) 1992; 83: 461–468 disequilibrium analysis of the CLN5 mutation in the Finnish
Donnet A, Habib M, Pellissier JF, Régis H, Farnarier G, Pelletier J, population. Am J Hum Genet 1996; 58: 506–512
Gosset A, Roger J, Khalil R. Kufs’ disease presenting as pro- Vesa J, Chin MH, Oelgeschläger K, Isosomppi J, DellAngelica EC,
gressive dementia with late-onset generalized seizures: a Jalanko A, Peltonen L. Neuronal ceroid lipofuscinoses are
clinicopathological and electrophysiological study. Epilep- connected at molecular level: interaction of CLN5 protein
sia 1992; 33: 65–74 with CLN2 and CLN3. Mol Biol Cell 2002; 13: 2410–2420
Gelot A, Maurage CA, Rodriguez D. Perrier-Pallissib D, Lar- Williams R, Santavuori P, Peltonen L, Gardiner RM, Järvelá I. A
mande P, Ruchoux MM. In vivo diagnosis of Kufs’ disease by variant form of late infantile neuronal ceroid lipofuscinosis
extracerebral biopsies. Acta Neuropathol (Berl) 1998; (CLN5) is not an allelic form of Batten (Spielmeyer-Vogt-
96:102–108 Sjögren, CLN3) disease: exclusion of linkage to the CLN3 re-
Martin J-J. Adult type of neuronal ceroid-lipofuscinosis. Dev gion of chromosome 16. Genomics 1994; 20: 289–290
Neurosci 1991; 13: 331–338 Wisniewski KE, Kida E, Connell F, Elleder M, Eviatar L, Konkol RJ.
Martin J-J.Adult type of neuronal ceroid-lipofuscinosis.J Inher- New subform of the late infantile form of neuronal ceroid
it Metab Dis 1993; 16: 237–240 lipofuscinosis. Neuropediatrics 1993; 24: 155–163
Tobo M, Misuyama Y, Ikari K, Itio K. familial occurrence of adult-
type neuronal ceroid lipofuscinosis. Arch Neurol 1984; 41: Neuronal Ceroid Lipofuscinosis: CLN 6
1091–1094
Auger KJ, Ajene A, Lerner T. Progress toward the cloning of
Neuronal Ceroid Lipofuscinosis: CLN 5 CLN6, the gene underlying a variant LINCL. Mol Genet
Metab 1999; 66: 332–336
Gao H, Boustany R-MN, Espanola JA, Cotman SL, Srinidhi L, An-
Autti T, Raininko R, Launes J, Nuutila A, Santavuori P, Janski-
tonellis KA, Gillis T, Qin X, Liu S, Donahue LR, Bronson RT,
Bielschowsky variant disease: CT, MRI, and SPECT findings.
Faust JR, Stout D, Haines JL, Lerner TJ, MacDonald ME. Muta-
Pediatr Neurol 1992; 2: 121–126
tions in a novel CLN6-encoded transmembrane protein
cause variant neuronal ceroid lipofuscinosis in man and
mouse. Am J Hum Genet 2002; 70: 324–335
930 References and Further Reading

Heine C, Koch B, Storch S, Kohlschutter A, Palmer DN, Braulke T. 16 Adult Polyglucosan Body Disease
Defective endoplasmic reticulum-resident membrane pro-
tein CLN6 affects lysosomal degradation of endocytosed Berkhoff M, Weis J, Schroth G, Sturzenegger M. Extensive
arylsulfatase A. J Biol Chem 2004; 279: 22347–22352 white-matter changes in case of adult polyglucosan body-
Pena HA, Cardozo JJ, Montiel CM, Molina OM, Boustany R-M. disease. Neuroradiology 2001; 43: 234–236
Serial MRI findings in the Costa Rican variant of neuronal Bigio EH,Weiner MF, Bonte FJ,White CL. Familial dementia due
ceroid-lipofuscinosis. Pediatr Neurol 2001; 25: 78–80 to adult polyglucosan bodydisease. Clin Neuropathol 1997;
Sharp JD,Wheeler RB, Lake BD, Fox M, Gardiner RM,Williams RE. 16: 227–234
Genetic and physical mapping of the CLN6 gene on chro- Boulan-Predseil P, Vital A, Brochet B, Darriet D, Henry P, Vital C.
mosome 15q21–23. Mol Genet Metab 1999; 66: 329–331 Dementia of frontal lobe due to adult polyglucosan body-
Wheeler RB, Sharp JD, Schultz RA, Joslin JM, Williams RE, Mole disease. J Neurol 1995; 242: 512–516
SE. The gene mutated in variant late-infantile neuronal Bruno C, Servidei S, Shanske S, Karpati G, Carpenter S, McKee D,
ceroid lipofuscinosis (CLN6) and nclf mutant mice encodes Barohn RJ,Hirano M,Rifai Z,DiMauro S.Glycogen branching
a novel predicted transmembrane protein. Am J Hum enzyme deficiency in adult polyglucosan bodydisease.Ann
Genet 2002; 70: 537–542 Neurol 1993; 33: 88–93
Busard HLSM, Gabreëls-Festen AAWM, Reinier WO, Gabreëls
FJM, Joosten EGM, van ‘t Hof MS, Rensing JBM. Adult
Neuronal Ceroid Lipofuscinosis: CLN 7 polyglucosan bodydisease: the diagnostic value of axilla
skin biopsy. Ann Neurol 1991; 29: 448–451
Mitchell WA,Wheeler RB, Sharp JD, Bate SL, Gardiner RM, Ranta Cafferty MS, Lovelace RE, Hays AP, Servidei S, DiMauro S, Row-
US, Lonka L,Williams RE, Lehesjoki AE, Mole SE.Turkish vari- land LP. Polyglucosan body disease. Muscle Nerve 1991; 14:
ant late infantile neuronal ceroid lipofuscinosis (CLN7) may 102–107
be allelic to CLN8. Eur J Paediatr Neurol 2001; 5: 21–27 Cavanagh JB. Corpora-amylacea and the family of polyglu-
Topçu M, Tan H, Yalnizoglu D, Usubütün A, Saatçi I, Aynaci M, cosan diseases. Brain Res Rev 1999; 29: 265–295
Anlar B, Topaloglu H, Turanli G, Köse G, Aysun S. Evaluation Gray F, Gherardi R, Mashall A, Path MRC, Janota I, Poirier J. Adult
of 36 patients from Turkey with neuronal ceroid lipofusci- polyglucosan body disease (APBD) J Neuropathol Exp Neu-
nosis: clinical, neurophysiological, neuroradiological and rol 1988; 47: 259–474
histopathologic studies.Turk J Pediatr 2004; 46: 1–10 Klein CM, Peter Bosch E, Dyck PJ. Probable adult polyglucosan
Wheeler RB, Sharp JD, Mitchell WA, Bate SL, Williams WA, Lake bodydisease. Mayo Clinic Proc 2000; 75: 1327–1331
BD, Gardiner RM. A new locus for variant late infantile neu- Lossos A, Baresh V, Soffer D, Argov Z, Gomori M, Ben-Nariah Z,
ronal ceroid lipofuscinosis-CLN7. Mol Genet Metab 1999; Abramsky O, Steiner I. Hereditary branching enzyme dys-
66: 337–378 function in adult polyglucosan bodydisease: a possible
metabolic cause in two patients. Ann Neurol 1991; 30:
655–662
Neuronal Ceroid Lipofuscinosis: CLN8 McDonald TD, Faust PL, Bruno C, DiMauro S, Goldman JE.
Polyglucosan bodydisease simulating amyotrophic lateral
sclerosis. Neurology 1993; 43: 785–790
Hirvasniemi A, Lang H, Lehesjoki A-E, Leisti KJ. Northern epilep-
Milde P, Guccion JG, Kelly J, Locatelli E, Jones RV. Adult polyglu-
sy syndrome: an inherited childhood-onset epilepsy with
cosan bodydisease. Diagnosis by sural nerve and skin biop-
associated mental deterioration. J Med Genet 1994; 31:
sy. Arch Pathol Lab Med 2001; 125: 519–522
177–182
Moses SW, Parvari R. The variable presentations of glycogen
Lonka L, Kyttälä A, Ranta S, Jalanko A, Lehesjoki A-E. The neu-
storage disease type IV: a review of clinical, enzymatic and
ronal ceroid lipofuscinosis CLN8 membrane protein is a res-
molecular studies. Curr Mol Med 2002; 2: 177–188
ident of the endoplasmic reticulum. Hum Mol Genet 2000;
Negishi C, Sze G. Spinal cord MRI in adult polyglucosan body
9: 1691–1697
disease. J Comput Assist Tomogr 1992; 16: 824–826
Ranta S, Lehesjoki A-E. Northern epilepsy, a new member of
Okamoto K, Llena JF, Hirano A. A type of adult polyglucosan
the NCL family. Neurol Sci 2000; 21: S43–S47
bodydisease. Acta Neuropathol (Berl) 1982; 58: 73–77
Ranta S, Lehesjoki A-E, Hirvasniemi A, Weissenbach J, Ross B,
Rifai Z, Klitzke M,Tawil R, Kazee AM, Shanske S, DiMauro S, Grig-
Leal SM, de la Chapelle A, Gilliam TC. Genetic and physical
gs RC. Dementia of adult polyglucosan body disease. Evi-
mapping of the progressive epilepsy with mental retarda-
dence of cortical and subcortical dysfunction. Arch Neurol
tion (EPMR) locus on chromosome 8p. Genome Res 1996; 6:
1994; 51: 90–94
351–360
Robertson NP, Wharton S, Anderson J, Scolding NJ. Adult
Ranta S, Zhang Y, Ross B, Lonka L, Takkunen E, Messer A, Sharp
polyglucosan body disease associated with an extrapyra-
J, Wheeler R, Kusumi K, Mole S. Liu W, Soares MB, de Fatima
midal syndrome. J Neurol Neurosurg Psychiatry 1998; 65:
Bonaldo M, Hirvasniemi A, de la Chapelle A, Gilliam TC,
788–790
Lehejoski A-E.The neuronal ceroid lipofuscinosis in human
Robitaille Y, Carpenter S, Karpati G, DiMauro S. A distinct form
EPMR and mnd mutant mice are associated with mutations
of adult polyglucosan bodydisease with massive involve-
in CLN8. Nat Genet 1999; 23: 233–236
ment of central and pheripheral neuronal processes and
Tahvanainen E,Ranta S,Hirvasniemi A.Karila E,Leisti J,Sistonen
astrocytes. A report of four cases and a review of the oc-
P, Weissenbach J, Lehesjoki E-E, de la Chapelle A. The gene
curence of polyglucosan bodies in other conditions such as
for a recessively inherited human childhood progressive
Lafora’s disease and normal ageing. Brain 1980; 103: 315–
epilepsy with mental retardation maps to the distal short
336
arm for chromosome 8. Proc Natl Acad Sci 1994; 91:
7267–7270
References and Further Reading 931

Sindern E, Patzold T, Vorgerd M, Shin YS, Podskarbi T, Schröder Moser HW, Smith KD, Watkins PA, Powers J, Moser AB. X-Linked
JM, Malin JP. Adult polyglucosan body disease. Report of a Adrenoleukodystrophy. In: Scriver CR, Beaudet AL, Sly WS,
case with predominant involvement of the central and pe- Valle D, eds. The metabolic and molecular bases of inherit-
ripheral nervous system and branching enzyme deficiency ed disease. New York: McGraw-Hill, 2001, pp 3257–3301
in leukocytes. Nervenarzt 1999; 70: 745–749 Motley AM, Brites P, Gerez L, Hogenhout EM, Haasjes J, Benne R,
Sindern E, Ziemssen F, Ziemssen T, Podskarbi T, Shin Y, Brasch F, Tabak HF,Wanders RJA,Waterham HR.Mutational spectrum
Müller KM, Schröder JM, Malin JP,Vorgerd M. Adult polyglu- in the PEX7 gene and functional analysis of mutant alleles
cosan body disease. A postmortem correlation study. Neu- in 78 patients with rhizomelic chondrodysplasia punctata
rology 2003; 61: 263–265 type 1. Am J Hum Genet 2002; 70: 612–624
Vos AJM, Joosten EGM, Gabreëls-Festen AAWM. Adult polyglu- Poll-The BT, Saudubray JM, Ogier HA, Odievre M, Scotto JM,
cosan body disease: clinical and nerve biopsy findings in Monnens L, Govaerts LC, Roels F, Cornelis A, Schutgens RBH.
two cases. Ann Neurol 1983; 13: 440–444 Infantile Refsum disease: an inherited peroxisomal disor-
Wierzba-Bobrowicz T, Stroinska-Kús B. Adult polyglucosan der. Comparison with Zellweger syndrome and neonatal
body disease. Folia Neuropathol 1994; 32: 37–41 adrenoleukodystrophy. Eur J Pediatr 1987; 146: 477–483
Ziemssen F, Sindern E, Michael Schröder J, Shin YS, Zange J, Kil- Setchell KD, Heubi JE, Bove KE, O’Connell NC, Brewsaugh T,
imann MW, Malin J-P,Vorgerd M. Novel missence mutations Steinberg SJ, Moser A, Squires RH Jr.Liver disease caused by
in the glycogen-branching enzyme gene in adult polyglu- failure to racemize trihydroxycholestanoic acid: gene mu-
cosan body disease. Ann Neurol 2000; 47: 536–540 tation and effect of bile acid therapy. Gastroenterology
2003; 124: 217–232
Shimozawa N,Suzuki Y,Orii T,Moser A,Moser HW,Wanders RJA.
Standardization of complementation grouping of peroxi-
17 Peroxisomes and Peroxisomal Disorders some-deficient disorders and the second Zellweger patient
with peroxisomal assembly factor-1 (PAF-1) defect. Am J
Aubourg P, Scotto J, Rocchiccioli F, Feldmann-Pautrat D, Robain Hum Genet 1993; 52: 843–844
O. Neonatal adrenoleukodystrophy. J Neurol Neurosurg Van den Brink DM, Brites P, Haasjes J, Wierzbicki AS, Mitchell J,
Psychiatry 1986; 49: 77–86 Lambert-Hamill M, de Belleroche J, Jansen GA, Waterham
Barth PG, Gootjes J, Bode H, Vreken P, Majoie CB, Wanders RJA. HR,Wanders RJA. Identification of PEX7 as the second gene
Late onset white matter disease in peroxisome biogenesis involved in Refsum disease. Am J Hum Genet 2003; 72:
disorder. Neurology 2001; 57: 1949–1955 471–477.
Barth PG, Majoie CB, Gootjes J,Wanders RJA,Waterham HR, van van der Knaap MS,Valk J.The MR spectrum of peroxisomal dis-
der Knaap MS, de Klerk JB, Smeitink J, Poll-The BT. Neuro- orders. Neuroradiology 1991; 33: 30–37
imaging of peroxisome biogenesis disorders (Zellweger Wanders RJA. Metabolic and molecular basis of peroxisomal
spectrum) with prolonged survival. Neurology 2004; 62: disorders: a review. Am J Med Genet 2004; 126: 355–375
439–444 Wanders RJA,Heymans HSA,Schutgens RBH,Barth PG,van den
Braverman N, Chen L, Lin P, Obie C, Steel G, Douglas P, Bosch H, Tager JM. Peroxisomal disorders in neurology. J
Chakraborty PK, Clarke JT, Boneh A, Moser A, Moser H, Valle Neurol Sci 1988; 88: 1–39
D. Mutation analysis of PEX7 in 60 probands with rhizomel- Wanders RJA, Barth PG, Heymans HSA. Single peroxisomal en-
ic chondrodysplasia punctata and functional correlations zyme deficiencies. In: Scriver CR, Beaudet AL, Sly WS,Valle D,
of genotype with phenotype. Hum Mutat 2002; 20: 284– eds. The metabolic and molecular bases of inherited dis-
297 ease. New York: McGraw-Hill, 2001, pp 3219–3256.
Corzo D, Gibson W, Johnson K, Mitchell G, LePage G, Cox GF, Wanders RJA, Jakobs C, Skjeldal OH. Refsum disease. In: Scriver
Casey R, Zeiss C, Tyson H, Cutting GR, Raymond GV, Smith CR, Beaudet AL, Sly WS,Valle D, eds.The metabolic and mol-
KD,Watkins PA, Moser AB, Moser HW, Steinberg SJ. Contigu- ecular bases of inherited disease.. New York: Mc Graw-Hill,
ous deletion of the X-linked adrenoleukodystrophy gene 2001, pp 3303–3321
(ABCD1) and DXS1357E: a novel neonatal phenotype simi- Wanders RJA,Vreken P, Ferdinandusse S, Jansen GA,Waterham
lar to peroxisomal biogenesis disorders. Am J Hum Genet HR, van Roermund CWT, van Grunsven EG. Peroxisomal fat-
2002; 70: 1520–1531 ty acid alpha- and beta-oxidation in humans: enzymology,
Ferdinandusse S, Denis S, Clayton PT, Graham A, Rees JE, Allen peroxisomal metabolite transporters and peroxisomal dis-
JT, Mclean BN, Brown AY, Vreken P, Waterham HR, Wanders eases. Biochem Soc Trans 2001; 29: 250–267
RJA. Mutations in the gene encoding peroxisomal alpha-
methylacyl-CoA racemase cause adult-onset sensory mo-
tor neuropathy. Nat Genet 2000; 24: 188–191
Gould SJ, Valle D. Peroxisome biogenesis disorders: genetics 18 Peroxisome Biogenesis Defects
and cell biology.Trends Genet 2000; 16: 340–345
Gould SJ, Raymond GV, Valle D. The peroxisomal biogenesis Agamanolis DP, Patre S. Glycogen accumulation in the central
disorder. In: Scriver CR, Beaudet AL, Sly WS, Valle D, eds. The nervous system in the cerebro-hepato-renal syndrome.
metabolic and molecular bases of inherited disease. New J Neurol Sci 1979; 41: 325–334
York: Mc Graw-Hill, 2001, pp 3181–3217 Agamanolis DP, Robinson HB, Timmons GD. Cerebro-hepato-
Kelley RI, Datta NS, Dobyns WB, Hajra AK, Moser AB, Noetzel MJ, renal syndrome.Report of a case with histochemical and ul-
Zackai EH, Moser HW. Neonatal adrenoleukodystrophy: trastructural observations. J Neuropathol Exp Neurol 1976;
new cases, biochemical studies, and differentiation from 35: 226–246
Zellweger and related peroxisomal polydystrophy syn- Aikawa J, Chen WW, Kelley RI,Tada K, Moser HW, Chen GL. Low-
dromes. Am J Med Genet 1986; 23: 869–901 density particles (W-particles) containing catalase in Zell-
Mannaerts GP, van Veldhoven PP. Functions and organization weger syndrome and normal fibroblasts. Proc Natl Acad Sci
of peroxisomal beta-oxidation. Ann NY Acad Sci 1996; 804: USA 1991; 88: 10084–10088
99–115
932 References and Further Reading

Aubourg P, Robain O, Rocchiccioli F, Dancea S, Scotto J. The Evrard P, Caviness VS, Prats-Vinas J, Lyon G. The mechanism of
cerebro-hepato-renal (Zellweger) syndrome: lamellar lipid arrest of neuronal migration in the Zellweger malforma-
profiles in adrenocortical, hepatic mesenchymal, astrocyte tion: an hypothesis based upon cytoarchitectonic analysis.
cells and increased levels of very long chain fatty acids and Acta Neuropathol (Berl) 1978; 41: 109–117
phytanic acid in the plasma. J Neurol Sci 1985; 69: 9–25 Ferninandusse S, Denis S, Mooijer PAW, Zhang Z, Reddy JK,
Aubourg P,Scotto J,Rocchiccioli R,Feldmann-Pautrat D,Robain Spector AA, Wanders RJA. Identification of the peroxisomal
O. Neonatal adrenoleukodystrophy. J Neurol Neurosurg b-oxidation enzymes involved in the biosynthesis of do-
Psychiatry 1986; 49: 77–86 cosahexaenoic acid. J Lipid Res 2001; 42: 1987–1995
Bader PI, Dougherty S, Cangany N, Raymond G, Jackson CE. In- Folz SJ, Trobe JD. The peroxisome and the eye. Surv Ophthal-
fantile Refsum disease in four Amish sibs. Am J Med Genet mol 1991; 35: 353–368
2000; 90: 110–114 Ghaedi K, Honsho M, Shimozawa N, Suzuki Y, Kondo N, Fujiki Y.
Barkovich AJ, Peck WW. MR of Zellweger syndrome. AJNR Am J PEX3 is the casual gene responsible for peroxisome mem-
Neuroradiol 1997; 18: 1163–1170 brane assembly-defective Zellweger syndrome of comple-
Barth PG, Schutgens RBH, Bakkeren JAJM, Dingemans KP, Hey- mentation group G. Am J Hum Genet 2000; 67: 976–981
mans HSA, Douwes AC, van der Klei-van Moorsel JM. A Goldfischer S, Powers JM, Johnson AB, Axe S, Brown FR, Moser
milder variant of Zellweger syndrome. Eur J Pediatr 1985; HW. Striated adrenocortical cells in cerebro-hepato-renal
144: 338–342 (Zellweger) syndrome. Virchows Arch 1983; 401: 355–361
Barth PG, Wanders RJA, Schutgens RBH, Bleeker-Wagemakers Gould SJ, Valle D. Peroxisome biogenesis disorders. Genetics
EM, van Heemstra D. Peroximal b-oxidation defect with de- and cell biology.Trends Genet 2000; 16: 340–345
tectable peroxisomes: a case with neonatal onset and pro- Govaerts L, Sippell WG, Monnens L. Further analysis of the dis-
gressive course. Eur J Pediatr 1990; 149: 722–726 turbed adrenocortical function in the cerebro-hepato-re-
Barth PG, Gootjes J, Bode H, Vreken P, Majoie CBLM, Wanders nal syndrome of Zellweger. J Inherit Metab Dis 1989; 12:
RJA. Late onset white matter disease in peroxisome bio- 423–428
genesis disorder. Neurology 2001; 57: 1949–1955 Groenendaal F, Bianchi MC, Battini R, Tosetti M, Boldrini A, de
Barth PG, Majoie CBLM, Gootjes J, Wanders RJA, Waterham HR, Vries LS, Cioni G. Proton magnetic resonance spectroscopy
van der Knaap MS,de Klerk JBC,Smeitink J,Poll-The BT.Neu- (1H-MRS) of the cerebrum in two young infants with Zell-
roimaging of peroxisome biogenesis disorders (Zellweger weger syndrome. Neuropadiatrics 2001; 32: 23–27
spectrum) with prolonged survival. Neurology 2004; 62: Heikoop JC, van den Berg M, Strijland A, Weijers PJ, Just WW,
439–444 Meijer AJ,Tager JM.Turnover of peroxisomal vesicles by au-
Beard ME, Moser AB, Sapirstein V, Holtzman E. Peroxisomes in tophagic proteolysis in cultured fibroblasts from Zellweger
infantile phytanic acid storage disease: a cytochemical patients. Eur J Cell Biol 1992; 59: 165–171
study of skin fibroblasts. J Inherit Metab Dis 1986; 9: 321– Heymans HSA, Schutgens RB, Tan R, van den Bosch H, Borst P.
334 Severe plasmalogen deficiency in tissues of infants without
Benke PJ, Reyes PF, Parker JC Jr. New form of adrenoleukodys- peroxisomes (Zellweger syndrome). Nature 1983; 306:
trophy. Hum Genet 1981; 58: 204–208 69–70
Bizzozero OA, Ziñiga G, Lees MB. Fatty acid composition of hu- Holmes RD, Wilson GN, Hajra A. Oral ether lipid therapy in pa-
man myelin proteolipid protein in peroxisomal disorders. tients with peroxisomal disorders. J Inherit Metab Dis 1987;
J Neurochem 1991; 56: 872–878 10: 239–241
Brosius U, Gärtner J. Cellular and molecular aspects of Zell- Imamura A, Shimozawa N, Suzuki Y, Zhang Z,Tsukamoto T, Fuji-
weger syndrome and other peroxisome biogenesis disor- ki Y, Osumi T, Wanders RJA, Kondo N.Temperature-sensitive
ders. Cell Mol Life Sci 2002; 59: 1058–1069 mutations of PEX6 in peroxisome biogenesis disorders in
Budden SS, Kennaway NG, Buist NRM, Poulos A, Weleber RG. complementation group C (CG-C): comparative study of
Dysmorphic syndrome with phytanic acid oxidase defi- PEX6 and PEX1. Pediatr Res 2000; 48: 541–545
ciency, abnormal very long chain fatty acids, and pipecolic Jaffe R, Crumrine P, Hashida Y, Moser HW. Neonatal adreno-
acidemia:studies in four children.J Pediatr 1986; 108:33–39 leukodystrophy. Clinical, pathologic, and biochemical de-
Burdette DE, Kremser K, Fink JK, Pahan K, Stanley W, Sing I. Late- lineation of a syndrome affecting both males and females.
onset generalized disorder of peroxisomes. Neurology Am J Pathol 1982; 108: 100–111
1996; 46: 829–831 Janssen A, Baes M, Gressens P, Mannaerts GP, Declerq P, van
Chow CW, Poulos A, Fellenberg AJ, Christodoulou J, Danks DM. Veldhoven PP. Docosahexaemoic acid deficit is not a major
Autopsy findings in two siblings with infantile Refsum dis- pathogenic factor in peroxisome-deficient mice. Lab Invest
ease. Acta Neuropathol (Berl) 1992; 83: 190–195 2000; 80: 31–35
De Leon GA, Grover WD, Huff DS, Moringo-Meure G, Punnett Kelley RI,Datta NA,Dobyns WB,Hajra AK,Moser AB,Noetzel MJ,
HH, Kistenmacher ML. Globoid cells, glial nodules, and pe- Zackal EH, Moser HW. Neonatal adrenoleukodystrophy:
culiar fibrillary changes in the cerebro-hepato-renal syn- new cases, biochemical studies, and differentiation from
drome of Zellweger. Ann Neurol 1977; 2: 473–484 Zellweger and related peroxisomal polydystrophy syn-
Dimmick JE, Applegarth DA. Pathology of peroxisomal disor- dromes. Am J Med Genet 1986; 23: 869–901
ders. Perspect Pediatr Pathol 1993; 17: 45–98 Liu HM, Bangaru BS, Kiddd J, Boggs J. Neuropathological con-
Dodt G, Braverman N, Wong C, Moser A, Moser HW, Watkins P, siderations in cerebro-hepato-renal syndrome (Zellweger’s
Valle D, Gould SJ. Mutations in the PTS1 receptor gene, syndrome) Acta Neuropathol (Berl) 1976; 34: 115–123
PXR1, define complementation group 2 of the peroxisome Manz HJ, Schuelein M, McCullough DC, Kishimoto Y, Eiben RM.
biogenesis disorders. Nat Genet 1995; 9: 115–125 New phenotypic variant of adrenoleukodystrophy. Patho-
Dubois J, Sebag G, Argyropoulou M, Brunelle F. MR findings in logic, ultrastructural, and biochemical study in two broth-
infantile Refsum disease: case report of two family mem- ers. J Neurol Sci 1980; 45: 245–260
bers. AJNR Am J Neuroradiol 1991; 12: 1159–1160 Martinez M. Restoring the DHA levels in the brain of Zellweger
patients. J Mol Neurosci 2001; 16: 309–316
References and Further Reading 933

Martinez M, Vasques E. MRI evidence that docosahexaenoic Preuss N, Brosius U, Biermanns M, Muntau AC, Conzelmann E,
acid ethyl ester improves myelination in generalized perox- Gärtner J. PEX1 Mutations in complementation group 1 of
isomal disorders. Neurology 1998; 51: 26–32 Zellweger spectrum patients correlate with severity of dis-
Matsumoto N, Tamura S, Moser A, Moser HW, Braverman N, ease. Pediatr Res 2002; 51: 706–714
Suzuki Y, Shimozawa N, Kondo N, Fujiki Y.The peroxin Pex6p Raafat F, Smith K, Halloran EA, Lacy D. Zellweger syndrome: a
gene is impaired in peroxisomal biogenesis disorders of histochemical diagnosis of two cases. Pediatr Pathol 1991;
complementation group 6. J Hum Genet 2001; 46: 273–277 11: 413–420
Matsumoto N, Tamura S, Furuki S, Miyata N, Moser A, Shimoza- Raas-Rothshild A, Wanders RJA, Mooijer PAW, Gootjes J, Water-
wa N, Moser HW, Suzuki Y, Kondo N, Fujiki Y. Mutations in ham HR, Gutman A, Suzuki Y, Shimozawa N, Kondo N, Eshel
novel peroxin gene PEX26 that causes peroxisome-biogen- G, Espeel M, Roels F, Korman SH. A PEX6-defective peroxiso-
esis disorders of complementation group 8 provide a mal biogenesis disorder with severe phenotype in an in-
genotype-phenotype correlation. Am J Hum Genet 2003; fant,versus mild phenotype resembling Usher syndrome in
73: 233–246 the affected parents. Am J Hum Genet 2002; 70: 1062–1068
Matsumoto N, Tamura S, Fujiki Y. The pathogenic peroxin Raymond GV. Peroxisomal disorders. Curr Opin Neurol 2001;
Pex26p recruits the Pex1p-Pex6p AAA ATPase complexes to 14: 783–787
peroxisomes. Nat Cell Biol 2003; 5: 454–60 Reuber BE, Germain-Lee E, Collins CS, Morrell JC, Ameritunga R,
Maxwell MA, Allen T, Solly PB, Svingen T, Paton BC, Crane DI. Moser HW,Valle D, Gould SJ. Mutations in PEX1 are the most
Novel PEX1 mutations and genotype-phenotype correla- common cause of peroxisome biogenesis disorders. Nat
tions in Australian peroxisome biogenesis disorder pa- Genet 1997; 17: 445–448
tients. Hum Mutat 2002; 20: 342–351 Robertson EF, Poulos A, Sharp P, Manson J,Wise G, Jaunzems A,
Mito T,Takada K, Akaboshi S,Takashima S,Takeshita K, Origuchi Carter R. Treatment of infantile phytanic acid storage dis-
Y. A pathological study of a peripheral nerve in a case of ease: clinical, biochemical and ultrastructural findings in
neonatal adrenoleukodystrophy. Acta Neuropathol (Berl) two children treated for 2 years. Eur J Pediatr 1988; 147:
1989; 77: 437–440 133–142
Muntau AC, Mayerhofer PU, Paton PC, Kammerer S, Roscher Roels F, Espeel M, de Craemer D. Liver pathology and immuno-
AA. Defective peroxisome membrane synthesis due to mu- cytochemistry in congenital peroxisomal diseases: a re-
tations in human PEX3 causes Zellweger syndrome, com- view. J Inherit Metab Dis 1991; 14; 853–875
plementation group G. Am J Hum Genet 2000; 67: 967–975 Roels F, Espeel M, Poggi F, Mandel H, van Maldergem L,
Nakai A, Shigematsu Y, Nishida K, Kikawa Y, Konishi Y. MRI find- Saudubray JM. Human liver pathology in peroxisomal dis-
ings of Zellweger syndrome. Pediatr Neurol 1995; 13: eases: a review including novel data. Biochimie 1993; 75:
346–348 281–292
Okumoto K, Fujiki Y.PEX12 encodes an integral membrane pro- Sarnat HB,Trevenen CL, Darwish HZ. Ependymal abnormalities
tein of peroxisomes. Nat Genet 1997; 17: 265–266 in cerebro-hepato-renal disease of Zellweger. Brain Dev
Panjan DJ, Megliè NP, Neubauer D. A case of Zellweger syn- 1993; 15: 270–277
drome with extensive MRI abnormalities and unusual EEG Schutgens RBH, Schrakamp G, Wanders RJA, Heymans HSA,
findings. Clin Electroencephalogr 2001; 32: 28–31 Tager JM, van den Bosch H. Prenatal and perinatal diagno-
Passarge E, McAdams AJ. Cerebro-hepato-renal syndrome. sis of peroxisomal disorders. J Inherit Metab Dis 1989; 12:
J Pediatr 1967; 71: 691–702 118–134
Poll-The BT, Poulos A, Sharp P, Boue J, Ogier H, Odièvre M, Scotto JM,Hadchouel M,Odievre M,Laudat MH,Saudubray JM,
Saudubray JM. Antenatal diagnosis of infantile Refsum’s Dulac O, Beucler I, Beaune P. Infantile phytanic acid storage
disease. Clin Genet 1985; 27: 524–526 disease, a possible variant of Refsum’s disease: three cases,
Poll-The BT,Gootjes J,Duran M,de Klerk JBC,Wenniger-Prick LJ, including ultrastructural studies of the liver. J Inherit Metab
Admiraal RJC, Waterham HR, Wanders JA, Barth PG. Peroxi- Dis 1982;5: 83–90
some biogenesis disorders with prolonged survival: phe- Sharp P, Johnson D, Poulos A. Molecular species of phos-
notypic expression in a cohort of 31 patients. Am J Med phatidylcholine containing very long chain fatty acids in
Genet [A] 2004; 126: 333–338 human brain: enrichment in X-linked adrenoleukodystro-
Portsteffen H, Beyer A, Becker E, Epplen C, Pawlak A, Kunau W- phy brain and diseases of peroxisome biogenesis brain.
H, Dodt G. Human PEX1 is mutated in complementation J Neurochem 1991; 56: 30–37
group 1 of the peroxisome biogenesis disorders. Nat Genet Shimozawa N, Suzuki Y, Zhang Z, Imamura A, Ghaedi K, Fujiki Y,
1997; 17: 449–452 Kondo N. Identification of PEX3 as the gene mutated in a
Poulos A, Sharp P, Johnson D. Plasma polyenoic very-long- Zellweger syndrome patient lacking peroxisomal remnant
chain fatty acids in peroxisomal disease: biochemical dis- structures. Hum Mol Genet 2000; 9: 1995–1999
crimination of Zellweger’s syndrome from other pheno- Slawekci ML, Dodt G, Steinberg S, Moser AB, Moser HW, Gould
types. Neurology 1989; 39: 44–47 SJ. Identification of three distinct peroxisomal protein im-
Powers JM. The pathology of peroxisomal disorders with port defects in patients with peroxisome biogenesis disor-
pathogenetic considerations. J Neuropathol Exp Neurol ders. J Cell Sci 1995; 108: 1817–1829
1995; 54: 710–719 Torvik A,Torp S, Kase BF, Ek J, Skjeldal O, Stokke O. Infantile Ref-
Powers JM, Tummons RC, Caviness VS, Moser AB, Moser HW. sum’s disease: a generalized peroxisomal disorder. Case re-
Structural and chemical alterations in the cerebral malde- port with postmortem examination. J Neurol Sci 1988; 85:
velopment of fetal cerebro-hepato-renal (Zellweger) syn- 39–53
drome. J Neuropathol Exp Neurol 1989; 48: 270–289 Ulrich J, Herschkowitz N, Heitz Ph, Sigrist Th, Baerlocher P.
Powers JM, Moser HG. Peroxisomal disorders: genotype, phe- Adrenoleukodystrophy. Preliminary report of a connatal
notype, major neuropathologic lesions, and pathogenesis. case. Light- and electron microscopical immunohisto-
Brain Pathol 1998; 8: 101–120 chemical and biochemical findings. Acta Neuropathol
(Berl) 1978: 43: 77–83
934 References and Further Reading

Vamecq J, Draye JP, van Hoof F, Misson JP, Evrard P, Verellen G, Clayton PT, Lake BD, Hjelm M, Stephenson JBP, Besley GTN,
Eyssen HJ, van Eldere J, Schutgens RH,Wanders RJA, Roels F, Wanders RJA, Schram AW, Tager JM, Schutgens RBH, Law-
Goldfischer SL. Multiple peroxisomal enzymatic deficiency son AM. Bile acid analysis in “pseudo-Zellweger”syndrome;
disorders. A comparative biochemical and morphologic clues to the defect in peroxisomal b-oxidation. J Inherit
study of Zellweger cerebrohepatorenal syndrome and Metab Dis 1988; 11: 165–168
neonatal adrenoleukodystrophy. Am J Pathol 1986; 125: Espeel M, Roels F, van Maldergem L, de Craemer D, Dacremont
524–535 G, Wanders RJA, Hashimoto T. Peroxisomal localization of
Van Roermund CWT, Brul S, Tager JM, Schutgens RBH, Wan- the immunoreactive b-oxidation enzymes in a neonate
ders RJA. Acyl-CoA oxidase, peroxisomal thiolase and dihy- with a b-oxidation defect. Pathological observations in liv-
droxyacetone phosphate acyltransferase: aberrant subcel- er, adrenal cortex and kidney. Virchows Arch [A] 1991; 419:
lular localization in Zellweger syndrome. J Inherit Metab 301–308
Dis 14: 152–164 Ferdinandusse S, van Grunsven EG, Oostheim W, Denis S,
Volpe JJ, Adams RD. Cerebro-hepato-renal syndrome of Zell- Hogenhout EM, Ijlst L, van Roermund CWT, Waterham HR,
weger: an inherited disorder of neuronal migration. Acta Goldfischer S,Wanders RJA. Reinvestigation of peroxisomal
Neuropathol (Berl) 1972; 20: 175–198 3-ketoacyl-CoA thiolase deficiency: identification of the
Walter C, Gootjes J, Mooijer PA, Portsteffen H, Klein C, Water- true defect at the level of D-bifunctional protein. Am J Hum
ham HR, Barth PG, Epplen JT, Kunau W-H, Wanders RJA, Genet 2002; 70: 1589–1593
Dodt G. Disorders of peroxisome biogenesis due to muta- Goldfischer S, Collins J, Rapin I, Neumann P, Neglia W, Spiro AJ,
tions in PEX1: phenotypes and PEX1 protein levels. Am J Ishii T, Roels F, Vamecq J, van Hoof F. Pseudo-Zellweger syn-
Hum Genet 2001; 69: 35–48 drome: deficiencies in several peroxisomal oxidative activi-
Wanders RJA, Boltshauser E, Steinmann B, Spycher MA, Schut- ties. J Pediatr 1986; 108: 25–32
gens RBH, Bosch van den H, Tager JM. Infantile phytanic Itoh M, Suzuki Y, Akaboshi S, Zhang Z, Miyabara S,Takashima S.
acid storage disease, a disorder of peroxisome biogenesis: Developmental and pathological expression of peroxiso-
a case report. J Neurol Sci 1990; 98: 1–11 mal enzymes: their relationship of D-bifunctional protein
Wanders RJA, Schutgens RBH, van den Bosch H, Tager JM, Klei- deficiency and Zellweger syndrome. Brain Res 2000; 858:
jer WJ. Prenatal diagnosis of inborn errors in peroxisomal 40–47
b-oxidation. Prenat Diagn 1991; 11: 253–261 Kaufman WE, Theda C, Naidu S, Watkins PA, Moser AB, Moser
Wanders RJA, Schutgens RBH, Barth PG, Tager JM, van den HW. Neuronal migration abnormality in peroxisomal bi-
Bosch H. Postnatal diagnosis of peroxisomal disorders: a functional enzyme defect. Ann Neurol 1996; 39: 268–271
biochemical approach. Biochimie 1993; 75: 269–279 McGuinness MC, Moser AB, Poll-The BT, Watkins PA. Comple-
Warren JS, Morrell JC, Moser HW, Valle D, Gould SJ. Identifica- mentation analysis of patients with intact peroxisomes and
tion of PEX10, the gene defective in complementation impaired peroxisomal b-oxidation. Biochem Med Metab Bi-
group 7 of the peroxisome-biogenesis disorders.Am J Hum ol 1993; 49: 228–242
Genet 1998; 63: 347–359 Möller G, van Grunsven EG,Wanders RJA, Adamski J. Molecular
Warren DS, Wolfe BD, Gould SJ. Phenotype-genotype relation- basis of D-bifunctional protein deficiency. Mol Cell En-
ships in PEX10-deficient peroxisome biogenesis disorder docrinol 2001; 171: 61–70
patients. Hum Mutat 2000; 15: 509–521 Naidu S, Hoefler G, Watkins PA, Chen WW, Moser AB, Hoefler S,
Wei H, Kemp S, McGuiness MC, Moser AB, Smith KD. Pharmaco- Rance NE, Powers JM, Beard M, Green WR, Hashimoto T,
logical induction of peroxisomes in peroxisome biogenesis Moser HW. Neonatal seizures and retardation in a girl with
disorders. Ann Neurol 2000; 47: 286–296 biochemical features of X-linked adrenoleukodystrophy: a
Wilson GN, Holmes RD, Hajra AK. Peroxisomal disorders: clinical possible new peroxisomal disease entity. Neurology 1988;
commentary and future prospects. Am J Med Genet 1988; 38: 1100–1107
30: 771–792 Nakada Y, Hyakuna N, Suzuki Y, Shimozawa N,Takaesu E, Ikema
Wolff J, Nyhan WL, Powell H, Takahashi D, Hutzler J, Hajra AK, R, Hirayama K. A case of pseudo-Zellweger syndrome with
Datta NS, Singh I, Moser HW. Myopathy in an infant with a a possible bifunctional enzyme deficiency but detectable
fatal peroxisomal disorder. Pediatr Neurol 1986; 2: 141–146 enzyme protein. Brain Dev 1993; 15: 453–456
Yahruas T,Braverman N,Dodt G,Kalish JE,Morrell JC,Moser HW, Nakano K, Zhang Z, Shimozawa N, Kondo N, Ishii N, Funatsuka
Valle D,Gould S.The peroxisome biogenesis disorder group M, Shirakawa S, Itho M, Takashima S, Une M, Kana-aki RR,
4 gene, PXAAA1, encodes a cytoplasmic ATPase required for Mukai K, Osawa M, Suzuki Y. D-Bifunctional protein defi-
stability of the PTS1 receptor. EMBO J 1996; 15: 2914–2923 ciency with fetal ascites, polyhydramnios, and contractures
of hands and toes. J Pediatr 2001; 139: 865–867
Paton BC, Solly PB, Nelson PV, Pollard AN, Sharp PC, Fietz MJ.
Molecular analysis of genomic DNA allows rapid, and accu-
19 Peroxisomal D-Bifunctional Protein rate, prenatal diagnosis of peroxisomal D-bifunctional pro-
Deficiency tein deficiency. Prenat Diagn 2002; 22: 38–41
Suzuki Y, Shimoxawa N, Yajima S, Tomatsu S, Kondo N, Nakada
Akaboshi S, Tomita Y, Suzuki Y, Une M, Sohma O, Takashima S, Y, Akaboshi S, Iai M, Tanabe Y, Hashimoto T, Wanders RJA,
Takeshita K. Peroxisomal bifunctional protein deficiency: Schitgens RBH, Moser HW, Orii T.Novel subtype of peroxiso-
serial neurophysiological examinations of a case. Brain Dev mal acyl-CoA oxidase deficiency and bifunctional protein
1997; 19: 295–299 enzyme deficiency with detectable enzyme protein: identi-
Clayton PT. Clinical consequences of defects in peroxisomal fication by means of complementation analysis. Am J Hum
b-oxidation. Biochem Soc Trans 2001; 29: 298–305 Genet 1994; 54: 35–43
References and Further Reading 935

Suzuki Y, Jiang LL, Souri M, Miyazawa S, Fukuda S, Zhang Z, Une 20 Acyl-CoA Oxidase Deficiency
M, Shimozawa N, Kondo N, Orii T, Hashimoto T. D-3-Hy-
droxy-CoA dehydratase / D-3-hydroxyacul-CoA dehydro- Clayton PT. Clinical consequences of defects in peroxisomal
genase bifunctional protein deficiency: a newly identified b-oxidation. Biochem Soc Trans 2001; 29: 298–305
peroxisomal disorder. Am J Hum Genet 1997; 61: 1153– Christensen E, Woldseth B, Hagve T-A, Poll-The BT, Wanders
1162 RJA, Sprecher H, Stokke O, Christophersen BO. Peroxisomal
Suzuki Y, Zhang Z, Shimozawa N, Muro M, Shono H, Toda S, b-oxidation of polyunsaturated long chain fatty acids in
Miyahara S-I,Hashimoto T,Usuda N,Ito M,Takashima S,Kon- human fibroblasts. The polyunsaturated and the saturated
do N. Prenatal diagnosis of peroxisomal D-3-hydroxyacyl- long chain fatty acids are retroconverted by the same acyl-
CoA dehydratase / D-3-hydroxyacyl-CoA dehydrogenase CoA oxidase. Scand J Clin Lab Invest 1993; 53: 61–74
bifunctional protein deficiency. J Hum Genet 1999; 44: Kurian MA,Ryan S,Besley GTN,Wanders RJA,King MD.Straight-
143–147 chain acyl-CoA oxidase deficiency presenting with dysmor-
Van Grunsven EG, van Roermund CWT, Denis S, Wanders RJA. phia, neurodevelopmental autistic type regression and a
Complementation analysis of fibroblasts from peroxisomal selective pattern of leukodystrophy. J Inherit Metab Dis
fatty acid oxidation deficient patients shows high frequen- 2004; 27: 105–108
cy of bifunctional protein enzyme deficiency plus intra- Mandel H, Berant M, Aizin A, Gershony R, Hemmli S, Schutgens
genic complementation: unequivocal evidence for differ- RBH, Wanders RJA. Zellweger-like phenotype in two sib-
ential defects in the same enzyme protein. Biochem Bio- lings: a defect in peroxisomal b-oxidation with elevated
phys Res Commun 1997; 235: 176–179 very long-chain fatty acids but normal bile acids. J Inherit
Van Grunsven EG, van Berkel E, Ijlst L, Vreken P, De Klerk JBC, Metab Dis 1992; 15: 381–384
Adamski J, Lemonde H, Clayton PT, Cuebas DA, Wanders Poll-The BT,Roels F,Ogier H,Scotto J,Vamecq J,Schutgens BBH,
RJA. Peroxisomal D-hydroxyacyl-CoA dehydrogenase defi- Wanders RJA, van Roermund CWT, van Wijland MJA,
ciency: resolution of the enzyme defect and its molecular Schram AW, Tager JM, Saudubray J-M. A new peroxisomal
basis in bifunctional protein deficiency. Proc Natl Acad Sci disorder with enlarged peroxisomes and a specific defi-
1998; 95: 2128–2133 ciency of acyl-CoA oxidase (pseudo-neonatal adrenoleuko-
Van Grunsven EG, van Berkel E, Lemonde H, Clayton PT, Wan- dystrophy). Am J Hum Genet 1988; 42: 422–434
ders RJA. Bifunctional protein deficiency: complementa- Su H-M, Moser AB, Moser HW,Watkins PA.Peroxisomal straight-
tion within the same group suggesting differential enzyme chain acyl-CoA oxidase and D-bifunctional protein are es-
defects and clues to the underlying basis. J Inherit Metab sential for the retroconversion step in docosahexaenoic
Dis 1998; 21: 298–301 acid synthesis. J Biol Chem 2001; 276: 38115–38120
Van Grunsven EG, van Berkel E, Mooijer PAW,Watkins PA, Moser Suzuki Y, Shimoxawa N, Yajima S, Tomatsu S, Kondo N, Nakada
HW, Suzuki Y, Jiang LL, Hashimoto T, Hoefler G, Adamski J, Y, Akaboshi S, Iai M, Tanabe Y, Hashimoto T, Wanders RJA,
Wanders RJA. Peroxisomal bifunctional protein deficiency Schitgens RBH, Moser HW, Orii T.Novel subtype of peroxiso-
revisited:resolution of its true enzymatic and molecular ba- mal acyl-CoA oxidase deficiency and bifunctional protein
sis. Am J Hum Genet 1999; 64: 99–107 enztme deficiency with detectable enzyme protein: identi-
Van Grunsven EG,Mooijer PAW,Aubourg P,Wanders RJA.Enoyl- fication by means of complementation analysis. Am J Hum
CoA hydratase deficiency: identification of a new type of D- Genet 1994; 54: 35–43
bifunctional protein deficiency. Hum Mol Genet 1999; 8: Suzuki Y, Iai M, Kamei A, Tanabe Y, Chida S,Yamaguchi S, Zhang
1509–1516 Z, Takamoto Y, Shimozawa N, Kondo N. Peroxisomal acyl
Van Maldergem L, Espeel M, Wanders RJA, Roels F, Gerard P, CoA oxidase deficiency. J Pediatr 2002; 140: 128–130
Scalais E, Mannaerts GP, Casteels M, Gillerot Y. Neonatal Watkins PA, McGuiness MC, Raymond GV, Hicks BA, Sisk JM,
seizures and severe hypotonia in a male infant suffering Moser AB, Mower HW. Distinction between peroxisomal bi-
from a defect in peroxisomal b-oxidation. Neuromusc Dis- functional enzyme and acyl-CoA oxidase deficiencies. Ann
ord 1992; 2: 217–234 Neurol 1995; 38: 472–477
Wanders RJA, van Roermund CWT, Schelen A, Schutgens RBH,
Tager JM, Stephenson JBP, Clayton PT. A bifunctional pro-
tein with deficient enzymatic activity: identification of a
new peroxisomal disorder using novel methods to mea- 21 X-Linked Adrenoleukodystrophy
sure the peroxisomal b-oxidation enzyme activities.J Inher-
it Metab Dis 1990; 13: 375–379 Afifi AK,Menezes AH,Reed LA,Bell WE.Atypical presentation of
Wanders RJA,van Roermund CWT,Brul S,Schutgens RBH,Tager X-linked childhood adrenoleukodystrophy with an unusu-
JM. Bifunctional protein deficiency: identification of a new al magnetic resonance imaging pattern. J Child Neurol
type of peroxisomal b-oxidation of unknown aetiology by 1996; 11: 497–499
means of complementation analysis. J Inherit Metab Dis Asano J-I, Suzuki Y, Yajima S, Iioue K, Shimozawa N, Kondo N,
1992; 15: 385–388 Murase M, Orii T. Effects of erucic acid therapy on Japanese
Watkins PA, Chen WW, Harris CJ, Hoefler G, Hoefler S, Blake DC patients with X-linked adrenoleukodystrophy. Brain Dev
Jr, Balfe A, Kelley RI, Moser AB, Beard ME, Moser HW. Peroxi- 1994; 16: 454–458
somal bifunctional protein deficiency.J Clin Invest 1989; 83: Aubourg P, Dubois-Dalcq M. X-linked adrenoleukodystrophy
771–777 enigma: how does the ALD peroxisomal transporter muta-
Watkins PA, McGuiness MC, Raymond GV, Hicks BA, Sisk JM, tion affect CNS glia? Glia 2000; 29: 186–190
Moser AB, Mower HW. Distinction between peroxisomal bi- Aubourg P, Blanche S, Jambaqué I, Rocchiccioli F, Kalifa G,
functional enzyme and acyl-CoA oxidase deficiencies. Ann Naud-Saudreau C, Rolland MO, Debré M, Chaussain JL,
Neurol 1995; 38: 472–477 Griscelli C, Fischer A, Bougnères P-F. Reversal of early neuro-
logic and neuroradiologic manifestations of X-linked
adrenoleukodystrophy by bone marrow transplantation.
N Engl J Med 1990; 322: 1860–1866
936 References and Further Reading

Aubourg P, Adamsbaum C, Lavallard-Rousseau M-C, Rocchicci- Edwin D,Speedie LJ,Kohler W,Naidu S,Kruse B,Moser HW.Cog-
oli F, Cartier N, Jambaqué I, Jakobezak C, Lemaitre A, nitive and brain magnetic resonance imaging findings in
Boureau F, Wolf C, Bougnères P-F. A two-year trail of oleic adrenomyeloneuropathy. Ann Neurol 1996; 40: 675–678
and erucic acids (“Lorenzo’s oil”) as treatment for adreno- Eichler FS, Barker PB, Cox C, Edwin D, Ulug AM, Moser HW, Ray-
myeloneuropathy. N Engl J Med 1993; 329: 745–752 mond GV.Proton MR spectroscopic imaging predicts lesion
Aubourg P, Mandel JL. X-linked adrenoleukodystrophy. Ann NY progression or MRI in X-linked adrenoleukodystrophy.Neu-
Acad Sci 1996; 804: 461–476 rology 2002; 58: 901–907
Barkovich AJ, Ferriero DM, Bass N, Boyer R. Involvement of the Eichler FS, Itoh R, Barker PB, Mori S, Garrett ES, van Zijl PCM,
pontomedullary corticospinal tracts: a useful finding in the Moser HW, Raymond GV, Melhem ER. Proton MR spectro-
diagnosis of X-linked adrenoleukodystrophy. AJNR Am J scopic and diffusion tensor brain MR imaging in X-linked
Neuroradiol 1997; 18: 95–100 adrenoleukodystrophy: initial experience. Radiology 2002;
Baumann M, Korenke GC, Widdige-Diedrichs A, Wilichowski E, 225: 245–252
Hunneman DH, Wilken B, Brockmann K, Klingebiel T, Ni- Elrington GM, Bateman DE, Jeffrey MJ, Flawton N. Adreno-
ethammer D, Kühl J, Ebell W, Hanefeld F. Haematopoietic leukodystrophy: heterogeneity in two brothers. J Neurol
stem cell transplantation in 12 patients with cerebral X- Neurosurg Psychiatry 1989; 52: 310–313
linked adrenoleukodystrophy. Eur J Pediatr 2003; 162: 6–14 Engelbrecht V, Rassek M, Gärtner J, Kahn T, Mödder U. The val-
Bekiesiñska-Figatowska M, Tylki-Szymañska A, Walekci J, ue of new MRI techniques in adrenoleukodystrophy. Pedi-
Stradomska TJ. MRI findings in a asymptomatic boy with X- atr Radiol 1997; 27: 207–215
linked adrenoleukodystrophy and his symptomatic moth- Farrell DF, Hamilton SR, Knauss TA, Sanocki E, Deeb SS. X-Linked
er. Neuroradiology 2001;43: 951–952 adrenoleukodystrophy: adult cerebral variant. Neurology
Bezman L, Moser AB, Raymond GV, Rinaldo P,Watkins PA, Smith 1993; 43: 1518–1522
KD, Kass NE, Moser HW. Adrenoleukodystrophy: incidence, Fatemi A,Barker PB,Uluğ AM,Nagae-Poetcher LM,Beauchamp
new mutation rate, and results of extended family screen- NJ, Moser AB, Raymond GV, Moser HW, Naidu S. MRI and
ing. Ann Neurol 2001; 49: 512–517 proton MRSI in women heterozygous for X-linked adreno-
Borker A, Yu LC. Unrelated allogeneic bone marrow transplant leukodystrophy. Neurology 2003; 60: 1301–1307
in adrenoleukodystrophy using CD34+ stem cell selection. Feigenbaum V, Gélot A, Casanova P, Daumas-Duport C,
Metab Brain Dis 2002; 17: 139–142 Aubourg P, Dubouis-Dalcq M. Apoptosis in the central ner-
Boutin B, Matsuguchi L, Lebon P, Ponsol G, Arthuis C. Immuno- vous system of cerebral adrenoleukodystrophy patients.
histochemical analysis of brain macrophages in adreno- Neurobiol Dis 2000; 7: 600–612
leukodystrophy. Neuropediatrics 1989; 20: 202–206 Gärtner J, Braun A, Holzinger A, Roerig P, Lenard H-G, Roscher
Brown FR, Chen WW, Kirschner DA, Frayer KL, Powers JM, Moser AA. Clinical and genetic aspects of X-linked adrenoleuko-
AB, Moser HW. Myelin membrane from adrenoleukodystro- dystrophy. Neuropediatrics 1998; 29: 3–13
phy brain white matter – biochemical properties. J Neuro- Garg BP, Markand ON, DeMeyer WE, Warren C Jr. Evoked re-
chem 1983; 41: 341–348 sponse studies in patients with adrenoleukodystrophy and
Cartier N, Guidoux S, Rocchiccioli F, Aubourg P. Simvastatin heterozygous relatives. Arch Neurol 1983; 40: 356–359
does not normalize very long chain fatty acids in Garside S, Rosebush PI, Levinson AJ, Mazurek MF. Late-onset
adrenoleukodystrophy mice. FEBS Lett 2000; 478: 205–208 adrenoleukodystrophy associated with long-standing psy-
Confort-Gouny S, Vion-Dury J, Chabrol B, Nicoli F, Cozzone PJ. chiatric symptoms. J Clin Psychiatry 1999; 60: 460–468
Localised proton magnetic resonance spectroscopy in X- Hershkovitz E, Narkis G, Shorer Z, Moser AB, Watkins PA, Moser
linked adrenoleukodystrophy. Neuroradiology 1995; 37: HW, Manor E. Cerebral X-linked adrenoleukodystrophy in a
568–575 girl with Xq27-Ter deletion. Ann Neurol 2002; 52: 234–237
Di Rocco M, Doria-Lamba L, Caruso U. Monozogotic twins with Hitomi T, Mezaki T, Tsujii T, Kinoshita M, Tomimoto H, Ikeda A,
X-linked adrenoleukodystrophy and different phenotypes. Shimohama S, Okazaki T, Uchiyama T, Shibasaki H. Improve-
Ann Neurol 2001; 50: 424 ment of central motor conduction after bone marrow
Dodd A, Rowland SA, Hawkes SLJ, Kennedy MA, Love DR. Muta- transplantation in adrenoleukodystrophy. J Neurol Neuro-
tions in the adrenoleukodystrophy gene. Hum Mutat 1997; surg Psychiatry 2003; 74: 373–375
9: 500–511 Hong-Magno ET,Muraki AS,Huttenlocher PR.Atypical CT scans
Domagk J, Linke I, Argyrakis A, Spaar FW, Rahlf G, Schulte FJ. in adrenoleukodystrophy. J Comput Assist Tomogr 1987;
Adrenoleukodystrophy. Neuropediatrics 1975; 6: 41–64 11: 333–336
Dubois-Dalcq M, Feigenbaum V, Aubourg P. The neurobiology Igarashi M, Schaumburg HH, Powers J, Kishimoto Y, Kolodny E,
of X-linked adrenoleukodystrophy, a demyelinating perox- Suzuki K. Fatty acid abnormality in adrenoleukodystrophy.
isomal disorder.Trends Neurosci 1999; 22: 4–12 J Neurochem 1976; 26: 851–860
Dumic M, Gubarev n, Sikic N, Roscher A, Plavsic V, Filipovic-Gr- Ito M, Blumberg BM, Mock DJ, Goodman AD, Moser AB, Moser
cic B. Sparse hair and multiple endocrine disorders in two HW,Smith KD,Powers JM.Potential environmental and host
women heterozygous for adrenoleukodystrophy. Am J participants in the early white matter lesions of adreno-
Med Genet 1992; 43: 829–832 leukodystrophy: morphologic evidence for CD8 cytotoxic T
Dunne E, Hyman NM, Huson SM, Németh AH. A novel point cells, cytolysis of oligodendrocytes, and CD-1 mediated
mutation in X-linked adrenoleukodystrophy presenting as lipid antigen presentation. J Neuropathol Exp Neurol 2001;
spinocerebellar degeneration. Ann Neurol 1999; 45: 60: 1004–1019
652–655 Ito R, Melhem ER, Mori S, Eichler FS, Raymond GV, Moser HW.
Dziewas R, Stögbauer F, Oelerich M, Ritter M, Husstedt IW. A Diffusion tensor brain MR imaging in X-linked cerebral
case of adrenomyeloneuropathy with unusual lesion pat- adrenoleukodystrophy. Neurology 2001; 56: 544–547
tern in magnetic resonance imaging. J Neurol 2001; 248: Jensen ME, Sawyer RW, Braun IF, Rizzo WB. MR imaging appear-
341–342 ance of childhood adrenoleukodystrophy with auditory,
visual, and motor pathway involvement. Radiographics
1990; 10: 53–66
References and Further Reading 937

Izquierdo M, Adamsbaum C, Benosman A, Aubourg P, BittounJ. Loes DJ, Stillman AE, Hite S, Shapiro E, Lockman L, Latchaw RE,
MR spectroscopic imaging of normal-appearing white Moser H, Krivit W. Childhood cerebral form of adreno-
matter in adrenoleukodystrophy. Pediatr Radiol 2000; 30: leukodystrophy: short-term effect of bone marrow trans-
621–629 plantation on brain MR observations. AJNR Am J Neurora-
Jorge P, Quelhas D, Oliveira P, Pinto R, Nogueira A. X-Linked diol 1994; 15: 1767–1771
adrenoleukodystrophy in patients with idiopathic Addison Loes DJ, Fatemi A, Melhem ER, Gupte N, Bezman L, Moser HW,
disease. Eur J Pediatr 1994; 153: 594–597 Raymond GV. Analysis of MRI patterns aids prediction of
Kano S, Watanabe M, Kanai M, Koike R, Onodera O, Tsuji S, progression in X-linked adrenoleukodystrophy. Neurology
Okamoto K, Shoji M. A Japanese family with adrenoleuko- 2003; 61: 369–374
dystrophy with a codon 291 deletion: a clinical, biochemi- Maier EM, Kammerer S, Muntau AC,Wichers M, Braun A, Rosch-
cal, pathological, and genetic report. J Neurol Sci 1998; 158: er AA. Symptoms in carriers of adrenoleukodystrophy re-
187–192 late to skewed X Inactivation.Ann Neurol 2002; 52: 683–688
Kaplan PW, Tusa RJ, Shankroff J, Heller J, Moser HW. Visual Maris T, Androulidakis EJ, Tzagournissakis M, Papavassiliou S,
evoked potentials in adrenoleukodystrophy: a trial with Moser H, Plaitakis A. X-linked adrenoleukodystrophy pre-
glycerol trioleate and Lorenzo oil. Ann Neurol 1993; 34: senting as neurologically pure familial spastic paraparesis.
169–174 Neurology 1994; 45: 1101–1104
Kemp S,Mooyer PAW,Bolhuis PA,van Geel BM,Mandel JL,Barth Marsh WW, Hurst DL. Variable phenotypes in a family kindred
PG, Aubourg P,Wanders RJA. ALDP Expression in fibroblasts with adrenoleukodystrophy. Pediatr Neurol 1991; 7: 50–52
of patients with X-linked adrenoleukodystrophy. J Inherit Martin JJ, Ceuterick C, Libert J. Skin and conjunctival nerve
Metab Dis 1996; 19: 667–674 biopsies in adrenoleukodystrophy and its variants. Ann
Kemp S, Wei H-M, Lu J-F, Braiterman LT, McGuinness MC, Moser Neurol 1980; 8: 291–295
AB,Watkins PA,Smith KD.Gene redundancy and pharmaco- McGuinness MC, Lu J-F, Zhang H-P, Dong G-X, Heinzer AK,
logical gene therapy: implications for X-linked adreno- Watkins PA, Powers J, Smith KD. Role of ALDP (ABCD1) and
leukodystrophy. Nat Med 1998; 4: 1261–1268 mitochondria in X-linked adrenoleukodystrophy. Mol Cell
Kemp S, Pujol A, Waterham HR, van Geel BM, Boehm CD, Ray- Biol 2003; 23: 744–753
mond GV, Cutting GR, Wanders RJA, Moser HW. ABCD1 Mu- Melhem ER, Breitner SN, Ulug AM, Raymond GV, Moser HW. Im-
tations and the X-linked adrenoleukodystrophy mutation proved tissue characterization in adrenoleukodystrophy
database: role in diagnosis and clinical correlations. Hum using magnetization transfer imaging. AJR Am J Roent-
Mutat 2001; 18: 499–515 genol 1996; 166: 689–695
Koike R,Tsuji S, Ohno T, Suzuki Y, Orii T, Miyatake T. Physiological Melhem ER, Loes DJ, Georgiades CG, Raymond GV, Moser HW.
significance of fatty acid elongation system in adreno- X-linked adrenoleukodystrophy: the role of contrast-en-
leukodystrophy. J Neurol Sci 1991; 103: 188–194 hanced MR imaging in predicting disease progression.
Kok F, Neumann S, Sarde C-O, Zheng S,Wu K-H,Wei H-M, Bergin AJNR Am J Neuroradiol 2000; 21: 839–844
J, Watkins PA, Gould S, Sack G, Moser H, Mandel J-L, Smith Melhem ER, Gotwald TF, Itho R, Zinreich SJ, Moser HW.T2 relax-
KD. Mutational analysis of patients with X-linked adreno- ation measurements in X-linked adrenoleukodystrophy
leukodystrophy. Hum Mutat 1995; 6: 104–115 performed using dual-echo fast fluid-attenuated inversion
Korenke GC, Christen H-J, Kruse B, Hunneman DH, Hanefeld F. recovery MR imaging. AJNR Am J Neuroradiol 2001; 22:
Progression of X-linked adrenoleukodystrophy under in- 733–776
terferon-b therapy. J Inherit Metab Dis 1997; 20: 59–66 Mo YH, Chen YF, Liu HM. Adrenomyeloneuropathy, a dynamic
Kukowski B. Magnetic transcranial brain stimulation and mul- disorder: brain magnetic resonance imaging of two cases.
timodality evoked potentials in an adrenoleukodystrophy Neuroradiology 2004; 46: 296–300
patient and members of his family.Electroencephalogr Clin Molzer B, Bernheimer H, Budka H, Pilz P,Toifl K. Accumulation of
Neurophysiol 1991; 78: 260–262 very long chain fatty acids is common to 3 variants of
Kumar AJ, Rosenbaum AE, Naidu S, Wener L, Citrin CM, Linden- adrenoleukodystrophy (ALD). J Neurol Sci 1981; 51: 301–
berg R, Kim WS, Zinreich SJ, Molliver ME, Mayberg HS, Moser 310
HW. Adrenoleukodystrophy: correlating MR imaging with Moser H, Dubey P, Fatemi A. Progress in X-linked adrenoleuko-
CT. Radiology 1987; 165: 497–504 dystrophy. Curr Opin Neurol 2004; 17: 263–269
Kumar AJ, Köhler W, Kruse B, Naidu S, Bergin A, Edwin D, Moser Moser HW. Adrenoleukodystrophy: natural history treatment
HW. MR findings in adult-onset adrenoleukodystrophy. and outcome. J Inherit Metab Dis 1995; 18: 435–447
AJNR Am J Neuroradiol 1995; 16: 1227–1237 Moser HW. Adrenoleukodystrophy. Curr Opin Neurol 1995; 8:
Kurihara M, Kumagai K, Yagishita S, Imai M, Watanabe M, Suzu- 221–226
ki Y, Orii T. Adrenoleukomyeloneuropathy presenting as Moser HW. Adrenoleukodystrophy: phenotype, genetics,
cerebellar ataxia in a young child: a probable variant of pathogenesis and therapy. Brain 1997; 120: 1485–1508
adrenoleukodystrophy. Brain Dev 1993; 15: 377–380 Moser HW, Moser AB, Frayer KK, Chen W, Schulman JD, O’Neill
Kurihara M, Kumagai K, Noda Y, Yagishita S. An autopsy case of BP, Kishimoto Y. Adrenoleukodystrophy: increased plasma
atypical adrenoleukomyeloneuropathy in childhood. Brain content of saturated very long chain fatty acids. Neurology
Dev 2000; 22: 394–397 1981; 31: 1241–1249
Kusaka H, Imai T. Ataxic variant of adrenoleukodystrophy: MRI Moser HW, Moser AB, Naidu S, Bergin A. Clinical aspects of
and CT findings. J Neurol 1992; 239: 307–310 adrenoleukodystrophy and adrenomyeloneuropathy. Dev
Loes DJ, Hite S, Moser H, Stillman AE, Shapiro E, Lockman L, Neurosci 1991; 13: 254–261
Latchaw RE, Krivit W. Adrenoleukodystrophy: a scoring Moser HW, Moser AB, Smith KD, Bergin A, Borel J, Shankroff J,
method for brain MR observations. AJNR Am J Neuroradiol Stine OC, Merette C, Ott J, Krivit W, Shapiro E.
1994; 15: 1761–1766 Adrenoleukodystrophy:Phenotypic variability and implica-
tions for therapy. J Inherit Metab Dis 1992; 15: 645–664
938 References and Further Reading

Moser HW, Loes DJ, Melhem ER, Raymond GV, Bezman L, Cox Powers JM, Schaumburg HH. Adreno-leukodystrophy (sex-
CS, Lu S-e. X-linked adrenoleukodystrophy: overview and linked Schilder’s disease). A pathogenetic hypothesis
prognosis as a function of age and brain magnetic reso- based on ultrastructural lesions in adrenal cortex, peripher-
nance imaging abnormality. A study involving 372 pa- al nerve and testis. Am J Pathol 1974; 76: 481–491
tients. Neuropediatrics 2000; 31: 227–239 Powers JM, Liu Y, Moser AB, Moser HW. The inflammatory
Moser HW, Brezman L, Lu SE, Raymond GV.Therapy of X-linked myelinopathy of adreno-leukodystrophy: cells, effector
adrenoleukodystrophy: prognosis based upon age and molecules, and pathogenetic implications. J Neuropathol
MRI abnormality and plans for placebo-controlled trails. J Exp Neurol 1992; 51: 630–643
Inherit Metab Dis 2000; 23: 273–277 Powers JM, DeCiero DP, Ito M, Moser AB, Moser HW. Adreno-
Moser HW, Raymond GV, Koehler W, Sokolowski P, Hanefeld F, myeloneuropathy: a neuropathologic review featuring its
Korenke GC, Green A, Loes DJ, Hunneman DH, Jones RO, Lu noninflammatory myelopathy. J Neuropathol Exp Neurol
SE,Uziel G,Giros ML,Roels F.Evaluation of the preventive ef- 2000; 59: 89–102
fect of glyceryl trioleate-trierucate (“Lorenzo’s oil”) therapy Powers JM, DeCiero DP, Cox C, Richfield EK, Ito M, Moser AB,
in X-linked adrenoleukodystrophy: results of two concur- Moser HW. The dorsal root ganglia in adrenomyeloneu-
rent trials. Adv Exp Med Biol 2003; 544: 369–387 ropathy: neuronal atrophy and abnormal mitochondria.
Mosser J, Douar AM, Sarde CO, Kioschis P, Feil R, Moser H, Poust- J Neuropathol Exp Neurol 2001; 60: 493–501
ka AM, Mandel JL, Aubourg P. Putative X-linked adreno- Rajanayagam V, Balthazor M, Shapiro EG, Krivit W, Lockman L,
leukodystrophy gene shares unexpected homology with Stillman AE. Proton MR spectroscopy and neuropsycholog-
ABC transporters. Nature 1993; 361: 726–730 ical testing in adrenoleukodystrophy. AJNR Am J Neurora-
Nishio H, Kodama S,Tsubota T,Takumi T,Takahashi T,Yokoyama diol 1997; 18: 1909–1914
S,Matsuo T.Adrenoleukodystrophy without adrenal insuffi- Ramsey RB, Banik NL, Scott T, Davison AN. Neurochemical find-
ciency and its magnetic resonance imaging. J Neurol 1985; ings in adreno-leukodystrophy. J Neurol Sci 1976; 29:
232: 265–270 277–294
Nowaczyk MJM, Saunders EF, Tein I, Blaser SI, Clarke JTR. Im- Ravid S, Diamond AS, Eviatar L. Coma as an acute presentation
munoablation does not delay the neurologic progression of adrenoleukodystrophy. Pediatr Neurol 2000; 22: 237–239
of X-linked adrenoleukodystrophy. J Pediatr 1997; 131: Restuccia D, Di Lazzaro V, Valeriani M, Oliviero A, Le Pera D,
453–455 Colosimo C, Brudi N, Cappa M, Bertini E, Di Biase A, Tonali P.
O’Neill BP, Marmion LC, Feringa ER.The adrenoleukomyeloneu- Neurophysiological abnormalities in adrenoleukodystro-
ropathy complex: expression in four generations. Neurolo- phy carriers. Evidence of different degrees of central ner-
gy 1981; 31: 151–156 vous system involvement. Brain 1997; 120: 1139–1148
O’Neill BP,Moser HW,Saxena KM.Familial X-linked Addison dis- Rizzo WB, Leshner RT, Odone A, Dammann AL, Craft BS, Jensen
ease as an expression of adrenoleukodystrophy (ALD): ele- ME,Jennings SS,Davis S,Jaitly R,Sgro JA .Dietary erucic acid
vated C26 fatty acid in cultured skin fibroblasts. Neurology therapy for X-linked adrenoleukodystrophy. Neurology
1982; 32: 543–547 1989; 39: 1415–1422
O’Neill GN, Aoki M, Brown RH Jr. ABCD1 translation-initiator Ronghe MD, Barton J, Jardine PE, Crowne EC, Webster MH, Ar-
mutation demonstrates genotype-phenotype correlation mitage M, Allen JT, Steward CG. The importance of testing
for AMN. Neurology 2001; 57: 1956–1962 for adrenoleukodystrophy in males with idiopathic Addi-
Pai GS, Khan M, Barbosa E, Key L, Craver JR, Curé JK, Betros R, son’s disease. Arch Dis Child 2002; 86: 185–189
Singh I. Lovastatin therapy for X-linked adrenoleukodystro- Samadja D, Cabre Ph, Kujas M,Vernant J-C, Rivierez M. Intracra-
phy: clinical and biochemical observations on 12 patients. nial hypertension as an unusual presentation of adreno-
Mol Genet Metab 2000; 69: 312–322 leukodystrophy. Eur J Neurol 1997; 4: 413–415
Paintlia AS, Gilg AG, Khan M, Singh AK, Barbosa E, Singh I. Cor- Schaumburg HH, Powers JM, Raine CS, Suzuki K, Richardson EP.
relation of very long chain fatty acid accumulation and in- Adrenoleukodystrophy: a clinical and pathological study of
flammatory disease progression in childhood X-ALD: impli- 17 cases. Arch Neurol 1975; 32: 577–591
cations for potential therapies. Neurobiol Dis 2003; 14: Schaumburg HH, Powers JM, Raine CS, Spencer PS, Griffin JW,
425–439 Prineas JW, Boehme DM. Adrenomyeloneuropathy: a prob-
Pasco A, Kalifa G, Sarrazin JL, Adamsbaum C, Aubourg P. Contri- able variant of adrenoleukodystrophy. General pathologic,
bution of MRI to the diagnosis of cerebral lesions of neuropathologic, and biochemical aspects. Neurology
adrenoleukodystrophy. Pediatr Radiol 1991; 21: 161–163 1977; 27: 1114–1119
Philips JP, Lockman LA, Shapiro EG, Blazar BR, Loes DJ, Moser Scholte W, Molzer B, Peiffer J, Poremba M, Schumm F, Harzer K,
HW, Krivit W. CSF findings in adrenoleukodystrophy: corre- Schnabel R, Bernheimer H. Adrenoleukodystrophy in an
lation between measures of cytokines, IgG production, and adult female. A clinical, morphological and neurochemical
disease severity. Pediatr Neurol 1994; 10: 289–294 study. J Neurol 1987; 235: 1–9
Poulos A, Gibson R, Sharp P, Beckman K, Grattan-Smith P. Very Schmidt S, Träber F, Block W, Keller E, Pohl C, von Oertzen J,
long chain fatty acids in X-linked adrenoleukodystrophy Schild H, Schlegel U, Klockgether T. Phenotype assignment
brain after treatment with Lorenzo’s oil. Ann Neurol 1994; in symptomatic female carriers of X-linked adrenoleuko-
36: 741–746 dystrophy. J Neurol 2001; 248: 36–44
Powell H, Tindall R, Schultz P, Paa D, O’Brien J, Lampert P. Schneider JFL, Il’yasov KA, Boltshauser E, Hennig J, Martin E.
Adrenoleukodystrophy. Electron microscopic findings. Diffusion tensor imaging in cases of adrenoleukodystro-
Arch Neurol 1975; 32: 250–260 phy: preliminary experience as a marker for early demyeli-
Powers JM, Moser HG. Peroxisomal disorders: genotype, phe- nation? AJNR Am J Neuroradiol 2003; 24: 819–824
notype, major neuropathologic lesions, and pathogenesis. Schöder JM, Mayer M, Weis J. Mitochondrial abnormalities and
Brain Pathol 1998; 8: 101–120 intrafamilial variability of sural nerve biopsy findings in
adrenomyeloneuropathy. Acta Neuropathol (Berl) 1996; 92:
64–69
References and Further Reading 939

Shapiro E, Krivit W, Lockman L, Jambaqué, Peters C, Cowan M, van Geel BM, Bezman L, Loes DJ, Moser HW, Raymond GV. Evo-
Harris R, Blanche S, Bordigoni P, Loes D, Ziegler R, Crittenden lution of phenotypes in adult male patients with X-linked
M, Ris D, Berg B, Cox C, Moser H, Fisher A, Aubourg P. Long- adrenoleukodystrophy. Ann Neurol. 2001; 49: 186–194
term effect of bone-marrow transplantation for childhood- Verrips A, Willemsen MAAP, Rubio-Gozalbo E, de Jong J,
onset cerebral X-linked adrenoleukodystrophy. Lancet Smeitink JAM. Simvastatin and plasma very-long-chain fat-
2000; 356: 713–718 ty acids in X-linked adrenoleukodystrophy. Ann Neurol
Singh I, Khan M, Key L, Pai S. Lovastatin for x-linked adreno- 2000; 47: 552–553
leukodystrophy. N Engl J Med 1998; 339: 702–703 Wanders RJA, van Roermund CWT, Lageweg W, Jakobs BS,
Smith KD, Kemp S, Braiterman LT, Lu J-F, Wei H-M, Geraghty M, Schutgens RBH, Nijenhuis AA, Tager JM. X-Linked adreno-
Stetten G, Bergin JS, Pevsner J, Watkins PA. X-linked leukodystrophy: biochemical diagnosis and enzyme de-
adrenoleukodystrophy: genes, mutations, and phenotypes. fect. J Inherit Metab Dis 1992; 15: 634–644
Neurochem Res 1999; 24: 521–535 Wanders RJA, Mooyer PW, Dekker C, Vreken P. X-Linked
Snyder RD, King JN, Keck GM, Orrison WW. MR imaging of the adrenoleukodystrophy: improved prenatal diagnosis using
spinal cord in 23 subjects with ALD–AMN complex. AJNR both biochemical and immunological methods. J Inherit
Am J Neuroradiol 1991; 12: 1095–1098 Metab Dis 1998; 21: 285–287
Sobue G, Ueno-Natsukari IU, Okamoto H, Connell TA, Aizawa I, Waragai M,Takaya Y, Hayashi M, Shibata N, Kobayashi M. MRI of
Mizoguchi K,Honma M,Ishakawa G,Mitusma T,Natsukari N. adrenoleukodystrophy involving predominantly the cere-
Phenotypic heterogeneity of an adult form of adreno- bellum and brain stem. Neuroradiology 1996; 38: 788–791
leukodystrophy in monozygotic twins. Ann Neurol 1994; Watkins PA, Gould JS, Smith MA, Braiterman LT, Wei H-M, Kok F,
36: 912–915 Moser AB, Moser HW, Smith KD. Altered expression of ALDP
Steinberg SJ, Kemp S, Braiterman LT, Watkins PA. Role of very- in X-linked adrenoleukodystrophy. Am J Hum Genet 1995;
long-chain-acyl-coenzyme A synthetase in X-linked 57: 292–301
adrenoleukodystrophy. Ann Neurol 1999; 46: 409–412 Weinhofer I, Forss-Petter S, Žigman M, Berger J. Cholesterol
Stephenson DJ, Bezman L, Raymond GV. Acute presentation of regulates ABCD2 expression: implications for the therapy of
childhood adrenoleukodystrophy. Neuropediatrics 2000; X-linked adrenoleukodystrophy. Hum Mol Genet 2002; 11:
31: 293–297 2701–2708
Tan E-K, Lim S-H, Chan L-L, Wong M-C, Tan K-P. X-linked Weller M, Liedtke W, Petersen D, Opitz H, Poremba M.Very-late-
adrenoleukodystrophy: spinocerebellar variant.Clin Neurol onset adrenoleukodystrophy: possible precipitation of de-
Neurosurg 1999; 101: 137–140 myelination by cerebral contusion. Neurology 1992; 42:
Tanaka K, Koyama A, Koike R, Ohno T, Atsumi T, Miyatake T. 367–370
Adrenomyeloneuropathy: report of a family and electron Wilken B, Dechent P, Brockmann K, Finsterbusch J, Baumann M,
microscopical findings in peripheral nerve. J Neurol 1985; Ebell W, Korenke GC, Pouwels PJW, Hanefeld FA, Frahm J.
232: 73–78 Quantitative proton magnetic resonance spectroscopy of
Terakawa H, Yanagihara T, Abe K. Unusual lateralization of children with adrenoleukodystrophy before and after
adrenoleukodystrophy lesions on neuroimaging. J Neu- hematopoietic stem cell transplantation. Neuropediatric
roimaging 1997; 7: 121–123 2003; 34: 237–246
Theda C, Moser AB, Powers JM, Moser HW. Phospholipids in X- Wilson R, Sargent JR. Lipid and fatty acid composition of brain
linked adrenoleukodystrophy white matter: fatty acid ab- tissue from adrenoleukodystrophy patients. J Neurochem
normalities before the onset of demyelination. J Neurol Sci 1993; 61: 290–297
1992; 110: 195–204 Zammarchi EM, Donati MA,Tucci F, Fonda C, Fanelli F, Pazzaglia
Uchiyama M, Hata Y,Tada S.MR imaging of adrenoleukodystro- R. Acute onset of X-linked adrenoleukodystrophy mimick-
phy. Neuroradiology 1991; 33: 25–29 ing encephalitis. Brain Dev 1994; 16: 238–240
Uyama E, Iwagoe H, Maeda J, Nakamura M, Terasaki T, Ando M. Zhang LX, Baski R, Fine E, Moser HW. Clinical and electrophysi-
Presenile-onset cerebral adrenoleukodystrophy present- ological improvement of adrenomyeloneuropathy with
ing as Balint’s syndrome and dementia.Neurology 1993;43: steroid treatment. J Neurol Neurosurg Psychiatry 2002; 74:
1249–1251 822–823
Uziel G, Bertini E, Bardelli P, Rimoldi M, Gambetti M. Experience
on therapy of adrenoleukodystrophy and adrenomyelo-
neuropathy. Dev Neurosci 1991; 13: 274–279
van der Knaap MS, Valk J. MR of adrenoleukodystrophy:
22 Refsum Disease
histopathologic correlations. AJNR Am J Neuroradiol 1989;
10: S12-S14 Baumgartner MR, Jansen GA, Verhoeven NM, Mooyer PAW,
van Geel BM, Assies J, Weverling GJ, Barth PG. Predominance Jakobs C, Roels F, Espeel M, Fourmaintraux A, Bellet H, Wan-
of the adrenomyeloneuropathy phenotype of X-linked ders RJA, Saudubray JM. Atypical Refsum disease with
adrenoleukodystrophy in the Netherlands: a survey of 30 pipecolic academia and abnormal catalase distribution.
kindreds. Neurology 1994; 44: 2343–2346 Ann Neurol 2000; 47: 109–113
van Geel BM, Assies J, Wanders RJA, Barth PG. X-linked Cammermeyer J. Neuropathological changes in hereditary
adrenoleukodystrophy: clinical presentation, diagnosis, neuropathies: manifestation of the syndrome here-
and therapy. J Neurol Neurosurg Psychiatry 1997; 63: 4–14 dopathia atactica polyneuritiformis in the presence of in-
van Geel BM, Assies J, Haverkort EB, Koelman JHTM, Verbeeten terstitial hypertrophic polyneuropathy. J Neuropathol Exp
B Jr,Wanders RJA, Barth PG. Progression of abnormalities in Neurol 1956; 15: 340–367
adrenomyeloneuropathy and neurologically asympto- Casteels M, Croes K, van Veldhoven PP, Mannaerts GP. Peroxiso-
matic X-linked adrenoleukodystrophy despite treatment mal localization of a-oxidation in human liver. J Inherit
with “Lorenzo’s oil.”J Neurol Neurosurg Psychiatry 1999; 67: Metab Dis 1997; 20: 665–673
290–299
940 References and Further Reading

Chahal A, Khan H, Pai SG, Barbosa E, Singh I. Restoration of phy- Millar JHD. Refsum disease – the effect of diet. Ulster Med J
tanic acid oxidation in Refsum disease fibroblasts from pa- 1985; 54: 41–45
tients with mutations in the phytanoyl-CoA hydroxylase Nadal N, Rolland MO, Tranchant C, Reutenauer L, Gyapay G,
gene. FEBS Lett 1998; 429: 119–122 Warter JM, Mandel JL, Koenig M. Localization of Refsum
Dick JPR, Gibberd FB, Meeran K, Rose CF. Hypokalaemia in disease with increased pipecolic acidaemia to chromo-
acute Refsum’s disease. J R Soc Med 1993; 86: 171–172 some 10p by homozygosity mapping and carrier testing in
Dickson N,Mortimer JG,Faed JM,Pollard AC,Styles M,Peart DA. a single nuclear family. Hum Mol Genet 1995; 4: 1963–1966
A child with Refsum’s disease: successful treatment with di- Oysu C, Aslan I, Basaran B, Baserer N.The site of the hearing loss
et and plasma exchange. Dev Med Child Neurol 1989; 31: in Refsum’s disease. Int J Pediatr Otorhinolaryngol 2001; 61:
81–97 129–134
Djupesland G, Flottorp G, Refsum S. Phytanic acid storage dis- Plant GR,Hansell DM,Gibberd FB,Sidey MC.Skeletal abnormal-
ease: hearing maintained after 15 years of dietary treat- ities in Refsum’s disease (heredopathia atactica polyneuri-
ment. Neurology 1983; 33: 237–240 tiformis). Br J Radiol 1990; 63: 537–541
Dotti MT, Rossi A, Rizzuto N, Hayek G, Bardelli N, Bardelli AM, Reese H, Bareta J. Heredopathia atactica polyneuritiformis.
Federico A. Atypical phenotype of Refsum’s disease: Clini- J Neuropathol Exp Neurol 1950; 9: 385–395
cal, biochemical, neurophysiological and pathological Singh I, Lazo O, Kalipada P, Singh AK. Phytanic acid a-oxidation
study. Eur Neurol 1985; 24: 85–93 in human cultured skin fibroblasts. Biochim Biophys Acta
Ferdinandusse S, Denis S, Clayton PT, Graham A, Rees JE, Allen 1992; 1180: 221–224
JT, McLean BN, Brown AY, Vreken P, Waterham HR, Singh I, Pahan K, Dhaunsi GS, Lazo O, Ozand P. Phytanic acid
Wanders RJA. Mutations in the gene encoding peroxisomal a-oxidation. J Biol Chem 1993; 268: 9972–9979
a-methylacyl-CoA racemase cause adult-onset sensory Skjeldal OH, Nyberg-Hansen R, Stokke O. Neurological disor-
motor neuropathy. Nat Genet 2000; 24: 188–191 ders and phytanic acid metabolism. Acta Neurol Scand
Flament-Durand J, Noel P, Rutsaert J, Toussaint D, Malmendier 1988; 78: 324–328
C, Lyon G. A case of Refsum’s disease: clinical, pathological, Tranchant C, Aubourg P, Mohr M, Rocchicciolo F, Zaenker C,
ultrastructural and biochemical study. Pathol Eur 1971; 6: Warter JM. A new peroxisomal disease with impaired phy-
172–191 tanic acid and pipecolic acid oxidation. Neurology 1993; 43:
Gibberd FB, Billimoria JD, Goldman JM, Clemens ME, Evans R, 2044–2048
Whitelaw MN, Retsas S, Sherratt RM. Heredopathia atactica Van den Brink DM, Brites P, Haasjes J, Wierzbicki AS, Mitchell J,
polyneuritiformis: Refsum’s disease. Acta Neurol Scand Lambert-Hamill M, de Belleroche J, Jansen GA, Waterham
1985; 72: 1–17 HR,Wanders RJA. Identification of PEX7 as the second gene
Gelot A, Vallat JM, Tabaraud F, Rocchiccioli F. Axonal neuropa- involved in Refsum disease. Am J Hum Genet 2003; 72:
thy and late detection of Refsum’s disease. Muscle Nerve 471–477
1995; 18: 667–670 Verhoeven NM, Schor DSM, Roe CR, Wanders RJA, Jakobs C.
Gordon N, Hudson REB. Refsum’s syndrome heredopathia at- Phytanic acid a-oxidation in peroxisomal disorders: studies
actica polyneuritiformis.A report of three cases, including a in cultured human fibroblasts. Biochim Biophys Acta 1997;
study of the cardiac pathology. Brain 1959; 82: 41–55 1361: 281–286
Harari D, Gibberd FB, Dick JPR, Sidey MC. Plasma exchange in Verhoeven NM, Schor DSM, ten Brink HJ, Wanders RJA, Jakobs
the treatment of Refsum’s disease (heredopathia atactica C. Resolution of the phytanic acid a-oxidation pathway:
polyneuritiformis). J Neurol Neurosurg Psychiatry 1991; 54: identification of pristanal as product of the decarboxyla-
614–617 tion of 2-hydroxyphytanoyl-CoA. Biochem Biophys Res
Hungerbühler JP, Meier C, Rousselle L, Quadri P, Bogousslavsky Commun 1997; 237: 33–36
J.Refsum’s disease: management by diet and plasmaphere- Verhoeven NM, Wanders RJA, Poll-The BT, Saudubray J-M,
sis. Eur Neurol 1985; 24: 153–159 Jakobs C. The metabolism of phytanic acid and pristanic
Jansen GA, Ofman R, Ferdinandusse S, Ijlst L, Muijsers AO, Skjel- acid in man: a review. J Inherit Metab Dis 1998; 21: 697–728
dal OH, Stokke O, Jakobs C, Besley GTN, Wraith JE, Wanders Wall WJH, Worthington BS. Skeletal changes in Refsum’s dis-
RJA. Refsum disease is caused by mutations in the phy- ease. Clin Radiol 1979; 30: 657–659
tanoyl-CoA hydroxylase gene. Nat Genet 1997; 17:190–193 Wanders RJA, Heymans HSA, Schutgens RBH, Poll-Thé BT,
Jansen GA, Hogenhout EM, Ferdinandusse S,Waterham HR, Of- Saudubray JM, Tager JM, Schrakamp G, van den Bosch H.
man R, Jakobs C, Skjeldal OH, Wanders RJA. Human phy- Peroxisomal functions in classical Refsum’s disease: com-
tanoyl-CoA hydroxylase: resolution of the gene structure parison with the infantile form of Refsum’s disease. J Neurol
and the molecular basis of Refsum’s disease. Hum Mol Sci 1988; 84: 147–155
Genet 2000; 9: 1195–1200 Wanders RJA, Jansen GA, Skjeldal OH. Refsum disease, peroxi-
Kuntzer T, Ochsner F, Schmid F, Regli F. Quantitative EMG analy- somes and phytanic acid oxidation: a review. J Neuropathol
sis and longitudinal nerve conduction studies in a Refsum’s Exp Neurol 2001; 60: 1021–1031
disease patient. Muscle Nerve 1993; 16: 857–863 Weinstein R. Phytanic acid storage disease: (Refsum’s disease):
Lemotte PK, Keidel S, Apfel CM. Phytanic acid is a retinoid X re- clinical characteristics, pathophysiology and the role of
ceptor ligand. Eur J Biochem 1996; 236; 328–333 therapeutic apheresis in its management. J Clin Apheresis
Leppert D, Schanz U, Burger J, Gmür J, Blau N,Waespe W. Long- 1999; 14: 181–184
term plasma exchange in a case of Refsum’s disease. Eur Wierzbicki AS, Mitchell J, Lambert-Hammill M, Hancock M,
Arch Psychiatry Clin Neurosci 1991; 241: 82–84 Greenwood J, Sidney MC, de Belleroche J, Gibberd FB. Iden-
MacBrinn M, O’Brien JS. Lipid composition of the nervous sys- tification of genetic heterogeneity in Refsum’s disease. Eur
tem in Refsum’s disease. J Lipid Res 1968; 9; 552–561 J Hum Genet 2000; 8: 649–651
Mihalik SJ, Morrell JC, Kim D, Sacksteder KA, Watkins PA, Gould Wierzbicki AS, Sankaralingam A, Lumb PJ, Hardman TC, Sidey
SJ. Identification of PAHX, a Refsum disease gene. Nat Genet MC, Gibberd FB. Transport of phytanic acid on lipoproteins
1997; 17: 185–189 in Refsum disease. J Inherit Metab Dis 1999; 22: 29–36
References and Further Reading 941

Wierzbicki AS, Lloyd MD, Schofield CJ, Feher MD, Gibberd FB. Coates PM, Tanaka K. Molecular basis of mitochondrial fatty
Refsum’s disease: a peroxisomal disorder affecting phytan- acid oxidation defects. J Lipid Res 1992; 33: 1099–1110
ic acid a-oxidation. J Neurochem 2002; 80: 727–735 Cuezva JM, Flores AI, Liras A, Santaren JF, Alconada A. Molecu-
Wills AJ, Manning NJ, Reilly MM. Refsum’s disease. Q J Med lar chaperones and the biogenesis of mitochondria and
2001; 94:403–406 peroxisomes. Biol Cell 1993; 77: 47–62
Young SP, Johnson AW, Muller DPR. Effects of phytanic acid on De Lonlay P, Valnot I, Barrientos A, Gorbatyuk M, Tzagoloff A,
the vitamin E status,lipid composition and physical proper- Taanman JW,Benayoun E,Chretien D,Kadhom N,Lombes A,
ties of retinal cell membranes: implications for adult Ref- de Baulny HO, Niaudet P, Munnich A, Rustin P, Rotig A. A mu-
sum disease. Clin Sci 2001; 101: 697–705 tant mitochondrial respiratory chain assembly protein
causes complex III deficiency in patients with tubulopathy,
encephalopathy and liver failure.Nat Genet 2001; 29: 57–60
DeVivo DC. The expanding clinical spectrum of mitochondrial
23 Mitochondria disorders. Brain Dev 1993; 15: 1–22
and Mitochondrial Disorders De Vries DD, Ruitenbeek W, de Wijs IJ, Trijbels JM, van Oost BA.
Enzymological versus DNA investigations in mitochondrial
Andreu AL, Bruno C, Dunne TC, Tanji K, Shanske S, Sue CM, (encephalo-myopathies).J Inherit Metab Dis 1993;16:534–536
Krishna S, Hadjigeorgiou GM, Shtilbans A, Bonilla E, DiMau- DiMauro S, Moraes CT. Mitochondrial encephalomyopathies.
ro S. A nonsense mutation (G15059A) in the cytochrome b Arch Neurol 1993; 50: 1197–1208
gene in a patient with exercise intolerance and myoglobin- DiMauro S, Schon EA. Mitochondrial respiratory-chain dis-
uria. Ann Neurol 1999; 45: 127–130 eases. N Engl J Med 2003; 348: 2656–2668
Andreu AL, Tanji K, Bruno C, Hadjigeorgiou GM, Sue CM, Jay C, Glick B, Schatz G. Import of proteins into mitochondria. Annu
Ohnishi T, Shanske S, Bonilla E, DiMauro S. Exercise intoler- Rev Genet 1991; 25: 21–44
ance due to a nonsense mutation in the mtDNA ND4 gene. Harding AE, Hammans SR. Deletions of the mitochondrial
Ann Neurol 1999; 45: 820–823 genome. J Inherit Metab Dis 1992; 15: 480–486
Astuti D, Latif F, Dallol A, Dahia PL, Douglas F, George E, Skold- Heuvel L, Smeitink JAM. The oxidative phosphorylation
berg F, Husebye ES, Eng C, Maher ER. Gene mutations in the (OXPHOS) system: nuclear genes and human genetic dis-
succinate dehydrogenase subunit SDHB cause susceptibil- eases. Bioessays 2001; 23: 518–525
ity to familial pheochromocytoma and to familial paragan- Hsieh RH, Li JY, Pang CY, Wei YH. A novel mutation in the mito-
glioma. Am J Hum Genet 2001; 69: 49–54 chondrial 16S rRNA gene in a patient with MELAS syn-
Benz R. Biophysical properties of porin pores from mitochon- drome, diabetes mellitus, hyperthyroidism and cardiomy-
drial outer membrane of eukaryotic cells. Experientia 1990; opathy. J Biomed Sci 2001; 8: 328–335
46: 131–137 Loeffen JL, Smeitink JAM, Trijbels JM, Janssen AJ, Triepels RH,
Bosbach S, Kornblum C, Schroder R, Wagner M. Executive and Sengers RC, van den Heuvel LP. Isolated complex I deficien-
visuospatial deficits in patients with chronic progressive cy in children: clinical, biochemical and genetic aspects.
external ophthalmoplegia and Kearns-Sayre syndrome. Hum Mutat 2000; 15: 123–134
Brain 2003; 126: 1231–1240 Poulton J. Duplications of mitochondrial DNA: implications for
Bourgeron T, Rustin P, Chretien D, Birch-Machin M, Bourgeois pathogenesis. J Inherit Metab Dis 1992; 15: 487–498
M, Viegas-Pequignot E, Munnich A, Rotig A. Mutation of a Rahman S,Taanman JW, Cooper JM, Nelson I, Hargreaves I, Me-
nuclear succinate dehydrogenase gene results in mito- unier B, Hanna MG, Garcia JJ, Capaldi RA, Lake BD, Leonard
chondrial respiratory chain deficiency. Nat Genet 1995; 11: JV, Schapira AH. A missense mutation of cytochrome oxi-
144–149 dase subunit II causes defective assembly and myopathy.
Brown MD,Voljavec AS, Lott MT,Torroni A,Yang CC,Wallace DC. Am J Hum Genet 1999; 65: 1030–1039
Mitochondrial DNA complex I and III mutations associated Rosenberg MJ, Agarwala R, Bouffard G, Davis J, Fiermonte G,
with Leber’s hereditary optic neuropathy. Genetics 1992; Hilliard MS, Koch T, Kalikin LM, Makalowska I, Morton DH,
130: 163–73 Petty EM, Weber JL, Palmieri F, Kelley RI, Schaffer AA,
Bruno C, Martinuzzi A, Tang Y, Andreu AL, Pallotti F, Bonilla E, Biesecker LG. Mutant deoxynucleotide carrier is associated
Shanske S, Fu J, Sue CM, Angelini C, DiMauro S, Manfredi G. with congenital microcephaly.Nat Genet 2002; 32: 175–179
A stop-codon mutation in the human mtDNA cytochrome Saada A, Shaag A, Mandel H, Nevo Y, Eriksson S, Elpeleg O. Mu-
c oxidase I gene disrupts the functional structure of com- tant mitochondrial thymidine kinase in mitochondrial DNA
plex IV. Am J Hum Genet 1999; 65: 611–620 depletion myopathy. Nat Genet 2001; 29: 342–344
Bruno C, Sacco O, Santorelli FM, Assereto S, Tonoli E, Bado M, Scholte HR, Busch HF, Luyt-Houwen IE, Vaandrager-Verduin
Rossi GA, Minetti C. Mitochondrial myopathy and respirato- MH, Przyrembel H, Arts WF. Defects in oxidative phosphory-
ry failure associated with a new mutation in the mitochon- lation. Biochemical investigations in skeletal muscle and
drial transfer ribonucleic acid glutamic acid gene. J Child expression of the lesion in other cells. J Inherit Metab Dis
Neurol 2003; 18: 300–303 1987; 10: 81–97
Cheng MY,Hartl FU,Martin J,Pollock RA,Kalousek F,Neupert W, Shoubridge EA. Cytochrome c oxidase deficiency. Am J Med
Hallberg EM, Hallberg RL, Horwich AL. Mitochondrial heat- Genet 2001; 106: 46–52
shock protein hsp60 is essential for assembly of proteins Simon DK, Friedman J, Breakefield XO, Jankovic J, Brin MF,
imported into yeast mitochondria. Nature 1989; 337: Provias J, Bressman SB, Charness ME, Tarsy D, Johns DR,
620–625 Tarnopolsky MA. A heteroplasmic mitochondrial complex I
Chol M, Lebon S, Benit P, Chretien D, de Lonlay P, Goldenberg A, gene mutation in adult-onset dystonia. Neurogenetics
Odent S, Hertz-Pannier L, Vincent-Delorme C, Cormier- 2003; 4: 199–205
Daire V, Rustin P, Rotig A, Munnich A. The mitochondrial Smeitink JA. Mitochondrial disorders: clinical presentation and
DNA G13513A MELAS mutation in the NADH dehydroge- diagnostic dilemmas. J Inherit Metab Dis 2003; 26: 199–207
nase 5 gene is a frequent cause of Leigh-like syndrome Smeitink J, van den Heuvel L, DiMauro S. The genetics and
with isolated complex I deficiency. J Med Genet 2003; 40: pathology of oxidative phosphorylation. Nat Rev Genet
188–191 2001; 2: 342–352
942 References and Further Reading

Smeitink JAM, van den Heuvel LWPJ, Koopman WJH, Nijtmans Balestri P, Grosso S. Endocrine disorders in two sisters affected
LGJ, Ugalde C,Willems PHGM. Cell biological consequences by MELAS syndrome. J Child Neurol 2000; 15: 755–758
of NADH: Ubiqunone oxidoreductase Deficiency. Curr Neu- Barak Y, Arnon S,Wolach B, Raz Y, Ashkenasi A, Glick B, Shapira Y.
rovasc Res 2004; 1:29–40 MELAS syndrome: peripheral neuropathy and cytochrome
Stadhouders AM, Sengers RCA. Morphological observations in c-oxidase deficiency: a case report and review of the litera-
skeletal muscle from patients with a mitochondrial myopa- ture. Isr J Med Sci 1995; 31: 224–229
thy. J Inherit Metab Dis 1987; 10: 62–80 Barisic N, Bernert G, Ipsiroglu O, Stromberger C, Muller T, Gru-
Suomalainen A, Kaukonen J. Diseases caused by nuclear genes ber S, Prayer D, Moser E, Bittner RE, Stöckler-Ipsiroglu S. Ef-
affecting mtDNA stability. Am J Med Genet 2001; 106: fects of oral creatine supplementation in a patient with
53–61 MELAS phenotype and associated nephropathy. Neurope-
Taylor RW, Morris AA, Hutchinson M, Turnbull DM. Leigh dis- diatrics 2002: 33: 157–161
ease associated with a novel mitochondrial DNA ND5 mu- Bataillard M, Chatzoglou E, Rumbach L, Sternberg D, Tounade
tation. Eur J Hum Genet 2002; 10: 141–144 A, Laforêt P, Jardel C, Maisonobe T, Lombès A. Atypical
Tiranti V, Corona P, Greco M,Taanman JW, Carrara F, Lamantea E, MELAS syndrome associated with a new mitochondrial
Nijtmans L, Uziel G, Zeviani M. A novel frameshift mutation tRNA glutamine point mutation. Neurology 2001; 56:
of the mtDNA COIII gene leads to impaired assembly of cy- 405–407
tochrome c oxidase in a patient affected by Leigh-like syn- Breningstall GN, Lockman LA. Massive focal brain swelling as a
drome. Hum Mol Genet 2000; 9: 2733–2742 feature of MELAS. Pediatr Neurol 1988; 4: 366–370
Tulinius MH, Holme E, Kristiansson B, Larsson NG, Oldfors A. Mi- Campos Y, Garcia-Silva T, Barrionuevo CR, Cabello A, Muley R,
tochondrial encephalomyopathies in childhood. I. Bio- Arenas J. Mitochondrial DNA deletion in a patient with mi-
chemical and morphologic investigations. J Pediatr 1991; tochondrial myopathy, lactic acidosis, and stroke-like
119: 242–250 episodes (MELAS) and Fanconi’s syndrome. Pediatr Neurol
Tulinius MH, Holme E, Kristiansson B, Larsson NG, Oldfors A. Mi- 1995; 13: 69–72
tochondrial encephalomyopathies in childhood. II. Clinical Castillo M, Kwock L, Green C. MELAS syndrome: imaging and
manifestations and syndromes. J Pediatr 1991; 119: 251– proton MR spectroscopic findings. AJNR Am J Neuroradiol
259 1995; 16: 233–239
Tulinius M, Moslemi AR, Darin N, Westerberg B, Wiklund LM, Chinnery PF, Howell N, Lightowlers RN,Turnbull DM. Molecular
Holme E, Oldfors A. Leigh Syndrome with cytochrome-c ox- pathology of MELAS and MERRF. The relationship between
idase deficiency and a single T insertion nt 5537 in the mi- mutation load and clinical phenotypes. Brain 1997; 120:
tochondrial tRNATrp gene. Neuropediatrics 2003; 34: 87–91 1713–1721
Visapaa I, Fellman V, Vesa J, Dasvarma A, Hutton JL, Kumar V, Chinnery PF, Howell N, Lightowlers RN,Turbull DM. MELAS and
Payne GS,Makarow M,Van Coster R,Taylor RW,Turnbull DM, MERRF. The relationship between maternal mutation load
Suomalainen A, Peltonen L. GRACILE syndrome, a lethal and the frequency of clinically affected offspring. Brain
metabolic disorder with iron overload, is caused by a point 1998; 121:1889–1894
mutation in BCS1L. Am J Hum Genet 2002; 71: 863–876 Ciafaloni E, Ricci E, Shanske S, Moraes CT, Silvestri G, Hirano M,
Vives-Bauza C, Del Toro M, Solano A, Montoya J, Andreu AL, Simonetti S, Servidei S, Zamarchi E, Bonilla E, DeVivo DC,
Roig M. Genotype-phenotype correlation in the 5703G>A Rowland LP, Schon EA, DiMauro S. MELAS: clinical features,
mutation in the tRNA (ASN) gene of mitochondrial DNA. J biochemistry, and molecular genetics. Ann Neurol 1992; 31:
Inherit Metab Dis 2003;26:507–508 391–398
Wallace DC. Mitochondrial genetics: a paradigm for aging and Clark JM, Marks MP, Adalsteinsson E, Spielman DM, Shuster D,
degenerative diseases? Science 1992; 256: 628–632 Horoupain D, Albers GW. MELAS: clinical and pathologic
Wallace DC,Lott MT,Shoffner JM,Brown MD.Diseases resulting correlations with MRI, xenon/CT, and MR spectroscopy.
from mitochondrial DNA point mutations. J Inherit Metab Neurology 1996; 46: 223–227
Dis 1992; 15: 472–479 Corona P, Antozzi C, Carrara F, D’Incerti L, Lamantea E, Tiranti V,
Zeviani M, DiDonato S. Neurological disorders due to muta- Zeviani M. A novel mtDNA mutation in the ND5 subunit of
tions of the mitochondrial genome. Neuromusc Disord complex I in two MELAS patients. Ann Neurol 2001; 49:
1991; 1: 165–172 106–110
Zeviani M, Spinazzola A, Carelli V. Nuclear genes in mitochon- Damian MS, Hertel A, Seibel P, Reichmann H, Bachmann G,
drial disorders. Curr Opin Genet Dev 2003; 13: 262–270 Sachenmayr W, Hoer G, Dorndorf W. Follow-in in carriers of
the ‘MELAS’ mutation without strokes. Eur Neurol 1998; 39:
9–15
De Coo IFM, Sistermans EA, de Wijs IJ, Catsman-Berrevoets C,
24 Mitochondrial myopathy, Busch HFM, Scholte HR, de Klerk JBC, van Oost BA, Smeets
encephalopathy, lactic acidosis HJM. A mitochondrial tRNAval gene mutation (G1642A) in a
and stroke-like episodes patient with mitochondrial myopathy, lactic acidosis, and
stroke-like episodes. Neurology 1998; 50: 293–295
Abe K, Fujimura H, Nishikawa Y, Yorifuji S, Mezaki T, Hirono N, De Coo IFM, Reinier WO, Ruitenbeek W, Ter Laak HJ, Bakker M,
Nishitani N, Kameyama M. Marked reduction in CSF lactate Schägger H, Van Oost BA, Smeets HJM. A 4-base pair dele-
and pyruvate levels after CoQ therapy in a patient with tion in the mitochondrial cytochrome b gene associated
mitochondrial myopathy, encephalopathy, lactic acidosis with Parkinsonism/MELAS overlap syndrome. Ann Neurol
and stroke-like episodes (MELAS). Acta Neurol Scand 1991; 1999; 45: 130–133
83: 356–359 Degoul F, Diry M, Pou-Serradell A, Lloreta J. Myo-leukoen-
Allard JC, Tilak S, Carter AP. CT and MR of MELAS syndrome. cephalopathy in twins: study of 3243-myopathy, en-
AJNR Am J Neuroradiol 1988; 9: 1234–1238 cephalopathy, lactic acidosis, and stokelike episodes mito-
chondrial DNA mutation. Ann Neurol 1994; 35: 365–370
References and Further Reading 943

Dubeau F, De Stefano N, Zifkin BG, Arnold DL, Shoubridge EA. Kamada K, Takeuchi F, Houkin K, Kitagawa M, Kuriki S, Ogata A,
Oxidative phosphorylation defect in the brains of carriers Tashiro K, Koyanagi I, Mitsumori K, Iwasaki Y. Reversible
of the tRNAleu(UUR) A3243G mutation in a MELAS pedigree. brain dysfunction in MELAS: MEG and 1H MRS analysis.
Ann Neurol 2000; 47: 179–185 J Neurol Neurosurg Psychiatry 2001; 70: 675–678
Fang W, Huang C-C, Lee C-C, Cheng S-Y, Pang C-Y,Wei Y-H. Oph- Kato T, Murashita J, Shiori T, Terada M, Inubushi T, Kato N. Phot-
thalmologic manifestations in MELAS syndrome. Arch Neu- ic stimulation-induced alteration of brain energy metabo-
rol 1993; 50: 977–980 lism measured by 31P-MR spectroscopy in patients with
Förster Ch, Hübner G, Müller-Höcker J, Pongratz D, Baierl P, Sen- MELAS. J Neurol Sci 1998; 155: 182–185
ger R, Ruitenbeek W. Mitochondrial angiopathy in a family Kim H-S, Kim D-I, Lee B-I, Lee B-I, Jeong E-K, Choi C, Lee JD,Yoon
with MELAS. Neuropediatrics 1992; 23: 165–168 P-H, Kim E-J, Yoon YK. Diffusion-weighted image and MR
Fujii T, Okuno T, Ito M, Motoh K, Hamazaki S, Okada S, Kusaka H, spectroscopic analysis of a case of MELAS with repeated at-
Mikawa H. CT, MRI, and autopsy findings in brain of a pa- tacks.Yonsei Med J 2001; 42: 128–133
tient with MELAS. Pediatr Neurol 1990; 6: 253–256 Kim I-O, Kim JY, Kim WS, Hwang WS, Yeon KM, Han MC. Mito-
Goto Y-I, Nonaka I, Horai S. A mutation in the tRNALeu(UUR) gene chondrial myopathy-encephalopathy-lactic acidosis-and
associated with the MELAS subgroup of mitochondrial en- strokelike episodes (MELAS) syndrome: CT and MRI find-
cephalomyopathies. Nature 1990; 348: 651–653 ings in seven children. AJR Am J Roentgenol 1996; 166:
Goto Y, Horai S, Matsuoka T, Koga Y, Nihei K, Kobayashi M, Non- 641–645
aka I. Mitochondrial myopathy, encephalopathy, lactic aci- Kimata KG, Goran L, Ajax ET, Davis PH, Grabowski T. A case of
dosis, and stroke-like episodes (MELAS): a correlative study late-onset MELAS. Arch Neurol 1998; 55: 722–725
of the clinical features and mitochondrial DNA mutation. Kimura M, Hasegawa Y,Yasuda K, Sejima H, Inoue M,Yamaguchi
Neurology 1992; 42: 545–550 S, Ando Y, Ohno S. Magnetic resonance imaging with fluid-
Hamazaki S, Okada S, Kusaka H, Fujii T, Okuno T, Kashu I, Mi- attenuated inversion recovery pulse sequences in MELAS
dorikawa O. Mitochondrial myopathy, encephalopathy, lac- syndrome. Pediatr Radiol 1997; 27: 153–154
tic acidosis, and stroke-like episodes. Acta Pathol Jpn 1989; Koga Y, Ishibashi M, Ueki I, Yatsuga S, Fukiyama R, Akita Y, Mat-
39: 599–606 suishi T. Effects of L-arginine on the acute phase of strokes
Hammans SR, Sweeney MG, Hanna MG, Brockington M, Mor- in three patients with MELAS. Neurology 2002; 58: 827–828
gan-Hughes JA, Marding AE.The mitochondrial DNA trans- Kolb SJ, Costello F, Lee AG, White M, Wong S, Schwartz ED,
fer RNALeu(UUR) AÆG(3243) mutation. A clinical and genetic Messé SR, Ellenbogen J, Kasner SE, Galetta SL. Distinguish-
study. Brain 1995; 118: 721–734 ing ischemic stroke from the stroke-like lesions of MELAS
Hanna MG, Nelson IP, Morgan-Hughes JA, Wood NW. MELAS: a using apparent diffusion coefficient mapping. J Neurol Sci
new disease associated mitochondrial DNA mutation and 2003; 216: 11–15
evidence for further genetic heterogeneity.J Neurol Neuro- Koo B, Becker LE, Chuang S, Merante F, Robinson BH, MacGre-
surg Psychiatry 1998; 65: 512–517 gor D, Tein I, Ho VB, McGreal DA, Wherrett JR, Logan WJ. Mi-
Hirano M, Ricci E, Koenigsberger R, Defendini R, Pavlakis ST, tochondrial encephalomyopathy, lactic acidosis, stroke-like
DeVivo DC, DiMauro S, Rowland LP. MELAS: an original case episodes (MELAS): clinical, radiological, pathological, and
and clinical criteria for diagnosis. Neuromusc Disord 1992; genetic observations. Ann Neurol 1993; 34: 25–32
2: 125–135 Kuriyama M, Umezaki H, Fukuda Y, Osame M, Koike K,Tateishi J,
Hirano M, Pavlakis SG. Mitochrondial myopathy, encephalo- Igata A. Mitochondrial encephalomyopathy with lactate-
pathy, lactic acidosis, and strokelike episodes (MELAS): cur- pyruvate elevation and brain infarctions. Neurology 1984;
rent concepts. J Child Neurol 1994; 9: 4–13 34: 72–77
Ihara Y, Namba R, Kuroda S, Sato T, Shirabe T. Mitochondrial Kaufmann P, Koga Y, Shanske S, Hirano M, DiMauro S, King MP,
encephalomyopathy (MELAS): pathological study and suc- Shon EA. Mitochondrial DNA and RNA processing in
cessful therapy with coenzyme Q10 and idebenone. J Neu- MELAS. Ann Neurol 1996; 40: 172–180
rol Sci 1989; 90: 263–271 Lee M-L. Chaou W-T, Yang AD, Jong Y-J, Tsai J-L, Pang C-Y, Wei
Iizuka T, Sakai F, Suzuki N, Hata T,Tsukahara S, Fukuda M,Takiya- Y-H. Mitochondrial myopathy, encephalopathy, lactic aci-
ma Y. Neuronal hyperexcitabilty in stroke-like episodes of dosis and strokelike episodes (MELAS); report of a sporadic
MELAS syndrome. Neurology 2002; 59: 816–824 case and review of the literature. Acta Paediatr Sin 1994; 35:
Iizuka T, Sakai F, Kan S, Suzuki N. Slowly progressive spread of 148–156
the stroke-like lesion in MELAS. Neurology 2003; 61: Lertrit P, Noer AS, Jean-Francois MJB, Kapsa R, Dennett X, Thya-
1238–1244 garajan D, Lethlean K, Byrne E, Marzuki S. A new disease-re-
Inui K, Fukushima H, Tsukamoto H, Taniike M, Midorikawa M, lated mutation for mitochondrial encephalopathy, lactic
Tanaka J, Nishigaki T, Okada S. Mitochondrial encephalo- acidosis, and strokelike episodes (MELAS) syndrome affects
myopathies with the mutation of the mitochondrial the ND4 subunit of the respiratory complex I. Am J Hum
tRNALeu(UUR) gene. J Pediatr 1992; 120: 62–66 Genet 1992; 51: 457–468
Johns DR, Stein AG, Wityk R. MELAS syndrome masquerading Liolitsa D, Rahman S, Benton S, Carr LJ, Hanna MG. Is the mito-
as herpes simplex encephalitis. Neurology 1993; 43: chondrial complex I ND5 gene a hot-spot for MELAS caus-
2471–2473 ing mutations? Ann Neurol 2003; 53:128–132
Joko T, Iwashige K, Hashimoto T, Ono Y, Kobayashi K, Sekiguchi Mariotti C, Saverese N, Soulalainen A, Rimoldi M, Comi G, Prelle
N, Kuroki T, Yanasa R, Takayanagi R, Umeda F, Nawata H. A A,Antozzi C,Servidei S,Jarre L,DiDonato S,Zeviani M.Geno-
case of mitochondrial encephalopathy, lactic acidosis and type to phenotype correlations in mitochondrial en-
stoke-like episodes associated with Diabetes Mellitus and cephalomyopathies associated with the A3224G mutation
hypothalamo-pituitary dysfunction. Endocr J 1997; 44: of mitochondrial DNA. J Neurol 1995; 242; 304–312
805–809
Kärppä M, Syrjälä P,Tolonen U, Majamaa K. Peripheral neuropa-
thy in patients with the 3243A>G mutation in mitochondr-
ial DNA. J Neurol 2003; 250: 216–221
944 References and Further Reading

Matsuzaki M, Izumi T, Shishikura K, Suzuki H, Hirayama Y. Hypo- Ravn K,Wibrand F,Hansen FJ,Horn N,Rosenberg T,Schwartz M.
thalamic growth hormone deficiency and supplementary An mtDNA muation, 1445GA, in the NADH dehydrogenase
GH therapy in two patients with mitochondrial myopathy, subunit 6 associated with severe MELAS syndrome. Eur J
encephalopathy, lactic acidosis and stroke-like-episodes. Hum Genet 2001; 9: 805–809
Neuropediatrics 2002; 33: 271–273 Rosen L, Philips S, Enzmann D. Magnetic resonance imaging in
Matthews PM, Tampieri D, Berkovic SF, Andermann F, Silver K, MELAS syndrome. Neuroradiology 1990; 32: 168–171
Chityat D, Arnold DL. Magnetic resonance imaging shows Sakuta R, Nonaka I. Vascular involvement in mitochondrial
specific abnormalities in the MELAS syndrome. Neurology myopathy. Ann Neurol 1989; 25: 594–601
1991; 41: 1043–1046 Sakuta R, Honzawa S, Murakami N, Goto Y, Nagai T. Atypical
Miyamoto A. Oki J, Takahashi S, Itoh J, Kusunoki Y, Cho K. Serial MELAS associated with mitochondrial tRNAlys gene
imaging in MELAS. Neuroradiology 1997; 39: 427–430 A8296G mutation. Pediatr Neurol 2002; 27: 397–400
Möller HE, Wiedermann D, Kurlemann G, Hilbich T, Schuierer G. Satoh M, Ishikawa N, Yoshizawa T, Takeda T, Akisada M. N-Iso-
Application of NMR spectroscopy to monitoring MELAS propyl-p-[123I]-iodoamphetamine SPECT in MELAS syn-
treatment: a case report. Muscle Nerve 2002; 25: 593–600 drome: comparison with CT and MR imaging. J Comput As-
Mongini T, Doriguzzi C, Chaiaò-Piat L, Silvestri G, Servidei S, Pal- sist Tomogr 1991; 15: 77–82
mucchi L. MERFF/MELAS overlap syndrome in a family with Serra G, Piccinnu R, Tondi M, Muntoni F, Zeviani M, Mastropao-
A3243G mtDNA mutation. Clin Neuropathol 2002; 21: lo C.Clinical and EEG findings in eleven patients affected by
72–76 mitochondrial encephalomyopathy with severe MERFF–
Mori O, Yamazaki M, Ohaki Y, Arai Y, Oguro T, Shimizu H, Asano MELAS overlap. Brain Dev 1996; 18: 185–191
G. Mitochondrial encephalomyopathy with lactic acidosis Seyama K, Suzuki K, Mizuno Y, Yoshida M, Tanaka M, Ozawa T.
and stroke like episodes (MELAS) with prominent degener- Mitochondrial encephalomyopathy with lactic acidosis
ation of the intestinal wall and cactus-like cerebellar atro- and stroke-like episodes with special reference to the
phy. Acta Neuropathol (Berl) 2000; 100: 712–717 mechanism of cerebral manifestations. Acta Neurol Scand
Mosewich RK, Donat JR, DiMauro S, Ciafaloni E, Shanske S, Eras- 1989; 80: 561–568
mus M, George D. The syndrome of mitochondrial en- Shoji Y, Sato W, Hayasaka K,Takada G.Tissue distribution of mu-
cephalomyopathy, lactic acidosis, and strokelike episodes tant mitochondrial DNA in mitochondrial myopathy, en-
presenting without stroke. Arch Neurol 1993; 50: 275–278 cephalopathy, lactic acidosis and stroke-like episodes
Nariai T, Ohno K, Ohta Y, Hirakawa K, Ishii K, Senda M. Discor- (MELAS). J Inherit Metab Dis 1993; 16: 27–30
dance between cerebral oxygen and glucose metabolism, Sparaco M, Simonati A, Cavellaro T, Bartelomei L, Grauso M, Pis-
and hemodynamics in a mitochondrial encephalomyo- cioli F, Morelli L, Rizzuto N. MELAS: clinical phenotype and
pathy, lactic acidosis, and strokelike episode patient. J Neu- morphological brain abnormalities. Acta Neuropathol
roimaging 2001; 11: 325–329 (Berl) 2003; 106: 202–121
Ohama E, Ohara S, Ikuta F, Tanaka K, Nishizawa M, Miyatake T. Sue CM, Crimmins DS, Soo YS, Pamphlett R, Presgrave CM, Kot-
Mitochondrial angiopathy in cerebral blood vessels of mi- simbos N, Jean-Fransois MJB, Morris JGL. Neuroradiological
tochondrial encephalomyopathy. Acta Neuropathol (Berl) features of six kindreds with MELAS tRNALeu A3243G point
1987; 74: 226–233 mutation: implications for pathogenesis. J Neurol Neuro-
Ohshita T, Oka M, Imon Y, Wanatabe C, Katayama S, Yamaguchi surg Psychiatry 1988; 65: 233–240
S,Kajima T,Momori Y,Nakamura S.Neuroradiology 2000; 42: Suzuki T, Koizumi J, Shiraishi H, Ishikawa N, Ofuku K, Sasaki M,
651–656 Hori T, Ohkoshi N, Anno I. Mitochondrial encephalomyopa-
Ooiwa Y, Uematsu Y, Terada T, Nakai K, Itakura T, Komai N, Mori- thy (MELAS) with mental disorder CT, MRI and SPECT find-
waki H.Cerebral blood flow in mitochondrial myopathy,en- ings. Neuroradiology 1990; 32: 74–76
cephalopathy, lactic acidosis, and strokelike episodes. Takahashi S, Tohgi H, Yonezawa H, Obara S, Nagane Y. Cerebral
Stroke 1993; 24: 304–309 blood flow and oxygen metabolism before and after
Oppenheim C, Galanaud D, Samson Y, Sahel M, Dormont D, stroke-like episode in patient with mitochondrial myo-
Wechsler B, Marsault C. Can diffusion weighted magnetic pathy, encephalopathy, lactic acidosis and stroke-like
resonance imaging help differentiate stroke from stroke- episodes (MELAS). J Neurol Sci 1998; 158: 58–64
like events in MELAS? J Neurol Neurosurg Psychiatry 2000; Tanahashi C, Nakayama A, Yoshida M, Ito M, Mori M, Mori N,
69: 248–250 Hashizume Y. MELAS with mitochondial DNA 3243 point
Park H, Davidson E, King MP.The pathogenic A3243G mutation mutation: a neuropathological study. Acta Neuropathol
in human mitochondrial tRNALeu(UUR) decreases the effi- (Berl) 2000; 99: 31–38
ciency of aminoacylation. Biochemistry 2003; 42: 958–964 Tanji K, Gamez J, Cervera C, Mearin F, Ortega A, de la Torre J,
Pavlakis SG, Phillips PC, DiMauro S, de Vivo DC, Rowland LP. Mi- Montoya J, Andreu AL, DiMauro S, Bonilla E. The A8344G
tochondrial myopathy, encephalopathy, lactic acidosis, and mutation in mitochondrial DNA associated with stroke-like
strokelike episodes: a distinctive clinical syndrome. Ann episodes and gastro-intestinal dysfunction. Acta Neu-
Neurol 1984; 16: 481–488 ropathol (Berl) 2003; 105: 69–75
Penn AMW,Lee JWK,Thuillier P,Wagner M,Maclure KM,Menard Taylor RW, Chinnery PF, Haldane F, Morris AAM, Bindoff FF, Wil-
MR, Hall LD, Kennaway NG. MELAS syndrome with mito- son J,Turnbull DM. MELAS associated with a transfer RNA in
chondrial tRNA Leu(UUR) mutation: correlation of clinical the valine transfer RNA gene of mitochondrial DNA. Ann
state, nerve conduction, and muscle 31P magnetic reso- Neurol 1996; 40: 459–462
nance spectroscopy during treatment with nicotinamide Terauchi A, Tamagawa K, Morimatsu Y, Kobayashi M, Sano T,
and riboflavin. Neurology 1992; 42: 2147–2152 Yoda S. An autopsy like case of mitochondrial encephalo-
Pulkes T, Eunson L, Patterson V, Siddiqui A,Wood NW, Nelson IP, myopathy, lactic acidosis and stroke-like episodes (MELAS)
Morgan-Hughes JA, Hanna MG. The mitochondrial DNA with a point mutation of mitochondrial DNA. Brain Dev
G13513A transition in ND5 is associated with a LHON/ 1996; 18: 224–229
MELAS overlap syndrome and may be a frequent cause of
MELAS. Ann Neurol 1999; 46: 916–919
References and Further Reading 945

Tokunaga M, Mita S, Murakami T, Kumamoto T, Uchino M, Non- Bu X, Rotter JI. X chromosome-linked and mitochondrial gene
aka I, Ando M. Single muscle fiber analysis of mitochondri- control of Leber hereditary optic neuropathy: evidence
al myopathy, encephalopathy, lactic acidosis, and stroke- from segregation analysis for dependence on X chromo-
like episodes (MELAS). Ann Neurol 1994; 35: 413–419 some inactivation. Proc Natl Acad Sci USA 1991; 88:
Tsuchiya K, Miyazaki H, Akebane H, Yamamoto M, Kondo H, 8198–8202
Mizusawa H, Ideka K. MELAS with prominent white matter Buhmann C, Ghadamosi J, Heesen C. Visual recovery in a man
gliosis and atrophy of the cerebellar granular layer: a clini- with the rare combination of mtDNA 11778m LHON muta-
cal, genetic, and pathological study. Acta Neuropathol tion and a MS-like disease after mitoxantrone therapy. Acta
(Berl) 1999; 97: 520–524 Neurol Scand 2002; 106: 236–239
Valenne L, Peatau A, Soumalainen A, Ketonen L, Pihko H. Lami- Chalmers RM, Harding AE. A case-control study of Leber’s
nar cortical necrosis in MELAS syndrome: MR and neu- hereditary optic neuropathy. Brain 1996; 119: 1481–1486
ropathological observations. Neuropediatrics 1996; 27: Cornelissen JC, Wanders RJA, Bolhuis PA, Bleeker-Wagemakers
154–160 E, Oostra RJ, Wijburg FA. Respiratory chain function in
Van Hellenberg Hubar JLM, Gabreëls FJM, Ruitenbeek W, Sen- Leber’s hereditary optic neuropathy: lack of correlation
gers RCA, Renier WO,Thijssen HOM, ter Laak HJ. MELAS syn- with clinical disease. J Inherit Metab Dis 1993; 16: 531–533
drome. Report of two patients, and comparison with data Cortelli P, Montagna P, Avoni P, Sangiorgi S, Bresolin N, Moggio
of 24 patients derived from the literature. Neuropediatrics M, Zaniol P, Mantovani V, Barboni P, Barbiroli B, Lugaresi E.
1991; 22: 10–14 Leber’s hereditary optic neuropathy: genetic, biochemical,
Wang XY, Noguchi K, Takashima S, Hayashi N, Ogawa S, Seto H. and phosphorus magnetic resonance spectroscopy study
Serial diffusion-weighted imaging in a patient with MELAS in an Italian family. Neurology 1991; 41: 1211–1215
and presumed cytotoxic oedema.Neuroradiology 2003; 45: De Vries DD,Went LN, Bruyn GW, Scholte HR, Hofstra RMW, Bol-
640–643 huis PA, van Oost BA. Genetic and biochemical impairment
Yanagihara C, Oyama A,Tanaka M, Nakaji K, Nishimura Y. An au- of mitochondrial complex I activity in a family with Leber
topsy case of mitochondrial encephalomyopathy with lac- hereditary optic neuropathy and hereditary spastic dysto-
tic acidosis and stroke-like episodes syndrome with chron- nia. Am J Hum Genet 1996; 58: 703–711
ic renal failure. Intern Med 2001; 40: 662–665 Dotti MT, Caputo N, Signorini E, Federico A. Magnetic reso-
Yoneda M, Maeda M, Kimura H, Fujii A, Katayama K, Kuriyama nance imaging findings in Leber’s hereditary optic neu-
M. Vasogenic edema on MELAS: a serial study with diffu- ropathy. Eur Neurol 1992; 32: 17–19
sion-weighted MR imaging. Neurology 1999; 53: 2182– Fauser S, Leo-Kottler B, Besch D, Luberichs J. Confirmation of
2184 the 14568 mutation in the mitochondrial ND6 gene as
Yonemura K, Hasegawa Y, Kimura K, Minematsu K,Yamaguchi T. causative in Leber’s hereditary optic neuropathy. Ophthal-
Diffusion-weighted MR imaging in a case of mitochondrial mic Genet 2002; 23: 191–197
myopathy, encephalopathy, lactic acidosis, and strokelike Flanigan KM, Johns DR. Association of the 11778 mitochondri-
episodes. AJNR Am J Neuroradiol 2001; 22: 269–272 al DNA mutation an dymyelinating disease. Neurology
1993; 43: 2720–2722
Funakawa I, Kato H, Terao A Ichihashi K, Kawashima S, Hayashi
T, Mitani K, Miyazaki S. Cerebellar ataxia in patients with
25 Leber Hereditary Optic Neuropathy Leber’s hereditary optic neuropathy. J Neurol 1995; 242:
75–77
Barbiroli B, Montagna P, Cortelli P, Lotti S, Lodi R, Barboni P, Funalot B, Reynier P, Vighetto A, Ranoux D, Bonnefont J-P,
Monari L, Lugaresi E, Frassineti C, Zaniol P. Defective brain Godinot C, Malthièry Y, Mas J-L. Leigh-like encephalopathy
and muscle energy metabolism shown by in vivo 31P mag- complicating Leber’s hereditary optic neuropathy. Ann
netic resonance spectroscopy in nonaffected carriers of Neurol 2002; 52: 374–377
11778 mtDNA mutation. Neurology 1995; 45: 1364–1369 Guy J, Qi X, Pallotti F, Schon EA, Manfredi G, Carelli V, Martinuzzi
Batioğlu F, Atilla H, Eryilmaz T. Chiasmal high signal on magnet- A, Hauswirth WW, Lewin AS. Rescue of a mitochondrial de-
ic resonance imaging in the atrophic phase of Leber hered- ficiency causing Leber hereditary optic neuropathy. Ann
itary optic neuropathy. J Neuroophthalmol 2003; 23: 28–30 Neurol 2002; 52: 534–542
Bet L,Moggio M,Comi GP,Mariani C,Prelle A,Checcarelli N,Bor- Harding AE, Sweeney MG, Miller DH, Mumford CJ, Kellar-Wood
doni A,Bresolin,Scarpini E,Scarlato G.Multiple sclerosis and H, Menard D, McDonald WI, Compston DAS. Occurrence of
mitochondrial myopathy: an unusual combination of dis- a multiple sclerosis-like illness in women who have a
eases. J Neurol 1994; 241: 511–516 Leber’s hereditary optic neuropathy mitochondrial DNA
Brown MD, Voljavec AS, Lott MT, MacDonald I, Wallace DC. mutation. Brain 1992; 115: 979–989
Leber’s hereditary optic neuropathy: a model for mito- Horváth R, Abicht A, Shoubridge EA, Karcagi V, Rózsa C, Komoly
chondrial neurodegenerative diseases. FASEB J 1992; 6: S, Lochmüller H. Leber’s hereditary optic neuropathy pre-
2791–2799 senting as multiple sclerosis-like disease of the CNS. J Neu-
Brown MD, Allan JC, van Stavern GP, Newman NJ, Wallace DC. rol 2000; 247: 65–67
Clinical, genetic, and biochemical characterization of a Huoponen K. Leber hereditary optic neuropathy: clinical and
Leber hereditairy optic neuropathy family containing both molecular genetic findings. Neurogenetics 2001; 3: 119–
the 11778 and 11484 primary mutations. Am J Med Genet 125
2001; 104: 331–338 Inglese M, Rovaris M, Bianchi S, La Mantia L, Mancardi GL,
Bruyn GW,Vielvoye GJ,Went LN. Hereditairy spastic dystonia: a Ghezzi A, Montagna P, Salvi F, Filippi M. Magnetic resonance
new mitochondrial encephalopathy? Putaminal necrosis as imaging, magnetisation transfer imaging, and diffusion
a diagnostic sign. J Neurol Sci 1991; 103: 195–202 weighted imaging correlates of optic nerve, brain, and cer-
vical cord damage in Leber’s hereditary optic neuropathy.
J Neurol Neurosurg Psychiatry 2001; 70: 444–449
946 References and Further Reading

Jansen PHP, van der Knaap MS, de Coo IFM. Leber’s hereditary Nikoskelainen EK, Marttilla RJ, Huoponen K, Juvonen V, Lammi-
optic neuropathy with the 11778 mtDNA mutation and nen T, Sonninen P. Savontaus M-L. Leber’s “plus”: neurologi-
white matter disease resembling multiple sclerosis: clinical, cal abnormalities in patients with Leber’s hereditary optic
MRI and MRS findings. J Neurol Sci 1996; 135: 176–180 neuropathy. J Neurol Neurosurg Psychiatry 1995; 59:
Kellar-Wood H, Robertson N, Govan GG, Compston DAS, Hard- 160–164
ing AE. Leber’s hereditary optic neuropathy mitochondrial Novotny EJ, Sing G, Wallace DC, Dorfman LJ, Louis A, Sogg RL,
DNA mutations in multiple sclerosis. Ann Neurol 1994; 36: Steinman L. Leber’s disease and dystonia: a mitochondrial
109–112 disease. Neurology 1986; 36:1053–1060
Kermode AG, Moseley IF, Kendall BE, Miller DH, MacManus DG, Parker WD Jr, Oley CA, Parks JK. A defect in mitochondrial elec-
McDonald WI.Magnetic resonance imaging in Leber’s optic tron-transport activity (NADH-coenzyme Q oxidoreduc-
neuropathy. J Neurol Neurosurg Psychiatry 1989; 52: tase) in Leber’s hereditary optic neuropathy. N Engl J Med
671–674 1989; 320: 1331–1333
Kim JY, Hwang J-M, Park SS. Mitochondrial DNA C4171A/ND1 is Paulus W, Straube A, Bauer W, Harding AE. Central nervous sys-
a novel primary mutation of Leber’s hereditary optic neu- tem involvement in Leber’s optic neuropathy. J Neurol
ropathy with a good prognosis. Ann Neurol 2002; 51: 1993; 240: 251–253
630–634 Riordan-Eva P, Sanders MD, Govan GG, Sweeney MG, da Costa
Larsson NG. Leber hereditary optic neuropathy: a nuclear solu- J, Harding AE.The clinical features of Leber’s hereditary op-
tion of a mitochondrial problem. Ann Neurol 2002; 52: tic neuropathy defined by the presence of a pathogenic
529–530 mitochondrial DNA mutation. Brain 1995; 118: 319–337
Larsson NG, Andersen O, Holme E, Oldfors A, Wahlström J. Sadun AA, Carelli V, Salomao SR, Berezovsky A, Quiros P, Sadun
Leber’s hereditary optic neuropathy and complex I defi- F, de Negri A-M, Andrade R, Schien S, Belfort R. A very large
ciency in muscle. Ann Neurol 1991; 30: 701–708 Brazilian pedigree with 11778 Leber’s hereditary optic neu-
Leo-Kottler B, Luberichs J, Besch D, Christ-Adler M, Fauser S. ropathy.Trans Am Ophthalmol Soc 2002; 100: 169–179
Leber’s hereditary optic neuropathy: clinical and molecular Shoffner JM, Brown MD, Stugard C, Jun AS, Pollock S, Haas RH,
genetic results in a patient with a point mutation at np Kaufman A, Koontz D, Kim Y, Graham JR, Smith E, Dixon J,
T11253C (isoleucine to threotine) in the ND4 gene and Wallace DC. Leber’s hereditary optic neuropathy plus dys-
spontaneous recovery. Graefes Arch Clin Exp Ophthalmol tonia is caused by a mitochondrial DNA point mutation.
2002; 240: 758–764 Ann Neurol 1995; 38: 163–169
Leuzzi V, Bertini E, de Negri AM, Gallucci M, Garavaglia B. Bilat- Sudoyo H, Suryadi H, Lertrit P, Pramoonjage P, Lyrawati D,
eral striatal necrosis, dystonia and optic atrophy in two sib- Marzuki S. Asian-specific mtDNA backgrounds associated
lings. J Neurol Neurosurg Psychiatry 1992; 55: 16–19 with the primary G11778A mutation of Leber’s hereditary
Lev D, Yanoov-Sharav M, Watemberg N, Leshinksky-Silver E, optic neuropathy. J Hum Genet 2002; 47: 594–604
Lerman-Sagie T. White matter abnormalities in Leber’s Valentino ML, Avoni P, Barboni P, Pallotti F, Rengo Ch,Torroni A,
hereditary optic neuropathy due to the 3460 mitochondri- Bellan M, Baruzzi A, Garrelli V. Mitochondrial DNA nu-
al DNA mutation. Eur J Paediatr Neurol 2002; 6: 121–123 cleotide changes C14482G and C14482A in the ND6 gene
Lodi R, Carelli V, Cortelli P, Lotti S,Valentino ML, Barboni P, Pallot- are pathogenic for Leber’s hereditary optic neuropathy.
ti F, Montagna P, Barbiroli B. Phosphorus MR spectroscopy Ann Neurol 2002; 51: 774–778
shows a tissue specific in vivo distribution of biochemical Vanopdenbosch L, Dubois B, D’Hooge M-B, Meire F, Carton H.
expression of the G3460A mutation in Leber’s hereditary Mitochondrial mutations of Leber’s hereditary optic neu-
optic neuropathy. J Neurol Neurosurg Psychiatry 2002; 72: ropathy: a risk factor for multiple sclerosis. J Neurol 2002;
805–807 247: 535–543
Luberichs J, Leo-Kottler B, Besch D, Fauser S. A mutation hot Wallace DC, Singh G, Lott MT, Hodge JA, Schurr TG, Lezza AMS,
spot in the mitochondrial ND6 gene in patients with Elsas LJ, Nikoskelainen EK. Mitochondrial DNA mutation as-
Leber’s hereditary optic neuropathy. Graefes Arch Clin Exp sociated with Leber’s hereditary optic neuropathy. Science
Ophthalmol 2002; 240: 96–100 1988; 242: 1427–1430
Man PYW, Turnbull DM, Chinnery PF. Leber hereditary optic Weiner NC, Newman NJ, Lessell S, Johns DR, Lott MT, Wallace
neuropathy. J Med Genet 2002; 39: 162–169 DC. Atypical Leber’s hereditary optic neuropathy with mol-
Man PYW, Griffiths PG, Brown DT, Howell N, Turnbull DM, Chin- ecular confirmation. Arch Neurol 1993; 50: 470–473
nery PF. The epidemiology of Leber hereditary optic neu-
ropathy in the North East of England. Am J Hum Genet
2003; 72: 333–339
Marsden CD, Lang AE, Quinn NP, McDonald WI, Abdallat A,
26 Kearns Sayre syndrome
Nimri S. Familial dystonia and visual failure with stiatal CT
lucencies. J Neurol Neurosurg Psychiatry 1986; 49: 500–509 Artuch R, Pavia C, Playán A, Vilaseca MA, Colomer J, Valls C, Ris-
Newman NJ. Leber’s hereditary optic neuropathy. Arch Neurol sech M, González MA, Pou A, Briones P,Montoya J, Pineda M.
1993; 50: 540–548 Multiple endocrine involvement in two pediatric patients
Newman NJ, Lott MT,Wallace DC.The clinical characteristics of with Kearns-Sayre syndrome. Horm Res 1998; 50: 99–104
pedigrees of Leber’s hereditary optic neuropathy with the Ashizawa T, Subramony SH. What is Kearns-Sayre syndrome
11778 mutation. J Ophthalmol 1991; 111: 750–762 after all? Arch Neurol 2001; 58: 1053–1054
Newman –Toker DE, Horton JC, Lessell S. Recurrent visual loss Barthélémy C, Ogier de Baulny H, Diaz J, Cheval MA, Franchon
in Leber hereditary optic neuropathy. Arch Ophthalmol P, Romero N, Goutières F, Fardeau M, Lombès A. Late-onset
2003; 121: 288–291 mitochondrial DNA depletion: DNA copy number, multiple
deletions, and compensation.Ann Neurol 2001; 49: 607–617
References and Further Reading 947

Becher MW, Wills ML, Noll WW, Hurko O, Price DL. Kearns-Sayre Marin-Garcia J, Goldenthal MJ. Sarnat HB. Kearns-Sayre syn-
syndrome with features of Pearson’s marrow-pancreas syn- drome with a novel mitochondrial DNA deletion. J Child
drome and a novel 2905-base pair mitochondrial DNA Neurol 2000; 15: 555–558
deletion. Hum Pathol 1999; 30: 577–581 Marín-García J, Goldenthal MJ, Flores-Sarnat L, Sarnat HB. Se-
Boles RG, Roe T, Sanadheera D, Mahnovski V, Wong LJC. Mito- vere mitochondrial cytopathy with complete A-V, PEO, and
chondrial DNA deletion with Kearns Sayre syndrome. Eur J mtDNA deletions. Pediatr Neurol 2002; 27: 213–216
Pediatr 1998; 157: 643–647 McShane MA,Hammans SR,Sweeney M,Holt IJ,Beattie TJ,Brett
Brockington M, Alsanjari N, Sweeney MG, Morgan-Hughes JA, EM, Harding AE. Pearson syndrome and mitochondrial en-
Scaravilli F, Harding AE. Kearns-Sayre syndrome associated cephalomyopathy in a patient with a deletion of mtDNA.
with mitochondrial DNA deletion or duplication: a molecu- Am J Hum Genet 1991; 48: 39–42
lar genetic and pathological study. J Neurol Sci 1995; 131: Mohri I, Taniike M, Fujimura H, Matsuoka T, Inui K, Nagai T, Oka-
78–87 da S. A case of Kearns-Sayre syndrome showing a constant
Chu BC, Terae S, Takahasi C, Kikuchi Y, Miyasaka K, Abe S, Mi- proportion of deleted mitochondrial DNA in blood cells
nowa K, Sawamura T. MRI of the brain in the Kearns-Sayre during 6 years of follow-up. J Neurol Sci 1998; 158: 106–109
syndrome: report of four cases and a review. Neuroradiolo- Muñoz A, Meteos F, Simón R, García-Silva MT, Cabello S, Arenas
gy 1999; 41: 759–764 J.Mitochondrial diseases in children: neuroradiological and
Coultier DL, Allen RJ. Abrupt neurological deterioration in chil- clinical features in 17 patients. Neuroradiology 1999; 41:
dren with Kearns-Sayre syndrome. Arch Neurol 1981; 38: 920–928
247–250 Nakagawa E, Hirano S, Yamanouchi H, Goto Y-I, Nonaka I,
Curless RG, Flynn J, Bachynski B, Gregorios JB, Benke P, Cullen R. Takashima S. Progressive brainstem and white matter le-
Fatal metabolic acidosis, hyperglycemia, and coma after sions in Kearns-Sayre syndrome: a case report. Brain Dev
steroid therapy for Kearns-Sayre syndrome. Neurology 1994; 16: 416–418
1986; 36: 872–873 Nakagawa E,Osari S-i,Yamanouchi H,Matsuda H,Goto Y-I,Non-
Daroff RB, Solitare GB, Pincus JH, Glaser GH. Spongiform en- aka I. Long-term therapy with cytochrome c, flavin
cephalopathy with chronic progressive external ophthal- mononucleotide and thiamine diphosphate for a patient
moplegia. Neurology 1966; 16: 161–169 with Kearns-Sayre syndrome. Brain Dev 1996; 18: 68–70
Demange P, Pham Gia H, Kalifa G, Sellier N. MR of Kearns-Sayre Nakano T,Imanaka K,Uchida H,Isaka N,Takezawa H.Myocardial
syndrome. AJNR Am J Neuroradiol 1989; 10: S91 ultrastructure in Kearns-Sayre syndrome. Angiology 1987;
Flynn JT, Bachynski BR, Rodrigues MM, Curless RG, Joshi B. Hy- 38: 28–35
perglycemic acidotic coma and death in Kearns-Sayre syn- Oldfors A, Fyhr I-M, Holme E, Larsson N-G, Tulinius M. Neu-
drome.Trans Am Ophthalmol Soc 1985; 83: 131–161 ropathology in Kearns-Sayre syndrome. Acta Neuropathol
Gallastegui J, Hairiman RJ, Handler B, Lev M, Bharati S. Cardiac (Berl) 1990; 80: 541–546
involvement in the Kearns-Sayre syndrome. Am J Cardiol Poulton J, Morten KJ, Weber K, Brown GK, Bindoff L. Are dupli-
1987; 60: 385–388 cations of mitochondrial DNA characteristic of Kearns-
Herzberg NH, van Schooneveld MJ, Bleeker-Wagemakers EM, Sayre syndrome? Hum Mol Genet 1994; 3: 947–951
Zwart R, Cremers FPM, van der Knaap MS, Bolhuis PA, de Provenzale JM,VanLandingham K.Cerebral infarction associat-
Visser M. Kearns-Sayre syndrome with a phenocopy of ed with Kearns-Sayre syndrome – related cardiomyopathy.
choroideremia instead of pigmentary retinopathy. Neurol- Neurology 1996; 46: 826–828
ogy 1993; 43: 218–221 Robertson Jr,WC,Viseskul C, Lee YE, Lloyd RV. Basal ganglia cal-
Ishikawa Y, Goto Y-I, Ishikawa Y, Minami R. Progression in a case cification in Kearns-Sayre syndrome. Arch Neurol 1979; 36:
of Kearns-Sayre syndrome.J Child Neurol 2000; 15: 750–755 711–713
Kamata Y, Mashima Y, Yokoyama M, Tanaka K, Goto Y-I, Oguchi Rötig A, Bourgeron T, Chretien D, Rustin P, Munnich A. Spec-
Y.Patient with Kearns-Sayre syndrome exhibiting abnormal trum of mitochondrial DNA arrangements in the Pearson
magnetic resonance image of the brain. J Neuroophthal- marrow-pancreas syndrome. Hum Mol Genet 1995; 4:
mol 1998; 18: 284–288 1327–1330
Kleber FX, Park J-W, Hübner G, Johannes A, Pongratz D, König E. Schröder R, Vielhaber S, Wiedemann FR, Kornblum C, Papas-
Congestive heart failure due to mitochondrial cardio- sotiropoulos A, Broich P, Zierz S, Elger CE, Reichmann H,
myopathy in Kearns-Sayre syndrome. Klin Wochenschr Seibel P, Klockgether T, Kunz WS. New insights into the
1987; 65: 480–486 metabolic consequences of large-scale mtDNA deletions: a
Klopstock T, Bishof F, Gerock K, Deuschl G, Seibel P, Ketelson UP, quantitative analysis of biochemical, morphological, and
Reichmann H. 3.1-kb Deletion of mitochondrial DNA in a genetic findings in human skeletal muscle. J Neuropathol
patient with Kearns-Sayre syndrome. Acta Neuropathol Exp Neurol 2000; 59: 353–360
(Berl) 1995; 90: 26–129 Seneca S, Verhelst H, De Meirleir L, Meire F, Ceuterick-De
Larsson NG, Holme E, Kristiansson B, Oldfors A, Tulinius M. Grootte C, Lissens W, Van Coster R. A new mitochondrial
Progressive increase of the mutated mitochondrial DNA point mutation in the transfer RNAleu gene in a patient with
fraction in Kearns-Sayre syndrome. Pediatr Res 1990; 28: clinical phenotype resembling Kearns-Sayre syndrome.
131–136 Arch Neurol 2001; 58: 1113–1118
Lertrit P, Imsumran A, Karnkirawattana P, Devahasdin V, San- Simaan EM, Mikati MA, Touma EH, Rötig A. Unusual presenta-
gruchi T, Atchaneeyasakul L-o, Mungkornkarn C, Neungton tion of Kearns-Sayre syndrome in early childhood. Pediatr
N. A unique 3.5-kb deletion of the mitochondrial genome Neurol 1999; 21: 830–831
in Thai patients with Kearns-Sayre syndrome. Hum Genet Valanne L, Ketonen L, Majander A, Soumalainen A, Pihko H.
1999; 105: 127–131 Neuroradiologic findings in children with mitochondrial
Leung TF, Hui J, Shoubridge E, Li CK, Chik KW, Shing MMK,Wong disorders. AJNR Am J Neuroradiol 1998; 19: 369–377
GWK, Yeung WL, Yeun PMP. Aplastic anaemia in association
with Kearns-Sayre syndrome. J Inherit Metab Dis 1999; 22:
86–87
948 References and Further Reading

Vásquez-Acevedo M, Vázquez-Memije ME, Mutchinick OM, Kocaefe YC, Erdem S, Özgüç M, Tan E. Four novel thymidine
Morales JJ, García-Ramos G, Gonzáles-Halphen D. A case of phosphorylase gene mutations in mitochondrial neuro-
Kearns-Sayre syndrome with the 4,977-bp common dele- gastrointestinal encephalomyopathy syndrome (MNGIE)
tion associated with a novel 7,704-bp deletion. Neurol Sci patients. Eur J Hum Genet 2003; 11: 102–104
2002; 23: 247–250 Labauge P, Durant R, Castelnovo G, Dubois A. MNGIE: diarrhea
Wilichowski E,Korenke GC,Ruitenbeek W,De Meirleir L,Hagen- and leukoencephalopathy. Neurology 2002; 25 :58
dorff A, Janssen AJM, Lissens W, Hanefeld F. Pyruvate dehy- Marti R, Spinazzola A, Nishino I, Andreu AL, Naini A. Tadesse S,
drogenase complex deficiency and altered respiratory Oliver JA, Hirano M. Mitochondrial neurogastrointestinal
chain function in a patient with Kearns-Sayre syndrome / encephalomyopathy and thymidine metabolism: results
MELAS overlap syndrome and A3243G mtDNA mutation. and hypothesis. Mitochondrion 2002; 2: 143–147
J Neurol Sci 1998; 157: 206–213 Martí R, Nishigaki Y, Hirano M. Elevated plasma deoxyuridine
Wray SH,Provenzale JM,Johns DR,Thulborn KR.MR of the brain in patients with thymidine phosphorlyase deficiency.
in mitochondrial myopathy AJNR Am J Neuroradiol 1995; Biochem Biophys Res Commun 2003; 303: 14–18
16: 1167–1173 Millar WS, Lignelli A, Hirano M. MRI of five patients with mito-
Yoda S,Terauchi A, Kitahara F, Akabane T. Neurologic deteriora- chondrial neurogastrointestinal encephalomyopathy. AJR
tion with progressive CT changes in a child with Kearns-Shy Am J Roentgenol 2004; 182: 1537–1541
syndrome. Brain Dev 1984; 6: 322–327 Nishino I, Spinazzola A, Hirano M. Thyamidine phosphorylase
Zanssen S, Molnar M, Buse G, Schröder JM. Mitochondrial cy- gene mutations in MNGIE, a human mitochondrial disor-
tochrome b gene deletion in Kearns-Sayre syndrome asso- der. Science 1999; 283: 689–692
ciated with a subclinical type of pheripheral neuropathy. Nishino I, Spinazzola A, Papadimitriou A, Hammans S, Steiner I,
Clin Neuropathol 1998; 17: 291–296 Hahn CD, Connolly AM, Verloes A, Guimarães J, Maillard I,
Zeviani M, Moraes CT, DiMauro S, Nakase H, Bonilla E, Schon EA, Hamano H, Donati MA, Semrad CE, Russell JA, Andreu AL,
Rowland LP. Deletions of mitochondrial DNA in Kearns- Hadjigeorgiou GM, Vu TH, Tadesse S, Nygaard TG, Nonaka I,
Sayre syndrome. Neurology 1988; 38: 1339–1346 Hirano I, Bonilla E, Rowland LP, DiMauro S, Hirano M. Mito-
chondrial neurogastrointestinal encephalomyopathy: an
autosomal recessive disorder due to thymidine phospho-
rylase mutations. Ann Neurol 2000; 47: 792–800
27 Myo-, Neuro-, Nishino I, Spinazzola A, Hirano M. MNGIE: from nuclear DNA to
Gastrointestinal Encephalopathy mitochondrial DNA. Neuromusc Disord 2001; 11: 7–10
Papadimitrou A, Comi GP, Hadjigeorgiou GM, Bordoni A, Sciac-
Bardosi A, Creutzfeldt W, DiMauro S, Felgenhauer K, Friede RL, co M, Napoli L, Prelle A, Moggio M, Fagiolari G, Bresolin N,
Goebel HH, Kohlschütter A, Mayer G, Rahlf G, Servidei S, van Salani S. Anastasopoulos I, Giassakis G, Divari R, Scarlao G.
Lessen G, Wetterling T. Myo-, neuro-, gastrointestinal en- Partial depletion and multiple deletions of muscle mtDNA
cephalopathy (MNGIE syndrome) due to partial deficiency in familial MNGIE syndrome. Neurology 1998; 51: 1086–
of cytochrome-c-oxidase. A new mitochondrial multisys- 1092
tem disorder. Acta Neuropathol (Berl) 1987; 74: 248–258 Simon LT, Horoupian DS, Dorf LJ, Marks M, Herrick MK, Wasser-
Carrozzo R, Davidson MM, Walker WF, Hiranio M, Miranda AF. stein P, Smith ME. Polyneuropathy, ophthalmoplegia, leuko-
Cellular and molecular studies in the muscle and cultures cenphalophathy and intestinal pseudo-obstruction: POLIP
from patients with multiple mitochondrial DNA deletions. syndrome. Ann Neurol 1990; 28: 349–360
J Neurol Sci 1999; 170: 24–31 Soykan I, Cetinkaya H, Erdem S,Tan E, Aydin F, Bahar K, Özden A.
Gamez J, Ferriero C, Accarino ML, Guarner L, Tadesse S, Marti Mitochondrial neurogastrointestinal encephalomyopathy.
RA, Andreu AL, Raguer N, Cervera C, Hirano M. Phenotypic Diagnostic features in two patients. J Clin Gastroenterol
variability in a Spanish family with MNGIE. Neurology 2002; 2002; 34: 446–448
59: 455–457 Spinazzola A, Marti R, Nishino I, Andreu AL, Naini AL Tadesse S,
Hirano M, Silvestri G, Blake DM, Lombes A, Minetti C, Bonilla E, Pela I, Zammarchi E, Donati MA, Oliver JA, Hirano M. Altered
Hays AP, Lovelace RE, Butler I, Bertorini TE,Threlkeld AB, Mit- thymidine metabolism due to defects of thymidine phos-
sumoto H, Salberg LM, Rowland LP, DiMauro S. Mitochondr- phorylase. J Biol Chem 2002; 277: 4128–4133
ial neurogastrointestinal encephalomyopathy (MNGIE): Suomalainen A, Kaukomen J. Diseases caused by nuclear
clinical, biochemical, and genetic features of an autosomal genes affecting mtDNA stability. Am J Med Genet 2001;
recessive mitochondrial disorder. Neurology 1994; 44: 106: 53–61
721–727 Teitelbaum JE, Berde CB, Nurko S, Buonomo C, Perez-Atayade
Hirano M, Garcia-de-Yebenes J, Jones AC, Nishino I, DiMauro S, AR, Vox VL. Diagnosis and management of MNGIE syn-
Carlo JR, Bender AN, Hahn AF, Salberg LM, Weeks DE, Ny- drome in children: case report and review of the literature.
gaard TG. Mitochondrial neurogastrointestinal encephalo- J Pediatr Gastroenterol Nutr 2002; 35: 377–383
myopathy syndrome maps to chromosome 22q13.32-qter. Uncini A, Servidei S, Silvestri G, Manfredi G, Sabatelli M, Di
Am J Hum Genet 1998; 63: 526–533 Muzio A, Ricci E, Mirabella M, DiMauro S, Tonali P. Ophthal-
Hirano M, Nishigaki Y, Marti R. Mitochondrial neurogastroin- moplegia, demyelinating neuropathy, leukoencephalopa-
testinal encephalopmyopathy (MNGIE): a disease of two thy, myopathy, and gastrointestinal dysfunction with multi-
genomes. Neurologist 2004; 10: 8–17 ple deletions of mitochondrial DNA: a mitochondrial multi-
Ionasescu V. Oculogastrointestinal muscular dystrophy. Am J system disorder in search of a name.Muscle Nerve 1994; 17:
Hum Med Genet 1983; 15: 103–112 667–674
Kaidar-Person O, Golz A, Netzer A, Goldsher D, Joachims HZ,
Goldenberg D. Rapidly progressive bilateral sensory neural
hearing loss as a presentation of mitochondrial neurogas-
trointestinal encephalomyopathy. Am J Otolaryngol 2003;
24: 128–130
References and Further Reading 949

28 Leigh Syndrome and Mitochondrial Kang PB, Hunter JV, Melvin JJ, Selak MA, Faerber EN, Kaye EM.
Leukoencephalopathies Infantile leukoencepathopathy owing to mitochondrial en-
zyme dysfunction. J Child Neurol 2002; 17: 421–428
Kimura S, Kobayashi T, Amemiya F. Myelin splitting in the
General spongy lesion in Leigh encephalopathy. Pediatr Neurol
1991; 7: 56–58
Absalon MJ, Harding CO, Fain DR, Li L, Mack KJ. Leigh syndrome Kirby DM, Kahler SG, Freckmann M-L, Reddihough D, Thorn-
in an infant resulting from mitochondrial DNA depletion. burn DR. Leigh disease caused by the mitochondrial DNA
Pediatr Neurol 2001; 24: 60–63 G14459A mutation in unrelated family. Ann Neurol 2000;
Anzil AP, Weindl A, Struppler A. Ultrastructure of a cerebral 48: 102–104
white matter lesion in a 41-year-old man with Leigh’s en- Kissel JT, Kolkin S, Chakeres D, Boesel C, Weiss K. Magnetic res-
cephalomyelopathy (LEM). Acta Neuropathol (Berl) 1981; onance imaging in a case of autopsy-proved adult suba-
Suppl VII: 233–238 cute necrotizing encephalomyelopathy (Leigh’s disease).
Arri J,Tanabe Y. Leigh syndrome: serial MR imaging and clinical Arch Neurol 1987; 44: 563–566
follow-up. AJNR Am J Neuroradiol 2000; 21: 1502–1509 Kohlschütter A, Kraus-Ruppert R, Rohrer T, Herschkowitz NN.
Barkovich AJ, Good WV, Koch TK, Berg BO. Mitochondrial disor- Myelin studies in a case of subacute necrotizing en-
ders: analysis of their clinical and imaging characteristics. cephalomyelopathy (SNE). J Neuropathol Exp Neurol 1978;
AJNR Am J Neuroradiol 1993; 14: 1119–1137 37: 155–164
Campos Y, Martin MA, Rubio JC, Solana LG, García-Benayas C, Krägeloh-Mann I, Grodd W, Nägele T. Proton spectroscopy in
Terradas JL, Arenas J. Leigh syndrome associated with the patients with Leigh’s disease and mitochondrial enzyme
T9176C mutation in the ATPase 6 gene of mitochondrial deficiency. Dev Med Child Neurol 1994; 36: 839–845
DNA. Neurology 1997; 49: 595–597 Majoie CB, Akkerman EM, Blank C, Barth PG, Poll-The BT, den
Coenen MJH, van den Heuvel LP, Smeitink JAM. Mitochondrial Heeten GJ. Mitochondrial encephalomyopathy: compari-
oxidative phosphorylation system assembly in man: recent son of conventional MR imaging with diffusion-weighted
achievements. Curr Opin Neurol 2001; 14: 777–781 and diffusion tensor imaging: case report. Am J Neuroradi-
Crompton MR. Spongiform subacute necrotising encephalo- ol 2002; 23: 813–816
myelopathy. Acta Neuropathol (Berl) 1969; 13: 204–208 Malandrini A, Palmeri S, Fabrizi GM, Villanova M, Berti G, Sal-
Davis PC, Hoffman JC, Braun IF, Ahmann P, Krawiecki N. MR of vadori C, Gardini G, Motti L, Solimé F, Guazzi GC. Juvenile
Leigh’s disease (subacute necrotizing encephalomyelopa- Leigh syndrome with protracted course presenting as
thy). AJNR Am J Neuroradiol 1987; 8: 71–75 chronic sensory motor neuropathy, ataxia, deafness and re-
De Lonlay-Debeney P, von Kleist-Retzow J-C, Hertz-Pannier L, tinitis pigmentosa: a clinicopathological report.J Neurol Sci
Peudenier S, Cormier-Daire V, Berquin P, Chrétien D, Rötig A, 1998; 155: 218–221
Saudubray J-M,Baraton J,Brunelle F,Rustin P,van der Knaap Masó E, Ferrer I, Herraiz J, Roquer J, Serrano S. Leigh’s syndrome
M, Munnich A. Cerebral white matter disease in children in an adult. J Neurol 1984; 231: 253–257
may be caused by mitochondrial respiratoiry chain defi- Medina L, Chi TL, DeVivo DC, Hilal SK. MR findings in patients
ciency. J Pediatr 2000; 136: 209–214 with subacute necrotizing encephalomyelopathy (Leigh
Egger J, Wynne-Williams CJE, Erdohazi M. Mitochondrial cy- syndrome): correlation with biochemical defect. AJNR Am J
topathy or Leigh’s syndrome? Mitochondrial abnormalities Neuradiol 1990; 11: 379–384
in spongiform encephalopathies.Neuropediatrics 1982; 13: Montpetit VJA, Andermann F, Carpenter S, Fawcett JS,
219–224 Zborowska-Sluis D, Giberson HR. Subacute necrotizing en-
Egger J, Pincott JR, Wilson J, Erdohazi M. Cortical subacute cephalomyelopathy. A review and a study of two families.
necrotizing encephalomyelopathy. A study of two patients Brain 1971; 94: 1–30
with mitochondrial dysfunction. Neuropediatrics 1984; 15: Moroni I, Bugiani M, Bizzi A, Castelli G, Lamantea E, Uziel G.
150–158 Cerebral white matter involvement in children with mito-
Feigin I, Kim HS. Subacute necrotizing encephalomyopathy in chondrial encephalopathies. Neuropediatrics 2002; 33:
a neonatal infant. J Neuropathol Exp Neurol 1977; 36: 79–85
364–372 Morris AAM, Leopard JV.The treatment of congenital lactic aci-
Filiano JJ, Goldenthal MJ, Mamourian AC, Hall CCC, Marin-Gar- doses. J Inherit Metab Dis 1996; 19: 573–580
cia J. Mitochondrial DNA depletion in Leigh syndrome . Pe- Nakagawa M, Kaminishi Y, Isashiki Y, Yamada H, Higuchi I, Uchi-
diatr Neurol 2002; 26: 239–242 da Y, Osame M. Familial mitochondrial encephalomyopathy
Goebel HH, Bardosi A, Friede RL, Kohlschütter A, Albani M, with deaf-mutism, ophthalmoplegia and leukodystrophy.
Siemes H. Sural nerve biopsy studies in Leigh’s subacute Acta Neurol Scand 1995; 92: 102–108
necrotizing encephalomyelopathy. Muscle Nerve 1986; Niers L, van den Heuvel L, Trijbels F, Sengers R, Smeitink J. Pre-
9:165–173 requisites and strategies for prenatal diagnosis of respira-
Greenberg SB, Faerber EN, Riviello JJ, de Leon G, Capitanio MA. tory chain deficiency in chorionoc villi. J Inherit Metab Dis
Subacute necrotizing encephalomyelopathy (Leigh dis- 2003; 26: 647–658
ease): CT and MRI appearances. Pediatr Radiol 1990; 21: 5–8 Rahman S. Blok RB, Dahl H-HM, Danks DM, Kirby DM, Chow CW,
Howell N, Kubacka I, Smith R, Frerman F, Parks JK, Parker WD Jr. Christodoulou J,Thorburn DR. Leigh syndrome: clinical fea-
Association of the mitochondrial 8344 MERRF mutation tures and biochemical and DNA abnormalities. Ann Neurol
with maternally inherited spinocerebellar degeneration 1996; 39: 343–351
and Leigh disease. Neurology 1996; 46: 219–222 Sandhu FS, Dillon WP. MR demonstration of leukoencephalo-
Jiang Y-W,Qin J,Yuan U,Qi Y,Wu X-R.Neuropathologic and clin- pathy associated with mitochondrial encephalomyopathy
ical features in eight Chinese patients with Leigh disease. (case report). AJNR Am J Neuroradiol 1991; 12: 375–379
J Child Neurol 2002; 17: 450–452
950 References and Further Reading

Santorelli FM, Barmada MA, Pons R, Zhang LL, DiMauro S. Dahl J-HM. Pyruvate dehydrogenase E1a deficiency: males
Leigh-type neuropathology in Pearson syndrome associat- and females differ yet again. Am J Hum Genet 1995; 56:
ed with impaired ATP production and a novel mtDNA dele- 553–557
tion. Neurology 1996; 47: 1320–1323 De Meirleir L, Lissens W, Denis R, Wayenberg J-L, Michotte A,
Santorelli FM, Tanji K, Shanske S, Krishna S, Schmidt RE, Green- Brucher J-M,Vamos E, Gerlo E, Liebaers I. Pyruvate dehydro-
wood RS, DiMauro S, De Vivo DC. The mitochondrial DNA genase deficiency:clinical and biochemical diagnosis.Pedi-
A8344G mutation in Leigh syndrome revealed by analysis atr Neurol 1993; 9: 216–220
in paraffin-embedded sections: revisiting the past. Ann De Meirleir L, Specola N, Seneca S, Lissens W. Pyruvate dehy-
Neurol 1998; 44: 962–964 drogenase E1a deficiency in a family: different clinical pre-
Seitz RJ, Langes K, Frenzel H, Kluitmann G,Wechsler W.Congen- sentation in two siblings. J Inherit Metab Dis 1998; 21:
ital Leigh’s disease: panencephalomyelopathy and periph- 224–226
eral neuropathy. Acta Neuropathol (Berl) 1984; 64: 167–171 De Meirleir L, Lissens W, Benelli C, Marsac C, de Klerk J, Scholte
Shtilbans A, Shanske S, Goodman S, Sue CM, Bruno C, Johnson J, van Diggelen O, Kleijer W, Seneca S, Liebaers I. Pyruvate
TL, Lava NS, Waheed N, DiMauro S. G8363A mutation in the dehydrogenase complex deficiency and absence of sub-
mitochondrial DNA transfer ribonucleic acidlys gene: anoth- unit X. J Inherit Metab Dis 1998; 21: 9–16
er case of Leigh syndrome. J Child Neurol 2000; 15: 759–761 Dey R, Mine M, Desguerre I, Slama A, van den Berghe L, Brivert
Valanne L, Ketonen L, Majander A, Suomalainen A, Pihko H. M, Aral B, Marsac C. A new case of pyruvate dehydrogenase
Neuroradiologic findings in children with mitochondrial deficiency due to a novel mutation in the PDX1 gene. Ann
disorders. AJNR Am J Neuroradiol 1998; 19: 369–377 Neurol 2003; 53: 273–277
van der Knaap MS, Jakobs C, Valk J. Magnetic resonance imag- Fujii T, van Coster RN, Old SE, Medori R, Winter S, Gubits RM,
ing in lactic acidosis. J Inherit Metab Dis 1996; 19: 535–547 Matthews PM, Brown RM, Brown GK, Dahl H-HM, de Vivo
Walter GF, Brucher JM, Martin JJ, Ceuterick C, Pilz P, Freund M. DC. Pyruvated dehydrogenase deficiency: molecular basis
Leigh’s disease – several nosological entities with an iden- for intrafamilial heterogeneity. Ann Neurol 1994; 36: 83–89
tical histopathological complex? Neuropathol Appl Neuro- Geoffroy V, Fouque F, Benelli C, Poggi F, Saudurbray JM, Lissens
biol 1986; 12: 95–107 W, Meirleir LD, Marsac C, Lindsay JG, Sanderson SJ. Defect in
Weinstock A, Giglio P, Cohen ME, Bakshi R, Januario J, Balos L. the X-lipoyl-containing component of the pyruvated dehy-
Diffuse magnetic resonance imaging white-matter drogenase complex in a patient with a neonatal lactic
changes in a 15-year-old boy with mitochondrial en- acidemia. Pediatrics 1996; 97: 267–272
cephalomyopathy. J Child Neurol 2002; 17: 47–49 Harada M,Tanouchi M, Arai K, Nishitani H, Miyoshi H, Hashimo-
to T.Therapeutic efficacy of a case of pyruvated dehydroge-
nase complex deficiency monitored by localized proton
Pyruvate Dehydrogenase Complex Deficiency magnetic resonance spectroscopy. Magn Res Imaging
1996; 14: 129–133
Aleck KA, Kaplan AM, Sherwood WG, Robinson BH. In utero Heckmann JM, Eastman R, Handler L, Wright M, Owen P. Leigh
central nervous system damage in pyruvate dehydroge- disease (subacute necrotizing encephalomyelopathy): MR
nase deficiency. Arch Neurol 1988; 45: 987–989 documentation of the evolution of an acute attack. AJNR
Bindhoff LA, Brich-Machin MA, Farnsworth L, Gardner-Medwin Am J Neuroradiol 1993; 14: 1157–1159
D, Lindsay JG, Turnbull DM. Familial intermittent ataxia due Kimura S, Osaka H, Saitou K, Ohtuki N, Kobayashi T, Nezu A. Im-
to defect of the E1 component of pyruvate dehydrogenase provement of lesions shown on MRI and CT scan by admin-
deficiency. J Neurol Sci 1989; 93: 311–318 istration of dichloroacetate in patients with Leigh syn-
Bonne G,Benelli C,de Meirleir L,Lissens W,Chaussain M,Diry M, drome. J Neurol Sci 1995; 134: 103–107
Clot J-P, Ponsot G, Geoffroy V, Leroux J-P, Marsac C. E1 pyru- Kimura S, Ohtuki N, Nezu A, Tanaka M, Takeshita S. Clinical and
vate dehydrogenase defiency in a child with motor neu- radiologic improvements in mitochondrial encephalo-
ropathy. Pediatr Res 1993; 33: 284–288 myelopathy following sodium dichloroacetate therapy.
Brown GK. Pyruvate dehydrogenase E1a deficiency. J Inherit Brain Dev 1997; 19: 535–540
Metab Dis 1992; 15: 625–633 Kinosita H, Sakuragawa N, Tada H, Naito E, Kuroda Y, Nonaka I.
Brown GK, Otero LJ, LeGris M, Brown RM. Pyruvate dehydroge- Recurrent muscle weakness and ataxia in thiamine-respon-
nase deficiency. J Med Genet 1994; 31: 875–879 sive pyruvated dehydrogenase complex deficiency. J Child
Cederbaum SD, Blass JP, Minkoff N, Brown WJ, Cotton ME, Har- Neurol 1997; 12: 141–144
ris SH. Sensitivity to carbohydrate in a patient with familial Kretzschmar HA, DeArmond SJ, Koch TK, Patel MS, Newth CJL,
intermittent lactic acidosis and pyruvate dehydrogenase Schmidt KA, Packman S. Pyruvate dehydrogenase complex
deficiency. Pediatr Res 1976; 10: 713–720 deficiency as a cause of subacute necrotizing encephalo-
Chabrol B, Mancini J, Benelli C, Gire C, Munnich A. Leigh syn- pathy (Leigh disease). Pediatrics 1987; 79: 370–373
drome: pyruvate dehydrogenase defect. A case with pe- Lie SO, Loken AChr, Strömme JH, Aagenaes O. Fatal congenital
ripheral neuropathy. J Child Neurol 1994; 9: 52–55 lactic acidosis in two siblings. Clinical and pathological
Chow CW, Anderson RMcD, Kenney GCT. Neuropathology in findings. Acta Paeditr Scand 1971; 60: 129–137
cerebral lactic acidosis. Acta Neuropathol (Berl) 1987; 74: Lissens W, de Meirleir L, Seneca S, Liebaers I, Brown GK, Brown
393–396 RM, Ito M, Naito E, Kuroda Y, Kerr DS, Wexler IS, Patel MS,
Cross JH, Connelly A, Gadian DG, Kendall BE, Brown GK, Robinson BH, Seyda A. Mutations in the X-linked pyruvate
Leonard JV. Clinical diversity of pyruvate dehydrogenase dehydrogenase (E1) asubunit gene (PDHA1) in patients
deficiency. Pediatr Neurol 1994; 10: 276–283 with a pyruvate dehydrogenase complex deficiency. Hum
Dahl H-HM, Hansen LL, Brown RM, Danks DM, Rogers JG, Brown Mutat 2000; 15: 209–219
GK. X-linked pyruvate dehydrogenase E1a subunit defi-
ciency in heterozygous females: variable manifestation of
the same mutation. J Inherit Metab Dis 1992; 15: 835–847
References and Further Reading 951

Marsac C, Stansbie D, Bonne G, Cousin J, Jehenson P, Benelli C, Weber TA, Antognetti MR, Stacpoole PW. Caveats when consid-
Leroux J-P, Lindsay G. Defect in the lipoyl-bearing protein X ering ketogenic diets for the treatment of pyruvate dehy-
subunit of the pyruvated dehydrogenase complex in two drogenase complex deficiency. J Pediatr 2001; 138: 390–
patients with encephalomyelopathy. J Pediatr 1993; 123: 395
915–920 Wexler ID, Hemalatha SG, McConnell J, Buist NRM, Dahl H-HM,
Matthews PM, Brown RM, Otero L, Marchington D, Leonard JV, Berry SA, Cederbaum SD, Patel MS, Derr DS. Outcome of
Brown GK. Neurodevelopmental abnormalities and lactic pyruvate dehydrogenase deficiency treated with keto-
acidosis in a girl with a 20-bp deletion in the X-linked pyru- genic diets. Studies in patients with identical mutations.
vate dehydrogenase E1a subunit gene. Neurology 1993; Neurology 1997; 49: 1655–1661
43: 2025–2030 Wijburg FA, Barth PG, Bindoff LA, Birch-Machin MA, van der Blij
Matthews PM, Brown RM, Otero LJ, Marchingon DR, LeGris M, JF, Ruitenbeek W, Turnbull DM, Schutgens RBH. Leigh syn-
Howes R, Meadows LS, Shevell M, Scriver CR, Brown GK. drome associated with a deficiency of the pyruvate dehy-
Pyruvate dehydrogenase deficiency. Clinical presentation drogenase complex: results of treatment with a ketogenic
and molecular genetic characterization of five new pa- diet. Neuropediatrics 1992; 23: 147–152
tients. Brain 1994; 117: 435–443 Zand DJ, Simon EM, Pulitzer SB, Wang DJ, Wang ZJ, Rorke LB,
Michotte A, de Meirleir L, Lissens W, Denis R, Wayenberg JL, Palmieri M, Berry GT. In vivo pyruvate detected by MR spec-
Liebears I, Brucher JM. Neuropathological findings of a pa- troscopy in neonatal dehydrogenase deficiency. AJNR Am J
tient with pyruvate dehydrogenase E1a deficiency pre- Neuroradiol 2003; 24: 1471–1474
senting as a cerebral lactic acidosis.Acta Neuropathol (Berl)
1993; 85: 674–678
Morten KJ, Beattie P, Brown GK, Matthews PM. Dichloroacetate Complex I Deficiency
stabilizes the mutant E1a subunit in pyruvate dehydroge-
nase deficiency. Neurology 1999; 53: 612–616 Andreu AL, Tanji K, Bruno C, Hadjigeorgiou GM, Sue CD, Jay C,
Naito E, Ito M, Yokota I, Saijo T, Chen S, Maehara M, Kuroda Y. Ohnishi T, Shanske S, Bonilla E, diMauro S. Exercise intoler-
Concomitant administration of sodium dichloroacetate ance due to a nonsense mutation in the mtDNA ND4 gene.
and thiamine in West syndrome caused by thiamine-re- Ann Neurol 1999; 45: 820–823
sponsive pyruvate dehydrogenase complex deficiency. Bénit P, Chretien D, Kadhom N, de Lonlay-Debeney P, Cormier-
J Neurol Sci 1999; 171: 56–59 Daire V, Cabral A, Peudenier S, Rustin P, Munnich A, Rötig A.
Otero LJ, Brown GK, Silver K, Arnold SL, Matthews PM. Associa- Large-scale deletion and point mutations of the nuclear
tion of cerebral dysgenesis and lactic acidemia with X- NDUFV1 and NDUFS1 genes in mitochondrial complex 1
linked PHD E1a subunit mutations in females. Pediatr Neu- deficiency. Am J Hum Genet 2001; 68: 1344–1352
rol 1995; 13: 327-332 Bentlage HACM, Wendel U, Schägger H, ter Lak HJ, Janssens
Robinson BH, MacMillan H, Petrova-Benedict R, Sherwood WG. AJM, Trijbels JMF. Lethal infantile mitochondrial disease
Variable clinical presentation in patients with defective E1a with isolated complex 1 deficiency in fibroblasts but with
component of pyruvate dehydrogenase complex. J Pediatr combined complex 1 and IV deficiencies in muscle. Neurol-
1987; 111: 525–533 ogy 1996: 47: 243–248
Robinson BH, MacKay N, Petrova-Benedict R, Ozalp I, Coskun T, Budde SMS, van den Heuvel LPWJ, Janssen AJ, Smeets RJP,
Stacpoole PW. Defects in the E2 lipoyl containing compo- Buskens CAF, deMeirleir L, van Coster R, Baethmann M, Voit
nent of the pyruvate dehydrogenase complex in patient T,Trijbels JMF,Smeitink JAM.Combined enzymatic complex
with lactic acidemia. J Clin Invest 1990; 85: 1821–1824 I and III deficiency associated with mutations in the nuclear
Robinson BH, MacKay N, Chun K, Ling M. Disorders of pyruvate encoded NDUFS4 gene. Biochem Biophys Res Commun
dehydrogenase carboxylase and the pyruvate dehydroge- 2000; 275: 63–68
nase complex. J Inherit Metab Dis 1996; 19: 452–462 Chol M, Lebon S, Bénit P, Chretien D, de Lonlay P, Goldenberg A,
Rubio-Gozalbo ME, Heerschap A,Trijbels JMF, de Meirleir L,Thi- Odent S, Hertz-Pennier L, Vincent-Delorme C, Cormier-
jssen HOM, Smeitink JAM. Proton MR spectroscopy in a Daire V, Rustin P, Rötig A, Munnich A. The mitochondrial
child with pyruvate dehydrogenase complex deficiency. DNA G13513A MELAS mutation in the NADH dehydroge-
Magn Res Imaging 1999; 17: 939–944 nase 5 gene is a frequent cause of Leigh-like syndrome
Shany E, Saada A, Landau D, Shaag A, Hershkovitz E, Elpeleg with isolated complex I deficiency. J Med Genet 2003; 40:
ON. Lipoamide dehydrogenase deficiency due to a novel 188–191
mutation in the interface domain. Biochem Biophys Res Cooper JM, Mann VM, Krige D, Shapira AHV. Human mitochon-
Commun 1999; 262: 163–166 drial complex I dysfunction. Biochim Biophys Acta 1992;
Shevell MI,Matthews PM,Scriver CR,Brown RM,Otero LJ,Legris 1101: 198–2003
M, Brown GK, Arnold DL. Cerebral dysgenesis and lactic Dionisi-Vici C, Ruitenbeek W, Fariello G, Bentlage H, Wanders
acidemia: an MRI/MRS phenotype associated with pyru- RJA, Schägger H, Bosman C, Piontadosi C, Sabetta G, Bertini
vate dehydrogenase deficiency. Pediatr Neurol 1994: 11: E. New familial mitochondrial encephalopathy with macro-
224–229 cephaly, cardiomyopathy, and complex I deficiency. Ann
Stacpoole PW, Barnes CL, Hurbanis MD, Cannon SL, Kerr DS. Neurol 1997; 42: 661–665
Treatment of congenital lactic acidosis with dichloroac- Fiellet F, Mousson Y, Grignon Y, Leonard JV, Vidailhet M. Necro-
etate. Arch Dis Child 1997; 77: 535–541 tizing encephalopathy with mitochondrial complex I defi-
Takahashi S, Oki J,Tokumitsu A, Obata M, Ogawa K,Tokusashi Y, ciency. Pediatr Neurol 1999; 20: 305–308
Saijo H, Okuno A. Autopsy findings in pyruvate dehydroge- Houshmand M, Larsson N-G, Oldfors A,Tulinius M, Holme E. Fa-
nase E1a deficiency: case report. J Child Neurol 1997; 12: tal mitochondrial myopathy, lactic acidosis and complex I
519–524 deficiency associated with a heteroplasmic AG mutation at
Wallace SJ. Deficiencies within the pyruvate dehydrogenase position 3251 in the mitochondrial tRNALeu(UUR) gene.
complex: clinical and pathological correlates. Dev Med Hum Genet 1996; 97: 369–273
Child Neurol 1985; 27: 249–260
952 References and Further Reading

Kirby DM, Crawford M, Cleary MA, Dahl H-HM, Dennett X, Van den Heuvel L, Ruitenbeek W, Smeets R, Gelman-Kohan Z,
Thornburn DR. Respiratiory chain complex I deficiency. An Elpeleg O, Loeffen J, Trijbels F, Mariman E, de Bruijn D,
underdiagnosed energy generation disorder. Neurology Smeitink J. Demonstration of a new pathogenic mutation
1999; 52: 1255–1264 in the human complex I deficiency: a 5-bp duplication in
Kirby DM, Boneh A, Chow CW, Ohtake A, Ryan MT, Thyagarajan the nuclear gene encoding the 18-kD (AQDQ) subunit. Am
D, Thorburn DR. Low mutant load of mitochondrial DNA J Hum Genet 1998; 62: 262–268
G13513A mutation can cause Leigh’s disease. Ann Neurol Vogel R, Nijtmans L, Ugalde C, van den Heuvel L, Smeitink J.
2003; 54: 473–478 Complex I assembly: a puzzling problem. Curr Opin Neurol
Loeffen J, Smeitink J,Triepels R, Smeets R, Schuelke M, Sengers 2004; 17: 179–168
R, Trijbels F, Hamel B, Mullaart R, van den Heuvel L. The first Wolf NI, Seitz A, Harting I, Smeitink JAM, Trijbels F, van den
nuclear-encoded complex I mutation in a patient with Heuvel LP, Schlemmer H, Ebinger F, Evert W, Rating D. New
Leigh syndrome. Am J Hum Genet 1998; 63: 1598–1608 pattern of brain MRI lesions in isolated complex I deficien-
Loeffen J, Smeets R, Smeitink J, Ruitenbeek W, Janssen A, Mari- cy. Neuropediatrics 2003; 34:156–159
man E, Sengers R, Trijbels F, van den Heuvel L. The X-chro-
mosomal NDUFA1 gene of complex I in mitochondrial en-
cephalomyopathies:tissue expression and mutation detec- Complex II Deficiency
tion. J Inherit Metab Dis 1998; 21: 210–215
Loeffen JLCM, Smeitink JAM,Trijbels JMF, Janssen AJM,Triepels Birch-Machin MA, Taylor RW, Cochran B, Ackrell BAC, Turnbull
RH, Sengers RCA, van den Heuvel LP. Isolated complex I DM. Late-Onset optic atrophy, ataxia, and myopathy associ-
deficiency in children: clinical, biochemical and genetic ated with a mutation of a complex II gene. Ann Neurol
aspects. Hum Mutat 2000; 15: 123–134 2000; 48: 330–335
Loeffen J, Elpeleg O, Smeitink J, Smeets R, Stockler-Ipsiroglu S, Bourgeois M, Goutieres F, Chretien D, Rustin P, Munnich A,
Mandel H, Sengers R,Trijbels F, van den Heuvel L. Mutations Aicardi J. Deficiency in complex II of the respiratory chain,
in the complex I NCUFS2 gene of patients with cardio- presenting as a leukodystrophy in two sisters with Leigh
myopathy and encephalomyopathy. Ann Neurol 2001; 49: syndrome. Brain Dev 1992; 14: 404–408
195–201 Bourgeron T, Rustin P, Chretien D, Birch-Machin M, Bourgeois
Morris AAM, Leonard JV, Brown GK, Bidouki SK, Bindhoff LA, M, Viegas-Péquignot E, Munnich A, Rötig A. Mutation of a
Woodward CE, Harding AE, Lake BD, Harding BN, Farrell MA, nuclear succinate dehydrogenase gene results in mito-
Bell JE, Mirakhur M, Turnbull DM. Deficiency of respiratoiry chondrial respiratoiry chain deficiency. Nat Genet 1995; 11:
chain complex I is a common cause of Leigh disease. Ann 144–148
Neurol 1996; 40: 25–30 Brockmann K, Bjornstad A, Dechent P, Korenke CG, Smeitink J,
Ogle RF, Christodoulou J, Fagan E, Blok RB, Kirby DM, Seller KL, Trijbels JMF, Athanassopoulos S, Villagran R, Sjkeldal AH,
Dahl H-HM,Thorburn DR. Mitochondrial myopathy wiht tR- Wilichowski E, Frahm J, Hanefeld F. Succinate in dystrophic
NALeu(uur) mutation and complex I deficiency responsive to white matter: a proton magnetic resonance spectroscopy
riboflavin. J Pediatr 1997; 130: 138–145 finding characteristic for complex II deficiency. Ann Neurol
Pitkänen S, Feigenbaum A, Laframboise R, Robinson BH. 2002: 52: 38–46
NADH-coenzyme Q reductase (complex I) deficiency: het- Taylor RW, Birch-Machin MA, Schaefer J, Taylor L, Shakir R, Ack-
erogeneity in phenotype and biochemical findings.J Inher- rell BAC, Cochran B, Bindhoff LA, Jackson MJ, Griffiths P,
it Metab Dis 1996; 19: 675–686 Turnbull DM. Deficiency of complex II of the mitochondrial
Dubio-Gozalbo ME, Ruitenbeek W, Wendel U, Sengers RCA, Tri- respiratoiry chain in late-onset optic atrophy and ataxia.
jbels JMF, Smeitink JAM. Systemic infantile complex I defi- Ann Neurol 1996; 39: 224–232
ciency with fatal outcome in two brothers. Neuropediatrics
1998; 29: 43–45
Schuelke M, Smeitink J, Mariman E, Loeffen J, Plecko B, Trijbels Complex IV Deficiency
F, Stockler-Ipsiroglu S, van den Heuvel L. Mutant
NDUFV1subunit of mitochondrial complex I causes Antonicka H, Mattman A, Carlson CG, Glerum DM, Hoffbuhr KC,
leukodystrophy and myoclonic epilepsy. Nat Genet 1999; Leary SC, Kennaway NG, Shoubridge EA. Mutations in
21: 260–261 COX15 produce a defect in the mitochondrial heme biosyn-
Smeitink JAM, Loeffen JLCM, Triepels RH, Smeets RJP, Trijbels thetic pathway, causing early-onset fatal hypertrophic car-
JMF, van den Heuvel LP. Nuclear genes of human complex I iomyopathy. Am J Hum Genet 2003; 72: 104–114
of the mitochondrial electron transport gene: state of the Antonicka H, Leary SC, Guercin G-H, Agar JN, Horvath R, Kenn-
art. Hum Mol Genet 1998; 7: 1573–1579 away NG, Harding CO, Jaksch M, Shoubridge EA. Mutations
Triepels RH, van den Heuvel LP, Loeffen JLCM, Buskens CAF, in COX10 result in a defect in mitochondrial heme A biosyn-
Smeets RJP, Rubio Gozalbo ME, Budde SMS, Mariman EC, thesis and account for multiple, early-onset clinical pheno-
Wijburg FA, Barth PG, Trijbels JMF, Smeitink JAM. Leigh syn- types associated with isolated COX deficiency. Hum Mol
drome associated with a mutation in the NDUFS7 (PSST) Genet 2003; 12: 2693–2702
nuclear encoded subunit of complex I.Ann Neurol 1999; 45: Bakker HD, van den Bogert C, Drewes JG, Barth PG, Scholte HR,
787–790 Wanders RJA, Ruitenbeek W. Progressive generalized brain
Triepels RH, van den Heuvel LP, Trijbels JM, Smeitink JA. Respi- atrophy and infantile spasms associated with cytochrome c
ratoiry chain complex I deficiency. Am J Med Genet 2001; oxidase deficiency. J Inherit Metab Dis 1996; 19: 153–156
106: 37–45 Bruno C, Martinuzzi A, Tang Y, Andreu AL, Pallotti F, Bonilla E,
Trijbels JMF, Ruitenbeek W, Sengers RCA, Janssen AJM, van Shanske S, Fu J, Sue CM, Angelini C, DiMauro S, Manfredi G.
Oost BA. Benign mitochondrial encephalomyopathy in a A stop-codon mutation in the human mtDNA cytochrome
patient with complex I deficiency. J Inherit Metab Dis 1996; c oxidase I gene disrupts the functional structure of com-
19: 149–152 plex IV. Am J Hum Genet 1999; 65: 611–620
References and Further Reading 953

Campos Y, García-Redondo A, Férnandez-Moreno MA, Morin C, Dubé J, Robinson BH, Lacroix J, Michaud J, De
Martínez-Pardo M, Goda G, Rubio JC, Martin MA, del Hoyo P, Braekeleer M, Geoffroy G, Lortie A, Blanchette C, Lambert
Cabello A, Bornstein B, Garesse R, Arenas J. Early-onset mul- MA, Mitchell GA. Stroke-like episodes in autosomal reces-
tisystem mitochondrial disorder caused by a nonsense mu- sive cytochrome oxidase deficiency. Ann Neurol 1999; 45:
tation in the mitochondrial DNA cytochrome C oxidase II 389–392
gene. Ann Neurol 2001; 50: 409–413 Papadopoulou LC, Sue CM, Davidson MM, Tanji K, Nishino I,
Clark KM, Taylor RW, Johnson MA, Chinnery PF, Chrzanowska- Sadlock JE, Krishna S, Walker W, Glerum DM, van Coster R,
Lightowlers ZMA, Andrews RM, Nelson IP, Wood NW, Lam- Lyon G, Scalais E, Lebel R, Kaplan P, Shanske S, de Vivo DC,
ont PJ, Hanna MG, Lightowlers RN,Turnbull DM. An mtDNA Bonilla E, Hirano M, DiMauro S, Schon EA. Fatal infantile car-
mutation in the initiation codon of the cytochrome C oxi- dioencephalomyopathy with COX deficiency and muta-
dase subunit II results in lower levels of the protein and a tions in SCO2, a COX assembly gene. Nat Genet 1999; 23:
mitochondrial encephalomyopathy. Am J Hum Genet 333–337
1999; 64: 1330–1339 Parfait B, Percheron A, Chretien D, Rustin P, Munnich A, Rötig A.
Comi GP, Bordoni A, Salani S, Franceschina L, Sciacco M, Prelle No mitochondrial cytochrome oxidase (COX) gene muta-
A, Fortunato F, Zeviani M, Napoli L, Bresolin N, Moggio M, tions in 18 cases of COX deficiency. Hum Genet 1997; 101:
Ausenda CD, Taanman J-W, Scarlato G. Cytochrome c oxi- 247–250
dase subunit I microdeletion in a patient wiht motor neu- Péquignot MO, Dey R, Zeviani M, Tiranti V, Godinot C, Payau A,
ron disease. Ann Neurol 1998; 43: 110–116 Sue C, Di Mauro S, Abitbol M, Marsac C. Mutations in the
DiMauro S, Nicholson JF, Hays AP, Eastwood AB, Papadimitriou SURF1 gene associated with Leigh syndrome and cyto-
A, Koenigsberger R, DeVivo DC. Benign infantile mitochon- chrome c oxidase deficiency. Hum Mutat 2001; 17: 374–381
drial myopathy due to reversible cytochrome c oxidase de- Rahman S,Taanman J-W, Cooper JM, Nelson I, Hargreaves I, Me-
ficiency. Ann Neurol 1983; 14: 226–234 unier B, Hanna MG, García JJ, Capaldi RA, Lake BD, Leonard
DiMauro S, Servidei S, Zeviani M, DiRocco M, DeVivo DC, DiDo- JV, Schapira AHV. A missense mutation of cytochrome oxi-
nato S, Uziel G, Berry K, Hoganson G, Johnsen SD, Johnson dase subunit II causes defective assembly and myopathy.
PC. Cytochrome c oxidase defiency in Leigh syndrome. Ann Am J Hum Genet 1999; 65: 1030–1039
Neurol 1987; 22: 498–506 Rahman S,Brown RM,Chong WK,Wilson CJ,Brown GK.A SURF1
Èaèiæ M, Wilichowski E, Mejaški-Bošnjak V. Cytochrome c oxi- gene mutation presenting as isolated leukodystrophy. Ann
dase partial deficiency-associated with Leigh disease pre- Neurol 2001; 49: 797–800
senting as an extrapyramidal syndrome. J Child Neurol Robinson BH. Human cytochrome oxidase deficiency. Pediatr
2001; 16: 616–619 Res 2000; 48: 581–585
Elia M Musumeci SA, Ferri R, Colamaria V, Azan G, Greco D, Ste- Rossi A, Biancheri R, Bruno C, Di Rocco M, Calvi A, Pessagno A,
fanini MC. Leigh syndrome and partial deficit of cyto- Tortori-Donati P. Leigh syndrome with COX deficiency and
chrome c oxidase associated with epilepsia partialis contin- SURF1 gene mutations: MR imaging findings. AJNR Am J
ua. Brain Dev 1996; 18: 207–211 Neuroradiol 2003; 24: 1188–1191
Farina L, Chiapparini L, Uziel G, Bugiani M, Zeviani M, Savoiardo Sacconi S, Salviati L, Sue CM, Shanske S, Davidson MM, Bonilla
M. MR findings in Leigh syndrome with COX deficiency and E, Niani AB, de Vivo DC, DiMauro S. Mutation screening in
SURF-1 mutations. AJNR Am J Neuroradiol 2002; 23: 1095– patients with isolated cytochrome c oxidase deficiency.
1100 Pediatr Res 2003; 53: 224–230
Goldenberg PC, Steiner RD, Merkens LS, Dunaway T, Egan RA, Salviati L, Sacconi S, Rasalan MM, Kronn DF, Braun A, Canoll P,
Zimmerman EA, Nesbit G, Robinson B, Kennaway NG. Re- Davidson M, Shanske S, Bonilla E, Hays AP, Schon EA, DiMau-
markable improvement in adult Leigh syndrome with par- ro S. Cytochrome c oxidase deficiency due to a novel SCO2
tial cytochome c oxidase deficiency. Neurology 2003; 60: mutation mimics Werdnig-Hoffmann disease. Arch Neurol
865–868 2002; 59: 862–865
Hanna MG, Nelson IP, Rahman S, Lane RJM, Land J, Heales S, Santoro L, Carrozzo R, Malandrini A, Piemonte F, Patrono C,
Cooper MJ, Shapira AHV, Morgan-Hughes JA,Wood NW. Cy- Villanova M, Tessa A, Palmeri S, Bertini E. Santorelli FM. A
tochrome c oxidase deficiency associated with the first novel SURF1 mutation results in Leigh syndrome with pe-
stop-codon point mutation in human mtDNA. Am J Hum ripheral neuropathy causes by cytochrome c oxidase defi-
Genet 1998; 63: 29–36 ciency. Neuromusc Disord 2000; 10: 450–453
Harpey J-P, Heron D, Prudent M, Charpentier C, Rustin P, Ponsot Savasta S, Comi GP, Perini MP, Lupi A, Strazzer S, Rognoni F,
G, Cormier-Daire V.Diffuse leukodystrophy in an infant with Rossoni R. Leigh disease: clinical, neuroradiologic, and bio-
cytochrome-c oxidase deficiency. J Inherit Metab Dis 1998; chemical study of three new cases with cytochrome c oxi-
21: 4748–752 dase deficiency. J Child Neurol 2001; 16: 608–613
Kaido M, Fujimura H,Taniike M,Yoshikawa H,Toyooka K,Yoirfu- Savoiardo M, Uziel G, Strada L,Visciani A, Grisoli M,Wang G. MRI
ji S, Inui K, Okada S, Sparaco M, Yanagihara T. Focal cy- findings in Leigh’s disease with cytochrome-c-oxidase defi-
tochrome c oxidase deficiency in the brain and dorsal root ciency. Neuroradiology 1991; 33: 507–508
ganglia in a case with mitochondrial encephalomyopathy Shoubridge EA. Cytochrome c oxidase deficiency. Am J Med
(tRNAIle 4269 mutation): histochemical, immunohisto- Genet 2001; 106: 45–52
chemical, and ultrastructural study. J Neurol Sci 1995; 131: Silvestri G, Mongini T, Odoardi F, Modoni A, deRosa G, Doriguzzi
170–176 C, Palmucci L, Tonali P, Servidei S. A new mtDNA mutation
Mootha VK, Lepage P, Miller K, Bunkenborg J, Reigh M, Hjerrild associated with a progressive encephalopathy and cyto-
M, Delmonte T, Villeneuve A, Sladek R, Xu F, Mitchell GA, chrome c oxidase deficiency. Neurology 2000; 54: 1693–
Morin C, Mann M, Hudson TJ, Robinson B, Rioux JD, Lander 1696
ES. Identification of a gene causing human cytochrome c
oxidase deficiency by integrative genomics. Proc Natl Acad
Sci U S A 2003; 100: 605–610
954 References and Further Reading

Sue CM, Karadimas C, Checcarelli N, Tanji K, Papadopuolou LC, ATPase 6 Gene Defect
Pallotti F, Guo FL, Shanske S, Hirano M, de Vivo DC, van
Coster R, Kaplan P, Bonilla E, DiMauro S. Differential features Baracca A, Bagori S, Carelli V, Lenaz G, Solaini G. Catalytic activ-
of patients with mutations in two COX assembly genes, ities of mitochondrial ATP synthase in patients with mito-
SURF-1 and SCO2. Ann Neurol 2000; 47: 589–595 chondrial DNA T8993G mutation in the ATPase 6 gene en-
Tiranti V, Hoertnagel K, Carrozzo R, Galimberti C, Munaro M, coding subunit a. J Biol Chem 2000; 275: 4177–4182
Granatiero M, Zelante L. Gasparini P, Marzella R, Rocchi M, Carelli V, Baracca A, Bagogi S, Pallotti F,Valentino ML, Montagna
Bayona-Bafaluy MP, Enriquez J-A, Uziel G, Bertini E, Dionisi- P, Zevaini M, Pini A, Lenaz G, Baruzzi A, Solaini G. Biochemi-
Vici C, Franco B, Meitinger T, Zeviani M. Mutations of SURF-1 cal-clinical correlation in patients with different loads of
in Leigh disease associated with cytochrome c oxidase de- the mitochondrial DNA T8993G mutation. Arch Neurol
ficiency. Am J Hum Genet 1998; 63: 1609–1621 2002; 59: 264–270
Tiranti V, Jaksch M, Hofmann S, Galimberti C, Hoertnagel K, Lul- Chowers I, Lerman-Sagie T, Elpeleg ON, Shaag A, Merin S. Cone
li L, Freisinger P, Bindhoff L Gerbitz KD, Comi G-P, Uziel G, Ze- and rod dysfunction in the NARP syndrome. Br J Ophthal-
viani M, Meitinger T. Loss-of-function mutations of SURF-1 mol 1999; 83: 190–193
are specifically associated with Leigh syndrome with cyto- Degoul F, François D, Diry M, Ponsot G, Desguerre I, Héron B,
chrome c oxidase deficiency. Ann Neurol 1999; 46: 161–166 Marsac C, Moutard ML. A near homoplasmic T8993G
Topçu M, Saatci I, Apak A, Söylemezoglu F, Akçören Z. Leigh mtDNA mutation in a patient with atypic Leigh syndrome
syndrome in a 3-year-old boy with unusual brain MR imag- not present in the mother’s tissues. J Inherit Metab Dis
ing and pathologic findings. AJNR Am J Neuroradiol 2000; 1997; 20: 49–53
21: 224–227 De Meirleir L, Senaca S, Lissens W, Schoentjes E, Desprechins B.
Tulinus M, Moslemi AR, Darin N, Westerberg B, Wiklund LM, Bilateral striatal necrosis with a novel point mutation in the
Holme E, Oldfors A. Leigh syndrome with cytochrome-c ox- mitochondrial ATPase 6 gene. Pediatr Neurol 1995; 13:
idase deficiency and a single T insertion nt 5537 in the mi- 242–246
tochondrial tRNAtrp gene. Neuropediatrics 2003; 34: 87–91 De Vries DD, van Engelen BGM, Gabreëls FJM, Ruitenbeek W,
Valnot I, Osmond S, Gigarel N, Mehaye B, Amiel J Cormier-Daire van Oost BA. A second missense mutation in the mitochon-
V, Munnich A, Bonnefont J-P, Rustin P, Rötig A. Mutations of drial ATPase 6 gene in Leigh’s syndrome. Ann Neurol 1993;
the SCO1 gene in mitochondrial cytochrome c oxidase 34: 410–412
deficiency with neonatal-onset hepatic failure and en- Dionisi-Vici C, Seneca S, Zeviani M, Fariello G, Rimoldi M, Berti-
cephalopathy. Am J Hum Genet 2000; 67: 1104–1109 ni E, de Meirleir L. Fulminant Leigh syndrome and sudden
Valnot I, von Kleist-Retzow J-C, Barrientos A, Gorbatyuk M, unexpected death in a family with the T9176C mutation of
Taanman J-W, Mehanye B, Rustin P, Tzagoloff A, Munnich A, the mitochondrial ATPase 6 gene. J Inherit Metab Dis 1998;
Rötig A. A mutation in the human heme A: farnesyltrans- 21: 2–8
ferase gene (COX10) causes cytochrome c oxidase deficien- Ferlin T, Landrieu P, Ramboud C, Fernandez H, Dumoulin R,
cy. Hum Mol Genet 2000; 9: 1245–1249 Rustin R, Mousson B. Segregation of the G8993 mutant mi-
Van Coster R, Lombes A, De Vivo DC, Chi TL, Dodson WE, Roth- tochondrial DNA trough generations and embryonic tis-
man S, Orrechio EJ, Grover W, Berry GT, Schwartz JF, Habib A, sues in a family at risk of Leigh syndrome. J Pediatr 1997;
DiMauro S. Cytochrome c oxidase-associated Leigh syn- 131: 447–449
drome: phenotypic features and pathogenetic specula- Fryer A, Appleton R, Sweeney MG, Rosenbloom L, Harding AE.
tions. J Neurol Sci 1991; 104: 97–111 Mitochondrial DNA 8993 (NARP) mutation presenting with
Varlamov DA, Kudin AP,Vielhaber S, Schröder R, Sassen R, Beck- a heterogeneous phenotype including ‘cerebral palsy’. Arch
er A, Kunz D, Haug K, Rebstock J, Heils A, Elger CE, Kunz WS. Dis Child 1994; 71: 419–422
Metabolic consequences of a novel missense mutation of Fujii T, Hattori H, Higuchi Y,Tsuji M, Mitsuyoshi I. Phenotypic dif-
the mtDNA CO 1 gene.Hum Mol Genet 2002; 11:1997–1805 ferences between T C and T G mutations at nt 8993 of mito-
Von Kleist-Retzow J-C,Yao J,Taanman J-W, Chantrel K, Chretien chondrial DNA in Leigh syndrome. Pediatr Neurol 1998; 18:
D, Cormier-Daire V, Rötig A, Munnich A, Rustin P, Shoubridge 275–277
EA. Mutations in SURF1 are not specifically associated with García JJ, Ogilvie I, Robinson BH, Capaldi RA. Structure, func-
Leigh syndrome. J Med Genet 2001; 2001: 109–113 tioning, and assembly of the ATP synthase in cells from pa-
Willis TA, Davidson J, Gray RGF, Poultron K, Ramani P, White- tients with the T8993G mitochondrial DNA mutation. J Biol
house W. Cytochrome oxidase deficiency presenting as Chem 2000; 275: 11075–11081
birth asphyxia. Dev Med Child Neurol 2000; 42: 414–417 Holt IJ, Harding AE, Petty RKH, Morgan-Hughes JA. A new mito-
Zafeiriou DI, Koletzko B, Mueller-Felber W, Paetzke I, Kueffer G, chondrial disease associated with mitochondrial DNA het-
Jensen M. Deficiency in complex IV (cytochrome c oxidase) eroplasmy. Am J Hum Genet 1990; 46: 428–433
of the respiratory chain, presenting as a leukodystrophy in Leshinsky-Sliver E, Perach M, Basilevsky E, Hershkovitz E,
two siblings with Leigh syndrome. Brain Dev 1995; 17: Yanoov-Sharav M, Lerman-Sagie T, Leve D. Prenatal exclu-
117–121 sion of Leigh syndrome due to T8993C mutation in the mi-
Zeviani M, Corona P, Nijtmans L,Tiranti V. Nuclear gene defects tochondrial DNA. Prenat Diagn 2003; 23: 31–33
in mitochondrial disorders. Ital J Nuerol Sci 1999; 20: 401– Lodi R, Montagna P, Iotti S, Zaniol P, Barboni P, Puddo P, Barbiroli
408 B. Brain and muscle energy metabolism studied in vivo by
Zhu Z, Yao J, Johns T, Fu K, de Bie I, Macmillan C, Cuthbert AP, 31P-magnetic resonance spectroscopy in NARP syndrome.
Newbold RF, Wang J-C, Brown GK, Brown RM, Shoubridge J Neurol Neurosurg Psychiatry 1994; 57: 1492–1496
AE. SURF1, encoding a factor involved in the biogenesis of Mak S-C, Chi C-S, Tsai C-R. Mitochondrial DNA 8993T >C Muta-
cytochrome c oxidase, is mutated in Leigh syndrome. Nat tion presenting as juvenile Leigh syndrome with respirato-
Genet 1998; 20: 337–343 ry failure. J Child Neurol 1998; 13: 349–351
References and Further Reading 955

Mäkelä-Bengs P, Suomalainen A, Majander A, Rapola J, Kalimo Wilson CJ, Wood NW, Leonard JV, Surtees R, Rahman S. Mito-
H, Nuutila A, Pihko H. Correlation between the clinical chondrial DNA point mutation T9176C in Leigh syndrome.
symptoms and the proportion of mitochondrial DNA carry- J Child Neurol 2000; 15: 830–833
ing the 8993 point mutation in the NARP syndrome.Pediatr Yamada T, Hayasaka S, Hongo K, Kubota H. Retinal dystrophy in
Res 1995; 37: 634–639 a Japanese boy harboring the mitochondrial DNA T8993G
Nagashima T, Mori M, Katayama K, Nunomura M, Nishihara H, mutation. Jpn J Ophthalmol 2002; 46: 460–462
Higara H, Tanaka S Goto Y-I, Nagashima K. Adult Leigh syn-
drome with mitochondrial DNA mutation at 8993. Acta
Neuropathol (Berl) 1999; 97: 416–422
Pastores GM, Santorelli FM, Shanske S, Gelb BD, Fyfe B,Wolfe D,
29 Pyruvate Carboxylase Deficiency
Willner JP.Leigh syndrome and hypertrophic cardiomyopa-
thy in an infant with a mitochondrial DNA point mutation Ahmad A, Kahler SG, Kishnani PS, Artigas-Lopez M, Pappu AS,
(T8993G). Am J Med Genet 1994; 50: 265–271 Steiner R, Millington DS, van Hove JLK. Treatment of pyru-
Porto FBO, Mack G, Sterboul M-J, Lewin P, Flament J, Sahel J, vate carboxylase deficiency with high doses of citrate and
Dofflus H. Isolated late-onset cone-rod dystrophy revealing aspartate. Am J Med Genet 1999; 87: 331–338
a familial neurogenic muscle weakness, ataxia, and retinitis Arnold GL, Griebel ML, Porterfield M, Brewster M. Pyruvate car-
pigmentosa syndrome with the T8993G mitochondrial mu- boxylase deficiency. Clin Pediatr 2001; 40: 519–521
tation. Am J Ophthalmol 2001; 132: 935–937 Atkin BM, Buist NRM, Utter MF, Leiter AB, Banker BQ. Pyruvate
Santorelli FM, Shanske S, Macaya A, DeVivo DC, DiMauro S.The carboxylase deficiency and lactic acidosis in a retarded
mutation at nt 8993 of mitochondrial DNA is a common child without Leigh’s disease. Pediatr Res 1979; 13: 109–116
cause of Leigh’s syndrome. Ann Neurol 1993; 34: 827–834 Baal MG, Gabreëls FJM, Renier WO, Hommes FA, Gijsbers THJ,
Santorelli FM, Shanske S, Jain KD, Tick D, Schon EA, DiMauro S. Lamers KJB, Kok JCN. A patient with pyruvate carboxylase
A T C mutation at nt 8993 of mitochondrial DNA in a child deficiency in the liver: treatment with aspartic acid and thi-
with Leigh syndrome. Neurology 1994; 44: 972–974 amine. Dev Med Child Neurol 1981; 23: 521–530
Santorelli FM, Mak S-C, Vazquez-Memije ME, Shanske S, Kranz- Bartlett K, Ghneim HK, Stirk JH, Dale G, Alberti GMM. Pyruvate
Eble P, Jain KD, Bleustone DL, de Vivo DC, DiMauro S. Clinical carboxylase deficiency. J Inherit Metab Dis 1984; 7: 74–78
heterogeneity associated with the mitochondrial DNA Brun N, Robitaille Y, Grignon A, Robinson A, Robinson BH,
T8993C point mutation. Pediatr Res 1996; 39: 914–917 Mitchell GA, Lambert M. Pyruvate carboxylase deficiency :
Shoffner JM, Fernhoff PM, Krawiecki NS, Caplan DB, Holt PJ, prenatal onset of ischemia-like brain lesions in two sibs
Koontz DA,Takei Y, Newman NJ, Ortiz RG, Polak M, Ballinger with the acute neonatal form. Am J Med Genet 1999; 84:
SW, Lott MT, Wallace DC. Subacute necrotizing encephalo- 94–101
pathy: oxidative phosphorylation defects and the ATPase 6 Carbone MA, MacKay N, Ling M, Cole DEC, Douglas C, Rigat B,
point mutation. Neurology 1992; 42: 2168–2174 Feigenbaum A, Clarke JTR, Haworth JC, Greenberg CR, Sear-
Suzuki Y,Wada T, Sakai T, Ishikawa Y, Minami R,Tachi N, Saitoh S. gant L, Robinson BH. Amerindian pyruvate carboxylase de-
Phenotypic variability in a family with a mitochondrial DNA ficiency is associated with two distinct missense mutations.
T8993C mutation. Pediatr Neurol 1998; 19: 283–286 Am J Hum Genet 1998; 62: 1312–1319
Takahashi S, Oki J, Miyamoto A, Okuno A. Proton magnetic res- Carbone MA, Applegarth DA, Robinson BH. Intron retention
onance spectroscopy to study the metabolic changes in and frameshift mutations result in severe pyruvate car-
the brain of a patient with Leigh syndrome. Brain Dev 1999; boxylase deficiency in two male siblings. Hum Mutat 2002;
21: 200–204 20: 48–56
Takanashi J-I, Sugita K, Tanabe Y, Maetomo T, Niimi H. Greter J, Gustafsson J, Holme E. Pyruvate-carboxylase deficien-
Dichloroacetate treatment in Leigh syndrome caused by cy with urea cycle impairment. Acta Paediatr Scand 1985;
mitochondrial DNA mutation. J Neurol Sci 1997; 145: 83–86 74: 982–986
Tatuch Y, Christodoulou J, Feigenbaum A, Clarke JTR,Wherret J, Hamilton J, Rae MD, Logan RW, Robinson PH. A case of benign
Smith C, Rudd N, Petrova-Benedict R, Robinson BH. Hetero- pyruvate carboxylase deficiency with normal develop-
plasmic mtDNA mutation (TÆG) at 8993 can cause Leigh ment. J Inherit Metab Dis 1997; 20: 401–403
disease when the percentage of abnormal mtDNA is high. Higgins JJ, Glasgow AM, Lusk M, Kerr DS. MRI, clinical, and bio-
Am J Hum Genet 1992; 50: 852–858 chemical features of partial pyruvate carboxylase deficien-
Tsao C-Y, Mendell JR, Bartholomew D. High mitochondrial DNA cy. J Child Neurol 1994; 9: 436–439
T8993G mutation (>90 %) without typical features of Higgins JJ, Ide SE, Oghalai JS, Polymeropoulos MH. Lack of mu-
Leigh’s and NARP syndromes. J Child Neurol 2001; 16: tations in the biotin-binding region of the pyruvate car-
533–535 boxylase (PC) gene in a family with partial PC deficiency.
Uziel G, Moroni I, Lemantea E, Fratta GM, Ciceri E, Carrara E, Ze- Clin Biochem 1997; 30: 79–81
viani M. Mitochondrial disease associated with the T8993G Murphy JV, Isohashi F, Weinberg MB, Utter MF. Pyruvate car-
mutation of the mitochondrial ATPase 6 gene: a clinical, boxylase deficiency: an alleged biochemical cause of
biochemical, and molecular study in six families. J Neurol Leigh’s disease. Pediatrics 1981; 368: 401–404
Neurosurg Psychiatry 1997; 63: 16–22 Oizumi J, Shaw KNF, Giudici TA, Carter M, Donnell GN, Ng WG.
Vászques-Memije ME, Shanske S, Santorelli FM, Franz-Elbe P, Neonatal pyruvate carboxylase deficiency with renal tubu-
DeVivo DC, DiMauro S. Comparative biochemical studies of lar acidosis and cysturia. J Inherit Metab Dis 1983; 6: 89–94
ATPases in cells from patients with the T8993G or T8993C Oizumi J, Donnell GN, Ng WG, Mulivor RA, Greene AE, Coriell LL.
mitochondrial DNA mutations. J Inherit Metab Dis 1998; 21: Congenital lactic acidosis associated with pyruvate car-
829–836 boxylase deficiency. Cytogenet Cell Genet 1984; 38: 81
White SL, Shanske S, Biros I,Warwick L, Dahl HM,Thornburn DR, Pineda M, Campistol J, Vilaseca MA, Briones P, Ribes A, Temudo
Di Mauro S. Two cases of prenatal analysis for the patho- T, Pons M, Cusi V, Rolland M-O. An atypical French form of
genic T to G substitution at nucleotide 8993 in mitochondr- pyruvate carboxylase deficiency. Brain Dev 1995;17: 276–
ial DNA. Prenat Diagn 1999; 19: 1165–1168 279
956 References and Further Reading

Robinson BH, Oei J, Sherwood WG, Applegarth D, Wong L, Ha- 30 Multiple Carboxylase Deficiency
worth J, Goodyer P, Casey R, Zaleski LA.The molecular basis
for the two different clinical presentations of classical pyru- Bakker HD, Westra M, Overweg-Plandsoen WCG, Waveren van
vate carboxylase deficiency. Am J Hum Genet 1984; 36: G, Sillevis-Smitt JH, Abeling NGGM, Wanders RJA, Schut-
283–294 gens RBH, Gennip van AH. Normalisation of severe cranial
Robinson BH, Toone JR, Benedict P, Dimmick JE, Oei J, Apple- CT scan abnormalities after biotin in a case of biotinidase
garth DA. Prenatal diagnosis of pyruvate carboxylase defi- deficiency. Eur J Pediatr 1994; 153: 861–866
ciency. Prenat Diagn 1985; 5: 67–71 Baumgartner ER, Suormala TM,Wick H, Probst A, Blauenstein U,
Robinson BH, Oei J, Saudubray JM, Marsac C, Bartlett K, Quan F, Bachmann C,Vest M. Biotinidase deficiency: a cause of sub-
Gravel R. The French and North American phenotypes of acute necrotizing encephalomyelopathy (Leigh syn-
pyruvate carboxylase deficiency, correlation with biotin drome). Report of a case with lethal outcome. Pediatr Res
containing protein by 3H-biotin incorporation, 35S-strepta- 1989; 26: 260–266
vidin labeling, and Northern blotting with a cloned cDNA Baumgartner ER, Suormala T. Multiple carboxylase deficiency:
probe. Am J Hum Genet 1987; 40: 50–59 inherited and acquired disorders of biotin metabolism. Int
Robinson BH, MacKay N, Chun K, Ling M. Disorders of pyruvate J Vitam Nutr Res 1997; 67: 377–384
carboxylase and the pyruvate dehydrogenase comples. J Bay CA, Berry GT, Glauser TA, Hayward JC, Wolf B, Sladky JT,
Inherit Metab Dis 1996; 19: 452–462 Kaplan P. Reversible metabolic myopathy in biotinidase
Rutledge SL, Snead OC, Kelly DR, Kerr DS, Swann JW, Spink DL, deficiency: its possible role in causing hypotonia. J Inherit
Martin DL. Pyruvate carboxylase deficiency: acute exacer- Metab Dis 1995; 18: 701–704
bation after ACTH treatment of infantile spasms. Pediatr Bousounis DP, Camfield PR, Wolf B. Reversal of brain atrophy
Neurol 1989; 5: 249–252 with biotin treatment in biotinidase deficiency. Neuropedi-
Sander J, Packman S, Berg BO, Hutchison HT, Caswell N. Pyru- atrics 1993; 24: 214–217
vate carboxylase activity in subacute necrotizing en- Casado de Frías E, Campos-Castellü J, Careaga Maldonado J,
cephalopathy (Leigh’s disease). Neurology 1984; 34: 515– Pérez Cerdá C. Biotinidase deficiency: result of treatment
516 with biotin from age 12 years. Eur J Paediatr Neurol 1997; 1:
Saudubray JM, Marsac C, Charpentier C, Cathelineau L, Leaud 173–176
MB, Leroux JP. Neonatal congenital lactic acidosis with Cornejo Navarro P, Guerra A, Alvarez JG, Ortiz FJ. Cutaneous
pyruvate carboxylase deficiency in two siblings.Acta Paedi- and neurologic manifestations of biotinidase deficiency.Int
atr Scand 1976; 65: 717–724 J Dermatol 2000; 39: 363–382
Stern HJ, Nayar R, Depalma L, Fifai N. Prolonged survival in Fuchshuber A, Suormala T, Roth B, Duran M, Michalk D, Baum-
pyruvate carboxylase deficiency : lack of correlation with gartner ER. Holocarboxylase synthetase deficiency: early
enzyme activity in cultured fibroblasts. Clin Biochem 1995; diagnosis and management of a new case. Eur J Pediatr
28: 85–89 1993; 152: 446–449
Tada K, Takada G, Omura K, Itokawa Y. Congenital lactic acido- Gibson KM, Bennett MJ, Nyhan WL, Mize CE. Late-onset holo-
sis due to pyruvate carboxylase deficiency: absence of an carboxylase synthetase deficiency. J Inherit Metab Dis
inhibitor of TPP-ATP phosphoryltransferase. Eur J Pediatr 1996; 19: 739–742
1978; 127: 141–147 Ginat-Israeli T, Hurvitz H, Klar A, Blinder G, Branski D, Amir N.
Tsuchiyama A, Oyanagi K, Hirano S, Tachi N, Sogawa H, Wagat- Deteriorating neurological and neuroradiological course in
suma K, Nakao T,Tsugawa S, Kawamura Y.A case of pyruvate treated biotinidase deficiency. Neuropediatrics 1993; 24:
carboxylase deficiency with later prenatal diagnosis of an 103–106
unaffected sibling. J Inherit Metab Dis 1983; 6: 85–88 Haagerup A, Brandt Andersen J, Blichfeldt S, Fjord Christensen
Van Coster RN, Fernhoff PM, De Vivo DC. Pyruvate carboxylase M. Biotinidase deficiency: two cases of very early presenta-
deficiency: a benign variant with normal development. tion. Dev Med Child Neurol 1997; 39: 832–835
Pediatr Res 1991; 30: 1–4 Honavar M, Janota I, Neville BGR, Chalmers RA. Neuropatholo-
Van Coster RN, Janssens S, Misson J-P, Verloes A, Leroy JG. Pre- gy of biotinidase deficiency. Acta Neuropathol (Berl) 1992;
natal diagnosis of pyruvate carboxylase deficiency by di- 84: 461–464
rect measurement of catalytic activity on chorionic villi Hymes J,Wolf B. Biotinidase and its roles in biotin metabolism.
samples. Prenat Diagn 1998; 18: 1041–1044 Clin Chim Acta 1996; 255: 1–11
Wallace JC, Jitrapakdee S, Chapman-Smith S. Pyruvate car- Kalayci Ö, Coskun T,Tokatli A, Demir E, Erdem G, Güngör C,Yük-
boxylase. Biochem Cell Biol 1998; 30: 1–5 selen A, Özalp I. Infantile spasms as the initial symptom of
Wexler ID,Du Y,Lisgaris MV,Mandal SK,Freytag SO,Yang B-S,Liu biotinidase deficiency. J Pediatr 1994; 124: 103–104
T-C, Kwon M, Patel MS, Kerr DS. Primary amino acid se- Kimura M, Fukui T, Tagami Y, Fujiwaki T, Yokoyama M, Ishioka C,
quence and structure of human pyruvate carboxylase. Kumasaka K, Terada N, Yamaguchi S. Normalization of low
Biochim Biophys Acta 1994; 1227: 46–52 biotinidase activity in a child with biotin deficiency after
Wexler ID, Kerr DS, Du Y, Kaung MM, Stephenson W, Lusk MM, biotin supplementation. J Inherit Metab Dis 2003; 26:
Wappner RS, Higgins JJ. Molecular characterization of pyru- 715–719
vate carboxylase deficiency in two consanguineous fami- Livne M, Gibson M, Amir N, Eshel G, Elpeleg ON. Holocarboxy-
lies. Pediatr Res 1998; 43: 579–584 lase synthetase deficiency: a treatable metabolic disorder
Wong LTK, Davidson GF, Applegarth DE, Dimmick JE, Norman masquerading as cerebral palsy. J Child Neurol 1994; 9:
MG, Toone JR, Pirie G, Wong J. Biochemical and histologic 170–172
pathology in an infant with cross-reacting material (nega- Mardach R, Zempleni J, Wolf B, Cannon MJ, Jennings ML, Cress
tive) pyruvate carboxylase deficiency. Pediatr Res 1986; 20: S, Boylan J, Roth S, Cederbaum S, Mock DM. Biotin depen-
274–279 dency due to a defect in biotin transport. J Clin Invest 2002;
109: 1617–1623
References and Further Reading 957

Mitchell G, Ogier H, Munnich A, Saudubray JM. Neurological Wolf B, Heard GS. Biotinidase deficiency. Adv Pediatr 1991; 38:
deterioration and lactic acidemia in biotinidase deficiency 1–21
– a treatable condition mimicking Leigh’s disease. Neuro- Wolf B, Hsia YE, Sweetman L, Feldman G, Boychuk RB, Bart RD,
pediatrics 1986; 17: 129–131 Crowell DH, Di Mauro RM, Nyhan WL. Multiple carboxylase
Möslinger D,Stöckler-Ipsiroglu S,Scheibenreiter S,Tiefenthaler deficiency: clinical and biochemical improvement follow-
M, Mühl A, Seidl R, Strobl W, Plecko B, Suormala T, Regula ing neonatal biotin treatment. Pediatrics 1981; 68: 113–118
Baumgartner ER. Clinical and neuropsychological outcome Wolf B, Heard GS, Weissbecker KA, Secor McVoy JR, Grier RE,
in 33 patients with biotinidase deficiency ascertained by Leshner RT. Biotinidase deficiency: initial clinical features
nationwide newborn screening and family studies in Aus- and rapid diagnosis. Ann Neurol 1985; 18: 614–617
tria. Eur J Pediatr 2001; 160: 277–282 Wolf B, Norrgard K, Pomponio RJ, Mock DM, Secor McVoy JR,
Pomponio RJ, Hymes J, Reynolds TR, Meyers GA, Fleischhauer Fleischhauer K, Shapiro S, Blitzer MG, Hymes J. Profound
K, Buck GA, Wolf B. Mutations in the human biotinidase biotinidase deficiency in two asymptomatic adults. Am J
gene that cause profound biotinidase deficiency in symp- Med Genet 1997; 73: 5–9
tomatic children:molecular,biochemical,and clinical analy- Wolf B, Pomponio RJ, Norrgard KJ, Lott IT, Regula Baumgartner
sis. Pediatr Res 1997; 42: 840–848 E, Suormala T, Ramaekers VTh, Coskun T, Tokatli A, Ozalp I,
Rahman S,Standing S,Dalton RN,Pike MG.Late presentation of Hymes J. Delayed-onset profound biotinidase deficiency.
biotinidase deficiency with acute visual loss and gait dis- J Pediatr 1998; 132: 362–365
turbance. Dev Med Child Neurol 1997; 39: 830–831 Wolf B, Spencer R, Gleason T. Hearing loss is a common feature
Ramaekers VT, Suormala TM, Brab M, Duran R, Heimann G, of symptomatic children with profound biotinidase defi-
Baumgartner ER. A biotinidase Km variant causing late ciency. J Pediatr 2002; 140: 242–246
onset bilateral optic neuropathy. Arch Dis Child 1992; 67: Wolf B, Jensen K, Hüner G, Demirkol M, Baykal T, Divry P, Rolland
115–119 MO, Perez-Cerdá C, Ugarte M, Straussberg R, Basel-Vana-
Ramaekers VT, Brab M, Rau G, Heimann G. Recovery from neu- gaite L, Baumgartner ER, Suormala T, Scholl S, Das AM,
rological deficits following biotin treatment in a bio- Schweitzer S, Pronicka E, Sykut-Cegielska J. Seventeen nov-
tinidase Km variant. Neuropediatrics 1993; 24: 98–102 el mutations that cause profound biotinidase deficiency.
Salbert BA, Astruc J, Wolf B. Ophthalmologic findings in bio- Mol Gen Metab 2002; 77: 108–111
tinidase deficiency. Ophthalmologica 1993; 206: 177–181
Salbert BA, Pellock JM, Wolf B. Characterization of seizures as-
sociated with biotinidase deficiency. Neurology 1993; 43:
1351–1355
31 Cerebrotendinous Xanthomatosis
Sander JE, Malamud N, Cowan MJ, Packman S, Amman AJ,Wara
DW. Intermittent ataxia and immunodeficiency with multi- Argov Z, Soffer D, Eisenberg S, Zimmerman Y. Chronic demyeli-
ple carboxylase deficiencies: a biotin-responsive disorder. nating peripheral neuropathy in cerebrotendinous xantho-
Ann Neurol 1980; 8: 544–547 matosis. Ann Neurol 1986; 20: 89–91
Schürmann M, Engelbrecht V, Lohmeier K, Lenard HG, Wendel Ballantyne CM, Vega GL, East C, Richards G, Grundy SM. Low-
U, Gärtner J. Cerebral metabolic changes in biotinidase de- density lipoprotein metabolism in cerebrotendinous
ficiency. J Inherit Metab Dis 1997; 20: 755–760 xanthomatosis. Metabolism 1987; 36: 270–276
Schultz PE, Weiner SP, Belmont JW, Fishman MA. Basal ganglia Barkhof F, Verrips A, van der Knaap MS, van Engelen BGM,
calcifications in a case of biotinidase deficiency. Neurology Gabreëls FJM, Keyser A, Wevers RA, Valk J. Cerebrotendi-
1988; 38: 1326–1328 nous xanthomatosis: the spectrum of imaging findings and
Secor McVoy JR, Levy HL, Lawler M, Schmidt MA, Ebers DD, Hart the correlation with neuropathologic findings. Radiology
PS, Dove Pettit D, Blitzer MG,Wolf B. Partial biotinidase defi- 2000; 217: 869–876
ciency: clinical and biochemical features. J Pediatr 1990; Bencze KS, van de Polder DR, Prockop LD. Magnetic resonance
116: 78–83 imaging of the brain and spinal cord in cerebrotendinous
Suormala TM, Baumgartner ER, Wick H, Scheibenreiter S, xanthomatosis. J Neurol Neurosurg Psychiatry 1990; 53:
Schweitzer S. Comparison of patients with complete and 166–167
partial biotinidase deficiency: biochemical studies. J Inher- Berginer VM, Berginer J, Salen G, Shefer S, Zimmerman RD.
it Metab Dis 1990; 13: 76–92 Computed tomography in cerebrotendinous xanthomato-
Suormala T, Ramaekers VTh, Schweitzer S, Fowler B, Laub MC, sis. Neurology 1981; 31: 1463–1465
Schwermer C, Bachmann J, Baumgartner ER. Biotinidase Berginer VM, Salen G, Shefer S. Long-term treatment of cere-
Km-variants: detection and detailed biochemical investiga- brotendinous xanthomatosis with chenodeoxycholic acid.
tions. J Inherit Metab Dis 1995; 18: 689–700 N Engl J Med 1984; 311: 1649–1652
Suormala T, Fowler B, Jakobs C, Duran M, Lehnert W, Raab K, Berginer VM, Salen G, Shefer S.Cerebrotendinous xanthomato-
Wick H, Baumgartner ER. Late-onset holocarboxylase syn- sis. Neurol Clin 1989; 7: 55–74
thetase-deficiency: pre- and post-natal diagnosis and eval- Berginer VM, Berginer J, Korczyn AD, Tamor R. Magnetic reso-
uation of effectiveness of antenatal biotin therapy. Eur J nance imaging in cerebrotendinous xanthomatosis: a
Pediatr 1998; 157: 570–575 prospective clinical and neuroradiological study. J Neurol
Sweetman L, Nyhan WL. Inheritable biotin-treatable disorders Sci 1994; 122: 102–108
and associated phenomena. Annu Rev Nutr 1986; 6: 317– Canelas HM, Quintao ECR, Scaff M,Vasconcelos KS, Brotto MWI.
343 Cerebrotendinous xanthomatosis: clinical and laboratory
Tsao CY, Kien CL. Complete biotinidase deficiency presenting study of 2 cases. Acta Neurol Scand 1983; 67: 305–311
as reversible progressive ataxia and sensorineural deaf- Chen W, Kubota S, Teramoto T, Ishida S, Ohsawa N, Katayama T,
ness. J Child Neurol 2002; 17: 146 Takeda T, Kuroda K,Yahara O, Kasuhara T, Neshige R, Seyama
Wiznitzer M, Bangert BA. Biotinidase deficiency: clinical and Y. Genetic analysis enables definite and rapid diagnosis of
MRI findings consistent with myelopathy. Pediatr Neurol cerebrotendinous xanthomatosis. Neurology 1998; 51:
2003; 29: 56–58 865–867
958 References and Further Reading

Claydon PT, Verrips A, Sisterman E, Mann A, Mieli-Vergani G, Leitersdorf E, Reshef A, Meiner V, Levitzki R, Pressman Schwartz
Wevers R. Mutations in the sterol 27-hydrolase gene S, Dann EJ, Berkman N, Cali JJ, Klapholz L, Berginer VM.
(CYP27A) cause hepatitis of infancy as well as cerebrotendi- Frameshift and splice-junction mutations in the sterol
nous xanthomatosis. J Inherit Metab Dis 2002; 25: 501–513 27-hydroxylase gene cause cerebrotendinous xan-
De Stefano N, Dotti MT, Mortilla M, Frederico A. Magnetic reso- thomatosis in Jews of Moroccan origin. J Clin Invest 1993;
nance imaging and spectroscopic changes in brains of pa- 91: 2488–2496
tients with cerebrotendinous xanthomatosis. Brain 2001; Lui C-C, Chang W-N. MRI changes in cerebrotendinous xan-
124: 121–131 thomatosis after treatment with chenodeoxycholic acid.
Dotti MT, Frederico A. Cerebrotendinous xanthomatosis with Riv Neuroradiol 1998; 11: 10–11
predominant parkinsonian syndrome:further confirmation Meiner V, Meiner Z, Reshef A, Björkem I, Leitersdorf E. Cerebro-
of the clinical heterogeneity. Mov Disord 2000; 15: 1017– tendinous xanthomatosis: molecular diagnosis enables
1019 presymptomatic detection of a treatable disease. Neurolo-
Dotti MT, Salen G, Federico A. Cerebrotendinous xanthomato- gy 1994; 44: 288–290
sis as a multisystem disease mimicking premature ageing. Moghadasian MH, Salen G, Frohlich JJ, Scudamore CH. Cere-
Dev Neurosci 1991; 13; 371–376 brotendinous xanthomatosis. A rare disease with diverse
Dotti MT, Federico A, Signorini E, Caputo N, Venturi C, Filosomi manifestations. Arch Neurol 2002; 59: 527–529
G, Guazzi GC. Cerebrotendinous xanthomatosis (van Bo- Mondelli M, Rossi A, Scarpini C, Dotti MT, Federico A. Evoked
gaert-Scherer-Epstein disease): CT and MR findings. AJNR potentials in cerebrotendinous xanthomatosis and effect
Am J Neuroradiol 1994; 15: 1721–1726 induce by chenodeoxycholic acid. Arch Neurol 1992; 49:
Dotti MT, Mondillo S, Plewnia K, Agricola E, Frederico A. Cere- 469–475
brotendinous xanthomatosis: evidence of lipomatous hy- Mondelli M, Sicurelli F, Scarpini C, Dotti MT, Frederico A. Cere-
pertrophy of the atrium septum. J Neurol 1998; 245: brotendinous xanthomatosis: 11-year treatment with
723–726 chenodeoxycholic acid in five patients. An electrophysio-
Dotti MT, Rufa A, Frederico A. Cerebrotendinous xanthomato- logical study. J Neurol Sci 2001; 190: 29–33
sis: heterogeneity of clinical phenotype with evidence of Ohno T, Kobayashi S, Hayasi M, Suakurai M, Kanazawa I.
previously undescribed ophthalmological findings. J Inher- Diphenylpyraline-responsive parkinsonism in cerebro-
it Metab Dis 2001; 24: 696–706 tendinous xanthomatosis: long-term follow up of three pa-
Elleder M, Michalec È, Jirásek A, Khun K, Havlová M, Ranný M. tients. J Neurol Sci 2001; 182: 95–97
Membranocystic lesion in the brain in cerebrotendinous Pedley TA, Emerson RG, Warner CL, Rowland LP, Salen G. Treat-
xanthomatosis. Histochemical and ultrastructural study ment of cerebrotendinous xanthomatosis with chen-
with evidence of its ceroid nature. Virchows Arch [B] 1989; odeoxycholic acid. Ann Neurol 1985; 18: 517–518
57: 367–374 Peynet J, Laurent A, de Liege P, Lecoz P, Gambert P, Legrand A,
Frederico A, Dotti MT. Cerebrotendinous xanthomatosis. Neu- Mikol J, Warnet A. Cerebrotendinous xanthomatosis: treat-
rology 2001; 57: 1743 ments with simvastatin, lovastatin, an chenodeoxycholic
Federico A,Dotti MT.Cerebrotendinous xanthomatosis:clinical acid in 3 siblings. Neurology 1991; 41: 434–436
manifestations, diagnostic criteria, pathogenesis and ther- Restuccia D, Di Lazzaro V, Servidei S, Colosimo C, Tonali P. So-
apy. J Child Neurol 2003; 18: 633–638 matosensory and motor evoked potentials in the assess-
Federico A, Dotti MT,Volpi N. Muscle mitochondrial changes in ment of cerebrotendinous xanthomatosis before and after
cerebrotendinous xanthomatosis. Ann Neurol 1991; 30: treatment with chenodeoxycholic acid: a preliminary
734–735 study. J Neurol Sci 1992; 112: 139–146
Fiorelli M, Di Piero V, Bastianello S, Bozzao L, Federico A. Cere- Salen G, Zaki G, Sabesin S, Boehme D, Sheper S, Mosbach EH.
brotendinous xanthomatosis: clinical and MRI study (case Intraphepatic pigment and crystal forms in patients with
report). J Neurol Neurosurg Psychiatry 1990; 53: 76–78 cerebrotendinous xanthomatosis (CTX). Gastroenterology
Hokezu Y, Kuriyama M, Kubota R, Nakagawa M, Fujiyama J, 1978; 74: 82–89
Osame M. Cerebrotendinous xanthomatosis: cranial CT and Salen G, Shefer S, Berginer V. Biochemical abnormalities in
MRI studies in eight patients. Neuroradiology 1992; 34: cerebrotendinous xanthomatosis. Dev Neurosci 1991; 13:
308–312 363–370
Javitt NB. Biologic role(s) of the 25(R),26-hydroxycholesterol Schimschock JR, Alvord EC, Swanson PD. Cerebrotendinous
metabolic pathway. Biochim Biophys Acta 2000; 1529: xanthomatosis. Arch Neurol 1968; 18: 688–698
136–141 Siebner HR, Berndt S, Conrad B. Cerebrotendinous xan-
Kawabata M, Kuriyama M, Mori S, Sakashita I, Osame M. Pul- thomatosis without tendon xanthomas mimichking Mari-
monary manifestations in cerebrotendinous xanthomato- nesco-Sjoegren syndrome: a case report. J Neurol Neuro-
sis. Intern Med 1998; 37: 922–926 surg Psychiatry 1996; 60: 582–585
Kuriyama M, Fujiyama J,Yoshidome H,Takenage S, Matsumuro Soffer D, Benharroch D, Berginer V. The neuropathology of
K, Kasama T, Fukuda K, Kuramoto T, Hoshita T, Seyama Y, cerebrotendinous xanthomatosis revisited: a case report
Okatu Y, Osame M. Cerebrotendinous xanthomatosis: clini- and review of the literature. Acta Neuropathol (Berl) 1995;
cal and biochemical evaluation of eight patients and re- 90: 213–220
view of the literature. J Neurol Sci 1991; 102: 225–232 Tai KS, Brockwell J, Chan FL, Janus ED, Lam KSL. Magnetic reso-
Lee M-H, Hazard S, Carpten DJ,Yi S, Cohen J, Gerhardt GT, Salen nance imaging of cerebrotendinous xanthomatosis. Aus-
G, Patel SB. Fine-mapping, mutation analysis, and structural tralas Radiol 1995; 39: 61–65
mapping of cerebrotendinous xanthomatosis in U.S. pedi- Tokimura Y, Kuriyama M, Arimura K, Fujiyama J, Osame M. Elec-
grees. J Lipid Res 2001; 42: 159–169 trophsiological studies in cerebrotendinous xanthomato-
sis. J Neurol Neurosurg Psychiatry 1992; 55: 52–55
References and Further Reading 959

Van Heijst AFJ,Wevers RA,Tangerman A,Cruysberg JRM,Renier Boltshauser E, Yalcinkaya C, Wichmann W, Reutter F, Prader A,
WO, Tolboom JJM. Chronic diarrhoea as a dominating Valavanis A. MRI in Cockayne syndrome type I. Neuroradiol-
symptom in two children with cerebrotendinous xan- ogy 1989; 31: 276–277
thomatosis. Acta Paediatr 1996; 85: 932–936 Castillo M, Thomas D, Mukherji SK. Facies to remember. Int J
Van Heijst AFJ,Verrips A,Wevers RA,Cruysberg JRM,Renier WO, Neuroradiol 1997; 3: 35–41
Tolboom JJM. Treatment and follow-up of childeren with Cirillo Silengo M, Franceschini P, Bianco R, Biagioli M, Pastorin L,
cerebrotendinous xanthomatosis. Eur J Pediatr 1998; 157: Vista N, Baldassar A, Benso L. Distinctive skeletal dysplasia
313–316 in Cockayne syndrome. Pediatr Radiol 1986; 16: 264–266
Van Rietvelde F, Lemmering M, Mespreuve M, Crevits L, De Citterio E, Van der Boom V, Schnitzler G, Kanaar R, Bonte E,
Reuck J, Kunnen M. MRI of the brain in cerebrotendinous Kingston RE, Hoeijmakers JHJ, Vermeulen W. ATP-depen-
xanthomatosis (van Bogaert-Scherer-Epstein disease). Eur dent chromatin remodeling by the Cockayne syndrome B
Radiol 2000; 10: 576–578 DNA repair–transcription-coupling factor. Mol Cell Biol
Verrips A, Lycklama à Nijeholt, GJ, Barkhof F, van Engelen BGM, 2000; 20: 7643–7653
Wesseling P, Luyten JAFM, Wevers RA, Stam J, Wokke JHJ, Colabucci F, Rossodivita A, Parigi A, Colavita N. A clinical and ra-
Van den Heuvel LPWJ, Keyser A, Gabreëls FJM. Spinal xan- diological study of two brothers affected by Cockayne syn-
thomatosis: a variant of cerebrotendinous xanthomatosis. drome type II. Rays 1987; 12: 57–63
Brain 1991; 122: 1589–1595 Dabbagh O, Swaiman KF. Cockayne syndrome: MRI correlates
Verrips A,Wevers RA, van Engelen BGM, Keyser A,Wolthers BG, of hypomyelination. Pediatr Neurol 1988; 4: 113–116
Barkhof F, Stalenhoef A, De Graaf R, Jansen-Zijlstra F, Van Del Bigio MR, Greenberg CR, Rorke LB, Schnur R, McDonald-
Spreeken A, Gabreëls FJM. Effect of Simvastatin in addition McGinn DM, Zackai EH.Neuropathological findings in eight
to chenoxycholic acid in patients with cerebrotendinous children wit cerebro-oculo-facio-skeletal (COFS) syndrome.
xanthomatosis. Metabolism 1999; 48: 233–238 J Neuropathol Exp Neurol 1997; 56: 1147–1157
Verrips A, van Engelen BGM, Ter Laak H, Gabreëls-Festen A, Demaerel P,Wilms G,Verdru P, Carton H, Baert AL. MRI in the di-
Janssen A, Zwarts M, Wevers RA, Gabreëls FJM. Cerebro- agnosis of Cockayne’s syndrome. One case. J Neuroradiol
tendinous xanthomatosis controversies about nerve and 1990; 17: 157–160
muscle: observations in ten patients. Neuromusc Disord Demaerel P, Kendall BE, Kingsley D. Cranial CT and MRI in dis-
2000; 10: 407–414 eases with DNA repair defects. Neuroradiology 1992; 34:
Verrips A, van Engelen BGM,Wevers RA, van Geel BM, Crusberg 117–121
JRM, van den Heuvel LPWJ, Keyser A, Gabreëls FJM. Pres- Friedberg EC. Cockayne syndrome – a primary defect in DNA
ence of diarrhea and abcense of tendom xanthomas in pa- repair, transcription, both or neither? Bioessays 1996; 18:
tients with cerebrotendinous xanthomatosis. Arch Neurol 731–738
2000; 57: 520–524 Graham JM Jr, Anyane-Yeboa K, Raams A, Appeldoorn E, Kleijer
Verrips A, Hoefsloot LH, Steenbergen GCH, Theelen JP, Wevers WJ, Garritsen VH, Busch D, Edersheim TG, Jaspers NGJ. Cere-
RA, Gabreëls JM, van Engelen BGM, Van den Heuvel LPWJ. bo-oculo-facio-skeletal syndrome with a nucleotide exci-
Clinical and molecular genetic characteristics of patients sion-repair defect and a mutated XPD gene, with prenatal
with cerebrotendinous xanthomatosis. Brain 2000; 123: diagnosis in a triplet pregnancy. Am J Hum Genet 2001; 69:
908–919 291–300
Wakamatsu N, Hayashi M, Kawai H, Kondo H, Gotoda Y, Nishida Grunnet ML, Zimmerman AW, Lewis RA. Ultrastructure and
Y, Kondo S, Matsumoto T. Mutations producing premature electrodiagnosis of peripheral neuropathy in Cockayne’s
termination of translation and an an amino acid substitu- syndrome. Neurology 1983; 33: 1606–1609
tion in the sterol 27-hydroxylase gene cause cerebrotendi- Hanawalt PC. The basis for Cockayne syndrome. Nature 2000;
nous xanthomatosis associated with parkinsonism. J Neu- 405: 415–416
rol Neurosurg Psychiatry 1999; 67: 195–198 Harbord MG, Finn JP, Hall-Craggs MA, Brett EM, Baraitser M. Ear-
Wevers RA,Cruysberg JRM,van Heijst AFJ,Janssen-Zijlstra FSM, ly onset leukodystrophy with distict facial features in 2 sib-
Renier WO,van Engelen BGM,Tolboom JJM.Paediatric cere- lings. Neuropediatrics 1989; 20: 154–157
brotendinous xanthomatosis. J Inherit Metab Dis 1992; 15: Hayashi M, Hayakawa K, Suzuki F, Sugita K, Satoh J, Morimatsu
374–376 Y. A neuropathological study of early onset Cockayne syn-
drome with chromosomal anomaly 47XXX. Brain Dev 1992;
14: 63–67
Hayashi M, Itoh M, Araki S, Kumada S, Shioda K, Tamagawa K,
32 Cockayne Syndrome Mizutaini T, Morimatsu Y, Minagawa M, Oda M. Oxidative
stress and disturbed glutamate transport in hereditary nu-
Balajee AS, De Santis LP, Brosh Jr RM, Selzer R, Bohr VA. Role of cleotide repair disorders. J Neuropathol Exp Neurol 2001;
the ATPase domain of the Cockayne syndrome group B 60: 350–356
protein in UV induced apoptosis. Oncogene 2000; 19: Houston CS, Zaleski WA, Rozdilsky B. Identical male twins and
477–489 brother with Cockayne syndrome. Am J Med Genet 1982;
Berneberg M, Lowe JE, Nardo T, Araújo S, Fousteri MI, Green 13: 211–223
MHL, Krutmann J, Wood RD, Stefanini M, Lehmann AR. UV Itoh M, Hayashi M, Shioda K, Minagawa M, Isa F, Tamagawaa K,
damage causes uncontrolled DNA breakage in cells from Morimatsu Y, Oda M. Neurodegeneration in hereditary nu-
patients with combined features of XP-D and Cockayne cleotide repair disorders. Brain Dev 1999; 21: 326–333
syndrome. EMBO J 2000; 19: 1157–1166 Kohji T, Hayashi M, Shioda K, Minagawa M, Morimatsu Y, Tama-
Berneburg M, Lehmann AR. Xeroderma pigmentosum and re- gawa K, Oda M. Cerebellar neurodegeneration in human
lated disorders: defect in DNA repair and transcription. Adv hereditary DNA repair disorders. Neurosci Lett 1998; 243:
Genet 2000; 43: 71–102 133–136
Bohr VA. Human premature aging syndrome and genomic in-
stability. Mech Ageing Dev 2002; 123: 987–993
960 References and Further Reading

Lee S-K, Yu S-L, Prakash L, Prakash S. Requirement of yeast Sakai T, Kikuchi F,Takashima S, Matsuda H,Watanabene N. Neu-
RAD2, a homolog of human XPG gene, for efficient RNA ropathological findings in the cerebro-oculo-facio-skeletal
polymerase II transcription: implications for Cockayne syn- (Pena-Shokeir II) syndrome. Brain Dev 1997; 19: 58–62
drome. Cell 2002; 109: 823–834 Sasaki K,Tachi N, Shinoda M, Satoh N, Minami R, Ohnishi A. De-
Leech RW, Brumback RA, Miller RH, Otsuka F, Tarone RE, Rob- myelinating peripheral neuropathy in Cockayne syndrome:
bins JH. Cockayne syndrome: clinicopathologic and tissue a histopathologic and morphometric study. Brain Dev
culture studies of affected siblings. J Neuropathol Exp Neu- 1992; 14: 114–117
rol 1985; 44: 507–519 Sato H, Saito T, Kurosawa K, Ootaka T, Furuyama T,Yoshinaga K.
Lehmann AR, Thompson AF, Harcourt SA, Stefanini M, Norris Renal lesions in Cockayne’s syndrome. Clin Nephrol 1988;
PG. Cockayne’s syndrome: correlation of clinical features 29: 206–209
with cellular sensitivity of RNA synthesis to UV irradiation. J Savary JB, Vasseur F, Deminatti MM. Routine autorediographic
Med Genet 1993; 30: 679–682 analysis of DNA excision-repair. Report of prenatal and
Licht CL, Stevsner T, Bohr VA. Cockayne syndrome group B cel- postnatal diagnosis in eleven families. Ann Genet 1991; 2:
lular and biochemical functions. Am J Hum Genet 2003; 73: 76–81
1217–1239 Smits MG, Gabreëls FJM, Renier WO, Joosten EMG, Gabreels-
Lindahl T, Karran P, Wood RD. DNA excision repair pathways. Festen AAWM, ter Laak HJ, Pinckers AJL, Hombergen GCJ,
Curr Opin Genet Dev 1997; 7: 159–169 Notermans SLH, Thijssen HOM. Peripheral and central
Lindenbaum Y, Dickson DW, Rosenbaum PS, Kraemer KH, Rob- myelinopathy in Cockayne’s syndrome. Neuropediatrics
bins JH, Rapin I. Xeroderma pigmentosum / Cockayne syn- 1982; 13: 161–167
drome complex: first neuropathological study and review Soffer D, Grotsky HW, Rapin I, Suzuki K. Cockayne syndrome:
of eight other cases. Eur J Pediatr Neurol 2001; 5: 225–242 unusual neuropathological findings and review of the liter-
Lowry RB. Early onset of Cockayne syndrome. Am J Med Genet ature. Ann Neurol 1979; 6: 340–348
1982; 13: 209–210 Sugita K,Takanashi J, Ishii M, Niimi H. Comparison of MRI white
Mallery DL,Tanganelli B, Colella S, Steingrimsdottir H,Van Gool matter changes with neuropsychologic impairment in
AJ,Troelstra C, Stefanini M, Lehmann AR. Molecular analysis Cockayne syndrome. Pediatr Neurol 1992; 8: 295–298
of mutations in the CSB (ERCC6) gene in patients with Cock- Takada K, Becker LE. Cockayne’s syndrome: report of two au-
ayne syndrome. Am J Hum Genet 1998; 62: 77–85 topsy cases associated with neurofibrillary tangles. Clin
Meira L, Graham Jr JM, Greenberg CR, Busch DB, Doughty ATB, Neuropathol 1986; 5: 64–68
Ziffer DW, Coleman DM, Savre-Train I, Friedberg EC. Mani- Talwar D, Smith SA. Camfak syndrome: a demyelinating inher-
tona aboriginal kindred with original cerebro-oculo-facio- ited disease similar to Cockayne syndrome. Am J Med
skeletal syndrome has a mutation in the Cockayne syn- Genet 1989; 34: 194–198
drome group B (CSB) gene. Am J Hum Genet 2000; 66: Traboulsi EI,de Becker I,Maumenee IH.Ocular findings in Cock-
1221–1228 ayne syndrome. Am J Ophthalmol 1992; 114: 579–583
Moyer DB, Marquis P, Shertzer ME, Burton BK. Cockayne syn- Van Gool AJ, van der Horst GTJ, Citterioo E, Hoeijmakers JHJ.
drome with early onset of manifestations. Am J Med Genet Cockayne syndrome: defective repair of transcription? EM-
1982; 13: 225–230 BO J 1997; 16: 4155–4162
Nance MA, Berry SA. Cockayne syndrome: review of 140 cases. Van Hoffen A, Kalle WHJ, De Jong-Versteeg A, Lehmann AR,Van
Am J Med Genet 1992; 42: 68–84 Zeeland AA, Mullenders LHF. Cells from XP-D and XP-D-CS
Nishio H, Kodama S, Matsuo T, Ichihashi M, Ito H, Fujiwara Y. patients exhibit equally inefficient of UV-repair damage in
Cockayne syndrome: magnetic resonance images of the transcribed genes but different capacity to recover UV-in-
brain in a severe form with early onset. J Inherit Metab Dis hibited transcription. Nucl Acids Res 1999; 27: 2898–2904
1988; 11: 88–102 Vos A, Gabreëls-Festen A, Joosten E, Gabreëls F, Renier W, Mul-
Norman RM, Tingey AH. Syndrome of micrencephaly, strio- laart R. The neuropathy of Cockayne syndrome. Acta Neu-
cerebellar calcifications, and leucodystrophy. J Neurol Neu- ropathol (Berl) 1983; 61: 153–156
rosurg Psychiatry 1966; 29: 157–163 Weliky-Conaway J, Conaway RC. Transcription elongation and
Ohnishi A, Mitsudome A, Murai Y. Primary segmental demyeli- human disease. Annu Rev Biochem 1999; 68: 301–319
nation in the sural nerve in Cockayne’s syndrome. Muscle Winter RM, Donna D, d’A Crawfurd M. Syndromes of micro-
Nerve 1987; 10: 163–167 cephaly, microphtalmia, cataracts, and joint contractures.
Özdirim E,Topçu M, Özön A, Cila A. Cockayne syndrome: review J Med Genet 1981; 18: 29–133
of 25 cases. Pediatr Neurol 1996; 15: 312–316 Zafeiriou DI,Thorel F, Andreou A, Kleijer WJ, Raams A, Garritsen
Patton MA, Giannelli F, Francis AJ, Baraiser M, Harding B, VH, Gombakis N, Jaspers NGJ, Clarkson SG. Xeroderma pig-
Williams AJ. Early onset Cockayne’s syndrome: case reports mentosum group G with severe neurological involvement
with neuropathological and fibroblast studies.J Med Genet and features of Cockayne syndrome in infancy. Pediatr Res
1989; 26: 154–159 2001; 49: 407–412
Pena SDJ, Shokeir MHK. Autosomal recessive cerbro-oculo-fa-
cio-skeletal (COFS) syndrome. Clin Genet 1974; 5: 285–293
Pena SDJ, Evans J, Hunter AGW. COFS syndrome revisited. Birth
Defects 1978; XIV: 205–213
33 Trichothiodystrophy
Rapin I. Lindenbaum Y, Dickson DW, Kraemer KH, Robbins JH. with Photosensitivity
Cockayne syndrome and xeroderma pigmentosum. DNA
repair disorders with overlaps and paradoxes. Neurology Battistella PA, Peserico A. Central nervous system dysmyelina-
2000; 55: 1442–1449 toion in PIBI(D)S syndrome: a further case. Childs Nerv Syst
Roy S, Srivastava RN, Gupta PC, Meyekar G. Ultrastructure of 1996; 12: 110–113
pheripheral nerve in Cockayne’s syndrome. Acta Neu- Bergmann E,Egly J-M.Trichothiodystrophy,a transcription syn-
ropathol (Berl) 1973; 24: 345–349 drome.Trends Genet 2001; 17: 279–286
References and Further Reading 961

Botta E, Nardo T, Broughton BC, Marinoni S, Lehmann AR, Ste- McGuaig C,Marcoux D,Rasmussen JE,Werner MM,Gentner NE.
fanini M. Analysis of mutations in the XPD gene in Italian Trichothiodystrophy associated with photosensitivity, go-
patients with trichothiodystrophy: site of mutation corre- nadal failure, and striking osteosclerosis. J Am Acad Derma-
lates with repair deficiency, but gene dosage appears to tol 1993; 28: 820–826
determine clinical severity. Am J Hum Genet 1998; 63: Murrin KL, Clarke DJ. Behavioral aspects of Pollitt syndrome: a
1036–1048 32-year follow-up of a case described by R.J. Pollitt and col-
Botta E, Nardo T, Lehmann AR, Egly JM, Pedrini AM, Stefanini M. leagues in 1968. J Intellect Disabil Res 2002; 46: 273–278
Reduced level of the repair/transcription factor TFIIH tri- Østergard JR, Christensen T. The central nervous system in Tay
chothiodystrophy. Hum Mol Genet 2002; 11: 2919–2928 syndrome. Neuropediatrics 1996; 27: 326–330
Bracun R, Hemmer W, Wolf-Abolvahab S, Focke M, Botzi C, Kil- Pescerico A, Battistella PA, Bertoli P. MRI of a very rare heredi-
lian W, Götz M, Jarisch R. Diagnosis of trichothiodystrophy tary ectodermal dysplasia: PIBI(D)S. Neuroradiology 1992;
in 2 siblings. Dermatology 1997; 194: 74–76 34: 316–317
Broughton BC, Steingrimsdottir H, Weber CA, Lehmann AR. Porto L, Weis R, Schulz C, Reichel P, Lanfermann H, Zanella FE.
Mutations in the xeroderma pigmentosum group D DNA Tay’s syndrome: MRI. Neuroradiology 2000; 42: 849–851
repair/transcription gene in patients with trichthiodystro- Queille S, Drougard C, Sarasin A, Daya-Grosjean L. Effects of
phy. Nat Genet 1994; 7: 189–194 XPD mutations on ultraviolet-induced apoptosis in relation
Broughton BC, Berneburg M, Fawcett H, Taylor EM, Arlett CF, to skin cancer-proneness in repair-deficient syndromes.
Nardo T, Stefanini M, Menefee E, Price VH, Queilli S, Sarasin J Invest Dermatol 2001; 117: 1162–1170
A, Bohnert E, Krutmann J, Davidson R, Kreamer KH, Racioppi L, Cancrini C, Romiti ML, Angelini F, di Cesare S, Berti-
Lehmann AR. Two individuals with features of both xero- ni E, Livadiotti S, Gambarara MG, Matarese G, Lago Paz F,
derma pigmentosum and trichothiodystrophy highlight Stefanini M, Rossi P. Defective dendritic cell maturation in a
the complexity of the clinical outcomes of mutations in the child with nucleotide excision repair deficiency and CD4
XPD gene. Hum Mol Genet 2001; 10: 2539–2547 lymphopenia. Clin Exp Immunol 2001; 126: 511–518
Cancrini C, Romiti ML, di Cesare S, Angelini F, Gigliotti D, Liva- Savary JB, Vasseur F, Vinatier D, Manouvrier S, Thomas P, Demi-
diotti S, Bertini E, Rossi R, Racioppi L. Restriction in T-cell re- natti MM. Prenatal diagnosis of PIBIDS. Prenat Diagn 1991;
ceptor repertoire in a patient affected by trichothiodystro- 11: 859–866
phy and CD4+ lymphopenia. Scand J Immunol 2002; 56: Stefanini M, Giliani S, Nardo T, Marinoni S, Nazzaro V, Rizzo R,
212–216 Trevisan G. DNA repair investigations in the nine Italian pa-
De Boer J, Hoeijmaker JHJ. Nucleotide excision repair and hu- tients affected by trichothiodystrophy. Mutat Res DNA Re-
man syndromes. Carcinogenesis 2000; 21: 453–460 pair 1992; 273: 119–125
Egly JM. TFIIH: from transcription to clinic. FEBS Lett 2001; Takayama K, Salazar EP, Brougthon BC, Lehmann AR, Sarasin A,
24884: 124–128 Thompson LH, Weber CA. Defects in the DNA repair and
Friedberg EC. Hot news: temperature-sensitive humans ex- transcription gene CRCC2(XPD) in trichothiodystrophy. Am
plain hereditary disease. Bioessays 2001; 23: 671–673 J Hum Genet 1996; 58: 263–270
Fortina AB, Alaibac M, Piaserico S, Pererico A. PIBI(D)S: clinical Taylor EM, Broughton BCM Botta E, Stefanini M, Sarasin A,
and molecular characterization of a new case. J Eur Acad Jaspers NGJ, Fawcett H, Harcourt SA, Arlett CF, Lehmann AR.
Dermatol Venereol 2001; 15: 65–69 Xeroderma pigmentosum and trichothiodystrophy are
Happle R, Traupe H, Gröbe H, Bonsmann G. The Tay syndrome associated with different mutations in the XPD (ERCC2)
(congenital ichthyosis with trichothiodystrophy). Eur J Pe- repair/transcription gene. Proc Natal Acad Sci 1997; 94:
diatr 1984; 141: 147–152 8658–8663
Itin PH, Sarasin A, Pittelkow MR. Trichothiodystrophy: update Vandenberghe K, Casteels I, Vandenbussche E, de Zegher F, de
on the sulfur-deficient brittle hair syndromes. J Am Acad Boeck K. Bilateral cataract and high myopia in a child with
Dermatol 2001; 44: 891–920 trichothiodystrophy: a case report. Bull Soc Belge Ophthal-
Jackson CE,Weiss L,Watson JHL.“Brittle”hair with short stature, mol 2001; 282: 15–18
intellectual impairment and decreased fertility: an autop- Vermeulen W, Bergmann E, Auriol J, Rademakers S, Frit P, Appel-
sied recessive syndrome in an Amish kindred. Pediatics doorn E, Hoeijmakers JHJ, Egly J-M. Sublimiting concentra-
1974; 54: 201–207 tion of TFIIH transcription/DNA repair factor causes TTD-A
Jaspers NGJ. Multiple involvement of nucleotide excision re- trichothiodystrophy disorder. Nat Genet 2000; 26: 307–313
pair enzymes: clinical manifestations of molecular intrica- Vermeulen W, Rademakers S, Jaspers NGJ, Appeldoorn E,
cies. Cytokines Mol Ther 1996; 2: 115–119 Raams A, Klein B, Kleijer WJ, hansen LK, Hoeijmakers JHJ. A
Jorizzo JL, Atherton DJ, Crounse RG,Wells RS. Ichthyosis, brittle temperature-sensitive disorder in basal transcription and
hair, impaired intelligence, decreased fertility and short DNA repair in humans. Nat Genet 2001; 27: 299–303
stature (IBIDS syndrome). Br J Dermatol 1982; 106: 705–710 Viprakasit V, Gibbons RJ, Broughton BC, Tolmie JL, Brown D,
King MD, Gummer CL, Stephenson JBP. Trichothiodystrophy- Lunt P, Winter RM, Marioni S, Stefanini M, Brueton L,
neurotrichocutaneous syndrome of Pollitt: a report of two Lehmann AR, Higgs DR. Mutations in the general transcrip-
unrelated cases. J Med Genet 1984; 21: 286–289 tion factor TFIIH result in b-thalassaemia in individuals with
Kousseff BG, Esterly NB. Trichothiodystrophy, IBIDS syndrome trichthiodystrophy. Hum Mol Genet 2001; 10: 2797–2802
or Tay syndrome? Birth Defects Orig Art Ser 1988; 24: Weeda G, Eveno E, Donker I, Vermeulen W, Chevallier-Lagente
169–181 O, Taïeb A, Stary A, Hoeijmakers JHJ, Mezzina M, Sarasin. A
Kousseff BG. Collodion baby, sign of Tay syndrome. Pediatrics mutation in the XPB/ERCC2 DNA repair transcription gene,
1991; 87: 571–574 associated with trichothiodystrophy. Am J Hum Genet
Lehmann AR. The xeroderma pigmentosum group D (XPD) 1997; 60: 320–329
gene: one gene, two functions, three cases. Gene Dev 2001;
15: 15–23
962 References and Further Reading

34 Pelizaeus-Merzbacher Disease Fanarraga ML, Griffiths IR, McCulloch MC, Barrie JA, Kennedy
PGE, Brophy PJ. Rumpshaker: an X-linked mutation causing
André M, Monin P, Moret C, Braun M, Picard L. Maladie de hypomyelination and glial cells between the optic nerve
Pelizaeus-Merzbacher. J Neuroradiol 1990; 17: 216–221 and spinal cord. Glia 1992; 5: 161–170
Apkarain P, Koetsveld-Baart JC, Barth PG. Visual evoked poten- Feldman JI, Kearns DB, Seid AB, Pransky SM, Jones MC.The oto-
tial characteristics and early diagnosis of Pelizaeus- laryngologic manifestations of Pelizaeus-Merzbacher dis-
Merzbacher disease. Arch Neurol 1993; 50: 981–985 ease. Arch Otolaryngol Head Neck Surg 1990; 116: 613–616
Battini R, Biancji MC, Boespflug-Tanguy O,Tosetti M, Bonanni P, Garbern JY, Cambi F, Lewis R, Shy M, Sima A, Kraft G, Vallat JM,
Canapicchi R, Cioni G. Unusual clinical and magnetic reso- Bosch EP, Hodes ME, Dlouhy S, Raskind W, Bird T, Macklin W,
nance imaging findings in a family with proteolipid protein Kamholz J. Peripheral neuropathy caused by proteolipid
gene mutation. Arch Neurol 2003; 60: 268–272 protein gene mutations. Ann NY Acad Sci 1999; 883:
Boltshauser E, Schinzel A, Wichmann W, Haller D, Valavanis A. 351–365
Pelizaeus-Merzbacher disease: identification of hetero- Garbern J, Cambi F, Shy M, Kamholz J.The molecular pathogen-
zygotes with magnetic resonance imaging? Hum Genet esis of Pelizaeus-Merzbacher disease. Arch Neurol 1999; 56:
1988; 80: 393–394 1210–1214
Bonavita S, Schiffmann R, Moore DF, Frei K, Choi B, Patronas N, Garbern J, Shy M, Krajewski K, Kamholz J, Hobson G, Cambi F.
Virta A, Boespflüg-Tanguy O, Tedeschi G. Evidence for neu- Evidence of neuroaxonal injury in patients with proteolipid
roaxonal injury in patients with proteolipid protein gene gene mutations. Neurology 2001; 57: 1938–1939
mutations. Neurology 2001; 56: 758–788 Garbern J,Yool DA, Moore GJ,Wilds IB, Faulk MW, Klugmann M,
Bond C, Si X, Crisp M,Wong P, Paulson GW, Boesel CP, Dlouhy SR, Nave K-A,Sistermans EA,van der Knaap MS,Bird TD,Shy ME,
Hodes ME. Family with Pelizaeus-Merzbacher disease / Kamholz JA, Griffiths IR. Patients lacking the major myelin
X-linked spastic paraplegia and a nonsense mutation in ex- protein, proteolipid protein 1, develop length-dependent
on 6 of the proteolipid protein gene.Am J Med Genet 1997; axonal degeneration in the absence of demyelination and
71: 357–360 inflammation. Brain 2002; 125: 551–561
Boulloche J, Aicardi J. Pelizaeus-Merzbacher disease: clinical Garg BP, Markand ON, DeMyer WE. Usefulness of BAER studies
and nosological study. J Child Neurol 1986; 1: 233–239 in the early diagnosis of Pelizaeus-Merzbacher disease.
Bourre JM, Jacque C, Nguyen-Legros J, Bornhofen JH, Araoz CA, Neurology 1983; 33: 955–956
Daudu O, Baumann NA. Pelizaeus-Merzbacher disease: bio- Gencic S, Abuelo D, Ambler M, Hudson LD. Pelizaeus-
chemical analysis of isolated myelin (electron-microscopy: Merzbacher disease: an X-linked neurologic disorder of
protein, lipid and unsubstituted fatty acids analysis). Eur myelin metabolism with a novel mutation in the gene en-
Neurol 1978; 17: 317–326 coding proteolipid protein. Am J Hum Genet 1989; 45:
Bridge PJ, MacLeod PM, Lillicrap DP. Carrier detection and pre- 435–442
natal diagnosis of Pelizaeus-Merzbacher disease using a Gow A, Lazzarini RA. A cellular mechanism governing the
combination of anonymous DNA polymorphisms and the severity of Pelizaeus-Merzbacher disease. Nat Genet 1996;
proteolipid protein (PLP) gene cDNA. Am J Med Genet 13: 422–428
1991; 38: 616–621 Griffiths I, Klugmann M, Anderson T, Yool D, Thomson C,
Cailloux F, Gauthier-Barichard F, Mimault C, Isabelle V, Courtois Schwab MH, Schneider A, Zimmermann F, McCulloch M,
V,Giraud G, Dastugue B,Boespflug-Tanguy O for Clinical Eu- Nadon N, Nave K-A. Axonal swellings and degeneration in
ropean Network on Brain Dysmyelinating Disease. Geno- mice lacking the major proteolipid of myelin. Science 1998;
type-phenotype correlation in inherited brain myelination 280: 1610–1613
defects due to proteolipid protein gene mutations. Eur J Griffiths I, Klugmann M, Anderson T,Thomson C,Vouyiouklis D,
Hum Genet 2000; 8: 837–845 Nave K-A. Current concepts of PLP and its role in the ner-
Cambi F,Tang X-M, Cordray P, Fain PR, Keppen LD, Barker DF. Re- vous system. Microsc Res Tech 1998; 41: 344–358
fined genetic mapping and proteolipid protein mutations Griffiths IR, Montague P, Dickinson P. The proteolipid protein
analysis in X-linked pure hereditary spastic paraplegia. gene. Neuropathol Appl Neurobiol 1995; 21: 85–96
Neurology 1996; 46: 1112–1117 Grinspan JB, Coulalaglou M, Beesley JS, Carpio DF, Scherer SS.
Cambi F,Tartaglino L, Lublin F, McCarren D. X-linked pure famil- Mutation-dependent apoptic cell death of oligodendro-
ial spastic parapresis. Characterization of a large kindred cytes in myelin-deficient rats. J Neurosci Res 1998; 54:
with magnetic resonance imaging studies. Arch Neurol 623–634
1995; 52: 665–669 Gutmann DH, Fischbeck KH, Kamholz J. Complicated heredi-
Caro PA, Marks HG. Magnetic resonance imaging and comput- tary spastic paraparesis with cerebral white matter lesions.
ed tomography in Pelizaeus-Merzbacher disease. Magn Re- Am J Med Genet 1990; 36: 251–257
son Im 1990; 8: 791–796 Hobson GM, Davis AP, Stowell NC, Kolodny EH, Sistermans EA,
Cuddon PA, Lipsitz D, Duncan ID. Myelin mosaicism and brain De Coo IFM, Funanage VL, Marks HG. Mutations in noncod-
plasticity in heterozygous females of a canine X-linked ing regions of the proteolipid protein gene in Pelizaeus-
trait. Ann Neurol 1998; 44: 771–779 Merzbacher disease. Neurology 2000; 55: 1089–1096
Ellis D, Malcom S. Proteolipid protein gene dosage effect in Hodes ME, DeMyer WE, Pratt VM, Edwards MK, Dlouhy SR. Girl
Pelizaeus-Merzbacher disease. Nat Genet 1994; 6: 333–334 with signs of Pelizaeus-Merzbacher disease heterozygous
Fanarraga ML, Griffiths IR, McCulloch MC, Barrie JA, Cattanach for a mutation in exon 2 of the proteolipid protein gene.
BM, Brophy PJ, Kennedy PGE. Rumpshaker: an X-linked mu- Am J Med Genet 1995; 55: 397–401
tation affecting CNS myelination.A study of the female het- Hodes ME, Blank CA, Pratt VM, Morales J, Napier J, Dlouhy SR.
erozygote. Neuropathol Appl Neurobiol 1991; 17: 323–334 Nonsense mutation in exon 3 of the proteollipid protein
gene (PLP) in a family with with an unusual form of
Pelizaeus-Merzbacher disease. Am J Med Genet 1997; 69:
121–125
References and Further Reading 963

Hodes ME, Zimmermann AW, Aydanian A, Naidu S, Miller NR, Merzbacher L. Eine eigenartige familiär-hereditäre Erkran-
Garcia Oller JL, Barker B, Aleck KA, Hurley TD, Dlouhy SR. Dif- kungsform (aplasia axialis extracorticalis congenita). Z Ges
ferent mutations in the same codon of the proteolipid pro- Neurol Psychiatrie 1910; 3: 1–138
tein gene, PLP, may help in correlating genotype with phe- Mimault C, Giraud G, Courtois V, Cailloux FN, Boire JY, Dastugue
notype in Pelizaeus-Merzbacher disease/X-linked spastic B, Boespflug-Tanguy O, The Clinical European Network on
paraplegia (PMD/SPG2).Am J Med Genet 1999; 82: 132–139 Brain Dysmyelination Disease. Proteolipoprotein gene
Hodes ME, Woodward K, Spinner NB, Emanuel BS, Enrico-Si- analysis in 82 patients with sporadic Pelizaeus-Merzbacher
mon A, Kamholz J, Stambolian D, Zackai EH, Pratt VM, disease: duplications, the major cause of the disease, origi-
Thomas IT, Crandall K, Dlouhy SR, Malcolm S. Additional nate more frequently in male germ cells, but point muta-
copies of the proteolipid protein gene causing Pelizaeus- tions do not. Am J Hum Genet 1999; 65: 360–369
Merzbacher disease arise by separate integration into the X Naidu S, Dlouhy SR, Geraghty MT, Hodes ME. A male child with
chromosome. Am J Hum Genet 2000; 67: 14–22 the rumpshaker mutation, X-linked spastic paraplegia/
Hudson LD. Pelizaeus-Merzbacher disease and spastic para- Pelizaeus-Merzbacher disease and lysinuria.J Inherit Metab
plegia type 2: two faces of myelin loss from mutations in Dis 1997; 20: 811–816
the same gene. J Child Neurol 2003; 18: 616–624 Nance MA,Boyadjiev S,Pratt VM,Taylor S,Hodes ME,Dlouhy SR.
Hudson LD, Puckett C, Berndt J, Chan J, Gencic S. Mutation of Adult-onset neurodegenerative disorder due to prote-
the proteolipid protein gene PLP in a human X chromo- olipid protein gene mutation in the mother of a man with
some-linked myelin disorder. Proc Natl Acad Sci 1989; 86: Pelizaeus-Merzbacher disease. Neurology 1996; 47: 1333–
8128–831 1335
Inoue K, Osaka H, Imaizumi K, Nezu A, Takanashi J-I, Arii J, Mu- Nezu A, Kimura S, Takeshita S, Osaka H, Kimura K, Inoue K. An
rayama K, Ono J, Kikawa Y, Mito T, Shaffer LG, Lupski JR. Pro- MRI and MRS study of Pelizaeus-Merzbacher disease. Pedi-
teolipid protein gene duplications causing Pelizaeus- atr Neurol 1998;18: 334–337
Merzbacher disease: molecular mechanism and phenotyp- Pelizaeus F. Ueber eine eigenartige familiäre Entwickelungs-
ic manifestations. Ann Neurol 1999; 45: 624–632 hemmung vornehmlich auf motorischem Gebiet. Arch
Inoue K,Tanaka H, Scaglia F, Araki A, Shaffer LG, Lupski JR. Com- Psychiatr Nervenkr 1899; 31: 100–104
pensating for central nervous system dysmyelination: fe- Plecko B, Stockler-Ipsiroglu S, Gruber S, Mlynarik V, Moser E,
males with a proteolipid protein gene duplication and sus- Simbrunner J, Ebner F, Bernert G, Harrer G, Gal A, Prayer D.
tained clinical improvement. Ann Neurol 2001; 50: 747–754 Degree of hypomyelination and magnetic resonance spec-
Journel H, Roussey M, Gandon Y, Allaire C, Carsin M, le Marec B. troscopy findings in patients with Pelizaeus Merzbacher
Magnetic resonance imaging in Pelizaeus-Merzbacher dis- phenotype. Neuropediatrics 2003; 34: 127–136
ease. Neuroradiology 1987; 29: 403–405 Readhead C, Schneider A, Griffiths I, Nave K-A. Premature arrest
Jung M, Sommer I, Schachner M, Nave K-A. Monoclonal anti- of myelin formation in transgenic mice with increased pro-
body 010 defines a conformationally sensitive cell-surface teolipid protein gene dosage. Neuron 1994; 12: 583–595
epitope of proteolipid protein (PLP): evidence that PLP mis- Renier WO, Gabreeels FJM, Hustinx TWJ, Jaspar HHJ, Geelen
folding underlies dysmyelination in mutant mice. J Neu- JAG, van Haelst UJG, Lommen EJP, ter Haar BGA. Connatal
rosci 1996; 16: 7920–7929 Pelizaeus-Merzbacher disease with congenital stridor in
Kaga M, Murakami T, Naitoh H, Nihei K. Studies on pediatric pa- two maternal cousins. Acta Neuropathol (Berl) 1981; 54:
tients with absent auditory brainstem response (ABR) later 11–17
components. Brain Dev 1990; 12: 380–384 Saugier-Veber P, Munnich A, Bonneau D, Rozet J-M, Le Merrer
Kagawa T, Ikenaka K, Inoue Y, Kuriyama S,Tsujii T, Nakao J, Naka- M, Gil R, Boespflug-Tanguy O. X-linked spastic paraplegia
jima K, Aruga J, Okano H, Mikoshiba K. Glial cell degenera- and Pelizaeus-Merzbacher disease are allelic disorders at
tion and hypomyelination caused by overexpression of the proteolipid protein locus. Nat Genet 1994; 6: 257–262
myelin proteolipid protein gene. Neuron 1994; 13: 427–442 Scheffer IE, Baraitser M,Wilson J, Harding B, Kendall B, Brett EM.
Karthigasan J, Evans EL,Vouyiouklis DA, Inouye H, Borenshteyn Pelizaeus-Merzbacher disease: classical or connatal? Neu-
N, Ramamurthy GV, Kirschner DA. Effects if Rumpsaker mu- ropediatrics 1991; 22: 71–78
tation on CNS myelin composition and structure. J Neu- Schneck L, Adachi M, Volk BW. Congenital failure of myeliniza-
rochem 1996; 66: 338–345 tion: Pelizaeus-Merzbacher disease? Neurology 1971; 21:
Kaye EM, Doll RF, Natowicz MR, Smith FI. Pelizaeus-Merzbacher 817–824
disease presenting as spinal muscular atrophy: clinical and Shy ME, Hobson G, Jain M, Boespflug-Tanguy O, Garbern J,
molecular studies. Ann Neurol 1994; 36: 916–919 Sperle K, Li W, Gow A, Rodrihuez D, Bertini E, Mancias P, Kra-
Kobayashi H, Hoffman EP. The rumpshaker mutation in spastic jewski K, Lewis R, Kamholz J. Schwann cell expression of
paraplegia. Nat Genet 1994; 7: 351–352 PLP1 but not DM 20 is necessary to prevent neuropathy.
Koeppen AH, Robitaille Y.Pelizaeus-Merzbacher disaese.J Neu- Ann Neurol 2003; 53: 354–365
ropathol Exp Neurol 2002; 61: 747–759 Silverstein AM, Hirsh DK, Trobe JD, Gebarski SS. MR imaging of
Koeppen AH, Ronca NA, Greenfield EA, Hans MB. Defective the brain in five members of a family with Pelizaeus-
biosynthesis of proteolipid protein in Pelizaeus-Merzbach- Merzbacher disease. AJNR Am J Neuroradiol 1990; 11:
er disease. Ann Neurol 1987; 21: 159–170 495–499
Komaki H Sasaki M, Yamamoto T, Iai M, Takanshima S. Con- Simons M, Krämer E-M, Macchi P, Rathke-Hartlieb S, Trotter J,
natal Pelizaeus-Merzbacher disease associated with the Nave K-A, Schulz JB. Overexpression of the myelin prote-
Jimpymsd mice mutation. Pediatr Neurol 1999; 20: 309–311 olipid protein leads to accumulation of cholesterol and
Learish RD, Brüstle O, Zhang S-C, Duncan ID. Intraventricular proteolipid protein in endosomes/lysosomes: implications
transplantation of oligodendrocyte progenitors into a fetal for Pelizaeus-Merzbacher disease. J Cell Biol 2002; 157:
myelin mutant results in widespread formation of myelin. 327–336
Ann Neurol 1999; 46: 716–722
964 References and Further Reading

Sistermans EA, De Wijs IJ, De Coo RFM, Smit LME, Menko FH, Woodward K, Kirtland K, Dlouhy S, Raskind W, Bird T, Malcolm S,
Van Oost BA. A (G-to-A) mutation in the intiation condon of Abeliovich D. X Inactive phenotype in carriers of Pelizaeus-
the proteolipid protein gene causing a reletively mild form Merzbacher disease: skewed in carriers of a duplication and
of Pelizaeus-Merzbacher disease in a Dutch family. Hum random in carriers of point mutations. Eur J Hum Genet
Genet 1996; 97: 337–339 2000; 8: 449–454
Sistermans EA, De Coo, RFM, De Wijs IJ, Van Oost BA. Duplica- Woodward K, Malcolm S. CNS myelination and PLP gene
tion of the proteolipid protein gene is the major cause of dosage. Pharmacogenomics 2001; 2: 263–272
Pelizaeus-Merzbacher disease. Neurology 1998; 50: 1749– Yool DA,Edgar JM,Montague P,Malcolm S.The proteolipid pro-
1754 tein gene and myelin disorders in man and animal models.
Sivakumar K, Sambuughin N, Selenge B, Nagle JW, Baasanjav D, Hum Mol Genet 2000; 9: 987–992
Hudson LD, Goldfarb LG. Novel exon 3B proteolipid protein Yousem DM, Gutmann DH, Milestone BN, Lenkinski RE. Inte-
gene mutation causing late-onset spastic paraplegia type grated MR imaging and proton nuclear magnetic reso-
2 with variable penetrance in female family members. Ann nance spectroscopy in a family with an X-linked spastic
Neurol 1999; 45: 680–683 paraparesis. AJNR Am J Neuroradiol 1991; 12: 785–789
Southwood CM, Garbern J, Jiang W, Gow A. The unfolded pro- Zeman W, Demyer W, Falls HF. Pelizaeus-Merzbacher disease.
tein response modulates disease severity in Pelizaeus- J Neuropathol Exp Neurol 1964; 23: 334–354
Merzbacher disease. Neuron 2002; 36: 585–596
Spalice A, Popolizio T, Parisi P, Scarabino T, Iannetti P. Proton MR
spectroscopy in connatal Pelizaeus-Merzbacher disease.
Pediatr Radiol 2000; 30: 171–175
35 18q– Syndrome
Spörkel O, Uschkureit T, Büssow H, Stoffel W. Oligodendrocytes
expressing exclusively the DM 20 isoform of the prote- Davis Ghidoni P, Hale DE, Cody JD, Gay CT, Thompson NM,
olipid protein gene: myelination and development. Glia McClure EB, Danny MM, Leach RJ, Kaye CI. Growth hormone
2002; 37: 19–30 deficiency associated in the 18q deletion syndrome. Am J
Takanashi J-I, Sugita K, Osaka H, Ishii M, Niimi H. Proton MR Med Genet 1997; 69: 7–12
spectroscopy in Pelizaeus-Merzbacher disease. AJNR Am J Felding I,Kristoffersson U,Sjöström H,Noren O.Contribution to
Neuroradiol 1997; 18: 533–535 the 18q– syndrome. A patient with del (18) (q22.3qter). Clin
Takanashi J-I, Sugita K,Tanabe Y, Nagasawa K, Inoue K, Osaka H, Genet 1987; 31: 206–210
Kohno Y. MR-Revealed myelination in the cerebral corti- Fryns JP, Logghe N, van Eygen M, van den Berghe H. 18q– Syn-
cospinal tract as a marker for Pelizaeus-Merzbacher’s dis- drome in mother and daughter. Eur J Pediatr 1979; 130:
ease with proteolipid protein gene duplication. AJNR Am J 189–192
Neuroradiol 1999; 20: 1822–1828 Gabrielli O, Coppa GV, Carloni I, Salvolini U. 18q– Syndrome and
Takanashi J, Inoue K, Tomita M, Kurihara A, Morita F, Ikehira H, white matter alterations. AJNR Am J Neuroradiol 1998; 39:
Tandana S,Yoshitome E, Kohno Y. Brain N-acetylaspartate is 398–399
elevated in Pelizaeus-Merzbacher disease with PLP1 dupli- Gay CT, Hardies LJ, Rauch RA, Lancaster JL, Plaetke R, DuPont
cation. Neurology 2002; 58: 237–241 BR, Cody JD, Cornell JE, Herndon RC, Ghidoni PD, Schiff JM,
Thomson CE,Montague P,Jung M,Nave K-A,Griffiths IR.Pheno- Kaye CI, Leach RJ, Fox PT. Magnetic Resonance Imaging
typic severity of murine PLp mutants reflects in vivo and in demonstrates incomplete myelination in 18q– syndrome:
vitro variations in transport of PLP isoproteins. Glia 1997; evidence for myelin basic protein haploinsufficiency. Am J
20: 322–332 Med Genet 1997; 74: 422–431
Ulrich J, Herschkowitz N. Seitelberger’s connatal form of Jayarajan V, Swan IRC, Patton MA. Hearing impairment in 18q
Pelizaeus-Merzbacher disease. Acta Neuropathol (Berl) deletion syndrome. J Laryngol Otol 2000; 114: 963–966
1977; 40: 129–136 Kamholz J, Spielman R, Gogolin K, Modi W, O’Brien S, Lazzarini
van der Knaap MS, Valk J. The reflection of histology in MR R. The human myelin-basic-protein gene: chromosomal lo-
imaging of Pelizaeus-Merzbacher disease. AJNR Am J Neu- calization and RFLP analysis. Am J Hum Genet 1987; 40:
roradiol 1989; 10: 99–103 365–373
Vaurs-Barriere C, Wong K, Weibel TD, Abu-Asab M, Weiss MD, Keppler-Noreuil KM, Carroll AJ, Finley SC, Descartes M, Cody JD,
Kaneski CR, Mixon TH, Bonavita S, Creveaux I, Heiss JD, DuPont BR, Gay CT, Leach RJ. Chromosome 18q paracentric
Tsokos M, Goldin E, Quarles RH, Boespflug-Tanguy O, Schiff- inversion in a family with mental retardation and hearing
mann R. Insertion of mutant proteolipid protein results in loss. Am J Med Genet 1998; 76: 372–378
missorting of myelin proteins. Ann Neurol 2003; 54: Kline AD,White ME,Wapner R, Rojas K, Biesecker LG, Kamholz J,
769–780 Zackai EH, Muenke M, Scott Jr CI, Overhauser J. Molecular
Watanabe I, McCaman R, Dyken P, Zeman W. Absence of cere- analysis of the 18q– syndrome – and correlation with phe-
bral myelin sheaths in a case of presumed Pelizaeus- notype. Am J Hum Genet 1993; 52: 895–906
Merzbacher disease. J Neuropathol Exp Neurol 1969; 28: Lemke G. Unwrapping the genes of myelin. Neuron 1988; 1:
243–256 535–543
Watanabe I, Patel V, Goebel HH, Siakotos AN, Zeman W, Demy- Linnankivi TT, Autti TH, Pikho SH, Somer MS, Tienari PJ,
er W, Schroder Dyer J. Early lesion of Pelizaeus-Merzbacher Wirtavuori KO,Valanne LK. 18q– syndrome: brain MRI shows
disease: electron microscopic and biochemical study. poor differentiation of gray and white matter on T2-
J Neuropathol Exp Neurol 1973; 32: 313–333 weighted images. J Magn Reson Imag 2003; 18: 414–419
Witter B, Debuch H, Klein H. Lipid investigation of central Loevner LA, Shapiro RM, Grossmann RI, Overhauser J, Kamholz
and peripheral nervous system in connatal Pelizaeus- J. White matter changes associated with deletions of the
Merzbacher’s disease. J Neurochem 1980; 34: 957–962 long arm of chromosome 18 (18q– syndrome): a dysmyeli-
nating disorder? AJNR Am J Neuroradiol 1996; 14: 1843–
1848
References and Further Reading 965

Mahr RM, Moberg PJ, Overhauser J, Strathdee G, Kamholz J, Lo- Breysem L, Smet M-H, Johannik K, van Hecke P, François B,
evner LA, Campbell H, Zackai EH, Reber ME, Mozley DP, Wilms B, Bosmans H. Marchal G, Jaeken J, Demaerel P. Brain
Brown L, Turetsky BI, Shapiro RM. Neuropsychiatry of 18q– MR imaging in dietary treatment phenylketonuria. Eur Ra-
syndrome. Am J Med Genet 1996; 67: 172–178 diol 1994: 4: 329–331
Miller G, Mowrey PN, Hopper KD, Frankel CA, Ladda RL. Neuro- Brismar J, Aqeel A, Gascon G, Ozand P. Malignant hyperpheny-
logic manifestations in 18q– syndrome. Am J Med Genet lalaninemia: CT and MR of the brain. AJNR Am J Neuroradi-
1990; 37: 128–132 ol 1990; 11: 135–138
Ono J, Harada K,Yamamoto T, Onoe S, Okada S. Delayed myeli- Burgard P. Development of intelligence in early treated
nation in a patient with 18q– syndrome. Pediatr Neurol phenylketonuria. Eur J Pediatr 2000; 159: S74-S79
1994; 11: 64–67 Burri R, Steffen CH, Stieger S, Brodbeck U, Colombo JP, Her-
Rodichok L, Miller G. A study of evoked potentials in the 18q– schkowitz N. Reduced myelinogenesis and recovery in hy-
syndrome which includes the absence of the gene locus for perphenylalaninemic rats. Mol Chem Neuropathol 1990;
myelin basic protein. Neuropediatrics 1992; 23: 218–220 13: 57–69
Stankiewicz P, Brozek I, Hélias-Rodzewicz Z, Wierzba J, Pilch J, Cerone R, Schiaffino MC, Di Stefano S, Veneselli E. Phenylke-
Bocian E, Balcerska A, Woÿniak A, Kardaœ I, Wirth J, Mazur- tonuria: diet for life or not? Acta Paediatr 1999; 88; 664–666
czak T, Limon J. Clinical and molecular–cytogenetic studies Chien Y-H, Peng S-F, Wang T-R, Hwu W-L. Cranial MR spec-
in seven patients with ring chromosome 18. Am J Med troscopy of tetrahydrobiopterin deficiency. AJNR Am J
Genet 2001; 101: 226–239 Neuroradiol 2002; 23: 1055–1058
Strathdee G, Zackai EH, Shapiro R, Kamholz J, Overhauser J. Cleary MA, Walter JH. Assessment of adult phenylketonuria.
Analysis of clinical variation seen in patients with 18q ter- Ann Clin Biochem 2001; 38: 450–458
minal deletations. Am J Med Genet 1995; 59: 476–483 Cleary MA, Walter JH, Wraith JE, Jenkins JPR, Alani SM, Tyler K,
Strathdee G, Sutherland R, Jonsson JJ, Sataloff R, Kohonen-Cor- Whittle D. Magnetic resonance imaging of the brain in
ish M, Grady D, Overhauser J. Molecular characterization of phenylketonuria. Lancet 1994; 334; 87–90
patients with 18q23 deletions. Am J Hum Genet 1997; 60: Cleary MA, Walter JH, Wraith JE, Jenkins JPR. Magnetic reso-
860–868 nance imaging in phenylketonuria: reversal of cerebral
Wang Z, Cody JD, Leach RJ, O’Connell P. Gene expression pat- white matter change. J Pediatr 1995; 127: 251–255
terns in cell lines from patients with 18q- syndrome. Hum Costa LG, Guizzetti M, Burry M, Oberdoerster J. Developmental
Genet 1999; 104: 347–475 neurotoxicity: do similar phenotypes indicate a common
Wertelecki W, Gerald PS. Clinical and chromosomal studies of mode of action? A comparison of fetal alcohol syndrome,
the 18q– syndrome. J Pediatr 1971; 78: 44–52 toluene embryopathy and maternal phenylketonuria.Toxi-
Wilson MG, Towner JW, Forsman I, Siris E. Syndromes associat- col Lett 2002; 127: 197–205
ed with deletion of the long arm of chromosome 18 [del Dezortová M, Hájek M, Tintra, Hejcmanová L, Syková E. MR in
(18q)]. Am J Med Genet 1979; 3: 155–174 phenylketonuria-related brain lesions.Acta Radiol 2001; 42:
459–466
Dhondt JL, Farriaux JP, Boudha A, Largillière C, Ringel J, Roger
MM, Leeming RJ. Neonatal hyperphenylalaninemia pre-
36 Phenylketonuria sumably caused by guanosine triphosphate-cyclohydro-
lase deficiency. J Pediatr 1985; 106: 954–956
Alvord EC, Stevenson LD, Vogel FS, Engle RL. Neuropathologi- Erlandsen H, Stevens RC. The structural basis of phenylke-
cal findings in phenyl-pyruvic oligophrenia (phenyl-ke- tonuria. Mol Genet Metab 1999; 68: 103–125
tonuria). J Neuropathol Exp Neurol 1950; 9: 298–310 Erlandsen H, Stevens RC. A structural hypothesis for BH4 re-
Anderson PJ, Wood SJ, Francis D, Coleman L, Warwick L, sponsiveness in patients with mild forms of hyperpheny-
Casanelia S, Anderson VA, Boneh A. Neuropsychological lalaninaemia and phenylketonuria. J Inherit Metab Dis
functioning in children with early-treated phenylketonuria: 2001; 24: 213–230
impact of white matter abnormalities. Dev Med Child Neu- Fisch RO, Burke B, Bass J, Ferrara TB, Mastri A. Maternal
rol 2004; 46: 230–238 phenylketonuria – chronology of the detrimental effects
Arnold GL,Vladutiu CJ, Kirby RS, Blakely EM, DeLuca JM. Protein on embryogenesis and fetal development: pathological re-
insufficiency and linear growth restriction in phenylke- port, survey, clinical application. Pediatr Pathol 1986; 5:
tonuria. J Pediatr 2002; 141: 243–246 449–461
Battistini S, de Stefano N, Parlanti S, Federico A. Unexpected Fisch RO, Chang P-N, Weisberg S, Guldberg P, Güttler F, Tsai MY.
white matter changes in an early treated PKU case and im- Phenylketonuric patients decades after diet. J Inherit
provement after dietary treatment. Funct Neurol 1991; 6: Metab Dis 1995; 18: 347–353
177–180 Gerstl B, Malamud N, Eng LF, Hayman RB. Lipid alterations in
Bick U, Fahrendorf G, Ludolph AC, Vassallo P, Weglage J, Ullrich human brains in phenylketonuria. Neurology 1967; 17:
K. Disturbed myelination in patients with treated hyper- 51–57
phenylalaninaemia: evaluation with magnetic resonance Giovannini M, Biasucci G, Brioschi M, Ghiglioni D, Riva E. Cofac-
imaging. Eur J Pediatr 1991; 150: 185–189 tor defects and PKU: diagnosis and treatment. Int Pediatr
Blau N, Branes I, Dhondt JL. International database of tetrahy- 1991; 6: 26–31
drobiopterin deficiencies.J Inherit Metab Dis 1996; 19: 8–14 Gudinchet F, Maeder P, Meuli RA, Deonna T, Mathieu JM. Cranial
Bonafé L, Blau N, Burlina AP, Romstad A, Gütter F, Burlina AB. CT and MRI in malignant phenylketonuria. Pediatr Radiol
Treatable neurotransmitter deficiency in mild phenylke- 1992; 22: 223–224
tonuria. Neurology 2001; 57: 908–911 Güttler F, Guldberg P. Mutation analysis anticipates dietary re-
Brenton DP, Pietz J. Adult care in phenylketonuria and hyper- quirements in phenylketonuria. Eur J Pediatr 2000; 159;
phenylalaninaemia: the relevance of neurological abnor- S150-S153
malities. Eur J Pediatr 2000; 159: S114-S120
966 References and Further Reading

Huijbreghts SCJ, de Sonneville LMJ, Licht R, van Spronsen FJ, Möller HE, Weglage J, Wiedermann D, Ullrich K. Blood-brain
Sergeant JA. Short-term dietary erventions in children and barrier phenylalanine transport and individual vulnerabili-
adolescents with treated phenylketonuria: effects on neu- ty in phenylketonuria. J Cereb Blood Flow Metab 1998; 18:
ropsychological outcome of a well-controlled population. J 1184–1191
Inherit Metab Dis 2002; 25: 419–430 Möller HE, Ullrich K, Weglage J. In vivo proton magnetic reso-
Huttenlocher PR.The neuropathology of phenylketonuria: hu- nance spectroscopy in phenylketonuria. Eur J Pediatr 2000;
man and animal studies. Eur J Pediatr 2000: 159; S102-S106 159: S121-S125
Jones SJ, Turano G, Kriss A, Shawkat F, Kendall B, Thompson AJ. Naylor EW, Ennis D, Davidson GF, Wong LTK, Applegarth DA,
Visual evoked potentials in phenylketonuria: association Niederwieser A. Guanosine triphosphate cyclohydrolase I
with brain MRI, dietary state, and IQ. J Neurol Neurosurg deficiency: early diagnosis by routine urine pteridine
Psychiatry 1995; 59: 260–265 screening. Pediatrics 1987; 79: 374–378
Knudden GM,Hasselbalch S,Toft PB,Christensen E,Paulson OB, Niederwieser A,Blau N,Wang M,Joller P,Atares M,Cardesa-Gar-
Lou H. Blood–brain barrier transport of amino acids in cia J. GTP cyclohydrolase I deficiency, a new enzyme defect
healthy controls and in patients with phenylketonuria. J In- causing hyperphenylalaninemia with neopterin, biopterin,
herit Metab Dis 1995; 18: 653–664 dopamine, and serotonin deficiencies and muscular hypo-
Koch R, Hanley W, Levy H, Matalon R, Rouse B, Trefz F, Guttler F, tonia. Eur J Pediatr 1984; 141: 208–214
Azen C, Friedman E, Platt L, de la Cruz F. maternal phenylke- Okano Y, Eisensmith RC, Güttler F, Lichter-Konecki U, Konecki
tonuria: an international study. Mol Genet Metab 2000; 71: DS, Trefz FK, Dasovich M, Wang T, Henriksen K, Lou H, Woo
233–239 SLC. Molecular basis of phenotypic heterogeneity in
Koch R, Burton B, Hoganson G, Peterson R, Rhead W, Rouse B, phenylketonuria. N Engl J Med 1991; 324: 1232–1238
Scott R, Wolff J, Stern AM, Guttler F, Nelson M, de la Cruz F, Pearsen KD, Gean-Marton AD, Levy HL, Davis KR. Phenylke-
Coldwell J, Erbe R, Geraghty MT, Shear C, Thomas J, Azen C. tonuria: MR imaging of the brain with clinical correlation.
Phenylketonuria in adulthood: a collaborative study. J In- Radiology 1990; 177: 437–440
herit Metab Dis 2002; 25: 333–346 Philips MD, McGraw P, Lowe MJ, Mathews VP, Hainline BE. Diffu-
Lässker U, Zschocke J, Blau N, Santer R.Tetrahydrobiopterin re- sion-weighted imaging of white matter abnormalities in
sponsiveness in phenylketonuria. Two new cases and a re- patients with phenylketonuria. AJNR Am J Neuroradiol
view of molecular genetic findings. J Inherit Metab Dis 2001; 22: 1583–1586
2002; 25: 65–70 Pietz J, Schmidt E, Matthis P, Kobialka B, Kutscha A, de Son-
Leuzzi V, Gualdi GF, Fabbrizi F, Trasimeni G, DiBiasi C, Antonozzi neville L. EEGs in phenylketonuria. I. Follow up to adult-
I.Neuroradiological (MRI) abnormalities in phenylketonuric hood. II. Short-term diet-related changes in EEGs and cog-
subjects: clinical and biochemical correlations. Neuropedi- nitive function. Dev Med Child Neurol 1993; 35: 54–64
atrics 1993; 24: 302–306 Pietz J, Kreis R, Boesch C, Penzien J, Rating D, Herschkowitz N.
Leuzzi V, Traisimeni G, Gualdi GF, Antonozzi I. Biochemical, clin- The dynamics of brain concentrations of phenylalanine
ical and neuroradiological (MRI) correlations in late-detect- and its clinical significance in patients with phenylke-
ed PKU patients. J Inherit Metab Dis 1995; 18: 624–634 tonuria determined by in vivo 1H magnetic resonance
Levy HL, Lobbregt D, Sansaricq C, Snyderman SE. Comparison spectroscopy. Pediatr Res 1995; 38: 657–663
of phenylketonuric and nonphenylketonuric sibs from un- Pietz J, Kreis R, Schmidt H, Meyding-Lamadé UK, Rupp A,
treated pregnancies in a mother with phenylketonuria. Am Boesch C. Phenylketonuria: findings at MR imaging and lo-
J Med Genet 1992; 44: 439–442 calized in vivo H-1 MR spectroscopy of the brain in patients
Longhi R,Valsasina R, Buttè C, Paccanelli S, Riva E, Giovannini M. with early treatment. Radiology 1996; 201: 413–420
Cranial computerized tomography in dihydropterine re- Pietz J,Dunckelmann R,Rupp A,Rating D,Meinck H-M.Schmidt
ductase deficiency. J Inherit Metab Dis 1985; 8: 109–112 H, Bremer HJ. Neurological outcome in adult patients with
Lou HC, Toft PB, Andressen J, Mikkelsen I, Olsen B, Güttler F, early-treated phenylketonuria. Eur J Pediatr 1998; 157:
Wieslander S, Henriksen O. An occipito-temporal syndrome 824–830
in adolescents with optimally controlled hyperphenylalan- Pietz J, Kreis R, Rupp A, Myatepek E, Rating D, Boesch C, Bremer
inaemia. J Inherit Metab Dis 1992; 15: 687–695 HJ. Large neutral amino acids block phenylalanine trans-
Magee AC, Ryan K, Moore A, Trimble ER. Follow up of fetal out- port into brain tissue in patients with phenylketonuria.
come in cases of maternal phenylketonuria in Northern Ire- J Clin Invest 1999; 103: 1169–1178
land. Arch Dis Child 2002; 87: F141-F143 Pietz J, Rupp A, Burgard P, Boesch C, Kreis R. No evidence for
Malamud N. Neuropathology of phenylketonuria. J Neuro- individual blood-brain barrier phenylalanine transport to
pathol Exp Neurol 1966; 25: 254–268 influence clinical outcome in typical phenylketonuria pa-
McCombe PA, McLaughlin DB, Chalk JB, Brown NN, McGill JJ, tients. Ann Neurol 2002; 52: 382–385
Pender MP. Spasticity and white matter abnormalities in Poser CM, van Bogaert L. Neuropathologic observations in
adult phenylketonuria. J Neurol Neurosurg Psychiatry phenylketonuria. Brain 1959; 82: 1–9
1992; 55: 359–361 Ris MD, Weber AM, Hunt MM, Berry HK, Williams SE, Leslie N.
Menkes JH. The pathogenesis of mental retardation in Adult psychosocial outcome in early-treated phenylke-
phenylketonuria and other inborn errors of amino acid me- tonuria. J Inherit Metab Dis 1997; 20: 499–508
tabolism. Pediatrics 1967; 39: 297–308 Rouse B, Azen C, Koch R, Matalon R, Hanley W, de la Cruz F,
Milandi N, Larnaout A, Dhondt J-L, Vincent M-F, Kaabachi N, Trefz F, Friedman E, Shifrin H. Maternal phenylketonuria col-
Hentati F. Dihydropteridine reductase deficiency in a large laborative study (MPKUCS) offspring: facial anomalities,
consanguineous Tunisian family: clinical, biochemical, and malformations, and early neurological sequelae. Am J Med
neuropathologic findings. J Child Neurol 1998; 13: 475–480 Genet 1997; 69: 89–95
Moats RA, Koch R, Moseley K, Guldberg P, Guttler F, Boles RG, Schmidt H, Ullrich K, Korinthenberg R, Peters PE.Basal ganglion
Nelson Jr, MD. Brain phenylalanine concentration of adults calcification in hyperphenylalaninemia due to deficiency
with phenylketonuria. J Inherit Metab Dis 2000, 23: 7–14 of dihydropteridine reductase. Pediatr Radiol 1988; 19:
54–56
References and Further Reading 967

Shaw DWW, Weinberger E, Maravilla KR. Cranial MR in phenyl- Weglage J, Wiedermann D, Denecke J, Feldmann R, Koch H-G,
ketonuria. J Comput Assist Tomogr 1990; 14: 458–460 Ullrich K, Harms E, Möller HE. Individual blood-brain barrier
Shaw DWW, Maravilla KR, Weinberger E, Garretson J, Trahms phenylalanine transport determines clinical outcome in
CM, Scott CR. MR imaging in phenylketonuria. AJNR Am J phenylketonuria. Ann Neurol 2001; 50: 463–467
Neuroradiol 1991; 12: 403–406 Weglage J, Wiedermann D, Denecke J, Fledmann R, Koch H-G,
Smith I. Review of neonatal screening programme for Ullrich K, Möller HE. Individual blood–brain barrier phenyl-
phenylketonuria. BMJ 1991; 303: 333–335 alanine transport in siblings with classical phenylketonuria.
Smith I. Treatment of phenylalanine hydroxylase deficiency. J Inherit Metab Dis 2002; 25: 431–436
Acta Paediatr Suppl 1994; 407: 60–65 Woody RC, Brewster MA, Glasier C. Progressive intracranial cal-
Smith I, Knowles J. Behavior in early treated phenylketonuria: a cification in dihydropteridine reductase deficiency prior to
systematic review. Eur J Pediatr 2000; 159; S89-S93 folinic acid therapy. Neurology 1989; 39: 683–675
Sugita R,Takahashi S, Ishii K, Matsumoto K, Ishibashi T, Sakamo-
to K, Narisawa K. Brain CT and MR findings in hyperphenyl-
alaninemia due to dihydropteridine reductase deficiency
(variant of phenylketonuria). J Comput Assist Tomogr 1990;
37 Glutaric Aciduria Type I
14: 699–703
Surtees R, Blau N. The neurochemistry of phenylketonuria. Eur Al-Essa M, Bakheet S, Patay Z, Al-Watban J, Powe J, Joshi S,
J Pediatr 2000; 159: S109-S113 Ozand PT. Fluoro-2-deoxyglucose (18FDG) PET scan of the
Takashima S, Chan F, Becker LE. Cortical dysgenesis in a variant brain in glutaric aciduria type I: clinical and MRI correla-
of phenylketonuria (dihydropteridine reductase deficien- tions. Brain Dev 1998; 20: 295–301
cy). Pediatr Pathol 1991: 11: 771–779 Alkan A, Baysal T,Yakinici C Siğerici A, Kutlu R. Glutaric aciduria
Thompson AJ, Smith I, Brenton D,Youl BD, Rylance G, Davidson type I diagnosed after poliovirus immunization: magnetic
DC, Kendall B, Lees AJ. Neurological deterioration in young resonance findings. Pediatr Neurol 2002; 26: 405–407
adults with phenylketonuria. Lancet 1990; 336: 602–605 Altman NR, Rovira MJ, Bauer M. Glutaric aciduria type 1: MR
Thompson AJ,Tillotson S, Smith I, Kendall B, Moore SG, Brenton findings in two cases. AJNR Am J Neuroradiol 1991; 12:
DP. Brain MRI changes in phenylketonuria. Brain 1993; 116: 966–968
811–821 Amir N, Peleg OE, Shalev RS, Christensen E. Glutaric aciduria
Toft PB, Lou HC, Krägeloh-Mann I, Anddresen J, Güttler F, Guld- type I: clinical heterogeneity and neuroradiologic features.
berg P, Henriksen O. Brain magnetic resonance imaging in Neurology 1987; 37: 1654–1657
children with optimally controlled hyperphenylalani- Amir N, Elpeleg ON, Shalev RS, Christensen E. Glutaric aciduria
naemia. J Inherit Metab Dis 1994; 17: 575–583 type I: enzymatic and neuroradiologic investigations of
Trefz FK, Cipcic-Schmidt S, Koch R. Final intelligence in late two kindreds. J Pediatr 1989; 114: 983–989
treated patients with phenylketonuria. Eur J Pediatr 2000; Bähr O, Mader I, Zschoke J, Dichgans J, Schultz JB. Adult onset
159: S145-S148 glutaric aciduria type I presenting with a leukoen-
Ullrich K, Weglage J, Schuierer G, Fünders B, Pietsch M. Koch cephalopathy. Neurology 2002; 59: 1802–1804
HG. Hahn-Ullrich H. Cranial MRI in PKU: evaluation of a criti- Bariæ I, Zschocke J, Christensen E, Duran M, Goodman SI,
cal treshold for blood phenylalanine.Neuropediatrics 1994; Leonard JV, Muller E, Morton DH, Superti-Furga A, Hoff-
25: 278–279 mann GF. Diagnosis and management of glutaric aciduria
Ullrich K, Möller H, Weglage J, Schuierer G, Bick U, Ludolph A. type I. J Inherit Metab Dis 1998; 21: 326–340
Hahn-Ullrich H, Fünders B, Koch D-G. White matter abnor- Baric I, Wagner L, Feyh P, Liesert M, Buckel W, Hoffmann GF.
malities in phenylketonuria: results of magnetic resonance Sensitivity and specificity of free and total glutaric acid and
measurements. Acta Paediatr Suppl 1994; 407: 78–82 3-hydroxyglutaric acid measurements by stable-isotope
Van Spronsen FJ, Smit PGA, Koch R. Phenylketonuria: tyrosine dilution assays for the diagnosis of glutaric aciduria type I.
beyond the phenylalanine-restricted diet. J Inherit Metab J Inherit Metab Dis 1999; 22: 867–882
Dis 2001; 24: 1–4 Bennett MJ, Marlow N, Pollitt RJ, Wales JKH. Glutaric aciduria
Waisbren SE, Zaff J, Personality disorder in young woman with type I: biochemical investigations and postmortem find-
treated phenylketonuria. J Inherit Metab Dis 1994; 17: ings. Eur J Pediatr 1986; 145: 403–405
584–592 Bennett MJ, Pollitt RJ, Goodman SI, Hale DE,Vamecq J. Atypical
Walter JH, Tyfield LA, Holton JB, Johnson C. Biochemical con- riboflavin-responsive glutaric aciduria, and deficient per-
trol, genetic analysis and magnetic resonance imaging in oxisomal glutaryl-CoA oxidase activity: a new peroxisomal
patients with phenylketonuria. Eur J Pediatr 1993; 152: disorder. J Inherit Metab Dis 1991; 14: 165–173
822–827 Bergman I, Finegold D, Gärtner JC, Zitelli BJ, Claassen D, Scara-
Walter JH, White F, Wraith JE, Jenkins JP, Wilson BPM. Complete no J, Roe CR, Stanley C, Goodman SI. Acute profound dysto-
reversal of moderate/severe brain MRI abnormalities in a nia in infants with glutaric acidemia. Pediatrics 1989; 83:
patient with classical phenylketonuria. J Inherit Metab Dis 228–234
1997; 20: 367–369 Bismar J, Ozand PT. CT and MR of the brain in glutaric aciduria
Weglage J, Bick U, Schuierer G, Pietsch M, Sprinz A, Zass R, Ull- type I: a review of 59 published cases and a report of 5 new
rich K. Progression of cerebral white matter abnormalities patients. AJNR Am J Neuroradiol 1995; 16: 675–683
in early treated patients with phenylketonuria during ado- Busquets C, Coll MR, Christensen E, Campistol J, Clusellas N,
lescence. Neuropediatrics 1997; 28: 239–240 Vilaseca MA, Ribes A. Feasibility of molecular prenatal diag-
Weglage J, Pietsch M, Denecke J, Sprintz A, Fledmann R, Gren- nosis of glutaric aciduria type I in chorionic villi. J Inherit
zeback M, Ullrich K. Regression of neuropsychological Metab Dis 1998; 21: 243–246
deficits in early-treated phenylketonurics during adoles- Busquets C, Coll MJ, Merinero B, Ugarte M, Ruiz MA, Martinez
cence. J Inherit Metab Dis 1999; 22: 693–705 Bermejo A, Ribes A. Prenatal molecular diagnosis of glutar-
ic aciduria type I by direct mutation analysis. Prenat Diagn
2000; 20: 761–764
968 References and Further Reading

Busquets C, Soriano M, Taveres de Almeida I, Garavaglia B, Ri- Hoffmann GF, Althanassopoulos S, Burlina AB, Duran M, de
moldi M,Rivera I,Uziel G,Cabral A,Coll MJ,Ribes A.Mutation Klerk BC, Lehnert W, Leonard JV, Moravari AA, Müller E,
analysis of the GCDH gene in Italian and Portuguese pa- Muntau AC, Naugten ER, Plecko-Starting B, Superti-Furga A,
tients with glutaric aciduria type I. Mol Genet Metab 2000; Zschocke J, Christensen E. Clinical course, early diagnosis,
71: 535–537 treatment, and prevention of disease in glutaryl-CoA dehy-
Busquets C, Merinero B, Christensen E, Gelpi JL, Campistol J, drogenase deficiency. Neuropediatrics 1996; 27: 115–123
Pineda M, Fernández-Alverez E, Prats JM, Sans A, Arteaga R, Hoffmann GF, Zschocke J. Glutaric aciduria type I: from clinical,
Martí M, Campos J, Martínez-Pardo M, Martinez-Bermejo A, biochemical and molecular diversity to successful therapy.
Ruiz-Falcó ML, Vaquerizo J, Orozco M, Ugarte M, Coll MJ, J Inherit Metab Dis 1999; 22: 381–391
Ribes A. Glutaryl-CoA dehydrogenase deficiency in Spain: Iafolla AK, Kahler SG. Megalencephaly in the neonatal period
evidence of two groups of patients, genetically, and bio- as the initial manifestation of glutaric aciduria type I. J Pedi-
chemically distinct. Pediatr Res 2000; 48: 315–322 atr 1989; 114: 1004–1006
Campistol J, Ribes A, Alvarez L, Christensen E, Millington DS. Jamjoom ZAB, Okamoto E, Jamjoom A-H B, Al Hajery O, Abu-
Glutaric aciduria type I: unusual biochemical presentation. Melha A. Bilateral arachnoid cysts of the sylvian region in
J Pediatr 1992; 121: 83–86 female siblings with glutaric aciduria type I. J Neurosurg
Chow CW, Haan EA, Goodman SI, Anderson RM, Evans WA, 1995; 82: 1078–1081
Kleinschmidt-de Masters BK, Wise G, McGill JJ, Danks DM. Kafil-Hussain NA, Monvarvi A, Bowell R, Thornton P, Naughten
Neuropathology in glutaric acidaemia type I. Acta Neuro- E, O’Keefe M. Ocular findings in glutaric aciduria type I.
pathol (Berl) 1988; 76: 590–594 J Pediatr Ophthalmol Strabismus 2000; 37: 289–293
Desai NK, Runge VM, Crisp DE, Crisp MB, Naul LG. Magnetic res- Kölker S, Ahlemeyer B, Kriegelstein J, Hoffmann GF. 3-Hydrox-
onance imaging of the brain in glutaric acidemia type I: a yglutaric and glutaric acids are neurotoxic through NMDA
review of the literature and a report of four new cases with receptors in vitro. J Inherit Metab Dis 1999; 22: 259–2623
attention to the basal ganglia and imaging technique. In- Kölker S, Ahlemeyer B, Kriegelstein J, Hoffmann GF. Evaluation
vest Radiol 2003; 38: 489–496 of trigger factors of acute encephalopathy in glutaric
Drigo P, Piovan S, Battistella PA, Della Puppa A, Burlina AB. aciduria type I: fever and tumour necrosis factor-a. J Inher-
Macrocephaly, subarachnoid fluid collection, and glutaric it Metab Dis 2000; 23: 359–362
aciduria type I. J Child Neurol 1996; 11: 414–417 Kölker S,Ahlemeyer B,Kriegelstein J,Hoffmann GF.Maturation-
Francois B, Jaeken J, Gillis P. Vigabatrin in the treatment of glu- dependent neurotoxicity of 3-hydroxyglutaric and glutaric
taric aciduria type I. J Inherit Metab Dis 1990; 13: 352–354 acids in vitro:a new pathophysiologic approach to glutaryl-
Goodman SE, Norenberg MD, Shikes RH, Breslich DJ, Moe PG. CoA dehydrognase deficiency. Pediatr Res 2000; 47: 495–
Glutaric aciduria: biochemical and morphologic considera- 503
tions. J Pediatr 1990; 90: 746–750 Kölker S, Ramaekers VT, Zschocke J, Hoffmann GF. Acute en-
Hald JK, Nakstad PH, Skjeldal OH, Strømme P. Bilateral arach- cephalopathy despite early therapy in a patient with ho-
noid cysts of the temporal fossa in four children with glu- mozygosity for E365 K in the glutaryl-coenzyme A dehy-
taric aciduria type I. AJNR Am J Neuroradiol 1991; 12: drogenase gene. J Pediatr 2001; 138: 277–279
407–409 Kölker S, Hoffmann GF, Schor DS, Feyh P, Wagner L, Jeffrey I,
Hartley LM, Khwaja OS, Verity CM. Glutatic aciduria type I and Pourfarzam M, Okun JG, Zschocke J, Baric I, Bain MD, Jakobs
nonaccidental head injury. Pediatrics 2001; 107: 174–175 C, Chalmers RA. Glutaryl-CoA dehydrogenase deficiency:
Hauser S, Peters H. Glutaric aciduria type I: an underdiagnosed region-specific analysis of organic acids and acylcarnitines
cause of encephalopathy and dystonia-dyskinesia syn- in post mortem brain predicts vulnerability of the puta-
drome in children. J Paediatr Child Health 1998; 34: 302– men. Neuropediatrics 2003; 34: 253–260
304 Kyllerman M, Skjeldal OH, Lundberg M, Holme I, Jellum E, von
Hauser SEP, Boneh A. Severe clincal cause with recurrent hy- Döbeln U, Fossen A, Carlsson G. Dystonia and dyskinesia in
perpyrexia in a patient with glutaric aciduria type I. Neuro- glutaric aciduria type I: clinical heterogeneity and thera-
pediatrics 1999; 30: 51–52 peutic considerations. Mov Dis 1994; 9: 22–30
Haworth JC, Booth FA, Chudley AE, de Groot GW, Dilling LA, Land JM, Goulder P, Johnson A, Hockaday J. Glutaric aciduria
Goodman SI,Greenberg CR,Mallory CJ,McClarty BM,Seshia type I. An atypical presentation together with some obser-
SS, Seargeant LE. Phenotypic variability in glutaric aciduria vations upon treatment and the possible cause of cerebral
type I: report of fourteen cases in five Canadian Indian kin- damage. Neuropediatrics 1992; 23: 322–326
dreds. J Pediatr 1991; 118: 52–58 Leibel RL, Shih VE, Goodman SI, Bauman ML, McCabe ERB, Zw-
Hoffmann GF, Trefz FK, Barth PG, Böhles HJ, Lehnert W, Chris- erdling RG, Bergman I, Costello C.Glutaric acidemia: a meta-
tensen E, Valk J, Rating D, Bremer HJ. Macrocephaly: an im- bolic disorder causing progressive choreoathetosis. Neu-
portant indication for organic acid analysis. J Inherit Metab rology 1980; 30: 1163–1168
Dis 1991; 14: 329–332 Lipkin PH, Roe CR, Goodman SI, Batshaw ML. A case of glutaric
Hoffmann GF, Trefz FK, Barth PG, Böhles HJ, Biggemann B, Bre- acidemia type I: effect of riboflavin and carnitine. J Pediatr
mer HJ, Christensen E, Frosch M, Hanefeld F, Hunneman DH, 1988; 112: 62–65
Jacobi H, Kurlemann G, Lawrenz-Wolf B, Rating D, Roe CR, Lütcherath V,Waaler PE, Jellum E,Wester K. Children with bilat-
Schutgens RBH, Ullrich K, Weisser J, Wendel U, Lehnert W. eral temporal arachnoid cysts may have glutaric aciduria
Glutaryl-coenzyme A dehydrogenase deficiency: a distinct type I (GATI); operation without knowing that may be
encephalopathy. Pediatrics 1991; 88: 1194–1203 harmful. Acta Neurochir (Wien) 2000; 142: 1025–1030
Hoffmann GF, Böhles HJ, Burlina A, Duran M, Herwig J, Lehnert Mandel H, Braun J, El-Peleg O, Christensen E, Berant M. Glutaric
W, Leonard JV, Muntau A, Plecko-Starting FK, Superti-Furga aciduria type I. Brain CT features and a diagnostic pitfall.
A,Trefz FK, Christensen E.Early signs and cause of disease of Neuroradiology 1991; 33: 75–78
glutaryl-CoA dehydrogenase deficiency.J Inherit Metab Dis
1995; 18: 173–176
References and Further Reading 969

Martinez-Lage JF, Casas C, Fernandez MA, Puche A, Rodriguez Vamecq J, de Hoffmann E, van Hoof F. Mitochondrial and per-
Costa T, Poza M. Macrocephaly, dystonia, and bilateral tem- oxisomal metabolism of glutaryl-CoA. Eur J Biochem 1985;
poral arachnoid cysts: glutaric aciduria type I. Childs Nerv 146: 663–669
Syst 1994; 10: 198–203 Woefle J, Kreft B, Emons D, Haverkamp F. Subdural hemorrhage
Martinez-Lage JF. Neurosurgical treatment for hydrocephalus, as an initial sign of glutaric aciduria type I: a diagnostic pit-
subdural hematomas, and arachnoid cysts in glutaric fall. Pediatr Radiol 1996; 26: 779–781
aciduria type I. Neuropediatrics 1996; 27: 335–336 Yager JY, McClarty BM, Seshia SS. CT scan findings in an infant
Merinero B, Pérez-Cerdá, Font LM, Garcia MJ, Aparicio M, Loren- with glutaric aciduria type I.Dev Med Child Neurol 1988; 30:
zo G, Martinez Pardo M, Garzo C, Martinez-Bermejo A, Cas- 808–820
troviejo IP, Christensen E, Ugarte M. Variable clinical and Zafeiriou DI, Zschocke J, Augoustidou-Savvopoulou P, Mauro-
biochemical presentation of seven Spanish cases with matis I, Sewell A, Kontopoulos E, Katzos G, Hoffmann GF.
glutaryl-CoA-dehydrogenase deficiency. Neuropediatrics Atypical and variable clinical presentation of glutaric
1995; 26: 238–242 aciduria type I. Neuropediatrics 2000; 31: 303–306
Monavari AA, Naughten ER. Prevention of cerebral palsy inglu- Zschocke J, Quak E, Guldberg P, Hoffmann GF. Mutation analy-
taric aciduria type I by dietary management. Arch Dis Child sis in glutaric aciduria type I.J Med Genet 2000; 37: 177–181
2000; 82: 67–70
Morton DH, Bennett MJ, Seargeant LE, Nichter CA, Kelley RI.
Glutaric aciduria type I: a common cause of episodic en-
cephalopathy and spastic paralysis in the Amish of Lancast-
38 Propionic Acidemia
er county, Pennsylvania. Am J Med Genet 1991; 4: 89–95
Nagasawa H, Yamaguchi S, Suzuki Y, Kobayashi M, Wada Y, Bergman AJIW,van der Knaap MS,Smeitink JAM,Duran M,Dor-
Shikura K, Shimao S, Okada T, Orii T. Neuroradiological find- land L,Valk J, Poll-The BT. Magnetic resonance imaging and
ings in glutaric aciduria type I: report of four Japanese pa- spectroscopy of the brain in propionic acidemia: clinical
tients. Acta Paediatr Jpn 1992; 34: 409–415 and biochemical considerations. Pediatr Res 1996; 40: 404–
Nyhan WL, Zschocke J, Hoffmann G, Stein DE, Bao L, Goodman 409
S. Glutaryl-CoA dehydrogenase deficiency presenting as Böhles H, Lehnert W. The effect of intravenous l-carnitine on
3-hydroxyglutaric aciduria. Mol Genet Metab 1999; 66: propionic acid excretion in acute propionic acidaemia. Eur
199–204 J Pediatr 1984; 143: 61–63
Osaka H, Kimura S, Nezu A, Yamazaki S, Saitoh K, Yamaguchi S. Brismar J, Ozand PT. CT and MR of the brain in disorders of the
Chronic subdural hematoma, as an initial manifestation of propionate and methylmalonate metabolism. AJNR Am J
glutaric aciduria type-I. Brain Dev 1993; 15: 125–127 Neuroradiol 1994; 15: 1459–1473
Pfluger T, Weil S, Muntau A, Willemsen UF, Hahn K. Glutaric Burlina AB, Dionisi-Vici C, Piovan S, Saponara I, Bartuli A, Sabet-
aciduria type I: a serious pitfall if diagnosed too late. Eur Ra- ta G, Zacchello F. Acute pancreatitis in propionic acidaemia.
diol 1997; 7: 1264–1266 J Inherit Metab Dis 1995; 18:169–172
Pineda M, Ribes A, Busquets C, Vilaseca MA, Aracil A, Chris- Chemelli AP, Schocke M, Sperl W, Trieb T, Aichner F, Felber S.
tensen E.Glutaric aciduria type I with high residual glutaryl- Magnetic resonance spectroscopy (MRS) in five patients
CoA dehydrogenase activity. Dev Med Child Neurol 1998; with treated propionic acidemia. J Magn Reson Imaging
40: 840–842 2000; 11: 596–600
Prevett MC, Howard RS, Dalton RN, Olpin SE. Glutaric aciduria Clavero S, Martínez MaA, Pérez B, Pérez-Cerdá C, Ugarte M,
type I in adutlhood.J Neurol Neurosurg Psychiatry 1996; 60: Desviat LR. Functional characterization of PCCA mutations
352–353 causing propionic acidemia. Biochim Biophys Acta 2002;
Renner C, Razeghi S, Überall MA, Hartmann P, Lehnert W. Clini- 1588: 119–125
cally asymptomatic glutaric aciduria type I in a 4 5/12-year- Gebarski SS, Gabrielsen TO, Knake JE, Latack JT. Cerebral CT
old girl with bilateral temporal arachnoid cysts. J Inherit findings in methylmalonic and propionic acidemias. AJNR
Metab Dis 1997; 20: 840–840 Am J Neuroradiol 1983; 4: 955–957
Schwartz M, Christensen E, Superti-Furga A, Brandt NJ.The hu- Gravel RA, Akerman BR, Lamhonwah AM, Loyer M, Léon-del-
man glutaryl-CoA dehydrogenase gene: report of intronic Rio A, Italiano I. Mutations participating in interallelic com-
sequences and of 13 novel mutations causing glutaric plementation in propionic acidemia. Am J Hum Genet
aciduria type I. Hum Genet 1998; 102: 452–458 1994; 55: 51–58
Soffer D, Amir N, Elpeleg ON, Gomori JM, Shalev RS, Gottschalk- Haas RH, Marsden DL, Capistrano-Estrada S, Hamilton R, Grafe
Sabag S.Striatal degeneration and spongy myelinopathy in MR, Wong W, Nyhand WL. Acute basal ganglia infarction in
glutaric acidemia. J Neurol Sci 1992; 107: 199–204 propionic acidemia. J Child Neurol 1995; 10: 18–22
Superti-Furga A, Hoffmann GF. Glutaric aciduria type I (glu- Hamilton RL, Haas RH, Nyhan WL, Powell HC, Grafe MR. Neu-
taryl-CoA–dehydrogenase deficiency):advances and unan- ropathology of propionic acidemia:a report of two patients
swered questions. Eur J Pediatr 1997; 156: 821–828 with basal ganglia lesions. J Child Neurol 1995; 10: 25–30
Twomey EL, Naughten ER, Donoghue VB, Ryan S. Neuroimag- Harding BN, Leonard JV, Erdohazi M. Propionic acidaemia: a
ing in glutaric aciduria type 1. Pediatr Radiol 2003; 33: neuropathological study of two patients presenting in in-
823–830 fancy. Neuropathol Appl Neurobiol 1991; 17: 133–138
Ullrich K, Flott-Rahmel B, Schluff P, Musshoff U, Das A, Lucke T, Hommes FA, Kuipers JRG, Elema JD, Jansen JF, Jonxis JHP. Propi-
Steinfeld R, Christensen E, Jakobs C, Ludolph A, Neu A, onicacidemia, a new inborn error of metabolism. Pediatr
Roper R.Glutaric aciduria type I:pathomechanisms of neuro- Res 1968; 2: 519–524
degeneration. J Inherit Metab Dis 1999; 22: 293–403 Inoue Y, Kuhara T. Rapid and sensitive method for prenatal
Vamecq J, van Hoof F. Implication of a peroxisomal enzyme in diagnosis of propionic acidemia using stable isotope dilu-
the catabolism of glutaryl-CoA. Biochem J 1984; 221: 203– tion gas chromatography-mass spectroscopy and urease
211 pretreatment. J Chromatogr B Biomed Sci Appl 2002; 776:
71–77
970 References and Further Reading

Kalloghlian A, Gleispach H, Ozand PT. A patient with propionic Agamanolis DP, Potter JL, Lundgren DW. Neonatal glycine en-
acidemia managed with continuous insulin infusion and cephalopathy: biochemical and neuropathologic findings.
total parenteral nutrition. J Child Neurol 1992; 7: S88-S91 Pediatr Neurol 1993; 9: 140–143
Kurczynski TW, Hoppel CL, Goldblatt PJ, Gunning WT. Metabol- Alejo J, Rincón P,Vaquerizo J, Gil C, Plasencia A, Gallardo R.Tran-
ic studies of carnitine in a child with propionic acidemia. sient nonketotic hyperglycinaemia: ultrasound, CT and MRI
Pediatr Res 1989; 26: 63–66 (case report). Neuroradiology 1997; 39: 658–660
Leonard JV. The management and outcome of propionic and Alemzadeh R, Gammeltoft K, Matteson K. Efficacy of low-dose
methylmalomic acidemia. J Inherit Metab Dis 1995; 18: dextromethorphan in the treatment of nonketotic hyper-
430–434 glycinemia. Pediatrics 1996; 97: 924–926
Leonard JV. Stable isotope studies in propionic and methyl- Aliefendioğlu D, Aslan AT, Colpkun T, Dursun A, Çakmak FN, Kes-
malonic acidaemia. Eur J Pediatr 1997; 156: S67–S69 imer M. Transient nonketotic hyperglycinemia: two case
Leonard JV, Walter JH, McKiernan PJ. The management of reports and literature review. Pediatr Neurol 2003; 28: 151–
organic acidaemias: the role of transplantation. J Inherit 155
Metab Dis 2001; 24: 309–311 Anderson JM. Spongy degeneration in the white matter of the
Muro S, Pérez B, Desviat R, Rodríguez-Pombo P, Pérez-Cerdá C, central nervous system in the newborn: pathological find-
Clavero S, Ugarte M. Effect of PCCB gene mutations on the ings in three infants, one with hyperglycinaemia. J Neurol
heteromeric and homomeric assambly of propionyl-CoA Neurosurg Psychiatry 1969; 32: 328–337
carboxylase. Mol Genet Metab 2001; 74: 476–483 Ando T, Nyhan WL, Bicknell J, Harris R, Stern J. Non-ketotic
Nyhan WL, Bay C, Webb Beyer E, Mazi M. Neurologic nonmeta- hyperglycinaemia in a family with an unusual phenotype.
bolic presentation of propionic acidemia. Arch Neurol J Inherit Metab Dis 1978; 1:79–83
1999; 56: 1153–1147 Appelgarth DA, Toone JR, Rolland MO, Black SH, Yim DKC, Be-
Ozand PT, Rahed M, Gascon GG, Youssef NG, Harfi H, Rahbeeni mis G. Non-concordance of CVS and live glycine cleavage
Z, Al Garawi S, Aqeel A Al. Unusual presentation of propi- enzyme in three families with non-ketotic hyperglyci-
onic acidemia. Brain Dev 1994; 16: 46–57 naemia (NKH) leading to false negative prenatal diagnosis.
Rolland MO,Divry P,Mandon G,Guibaud P,Mathieu M,Sournies Prenat Diagn 2000; 20: 367–370
G, Thoulon JM. Early prenatal diagnosis of propionic aci- Arnold GL, Griebel ML, Valentine JL, Koroma DM, Kearns GL.
daemia with simultaneous sampling of chorionic villus and Dextromethorphan in nonketotic hyperglycinaemia: mata-
amniotic fluid. J Inherit Metab Dis 1990; 13: 345–348 bolic variation confounds the dose–response relationship.
Schlenzig JS, Poggi-Travert F, Laurent J, Rabier D, Jan D,Wendel J Inherit Metab Dis 1997; 20: 28–38
U, Sewell AC, Revellion Y, Kamoun P, Saudubray JM. Liver Bank WJ. A familial spinal cord disorder with hyperglycinemia.
transplantation in two cases of propionic acidaemia. Arch Neurol 1972; 27: 136–144
J Inherit Metab Dis 1995; 18: 448–461 Bekiesniñska-Figatowska M, Rokicki D,Walecki J. MRI in nonke-
Sethi KD, Ray R, Roesel RA, Carter AL, Callagher BB, Loring DW, totic hyperglycinaemia: case report. Neuroradiology 2001;
Hommes FA. Adult-onset chorea and dementia with propi- 43: 792–793
onic acidemia. Neurology 1989; 39: 1343–1345 Boneh A, Degani Y, Harari M. Prognostic clues and outcome of
Shigematsu Y, Mori I, Nakai A, Kikawa Y, Kuriyama M, Konishi Y, early treatment of nonketotic hyperglycenemia. Pediatr
Fuji T, Sudo M. Acute infantile hemiplegia in a patient with Neurol 1996; 15: 137–141
propionic acidaemia. Eur J Pediatr 1990; 149: 659–660 Brun A, Börjeson M, Hultberg B, Sjöblad S, Akesson H, Litwin E.
Steinman L, Clancy RR, Cann H, Urich H.The neuropathology of Neonatal non-ketotic hyperglycinemia a clinical, biochem-
propionic acidemia. Dev Med Child Neurol 1983; 25: 87–94 ical and neuropathological study including electronmicro-
Surtees RAH, Matthews EE, Leonard JV. Neurologic outcome of scopic findings. Neuropädiatrie 1979; 10: 195–205
propionic acidemia. Pediatr Neurol 1992; 8: 333–337 Choi C-G,Lee HHK,Yoon J-H.Localized proton MR spectroscop-
Ugarte M,Pérez-Cerdá C,Rodriguez-Pombo P,Desviat LR,Pérez ic detection of nonketotic hyperglycinemia in an infant.
B, Richard E, Muro S, Campeau E, Ohura T, Gravel RA. Korean J Radiol 2001; 2: 239–242
Overview of mutations in the PCCA and PCCB genes caus- Christodoulou J,Kure S,Hayasaka K,Clarke JTR.Atypical nonke-
ing propionic acidemia. Hum Mutat 1999; 14: 275–282 totic hyperglycinemia confirmed by assay of the glycine
Wolf B, Hsia YE, Sweetman L, Gravel R, Harris DJ, Nyhan WL. cleavage system in lymphoblasts. J Pediatr 1993; 123: 100–
Propionic acidemia: a clinical update. J Pediatr 1981; 99: 102
835–846 Dalla Bernardina B, Aicardi J, Goutières F, Plouin P. Glycine
Yorifuji T, Kawai M, Muroi J, Mamada M, Kurokawa K, Shige- encephalopathy. Neuropädiatrie 1978; 10: 209–225
matsu Y, Hirano S, Sakura N, Yoshida I, Kuhara T, Endo F, Deutsch SI, Rosse RB, Mostropaolo J. Current status of NMDA
Mishubushi H, Nakahata T. Unexpectedly high prevalence antagonist interventions in the treatment of nonketotic
of the mild form of propionic acidemia in Japan: presence hyperglycinemia. Clin Neuropharmacol 1998; 21: 71–79
of a common mutation and possible clinical implications. Dobyns WB. Agenesis of the corpus callosum and gyral malfor-
Hum Genet 2002; 111: 161–165 mations are frequent manifestations of nonketotic hyper-
glycinemia. Neuroradiology 1989; 39: 817–820
Eyskens FJM, van Doorn JWD, Mariën P. Clinical and laboratory
observations. Neuroradiologic sequelae in transient non-
39 Nonketotic Hyperglycinemia ketotic hyperglycinemia of the neonate.J Pediatr 1992; 121:
620–621
Agamanolis DP, Potter JL, Herrick MK, Sternberger NH.The neu- Flannery DB, Pellock J, Bousounis D, Hunt P, Nance C, Wolf B.
ropathology of glycine encephalopathy: a report of five Nonketotic hyperglycinemia in two retarded adults: a mild
cases with immunohistochemical and ultrastructural ob- form of infantile nonketotic hyperglycinemia. Neurology
servations. Neurology 1982; 32: 975–985 1983; 33: 1064–1066
References and Further Reading 971

Fletcher JM, Bye AME, Nayanar V,Wilcken B. Non-ketotic hyper- Luder AS, Davidson A, Goodman SI, Greene CL. Transient non-
glycinaemia presenting as pachygyria. J Inherit Metab Dis ketotic hyperglycinemia in neonates. J Pediatr 1989; 114:
1995; 18: 665–668 1013–1015
Frazier DM, Summer GK, Chamberlin HR. Hyperglycinuria and Matsuo S, Inoue F, Takeuchi Y, Yoshioka H, Kinugasa A, Sawada
hyperglycinemia in two siblings with mild developmental T. Efficacy of tryptophan for the treatment of nonketotic
delays. Am J Dis Child 1978; 132: 777–781 hyperglycinemia: a new therapeutic approach for modulat-
Gabis L, Parton P, Roche P, Lenn N, Tudorica A, Huang W. In vivo ing the N-methyl-D-aspartate receptor. Pediatrics 1995; 95:
1H magnetic resonance spectroscopic measurement of
142–146
brain glycine levels in nonketotic hyperglycinemia. J Neu- Nanao K, Okamura-Ikeda K, Motokawa Y, Danks DM, Baumgart-
roim 2001; 11: 209–211 ner ER, Takada G, Hayasaka K. Identification of the muta-
Hamosh A, Maher JF, Bellus GA, Rasmussen SA, Johnston MV. tions in the T-protein gene causing typical and atypical
Long-term use of high-dose benzoate and dextromethor- nonketotic hyperglycinemia. Hum Genet 1994; 93: 665–658
phan for the treatment of nonketotic hyperglycinemia. Neuberger JM, Schweitzer S, Rolland M-O, Burghard R. Effect of
J Pediatr 1998; 132: 709–713 sodium benzoate in the treatment of atypical nonketotic
Hayasaka K,Tada K, Fueki N, Kakamura Y, Nyhan WL, Schmidt K, hyperglycinaemia. J Inherit Metab Dis 2000; 23: 22–26
Packman S, Seashore MR, Haan E, Danks DM, Schutgens Nightingale S, Barton ME. Intermittent vertical supranuclear
RBH. Nonketotic hyperglycinemia: analysis of glycine cleav- ophthalmoplegia and ataxia. Mov Disord 1991; 6: 76–78
age system in typical and atypical cases.J Pediatr 1987; 110: Ohya Y, Ochi N, Mizutani N, Hayakawa C, Watanabe K. Non-
873–877 ketotic hyperglycinemia: treatment with NMDA antagonist
Heindel W, Kugel H, Roth B. Noninvasive detection of increased and consideration of neuropathogenesis. Pediatr Neurol
glycine content by proton MR spectroscopy in the brains of 1991; 7: 65–68
two infants with nonketotic hyperglycinemia. AJNR Am J Press GA, Barshop BA, Haas RH, Nyhan WL, Glass RF, Hesselink
Neuroradiol 1993; 14: 629–635 JR. Abnormalities of the brain in nonketotic hyperglycine-
Holmgren G, Kison Blomquist H. Non-ketotic hyperglycinemia mia: MR manifestations. AJNR Am J Neuroradiol 1989; 10:
in two sibs with mild psycho-neurological symptoms. 315–321
Neuropädiatrie 1977; 8: 67–72 Rushton DI. Spongy degeneration of the white matter of the
Huisman TAGM,Thiel T, Steinmann B, Zeilinger G, Martin E. Pro- central nervous system associated with hyperglycinuria.
ton magnetic resonance spectroscopy of the brain of a J Clin Pathol 1968; 21: 246–462
neonate with nonketotic hyperglycinemia: in vivo-in vitro Schiffmann R, Kaye EM, Willis JK III, Africk D, Ampola M. Tran-
(ex vivo) correlation. Eur Radiol 2002; 12: 858–861 sient neonatal hyperglycinemia. Ann Neurol 1989; 25:
Khong PL, Lam PCC, Chung BHY, Wong KY, Ooi GC. Diffusion- 201–201
weighted MR imaging in neonatal nonketotic hyperglycin- Schmitt B, Steinmann B, Gitzelmann R, Thun-Hohenstein L,
emia. AJNR Am J Neuroradiol 2003; 24: 1181–1183 Mascher H, Dumermuth G. Nonketotic hyperglycinemia:
Kish SJ, Dixon LM, Burnham WM, Perry TL, Becker L, Cheng J, clinical and electrophysiologic effects of dextromethor-
Chang LJ, Rebbetoy M. Brain neurotransmitters in glycine phan, an antagonist of the NMDA receptor. Neurology
encephalopathy. Ann Neurol 1988; 24: 458–461 1993; 43: 421–424
Korman SH, Gutman S. Pitfalls in the diagnosis of glycine Shuman RM, Leech RW, Scott CR. The neuropathology of the
encephalopathy (non-ketotic hyperglycinemia). Dev Med nonketotic and ketotic hyperglycinemias: three cases. Neu-
Child Neurol 2002; 44: 712–720 rology 1978; 28: 139–146
Kure S, Narisawa K, Tada K. Enzymatic diagnosis of nonketotic Singer HS,Valle D, Hayasaka K,Tada K. Nonketotic hyperglycin-
hyperglycinemia with lymphoblasts. J Pediatr 1992; 120: emia: studies in an atypical variant. Neurology 1989; 39:
95–98 286–288
Kure S,Takayanagi M Nirisawa K Tada K,Leisti J.Identification of Slager UT, Berggren RL, Marubayashi S. Nonketotic hyper-
a common mutation in Finnish patients with nonketotic glycinemia: report of a case and review of the clinical,
hyperglycinemia. J Clin Invest 1992; 90: 160–164 chemical, and pathological changes. Ann Neurol 1977; 1:
Kure S, Tada K, Narisawa K. Nonketotic hyperglycenemia: bio- 399–402
chemical, molecular, and neurological aspects. Jpn J Hum Steiner RD, Sweetser DA, Rohrbaugh JR, Dowton B, Toone JR,
Genet 1997; 42: 13–22 Applegarth DA. Nonketotic hyperglycinemia: atypical clini-
Kure S,Rolland M-O,Leisti J,Mandel H,Sakata Y,Tada K,Matsub- cal and biochemical manifestations. J Pediatr 1996; 128:
ara Y, Narisawa K. Prenatal diagnosis of non-ketotic hyper- 243–246
glycenaemia: enzymatic diagnosis in 28 families and DNA Steinman GS, Yudkoff M, Berman PH, Blazer-Yost B, Segal S.
diagnosis detecting prevalent Finnish and Israeli-Arab mu- Late-onset nonketotic hyperglycinemia and spinocerebel-
tations. Prenat Diagn 1999; 19: 717–720 lar degeneration. J Pediatr 1979; 94: 907–911
Kure S, Kojima K, Kudo T, Kanno K, Aoiki Y, Suzuki Y, Shinka T, Tada K, Hayasaka K. Non-ketotic hyperglycinaemia: clinical and
Sakata Y, Narisawa K, Matsubara Y. Chromosomal localiza- biochemical aspects. Eur J Pediatr 1987; 146: 221–227
tion, structure, single-nucleotide polymorphisms, and ex- Tada K, Kure S. Non-ketotic hyperglycinaemia: molecular le-
pression of the human H-protein gene of the glycine cleav- sion, diagnosis and pathophysiology. J Inherit Metab Dis
age system (GCSH), a candidate gene for nonketotic hyper- 1993; 16: 691–703
glycinemia. J Hum Genet 2001; 46: 378–384 Takayanagi M, Kure S, Sakata Y, Kurihara Y, Ohya Y, Kajita M,
Kure S, Kojima K, Ichinohe A, Maeda T, Kalmanchey R, Feteke G, Tada K, Matsubara Y, Narisawa K.Human glycine decarboxy-
Berg SZ, Filiano J, Aoki Y, Suzuki Y, Izumi T, Matsubara Y. Het- lase gene (GLDC) and its highly conserved processed
erozygous GLDC and GCSH gene mutations in transient pseudogene (øGLDC): their structure and expression, and
neonatal hyperglycinemia. Ann Neurol 2002; 52: 643–646 the identification of a large deletion in a family with non-
Lu FL, Wang P-J, Hwu W-L. Taou Yau K-I, Wang T-R. Neonatal ketotic hyperglycinemia. Hum Genet 2000; 106: 298–305
type of nonketotic hyperglycinemia. Pediatr Neurol 1999;
20: 295–300
972 References and Further Reading

Toone JR, Applegarth DA, Coulter-Mackie M, James ER. Identifi- Felber SR, Sperl W, Chemelli A, Murr Ch,Wendel U. Maple syrup
cation of the first reported splice site mutation (IVS7–1GA) urine disease: metabolic decompensation monitored by
ion th aminomethyltransferase (T-protein) gene (AMT) of proton magnetic resonance imaging and spectroscopy.
the glycine cleavage complex in 3 unrelated families with Ann Neurol 1993; 33: 396–401
nonketotic hyperglycinemia. Hum Mutat 2000; 17: 76–81 Giacoia GP, Berry GT. Acrodermatitis enteropathica-like syn-
Tauner DA, Page T, Green C, Sweetman L, Kulovich S, Nyhan WL. drome secondary to isoleucine deficiency during treat-
Progressive neurodegenerative disorder in a patient with ment of maple syrup urine disease. Am J Dis Child 1993;
nonketotic hyperglycinemia. J Pediatr 1981; 98: 272–275 147: 954–956
Van Hove JLK, Kishnani PS, Damaerel P, Kahler SG, Miller C, Gouyon JB, Semama D, Prévot A, Desgres J. Removal of
Jaeken J, Rutledge SL. Acute hydrocephalus in nonketotic branched-chain amino acids and a-ketoisocaproate by
hyperglycinemia. Neurology 2000; 54: 754–756 haemofiltration and haemodiafiltration. J Inherit Metab Dis
Wiltshire EJ, Poplawski NK, Harrison JR, Fletcher JM. Treatment 1996;19: 610–620
of late-onset nonketotic hyperglycinaemia:effectiveness of Ha JS, Kim TK, Eun BL, Lee HS, Lee KY, Seol HY, Cha SH. Maple
imipramine and benzoate. J Inherit Metab Dis 2000; 23: syrup urine disease encephalopathy: a follow-up study in
15–21 the acute stage using diffusion-weighted MRI. Pediatr
Wraith JE. Non-ketotic hyperglycinemia: prolonged survival in Radiol 2004; 34: 163–166
a patient with a mild variant. J Inherit Metab Dis 1996; 19: Hilliges C, Awiszus D, Wendel U. Intellectual performance of
695–696 children with maple syrup urine disease. Eur J Pediatr 1993;
Zammarchi E, Donati MA, Ciani F, Pasquini E, Pela I, Fiorini P. Fail- 152: 144–147
ure of early dextromethorphan and sodium benzoate ther- Indo Y, Akaboshi I, Nobukumi Y, Endo F, Matsuda I. Maple syrup
apy in an infant with nonketotic hyperglycinemia. Neuro- urine disease: a possible biochemical basis for the clinical
pediatrics 1994; 25: 274–276 heterogeneity. Hum Genet 1988; 80: 6–10
Zammarchi E, Donati MA, Ciani F.Transient neonatal nonketot- Jan W, Zimmerman RA, Wang ZJ, Berry GT, Kaplan PB, Kaye EM.
ic hyperglycinemia: a 13-year follow-up. Neuropediatrics MR diffusion imaging and MR spectroscopy of maple syrup
1995; 26: 328–330 urine disease during acute metabolic decompensation.
Neuroradiology 2003; 45: 393–399
Jouvet P, Poggi F, Rabier D, Michel JL, Hubert P, Sposito M,
Saudurbray JM, Man NK. Continuous venovenous haemo-
40 Maple Syrup Urine Disease diafiltration in the acute phase of neonatal maple syrup
disease. J Inherit Metab Dis 1997; 20: 463–477
Backhouse O, Leitch RJ,Thompson D, Kriss A, Charris D, Clayton Jouvet P, Rustin P, Taylor DL, Pocock JM, Felderhoff-Mueser U,
P, Russel-Eggitt I. A case of reversible blindness in maple Mazarakis ND, Sarraf C, Joashi U, Kozma M, Greenwood K,
syrup urine disease. Br J Ophthalmol 1999; 83: 250–251 Edwards DA, Mehmet H. Branched chain amino acids in-
Berry GT, Heidenreich R, Kaplan P, Levine F, Mazur A, Palmieri duce apoptosis in neural cells without mitochondrial mem-
MJ,Yudkoff M,Segal S.Branched-chain amino acid-free par- brane depolarization or cytochrome c release: implications
enteral nutrition in the treatment of acute metabolic de- for neurological impairment associated with maple syrup
compensation in patients with maple syrup urine disease. urine disease. Mol Biol Cell 2000; 11: 1919–1932
N Engl J Med 1991; 324: 175–179 Jouvet P, Jugie M, Rabier D, Desgrès J, Hubert P, Saudubray JM,
Biggemann B, Zass R, Wendel U. Postoperative metabolic de- Man NK. Combined nutritional support and continuous
compensation in maple syrup urine disease is completely extracorporeal removal therapy in the severe acute phase
prevented by insulin. J Inherit Metab Dis 1993; 16: 912–913 of maple syrup urine disease. Intensive Care Med 2001; 27:
Brismar J, Aqeel A, Brismar G, Coates R, Gascon G, Ozand P. 1798–1806
Maple syrup urine disease: findings on CT and MR scans of Kamei A, Takashima S, Chan F, Becker LE. Abnormal dendritic
the brain in 10 infants. AJNR Am J Neuroradiol 1990; 11: development in maple syrup urine disease. Pediatr Neurol
1219–1228 1992; 8: 145–147
Cavelleri F, Berardi A, Burlina AB, Ferrari F, Mavilla L. Diffusion- Kaplan P, Mazur A, Field M, Berlin JA, Berry GT, Heidenreich R,
weighted MRI of maple syrup urine disease encephalopa- Yudkoff M, Segal S. Intellectual outcome in children with
thy. Neuroradiology 2002; 44: 449–502 maple syrup urine disease. J Pediatr 1991; 119: 46–50
Chuang DT, Davie JR, Max Wynn R, Chuang JL, Koyata H, Cox RP. Kleopa KA, Raizen DM, Friedrich CA, Brown MJ, Bird SJ. Acute
Molecular basis of maple syrup urine disease and stable axonal neuropathy in maple syrup urine disease. Muscle
correction by retroviral gene transfer. J Nutr 1995; 125: Nerve 2001; 24: 284–287
1766S–1772S Menkes JH, Philippart M, Fiol RE. Cerebral lipids in maple syrup
Chuang JL, Chuang DT. Diagnosis and mutational analysis of disease. J Pediatr 1965; 66: 584–594
maple syrup urine disease using cell cultures. Methods En- Menkes JH, Solcher H. Maple syrup disease. Arch Neurol 1967;
zymol 2000; 324: 413–464 16: 486–491
Delis D, Michelakakis H, Katsarou E, Bartsocas CS. Thiamin-re- Morton DH, Strauss KA, Robinson DL, Puffenberger EG, Kelly RI.
sponsive maple syrup urine disease: seizures after 7 years Diagnosis and treatment of maple syrup disease: a study of
of satisfactory metabolic control. J Inherit Metab Dis 2001; 36 patients. Pediatrics 2002; 109: 999–1008
24: 683–684 Müller K, Kahn T,Wendel U. Is demyelination a feature of maple
Fariello G, Dionisi-Vice C, Orazi C, Malena S, Bartuli A, Schingo P, syrup urine disease? Pediatr Neurol 1993; 9: 375–382
Carnavale E, Saponara I, Sabetta G. Cranial ultrasonography Nellis MM, Danner DJ. Gene preference in maple syrup urine
in maple syrup urine disease. AJNR Am J Neuroradiol 1996; disease. Am J Hum Genet 2001; 68: 232–237
17: 311–315 Nobukuni Y, Mitsubuchi H, Akaboshi I, Indo Y, Endo F, Matsuda
I.Maple syrup urine disease: clinical and biochemical signif-
icance of gene analysis. J Inherit Metab Dis 1991; 14: 787–
792
References and Further Reading 973

Nord A, van Doorninck WJ, Greene C. Developmental profile of Tsuruta M, Mitsubuchi H, Mardy S, Miura Y, Hayashida Y, Kinu-
patients with maple syrup urine disease.J Inherit Metab Dis gasa A, Ishitsu T, Matsuda I, Indo Y. Molecular basis of inter-
1991; 14: 881–889 mittent maple syrup urine disease: novel mutations in the
Northrup H, Sigman ES, Hebert AA. Exfoliative erythroderma E2 gene of the branched-chain (ALPHA)-keto acid dehydro-
resulting from inadequate intake of branched-chain amino genase complex. J Hum Genet 1998; 43: 91–100
acids in infants with maple syrup urine disease. Arch Der- Uziel G, Savoiardo M, Nardocci N. CT and MRI in maple syrup
matol 1993; 129: 384–385 urine disease. Neurology 1988; 38: 486–488
Nyhan WL, Rice-Kelts M, Klein J, Barshop BA. Treatment of the Verdu A, Lopez-Herce J, Pascual-Castroviejo I, Martinez-Berme-
acute crisis in maple syrup urine disease. Arch Pediatr jo A, Ugarte M, Garcia MJ. Maple syrup urine disease variant
Adolesc Med 1998; 152: 593–598 form: presentation with psychomotor retardation and CT
Ogier de Baulny H, Saudurbray JM. Branched-chain organic scan abnormalities. Acta Paediatr Scand 1985; 74: 815–818
acidurias. Semin Neonatol 2002; 7: 65–74 Wanjer M, Coelho DM, Barschak AG, Araújo PR, Pires RF, Lulhier
Parini R, Sereni LP, Bagozzi DC, Corbetta C, Rabier D, Narcy C, FLG, Vargas CR. Reduction of large neutral amino acid con-
Hubert P, Saudubray JM. Nasogastric drip feeding as the centrations in plasma and CSF of patients with maple syrup
only treatment of neonatal maple syrup urine disease. urine disease during crisis. J Inherit Metab Dis 2000; 23:
Pediatrics 1993; 92: 280–283 505–512
Parmar H, Sitoh YY, Ho L. Maple syrup urine disease. Diffusion- Wendel U, Saudubray JM, Bodner A, Schadewaldt P. Liver trans-
weighted and diffusion-tensor magnetic resonance imag- plantation in maple syrup disease. Eur J Pediatr 1999; 158:
ing findings. J Comput Assist Tomogr 2004; 28: 93–97 S60-S64
Parsons HG, Carter RJ, Unrath M, Snyder FF. Evaluation of Wynn RM, Ho R, Chuang JL, Chuang DT. Roles of active site and
branched-chain amino acid intake in children with maple novel K+ ion-binding site residues in human mitochondrial
syrup urine disease and methylmalonic aciduria. J Inherit branched chain a-ketoacid decarboxylase/dehydroge-
Metab Dis 1990; 13: 125–136 nase. J Biol Chem 2001; 9: 4168–4174
Prensky AL, Moser HW. Brain lipids, proteolipids, and free Yoshino M,Aoki K,Akeda H,Hashimoto K,Ikeda T,Inoue F,Ito M,
amino acids in maple syrup urine disease. J Neurochem Kawamura M, Kohno Y, Koga Y, Kuroda Y, Maesaka H, Mu-
1966; 13: 863–874 rakami-Soda H, Sugiyama N, Suzuki Y, Yano S, Yoshioka A.
Puliyanda DP, Harmon WE, Peterschmitt MJ, Irons M, Somers Management of acute metabolic decompensation in
MJG. Utility of hemodialysis in maple syrup urine disease. maple syrup urine disease: a multi-center study. Pediatr Int
Pediatr Nephrol 2002; 17: 239–242 1999; 41: 132–173
Riviello JJ, Rezvani I, DiGeorge AM, Foley CM. Cerebral edema Zneimer SM, Lau KS, Eddy RL, Shows TB, Chuang JL, Chuang DT,
causing death in children with maple syrup urine disease. Cox RD. Regional assignment of two genes of the human
J Pediatr 1991; 119: 42–45 branched-chain (ALPHA)-keto acid dehydrogenase com-
Schönberger S, Schweiger B, Schwahn B, Schwarz M,Wendel U. plex: the E1b gene (BCKDHB) to chromosome 6p21–22 and
Dysmyelination in the brain of adolescents and young the E2 gene (DTB) to chromosome 1p31. Genomics 1991;
adults with maple syrup urine disease. Mol Genet Metab 10: 740–747
2004; 82: 69–75
Scriver CR, Clow CL, Mackenzie S, Delvin E. Thiamine-respon-
sive maple-syrup-urine disease. Lancet 1971; I: 310–312
Scriver CR, Clow CL, George H. So-called thiamin-responsive
41 3-Hydroxy-3-Methylglutaryl CoA Lyase
maple syrup urine disease: 15-year follow-up of the original Deficiency
patient. J Pediatr 1985; 107: 763–765
Sener RN.Diffusion magnetic resonance imaging in intermedi- Bakker HD, Wanders RJA, Schutgens RBH, Abeling NGGM, van
ate form of maple syrup urine disease. J Neuroimaging Gennip AH. 3-Hydroxy-3-methylglutaryl-CoA lyase defi-
2002; 12: 368–390 ciency: absence of clinical symptoms due to a self-imposed
Silberberg DH. Maple syrup urine disease metabolites studies dietary fat and protein restriction. J Inherit Metab Dis 1993;
in cerebellum cultures. J Neurochem 1969; 16: 1141–1146 16: 1061–1062
Silberman J, Dancis J, Feigin I. Neuropathological observations Ferris JN,Tien RD. Cerebral MRI in 3-hydroxy-3-methylglutaryl-
in maple syrup urine disease. Arch Neurol 1961; 5: 351–363 coenzyme A lyase deficiency: case report. Neuroradiology
Taccone A, Schiaffino MC, Cerone R, Fondelli MP, Romano C. 1993; 35: 559–560
Computed tomography in maple syrup urine disease. Eur J Gibson KM, Breuer J, Kaiser K, Nyhan WL, McCoy EE, Ferreira P,
Radiol 1992; 14: 207–212 Greene CL, Blitzer MG, Shapira E, Reverte F, Conde C, Bagnell
Tharp BR. Unique EEG pattern (comb-like rhythm) in neonatal P, Cole DEC. 3-Hydroxy-3-methylglutaryl-coenzyme A lyase
maple syrup urine disease. Pediatr Neurol 1992; 8: 65–68 deficiency: report of five new patients. J Inherit Metab Dis
Thompson GN, Francis DEM, Halliday D. Acute illness in maple 1988; 11: 76–87
syrup urine disease: dynamics of protein metabolism and Gibson KM, Breuer J, Nyhan WL. 3-Hydroxy-3-methylglutaryl-
implications for management. J Pediatr 1991; 119: 35–41 coenzyme A lyase deficiency: review of 18 reported pa-
Tornqvist K, Tornqvist H. Corneal de-epithelialization caused tients. Eur J Pediatr 1988: 148: 180–186
by acute deficiency of isoleucine during treatment of a pa- Gibson KM, Cassidy SB, Seaver LH,Wanders RJA, Kennaway NG,
tient with maple syrup urine disease. Acta Ophthalmol Mitchell GA, Sprak RP. Fatal cardiomyopathy associated
Scand 1996; 74: 48–49 with 3-hydroxy-3-methylglutaryl-CoA lyase deficiency.
Treacy E, Clow CL, Reade TR, Chitayat D, Mamer OA, Scriver CR. J Inherit Metab Dis 1994: 17: 291–294
Maple syrup urine disease: interrelations between Gordon K, Riding M, Camfield P, Bawden H, Ludman M, Bagnell
branched-chain amino, oxo- and hydroxyacids; implica- P. CT and MR of 3-hydroxy-3-methylglutaryl-coenzyme A
tions for treatment; associations with CNS dysmyelination. lyase deficiency. AJNR Am J Neuroradiol 1994: 15: 1474–
J Inherit Metab Dis 1992; 15: 121–135 1476
974 References and Further Reading

Huemer M, Muehl A, Wandl-Vergesslich K, Strobl W, Wanders Adachi M, Schneck L, Cara J, Volk BW. Spongy degeneration of
RJA, Stoeckler-Ipsiroglu S. Stroke-like encephalopathy in an the central nervous system (Van Bogaert and Bertrand
infant with 3-hydroxy-3-methylglutaryl-coenzyme A lyase type; Canavan’s disease). Hum Pathol 1973; 4: 331–347
deficiency. Eur J Pediatr 1998; 157: 743–746 Austin SJ,Connelly A,Gadian DG,Benton JS,Brett EM.Localized
1H NMR spectroscopy in Canavan’s disease: a report of two
Leupold D, Bojasch M, Jacobs C. 3-Hydroxy-3-methylglutaryl-
coenzyme A lyase deficiency in an infant with macro- cases. Magn Reson Med 1991; 19: 439–445
cephaly and mild metabolic acidosis. Eur J Pediatr 1982; Banker BQ, Robertson JT, Victor M. Spongy degeneration of
138: 73–76 the central nervous system in infancy. Neurology 1964; 14:
Lisson G, Leupold D, Bechinger D, Wallesch C. CT findings in a 981–1001
case of deficiency of 3-hydroxy-3-methylglutaryl-CoA Barker PB, Bryan RN, Kumar AJ, Naidu S. Proton NMR spec-
lyase. Neuroradiology 1981; 22: 99–101 troscopy of Canavan’s disease. Neuropediatrics 1992; 23:
Mitchell GA, Jakobs C, Gibson KM, Robert M-F, Burlina A, Dion- 263–267
isi-Vici C, Dallaire L. Molecular prenatal diagnosis of 3-hy- Baslow MH. Molecular water pumps and the aetiology of
droxy-3-methylglutaryl-CoA lyase deficiency. Prenat Diagn Canavan’s disease: a case of sorcerer’s apprentice. J Inherit
1995; 15: 725–729 Metab Dis 1999; 22: 99–101
Ozand PT, Al Aqeel A, Gascon G, Brismar J,Thomas E, Gleispach Baslow MH. Canavan’s spongiform leukodystrophy. A clinical
H. 3-Hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) anatomy of a genetic metabolic CNS disease. J Mol Neu-
lyase deficiency in Saudi Arabia. J Inherit Metab Dis 1991; rosci 2000; 15: 61–69
14: 174–188 Baslow MH. Evidence supporting a role for N-acetyl-L-aspar-
Robinson BH,Oei J,Sherwood G,Slyper AH,Heininger J,Mamer tate as a molecular water pump in myelinated neurons in
OA. Hydroxymethylglutaryl CoA lyase deficiency: features the central nervous system. An analytical review. Neuro-
resembling Reye syndrome. Neurology 1980; 30: 714–718 chem Int 2002; 40: 295–300
Thompson GN, Chalmers RA, Halliday D. The contribution of Baslow MH. Brain N-acetylaspartate as a molecular water
protein catabolism to metabolic decompensation in 3-hy- pump and its role in the etiology of Canavan disease. J Mol
droxy-3-methylglutaric aciduria. Eur J Pediatr 1990; 149: Neurosci 2003; 21: 185–189
346–350 Baslow MH, Resnik TR. Canavan Disease. Analysis of the nature
van der Knaap MS, Bakker HD, Valk J. MR imaging and proton of the metabolic lesions responsible for development of
spectroscopy in 3-hydroxy-3-methylglutaryl-coenzyme A the observed clinical symptoms. J Mol Neurosci 1997; 9:
lyase deficiency. AJNR Am J Neuroradiol 1998; 19: 378–382 109–125
Walter JH, Clayton PT, Leonard JV. 3-Hydroxy-3-methylglutaryl- Baslow MH, Suckow RF,Sapirstein V,Hungund BL.Expression of
coenzyme A lyase deficiency. J Inherit Metab Dis 1986; 9: aspartoacylase activity in cultured rat macroglial cells is
287–288 limited to oligodendrocytes.J Mol Neurosci 1999; 13: 47–53
Wang SP, Robert M-F, Gibson KM, Wanders RJA, Mitchell GA. 3- Bennett MJ, Gibson KM, Sherwood WG, Divry P, Rolland MO,
Hydroxy-3-methylglutaryl-CoA lyase (HL): mouse and hu- Elpeleg ON, Rinaldo P, Jakobs C. Reliable prenatal diagnosis
man HL gene (HMGCL) cloning and detection of large gene of Canavan disease (aspartoacylase deficiency): compari-
deletions in two unrelated HL-deficient patients. Genomics son of enzymatic and metabolite analysis. J Inherit Metab
1996; 33: 99–104 Dis 1993; 16: 831–836
Wilson WG, Cass MB, Søvik O, Gibson KM, Sweetman L. A child Besley GTN, Elpeleg ON, Shaag A, Manning NJ, Jakobs C,Walter
with acute pancreatitis and recurrent hypoglycemia due to JH. Prenatal diagnosis of Canavan disease. Problems and
3-hydroxy-3-methylglutaryl-CoA lyase deficiency. Eur J Pe- dilemmas. J Inherit Metab Dis 1999; 22: 263–366
diatr 1984; 142: 289–291 Brismar J, Brismar G, Gascon G, Ozand P. Canavan disease: CT
Wysocki SJ, Hähnel R. 3-Hydroxy-3-methylglutaryl-coenzyme and MR imaging of the brain. AJNR Am J Neuroradiol 1990;
A lyase deficiency: a review. J Inherit Metab Dis 1986; 9: 11: 805–810
225–233 Brown LW, Rorke LB, Deray MJ, Smith SB, Altman N. Psychomo-
Yalçinkaya C, Dinçer A, Gündüz E, Fiçicioğlu C, Koçer N, Aydin A. tor retardation and macrocephaly in an infant. Pediatr Neu-
MRI and MRI in HMG-CoA lyase deficiency. Pediatr Neurol rosci 1985–1986; 12: 266–271
1999; 20: 375–380 Burlina AP, Ferrari V, Divry P, Gradowska W, Jakobs C, Bennett
Zoghbi HY, Spence JE, Beaudet AL, O’Brien WE, Goodman CJ, MJ, Sewell AC, Dionisi-Vici C, Burlina AB. N-Acetylaspartyl-
Gibson KM. Atypical presentation and neuropathological glutamate in Canavan disease: an adverse effector? Eur J
studies in 3-hydroxy-3-methylglutaryl-CoA layase deficien- Pediatr 1999; 158: 406–409
cy. Ann Neurol 1986; 20: 367–369 De Coo IFM, Gabreëls FJM, Renier WO, de Pont JJHHM, Hälst
UJGM, Veerkamp JH, Trijbels JMF, Jaspar HHJ, Renkawek.
Canavan disease: neuromorphological and biochemical
analysis of a brain biopsy specimen.Clin Neuropathol 1991;
42 Canavan Disease 10: 73–78
Echenne B, Divry P, Vianey-Liaud C. Spongy degeneration of
Adachi M, Wallace BJ, Schneck L, Volk BW. Fine structure of the neuraxis (Canavan-Van Bogaert disease) and N-acetyl-
spongy degeneration of the central nervous system (van aspartic aciduria. Neuropediatrics 1989; 20: 70–81
Bogaert and Betrand type). J Neuropathol Exp Neurol 1966; Engelbrecht V, Scherer A, Rassek M, Witsack HJ, Mödder U. Dif-
25: 598–616 fusion-weighted MR imaging in the brain in children: find-
Adachi M,Volk BW. Protracted form of spongy degeneration of ings in the normal brain and in the brain with white matter
the central nervous system.Neurology 1968; 18:1084–1092 diseases. Radiology 2002; 222: 410–418
Adachi M,Torii J, Schneck L, Volk BW. Electron microscopic and Gascon GG, Ozand PT, Mahdi A, Jamil A, Haider A, Brismar J,
enzyme histochemical studies of the cerebellum in spongy Al-Nasser M. Infantile CNS spongy degeneration–14 cases:
degeneration. Acta Neuropathol (Berl) 1972; 20: 22–31 clinical update. Neurology 1990; 40: 1876–1882
References and Further Reading 975

Gordon N. Canavan disease: a review of recent developments. Matalon R, Kaul R, Gao GP, Michals K, Gray RGF, Bennett-Briton
Eur J Paediatr Neurol 2000; 5: 65–69 S,Norman A,Smith M,Jakobs C.Prenatal diagnosis for cana-
Grod W, Krägeloh-Mann I, Petersen D,Trefz FK, Harzer K. In vivo van disease: the use of DNA markers. J Inherit Metab Dis
assessment of N-acetylaspartate in brain in spongy degen- 1995; 18: 215–217
eration (Canavan’s disease) by proton spectroscopy. Lancet Matalon R, Rady PL, Platt KA, Skinner HB, Quast MJ, Campbell
1990; 336: 437–438 GA, Metalon K, Ceci JD,Tyring SK, Nehls M, Surendran S,Wei
Hagenfeldt L, Bollgren I, Venizelos N. N-Acetylaspartic aciduria J, Ezell EL, Szucs S. Knock-out mouse for Canavan disease: a
due to aspartoacylase deficiency – a new aetiology of model for gene transfer tot the central nervous system.
childhood leucodystrophy. J Inherit Metab Dis 1987; 10: J Gene Med 2000; 2: 165–175
135–141 Meyding-Lamadé U, Sartor K. Magnetresonanztomographie
Hamaguchi H, Nihei K, Nakamoto N, Ezoe T, Naito H, Hara M, bei neurodegenerativen Erkrankungen im Kindesalter. Klin
Yokota K, Inoue Y, Matsumoto I. A case of Canavan disease: Neuroradiol 1993; 3: 52–61
the first biochemically proven case in a Japanese girl. Brain Toft PB, Geiß-Holtorff R, Roland MO, Pryd SO, Mueller-Forell W,
Dev 1993; 15: 367–371 Christensen E,Lehnert W,Lou HC,Ott D,Hennig J,Henriksen
Janson C, McPhee S, Bilaniuk L, Haselgrove J,Testaiuti M, Freese O. Magnetic resonance imaging in juvenile Canavan dis-
A, Wang D-J, Shera D, Hurh P, Rupin J, Saslow E, Goldfarb O, ease. Eur J Pediatr 1993; 152: 750–753
Goldberg M, Larijani G, Sharrar W, Liouterman L, Camp A, Topçu M, Erdem G, Saatçi I, Aktan G, Şimşek A A, Renda Y, Schut-
Kolodny E, Samulski J, Leone P. Gene therapy of Canavan gens RBH,Wanders RJA,Jacobs C.Clinical and magnetic res-
disease: AAV-2 vector for neurosurgical delivery of aspar- onance imaging features of L-2-hydroxyglutaric academia:
toacylase gene (ASPA) to the human brain. Hum Gene Ther report of three cases in comparison with Canavan disease.
2002; 13: 1391–1412 J Child Neurol 1996; 11: 373–377
Kamoshita S, Rapin I, Suzuki K, Suzuki K. Spongy degeneration Traeger E, Rapin I.The clinical course of Canavan disease. Pedi-
of the brain. Neurology 1968; 18: 975–985 atr Neurol 1998; 18: 207–212
Kaul R, Gao GP, Balamurugan K, Matalon R. Cloning of the hu- Van Bogaert L, Bertrand I. Les leucodystrophies progressives
man aspartoacylase cDNA and a predominant missense familiales. Rev Neurol 1933; 2: 249–286
mutation in Canavan disease. Nat Genet 1993; 5: 118–123 Wittsack HJ, Kugel H, Roth B, Heindel W. Quantitative measure-
Kendall BE. Disorders of lysosomes, peroxisomes, and mito- ments with localized 1H MR spectroscopy in children with
chondria. AJNR Am J Neuroradiol 1992; 13: 621–653. Canava’s disease. J Magn Reson Imaging 1996; 6: 889–893
Kirmani BF,Jocobowitz DM,Kallarakal AT,Namboodiri MAA.As- Zafeiriou D, Kleijer WJ, Maropoulos G, Anastasiou AL, Augousti-
partoacylase is restricted primarily to myelin synthesizing dou-Savvopoulou P, Papadopoulou F, Kontopuolos EE,
cells in the CNS: therapeutic implications for Canavan dis- Fagan E, Payne S. Protracted course of N-acetylaspartic
ease. Mol Brain Res 2002; 107: 176–183 aciduria in two non-Jewish siblings: identical clinical and
Kvittingen EA, Guldal G, Borsting S, Skalpe IO, Stokke O, Jellum magnetic resonance imaging findings. Brain Dev 1999; 21:
E. N-acetylaspartic aciduria in a child with a progressive 205–208
cerebral atrophy. Clin Chim Acta 1986; 158: 217–227 Zelnik N, Amir N, Luder AS, Hemli JA, Elpeleg ON, Fatal A, Gross-
Leone P, Janson CG, Bilianuk L, Wang Z, Sorgi F, Huang L, Mat- Tsur V, Harel S. Protracted clinical course for patients with
alon R, Kaul R, Zeng Z, Freese A, McPhee SW, Mee E, During Canavan disease. Dev Med Child Neurol 1993; 35: 346–358
MJ.Aspartoacylase gene transfer to the mammalian central
nervous system with therapeutic implications for Canavan
disease. Ann Neurol 2000; 48: 27–38
Mahloudji M, Daneshbod K, Karjoo M. Familial spongy degen-
43 L-2-Hydroxyglutaric Aciduria
eration of the brain. Arch Neurol 1970; 22: 294–298
Marks HG, Caro PA, Wang Z, Detre JA, Bogdan AR, Gusnard DA, Aydin K, Ozmen M, Tatli B, Sencer S. Single-voxel MR spectro-
Zimmerman RA. Use of computed tomography, magnetic scopy and diffusion-weighted MRI in two patients with L-2-
resonance imaging, and localized 1H magnetic resonance hydroxyglutaric aciduria. Pediatr Radiol 2003; 33: 872–876
spectroscopy in Canavan’s disease: a case report. Ann Neu- Barbot C, Fineza I, Diogo L, Maia M, Melo J, Guimarães A, Pires
rol 1991;30: 106–110 MM, Cardoso ML, Vilarinho L. L-2-Hydroxyglutaric aciduria:
Matalon R, Michals-Matalon K. Prenatal diagnosis of Canavan clinical biochemical and magnetic resonance imaging in
disease. Prenat Diagn 1999; 19: 669–670 six Portuguese pediatric patients. Brain Dev 1997; 19: 268–
Matalon R, Michals-Matalon K. Spongy degeneration of the 273
brain, Canavan disease: biochemical and molecular find- Barth PG, Hoffmann GF, Jaeken J, Lehnert W, Hanefeld F, van
ings. Pediatr Pathol Mol Med 2000; 18: 471–481 Gennip AH, Duran M, Valk J, Schutgens RBH, Trefz FK,
Matalon R, Michals K, Sebesta D, Deanching M, Gashkoff P, Reimann G, Hartung HP. L-2-Hydroxyglutaric acidemia: a
Casanova J.Aspartoacylase deficiency and N-acetylaspartic novel inherited neurometabolic disease. Ann Neurol 1992;
aciduria in patients with Canavan disease. Am J Med Genet 32: 66–71
1988; 29: 463–471 Barth PG, Hoffmann GF, Jaeken J, Wanders RJA, Duran M,
Matalon R, Kaul R, Casanova J, Michals K, Johnson A, Rapin I, Jansen GA, Jakobs C, Lehnert W, Hanefeld F, Valk J, Schut-
Gashkoff P, Deanching M. Aspartoacylase deficiency: the gens RBH, Trefz FK, Hartung HP, Chamoles NA, Sfaello Z,
enzyme defect in Canavan disease. J Inherit Metab Dis Caruso U. L-2-Hydroxyglutaric acidaemia: clinical and bio-
1989; 12: 329–331 chemical findings in 12 patients and preliminary report on
L-2-hydroxyacid dehydrogenase. J Inherit Metab Dis 1993;
Matalon R, Kaul R, Michals K. Canavan disease: biochemical and
molecular studies. J Inherit Metab Dis 1993; 16: 744–752 16: 753–761
Matalon R, Michals K, Kaul R. Canavan disease: from spongy Barth PG,Wanders RJA, Scholte HR, Abeling N, Jakobs C, Schut-
degeneration to molecular analysis. J Pediatr 1995; 127: gens RBH, Vreken P. L-2-Hydroxyglutaric aciduria and lactic
511–517 acidosis. J Inherit Metab Dis 1998; 21: 251–254
976 References and Further Reading

Chen E, Nyhan WL, Jakobs C, Greco CM, Barkovich AJ, Cox VA, Topçu M, Erdem G, Saatçi I, Aktan G, Şimşek A A, Renda Y, Schut-
Packman P. L-2-Hydroxyglutaric aciduria: neuropathologi- gens RBH,Wanders RJA,Jacobs C.Clinical and magnetic res-
cal correlations and first report of severe neurodegenera- onance imaging features of L-2-hydroxyglutaric aciduria
tive disease and neonatal death. J Inherit Metab Dis 1996; academia: report of three cases in comparison with Cana-
19: 335–343 van disease. J Child Neurol 1996; 11: 373–377
Clerc C,Bataillard M,Richard P,Divry P,Kreahenbuhl J,Rumbach Topçu M, Jobard F, Halliez S, Coskun T, Yalçinkayal C, Gerceker
L. An adult form of L-2-hydroxyglutaric aciduria revealed by FO, Wanders RJA, Prud’homme JF, Lathrop M, Özguc M, Fis-
tremor. Eur Neurol 2000; 43: 119–120 cher J. L-2-Hydroxyglutaric aciduria: identification of a mu-
De Klerk JBC, Huijmans JGM, Stroink H, Robben SGF, Jakobs C, tant gene C14orf160, localized on chromosome 14q22.1.
Duran M. L-2-Hydroxyglutaric aciduria: clinical heterogene- Hum Mol Genet 2004; 13: 2803–2811
ity versus biochemical homogeneity in a sibship. Neurope- Wanders RJA, Vilarinho L, Hartung HP, Hoffmann GF, Mooijer
diatrics 1997; 28: 314–317 PAW, Jansen GA, Huijmans JGM, de Klerk JBC, ten Brink HJ,
D’Incenti, Farina L, Moroni I, Uziel G, Savoiardo M. L-2-Hydrox- Jakobs C, Duran M. L-2-Hydroxyglutaric aciduria: normal L-
yglutaric aciduria: MRI in seven cases. Neuropathology 2-hydroxyglutarate dehydrogenase activity in liver from
1998; 40: 727–733 two new patients. J Inherit Metab Dis 1997; 20: 725–726
Diogo L, Fineza I, Canha J, Borges L, Cardoso ML, Vilarinho L. Wilcken B, Pitt J, Heath F,Walsh P,Wilson G, Bachanan N. L-2-Hy-
Macrocephaly as the presenting feature of L-2-hydroxyglu- droxyglutaric aciduria: three Australian cases. J Inherit
taric aciduria in a 5-month-old-boy. J Inherit Metab Dis Metab Dis 1993; 16: 501–504
1996; 19: 369–370 Zafeiriou DI, Sewell A, Savvopoulou-Augoustidou P, Gombakis
Divry P, Jakobs C, Vianey-Saban C, Gibson KM, Michelakakis H, N, Katzos G. L-2-Hydroxyglutaric aciduria presenting as a
Papadimitriou A, Divari R, Chabrol B, Cournelle MA, Livet status epilepticus. Brain Dev 2001; 23: 255–257
MO. L-2-Hydroxyglutaric aciduria: two further cases. J Inher-
it Metab Dis 1993; 16: 505–507
Duran M,Kamerling JP,Bakker HD,van Gennip AH,Wadman SK.
L-2-Hydroxyglutaric aciduria: an inborn error of metabo-
44 D-2-Hydroxy Glutaric Aciduria
lism. J Inherit Metab Dis 1980: 3:109–112
Fujitake J, Ishikawa Y, Fujii H, Nishimura K, Inoue F, Terada N, Achouri Y, Noël G,Vertommen D, Rider MH,Veiga-Da-Cunha M,
Okochi M, Tatsuoka Y. L-2-Hydroxyglutaric aciduria: two Van Schaftingen E. Identification of a dehydrogenase act-
Japanese adult cases in one family. J Neurol 1999; 246: ing on D-2-hydroxyglutarate. Biochem J 2004; 381: 35-42
378–382 Amiel J, de Lonay P, Francannet C, Picard A, Bruel H, Rabier D, Le
Hoffmann GF, Jakobs C, Holmes B, Mitchell L, Becker G, Hartung Merrer M, Verhoeven N, Jakobs C, Lyonnet S Munnich A.
H-P, Nyhan WL. Organic acids in cerebrospinal fluid and Facial anomalies in D-2-hydroxyglutaric aciduria. Am J Med
plasma of patients with L-2-hydroxyglutaric aciduria. J In- Genet 1999; 86: 124–129
herit Metab Dis 1995; 18: 189–193 Baker NS, Sarnat HB, Jack RM, Patterson K, Shaw DW, Herndon
Jansen GA, Wanders RJA. L-2-Hydroxyglutarate dehydroge- SP. D-2-hydroxyglutaric aciduria: hypertonia, cortical blind-
nase:identification of a novel enzyme activity in rat and hu- ness, seizures, cardiomyopathy and cylindrical spirals in
man liver. Implications for L-2-hydroxyglutaric aciduria. skeletal muscle. J Child Neurol 1997; 12: 31–36
Biochim Biophys Acta 1993; 1225: 53–56 Chalmers RA, Lawson AM, Watts WE, Tavill AS. D-2-Hydroxyglu-
Kaabachi N, Larnaout A, Rabier D, Jakobs C, Belal S, Hentati F, taric aciduria: case report and biochemical studies. J Inher-
Parvey P, Bardet J, Ben Hamida M, Mebazaa A, Kamoun P. it Metab Dis 1980; 3: 11–15
Familial encephalopathy and L-2-hydroxyglutaric aciduria. Clarcke NF,Andrews I,Carpenter K,Jakobs C,van der Knaap MS,
J Inherit Metab Dis 1993; 16: 893 Kirk EP. D-2-Hydroxyglutaric aciduria: a case with an inter-
Kossoff EH, Keswani SC, Raymond GV. L-2-Hydroxyglutaric mediate phenotype and prenatal diagnosis of two affected
aciduria presenting as migraine. Neurology 2001; 57: 1731– fetuses. Am J Med Genet 2003; 120A: 523–527
1732 Craigen WJ, Jakobs C, Sekul EA, Levy ML, Gibson KM, Butler IJ,
Larnaout A, Hentati F, Belal S, Ben Hamida C, Kaabachi N, Ben Herman GE. D-2-Hydroxyglutaric aciduria in neonate with
Hamida M. Clinical and pathological study of three Tunisian seizures and CNS dysfunction. Pediatr Neurol 1994; 10:
siblings with L-2-hydroxyglutaric aciduria. Acta Neu- 49–53
ropathol (Berl) 1994; 88: 367–370 Da Silva CG, Ribeiro CAJ, Leipnitz G, Duntra-Filho CS,Wyse ATS,
Moroni I, D’Incerti L, Farina L, Rimoldi M, Uziel G. Clinical, bio- Wannmacher CMD, Sarkis JJF, Jakobs C,Wajner M.Inhibition
chemical and neuroradiological findings in L-2-hydroxyglu- of cytochrome c oxidase activity in rat cerebral cortex and
taric aciduria. Neurol Sci 2000; 21: 103–108 human skeletal muscle by D-2-hydroxyglutaric acid in vitro.
Moroni I, Bugiani M, D’Incerti L, Maccagnano C, Rimoldi M, Bis- Biochim Biophys Acta 2002; 1586: 81–91
sola L, Pollo B, Finocchiaro G, Uziel G. L-2-Hydroxyglutaric Eeg-Olofsson O, Zhang WW, Olsson Y, Jagell S, Hagenfeldt L.
D-2-Hydroxyglutaric aciduria with cerebral, vascular, and
aciduria and brain malignant tumors. A predisposing con-
dition? Neurology 2004; 62: 1882–1884 muscular abnormalities in a 14-year-old boy. J Child Neurol
Rzem R, Veiga-da-Cunha M, Noël G, Goffette S, Nassogne MC, 2000; 15: 488–492
Tabarki B, Schöller C, Marquardt T, Vikkula M, Van Schaftin- Geerts Y, Renier WO, Bakkeren J, de Jong J. 2-Hydroxyglutaric
gen E. A gene encoding a putative FAD-dependent L-2-hy- aciduria:a case report on an infant with the D-isomeric form
droxyglutarate dehydrogenase is mutated in L-2-hydroxy- with review of the literature. J Neurol Sci 1996; 143:
glutaric aciduria. Proc Natl Acad Sci USA 2004; 101: 166–169
16849–16854 Gibson KM, Craigen W, Herman GE, Jakobs C. D-2-Hydroxyglu-
Sztriha L, Gururaj A, Vreken P, Nork M, Lestringant GG. L-2-Hy- taric aciduria in a newborn with neurological abnormali-
droxyglutaric aciduria in two siblings. Pediatr Neurol 2002; ties: a new neurometabolic disorder? J Inherit Metab Dis
27: 141–144 1993; 16: 497–500
References and Further Reading 977

Gibson KM, ten Brink HJ, Schor SM, Kok RM, Bootsma AH, Hoff- 45 Hyperhomocysteinemias
mann GF, Jakobs C. Stable isotope dilution analysis of
D- and L-2-hydroxyglutaric acid: application to the detec-
Defects in the Transsulfuration Pathway
tion and prenatal diagnosis of D- and L-2-hydroxyglutaric
acidemias. Pediatr Res 1993; 34: 277–280
Kwong KL, Mak T, Fong CM, Poon KH, Wong SN, So KT. D-2-Hy- Abeling NGGM, van Gennip AH, Blom H, Wevers RA, Vreken P,
droxyglutaric aciduria and subdural haemorrhage. Acta van Tinteren HLG, Bakker HD. Rapid diagnosis and methio-
Paediatr 2002; 91: 716–718 nine administration: basis for a favourable outcome in a pa-
Muntau AC, Röschinger W, Merkenschlager A, van der Knaap tient with methylene tetrahydrofolate reductase deficien-
MS, Jakobs C, Duran M, Hoffmann GF, Roscher AA. Com- cy. J Inherit Metab Dis 1999; 22: 240–242
bined D-2- and L-2-hydroxyglutaric aciduria with neonatal Al-Essa MA, Al Amir A, Rashed M, Al Jishi E, Abutaleb A,
onset encephalopathy: a third biochemical variant of 2-hy- Mobaireek K, Shin YS, Ozand PT. Clinical, flurorine-18 la-
droxyglutaric aciduria? Neuropediatrics 2000; 31: 137–140 beled 2-fluoro-2-deoxyglucose positron emission tomo-
Nyhan WL, Shelton D, Jakobs C, Holmes B, Bowe C, Curry CJR, graphy of the brain, MR spectroscopy, and therapeutic at-
Vance C, Duran M, Sweetman L. D-2-Hydroxyglutaric tempts in methylenetetrahydrofolate reductase deficiency.
aciduria. J Child Neurol 1995; 10: 137–142 Brain Dev 1999; 21: 345–349
Struys EA, Verhoeven NM, Roos B, Jakobs C. Disease-related Baethmann M, Wendel U, Hoffmann GF, Göhlich-Ratmann G,
metabolites in culture medium of fibroblasts from pa- Kleinlein B, Seiffert P, Blom H,Voit T. Hydrocephalus internus
tients with D-hydroxyglutaric aciduria, L-2-hydroxyglutaric in two patients with 5,10-mehylenetetrahydrofolate reduc-
aciduria and combined D-/L-2-hydroxyglutaric aciduria. tase deficiency. Neuropediatrics 2000; 31: 314–317
Clin Chem 2003; 49: 1133–1138 Beckman DR, Hoganson G, Berlow S, Gilbert EF. Pathological
Struys EA,Verhoeven NM, Brunengraber H, Jakobs C. Investiga- findings in 5,10-methylene tetrahydrofolate reductase de-
tions by mass isotopomer analysis of the formation of D-2- ficiency. Birth Defects 1987; 23: 47–64
hydroxyglutarate by cultured lymphoblasts from two pa- Beradelli A,Thompson PD, Zaccagnini M, Giardini O, D’Eufemia
tients with D-2-hydroxyglutaric aciduria. FEBS Lett 2004; P, Massoud R, Manfredi M.Two sisters with generalized dys-
557: 115–120 tonia associated with homocystinuria. Mov Disord 1991; 6:
Struys EA, Salomons GS, Achouri Y, van Schaftingen E, Grosso S, 163–165
Craigen WJ, Verhoeven NM, Jakobs C. Mutations in the D-2- Carson NAJ, Dent CE, Field CMB, Gaull GE. Homocystinuria.
hydroxyglutarate dehydrogenase gene cause D-2-hydrox- J Pediatr 1965; 66: 565–583
yglutaric aciduria. Am J Hum Genet 2005; 76 358–360 Chou SM, Waisman HA. Spongy degeneration of the central
Sugita K, Kakinuma H, Okajima Y, Ogawa A, Watanabe H, Niimi nervous system. Arch Pathol Lab Med 1965; 79: 357–363
H. Clinical and MRI findings in a case of D-2-hydroxyglutaric Clayton PT, Smith I, Harding B, Hyland K, Leonard JV, Leeming
aciduria. Brain Dev 1995; 17: 139–141 RJ. Subacute combined degeneration of the cord, demen-
van der Knaap MS, Jakobs C, Hoffmann GF, Nyhan WL, Renier tia and Parkinsonism due to an inborn error of folate me-
WO, Smeitink JAM, Catsman-Berrevoets CE, Hjalmarson O, tabolism. J Neurol Neurosurg Psychiatry 1986; 49: 920–927
Vallance H, Sugita K, Bowe CM, Herrin JT, Caigen WJ, Buist Dunn HG, Perry TL, Dolman CL. Homocystinuria. A recently
NRM, Brookfield DSK, Cahlmers RA. D-2-Hydroxyglutaric discovered cause of mental defect and cerebrovascular
aciduria: biochemical marker or clinical disease entity? Ann thrombosis. Neurology 1966; 16: 407–420
Neurol 1999a; 45: 111–119 Engelbrecht V,Rassek M,Huismann J,Wendel U.MR and proton
van der Knaap MS, Jakobs C, Hoffmann GF, Duran M, Muntau MR spectroscopy of the brain in hyperhomocysteinemia
AC, Schweitzer S, Kelley RI, Parrot-Rouland F, Amiel J, De caused by methylenetetrahydrofolate reductase deficien-
Lonay P, Rabier D, Eeg-Olofsson O. D-2-Hydroxyglutaric cy. AJNR Am J Neuroradiol 1997; 18: 536–539
aciduria: further clinical delineation. J Inherit Metab Dis Fattal-Valevski A, Bassan H, Korman SH, Lerman-Sagie T, Gut-
1999b; 22: 404–413 man A, Harel S. Methylenetetrahydrofolate reductase defi-
Wagner L, Hoffmann GF, Jakobs C. D-2-Hydroxyglutaric ciency: importance of early diagnosis. J Child Neurol 2000;
aciduria: evidence of clinical and biochemical heterogene- 15: 539–543
ity. J Inherit Metab Dis 1998; 21: 247–250 Fowler B. Disorders of homocysteine metabolism. J Inherit
Wajner M, Vargas CR, Funayama C, Fernandez A, Elias MLC, Metab Dis 1997; 20: 270–285
Goodman SI, Jakobs C, van der Knaap MS. D-2-Hydroxyglu- Gerritsen T, Waisman HA. Homocystinuria, an error in the me-
taric aciduria in a patient with a severe clinical phenotype tabolism of methionine. Pediatrics 1964; 33: 413–420
and unusual MRI findings. J Inherit Metab Dis 2002; 25: Haworth JC, Dilling LA, Surtees RAH, Seargeant LE, Shing HL,
28–34 Cooper BA, Rosenblatt DS. Symptomatic and asympto-
Wanders RJA, Mooyer P. D-2-Hydroxyglutaric aciduria: identifi- matic methylenetetrahydrofolate reductase deficiency in
cation of a new enzyme, D-2-hydroxyglutarate dehydroge- two adult brothers. Am J Med Genet 1993; 45: 572–576
nase, localized in mitochondria. J Inherit Metab Dis 1995; Hyland K, Smith I, Bottiglieri T, Perry J, Wendel U, Clayton PT,
18: 194–196 Leonard JV. Demyelination and decreased S-adenosylme-
thionine in 5,10-methylenetetrahydrofolate reductase de-
ficiency. Neurology 1988; 38: 459–462
Kelly PJ, Furie KL, Kistler JP, Barron M, Picard EH, Mandell R, Shih
VE. Stroke in young patients with hyperhomocsyteinemia
due to cystathionine beta-synthase deficiency. Neurology
2003; 60: 275–279
Keskin S, Yalcin E. Case report of homocystinuria: clinical, elec-
troencephalographic, and magnetic resonance imaging
findings. J Child Neurol 1994; 9: 210–211
978 References and Further Reading

Kishi T, Kawamura I, Harada Y, Eguchi T, Sakura N, Ueda K, Nari- Bellamy MF, McDowell IFW. Putative mechanisms for vascular
sawa K, Rosenblatt DS. Effect of betaine on S-adenosylme- damage by homocysteine. J Inherit Metab Dis 1997; 20:
thionine levels in the cerebrospinal fluid in a patient with 307–315
methylenetetrahydrofolate reductase deficiency and pe- Broers GHJ.The case for mild hyperhomocysteinaemia as a risk
ripheral neuropathy. J Inherit Metab Dis 1994; 17: 560–565 factor. J Inherit Metab Dis 1997; 20: 301–306
Kraus JP. Molecular basis of phenotype expression in homo- Bushnell CD, Goldstein LB. Homocysteine testing in patients
cystinuria. J Inherit Metab Dis 1994; 17: 383–390 with acute ischemic stroke. Neurology 2002; 59: 1541–1546
Lee C-C, Surtees R, Duchen LW. Distal motor axonopathy and Cottington EM, LaMantia C, Stabler SP, Allen RH, Tangerman A,
central nervous system myelin vacuolation caused by Wagner C, Zeisel SH, Mudd SH. Adverse event associated
cycloleucine, an inhibitor of methionine adenosyltrans- with methionine loading test. Vasc Biol 2002; 22: 1046–
ferase. Brain 1992; 115: 935–955 1050
Li SCH, Stewart PM. Homocystinuria and psychiatric disorder: Diaz-Arrastia R. Homocysteine and neurologic disease. Arch
a case report. Pathology 1999; 31: 221–224 Neurol 2000; 57: 1422–1428
Ludolph AC, Ullrich K, Bick U, Fahrendorf G, Pzyrembel H. Func- Dufouil C, Alpérovitch A, Ducros V, Tzourio C. Homocysteine,
tional and morphological deficits in late-treated patients white matter hyperintensities, and cognition in healthy
with homocystinuria: a clinical, electrophysiologic and MRI elder people. Ann Neurol 2003; 53: 214–221
study. Acta Neurol Scand 1991; 83: 161–165 Ganesan V, Prengler M, McShane MA, Wade AM, Kirkham FJ.
Nishimura M, Yoshino K, Tomita Y, Takashima S, Tanaka J, Nari- Investigation of risk factors in children with arterial is-
sawa K, Kurobane I. Central and peripheral nervous system chemic stroke. Ann Neurol 2003; 53: 167–173
pathology of homocystinuria due to 5,10-methylenete- Hogeveen M, Blom HJ, van Amerongen M, Boogmans B, van
trahydrofolate reductase deficiency. Pediatr Neurol 1985; 1: Beynum IM, van de Bor M. Hyperhomocysteinemia as risk
375–378 factor for ischemic and hemorrhagic stroke in newborn
Ruano MM, Castillo M, Thompson JE. MR imaging in a patient infants. J Pediatr 2002; 141: 429–431
with homocystinuria. AJR Am J Roentgenol 1998; 171: Kelly PJ, Rosand J, Kistler JP, Shih VE, Silveira S, Plomaritoglu A,
1147–1149 Furie KL. Homocysteine, MTHFR 677CT polymorphism, and
Sakura N, Ono H, Nomura S, Ueda H, Fujita N. Betaine dose risk of ischemic stroke. Results of a meta-analysis. Neuro-
and treatment intervals in therapy for homocystinuria due logy 2002; 59: 529–536
to 5,10-methylenetetrahydrofolate reductase deficiency. Kim NK, Choi BO, Jung WS, Choi YJ, Choi KG. Hyperhomo-
J Inherit Metab Dis 1998; 21: 84–85 cysteinemia as an independent risk factor for silent brain
Surtees R, Leonard J, Austin S. Association of demyelination infarction. Neurology 2003; 61: 1595–1599
with deficiency of cerebrospinal-fluid-S-adenosylmethion- Kohara K, Fujisawa M, Ando F,Tabara Y, Niino N, Miki T, Shimoka-
ine in inborn errors of methyl-transfer pathway. Lancet ta H. MTHFR gene polymorphism as a risk factor for silent
1991; 338: 1550–1554 brain infarcts and white matter lesions in the Japanese
Surtees R, Heales S, Bowron A. Association of cerebrospinal fluid general population. The NILS-LSA study. Stroke 2003; 34:
deficiency of 5-methyltetrahydrofolate, but not S-adeno- 1130–1135
sylmethionine, with reduced concentrations of the acid Madonna P, de Stefano V, Coppola A, Cirillo F, Cerbone AM
metabolites of 5-hydroxytryptamine and dopamine. Clin Orefice G, Di Minno G. Hyperhomocysteinemia and other
Sci 1994; 86: 697–702 inherited prothrombotic conditions in young adults with a
Visy JM, le Coz P, Chadefaux B, Fressinaud C, Woimant F, Mar- history of ischemic stoke. Stroke 2002; 33: 51–56
quet J, Zittoun J,Visy J,Vallat JM, Haguenau M. Homocystin- Meiklejohn DJ,Vickers MA,Dijkhuisen R,Greaves M.Plasma ho-
uria due to 5,10-methylenetetrahydrofolate reductase defi- mocysteine concentrations in the acute convalescent peri-
ciency revealed by stroke in adult siblings. Neurology 1991; ods of atherothrombotic stroke. Stroke 2001; 32: 57–62
41: 1313–1315 Pezzini A, del Zotto E, Archetti S, Negrini R, Bani P, Albertini A,
Walk D, Kang SS, Horwitz A. Intermittent encephalopathy, re- Grassi M, Assanelli D, Gasparotti R, Vignolo LA, Magoni M,
versible nerve conduction slowing, and MRI evidence of Padovani A. Plasma homocysteine concentration, C677T
cerebral white matter disease in methylenetatrahydrofo- MTHFR genotype, and 844ins68 bp CBS genotype in young
late reductase deficiency. Neurology 1994; 44: 344–347 adults with spontaneous cervical artery dissection and
White HH, Rowland LP, Araki S,Thompson HL, Cowen D. Homo- atherothrombotic stroke. Stroke 2002; 33: 664–669
cystinuria. Arch Neurol 1965; 13: 455–470 Prins ND, den Heijer T, Hofman A, Koudstaal PJ, Jolles J, Clarke
Yap S. Classical homocystinuria: vascular risk and its preven- R, Breteler MMB. Homocysteine and cognitive function in
tion. J Inherit Metab Dis 2003; 26: 259–265 the elderly. The Rotterdam scan study. Neurology 2002; 59:
Yap S, Naughten E. Homocystinuria due to cystathionine 1375–1380
b-synthase deficiency in Ireland: 25 years’ experience of a Rosengarten B, Osthaus S, Auch D, Kaps M. Effects of acute
newborn screened and treated population with reference hyperhomocysteinemia on the neurovascular coupling
to clinical outcome and biochemical control. J Inherit mechanism in healthy young adults. Stroke 2003; 34:
Metab Dis 1998; 21: 738–747 446–451
Sarkar PK, Lambert LA. Aetiology and treatment of hyperho-
mocysteinaemia causing ischaemic stroke. Int J Clin Pract
Mild Hyperhomocysteinemia 2001; 55: 262–268
Tan NC-K,Venketasubramanian N, Saw S-M,Tjia HT-L.Hyperho-
Aronow WS, Ahn C, Gutstein H. Increased plasma homocys- mocyst(e)inemia and risk of ischemic stroke among young
teine is an independent predictor of new atherotrombotic Asian adults. Stoke 2002; 33: 1956–1962
brain infarction in older persons. Am J Cardiol 2000; 86: Van Beynum IM, Smeitink JAM, den Heijer M, te Poele Pothoff
585–586 MTWB,Blom HJ.Hyperhomocysteinemia.A risk factor for is-
chaemic stroke in children. Circulation 1999; 99: 2070–2072
References and Further Reading 979

Van Diemen-Steenvoorde R, van Nieuwenhuizen O, de Klerk Bianheri R, Cerone R, Schiaffino MC, Caruso U, Veneselli E, Per-
JBC, Duran M. Quasi-Moyamoya disease and heterozygosi- rone MC, Rossi A, Gatti R. Cobalamin (Cbl) C/D deficiency:
ty for homocystinuria in a five-year-old girl. Neuropedi- clinical, neurophysiological and neuroradiological findings
atrics 1990; 21: 110–112 in 14 cases. Neuropediatrics 2001; 32: 14–22
Vermeer SE, van Dijk EJ, Koudstaal PJ, Oudkerk M, Hofman A, Bibi H, Gelman-Kohan Z, Baumgartner ER, Rosenblatt DS.
Clarke R, Breteler MMB. Homocysteine, silent brain infarcts, Transcobalamin II deficiency with methylmalonic aciduria
and white matter lesions: the Rotterdam scan study. Ann in three sisters. J Inherit Metab Dis 1999; 22: 765–772
Neurol 2002; 51: 285–289 Carmel R, Watkins D, Goodman SI, Rosenblatt DS. Hereditary
Vermeulen EGJ, Stehouwer CDA, Valk J, van der Knaap MS, van defect of cobalamin metabolism (cblG mutation) present-
den Berg M,Twisk JWR,Prevoo W,Rauwerda JA.Effect of ho- ing as a neurologic disorder in adulthood. N Engl J Med
mocysteine-lowering treatment with folic acid plus vita- 1988; 318: 1738–1741
min B6 on cerebrovascular atherosclerosis and white mat- Dayan AD, Ramsey RB. An inborn error of vitamin B12 metabo-
ter abnormalities as determined by MRA and MRI: a place- lism associated with cellular deficiency of coenzyme forms
bo-controlled, randomized trial. Eur J Clin Invest 2004; 34: of the vitamin. J Neurol Sci 1974; 23: 117–128
256–261 Enns GM, Barkovich AJ, Rosenblat DS, Fredrick DR, Weisiger K,
Welch GN, Loscalzo J. Homocysteine and atherothrombosis. Ohnstad C, Packman S. Progressive neurological deteriora-
N Engl J Med 1998; 338: 1042–1050 tion and MRI changes in clbC methylmalonic acidaemia
Wilcken DEL, Wilcken B.The natural history of vascular disease treated with hydroxocobalamin. J Inherit Metab Dis 1999;
in homocystinuria and the effects of treatment. J Inherit 22: 599–607
Metab Dis 1997; 20: 295–300 Fenton WA, Rosenberg LE. Genetic and biochemical analysis of
human cobalamin mutants in cell culture. Annu Rev Genet
1978; 12: 223–248
Folate Deficiency Linnell JC, Ray Bhatt H. Inherited errors of cobalamin metabo-
lism and their management. Balliere’s Clin Haematol 1995;
Giles WH, Kittner SJ, Anda RF, Croft JB, Casper ML. Serum folate 8: 567–601
and risk factor for ischemic stroke. First national health and Mitchell GA, Watkins D, Melançon SB, Rosenblatt DS, Geoffroy
nutrition examination survey. Epidemiologic follow-up G, Orquin J, Homsy MB, Dallaire L. Clinical heterogeneity in
study. Stroke 1995; 26: 1166–1170 cobalamin C variant of combined homocystinuria and
Hoffbrand AV, Weir DG. The history of folic acid. Br J Haematol methylmalonic aciduria. J Pediatr 1986; 108: 410–415
2001; 113: 579–589 Powers JM, Rosenblatt DS, Schmidt RE, Cross AH, Black JT,
Lever EG, Elwes RDC, Williams A, Reynolds EH. Subacute com- Moser AB, Moser HW, Morgan DJ. Neurological and neu-
bined degeneration of the cord due to folate deficiency: re- ropathological heterogeneity in two brothers with cobal-
sponse to methyl folate treatment. J Neurol Neurosurg Psy- amin C deficiency. Ann Neurol 2001; 49: 396–400
chiatry 1986; 49: 1203–1207 Rossi A, Biancheri R, Tortori-Donati P. The pathogenesis of hy-
Robertson DM, Dinsdale HB, Campbell RJ, Kingston ChB. Suba- drocephalus in inborn errors of the single carbon transfer
cute combined degeneration of the spinal cord. No associ- pathway. Neuropediatrics 2001a; 32: 335–336
ation with vitamin B12 deficiency. Arch Neurol 1971: 24: Rossi A, Cerone R, Biancheri R, Gatti R, Schiaffino MC, Fonda C,
203–207 Zammarchi E, Tortori-Donati P. Early-onset combined
Scott JM, Weir DG. The methyl folate trap. Lancet 1981: 2: 337– methylmalonic aciduria and homocystinuria: neuroradio-
340 logic findings. AJNR Am J Neuroradiol 2001b; 22: 554–563
Selhub J. Folate, vitamin B12 and vitamin B6 and one carbon Schuh S, Rosenblatt DS, Cooper BA, Schroeder M-L, Bishop AJ,
metabolism. J Nutr Health Aging 2002; 6: 39–42 Seargeant LE, Haworth JC. Homocystinuria and mega-
Van der Weyden MB, Hayman RJ, Rose IS, Brumley J. Folate-de- loblastic anemia responsive to vitamin B12 therapy. An
ficient human lymphoblasts: changes in deoxynucleotide inborn error of metabolism due to a defect in cobalamin
metabolism and thymidylate cycle activities. Eur J Haema- metabolism. N Engl J Med 1984; 310: 686–690
tol 1991: 47: 109–114 Shinnar S, Singer HS. Cobalamin C mutation (methylmalonic
Weir DG, Scott JM. Brian function in the elderly: role of vitamin aciduria and homocystinuria) in adolescence. N Engl J Med
B12 and folate. Br Med Bull 1999; 55: 669–682 1984; 311: 451–454
Steen C, Rosenblat DS, Scheying H, Braeuer JC, Kohlstütter A.
Cobalamin E (clbE) disease: a severe neurological disorder
Inborn Errors of Cobalamin Metabolism with megaloblastic anaemia, homocystinuria and slow
serum mehionine. J Inherit Metab Dis 1997; 20: 705–706
Augoustides-Savvopouloou P, Mylonas I, Sewell AC, Rosenblatt Surtees R. Demyelination and inborn errors of the single car-
DS.Reversible dementia in an adolescent with clbC disease: bon transfer pathway. Eur J Pediatr 1998; 157: S118-S121
clinical heterogeneity within the same family. J Inherit Thomas PK, Hoffbrand AV. Hereditary transcobalamin II defi-
Metab Dis 1999; 22: 759–758 ciency: a 22 year follow up. J Neurol Neurosurg Psychiatry
Bartholomew DW, Batshaw ML, Allen RH, Roe CR, Rosenblatt D, 1997; 62: 197
Valle DL, Francomano CA. Therapeutic approaches to Tuchman M, Kelly P, Watkins D, Rosenblatt DS. Vitamin B12-re-
cobalamin-C methylmalonic acidemia and homocystin- sponsive megaloblastic anemia, homocystinuria, and tran-
uria. J Pediatr 1988; 112: 32–39 sient methylmalonic aciduria in cblE disease. J Pediatr
Bellini C, Cerone R, Bonacci W, Caruso U, Magliano CP, Serra G, 1988;113: 1052–1055
Fowler B, Romano C. Biochemical diagnosis and outcome Watkins D, Rosenblatt DS. Genetic heterogeneity among pa-
of 2 years’ treatment in a patient with combined methyl- tients with methylcobalamin deficiency. Definition of two
malonic aciduria and homocystinuria. Eur J Pediatr 1992; complementation groups: cblE and cblG. J Clin Invest 1988;
151: 818–820 81: 1690–1694
980 References and Further Reading

Zavad’ákoa P, Fowler B, Zeman J, Suromala T, Pøistoupilová K, Higginbottom MC, Sweetman L, Nyhan WL. A syndrome of
Kozich V. CblE type of homocystinuria due to methionine methylmalonic aciduria, homocystinuria, megalobastic
synthase reductase deficiency: clinical and molecular stud- anemia and neurologic abnormalities in a vitaman B12 defi-
ies and prenatal diagnosis in two families. J Inherit Metab cient breast-fed infant of a strict vegetarian. N Engl J Med
Dis 2002; 25: 461–476 1978; 299: 317–323
Hoffbrand AV, Jackson BFA. Correction of the DNA synthesis
defect in vitamin B12 deficiency by tetrahydrofolate: evi-
Cobalamin Deficiency dence in favour of the methyl-folate trap hypothesis as the
cause of megaloblastic anaemia in vitamin B12 deficiency.
Bassi SS, Bulundwe KK, Greeff GP, Labuscagne JH, Gledhill RF. Br J Haematol 1993; 83: 643–647
MRI of the spinal cord in myelopathy complicating vitamin Holloway KL, Alberico AM. Postoperative myeloneuropathy: a
B12 deficiency: two additional cases and a review of the lit- preventable complication in patients with B12 deficiency.
erature. Neuroradiology 1999; 41: 271–274 J Neurosurg 1990; 72: 732–736
Brattström L, Israelsson B, Lindgärde F, Hultberg B. Higher total Karnaze DS, Carmel R. Neurologic and evoked potential abnor-
plasma homocysteine in vitamin B12 deficiency than in malities in subtle cobalamin deficiency states, including
heterozygosity for homocystinuria due to cystathionine deficiency without anemia and with normal absorption of
b-synthase deficiency. Metabolism 1988;37: 175–178 free cobalamin. Arch Neurol 1990; 47: 1008–1012
Carmel R. Pernicious anemia. Arch Intern Med 1988; 148: Katsaros VK, Glocker FX, Hemmer B, Schumacher M. MRI of
1712–1714 spinal cord and brain lesions in subacute combined degen-
Chatterjee A,Yapundich R, Palmer CA, Marson DC, Mitchell GW. eration. Neuroradiology 1998; 40: 716–719
Leukoencephalopathy associated with cobalamin defi- Kealey SM, Provenzale JM. Tensor diffusion imaging in B12
ciency. Neurology 1996; 16: 832–834 leukoencephalopathy. J Comput Assist Tomogr 2002; 26:
Clementz CL, Schade SG.The spectrum of vitamin B12 deficien- 952–955
cy. Am Fam Physician 1999; 41:150–162 Kühne T, Bubl R, Baumgartner R. Maternal vegan diet causing a
Duprez TP, Gille M, vande Berg BC, Malghem J, Grandin CB, serious infantile neurological disorder due to vitamin B12
Michel P, Ghariani S, Maldague BE. MRI of the spine in cobal- deficiency. Eur J Pediatr 1991; 150: 205–208
amin deficiency: the value of examining both spinal cord Küker W, Hesselmann V, de Simone A. MRI demonstration of re-
and bone marrow. Neuroradiology 1996; 38: 511–515 versible impairment of the blood-CNS barrier function in
Fine EJ, Soria E, Paroski MW, Petryk D, Thomasula L. The neuro- subacute combined degeneration of the spinal cord.J Neu-
physiological profile of vitamin B12 deficiency. Muscle rol Neurosurg Psychiatry 1997; 62: 298–299
Nerve 1990; 13: 158–164 Lindenbaum J, Healton EB, Savage DG, Brust JCM, Garrett TJ,
Flippo TS, Holder WD. Neurologic degeneration associated Podell ER, Marcell PD, Stabler SP, Allen RH. Neuropsychiatric
with nitrous oxide anesthesia in patients with vitamin B12 disorders caused by cobalamin deficiency in the absence
deficiency. Arch Surg 1993; 128: 1391–1395 of anemia or macrocytosis. N Engl J Med 1988; 318:
Garewal G, Narang A, Das KC. Infantile tremor syndrome: a vit- 1720–1728
amin B12 deficiency syndrome in infants. J Trop Pediatr Lövblad K-O, Ramelli G, Remonda L, Nirkko AC, Ozdoba C,
1988; 34: 174–178 Schroth G. Retardation of myelination due to dietary vita-
Gilois C,Wierzbicki AS, Hirani N, Norman PM, Jones SJ, Ponsford min B12 deficiency: cranial MRI findings. Pediatr Radiol
S, Alani SM, Kriss A. The hematological and electrophysio- 1997; 27: 155–158
logical effects of cobalamin. Deficiency secondairy to veg- Morita S, Miwa H, Kihira T, Kondo T. Cerebellar ataxia and
etarian diets. Ann N Y Acad Sci 1992; 669: 345–348 leukoencephalopathy associated with cobalamin deficien-
Graham SM, Arvela OM,Wise GA. Long-term neurologic conse- cy. J Neurol Sci 2003; 216: 183–184
quences of nutritional vitamin B12 deficiency in infants. Murata S, Naritomi H, Sawada T. MRI in subacute combined de-
J Pediatr 1992; 121: 710–714 generation. Neuroradiology 1994; 36: 408–409
Grattan-Smith PJ, Wilcken B, Procopis PG, Wise GA. The neuro- Narayanan MN, Dawson DW, Lewis MJ. Dietary deficiency of
logical syndrome of infantile cobalamin deficiency: devel- vitamin B12 is associated with low serum cobalamin levels
opmental regression and involuntary movements. Mov in non-vegetarians. Eur J Haematol 1991; 47: 115–118
Disord 1997; 12: 39–46 Perry J, Chanarin I, Deacon R, Lumb M. Methylation of DNA in
Green R,Kinsella LJ.Current concepts in the diagnosis of cobal- megaloblastic anaemia. J Clin Pathol 1990; 43: 211–212
amin deficiency. Neurology 1995; 45: 1435–1440 Rasmussen SA, Fernhoff PM, Scanlon KS. Vitamine B12 defi-
Healton EB, Savage DG, Brust JCM, Garrett TJ, Lindenbaum J. ciency in children and adolescents. J Pediatr 2001; 138:
Neurologic aspects of cobalamin deficiency. Medicine 10–17
1991; 70: 229–245 Reynolds EH, Bottiglieri T, Laundy M, Stern J, Payan J, Linnell J,
Heckmann JG, Lang CJG, Ganslandt O, Tomandl B, Neundörfer Faludy J. Subacute combined degeneration with high
B. Reversible leukoencephalopathy due to vitamin B12 de- serum vitamin B12 level and abnormal vitamin B12 binding
ficiency in an acromegalic patient. J Neurol 2003; 250: protein. Arch Neurol 1993;50: 739–742
366–368 Saracaceanu E, Tramoni AV, Henry JM. An association between
Hector M,Burton JR.What are the psychiatric manifestations of subcortical dementia and pernicious anemia. A psychiatric
vitamin B12 deficiency? J Am Geriatr Soc 1988; 36: 1105– mask. Compr Psychiatry 1997; 38: 349–351
1112 Scott JM, Wilson P, Dinn JJ, Weir DG. Pathogenesis of subacute
Hemmer B, Glocker FX, Schumacher M, Deuschl G, Lücking CH. combined degeneration: a result of methyl group deficien-
Subacute combined deterioration: clinical, electrophysio- cy. Lancet 1981; 2: 334–337
logical, and magnetic resonance imaging findings. J Neurol Shevell MI, Rosenblatt DS. The neurology of cobalamin. Can J
Neurosurg Psychiatry 1998; 65: 822–827 Neurol Sci 1992; 19: 472–486
References and Further Reading 981

Soria ED, Fine EJ. Somatosensory evoked potentials in the neu- Christodoulou J, Qureshi IA, McInnes RR, Clarke JTR. Ornithine
rological sequelae of treated vitamin B12 deficiency. Elec- transcarbamylase deficiency presenting with strokelike
tromyogr Clin Neurophysiol 1992; 32: 63–71 episodes. J Pediatr 1993; 122: 423–425
Stabler SP, Allen RH, Savage DG, Lindenbaum J. Clinical spec- Connelly A, Cross JH, Gadian DG, Hunter JV, Kirkham FJ,
trum and diagnosis of cobalamin deficiency. Blood 1990; Leonard JV. Magnetic resonance spectroscopy shows in-
76: 871–881 creased brain glutamine in ornithine carbamoyl trans-
Steiner I, Kidron D, Soffer D, Wirguin I, Abramsky O. Sensory ferase deficiency. Pediatr Res 1993; 33: 77–81
peripheral neuropathy of vitamin B12 deficiency: a primary Dolman CL, Clasen RA, Dorovini-Zis K. Severe cerebral damage
demyelinating disease? J Neurol 1988; 235: 163–164 in ornithine transcarbamylase deficiency. Clin Neuropathol
Stojsavlejeviæ N, Leviæ Z, Druloviæ J, Dragutinoviæ G. A 44- 1988; 7: 10–15
month clinical-brain MRI follow-up in a patient with B12 Donn SM, Thoene JG. Prospective prevention of neonatal hy-
deficiency. Neurology 1997; 49: 878–881 perammonaemia in argininosuccinic acidura by arginine
Surtees R. Biochemical pathogenesis of subacute combined therapy. J Inherit Metab Dis 1985; 8: 18–20
degeneration of the spinal cord and brain. J Inherit Metab Finkelstein JE, Hauser ER, Leonard CO, Brusilow SW. Late-onset
Dis 1993; 16: 762–770 ornithine transcarbamylase deficiency in male patients.
Timms SR, Curé JK, Kurent JE. Subacute combined degenera- J Pediatr 1990; 117: 897–902
tion of the spinal cord: MR findings. AJNR Am J Neuroradiol Gallagher JV, Rifai N, Conry J, Soldin SJ. Role of the clinical lab-
1993; 14: 1224–1227 oratory in evaluation of argininosuccinate lyase deficiency.
Ubbink JB. The role of vitamins in the pathogenesis and treat- Clin Chem 1991; 37: 1384–1389
ment of hyperhomocyst(e)inaemia. J Inherit Metab Dis Gerrits GPJM, Gabreëls FJM, Monnens LAH, De Abreu RA, van
1997; 20: 316–325 Raaij-Selten B, Niezen-Koning KE, Trijbels JMF. Arginiosuc-
Yamada K, Shrier DA, Tanaka H, Numaguchi Y. A case of suba- cinic aciduria: clinical and biochemical findings in three
cute combined degeneration: MRI findings. Neuroradiolo- children with the late onset form, with special emphasis on
gy 1998; 40: 398–400 cerebrospinal fluid findings of amino acids and pyrim-
Zegers de Beyl D, Delecluse F, Veranck P, Borestein S, Capel P, idines. Neuropediatrics 1993; 21: 15–18
Brunko E. Somatosensory conduction in vitamin B12 defi- Grody WW, Kern RM, Klein D, Dodson AE, Wissman PB, Barsky
ciency. Electroencephalogr Clin Neurophysiol 1988; 69: SH, Cederbaum SD. Arginase deficiency manifesting de-
313–318 layed clinical sequelae and induction of a kidney arginase
isozyme. Hum Genet 1993; 91: 1–5
Grompe M, Caskey CT, Fenwick RG. Improved molecular diag-
nostics for ornithine transcarbamylase deficiency. Am J
46 Urea Cycle Defects Hum Genet 1991; 48: 212–222
Harding BN, Leonard JV, Erdohazi M. Ornithine carbamoyl
Aida S, Ogata T, Kamota T, Nakamura N. Primary ornithine tran- transferase deficiency: a neuropathological study. Eur J Pe-
scarbamylase deficiency. Acta Pathol Jpn 1989; 39: 451–456 diatr 1984; 141: 215–220
Albayram S, Murphy KJ, Gailloud P, Moghekar A, Brunberg JA. Hommes FA, de Groot CJ, Wilmink CW, Jonxis JHP. Carb-
CT findings in the infantile form of citrullinemia. AJNR Am J amylphosphate synthetase deficiency in an infant with se-
Neuroradiol 2002; 23: 334–336 vere cerebral damage. Arch Dis Childh 1969; 44: 688–693
Bachmann C.Ornithine carbamoyl transferase deficiency: find- Honeycutt D, Callahan K, Rutledge L, Evans B. Heterozygote or-
ings, models and problems. J Inherit Metab Dis 1992;15: nithine transcarbamylase deficiency presenting as sympto-
578–591 matic hyperammonemia during initiation of valproate
Bajaj SK, Kurlemann G, Schuierer G, Peters PE. CT and MRI find- therapy. Neurology 1992; 42: 666–668
ings in a girl with late-onset ornithine transcarbamylase Horiuchi M, Imamura Y, Nakamura N, Maruyama I, Saheki T. Car-
deficiency: case report. Neuroradiology 1996; 38: 796–799 bamoylphosphate synthetase deficiency in an adult: dete-
Batshaw ML. Inborn errors of urea synthesis. Ann Neurol 1994; rioration due to administration of valproic acid. J Inherit
35: 133–141 Metab Dis 1993;16: 39–45
Brockstedt M, Smit LME, de Grauw AJC, van der Klei-van Kleijer WJ,Garritsen VH,Linnebank M,Mooyer P,Huijmans JGM,
Moorsel JM, Jakobs C. A new case of hyperargininaemia: Mustonen A, Simola KOJ, Arslan-Kirchner M, Battini R,
neurological and biochemical findings prior to and during Briones P,Cardo E,Mandel H,Tschiedel E,Wanders RJA, Koch
dietary treatment. Eur J Pediatr 1990; 149: 341–343 HG. Clinical, enzymatic, and molecular genetic characteri-
Brusilow SW. Arginine, an indespensible amino acid for pa- zation of a biochemical variant type of argininosuccinic
tients with inborn errors of urea synthesis. J Clin Invest aciduria: prenatal and postnatal diagnosis in five unrelated
1984; 74: 2144–2148 families. J Inherit Metab Dis 2002; 25: 399–410
Bruton CJ, Corsellis JAN, Russell A. Hereditary hyperammon- Kornfeld M, Woodfin BM, Papile L, Davis LE, Bernard LR. Neu-
aemia. Brain 1970; 93: 423–434 ropathology of ornithine carbamyl transferase deficiency.
Cerone R, Caruso U, Barabino A. Gatt R, Jakobs C, Jacquemyn I, Acta Neuropathol (Berl) 1985; 65: 261–264
Marescau B, De Deyn PP. Hyperargininemia: pre-natal diag- Kurihara A, Takanashi J-i, Tomita M, Kobayashi K, Ogawa A,
nosis and treatment from birth. In: De Deyn PP, Marescau B, Kanazawa M, Yamamoto S, Kohno Y. Magnetic resonance
Qureshi IA, Mori A, eds. Guanidino compounds. Eastleigh: imaging in late-onset ornithine transcarbamylase deficien-
John Libbey 1997, pp 71–76 cy. Brain Dev 2003; 25: 40–44
Chen Y-F, Huang Y-C, Liu H-M, Hwu W-L. MRI in a case of adult Lee B, Goss J. Long-term correction of urea cycle disorders.
onset citrullinemia. Neuroradiology 2001; 43: 845–847 J Pediatr 2001; 138: S62-S71
Choi C-G,Yoo HW.Localized proton MR spectroscopy in infants Legras A, Labarthe F, Maillot F, Garrigue M-A, Kouatchet A, Ogi-
with urea cycle defect. AJNR Am J Neuroradiol 2001; 22: er de Baulny H. Late diagnosis of ornithine transcarbamy-
834–837 lase defect in three related female patients: polymorphic
presentations. Crit Care Med 2002; 30: 241–244
982 References and Further Reading

Maestri NE, Hauser ER, Bartholomew D, Brusilow SW. Prospec- Picker JD, Puga AC, Levy HL, Marsden D, Shih VE, DeGirolami U,
tive treatment of urea cycle disorders. J Pediatr 1991; 119: Ligon KL, Cederbaum SD, Kern RM, Cox GF. Arginase defi-
923–928 ciency with lethal neonatal expression: evidence for the
Maestri NE, McGowan KD, Brusilow SW. Plasma glutamine con- glutamine hypothesis of cerebral edema. J Pediatr 2003;
centration: a guide in the management of urea cycle disor- 142: 349–352
ders. J Pediatr 1992; 121: 259–261 Potter M, Hammond JW, Sim K-G, Green AK, Wilcken B. Or-
Maestri NE, Brusilow SW, Clissold DB, Bassett SS. Long-term nithine carbamoyltransferase deficiency: improved sensi-
treatment of girl with ornithine transcarbylase defiency. N tivity of testing for protein tolerance in the diagnosis of
Engl J Med 1996; 335: 855–859 heterozygotes. J Inherit Metab Dis 2001; 24: 5–14
Mamourian AC, du Plessis A. Urea cycle defect: a case with MR Prasad AN, Breen JC, Ampola MG, Rosman NP. Argininemia: a
and CT findings resembling infarct. Pediatr Radiol 1991; 21: treatable genetic cause of progressive spastic diplegia sim-
594–595 ulating cerebral palsy: case reports and literature review.
Marescau B, de Deyn PP, Lowenthal A, Qureshi IA, Antonozzi I, J Child Neurol 1997; 12: 301–309
Bachmann C, Cederbaum SD, Cerone R, Chamoles N, Pridmore CL, Clarke JTR, Blaser S. Ornithine transcarbamylase
Colombo JP,Hyland K,Gatti R,Kang SS,Letarte J,Lambert M, deficiency in females: an often overlooked cause of treat-
Mizutani N, Possemiers I, Rezvani I, Snyderman SE, Ter- able encephalopathy. J Child Neurol 1995; 10: 369–374
heggen HG, Yoshino M. Guanidino compound analysis as a Solitare GB, Shih VE, Nelligan DJ, Dolan Jr DJ, Arigininosuccinic
complementary diagnostic parameter for hyperarginine- aciduria: clinical, biochemical, anatomical and neuropatho-
mia: follow-up of guanidino compound levels during ther- logical observations. J Ment Defic Res 1969; 13: 153–170
apy. Pediatr Res 1990; 27: 297–303 Sperl W, Felber S, Skladal D, Wermuth B. Metabolic stroke in
Martin JJ, Farriaux JP, de Jonghe P. Neuropathology of citrulli- carbamyl phosphate synthetase deficiency. Neuropedi-
naemia. Acta Neuropathol (Berl) 1982; 56: 303–306 atrics 1997; 28: 229–234
Mathias RS, Kostiner D, Packman S. Hyperammonemia in urea Takanashi J, Kurihara A, Tomita M, Kanazawa M, Yamamoto S,
cycle disorders: role of the nephrologist. Am J Kidney Dis Morita F, Ikehira H, Tanada H, Tanada S, Kohno Y. Distinctly
2001; 37: 1069–1080 abnormal brain metabolism in late-onset ornithine trans-
Matsuda I, Nagata N, Matsuura T, Oyanagi K,Tada K, Narisawa K, carbamylase deficiency. Neurology 2002; 59: 210–214
Kitagawa T, Sakiyama T, Yamashita F, Yoshino M. Retrospec- Takanashi J-I, Barkovic AJ, Cheng SF, Weisiger K, Zlatunich CO,
tive survey of urea cycle disorders. 1. Clinical and laborato- Mudge C, Rosenthal P, Tuchman M, Packman S. Brain MR
ry observations of thirty-two Japanese male patients with imaging in neonatal hyperammonemic encephalopathy
ornithine transcarbamylase deficiency. Am J Med Genet resulting from proximal urea cycle disorders. AJNR Am J
1991; 38: 85–89 Neuroradiol 2003; 24: 1184–1187
Matsuura T, Hoshide R, Fukushima M, Sakiyama T, Owada M, Takanashi J-I, Barkovic AJ, Cheng SF, Kostiner D, Baker JC, Pack-
Matsuda I. Prenatal monitoring of ornithine transcarb- man S. Brain MR imaging in acute hyperammonemic en-
amoylase deficiency in two families by DNA analysis. J In- cephalopathy arising form late-onset ornithine transcar-
herit Metab Dis 1993; 16: 31–38 bamylase deficiency. AJNR Am J Neuroradiol 2003; 24:
Mattson LR, Lindor NM, Goldman DH, Goodwin JT, Groover RV, 390–393
Vockley J. Central pontine myelinosis as a complication of Takeoka M, Soman TB, Shih VE, Caviness VS, Krishnamoorthy
partial ornithine carbamoyl transferase deficiency. Am J KS.Carbamyl phosphate synthetase 1 deficiency: a destruc-
Med Genet 1995; 60: 210–213 tive encephalopathy. Pediatr Neurol 2001; 24: 193–199
McCullough BA, Yudkoff M, Batshaw ML, Wilson JM, Raper SE, Tomomasa T, Kobayashi K, Kaneko H, Shimura H, Fukusato T,
Tuchman M. Genotype spectrum of ornithine transcar- Tabata M, Ioue Y, Ohwada S, Kasahara M, Morishita Y, Kimu-
bamylase deficiency: correlation with the clinical and bio- ra M, Saheki T, Morikawa A. Possible clinical and histological
chemical phenotype. Am J Med Genet 2000; 93: 313–319 manifestations of adult-onset type II citrullinemia in early
Msall M, Batshaw ML, Suss R, Brusilow SW, Mellits ED. Neuro- infancy. J Pediatr 2001; 138: 741–743
logic outcome in children with inborn errors of urea syn- Travers H, Reed JS, Kennedy JA. Ultrastructural study of the liv-
thesis. N Engl J Med 1984; 310: 1500–1505 er in argininosuccinase deficiency. Pediatr Pathol 1986; 5:
Oechsner M, Steen C, Stürenberg HJ, Kohlschütter A. Hyperam- 307–318
monaemic encephalopathy after initiation of valproate Tuchman M.The clinical, biochemical, and molecular spectrum
therapy in unrecognized ornithine transcarbamylase defi- of ornithine transcarbamylase deficiency. J Lab Clin Med
ciency. J Neurol Neurosurg Psychiatry 1998; 64: 680–682 1992; 120: 836–850
Olier J, Gallego J, Digon E. Computerized tomography in pri- Tuchman M. Mutations and polymorphisms in the human
mary hyperammonemia. Neuroradiology 1989; 31: 356– ornithine transcarbamylase gene. Hum Mutat 1993; 2: 174–
257 178
Osafune K, Ichikawa K, Yasui Y, Sekikawa A, Takeoka H, Kanatsu Tuchman M, Mauer SM, Holzknecht RA, Summar ML, Vnencak-
K, Kohigashi K, Koshiyama H. An adult-onset case of argini- Jones CL. Prospective versus clinical diagnosis and therapy
nosuccinate synthetase deficiency presenting with atypi- of acute neonatal hyperammonaemia in two sisters with
cal citrullinemia. Intern Med 1999; 38: 590–596 carbamyl phosphate synthetase deficiency. J Inherit Metab
Oshiro S, Kochinda T, Tana T,Yamazato M, Kobayashi K, Komine Dis 1992; 15: 269–277
Y, Muratani H, Saheki T, Iseki K, Takishita S. A patient with Whitington PF, Alonso EM, Boyle JT, Molleston JP, Rosenthal P,
adult-onset type II citrullinemia on long-term hemodialy- Emond JC, Millis JM. Liver transplantation for the treatment
sis: reversal of clinical symptoms and brain MRI findings. of urea cycle disorders. J Inherit Metab Dis 1998; 21:
Am J Kidney Dis 2002; 39: 189–192 112–118
References and Further Reading 983

Widhalm K, Koch S, Scheibenreiter S, Knoll E, Colombo JP, Bach- 48 Molybdenum Cofactor Deficiency
mann C, Thalhammer O (1992) Long-term follow-up of 12 and Isolated Sulfite Oxidase Deficiency
patients with the late-onset variant of argininosuccinic acid
lyase deficiency: no impairment of intellectual and psy-
Appignani BA, Kaye EM,Wolpert SM. CT and MR appearance of
chomotor development during therapy. Pediatrics 87:
the brain in two children with molybdenum cofactor defi-
1182–1184
ciency. AJNR Am J Neuroradiol 1996; 17: 317–320
Arslanoglu S, Yalaz M, Göklpen, Çoker M, Tütüncüoğlu S, Akisu
M, Darcan Ŝ, Kultursay N, Çiris M, Demirtaŝ E. Molybdenum
47 Serine Synthesis Defect cofactor deficiency associated with Dandy-Walker com-
plex. Brain Dev 2001; 23: 815–818
De Koning TJ. 3-Phosphoglycerate dehydrogenase in disease Barbot C, Martins E, Vilarinho L, Dorche C, Cardoso ML. A mild
and development. Thesis, University of Utrecht. Utrecht: form of infantile isolated sulphite oxidase deficiency. Neu-
Zuidam en Uithof, 2001 ropediatrics 1995; 322: 322–324
De Koning TJ, Duran M, Dorland M, Dorland L, Gooskens R, van Brown GK, Scholem RD, Croll HB,Wraith JE, McGill JJ. Sulfite ox-
Schaftingen E, Jeaken J, Blau N, Berger R, Poll-The BT. Bene- idase deficiency: clinical, neuroradiologic, and biochemical
ficial effects of l-serine and glycine in the management of features in two new patients. Neurology 1989; 39: 252–257
seizures in 3-phosphoglycerate dehydrogenase deficiency. Dublin AB, Hald JK,Wootton-Gorges SL. Isolated sulfite oxidase
Ann Neurol 1998; 44: 261–265 deficiency: MR images features. AJNR Am J Neuroradiol
De Koning TJ, Poll-The BT, Jaeken J. Continuing education in 2002; 23: 484–485
neurometabolic disorders – serine deficiency disorders. Duran M, Beemer FA, van der Heijden C, Korteland J, de Bref PK,
Neuropediatrics 1999; 30: 1–4 Brink M, Wadman SK. Combined deficiency of xanthine ox-
De Koning TJ, Jaeken J, Pineda M, van Maldergem L, Poll-The idase and sulphite oxidase: a defect of molybdenum me-
BT, van der Knaap MS. Hypomyelination and reversible tabolism or transport. J Inherit Metab Dis 1978; 1; 175–178
white matter attenuation in 3-phosphoglycerate dehydro- Endres W, Shin YS, Günther R, Ibel H, Duran M, Wadman SK. Re-
genase deficiency. Neuropediatrics 2000; 31: 287–292 port on a new patient with combined deficiencies of sul-
De Koning TJ, Duran M, van Maldergem L, Pineda M, Dorland L, phite oxidase and xanthine dehydrogenase due to molyb-
Gooskens R, Jaeken J, Poll-The BT. Congenital microcephaly denum cofactor deficiency. Eur J Pediatr 1988; 148:
and seizures due to 3-phosphoglycerate dehydrogenase 246–249
deficiency: outcome of treatment with amino acids. J In- Feng G,Tintrup H, Kirsch J, Nichol MC, Kuhse J, Betz H, Sanes JR.
herit Metab Dis 2002; 25: 119–125 Dual requirement for gephyrin in glycine receptor cluster-
De Koning TJ, Snell K, Duran M, Berger R, Poll-The BT, Surtees R. ing and molybdoenzyme activity. Science 1998; 282:
L-Serine in disease and development. Biochem J 2003; 371: 1321–1324
653–661 Hänzelmann P, Schwartz G, Mendel RR. Functionality of alter-
Häusler MG, Jaeken J, Mönch E, Ramaekers VT. Phenotypic native splice forms of the first enzymes involved in human
heterogeneity and adverse effects of serine treatment in molybdenum cofactor biosynthesis. J Biol Chem 2002; 227:
3-phosphoglycerate dehydrogenase deficiency: report on 18303–18312
two siblings. Neuropediatrics 2001; 32: 191–195 Hughes EF,Fairbanks L,Simmonds HA,O’Robinson R.Molybde-
Jaeken J, Detheux M, van Maldegem L, Foulon M, Carchon H, num cofactor deficiency – phenotypic variability in a fami-
van Schaftingen E.3-Phosphoglycerate dehydrogenase de- ly with late-onset variant. Dev Med Child Neurol 1998; 40:
ficiency: an inborn error of serine biosynthesis. Arch Dis 57–61
Child 1996; 74: 542–545 Johnson JL, Coyne E, Rajagopalan KV, van Hove JLK, Mackay M,
Jaeken J, Detheux M, van Maldegem L, Frijns JP, Alliet P, Foulon Pitt J,Boneh A.Molybdopterin synthase mutations in a mild
M, Carchon H, van Schaftingen E. 3-Phosphoglycerate de- case of molybdenum cofactor deficiency. Am J Med Genet
hydrogenase deficiency and 3-phosphosine phosphatase 2001; 104: 169–173
deficiency: inborn errors of serine biosynthesis. J Inherit Johnson JL, Coyne KE, Garret RM, Zabot M-T, Dorche C, Kisker C,
Metab Dis 1996; 19: 223–226 Rajagopalan KV. Isolated sulfite oxidase deficiency: identifi-
Klomp LWJ, de Koning TJ, Malingré HEM, van Beurden EACM, cation of 12 novel SUOX mutations in 10 patients. Hum
Brink M, Opdam FL, Duran M, Jaeken J, Pineda M, van Mutat 2002; 20: 74–79
Maldergem L, Poll-The BT, Van den Berg IET, Berger R. Mole- Lee HF, Mak BSC, Chi CS, Tsai CR, Chen CH, Shu SG. A novel mu-
cular characterization of 3-phosphoglycerate dehydroge- tation in neonatal isolated sulphite oxidase deficiency.
nase deficiency – a neurometabolic disorder associated Neuropediatrics 2002; 33: 174–179
with reduced l-serine biosynthesis. Am J Hum Genet 2000; Mize C, Johnson JL, Rajagopalan KV. Defective molybdopterin
67: 1389–1399 biosynthesis: clinical heterogeneity associated with molyb-
Pineda M,Vilaseca MA,Artuch R,Santos S,Garcia González MM, denum cofactor deficiency. J Inherit Metab Dis 1995; 18:
Sua I, Aracil A, van Schaftingen E, Jaeken J. 3-Phosphoglyc- 283–290
erate dehydrogenase deficiency in a patient with West syn- Reiss J. Genetics of molybdenum cofactor deficiency. Hum
drome. Dev Med Child Neurol 2000; 42: 629–633 Genet 2000; 106: 157–163
Reiss J, Johnson LJ. Mutations in the molybdenum cofactor
biosynthetic genes MOCS1, MOCS2, and GEPH. Hum Mutat
2003; 21: 569–576
Reiss J, Cohen N, Dorche C, Mandel H, Mendel RR, Stallmeyer B,
Zabot MT, Dierks T. Mutations in a polycistronic nuclear
gene associated with molybdenum cofactor deficiency.
Nat Genet 1998; 20: 51–53
984 References and Further Reading

Reiss J, Christensen E, Kurlemann G, Zabot M-T, Dorche C. Ge- Bosch AM,Bakker HD,van Gennip AH,van Kempen JV,Wanders
nomic structure and mutation spectrum of the bicistronic RJA, Wijburg FA. Clinical features of galactokinase deficien-
MOCS1 gene defective in molybdenum cofactor deficiency cy: a review of the literature. J Inherit Metab Dis 2002; 25:
type A. Hum Genet 1998; 103: 639–644 629–634
Reiss J, Dorche C, Stallmeyer B, Mendel RR, Cohen N, Zabot MT. Burke JP, O’Keefe M, Bowell R, Naughten ER. Ophthalmic find-
Human molybdopterin synthase gene: genomic structure ings in classical galactosemia – a screened population.
and mutations in molybdenum cofactor deficiency type B. J Pediatr Ophthalmol Strabismus 1989; 26: 165–168
Am J Hum Genet 1999; 64: 706–711 Cleary MA, Heptinstall LE, Wraith JE, Walter JH. Galactosemia:
Reiss J, Gross-Hart S, Christensen E, Schmidt P, Mendel RR, relationship of IQ to biochemical control and genotype.
Schwartz G. A mutation in the gene for the neurotransmit- J Inherit Metab Dis 1995; 18: 151–152
ter receptor-clustering protein gephyrin causes a novel Crome L. A case of galactosaemia with the pathological and
form of molybdenum cofactor deficiency. Am J Hum Genet neuropathological findings. Arch Dis Child 1962; 37: 415–
2001; 68: 208–213 429
Rupar CA, Gillett J, Gordon BA, Ramsay DA, Johnson JL, Garrett Friedman JH,Levy HL,Boustany R-M.Late onset of distinct neu-
RM, Rajagopalan KV, Jung JH, Bacheyie GS, Sellers AR. Isolat- rologic syndromes in galactosemic siblings. Neurology
ed sulfite oxidase deficiency. Neuropediatrics 1996; 27: 1989; 39: 741–742
299–304 Gitzelmann R. Galactose-1-phosphate in the pathophysiology
Salvan AM, Chambrol B, Lamoureux S, Confort-Gouny S, Coz- of galactosemia. Eur J Pediatr 1995; 154: S45–S49
zone PJ, Vion-Dury J. In vivo brain proton MR spectroscopy Gitzelmann R, Steinmann B, Mitchell B, Haigis E. Uridine
in a case of molybdenum cofactor deficiency. Pediatr Radi- diphosphate galactose 4-epimerase deficiency. Report of
ol 1999; 29: 846–848 eight cases in three families. Helv Paediatr Acta 1976; 31:
Schuierer G, Kurlemann G, Bick U, Stephani U. Molybdenum- 441–452
cofactor deficiency; CT and MR findings. Neuropediatrics Haberland C, Perou M, Brunngraber EG, Hof H. The neuro-
995; 26: 5–54 pathology of galactosemia. J Neuropathol Exp Neurol
Shalata A, Mandel H, Reiss J, Szargel R, Cohen-Akenine A, 1971; 30: 431–447
Dorche C,Zabot M-T,Van Gennip A,Abeling N,Berant M,Co- Henderson MJ, Holton JB. Further observations in a case of
hen N. Localisation of a gene for molybdenum cofactor de- uridine diphosphate galactose-4-epimerase deficiency
ficiency, on the short arm of chromosome 6, by homozy- with a severe clinical presentation.J Inherit Metab Dis 1983;
gosity mapping. Am J Hum Genet 1998; 63: 148–154 6: 17–20
Shih VE, Abroms IF, Johnson JL, Carney M, Mandell R, Robb RM, Holton JB. Effects of galactosemia in utero. Eur J Pediatr 1995;
Cloherty JP, Rajagopalan KV. Sulfite oxidase deficiency. Bio- 154: S77–S81
chemical and clinical investigations of a hereditary meta- Holton JB. Galactosemia: pathogenesis and treatment. J In-
bolic disorder in sulfur metabolism. N Engl J Med 1977; 207: herit Metab Dis 1996; 19: 3–7
1022–1028 Holton JB, Allen JT, Gillett MG. Prenatal diagnosis of disorders
Topçu M, Coskun T, Haliloglu G, Saatci I. Molybdenum cofactor of galactose metabolism. J Inherit Metab Dis 1989; 12
deficiency: report of three cases presenting as hypoxic-is- (suppl 1): 202–206
chemic encephalopathy. J Child Neurol 2001; 16: 264–270 Keevill NJ, Holton JB, Allen JT. UDP-glucose and UDP-galactose
Touati G, Rusthoven E, Depondt, Dorche C, Duran M, Héron B, concentrations in cultured skin fibroblasts of patients with
Rabier D, Russo M, Saudubray JM. Dietary therapy in two classical galactosaemia. J Inherit Metab Dis 1994; 17: 23–26
patients with a mild form of sulphite oxidase deficiency. Ev- Kliegman RM, Sparks JW. Perinatal galactose metabolism. J Pe-
idence for clinical and biochemical improvement. J Inherit diatr 1985; 107: 831–841
Metab Dis 2000; 23: 45–53 Koch TK,Schmidt KA,Wagstaff JE,Won G,Packman S.Neurolog-
Van der Klei-van Moorsel JM, Smit LM, Brockstedt M, Jacobs C, ic complications in galactosemia. Pediatr Neurol 1992; 8:
Dorche C, Duran M. Infantile isolated sulphite oxidase defi- 217–220
ciency: report of a case with negative sulphite test and nor- Landing BH, Ang SM, Villarreal-Engelhardt G, Donnell GN.
mal sulphate excretion. Eur J Pediatr 1991; 150: 196–197 Galactosemia: clinical and pathologic features, tissue stain-
ing patterns with labeled galactose- and galactosamine-
binding lectins, and possible loci of nonenzymatic galacto-
sylation. Perspect Pediatr Pathol 1993; 17: 99–124
49 Galactosemia Lesgold Belman A, Moshe SL, Zimmerman RD. Computed to-
mographic demonstration of cerebral edema in a child
Acosta PH, Gross KC. Hidden sources of galactose in the envi- with galactosemia. Pediatrics 1986; 78: 606–609
ronment. Eur J Pediatr 1995; 154: S87-S92 Liu G, Hale GE, Hughes CL. Galactose metabolism and ovarian
Beigi B, O’Keefe M, Bowell R, Naughten E, Badawi N, Lanigan B. toxicity. Reprod Toxicol 2000; 14: 377–384
Ophthalmic findings in classical galactosaemia – prospec- Nelson CD, Waggoner DD, Donnell GN, Tuerck JM, Buist NRM.
tive study. Br J Ophthalmol 1993; 77: 162–164 Verbal dyspraxia in treated galactosemia. Pediatrics 1991;
Berry GT. The role of polyols in the pathophysiology of hyper- 88: 346–350
galactosemia. Eur J Pediatr 1995; 154: S53-S64 Nelson MD,Wolff JA, Cross CA, Donnell GN, Kaufman FR. Galac-
Berry GT, Palmieri M, Gross KC, Acosta PB, Henstenburg JA, tosemia: evaluation with MR imaging. Radiology 1992; 184:
Mazur A, Reynolds R, Segal S.The effect of dietary fruits and 255–261
vegetables on urinary galactitol excretion in galactose-1- Olambiwonnu NO, McVie R, Won G, Frasier SD, Donnell GN.
phosphate uridyltransferase deficiency. J Inherit Metab Dis Galactokinase deficiency in twins: clinical and biochemical
1993; 16: 91–100 studies. Pediatrics 1974; 53: 314–318
Böhles H, Wenzel D, Shin YS. Progressive cerebellar and ex-
trapyramidal motor disturbances in galactosaemic twins.
Eur J Pediatr 1986; 145: 413–417
References and Further Reading 985

Ornstein KS, McGuire EJ, Berry GT, Roth S, Segal S. Abnormal Gomori JM, Leibovici V, Zlotogorski A, Wirguin I, Haham-Zadeh
galactosylation of complex carbohydrates in cultured S. Computed tomography in Sjögren-Larsson syndrome.
fibroblasts from patients with galactose-1-phosphate Neuroradiology 1987; 29: 557–559
uridyltransferase deficiency. Pediatr Res 1992; 31: 508–511 Hussain MZ, Aihara M, Oba H, Ohtomo K, Uchiyama G,
Ratner Kaufman F,McBride-Chang C, Manis FR,Wolff JA, Nelson Hayashibe H, Nakazawa S. MRI of white matter changes in
MD. Cognitive functioning, neurologic status and brain the Sjögren-Larsson syndrome. Neuroradiology 1995; 37:
imaging in classical galactosemia. Eur J Pediatr 1995; 154: 576–577
S2–S5 Kelson TL, Craft DA, Rizzo WB. Carrier detection for Sjögren-
Ratner Kaufman F, Horton EJ, Gott P, Wolff JA, Neson Jr. MD, Larsson syndrome. J Inherit Metab Dis 1992; 15: 105–111
Azen C. Manis FR. Abnormal somatosensory evoked poten- Kelson TL, Secor McVoy JR, Rizzo WB. Human liver fatty alde-
tials in patients with classic galactosemia: correlation with hyde dehydrogenase: microsomal localization, purification,
neurologic outcome. J Child Neurol 1995; 10: 32–36 and biochemical characterization. Biochim Biophys Acta
Sardharwalla IB,Wraith JE, Bridge C, Fowler B, Roberts SA. A pa- 1997; 1335: 99–110
tient with severe type of epimerase deficiency galac- Lake BD, Smith VV, Judge MR, Harper JI, Besley GTN. Hexanol
tosaemia. J Inherit Metab Dis 1988; 11 (suppl 2): 249–251 dehydrogenase activity shown by enzyme histochemistry
Schwarz HP, Schaefer T, Bachmann C. Galactose and galactitol on skin biopsies allows differentiation of Sjögren-Larsson
in the urine of children with compound heterozygosity for syndrome from other ichthyoses. J Inherit Metab Dis 1991;
Duarte variant and classical galactosemia (GtD/gt) after an 14: 338–340
oral galactose load. Clin Chem 1985; 31: 420–422 Mano T, Ono J, Taminaga T, Imai K, Sakurai K, Harada K, Nagai T,
Schweitzer S, Shin Y,Jakobs C, Brodehl J.Long-term outcome in Rizzo WB, Okada S. Proton MR spectroscopy of Sjögren-
134 patients with galactosaemia. Eur J Pediatr 1993; 152: Larsson’s syndrome. AJNR Am J Neuroradiol 1999; 20:
36–43 1671–1673
Segal S. Galactosemia unsolved. Eur J Pediatr 1995; 154: Miyanomae Y,Ochi M,Yoshioka H,Takaya K,Kizaki Z,Inoue F,Fu-
S97–S102 ruya S, Naruse S. Cerebral MRI and spectroscopy in Sjögren-
Segal S, Rutman JY, Frimptr GW. Galactokinase deficiency and Larsson syndrome: case report. Neuroradiology 1995; 37:
mental retardation. J Pediatr 1979; 95: 750–753 225–228
Smetana HF, Olen E. Hereditary galactose disease. Am J Clin Mulder LJMM, Oranje AP, Loonen MCB. Cranial CT in the Sjö-
Pathol 1962; 38: 3–25 gren-Larsson syndrome. Neuroradiology 1987; 29: 560–561
Sokol RJ, McCabe ERB, Kotzer AM, Langendoerfer SI. Pitfalls in Rizzo WB. Inherited disorders of fatty alcohol metabolism. Mol
diagnosing galactosemia: false negative newborn screen- Genet Metab 1998; 65: 63–73
ing following red blood cell transfusion. J Pediatr Gastroen- Rizzo WB. Sjögren-Larsson syndrome. Explaining the skin–
terol Nutr 1989; 8: 266–268 brain connection. Neurology 1999; 52: 1307–1308
Tyfield LA,Reichardt J,Fridovich-Keil J,Croke Dt,Elas II LJ,Strobl Rizzo WB, Craft DA. Sjögren-Larsson syndrome. Deficient activ-
W, Kozak L, Coskun T, Novelli G, Okano Y, Zekanowski C, Shin ity of the fatty aldehyde dehydrogenase component of
Y, Dolores Boleda M. Classical galactosemia and mutations fatty alcohol: NAD+ oxidoreductase in cultured fibroblasts.
at the galactose-1-phosphate uridyl transferase (GALT) J Clin Invest 1991; 88: 1643–1648
gene. Hum Mutat 1999; 13: 417–430 Rizzo WB, Craft DA. Sjögren-Larsson syndrome: accumulation
Tyfield LA. Galactosemia and allelic variation at the galac- of free fatty alcohols in cultured fibroblasts and plasma.
toseme-1-phosphate uridyltransferase gene: a complex re- J Lipid Res 2000; 41: 1077–1081
lationship between genotype and phenotype. Eur J Pediatr Rizzo WB, Carney G, Lin Z.The molecular basis of Sjögren-Lars-
2000; 159: S204–S207 son syndrome: mutation analysis of the fatty aldehyde de-
Waggoner DD, Buist NRM. Long-term complications in treated hydrogenase gene. Am J Hum Genet 1999; 65: 1547–1560
galactosemia. Int Pediatr 1993; 8: 97–100 Rizzo WB, Heinz E, Simon M, Craft DA. Microsomal fatty alde-
Waggoner DD, Buist NRM, Donnell GN. Long-term prognosis in hyde dehydrogenase catalyzes the oxidation of aliphatic
galactosaemia: results of a survery of 350 cases. J Inherit aldehyde derived form ether glycerolipid catabolism: im-
Metab Dis 1990; 13: 802–818 plications for Sjögren-Larsson syndrome. Biochim Biophys
Acta 2000; 1535: 1–9
Rizzo WB, Lin Z, Carney G. Fatty aldehyde dehydrogenase:
genomic structure, expression and mutation analysis in
50 Sjögren-Larsson Syndrome Sjögren-Larsson syndrome. Chem Biol Interact 2001;
130–132: 297–307
Altmok D,Yildiz YT, Seçkin D, Altmok G, Tacal T, Eryilmaz M. MRI Sillén A, Anton-Lamprecht I, Braun-Quentin C, Kraus CS, Sitki
of three siblings with Sjögren-Larsson syndrome. Pediatr Sayli B, Ayuso C, Jagell S, Küster W, Waledius C. Spectrum of
Radiol 1999; 29: 776–769 mutations and sequence variants in the FALDH gene in pa-
Auada MP, Taube MBZ, Collares EF, Tanaka AMU, Cintra ML. tients with Sjögren-Larsson syndrome. Hum Mutat 1998;
Sjögren-Larsson syndrome: biochemical defects and follow 12: 377–384
up in three cases. Eur J Dermatol 2002; 12: 263–266 Sylvester PE. Pathological findings in Sjögren-Larsson syn-
De Laurenzi V, Rogers GR, Hamrock DJ, Marekov LN, Steinert SP, drome. J Ment Defic Res 1969; 13: 267–275
Compton JG, Markova N, Rizzo WB. Sjögren-Larsson syn- Tabsh K, Rizzo WB, Holbrook K,Theroux N.Sjögren-Larsson syn-
drome is caused by mutations in the fatty aldehyde dehy- drome: technique and timing of prenatal diagnosis. Obstet
drogenase gene. Nat Genet 1996; 12: 52–57 Gynecol 1993; 82 (suppl II): 700–703
Di Rocco M,Filocamo M,Tortori-Donati P,Veneselli E,Borrone C, Taube B, Billeaud C, Labrèze C, Entressangles B, Fontan D,Taïeb
Rizzo WB. Sjögren-Larsson syndrome: nuclear magnetic A. Sjögren-Larsson syndrome: early diagnosis, dietary man-
resonance imging of the brain in a 4-year-old boy. J Inherit agement and biochemical studies in two cases. Dermatol-
Metab Dis 1994; 17: 112–114 ogy 1999; 198: 340–345
986 References and Further Reading

Tsukamoto N, Chang C, Yoshida A. Mutations associated with Attree O, Olivos IM, Okabe I, Bailey LC, Nelson DL, Lewis RA,
Sjögren-Larsson syndrome. Ann Hum Genet 1997; 61: McInnes RR, Nussbaum RL. The Lowe’s oculocerebrorenal
235–242 syndrome gene encodes a protein highly homologous to
Van Domburg PHMF, Willemsen MAAP, Rotteveel JJ, de Jong inositol polyphosphate-5-phosphatase. Nature 1992; 358:
JGN, Thijssen HOM, Heerschap A, Cruysberg JRM, Wanders 239–242
RJA, Gabreëls FJM, Steijlen PM. Sjögren-Larsson syndrome. Carroll WJ, Woodruff WW, Cadman TE. MR findings in oculo-
Clinical and MRI/MRS findings in FALDH-deficient patients. cerebrorenal syndrome. AJNR Am J Neuroradiol 1993; 14:
Neurology 1999; 52: 1345–1352 449–451
Van Mieghem F, van Goethem JWM, Parizel PM, Cras P, van den Charnas LR, Gahl WA. The oculocerebrorenal syndrome of
Hauwe L, de Meirleire J, de Schepper AM. MR of the brain in Lowe. Adv Pediatr 1991; 38: 75–107
Sjögren-Larsson syndrome. AJNR Am J Neuroradiol 1997; Charnas L, Bernar J, Pezeshkpour GH, Dalakas M, Harper GS,
18: 1561–1563 Gahl WA. MRI findings and peripheral neuropathy in Lowe’s
Verhoeven NM, Jacobs C, Carney G, Somers MP, Wanders RJA, syndrome. Neuropediatrics 1988; 19: 7–9
Rizzo WB. Involvement of microsomal fatty aldehyde dehy- Charnas LR, Bernardini I, Rader D, Hoeg JM, Gahl WA. Clinical
drogenase in the a-oxidation of phytanic acid. FEBS Lett and laboratory findings in the oculocerebrorenal syn-
1998; 429: 225–228 drome of Lowe, with special reference to growth and renal
Wester P, Bergström U, Brun A, Jagell S, Karlsson B, Eriksson A. function. N Engl J Med 1991; 324: 1318–1325
Monoaminergic dysfunction in Sjögren-Larsson syndrome. Cibis GW, Waeltermann JM, Whitcraft CT, Tripathi RC, Harris DJ.
Mol Chem Neuropathol 1991; 15: 13–28 Lenticular opacities in carriers of Lowe’s syndrome. J Oph-
Willemsen MAAP, Rotteveel JJ, Steijlen PM, Heerschap A, May- thalmol 1986; 93: 1041–1045
atepek E. 5-Lipoxygenase inhibition: a new treatment strat- De Camilli P, Emr SD, McPherson PS, Novick P. Phosphoinosi-
egy for Sjögren-Larsson Syndrome. Neuropediatrics 1999; tides as regulators in membrane traffic. Science 1996; 271:
31: 1–3 1533–1539
Willemsen MAAP, Rotteveel JJ, Domburg v PHMF, Gabreëls Demmer LA, Wippold FJ, Dowton SB. Periventricular white
FJM, Mayatepek E, Sengers RCA. Preterm birth in Sjogren- matter cystic lesions in Lowe (oculocerebrorenal) syn-
Larsson syndrome. Neuropediatrics 1999; 30: 325–327 drome. Pediatr Radiol 1992; 22: 76–77
Willemsen MAAP, de Jong JGN, van Domburg PHMF, Rotteveel Dressman MA, Olivos-Glander IM, Nussbaum RL, Suchy SF.
JJ, Wanders RJA, Mayatepek E. Defective inactivation of Ocrl1, a PtdIns(4,5)P2 5-phosphatase, is localized to the
leukotriene B4 in patients with Sjögren-Larsson syndrome. trans-Golgi network of fibroblasts and epithelial cells. J His-
J Pediatr 1999; 136: 258–260 tochem Cytochem 2000; 48: 179–189
Willemsen MAAP, Cruysberg RM, Rotteveel JJ, Aandekerk AL, Fivush BA, Rausen C, Christenson MJ, Olson JL. Acute tubular
van Domburg PHMF, Deuteman AF. Juvenile Macular dys- necrosis associated with Lowe’s syndrome: possible role of
trophy associated with deficient activity of fatty aldehyde rhabdomyolysis. Am J Kidney Dis 1992; 20: 396–399
dehydrogenase in Sjögren-Larsson syndrome. Am J Oph- Garzuly F, Jellinger K, Szabo L,Toth K. Morbid changes in Lowe’s
thalmol 2000; 130: 782–789 oculo-cerebro-renal syndrome. Neuropediatrics 1973; 4:
Willemsen MAAP,Lutt MAJ,Steijlen PM,Cruysberg JRM,van der 304–313
Graaf M, Nijhuis-van der Sanden MWG, Pasman JW, Mayate- Giannakopoulos P, Bouras C, Vallet P, Constantinidis J. Lowe
pek E, Rotteveel JJ. Clinical and biochemical effects of zileu- syndrome: clinical and neuropathological studies of an
ton in patients with the Sjögren-Larsson syndrome. Eur J adult case. J Ment Defic Res 1990; 34: 491–500
Pediatr 2001; 160: 711–717 Irvine R. Second messengers and Lowe syndrome. Nat Genet
Willemsen MAAP, Rotteveel JJ, de Jong JGN, Wanders RJA, Ijlst 1992; 1: 315–316
L, Hoffmann GF, Mayatepek E. Defective metabolism of Kenworthy L, Park T, Charnas LR. Cognitive and behavioral pro-
leukotriene B4 in the Sjögren-Larsson syndrome. J Neurol file of the oculocerebrorenal syndrome of Lowe. Am J Med
Sci 2001; 183: 61–67 Genet 1993; 46: 297–303
Willemsen MAAP, IJlst L, Steijlen PM, Rotteveel JJ, de Jong JGN, Kohyama J, Niimura F, Kawashima K, Iwakawa Y, Nonaka I. Con-
van Domburg PHMF, Mayatepek E, Gabreëls FJM, Wanders genital fiber type disproportion myopathy in Lowe syn-
RJA. Clinical, biochemical and molecular genetic character- drome. Pediatr Neurol 1989; 5: 373–376
istics of 19 patients with the Sjögren-Larsson syndrome. Lin T, Orrison BM, Leahey A-M, Suchy SF, Bernard DJ, Lewis RA,
Brain 2001; 124: 1426–1437 Nussbaum RL. Spectrum of mutations in the OCRL1 gene in
Willemsen MAAP, van der Graaf M, van der Knaap MS, Heer- the Lowe oculocerebrorenal syndrome. Am J Hum Genet
schap A, van Domburg PHMF, Gabreëls FJM, Rotteveel JJ. 1997; 60: 1384–1388
MR imaging and proton MR spectroscopic studies in Sjö- Lin T, Orrison BM, Suchy SF, Lewis RA, Nussbaum RL. Mutations
gren-Larsson syndrome: characterization of the leukoen- are not uniformly distributed throughout the OCRL1 gene
cephalopathy. AJNR Am J Neuroradiol 2004; 25: 649–657 in Lowe syndrome patients. Mol Genet Metab 1998; 64:
58–61
Monnier N, Satre V, Lerouge E, Berthoin F, Lunardi J. OCRL1 mu-
tation analysis in French Lowe syndrome patients: implica-
51 Lowe Syndrome tions for molecular diagnosis strategy and genetic counsel-
ing. Hum Mutat 2000; 16: 157–165
Athreya BH, Schumacher HR, Getz HD, Norman ME, Borden S, Mueller OT, Hartsfield JK, Gallardo LA, Essig YP, Miller KL, Papen-
Witzleben CL. Arthropathy of Lowe’s (oculocerebrorenal) hausen PR, Tedesco TA. Lowe oculocerebrorenal syndrome
syndrome. Arthritis Rheum 1983; 26: 728–735 in a female with a balanced X;20 translocation: mapping of
the X chromosome breakpoint. Am J Hum Genet 1991; 49:
804–810
References and Further Reading 987

Nielsen KF, Steffensen GK. Congenital nephritic syndrome as- Brewer GJ. Recognition, diagnosis, and management of Wil-
sociated with Lowe’s syndrome. Child Nephrol Urol 1990; son’s disease. Proc Soc Exp Biol Med 2000; 233: 39–46
10: 92–95 Brewer GJ, Askari F. Transplant livers in Wilson’s disease for he-
Olivos-Glander IM, Jänne PA, Nussbaum RL. The oculocere- patic, not neurologic, indications. Liver Transplant 2000; 6:
brorenal syndrome gene product is a 105-kD protein local- 662–664
ized tot the Golgi complex. Am J Hum Genet 1995; 57: Brewer GJ, Yuzbasiyan-Gurkan V. Wilson disease. Medicine
817–823 1992; 71: 139–164
Ono J,Harada K,Mano T,Yamamoto T,Okada S.MR findings and Brewer GJ, Fink JK, Hedera P. Diagnosis and treatment of Wil-
neurologic manifestations in Lowe oculocerebrorenal syn- son’s disease. Semin Neurol 1999; 19: 261–270
drome. Pediatr Neurol 1996; 14: 162–164 Brewer GJ, Johnson VD, Dick RD, Hedera P, Fink JK, Kluin KJ.
O’Tuama LA, Laster DW. Oculocerebrorenal syndrome: case re- Treatment of Wilson’s disease with zinc.XVII.Treatment dur-
port with CT and MR correlates. AJNR Am J Neuroradiol ing pregnancy. Hepatology 2000; 31: 364–370
1987; 8: 555–557 Brewer GJ, Dick RD, Johnson VD, Fink JK, Kluin KJ, Daniels S.
Pueschel SM, Brem AS, Nittoli P. Central nervous system and re- Treatment of Wilson’s disease with zinc. XVI.Treatment dur-
nal investigations in patients with Lowe syndrome. Childs ing the pediatric years. J Lab Clin Med 2001; 137: 191–198
Nerv Syst 1992; 8: 45–48 Brewer GJ, Hedera P, Kluin KJ, Carlson M, Askari F, Dick RB, Sitter-
Satre V, Monnier N, Berthoin F, Ayuso C, Joannard A, Jouk P-S, ly J, Fink JK. Treatment of Wilson disease with ammonium
Lopex-Pajares I, Megabarne A, Philipe HJ, Plauchu H, Torres tetrathiomolybdate. Arch Neurol 2003; 60: 379–385
ML, Lunardi J. Characterization of a germline mosaicism in Bingle CD, Srai SKS, Epstein O. Copper metabolism in hyper-
families with Lowe syndrome, and identification of seven cupremic human livers. Studies of its subcellular distribu-
novel mutations in the OCRL1 gene. Am J Hum Genet 1999; tion, association with binding proteins and expressions of
65: 68–76 mRNAs. J Hepatol 1992; 15: 94–101
Savolaine ER,Bielke DJ.Cranial magnetic resonance imaging in Brugieres P, Combes C, Ricolfi F, Degos JD, Poirier J, Gaston A.
Lowe’s syndrome. Clin Imaging 1993; 17: 133–136 Atypical MR presentation of Wilson disease: a possible con-
Schneider JF, Boltshauser E, Neuhaus TJ, Rauscher C, Martin E. sequence of paramagnetic effect of copper? Neuroradiolo-
MRI and proton spectroscopy in Lowe syndrome. Neurope- gy 1992; 34: 222–224
diatrics 2001; 32: 45–48 Bull PC,Thomas GR, Rommens JM, Forbes JR,Wilson Cox D.The
Shields D, Arvan P. Disease models provide insight into post- Wilson disease gene is a putative copper transporting
Golgi protein trafficking, localization and processing. Curr P-type ATPase similar to the Menkes gene. Nat Genet 1993;
Opin Cell Biol 1999; 11: 489–494 5: 327–337
Suchy SF, Nussbaum RL. The deficiency of PIP2 5-phosphatase Castilla-Higuero L,Romero-Gomez M,Suarez E,Castro M.Acute
in Lowe syndrome affects actin polymerization. Am J Hum hepatitis after starting zinc therapy in a patient with
Genet 2002; 71: 1420–1427 presymptomatic Wilson’s disease. Hepatology 2000; 32: 877
Suchy SF, Olivos-Glander IM, Nussbaum RL. Lowe syndrome, a Chelly J, Monaco AP. Cloning the Wilson disease gene. Nat
deficiency of a phosphatidyl-inositol 4,5-biphosphate 5- Genet 1993; 5: 317–318
phosphatase in the Golgi apparatus. Hum Mol Genet 1995; De Haan J, Grossman RI, Civitello L, Hackney DB, Golberg HI,
4: 2245–2250 Bilaniuk LT, Zimmerman RA. High-field magnetic resonance
Terslev E. Two cases of aminoaciduria, ocular changes and re- imaging of Wilson’s disease. J Comput Assist Tomogr 1987;
tarded mental and somatic development (Lowe’s syn- 11: 132–135
drome). Acta Paediatr Scand 1960; 49: 635–644 Demirkiran M, Jankovic J, Lewis RA, Cox DW. Neurologic pre-
Tripathi RC, Cibis GW, Tripathi BJ. Pathogenesis of cataracts in sentation of Wilson disease without Kayser-Fleischer rings.
patients with Lowe’s syndrome. J Ophthalmol 1986; 93: Neurology 1996; 46: 1040–1043
1046–1051 Dening TR, Berrios GE, Walshe JM. Wilson’s disease and epilep-
Zhang X,Majerus PW.Phosphatidylinositol signaling reactions. sy. Brain 1988; 111: 1139–1155
Semin Cell Dev Biol 1998; 9: 153–160 Di Donato M, Sarkar B. Copper transport and its alterations in
Zhang X, Jefferson AB, Auethavekiat V, Majerus PW.The protein Menkes and Wilson diseases. Biochim Biophys Acta 1997;
deficient in Lowe syndrome is a phosphatidylinositol-4,5- 1360: 3–16
biphosphate 5-phosphatase. Proc Natl Acad Sci 1995; 92: Emre S, Atillasoy EO, Ozdemir S, Schilsky M, Rathna Varma CVR,
4853–4856 Thung SN, Sternlieb I, Guy SR, Sheiner PA, Schwartz ME,
Miller CM. Orthotopic liver transplantation for Wilson’s dis-
ease: a single-center experience. Transplantation 2001; 72:
1232–1236
52 Wilson Disease Engelbrecht V, Schlaug G, Hefter H, Kahn T, Mödder U. MRI of
the brain in Wilson disease: T2 signal loss under therapy.
Aisen AM, Martel W, Gabrielsen TO, Glazer GM, Brewer G,Young J Comput Assist Tomogr 1995; 19: 635–638
AB, Hill G.Wilson disease of the brain: MR imaging. Radiolo- Gaffney D,Walker JL, O’Donnell JG, Fell GS, O’Neill KF, Park RHR,
gy 1985; 157: 137–141 Russell RI. DNA-based presymptomatic diagnosis of Wilson
Albernaz VS, Castillo M, Mukherji SK, Siatkowski M, Naidich TP. disease. J Inherit Metab Dis 1992; 15: 161–170
Facies to remember. Int J Neuroradiol 1997; 3: 206–217 Giagheddu M, Tamburini G, Piga M, Tacconi P, Giagheddu A,
Bertrand E, Lewandowska E, Szpak GM, Hoogenraad T, Serra A, Siotto P, Satta L, Demilia L, Marrosu F.Comparison of
Blaauwgers HG, Czlonkowska A, Dymecki J. Neuropatho- MRI, EEG, EPS and ECD-SPECT in Wilson’s disease. Acta Neu-
logical analysis of pathological forms of astroglia in Wil- rol Scand 2001; 103: 71–81
son’s disease. Folia Neuropathol 2001; 39: 73–79 Gow PJ, Smallwood RA, Angus PW, Smith AL,Wall AJ, Sewell RB.
Brewer GJ. Penicillamine should not be used as initial therapy Diagnosis of Wilson’s disease: an expience over three
in Wilson’s disease. Mov Disord 1999; 14: 551–554 decades. Gut 2000; 46: 415–419
988 References and Further Reading

Grimm G, Madl C, Katzenschlager R, Oder W, Ferenci P, Gangl A. McQuaid A, Lamand M, Mason J. The interactions of penicil-
Detailed evaluation of evoked potentials in Wilson’s lamine with copper in vivo and the effect on hepatic met-
disease. Electroencephalogr Clin Neurophysiol 1992; 82: allothionein levels and copper/zinc distribution: the impli-
119–124 cations for Wilson’s disease and arthritis therapy. J Lab Clin
Heckmann JM, Eastman RW.Wilson’s disease: neurological and Med 1992; 119: 744–750
magnetic resonance imaging improvement on zinc treat- Mercer FJB. The molecular basis of copper-transport diseases.
ment. J Neurol Neurosurg Psychiatry 1994; 57: 1273–1274 Trends Mol Med 2001; 7: 64–69
Hedera P, Brewer GJ, Arbor A, Fink JK. White matter changes in Mochizuki H, Kamakura K, Masaki T, Okano M, Nagata N, Inui A,
Wilson disease. Arch Neurol 2002; 59: 866–867 Fujisawa T, Kaji T. Atypical MRI features of Wilson’s disease:
Hefter H, Rautenberg W, Kreuzpainter G, Arendt G, Freund H-J, high signal in globus pallidus on T1-weighted images. Neu-
Pichlmayr R, Strohmeyer G. Does orthopic liver transplanta- roradiology 1997; 39: 171–174
tion heal Wilson’s disease? Clinical follow-up of two liver- Oder W, Grimm G, Kollegger H, Ferenci P, Schneider B, Deecke L
transplanted patients.Acta Neurol Scand 1991; 84: 192–196 . Neurological and neuropsychiatric spectrum of Wilson’s
Hoogenraad TU, Koevoet R, de Ruyter-Korver EGWM. Oral zinc disease: a prospective study of 45 cases. J Neurol 1991; 238:
sulphate as long term treatment in Wilson’s disease. Eur 281–287
Neurol 1979; 18: 205–211 Oder W, Prayer L, Grimm G, Spatt J, Ferenci P, Kollegger H,
Houwen RHJ,Roberts EA,Thomas GR,Cox DW.DNA markers for Schneider B, Gangl A, Deecke L. Wilson’s disease: evidence
the diagnosis of Wilson disease. J Hepatol 1993; 17: 269– of subgroups derived from clinical findings and brain le-
276 sions. Neurology 1993; 43: 120–124
Huang C-C, Chu N-S. Acute dystonia with thalamic and brain- Porzio S, Iorio R,Vajro P, Pensati P,Vegnente A. Penicillamine-re-
stem lesions after initial penicillamine treatment in Wilson’s lated neurologic syndrome in a child affected by Wilson
disease. Eur Neurol 1998; 39: 32–37 disease with hepatic presentation. Arch Neurol 1997; 54:
Huang C-C, Chu N-S.Wilson’s disease: resolution of MRI lesions 1166–1168
following long-term oral zinc therapy. Acta Neurol Scand Prayer L, Wimberger D, Kramer J, Grimm G, Oder W, Imhof H.
1996; 93: 215–218 Cranial MRI in Wilson’s disease. Neuradiology 1990; 32:
Imlya M, Ichikawa K, Matsushima H, Kageyama Y, Fujioka A. MR 211–214
of the base of the pons in Wilson disease. AJNR Am J Neu- Richard VS, Harris VK, Shankar V, Loganathan G, Chandy GM.
roradiol 1992; 13: 1009–1012 Clinical manifestations and survival pattern of Wilson’s dis-
Ishino H,Mii T,Hayashi Y,Saito A,Otsuki S.A case of Wilson’s dis- ease. Natl Med J India 2000; 13: 301–303
ease with enormous cavity formation of cerebral white Riordan SM, Williams R. The Wilson’s disease gene and pheno-
matter. Neurology 1972; 22: 905–909 typic diversity. J Hepatol 2001; 34: 165–171
Keller R, Torta R, Lagget M, Crasto S, Bergamosso B. Psychiatric Roh JK,Lee TG,Wie BA,Lee SB,Park SH,Chang KH.Initial and fol-
symptoms as late onset of Wilson’s disease: neuroradiolog- low-up brain MRI findings and correlation with the clinical
ical findings,clinical features and treatment.Ital J Neurol Sci course in Wilson’s disease. Neurology 1994; 44: 1064–1068
1999; 20: 49–54 Saatci I,Topcu M, Baltaoglu FF, Köse G,Yalaz K, Renda Y, Besim A.
Kobayashi S, Ochiai T, Hori S, Suzuki T, Shimizu T, Gunji Y, Shima- Cranial MR findings in Wilson’s disease. Acta Radiol 1997;
da H, Yamamoto S, Ogawa A, Kohno Y, Sunaga M, Shimazu 38: 250–258
M, Takena K. Copper metabolism after living donor liver Saha M, Kumar S, Das A, Gupta RK. Similarities and differences
transplantation for hepatic failure of Wilson’s disease from of MR findings between Japanese encephalitis and Wilson’s
a gene mutated donor. Hepatogastroenterology 2001; 48: disease. Eur Radiol 2002; 12: 872–876
1259–1261 Sarkar B. Copper transport and its defect in Wilson disease:
Komatsu H, Fujisawa T, Inui A, Soga T, Sekine I, Kodama H, Ue- characterization of the copper-binding domain of Wilson
moto S,Tanaka K. Hepatic copper concentration in children disease in ATPase. J Inorg Biochem 2000; 79: 187–191
undergoing living related liver transplantation due to Schagen van Leeuwen JH, Christiaens GCML, Hoogenraad TU.
Wilsonian fulminant hepatic failure. Clin Transplant 2002; Recurrent abortion and the diagnosis of Wilson disease.
16: 227–232 Obstet Gynecol 1991; 78: 547–549
Lang CJG, Rabas-Kolominsky P, Engelhart A. Fatal deterioration Schilsky ML. Diagnosis and treatment of Wilson’s disease. Pedi-
of Wilson’s disease after institution of oral zinc therapy. atr Transplant 2002; 6: 15–19
Arch Neurol 1993; 50: 1007–1008 Schlaug G, Hefter H, Engelbrecht V, Kluwert T, Arnold S, Stöcklin
Longhi R, Riva E, Rottoli A, Valsasina R, Pinelli P, Giovannini M. G, Seitz RJ. Neurological impairment and recovery in Wil-
Nuclear magnetic resonance brain study in a case of Wilson son’s disease: evidence from PET and MRI. J Neurol Sci 1996;
disease. J Inherit Metab Dis 1989; 12 (suppl) 386–388 136: 129–139
Lui CC, Chen CL, Cheng YF, Lee TY. Recovery of neurological Schulman S, Barbeau A.Wilson’s disease: a case with almost to-
deficits in a case of Wilson’s disease after liver transplanta- tal loss of cerebral white matter. J Neuropathol Exp Neurol
tion.Transplant Proc 1998; 30: 3324–3325 1963; 22: 105–119
Magalhaes ACA, Caramelli P, Menezes JR, Lo LS, Bacheschi LA, Schumacher G, Platz KP, Mueller AR, Neuhaus R, Luck W, Lan-
Barbosa ER, Rosemberg LA. Wilson’s disease: MRI with clini- grehr JM, Settmacher U, Steinmuller T, Becker M, Neuhaus P.
cal correlation. Neuroradiology 1994; 36: 97–100 Liver transplantation in neurologic Wilson’s disease. Trans-
Mason AL, Marsh W, Alpers DH. Intractable neurological Wil- plant Proc 2001; 33: 1518–1519
son’s disease treated with orthotopic liver transplantation. Scully RE, Mark EJ, McNelly WF, Ebeling SH. Presentation of a
Dig Dis Sci 1993; 38: 1746–1750 case. N Engl J Med 1997; 336: 118–125
Matsuura T, Sasaki H,Tashiro K. Atypical MR findings in Wilson’s Selwa LM,Vanderzant CW, Brunberg JA, Brewer GJ, Drury I, Bey-
disease: pronounced lesions in the dentate nucleus caus- doun A.Correlation of evoked potential and MRI findings in
ing tremor. J Neurol Neurosurg Psychiatry 1998; 64: 160 Wilson’s disease. Neurology 1993; 43: 2059–2064
References and Further Reading 989

Selwa LM,Vanderzant CW, Brunberg JA, Brewer GJ, Drury I, Bey- Yüce A, Koçak N, Özen H, Gürakan F. Wilson’s disease patients
doun A.Correlation of evoked potential and MRI findings in with normal ceruloplasmin levels. Turk J Pediatr 1999; 41:
Wilson’s disease. Neurology 1993; 43: 2059–2064 99–102
Sener RN.Wilson’s disease: MRI demonstration of cavitations in Yuzbasiyan-Gurkan V, Grider A, Nostrant T, Cousins RJ, Brewer
basal ganglia and thalami. Pediatr Radiol 1993; 23: 157 GJ. Treatment of Wilson’s disease with zinc. X. Intestinal
Sener RN. The claustrum on MRI: normal anatomy, and the metallothionein induction. J Lab Clin Med 1992; 120:
bright claustrum as a new sign in Wilson’s disease. Pediatr 380–386
Radiol 1993; 23: 594–596
Shimizu N,Yamaguchi Y, Aoki T.Treatment and management of
Wilson’s disease. Pediatr Int 1999; 41: 419–422
Siegmund R, Lößner, Günther K, Kühn H-J, Bachmann H. Mode
53 Menkes Disease
of action of triethylenetetramine dihydrochloride on cop-
per metabolism in Wilson’s disease. Acta Neurol Scand Ambrosini L, Mercer JFB. Defective copper-induced trafficking
1991; 83: 356–359 and localization of the Menkes protein in patients with
Singcharoen T, Chakkaphak K, Udompanich O. Unusual mag- mild and copper treated classical Menkes disease.Hum Mol
netic resonance findings in Wilson’s disease. Br J Radiol Genet 1999; 8: 1545–1555
1991; 64: 752–754 Aynaci FM, Mocan H, Bahadir S, Sari A, Aksoy A. A case of
Song Y-M, Chen M-D. A single determination of liver copper Menkes’ syndrome associated with deafness and inferior
concentration may misdiagnose Wilson’s disease. Clin cerebellar vermian hypoplasia. Acta Paediatr 1997; 87:
Biochem 2000; 33: 589–590 121–123
Starosta-Rubinstein S, Young AB, Kluin K, Hill G, Aisen AM, Barnard RO, Best PV, Erdohazi M. Neuropathology of Menkes’
Gabrielsen T, Brewer GJ. Clinical assessment of 31 patients disease. Dev Med Child Neurol 1978; 20: 586–597
with Wilson’s disease. Correlations with structural changes Blaser SI, Berns DH, Ross JS, Lanska MJ,Weissman BM. Serial MR
on magnetic resonance imaging. Arch Neurol 1987; 44: studies in Menkes disease. J Comput Assist Tomogr 1989;
365–370 13: 113–115
Sternlieb I. The outlook for the diagnosis of Wilson’s disease. Camakaris J, Voskoboinik I, Mercer JF. Molecular mechanisms
J Hepatol 1993; 17: 263–264 of copper homeostasis. Biochem Biophys Res Commun
Suzuki M, Gitlin JD. Intracellular localization of the Menkes and 1999; 261: 225–232
Wilson’s disease proteins and their role in intracellular cop- Chelly J, Tümer Z, Tønnesen T, Petterson A, Ishikawa-Brush Y,
per transport. Pediatr Int 1999; 41: 436–442 Tommerup N, Horn N, Monaco AP. Isolation of a candidate
Tanzi RE, Petrukhin K, Chernov I, Pellequer JL, Wasco W, Ross B, gene for Menkes disease that encodes a potential heavy
Romano M, Parano E, Pavone L, Brzustowicz LM, Devoto M, metal binding protein. Nat Genet 1993; 3: 14–19
Peppercorn J, Bush AI, Sternlieb I, Pirastu M, Gusella JF, Christodoulou J, Danks DM, Sarkar B, Baerlocher KE, Casey R,
Evgrafov O, Penchaszaeh GK, Honig B, Eelman IS, Soares MB, Horn N,Tümer Z, Clarke JTR. Early treatment of Menkes dis-
Scheinberg IH, Gilliam TC.The Wilson disease gene is a cop- ease with parenteral cooper-histidine: long-term follow-up
per transporting ATPase with homology to the Menkes dis- of four treated patients. Am J Med Genet 1998; 76: 154–164
ease gene. Nat Genet 1993; 5: 344–350 Dagenais SL, Adam AN, Innis JW, Glover TW. A novel frameshift
Thuomas KA, Aquilonius SMA, Bergström K, Westermark K. mutation in exon 23 of ATP7A (MNK) results in occipital
Magnetic resonance imaging of the brain in Wilson’s dis- horn syndrome and not in Menkes disease. Am J Hum
ease. Neuroradiology 1993; 35: 134–141 Genet 2001; 69: 420–427
Van Wassenaar-van Hall HN, van den Heuvel AG, Jansen GH, DiDonato M, Sarkar B. Copper transport and its alterations in
Hoogenraad TU, Mali WPTM. Cranial MR in Wilson disease: Menkes and Wilson diseases. Biochim Biophys Acta 1997;
abnormal white matter in extrapyramidal and pyramidal 1360: 3–16
tracts. AJNR Am J Neuroradiol 1995; 16: 2021–2027 Faerber EN, Grover WD, DeFilipp GJ, Capitanio MA, Lui T-H,
Van Wassenaar-van Hall HN, van den Heuvel AG, Algra A, Swartz JD. Cerebral MR of Menkes kinky-hair disease. AJNR
Hoogenraad TU, Mali WPTM.Wilson disease: findings at MR Am J Neuroradiol 1989; 10: 190–192
imaging and CT of the brain with clinical correlation. Radio- Gasch AT, Caruso RC, Kaler SG, Kaiser-Kupfer M. Menkes’ syn-
logy 1996; 198: 531–536 drome. Ophthalmic findings. Ophthalmology 2002; 109:
Walker JM, Tsivkovskii R, Lutsenko S. Metallochaperone Atox1 1477–1483
transfers copper to the NH2-terminal domain of the Wil- Geller TJ, Pan Y, Martin DS. Early neuroradiologic evidence of
son’s disease protein and regulates its catalytic activity. degeneration in Menkes’ disease. Pediatr Neurol 1997; 17:
J Biol Chem 2002; 277: 27953–27959 255–258
Walshe JM. Penicillamine: the treatment of first choice for pa- George DH,Casey RE.Menkes disease after copper histidine re-
tients with Wilson’s disease. Mov Disord 1999; 14: 545–550 placement therapy: case report. Pediatr Dev Pathol 2001; 4:
Walshe JM, Yealland M. Wilson’s disease: the problem of de- 281–288
layed diagnosis. J Neurol Neurosurg Psychiatry 1992; 55: Gu Y-H, Kodoma H, Murata Y, Mochizuki D, Yanagawa Y, Ushiji-
692–696 ma H, Shiba T, Lee C-C. ATP7A Gene mutations in 16 patients
Wu J-C, Huang C-C, Jeng L-B, Chu N-S. Correlation of neurolog- with Menkes disease and a patient with occipital horn syn-
ical manifestations an MR images in a patient with Wilson’s drome. Am J Med Genet 2001; 99: 217–222
disease after liver transplantation. Acta Neurol Scand 2000; Guimarães Santos LM, da Silva Teixeira C, Pereira Vilanova LC,
102: 135–139 Micheletti C, Curiati Mendes CS, Borri ML, Martins AM.
Yarze JC, Martin P, Munoz SJ, Friedman LS.Wilson’s disease: cur- Menkes disease. Case report of an uncommon presenta-
rent status. Am J Med 1992; 92: 643–654 tion with white matter lesions.Arq Neuropsiquiatr 2001; 59:
Yoshi F, Takahashi W, Shinohara Y. A Wilson’s disease patient 125–127
with prominent cerebral white matter lesions: five-year fol-
low-up by MRI. Eur Neurol 1996; 36: 392–393
990 References and Further Reading

Harrison MD, Dameron CT. Molecular mechanisms of copper Linder MC, Hazegh-Azam. Copper biochemistry and molecular
metabolism and the role of Menkes disease protein. biology. Am J Clin Nutr 1996; 63: 797S-811S
J Biochem Mol Toxicol 1999; 13: 93–106 Llanos RM, Mercer JFB. The molecular basis of copper home-
Hsisch GE,Robertson RL,Irons M,Soul JS,du Plessis AJ.Cerebral ostasis and copper-related disorders. DNA Cell Biol 2002;
infarction in Menkes’ disease. Pediatr Neurol 2000; 23: 21: 259–270
425–428 Lou HC,Hølmer GK,Reske-Nielsen E,Vagn-Hansen P.Lipid com-
Ichihashi K,Yano S, Kobayashi S, Miyao M,Yanagisawa M. Serial position in gray and white matter of the brain in Menkes’
imaging of Menkes disease. Neuroradiology 1990; 32: disease. J Neurochem 1974; 22: 377–381
56–59 Martin JJ, Leroy JG. Thalamic lesions in a patient with Menkes
Jacobs DS, Smith AS, Finelli DA, Lanzieri CF, Wiznitzer M. kinky-hair disease. Clin Neuropathol 1985; 4: 206–209
Menkes kinky hair disease: characteristic MR angiographic Martin JJ, Flament-Durand J, Farriaux JP,Buyssens N, Ketelbant-
findings. AJNR Am J Neuroradiol 1993; 14: 1160–1163 Balasse P, Jansen C. Menkes kinky-hair disease. A report on
Jankov RP, Boerkoel CF, Hellmann J, Sirkin WL,Tümer Z, Horn N, its pathology. Acta Neuropathol (Berl) 1978; 42: 25–32
Feigenbaum A. Lethal neonatal Menkes’ disease with se- Martins C, Gonçalves C, Moreno A, Poiares Baptista A, Bairos V.
vere vasculopathy and fractures. Acta Paediatr 1998; 87: Menkes’ kinky hair syndrome: ultrastructural cutaneous
1297–1300 alterations of the elastic fibers. Pediatr Dermatol 1997; 14:
Jayawant S, Halpin S,Wallence S. Menkes kinky hair disease: an 347–350
unusual case. Eur J Paediatr Neurol 2000; 4: 131–134 Menkes JH. Menkes disease and Wilson disease: two sides of
Johnson DE, Coleman L, Poe L. MR of progressive neurodegen- the same copper coin.I.Menkes disease.Eur J Paediatr Neu-
erative change in treated Menkes’ kinky hair disease. Neu- rol 1999; 3: 147–158
roradiology 1991; 33: 181–182 Menkes JH. Subdural haematoma, non-accidental head injury
Kaler SG. Menkes disease. Adv Pediatr 1994; 41: 263–304 or ...? Eur J Paediatr Neurol 2001; 5: 175–176
Kaler SG. Diagnosis and therapy of Menkes syndrome, a genet- Mercer JFB. The molecular basis of copper-transport diseases.
ic form of copper deficiency.Am J Clin Nutr 1998; 67: 1029S- Trends Mol Med 2001; 7: 64–69
1034S. Mercer JFB, Livingston J, Hall B, Paynter JA, Begy C, Chan-
Kaler SG. Metabolic and molecular bases of Menkes disease drasekharappa S, Lockhart P, Grimes A, Bhave M, Siemieniak
and occipital horn syndrome. Pediatr Dev Pathol 1998; 1: D, Glover TW. Isolation of a partial candidate gene for
85–98 Menkes disease by positional cloning. Nat Genet 1993; 3:
Kaler SG, Tümer Z. Prenatal diagnosis of Menkes disease. Pre- 20–25
nat Diagn 1998; 18: 287–289 Møller LB, Tümer Z, Lund C, Petersen C, Cole T, Hanusch R, Sei-
Kaler SG, Goldstein DS, Holmes C, Salerno JA, Gahl WA. Plasma del J, Jensen LR, Horn N. Similar splice-site mutations of the
and cerebrospinal fluid neurochemical pattern in Menkes ATP7A gene lead to different phenotypes: classical Menkes
disease. Ann Neurol 1993; 33: 171–175 disease or occipital horn syndrome. Am J Hum Genet 2000;
Kaler SG, Gallo LK, Proud VK, Percy AK, Mark Y, Segal NA, Gold- 66: 1211–1220
stein DS, Holmes CS, Gahl WA. Occipital horn syndrome and Morgello S, Peterson HDC, Kahn LJ, Laufer H. Menkes kinky hair
a mild Menkes phenotype associated with splice site muta- disease with ‘ragged red’ fibers. Dev Med Child Neurol
tions at the MNK locus. Nat Genet 1994; 8: 195–202 1988; 30: 808–820
Kaler SG, Buist NRM, Holmes CS, Goldstein DS, Miller RC, Gahl Okeda R, Gei S, Chen I, Okaniwa M, Shinomiya, Matsubara O.
WA.Early copper therapy in classic Menkes disease patients Menkes’kinky hair disease: morphological and immunohis-
with a novel splicing mutation. Ann Neurol 1995; 38: tochemical comparison of two autopsied patients. Acta
921–928 Neuropathol (Berl) 1991; 81: 450–457
Kaler SG, Das S, Levinson B, Goldstein DS, Holmes CS, Patronas Oshio T, Hino M, Kirino A, Matsumura C, Fukuda K. Urologic ab-
NJ, Packman S, Gahl WA. Successful early copper therapy in normalities in Menkes’ kinky hair disease: report of three
Menkes disease associated with a mutant transcript con- cases. J Pediatr Surg 1997; 32: 782–784
taining a small in-frame deletion. Biochem Mol Med 1996; Ozawa H, Kodama H, Murata Y, Takashima S, Noma S. Transient
57: 37–46 temporal lobe changes and a novel mutation in a patient
Kanumakala S, Boneh A, Zacharin M. Pamidronate treatment with Menkes disease. Pediatr Int 2001; 43: 437–440
improves bone mineral density in children with Menkes Pasquali-Ronchetti I, Baccarani-Contri M, Young RD, Vogel A,
disease. J Inherit Metab Dis 2002; 25: 391–398 Steinmann B, Royce PM. Ultrastructural analysis of skin and
Kim BE, Smith K, Meagher CK, Petris MJ. A conditional mutation aorta from a patient with Menkes disease. Exp Mol Pathol
affecting localization of the Menkes disease copper ATPase. 1994; 61: 36–57
Suppression by copper supplementation. J Biol Chem Petris MJ, Mercer JFB, Culvenor JG, Lockhart P, Gleeson PA, Ca-
2002; 227: 44079–44084 makaris J. Ligand-regulated transport of the Menkes cop-
Kim OH, Suh JH. Intracranial and extracranial MR angiopathy in per P-type ATPase efflux pump from the Golgi apparatus to
Menkes disease. Pediatr Radiol 1997; 7: 782–784 the plasma membrane: a novel mechanism of regulated
Kodoma H, Murata Y, Kobayashi M. Clinical manifestations and trafficking. EMBO J 1996; 15: 6084–6095
treatment of Menkes disease and its variants. Pediatr Int Petris MJ, Strausak D, Mercer JFB. The Menkes copper trans-
1999; 41: 423–429 porter is required for the activation of tyrosinase. Hum Mol
Kreuder J, Otten A, Fuder H, Tümer Z, Tønnesen, Horn N, Dralle Genet 2000; 9: 2845–2851
D. Clinical and biochemical consequences of copper-histi- Pinto F, Calderazzi A, Canapicchi R,Taddeucci G,Tarantino E.Ra-
dine therapy in Menkes disease. Eur J Pediatr 1993; 152: diological findings in a case of Menkes’disease. Childs Nerv
828–832 Syst 1995; 11: 112–114
Leventer RJ, Kornberg AJ, Phelan EM, Kean MJ. Early magnetic Proud VK,Mussell HG,Kaler SG,Young DW,Percy AK.Distinctive
resonance imaging findings in Menkes’ disease. J Child Menkes disease variant with occipital horns: delineation of
Neurol 1997; 12: 222–224 natural history and clinical phenotype. J Med Genet 1996;
65: 44–51
References and Further Reading 991

Robain O, Aubourg P, Routon MC, Dulac O, Ponsot G. Menkes Devys D, Lutz Y, Rouyer N, Bellocq J-P, Mandel JL. The FMR-1
disease: a Golgi and electron microscopic study of the cere- protein is cytoplasmic, most abundant in neurons and ap-
bellar cortex. Clin Neuropathol 1988; 7: 47–52 pears normal in carriers of a fragile X premutation. Nat
Rotilio G, Carri MT, Rossi L, Ciriolo MR. Copper-dependent ox- Genet 1993; 4: 335–340
idative stress and neurodegeneration. IUBMB Life 2000; 50: Dorn MB, Mazzocco MMM, Hagerman RJ. Behavioral and psy-
309–314 chiatric disorders in adult male carriers of fragile X. J Am
Sherwood G, Sarkar B, Sass Kortsak A. Copper histidinate ther- Child Adolesc Psychiatry 1994; 33: 256–264
apy in Menkes’disease: prevention of progressive neurode- Franke P, Maier W, Hautzinger M, Weiffenbach O, Gänsicke M,
generation. J Inherit Metab Dis 1989; 12: 393–396 Iwers B, Poustka F, Schwab SG, Froster U. Fragile-X carrier
Sparaco M, Hirano A, Hirano M, DiMauro S, Bonnilla E. Cyto- females: evidence for a distinct psychopathological pheno-
chrome C oxidase deficiency and neuronal involvement in type? Am J Med Genet 1996; 64: 334–339
Menkes’ kinky hair disease: immunohistochemical study. Franke P, Leboyer M, Gänsicke M, Weiffenbach O, Biancalana V,
Brain Pathol 1993; 3: 349–354 Cornillet-Lefebre P, Croquette MF, Froster U, Schwab SG,
Sugio Y, Sugio Y, Kuwano A, Miyoshi O, Yamada K, Niikawa N, Poustka F, Hautzinger M, Maier W. Genotype-phenotype
Tsukahara M. Translocation t(X;21)(q13.3; p11.1) in a girl relationship in female carriers of the premutation and full
with Menkes disease. Am J Med Genet 1998; 79: 191–194 mutation of FMR-1. Psychiatry Res 1998; 80: 113–127
Sztriha L, Janáky M, Kiss J, Buga K. Electrophysiological and Greco CM, Hagerman RJ, Tassone F, Chudley AE, Del Bigio MR,
99mTc-HMPAO-SPECT studies in Menkes disease. Brain Dev
Jacquemont S, Leehey M, Hagerman PJ. Neuronal intranu-
1994; 16: 224–228 clear inclusions in a new cerebellar tremor/ataxia syn-
Takahashi S, Matsumoto K, Higano S, Ishibashi T, Zuguchi M, drome among fragile X carriers. Brain 2002; 125:1760–1771
Maruoka S, Sakamoto K, Kondo Y. Cranial MRI and MR an- Hagerman PJ, Hagerman RJ. The fragile-X premutation: a ma-
giography in Menkes’ disease. Neuroradiology 1993; 35: turing perspective. Am J Hum Genet 2004; 74: 805–816
556–558 Hagerman PJ, Hagerman RJ. Fragile X-associated tremor/atax-
Tümer Z,Horn N.Menkes disease:recent advances and new as- ia syndrome (FXTAS). Ment Retard Dev Disabil Res Rev
pects. J Med Genet 1997; 34: 265–274 2004; 10: 25–30
Tümer Z, Horn N. Menkes disease: underlying genetic defect Hagerman RJ, Leehey M, Heinrichs W, Tassone F, Wilson R, Hills
and new diagnostic possibilities. J Inherit Metab Dis 1998; J, Grigsby J, Gage B, Hagerman PJ. Intention tremor, parkin-
21: 604–612 sonism, and generalized brain atrophy in male carriers of
Tümer Z, Lund C, Tolshave J, Vural B, Tønnesen T, Horn N. Iden- fragile X. Neurology 2001; 57: 127–130
tification of point mutations in 41 unrelated patients af- Hagerman RJ, Leavitt BR, Farzin F, Jacquemont S, Greco CM,
fected with Menkes disease. Am J Hum Genet 1997; 60: Brunberg JA, Tassone F, Hessl D, Harris SW, Zhang L, Jardini
63–71 T, Gane LW, Ferranti J, Ruiz L, Leehey MA, Grigsby J, Hager-
Vulpe C, Levinson B,Whitney S, Packman S, Gitschier J. Isolation man PJ. Fragile-X-associated tremor/ataxia syndrome
of a candidate gene for Menkes disease and evidence that (FXTAS) in females with the FMR1 premutation. Am J Hum
it encodes a copper-transporting ATPase. Nat Genet 1993; Genet 2004; 74: 1051–1056
3: 7–13 Jacquemont S, Hagerman RJ, Leehey M, Grigsbey J, Zhang L,
Waslen TA, Stuart Houston C, Tchang S. Menkes’ kinky-hair dis- Brunberg JA,Gredo C,Des Portes V,Jardini T,Levine R,Berry-
ease: radiologic findings in a patient treated with copper Kravis E,Ted Brown W, Schaeffer S, Kissel J,Tassone F, Hager-
histidinate. Can Assoc Radiol J 1995; 46: 114–117 man PJ. Fragile X premutation tremor/ataxia syndrome:
Yamaguchi Y, Heiny ME, Suzuki M. Gitlin JD. Biochemical char- molecular, clinical, and neuroimaging correlates. Am J Hum
acterization and intracellular localization of the Menkes Genet 2003; 72: 869–878
disease protein. Proc Natl Acad Sci 1996; 93: 14030–14035 Jacquemont S, Hagerman RJ, Leehey MA, Hall DA, Levine RA,
Brunberg JA, Zhang L, Jardini T, Gane LW, Harris SW, Herman
K, Grigsby J, Greco CM, Berry-Kravis E, Tassone F, Hagerman
PJ. Penetrance of the fragile X-associated tremor/ataxia
54 Fragile X Premutation syndrome in a premutation carrier population. JAMA 2004;
291: 460–469
Beilina A,Tassone F,Schwartz PH,Sahota P,Hagerman PJ.Redis- Murphy DGM, Mentis MJ, Pietrini P, Grady CL, Moore CJ, Hor-
tribution of transcription starts sites within the FMR1 pro- withz B, Hinton V, Dobkin CS, Schapiro MB, Rapoport SI. Per-
motor region with expansion of the downstream CGG-re- mutation female carriers of fragile X syndrome: A pilot
peat element. Hum Mol Genet 2004; 13: 543–549 study on brain anatomy and metabolism. J Am Acad Child
Berry-Kravis E, Lewin F, Wuu J, Leehey M, Hagerman R, Hager- Adolesc Psychiatry 1999; 38: 1294–1301
man P, Goetz CG.Tremor and ataxia in fragile X premutation Riddle JE, Cheema A, Sobesky WE, Gardner SC. Phenotypic in-
carriers: blinded videotape study. Ann Neurol 2003; 53: volvement in females with the FMR1 gene mutation. Am J
616–623 Ment Retard 1998; 102: 590–601
Brunberg JA, Jacquemont S, Hagerman RJ, Berry-Kravis EM, Tassone F, Hagerman RJ,Taylor AK, Gane LW, Godfrey TE, Hager-
Grigsby J, Leehey MA, Tassone F, Brown TW, Greco CM, man PJ. Elevated levels of FMR1 mRNA in carrier males: a
Hagerman PJ. Fragile X premutation carriers: characteristic new mechanism of involvement in the fragile-X syndrome.
MR imaging findings of adult male patients with progres- Am J Hum Genet 2000; 66: 6–15
sive cerebellar and cognitive dysfunction. AJNR Am J Neu- Tassone F, Hagerman RJ, Hagerman PJ. A majority of fragile X
roradiol 2002; 23: 1757–1766 males with methylated, full mutation alleles have signifi-
Chen L-S, Tassone F, Sahota P, Hagerman PJ. The (CGG)n repeat cant levels of FMR1 messenger RNA. J Med Genet 2001; 38:
element within the 5’untranslated region of the FMR1 mes- 453–456
sage provides both positive and negative cis effects on in
vivo translation of a downstream reporter. Hum Mol Genet
2003; 12: 3067–3074
992 References and Further Reading

Tassone F, Hagerman RJ, Garcia-Arocena D, Khandjian EW, Gre- Pini G, Faulkner LB. Cerebellar involvement in hypomelanosis
co CM, Hagerman PJ. Intranuclear inclusions in neural cells of Ito. Neuropediatrics 1995; 26: 208–210
with premutation alleles in fragile X associated tremor/ Ross DL, Liwnicz BH, Chun RWM, Gilbert E. Hypomelanosis of
ataxia syndrome. J Med Genet 2004; 41: e43 Ito (incontinetia pigmenti achromians) – a clinicopatholog-
ic study: macrocephaly and gray matter heterotopias. Neu-
rology 1982; 32: 1013–1016
Rott H-D, Lang GE, Huk W, Pfeiffer RA. Hypomelanosis of Ito (in-
55 Hypomelanosis of Ito continentia pigmenti achromians). Ophthalmological evi-
dence for somatic mosaicism. Ophthalmic Paediatr Genet
Ardinger HH, Bell WE. Hypomelanosis of Ito. Wood’s light and 1990; 4: 273–279
magnetic resonance imaging as diagnostic measures. Arch Ruggieri M, Tigano G, Mazzone D, Tine A, Pavone L. Involve-
Neurol 1986; 43: 848–850 ment of the white matter in hypomelanosis of Ito (inconti-
Auriemma A, Agostinis C, Bianchi P, Bellan C, Salvoni L, Manara nentia pigmenti achromiens). Neurology 1996; 46: 485–492
O, Colombo A. Hemimegalencephaly in hypomelanosis of Steiner J, Adamsbaum C, Desguerres I, Lalande G, Raynaud F,
Ito: early sonographic pattern and peculiar MR findings in a Ponsot G, Kalifa G. Hypomelanosis of Ito and brain abnor-
newborn. Eur J Ultrasound 2000; 12: 61–67 malities: MRI findings and literature review. Pediatr Radiol
Battistella PA, Peserico A, Bertoli P, Drigo P, Laverda AM, Casara 1996; 26: 763–768
GL. Hypomelanosis of Ito and hemimegalencephaly. Childs Tagawa T, Futagi Y, Arai H, Mushiake S, Nakayama M. Hypome-
Nerv Syst 1990; 6: 421–423 lanosis of Ito associated with hemimegalencephaly: a clini-
Bermejo F, Dooms G. Cerebral magnetic resonance imaging in copathological study. Pediatr Neurol 1997; 17: 180–184
a 2-year-old Caucasian girl, with hypomelanosis of Ito. Thomas IT, Frias JL, Cantu ES, Lafer CZ, Flannery DB, Graham JG
J Neuroradiol 1996; 23: 248–250 Jr.Association of pigmentary anomalies with chromosomal
Bhushan V, Gupta RR, Weinreb J, Kairam R. Unusual brain MRI and genetic mosaicism and chimerism. Am J Hum Genet
findings in a patient with hypomelanosis of Ito. Pediatr 1989; 45: 193–205
Radiol 1989; 20: 104–106 Traupe H. Functional X-Chromosomal mosaicism of the skin:
Echenne BP, Leboucq N, Humbertclaude V. Ito hypomelanosis Rudolf Happle and the lines of Alfred Blaschko. Am J Med
and moyamoya diasease. Pediatr Neurol 1995; 13: 169–171 Genet 1999; 85: 324–329
Fryburg JS, Lin KY, Matsumoto J. Abnormal head MRI in a neu- Tunca Y, Wilroy RS, Kadandale JS, Martens PR, Gunther WM,
rologically normal boy with hypomelanosis of Ito. Am J Tharapel AT. Hypomelanosis of Ito and a “mirror image”
Med Genet 1996; 66: 200–203 whole chromosome duplication resulting in trisomy 14
Fujino O, Hashimoto K, Fujita T, Enokido H, Komatsuzaki H, mosaicism. Ann Genet 2000; 43: 39–43
Asano G, Sato J, Morimatsu Y. Clinico-neuropathological Williams DW, Elster AD. Cranial MR imaging in hypomelanosis
study of incontinentia pigmenti achromians – an autopsy of Ito. J Comput Assist Tomogr 1990; 14: 981–983
case. Brain Dev 1995; 17: 425–427 Zappela M.Autism and hypomelanosis of Ito in twins.Dev Med
Glover MT, Brett EM, Atherton DJ. Hypomelanosis of Ito: spec- Child Neurol 1993; 35: 826–832
trum of the disease. J Pediatr 1989; 115: 75–80
Griebel V, Krägeloh-Mann I, Michaelis R. Hypomelanosis of Ito –
report of four cases and survey of the literature.Neuropedi-
atrics 1989; 20: 234–237 56 Incontinentia Pigmenti
Happle R. Mosaicism in human skin. Understanding the pat-
terns and mechanisms. Arch Dermatol 1993; 129: 1460 Aydingöz Ü, Midia N. Central nervous system involvement in
–1470 incontinentia pigmenti: cranial MRI of two siblings. Neuro-
Happle R. New aspects of cutaneous mosaicism. J Dermatol radiology 1998; 40: 364–366
2002; 29: 681–692 Chatkupt S, Gozo AO, Wolansky LJ, Sun S. Characteristic MR
Kimura M,Yoshino K,Maeoka Y,Suzuki N.Hypomelanosis of Ito: findings in a neonate with incontinentia pigmenti.Am J Ra-
MR findings. Pediatr Radiol 1994; 24: 68–69 diol 1993; 160: 372–374
Kuwahara RT, Henson T, Tunca Y, Wilroy SW. Hyperpigmenta- Cohen BA. Incontinentia pigmenti. Neurol Clin 1987; 5:
tion along the lines of Blaschko with associated chromo- 361–377
some 14 mosaicism. Pediatr Dermatol 2001; 18: 360–361 Fiorillo L, Sinclair DB, O’Byrne ML, Krol AL. Bilateral cerebrovas-
Malherbe V,Pariente D,Tardieu M,Lacroix C,Venencie PY,Hibon cular accidents in incontinentia pigment. Pediatr Neurol
D,Vedrenne J, Landrieu P. Central nervous system lesions in 2003; 29: 66–68
hypomelanosis of Ito: an MRI and pathological study.J Neu- Goldberg MF. The blinding mechanisms of incontinentia pig-
rol 1993; 240: 302–304 menti. Ophthalmic Genet 1994; 2: 69–76
Mendiratta V,Sharma RC,Arya L,Sardana K.Linear and whorled Goldberg MF, Custis PH. Retinal and other manifestations of in-
nevoid hypermelanosis. J Dermatol 2001; 28: 58–59 continentia pigmenti (Bloch-Sulzberger syndrome). Oph-
Montagna P, Procaccianti G, Galli G, Ripamonti L, Patrizi A, thalmology 1993; 100: 1645–1654
Baruzzi A. Familial hypomelanosis of Ito. Eur Neurol 1991; Hennel SJ, Ekert PG, Volpe JJ, Inder TE. Insights into the patho-
31: 345–347 genesis of cerebral lesions in incontinentia pigmenti: Pedi-
Pascual-Castroviejo I, López-Rodriquez L, de la Cruz Medina M, atr Neurol 2003; 29: 148–150
Salamanca-Maesso C, Herrero CR. Hypomelanosis of Ito. Kasai T, Kato Z, Matsui E, Sakai A, Nishida T, Kondo N, Taga T.
Neurological complications in 34 cases. Can J Neurol Sci Cerebral infarction in incontinentia pigmenti: the first re-
1988; 15: 124–129 port of a case evaluated by single photon emission com-
Pascual-Castroviejo I, Roche C, Martinez-Bermejo A, Arcas J, puted tomography. Acta Paediatr 1997; 86: 665–667
Lopez-Martin V,Tendero A, Esquiroz JLH, Pascual-Pascual S- Mangano S, Barbagallo A. Incontinentia pigmenti: clinical and
I. Hypomelanosis of Ito. A study 76 infantile cases. Brain Dev neuroradiologic features. Brain Dev 1993; 15: 362–366
1998; 20: 36–43
References and Further Reading 993

Pascual-Castroviejo I, Roche MC, Fernández VM, Perez-Romero Farrell K, Chuang S, Becker LE. Computed tomography in
M, Escudero RM, Garcia-Peñas JG, Sanchez M. Incontinentia Alexander’s disease. Ann Neurol 1984; 15: 605–607
pigmenti: MR demonstration of brain changes. AJNR Am J French TA, Bower BD, Cameron AH. Alexander’s disease pre-
Neuroradiol 1994; 15: 1521–1527 senting as astrocytoma. J Neurol Neurosurg Psychiatry
Pellegrino RJ, Shah AJ. Vascular occlusion associated with in- 1976; 39: 803–809
continentia pigmenti. Pediatr Neurol 1994; 10: 73–74 Friede RL. Alexander’s disease. Arch Neurol 1964; 11: 414–422
Sha SN,Gibbs S,Upton CJ,Pickworth FE,Garioch JJ.Incontinen- Friedman JH, Ambler M Progressive parkinsonism associated
tia pigmenti associated with cerebral palsy and cerebral with Rosenthal fibers: senile-onset Alexander’s disease?
leukomalacia: a case report and literature review. Pediatr Neurology 1992; 42: 1733–1735
Dermatol 2003; 20: 491–494 Garcia L, Gascon G, Ozand P, Yaish H. Increased intracranial
Shuper A, Bryan RN, Singer HS. Destructive encephalopathy in pressure in Alexander disease: a rare presentation of white-
incontinentia pigmenti: a primary disorder? Pediatr Neurol matter disease. J Child Neurol 1992; 7: 168–171
1990; 6: 137–140 Garret R,Ames RP.Alexander disease.Case report with electron
Siemes H, Schneider D, Dening D, Hanefeld F. Encephalitis in microscopical studies and review of the literature. Arch
two members of a family with incontinentia pigmenti Pathol 1974; 98: 379–385
(Bloch-Sulzberger syndrome). Eur J Pediatr 1978; 129: 102– Gingold MK, Bodensteiner JB, Schochet SS, Jaynes M. J Child
115 Neurol 1999; 14: 325–329
Tekin N, Uçar B, Saraçoglu ZN, Koçak AK, Ürer S, Yakut A. Diag- Goebel HH, Bode G. Ceasar R, Kohlschütter A. Bulbar palsy with
noses and follow up in four cases of incontinentia pigmen- Rosenthal fiber formation in the medulla of a 15-year-old
ti. Pediatr Int 2000; 42: 557–560 girl.Localized form of Alexander’s disease? Neuropediatrics
Yang J-H, Ma S-Y, Tsai C-H. Destructive encephalaopathy in in- 1981; 12: 382–391
continentia pigmenti: a case report. J Dermatol 1995; 22: Gorespe JR, Naidu S, Johnson AB, Puri V, Raymond GV, Jenkins
340–343 SD, Pedersen RC, Lewis D, Knowles P, Fernandez R, de Vivo D,
Yoshikawa H, Uehara Y, Abe T, Oda Y. Disappearance of a white van der Knaap MS, Messing A, Brenner M, Hoffman EP.
matter lesion in incontinentia pigmenti. Pediatr Neurol Molecular findings in symptomatic and pre-symptomatic
2000; 23: 364–367 Alexander disease patients.Neurology 2002; 58:1494–1500
Guthrie SO, Burton EM, Knowles P, Marshall R. Alexander’s dis-
ease in a neurologically normal child: a case report. Pediatr
Radiol 2003; 33: 47–49
57 Alexander Disease Habib M, Hassoun J, Ali-Cherif A, Alonzo B,Toga M, Khalil R.Mal-
adie d’Alexander de l’adulte.Rev Neurol 1984; 140:179–189
Aoki Y, Haginoya K, Munakata M,Yokoyama H, Nishio T,Tagashi Honnorat J, Flocard F, Ribot C, Saint-Pierre G, Pineau D, Peysson
N, Ito T, Suzuki Y, Kure S, Linuma K, Brenner M, Matsubara Y. P, Kopp N. Maladie d’Alexander de l’adulte et gliomatose
A novel mutation in glial fibrillary acidic protein gene in a cérébrale diffuse chez deux members d’une famille. Rev
patient with Alexander disease. Neurosci Lett 2001; 312: Neurol 1993; 149: 781–787
71–74 Imamura A, Orii KE, Mizuno S, Hoshi H, Kondo T. MR imaging
Bobele GB, Garnica A, Schaefer GB, Leonard JC,Wilson D, Marks and 1H-MR spectroscopy in a case of juvenile Alexander
WA, Leech RW, Brumback RA. Neuroimaging findings in disease. Brain Dev 2002; 24: 723–726
Alexander’s disease. J Child Neurol 1990; 5: 253–258 Jacob J, Robertson NJ, Hilton DA. Short report. The clinico-
Borrett D, Becker LE. Alexander’s disease. A disease of astro- pathological spectrum of Rosenthal fibre encephalopathy
cytes. Brain 1985; 108: 367–385 and Alexander’s disease: a case report and review of the lit-
Brenner M, Johnson AB, Boespflug-Tanguy O, Rodriquez D, erature. J Neurol Neurosurg Psychiatry 2003; 74: 807–810
Goldman JE, Messing A. Mutations in GFAP, encoding glial Johnson AB. Alexander disease: a review and the gene. Int J
fibrillary acidic protein, are associated with Alexander dis- Dev Neurosci 2002; 20: 391–394
ease. Nat Genet 2001; 27: 117–120 Johnson AB, Brenner M. Alexander’s disease: clinical, patholog-
Brockmann K, Dechent P, Meins M, Haupt M, Sperner J, ic, and genetic features. J Child Neurol 2003; 18: 625–632
Stephani U, Frahm J, Hanefeld F. Cerebral proton magnetic Klein EA, Anzil AP. Prominent white matter cavitation in an in-
resonance spectroscopy in infantile Alexander disease. fant with Alexander’s disease. Clin Neuropathol 1994; 13:
J Neurol 2003; 250: 300–306 31–38
Clifton AG, Kendall BE, Kingsley DPE, Cross JH, Andar U. Com- Li R, Messing A, Goldman JE, Brenner M. GFAP mutations in
puted tomography in Alexander’s disease. An atypical case Alexander disease. Int J Dev Neurosci 2002;20: 259–268
with extensive low density in both frontal lobes. Neurora- Liedtke W, Edelmann W, Bieri PL, Chiu F-C, Cowan NJ, Kucher-
diology 1991; 33: 438–440 lapati R, Raine CS. GFAP is necessary for the integrity of CNS
Cole G, de Villiers F, Proctor NSF, Freiman I, Bill P.Alexander’s dis- white matter architecture and long-term maintenance of
ease: case report including histopathological and electron myelination. Neuron 1996; 17: 607–615
microscopic features. J Neurol Neurosurg Psychiatry 1979; Madsen JR, Partington MD, Hay TC, Tyson RW. A 2-month-old
42: 619–624 female infant with progressive macrocephaly and irritabili-
Crome L. Megalencephaly associated with hyaline pan-neu- ty. Pediatr Neurosurg 1999; 30: 157–163
ropathy. Brain 1953; 76: 215–228 Martidis A,Yee RD, Azzarelli B, Biller J. Neuro-ophthalmic, radio-
Deprez M, D’Hooge M, Misson JP, de Leval L, Ceuterick C, Reznik graphic, and pathologic manifestations of adult-onset
M, Martin JJ. Infantile and juvenile presentations of Alexan- Alexander disease. Arch Ophthalmol 1999; 117: 265–267
der’s disease:a report of two cases.Acta Neurol Scand 1999; Mastri AR, Sung JH. Diffuse Rosenthal fiber formation in the
99: 158–165 adult:a report of four cases.J Neuropathol Exp Neurol 1973;
Duckett S, Schwartzman RJ, Osterholm J, Rorke LB, Friedman D, 32: 424–436
McLellan TL. Biopsy diagnosis of familial Alexander’s dis-
ease. Pediatr Neurosurg 1992; 18: 134–138
994 References and Further Reading

Meins M, Brockmann K, Yadav S, Haupt M, Sperner J, Stephani Shah M, Ross JS. Infantile Alexander disease: MR appearance of
U, Hanefeld F. Infantile Alexander disease: a GFAP mutation a biopsy-proved case. AJNR Am J Neuroradiol 1990; 11:
in monozygotic twins an novel mutations in two other 1105–1106
patients. Neuropediatrics 2002; 33: 194–198 Sherwin RM, Berthrong M. Alexander’s disease with su-
Messing A, Head MW, Galles K, Galbreath EJ, Goldman JE, Bren- danophilic leukodystrophy. Arch Pathol 1970; 89: 321–328
ner M. Fatal encephalopathy with astrocytes inclusions in Shiroma N, Kanazawa N, Izumi M, Sugai K, Fukumizu M, Sasaki
GFAP transgenic mice. Am J Pathol 1998; 152: 391–398 M,Hanaoka S,Kaga M,Tsujino S.Diagnosis of Alexander dis-
Messing A, Goldman JE, Johnson AB, Brenner M. Alexander dis- ease in a Japanese patient by molecular genetic analysis.
ease: new insights from genetics. J Neuropathol Exp Neurol J Hum Genet 2001; 46: 579–582
2001; 60: 563–573 Soffer D, Horoupian DS. Rosenthal fibers formation in the cen-
Mignot C, Boespflug-Tanguy O, Gelot A, Dautigny A, Pham- tral nervous system. Its relation to Alexander’s disease. Acta
Dinh D, Rodriguez D. Alexander disease: putative mecha- Neuropathol (Berl) 1979; 47: 81–84
nisms of an astrocytic encephalopathy. Cell Mol Life Sci Spalke G, Mennel HD. Alexander’s disease in an adult: clinico-
2004; 61: 369–385 pathologic study of a case and review of the literature. Clin
Namekawa M, Takiyama Y, Aoki Y, Takayashiki N, Sakoe K, Shi- Neuropathol 1982; 1: 106–112
mazaki H,Taguchi T,Tanaka Y, Nishizawa M, Saito K, Matsub- Springer S, Erlewein R, Naegele T, Becker I, Auer D, Grodd W,
ara Y, Nakano I. Identification of GFAP gene mutation in Krägeloh-Mann I. Alexander disease – classification revisit-
hereditary adult-onset Alexander’s disease. Ann Neurol ed and isolation of a neonatal form. Neuropediatrics 2000;
2002; 52: 779–785 31: 86–92
Neal JW, Cave EM, Singhrao SK, Cole G, Wallace SJ. Alexander’s Stumpf E, Masson H, Duquette A, Berthelet F, McNabb J, Lortie
disease in infancy and childhood: a report of two cases. A, Lesage J, Montplaisir J, Brais B, Cossette P. Adult Alexan-
Acta Neuropathol (Berl) 1992; 84: 322–327 der disease with autosomal dominant transmission. A dis-
Neumaier Probst E, Hagel C, Weisz V, Nagel S, Wittkugel O, tinct entity caused by mutation in the glial fibrillary acid
Zeumer H, Kohlschütter A. Atypical focal MRI lesions in a protein gene. Arch Neurol 2003; 60: 1307–1312
case of juvenile Alexander’s disease. Ann Neurol 2003; 53: Takanashi J-I, Sugita K, Tanabe Y, Niimi H. Adolescent case of
118–120 Alexander disease: MR imaging an MR spectroscopy. Pedi-
Ni Q, Johns GS, Manepalli A, Martin DS, Geller TJ, Infantile atr Neurol 1998; 18: 67–70
Alexander’s disease: serial neuroradiologic findings. J Child Torreman M, Smit LME, van der Valk P, Valk J, Scheltens Ph. A
Neurol 2002; 17: 463–466 case of macrocephaly, hydrocephalus, megacerebellum,
Okamoto Y, Mitsuyama H, Jonosono M, Hirata K, Arimura K, white matter abnormalities and Rosenthal fibres. Dev Med
Osame M, Nakagawa M. Autosomal dominant palatal myo- Child Neurol 1993; 35: 732–736
clonus and spinal cord atrophy. J Neurol Sci 2002; 195: Towfighi J, Young R, Sassani J, Ramer J, Horoupian DS. Alexan-
71–76 der’s disease: further light- and electron-microscopic ob-
Pridmore CL, Baraitser M, Harding B, Boyd SG, Kendall B, Brett servations. Acta Neuropathol (Berl) 1983; 61: 36–42
EM. Alexander’s disease: clues to diagnosis. J Child Neurol van der Knaap MS, Naidu S, Breiter SN, Blaser S, Stroink H,
1993; 8: 134–144 Springer S, Begeer JC, van Coster R, Barth PG, Thomas NH,
Reichard EAP, Ball WS Jr, Bove KE. Alexander disease: a case re- Valk J, Powers JM. Alexander disease: diagnosis with MR
port and review of the literature. Pediatr Pathol Lab Med imaging. AJNR Am J Neuroradiol 2001; 22: 541–552
1996; 16: 327–343 Vogel PS, Hallervorden J. Leukodystrophy with diffuse Rosen-
Riggs JE, Schochet SS Jr, Nelson J. Asymptomatic adult Alexan- thal fiber formation. Acta Neuropathol (Berl) 1962; 2:
der’s disease: entity or nosological misconception? Neuro- 126–143
logy 1988; 38: 152–154 Walls TJ, Jones RA, Cartlidge NEF, Saunders M. Alexander’s dis-
Rodriguez D, Gauthier F, Bertini E, Bugiani M, Brenner M, ease with Rosenthal fibre formation in an adult. J Neurol
N’guyen S, Goizet C, Gelot A, Surtees R, Predespan J-M, Her- Neurosurg Psychiatry 1984; 47: 399–403
nandorena X, Troncoso M, Uziel G, Messing A, Ponsot G, Wohlwill FJ, Bernstein J, Yakovlev PI. Dysmyelinogenic leuko-
Pham-Dinh D, Dautigny A, Boespflug-Tanguy O. Infantile dystrophy. J Neuropathol Exp Neurol 1959; 18: 359–383
Alexander disease: spectrum of GFAP mutations and geno-
type-phenotype correlation. Am J Hum Genet 2001; 69:
1134–1140
Russo LS Jr, Aron A, Anderson PJ. Alexander’s disease: a report
58 Giant Axonal Neuropathy
and reappraisal. Neurology 1976; 26: 607–614
Sawaishi Y, Yano T, Takaku I, Takada G. Juvenile Alexander dis- Bomont P, Cavalier L, Blondeau F, Ben Hamida C, Belal S,Tazir M,
ease with a novel mutation in glial fibrillary acidic protein Demir E, Topaloglu H, Koninthenberg R,Tüysüz B, Landrieu
gene. Neurology 2002; 58: 1541–1543 P, Hentati F, Koenig M.The gene encoding gigaxonin, a new
Schochet CSS Jr, Lampert PW, Earle KM. Alexander’s disease. A member of the cytoskeletal BTB / kelch repeat family, is
case report with electron microscopic observations.Neuro- mutated in giant axonal neuropathy. Nat Genet 2000; 26:
logy 1968; 18: 543–549 370–374
Schuster V, Horwitz AE, Kreth HW. Alexander’s disease: cranial Bomont P, Ioos C, Yalcinkaya C, Korinthenberg R, Vallat JM,
MRI and ultrasound findings. Pediatr Radiol 1991; 21: 133– Assami S, Munnich A, Chabrol B, Kurlemann G, Tazir M,
134 Koenig M. Identification of seven novel mutations in the
Schwankhaus JD, Parisi JE, Gulledge WR, Chin L, Currier RD. GAN gene. Hum Mutat 2003; 21: 446
Hereditary adult-onset Alexander’s disease with palatal Brockmann K, Pouwels PJW, Dechent P, Flanigan KM, Frahm J,
myoclonus, spastic paraparesis, and cerebellar ataxia. Neu- Hanefeld F. Cerebral proton magnetic resonance spec-
rology 1995; 45: 2266–2271 troscopy of a patient with giant axonal neuropathy. Brain
Seil FJ,Schochet SS Jr,Earle KM.Alexander’s disease in an adult. Dev 2003; 25: 45–50
Report of a case. Arch Neurol 1968; 19: 494–502
References and Further Reading 995

Bruno C, Bertini E, Federico A, Tonoli E, Lispi ML, Cassandrini D, 59 Megalencephalic Leukoencephalopathy


Pedemonte M, Santorelli FM, Filocamo M, Dotti MT, with Subcortical Cysts
Schenone A, Malandrini A, Minetti C. Clinical and molecular
findings in patients with giant axonal neuropathy (GAN).
Aoki K, Uchihara T,Tsuchiya K, Nakamura A, Ikeda K,Wakayama
Neurology 2004; 62: 13-16
Y.Enhanced expression of aquaporin 4 in human brain with
Ding J, Liu JJ, Kowal AS, Nardine T, Bhattacharya P, Lee A,Yang Y.
infarction. Acta Neuropathol (Berl) 2003; 106: 121–124
Microtubule-associated protein 1B: a neuronal binding
Ben-Zeev B, Gross V, Kushnir T, Shalev R, Hoffman C, Shinar Y,
partner for gigaxonin. J Cell Biol 2002; 158: 427–433
Pras E, Brand N. Vacuolating megalencephalic leukoence-
Donaghy M, Brett EM, Ormerod IEC, King RHM. Giant axonal
phalopathy in 12 Israeli patients. J Child Neurol 2001; 16:
neuropathy: observations on a further patient. J Neurol
93–99
Neurosurg Psychiatry 1988; 51: 991–994
Ben-Zeev B, Levy-Nissenbaum E, Lahat H, Anikster Y, Shinar Y,
Griffiths IR, Duncan ID. The central nervous system in canine
Brand N, Gross-Tzur V, MacGregor D, Sidi R, Kleta R, Frydman
giant axonal neurophathy. Acta Neuropathol (Berl) 1979;
M, Pras E.Megalencephalic leukoencephalopathy with sub-
46: 169–172
cortical cysts; a founder effect in Israeli patients and a high-
Igisu H, Ohta M, Tabira T, Hosokawa S, Goto I, Kuroiwa Y. Giant
er than expected carrier rate among Libyan Jews. Hum
axonal neuropathy; a clinical entity affecting the central as
Genet 2002; 111: 214–218
well as the peripheral nervous system. Neurology 1975; 25:
Bešenski N, Bošnjak V, Cop S, Pavic D, Mikulic D, Oroliæ K. Neu-
717–721
roimaging and clinically distinctive features in van der
Kretzschmar HA, Berg BO, Davis RL. Giant axonal neuropathy.
Knaap megalencephalic leukoencephalopathy.Int J Neuro-
Acta Neuropathol (Berl) 1987; 73: 138–144
radiol 1997; 3: 244–249
Kuhlenbäumer G,Young P, Oberwittler C, Hünermund G, Schir-
Biancheri R, Pisaturo C, Perrone MV, Passagno A, Rossi A, Vene-
macher A, Domschke K, Ringelstein B, Stögbauer F. Giant
selli E. Presence of delayed myelination and macrocephaly
axonal neuropathy (GAN): case report and two novel muta-
in the sister of a patient with vacuolating leukocen-
tions in the gigaxonin gene. Neurology 2002; 58: 1273–
cephalopathy with subcortical cysts. Neuropediatrics 2000;
1276
31: 321–324
Kumar K, Barre P, Nigro M, Jones MZ. Giant axonal neuropathy:
Blattner R, von Moers A, Leegwater PAJ, Hanefeld FA, van der
clinical, electrophysiologic, and neuropathologic features
Knaap MS, Köhler W. Clinical and genetic heterogeneity in
in two siblings. J Child Neurol 1990; 5: 229–234
megalencepahlic leukoencephalopathy with subcortical
Lampl Y, Eshel Y, Ben-David E, Gilad R, Sarova-Pinhas I, Sand-
cysts (MLC). Neuropediatrics 2002; 34: 215–218
bank U. Giant axonal neuropathy with predominant central
Boor PKI, de Groot K, Waisfisz Q, Kamphorst W, Oudejans CBM,
nervous system manifestations. Dev Med Child Neurol
Powers JM, Pronk JC, Scheper GC, van der Knaap MS. MLC1:
1992; 34: 164–181
a novel protein in distal astroglial processes. J Neuropathol
Majnemer A, Rosenblatt B, Watters G, Andermann F. Giant
Exp Neurol 2005 (in press)
axonal neuropathy: central abnormalities demonstrated by
Brockmann K, Finsterbusch J, Terwey B, Frahm J, Hanefeld F.
evoked potentials. Ann Neurol 1986; 19: 394–396
Megalencephalic leukoencephalopathy with subcortical
Malandrini A, Dotti MT, Battisti C, Villanova M, Capocchi G,
cysts in an adult: quantitative proton MR spectroscopy and
Federico A. Giant axonal neuropathy with subclinical in-
diffusion tensor MRI. Neuroradiology 2003; 45: 137–142
volvement of the central nervous system (case report).
Bugiani M, Moroni I, Bizzi A, Nardocci N, Bettecken T, Gärtner J,
J Neurol Sci 1998; 158: 232–235
Uziel G. Consciousness disturbances in megalencephalic
Ouvrier RA. Giant axonal neuropathy – a review. Brain Dev
leukoencephalopathy with subcortical cysts. Neuropedi-
1989; 11: 207–214
atrics 2003; 34: 211–214
Peiffer J, Schlote W, Bischoff A, Boltshauser E, Müller G. General-
Chandrashekar HS, Guruprasad AS, Jayakumar PN, Srikanth SG,
ized giant axonal neuropathy – a filament-forming disease
Taly AB. Megalencephalic leukoencephalopathy with sub-
of neuronal, endothelial, glial, and schwann cells in a pa-
cortical cysts: MRI and proton spectroscopic features. Neu-
tient without kinky hair. Acta Neuropathol (Berl) 1977; 40:
rol India 2003; 51: 525–527
213–218
De Stefano N, Balestri P, Dotti MT, Grosso S, Mortilla M, Morgese
Richen P, Tandan R. Giant axonal neuropahty: progressive clin-
G, Frederico A. Severe metabolic abnormalities in the white
ical and radiologic CNS involvement. Neurology 1992; 42:
matter of patients with vacuolating megalencephalic
2220–2222
leukoencephalopathy with subcortical cysts. A proton MR
Stollhoff K, Albani M, Goebel HH. Giant axonal neuropathy and
spectroscopic imaging study. J Neurol 2001; 248: 403–409
leukodystrophy. Pediatr Neurol 1991; 7: 69–71
Gelal F, Call C, Apaydin M, Erdem G. Van der Knaap’s leukoen-
Thomas C, Love S, Powell HC, Schultz P, Lampert PW. Giant
cephalopathy: report of five new cases with emphasis on
axonal neuropathy: correlation of clinical findings with
diffusion-weighted MRI findings. Neuroradiology 2002; 4:
postmortem neuropathology. Ann Neurol 1987; 22: 79–84
625–630
Treiber-Held S, Budjarjo-Welim H, Riemann D, Richter J, Kret-
Gorospe JR, Singhal BS, Kainu T, Wu F, Stephan D, Trent J, Hoff-
zschmar HA, Hanefeld F. Giant axonal neuropathy: a gener-
man EP, Naidu S. Indian Agarwal megalencephalic leukody-
alized disorder of intermediate filaments with longitudinal
strophy with cysts is caused by a common MLC1 mutation.
grooves in the hair. Neuropediatrics 1994; 25: 89–93
Neurology 2004; 62: 878–882
Goutières F, Boulloche J, Bourgeois M, Aicardi J. Leukoen-
cephalopathy, megalencephaly, and mild clinical course. A
recently individualized familial leukodystrophy. Report of
five new cases. J Child Neurol 1996; 11: 439–444
Gulati S, Kabra M, Gera S, Ghosh M, Menon PSN, Kalra V. Infan-
tile-onset leukoencephalopathy with discrepant mild clini-
cal course. Indian J Pediatr 2000; 67: 769–773
996 References and Further Reading

Harbord MG, Harden A, Harding B, Brett EM, Baraitser M. Mega- Singhal BS, Gursahani RD, Udami VPk Biniwale AA. Megalen-
lencephaly with dysmyelination, spasticity, ataxia, seizures cephalic leukodystrophy in an Asian Indian Ethnic Group.
and distinctive neurophysiological findings in two siblings. Pediatr Neurol 1996; 14: 291–296
Neuropediatrics 1990; 21: 164–168 Singhal BS, Gorospe JR, Naidu S. Megalencephalic leukoen-
Higuchi Y,Hattori H,Tsuji M,Asato R,Nakahata T.Partial seizures cephalopathy with subcortical cysts. J Child Neurol 2003;
in leukoencephalopathy with swelling and a discrepantly 18: 646–652
mild clinical course. Brain Dev 2000; 22: 287–389 Takanashi J-I, Sugita K, Kohno Y. Vacuolating leukoencephalo-
Koeda T, Takeshita K. Slowly progressive cystic leukoen- pathy with subcortical cysts with late onset athetotic
cephalopathy with megalencephaly in a Japanese boy. movements. J Neurol Sci 1999; 165: 90–93
Brain Dev 1998; 20: 245–249 Teijido O, Martinez A, Pusch M, Zorzano A, Soriano E, Del Rio JA,
Leegwater PAJ, Qiang Yuan B, van der Steen J, Mulders J, Könst Palacin M,Estevez R.Localization and functional analyses of
AAM, Ilja Boor PK, Mejaski-Bosnjak V, Van der Maarel SM, the MLC1 protein involved in megalencephalic leukoen-
Frants RR, Oudejans CBM, Schutgens RBH, Pronk JC, van der cephalopathy with subcortical cysts. Hum Mol Genet 2004;
Knaap MS. Mutations of MCL1 (KIAA0027), encoding a puta- 13: 2581–2594
tive membrane protein cause megalencephalic leukoen- Thele T, Balslev T, Christenen T. Van der Knaap’s vacuolating
cephalopathy with subcortical cysts. Am J Hum Genet leukoencephalopathy: two additional cases. Eur J Pediatr
2001; 68: 831–838 Neurol 1999; 3: 83–86
Leegwater PAJ, Boor PKI, Yuan BQ, van der Steen J, Visser A, Topçu M, Saatci I, Topcuoglu MA, Kose G, Kunak B. Megalen-
Könst AAM, CBM Oudejans, Schutgens RBH, Pronk JC, van cephaly and leukodystrophy with mild clinical course: a re-
der Knaap MS. Identifications of novel mutations in MLC1 port on 12 new cases. Brain Dev 1998; 20: 142–153
responsible for megalencephalic leukoencephalopathy Topçu M, Gartioux C, Ribierre F,Yalçinkaya C,Tokus E, Öztekin N,
with subcortical cysts. Hum Genet 2002; 110: 279–283 Beckmann JS, Ozguc M, Seboun E. Vacuolating megalen-
Mejaški-Bošnjak V, Beenski N. Megalencephalic leukoen- cephalic leukoencephalopathy with subcortical cysts,
cephalopathy: a further case of a new neurodegenerative mapped to chromosome 22qtel. Am J Hum Genet 2000; 66:
white matter disease. Dev Med Child Neurol 1997; 39: 733–739
561–563 Tsujino S, Kanazawa N, Yoneyama H, Shimono M, Kawakami A,
Mejaški-Bošnjak V, Bešenski N, Brockmann K, Pouwels PJW, Hatanaka Y, Shimizu T, Oba H. A common mutation and a
Frahm J, Hanefeld FA. Cystic leukoencephalopathy in a novel mutation in Japanese patients with van der Knaap
megalencephalic child: clinical and magnetic resonance disease. J Hum Genet 2003; 48: 605–608
imaging/magnetic resonance spectroscopy findings. Pedi- van der Knaap MS, Barth PG, Stroink H, van Nieuwenhuizen O,
atr Neurol 1997; 16: 347–350 Arts WFM,Hoogenraad F,Valk J.Leukoencephalopathy with
Neely JD, Amiry-Moghaddam M, Ottersen OP, Froehner SC, swelling and a discrepantly mild clinical course in 8 chil-
Agre P, Adams ME. Syntrophin-dependent expression and dren. Ann Neurol 1995; 37: 324–334
localization of aquaporin-4 water channel protein. Proc van der Knaap, Valk J, Barth PG, Smit LME, Van Engelen BGM,
Natl Acad Sci USA 2001; 98: 14108–14113 Tortori Donati P. Leukoencephalopathy with swelling in
Patrono C, di Giacinto G, Eymard-Pierre E, Santorelli FM, Ro- children and adolenscents: MRI pattern and differential di-
driguez D, de Stefano N, Federico A, Gatti R, Benigo V, agnosis. Neuroradiology 1995; 37: 679–686
Megarbané A, Tabarki B, Boespflug-Tanguy O, Bertini E. van der Knaap, Barth PG, Vrensen GFJM, Valk J. Histopathology
Genetic heterogeneity of megalencephalic leukoence- of an infantile-onset spongiform leukoencephalopathy
phalopathy with subcortical cysts. Neurology 2003; 61: with a discrepantly mild clinical course. Acta Neuropathol
534–537 (Berl) 1996; 92: 206–212
Rubie C, Lichtner P, Gärtner J, Siekiera M, Uziel G, Kohlmann B, Yakinci C, Soylu H, Kutlu NO, Sener RN. Leukoencephalopathy
Kohlschütter A,Meitinger T, Stöber G, Bettecken T. Se- with a mild clinical course: a case report. Comput Med
quence diversity of KIAA0027/MLC1: are megalencephalic Imaging Graph 1999; 23: 169–172
leukoencephalopathy and schizophrenia allelic disorders? Yalçinkaya C, Çomu S, Koçer N, Yüksel A, Güdüz E, Dermirbilek
Hum Mutat 2002; 21: 45–53 V, Öcal A. Siblings with cystic leukoencephalopathy and
Saijo H, Nakayama H, Ezoe T, Araki K, Sone S, Hamaguchi H, megalencephaly. J Child Neurol 2000; 15: 690–693
Suzuki H, Shiroma N, Kanazawa N, Tsujino S, Hirayama Y, Yalçinkaya C, Yüksel A, Çomu S, Kiliç G, Çokar Ö, Dervent A.
Arima M. A case of megalencephalic leukoencephalopathy Epilepsy in vacuolating megalencephalic leukoencephalo-
with subcortical cysts (van der Knaap disease): molecular pathy with subcortical cysts. Seizure 2003; 12: 388–396
genetic study. Brain Dev 2003; 25: 362–366
Schmitt A, Gofferje V, Weber M, Meyer J, Mössner R, Lesch K-P.
The brain-specific protein MLC1 implicated in megal-
encephalic leukoencephalopathy with subcortical cysts is
60 Congenital Muscular Dystrophies
expressed in glial cells in the murine brain. Glia 2003; 44: Walker-Warburg Syndrome
283–295
Sener RN. Van der Knaap syndrome: MR imaging findings in- Asano Y, Minagawa K, Okuda A, Matsui T, Ando K, Kondo-Iida E,
cluding FLAIR, diffusion imaging, and proton MR spec- Kobayashi O,Toda T, Nonaka I, Tanizawa T. A case of Walker-
troscopy. Eur Radiol 2000; 10: 1452–1455 Warburg syndrome. Brain Dev 2000; 22: 454–457
Shinar Y, Ben-Zeev B, Brand N, Lahat H, Gross-Zur V, MacGregor Barkovic AJ. Neuroimaging manifestations and classification
D, Bahan T, Kastner DL, Pras E. A common ancestral haplo- of congenital muscular dystrophies. AJNR Am J Neuroradi-
type in carrier chromosomes form different ethnic back- ol 1998; 19: 1389–1396
grounds in vacuolating megalencephalic leukoencephalo-
pathy with subcortical cysts. J Med Genet 2002; 39: 54–57
References and Further Reading 997

Bertrán-Valero de Bernabé D, Currier S, Steinbrecher A, Celli J, Voit T,Sewry CA,Meyer K,Hermann R,Straub V,Muntoni F,Kahn
van Beusekom E, van der Zwaag B, Kayserili H, Merlini L, T, Unsold R, Helliwell TR, Appleton R, Lenard HG. Preserved
Chitayat D, Dobyns WB, Cormand B, Lehesjoki A-E, Cruces J, merosin M-chain (or laminin-a2) expression in skeletal
Voit Th, Walsh CA, van Bokhoven H, Brunner HG. Mutations muscle distinguishes Walker-Warburg syndrome from
in the O-mannosyltransferase gene POMT1 give rise to the Fukuyama muscle dystrophy and merosin-deficient con-
severe neuronal migration disorder Walker-Warburg syn- genital muscular dystrophy. Neuropediatrics 1995; 26:
drome. Am J Hum Genet 2002; 71: 1033–1043 148–155
Di Rocco M, Leveratto L, Cama A, Bado M, Tortori Donati P, Williams RS, Swisher CN, Jennings M, Ambler M, Caviness Jr VS.
Andreussi L, Borrone C. Report on a patient with congenital Cerebro-ocular dysgenesis (Walker-Warburg syndrome):
muscular distrophy, hydrocephalus, Dandy-Walker malfor- neuropathologic and etiologic analysis. Neurology 1984;
mation and leukodystrophy. Genet Couns 1993; 4: 295–298 34: 1531–1541
Dobyns WB, Kirkpatrick JB, Hittner HM, Roberts RM, Kretzer FL. Zaleski CG, Abdenour GE. Pediatric case of the day. Radio-
Syndromes with lissencephaly.II.Walker-Warburg and cere- graphics 1997; 17: 1319–1323
bro-oculo-muscular syndromes and a new syndrome with
type II lissencephaly. Am J Med Genet 1985; 22: 157–195
Dobyns WB, Pagon RA, Armstrong D, Curry CJR, Greenberg F, Fukuyama Type
Grix A, Holmes LB, Laxova R, Michels VV, Robinow M, Zim- of Congenital Muscular Dystrophy
merman RL. Diagnostic criteria for Walker-Warburg syn-
drome. Am J Med Genet 1989; 32: 195–210 Aida N. Fukuyama congenital muscular dystrophy: a neurora-
Heyer R, Ehrich J, Goebel HH, Christen HJ, Hanefeld F. Congeni- diologic review. J Magn Reson Imaging 1998; 8: 317–326
tal muscular dystrophy with cerebral and ocular malforma- Aida N, Yagishita A, Takada K, Katsumata Y. Cerebellar MR in
tions (cerebro-oculo-muscular syndrome). Brain Dev 1986; Fukuyama congenital muscular dystrophy: polymicrogyria
8: 614–619 with cystic lesions. AJNR Am J Neuroradiol 1994; 15: 1755–
Kanoff RJ, Curless RG, Petito C, Falcone S, Siatkowski RM, Pego- 1759
raro E. Walker-Warburg syndrome: neurologic features and Aida N,Tamagawa K,Takada K,Yagishita A, Kobayashi N, Chiku-
muscle membrane structure. Pediatr Neurol 1998; 18: maru K, Iwamoto H. Brain MR in Fukuyama congenital mus-
76–80 cular dystrophy. AJNR Am J Neuroradiol 1996; 17: 605–613
Kim DS, Hayashi YK, Matsumoto H, Ogawa M, Noguchi S, Mu- Aihara M, Tanabe Y, Kato K. Serial MRI in Fukuyama type con-
rakami N, Sakuta R, Michozuki M, Michele DE, Campbell KP, genital muscular dystrophy. Neuroradiology 1992; 34: 396–
Nonaka I, Nishino I. POMT1 mutation results in defective 398
glycosylation and loss of laminin-binding activity in a-DG. Aravind L, Koonin EV. The fukutin protein family – predicted
Neurology 2004; 62: 1009–1011 enzymes modifying cell-surface molecules. Curr Biol 1999;
Kimura S, Sasaki Y, Kobayashi Y, Ohtsuki N, Tanaka Y, Hara M, 9: R836-R387
Miyake S, Yamada M, Iwamoto H, Misugi N. Fukuyama-type Beltrán-Valero de Barnabé D, van Bokhoven H, van Beusekom
congenital muscular dystrophy and the Walker-Warburg E, van den Akker W, Kant S, Dobyns WB, Cormand B, Currier
syndrome. Brain Dev 1993;15:182–191 S, Hamel B, Talim B, Topaloglu H, Brunner HG. A homo-
Kükner S, Gürer Y, Saatçi I, Akçören Z, Topaloğlu H. Laminin-a2 zygous nonsense mutation in the Fukutin gene causes a
chain (merosin M) is preserved in the Walker-Warburg syn- Walker-Warburg syndrome phenotype. J Med Genet 2003;
drome. Neuropediatrics 1996; 27: 279–280 40: 845–848
Pabuöçu Y, Bulakbaöý N, Koaoğlu M, Uçöz T. Walker-Warburg Chijiiwa T, Nishimura M, Inomata H, Yamana T, Yamana T,
syndrome variant. Comput Med Imaging Graph 2002; 26: Narazaki O, Kurokawa T. Ocular manifestations of congeni-
453–458 tal muscular dystrophy (Fukuyama type). Ann Ophthalmol
Rhodes RE, Hatten HP, Ellington KS.Walker-Warburg syndrome. 1983; 15: 921–923 and 926–928
AJNR Am J Neuroradiol 1992; 13: 123–126 Colombo R, Bignamini AA, Carobene A, Sasaki J, Tachikawa M,
Sabatelli P, Columbaro M, Mura I, Capanni C, Lattanzi G, Maraldi Kobayashi K,Toda T.Age and origin of the FCMD 3’-untrans-
NM, Beltràn-Valero de Barnabè D, van Bokhoven H, Squar- lated gene-region retrotransposal insertion mutation caus-
zoni S, Merlini L. Extracellular matrix and nuclear abnormal- ing Fukuyama-type congenital muscular dystrophy in the
ities in skeletal muscle of a patient with Walker-Warburg Japanese population. Hum Genet 2000; 107: 559–567
syndrome caused by POMT1 mutation. Biochim Biophys Fukuyama Y, Ohsawa M. A genetic study of the Fukuyama type
Acta 2003; 1638: 57–62 congenital dystrophy. Brain Dev 1984; 6: 373–390
Simma B, Felber S, Maurer H, Gassner I, Krassnitzer S. MR and ul- Fukuyama Y, Osawa M, Suzuki H. Congenital progressive mus-
trasound findings in a case of cerebro-oculo-muscular-syn- cular dystrophy of the Fukuyama type – clinical, genetic
drome. Pediatr Radiol 1990; 20: 554–555 and pathological considerations. Brain Dev 1981; 3: 1–29
Towfighi J, Sassani JW, Suzuki K, Ladda RL. Cerebro-ocular dys- Hayashi YK, Engvall E, Arikawa-Hirasawa E, Goto K, Koga R, Non-
plasia – muscular dystrophy (COD-MD) syndrome. Acta aka I, Sugita H, Arahata K. Abnormal localization of laminin
Neuropathol (Berl) 1984; 65: 110–123 subunits in muscular dystrophies. J Neurol Sci 1993; 119:
Vajsar J, Ackerley C, Chitayat D, Becker LE. Basal lamina skeletal 53–64
muscle of Walker-Warburg syndrome. Pediatr Neurol 2000; Hayashi YK, Ogawa W, Tagawa K, Noguchi S, Ishihara T, Nonaka
22: 139–143 I, Arahata K. Selective deficiency of a-dystroglycan in
Villanova M, Sabatelli P, He Y, Malandrini A, Petrini S, Maraldi Fukuyama-type congenital muscular dystrophy. Neurology
NM, Merlini L. Immunofluorescence study of a muscle biop- 2001; 57: 115–121
sy from a 1-year-old patient with Walker-Warburg syn- Ishii H, Hayashi YK, Nonaka I, Arahata K. Electron microscopic
drome. Acta Neuropathol (Berl) 1998; 96: 651–654 examination of basal lamina in Fukuyama congenital mus-
cular dystrophy. Neuromusc Disord 1997; 7: 191–197
998 References and Further Reading

Kato T, Funahashi M, Matsui A,Takashima S, Suzuki Y. MRI of dis- Toda T, Segawa M, Nomura Y, Nonaka I, Masuda K, Ishihara T,
seminated development dysmyelination in Fukuyama type Suzuki M, Tomita I, Origuchi Y, Ohno K, Misugi N, Sasaki Y,
of CMD. Pediatr Neurol 2000; 23: 385–388 Takada K, Kawai M, Otani K, Murakami T, Saito K, Fukuyama
Kihira S, Nonaka I. Congenital muscular dystrophy. A histo- Y, Shimizu T, Kanazawa I, Nakamura Y. Localization of a gene
chemical study with morphometric analysis on biopsied for Fukuyama type congenital muscular dystrophy to chro-
muscles. J Neurol Sci 1985; 70: 139–149 mosome 9q31–33. Nat Genet 1993; 5: 283–286
Kobayashi K, Nakahori Y, Miyake M, Matsumara K, Kodo-Lida E, Toda Y,Watanabe T, Matsumura K, Sunada Y,Yamada H, Nakano
Nomura Y, Segawa M, Yoshioka M, Saito K, Osawa M, I, Mannen T, Kanazawa I, Shimizu T. Three-dimension MR
Hamano K, Sakikahara Y, Nonaka I, Nakagome Y, Kanazawa I, imaging of brain surface anomalies in Fukuyama-type con-
Nakamura Y,Tokunaga K,Toda T. An ancient retrotransposal genital muscular dystrophy. Muscle Nerve 1995; 18: 508–
insertion causes by Fukuyama-type congenital muscular 517
dystrophy. Nature 1998; 394: 388–392 Toda T, Kobayashi K, Kondo-Iida E, Sasaki J, Nakamura Y. The
Kumada S,Tsuchiya K,Takahasi M,Takesue M, Shiotsu H, Nomu- Fukuyama congenital muscular dystrophy story. Neuro-
ra Y, Segawa M, Ikeda K, Hayashi M. The cerebellar and thal- musc Disord 2000; 10: 153–159
amic degeneration in Fukuyama-type congenital muscular Tsutsumi A, Uchida Y, Osawa M, Fukuyama Y. Ocular finings in
dystrophy. Acta Neuropathol (Berl) 2000; 99: 209–213 Fukuyama type congenital muscular dystrophy. Brain Dev
Nakano I, Funahashi M, Takada K, Toda T. Are breaches in the 1989; 11: 413–419
glia limitans the primary cause of the micropolygyria in Wang Z-P, Osawa M, Fukuyama Y. Morphometric study of the
Fukuyama-type congenital muscular dystrophy (FCMD)? – corpus callosum in Fukuyama type congenital muscular
Pathological study of the cerebral cortex of an FCMD fetus. dystrophy by magnetic resonance imaging. Brain Dev
Acta Neuropathol (Berl) 1996; 91: 313–321 1995; 17: 104–110
Osawa M, Arai Y, Ikenake H, Murasugi H, Sugahara N, Sumida S, Yamamoto T, Toyoda C, Kobayashi M, Kondo E, Saito K, Osawa
Okada N, Shishikura K, Suzuki H, Hirayama Y, Hirasawa K, M. Pial-glial barrier abnormalities in fetuses with Fukuyama
Fukuyama Y,Tsutsumi A,Ito K,Uchida Y.Fukuyama type con- congenital muscular dystrophy. Brain Dev 1997; 19: 35–42
genital progressive muscular dystrophy. Acta Paediatr Jpn Yamamoto T, Armstrong D, Shibata N, Kanazawa M, Kobayashi
1991; 33: 261–269 M. Immature astrocytes in Fukuyama congenital muscular
Saito K, Osawa M, Wang Z-P, Ikeya K, Fukuyama Y, Kondo-Iida E, dystrophy: an immunohistochemical study. Pediatr Neurol
Toda T, Ohashi H, Kurosawa K, Wakai S, Kanedo K-i. Haplo- 1999; 20: 31–37
type-phenotype correlation in Fukuyama congenital mus- Yoshioka M, Kuroki S. Clinical spectrum and genetic studies of
cular dystrophy. Am J Med Genet 2000; 92: 184–190 Fukuyama congenital muscular dystrophy. Am J Med
Saito Y, Murayama S, Kawai M, Nakano I. Breached cerebral glia Genet 1994; 53: 245–250
limitans-basal lamina complex in Fukuyama-type congeni- Yoshioka M, Kuroki S, Kondo T. Ocular manifestations in
tal muscular dystrophy. Acta Neuropathol (Berl) 1999; 98: Fukuyama type congenital muscular dystrophy. Brain Dev
330–336 1990; 12: 423–426
Silan F,Yoshioka M,Kobayashi K,Simsek E,Tunc M,Alper M,Cam Yoshioka M, Saiwai S, Kuroki S, Nigami H. MR imaging of the
M, Guven A, Fukuda Y, Kinoshita M, Kocabay K,Toda T. A new brain in Fukuyama-type congenital muscular dystrophy.
mutation of the fukutin gene in a non-Japanese patient. AJNR Am J Neuroradiol 1991; 12: 63–65
Ann Neurol 2003; 53: 392–396 Yoshioka M, Toda T, Kuroki S, Hamado K. Broader clinical spec-
Takada K. Fukuyama congenital muscular dystrophy as a trum of Fukuyama congenital muscular dystrophy mani-
unique disorder of neuronal migration: a neuropathologi- fested by haplotype analysis. J Child Neurol 1999; 14:
cal review and hypothesis.Yonago Acta Med 1988; 31: 1–16 711–715
Takada K, Nakamura H. Cerebellar micropolygyria in Fukuyama Zanoteli E, Rocha JCC, Narumia LK, Fireman MAT, Moura LS,
congenital muscular dystrophy: observations in fetal and Oliveira ASB, Gabbai AA, Fukuda Y, Kinoshita M, Toda T.
pediatric cases. Brain Dev 1990; 12: 774–778 Fukuyama-type congenital muscular dystrophy: a case re-
Takada K, Nakamura H, Tanaka J. Cortical dysplasia in congeni- port in the Japanese population living in Brazil. Acta Neurol
tal muscular dystrophy with central nervous system in- Scand 2002; 106: 117–121
volvement (Fukuyama type). J Neuropathol Exp Neurol
1984; 43: 395–407 Muscle-Eye-Brain Disease
Takada K, Nakamura H, Suzumori K, Ishikawa T, Sugiyama N.
Cortical dysplasia in a 23-week fetus with Fukuyama con-
Auranen M, Rapola J, Pihki H, Haltia M, Leivo I, Soinila S, Virta-
genital muscular dystrophy (FCMD). Acta Neuropathol
nen I, Kalimo H, Anderson LVB, Santavuori P, Somer H. Mus-
(Berl) 1987; 74: 300–306
cle membrane–skeleton protein changes and histopatho-
Takada K, Nakamura H, Takashima S. Cortical dysplasia in
logical characterization of muscle-eye-brain disease. Neu-
Fukuyama congenital muscular dystrophy (FCMD): a Golgi
romusc Disord 2000; 10: 16–23
and angioarchitectonic analysis. Acta Neuropathol (Berl)
Chiba A, Matsumura K, Yamada H, Inazu T, Shimizu T, Kusunoki
1988; 76: 170–178
S, Kanazawa I, Kobata A, Endo T. Structures of sialylated O-
Takeda S, Kondo M, Sasaki J, Kurahashi H, Kano H, Arai K, Misaki
linked oligosaccharides of bovine peripheral nerve a-dys-
K, Fukui T, Kobayashi K, Tachikawa M, Imamura M, Nakamu-
troglycan. The role of a novel O-manniosyl-type oligosac-
ra Y, Shimizu T, Murakami T, Sunada Y, Fujikado T, Matsumu-
charide in the binding of a-dystroglycan with laminin. J Bi-
ra K, Terashima T, Toda T. Fukutin is required for mainte-
ol Chem 1997; 272: 2216–262
nance of muscle integrity, cortical histiogenesis and nor-
Cormand B, Avela K, Pihko H, Santavouri P, Talin B, Topalogly H,
mal eye development.Hum Mol Genet 2003;12:1449–1459
de la Chapelle A, Lehesjoki A-E. Assignment of the muscle-
Terasawa K. Muscle regeneration and satellite cells in Fukuya-
eye-brain disease gene to 1p32-p34 by linkage analysis
ma-type congenital muscular dystrophy. Muscle Nerve
and homozygosity mapping. Am J Hum Genet 1999; 64:
1986; 9: 465–470
126–135
References and Further Reading 999

Goebel HH, Fidzianska A, Lenard HG, Osse G, Hori A. A morpho- Zervos A, Hunt KE,Tong G-Q, Avallone J, Morales J, Friedman N,
logical study of non-Japanese congenital muscular dystro- Cohen BH, Clark B, Guo S, Gazda H, Beggs AH, Traboulsi EI.
phy associated with cerebral lesions. Brain Dev 1983; 5: Clinical, genetic and histopathological findings in two sib-
292–301 lings with muscle-eye-brain disease. Eur J Ophthalmol
Haltia M, Leivo I, Somer H, Pikho H, Paetau A Kivelä T,Tarkkanen 2002; 12: 253–261
A,Tomé F, Engvall E, Santavuori P. Muscle-eye-brain disease:
a neuropathological study. Ann Neurol 1997; 41: 173–180
Kano H, Kobayashi K, Herrmann R, Tachikawa M, Manya H, Merosin-deficient Congenital
Nishino I, Nonaka I, Straub V, Talim B, Voit T, Topaloglu H, Muscular Dystrophy
Endo T,Yoshikawa H,Toda T.Deficiency of a-dystroglycan in
muscle-eye-brain disease. Biochem Biophys Res Commun Brett FM, Costigan D, Farrell MA, Heapy P, Thornton J, King MD.
2002; 291: 1283–1286 Merosin-deficient congenital muscular dystrophy and cor-
Korinthenberg R, Palm D, Schlake W, Klein J. Congenital muscu- tical dysplasia. Eur J Pediatr Neurol 1998; 2: 77–82
lar dystrophy, brain malformation and ocular problems Caro PA, Scavina M, Hoffman E, Pegoraro E, Marks HG. MR imag-
(muscle, eye and brain disease) in two German families. Eur ing findings in children with merosin-deficient congenital
J Pediatr 1984; 142: 64–68 muscular dystrophy. AJNR Am J Neuroradiol 1999; 20: 324–
Manya H, Sakai K, Kobayashi K,Taniguchi K, Kawakita M,Toda T, 326
Endo T. Loss-of-function of an N-acetylglucosaminyltrans- Cohn RD, Herrmann R, Sorokin L,Wewer UM,Voit T. Laminin a2
ferase, POMGnT1, in muscle–eye–brain disease. Biochem chain-deficient congenital muscular dystrophy. Neurology
Biophys Res Commun 2003; 306: 93–97 1998; 51: 94–101
Michele DE, Barresi R, Kanagawa M, Saito F, Cohn RD, Satz JS, Coral-Vazquez RM, Rosas-Vargas H, Meza-Espinosa P, Mendoza
Dollar J, Nishino I, Kelley RI, Somer H, Straub V, Mathews KD, I, Huicochea JC, Ramon G, Salamanca F. Severe congenital
Moore SA, Campbell KP. Post-translational disruption of dy- muscular dystrophy in a Mexican family with a new non-
stroglycan-ligand interactions in congenital muscular dys- sense mutation (R2578X) in the laminin a-2 gene. J Hum
trophies. Nature 2002; 418: 417–422 Genet 2003; 48: 91–95
Moore SA, Saito F, Chen J, Michele DE, Henry MD, Messing A, Di Blasi C, Mora M, Pareyson D, Farina L, Sghirlanzoni A, Vignier
Cohn RD, Ross-Barta SE, Westra S, Williamson RA, Hoshi T, N, Blasevich F, Cornelio F, Guicheney P, Morandi L. Partial
Campbell KP. Deletion of brain dystroglycan recapitulates laminin a2 chain deficiency in a patient with myopathy re-
aspects of congenital muscular dystrophy. Nature 2002; sembling inclusion body myositis. Ann Neurol 2000; 47:
418: 422–425 811–816
Muntoni F, Brockington M, Blake DJ, Torelli S, Brown SC. Defec- Di Muzio A, de Angelis MV, di Fulvio P, Ratti A, Pizzuti A, Stuppia
tive glycosylation in muscular dystrophy. Lancet 2002; 360: L, Gambi D, Unchini A. Dysmyelinating sensory-motor neu-
1419–1421 ropathy in merosin-deficient congenital muscular dystro-
Pihko H, Lappi M, Raitta C, Sainio K, Valanne L, Somer H, San- phy. Muscle Nerve 2003; 27: 500–506
tavuori P. Ocular findings in muscle-eye-brain (MEB) dis- Echenne B, Pages M, Marty-Double C. Congenital muscular dy-
ease: a follow-up study. Brain Dev 1995; 17: 57–61 strophy with cerebral white matter spongiosis. Brain Dev
Ross ME.Full circle to cobbled brain.Nature 2002; 418:376–377 1984; 6: 491–495
Santavuori P, Somer H, Sainio K, Rapola J, Kruus S, Nikitin T, Ke- Farina L, Morandi L, Milanesi I, Ciceri E, Mora M, Moroni I, Panta-
tonen L, Leisti J. Muscle-eye-brain disease (MEB). Brain Dev leoni C, Savoiardo M. Congenital muscular dystrophy with
1989; 11: 147–153 merosin deficiency: MRI findings in five patients. Neurora-
Santavuori P,Valanne L, Autti T, Haltia M, Pihko H, Sianio K. Mus- diology 1998; 40: 807–811
cle-eye-brain disease. Clinical features, visual evoked po- Gilhuis HJ, ten Donkelaar HJ,Tanke RB,Vingerhoets DM, Zwarts
tentials and brain imaging in 20 patients. Eur J Pediatr Neu- MJ, Verrips A, Gabreëls FJM. Nonmuscular involvement in
rol 1998; 1: 41–47 merosin-negative congenital muscular dystrophy. Pediatr
Taniguchi K, Kobayashi K, Saito K, Yamanouchi H, Ohnuma A, Neurol 2002; 26: 30–36
Hayashi YK, Manya H, Jin DK, Lee M, Parana E, Falsaperia R, Guicheney P, Vignier N, Helbling-Leclerc A, Nissinen M, Zhang
Pavone P, van Coter R, Talim B, Steinbrecher A, Straub V, X, Cruaud C, Lambert J-C, Richelme C, topaloglu H, Merlini L,
Nishino I, Topaloglu H, Voit T, Endo T, Toda T. Worldwide dis- Barois A, Schwartz K, Tome FMS, Tryggvason K, Fardeau M.
tribution and broader clinical spectrum of muscle-eye- Genetics of laminin a2 chain (or merosin) deficient con-
brain disease. Hum Mol Genet 2003; 12: 527–534 genital muscular dystrophy: from identification of muta-
Valanne L,Pihko H,Katevuo K,Karttunen P,Somer H,Santavuori tions to prenatal diagnosis. Neuromusc Disord 1997; 7:
P. MRI of the brain in muscle-eye-brain (MEB) disease. Neu- 180–186
roradiology 1994; 36: 473–476 Hillaire D, Leclerc A, Fauré S, Topaloglu H, Chiannilkulchaï N,
van der Knaap MS,Smit LME,Barth PG,Catsman-Berrevoets CE, Guicheney P, Grinas L, Legos P, Philpot J, Evangelista T, Rou-
Brouwer OF, Begeer JH, de Coo IFM, Valk J. Magnetic reso- ton M-C,Mayer M,Pellissier J-F,Estournet B,Barois A,Hentati
nance imaging in classification of congenital muscular dy- F, Feingold N, Beckmann JS, Dubowitz V,Tomé FMS, Fardeau
strophies with brain abnormalities. Ann Neurol 1997; 42: M. Localization of merosin-negative congenital muscular
50–59 dystrophy to chromosome 6q2 by homozygosity mapping.
Yoshida A, Kobayashi K, Manya H, Tainguchi K, Kano H, Mizuno Hum Mol Genet 1994; 3: 1657–1661
M, Inazu T, Matsuhashi H, Takahashi S, Takeuchi M, Her- Jones KJ, Morgan G, Johnston H,Tobias V, Ouvrier RA,Wilkinson
rmann R, Straub V, Talim B, Voit T, Topaloglu H, Toda T, Endo I, North KN. The expanding phenotype of laminin a2 chain
T. Muscular dystrophy and neuronal migration disorder (merosin) abnormalities: case series and review. J Med
caused by mutations in a glycosyltransferase, POMGnT1. Genet 2001; 38: 649–657
Develop Cell 2001; 1: 717–724
1000 References and Further Reading

Lamer S, Carlier R-Y, Pinard J-M, Mompoint D, Bagard C, Burda- Pegoraro E, Marks H, Garcia CA, Crawford T, Mancias P, Connol-
iron E, Estournet B, Barois A, Vallée C. Congenital muscular ly AM, Fanin M, Martinello F,Trevisan CP, Angelini C, Stella A,
dystrophy: use of brain MR imaging findings to predict Scavina M, Munk RL, Servidei S, Bönnemann CC, Bertorini T,
merosin deficiency. Radiology 1998; 206: 811–816 Acsadi G, Thompson CE, Gagnon D, Hoganson G, Carver V,
Mackay MT, Kornberg AJ, Shield L, Phelan E, Kean MJ, Coleman Zimmerman RA, Hoffman EP. Laminin a2 muscular dystro-
LT. Dennett X. Congenital muscular dystrophy, white matter phy genotype/phenotype studies of 22 patients. Neurolo-
abnormalities, and neuronal migration disorders: the ex- gy 1998; 51: 101–110
panding concept. J Child Neurol 1998; 13: 481–487 Pegoraro E, Fanin M,Trevisan CP, Angelini C, Hoffman EP. A nov-
Marbini A, Bellanova MF, Ferrari A, Lodesani M, Gemignani F. el laminin a2 isoform in severe laminin a2 deficient con-
Immunohistochemical study of merosin-negative congen- genital muscular dystrophy. Neurology 2000; 55: 1128–
ital muscular dystrophy: laminin a2 deficiency in skin biop- 1134
sy. Acta Neuropathol (Berl) 1997; 94: 103–108 Philpot J,Sewry C,Pennock J,Dubowitz V.Clinical phenotype in
Martinello F, Angelini C, Trevisan CP. Congenital muscular congenital muscular dystrophy: correlation with expres-
dystrophy with partial merosin deficiency and late onset sion of merosin in skeletal muscle.Neuromusc Disord 1995;
epilepsy. Eur Neurol 1998; 40: 37–45 5: 301–305
Mercuri E, Pennock J, Goodwin F, Sewry C, Cowan F, Dubowitz Philpot J, Cowan F, Pennock J, Sewry C, Dubowitz V, Bydder G,
L, Dubowithz V, Muntoni F. Sequential study of central and Muntoni F. Merosin-deficient congenital muscular dystro-
peripheral nervous system involvement in an infant with phy: the spectrum of brain involvement on magnetic reso-
merosin-deficient congenital muscular dystrophy. Neuro- nance imaging. Neuromusc Disord 1999; 9: 81–85
musc Disord 1996; 6: 425–429 Pini A,Merlini L,Tomé FMS,Chevallay M,Gobbi G.Merosin-neg-
Mercuri E, Anker S, Philpot J, Sewry C, Dubowitz V, Muntoni F. ative congenital muscular dystrophy, occipital epilepsy
Visual function in children with merosin-deficient and with periodic spasms and focal cortical dysplasia. Report of
merosin-positive congenital muscular dystrophy. Pediatr three Italian cases in two families. Brain Dev 1996; 18:
Neurol 1998; 18: 339–401 316–322
Mercuri E, Grunter-Andrew J, Philpot J, Sewry C, Counsell S, Reed U,Marie SK,Vainzof M,Salum PB,Levy JA,Zatz M,Diament
Henderson S, Jensen A, Naom I, Bydder G, Dubowitz V, A. Congenital muscular dystrophy with cerebral white
Muntoni F. Cognitive abilities in children with congenital matter hypodensity. Correlation of clinical features and
muscular dystrophy: correlation with brain MRI and merosin deficiency. Brain Dev 1996; 18: 53–58
merosin status. Neuromusc Disord 1999; 9: 383–387 Sanuda Y, Edgar TS, Lotz BP, Rust RS, Campbell KP. Merosin-neg-
Minetti C Bado M, Morreale G, Pedemonte M, Cordone G. Dis- ative congenital muscular dystrophy associated with ex-
ruption of muscle basal lamina in congenital muscular tensive brain abnormalities. Neurology 1995; 45: 2084–
dystrophy with merosin deficiency. Neurology 1996; 46: 2089
1354–1358 Schorer Z, Philpot J, Muntoni F, Sewry C, Dubowitz V. Demyeli-
Miyagoe-Suzuki Y, Nakagawa M, Takeda S. Merosin and con- nating peripheral neuropathy in merosin-deficient con-
genital muscular dystrophy. Microsc Res Tech 2000; 48: genital muscular dystrophy. J Child Neurol 1995; 10: 472–
181–191 475
Mora M, Moroni I, Uziel G, di Balsi C, Barresi R, Farina L, Morandi Sewrey CA, Philpot J, Mahonoy D, Wilson LA, Muntoni F,
L. Mild clinical phenotype in a 12-year-old boy with partial Dubowitz V. Expression of laminin subunits in congenital
merosin deficiency and central and peripheral nervous sys- muscular dystrophy. Neuromusc Disord 1995; 5: 307–316
tem abnormalities. Neuromusc Disord 1996; 6: 377–381 Sewrey CA, Philpot J, Sorokin LM, Wilson LA, Naom I, Goodwin
Mrak RE. The pathologic spectrum of merosin deficiency. F,D’Allesandro M,Muntoni F.Diagnosis of merosin (laminin-
J Child Neurol 1998; 13: 513–515 2) deficient congenital muscular dystrophy by skin biopsy.
Nissinen M, Helbling-Leclerc A, Zhang X, Evangelista T, Lancet 1996; 347: 582–584
Topaloglu H, Cruaud C, Weissenbach J, Fardeau M, Tomé Sewrey CA, Naom I, D’Allesandro M, Sorokin L, Bruno S, Wildon
FMS,Schwartz K,Tryggvason K,Guicheney P.Substitution of LA, Dubowitz V, Muntoni F. Variable clinical phenotype in
a conserved cysteine-996 in a cysteine-rich motif of the merosin-deficient congenital muscular dystrophy associat-
laminin a2-chain in congenital muscular dystrophy with ed with differential immunolabelling of two fragments of
partial deficiency of the protein. Am J Hum Genet 1996; 58: the laminin a2 chain. Neuromusc Disord 1997; 7: 169–175
1177–1184 Sewrey CA, D’Alessandro M, Wilson LA, Sorokin LM, Naom I,
North KN, Specht LA, Sethi RK, Shapiro F, Beggs AH. Congenital Bruno S, Ferlini A, Dubowitz V, Muntoni F. Expression of
muscular dystrophy associated with merosin deficiency. laminin chains in skin in merosin-deficient congenital mus-
J Child Neurol 1996; 11: 291–295 cular dystrophy. Neuropediatrics 1997; 28: 217–222
Osari S-i, Kobayashi O,Yamashita Y, Matsuishi T, Goto M,Tanabe Sewrey CA, Naom I, D’Allesandro M, Sorokin L, Bruno S, Wilson
Y, Migita T, Nonaka I. Basement membrane abnormality in LA, Dubowitz V, Muntoni F. Variable clinical phenotype in
merosin-negative congenital muscular dystrophy. Acta merosin-deficient congenital muscular dystrophy associat-
Neuropathol (Berl) 1996; 91: 332–336 ed with differential immunolabelling of two fragments of
Pegoraro E, Mancias P, Swerdlow SH, Raikow RB, Garcia C, Marks the laminin a2 chain. Neuromusc Disord 1997; 7: 169–175
H, Crawford T, Carver V, di Cianno B, Hoffman EP. Congenital Tachi N,Kamimura S,Ohya K,Chiba S,Sasaki K.Congenital mus-
muscular dystrophy with primary laminin a2 (merosin) de- cular dystrophy with partial deficiency of merosin. J Neu-
ficiency presenting as inflammatory myopathy. Ann Neurol rolog Sci 1997; 151: 25–27
1996; 40: 782–791 Tan E,Topaloglu H, Sewry C, Zorlu Y, Naom I, Erdem S, D’Allesan-
dro M, Muntoni F, Dubowitz V. Late onset muscular dystro-
phy with cerebral white matter changes due to partial
merosin deficiency. Neuromusc Disord 1997; 7: 85–89
References and Further Reading 1001

Taratuto AL, Lubieniecki F, Díaz D, Schultz M, Ruggieri V, Saccol- Brown SC, Torelli S, Brockington M, Yuva Y, Jimenez C, Feng L,
iti M, Dubrovski A. Merosin-deficient congenital muscular Anderson L, Ugo I, Kroger S, Bushby K, Voit T, Sewry C,
dystrophy associated with abnormal cerebral cortical gyra- Muntoni F. Abnormalities in a-dystroglycan expression in
tion: an autopsy study. Neuromusc Disord 1999; 9: 86–94 MDC1C and LGMD2I muscular dystrophies. Am J Pathol
Tezak Z, Prandini P, Boscaro M, Marin A, Devaney J, Marino M, 2004; 164: 727–737
Fanin M, Trevisan CP, Park J, Tyson W, Finkel R, Garcai C, Al- De Paula F, Vieira N, Starling A, Uraco Yamamoto L, Lima B, De
gelini C, Hoffman EP, Pagoraro E. Clinical and molecular Cássia Pavanello R, Vainzof M, Nigro V, Zatz M. Asympto-
study in congenital muscular dystrophy with partial matic carriers for homozygous novel mutations in the FKRP
laminin a2 (LAMA2) deficiency. Hum Mutat 2003; 21: 103– gene: the other end of the spectrum. Eur J Hum Genet
111 2003; 11: 923–930
Tomé FMS, Evangelista T, Leclerc A, Sunada Y, Manole E, Estour- Driss A, Noguchi S, Amouri R, Kefi M, Sasaki T, Surgie K, Souilem
net B, Barois A, Campbell KP, Fardeau M. Congenital muscu- S, Hayashi YK, Shimizu N, Minoshima S, Kudoh J, Hentati F,
lar dystrophy with merosin deficiency. Life Sci 1994; 317: Nishino I. Fukutin-related protein gene mutated in the
351–357 original kindred limb-girdle MD 2I. Neurology 2003; 60:
Trevisan CP, Martinello F, Ferruzza E, Angelini C. Divergence of 1341–1344
central nervous system involvement in 2 Italian sisters with Harel T,Goldberg Y,Shalev SA, Chervinski I, Ofir R, Birk OS.Limb-
congenital muscular dystrophy: a clinical and neuroradio- girdle muscular dystrophy 2I: phenotypic variability within
logical follow-up. Eur Neurol 1995; 35: 230–235 a large consanguineous Bedouin family associated with a
Trevisan CP,Martinello F,Ferruzza E,Fanin M,Chevallay M,Tomé novel FKRP mutation. Eur J Hum Genet 2004; 12: 38–43
FMS. Brain alterations in the classical form of congenital Louhichi N,Triki C, Quijano-Roy S, Richard P, Makri S, Méziou M,
muscular dystrophy. Child Nerv Syst 1996; 12: 604–610 Estournet B, Mrad S, Romero NB, Ayadi H, Guicheney P,
Tsao CY, Mendell JR, Rusin J, Luquette M. Congenital muscular Fakhfakh F. New FRKP mutations causing congenital mus-
dystrophy with complete laminin-a2-deficiency, cortical cular dystrophy associated with mental retardation and
dysplasia, and cerebral white-matter changes in children. central nervous system abnormalities. Identification of a
J Child Neurol 1998; 13: 253–256 founder mutation in Tunesian families. Neurogenetics
Van Engelen BGM, Leyten QH, Bernsen PLJA, Gabreëls FJM, 2004; 5: 27–34
Barkhof F, Joosten EMG, Hamel BCJ, ter Laak HJ, Ruijs MBM, Mercuri E, Sewry CA, Brown SC, Brockington M, Jungbluth H,
Cruysberg JRM,Valk J.Familial adult-onset muscular dystro- DeVille C, Counsell S, Muntioni F. Congenital muscular dys-
phy with leukoencephalopathy. Ann Neurol 1992; 32: trophy with secondary merosin deficiency and normal
577–580 brain MRI: a novel entity? Neuropediatrics 2000; 31:
Villanova M, Malandrini A, Toti P, Salvestroni R, Six J, Martin JJ, 186–189
Guazzi GC. Localization of merosin in the normal human Mercuri E, Brockington M, Straub V, Quijano-Roy S, Yuva Y, Her-
brain: implications for congenital muscular dystrophy with mann R, Brown SC, Torelli S, Dubowitz V, Blake DJ, Romero
merosin deficiency. J Submicrosc Cytol Pathol 1996; 28: 1–4 NB, Estournet B, Sewry CA, Guicheney P, Voit T, Muntoni F.
Villanova M, Malandrini A, Sabatelli P, Sewry CA, Toti P, Torelli S, Phenotype spectrum associated with mutations in the
Six J, Scarfó G, Palma L, Muntoni F, Squarzoni S,Tosi P, Maral- Fukutin-related protein gene. Ann Neurol 2003; 53: 537–
di NM, Guazi GC. Localization of laminin a2 chain in normal 542
human central nervous system: an immunofluorescence Topaloglu H, Brockington M, Yuva Y, Talim B, Haliloglu G, Blake
and ultrastructural study. Acta Neuropathol (Berl) 1997; 94: D, Torelli S, Brown SC, Muntoni F. FKRP gene mutations
567–571 cause congenital muscular dystrophy, mental retardation,
Wewer UM, Engvall E. Merosin/laminin-2 and muscular dystro- and cerebellar cysts. Neurology 2003; 60: 988–992
phy. Neuromusc Disord 1996; 6: 409–418
Yurchenco PD, Cheng Y-S, Campbell K, Li S. Loss of basement
membrane, receptor and cytoskeletal lattices in a laminin- MDC1D
deficient muscular dystrophy. J Cell Sci 2004; 117: 735–742
Endo T, Toda T. Glycosylation in congenital muscular dystro-
phies. Biol Pharm Bull 2003; 26: 1641–1647
Grewal PK, Hewitt JE. Glycosylation defects: a new mechanism
MDC1C
for muscular dystrophy? Hum Mol Genet 2003; 12: R259-
R264
Brockington M, Blake DJ, Prandini P, Brown SC,Torelli S, Benson Hewitt JE, Grewal PK, Glycosylation defects in inherited muscle
MA, Ponting CP, Estournet B, Romero NB, Mercuri E, Voit T, disease. Cell Mol Life Sci 2003; 60: 251–258
Sewry CA, Guicheney P, Muntoni F. Mutations in the Longman C, Brockington M, Torelli S, Jimenez-Mallebrera C,
Fukutin-related protein gene (FKRP) cause a form of con- Kennedy C, Khalil N, Feng L, Saran RK,Voit T, Merlini L, Sewry
genital muscular dystrophy with secondary laminin a2 de- CA, Brown SC, Muntoni F. Mutations in the human LARGE
ficiency and abnormal glycosylation of a-dystroglycan. Am gene caus MDC1D, a novel form of congenital muscular dy-
J Hum Genet 2001; 69: 1198–1209 strophy with severe mental retardation and abnormal gly-
Brockington M, Yuva Y, Prandini P, Brown SC, Torelli S, Benson cosylation of a-dystroglycan. Hum Mol Genet 2003; 12:
MA, Herrmann R, Anderson LVB, Bashir R, Burgunder J-M, 2853–2861
Fallet S, Romero N, Fardeau M, Straub V, Storey G, Pollitt C, Martin PT, Freeze HH. Glycobiology of neuromuscular disor-
Richard I, Sewry CA, Bushby K, Voit T, Blake DJ, Muntoni F. ders. Glycobiology 2003; 13: 67R-75R
Mutations in the Fukutin-related protein gene (FKRP) iden- Martin-Rendon E, Blake DJ. Protein glycosylation in disease:
tify limb girdle muscular dystrophy 2 l as a milder allelic new insights into the congenital muscular dystrophies.
variant of congenital muscular dystrophy MDC1C. Hum Trends Pharmacol Sci 2003; 24: 178–183
Mol Genet 2001; 10: 2851–2859
1002 References and Further Reading

Muntoni F, Valero de Bernabe B, Bittner R, Blake D, van Brunner HG, Jansen G, Nillesen W, Nelen MR, de Die CEM,
Bokhoven H, Brockington M, Brown S, Bushby K, Campbell Höweler CJ, van Oost BA, Wieringa B, Ropers HH, Smeets
KP, Fiszman M, Gruenewald S, Merlini L, Quijano-Roy S, HJM. Brief report: Reverse mutation in myotonic dystrophy.
Romero N, Sabatelli P, Sewry CA, Straub V,Talim B,Topaloğlu N Engl J Med 1993; 328: 476–480
H, Voit T, Yurchenco PD, Urtizberea JA, Wewer UM, Guich- Censori B, Provinciali L, Danni M, Chiaramoni I, Maricotti M,
eney P. 114th ENMC International Workshop on Congenital Foschi N, Del Pesce M, Salvolini U. Brain involvement in
Muscular Dystrophy (CMD) (8th Workshop of the Interna- myotonic dystrophy: MRI features and their relationship to
tional Consortium on CMD; 3rd Workshop of the Myo-clus- clinical and cognitive conditions. Acta Neurol Scand 1994;
ter Project GENRE). Neuromusc Disord 2003; 13: 579–588 90: 211–217
Muntoni F, Brockington M,Torelli S, Brown SC. Defective glyco- Damian MS, Bachmann G, Herrmann D, Dorndorf W. Magnetic
sylation in congenital muscular dystrophies. Curr Opin resonance imaging of muscle and brain in myotonic dys-
Neurol 2004; 17: 205–209 trophy. J Neurol 1993; 240: 8–12
Damian MS, Schilling G, Bachman G, Simon C, Stöppler S, Dorn-
dorf W. White matter lesions and cognitive deficits: rele-
Rest vance of lesion pattern? Acta Neurol Scand 1994; 90:
430–436
Arahata K, Brockington M, Bushby K, Cormand B, Dubowitz V, Di Costanzo A, Di Salle F, Santoro L, Bonavita V, Tedeschi G. T2
Engvall E, Flanigan K, Guicheney P, Moghadaszadeh B, relaxometry of brain in myotonic dystrophy. J Neuroradiol
Morandi L, Nuntoni F, Naom I, Pihko H, Sewry C, Soininen R, 2001; 43: 198–204
Straub V, Toda T, Tomé F, Topaloğlu H, Urizberea A, Villanova Di Costanzo A, Di Salle F, Santoro L, Tessitore A, Bonavita V,
M, Vilquin J-T, Voit T. 68th ENMC International Workshop Tedeschi G. Pattern and significance of white matter abnor-
(5th International Workshop): on congenital muscular dys- malities in myotonic dystrophy type 1: an MRI study. J Neu-
trophy. Neuromusc Disord 1999; 9: 446–454 rol 2002; 249: 1175–1182
De Stefano N, Dotti MT, Villanova M, Scarano G, Federico A. Di Costanzo A,Di Salle F,Santoro L,Bonavita V,Tedeschi G.Brain
Merosin positive congenital muscular dystrophy with se- MRI features of congenital and adult-form myotonic dys-
vere involvement of the central nervous system. Brain Dev trophy type 1: case-control study. Neuromusc Disord 2002;
1996; 18: 323–326 12: 476–483
Echenne B, Rivier F, Jallali AJ, Azais M, Mornet D, Pons F. Merosin Fierro B, Daniele O, Aloisio A, Buffa D, La Bua V, Oliveri M, Man-
positive congenital muscular dystrophy with mental defi- frè L, Brighina F. Neurophysiological and radiological find-
ciency, epilepsy and MRI changes in the cerebral white ings in myotonic dystrophy patients. Eur J Neurol 1998; 5:
matter. Neuromusc Disord 1997; 7: 187–190 89–94
Kobayashi O, Hayashi Y, Arahata K, Ozawa E, Nonaka I. Congen- Garcia-Alix A, Cabanas F, Morales C, Pellicer A, Echevarria J,
ital muscular dystrophy: clinical and pathologic study of 50 Paisan L, Quero J. Cerebral abnormalities in congenital
patients with the classical (occidental) merosin-positive myotonic dystrophy. Pediatr Neurol 1991; 7: 28–32
form. Neurology 1996; 46: 815–818 Glantz RH, Wright RB, Huckman MS, Garron DC, Siegel IM. Cen-
Topaloğlu H, Kale G, Yalnizoğlu D, Taödemir AH, Karaduman A, tral nervous system magnetic resonance imaging findings
Topçu M, Kotioğlu E. Analysis of “pure” congenital muscular in myotonic dystrophy. Arch Neurol 1998; 45: 36–37
dystrophies in thirty-eight cases. How different is the clas- Groenen P, Wieringa B. Expanding complexity in myotonic
sical type I from the occidental type cerebromuscular dys- dystrophy. BioEssays 1998; 20: 901–912
trophy? Neuropediatrics 1994; 25: 94–100 Hashimoto T, Tayama M, Miyazaki M, Murakawa K, Kawai H,
Triki C, Louhichi N, Méziou M, Choyakh F, Kéchaou MS, Jlidi R, Nishitani H, Kuroda Y. Neuroimaging study of myotonic
Mhiri C, Fakhfakh F, Ayadi H. Merosin-deficient congenital dystrophy. I. Magnetic resonance imaging of the brain.
muscular dystrophy with mental retardation and cerebellar Brain Dev 1995; 17: 24–7
cysts, unlinked to the LAMA2, FCMD, MEB and CMD1B loci, Hashimoto T, Tayama M, Miyazaki M, Murakawa K, Kawai H,
in three Tunesian patients. Neuromusc Disord 2003; 13: Nishitani H, Kuroda Y. Neuroimaging study of myotonic
4–12 dystrophy. II. MRI measurements of the brain. Brain Dev
Voit T, Cohn RD, Sperner J, Leube B, Sorokin L,Toda T, Herrmann 1995; 17: 28–32
R. Merosin-positive congenital muscular dystrophy with Huber SJ, Kissel JT, Shuttleworth EC, Chakeres DW, Clapp LE,
transient brain dysmyelination, pontocerebellar hypopla- Brogan MA. Magnetic resonance imaging and clinical cor-
sia and mental retardation. Neuromusc Disord 1999; 9: relates of intellectual impairment in myotonic dystrophy.
95–101 Arch Neurol 1989; 46: 536–540
Martinello F, Piazza A, Pastorello E, Angelini C,Trevisan CP. Clin-
ical and neuroimaging study of central nervous system in
congenital myotonic dystrophy. J Neurol 1999; 246: 186–
61 Myotonic Dystrophy Type 1 192
Miaux Y, Chiras J, Eymard B, Lauriot-Prevost MC, Radvanyi H,
Abe K, Fujimura H, Toyooka K, Yorfuji S, Nishikawa Y, Hazama T, Martin-Duverneuil N, Delaporte C. Cranial MRI findings in
Yanagihira T. Involvement of the central nervous system in myotonic dystrophy. J Neuroradiol 1997; 39: 166–170
myotonic dystrophy. J Neurol Sci 1991; 127: 179–185 Mitzukami K, Sasaki M, Baba A, Suzuki T, Shiraishi H. An autop-
Abe K, Fujimura H, Soga F. The fluid-attenuated inversion-re- sy case of myotonic dystrophy with mental disorders and
covery pulse sequence in assessment of central nervous various neuropathologic features. J Psychiatry Clin Neu-
system involvement in myotonic dystrophy. J Neuroradiol rosci 1999; 53: 51–55
1998; 40: 32–35 Moxley R III. Myotonic disorders in childhood: diagnosis and
Bachman G, Damian MS, Koch M, Schilling G, Fach B, Stöppler S. treatment. J Child Neurol 1997; 12: 116–129
The clinical and genetic correlates of MRI findings in
myotonic dystrophy. J Neuroradiol 1996; 38: 629–635
References and Further Reading 1003

Naka H, Imon Y, Ohshita T, Honjo K, Kitamura T, Mimori Y, Naka- Meola G, Sansone V. A newly-described myotonic disorder
mura S. Magnetezation transfer measurements of cerebral (proximal myotonic myopathy – PROMM): personal experi-
white matter in patients with myotonic dystrophy. J Neurol ence and review of the literature. Ital J Neurol Sci 1996; 17:
Sci 2002; 193: 111–116 347–353
Ogata A, Terae S, Fujita M, Tashiro K. Anterior temporal white Meola G, Sansone V, Perani D, Colleluori A, Cappa S, Cotelli M,
matter lesions in myotonic dystrophy with intellectual im- Fazio F, Thornton CA, Moxley RT. Reduced cerebral blood
pairment: an MRI and neuropathological study. J Neurora- flow and impaired visual-spatial function in proximal myo-
diol 1998; 40: 411–415 tonic myopathy. Neurology 1999; 53: 1042–1050
Rosman NP,Rebeiz JJ.The cerebral defect and myopathy in my- Meola G, Sansone V, Marinou K, Cotelli M, Moxley III RT, Thorn-
otonic dystrophy. Neurology 1967; 17: 1106–1112 ton CA, De Ambroggi L. Proximal myotonic myopathy: a
Spranger M, Spranger S,Tischendorf M, Meinck HM, Cremer M. syndrome with a favourable prognosis? J Neurol Sci 2002;
Myotonic dystrophy the role of large triplet repeat length 193: 89–96
in the development of mental retardation. Arch Neurol Moxley RT III, Udd B, Ricker K. 54th ENMC International Work-
1997; 54: 251–254 shop: PROMM (proximal myotonic myopathies) and other
Tanabe Y, Iai M, Tamai K, Fujimoto N, Sugita K. Neuroradiologi- myotonic syndromes. 10–12th October 1997. Neuromusc
cal findings in children with congenital myotonic dystro- Disord 1998; 8: 508–518
phy. Acta Paediatr 1992; 81: 613–617 Moxley RT III, Meola G, Udd B, Ricker K. Report of the 84th
Timchenko L. Trinucleotide repeats myotonic dystrophy: the ENMC workshop: PROMM (proximal myotonic myopathy)
role of RNA CUG triplet repeats. Am J Hum Genet 1999; 64: and other myotonic dystrophy-like syndromes: 2nd work-
360–364 shop. 13–15th October, 2000. Neuromusc Disord 2002; 12:
Timchenko L, Caskey CT. Triplet repeat disorders: discussion 306–317
of molecular mechanisms. Cell Mol Life Sci 1999; 55: Nagamitsu S, Ashizawa T. 18 Myotonic dystrophies. Adv Neurol
1432–1447 2002; 88: 293–314
Timchenko L, Monckton DG, Caskey CT. Myotonic dystrophy: Ricker K, Grimm T, Koch MC, Schneider C, Kress W, Reimers CD,
an unstable CTG repeat in a protein kinase gene. Cell Biol Schulte-Mattler W, Mueller-Myshok B, Toyka KV, Mueller CR.
1995; 6: 13–19 Linkage of proximal myotonic myopathy to chromosome
Uluc K, Murat Arsava E, Erdem S,Tan E. Proximal myopathy and 3q. Neurology 1999; 52: 170–171
diffuse white matter involvement in myotonic dystrophy Schneider C, Ziegler A, Ricker K, Grimm T, Kress W, Reimers CD,
type I. J Neurol 2002; 249: 629–630 Meinck H-M, Reiners K, Toyka KV. Proximal myotonic myo-
pathy. Evidence for anticipation in families with linkage to
chromosome 3q. Neurology 2000; 55: 383–388
Wieser T, Bönsch D, Eger K, Schulte-Mattler W, Zierz S. A family
62 Myotonic Dystrophy Type 2 with PROMM not linked to the recently mapped PROMM
locus DM 2. Neuromusc Disord 2000; 10: 141–143
Bönsch D, Neumann C, Lang-Roth R, Witte O, Lamprecht-Din-
nesen A, Deufel T. PROMM and deafness: excluding of ZNF9
as the disease in DFNA18 suggests a polygenic origin of the
PROMM/DM 2 phenotype. Clin Genet 2003; 63: 73–75 63 X-linked Charcot-Marie-Tooth Disease
Day JW, Ricker K, Jacobsen JF, Rasmussen LJ, Dick KA, Kress W,
Schneider C, Koch MC, Beilman GJ, Harrison AR, Dalton JC, Abrams CK, Oh S, Ri Y, Bargiello TA. Mutations in connexin 32:
Ranum LPW. Myotonic dystrophy type 2. Molecular, diag- the molecular and biophysical bases for the X-linked form
nostic and clinical spectrum. Neurology 2003; 60: 657–664 of Charcot-Marie-Tooth disease. Brain Res Rev 2000;
Finsterer J, Monotonic dystrophy type 2. Eur J Neurol 2002; 9: 32:203–214
441–447 Abrams CK, Freidin MM, Verselis VK, Bennett MVL, Bargiello TA.
Hund E, Jansen O, Koch MC, Ricker K, Fogel W, Niedermaier N, Functional alterations in gap junction channels formed by
Otto M, Kuhn E, Meinck HM. Proximal myotonic myopathy mutant forms of connexin 32: evidence for loss of function
with MRI white matter abnormalities of the brain. Neurolo- as a pathogenic mechanism in the X-linked form of Char-
gy 1997; 48: 33–37 cot-Marie-Tooth disease. Brain Res 2001; 300: 9–25
Kress W,Mueller-Myhsok B,Ricker K,Schneider C,Koch MC,Toy- Abrams CK, Bennett MVL, Verselis VK, Bargiello TA. Voltage
ka KV, Mueller CR, Grimm T. Proof of genetic heterogeneity opens unopposed gap junction hemichannels formed by a
in the proximal myotonic myopathy syndrome (PROMM) connexin 32 mutant associated with X-linked Charcot-
and its relationship to myotonic dystrophy type 2 (DM). Marie-Tooth disease. Proc Natl Acad Sci USA 2002; 99:
Neuromusc Disord 2000; 10: 478–480 3980–3984
Liquiori CL, Ricker K, Moseley ML, Jacobsen JF, Kress W, Naylor Bähr M, Andres F, Timmerman V, Nelis ME, Van Broeckhoven C,
SL, Day JW, Ranum LPW. Myotonic dystrophy type 2 caused Dichgans J. Central visual, acoustic, and motor pathway in-
by a CCTG expansion in Intron 1 of ZNF9. Science 2001; 293: volvement in a Charcot-Marie-Tooth family with an
864–867 Asn205Ser mutation in the connexin 32 gene. J Neurol
Mastaglia FL, Harker N, Philips BA, Day TJ, Hankey GJ, Laing NG, Neurosurg Psychiatry 1999; 66: 202–206
Fabian V, Kakulas BA. Dominantly inherited proximal my- Bergoffen J, Scherer SS, Wang S, Oronzi Scott M, Bone LJ, Paul
otonic myopathy and leukoencephalopathy in a family DL, Chen K, Lensch MW, Chance PF, Fischbeck KH. Commex-
with an incidental CLCN1 mutation. J Neurol Neurosurg in mutations in X-linked Charcot-Marie-Tooth disaese. Sci-
Psychiatry 1998; 64: 543–547 ence 1993; 262: 2039–2042
Meola G. Clinical and genetic heterogeneity in myotonic dys-
trophies. Muscle Nerve 2000; 23: 1789–1799
Meola G. Motonic dystrophies. Curr Opin Neurol 2000; 13:
519–525
1004 References and Further Reading

Birouk N, LeGuern E, Maisonobe T, Rouger H, Gouider R,Tardieu Senderek J, Hermanns B, Bergmann C, Boroojerdi B, Bajbouj M,
S, Gugenheim M, Routon MC, Léger JM, Agid Y, Brice A, Hungs M, Ramaekers VT, Quasthoff S, Karch D, Micheal
Bouche P. X-linked Charcot-Marie-Tooth disease with con- Schröder J. X-linked dominant Charcot-Marie-Tooth neu-
nexin 32 mutations. Clinical and electrophysiologic study. ropathy: clinical electrophysiological, and morphological
Neurology 1998; 50: 1074–1082 phenotype in four families with different connexin32 mu-
Bondurand N, Girard M, Pingault V, Lemort N, Dubourg O, tations. J Neurologic Sci 1999; 167: 90–101
Goossens M. Human Connexin 32, a gap junction protein Stojkovic T, Latour P. Vanderberghe A, Hurtevent JF, Vermersch
altered in the X-linked form of Charcot-Marie-Tooth dis- P. Sensorineural deafness in X-linked Charcot-Marie-Tooth
ease, is directly regulated by transcription factor SOX10. disease with connexin 32 mutation (R142Q). Neurology
Hum Mol Genet 2001; 10: 2783–2795 1999; 52: 1010–1014
Goto H, Matsuo H, Ohnishi A, Fukudome T, Shibuya N. X-linked Sutor B, Schmolke C, Teubner B, Schirmer C, Willecke K. Myeli-
motor and sensory neuropathy with pyramidal signs and nation defects and neuronal hyperexcitability in the neo-
cerebral white matter lesions. Muscle Nerve 2003; 28: cortex of connexin 32-deficient mice. Cerebr Cortex 2000;
623–625 10: 684–697
Hanemann CO, Bergmann C, Senderek J, Zerres K, Sperfeld A-D.
Transient, recurrent, white matter lesions in X-linked Char-
cot-Marie-Tooth disease with novel connexin 32 muta-
tions. Arch Neurol 2003; 60: 605–609
64 Oculodentodigital Dysplasia
Ionasescu V, Ionasescu R, Searby C. Correlation between 32
gene mutations and clinical phenotype in X-linked domi- Ginsberg LE, Jewett T, Grub R, McLean WT. Oculodental digital
nant Charcot-Marie-Tooth neuropathy. Am J Med Genet dysplasia: neuroimaging in a kindred. Neuroradiology
1996; 63: 486–491 1996; 38: 84–86
Kawakami H, Inoue K, Sakakihara I, Nakamura S. Novel muta- Gladwin A, Donnai D, Metcalfe K, Schrander-Stumpel C, Brue-
tion in X-linked Charcot-Marie-Tooth disease associated ton L, Verloes A, Aylsworth A, Torrielo H, Winter R, Dixon M.
with CNS impairment. Neurology 2002; 59: 923–926 Localization of a gene for oculodentodigital syndrome to
Kleopa KA, Yum SW, Scherer SS. Cellular mechanisms of con- human chromosome 6q22-q24. Hum Mol Genet 1997; 6:
nexin32 associated with CNS manifestations.J Neurosci Res 123–127
2002; 68: 522–534 Gutmann DH,Zackai EH,McDonald-McGinn DM,Fischbeck KH,
Krutovskikh V,Yamasaki H.Connexin gene mutations in human Kamholz J. Oculodentodigital dysplasia syndrome associ-
genetic diseases. Mutat Res 2000; 462: 197–207 ated with abnormal cerebral white matter. Am J Med Genet
Lee M-J, Nelson I, Houlden H, Sweeney MG, Hilton-Jones D, 1991; 41: 18–20
Blake J, Wood NW, Reilly MM. Six novel connexin32 (GJB1) Loddenkemper T, Grote K, Evers S, Oelerich M, Stögbauer F.
mutations in X-linked Charcot-Marie-Tooth disease. J Neu- Neurological manifestations of the oculodentodigital dys-
rol Neurosurg Psychiatry 2002; 73: 304–306 plasia syndrome. J Neurol 2002; 249: 584–595
Marques W Jr, Sweeney MG, Wood NW, Wroe SJ, Marques W. Norton KK, Carey JC, Gutmann DH. Oculodentodigital dyspla-
Central nervous system involvement in a novel connexin sia with cerebral white matter abnormalities in a two-gen-
32 mutation affecting identical twins. J Neurol Neurosurg eration family. Am J Med Genet 1995; 57: 458–461
Psychiatry 1999; 66: 803–804 Patton MA, Laurence KM. Three new cases of oculodentodigi-
Nicholson GA, Yeung L, Corbett A. Efficient neurophysiologic tal (ODD) syndrome: development of the facial phenotype.
selection of X-linked Charcot-Marie-Tooth families. Ten J Med Genet 1985; 22: 386–389
novel mutations. Neurology 1998; 51: 1412–1416 Paznekas WA, Boyadjiev SA, Shapiro RE, Daniels O, Wollnik B,
Panas M, Karadimas C, Avramopoulos D, Vassillopoulos D. Cen- Keegan CE, Innis JW, Dinulos MB, Christian C, Hannibal MC,
tral nervous system involvement in four patients with Char- Jabs EW. Connexin 43 (GJA1) mutations cause the
cot-Marie-Tooth disease with connexin 32 extracellular pleiotropic phenotype of oculodentodigital dysplasia.Am J
mutations. J Neurol Neurosurg Psychiatry 1998; 65: 947– Hum Genet 2003; 72: 408–418
962 Pizzuti A, Flex E, Mingarelli R, Salpietro C, Zelante L, Dallapicco-
Paulson HL, Garbern JY, Hoban TF, Krajewski KM, Lewis RA, Fis- la B. A homozygous GJA1 gene mutation causes a Haller-
chbeck KH, Grossmann RI, Lenkinski R, Kamholz JA, Shy ME. man-Streiff/ODDD spectrum phenotype. Hum Mutat 2004;
Transient central nervous system white matter abnormali- 23: 286
ty in X-linked Charcot-Marie-Tooth disease. Ann Neurol Richardson RR, Donnai D, Meire F, Dixon MJ. Expression of Gja1
2002; 52: 429–434 correlates with the phenotype observed in oculodento-
Ressot C, Gomès D, Dautigny A, Pham-Dihn D, Burzzone R. Con- digital syndrome/type III syndactyly. J Med Genet 2004; 41:
nexin32 mutations associated with X-linked Charcot- 60–67
Marie-Tooth disease show two distinct behaviors: loss of Schrander-Stumpel CTRM, Franke CL. Central nervous system
function and altered gating properties. J Neurosci 1998; 18: abnormalities in oculodentodigital dysoplasia. Genet
4063–4075 Couns 1996; 7: 233–235
Schelhaas HJ, Van Engelen BMG, Gabreëls-Festen AAWM, Schrander-Stumpel CTRM, de Groot-Wijnands JBG, de Die-
Hageman G, VLiegen JHR, Van der Knaap MS, Zwarts MJ. Smulders C, Fryns JP. Type III syndactyly and odulodento-
Transient cerebral white matter lesions in a patient with digital dysplasia: a clinical spectrum. Genet Couns 1993; 4:
connexin 32 missence mutation. Neurology 2002; 59: 271–276
2007–2008 Shapiro RE, Griffin JW, Stine OC. Evidence for genetic anticipa-
tion in the oculodentodigital syndrome. Am J Med Genet
1997; 71: 36–41
References and Further Reading 1005

Spaepen A, Schrander-Stumpel C, Fryns J-P, de Die-Smulders C, Fogli A, Rodriguez D, Eymard-Pierre E, Bouhour F, Labauge P,
Borghgraef M, van den Berghe H. Hallerman-Streiff syn- Meaney BF, Zeesman S, Kaneski CR, Schiffmann R, Boes-
drome: clinical and psychological findings in children. pflug-Tanguy O. Ovarian failure related to eukaryotic initia-
Nosologic overlap with oculodentaldigital dysplasia? Am J tion factor 2B mutations. Am J Hum Genet 2003 72:
Hum Genet 1991; 41: 517–520 1544–1550
Fogli A, Schiffmann R, Hugendubler L, Combes P, Bertini E, Ro-
driguez D, Kimball SR, Boespflug-Tanguy O. Decreased gua-
nine nucleotide exchange factor activity in eIF2B-mutated
65 Vanishing White Matter patients. Eur J Hum Genet 2004; 12: 561–566
Fogli A, Schiffmann R, Bertini E, Ughetto S, Combes P, Eymard-
Abbott CM, Proud CG. Translation factors: in sickness and in Pierre E, Kaneski CR, Pineda M, Troncoso M, Uziel G, Surtees
health.Trends Biochem Sci 2004; 29: 25–31 R, Pugin D, Chaunu M-P, Rodriguez D, Boespflug-Tanguy O.
Alorainy IA, Patenaude YG, O’Gorman AM, Black DN, Meagher- The effect of genotype on the natural history of eIF2B-relat-
Villemure K. Cree leukoencephalopathy: neuroimaging ed leukodystrophies. Neurology 2004; 62: 1509–1517
findings. Radiology 1999; 213: 400–406 Francalanci P, Eymard-Pierre E, Dionisi-Vici C, Bolderini R,
Anzil AP, Gessaga E. Late-life cavitating dystrophy of the cere- Piemonte F, Virgili R, Fariello G, Bosman C, Santorelli FM,
bral and cerebellar white matter. Eur Neurol 1972; 7: 79–94 Boespflug-Tanguy O, Bertini E. Fatal infantile leukodystro-
Biancheri R, Rossi A, Di Rocco M, Filocamo M, Pronk JC, van der phy: a severe variant of CACH/VWM syndrome, allelic to
Knaap MS, Tortori-Domati P. Leukoencephalopathy with chromosome 3q27. Neurology 2001; 57:265–70
vanishing white matter: an adult onset case. Neurology Gallo A, Rocca MA, Falini A, Scaglione C, Salvi F, Gambini A,
2003; 61: 1818–1819 Guerrini L, Mascalchi M, Pronk JC, van der Knaap MS, Filippi
Black DN, Booth F, Watters GV, Andermann E, Dumont C, Halli- M. Multiparametric MRI in a patient with adult-onset
day WC, Hoogstraten J, Kabay ME, Kaplan P, Meagher-Ville- leukoencephalopathy with vanishing white matter.Neurol-
mure K, Michaud J, O’Gorman G. Leukoencephalopathy ogy 2004; 62: 323–326
among native Indian infants in northern Quebec and Man- Gautier JC, Gray F, Awada A, Escourolle R. Leucodystrophie
itoba. Ann Neurol 1988; 24: 490–496 orthochromatique cavitaire de l’adulte. Prolifération et in-
Blüml S, Philippart M, Schiffmann R, Seymour K, Ross BD. Mem- clusion oligodendrogliales. Rev Neurol 1984; 140: 493–501
brane phospholipids and high-energy metabolites in child- Girard PF, Tommassi M, Rochet M, Boucher M. Leuco-
hood ataxia with CNS hypomyelination. Neurology 2003; encéphalopathie avec cavitations massives, bilatérales et
61: 648–654 symétriques. Syndrome de décortication post-trauma-
Boesen T, Mohammed SS, Pavitt GD, Andersen GR. Structure of tique. Presse Med 1968; 76: 163–166
the catalytic fragment of translation initiation factor 2B and Gow A,Sharma R.The unfolded protein response in protein ag-
identification of a critically important catalytic residue. gregating diseases. Neuromol Med 2003; 4: 73–94
J Biol Chem 2004; 279: 10584–10592 Graveleau P, Gray F, Plas J, Graveleau J, Brion S.Leucodystrophie
Boltshauser E, Barth PG,Troost D, Martin E, Stallmach T.“Vanish- orthochromatique cavitaire avec modifications oligoden-
ing white matter” and ovarian dysgenesis in an infant with drogliales. Un cas sporadique adulte. Rev Neurol 1985; 141:
cerebro-oculo-facio-skeletal phenotype. Neuropediatrics 713–718
2002; 33: 57–62 Hanefeld F, Holzbach U, Kruse B, Wilichowski E, Christen HJ,
Brück W, Herms J, Brockmann K, Schulz-Schaeffer W, Hanefeld Frahm J. Diffuse white matter disease in three children: an
F. Myelinopathia centralis diffusa (vanishing white matter encephalopathy with unique features on magnetic reso-
disease): evidence of apoptotic oligodendrocyte degener- nance imaging and proton magnetic resonance spec-
ation in early lesion development. Ann Neurol 2001; 50: troscopy. Neuropediatrics 1993; 24: 244–248
532–536 Harding HP, Zhang Y, Ron D. Protein translation and folding are
Deisenhammer E, Jellinger K. Höhlenbildende Neurtralfett- coupled by an endoplasmic-reticulum-resident kinase.
leukodystrophie mit Schubverlauf. Neuropediatrics 1976; Nature 1999; 397:271–274
7: 111–121 Harding HP, Novoa I, Zhang Y, Zeng H,Wek R, Schapira M, Ron D.
Duncan RF, Hershey JW. Protein synthesis and protein phos- Regulated translation initiation controls stress-induced
phorylation during heat stress, recovery, and adaptation. gene expression in mammalian cells. Mol Cell 2000; 6:
J Cell Biol 1989; 109: 1467–1481 1099–1108
Eicke WJ. Polycystische Umwandlung des Marklagers mit pro- Hershey JWB, Merrick WC.The pathway and mechanism of ini-
gredientem Verlauf. Atypische diffuse Sklerose? Arch Psy- tiation of protein synthesis. In: Sonenberg N, Hershey JWB,
chiatr Nervenkr 1962; 203: 599–602 Mathews MB, eds.Translational control of gene expression.
Espay AJ, Bodensteiner JB, Patel H. Episodic coma in a new Cold Spring Harbor: Cold Spring Harbor Laboratory Press,
leukodystrophy. Pediatr Neurol 2002; 26: 139–142 2000, pp 33–88
Fogli A, Dionisi-Vici C, Deodato F, Bartuli A, Boespflug-Tanguy Hinnebusch AG. Mechanism and regulation of initiator me-
O, Bertini E. A severe variant of childhood ataxia with thionyl-tRNA binding to ribosomes. In: Sonenberg N, Her-
central hypomyelination / vanishing white matter leuko- shey JWB, Mathews MB, eds. Translational control of gene
encephalopathy related to EIF21B5 mutation. Neurology expression. Cold Spring Harbor: Cold Spring Harbor Labo-
2002; 59: 1966–1968 ratory Press, 2000, pp 185–243
Fogli A, Wong K, Eymard-Pierre E, Wenger J, Bouffard JP, Goldin Kleijn M, Scheper GC, Voorma HO, Thomas AA. Regulation of
E, Black DN, Boespflug-Tanguy O, Schiffmann R. Cree translation initiation factors by signal transduction. Eur J
leukoencephalopathy and CACH/VWM disease are allelic at Biochem 1998; 253: 531–544
the EIF2B5 locus. Ann Neurol 2002; 52: 506–510
1006 References and Further Reading

Krishnamoorthy T, Pavitt GD, Zhang F, Dever TE, Hinnebusch Scheper GC, Mulder J, Kleijn M, Voorma HO, Thomas AAM,
AG. Tight binding of the phosphorylated alpha subunit of van Wijk R. Inactivation of eIF2B and phosphorylation of
initiation factor 2 (eIF2alpha) to the regulatory subunits of PHAS-I in heat-shocked rat hepatoma cells. J Biol Chem
guanine nucleotide exchange factor eIF2B is required for 1997; 272: 26850–26856
inhibition of translation initiation. Mol Cell Biol 2001; 21: Schiffmann R, Moller JR, Trapp BD, Shih HH, Farrer RG, Katz DA,
5018–5030 Alger JR, Parker CC, Hauer PE, Kaneski CR, Heiss JD, Kaye EM,
Leegwater PA, Konst AA, Kuyt B, Sandkuijl LA, Naidu S, Oude- Quarles RH,Brady RO,Barton NW.Childhood ataxia with dif-
jans CB, Schutgens RB, Pronk JC, van der Knaap MS. The fuse central nervous system hypomyelination. Ann Neurol
gene for leukoencephalopathy with vanishing white mat- 1994; 35: 331–340
ter is located on chromosome 3q27. Am J Hum Genet 1999; Schiffmann R, Tedeschi G, Kinkel P, Trapp BD, Frank JA, Kaneski
65: 728–734 CR, Brady RO, Barton NW, Nelson L, Yanovski JA. Leucodys-
Leegwater PA, Vermeulen G, Konst AA, Naidu S, Mulders J, Viss- trophy in patients with ovarian dysgenesis. Ann Neurol
er A, Kersbergen P, Mobach D, Fonds D, van Berkel CG, Lem- 1997; 41: 654–661
mers RJ, Frants RR, Oudejans CB, Schutgens RB, Pronk JC, Schneider RJ. Translational control during heat shock. In: So-
van der Knaap MS. Subunits of the translation initiation fac- nenberg N, Hershey JWB, Mathews MB (eds) Translational
tor eIF2B are mutant in leukoencephalopathy with vanish- Control of Gene Expression. Cold Spring Harbor Laboratory
ing white matter. Nat Genet 2001; 29: 383–388 Press, Cold Spring Harbor, 2000, pp 581–593
Leegwater PAJ, Pronk JC, van der Knaap MS. Leukoencephalo- Şenol U, Haspolat S, Karaali K, Lüleci E.MR imaging of vanishing
pathy with vanishing white matter: from magnetic reso- white matter. AMJ Am J Roentgenol 2000; 175; 826–828
nance imaging pattern to five genes. J Child Neurol 2003; Sugiura C, Miyata H, Oka A, Takashima S, Ohama E, Takeshita K.
18: 639–645 A Japanese girl with leukoencephalopathy with vanishing
Li W,Wang X, van der Knaap MS, Proud CG. Mutations linked to white matter. Brain Dev 2001; 23: 56–61
leukoencephalopathy with vanishing white matter impair Tedeschi G, Schiffmann R, Barton NW, Shin HH-L, Gospe SM,
the function of the eukaryotic initiation factor 2B complex Brady RO, Alger JR, di Chiro G. Proton magnetic resonance
in diverse ways. Mol Cell Biol 2004; 24: 3295–3306 spectroscopy imaging in childhood ataxia with diffuse cen-
Matsui M, Mizutani K, Miki Y, Mezaki T,Takahashi Y, Shibasaki H. tral nervous system hypomyelination. Neurology 1995; 45:
Adult-onset leukoencephalopathy with vanishing white 1526–1532
matter. Eur J Radiol Extra 2003; 46: 90–92 Topçu I,Saatci I,Anil Apak R,Söylemezoglu F.A case of leukoen-
Ohtake H,Shimohata T,Terajima K,Kimura T,Jo R,Kaseda R,Iizu- cephalopathy with vanishing white matter. Neuropadi-
ka O, Takano M, Akaiwa Y, Goto H, Kobayashi H, Sugai T, atrics 2000; 31: 100–103
Muratake T, Hosoki T, Shioiri T, Okamoto K, Onodera O,Tana- van der Knaap MS, Barth PG, Gabreels FJ, Franzoni E, Begeer JH,
ka K, Someya T, Nakada T, Tsuji S. Adult-onset leuko- Stroink H, Rotteveel JJ, Valk J. A new leukoencephalopathy
encephalopathy with vanishing white matter with a mis- with vanishing white matter. Neurology 1997; 48: 845–855
sense mutation in EIF2B5. Neurology 2004; 62: 1601–1603 van der Knaap MS, Kamphorst W, Barth PG, Kraaijeveld CL, Gut
Oldfield S, Jones BL, Tanton D, Proud CG. Use of monoclonal E,Valk J. Phenotypic variation in leukoencephalopathy with
antibodies to study the structure and function of eukaryot- vanishing white matter. Neurology 1998; 51: 540–547
ic protein synthesis initiation factor eIF-2B. Eur J Biochem van der Knaap MS,Wevers RA, Kure S, Gabreëls FJM,Verhoeven
1994; 221: 399–410 NM, van Raaij-Selten B, Jaeken J. Increased cerebrospinal
Prass K, Brück W, Schröder NWJ, Bender A, Prass M, Wolf T, fluid glycine: a biochemical marker for a leukoencepo-
van der Knaap MS, Zschenderlein R. Adult-onset leukoen- halopathy with vanishing white matter. J Child Neurol
cephalopathy with vanishing white matter presenting with 1999; 14: 728–731
dementia. Ann Neurol 2001; 50: 665–668 van der Knaap MS, Leegwater PA, Konst AA, Visser A, Naidu S,
Proud CG. Regulation of eukaryotic initiation factor eIF2B. Prog Oudejans CB, Schutgens RB, Pronk JC. Mutations in each of
Mol Subcell Biol 2001; 26: 95–114 the five subunits of translation initiation factor eIF2B can
Proud CG. Regulation of mammalian translation factors by cause leukoencephalopathy with vanishing white matter.
nutrients. Eur J Biochem 2002; 269:5338–5349 Ann Neurol 2002; 51: 264–270
Richardson JP, Mohammad SS, Pavitt GD. Mutations causing van der Knaap MS, van Berkel CGM, Herms J, van Coster R,
childhood ataxia with central nervous system hypomyeli- Baethmann M, Naidu S, Boltshauser E, Willemsen MAAP,
nation reduce eukaryotic initiation factor 2b complex for- Plecko B, Hoffmann GF, Proud CG, Scheper GC, Pronk JC.
mation and activity. Mol Cel Biol 2004; 24: 2352–2363 eIF2B Related Disorders: Antenatal Onset and Involvement
Rodriguez D, Gelot A, della Gaspera B, Robain O, Ponsot G, Sar- of Multiple Organs. Am J Hum Genet 2003; 73: 1199–1207
liève LL, Ghandour S, Pompidou A, Dautigny A, Aubourg P, van der Knaap MS, Leegwater PAJ, van Berkel CGM, Brenner C,
Pham-Dinh D. Increased density of oligodendrocytes in Storey E, di Rocco M, Salvi F, Pronk J. Arg113His mutation in
childhood ataxia with diffuse central hypomyelination eIF2B0 as cause of leukoencephalopathy in adults. Neurol-
(CACH) syndrome: neuropathological and biochemical ogy 2004; 62: 1598–1600
study of two cases. Acta Neuropathol (Berl)1999; 97: 469– Van Haren K, van der Voorn JP, Peterson DR, van der Knaap MS,
480 Powers JM. The life and death of oligodendrocytes in van-
Rosemberg S, Leite C da C, Arita FN, Kliemann SE, Lacerda MT. ishing white matter disease. J Neuropathol Exp Neurol
Leukoencephalopathy with vanishing white matter: report 2004; 63: 631–640
of four cases from three unrelated Brazilian families. Brain Watanabe I, Muller J. Cavitating “diffuse sclerosis” J Neu-
Dev 2002; 24: 250–256 ropathol Exp Neurol 1976; 26: 437–455
Rowlands AG, Montine KS, Henshaw EC, Panniers R. Physio- Welch WJ. Mammalian stress response: cell physiology, struc-
logical stresses inhibit guanine-nucleotide-exchange fac- ture/function of stress proteins, and implications for medi-
tor in Ehrlich cells. Eur J Biochem 1988; 175: 93–99 cine and disease. Physiol Rev 1992; 72: 1063–1081
References and Further Reading 1007

Wong K, Armstrong RC, Gyure KA, Morrison AL, Rodriguez D, Bönnemann CG, Meinecke P. Encephalopathy of infancy with
Matalon R, Johnson AB, Wollmann R, Gilbert E, Le TQ, intracerebral calcification and chronic spinal fluid lympho-
Bradley CA, Crutchfield K, Schiffmann R. Foamy cells with cytosis – another case of the Aicardi-Goutières syndrome.
oligodendroglial phenotype in childhood ataxia with dif- Neuropediatrics 1992; 23: 157–161
fuse central nervous system hypomyelination syndrome. Bönnemann CG, Meinecke P, Reich H. Encephalopathy with in-
Acta Neuropathol (Berl) 2000; 100: 635–646 tracerebral calcification, white matter lesions, growth hor-
mone deficiency, microcephaly, and retinal degeneration:
two sibs confirming a probably distinct entity. J Med Genet
1991; 28: 708–711
66 Aicardi-Goutières Syndrome Burn J,Wickramasinghe HT,Harding B,Baraitser M.A syndrome
with intracranial calcification and microcephaly in two sibs,
Abdel-Salam GMH, Zaki MS, Lebon P, Meguid NA. Aicardi- resembling intrauterine infection. Clin Genet 1986; 30:
Goutières syndrome: clinical and neuroradiological find- 112–116
ings of 10 new cases. Acta Paediatr 2004; 93: 929–936 Campbell IL, Krucker T, Steffensen S, Akwa Y, Powell HC, Lane T,
Aicardi J. Aicardi-Goutières syndrome: special type early-onset Carr DJ, Gold LH, Henriksen SJ, Siggins GR. Structural and
encephalopathy. Eur J Paediatr 2002; 6: A1-A7 functional neuropathology in transgenic mice with CNS
Aicardi J, Goutières F. A progressive familial encephalopathy in expression of IFN-a. Brain Research 1999; 835: 46–61
infancy with calcifications of the basal ganglia and chronic Crow Y. The genetics of Aicardi-Goutières syndrome. Eur J
cerebrospinal fluid lymphocytosis. Ann Neurol 1984; 15: Paediatr Neurol 2002; 6: A33-A35
49–54 Crow YJ, Jackson AP, Roberts E, van Beusekom E, Barth P, Corry
Aicardi J, Goutières F. Systemic lupus erythematosus or Aicar- P, Ferrie CD, Hamel BCJ, Jayatunga R, Karbani G, Kálmánchey
di-Goutières syndrome? Neuropediatrics 2000; 31: 113 R, Kelemen A, King M, Kumar R, Livingstone J, Massey R,
Akwa Y, Hassett DE, Eloranta ML, Sandberg K, Masliah E, Powell McWilliam R, Meager A, Rittey C, Stephenson JBP,Tolmie JL,
H, Lindsay Whitton J, Bloom FE, Campbell IL.Transgenic ex- Verrips A, Voit T, van Bokhoven H, Brunner HG, Woods CG.
pression of IFN-a the central nervous system of mice pro- Aicardi-Goutières syndrome displays genetic heterogene-
tects against lethal neurotropic viral infection but induces ity with one locus (AGS1) on chromosome 3p21. Am J Hum
inflammation and neurodegeneration. J Immunol 1998; Genet 2000; 67: 213–221
161: 5016–5026 Dale RC, Ping Tang S, Heckmatt JZ,Tatnall FM. Familial systemic
Al-Dabbous R, Sabry MA, Farah S, Al-Awadi SA, Sineonov S, lupus erythematosus and congenital infection-like syn-
Farag TI. The autosomal recessive congenital intrauterine drome. Neuropediatrics 2000; 31: 155–158
infection-like syndrome of microcephaly, intracranial calci- Fauré S, Bordelais I, Marquette C, Rittey C, Campos-Castello J,
fication, and CNS disease: report of another Bedouin fami- Goutières F, Ponsot G, Weissenbach J, Lebon P. Aicardi-
ly. Clin Dysmorphol 1998; 7: 127–130 Goutières syndrome: monogenic recessive disease, geneti-
Babitt DP, Tang T, Dobbs J, Berk R. Idiopathic familial cere- cally heterogeneous disease, or multifactorial disease? Clin
brovascular ferrocalcinosis (Fahr’s disease) and review of Genet 1999; 56: 149–153
differential diagnosis of intracranial calcification in chil- Goutières F, Aicardi J, Barth PG, Lebon P. Aicardi-Goutières syn-
dren. Am J Roentgenol Radiat Ther Nucl Med 1969; 105: drome: an update and results of interferon- a studies. Ann
352–358 Neurol 1998; 44: 900–907
Baraitser M, Brett EM, Piesowicz AT. Microcephaly and intracra- Jervis GA. Microcephaly with extensive calcium deposits and
nial calcification in two brothers. J Med Gen 1983; 20: 210– demyelination. J Neuropathol Exp Neurol 1954; 13: 318–
212 329
Barth PG.The neuropathology of Aicardi-Goutières syndrome. Kato M, Ishii R, Honma A, Ikeda H, Hayasaka K. Brainstem lesion
Eur J Paediatr Neurol 2002; 6: A27-A31 in Aicardi-Goutières syndrome. Pediatr Neurol 1998; 19:
Barth PG, Walter A, van Gelderen I. Aicardi-Goutières syn- 145–147
drome: a genetic microangiopathy? Acta Neuropathol Koul R, Chacko A, Surendranath J, Sankhla D. Aicardi-Goutières
(Berl) 1999; 98: 212–216 syndrome in siblings. J Child Neurol 2001; 16: 759–761
Billard C, Dulac O, Bouloche J, Echenne B, Lebon P, Motte J, Kuijpers TW. Aicardi-Goutières syndrome: immunophenotyp-
Robain O, Santini JJ. Encephalopathy with calcifications of ing in relation to interferon-alpha. Eur J Paediatr Neurol
the basal ganglia in children. A reappraisal of Fahr’s syn- 2002;6: A59-A64
drome with respect to 14 new cases. Neuropediatrics 1989; Kumar D, Rittey C, Cameron AH,Variend S. Recognizable inher-
20: 12–19 ited syndrome of progressive central nervous system de-
Black DN, Watters GV, Andermann E, Dumont C, Kabay ME, Ka- generation and generalized intracranial calcification with
plan P,Meagher-Villemure K,Michaud J,O’Gorman G,Reece overlapping phenotype of the syndrome of Aicardi and
E, Tsoukas C, Wainberg MA. Encephalitis among cree chil- Goutières. Am J Med Genet 1998; 75: 508–515
dren in Northern Quebec. Ann Neurol 1988; 24: 483–489 Lanzi G, Fazzi E, D’Arrigo S. Aicardi-Goutières syndrome: a de-
Blau N, Bonafé L, Krägeloh-Mann I,Thöny B, Kierat L, Häusler M, scription of 21 new cases and a comparison with the litera-
Ramaekers V. Cerebrospinal fluid pterins and folates in ture. Eur J Paediatr Neurol 2002; 6: A9-A22
Aicardi-Goutières syndrome. A new phenotype. Neurology Lebon P, Badoual J, Ponsot G, Goutières F, Hémeury-Cukier F,
2003; 61: 642–647 Aicardi J.Intrathecal synthesis of interferon-alpha in infants
Boltshauser E,Steinlin M, Boesch C, Martin E,Schubiger G.Mag- with progressive familial encephalopathy. J Neurol Sci
netic resonance imaging in infantile encephalopathy with 1988; 84: 201–208
cerebral calcification and leukodystrophy. Neuropediatrics Lebon P, Meritet JF, Krivine A, Rozenberg F. Interferon and
1991; 22: 33–35 Aicardi-Goutières syndrome. Eur J Paediatr Neurol 2002; 6:
A47-A53
1008 References and Further Reading

McEntagart M, Kamel H, Lebon P, King MD. Aicardi-Goutières Gayatri NA, Hughes MI, Lloyd IC, Wynn RF. Association of the
syndrome: an expanding phenotype. Neuropediatrics congenital bone marrow failure syndromes with retinopa-
1998; 29: 163–167 thy, intracerebral calcification and progressive neurological
Mehta L, Trounce JQ, Moore JR, Young ID. Familial calcification impairment. Eur J Paediatr Neurol 2002; 6: 125–128
of the basal ganglia with cerebrospinal fluid pleocytosis. Goutières F, Dollfus H, Becquet F, Dufier J-L. Extensive brain cal-
J Med Genet 1986; 23: 157–160 cification in two children with bilateral Coats’ disease. Neu-
Monastiri K, Salem N, Korbi S, Snoussi N. Microcephaly and in- ropediatrics 1990; 30: 19–21
tracranial calcification: two new cases. Clin Genet 1997; 51: Kajtár P, Méhes K. Bilateral coats retinopathy associated with
142–143 aplastic anaemia and mild dyskeratotic signs. Am J Med
Østergaard JR, Christensen T, Nehen AM. A distinct difference Genet 1994; 49: 374–377
in clinical expression of two siblings with Aicardi-Goutières Labrune P, Lacroix C, Goutières F, de Laveaucoupet J, Chevalier
syndrome. Neuropediatrics 1999; 30: 38–41 P, Zerah M, Husson B. Landrieu P. Extensive brain calcifica-
Polizzi A, Pavone P, Parana E, Incorpora G, Ruggieri M. Lack of tions, leukodystrophy, and formation of parenchymal cysts:
progression of brain atrophy in Aicardi-Goutières syn- a new progressive disorder due to diffuse cerebral mi-
drome. Pediatr Neurol 2001; 24: 300–302 croangiopathy. Neurology 1996; 46: 1297–1301
Razavi-Encha F, Larroche JC, Gaillard D. Infantile familial en- Nagae-Poetscher LM, Bibat G, Philippart M, Rosemberg S, Fate-
cephalopathy with cerebral calcifications and leukodystro- mi A, Lacerda MTC, Costa MOR, Kok F, Costa Leite C, Horská
phy. Neuropediatrics 1988; 19: 72–79 A, Barker PB, Naidu S. Leukoencephalopathy, cerebral calci-
Reardon W,Hockey A,Silberstein P,Kendall B,Farag TI,Swash M, fications, and cysts. New observations. Neurology 2004; 62:
Stevenson R, Baraitser M. Autosomal recessive congenital 1206–1209
intrauterine infection-like syndrome of microcephaly, in- Niedermeyer I, Reiche W, Graf N, Mestres P, Feiden W. Cere-
tracranial calcification, and CNS disease. Am J Med Genet broretinal vasculopathy and leukoencephalopathy mimic-
1994; 52: 58–65 king a brain tumor. Clin Neuropathol 2000; 19: 285–295
Schwarz KB, Ferrie CD, Woods CG. Two siblings with a new Revesz T, Fletcher S, Al-Gazali LI, DeBuse P. Bilateral retinopathy,
Aicardi-Goutières -like syndrome. Dev Med Child Neurol aplastic anaemia, and central nervous system abnormali-
2002; 44: 422–425 ties: a new syndrome? J Med Genet 1992; 29: 673–675
Slee J, Lam G, Walpole I. Syndrome of microcephaly, micro- Sazgar M, Leonard NJ, Renaud DL, Bhargava R, Sinclair DB. In-
phthalmia, cataracts, and intracranial calcification. Am J tracranial calcification, retinopathy, and osteopenia: a new
Med Genet 1999; 84: 330–333 syndrome? Pediatr Neurol 2002; 26: 324–328
Tolmie JL, Shillito P, Hughes-Benzie R, Stephenson JBP. The Tolmie JL, Browne BH, McGettrick PM, Stephenson JBP. A famil-
Aicardi-Goutières syndrome (familial, early onset en- ial syndrome with Coats’ reaction retinal angiomas, hair
cephalopathy with calcifications of the basal ganglia and and nail defects and intracranial calcification. Eye 1988; 2:
chronic cerebrospinal fluid lymphocytosis). J Med Genet 297–303
1995; 32: 881–884
Troost D, van Rossum A, Veiga Pires J, Willemse J. Cerebral cal-
cifications and cerebellar hypoplasia in two children: clini-
cal, radiologic and neuropathological studies – a separate
68 Leukoencephalopathy with Brain Stem
neurodevelopmental entity. Neuropediatrics 1984; 15: and Spinal Cord Involvement
102–109 and Elevated Lactate
Verrips A, Hiel JAP, Gabreëls FJM, Wesseling P, Rotteveel JJ. The
Aicardi-Goutières syndrome: variable clinical expression in Linnankivi T, Lundbom N, Autti T, Häkkinen AM, Koillinen H,
two siblings. Pediatr Neurol 1997; 16: 323–325 Kuusi T, Lönnqvist T, Saino K, Valanne L, Äärimaa T, Pihko H.
Vivarelli R, Grosso S, Cioni M, Galluzzi P, Monti L, Morgese G, Five new cases of a recently described leukoencephalopa-
Balestri P. Pseudo-TORCH syndrome or Baraitser-Reardon thy with high brain lactate. Neurology 2004; 63: 688–693
syndrome: diagnostic criteria. Brain Dev 2001; 23: 18–23 Serkov SV,Pronin IN,Bykova OV,Maslova OI,Arutyunov NV,Mu-
Wieczorek D, Gillessen-Kaesbach G, Passarge E. A nine-month- ravina TI, Kornienko VN, Fadeeva LM, Marks H, Bönnemann
old boy with microcephaly, cataracts, intracerebral calcifi- C, Schiffmann R, van der Knaap MS. Five patients with a re-
cations and dysmorphic signs: an additional observation of cently described novel leukoencephalopathy with brain-
an autosomal recessive congenital infection-like syn- stem and spinal cord involvement and elevated lactate.
drome? Genet Couns 1995; 6: 297-302 Neuropediatrics 2004; 35: 1–5
van der Knaap MS, van der Voorn P, Barkhof F, van Coster R,
Krägeloh-Mann I, Feigenbaum A, Blaser S, Vles JSH, Rieck-
67 Leukoencephalopathy mann P, Pouwels PJW. A new leukoencephalopathy with
brainstem and spinal cord involvement and high lactate.
with Calcifications and Cysts Ann Neurol 2003; 53: 252–258

Aynaci FM, Celep F, Ahmetoglu A. Encephalopathy with in-


tracranial calcification, dwarfism, leucodystrophy and neu-
ropathy: a new clinical entity? Brain Dev 2002; 24: 639–640
Crow YJ, McMenamin J, Haenggeli CA, Hadley DM, Tirupathi S,
Tracy EP, Zuberi SM, Browne BH, Tolmie JL, Stephanson JBP.
Coats’ plus: a progressive syndrome of bilateral Coats’
disease, characteristic cerebral calcification, leukoen-
cephalopathy, slow pre- and post-natal linear growth and
defects of bone marrow and integument. Neuropediatrics
2004; 35: 10–19
References and Further Reading 1009

69 Hypomyelination with Atrophy Burke JR, Wingfield MS, Lewis KE, Roses AD, Lee JE, Hulette C,
of the Basal Ganglia and Cerebellum Pericak-Vance MA,Vance JM.The Haw River syndrome: den-
tatorubral-pallidoluysian atrophy (DRPLA) in an African-
American family. Nat Genet 1994: 7: 521–524
van der Knaap MS, Naidu S, Pouwels PJW, Bonavita S, van
Filla A, Mariotti C, Caruso G, Coppola G, Cocozza S, Castaldo I,
Coster R, Lagae L, Sperner J, Surtees R, Schiffmann R, Valk J.
Calabrese O, Salvatore E, De Michelle G, Riggio MC,
New syndrome characterized by hypomyelination with
Pareyson D, Gellera C, Di Donato S. Relative frequencies of
atrophy of the basal ganglia and cerebellum. AJNR Am J
CAG expansions in spinocereballar ataxia and denta-
Neuroradiol 2002; 23: 1466–1474
torubropallidoluysian atrophy in 116 Italian families. Eur
Neurol 2000; 44: 31–36
Imamura A,Ito R,Tanaka S,Fukutomi O,Shimozawa N,Nishimu-
70 Hereditary Diffuse Leukoencephalo- ra M, Suzuki Y, Kondo N, Yamada M, Orii T. High-intensity
pathy with Neuroaxonal Spheroids proton and T2-weighted MRI signals in the globus pallidus
juvenile-type of dentatorubral and pallidoluysian atrophy.
Neuropediatrics 1994; 25: 234–237
Axelsson R, Röyttä M, Sourander P, Åkesson HO, Andersen O.
Koide R, Onodera O, Ikeuchi T, Kondo R, Tanaka H, Tokiguchi S,
Hereditary diffuse leucoencephalopathy with spheroids.
Tomoda A, Miike T, Isa F, Beppu H, Shimizu N, Watanabe Y,
Acta Psychiatr Scand 1984; 69 (suppl 314): 7–65
Horikawa Y, Shimohata T; Hirota K, Ishikawa A, Tsuji S. Atro-
Browne L, Sweeney BJ, Farrell MA. Late-onset neuraxonal leu-
phy of the cerebellum and brain stem in dentatorubral-pal-
coencephalopathy with spheroids and vascular amyloid.
lidoluysian atrophy. Influence of GAG repeat size on MR
Eur Neurol 2003; 50: 85–90
findings. Neurology 1997; 49: 1605–1612
Goodman LA, Dickson DW. Nonhereditary diffuse leukoen-
Lee I-H, Soong B-W, Lu Y-C, Chang Y-C. Dentatorubral-palli-
cephalopathy with spheroids presenting as early-onset
doluysian atrophy in Chinese. Arch Neurol 2001; 58:
rapidly progressive dementia. J Neuropathol Exp Neurol
1905–1908
1995; 54: 471
Lopez-Cendez I, Teive HGA, Calcagnotto ME, Da Costa JC, Car-
Hancock N, Poon M, Taylor B, McLean C. Hereditary diffuse
doso F, Viana E, Maciel JA, Radvany J, Arruda WO, Trevisol-
leukoencephalopathy with spheroids. J Neurol Neurosurg
Bittencourt PC,Neto PR,Silveira I,Steiner CE,Pinto Jr.W,San-
Psychiatry 2003; 74: 1345–1347
tos AS, Neto YC, Werneck LC, Araújo AQC, Carakushansky G,
Lampert PW. A comparative electron microscopic study of re-
Mello LR, Jardim LB, Rouleau GA. Frequency of the different
active, degenerating, regenerating, and dystrophic axons. J
mutations causing spinobellar ataxia (SCA1, SCA2, MJD/
Neuropathol Exp Neurol 1967; 26: 345–368
SCA3 and DRPLA) in a large group of Brazilian patients. Arq
Matsuyama H, Watanabe I, Mihm MC, Richardson EP. Derma-
Neuropsiquiatr 1997; 55: 519–529
toleukodystrophy with neuroaxonal spheroids. Arch Neu-
Melberg A, Dahl N, Hetta J,Valind S, Nennesmo I, Lundberg PO,
rol 1978; 35: 329–336
Raininko R. Neuroimaging study in autosomal dominant
Moro-de-Casillas ML, Cohen ML, Riley DE. Leucoencephalopa-
cerebellar ataxia, deafness, and narcolepsy. Neurology
thy with neuroaxonal spheroids (LENAS) presenting as the
1999; 53: 2190–2192
cerebellar subtype of multiple system atrophy. J Neurol
Miyazaki M, Hashimoto T, Yoneda Y, Tayama M, Harada M,
Neurosurg Psychiatry 2004; 75: 1070–1072
Miyoshi H, Kawano N, Murayama N, Kondo I, Kuroda Y. Pro-
Seitelberger F. Neuropathological conditions related to neu-
ton magnetic resonance spectroscopy in childhood-onset
roaxonal dystrophy. Acta Neuropathol (Berl) 1971; 21 (sup-
dentatorubral-pallidoluysian atrophy (DRPLA). Brain Dev
pl V); 17–29
1966; 18: 142–146
Torack R, Hughes CP. Neuroaxonal dystrophy in subacute de-
Mizukami K, Sasaki M, Shiraishi H, Koizumi J, Ogata T, Kosaka K.
mentia. Acta Neuropathol (Berl) 1972; 22: 267–268
An autopsied case of dentatorubropallidoluysian atrophy
van der Knaap MS, Naidu S, Kleinschmidt-DeMasters BK, Kam-
with atypical pathological features. Jpn J Psy Neurol 1992;
phorst W, Weinstein HC. Autosomal dominant diffuse
46: 749–754
leukoencephalopathy with neuroaxonal spheroids.Neurol-
Miyazaki M, Kato T, Hashimoto T, Harada M, Kondo I, Kuroda Y.
ogy 2000; 54: 463–468
MR of childhood-onset dentatorubral-pallidoluysian atro-
Yamashita M,Yamamoto T. Neuroaxonal leukoencephalopathy
phy. AJNR Am J Neuroradiol 1995; 16: 1834–1836
with axonal spheroids. Eur Neurol 2002; 48: 20–25
Muñoz E, Milà M, Sánchez A, Latorre P, Ariza A, Codina M, Balles-
Yazawa I, Nakano I,Yamada H, Oda M. Long tract degeneration
ta F. Tolosa E. Dentatorubral-pallidoluysian atrophy in a
in familial sundanophilic leukodystrophy with prominent
Spanish family: a clinical, radiological, pathological, and
spheroids. J Neurol Sci 1997; 147: 185–191
genetic study. J Neurol Neurosurg Psychiatry 1999; 67:
811–814
Nagafuchi S, Yanagisawa H, Sato K, Shirayama T, Ohsaki E, Bun-
71 Dentatorubropallidoluysian Atrophy do M,Takeda T,Tadokoro K, Kondo I, Murayama N,Tanaka Y,
Kikushima H, Umino K, Kurosawa H, Furukawa T, Nihei K, In-
Becher MW, Rubinztein DC, Loggo J, Wagster MV, Stine OC, Ra- oue T, Sano A, Komue O, Takahashi M, Yoshizawa T, Kanaza-
nen NG,Franz ML,Abbott MH,Sherr M,MacMillan JC,Barron wa I, Yamada M. Detatorubral and pallidoluysian atrophy
L, Porteous M, Harper PS, Ross CA. Dentatorubral-palli- expansion of an unstable CAG trinucleotide on chromo-
doluysian atrophy (DRPLA). Clinical and neuropathological some 12p. Nat Genet 1994; 6: 14–18
findings in genetically confirmed North American and Eu- Nagaoka U, Suzuki Y, Kawanami T, Kurita K, Shikama Y, Honda K,
ropean Pedigrees. Mov Disord 1997; 12: 519–530 Abe K, Nakajima T, Kato T. Regional differences in genetic
subgroup frequency in hereditary cerebellar ataxia, and a
morphometrical study of brain MR images in SCA1, MJD
and SCA6. J Neurol Sci 1999; 164: 187–194
1010 References and Further Reading

Robitaille Y,Lopez-Cendez I,Becher M,Rouleau G,Clark AW.The Benyamini H, Gunasekaran K,Wolfson H, Nussinov R. Conserva-
neuropathology of CAG repeat diseases: review and up- tion and amyloid formation: a study of the Gelsosin-like
date of genetic and molecular features. Brain Pathol 1997; family. Proteins 2003; 51: 266–282
7: 901–926 Bornebroek M, van Buchem MA, Haan J, Brand R, Lanser JBK, de
Silveira I, Miranda C, Guimarães, Moreira M-C, Alonso I, Men- Bruïne FT, Roos RAC. Hereditary cerebral hemorrhage with
donça P, Ferro A, Pinto-Basto J, Coelho J, Ferreirinha F, Poiri- amyloidosis-Dutch type: better correlation of cognitive de-
er J, Parreira E, Vale J, Januário C, Barbot C, Tuna A, Barros J, terioration with advancing age than with number of focal
Koide R, Tsuji S, Holmes SE, Margolis RL, Jardim L, Pandolfo lesions or white matter hyperintensities. Alzheimer Dis As-
M, Coutinho P, Seqeiros J.Trinucleide repeats in 202 families soc Disord 1996; 10: 224–231
with ataxia. Arch Neurol 2002; 59: 623–629 Bornebroek M, Haan J, Maat-Schieman MLC, van Duinen SG,
Smith JK, Gonda VE, Malamud N. Unusual form of cerebellar Roos RAC. Hereditary cerebral hemorrhage with amyloido-
ataxia: combined dentatorubral and pallidoluysian degen- sis-Dutch type (HCHWA-D). 1. A review of clinical, radiolog-
eration. Neurology 1958; 13: 266–269 ic and genetic aspects. Brain Pathol 1996; 6: 111–114
Tsuchiya K, Oyanani S, Arima K, Ikeda K, Akashi T, Ando S. Kuro- Bornebroek M,Haan J,Backhovens H,Deutz P,van Buchem MA,
sawa T, Ikeuchi T, Tsuji S. Dentatorubropallidoluysian atro- van der Broeke M, Bakker E, Roos RAC, van Broeckhoven C.
phy: clinicopathological study of dementia and involve- Presenilin-1 polymorphism and hereditary cerebral hemor-
ment of the nucleus basalis of Meynert in seven autopsy rhage with amyloidosis, Dutch type. Ann Neurol 1997; 42:
cases. Acta Neuropathol (Berl) 1998; 96: 502–508 108–110
Uyama E, Kondo I, Uchino M, Fukushima T, Murayama N, Bots GTAM. Neuropathological findings in cerebral B-protein
Kuwano A, Inokuchi N, Ohtani Y, Ando M. Dentatorubral- amyloidosis. Differences and similarities in those cases pre-
pallidoluysian atrophy (DRPLA): clinical, genetic and neuro- senting as a cerebral hemorrhage and those presenting as
radiologic studies in a family. J Neurol Sci 1995; 130: a dementia of the Alzheimer type. Clin Neurol Neurosurg
146–153 1992; 94 (suppl): S52-S53
Violante V, Luongo A, Pepe S, Annunziata S, Gentile V.Transglu- Burgermeister P, Calhoun ME, Winkler DT, Jucker M. Mecha-
taminase-dependent formation of protein aggregates as nisms of cerebrovascular amyloid deposition. Lessons from
possible biochemical mechanism for polyglutamine dis- mouse models. Ann NY Acad Sci 2000; 903: 307–316
eases. Brain Res Bull 2001; 56: 169–172 Caulo M, Tampieri D, Brassard R, Guiot MC, Malanson D. Cere-
Warner TT, Lennox GC, Janota I, Harding E. Autosomal-domi- bral amyloid angiopathy presenting as nonhemorrhagic
nant dentatorubropallidoluysian atrophy in the United diffuse encephalopathy: neuropathologic and neuroradio-
Kingdom. Mov Disord 1994; 3: 289–296 logic manifestations in one case. AJNR Am J Neuroradiol
Warner TT,Williams LD,Walker RWH, Flinter F, Robb SA, Bundey 2001; 22: 1072–7076
SE, Honavar M, Harding AE. A clinical and molecular genet- Crooks DA. Cerebral amyloid angiopathy. J Neurol Neurosurg
ic study of dentatorubropallidoluysian atrophy in four Psychiatry 1994; 57: 1457
European families. Ann Neurol 1995; 37: 452–459 Cuny E, Loiseau H, Rivel J, Vital C, Castel J-P. Amyloid angiopa-
Yabe I, Sasaki H, Kikuchi S, Nonaka M, Moriwake F, Tashiro K. thy-related cerebellar hemorrhage. Surg Neurol 1996; 46:
Late onset ataxia phenotype in dentatorubro-palli- 235–239
doluysian atrophy (DRPLA). J Neurol 2002; 249: 432–436 De la Monte S, Sohn Y, Etienne D, Kraft J,Wands JR. Role of aber-
Yamada M, Sato T,Tsuji S,Takahashi H. Oligodendrocytic polyg- rant nitric oxide synthase-3 expression in cerebrovascular
lutamine pathology in dentatorubral-pallidoluysian atro- degeneration and vascular-mediated injury in Alzheimer’s
phy. Ann Neurol 2002; 52: 670–674 disease. Ann NY Acad Sci 2000; 903: 61–71
Yoshii F, Tomiyashu H, Shinohara Y. Fluid attenuation inversion Fountain NB, Eberhart DA. Primary angiitis of the cerebral ner-
recovery (FLAIR) images of dentatorubropallidoluysian at- vous system associated with cerebral amyloid angiopathy.
rophy: case report. J Neurol Neurosurg Psychiatry 1998; 65: Neurology 1996; 46: 190–197
396–399 Fountain NB, Lopez MB. Control of primary angiitis of the CNS
Zoghbi HY, Orr HT. Glutamine repeats and neurodegeneration. associated with cerebral amyloid angiopathy by cyclo-
Ann Rev Neurosci 2000; 23: 217–247 phosphamide alone. Neurology 1999: 52: 660–662
Frangione B,Vidal R,Rostagno A,Ghiso J.Familial cerebral amy-
loid angiopathies and dementia. Alzheimer Dis Assoc Dis-
ord 2000; 14 (suppl 1): S25-S29
72 Cerebral Amyloid Angiopathy Frangione B, Révész T, Vidal R, Holton J, Lashley T, Houlden H,
Wood N, Rostagno A, Plant G, Ghiso J. Familial cerebral amy-
Abrahamson M, Jonsdottir S, Olafsson I, Jensson O, Grubb A. loid agiopathy related to stroke and dementia. Amyloid
Hereditary cystin C amyloid angiopathy: indentification of 2004; 8 (suppl 1): 36–42
the disease-causing mutation and specific diagnosis by Friedland RP, Majocha RE, Reno JM, Lyle LR, Marotta CA. Devel-
polymerase chain reaction based analysis. Hum Genet opment of an anti-ab monoclonal antibody for in vivo
1992; 89: 377–380 imaging of amyloid angiopathy in Alzheimer’s disease. Mol
Atwood CS, Bishop GM, Perry G, Smith MA. Amyloid-b: a vascu- Neurobiol 1994; 9: 107–113
lar sealant that protects against hemorrhage? J Neurosci Gambetti P, Russo C. Human brain amyloidoses. Nephrol Dial
Res 2002; 70: 356 Transplant 1998; 13: 33–40
Baumann MH, Wisniewski T, Levy E, Plant GT, Ghiso J. C-Termi- Good CD, NG VWK, Clifton A, Britton JA, Hart Y, Wilkins P. Amy-
nal fragments of a- and b-tubulin form amyloid fibrils in vit- loid angiopathy causing widespread miliary haemorrhages
ro and associate with amyloid deposits of familial cerebral within the brain evident on MRI. Neuroradiology 1998; 40:
amyloid angiopathy, British type. Biochem Biophys Res 308–311
Commun 1996; 219: 238–242
References and Further Reading 1011

Graffagnino C, Herbstreith MH, Schmechel DE, Levy E, Roses Imaoka K, Kobayashi S, Fujihara S, Shimode K, Nagasaki M.
AD, Alberts MJ. Cystatin C mutation in an eldery man with Leukoencephalopathy with cerebral amyloid angiopathy: a
sporadic amyloid angiopathy and intracerebral hemor- semiquantitive and morphometric study. J Neurol 1999;
rhage. Stroke 1995; 26: 2190–2193 246: 661–666
Gray F, Dubas F, Roullet E, Escourolle R. Leukoencephalopathy Itoh Y,Yamanda M, Hayakawa M, Otomo E, Miyatake T. Cerebral
in diffuse hemorrhagic cerebral amyloid angiopathy. Ann amyloid angiopathy: a significant cause of cerebellar as
Neurol 1985; 18: 54–59 well as lobar cerebral hemorrhage in the elderly. J Neurol
Greenberg SM. Cerebral amyloid angiopathy. Prospects for Sci 1993; 116: 135–141
clinical diagnosis and treatment. Neurology 1998; 51: Itoh Y, Yamada M, Suematsu N, Matsushita M, Otomo E. Influ-
690–694 ence of apolipoprotein E genotype on cerebral amyloid an-
Greenberg SM. Cerebral amyloid angiopathy and dementia. giopathy in the eldery. Stroke 1996; 27: 216–218
Two amyloids are worse than one. Neurology 2002; 58: Kim D-E, Bae H-J, Lee S-H, Kim H, Yoon B-W, Roh J-K. Gradient
1587–1588 echo magnetic resonance imaging in the prediction of
Greenberg SM. Cerebral amyloid angiopathy and vessel dys- hemorrhagic vs ischemic stroke. A need for the considera-
function. Cerebrovasc Dis 2002; 13 (suppl 2): 42–47 tion of the extent of leukoaraiosis. Arch Neurol 2002: 59:
Greenberg SM, Hyman BT. Cerebral amyloid angiopathy and 425–429
apoliprotein E: bad news for the good allele? Ann Neurol Knudson KA, Rosand J, Karluk D, Greenberg SM. Clinical diag-
1997; 41: 701–702 nosis of cerebral amyloid angiopathy: validation of the
Greenberg SM, Vonsattel JPG. Diagnosis of cerebral amyloid Boston Criteria. Neurology 2001; 56: 537–539
angiopathy. Sensitivity and specificity of cortical biopsy. Levey AI, Heilman CJ, Lah JJ, Nash NR, Rees HD, Wakai M, Mirra
Stroke 1997; 28: 1418–1422 SS, Rey DB, Nochlin D, Brid TD, Mufson EJ. Preselin-1 protein
Greenberg SM, Vonsattel JPG, Stakes JW, Gruber M, Finklestein expression in familial and sporadic Alzheimer’s disease.
SP. The clinical spectrum of cerebral amyloid angiopathy: Ann Neurol 1997; 41: 742–753
presentations without lobar hemorrhage. Neurology 1993; Liberski PP, Barcikowska M. Pathology of the vessels in cerebral
43; 2073–2079 amyloid angiopathy. Folia Neuropathol 1995; 33: 207–214
Greenberg SM, Rebeck W,Vonssattel JPG, Gomez-Isla T, Hyman Loes DJ, Biller J, Yuh WTC, Hart MN, Godersky JC, Adams Jr HP,
BT. Apolipoprotein E e4 and cerebral hemorrhage associat- Keefauver SP, Tranel D. Leukoencephalopathy in cerebral
ed with amyloid angiopathy. Ann Neurol 1995; 38: 254–259 amyloid angiopathy: MR imaging in four cases. AJNR Am J
Greenberg SM, Briggs ME, Hyman BT, Kokoris GH, Takis C, Kan- Neuroradiol 1990; 11: 485–488
ter DS, Kase CS, Pessin MS. Apolipoprotein E e4 is associated Maat-Schieman MLC, Radder CM, van Duinen SG, Roos RAC.
with the presence and earlier onset of hemorrhage in cere- Hereditary cerebral hemorrhage with amyloidosis (Dutch):
bral amyloid angiopathy. Stroke 1996; 27: 1333–1337 a model for congophilic plaque formation without neuro-
Haan J, Roos RAC. Comparison between the Icelandic and fabrillary pathology. Acta Neuropathol (Berl) 1994; 88: 371–
Dutch forms of hereditary cerebral amyloid angiopathy. 378
Clin Neurol Neurosurg 1992; 94 (suppl): S82-S83 Maury CPJ, Liljeström M, Boysen G, Törnroth T, de la Capelle A,
Haan J, Bakker E, Jennekens-Schinkel A, Roos RAC. Progressive Nurmiaho-Lassila E-L. Danish type gelsolin related amyloi-
dementia, without cerebral hemorrhage, in a patient with dosis: 654G-T mutation is associated with a disease patho-
hereditary cerebral amyloid angiopathy.Clin Neurol Neuro- genetically and clinically similar to that caused by the
surg 1992; 94: 317–318 654G-A mutation (familial amyloidosis of the Finnish type).
Haan J, Maat-Schieman MLC, van Duinen MLC, Jensson O, J Clin Pathol 2000; 53: 95–99
Thorsteinsson L, Roos RAC. Co-localization of b/A4 and cys- McCarron MO, Nicoll JAR. Apolipoprotein E genotype and
tatin C in cortical blood vessels in Dutch, but not in Ice- cerebral amyloid angiopathy-related hemorrhage. Ann NY
landic hereditary cerebral hemorrhage with amyloidosis. Acad Sci 2000; 903: 176–179
Acta Neurol Scand 1994; 89: 367–371 Mehdorn HM, Gerhard L, Müller SP, Olbirch HM. Clinical and
Haan J,Maat-Schieman MLC,Roos RAC.Clinical aspects of cere- cerebral blood flow studies in patients with intracranial he-
bral amyloid angiopathy. Dementia 1994; 5: 210–213 morrhage and amyloid angiopathy typical of Alzheimer’s
Haan J, Roos RAC, Bakker E. No prospective effect of apolipo- disease. Neurosurg Rev 1992;15: 111–116
protein E e2 allele in Dutch hereditary cerebral amyloid an- Natté R, Maat-Schieman MLC, Haan J, Bornebroek M, Roos RAC,
giopathy. Ann Neurol 1995; 37: 282 van Duinen SG. Dementia in hereditary cerebral hemor-
Hendricks HT, Franke CL, Theunissen PHMH. Cerebral amyloid rhage with amyloidosis-Dutch type is associated with cere-
angiopathy: diagnosis by MRI and brain biopsy. Neurology bral amyloid angiopathy but is independent of plaques
1991; 40: 1308–1310 and neurofibrillary tangles. Ann Neurol 2001; 50: 765–771
Holton JL, Ghiso J, Lashley T, Rostagno A, Guerin CJ, Gibb G, Nicoll JAR,Burnett C,Love S,Graham DI,Ironside JW,Vinters HV.
Houlden H, Ayling H, Martinain L, Anderton BH, Wood NW, High frequency of apolipoprotein E e2 in patients with
Vidal R, Plant G, Frangione B, Revesz T. Regional distribution cerebral hemorrhage due to cerebral amyloid angiopathy.
of amyloid-bri deposition and its association with neu- Ann Neurol 1996; 396: 682–683
rofibrillary degeneration in familial British dementia. Am J Nicoll JAR, Burnett C, Love S, Graham DI, Dewar D, Ironside JW,
Pathol 2001; 158: 515–526 Stewart J, Vinters HV. High frequency of apoliprotein E e2
Holtzman DM. Role of apoE/Ab interactions in the pathogene- allele in hemorrhage due to cerebral amyloid angiopathy.
sis of Alzheimer’s disease and cerebral amyloid angiopathy. Ann Neurol 1997; 41: 716–721
J Mol Neurosci 2001; 17: 147–155 Oh U, Gupta R, Krakauer JW, Khandjii AG, Chin Ss, Elkind MSV.
Hyman BT, Tanzi RE. Amyloid, dementia and Alzheimer’s dis- Reversible leukoencephalopathy associated with cerebral
ease. Curr Opin Neurol Neurosurg 1992; 5: 88–93 amyloid angiopathy. Neurology 2004; 62: 494–497
1012 References and Further Reading

Oide T, Takahashi H, Yutani C, Ishihara T, Ikeda S-I. Relationship Vonsattel JPG, Myers RH, Hedley-Whyte ET, Ropper AH, Bird ED,
between lobar intracerebral hemorrhage and leukoen- Richardson EP Jr.Cerebral amyloid angiopathy without and
cephalopathy associated with cerebral amyloid angio- with cerebral hemorrhages: a comparative histological
pathy: clinicopathological study of 64 Japanese patients. study. Ann Neurol 1991; 30: 637–649
J Protein Folding Disord 2003; 10: 136–143 Walker LC. Animal models of cerebral ß-amyloid angiopathy.
Ólafsson Í, Grubb A.Hereditary cystatin C amyloid angiopaghy. Brain Res Rev 1997: 25: 70–84
Int J Exp Clin Invest 2000; 7: 70–79 Wattendorf AR, Frangione B, Luyendijk W, Bots GTAM. Heredi-
Olichney JM, Hansen LA, Hofstetter R, Grundman M, Katzman tary cerebral hemorrhage with amyloidosis, Dutch type
R, Thal LJ. Cerebral infarction in Alzheimer’s disease is asso- (HCHWA-D): clinicopathological studies. J Neurol Neuro-
ciated with severe amyloid angiopathy and hypertension. surg Psychiatry 1995; 58: 699–705
Arch Neurol 1995; 52: 702–708 Weller RO, Massey A, Kuo Y-M, Rohler AE. Cerebral amyloid an-
Olichney JM, Hansen LA, Lee JH, Hofstetter CR, Katzman R,Thal giopathy: accumulation of Aß in interstitial fluid drainage
LJ. Relationship between severe amyloid angiopathy, pathways in Alzheimer’s disease. Ann NY Acad Sci 2000;
apolipoprotein E genotype and vascular lesions in 903: 110–117
Alzheimer’s disease. Ann NY Acad Sci 2000; 903: 138–143 Yamada M. Cerebral amyloid angiopathy: an overview. Neu-
O’Riordan S, McMonagle P, Janssen JC, Fox NC, Farell M, ropathology 2000; 1: 8–22
Collinge J, Rossor MN, Hutchinson M. Preselin-1 mutation Yoshimura M, Yamanouchi H, Kuzuhara S, Mori H, Sugiura S,
(E280G), spastic paraparesis, and cranial MRI white matter Mizutani T, Shimada H, Tomonaga M, Toyokura Y. Dementia
abnormalities. Neurology 2002; 59: 1108–1110 in cerebral amyloid angiopathy: a clinicopathological
Osumi AK, Tien RD, Felsberg GJ, Rosenbloom M. Cerebral amy- study. J Neurol 1992; 239: 441–450
loid angiopathy presenting as a brain mass. AJNR Am J
Neuroradiol 1995; 16: 911–915
Pfeifer LA, White LR, Ross GW, Petrovitch H, Launer LJ. Cerebral
amyloid angiopathy and congnitive function. Neurology
73 Cerebral Autosomal Dominant
2002; 58: 1629–1634 Arteriopathy with Subcortical Infarcts
Prior R, Wihl G, Urmoneit B. Apolipoprotein E, smooth muscle and Leukencephalopathy
cells and the pathogenesis of cerebral amyloid angiopathy:
the potential role of impaired cerebrovascular Ab clear- Abe K, Murakami T, Matsubara E, Manabe Y, Nagano I, Shoji M.
ance. Ann NY Acad Sci 2000; 903: 180 – 186 Clinical features of CADASIL. Ann NY Acad Sci 2002; 977:
Revasz T, Holton JL, Lashley T, Plant G, Rostagno A, Ghiso J, Fran- 266–272
gione B. Sproradic and familial cerebral amyloid an- Arbodela-Valasquez JF, Lopera F, Lopez E, Frosch MP, Sepulve-
giopathies. Brain Pathol 2002; 12: 343–357 da-Falla D,Gutierrez JE,Vargas S,Medina M,Martinez de Ari-
Roob G, Fazekas F. Magnetic resonance imaging of cerebral eta C, Lebo RV, Slangenhaupt SA, Betensky RA, Villegas A,
microbleeds. Curr Opin Neurol 2000; 13: 69–73 Arcos-Burgos M, Rivers D, Restrepo JC, Kosik KS. C455R
Sarazin M, Amarenco P, Mikol J, Dimitri D, Lot G, Bousser MG. notch3 mutation in a Colombian CADASIL kindred with ear-
Reversible leukoencephalopathy in cerebral amyloid an- ly onset of stroke. Neurology 2002; 59: 277–279
giopathy presenting as subacute dementia. Eur J Neurol Auer DP, Schirmer T, Heidenreich JO, Herzog J, Pütz B, Dichgans
2002; 9: 353–358 M. Altered white and gray matter metabolism in CADASIL.
Schubert P, Morino T, Miyazaki H, Ogata T, Nakamura Y, Marchi- A proton MR spectroscopy and 1H-MRSI study. Neurology
ni C, Ferroni S. Cascading glia reactions: a common patho- 2001; 56: 635–642
mechanism and its defferentiated control by cycling neu- Auer DP, Pütz B, Gössl C, Elbel G-K, Gasser T, Dichgans M. Differ-
cleotide signaling. Ann NY Acad Sci 2000; 903: 24–33 ential lesion patterns in CADASIL and sporadic subcortical
Scully RE, Mark EJ, McNeely WF, Ebeling SH, Ellender SM. Pre- arteriosclerotic encephalopathy: MR imaging study with
sentation of case. N Engl J Med 2000; 342: 957–965 statistical parametric group comparison. Radiology 2001;
Shimode K, Kobayashi S, Imaoka K, Umegae N, Nagai A. 218: 443–451
Leukoencephalopathy-related cerebral amyloid angio- Bruening R, Dichgans M, Berchtenbreiter C,Yousry T, Seelos KC,
pathy with cystatin C deposition. Stroke 1996; 27; 1417– Wu RH, Mayer M, Brix G, Reiser M. Cerebral autosomal dom-
1419 inant arteriopathy with subcortical infarcts and leukoen-
Sillus M, Saeger W, Linke RP, Müller D,Voigt C. Cerebral amyloid cephalopathy: decrease in regional cerebral blood volume
angiopathy. Frequency, significance and immunohisto- in hyperintense subcortical lesions inversely correlates
chemistry. Zentrabl Pathol 1993; 139: 2074–215 with disability and cognitive performance. AJNR Am J
Sveinbjörnsdóttir S, Blöndal H, Gudmundsson G, Kjartansson Neuroradiol 2001; 22: 1268–1274
O, Jónsdóttir S, Gudmundsson G. Progressive dementia and Brulin P, Godfraind C, Leteurtre E, Ruchoux M-M. Morphometric
leukoencephalopathy as the initial presentation of late on- analysis of ultrastructural vascular changes in CADASIL:
set hereditary cystatin-C amyloidosis. Clinical presentation analysis of 50 skin biopsy specimens and pathogenic impli-
of two cases. J Neurol Sci 1996; 140: 101–108 cations. Acta Neuropathol (Berl) 2002; 104: 241–248
Van Duinen SG, Maat-Schieman MLC, Bruijn JA, Haan J, Roos Ceroni M, Poloni TE, Tonietti S, Fabozzi D, Uggetti C, Frediani F,
RAC. Cortical tissue of patients with hereditary cerebral he- Simonetti F, Malaspina A, Alimonti D, Celano M, Ferrari M,
morrhage with amyloidosis (Dutch) contains various extra- Carrera P. Migraine with aura and white matter abnormali-
cellular matrix deposits. Lab Invest 1995; 73: 183–189 ties: notch3 mutation. Neurology 2000: 54: 1869–1871
Van Nostrand WE, Melchor J, Wagner M, Davis J. Cerebrovascu- Chabriat H, Mrissa R, Levy C, Vahedi K, Taillia H, Iba-Zizen MT,
lar smooth muscle cell surface fibrillar Ab. Alteration of the Joutel A, Tournier-Lasserve E, Bousser MG. Brian stem MRI
proteolytic environment in the cerebral vessel wall. Ann NY signal abnormalities in CADASIL. Stroke 1999; 30: 457–459
Acad Sci 2000; 903: 89 – 96
Vinters HV. Cerebral amyloid angiopathy and Alzheimer’s dis-
ease: two entities or one? J Neurol Sci 1992; 112: 1–3
References and Further Reading 1013

Chabriat H, Pappata S, Poupon F, Clark CA, Vahedi K, Poupon F, Furby A, Vahedi K, Force, Larrouy S, Ruchoux M-M, Joutel A,
Magin JF, Pachot-Clouard M, Jobert A, le Bihan D, Bousser Tournier-Lasserve E. Differential diagnosis of a vascular
M-G. Clinical severity in CADASIL related to ultrastructural leukoencephalopathy within a CADASIL family: use of skin
damage in white matter. In vivo study with diffusion tensor biopsy electron microscopy study and direct genotypic
MRI. Stroke 1999; 30: 2637–2643 screening. J Neurol 1998; 245: 734–740
Chabriat H, Pappata S, Ostergaard L, Clark CA, Pachot-Clouard Iannucci G, Dichgans M, Rovaris M, Brüning R, Gasser T, Giaco-
M, Valhedi K, Jobert A, le Bihan D, Bousser MG. Cerebral he- motti L, Yousry TA, Filippi M. Correlations between clinical
modynamics in CADASIL before and after acetazolamide findings and magnetization transfer imaging metrics of tis-
challenge assessed with MRI bolus tracking. Stroke 2000; sue damage in individuals with cerebral autosomal domi-
31: 1904–1912 nant arteriopathy with subcortical infarcts and leukoen-
Chawda SJ, de Lange RPJ, Hourihan MD, Halpin SFS, St Clair D. cephalopathy. Stroke 2001; 32: 643–648
Diagnosing CADASIL using MRI: evidence from families Ito D, Tanahasi N, Murata M, Sato H, Saito I, Wanatabe K,
with known mutations of Notch 3 gene. Neuroradiology Fukkuchi Y. Notch3 gene polymorphism and ischaemic
2000; 42: 249–255 cerebrovascular disease. J Neurol Neurosurg Psychiatry
Coulthard A, Blank SC, Bushby K, Kalaria RN, Burn DJ. Distribu- 2002; 72: 382–384
tion of cranial MRI abnormalities in patients with sympto- Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton
matic and subclinical CADASIL. Br J Radiol 2000; 73: P, Alamowitch S, Domenga V, Cécillion M, Maréchal E, Maci-
256–265 azek J, Vayssière C, Cruaud C, Cabanis E-A, Ruchoux MM,
Davous P. CADASIL; a review with proposed diagnostic criteria. Weissenbach J, Bach JF, Bousser MG, Tournier-Lasserve E.
Eur J Neurol 1998; 5: 219–233 Notch3 mutations in CADASIL, a hereditary adult-onset
De Lange RPJ, Bolt J, Reid E, da Silva R, Shaw DJ, St Clair DM. condition causing stroke and dementia. Nature 1996; 383:
Screening British CADASIL families for mutations in the 707–710
NOTCH3 gene. J Med Genet 2000; 37: 224–225 Joutel A, Vahedi K, Corpechot C, Troesch A, Chabriat H,
Desmond DW, Moroney JT, Lynch T, Chan S, Mohr JP.The natur- Vayssière C, Cruaud C, Maciazek M, Weissenbach J, Bousser
al history of CADASIL. A pooled analysis of previously pub- M-G, Bach J-F, Tournier-Lasserve E. Strong clustering and
lished cases. Stroke 1999; 30: 1230–1233 stereotyped nature of Notch3 mutations in CADASIL pa-
Dichans M, Mayer M, Uttner I, Brüning R, Müller-Höcker J, Rung- tients. Lancet 1997; 350: 1511–1515
ger G,Ebke M,Klockgether T,Gasser T.The phenotypic spec- Joutel A, Andreux F, Gaulis S, Domenga V, Cecillon M, Battail N,
trum of CADASIL: clinical findings in 102 cases. Ann Neurol Piga N, Chapon F, Godfrain C, Tournier-Lasserve E. The
1998; 44: 731–739 ectodomain of the Notch3 receptor accumulates within the
Dichgans M, Filippi M, Brüning R, Iannucci G, Berchtenbreiter C, cerebrovasculature of CADASIL patients. J Clin Invest 2000;
Minicucci L,Uttner I,Crispin A,Ludwig H,Gasser T,Yousry TA. 105: 597–605
Quantitative MRI and CADASIL. Correlation with disability Joutel A, Dodick DD, Parisi JE, Cecillon M, Tournier-Lasserve E,
and cognitive performance. Neurology 1999; 52: 1361– Bousser MG. De novo mutation in the notch3 gene causing
1367 CADASIL. Ann Neurol 2000; 47: 388–391
Dichgans M, Holtmannspötter M, Herzog M, Herzog J, Peters N, Joutel A, Monet M, Domega V, Riant F, Tournier-Lasserve E.
Bergmann M, Yousry TA. Cerebral microbleeds in CADASIL. Pathogenic mutations associated with cerebral autosomal
A gradient-echo magnetic resonance imaging and autopsy dominant arteriopathy with subcortical infarcts and
study. Stroke 2002; 33: 67–71 leukoencephalopathy differently affect jagged1 binding
Donahue CP, Kosik KS. Distribution patter of Notch3 mutations and Notch3 activity via the RBP/JK signaling pathway. Am J
suggests a gain-of-function mechanisms for CADASIL. Hum Genet 2004; 74: 338–347
Genomics 2004; 83: 59–65 Kalimo H,Viitanen M, Amberlat K, Juvonen V, Marttila R, Pöyhö-
Ducros A, Nagy T, Alamowitch S, Nibbio A, Joutel A, Vahedi K, nen M, Rinne JO, Savontaus M-L, Tuisku S, Winblad B.
Chabriat H, Iba-Zizen MT, Julien J, Davous P, Goas JY, Lyon- CADASIL: hereditary disease of arteries causing brain in-
Caen O, Dubois B, Ducrocq X, Salsa F, Ragno M, Burkhard P, farcts and dementia. Neuropathol Appl Neurobiol 1999; 25:
Bassetti C, Hutchinson M, Vérin M, Viader F, Chapon F, 257–265
Levasseur M, Mas JL, Delrieu O, Maciazek J, Prieur M, Karlström H, Beatus P, Dannaeus K, Chapman G, Lendahl U,
Mohrenweiser H, Bach JF, Bousser MG, Tournier-Lasserve E. Lundkvist J. A CADASIL-mutated Notch 3 receptor exhibits
Cerebral autosomal dominant arteriopathy with subcorti- impaired intracellular trafficking and maturation but nor-
cal infarcts and leukoencephalopathy, genetic homogene- mal ligand-induced signaling. Proc Natl Acad Sci 2002; 99:
ity, and mapping of the locus within a 2-cM interval. Am J 17119–17124
Hum Genet 1996; 58: 171–181 LaPoint SF, Patel U, Rubio A. Cerebral autosomal dominant
Engelter ST, Rueegg S, Kirsch EC, Fluri F, Probst A, Steck AJ, arteriopathy with subcortical infarcts and leukoencephalo-
Lyrer PA. CADASIL mimicking primary angiitis of the central pathy (CADASIL). Adv Anat Pathol 2000; 7: 307–321
nervous system. Arch Neurol 2002; 59: 1480–1483 Le Bler I, Carluer L, Derache N, Lalevée C, Ledoze F, Defer GL.
Feuerhake F, Volk B, Ostertag CB, Jungling FD, Kassubek J, Unusual presentation of CADASIL with reversible coma
Orszagh M, Dichgans M. Reversible coma with raised and confusion. Neurology 2002; 59: 1115–1116
intracranial pressure: an unusual clinical manifestation of Lesnik Oberstein SAJ, van den Boom R, Middelkoop HAM, Fer-
CADASIL. Acta Neuropathol (Berl) 2002; 103: 188–192 rari MD, Knaap YM, van Houwelingen HC, Breuning MH, van
Filley CM, Thompon LL, Sze C-I, Simon JA, Paskavitz JF, Klein- Buchem MA, Haan J. Incipient CADASIL. Arch Neurol 2003;
schmidt-deMasters BK.White matter dementia in CADASIL. 60: 707–712
J Neurol Sci 1999; 163: 163–167 Lesnik Oberstein SAJ, van Duinen SG, van den Boom R, Maat-
Schieman MLC, van Buchem MA, van Houwelingen HC,
Hegeman-Kleinn IM, Ferrari MD, Breuning MH, Haan J. Eval-
uation of diagnostic NOTCH3 immunostaining in CADASIL.
Acta Neuropathol (Berl) 2003; 106: 107–111
1014 References and Further Reading

Malandrini A, Carrera P, Palmeri S, Cavellaro T, Fabrizi GM, Vil- Santa Y, Uyama E, Chui de H, Arima M, Kotorii S, Takahashi K,
lanova M, Fattapposta M, Vismara L, Brancolini V, Tanganelli Tabira T. Genetic, clinical and pathological studies of
P, Calì A, Morocutti C, Zeviani M, Ferrari M, Guazzi GC. Clini- CADASIL in Japan: a partial contribution of Notch3 muta-
copathological and genetic studies of two further Italian tions and implications of smooth muscle cell degeneration
families with cerebral dominant arteriopathy. Acta Neu- for the pathogenesis. J Neurol Sci 2003; 212: 79–84
ropathol (Berl) 1996; 92: 115–122 Schon F, Martin RJ, Prevett M, Clough C, Enevoldson TP, Markus
Malandrini A, Carrera P, Giacci G, Gonelli Villanova M, Palmeri S, HS.“CADASIL coma”: an underdiagnosed acute encephalo-
Vismara L, Brancolini V, Signorini E, Ferrari, Guazzi GC. Un- pathy. J Neurol Neurosurg Psychiatry 2003: 74: 249–252
usual clinical features and early brain MRI lesions in a fami- Schultz A, Santoianni R, Hewan-Lowe D. Vasculopathic
ly with cerebral autosomal dominant arteriopathy. Neurol- changes of CADASIL can be focal in skin biopsies. Ultra-
ogy 1997; 48: 1200–1203 struct Pathol 1999; 23: 241–247
Malandrini A, Albani F, Palmeri S, Fattapposta F, Gambelli S, Thomas NJ, Morris CM, Scaravilli F, Johansson J, Rossor M, de
Berti G, Bracco A, Tammaro A, Calzavara S, Villanova M, Fer- Lange R, St Clair D, Nicoll J, Blank C, Coulthard A, Bushby K,
rari M, Rossi A, Carrera P. Asymptomatic and paracrystalline Ince PG, Burn D, Kalaria RN. Hereditary vascular dementia
mitochondrial inclusions in CADASIL. Neurology 2002; 59: linked to notch 3 mutations. CADASIL in British families.
617–620 Ann N Y Acad Sci 2000; 903: 293–298
Markus HS, Martin RJ, Simpson MA, Dong YB, Ali N, Crosby AH, Tournier-Lasserve E, Joutel A, Melki J, Weissenbach J, Lathrop
Powell JF. Diagnostic strategies in CADASIL. Neurology GM, Chabriat H, Mas J-L, Cabanis E-A, Baudrimont M, Maci-
2002; 59: 1134–1138 azek J, Bach M-A, Bousser M-G. Cerebral autosomal domi-
Mayer M, Straube A, Bruening R, Uttner I, Pongratz D, Gasser T, nant arteriopathy with subcortical infarcts and leukoen-
Dichgans M, Müller-Höcker J. Muscle and skin biopsies are cephalopathy maps to chromosome 19q12. Nat Genet
sensitive diagnostic tool in the diagnosis of CADASIL. 1993; 3: 256–259
J Neurol 1999; 246: 526–532 Trojano L, Ragno M, Manca A, Caruso G. A kindred affect by
Molko N, Pappata S, Magnin JF, Poupon C, Vahedi K, Jobert A, cerebral autosomal dominant arteriopathy with subcorti-
LeBihan D, Bousser MG, Chabriat H. Diffusion tensor imag- cal infarcts and leukoencephalopathy (CADASIL). A 2-year
ing study of subcortical gray matter in CADASIL. Stroke neuropsychological follow-up. J Neurol 1998; 245: 217–222
2001; 32: 2049–2054 Tuominen S, Juvonen V, Amberla K, Jolma T, Rinne JO, Tuisku S,
Molko N, Pappata S, Mangin J-F, Poupon F, LeBihan D, Bousser Kurki T, Marttila R, Pöyhönen M, Savontaus M-L, Viitanen M,
M-G, Chabriat H. Monitoring disease progression in Kalimo H. Phenotype of a homozygous CADASIL patient in
CADASIL with diffusion magnetic resonance imaging. A comparison to 9 age-matched heterozygous patients with
study with whole brain histogram analysis. Stroke 2002; 33: the same R113C Notch3 mutation. Stroke 2001; 32: 1767–
2902–2908 1774
Okeda R,Arima K,Kawai M.Arterial changes in cerebral autoso- Utatsu Y,Takashima H,Michizono K,Kanda N,Endou K,Mitsuya-
mal dominant arteriopathy with subcortical infarcts and ma Y, Fujimoto T, Nagai M, Umehara F, Higuchi I, Arimura K,
leukoencephalopathy (CADASIL) in relation to pathogene- Nakagawa M, Osame M. Autosomal dominant early onset
sis of diffuse myelin loss of cerebral white matter. Examina- dementia and leukoencephalopathy in a Japanese family:
tion of cerebral medullary arteries by reconstruction of se- clinical, neuroimaging and genetic studies. J Neurol Sci
rial sections of an autopsy case. Stroke 2002; 33: 2565–2569 1997; 147: 55–62
O’Sullivan, M Jarosz JM, Martin RJ, Deasy N, Powell JF, Markus Van den Boom R, Lesnik Oberstein SAJ, van Duinen SG, Borne-
HS. MRI hyperintensities of the temporal lobe and external broek M, Ferrari MD, Haan J, van Buchem MA. Subcortical la-
capsule in patients with CADASIL. Neurology 2001; 56: cunar lesions: an MR imaging finding in patients with cere-
628–634 bral autosomal dominant arteriopathy with subcortical in-
O’Sullivan M, Rich PM, Barrick TH, Clark CA, Markus HS. Fre- farcts and leukoencephalopathy. Radiology 2002; 224:
quency of subclinical lacunar infarcts in ischemic leuko- 791–796
araiosis and cerebral autosomal dominant arteriopathy Van den Boom R, Lesnik Oberstein SAJ, Ferrari MD, Haan J, van
with subcortical infarcts and leukoencephalopathy. AJNR Buchem MA. Cerebral autosomal dominant arteriopathy
Am J Neuroradiol 2003; 24: 1348–1354 with subcortical infarcts and leukoencephalopathy: MR
Robinson W, Galetta SL, McCluskey L, Forman MS, Balcer LJ. findings at different ages – 3rd–6th decades. Radiology
Retinal findings in cerebral autosomal dominant arterio- 2003; 229: 683–690
pathy with subcortical infarcts and leukoencephalpathy Viitanen M, Kalimo H. CADASIL: hereditary arteriopathy lead-
(CADASIL). Surv Ophthalmol 2001; 45: 445–448 ing to multiple brain infarcts and dementia. Ann N Y Acad
Rocca MA, Filippi M, Herzog J, Sormani MP, Dichgans M,Yousry Sci 2000; 903: 273–284
TA. A magnetic resonance imaging study of the cervical Villa N, Walker L, Lindsell CE, Gasson J, Iruela-Arispe ML, Wein-
cord of patients with CADASIL. Neurology 2001; 56: 1392– master G. Vascular expression of Notch pathway receptors
1394 and ligands is restricted to arterial vessels. Mech Dev 2001;
Rubio A, Rifkin D, Powers JM, Patel U, Stewart J, Faust P, Gold- 108: 161–164
man JE, Mohr JP, Numaguchi Y, Jensen K. Phenotypic vari- Wang T, Sharma SD, Fox N, Rossor M, Brown MJ, Sharma P. De-
ability of CADASIL and novel morphologic findings. Acta scription of a simple test for CADASIL disease and determi-
Neuropathol (Berl) 1997; 94: 247–254 nation of mutation frequencies in sporadic ischaemic
Ruchoux M-M, Muarage C-A. CADASIL: cerebral autosomal stroke and dementia patients. J Neurol Neurosurg Psychia-
dominant arteriopathy with subcortical infarcts and leuko- try 2000; 69: 652–654
encephalopathy. J Neuropathol Exp Neurol 1997; 56: 947–
964
References and Further Reading 1015

Wielaard R, Bornebroek M, Ophoff RA, Winter-Warnars HAO, Bianchin MM, Capella HM, Chaves DL, Steindel M, Grisard EC,
Scheltens P, Frants RR, Ferrari MD, Haan J. A four-generation Ganev GG, Péricles da Silva J, Neto ES, Poffo MA,Walz R, Car-
Dutch family with cerebral autosomal dominant arteriopa- lotti CG, Sakamoto AC. Nasu-Hakola disaease (polycystic
thy with subcortical infarcts and leukoencephalopathy lipomembranous osteodysplasia with sclerosing leukoen-
(CADASIL), linked to chromosome 19p13. Clin Neurol Neu- cephalopathy – PLOSL): a dementia associated with bone
rosurg 1995; 97: 307–313 cystic lesions. From clinical to genetic and molecular as-
Yousry TA, Seelos K, Mayer M, Brüning R, Uttner I, Dichgans M, pects. Cell Mol Neurobiol 2004; 24: 1–24
Mammi S, Straube A, Mai N, Filippi M. Characteristic MR le- Bird TD, Koerker RM, Leaird BJ, Vlcek BW, Thorning DR.
sion pattern and correlation of T1 and T2 lesion volume Lipomembranous polycystic osteodysplasia (brain, bone,
with neurologic and neuropsychological findings in cere- and fat disease): a genetic cause of presenile dementia.
bral autosomal dominant arteriopathy with subcortical Neurology 1983; 33: 81–86
infarcts and leukoencephalopathy (CADASIL). AJNR Am J Chaabane M, Larnaout A, Sebai R, Nagi S, Touibi S, Hentati F.
Neuroradiol 1996; 20: 91–100 Nasu-Hakola disease in two Tunisian siblings: new radio-
logical findings. Neuroradiology 2000; 42: 375–378
Hakola HPA, Järvi OH, Sourander P. Osteodysplasia polycystica
hereditaria combined with sclerosing leukoencephalopa-
74 Cerebral Autosomal Recessive thy, a new entity of the dementia praesenilis group. Acta
Arteriopathy with Subcortical Infarcts Neurol Scand 1970; 46: 79–80
and Leukencephalopathy (CARASIL) Hakola HPA. Neuropsychiatric and genetic aspects of a new
hereditary disease characterized by progressive dementia
Fukutake T. Young-adult-onset hereditary subcortical vascular and lipomembranous polycystic osteodysplasia. Acta Psy-
dementia: cerebral autosomal recessive arteriosclerosis chiatr Scand 1972; 232: 1–173
with subcortical infarcts and leukoencephalopathy Hakola HPA, Livanainen M. A new hereditary disease with pro-
(CARASIL). Clin Neurol 1999: 39: 50–52 gressive dementia and polycystic osteodysplasia: neurora-
Fukutake T, Hirayama K. Familial young-adult-onset arterio- diological analysis of seven cases. Neuroradiology 1973; 6:
sclerotic leukoencephalopathy with alopecia and lumbago 162–168
without arterial hypertension. Eur Neurol 1995; 35: 69–79 Hakola HPA, Partanen VSJ. Neurophysiological findings in the
Maeda S, Nakayama H, Isaka K, Aihara Y, Nemoto S. Familial un- hereditary presenile dementia characterized by polycystic
usual encephalopathy of Binswanger’s type without hyper- lipomembranous osteodysplasia and sclerosing leukoen-
tension. Folia Psychiatr Neurol Jap 1976; 30: 165–177 cephalopathy. J Neurol Neurosurg Psychiatry 1983; 46:
Uchino M, Hirano T, Uyama E, Hashimoto Y. Cerebral autosomal 515–520
dominant arteriopathy with subcortical infarcts and Hakola HPA, Puranen M. Neuropsychiatric and brain CT find-
leukoencephalopathy (CARASIL) and CADASIL-like disor- ings in polycystic lipomembranous osteodysplasia with
ders in Japan. Ann NY Acad Sci 2002; 977: 273–278 sclerosing leukoencephalopathy. Acta Neurol Scand 1993;
Yamamura T, Nishimura M, Shirabe T, Fujita M. Subcortical vas- 88: 370–375
cular encephalophy in a normotensive, young adult with Haruta K, Matsunaga S, Ito H, Hayashi K, Yokouchi M, Onishi T,
premature baldness and spondylitis deformans. A clinoco- Komiya S. Membranous lipodystrophy (Nasu-Hakola dis-
pathological study and review of the literature. J Neurol Sci ease) presenting an unusually benign clinical course. Oncol
1987; 78: 175–188 Rep 2003; 10: 1007–1010
Yanagawa S, Ito N, Arima K, Ikeda S-I. Cerebral autosomal reces- Livanainen M, Hakola P, Erkinjuntti T, Sipponen JT, Ketonen L,
sive arteriopathy with subcortical infarcts and leuko- Sulkava R, Sepponen RE. Cerebral MR and CT imaging in
encephalopathy. Neurology 2002; 58: 817–820 polycystic lipomembranous osteodysplasia with sclerosing
Yokoi S, Nakayama H. Chronic progressive leukoencephalo- leukoencephalopathy. J Comput Assist Tomogr 1984; 8:
pathy with systemic arteriosclerosis in young adults. Clin 940–943
Neuropathol 1985; 4: 165–173 Kalimo H, Sourander P, Järvi O, Hakola P. Vascular changes and
blood-brain barrier damage in the pathogenesis of poly-
cystic lipomembranous osteodysplasia with sclerosing
leukoencephalopathy (membranous lipodystrophy). Acta
75 Polycystic Lipomembranous Osteo- Neurol Scand 1994; 89: 353–361
dysplasia with Sclerosing Leuko- Kitajima I, Suganuma T, Murata F, Nagamatsu K. Ultrastructural
encephalopathy (Nasu-Hakola Disease) demonstration of Maclura pomifera agglutinin bindings
sites in the membranocystic lesions of membranous lipo-
Amano N, Iwabuchi K, Sakai H,Yaghishita S, Itho Y, Eseki E,Yokoi dystrophy (Nasu-Hakola disease). Virchows Arch [A] 1988;
S, Arai N, Kinoshita J. Nasu-Hakola’s disease (membranous 413: 475–483
lipodystrophy). Acta Neuropathol (Berl) 1987; 74: 294–299 Kitajima I, Kuriyama M, Usuki F, Izumo S, Osame M, Suganuma
Araki T,Ohba H,Monzawa S,Sakuyama K,Hachiya J,Seki T,Taka- T, Murata F, Nagamatsu K. Nashu-Hakola disease (membra-
hashi Y, Yamaguchi M. Membranous lipodystrophy: MR nous lipodystrophy). Clinical, histopathological and bio-
imaging appearance of the brain. Radiology 1991; 180: chemical studies of three cases. J Neurol Sci 1989; 91: 35–52
793–797 Kobayashi K, Kobayashi E, Miyazu K, Muramori F, Hiramatsu S,
Aoki T, Nakamura I, Koshino Y. Hypothalamic haemorrhage
and thalamus degeneration in a case of Nasu-Hakola dis-
ease with hallucinatory symptoms and central hypother-
mia. Neuropathol Appl Neurobiol 2000; 26: 98–101
1016 References and Further Reading

Malandrini A, Scarpini C, Palmeri S, Villanova M, Parrotta E, Ueki Y, Kohara N, Oga T, Fukuyama H, Akiguchi I, Kimura J,
Tripodi S, Giani S, DeFalco D, Guazzi GC. Palatal myoclonus Shibasaki H. Membranous lipodystrophy presenting with
and unusual MRI findings in a patient with membranous palilalia: a PET study of cerebral glucose metabolism. Acta
lipodystrophy. Brain Dev 1996; 18: 59–63 Neurol Scand 2000; 102: 60–64
Matsushita M, Oyanagi S, Hanawa S, Shiraki H, Kosaka K. Nasu- Verloes A, Maquet P, Sadzot B, Vivario M, Thiry A, Franck G. Na-
Hakola’s disease (membranous lipodystrophy). A case re- su-Hakola syndrome: polycystic lipomembranous os-
port. Acta Neuropathol (Berl) 1981; 54: 89–93 teodysplasia with sclerosing leukoencephalopathy and
Mii Y, Miyauchi Y, Yoshikawa T, Honoki K, Aoki M, Tsutsumi M, presenile dementia. J Med Genet 1997; 34: 753–757
Maruyama H, Funauchi M, Konishi Y,Tamai S. Ultrastructural Yagishita S, Ito Y, Sakai H, Amano N. Membranocystic lesions of
lipid and glycoconjugate cytochemistry of membranous the lung in Nasu-Hakola disease. Virchows Arch [A] 1985;
lipodystrophy (Nasu-Hakola disease). Virchows Archiv [A] 408: 211–217
1991; 419: 137-142
Minagawa M, Maeshiro H, Kato K, Shioda K. A rare case of leu-
codystrophy-neuroaxonal leucodystrophy (Seitelberger).
Psychiatry Neurol Jpn 1980; 82: 488–503
76 Pigmentary Orthochromatic
Minagawa M, Maeshiro H, Shioda K, Hirano A. membranous Leukodystrophy
lipodystrophy (Nasu disease): clinical and neuropathologi-
cal study of a case. Clin Neuropathol 1985; 4: 38–45 Belec L, Gray F, Louarn F, Gherardi R, Morelot D, Destée A, Poiri-
Miyazu K, Kobayashi K, Fukutani Y, Nakamura I, Hasegawa H,Ya- er J, Castaigne P. Leucodystrophie orthochromatique pig-
maguchi N, Saitoh T. Membranous lipodystrophy (Nasu- mentaire. Rev Neurol 1988; 144: 347–357
Hakola disease) with thalamic degeneration: report of an Constantinidis J,Wisniewski TM.The dominant form of the pig-
autopsied case. Acta Neuropathol (Berl) 1991; 82: 414–419 mentairy orthochromatic leukodystrophy. Acta Neu-
Motohasi N, Shinohara M, Shioe K, Fukuzawa H, Akiyama Y, ropathol (Berl) 1991; 82: 483–487
Kariya T. A case of membranous lipodystrophy (Nasu-Hako- Dousset V,Tison F,Vital A, Henry P. Leucodystrophie orthochro-
la disease) with unique MRI findings. Neuroradiology 1995; matique d’évolution rapide chez l’adulte. Rev Neurol 1998;
37: 549–550 154: 415–418
Nasu T, Tsukahara Y, Terayama K. A lipid metabolic disease – Gray F,Destee A,Bourre J-M,Gherardi R,Krivosic I,Warot P,Poiri-
“membranous lipodystrophy” – an autopsied case demon- er J. Pigmentary type of orhtochromatic leukodystrophy
strating numerous peculiar membrane-structures com- (OLD); a new case with ultrastructural and biochemical
posed of compound lipid in bone and bone marrow and study. J Neuropathol Exp Neurol 1987; 46: 585–596
various adipose tissues. Acta Pathol Jpn 1973; 23: 539–558 Knopman D,Sung JH,Davis D.Progressive familial leukodystro-
Oishi M, Mori N, Takasu T, Osaka S, Yamamoto M, Uchiyama T, phy of late onset. Neurology 1996; 46: 429–434
Sawada S.Nasu-Hakola disease.A case accompanied by ab- Marotti JD, Tobias S, Fratkin JD, Powers JM, Rhodes CH. Adult
normalities in fatty acid composition of serum total lipids onset leukodystrophy with neuroaxonal spheroids and
and amino acid analysis. Acta Neurol (Napoli) 1993; 15: pigmented glia: report of a family, historical perspective,
53–61 and review of the literature. Acta Neuropathol (Berl) 2004;
Paloneva J, Kestilä M,Wu J, Salminen A, Böhling T, Ruotsalainen 107: 481–488
V, Hakola P, Bakker ABH, Phillips JH, Pekkarinen, Lanier LL,Ti- Okeda R,Matsuo T,Kawahara Y,Eishi Y,Tamai Y,Tanaka M,Kama-
monen T, Peltonen L. Loss-of-function mutations in TYROBP ki M, Tsubota N, Yamadera H. Adult pigment type (Pfeiffer)
(DAP12) result in a presenile dementia with bone cysts. Nat of sudanophilic leukodystrohy. Pathological and morpho-
Genet 2000; 25: 357–361 metrical studies on two autopsied cases of siblings. Acta
Paloneva J, Autti T, Raininko R, Partanen J, Salonen O, Puranen Neuropathol (Berl) 1989; 78: 433–542
M, Hakola P, Haltia M. CNS manifestations of Nasu-Hakola Pietrini V, Tagliavini F, Pilleri G, Trabattoni CR, Lechi A. Or-
disease. A frontal dementia with bone cysts. Neurology thochromatic leukodystrophy with pigmented glial cells.
2001; 56: 1552–1558 An adult case with clinical-anatomical study. Acta Neurol
Paloneva J, Manninen T, Christman G, Hovanes K, Mandelin J, Scand 1979; 59: 140–147
Adolfsson R, Bianchin M, Bird T, Miranda R, Salmaggi A, Seiser A,Jellinger K,Brainin.Pigmentary type of orthochromat-
Tranebjærg Y, Konttinen Y, Peltonen L. Mutations in two ic leukodystrophy with early onset and protracted course.
genes encoding different subunits of a receptor signaling Neuropediatrics 1990; 21: 48–52
complex results in an identical disease phenotype. Am J Shannon P, Wherrett JR, Nag S. A rare form of adult onset
Hum Genet 2002; 71: 656–662 leukodystrophy: orthochromatic leukodystrophy with pig-
Pekkarinen P, Hovatta I, Hakola P, Järvi O, Kestilä M, Lenkkeri U, mented glia. Can J Neurol Sci 1997; 24: 146–150
Adolfsson R, Holmgren G, Nylander P-O, Tranebjærg L, Ter- Verghese J, Weidenheim K, Malik S, Rapin I. Adult onset pig-
williger JD, Lönnqvist J, Peltonen L. Assignment of the locus mentary orthochromatic leukodystrophy with ovarian dys-
for PLO-SL, a frontal lobe dementia with bone cysts, to genesis. Eur J Neurol 2002; 9: 663–670
19q13. Am J Hum Genet 1998; 62: 362–372
Snow JL, Su WPD, Gibson LE. Lipomembranous (membra-
nocystic) changes associated with morphea: a clinico-
77 Adult-onset Autosomal Dominant
pathologic review of three cases. J Am Acad Dermatol
1994; 31: 246–250 Leukoencephalopathies
Tanaka J. Leukoencephalopathic alteration in membranous
lipodystrophy. Acta Neuropathol (Berl) 1980; 50: 193–197 Abe K, Ikeda M,Watase K,Tanabe H, Fujimura H,Yorifuji S, Ueno
Tanaka J. Nasu-Hakola disease: a review of its leukoencephalo- S, Mezaki T, Mori T. A kindred of hereditary adult-onset
pathic and membranolipodystrophic features. Neu- leukodystrophy with sparing of the optic radiations.Neuro-
ropathology 2000; 20: S25-S29 radiology 1993; 35: 281–283
References and Further Reading 1017

Bergui M, Bradac GB, Leombruni S, Vaula G, Quattrocolo G. MRI Delves PJ, Roitt IM. The immune system. Part 1. N Engl J Med
and CT in autosomal-dominant adult-onset leukodystro- 2000; 343: 37–49
phy. Neuroradiology 1997; 39: 423–426 Delves PJ, Roitt IM. The immune system. Part 2. N Engl J Med
Calandriello L, Matteucci C, Bertini E, Medolago Albani L, An- 2000; 343: 108–117
tonelli A, Manfredi M, Pelladini G. Biopsy diagnosis of a case Dopp JM, de Velllis J. Strategies for the therapeutic manipula-
of adult onset orthochromatic leukodystrophy. Clinical and tion of cytokines and their receptors in inflammatory neu-
brain biopsy findings. Ital J Neurol Sci 1992; 13: 787–792 rodegenerative diseases. Ment Retard Dev Disabil Res Rev
Coffeen CM,McKenna C,Koeppen AH,Plaster NM,Maragakis N, 1998; 4: 200–211
Mihalopoulos J, Schwankhaus JD, Flanigan KM, Gregg RG, Ghirnikar RS, Lee YL, Eng LF. Inflammation in traumatic brain
Ptácek LJ, Fu Y-H. Genetic localization of an autosomal injury: role of cytokines and chemokines. Neurochem Res
dominant leukodystrophy mimicking chronic progressive 1998; 23: 329–340
multiple sclerosis to chromosome 5q31. Hum Mol Genet Gonzalez-Scarano F, Tyler KC. Molecular pathogenesis of neu-
2000; 9: 787–793 rotropic viral infections. Ann Neurol 1987; 22: 565–574
Eldridge R, Anayiotos CP, Schlesinger S, Cowen D, Bever C, Pa- Gonzalez-Scavano F, Tyler KC. Molecular pathogenesis of neu-
tronas N, McFarland H. Hereditary adult-onset leukodystro- rotropic viral infections. Ann Neurol 1987; 22: 565–574
phy simulating chronic progressive multiple sclerosis.N En- Hartung H-P, Archelos JJ, Zielasek J, Gold R, Koltzenburg M,
gl J Med 1984; 311: 948–953 Reiners K-H, Toyka KV. Circulating adhesion molecules and
Fukazawa T, Sasaki H, Kikuchi S, Hamada K, Hamada T,Tashiro K. inflammatory mediators in demyelination: a review. Neu-
Dominantly inherited leukodystrophy showing cerebellar rology 1995; 45: S22-S32
deficits and spastic paraparesis: a new entity? J Neurol Hartung HP, Jung S, Stoll G, Zielasek J, Schmidt B, Archelos JJ,
1997; 244: 446–449 Toyka KV. Inflammatory mediators in demyelinating disor-
Letournel F, Etcharry-Bouyx F, Verny C, Barthelaix A, Dubas F. ders of the CNS and PNS. J Neuroimmunol 1992; 40: 197–
Two clinicopathological cases of a dominantly inherited, 210
adult onset orthochromatic leucodystrophy. J Neurol Neu- Henson PM, Murphy RC, eds. Mediators of the inflammatory
rosurg Psychiatry 2003; 74: 671–673 process. Handbook of inflammation, vol 6, 3rd ed. Amster-
Quattrocolo G, Leombruni S, Vaula G, Bergui M, Riva A, Bradac dam: Elsevier, 1989
GB, Bergamini L. Autosomal dominant late-onset leuko- Johnson RT. Selective vulnerability of neural cells to viral infec-
encephalopathy. Eur Neurol 1997; 37: 53–61 tions. Brain 1980; 103: 447–472
Schwankhaus JD, Patronas N, Dorwart R, Eldridge R, Johnson RT. The virology of demyelinating diseases. Ann Neu-
Schlesinger S, McFarland H. Computed tomography and rol 1994; 36: S54-S60
magnetic resonance imaging in adult-onset leukodystro- Johnson RT, Mims CA. Pathogenesis of viral infections of the
phy. Arch Neurol 1988; 45: 1004–1008 nervous system. N Engl J Med 1968; 278: 23–30
Schwankhaus JD, Katz DA, Eldridge R, Schlesinger S, McFarland Kieseier BC, Storch MK, Archelos JJ, Martino G, Hartung H-P. Ef-
H. Clinical and pathological features of an autosomal dom- fector pathways in immune mediated central nervous sys-
inant, adult-onset leukodystrophy simulating chronic pro- tem demyelination. Curr Opin Neurol 1999; 12: 323–336
gressive multiple sclerosis. Arch Neurol 1994; 51: 757–766 Klein J, Sato A.The HLA system. First of two parts. N Engl J Med
Tagawa A, Ono S, Inoue K, Hosoi N, Kaneda K, Suzuki M, Nagao 2000; 343: 702–709
K, Shimizu N. A new familial adult-onset leukodystrophy Klein J, Sato A. The HLA system. Second of two parts. N Engl J
manifesting as cerebellar ataxia and dementia. J Neurol Sci Med 2000; 343: 782–786
2001; 183: 47–55 Kuroda Y,Shimamoto Y.Human tumor necrosis factor-alpha ar-
guments experimental allergic encephalomyelitis in rats. J
Neuroimmunol 1991; 34: 159–165
Ledeen RW, Chakraborty G. Cytokines, signal transduction, and
78 Inflammatory and Infectious Disorders inflammatory demyelination: review and hypothesis. Neu-
rochem Res 1998; 23: 277–289
Albert LJ, Inman RD. Molecular mimicry and autoimmunity. Lewis RA, Austen KF, Soberman RJ. Leukotrienes and other
N Engl J Med 1999; 341: 2068–2074 products of the 5-lipoxygenase pathway. N Engl J Med
Beckman JS. The double edged role of nitric oxide in brain 1990; 323: 645–655
function and super-oxide mediated injury. J Dev Physiol Linington C, Morgan BP, Scolding NJ, Wilkins P, Pidleston S,
1991; 15: 53–59 Compston DAS.The role of complement in the pathogene-
Bradl M, Flügel A. The role of T cells in brain pathology. Prot sis of experimental allergic encephalomyelitis. Brain 1989;
Pathol Immune Respons CNS 2002; 265: 141–162 112: 895–911
Coe-Clough N, Roth JA. Understanding immunology. London, Luster AD. Chemokines – chemotactic cytokines that mediate
Mosby, 1997 inflammation. N Engl J Med 1998; 388: 436–445
Cuzner ML, Loughlin AJ, Mosley K, Woodroofe MN. The role of MacMicking JD, Willenborg DO, Weidemann MJ, Rockett KA,
microglia macrophages in the processes of inflammatory Cowden WB. Elevated secretion of nitrogen and oxygen in-
demyelination and remyelination. Neuropathol Appl Neu- termediates by inflammatory leukocytes in hyperacute ex-
robiol 1994; 20: 200–201 perimental autoimmune encephalomyelitis: enhancement
Dak Canto M, Rabinowitz SG. Experimental models of virus-in- by the soluble products of encephalitogenic T cells. J Exp
duced demyelination of the central nervous system. Ann Med 1992; 176: 303–307
Neurol 1982; 11: 109–127 Maimone D, Gregory S, Arnason BGW, Reder AT. Cytokine levels
De Jong R, Brouwer M, Kuiper HM, Hooibrink B, Miedema F, van in the cerebrospinal fluid and serum of patients with multi-
Lier RAW. Maturation and differentiation dependent re- ple sclerosis. J Neuroimmunol 1991; 32: 67–74
sponsiveness of human CD4+ T helper subits. J Immunol Matyszak MK. Inflammation in the CNS: balance between im-
1992; 149: 2795–2801 munological privilege and immune response. Prog Neuro-
biol 1998; 56: 19–35
1018 References and Further Reading

Mayatepek E, Hoffmann GF. Leukotrienes: biosynthesis, metab- Barkhof F, Filippi M, van Waesberghe JHTM, Campi A, Miller DH,
olism, and pathophysiologic significance. Pediatr Res 1995; Adèr HJ. Interobserver agreement for diagnostic MRI crite-
37: 1–9 ria in suspected multiple sclerosis. Neurology 1999; 41:
Merrill JE, Gerner RH, Myers LW, Ellison GW. Regulation of nat- 347–350
ural killer cell cytotoxicity by prostaglandin E in the periph- Barkhof F, Karas GB, van Walderveen MAA. T1 Hypointensities
eral blood and cerebrospinal fluid of patients with multiple and axonal loss.Neuroimaging Clin N Am 2000;10:739–752
sclerosis and other neurological diseases. J Neuroimmunol Barkhof F, Rocca M, Francis G, van Waesberghe J-HTM, Uitde-
1983; 4: 223–237 haag BMJ, Hommes OR, Hartung H-P, Durelli L, Edan G, Fer-
Merrill JE, Strom SR, Ellison GW, Myers LW. In vitro study of nández O, Seeldrayer P, Sørensen P, Margrie S, Rovaris M, Co-
mediators of inflammation in multiple sclerosis. J Clin Im- mi G, Filippi M. Validation of diagnostic magnetic reso-
munol 1989; 9: 84–96 nance imaging criteria for multiple sclerosis and response
Mims CA, Nash A, Stephen J. MIMS pathogenesis of infectious to interferon b1a. Ann Neurol 2003; 53: 718–724
disease, 5th ed. London: Academic Press, 2000 Barkhof F, van Waesberghe J-HTM, Filippi M,Yousry T, Miller DH,
Powrie F, Coffman RC. Cytokine regulation of T-cell function: Hahn D, Thompson AJ, Kappos L, Brex P, Pozzilli C, Polman
potential for therapeutic intervention. Immunol Today CH.T1 Hypointense lesions in secondary progressive multi-
1993; 14: 270–276 ple sclerosis: effect of interferon beta-1b treatment. Brain
Starzl TE, Zinkernagel RM. Antigen localization and migration 2001; 124: 1396–1402
in immunity and tolerance. N Engl J Med 1998; 339: Barkhof F, van Walderveen M. Characterization of tissue dam-
1905–1913 age in multiple sclerosis by nuclear magnetic resonance.
Von Andrian UH, Mackay CR. T-Cell function and migration. Phil Trans R Soc Lond B 1999; 354: 1675–1686
Two sides of the same coin. N Engl J Med 2000; 14: Barkhof F. Imaging of remyelination. Mult Scler 1997; 3:
1020–1034 129–132
Walport MJ.Complement.First of two parts.N Engl J Med 2001; Barkhof F.The clinico-radiological paradox in multiple sclerosis
344: 1058–1066 revisited. Curr Opin Neurol 2002; 15: 239–245
Walport MJ. Complement. Second of two parts. N Engl J Med Barnes G, Benjamin S, Bowen JD,Cutter N, de Lateur BJ, Dietrich
2001; 344: 1140–1144 WD, Dowling MM, Griffin JW, Hummers L, Irani D, Jorens J,
Weiner LP, Johnson RT, Herndon RM. Viral infections and de- Kaplin AI, Kratz JD. Kerr DA, Krishnan C, Lecy CE, Lucchinetti
myelinating diseases. N Engl J Med 1973; 288: 1103–1110 C, Lynn DJ, Manler RN, McArthur JC, McDonald JW III, Morri-
Whitley RJ. Viral encephalitis. N Engl J Med 1990; 323: 242–250 son L, Pardo-Villamizar C, Pidcock FS, Ransohoff R, Roos KL,
Wright SA, Unkeless JC. Innate immunity. Fatal attraction: Trovato MK, Vollmer TL, Wegener ST, Weinschenker BG,
recognition and killing mechanisms of innate immunity. Wingerchuck DM. Proposed diagnostic criteria and noso-
Curr Opin Immunol 1993; 5: 57–61 logy of acute transverse myelinitis. Neurology 2002; 59:
499–505
Beer S, Rösler KM, Hess CW. Diagnostic value of paraclinical
tests in multiple sclerosis: relative sensitivities and specifici-
79 Multiple Scerosis ties for reclassification according to the Poser committee
criteria. J Neurol Neurosurg Psychiatry 1995; 59: 152–159
Achiron A, Gabbay U, Gilad R, Hassin-Baer S, Barak Y, Gornish M, Belássy C, Bernert G,Wöber-Bingöl C, Csapó B, Kornek B, Széles
Elizur A, Goldhammer Y, Sarova-Pinhas I. Intravenous im- J, Fleischmann D, Prayer D. Long-term MRI observations of
munoglobulin treatment in multiple sclerosis. Effect on childhood-onset relapsing-remitting multiple sclerosis.
relapses. Neurology 1998; 50: 398–402 Neuropediatrics 2001; 32: 28–37
Álvarez-Lafuente R, Martín-Estefanía C, de las Heras V, Castrillo Berger T, Rubner P, Schautzer F, Egg R, Ulmer H, Mayringer I,
C, Picazo JJ, Varela de Seijas E, González RA. Active human Dilitz E, Deisenhammer F, Reindl M. Antimyelin antibodies
herpesvirus 6 infection in patients with multiple sclerosis. as a predictor of clinically definite multiple sclerosis after a
Arch Neurol 2002; 59: 929–933 first demyelinating event. N Engl J Med 2003; 349: 139–145
Arnold DL,Wolinsky JS, Matthews PM, Falini A.The use of mag- Bergers E, Bot JCJ, van der Valk P, Castelijns JA, Lycklama A, Ni-
netic resonance spectroscopy in the evaluation of the nat- jeholt GJ, Kamphorst W, Polman CH, Blezer EIA, Nicolay K,
ural history of multiple sclerosis.J Neurol Neurosurg Psychi- Ravid R, Barkhof F. Diffuse signal abnormalities in the spinal
atry 1998; 64: S94-S101 cord in multiple sclerosis: direct postmortem in situ mag-
Barcellos LF, Klitz W, Field LL, Tobias R, Bowcock AM, Wilson R, netic resonance imaging correlated with in vitro high reso-
Nelson MP, Nagotomi J, Thomson G. Association mapping lution magnetic resonance imaging and histopathology.
of disease loci using a pooled DNA genomic screen. Am J Ann Neurol 2002; 41: 652–656
Hum Genet 1997; 61: 734–747 Bitsch A, Bruhn H, Vougioukas V, Stringaris A, Lassmann H,
Barkhof F, Brück W, de Groot CJA, Bergers E, Hulshof S, Geurts J, Frahm J, Brück W. Inflammatory CNS demyelination:
Polman CH, van der Valk P. Remyelinated lesions in multiple histopathologic correlation with in vivo quantitative pro-
sclerosis. Magnetic resonance image appearance. Arch ton MR spectroscopy. AJNR Am J Neuroradiol 1999; 20:
Neurol 2003; 60: 1073–1081 1619–1627
Barkhof F, Brück W, de Groot JA, Bergers E, Hulshof S, Geurts J, Bjartmar C, Kidd G, Mörk S, Ruddick R, Trapp BD. Neurological
Olman CH, van der Valk P. Remyelinated lesions in multiple disability correlates with spinal cord axonal loss and re-
sclerosis. Arch Neurol 2003; 60: 1073–1081 duced N-acetyl aspartate in chronic multiple sclerosis pa-
Barkhof F, Filippi M, Miller DH, Scheltens P, Campi A, Polman CH, tients. Ann Neurol 2000; 48: 893–901
Comi G, Adèr HJ, Losseff N, Valk J. Comparison of MRI crite- Bjartmar C, Kinkel RP, Kidd G, Rudick RA, Trapp BD. Axonal loss
ria at first presentation to predict conversion to clinically in normal-appearing white matter in a patient with acute
definite multiple sclerosis. Brain 1997; 120: 2059–2069 MS. Neurology 2001; 57: 1248–1252
References and Further Reading 1019

Bjartmar C, Trapp, BD. Axonal and neuronal degeneration in Confavreax C, Vukusic S, Moreau T, Adelaine P. Relapses and
multiple sclerosis: mechanisms and functional conse- progression of disability in multiple sclerosis. N Engl J Med
quences. Curr Opin Neurol 2001; 14: 271–278 2000; 343: 1430–1438
Bjartmar C,Wujek JR,Trapp BD. Axonal loss in the pathology of Cossins JA, Clements JM, Ford J, Miller KM, Pigott R, Vos W, van
MS: consequences for understanding the progressive der Valk P, de Groot CJA. Enhanced expression of MMP-7
phase of the disease. J Neurol Sci 2003; 206:165–171 and MMP-9 in demyelinating multiple sclerosis lesions. Ac-
Bö L,Vedeler CA,Nyland HI,Trapp BD,Mørk SJ.Subpial demyeli- ta Neuropathol (Berl) 1997; 94: 590–598
nation in the cerebral cortex of multiple sclerosis patients. Dalton CM, Brax PA, Miszkiel KA, Hickman SJ, MacManus DG,
J Neuropathol Exp Neurol 2003; 62: 723–732 Plant GT, Thompson AJ, Miller DH. Application of the new
Bö L, Geurts JJ, Ravid R, Barkhof F.Magnetic resonance imaging McDonald criteria to patients with clinically isolated syn-
as a tool to examine the neuropathology of multiple sclero- dromes suggestive of multiple sclerosis. Ann Neurol 2002;
sis. Neuropathol Appl Neurobiol 2004; 30: 106–117 52: 47–53
Boonetti B, Raine CS. Multiple sclerosis: oligodendrocytes dis- Dalton CM, Brex PA, Mitzkiel KA, Fernando K, MacManus DG,
play cell death-related molecules in situ but do not under- Plant GT,Thompson AJ, Miller DH. New T2 lesions enable an
go apoptosis. Ann Neurol 1997; 42: 74–84 earlier diagnosis of multiple sclerosis in clinically isolated
Bot JC, Barkhof F, Lycklama à Nijeholt G, van Schaardenburg D, syndromes. Ann Neurol 2003; 53: 673–676
Voskuyl AE, Ader HJ, Pijnenburg JA, Polman CH, Uitdehaag Degaonkar NM, Jayasundar R, Jagannathan NR. Sequential dif-
BM, Vermeulen EG, Castelijns JA. Differentiation of multiple fusion-weighted magnetic resonance study of lysophos-
sclerosis from other inflammatory disorders and cere- phatidyl choline-induced experimental demyelinating le-
brovascular disease: value of spinal MR imaging. Radiology sion: an animal model of multiple sclerosis. J Magn Reson
2002; 223: 46–56 Imaging 2002; 16: 153–159
Bot JC, Barkhof F, Lycklama a Nijeholt GJ, Bergers E, Polman CH, De Groot CJA, Bergers E, Kamphorst W, Ravid R, Polman CH,
Ader HJ, Castelijns JA. Comparison of a conventional car- Barkhof F, van der Valk P. Post-mortem MRI-guided sam-
diac-triggered dual spin-echo and a fast STIR sequence in pling of multiple sclerosis brain lesions. Increased yield of
detection of spinal cord lesions in multiple sclerosis.Eur Ra- active demyelinating and (p)reactive lesions. Brain 2001:
diol 2000; 10: 753–758 124: 1635–1645
Bot JCJ,Barkhof F,Lycklama à Nijeholt G,van Schaardenburg D, De Seze J, Stojkovic T, Ferriby D, Gauvrit JY, Montagne C,
Voskuyl AE, Ader HJ, Pijnenburg JAL, Polman CH, Uitdehaag Mounier-Vehier F, Verier A, Pruvo JP, Hache JC, Vermersch P.
BMJ, Vermeulen EGJ, Castelijns JA. Differentiation of multi- Devic’s neuromyelitis optica: clinically, laboratory, MRI and
ple sclerosis from other inflammatory disorders and cere- outcome profile. J Neurol Sci 2002; 197: 57–61
brovascular disease: value of spinal MR imaging. Radiology De Stefano N, Narayanan S, Francis GS, Arnaoutelis R, Tartaglai
2002; 223: 46–56 MC, Antel JP, Matthews PM, Arnold DL. Evidence of axonal
Brex PA, Miszkiel KA, O’Riordan JI, Plant GT, Moseley IF, Thomp- damage in the early stages of multiple sclerosis and its
son AJ, Miller DH. Assessing the risk of early multiple sclero- relevance to disability. Arch Neurol 2001; 58: 65–70
sis in patients with clinically isolated syndromes: the role of Dhib-Jablut S. Glatiramer acetate (Copaxone) therapy for mul-
a follow-up MRI. J Neurol Neurosurg Psychiatry 2001; 70: tiple sclerosis. Pharmacol Ther 2003; 98: 245–255
390–393 Dyment DA, Cader MZ, Willer CJ, Risch N, Sadovnick AD, Ebers
Buljevac D, Flach HZ, Hop WCJ, Hijdra D, Laman JD, Savekoul GC. A multigenerational family with multiple sclerosis.
HFJ, van der Meché FGA, van Doorn PA, Hintzen RQ. Brain 2002; 125: 1474–1482
Prospective study on the relationship between infections Ebers GC. Treatment of multiple sclerosis. Lancet 1994; 343:
and multiple sclerosis exacerbations. Brain 2002; 125: 275–279
952–960 Epplen C, Jäckel S, Santos EJM, D’Souza M, Poehlau D, Dortz-
Caramia F, Pantano P, di Legge S, Piattella MC, Lenzi D, Paolillo auer B, Sindern E, Haupts M, Rüde K-P, Weber F, Stöver J,
A, Nucciarelli W, Lenzi GL, Bozzao L, Pozzilli C. A longitudinal Poser S, Gehler W, Malin J-P, Przuntek H, Epplen JT. Genetic
study of MR diffusion changes in normal appearing white predisposition to multiple sclerosis as revealed by im-
matter of patients with early multiple sclerosis. Magn Re- munoprinting. Ann Neurol 1997 41: 341–352
son Imaging 2002; 20: 383–388 Filippi M. In-vivo tissue characterization of multiple sclerosis
Carcciolo JT, Murtagh RD, Rojiani AM, Murtagh FR. Pathogno- and other white matter diseases using magnetic resonance
monic MR imaging findings in Balo concentric sclerosis. based techniques. J Neurol 2001; 248: 1019–1029
AJNR Am J Neuroradiol 2001; 22: 292–293 Filippi M, Rocca MA. Disturbed functional and plasticity in mul-
Chard DT, Griffin CM, McLean MA, Kapeller P, Kapoor R,Thomp- tiple sclerosis as gleaned from functional magnetic reso-
son AJ, Miller DH. Brain metabolite changes in cortical gray nance imaging. Curr Opin Neurol 2003; 16: 275–282
and normal-appearing white matter in clinically early re- Filippi M, Dousset V, McFarland HF, Miller DH, Grossman RI. Role
lapsing-remitting multiple sclerosis. Brain 2002; 125: of magnetic resonance imaging in the diagnosis and mon-
2342–2352 itoring of multiple sclerosis: consensus report of the white
Ciccarelli O, Giugni E, Paolilo A, Mainero C, Gasperini C, Bas- matter study group. J Magn Reson Imaging 2002; 15: 499–
tianello S, Pozzeli. Magnetic resonance outcome of new en- 504
hancing lesions in patients with relapsing-remitting multi- Filippi M, Bozzali M, Rovaris M, Gonen O, Kesavadas C, Ghezzi A,
ple sclerosis. Eur J Neurol 1999; 6: 455–459 Martinelli V, Grossman RI, Scotti G, Comi G, Falini A.Evidence
Comi G, Filippi M, Wolinsky JS. European/Canadian multicen- for widespread axonal damage at the earliest clinical stage
ter, double-blind, randomized, placebo-controlled study of of multiple sclerosis. Brain 2003; 126: 433–437
the effects of glatiramer acetate on magnetic resonance Finsen B, Antel J, Owens T. TNFa: kill or cure for demyelinating
imaging – measured disease activity and burden in pa- disease? Mol Psychiatry 2002; 7: 820–821
tients with relapsing multiple sclerosis. Ann Neurol 2001;
49: 290–297
1020 References and Further Reading

Fisher E, Rudick RA, Simon JH, Cutter G, Baier M, Lee J-C, Miller Ibrahim SM, Mix E, Böttcher T, Koczan D, Gold R, Rolfs A,Thiesen
D, Weinstock-Guttman B, Mass MK, Dougherty DS, Simon- H-J. Gene expression profiling of the nervous system in
ian NA. Eight-year follow-up study of brain atrophy in pa- murine experimental autoimmune encephalitis. Brain
tients with MS. Neurology 2002; 59: 1412–1420 2001; 124: 1927–1938
Frederiksen JL. Bilateral acute optic neuritis: prospective clini- Jacobs JD, Cookfair DL, Rudick RA, Herndon RM, Richert JR,
cal, MRI, CSF, neurophysiological and HLA findings. Neu- Salazar AM, Fischer JS, Goodkin DE, Granger CV, Simon JH,
roophthalmology 1997; 17: 175–183 Alam JJ, Bartozak DM, Bourdette DN, Brainam J, Brown-
Gasperini C, Pozzilli C, Bastianello S, Koudriavssteva T, Galgani scheidle CM, Coats ME, Cohan SL, Dougherty DS, Kinkel RP,
S, Millefiorini E, Paolillo A, Horsfield MA, Bozzao L, Fieschi C. Mass MK, Munschauer FE, Priore RL, Pullicino PM Scherok-
Effect of steroids on Gd-enhancing lesions before and dur- man BJ, Weinstock-Guttman B, Whitham RH. Intramuscular
ing recombinant beta interferon 1a treatment in relapsing interferon beta-1a for disease progression in relapsing
remitting multiple sclerosis. Neurology 1998; 50: 403–406 multiple sclerosis. Ann Neurol 1996; 39: 285–294
Ghorpade A, Holter S, Borgmann K, Persidsky R,Wu L.HIV-1 and Jacobsen M, Schweer D, Ziegler A, Gaber R, Schock S, Schwinz-
IL-1b regulate Fas ligand expression in human astrocytes er R, Wonigeit K, Lindert R-B, Kantarci O, Schaefer-Klein J,
through the NF-B pathway. J Neuroimmunol 2003; 141: Schipper HI, Oertel WH, Heidenreich F, Weinschenker BG,
141–149 Sommer N, Hemmer B. A point mutation in PTPRC is associ-
Giovannoni G, Lai M, Thorpe J, Kidd D, Chamoun V, Thompson ated with the development of multiple sclerosis. Nat Genet
AJ, Miller DH, Feldmann M, Thompson EJ. Longitudinal 2000; 26: 495–499
study of soluble adhesion molecules in multiple sclerosis: Johnson KP, Brooks BR, Cohen JA, Ford CC, Goldstein J, Lisak RP,
correlation with gadolinium enhanced magnetic reso- Myers LW, Panitch HS, Rose JW, Schiffer RB, Vollmer T, Wein-
nance imaging. Neurology 1997; 48: 1557–1565 er LP,Wolinsky JS.Copolymer 1 reduces relapse rate and im-
Glasier CM, Robbins MB, Davis MB, Davis PC, Ceballos E, Bates proves disability in relapsing-remitting multiple sclerosis:
SR. Clinical, neurodiagnostic, and MR findings in children results of a phase III multicenter, double-blind, placebo-
with spinal and brain stem multiple sclerosis. AJNR Am J controlled trial. Neurology 1995; 45: 1268–1276
Neuroradiol 1995; 16: 87–95 Johnson KP, Brooks BR, Ford CC, Goodman A, Guarnaccia J,
Gold R. Linington C. Devic’s disease: bridging the gap between Lisak RP, Myers LW, Panitch HS, Pruitt A, Rose JW, Kachuck N,
laboratory and clinic. Brian 2002; 125: 1425–1427 Wolinsky JS. Sustained clinical benefits of glatiramer ac-
Gonen O, Moriarty DM, Li BSY, Babb JS, He J, Listerud J, Jacobs etate in relapsing multiple sclerosis patients observed for 6
D,Markowitz CE,Grossman RI.Relapsing-remitting multiple years. Mult Scler 2000; 6: 255–266
sclerosis and whole-brain N-acetylaspartate measurement: Kalkers NF, Bergers E, Castelijns JA, van Walderveen MAA, Bot
evidence for different clinical cohorts – initial observations. JCJ, Adèr HJ, Polman CH, Barkhof F. Optimizing the associa-
Radiology 2002; 255: 261–268 tion between disability and biological markers in MS. Neu-
Goodin DS, Arnason BG, Coyle PK, Frohman EM, Paty DW. The rology 2001; 57: 1253–1528
use of mitoxantrone (Novantrone) for the treatment of Kapeller P, McLean MA, Griffin CM, Chard D, Parker GJM, Barker
multiple sclerosis. Neurology 2003; 61: 1332–1338 GJ, Thompson AJ, Miller DH. Preliminary evidence for neu-
Goodin DS, Arnason BG, Coyle PK, Frohman EM, Paty DW; Ther- ronal damage in cortical grey matter and normal appear-
apeutics and Technology Assessment Subcommittee of ing white matter in short duration relapsing-remitting mul-
the American Academy of Neurology. The use of mitox- tiple sclerosis: a quantitative MR spectroscopic imaging
antrone (Novantrone) for the treatment of multiple sclero- study. J Neurol 2001; 248: 131–138
sis: report of the Therapeutics and Technology Assessment Kappos L, Moeri D, Radue EW, Schoetzau A, Schweikert K, Bark-
Subcommittee of the American Academy of Neurology. hof F,Miller D,Guttman CR,Weiner HL,Gasperini C,Filippi M.
Neurology 2003; 61: 1332–1338 Predictive value of gadolinium-enhanced magnetic reso-
Halfpenny C, Benn T, Scolding N. Cell transplantation, myelin nance imaging for relapse rate and changes in disability or
repair, and multiple sclerosis. Lancet [Neurol] 2002; 1: 31–40 impairment in multiple sclerosis: a meta-analysis. Gadolini-
Hartung H-P.Pathogenesis of inflammatory demyelination:im- um MRI Meta-analysis Group. Lancet 1999; 353: 964–969
plications for therapy. Curr Opin Neurol 1995; 8: 191–199 Karaarslan E, Altintas A, Senol U, Yeni N, Dincer A, Bayindir C,
He J, Grossman RI, Ge Y, Mannon LJ. Enhancing patterns in mul- Karaagac N, Siva A. Baló’s concentric sclerosis: clinical and
tiple sclerosis: evolution and persistence.AJNR Am J Neuro- radiologic features of five cases. AJNR Am J Neuroradiol
radiol 2001; 22: 664–669 2001; 22: 1362–1367
Hermans G, Stinissen P, Hauben L, van den Berg-Loonen E, Raus Killestein J, Kalkers NF, Meilof JF, Barkhof F, van Lier RA, Polman
J, Zhang J. Cytokine profile of myelin basic protein reactive CH. TNFalpha production by CD4(+) T celIs predicts long-
T cells in multiple sclerosis and healthy individuals. Ann term increase in lesion load on MRI in MS. Neurology 2001;
Neurol 1997; 42: 18–27 57: 1129–1131
Herrera BM, Ebers GC. Progress in deciphering the genetics of Killestein J, Eikelenboom MJ, Izeboud T, Kalkers NF, Ader HJ,
multiple sclerosis. Curr Opin Neurol 2003; 16: 253–258 Barkhof F, van Lier RA, Uitdehaag SM, Polman CH. Cytokine
Horsfield MA, Barker GJ, Barkhof F, Miller DH,Thompson AJ, Fil- producing CD8+ T celIs are correlated to MRI features of tis-
ippi M. Guidelines for using quantitative magnetization sue destruction in MS. J Neuroimmunol 2003; 142: 141–148
transfer resonance imaging for monitoring treatment of Kornek B, Bernert G, Balassy C, Geldner J, Prayer D, Feucht M.
multiple sclerosis.J Magn Reson Imaging 2003; 17: 389–397 Glatiramer acetate treatment in patients with acute child-
Iannucci G, Rovaris M, Giacomotti L, Comi G, Filippi M. Correla- hood and juvenile onset multiple sclerosis.Neuropediatrics
tion of multiple sclerosis measures derived from T2- 2003; 34: 120–126
weighted, T1-weighted, magnetization transfer, and diffu- Kotil K, Kalayci M, Köseoğlu T, Tuğrul A. Myelinoclastic diffuse
sion tensor MR imaging. AJNR Am J Neuroradiol 2001; 22: sclerosis (Schilder’s disease): a report of a case and review
1462–1467 of the literature. Br J Neurosurg 2002; 16: 516–519
Lassmann H. Neuropathology in multiple sclerosis: new con-
cepts. Multiple Sclerosis 1998; 4: 93–98
References and Further Reading 1021

Lassmann H, Bruck W, Lucchinetti C. Heterogeneity of multiple McDonald WI, Composton A, Edan G, Goodkin D, Hartung H-P,
sclerosis pathogenesis: implications for diagnosis and ther- Lublin FD, McFarland HF, Paty DW, Polman CH, Reingold SC,
apy.Trends Mol Med 2001; 7: 115–121 Sandberg-Wolheim M, Sibley W, Thompson A, van der
Li BSY, Regal J, Soher BJ, Mannon LJ, Grossman RI, Gonen O. Bri- Noort S,Weinshenker BY,Wolinsky JS. Recommended diag-
an metabolite profiles of T1-hypointense lesions in relaps- nostic criteria for multiple sclerosis: guidelines from the In-
ing-remitting multiple sclerosis. AJNR Am J Neuroradiol ternational Panel on the diagnosis of multiple sclerosis.
2003; 24: 68–74 Ann Neurol 2001; 50: 121–127
Loevner LA, Grossman RI, Cohen JA, Lexa FJ, Kessler D, Kolson Medena IM, Esiri MM. Axonal damage: a key predictor of out-
DL. Microscopic disease in normal-appearing white matter come in human CNS diseases. Brain 2003; 126: 515–530
on conventional MR images in patients with multiple scle- Merimsky O, Reider I, Merimsky E, Chaitchik S. Interferon-relat-
rosis: assessment with magnetization-transfer measure- ed leukoencephalopathy in a patient with renal cell carci-
ments. Radiology 1995; 196: 511–515 noma.Tumori 1991; 77: 361–362
Lublin FD, Reingold SC. Defining the clinical course of multiple Miller DH, Khan OA, Sheremata WA, Blumhardt LD, Rice GP,
sclerosis: results of an international study. Neurology 1996; Libonati MA, Willmer-Hulme AJ, Dalton CM, Miszkiel KA,
46: 907–911 O’Connor PW. A controlled trial of natalizumab for relaps-
Lucchinetti CF, Rodriguez M. The controversy surrounding the ing multiple sclerosis. N Engl J Med 2003; 348: 15–23
pathogenesis of the multiple sclerosis lesion. Mayo Clin Milligan NM, Newcombe R, Compston DAS. A double-blind
Proc 1997; 72: 665–678 controlled trial of high dose methylprednisolone in pa-
Lucchinetti CF, Mandler RN, McGavern D, Bruck W, Gleich G, tients with multiple sclerosis: 1. clinical effects. J Neurol
Ransohoff RM, Trebst C, Weinschenker B, Wingerchuck D, Neurosurg Psychiatry 1987; 50: 511–516
Parisi JE, Lassmann H. A role for humoral mechanisms in the Molyneux PD, Tubridy N, Parker GJM, MacManus DG, Tofts PS,
pathogenesis of Devic’s neuromyelitis optica. Brain 2002; Moseley IF, Miller DH.The effect of section thickness on MR
125: 1450–1461 lesions detection and quantification in multiple sclerosis.
Lycklama à Nijeholt GJ,Thompson A, Filippi M, Miller D, Polman AJNR Am J Neuroradiol 1998; 19: 1715–1720
C, Fazekas F, Barkhof F. Spinal-cord MRI in multiple scle- Muraro PA, Cassiani R, Ingoni RC, Martin R. Hematopoietic stem
rosos. Lancet Neurol 2003: 2: 555–562 cell transplantation for multiple sclerosis: current status
Lycklama à Nijeholt GJ, Barkhof F, Castelijns JA, Van Waes- and future challenges. Curr Opin Neurol 2003; 16: 299–305
berghe JHTM,Valk J, Jongen PJH, Hommes OR. Comparison Neumann H. Molecular mechanisms of axonal damage in in-
of two MR sequences for the detection of multiple sclerosis flammatory central nervous system diseases. Curr Opin
lesions in the spinal cord. AJNR Am J Neuroradiol 1996; 17: Neurol 2003: 16: 267–273
1533–1538 Noseworthy JH, Gold R, Hartung H-P. Treatment of multiple
Lycklama à Nijeholt GJ, van Walderveen MAA, Castelijns JA, van sclerosis: recent trails and future perspectives. Curr Opin
Waesberghe JHTM, Polman C, Scheltens P, Rosier PFWM, Neurol 1999; 12: 279–293
Jongen PJH, Barkhof F. Brain and spinal cord abnormalities Pathy DW, Arnold DL.The lesions of multiple sclerosis. N Engl J
in multiple sclerosis. Correlation between MRI parameters, Med 2002; 346: 199–200
clinical subtypes and symptoms. Brain 1998; 121: 687–697 Pathy DW, Oger JJF, Kastrukoff LF, Hashimoto SA, Hooge JP,
Lycklama à Nijeholt GJ, Bergers E, Kamphorst W, Bot J, Nicolay Eisen AA, Eisen KA, Purves SJ, Low MD, Brandejs V, Robert-
K, Castelijns JA, van Waesberghe JHTM, Ravid R, Polman CH, son WD, Li DKB. MRI in the diagnosis of MS: a prospective
Barkhof F. Post-mortem high-resolution MRI of the spinal study with comparison of clinical evaluation, evoked po-
cord in multiple sclerosis. A correlative study with conven- tentials, oligoclonal banding, and CT. Neurology 1988; 38:
tional MRI, histopathology and clinical phenotype. Brain 180–185
2001; 124: 154–166 Peterson JW, Bo L, Mork S, Chang A, Trapp BD. Transected neu-
Mader I, Roser W, Kappos L, Hagberg G, Seelig J, Radue EW, rites, apoptotic neurons, and reduced inflammation in
Steinbrich W. Serial proton MR spectroscopy of contrast- cortical multiple sclerosis lesions. Ann Neurol 2001; 50:
enhancing multiple sclerosis plaques: absolute metabolic 389–400
values over 2 years during a clinical pharmacological study. Polman CH, Hartung H-P. The treatment of multiple sclerosis:
AJNR Am J Neuroradiol 2000; 21: 1220–1227 current and future. Curr Opin Neurol 1995; 8: 200–209
Mader I, Seeger U, Weissert R, Klose U, Naegele T, Melms A, Poppe M, Bruck W, Hahn G, Weissbrich B, Heubner G, Goebel
Grodd W. Proton MR spectroscopy with metabolite-nulling HH, Todt H. Fulminant course in a case of diffuse myelin-
reveals elevated macromolecules in acute multiple sclero- oclastic encephalitis– a case report. Neuropediatrics 2001;
sis. Brain 2001; 124: 953–961 32: 41–44
Martino G, Adorini L, Rieckmann P, Hillerts J, Kallmann B, Comi Poser CM. The pathogenesis of multiple sclerosis: a commen-
G, Filippi M. Inflammation in multiple sclerosis: the good, tary. Clin Neurol Neurosurg 2000; 102: 191–194
the bad and the complex. Lancet [Neurol] 2002; 1: 499–509 Poser CM, Brinar VV. Diagnostic criteria for multiple sclerosis.
Masden JC, Quinto C, Olivera C,Tenner M, Leslie D,Visintainer P. Clin Neurol Neurosurg 2001; 103: 1–11
Open-ring imaging sign. Neurology 2000; 54: 1427–1433 Pretorius M-L, Loock DB, Ravenscoft A, Schoeman JF. Demyeli-
Masden JC, Quinto C, Olivera C,Tenner M, Leslie D,Visintainer P. nating disease of Schilder type in three young South
Open-ring imaging sign. Highly specific for atypical brain African children: dramatic response to corticosteroids.
demyelination. Neurology 2000; 54: 1427–1433 J Child Neurol 1998; 13: 197–201
McAdam LC, Blaser SI, Banwell BL. Pediatric tumefactive de- Pretorius PM, Quaghebeur G. The role of MRI in the diagnosis
myelination: case series and review of the literature. Pediatr of MS. Clin Radiol 2003; 58: 434–448
Neurol 2002; 26: 18–25 Rieckman P, Altenhofen B, Riegel A, Baudewig J, Felgenhauer K.
McDonald WI, Ron MA. Multiple sclerosis: the disease and its Soluble adhesion molecules (sVCAM-1 and sICAM-1) in
manifestations. Phil Trans R Soc Land B 1999; 354: 1615– cerebrospinal fluid and serum correlate with MRI activity in
1622 multiple sclerosis. Ann Neurol 1997; 41: 326–333
1022 References and Further Reading

Rovaris M, Bozzali M, Iannucci G, Ghezzi A, Caputo D, Montanari Stevenson VL, Leary SM, Losseff NA, Parker GJM, Barker GJ, Hu-
E, Bertolotto A, Bergamaschi R, Capra R, Mancardi GL, Mar- mani Y, Miller DH, Thompson AJ. Spinal cord atrophy and
tinelli V, Comi G, Filippi M. Assessment of normal-appearing disability in MS. A longitudinal study. Neurology 1998; 51:
white and gray matter in patients with primary progressive 234 – 238
multiple sclerosis diffusion – tensor magnetic resonance Stevenson VL, Smith SM, Matthews PM, Miller FH, Thompson
imaging study. Arch Neurol 2002; 59: 1406–1412 AJ. Monitoring disease activity and progression in primary
Rudick RA, Goodkin DE, Jacobs LD, Cookfair DL, Herndon RM, progressive multiple sclerosis using MRI: sub-voxel regis-
Richert JR,Salazar AM,Fisher JS,Granger CV,Simon JH,Alam tration to identify lesion changes and to detect cerebral at-
JJ, Simonian NA, Campion MK, Bartosak DM, Bourdette DN, rophy. J Neurol 2002; 249: 171–177
Braimain J, Brownscheidle CM, Coats ME, Cohand SL, Stinissen P, Medaer R, Raus J. Myelin reactive T cells in the au-
Dougherty DS, Kinkel RP, Mass MK, Munschauer RL, Pullici- toimmune pathogenesis of multiple sclerosis. Mult Scler
no PM, Scherokman BJ,Weistock-Guttman BJ,Whitham RH. 1998; 4: 203–211
Impact of interferon beta-1a on neurologic disability in re- Strangel M, Hartung H-P. Remyelinating strategies for the
lapsing multiple sclerosis. Neurology 1997; 49: 358–363 treatment of multiple sclerosis. Prog Neurobiol 2002; 68:
Sarchielli P, Presciutti O, Pelliccioli GP, Tarducci R, Gobbi G, 361–376
Chiarini P, Alberti A, Vicinanza F, Gallai V. Absolute quantifi- Tan IL, van Schijndel RA, Pouwels PJ, van Walderveen MA, Re-
cation of brain metabolites by proton magnetic resonance ichenbach JR, Manoliu RA, Barkhof F. MR venography of
spectroscopy in normal appearing white matter of multi- multiple sclerosis. AJNR Am J Neuroradiol 2000; 21 6
ple sclerosis patients. Brain 1999; 122: 513–521 1039–1042
Sarchielli P, Presciutti O, Tarducci R, Gobbi G, Alberti A, Pellicci- Tan IL, van Schijndel RA, Pouwels PJW, van Walderveen MAA,
oli GP, Chiarini P, Gallai V. Localized 1H magnetic resonance Reichenbach JR, Manoliu RA, Barkhof F. MR Venography
spectroscopy in mainly cortical gray matter of patients of multiple sclerosis. AJNR Am J Neuroradiol 2000; 21:
with multiple sclerosis. J Neurol 2002; 249: 902–910 1039–1042
Sawcer S, Compston A.The genetic analysis of multiple sclero- Tan IL, van Schijndel RA, van Walderveen MAA, Quist M, Bos R,
sis in Europeans: concepts and design. J Neuroimmunol Pouwels PJW, Desmedt P, Adèr HJ, Barkhof F. Magnetic reso-
2003; 143: 13–16 nance image registration in multiple sclerosis: comparison
Scarisbrick IA, Blaber SI, Lunchinetti CF, Genain CP, Blaber M, with repositioning error and observer-based variability.
Rodriguez M. Activity of a newly identified serine protease J Magn Reson Imaging 2002; 15: 505–510
in CNS demyelination. Brain 2002; 125: 1283–1296 Tartaglia MC, Narayanan S, de Stefano N, Arnaoutelis R, Antel
Schapiro RT, Langer SL. Symptomatic therapy of multiple scle- SB, Francis SJ, Santos AC, Lapierre Y, Arnold DL. Choline is in-
rosis. Curr Opin Neurol 1994; 7: 229–233 crease in pre-lesional normal appearing white matter in
Schönrock LM, Kuhlmann T, Adler S, Bitsch A, Brück W. Identifi- multiple sclerosis. J Neurol 2002; 249: 1382–1390
cation of glial cell proliferation in early multiple sclerosis Tartaglino LM, Friedman DP, Flanders AE, Lublin FD, Knober RL,
lesions. Neuropathol Appl Neurobiol 1998; 24: 320–330 Liem M. Multiple sclerosis in the spinal cord: MR appear-
Scully RE, Mark EJ, McNeely WF, Ebeling SH. Case 26–1998. ance and correlation with clinical parameters. Radiology
N Engl J Med 1998; 339: 542–549 1995; 195: 725–732
Shields DC, Banik NL. Pathophysiological role of calpain in Tas MW, Barkhof F, van Walderveen MAA, Polman CH, Hommes
experimental demyelination. J Neurosci Res 1999; 55: 533– OR, Valk J. The effect of gadolinium on the sensitivity and
541 specificity of MR in the initial diagnosis of multiple sclero-
Silver N, Lai M, Symms M, Barker G, McDonald I, Miller D. Serial sis. AJNR Am J Neuroradiol 1995; 16: 259–264
gadolinium-enhanced and magnetization transfer imag- Thompson AJ, Montalban X, Barkhof F, Brochet B, Filippi M,
ing to investigate the relationship between the duration of Miller DH, Polman CH, Stevenson VL, McDonald WI. Diag-
blood–brain barrier disruption and extent of demyelina- nostic criteria for primary progressive multiple sclerosis: a
tion in new multiple sclerosis lesions. J Neurol 1999; 246: position paper. Ann Neurol 2000; 47: 831–835
728–730 Thorpe JW, Kidd D, Mosely IF, Kendall BE, Thompson AJ, Mac-
Šišková D, Hadaè J. Multiple sclerosis in children. Eur J Paediatr Manus DG, McDonald WI, Miller DH. Serial gadolinium-en-
Neurol 1999; 3; A82 hanced MRI of the brain and spinal cord in early relapsing-
Smith KJ, Hall SM. Factors directly affecting impulse transmis- remitting multiple sclerosis. Neurology 1996; 46: 373–378
sion in inflammatory demyelinating disease: recent ad- Tintoré M, Rovira A, Martínez MJ, Rio J, Díaz-Valloslada P, Brieva
vances in our understanding. Curr Opin Neurol 2001; 14: L, Borrás C, Grivé E, Capellades J, Montalban X. Isolated de-
2389–298 myelinating syndromes: comparison of different MR imag-
Smith KJ, Lassmann H. The role of nitric oxide in multiple scle- ing criteria to predict conversation to clinically definite
rosis. Lancet [Neurol] 2002; 1: 232–241 multiple sclerosis. AJNR Am J Neuroradiol 2000; 21: 702–
Söderström M, Ya-Ping J, Hillert J, Link H. Optic Neuritis. Prog- 706
nosis for multiple sclerosis from MRI, CSF, and HLA findings. Tintoré M, Rovira A, Río J, Nos C, Grivé E, Sastre-Garriga J, Peri-
Neurology 1998; 50: 708–714 cot I, Sánchez E, Comabella M, Montalban X. New diagnos-
Soramani MP, Bruzzi P, Beckmann K, Wagner K, Miller DH, Kap- tic criteria for multiple sclerosis. Application in first de-
pos L, Filippi M. MRI metrics as surrogate endpoints for myelinating episode. Neurology 2003; 60: 27–30
EDSS progression in SPMS patients treated with IFN b-1b. Tourbah A, Stievenart J-L, Abanou AA, Iba-Zizen M-T, Hamard
Neurology 2003; 60: 1462–1466 H, Lyon-Caen O, Cabanis EA. Normal-appearing white mat-
Steinman L, Martin R, Bernard C, Conlon P, Oksenberg JR. Multi- ter in optic neuritis and multiple sclerosis: a comparative
ple sclerosis: deeper understanding of its pathogenesis re- proton spectroscopy study. Neuroradiology 1999; 41: 738–
veals new targets for therapy. Annu Rev Neurosci 2002; 25: 743
491–505
References and Further Reading 1023

Trojano M, Avolio C, Ruggieri, de Robertis F, Giuliani F, Paolicelli Yao S-Y, Stratton CW, Mitchell WM, Sriram S. CSF Oligoclonal
D, Livrea P. Soluble intracellular adhesion molecule-1 bands in MS include antibodies against Chlamydophila
(sICAM-1) in serum and cerebospinal fluid of demyelinat- antigens. Neurology 2001; 56: 1168–1176
ing diseases of the central and peripheral nervous system. Zivadinov R, Locatelli L, Stival B, Bratina A, Grop A, Nasuelli D,
Mult Scler 1998; 4: 39–44 Brnabic-Razmilic O, Zorzon M. Normalized regional brain
Ugawa Y, Shirouzu I,Terao Y, Hanajima R, Machii K, Mochizuki H, atrophy measurements in multiple sclerosis. Neuroradiolo-
Furybayashi T, Kanazawa I. Physiological analysis of a pa- gy 2003; 45: 793–798
tient with extreme widening of Virchow-Robin spaces.
J Neurol Sci 1998; 159: 25–27
Van Buchem MA, Udupa JK, McGowan, Miki Y,Heyning FH, Bon-
coeur-Martel M-P, Kolson DL, Polansky M, Grossman RI.
80 Acute Disseminated Encephalomyelitis
Global volumetric estimation of disease burden in multiple and Acute Hemorrhagic
sclerosis based on magnetization transfer imaging. AJNR Encephalomyelitis
Am J Neuroradiol 1997; 18: 1287–1290
Van Waesberghe JH, Kamphorst W, de Groot CJ, van An SF, Groves M, Martinian L, Kuo LT, Scaravilli F.Detection of in-
Walderveen MA, Castelijns JA, Ravid R, Lycklama à Nijeholt fectious agents in brain of patients with acute hemorrhag-
GJ, van der Valk P, Polman CH, Thompson AJ, Barkhof F. ic leukoencephalitis. J Neurovirol 2002; 8: 439–446
Axonal loss in multiple sclerosis lesions: magnetic reso- Andreula CD, Recchia Luciani ANM, Milella D. Magnetic reso-
nance imaging insights into substrates of disability. Ann nance imaging in the diagnosis of acute disseminated en-
Neurol 1999; 46: 747–754 cephalitis (ADEM). Int J Neuroradiol 1997; 3: 21–34
Van Walderveen MA, Barkhof F, Hommes OR, Polman CH, Tobi Anlar B, Basaran C, Kose G, Guven A, Haspolat S, Yakut A,
H, Frequin STFM,Valk J. Correlating MR imaging and clinical Sedaroglu A, Senbil N, Tan H, Karaagaoglu E, Karli Oguz K.
disease activity in multiple sclerosis: relevance of hypo- Acute disseminated encephalomyelitis in children: out-
intense lesions on short TR/short TE (T1-weighted) spin come and prognosis. Neuropediatrics 2003; 34: 194–199
echo images. Neurology 1995; 45: 1684–1690 Atlas SW, Grossman RI, Goldberg HI, Hackney DB, Bilaniuk LT,
Van Walderveen MA, Lycklama à Nyeholt GJ, Ader HJ, Jongen Zimmerman RA. MR diagnosis of acute disseminated en-
PJ, Polman CH, Castelijns JA, Barkhof F. Hypointense lesions cephalomyelitis. J Comput Assist Tomogr 1986; 10: 798–801
on T1 weighted spin-echo magnetic resonance imaging: Archer H, Wall R. Acute haemorrhagic leukoencephalopathy:
relation to clinical characteristics in subgroups of patients two case reports and review of the literature. J Infect 2003;
with multiple sclerosis. Arch Neurol 2001; 58: 76–81 46: 133–137
Van Walderveen MAA, Barkhof F, Hommes OR, Polman CH,Tobi Battistella PA, Laverda AM, Drigo P, Salandin M, Carollo C. Leu-
H, Frequin STFM,Valk J. Correlating MRI and clinical disease coencefalite acuta disseminata: aspetti clinici e neuroradio-
activity in multiple sclerosis: relevance of hypointense logici in 9 casi pediatrici. Riv Neuroradiol 1996; 9: 649–657
lesions on short-TR/short-TE (T1-weighted) spin-echo im- Baum PA, Barkovich AJ, Koch TK, Berg BO. Deep gray matter in-
ages. Neurology 1995; 45: 1684–1690 volvement in children with acute disseminated en-
Van Walderveen MAA, Kamphorst W, Scheltens P, van Waes- cephalomyelitis. AJNR Am J Neuroradiol 1994; 15: 1275–
berghe JHTM, Ravid R, Valk J, Polman CH, Barkhof F. 1283
Histopathologic correlate of hypointense lesions on T1- Bernarding J, Braun J, Koennecke HC. Diffusion- and perfusion-
weighted spin-echo MRI in multiple sclerosis. Neurology weighted MR imaging in a patient with acute demyelinat-
1998; 50: 1282–1288 ing encephalomyelitis (ADEM). J Magn Reson Imaging
Van Walderveen MAA, van Schijndel RA, Pouwels PJW, Polman 2002; 15: 96–100
CH, Barkhof F. Multislice T1 relaxation time measurements Bizzi A, Uluğ AM, Crawford TO, Passe T, Bugiani M, Bryan N,
in the brain using IR-EPI: reproducibility: normal values, and Barker PB. Quantitative proton MR spectroscopic imaging
histogram analysis in patients with multiple sclerosis. in acute disseminated encephalomyelitis.AJNR Am J Neuro-
J Magn Reson Imag 2003; 18: 656–664 radiol 2001; 22: 1125–1130
Wang CH, Walsh K. Multiple ring-enhancing lesions in a child Dale RC, Sousa de C, Chong WK, Cox TCS, Harding B, Neville
with relapsing multiple sclerosis. J Child Neurol 2002; 17: BGR. Acute disseminated encephalomyelitis, multiphasic
69–72 disseminated encephalomyelitis and multiple sclerosis in
Wang P-Y,Tseng C-L,Young C,Liu H-M,Chang Y-C,Shen Y-Z,Lee children. Brain 2000; 123: 2407–2422
C-Y. Multiple sclerosis in children: clinical, neuroimaging, Feydy A, Carlier R, Mompoint D, Clair B, Chillet P, Vallee C. Brain
and neurophysiological correlations. Acta Paediatr Sin and spinal cord MR imaging in a case of acute disseminat-
1995; 36: 93–100 ed encephalomyelitis. Eur Radiol 1997; 7: 415–417
Wasman SG. Do ‘demyelinating’ diseases involve more than Gupte G, Stonehouse M, Wassmer E, Coad NAG, Whitehouse
myelin? Nat Am 2000; 6: 738–739 WP.Acute disseminated encephalitis:a review of 18 cases in
Wingerchuck DM, Hogancamp WF, O’Brien PC, Weinschenker childhood. J Paediatr Child Health 2003; 39: 336–342
BG.The clinical course of neuromyelitis optica (Devic’s syn- Haase CG, Faustmann PM, Diener HC. Idiopathic inflammatory
drome). Neurology 1999; 53: 1107–1114 demyelinating diseases of the central nervous system: dif-
Wood DD, Bilbao JM, O’Connors P, Moscarello MA. Acute multi- ferentiating between acute disseminated encephalo-
ple sclerosis (Malburg type) is associated with develop- myelitis and malignant multiple sclerosis. J Clin Neurosci
mentally immature myelin basic protein. Ann Neurol 1996; 1999; 6: 221–226
40: 18–24 Hartung HP, Grossman RI. ADEM. Distinct disease or part of the
Wuerfel J,Bellmann-Strobl J,Brunecker P,Aktas O,McFarland H, MS spectrum? Neurology 2001; 56: 1257–1260
Villiringer A, Zipp F. Changes in cerebral perfusion precede
plaque formation in multiple sclerosis: a longitudinal per-
fusion MRI study. Brain 2004; 127: 111–119
1024 References and Further Reading

Höllinger P, Sturzenegger M, Mathis J, Schroth G, Hess CW. Shoji H, Kusuhara T, Honda Y, Hino H, Kojima K, Abe T,Watanabe
Acute disseminated encephalomyelitis in adults: a reap- M. Relapsing acute disseminated encephalomyelitis associ-
praisal of clinical, CSF, EEG, and MRI findings. J Neurol 2002; ated with chronic Epstein-Barr virus infection: MRI findings.
249: 320–329 Neuroradiology 1992; 34: 340–342
Hynson JL, Kornberg AJ, Coleman LT, Shield L, Harvey AS, Kean Tachi N,Watanabe T,Wakai S, Sato T, Chiba S. Acute disseminat-
MJ. Clinical and neuroradiologic features of acute dissemi- ed encephalomyelitis following HTLV-I associated myelo-
nated encephalomyelitis in children. Neurology 2001; 56: pathy. J Neurol Sci 1992; 110: 234–235
1308–1312 Tenembaum S, Chamoles N, Fejerman N. Acute disseminated
Inglese M. Salvi F, Iannucci G, Mancardi GL, Mascalchi M, Filippi encephalomyelitis, a long-term follow-up study of 84 pedi-
M. Magnetization transfer and diffusion tensor MR imaging atric patients. Neurology 2002; 59: 1224–1231
of acute disseminated encephalomyelitis. AJNR Am J Neu- Yapici Z, Eraksoy M. Bilateral demyelinating tumefactive le-
roradiol 2002; 23: 267–272 sions in three children with hemiparesis. J Child Neurol
Jaing TH,Lin KL,Chiu CH,Lo WC,Wu PL.Acute disseminated en- 2002; 17: 655–660.
cephalomyelitis in autoimmune hemolytic anemia. Pediatr Yoshikawa H, Oda Y. Acquired aphasia in acute disseminated
Neurol 2001; 24: 303–305 encephalomyelitis. Brain Dev 1999; 21: 341–344
Johnson RT, Griffin DE, Hirsch RL, Wolinsky JS, Roedenbeck S,
Lindo de Soriano I, Vaisberg A. Measles encephalomyelitis-
clinical and immunologic studies. N Engl J Med 1984; 310:
137–141
81 Acquired Immunodeficiency Syndrome
Kepes JJ. Large focal tumor-like demyelinating lesions of the
brain: intermediate entity between multiple sclerosis and Abdoulafia DM, Taylor L. Vacuolar myelopathy and vacuolar
acute disseminated encephalomyelitis? A study of 31 pa- cerebellar leukoencephalopathy: a late complication of
tients. Ann Neurol 1993; 33: 18–27 AIDS after highly active antiretroviral therapy-induced im-
Khong PL, Ho HK, Cheng PW,Wong VCN, Goh W, Chan FL. Child- mune reconstruction. AIDS Pat Care 2002; 16: 579–584
hood acute disseminated encephalomyelitis: the role of Albright AV, Soldan SS, Gonzalez-Scarano F. Pathogenesis of
brain and spinal cord MRI. Pediatr Radiol 2002; 32: 59–66 human immunodeficiency virus-induced neurological dis-
Kimura S, Nezu A, Ohtsuki N, Kobayashi T, Osaka H, Uehara S. ease. J Neurovirol 2003; 9: 222–227
Serial magnetic resonance imaging in children with postin- Anderson E, Zink W, Xiong H, Gendelman HE. HIV-1-associated
fectious encephalitis. Brain Dev 1996; 18: 461–465 dementia: a metabolic encephalopathy perpetrated by
Klein CJ, Wijdicks EFM, Earnest IV F. Full recovery after acute virus-infected and immune-competent mononuclear
hemorrhagic leukoencephalitis (Hurst’s disease). J Neurol phagocytes.J Acquir Immune Defic Syndr 2002; 31:S43-S54
2000; 247: 977–979 Annunziata P. Blood–brain barrier changes during invasion of
Mader I, Wolff M, Niemann G, Küker W. Acute haemorrhagic the central nervous system by HIV-1. J Neurol 2003; 250:
encephalomyelitis (ADEM): MRI findings. Neuropediatrics 901–906
2004; 35: 143–146 Avison MJ,Nath A,Berger JR.Understanding pathogenesis and
Murthy JMK. MRI in acute disseminated encephalomyelitis fol- treatment of HIV dementia: a role for magnetic resonance?
lowing Semple antirabies vaccine. Neuroradiology 1998; Trends Neurosci 2002; 25: 468–473
40: 420–423 Barret B, Tardieu M, Rustin P, Lacroix C, Chabrol B, Desguerre I,
Murthy SNK, Faden HS, Cohen ME, Bakshi R. Acute disseminat- Dollfus C, Mayaux M-J, Blance S. Persistent mitochondrial
ed encephalomyelitis in children. Pediatrics 2002; 110: e21 dysfunction in HIV-1-exposed but uninfected infants: clini-
Nagai K, Mori T. Acute disseminated encephalomyelitis with cal screening in a large prospective cohort. AIDS 2003; 17:
probable measles vaccine failure. Pediatr Neurol 1999; 20: 1769–1785
399–402 Belman AL, Ultmann MH, Horoupian D, Novick B, Spiro AJ, Ru-
O’Riordan JI, Gomez-Anson B, Moseley IF, Miller DH. Long term binstein A, Kurtzberg D, Cone-Wesson B. Neurological com-
MRI follow-up of patients with post infectious en- plications in infants and children with acquired immune
cephlomyelitis: evidence for a monophasic disease. J Neu- deficiency syndrome. Ann Neurol 1985; 18: 560–566
rol Sci 1999; 167: 132–136 Bencherif B, Rottenberg DA. Neuroimaging of the AIDS de-
Piyasirisilp S, Hemachudha T. Neurological adverse events as- mentia complex. AIDS 1998; 12: 233–244
sociated with vaccination. Curr Opin Neurol 2002; 15: Berger JR, Sheremata WA, Resnick L, Atherton S, Fletcher MA,
333–338 Norenberg M. Multiple sclerosis-like illness occurring with
Reich H, Lin SR, Goldblatt D. Computerized tomography in human immunodeficiency virus infection. Neurology 1989;
acute hemorrhagic leukoencephalopathy: a case report. 39: 324–329
Neurology 1979; 29: 255–258 Blanche S, Tardieu M, Rustin P, Slama A, Barret B, Firtion G,
Schwarz S, Mohr A, Knauth M, Wildemann B, Storch-Hagen- Ciraru-Vigneron N, Lacroix C, Rouzioux C, Mabdelbrot L,
locher B. Acute disseminated encephalomyelitis, a follow- Desguerre I, Rötig A, Mayaux M-J, Delfraissy JF. Persistent
up study of 40 adult patients. Neurology 2001; 56: mitochondrial dysfunction and perinatal exposure to anti-
1313–1318 retroviral nucleoside analogues. Lancet 1999; 354: 1084–
Scully RE, Mark EJ, McNeely WF, Ebeling SH. Case records of the 1089
Massachusetts General Hospital. N Engl J Med 1996; 334: Camacho DLA, Smith JK, Castillo M. Differentiation of toxoplas-
715–720 mosis and lymphoma in AIDS patients by using apparent
Scully RE, Mark EJ, McNeely WF, Ebeling SH. Case records of the diffusion coefficients. AJNR Am J Neuroradiol 2003; 24:
Massachusetts General Hospital. N Engl J Med 1999; 340: 633–637
127–135 Cedis Meltzer C,Wells SW, Bechner MW, Flanigan KM, Oyler GA,
Lee RR. AIDS-related MR hyperintensity of the basal gan-
glia. AJNR Am J Neuroradiol 1998; 19: 83–89
References and Further Reading 1025

Chamberlain MC,Nichols SL,Chase CH.Pediatric AIDS:compar- Geremia GK, McCluney KW, Adler SS, Charletta DA, Hoile RD,
ative cranial MRI and CT scans. Pediatr Neurol 1991; 7: Huckman MS, Ramsey RG. The magnetic resonance hy-
357–362 pointense spine of AIDS. J Comput Assist Tomogr 1990; 14:
Chang L. In vivo magnetic resonance spectroscopy in HIV and 785–789
HIV-related brain diseases. Rev Neurosci 1995; 6: 365–378 Grafe MR, Press GA, Berthoty DP, Hesselink JR,Wiley CA. Abnor-
Chong J, di Rocco A,Tagliati M, Danisi F, Simpson DM, Atlas SW. malities of the brain in AIDS patients: correlation of post-
MR findings in AIDS–associated myelopathy. AJNR Am J mortem MR findings with neuropathology.AJNR Am J Neu-
Neuroradiol 1999; 20: 1412–1416 roradiol 1990; 11: 905–911
Chryskikopoulos HS, Press GA, Grafe MR, Hesselink JR, Wiley Goldstick L, Mandybur TI, Bode R. Spinal cord degeneration in
CA. Encephalitis caused by human immunodeficiency AIDS. Neurology 1985; 35: 103–106
virus; CT and MR imaging manifestations with clinical and Gray F, Gherardi R, Scaravilli F. The neuropathology of the ac-
pathologic correlation. Radiology 1990; 175: 185–191 quired immune deficiency syndrome (AIDS).A review.Brain
Church JA. Reversible leukoencephalopathy in a patient with 1988; 111: 245–266
nucleoside analogue-associated mitochondrial depletion Greene WC. Mechanisms of disease. The molecular biology of
and metabolic disease. AIDS 2002;16:2366–2367 human immunodeficiency virus type 1 infection. N Engl J
Cinque P, Bossolasco S, Bestetti A, Sala S, Pierotti C, Lazzarin A. Med 1991; 324: 308–317
Molecular studies of cerebrospinal fluid in human immun- Hénin D, Smith TW, de Girolami U, Sughayer M, Hauw J-J. Neu-
odeficiency virus type 1-associated opportunistic central ropathology of the spinal cord in the acquired immunode-
nervous system diseases – an update. J Neurovirol 2002; 8: ficiency syndrome. Hum Pathol 1992; 23: 1106–1114
122–128 Ho DD, Pomarantz RJ, Kaplan JC. Pathogenesis of infection
Corral I, Quereda C, Hellin T, Navas E, García-Villanueva M. Re- with human immunodeficiency virus. N Engl J Med 1987;
lapsing and remitting leukoencephalopathy associated 317: 278–286
with chronic HIV infection. Eur Neurol 2002; 48: 39–41 Holland NR, Power C, Mathews VP, Glass JD, Forman M,
Côté HCF, Brumme ZL, Craib KJP, Alexander CS, Wynhoven B, McArthur JC. Cytomegalovirus encephalitis in acquired im-
Ting L,Wong H, Harris M, Harrigan PR, O’ Shaunghnessy MV, munodeficiency syndrome (AIDS). Neurology 1994; 44:
Montaner JSG. Changes in mitochondrial DNA as a marker 507–514
of nucleoside toxicity in HIV-infected patients.N Engl J Med Holliday RA. Manifestations of AIDS in the oromaxillofacial re-
2002; 346: 811–820 gion.The role of imaging. Radiol Clin N Am 1993; 31: 45–60
Curless RG. Congenital AIDS: review of neurologic problems. Ito M, Baker JV, Mock DJ, Goodman AD, Blumberg BM, Shrier
Childs Nerv Syst 1989; 5: 9–11 DA, Powers JM. Human herpes 6-meningoencephalitis in
Da Chuna A, Mintz M, Eiden LE, Sharer LR. A neuronal and neu- an HIV patient with progressive multifocal leukoen-
roanatomical correlate of HIV-1 encephalopathy relative to cephalopathy. Acta Neuropathol (Berl) 2000; 100: 337–341
HIV-1 in HIV-1-infected children. J Neuropathol Exp Neurol Jones HR Jr, Ho DD, Forgacs P, Adelman LS, Silverman ML, Bak-
1997; 56: 974–987 er RA, Locuratolo P. Acute fulminating fatal leukoen-
Delisle MB, Bouissou H, Saidi A. What’s new in cerebral pathol- cephalopathy as the only manifestation of human immun-
ogy in acquired immune deficiencies? Pathol Res Pract odeficiency virus infection. Ann Neurol 1988; 3: 519–522
1986; 181: 85–92 Kauffman WM, Sivit CJ, Fitz CR, Rakusan TA, Herzog K, Chandra
Diaz-Marchan PJ, Huang ML, Jackson EF, Norton RE, Hayman RS. CT and MR evaluation of intracranial involvement in pe-
LA. Triple-dose contrast-enhanced images in neurological- diatric HIV infection: A clinical-imaging correlation. AJNR
ly symptomatic HIV-positive patients. Neuroradiology Am J Neuroradiol 1992; 13: 949–957
2000; 42: 256–260 Kirshenbaum KJ, Nadimpalli SR, Friedman M, Kirshenbaum GL,
Di Rocco A, Bottiglieri T, Werner P, Geraci A, Simpson D, God- Cavallino RP. Benign lymphoepithelial parotid tumors in
bold J, Morgello S. Abnormal cobalamin-dependent trans- AIDS patients: CT and MR findings in nine cases. AJNR Am J
methylation in AIDS-associated myelopathy. Neurology Neuroradiol 1991; 12: 271–274
2002; 58: 730–735 Kovacs A, Schluchter M, Easley K, Demmler G, Shearer W, la Rus-
Eggers C. HIV-1 associated encephalopathy and myelopathy. sa P,Pitt J,Cooper E,Goldfarb J,Hodes D,Kattan M,McIntosh
J Neurol 2002; 249: 1132–1136 K. Cytomegalovirus infection and HIV-1 disease progres-
Ellis RJ, Moore DJ, Childers ME, Letendre S, McCutchan JA,Wolf- sion in infants born to HIV-1-infected women. N Engl J Med
son T, Spector SA, Hsia K, Heaton RK, Grant I. Progression to 1999; 341: 77–84
neuropsychological impairment in human immunodefi- Kupfer MC, Zee CS, Colletti PM, Boswell WD, Rhodes R.MRI eval-
ciency virus infection predicted by elevated cerebrospinal uation of AIDS-related encephalopathy: toxoplasmosis vs.
fluid levels of human immunodeficiency virus RNA. Arch lymphoma. Magn Reson Imaging 1990; 8: 51–57
Neurol 2002; 59: 923–928 Langford TD, Letendre SL, Marcotte TD, Ellis RJ, McCuthan JA,
Filippi CG, Sze G, Farber SJ, Shahmanesh M, Selwyn PA. Regres- Grant I, Mallory ME, Hansen LA, Archibald S, Jernigan T,
sion of HIV encephalopathy and basal ganglia signal inten- Masliah E. Severe, demyelinating leukoencephalopathy in
sity abnormality at MR imaging in patients with AIDS after AIDS patients on antiretroviral therapy. AIDS 2002; 16:
the initiation of protease inhibitor therapy. Radiology 1998; 1019–1029
206: 491–498 Langford TD, Letendre SL, Larrea GJ, Masliah E. Changing pat-
Fliss DM, Parikh J, Freeman JL.AIDS-related Kaposi’s sarcoma of terns in the neuropathogenesis of HIV during the HAART
the sphenoid sinus. J Otolaryngol 1992; 21: 235–237 era. Brain Pathol 2003; 13: 195–210
Flowers CH, Mafee MF, Crowell R, Raofi B, Arnold P, Dobben G, Leger JM,Bouche P,Bolgert F,Chaunu MP,Rosenheim M,Catha-
Wycliffe N. Encephalopathy in AIDS patients: evaluation la HP, Gentilini M, Hauw JJ, Brunet P. The spectrum of
with MR imaging. AJNR Am J Neuroradiol 1990; 11: polyneuropathies in patients infected with HIV. J Neurol
1235–1245 Neurosurg Psychiatry 1989; 52: 1369–1374
1026 References and Further Reading

Levy RM, Pons VG, Rosenblum ML. Central nervous system Thurnher MM, Schindler EG, Thurnher SA, Pernerstorfer-Schön
mass lesions in the acquired immunodeficiency syndrome H, Kleibl-Popov C, Rieger A. Highly active antiretroviral ther-
(AIDS). J Neurosurg 1984; 61: 9–16 apy for patients with AIDS dementia complex: effect on MR
Lindegren ML, Rhodos P, Gordon L, Fleming P. Lack of mortality imaging findings and clinical course. AJNR Am J Neuroradi-
related to mitochondrial dysfunction among perinatally ol 2000; 21: 670–678
HIV-exposed children in pediatric HIV surveillance. Ann N Y Thurnher MM, Thurnher SA, Schindler E. CNS involvement in
Acad Sci 2000; 918: 222–235 AIDS: spectrum of CT and MR findings. Eur Radiol 1997; 7:
Lüer W, Gerhards J, Poser S, Weber T, Felgenhauer K. Acute dif- 1091–1097
fuse leukoencephalitis in HIV-1 infection. J Neurol Neuro- Trenkwalder P, Trenkwalder C, Feiden W, Vogl TJ, Einhäupl KM,
surg Psychiatry 1994; 57: 105–107 Lydtin H. Toxoplasmosis with early intracerebral hemor-
Mamidi A,DeSimone JA,Pomerantz RJ.Central nervous system rhage in a patient with the acquired immunodeficiency
infections in individuals with HIV-1 infection. J Neurovirol syndrome. Neurology 1992; 42: 436–438
2002;8:158–167 Tucker T. Central nervous system AIDS. J Neurol Sci 1989; 89:
Morris AA, Carr A. HIV nucleoside analogues: new adverse ef- 119–133
fects on mitochondria? Lancet 1999; 354: 1046–1047 Vago L, Bonetto S, Nebuloni M, Duca P, Carsana L, Zerbi P,
Navia BA, Cho E-S, Petito CK, Price RW.The AIDS dementia com- D’Arminio-Monforcte. Pathological findings in the central
plex. II. Neuropathology. Ann Neurol 1986; 19: 525–535 nervous system of AIDS patients on assumed antiretroviral
Nielsen SL, Petito CK, Urmacher CD, Posner JB. Subacute en- therapeutic regiments: retrospective study of 1597 autop-
cephalitis in acquired immune deficiency syndrome: a sies. AIDS 2002; 16: 1925–1928
postmortem study. Am J Clin Pathol 1984; 82: 678–682 Visudtibhan A, Visudhipan P, Chiemchanya S. Stroke and
Portegies P, Enting RH, Gans de J, Algra PR, Derix MMA, Lange seizures as the presenting signs of pediatric HIV infection.
JMA, Goudsmit J. Presentation and course of AIDS demen- Pediatr Neurol 1999; 20: 53–56
tia complex: 10 years of follow-up in Amsterdam, the Vittecoq S, Jardel C, Barthélémy C, Escout L, Cheminot N,
Netherlands. AIDS 1993; 7: 669–675 Chapin S, Sternberg D, Maisonobe T, Lombès SA. Mito-
Quencer RM. AIDS-associated myelopathy: clinical severity, MR chondrial damage associated with long-term antiretro-
findings, and underlying etiologies. AJNR Am J Neuroradiol viral treatment: associated alteration or causal disorder?
1999; 20: 1387–1388 J Acquir Immune Defic Syndr 2002; 31: 299–308
Sacktor N. The epidemiology of human immunodeficiency Walker UA, Setzer B,Venhoff N. Increased long-term mitochon-
virus-associated neurological disease in the era of highly drial toxicity in combinations of nucleoside analogue re-
active antiretroviral therapy. J Neurovirol 2002; 8 Suppl 2: verse-transcriptase inhibitors. AIDS 2002; 16: 2165–2173
115–1121 Wang H, Lemire BD,Cass CE,Weiner JH, Michalak M, Penn AMW,
Sánchez-Ramón S,Cantó-Nogués C,Muñoz-Fernández MA.Re- Fliegel L. Zidovudine an dideoxynucleosides deplete wild-
construction of the course of HIV-1-associated progressive type mitochondrial DNA levels and increase deleted mito-
encephalopathy in children. Med Sci Monit 2002; 8: RA249- chondrial DNA levels in cultured Kearns-Sayre syndrome
RA252 fibroblasts. Biochim Biophys Acta 1996; 1316: 51–59
Shugar JMA, Som PM, Jacobson AL, Ryan JR, Bernard PJ, Dick- Wehn SM, Heinz ER, Burger PC, Boyko OB. Dilated Virchow-
man SH. Multicentric parotid cysts and cervical adenopa- Robin spaces in cryptococcal meningitis associated with
thy in AIDS patients. A newly recognized entity: CT and MR AIDS: CT and MR findings.J Comput Assist Tomogr 1989; 13:
manifestations. Laryngoscope 1988; 98: 772–775 756–762
Snider WD, Simpson DM, Nielsen S, Gold JWM, Metroka CE, Pos- Zimmer C, Märzheuser S, Patt S, Rolfs A, Gottschalk J, Weigel K,
ner JB.Neurological complications of acquired immune de- Gosztonyi G. Stereotactic brain biopsy in AIDS. J Neurol
ficiency syndrome: analysis of 50 patients. Ann Neurol 1992; 239: 394–400
1983; 14: 403–418
Stankoff B, Tourbah A, Suarez S, Turell E, Stievenart JL, Payan C,
Coutellier A, Herson S, Baril L, Bricaire F, Calvez V, Cabanis EA,
Lacomblez L, Lubetzki C. Clinical and spectroscopic im-
82 Progressive Multifocal
provement in HIV-associated impairment. Neurology 2001; Leukoencephalopathy
56: 112–115
Starr SE, Fletcher CV, Spector SA, Yong FH, Fenton T, Brundage Agostini HT, Ryschkewitsch CF, Baumhefner RW, Tourtelotte
RC, Manion D, Ruiz NM, Gersten M, Becker M, McNamara J, WW, Singer EJ, Komoly S, Stoner GL. Influence of JC virus
Mofenson LM. Combination therapy with efavirenz, nelfi- coding region genotype on risk of multiple sclerosis and
navir, and nucleoside reverse-transcript inhibitors in chil- progressive multifocal leukoencephalopathy. J Neurovirol
dren infected with human immunodeficiency type 1. N En- 2000; 6: S101-S108
gl J Med 1999; 341: 1874–1881 Aksamit AJ, Gendelman HE, Orenstein JM, Pezeshkpour GH.
Steinbrook R, Drazen JM.AIDS – will the next 20 years be differ- AIDS-associated progressive multifocal leukoencephalo-
ent? N Engl J Med 2001; 344: 1781–1782 pathy (PML): comparison to non-AIDS PML with situ hy-
Swindells S, Zheng J, Gendelman HE. HIV-associated dementia: bridization and immunohistochemistry. Neurology 1990;
new insights into disease pathogenesis and therapeutic in- 40: 1073–1078
terventions. AIDS Pat Care 1999; 13: 153–163 Arai Y, Tsutsui Y, Nagashima K, Shinmura Y, Kosugi T, Wakai M,
Talpos D, Tien RD, Hesselink JR. Magnetic resonance imaging Nishikage H, Yamamoto J. Autopsy case of the cerebellar
of AIDS-related polyradiculopathy. Neurology 1991; 41: form of progressive multifocal leukoencephalopathy with-
1996–1997 out immunodeficiency. Neuropathology 2002; 22: 48–56
Tardieu M. HIV-1-related central nervous system diseases. Curr Berger JR, Pall L, Lanska D,Whiteman M. Progressive multifocal
Opin Neurol 1999; 12: 377–381 leukoencephalopathy in patients with HIV infection. J Neu-
Thurnher MM, Rieger A, Kleibl-Popov C, Settinek U, Henk C, rovirol 1998; 4: 59–68
Haberler C, Schindler E. Neuroradiology 2001; 43: 29–35
References and Further Reading 1027

Berger JR, Major EO.Progressive multifocal leukoencephalopa- Houff SA, Major EO, Katz DA, Kufta CV, Sever JL, Pittaluga S,
thy. Semin Neurol 1999; 19: 193–200 Roberts JR, Gitt J, Saini N, Lux W. Involvement of JC virus-in-
Bergui M, Bradac GB, Oguz KK, Boghi A, Geda C, Gatti G, Schiffer fected mononuclear cells from the bone marrow and
D. Progressive multifocal leukoencephalopathy: diffusion- spleen in the pathogenesis of progressive multifocal
weighted imaging and pathological correlations. Neurora- leukoencephalopathy. N Engl J Med 1988; 318: 301–305
diology 2004; 46: 22–25 Iida T, Kitamura T, Guo J, Taguchi F, Aso Y, Nagashima K, Yogo Y.
Cardenas RL, Cheng KH, Sack K.The effects of cidofovir on pro- Origin of JC polyomavirus variants associated with pro-
gressive multifocal leukoencephalopathy: an MRI case gressive multifocal leukoencephalopathy. Proc Natl Acad
study. Neuroradiology 2001; 43: 379–382 Sci 1993; 90: 5062–5065
Del Valle L, Gordon J, Assimakopoulou M, Enam S, Geddes JF, Kasner SE, Gelatta SL, McGowan JC, Grossman RI. Magnetiza-
Varakis JN, Katsetos CD, Croul S, Khalili K. Detection of JC tion transfer imaging in progressive multifocal leukoen-
virus DNA sequences and expression of the viral regulatory cephalopathy. Neurology 1997; 48: 534–536
protein T-antigen in tumors of the central nervous system. Koralnik IJ. Overview of the cellular immunity against JC virus
Cancer Res 2001; 61: 4287–4293 in progressive multifocal leukoencephalopathy. J Neurovi-
Donovan Post MJ, Yiannoutsos C, Simpson D, Booss J, Clifford rol 2002; 2: 59–65
DB, Cohen B, McArthur JC, Hall CD. Progressive multifocal Kuchelmeister K, Gullotta F, Bergmann M, Angeli G, Masini T.
leukoencephalopathy in AIDS: are there any MR findings Progressive multifocal leukoencephalopathy (PML) in the
useful to patient management and predictive of patient acquired immunodeficiency syndrome (AIDS). Pathol Res
survival? AJNR Am J Neuroradiol 1999; 20: 1896–1906 Pract 1993; 189: 163–173
Dousset V, Armand J-P, Lacoste D, Mièze S, Letenneur L, Dar- Mark AS, Atlas SW. Progressive multifocal leukoencephalopa-
tigues J-F, Caillé J-M. Magnetization transfer study of HIV thy in patients with AIDS:appearance on MR images.Radio-
encephalitis and progressive multifocal leukoencephalo- logy 1989; 173: 517–520
pathy. AJNR Am J Neuroradiol 1997; 18: 895–901 Marra CM, Rajicic N, Barker DE, Cohen BA, Clifford D, Donovan
Du Pasquier RA, Koralnik IJ. Inflammatory reaction in progres- Post MJ, Ruiz A, Bowen BC, Huang M-L, Queen-Baker J, An-
sive multifocal leukoencephalopathy: harmful or benefi- dersen J, Kelly S, Shriver S. A pilot study of cidofovir for pro-
cial? J Neurovirol 2003; 9 (Suppl 1): 25–31 gressive multifocal leukoencephalopathy in AIDS. AIDS
Enting RH, Portegies P. Cytarabine and highly active antiretro- 2002; 16: 1791–1797
viral therapy in HIV-related progressive multifocal leuko- Mock DJ, Powers JM, Goodman AD, Blumenthal SR, Ergin N,
encephalopathy. J Neurol 2000; 247: 134–138 Baker JV, Mattson DH, Assouline JG, Bergey EJ, Chem N. Ep-
García de Viedma D, Diaz Infantes M, Miralles P, Berenguer J, stein LG, Blumberg BM. J Neurovirol 1999; 5: 363–373
Marin M,Muñoz L,Bouza E.JC Virus load in progressive mul- Newton HB, Makley M, Slivka AP, Li J. progressive multifocal
tifocal leukoencephalopathy:analysis of the correlation be- leukoencephalopathy presenting as multiple enhancing
tween the viral burden in cerebrospinal fluid, patient sur- lesions on MRI: case report and literature review. J Neu-
vival, and the volume of neurological lesions. Clin Infect Dis roimaging 1995; 5: 125–125
2002; 34: 1568–1575 Newton P, Aldridge RD, Lessells AM, Best PV. Progressive multi-
Gentile S, Sacerdote L, Roccatello D, Giordana MT. Progressive focal leukoencephalopathy complicating systemic lupus
multifocal leukoencephalopathy during cyclosporine erythematosus. Arthritis Rheum 1986; 29: 337–343
treatment. A case report. Ital J Neurol Sci 1996; 17: 363–366 Okada Y, Sawa H, Endo S, Orba Y, Umemura T, Nishihara H, Stan
Gordon J, Gallia GL, del Valle L, Amini S, Khalili K. Human poly- AC, Takana S, Takahashi H, Nagashima K. Expression of JC
omavirus JCV and expression of myelin genes. J Neurovirol virus agnoprotein in progressive multifocal leukoen-
2000; 6: S92-S97 cephalopathy brain. Acta Neuropathol (Berl) 2002; 104:
Hou J, Major EO. Progressive multifocal leukoencephalopathy: 130–136
JC Virus-induced demyelination in the immune compro- Padgett BL, Walker DL, ZuRhein GM, Hodach AE, Chou SM. JC
mised host. J Neurovirol 2000; 6: S98-S100 papovavirus in progressive multifocal leukoencephalo-
Hal CD, Dafni U, Simpson D, Clifford D, Wetherill PE, Cohen B, pathy. J Infect Dis 1976; 133: 686–690
McArthur J, Hollander H, Yainnoutsos C, Major E, Millar L, Peters ACB,Versteeg J, Bots GTAM, Boogerd W,Vielvoye GJ. Pro-
Timpone J. Failure of cytarabine in progressive multifocal gressive multifocal leukoencephalopathy. Immunofluores-
leukoencephalopathy associated with human immunode- cent demonstration of simian virus 40 antigen in CSF cells
ficiency virus infection. N Engl J Med 1998; 338: 1345–1351 and response to cytarabine therapy. Arch Neurol 1980; 37:
Hansman Whiteman ML, Donovan Post MJ, Berger JR, Tate LG, 497–501
Bell MD, Limonte LP. Progressive multifocal leukoen- Port JD, Miseljic S, Lee RR, Ali SZ, Nicol TL, Royal W, Chin BB. Pro-
cephalopathy in 47 HIV-seropositive patients: neuroimag- gressive multifocal leukoencephalopathy demonstrating
ing with clinical and pathologic correlation. Radiology contrast enhancement on MRI and uptake of thallium-201:
1993; 187: 233–240 a case report. Neuroradiology 1999; 41: 895–898
Hawkins CP, McLaughlin JE, Kendall BE, McDonald WI. Patho- Roberts MTM, Carmichael A, Lever AML. Prolonged survival in
logical findings correlated with MRI in HIV infection. Neuro- AIDS-related progressive multifocal leukoencephalopathy
radiology 1993; 35: 264–268 following anti-retroviral therapy and cidofovir. Int J Antimi-
Henson J, Saffer J, Furneaux H. The transcription factor Sp 1 crob Agents 2003; 21: 347–349
binds to the JC virus promoter and is selectively expressed Rockwell D, Ruben FL, Winkelstein A, Mendelow H. Absence of
in glial cells in human brain. Ann Neurol 1992; 32: 72–77 immune deficiencies in a case of progressive multifocal
Ho JL, Poldre PA, McEniry D, Howley PM, Snydman DR, Rudders leukoencephalopathy. Am J Med 1976; 61: 433–436
RA,Worthington M.Acquired immunodeficiency syndrome Rubin DI, Norris S, Flint R. Isolated pontine progressive multifo-
with progressive multifocal leukoencephalopathy and cal leukoencephalopathy: unusual magnetic resonance
monoclonal B-cell proliferation. Ann Intern Med 1984; 100: imaging features. J Neuroimaging 2002; 12: 63–66
693–696
1028 References and Further Reading

Sabath BF, Major EO.Traffic of JC virus from sites of initial infec- Bahemuka M, Rahman Shemena A, Panayiotopoulos CP,
tion to the brain: the path to progressive multifocal Al-Aska AK, Obeid T, Daif AK. Neurological syndromes of
leukoencephalopathy. J Infect Dis 2002; 186: S180-S186 brucellosis. J Neurol Neurosurg Psychiatry 1988; 51: 1017–
Safdar A, Rubocki J, Horvath JA, Narayan K, Waldron RL. Fatal 1021
immune restoration disease in human immunodeficiency Bahemuka M, Babiker MA, Wright SF, Al Orainey I, Obeid T.
virus type 1-infected patients with progressive multifocal The pattern of infections in the nervous system in Riyadh:
leukoencephalopathy: impact of antiretroviral therapy-as- a review of 121 cases. Q J Med 1988; 68: 517–524
sociated immune reconstruction. Clin Infect Dis 2002; 35: Bashir R, Al-Kawi MZ, Harder EJ, Jinkins J. Nervous system bru-
1250–1257 cellosis: diagnosis and treatment. Neurology 1985; 35:
Seth P, Diaz F, Major EO. Advances in the biology of JC virus and 1576–1581
induction of progressive multifocal leukoencephalopathy. Belman AL,Coyle PK,Roque C,Cantos E.MR findings in children
J Neurovirol 2003; 9: 236–246 infected by Borrelia burgdorferi. Pediatr Neurol 1992; 8:
Shapiro RA, Mullane KM, Camras L, Flowers C, Sutton S. Clinical 428–431
and magnetic resonance imaging regression of progres- Bussone G, La Manita L, Grazzi L, Lamperti E, Salmaggi A,
sive multifocal leukoencephalopathy in an AIDS patient af- Strada L. Neurobrucellosis mimicking multiple sclerosis: a
ter intensive antiretroviral therapy. J Neuroimaging 2001; case report. Eur Neurol 1989; 29: 238–240
·
11: 336–339 Çiftçi E, Erden I, Akyar S. Brucellosis of the pituitary region: MRI.
Sima AAF, Finkelstein SD, McLachlan DR. Multiple malignant Neuroradiology 1998; 40: 383–384
astrocytomas in a patient with spontaneous progressive Coskun E, Züzer T,Yalçin N,Tahta K. Spinal extradural compres-
multifocal leukoencephalopathy. Ann Neurol 1983; 14: sion caused by granuloma of brucellosis. Scand J Infect Dis
183–188 1998; 30: 311–312
Steiger MJ, Tarnesby G, Gabe S, McLaughlin J, Schapira AHV. Galanakis E, Bourantas KL, Leveidotou S, Lapatsanis PD. Child-
Successful outcome of progressive multifocal leukoen- hood brucellosis in north-west Greece: a retrospective
cephalopathy with cytarabine and interferon. Ann Neurol analysis. Eur J Pediatr 1996; 155: 1–6
1993; 33: 407–411 Güven MB, Cirak B, Kutluhan A, Urgras S. Pituitary abscess sec-
Stohlman SA, Hinton DR. Viral induced demyelination. Brain ondairy to neurobrucellosis. J Neurosurg 1999; 90: 1142
Pathol 2001;11:92–106 Guvenc H, Kocabay K, Okten A, Bektas S. Brucellosis in a child
Sugimoto C, Ito D, Matsuda H, Saito H, Sakai H, Fujihara K, Itoya- complicated with multiple brain abscesses. Scand J Infect
ma Y, Yamada T, Kira J, Matsumoto R, Mori M, Nagashima K, Dis 1989; 21: 333–336
Yogo Y. Amplification of JC virus regulatory DNA sequences Martínez-Chamorro E, Muñoz A, Esparza J, Muñoz MJ, Gian-
from cerebrospinal fluid: diagnostic value for progressive gaspro E. Focal cerebral involvement by neurobrucellosis:
multifocal leukoencephalopathy. Arch Virol 1998; 143: pathological and MRI findings. Eur J Radiol 2002; 43: 28–30
249–262 McLean DR, Russell N, Khan MY. Neurobrucellosis: clinical and
Thurnher MM, Donovan Post MJ, Rieger A, Kleibl-Popov C, therapeutic features. Clin Infect Dis 1992; 15: 582–590
Loewe C, Schindler E.Initial and follow-up MR imaging find- Oliveri RL, Materna G, Focà A, Zappia M, Aguglia U, Quattrone
ings in AIDS-related progressive multifocal leukoen- A. Polyradiculoneuropathy with cerebrospinal fluid albu-
cephalopathy treated with highly active antiretroviral ther- minocytological dissociation due to neurobrucellosis. Clin
apy. AJNR Am J Neuroradiol 2001; 22: 977–984 Infect Dis 1996; 23: 833–834
Trotot PM Vazeux R, Yamashita HK, Sandoz-Tronca C, Mikol J, Piampiano P, McLeary M, Young LW, Janner D. Brucellosis: un-
Vedrenne C, Thiébaut JB, Gray F, Cikurel M, Pialoux G, Levil- usual presentations in two adolescent boys. Pediatr Radiol
lain R. MRI pattern of progressive multifocal leukoen- 2000; 30: 355–357
cephalopathy (PML) in AIDS. J Neuroradiol 1990; 17: 233– Samdani PG, Patil S. Neurobrucellosis. Indian Pediatr 2003; 40:
254 565–568
Weber T, Major EO. Progressive multifocal leukoencephalopa- Seidel G, Pardo CA, Newman-Toker D, Olivi A, Eberhart CG. Neu-
thy: molecular biology, pathogenesis and clinical impact. robrucellosis presenting as leukoencephalopathy. The role
Intervirology 1997; 40: 98–111 of cytotoxic T lymphocytes. Arch Pathol Lab Med 2003; 127:
Zachoval R, Hunstein W, Ho AD. Progressive multifocal e374-e377
leukoencephalopathy in a patient with Hodgkin’s disease. Shakir RA, Al-Din ASN, Araj GF, Lulu AR, Mousa AR, Saadah MA.
Blut 1980; 41: 451–454 Clinical categories of neurobrucellosis. A report on 19 cas-
es. Brain 1987; 110: 213–223
Sharif HS, Clark DC, Aabed MY, Haddad MC, Al Deeb SM,Yaqub
B, Al Moutaery KR. Granulomatous spinal infections: MR
83 Brucellosis imaging. Radiology 1990; 177: 101–107
Trifiletti RR,Restivo DA,Pavone P,Giuffrida S,Parano E.Diabetes
Akdeniz H, Irmak H, Anlar O, Demiroz AP. Central nervous insipidus in neurobrucellosis. Clin Neurol Neurosurg 2000;
system brucellosis: presentation, diagnosis and treatment. 102: 163–165
J Infect 1998; 36: 297–301 Young EJ. Utility of the enzyme-linked immunosorbent assay
Al Deeb SM, Yaqub BA, Sharif HS, Phadke JG. Neurobrucellosis: for diagnosing neurobrucellosis. Clin Infect Dis 1998; 26:
clinical characteristics, diagnosis, and outcome. Neurology 1481
1989; 39: 498–501 Zaidan R, Al Tahan A-R.Central venous thrombosis: a new man-
Al-Eissa YA. Unusual suppurative complications of brucellosis ifestation of neurobrucellosis. Clin Infect Dis 1999; 28:
in children. Acta Paediatr 1993; 82: 987–992 399–400
Al-Sous MW, Bohlega S, Al-Kawi MZ, Alwatban J, McLean DR.
Neurobrucellosis: clinical and neuroimaging correlation.
AJNR Am J Neuroradiol 2004; 25: 395–401
References and Further Reading 1029

84 Subacute Sclerosing Panencephalitis Dyken PR, Cunningham SC, Charles Ward L. Changing charac-
ter of subacute sclerosing panencephalitis in the United
Akdal G, Baklan B, Çakmakçi H, Kovanlikaya A. MRI follow-up States. Pediatr Neurol 1989; 5: 339–341
of basal ganglia involvement in subacute sclerosing pan- Ehrengruber MU, Ehler E, Billeter MA, Naim HY. Measles virus
encephalitis. Pediatr Neurol 2001; 24: 393–395 speads in rat hippocampal neurons by cell-to-cell contact
Alexander M, Singh S, Gnanamuthu C, Chandi S, Korah IP. Sub- and in a polarized fashion. J Virol 2002; 76: 5720–5728
acute sclerosing panencephalitis: CT and MR imaging in a Fayad MN, Yamout BI, Mroueh S. Alpha-interferon in the treat-
rapidly progressive case. Neurol India 1999; 47: 304–307 ment of subacute sclerosing panencephalitis. J Child Neu-
Alkan A, Sarac K, Kutlu R, Yakinci C, Sigirci A, Aslan M, Baysal T. rol 1997; 12: 486–488
Early- and late-state subacute sclerosing panencephalitis; Gascon GG. Subacute sclerosing panencephalitis. Sem Pediatr
chemical shift imaging and single-voxel MR spectroscopy. Neurol 1996; 3: 260–269
AJNR Am J Neuroradiol 2003; 24: 501–506 Gascon GG, International Consortium on Subacute Sclerosing
Allen IV, McQuaid S, McMaron J, Kirk J, McConnell R.The signif- Panencephalitis. Randomized treatment study of inosiplex
icance of measles virus antigen and genome distribution in versus combined inosiplex and intraventricular interferon-
the CNS in SSPE for mechanisms of viral spread and alpha in subacute sclerosing panencephalitis (SSPE): inter-
dymyelination.J Neuropathol Exp Neurol 1996;55:471–480 national multicenter study. J Child Neurol 2003; 18: 819–
Anlar B, Saatçi I, Köse G, Yalaz K. MRI findings in subacute scle- 827
rosing panencephalitis. Neurology 1996; 47: 1278–1283 Geller TJ, Vern BA, Sarwar M. Focal MRI findings in early SSPE.
Anlar B, Yalaz K, Oktem F, Kose G. Long-term follow-up of pa- Pediatr Neurol 1987; 3: 310–312
tients with subacute sclerosing panencephalitis treated Gokcil Z, Odabasi Z, Demirkaya S, Eroglu E, Vural O. a-Interfer-
with intraventricular a-interferon. Neurology 1997; 48: on and isoprinosine in adult-onset subacute sclerosing
526–528 panencephalitis. J Neurol Sci 1999; 162: 62–64
Anlar B, Yalaz K, Köse G, Saygi S. b-Interferon plus inosiplex in Gürer YKY, Kükner S, Sarica B. Intravenous g-globulin treatment
the treatment of subacute sclerosing panencephalitis. J in a patient with subacute sclerosing panencephalitis.Pedi-
Child Neurol 1998; 13: 557–559 atr Neurol 1996; 14: 72–74
Anlar B, Söylemenzoğlu F, Aysun S, Köse G, Belen D,Yalaz K.Tis- Haspolat S, Anlar B, Köse G, Coskun M, Yegin O. Interleukin-1b,
sue inflammatory response in subacute sclerosing panen- interleukin-1 receptor antagonist levels in patients with
cephalitis (SSPE). J Child Neurol 2001; 16: 895–900 subacute sclerosing panencephalitis and the effects of
Arora SC, Al-Tahan AR, Al-Zeer A, Al-Tahan F, Ozo CO, ur-Rah- different treatment protocols. J Child Neurol 2001; 16:
man N. Subacute sclerosing panencephalitis presenting 417–420
as acute disseminated encephalomyelitis: a case report. Hayashi M, Arai N, Satoh J, Suzuki H, Katayama K, Tamagawa K,
J Neurol Sci 1997; 146: 13–18 Morimatsu Y. Neurodegenerative mechanisms in subacute
Barthez Carpentier MA, Billard C, Maheut J, Jourdan ML, sclerosing panencephalitis. J Child Neurol 2002; 17: 725–
Degenne D, Ruchoux MM, Goudeau A, Santini JJ. Acute 730
measles encephalitis of the delayed type: neuroradiologi- Inoue T, Kira R, Nakao F, Ihara K, Bassuny WM, Kusuhara K, Nihei
cal and immunological findings. Eur Neurol 1992; 32: K,Takeshita K, Hara T. Contribution of the interleukin 4 gene
235–237 to susceptibility of subacute sclerosing panencephalitis.
Bismar J, Gascon GG, Vult von Steyern K, Bohlega S. Subacute Arch Neurol 2002; 59: 822–827
sclerosing panencephalitis; evaluation with CT and MR. Jin L, Beard S, Hunjan R, Brown DWG, Miller E. Characterization
AJNR Am J Neuroradiol 1996; 17: 761–772 of measles virus strains causing SSPE: a study of 11 cases.
Bohlega S, Al-Kawi MZ. Subacute sclerosing panencephalitis. J Neurovirol 2002; 8: 335–344
J Neuroimaging 1994; 4: 71–76 Johnson KP,Byington DP,Gaddis L.Subacute sclerosing panen-
Callebaut DP,Cras P,Martin JJ.Prolonged and atypical course in cephalitis. Adv Neurol 1974; 6: 77–86
some cases of subacute sclerosing panencephalitis. Acta Kai C, Yamanouchi K, Sakata H, Miyashita N, Takahashi H,
Neurol Belg 1997; 97: 39–44 Kobune F. Nucleotide sequences of the M gene of prevail-
Cathomen T, Naim HY, Cattaneo R. Measles viruses with altered ing wild measles viruses and a comparison with subacute
envelope protein cytoplasmic tails gain cell infusion com- sclerosing panencephalitis virus. Virus Genes 1996; 12:
petence. J Virol 1998; 72: 1224–1234 85–87
Cruzado D, Massery-Spicher V, Roux L, Delavelle J, Picard F, Kato Z, Siato K, Yamada M, Asano T, Kondo N. Proton magnetic
Haenggeli C-A. Early onset and rapidly progressive sub- resonance spectroscopy in a case of subacute sclerosing
acute sclerosing panencephalitis after congenital measles panencephalitis. J Child Neurol 2002; 17: 788–790
infection. Eur J Pediatr 2002; 161: 438–441 Koppel BS, Poon TP, Khandji A, Pavlakis SG, Pedley TA. Subacute
Dimova P, Bojinova V. Subacute sclerosing panencephalitis sclerosing panencephalitis and acquired immunodeficien-
with atypical onset: clinical, computed tomographic, and cy syndrome: role of electroencephalography and magnet-
magnetic resonance imaging correlations. J Child Neurol ic resonance imaging. J Neuroimaging 1996; 6: 122–125
2000; 15: 258–260 Lawrence DMP, Patterson CE, Gales TL, D’Orazio JL, Vuaghn M,
Dlugos DJ, Liu GT. Subacute sclerosing panencephalitis in an Rall GF. Measles virus spread between neurons requires cell
American-born adult. Clin Infect Dis 2001; 32: 173–174 contact but not CD46 expression, syncytium formation, or
Dyken PR. Subacute sclerosing panencephalitis. Neurol Clin extracellular virus production. J Virol 2000; 74: 1908–1918
1985; 3: 179–196 Lee WY, Cho WH, Kim SH, Kim HD, Kim IO. Acute encephalitis as-
Dyken PR. Neuroprogressive disease of post-infectious origin: sociated with measles: MRI features. Neuroradiology 2003;
a review of a resurging subacute sclerosing panencephali- 45: 100–106
tis (SSPE). Ment Retard Dev Disabl Res Rev 2001; 7: 217–225 Lewandowska E, Szpak Gm, Lechowicz W, Pasennik E, Sobczyk
W.Ultrastructural changes in neuronal and glial cells in sub-
acute sclerosing panencephalitis: correlation with disease
duration. Folia Neuropathol 2001; 39: 193–202
1030 References and Further Reading

Lewandowska E, Lechowicz W, Szpak GM, Sobczyk W. Quanta- Schneider-Schaulies J, Niewiesk S, Schneider-Schaulies S, ter
tive evaluation of intranuclear inclusions in SSPE: correla- Meulen V.Measles virus in the CNS: the role of viral and host
tion with disease duration. Folia Neuropathol 2001; 39: factors for the establishment and maintenance of a persis-
237–241 tent infection. J Neurovirol 1999; 5: 613–622
Lum GB,Williams JP, Dyken PR, Machen BC, Dotson PM, Harpen Senol U, Haspolat S, Cevikol C, Saatçi I. Subacute sclerosing pa-
MD, McLeod N. Magnetic resonance and CT imaging corre- nencephalitis: brain stem involvement in a peculiar pat-
lated with clinical status in SSPE. Pediatr Neurol 1986; 2: tern. Neuroradiology 2000; 42: 913–916
75–79 Takayama S, Iwasaki Y, Yamanouchi H, Sugai K, Takashima S,
Mawrin C, Lins H, Koenig B, Heinrichs T, Murayama S, Kirches E, Iwasaki A. Characteristic clinical features in a case of fulmi-
Boltze C, Dietzmann K. Spatial and temporal disease pro- nant subacute sclerosing panencephalitis. Brain Dev 1994;
gression of adult-onset subacte sclerosing panencephali- 16: 132–135
tis. Neurology 2002; 58: 1568–1571 Tomoda A, Shiraishi S, Hosoya M, Hamada A, Miike T. Combined
McCarron MO, McDonnell GV, Gibson JM. Rapid neurological treatment with interferon-alpha and ribavirin for subacute
deterioration in a 22-year-old man. Postgrad Med J 1997; sclerosing panencephalitis. Pediatr Neurol 2001; 24: 54–59
73: 344–347 Tsuchiya K,Yamauchi T, Furui S, Suda Y,Takenaka E. MR imaging
McQuaid S, Campbell S, Wallace IJC, Kirk J, Cosby SL. Measles vs CT in subacute sclerosing panencephalitis. AJNR Am J
virus infection and replication in undifferentiated and dif- Neuroradiol 1988; 9: 943–946
ferentiated human neuronal cells in culture. J Virol 198; 72: Tuncay R, Akman-Demir G, Gökyigit A, Eraksoy M, Barlas M,Tol-
5245–5250 un R, Gürsoy G. MRI in subacute sclerosing panencephalitis.
Miki K, Komase K, Mgone CS, Kawanishi R, Iijima M, Mgone JM, Neuroradiology 1996; 38: 636–640
Asuo PG, Alpers MP, Takasu T Mizutani T. Molecular analysis Voudris KA, Skardoutsou A, Hasiotou M, Theodoropoulos B,
of measles virus infection genome derived from SSPE and Vagiakou EA. Long-term findings on brain magnetic reso-
acute measles patients in Papua, New Guinea. J Med Virol nance imaging in acute encephalopathy with bilateral stri-
2002; 68: 105–112 atal necrosis associated with measles. J Child Neurol 2002;
Modi G, Campbell H, Bill P. Subacute sclerosing panencephali- 17: 776–777
tis: changes on CT scan during acute relapse. Neuroradio- Winer JB, Pires M, Kermode A, Ginsberg L, Rossor M. Resolving
logy 1989; 31: 433–434 MRI abnormalities with progression of subacute sclerosing
Norrby E, Kristensson K. Measles virus in the brain. Brain Res panencephalitis. Neuroradiology 1991; 33: 178–180
Bull 1997; 44: 213–220 Woelk CH, Pybus OG, Jin L, Brown DWG, Holmes EC. Increased
Ogura H, Ayata M, Hayashi K, Seto T, Matsuoka O, Hattori H, positive selection pressure in persistent (SSPE) versus acute
Tanaka K, Tanaka K, Takano Y, Murata R. Efficient isolation of measles virus infections. J Gen Virol 2002; 83: 1419–1430
subacte sclerosing panencephalitis virus from patient Yalaz K, Anlar B, Oktem F, Aysun S, Ustacelebi S, Gurcay O, Gur-
brains by reference to magnetic resonance and computed cay O, Gucuyener K, Renda Y. Intraventricular interferon and
tomographic images. J Neurovirol 1997; 3: 304–309 oral inosiplex in the treatment of subacute sclerosing pa-
Ohya T, Martinez AJ, Jabbour JT, Lemmi, Duenas DA. Subacute nencephalitis. Neurology 1992; 42: 488–491
sclerosing panencephalitis: correlation of clinical, neuro- Zilber N, Kahana E. Environmental risk factors for subacute
physiologic and neuropathologic findings. Neurology sclerosing panencephalitis (SSPE). Acta Neurol Scand 1998;
1974; 24: 211–218 98: 49–54
Özturk A, Gürses C, Baykan B, Gökyiğit A, Eraksoy M. Subacute
sclerosing panencephalitis: clinical and magnetic reso-
nance imaging evaluation of 36 patients. J Child Neurol
2002; 17: 25–29
85 Congenital and Perinatal
Panagariya A, Sureka RK, Aurora A. Current developments in Cytomegalovirus Infection
the management of subacute sclerosing panencephalitis.
J Assoc Physicians India 1998; 46: 218–222 Adler SP. Immunoprophylaxis against cytomegalovirus dis-
Patterson JB, Cornu TI, Redwine J, Dales S, Lewicki H, Holz A, ease. Scand J Infect Dis Suppl 1995; 99: 105–109
Thomas D, Billeter MA, Oldstone MBA. Evidence that the Adler SP. Current prospects for immunization against cy-
hypermutated M protein of a subacute sclerosing pan- tomegalovirus disease. Infect Agents Dis 1996; 85: 29–35
encephalitis measles virus actively contributes to the Ahlfors K, Ivarsson S-A, Harris S. Report on a long-term study of
chronic progressive CNS disease. Virology 2001; 291: 215– maternal and congenital cytomegalovirus infection in
225 Sweden. Review of prospective studies available in the lit-
Salvan AM, Confort-Gouny S, Cozzone PJ, Vion-Dury J, Chabrol erature. Scand J Infect Dis 1999; 31: 443–457
B, Manchini J. In vivo cerebral proton MRS in a case of sub- Alford CA, Stagno S, Pass RF, Britt WJ. Congenital and perinatal
acute sclerosing panencephalitis. J Neurol Neurosurg Psy- cytomegalovirus infections. Rev Infect Dis 1990; 12: 45–53
chiatry 1999; 66: 547–555 Allen RD, Pellett PE, Stewart JA, Koopmans M. Nonradioactive
Santoshkumar B, Radhakrishnan K. Substantial spontaneous PCR-enzyme-linked immunosorbent assay method for de-
long-term remission in subacute sclerosing panencephali- tection of human cytomegalovirus DNA. J Clin Microbiol
tis (SSPE). J Neurol Sci 1998: 154: 83–88 1995; 33: 725–728
Sawaiski Y, Yano Y, Watanabe Y, Takada G. Migratory basal gan- Anderson KS, Amos CS, Boppana S, Pass R. Ocular abnormali-
glia lesions in subacute sclerosing panencephalitis (SSPE): ties in congenital cytomegalovirus infection. J Am Optom
clinical implications of axonal spread. J Neurol Sci 1999; Assoc 1996; 67: 273–278
168: 137–140 Atkins JT, Demmler GJ, Williamson WD, McDonald JM, Istas AS,
Sawaishi Y, Abe T, Yano T, Ishikawa K, Takada G. SSPE following Buffone GJ. Polymerase chain reaction to detect cy-
neonatal measles infection. Pediatr Neurol 1999; 20: 63–65 tomegalovirus DNA in the cerebrospinal fluid of neonates
with congenital infection. J Infect Dis 1994; 169: 1334–1337
References and Further Reading 1031

Bale JR Jr, Bray PF, Bell WE. Neuroradiographic abnormalties in Damato EG, Winnen CW. Cytomegalovirus infection: prenatal
cogentical cytomegalovirus infection. Pediatr Neurol 1985; implications. J Obstet Gynecol Neonatal Nurs 2002; 31:
1: 42–47 86–92
Bale JR Jr, Sato Y, Eisert D. Progressive postnatal subependymal Daniel Y, Gull I, Reuben Peyser M, Lessing JB. Congenital cyto-
necrosis in an infant with congenital cytomegalovirus in- megalovirus infection. Eur J Obstet Gynecol Reprod Biol
fection. Pediatr Neurol 1986; 2: 367–370 1995; 63: 7–16
Bale JR Jr, Blackman JA, Sato Y. Outcome in children with symp- Darin N, Bergström T, Fast A, Kyllerman M. Clinical, seriological
tomatic congenital cytomegalovirus infection. J Child Neu- and PCR evidence of cytomegalovirus infection in the cen-
rol 1989; 4: 131–136 tral nervous system in infancy and childhood. Neuropedi-
Bale JR Jr, Zimmerman B, Dawson JD, Souza IE, Petheram SJ, atrics 1994; 25: 316–322
Murph JR. Cytomegalovirus transmission in child care Das VK. Aetiology of bilateral sensorineural hearing impair-
homes. Arch Pediatr Adolesc Med 1999; 153; 75–79 ment in children: a 10 year study. Arch Dis Child 1996; 74:
Barbi M, Binda S, Primache V, Caroppo S, Didò P, Guidotti P, Cor- 8–12
betta C, Melotti D. Cytomegalovirus DNA detection in Davis LE, Stewart JA, Rarey KE, McLaren LC. Recovery and prob-
Guthrie cards: a powerful tool for diagnosing congenital in- able persistence of cytomegalovirus in human inner ear
fection. J Clin Virol 2000; 17: 159–165 fluid without cochlear damage.Ann Otol Laryngol 1987; 96:
Barkovich AJ, Lindan CE.Congenital cytomegalovirus infection 380–383
of the brain: imaging analysis and embryologic considera- Demmler GJ. Infectious diseases society of America and Cen-
tions. AJNR Am J Neuroradiol 1994; 15: 703–715 ters for disease control. Summary of a workshop on surveil-
Boesch C, Issakainen J, Kewitz G, Kikinis R, Martin E, Boltshauser lance for congenital cytomegalovirus disease. Rev Infect
E. Magnetic resonance imaging of the brain in congenital Dis 1991; 13: 315–329
cytomegalovirus infection. Pediatr Radiol 1989; 19: 91–93 Demmler GJ. Congenital cytomegalovirus infection and dis-
Boom R, Sol C,Weel J, Gerrits Y, de Boer M, Ertheim-van Dillen P. ease. Adv Pediatr infect Dis 1996; 11: 135–162
A highly sensitive assay for detection and quantitation of De Vries LS, Gunardi H, Barth PG, Bok LA, Verboon-Maciolek
human cytomegalovirus DNA in serum and plasma by PCR MA, Groenendaal F.The spectrum of cranial ultrasound and
and electrochemiluminescence. J Clin Microbiol 1999; 37: magnetic resonance imaging abnormalities in congenital
1489–1497 cytomegalovirus infection.Neuropediatrics 2004; 35:113–9
Boppana S, Amos C, Britt W, Stagno S, Alford C, Pass R. Late on- Eggers M, Metzger C, Enders G. Differentiation between acute
set and reactivation of chorioretinitis in children with con- primary and recurrent human cytomegalovirus infection in
genital cytomegalovirus infection. Pediatr Infect Dis J 1994; pregnancy, using a microneutralization assay. J Med Virol
13: 1139–1142 1998; 56: 351–358
Boppana SB,Pass RF,Britt WJ,Stagno S,Alford CA.Symptomatic Fischler B, Rodensjö P, Nemeth A, Forsgren M, Lewensohn-
congenital cytomegalovirus infection: neonatal morbidity Fuchs I. Cytomegalovirus DNA detection on Guthrie cards
and mortality. Pediatr Infect Dis J 1992; 11: 93–99 in patients with prenatal cholestasis. Arch Dis Child Fetal
Boppana SB, Fowler KB, Viad Y, Hedlund G, Stagno S, Britt WJ, Neonat Ed 1999; 80: F130-F134
Pass RF. Neuroradiographic findings in the newborn period Fowler KB, McCollister FP, Dahle AJ, Boppana S, Britt WJ, Pass RF.
and long-term outcome in children with symptomatic con- Progressive and fluctuating sensorineural hearing loss in
genital cytomegalovirus infection. Pediatrics 1997; 99: children with asymptomatic congenital cytomegalovirus
409–414 infection. J Pediatr 1997; 130: 624–630
Boppana SB, Fowler KB, Britt WJ, Stagno S, Pass RF. Sympto- Fowler SL. A light in the darkness: predicting outcomes for
matic congenital cytomegalovirus infection in infants born congenital cytomegalovirus infections. J Pediatr 2000; 137:
to mothers with preexisting immunity to cytomegalovirus. 4–6
Pediatrics 1999; 104: 55–60 Fritschy J-M, Brandner S, Aguzzi A, Koedood M, Lüscher B,
Boppana SB,Rivera LB,Fowler KB,Mach M,Britt WJ.Intrauterine Mitchell PJ.Brain cell type specification and gliosis-induced
transmission of congenital cytomegalovirus to infants of activation of the human cytomegalovirus immediate-early
women with preconceptional immunity. N Engl J Med promotor in transgenic mice. J Neurosci 1996; 16: 2275–
2001; 344: 1366–1371 2282
Brown HL, Abernathy MP. cytomegalovirus infection. Semin Gaytant MA, Steegers EAP, Semmekrot BA, Merkus HMMW,
Perinatol 1998; 22: 260–266 Galama JMD. Congenital cytomegalovirus infection: review
Bryant P, Morley C, Garland S, Curtis N. Cytomegalovirus trans- of the epidemiology and outcome. Obstet Gynecol Surv
mission for breast milk in premature babies: does it matter? 2002; 67: 245–256
Arch Dis Child Fetal Neonatal Ed 2002; 87: F75-F77 Gomes AL, Vieira JP, Saldanha J. Non-progressive leuko-
Ceballos R, Ch’ien LT, Whitley RJ, Brans YW. Cerebellar hypopla- encephalopathy with bilateral temporal cysts. Eur J Pediatr
sia in an infant with congenital cytomegalovirus infection. Neurol 2001; 5: 121–125
Pediatrics 1976; 57: 155–157 Grangeot-Keros L, Cointe D. Diagnosis and prognostic markers
Cinque P, Marenzi R, Ceresa D. Cytomegalovirus infections of of HCMV infection. J Clin Virol 2001; 21: 213–221
the nervous system. Intervirology 1997; 40: 85–97 Grant EG, Williams AL, Schellinger D, Slovis TL. Intracranial cal-
Conboy TJ, Pass RF, Stagno S, Alford CA, Myers GJ, Britt WJ, cification in the infant and neonate: evaluation by sono-
McCollister FP, Summers MN, McFarland CE, Boll TJ. Early graphy and CT. Radiology 1985; 157: 63–68
clinical manifestations and intellectual outcome in children Griffith BP, Booss J. Neurological infections of the fetus and
with symptomatic congenital cytomegalovirus infection. newborn. Neurol Clin 1994; 12: 541–564
J Pediatr 1987; 111: 343–348 Hagay ZJ, Biran G, Ornoy A, Reece EA. Congenital cyto-
Connolly PK, Jerger S, Williamson WD, Smith RJH, Demmler G. megalovirus infection: a long-standing problem still seek-
Evaluation of higher-level auditory function in children ing a solution. Am J Obstet Gynecol 1996; 174: 241–245
with asymptomatic congenital cytomegalovirus infection.
Am J Otol 1992; 13: 185–193
1032 References and Further Reading

Haginoya K, Ohura T, Kon K,Yagi T, Sawaishi Y, Ishii KK, Funato T, Mahlm G, Gröndahl EH, Lewensohn-Fuchs I. Congenital cyto-
Higano S,Takahashi S, Iinuma K. Abnormal white matter le- megalovirus infection: a retrospective diagnosis in a child
sions with sensorineural hearing loss caused by congenital with pachygyria. Pediatr Neurol 2000; 22: 407–408
cytomegalovirus infection: retrospective diagnosis by PCR Malinger G, Lev D, Zahalka N, Aroia ZB,Watemberg N, Kidron D,
using Guthrie cards. Brain Dev 2002; 24: 710–714 Sira LB, Lerman-Sagie T. AJNR Am J Neuroradiol 2003; 24:
Hayward JC, Titelbaum DS, Clancy RR, Zimmerman RA. 28–32
Lissencephaly-pachygyria associated with congenital cy- Marques Dias MJ, Harmant-van Rijckevorsel G, Landrieu P, Lyon
tomegalovirus infection. J Child Neurol 1991; 6: 109–114 G. Prenatal cytomegalovirus disease and cerebral micro-
Istas AS, Demmler GJ, Dobbins JG, Stewart JA. Surveillance for gyria: evidence for perfusion failure, not disturbance of his-
congenital cytomegalovirus disease: a report from the Na- togenesis, as the major cause of fetal cytomegalovirus en-
tional Congenital Cytomegalovirus Disease Registry. Clin cephalopathy. Neuropediatrics 1984; 15: 18–24
Infect Dis 1995; 20: 665–670 Martinez A, Castro A, Gil C, Perez C. Recent strategies in the de-
Jauniaux E, Jurkovic D, Gulbis B, Liesnard C, Lees C, Campbell S. velopment of new human cytomegalovirus inhibitors. Med
Materno-fetal immunoglobulin transfer and passive immu- Res Rev 2001; 3: 227–244
nity during the first trimester of human pregnancy. Hum Michaels MG, Greenberg FP, Sabo DL, Wald ER. Treatment of
Reprod 1995; 10: 3297–330 children with congenital cytomegalovirus infection with
Johansson PJH, Jönsson M, Ahlfors K, Ivarsson SA, Svanberg L, ganciclovir. Pediatr Infect Dis J 2003; 22: 504–508
Guthenberg C. Retrospective diagnostics of congenital cy- Morris DJ, Sims S, Chiswick M, Das VK, Newton VE. Symptomatic
tomegalovirus infection performed by polymerase chain congenital cytomegalovirus infection after maternal recur-
reaction in blood stored on filter paper. Scand J Infect Dis rent infection. Pediatr Infect Dis J 1994; 13: 61–64
1997; 29: 465–468 Murph JR, Souza IE, Dawson JD, Benson P, Petheram SJ, Pfab D,
Jones CA, Isaacs D. Predicting the outcome of symptomatic Gregg A, O’Neill ME, Zimmerman B, Bale JF Jr. Epidemiology
congenital cytomegalovirus infection. J Paediatr Child of congenital cytomegalovirus infection: maternal risk fac-
Health 1995; 31: 70–71 tors and molecular analysis of cytomegalovirus strains. Am
Kimberlin DW,Lin C-Y,Sanches PJ,Demmler GJ,Danker W,Shel- J Epidemol 1998; 147: 940–947
ton M,Jacobs RF,Vaudry W,Pass RF,Kiell JM,Soong S-J,Whit- Mussi-Pinhata MM, Yamamoto AY, Figueiredo LTM, Cervi MC,
ley RJ. Effect of ganciclovir therapy on hearing in sympto- Duarte G. Congenital and perinatal cytomegalovirus infec-
matic congenital cytomegalovirus disease involving the tion in infants born to mothers infected with human im-
central nervous system: a randomized, controlled trial. munodeficiency virus. J Pediatr 1998; 132: 285–290
J Pediatr 2003; 143: 16–25 Negishi H, Yamada H, Hirayama E, Okuyama K, Sagawa T, Mat-
Koedood M, Fichtel A, Meier P, Mitchell PJ. Human cyto- sumoto Y, Fujimoto S. Intraperitoneal administration of
megalovirus (HCMV) immediate-early enhancer/promoter cytomeglovirus hyperimmunoglobulin to the cytomegalo-
specificity during embryogenesis defines target tissues virus-infected fetus. J Perinatol 1998; 18: 466–469
of congenital HCMV infection. J Virol 1995; 69: 2194– Nelson CT, Istas AS, Wilkerson MK, Demmler GJ. PCR detection
2207 of cytomegalovirus DNA in serum as a diagnostic test for
Kohyama J, Kajiwara M, Shimohira M, Iwakawa Y, Okawa H. congenital cytomegalovirus infection. J Clin Microbiol
Human cytomegalovirus DNA in cerebrospinal fluid. Arch 1995; 33: 3317–3318
Dis Child 1994; 71: 414–418 Noyola DE, Demmler GJ, Williamson WD, Griesser C, Sellers S,
Koi H, Zhang J, Parry S.The mechanisms of placental viral infec- Llorente A, Littman T, Williams S, Jarrett L, Yow MD. Cyto-
tion. Ann NY Acad Sci 2001; 943: 148–156 megalovirus urinary excretion and long term outcome in
Koopmans M, Sánchez-Martinéz D, Patton J, Stewart J. Evalua- children with congenital cytomegalovirus infection. Pedi-
tion of antigen and antibody detection in urine specimens atr Infect Dis J 2000; 19: 505–510
from children with congenital human cytomegalovirus in- Noyola DE, Demmler GJ, Nelson CT, Griesser C, Williamson WD,
fection. J Med Virol 1995; 46: 321–328 Atkins JT, Rozelle J,Turcich M, Llorente AM, Sellers-Vinson S,
Lagasse N, Dhooge I, Govaert P. Congenital CMV-infection and Reynolds A, Bale JF Jr, Gerson P,Yow MD. Early predictors of
hearing loss. Acta Otolaryngol Belg 2000; 54: 431–436 neurodevelopmental outcome in symptomatic congenital
Lamy ME, Mulongo KN, Gadisseux J-F, Lyon G, Gaudy V, van cytomegalovirus infection. J Pediatr 2001; 138: 325–331
Lierde M. Prenatal diagnosis of fetal cytomegalovirus infec- Numazaki K. Human cytomegalovirus infection of breast milk.
tion. Am J Obstet Gynecol 1992; 166: 91–94 FEMS Immunol Med Microbiol 1997; 18: 91–98
Lazzarotto T, Guerra B, Spezzacatena P, Varani S, Gabrielli L, Namazaki K, Chiba S. Current aspects of diagnosis and treat-
Pradelli P, Rumpianesi F, Banzi C, Bovicelli L, Landini MP. Pre- ment of cytomegalovirus infections in infants. Clin Diagn
natal diagnosis of congenital cytomegalovirus infection. Virol 1997; 8: 169–181
J Clin Microbiol 1998; 36: 3540–3544 Namazaki K, Asanuma H, Ikehata M, Chiba S. Detection of cyto-
Lazzarotto T, Spezzacatena P, Varani S, Gabrielli L, Pradelli P. kines and cytomegalovirus DNA in serum as test for con-
Guerra B, Landini MP. Anticytomegalovirus (anti-CMV) im- genital infection. Early Hum Dev 1998; 52: 43–48
munoglobulin G avidity in identification of pregnant Olivier M, Lenard HG, Aksu F, Gärtner J. A new leukoen-
woman at risk of transmitting congenital CMV infection. cephalopathy with bilateral anterior temporal lobe cysts.
Clin Diagn Lab Immunol 1999; 6: 127–129 Neuropediatrics 1998; 29: 225–228
Lazzarotto T Varani S, Gabrielli L, Spezzacatena P, Landini MP. Pass RF. Immunization strategy for prevention of congenital
New advances in the diagnosis of congenital cytomegalo- cytomegalovirus infection. Infect Agents Dis 1996; 5:
virus infection. II. Diagnostics and antiviral therapy. Intervi- 240–244
rology 1999; 42: 390–397 Pass RF. Cytomegalovirus infection. Pediatr Rev 2002; 23:
Lazzarotto T,Varani S, Guerra B, Nicolosi A, Lanari M, Landini MP. 163–169
Prenatal indicators of congenital cytomegalovirus infec- Peckham C, Tookey P, Logan S, Giaquinto C. Screening options
tion. J Pediatr 2000; 137: 90–95 for prevention of congenital cytomegalovirus infection.
J Med Screen 2001; 8: 119–124
References and Further Reading 1033

Peckham CS, Coleman JC, Hurley R, Chin KS, Henderson K, Sugita K, Ando M, Makino M, Takanashi J, Fujimoto N, Niimi H.
Preece PM. Cytomegalovirus infection in pregnancy: pre- Magnetic resonance imaging of the brain in congenital
liminary findings from a prospective study. Lancet 1983; I: rubella virus and cytomegalovirus infections. Neuroradio-
1352–1355 logy 1991; 33: 239–242
Poland SD, Bambrick LL, Dakaban GA, Rice GPA. The extent of Titelbaum DS Hayward JC, Zimmerman RA. Pachygyriclike
human cytomegalovirus replication in primary neurons is changes: topographic appearance at MR imaging ant CT
dependent on host cell differentiation. J Infect Dis 1994; and correlation with neurologic status. Radiology 1989;
170: 1267–1271 173: 663–667
Raine CS, Fields BN. Neurotropic viruses and the developing van der Knaap MS, Vermeulen G, Barkhof F, Hart AAM, Loeber
brain. N Y State Med 1973; 73: 1169–1179 JG, Weel JFL. A pattern of white matter abnormalities on
Ramenghi LA, Domizio S, Quartulli L, Sabatino G. Prenatal MRI: polymerase chain reaction on Guthrie cards to link the
pseudocysts of the germinal matrix in preterm infants. pattern with congenital CMV.Radiology 2004; 230:529–536
J Clin Ultrasound 1997; 25: 169–173 Weinberg A. The role of immune reconstruction in cyto-
Revello MG, Baldanti F, Furione M, Sarasini A, Percivalle E, Zavat- megalovirus infection. Biodrugs 2002; 16: 89–95
toni M, Gerna G. Polymerase chain reaction for prenatal Williamson WD, Percy AK, Yow MD, Gerson P, Catlin FI, Koppel-
diagnosis of congenital human cytomegalovirus infection. man ML,Thurber S. Asymptomatic congenital cytomegalo-
J Med Virol 1995; 47: 462–466 virus infection. Audiologica, Neuroradiologic, and neurode-
Revello MG, Sarasini A, Zavattoni M, Baldanti F, Gerna G. Im- velopmental abnormalities during the first year. Am J Dis
proved prenatal diagnosis of congenital human cyto- Child 1990; 144: 1365–1368
megalovirus infection by a modified nested polymerase Williamson WD, Demmler GJ, Percy AK, Catlin FI. Progressive
chain reaction. J Med Virol 1998; 56: 99–103 hearing loss in infants with asymptomatic congenital cyto-
Schackelford GD, Fulling KH, Glasier CM Maj. Cysts of the megalovirus infection. Pediatrics 1992; 90: 862–866
subependymal germinal matrix: sonographic demonstra- Yamashita Y, Outani Y, Kawano Y, Horikwawa M, Matsuishi T,
tion with pathologic correlation. Radiology 1983; 149: Hashimoto T. Clinical analysis and short-term progress of
117–121 neonates with subependymal cysts. Pediatr Neurol 1990; 6:
Schendel DE. Infection in pregnancy and cerebral palsy. J Am 375–378
Med Womens Assoc 2001; 56:105–108 Zaia JA, Lang DJ. Cytomegalovirus infection of the fetus and
Schneeberger PM, Groenendaal F, de Vries LS, van Loon AM, neonate. Neurol Clin 1984; 2: 387–410
Vroom TM. Variable outcome of a congenital cytomegalo-
virus infection in a quadruplet after primary infection of
the mother during pregnancy. Acta Paediatr 1994; 83:
986–989
86 Whipple Disease
Schopper K, Lauber E, Krech U. Congenital cytomegalovirus in-
fection in newborn infants of mothers infected before Anderson M. Neurology of Whipple’s disease. J Neurol Neuro-
pregnancy. Arch Dis Child 1978; 53: 536–539 surg Psychiatry 2000; 68: 2–5
Schwebke K, Henry K, Balfour HH Jr, Olson D, Crane RT, Jardan Brändle M, Ammann P, Spinas GA, Dutley F, Galeazzi RL, Schmid
C. Clinical and laboratory observations. Congenital cyto- C, Altwegg M. Relapsing Whipple’s disease presenting with
megalovirus infection as a result of nonprimary cyto- hypopituitarism. Clin Endocrinol 1999; 50: 399–403
megalovirus disease in a mother with acquired immunod- Brown AP, Lane JC, Murayama S, Vollmer DG.Whipple’s disease
eficiency syndrome. J Pediatr 1995; 126: 293–295 presenting with isolated neurological symptoms. J Neuro-
Sener RN. Schizencephaly and congenital cytomegalovirus in- surg 1990; 73: 623–627
fection. J Neuroradiol 1998; 25: 151–152 Carella F, Valla P, Bernardi G, Parente F, Costa A, Lodrini S. Cere-
Shaw C-M, Alvord EC Jr. Subependymal germinolysis. Arch bral Whipple’s disease: clinical and cerebrospinal fluid find-
Neurol 1974; 31: 374–381 ings. Ital J Neurol Sci 1998; 19: 101–105
Soussotte C, Maugey-Laulom B, Charles D, Diard F. Contribu- Coene De B, Gilliard C, Indekeu P, Duprez T,Trigaux JP.Whipple’s
tion of transvaginal ultrasonography and fetal cerebral MRI disease confined to the central nervous system. Neuroradi-
in a case of congenital cytomegalovirus infection. Fetal Di- ology 1996; 38: 325–327
agn Ther 2000; 15: 219–223 Cooper GS,Blades EW,Remler BF,Salata RA,Bennert KW,Jacobs
Souza IE, Gregg A, Pfab D, Dawson JD, Benson P. O’Neill ME, GH. Central nervous system Whipple’s disease: relapse dur-
Murph JR, Petheram SJ, Bale FJ Jr. Cytomegalovirus infec- ing therapy with trimethoprim-sulfamethoxazole and re-
tion in newborns and their family members: polymerase mission with cefixime. Gastroenterology 1994; 106: 782–
chain reaction analysis of isolates. Infection 1997; 25: 786
144–149 Davion T, Rosat P, Sevestre H, Desablens B, Debussche C, Dela-
Stamos JK, Rowley AH. Timely diagnosis of congenital infec- marre J, Capron JP. MR Imaging of CNS relapse of Whipple
tions. Pediatr Clin North Am 1994; 41: 1017–1033 disease. J Comput Assist Tomogr 1990; 14: 815–817
Steinlin M. Non-progressive congenital ataxias. Brain Dev Duprez TPJ, CrandinCBG, Bonnier C,Thauvoy CW, Gadisseux JF,
1998;20:199–208 Dutrieux JL, Evrard P. Whipple disease confined to the cen-
Steinlin M, Zürrer M, Marin E, Boesch CH, Largo RH, Boltshauser tral nervous system in childhood. AJNR Am J Neuroradiol
E. Contribution of magnetic resonance imaging in the eval- 1996; 17: 1589–1591
uation of microcephaly. Neuropediatrics 1991; 22: 184–189 Erdem E, Carlier R, Delvalle A, Caquet R, Etienne JP, Doyon D.
Steinlin MI, Nadal D, Eich GE, Martin E, Boltshauser EJ. Late in- Gadolinium-enhanced MRI in cerebral Wipple’s disease.
trauterine cytomegalovirus infection: Clinical and neu- Neuroradiology 1993; 35: 581–583
roimaging findings. Pediatr Neurol 1996;15:249–253 Gérard A, Sarrot-Reynauld F, Liozon E, Cathebras P, Besson G,
Stronati M, Revello MG, Cerbo RM, Furione M, Rondini G, Gerna Robin C, Vighetto A, Mosnier JF, Durieu I, Durand DV, Rous-
G. Ganciclovir therapy of congenital human cytomegalo- set H. Neurologic presentation of Whipple disease. Medi-
virus hepatitis. Acta Paediatr 1995; 84: 340–341 cine 2002; 81: 443–457
1034 References and Further Reading

Lynch T, Odel J, Fredricks DN, Louis ED, Forman S, Rotterdam H, Cohen JA, Fisher RS, Brigell MG, Peyster RG, Sze G.The potential
Fahn S, Redman DA. Polymerase chain reaction-based de- for vigabatrin-induced intramyelinic edema in humans.
tection of Tropheryma whippelii in central nervous system Epilepsia 2000; 41: 148–157
Whipple’s disease. Ann Neurol 1997; 42: 120–124 Crapper McLachlan DR, De Boni U. Aluminium in human brain
Manzel K, Tranel D, Cooper G. Cognitive and behavioral abnor- disease – an overview. Neurotoxicology 1980; 1: 3–16
malities in a case of central nervous system Whipple dis- Davis LE, Kornfeld M, Mooney HS, Fiedler KJ, Haaland KY, Orri-
ease. Arch Neurol 2000; 57: 399–403 son WW, Cernichiari E, Clarkson TW. Methylmercury poison-
Mendel E, Khoo LT, Go JL, Hinton D, Zee CS, Apuzzo MLJ. Intra- ing: long-term clinical, radiological, toxicological, and
cerebral Whipple’s disease diagnosed by stereotactic biop- pathological studies of an affected family.Ann Neurol 1994;
sy: a case report and review of the literature. Neurosurgery 35: 680–688
1999; 44: 203–209 Del Amo M, Berenguer J, Pujol T, Mercader JM. MR in
Messori A, Di Bella P, Polonara G, Logullo F, Pauri P, Haghigh- trichloroethane poisoning. AJNR Am J Neuroradiol 1995;
ipour R, Salvolini U. An unusual spinal presentation of 17: 1180–1182
Whipple disease. AJNR Am J Neuroradiol 2001; 22: Díez-Tejedor E, Frank A, Gutiérrez M, Barreiro P. Encephalopa-
1004–1008 thy and biopsy-proven cerebrovascular inflammatory
Peters G, du Plessis DG, Humphrey PR. Cerebral Whipple’s dis- changes in a cocaine abuser. Eur J Neurol 1998; 5: 103–107
ease with a stroke-like presentation and cerebrovascular Dobbing J.Vulnerable periods in developing brain. In: Davison
pathology. J Neurol Neurosurg Psychiatry 2002; 73: 336– AN, Dobbing J, eds. Applied neurochemistry. Oxford: Black-
339 well, 1968, pp 287–316
Raoult D, Birg ML, Scola la B, Fournier PE, Enea M, Lepidi H, Roux Donaldson J.The physiopathologic significance of manganese
V, Piette JC,Vandenesch F,Vital-Durand D, Marrie TJ. Cultiva- in brain: its relation to schizophrenia an neurodegenerative
tion of the bacillus of Whipple’s disease. N Engl J Med 2000; disorders. Neurotoxicology 1987; 8: 451–462
342: 620–625 Escobar A, Aruffo C. Chronic thinner intoxication: clinico-
Schnider P,Trattnig S, Kollegger H, Auff E. MR of cerebral Whip- pathologic report of a human case.J Neurol Neurosurg Psy-
ple disease. AJNR Am J Neuroradiol 1995; 16: 1328–1329 chiatry 1980; 43: 986–994
Scully RE, Mark EJ, McNeely WF, Ebeling SH, Phillips LD. Case Filley CM. Toxic leukoencephalopathy. Clin Neuropharmacol
records of the Massachusetts General Hospital. N Engl J 1999; 22: 249–260
Med 1997; 37: 1612–1620 Filley CM, Heaton RK, Rosenberg NL.White matter dementia in
Süzer T, Demirkan N, Tahta K, Coskun E, Cetin B. Whipple’s dis- chronic toluene abuse. Neurology 1990; 40: 532–534
ease confined to the central nervous system: case report Gacouin A, Lavoue S, Signouret T, Person A, Dinard MD, Shpak
and review of the literature. Scand J Infect Dis 1999; 31: N, Thomas R. Reversible spongiform leukoencephalopathy
411–414 after inhalation of heroin. Intensive Care Med 2003; 29:
Swartz MN. Whipple’s disease – past, present, and future. 1012–1015
N Engl J Med 2000; 342: 648–650 Galle P, Meyrignac C, Heine P. Presence of high concentrations
Verhagen WIM, Huygen PLM, Dalman JE, Schuurmans MMJ. of aluminium in neurons of a patient dying from a rapidly
Whipple’s disease and the central nervous system: a case developing dementia. C R Acad Sci III 1984; 299: 535–539
report and a review of the literature. Clin Neurol Neurosurg Gaul HP,Wallace CJ, Auer RN, Fong TC.MR Findings in methanol
1996; 98: 299–304 intoxication. AJNR Am J Neuroradiol 1995; 16: 1783–1786
Wroe SJ, Pires M, Harding B, Youl BD, Shorvon S. Whipple’s dis- Greger JL. Nutrition versus toxicology of manganese in hu-
ease confined to the CNS presenting with multiple intrac- mans: evaluation of potential biomarkers. Neurotoxicology
erebral mass lesions. J Neurol Neurosurg Psychiatry 1991; 1999; 20: 205–212
54: 989–992 Heier LA, Carpanzano CR, Mast J, Brill PW, Winchester P, Deck
MDF. Maternal cocaine abuse: the spectrum of radiologic
abnormalities in the neonatal CNS. AJNR Am J Neuroradiol
1991; 12: 951–956
87 Toxic Encephalopathies Henry JA. Ecstasy and the dance of death. Severe reactions are
unpredictable. BMJ 1992; 304: 5–6
Andreas K, Blaschke M, Bergsträsser E, Fisher H-D, Schmidt J. Hormes JT, Filley CM, Rosenberg NL. Neurologic sequelae of
Neurochemical characterization of hexachlorophene in- chronic solvent vapor abuse. Neurology 1986; 36: 698–702
duced brain oedema. Arch Toxicol 1991; 14: 30–32 Hsu HH, Chen CY, Chen FH, Lee CC, Chou TY, Zimmerman RA.
Ashikaga R, Araki Y, Miura K, Ishida O. Cranial MRI in chronic Optic atrophy and cerebral infarcts caused by methanol in-
thinner intoxication. Neuroradiology 1995; 37: 443–444 toxication: MRI. Neuroradiology 1997; 39: 192–194
Aydin K, Sencer S, Demir T, Ogel K,Tunaci A, Minareci O. Cranial Huang CC, Chu CC, Chen RS, Lin SK, Shih TS. Chronic carbon
MR findings in chronic toluene abuse by inhalation. AJNR disulfide encephalopathy. Eur Neurol 1996; 36: 364–368
Am J Neuroradiol 2002; 23: 1173–1179 Huang CC, Chu CC, Chu NS,Wu TN. Carbon disulfide vasculopa-
Brown GK. Metabolic disorders of embryogenesis. J Inherit thy: a small vessel disease. Cerebrovasc Dis 2001; 11: 245–
Metab Dis 1994; 17: 448–458 250
Brust JCM. Neurologic complications of substance abuse. Kamran S, Bakshi R. MRI in chronic toluene abuse: low signal in
J AIDS 2002; 31: S29-S34 the cerebral cortex on T2-weighted images. Neuroradiolo-
Burg JR, Gist GL. Health effects of environmental contaminant gy 1998; 40: 519–521
exposure: an intrafile comparison of the Trichloroethylene Keogh CF, Andrews GT, Spacey SD, Forkheim KE, Graeb DA.
Subregistry. Arch Environ Health 1999; 54: 231–241 Neuroimaging features of heroin inhalation toxicity:“chas-
Cho SK, Kim RH, Yim SH, Tak SW, Lee YK, Son MA. Long-term ing the dragon”. AJR Am J Roentgenol 2003; 180: 847–850
neuropsychological effects and MRI findings in patients
with CS2 poisoning. Acta Neurol Scand 2002; 106: 269–275
References and Further Reading 1035

Kim JH, Chang KH, Song IC, Kim KH, Kwon BJ, Kim HC, Kim JH, Sedman AB, Wilkeling GN, Warady BA, Lum GM, Alfrey AC. En-
Han MH. Delayed encephalopathy of acute carbon monox- cephalopathy in childhood secondary to aluminium toxici-
ide intoxication: diffusivity of cerebral white matter lesions. ty. J Pediatr 1984; 105: 836–838
AJNR Am J Neuroradiol 2003; 24: 1592–1597 Tan TP, Algra PG, Valk J, Wolters EC.Toxic leukoencephalopathy
Kimbrough RD, Gaines TB. Hexachlorophene effects on the rat after inhalation of poisoned heroin: MR findings. AJNR Am
brain. Study of high doses by light and electron micro- J Neuroradiol 1994; 15: 175–178
scopy. Arch Environ Health 1971; 23: 114–118 Tokuomi H, Okajima T, Kanai J,Tsunoda M, Ichiyasu Y, Misumi H,
Kondo K. Congenital Minamata disease: warnings from Japan’s Shimomura K, Takaba M. Minamata disease – an unusual
experience. J Child Neurol 2000; 15: 458–464 neurological disorder occurring in Minamata, Japan.
Kornfeld M, Moser AB, Moser HW, Kleinschmidt-DeMasters BK, Kumamoto Med J 1691; 14: 47–64
Nolte K, Phelps A. Solvent vapor abuse leukoencephalopa- Valk J, van der Knaap MS. Toxic encephalopathy. AJNR Am J
thy. Comparison to adrenoleukodystrophy. J Neuropathol Neuroradiol 1992; 13: 747–760
Exp Neurol 1994; 53: 389–398 Valk J, van der Knaap MS. Selective vulnerability in toxic
Korogi Y,Takahashi M, Shinzato J, Okajima T.MR findings in sev- encephalopathies and metabolic disorders. Riv Neuro-
en patients with organic mercury poisoning (Minamata radiol 1996; 9: 749–760
disease). AJNR Am J Neuroradiol 1994; 15: 1575–1578 Vella S, Kreis R, Lovblad KO, Steinlin M. Acute leuko-
Korogi Y,Takahashi M, Okajima T, Eto K. MR findings in Minama- encephalopathy after inhalation of a single dose of heroin.
ta disease – organic mercury poisoning. J Magn Reson Neuropediatrics 2003; 34: 100–104
Imaging 1998; 3: 308–316 Wolters EC, Stam FC, Lousberg RJ, van Wijngaarden CK, Ren-
Kriegstein AR, Shungru DC, Millar WS, Armitage BA, Brust JC, gelink H, Schipper MEI, Verbeten B. Leucoencaphalopathy
Chilllrud S, Goldman J, Lynch T. Leukoencephalopathy and after inhaling “heroin” pyrolysate. Lancet 1982; II, 1233–
raised brain lactate from heroin vapor inhalation (“chasing 1237
the dragon”) Neurology 1999; 53: 1765–1773 Xiong L, Matthes JD, Jinkins JR. MRI imaging of “spray heads”:
Ku M-C, Huang C-C, Kuo H-C,Yen T-C, Chen C-J, Shih T-S, Chang toluene abuse via aerosol paint inhalation. AJNR Am J
H-Y. Diffuse white matter lesions in carbon disulfide intoxi- Neuroradiol 1993; 14: 1195–1199
cation: microangiopathy or demyelination. Eur Neurol Yamanouchi N, Okada S-I, Kodama K, Hirai S, Sekine H, Muraka-
2003; 50: 220–224 mi A, Komatsu N, Sakamoto T, Sato T.White matter changes
Kuteifan K, Oesterlé H, Tajahmady T, Gutbub AM, Lapatte G. caused by chronic solvent abuse. AJNR Am J Neuroradiol
Necrosis and haemorrhage of the putamen in methanol 1995; 16: 1643–1649
poisoning shown on MRI. Neuroradiology 1998; 40:
158–160
Lee JC, Bakay L. Ultrastructural changes in the edematous cen-
tral nervous system. Arch Neurol 1965; 13: 48–57
88 Iatrogenic Toxic Encephalopathies
Long H, Deore K, Hoffman RS, Nelson LS. A fatal case of spongi-
form leukoencephalopathy linked to “chasing the dragon”. Appignani BA, Bhadelia RA, Blacklow SC, Wang AK, Roland SF,
J Toxicol Clin Toxicol 2003; 41: 887–891 Freeman RB Jr. Neuroimaging findings in patients on im-
Martin-Bouyer G, toga M, Lebreton R, Stolley PD. Outbreak of munosuppressive therapy: experience with tacrolimus tox-
accidental hexachlorophene poisoning in France. Lancet icity. AJR Am J Roentgenol 1996; 166: 683–688
1982; I: 91–95 Bargállo N, Burrel M, Berenguer J, Cofan F, Buñesch L, Mercader
McCreary M, Emerman C, Hanna J, Simon J. Acute myelopathy JM. Cortical laminar necrosis caused by immunosuppres-
following intranasal infusion of heroin; a case report. Neu- sive therapy and chemotherapy. AJNR Am J Neuroradiol
rology 2000; 55: 316–317 2000; 21: 479–484
Maxwell IC, Le Quesne PM. Conduction velocity in hexachloro- Bartynski WS,Grabb BC,Zeigler Z,Lin L,Andrews DF.Watershed
phane neuropathy. Correlation between electrophysiolog- imaging features and clinical vascular injury in cyclosporin
ical and histological neuropathy. J Neurol Sci 1979; 43: A neurotoxicity. J Comput Assist Tomogr 1997; 21: 872–880
95–110 Bartynski WS, Zeigler Z, Spearman MP, Lin L, Shadduck RK, Lis-
Nanan R, von Stockhausen HB, Petersen B, Solymosi L, War- ter J. Etiology of cortical and white matter lesions in cyclo-
muth-Metz M. Usual pattern of leukoencephalopathy after sporin-A and FK-506 neurotoxicity. AJNR Am J Neuroradiol
morphine sulphate intoxication. Neuroradiologie 2000; 42: 2001; 22: 1901–1914
845–848 Bova D, Shownkeen H, Goldberg K, Horowitz S, Azar-Kia B.
Ohnuma A, Kimura I, Saso S. MRI in chronic paint-thinner intox- Delayed transient neurologic toxicity due to tacrolimus:
ication. Neurology 1995; 37: 445–446 CT and MRI. Neuroradiology 2000; 42: 666–668
Pal KP, Samii A, Calne DB. Manganese neurotoxicity: a review of Bushara KO, Rust RS. Reversible MRI lesions due to pegaspar-
clinical features, imaging and pathology. Neurotoxicology gase treatment of non-Hodgkin’s lymphoma. Pediatr Neu-
1999; 20: 227–238 rol 1997; 17: 185–187
Palmer K, Inskip H, Martyn C, Coggon D. Dementia and occupa- Casanova B, Prieto M, Deya E, Gisbert C, Mir J, Berenguer J,
tional exposure to organic solvents. Occup Environ Med Vilchez JJ. Persistent cortical blindness after cyclosporine
1998; 55: 712–715 leukoencephalopathy. Liver Transplant Surg 1997; 3: 638–
Powell H, Swarner O, Gluck L, Lampert P. Hexachlorophene 640
myelinopathy in premature infants. J Pediatr 1973; 82: Coley SC, Porter DA, Calamante F, Chong WK, Connelly A. Quan-
976–981 titative MR diffusion mapping and cyclosporine-induced
Rosenberg NL, Kleinschmidt-DeMasters BK, Davis KA, Dreis- neurotoxicity. AJNR Am J Neuroradiol 1999; 20: 1507–1510
bach JN, Hormes JT, Filley CM.Toluene abuse causes diffuse Franco DA, Greenberg HS. 5-FU multifocal inflammatory
central nervous system white matter changes. Ann Neurol leukoencephalopathy and dihydropyrimidine dehydroge-
1988; 23: 611–614 nase deficiency. Neurology 2001; 56: 110–112
1036 References and Further Reading

Furukawa M,Terae S, Chu BC, Kaneko K, Kamada H, Miyasaka K. Ohara S,Hayashi R,Hata S,Itoh N,Hanyu N.Leukoencephalopa-
MRI in seven cases of tacrolimus (FK-506) encephalopathy: thy induced by chemotherapy with tegafur, a 5-fluorouracil
utility of FLAIR and diffusion-weighted imaging. Neuro- derivative. Acta Neuropathol (Berl) 1998; 96: 527–531
radiology 2001; 43: 615–621 Pace MT, Slovis TL, Kelly JK, Abella SD. Cyclosporin A toxicity:
Gleeson JG, duPlessis AJ, Barnes PD, Riviello JJ Jr. Cyclosporin A MRI appearance of the brain. Pediatr Radiol 1995; 25:
acute encephalopathy and seizure syndrome in childhood: 180–183
clinical features and ris of seizure recurrence. J Child Neurol Provenzale JM, Graham ML. Reversible leukoencephalopathy
1998; 13: 336–344 associated with graft-versus-host disease: MR findings.
Honkaniemi J, Kähärä V, Dastidar P, Latvala M, Hietaharju A, AJNR Am J Neuroradiol 1996; 17: 1290–1294
Salonen T, Keskinen L, Ollikainen J, Vahamaki L, Kello- Sánchez-Carpintero R, Narbona J, López de Mesa R, Arbizu J,
kumpu-Lehtinen P, Frey H. Reversible posterior leuko- Sierrasesúmaga L. Transient posterior encephalopathy in-
encephalopathy after combination chemotherapy. Neuro- duced by chemotherapy in children. Pediatr Neurol 2001;
radiology 2000; 42: 895–899 24: 145–148
Hook CC, Kimmel DW, Kvols LK, Scheithauer BW, Forsyth PA, Savarese DM, Gordon J, Smith TW, Litofsky NS, Licho R, Ragland
Rubin J, Moertel CG, Rodriquez M. Multifocal inflammatory R, Recht L. Cerebral demyelination syndrome in a patient
leukoencephalopathy with 5-fluorouracil an levamisole. treated with 5-fluorouracil and levamisole. The use of thal-
Ann Neurol 1992; 31: 262–267 lium SPECT imaging to assist in noninvasive diagnosis – a
Hwang YH, Suh CK, Park SP. Multifocal inflammatory leukoen- case report. Cancer 1996; 77: 387–394
cephalopathy: use of thallium-201 SPECT and proton MRS. Schuuring J,Wesseling P,Verrips A. Severe tacrolimus leukoen-
J Korean Med Sci 2003; 18: 621–624 cephalopathy after liver transplantation. AJNR Am J Neuro-
Israel ZH, Lossos A, Barak V, Soffer D, Siegal T. Multifocal de- radiol 2003; 24: 2085–2088
myelinative leukoencephalopathy associated with 5-fluo- Schwartz RB, Bravo SM, Klufas RA, Hsu L, Barnes PD, Robson CD,
rouracil and levamisole. Acta Oncol 2000; 39: 117–120 Antin JH. Cyclosporine neurotoxicty and its relationship to
Ito Y, Arahata Y, Goto Y, Hirayama M, Nagamutsu M, Yasuda T, hypertensive encephalopathy: CT and MR findings in 16
Yanagi T, Sobue G. Cisplatin neurotoxicity presenting as re- cases. AJR Am J Roentgenol 1995; 165: 627–631
versible posterior leukoencephalopathy syndrome. AJNR Scully RE, Mark EJ, McNeely WF, Ebeling SH, Phillips LD, Ellender
Am J Neuroradiol 1998; 19: 415–417 SM. Case 24–1999. N Eng J Med 1999; 341: 512–519
Jäger HR, Williams EJ, Savage DG, Rule SAJ, Hajnal JV, Sikora K, Shah AK. Cyclosporine A neurotoxicity among bone marrow
Goldman JM, Bydder GM. Assessment of brain changes transplant recipients. Clin Neuropharmacol 1999; 22: 67–73
with registered MR before and after bone marrow trans- Small SL, Fukui MB, Bramblett GT, Eidelman BH. Immunosup-
plantation for chronic myeloid leukemia. AJNR Am J Neuro- pression-induced leukoencephalopathy from tacrolimus
radiol 1996; 17: 1275–1282 (FK506). Ann Neurol 1996; 40: 575–580
Jansen O, Krieger D, Krieger S, Sartor K. Cortical hyperintensity Stemmer SM, Stears JC, Burton BS, Jones RB, Simon JH. White
on proton density-weighted images: an MR sign of cyclo- matter changes in patients with breast cancer treated with
sporine-related encephalopathy. AJNR Am J Neuroradiol high-dose chemotherapy and autologous bone marrow
1996; 17: 337–344 support. AJNR Am J Neuroradiol 1994; 15: 1267–1273
Jarosz JM, Howlett DC, Cox TCS, Bingham JB. Cyclosporine-re- Stone JA, Castillo M, Mukherji SK. Leukoencephalopathy com-
lated reversible posterior leukoencephalopathy: MRI. Neu- plicating an ommaya reservoir and chemotherapy. Neuro-
roradiology 1997; 39: 711–715 radiology 1999; 41: 134–136
Kelly P, Staunton H, Lawler M, Brennan P, Jennings S, Unger ER, Tahsildar HI, Remler BF, Creger RJ, Cooper BW, Snodgrass SM,
Sung JH, Farrell MA. Multifocal remitting-relapsing cerebral Tarr RW, Lazarus HM. Delayed, transient encephalopathy af-
demyelination twenty years following allogeneic bone ter marrow transplantation: case reports and MRI findings
marrow transplantation. J Neuropathol Exp Neurol 1996; in four patients. J Neurol-Oncol 1996; 27: 241–250
55: 992–998 Tha KK, Terae S, Sugiura M, Nishioka T, Oka M, Kudoh K, Kaneko
Kilinç M, Benli SU, Can U,Yilmaz A, Karakayali H, Çolak T,Tarhan K, Miyasaka K. Diffusion-weighted magnetic resonance
C, Özdemir BH. FK 506-induced fulminant leukoen- imaging in early stage of 5-fluorouracil-induced leuko-
cephalopathy after kidney transplantation: case report. encephalopathy. Acta Neurol Scand 2002; 106: 379–386
Transplant Proc 2002; 34: 1182–1184 Tomura N,Kurowawa R,Kato K,Takahashi S,Watarai J,Takeda O,
Kimmel DW,Wijdicks EFM, Rodiquez M. Multifocal inflammato- Watanabe A, Takada G. J Comput Assist Tomogr 1998; 22:
ry leukoencephalopathy associated with levamisole thera- 505–507
py. Neurology 1995; 45: 374–376 Trullemans F, Grignard F, van Camp B, Schots R. Clinical findings
Kuzuhara S, Ohkoshi N, Kanemaru K, Hashimoto H, Nakanishi T, and magnetic resonance imaging in severe cyclosporine-
Toyokura Y. Original investigations. Subacute leukoen- related neurotoxicity after allogeneic bone marrow trans-
cephalopathy induced by carmofur, a 5-fluorouracil deriva- plantation. Eur J Haematol 2001; 67: 94–99
tive. J Neurol 1987; 234: 365–370 Wilson DA, Nitschke R, Bowman ME, Chaffin MJ, Sexauer CL,
Lanzino G, Cloft H, Hemstreet MK, West K, Alston S, Ishitani M. Prince JR.Transient white matter changes on MR images in
Reversible posterior leukoencephalopathy following organ children undergoing chemotherapy for acute lymphocytic
transplantation. Description of 2 cases. Clin Neurol Neuro- leukemia: correlation with neuropsychologic deficiencies.
surg 1997; 99: 222–226 Radiology 1991; 180: 205–209
Lyass O, Lossos A, Hubert A, Gips M, Peretz T. Cisplatin-induced
non-convulsive encephalopathy. Anti-Cancer Drugs 1998;
9: 100–104
Matsumoto S, Nishizawa S, Murakami M, Noma S, Sano A, Kuro-
da Y. Carmofur-induced leukoencephalopathy: MRI. Neuro-
radiology 1995; 37: 649–652
References and Further Reading 1037

89 Central Pontine Laubenberger J, Schneider B, Ansorge O, Götz F, Häussinger D,


and Extrapontine Myelinolysis Volk B, Langer M. Central pontine myelinolysis: clinical pre-
sentation and radiologic findings. Eur Radiol 1996; 6: 177–
183
Adams RD, Victor M, Mancall EL. Central pontine myelinolysis.
Laureno R,Karp BI.Myelinolysis after corection of hyponatrem-
Arch Neurol Psychiatry 1959; 81: 154–172
ia. Ann Intern Med 1997; 126: 57–62
Adler S, Verbalis JG, Williams D. Effect of rapid correction of
Lien YH. Role of organic osmolytes in myelinolysis. J Clin Invest
hyponatremia on the blood-brain barrier of rats. Brain Res
1995; 95: 1579–1586
1995; 679:135–143
Maraganore DM, Folger WN, Swanson JW, Ahlskog JE. Move-
Ağildere AM, Benli S, Erten Y, Colpkun M, Boyvat F, Özdemir N.
ment disorders as sequelae of central pontine myelinolysis:
Osmotic demyelination syndrome with a dysequilibrium
report of three cases. Mov Disord 1992; 7: 142–148
syndrome: reversible MRI findings. Neuroradiology 1998;
Murdoch M, Chang M, McVicar J. Central pontine myelinolysis
40: 228–232
after liver transplantation: a case report. Transplant Int
Ashrafian H, Davey P A review of the causes of central pontine
1995; 8: 399–402
myelinosis: yet another apoptotic illness? Eur J Neurol
Menakaya JO, Wassmer E, Bradshaw K, Seri S, Whitehouse WP.
2001; 8: 103–109
Idiopathic central pontine myelinolysis in childhood. Dev
Ayus JC, Arieff AI. Brain damage and postoperative hypona-
Med Child Neurol 2001; 43: 697–700
tremia: the role of gender. Neurology 1996; 46: 323–328
Menger H, Jörg J. Outcome of central pontine and extrapon-
Ayus JC, Krothapalli RK, Arieff AI. Treatment of symptomatic
tine myelinolysis (N = 44). J Neurol 1999; 246: 700–705
hyponatremia and its relation to brain damage. N Engl J
Moore K, Midha M. Extra pontine myelinolysis in a tetraplegic
Med 1987; 317: 1190–1195
patient: case report. Spinal Cord 1997; 35: 332–334
Brown WD. Osmotic demyelination disorders: central pontine
Niehaus L, Kulozik A, Lehmann R. Reversible central pontine
and extrapontine myelinolysis. Curr Opin Neurol 2000; 13:
and extrapontine myelinolysis in a 16-year-old girl. Childs
691–697
Nerv Syst 2001; 17: 294–296
Brunner JE, Redmond JM, Haggar AM, Kruger DF, Elias SB. Cen-
Okeda R, Kitano M, Sawabe M, Yamada I, Yamada M. Distribu-
tral pontine myelinolysis and pontine lesions after rapid
tion of demyelinating lesions in pontine and extrapontine
correction of hyponatremia: a prospective magnetic reso-
myelinolysis – three autopsy cases including one case de-
nance imaging study. Ann Neurol 1990; 27: 61–66
void of central pontine myelinolysis. Acta Neuropathol
Choe WJ,Cho BK,Kim IO,Shin HY,Wang KC.Extrapontine myeli-
(Berl) 1986; 69: 259–266
nolysis caused by electrolyte imbalance during the man-
Pradhan S, Jha R, Singh MN, Gupta S, Phadke RV, Kher V. Central
agement of suprasellar germ cell tumors. Childs Nerv Syst
pontine myelinolysis following “slow” correction of hy-
1998; 14: 155–158
ponatremia. Clin Neurol Neurosurg 1995; 97: 340–343
Chu K, Kang DW, Ko SB, Kim M. Diffusion-weighted MR findings
Rouanet F, Tison F, Dousset V, Corand V, Orgogozo JM. Early T2
of central pontine and extrapontine myelinolysis. Acta
hypointense signal abnormality preceding clinical mani-
Neurol Scand 2001; 104: 385–388
festations of central pontine myelinolysis. Neurology 1995;
Chua GC, Sitoh YY, Lim CC, Chua HC, Ng PY. MRI findings in
44: 979–980
osmotic myelinolysis. Clin Radiol 2002; 57: 800–806
Salvesen R. Extrapontine myelinolysis after surgical removal of
Cramer SC, Stegbauer KC, Schneider A, Mukai J, Maravilla KR.
a pituitary tumour. Acta Neurol Scand 1998; 98: 213–215
Decreased diffusion in central pontine myelinolysis. AJNR
Susa S, Daimon M, Morita Y, Kitagawa M, Hirata A, Manaka H,
Am J Neuroradiol 2001; 22: 1476–1479
Sasaki H, Kato T. Acute intermittent porphyria with central
DeLuca GC, Nagy Zs, Esiri MM, Davey P. Evidence for a role for
pontine myelinolysis and cortical laminar necrosis. Neuro-
apoptosis in central pontine myelinolysis. Acta Neuro-
radiology 1999; 41: 835–839
pathol (Berl) 2002; 103: 590–598
Tien R, Arieff AL, Kucharczyk W,Wasik A, Kucharczyk J. Hypona-
Giannetti AV, Pittella JEH. Ischemic and hemorrhagic necrosis
tremic encephalopathy: is central pontine myelinolysis a
of the pons with anatomical location similar to that of cen-
component? Am J Med 1992; 92: 513–522
tral pontine myelinolysis in a chronic alcoholic patient. Clin
Udani VP, Dharnidharka VR, Gajendragadkar AR, D’Souza B. Ex-
Neuropathol 1993; 12: 156–159
tra and central pontine myelinolysis in a child with adrenal
Hadfield MG, Kubal WS. Extrapontine myelinolysis of the basal
insufficiency. Pediatr Neurol 1997; 17: 158–160
ganglia without central pontine myelinolysis. Clin Neu-
ropathol 1996; 15: 96–100
Karp BI, Laureno R. Pontine and extrapontine myelinolysis: a
neurologic disorder following rapid correction of hypona- 90 Hypernatremia
tremia. Medicine 1993; 72: 359–373
Karp BI, Laureno R. Central pontine and extrapontine myelino- Al Orainy IA, I’Gorman A, Decell MK. Cerebral bleeding, infarcts,
lysis after correction of hyponatremia. Neurologist 2000; 6: and presumed extrapontine myelinosis in hypernatraemic
255–266 dehydration. Neuroradiology 1999; 41: 144–146
Koch KJ, Smith RR. Gd-DTPA enhancement in MR imaging of Arieff AI, Guisado R. Effects on the cerebral nervous system of
central pontine myelinolysis. AJNR Am J Neuroradiol 1989; hypernatremic and hyponatremic states. Kidney Int 1976;
10 (suppl 5): S58 10: 104–116
Koci TM, Chiang F, Chow P, Wang A, Chiu LC, Itabashi H, Barer J, Leighton L, Hill RM, Martinez WM. Fatal poisoning from
Mehringer CM. Thalamic extrapontine lesions in central salt used as an emetic. Am J Dis Child 1973; 125: 889–890
pontine myelinolysis. AJNR Am J Neuroradiol 1990; 11: Baugh JR, Krug EF, Weir MR. Punishment by salt poisoning.
1229–1233 South Med J 1963; 76: 540–541
Brown WD, Caruso JM. Extrapontine myelinolysis with involve-
ment of the hippocampus in three children with severe hy-
ponatremia. J Child Neurol 1999; 14: 428–433
1038 References and Further Reading

Burnett LS,Wentz AC, King TM.Techniques of pregnancy termi- Samir El-Dahr,Gomez RA,Campbell FG,Chevalier RL.Rapid cor-
nation. I, II. Obstet Gynecol Surv 1974; 29: 6–42 rection of acute salt poisoning by peritoneal dialysis. Pedi-
Calam J, Krasner N, Haqqani M.Extensive gastrointestinal dam- atr Nephrol 1987; 1: 602–604
age following a saline emetic. Dig Dis Sci 1982; 27: 936–946 Saunders N, Balfe JW,Laski B.Severe salt poisoning in an infant.
Calvin ME, Knepper R, Robertson WO. Hazards to health. J Pediatr 1976; 88: 258–261
N Engl J Med 1964; 270: 625–626 Schiffer MA, Pakter J, Clahr J. Mortality associated with hyper-
Carter RF, Fotheringham BJ. Fatal salt poisoning due to gastric tonic saline abortion. Obstet Gynecol 1973; 42: 759–764
lavage with hypertonic saline. Med J Aust 1971; 1: 539–541 Soupart A, Penninckx R, Namias B, Seinuit A, Perier O, Decaux G.
Dine MS, McGovern ME. Intentional poisoning of children – an Brain myelinolysis following hypernatremia in rats. J Neu-
overlooked category of child abuse: report of seven cases ropathol Exp Neurol 1996; 55: 106–113
and review of the literature. Pediatrics 1982; 70: 32–35 Soupart A, Penninckx R, Stenuit A, Perier O, Deaux G. Reinduc-
Dockery WK. Fatal intentional salt poisoning associated with a tion of hyponatremia improves survival in rats with myeli-
radiopaque mass. Pediatrics 1992; 89: 964–965 nolysis-related neurologic symptoms. J Neuropathol Exp
Elton NW, Elton WJ, Nazereno JP. Pathology of acute salt poi- Neurol 1996; 55: 594–601
soning in infants. Am J Clin Pathol 1963; 39: 252–264 Walter GF, Maresch W. Irrtümliche kochsalzintoxikation bei
Finberg L. Pathogenesis of lesions in the nervous system in hy- neugeborenen. Morphologische befunde und patho-
pernatremic states. Clinical observations of infants. Pedi- genetische discussion. Klin Padiatr 1987; 1999: 269–273
atrics 1959; 23: 40–44 Winter M, Taylor DJE. Danger of saline emetics in first-aid for
Finberg L. Unforgettable patients. J Pediatr 1992; 121: 323–324 poisoning. BMJ 1974; III: 802
Finberg L,Harrison HE.Hypernatremia in infants.An evaluation
of the clinical and biochemical findings accompanying this
state. Pediatrics 1955; 16: 1–12
Finberg L, Luttrell C, Redd H. Pathogenesis of lesions in the ner-
91 Marchiafava-Bignami Syndrome
vous system in hypernatremic states. Experimental studies
of gross anatomic changes and alterations of chemical Arbelaez A,Pajon A,Castillo M.Acute Marchiafava-Bignami dis-
composition of the tissues. Pediatrics 1959; 23: 46–53 ease: MR findings in two patients. AJNR Am J Neuroradiol
Fried LF, Palevsky PM. Hyponatremia and hypernatremia. Med 2001; 24: 1955–1957
Clin North Am 1997; 81: 585–609 Berek K,Wagner M, Chemelli AP, Aichner F, Benke T. Hemispher-
Friis-Hansen B, Buchthal F. EEG findings in an infant with water ic disconnection in Marchiafava-Bignami disease: clinical,
intoxication and convulsions incident to hypernatraemia. neuropsychological and MRI findings. J Neurol Sci 1994;
Electroencephalogr Clin Neurophysiol 1965; 19: 387–390 123: 2–5
Golder W, Felgenhauer N, von Einsiedel H, von Clarmann M. Caparros-Lefebvre D, Pruvo JP, Josien E, Pertuzon B, Clarisse J,
Ausgedehnte Kleinhirblutungen nach Kochsalzvergiftung. Petit H.Marchiafava-Bignami disease: use of contrast media
Klin Neuroradiol 1992; 2: 21–23 in CT and MRI. Neuroradiology 1994; 36: 509–511
Götze H. Kochsalzvergiftung infolge übergrosser parenteraler Çelik Y, Kaya M, Šengün S, Utku U. Marchiafava-Bignami dis-
Zufuhr. Arch Toxikol 1962; 19: 284–292 ease:cranial MRI and SPECT findings.Clin Neurol Neurosurg
Habbick BF, Hill A,Tchang SPK. Computer tomography in an in- 2002; 104: 339–341
fant with salt poisoning: relationship of hypodense areas in Clavier E, Thiebot J, Delangre T, Hannequin D, Samson M,
basal ganglia to serum sodium concentration. Pediatrics Benozio M. Marchiafava-Bignami disease. A case studied by
1984; 74: 1123–1125 CT and MRI imaging. Neuroradiology 1986; 28: 376
Hartfield SD, Loewy JA,Yager JY.Transient thalamic changes on Ferracci F, Conte F, Gentile M, Candeago R, Foscolo L, Bendini M,
MRI in a child with hypernatremia. Pediatr Neurol 1999; 20: Fassetta G. Marchiafava-Bignami disease. Computed tomo-
60–62 graghic scan, 99mTc HMPAO-SPECT, and FLAIR MRI findings
Klein Schwartz W, Finke HB. Saltwater intoxication. JAMA 1978; in a patient with subcortical aphasia, alexia, bilateral
240: 1338–1339 agraphia, and left-handed deficit of constructional ability.
Korkmaz A, Yiğit S, Firat M, Oran O. Cranial MRI in neonatal Arch Neurol 1999; 56: 107–110
hypernatraemic dehydration. Pediatr Radiol 2000; 30: 323– Friese SA, Bitzer M, Freudenstein D, Voigt K, Küker W. Classifica-
325 tion of acquired lesions of the corpus callosum with MRI.
Meadow R. Non-accidental salt poisoning. Arch Dis Child 1993; Neuroradiology 2000; 42: 795–802
68: 448–452 Gambini A,Falini A,Moiola L,Comi G,Scotti G.Marchiafava-Big-
Paut O, André N, fabre P, Sobraquès P, Drouet G, Arditti J, Cam- nami disease: longitudinal MR imaging and MR spec-
boulives J. The management of extreme hypernatraemia troscopy study. AJNR Am J Neuroradiol 2003; 24: 249–253
secondary to salt poisoning in an infant. Paediatr Anaesth Gass A, Birtsch C, Oster M, Schwartz A, Hennerici MG. Marchi-
1999; 9: 171–174 afava-Bignami disease: reversibility of neuroimaging ab-
Peer G, Wigler I, Weintraub M, Liron M, Aviram A. Acute psy- normality. J Comput Assist Tomogr 1998; 22: 503–504
chosis and hypertensive crisis following oral salt replace- Hayashi T, Tanohata K, Kunimoto M, Inoue K. Marchiafava-Big-
ment. Lancet 1983; II: 634–635 nami disease with resolving symmetrical putaminal lesion.
Postuma R.Whole bowel irrigation in pediatric patients. J Pedi- J Neurol 2002; 249: 227–228
atr Surg 1982; 17: 350–352 Helenius J, Tatlisumak T, Soinne L, Valanne L, Kaste M. Marchi-
Roberts CJC, Noakes MJ. Fatal outcome from administration of afava-Bignami disease: two cases with favourable out-
a salt emetic. Postgrad Med J 1974; 50: 513–515 come. Eur J Neurol 2001; 8: 269–272
Roberts CJC, Noakes MJ. Danger of emetics in first-aid for poi- Ishii K, Ikejiri Y, Sasaki M, Kitagaki H, Mori E. Regional cerebral
soning. BMJ 1974; III: 683 glucose metabolism and blood flow in a patient with
Roscelli JD,Yu CE, Southgate WM. Management of salt poison- Marchiafava-Bignami disease.AJNR Am J Neuroradiol 1999;
ing in an extremely low birth weight infant.Pediatr Nephrol 20: 1249–1251
1994; 8: 172–174
References and Further Reading 1039

Izquierdo G, Quesada MA, Chacon J, Martel J. Neuroradiologic Holdcroft A, Oatridge A, Fusi L, Hajnal JV, Saeed N, Bydder GM.
abnormalities in Marchiafava-Bignami disease of benign Magnetic resonance imaging in preeclampsia and eclamp-
evolution. Eur J Radiol 1992; 15: 71–74 sia by visual disturbance and other neurological abnormal-
Kohler CG, Ances BM, Coleman AR, Ragland JD, Lazarev M, Gur ities. Int J Obstet Anesth 2002; 11: 255–259
RC. Marchiafava-Bignami disease: literature review and Karampekios SK, Contopoulou E, Basta M, Tzagournissakis M,
case report. Neuropsychiatry Neuropsychol Behav Neurol Gourtsoyiannis N. Hypertensive encephalpathy with pre-
2000; 13: 67–76 dominant brain stem involvement: MRI findings. J Hum
Mayer JW, de Liège P, Netter JM, Danzé F, Reizine D. Computer- Hypertens 2004: 18: 133–134
ized tomography and nuclear magnetic resonance imag- Kastrup O, Maschke M,Wanke I, Diener HC. Posterior reversible
ing in Marchiafava-Bignami disease. J Neuroradiol 1987; 14: encephalopathy syndrome due to severe hypercalcemia.
152–158 J Neurol 2002; 249: 1563–1566
Ruiz-Martínez J, Perez-Balsa AM, Ruibal M, Urtasun M, Villanua King PH, Bragdon AC. MRI reveals multiple reversible cerebral
J, Massó JFM. Marchiafava-Bignami disease with wide- lesions in an attack of acute intermittent porphyria. Neuro-
spread extracallosal lesions and favourable course. Neuro- logy 1991; 41: 1300–1302
radiology 1999; 41: 40–43 Kumai Y, Toyoda K, Fujii K, Ibayashi S. Hypertensive encephalo-
Sano M, Ishii K, Suzuki T, Uchigata M, Senda M. Reductions in pathy extending into the whole brainstem and deep struc-
the bilateral parietal and occipital cerebral blood flow and tures. Hypertens Res 2002; 25: 797–800
metabolism in a patient with Marchiafava-Bignami disease. Loureiro R, Leite CC, Kahhale S, Freire S, Sousa B, Cardoso EF,
J Neurol 1999; 246: 607–607 Alves EA, Borba P, Cerri GG, Zugaib M. Diffusion imaging
Tarnowska-Dziduszko E, Bertrand E, Szpak GM. Morphological may predict reversible brain lesions in severe pre-eclamp-
changes in the corpus callosum in chronic alcoholism. Folia sia: initial experience. Am J Obstet Gynecol 2003; 189:
Neuropathol 1995; 33: 25–29 1350–1355
Yamamoto T, Ashikaga R, Araki Y, Nishimura Y. A case of Marchi- Moore KR, Osborn AG, Townsend JJ, Boyer RS, Orrison WW Jr,
afava-Bignami disease: MRI findings on spin-echo and fluid Ablin DS. The imaging and pathologic spectrum of he-
attenuated inversion recovery (FLAIR) images. Eur J Neurol molytic-uremic syndrome-thrombotic trombocytopenic
2000; 34: 141–143 purpura (HUS-TTP syndrome). Int J Neuroradiol 1997; 3:
147–159
Morello F, Marino A, Cigolini M, Cappellari F.Hypertensive brain
stem encephalopathy: clinically silent massive edema of
92 Posterior Reversible Encephalopathy the pons. Neurol Sci 2001; 22: 317–320
Syndrome Mukherjee P, McKinstry RC. Reversible posterior leukoen-
cephalopathy syndrome: evaluation with diffuse – tensor
Çelik M, Forta H, Dalkiliç T, Babacan G. MRI reveals reversible MR imaging. Radiology 2001; 219: 756–765
lesions resembling posterior reversible encephalopathy in Pavlakis SG, Frank Y, Chusid R. Hypertensive encephalopathy,
porphyria. Neuroradiology 2002; 44: 839–841 reversible occipitoparietal encephalopathy, or reversible
Chakravarty A, Chakrabarti SD. The neurology of eclampsia: posterior leukoencephalopathy: three names for an old
some observations. Neurol India 2002; 50: 128–135 syndrome. J Child Neurol 1999; 14: 277–281
Covarrubias DJ, Luetmer PH, Campeau NG. Posterior reversible Primavera A, Audenino D, Mavilio N, Cocito L. Reversible poste-
encephalopathy syndrome: prognostic utility of quantita- rior leukoencephalopathy syndrome in systemic lupus and
tive diffusion-weighted MR images. AJNR Am J Neuroradiol vasculitis. Ann Rheum Dis 2001; 60: 534–537
2002; 23: 1038–1048 Provenzale JM, Petrella JR, Cruz LCH Jr,Wong JC, Engelter S, Bar-
Delanty N, Vaughan C, Frucht S, Stubgen P. Erythropoietin-as- boriak DP. Quantitative assessment of diffusion abnormali-
sociated hypertensive posterior leukoencephalopathy. ties in posterior reversible encephalopathy syndrome.
Neurology 1997; 49: 686–689 AJNR Am J Neuroradiol 2001; 22: 1455–1461
De Seze J, Mastain B, Stojkovic T, Ferriby D, Pruvo JP, Destée A, Schwartz RB, Mulkern RV, Gudbjartsson H, Jolesz F. Diffusion-
Vermersch P. Unusual MR findings of the brain stem in arte- weighted MR imaging in hypertensive encephalopathy:
rial hypertension.AJNR Am J Neuroradiol 2000; 21:391–394 clues to pathogenesis. AJNR Am J Neuroradiol 1998; 19:
Digre KB, Varner MW, Osborn AG, Crawford S. Cranial magnetic 859–862
resonance imaging in severe preeclampsia vs eclampsia. Schwartz RB, Feske SK, Polak JF, Degirolami U, Iaia A, Beckner
Arch Neurol 1993; 50: 399–406 KM, Bravo SM, Klufas RA, Chai RYC, Repke JT. Preeclampsi-
Eguchi K, Kasahara K, Nagashima A, Mori T, Nii T, Ibaraki K, Kario eclampsia: clinical and neuroradiographic correlates and
K, Shimada K.Two cases of malignant hypertension with re- insights into the pathogenesis of hypertensive en-
versible diffuse leukoencephalopathy exhibiting a re- cephalopathy. Radiology 2000; 217: 371–376
versible nocturnal blood pressure “riser”pattern.Hypertens Sengar AR, Gupta RK, Dhanuka AK, Das K. MR imaging, MR an-
Res 2002; 25: 467–473 giography, and MR spectroscopy of the brain in eclampsia.
Eichler FS,Wang P,Wityk RJ,Beauchamp NJ Jr,Barker PB.Diffuse AJNR Am J Neuroradiol 1997; 18: 1485–1490
metabolic abnormalities in reversible posterior leukoen- Shimono T, Miki Y, Toyoda H, Egawa H, Eumoto S, Tanaka K,
cephalopathy syndrome. AJNR Am J Neuroradiol 2002; 23: Hattori H, Kanagaki M, Itoh K, Konishi J. MR imaging with
833–837 quantitative diffusion mapping of tracolimus-induced
Hauser RA, Lacey M, Knight MR.Hypertensive encephalopathy: neurotoxicity in organ transplant patients. Eur Radiol 2003;
magnetic resonance description of reversible cortical and 13: 986–993
white matter lesions. Arch Neurol 1988; 45: 1078–1083 Susa S, Daimon M, Morita Y, Kitagawa M, Hirata A, Manaka H,
Hinchey J, Chaves C, Appignani B, Breen J, Pao L,Wang A, Pessin Sasaki H, Kato T. Acute intermittent porphyria with central
MS, Lamy C, Mas JL, Caplan LR. A reversible posterior pontine myelinolysis and cortical laminar necrosis. Neuro-
leukoencephalopathy syndrome. N Engl J Med 1996; 334: radiology 1999; 41: 835–839
494–500
1040 References and Further Reading

Tajima Y, Isonishi K, Kashiwaba T,Tashiro K.Two similar cases of Grois N, Barkovich AJ, Rosenau W, Ablin AR. Central nervous
encephalopathy, possibly a reversible posterior leukoen- system disease associated with Langerhans’ cell histiocyto-
cephalopathy syndrome: serial findings of magnetic reso- sis. Am J Pediatr Hematol Oncol 1993; 15: 245–254
nance imaging, SPECT and angiography. Int Med 1999; 38: Grois NG, Favara BE, Mostbeck GH, Prayer D. Central nervous
54–58 system disease in Langerhans cell histiocytosis. Hematol
Urushitani M, Seriu N, Udaka F, Kameyama M, Nishinaka K, Ko- Oncol Clin North Am 1998; 12: 287305
dama M. MRI Demonstration of a reversible lesion in cere- Herzog KM, Tubbs RR. Langerhans cell histiocytosis. Adv Anat
bral deep white matter in thrombotic trombocytopenic Pathol 1998; 5: 347–358
purpura. Neuroradiology 1996; 38: 137–138 Hund E, Steiner H-H, Jansen O, Sieverts H, Sohl G, Essig M.Treat-
Taylor MB, Jackson A, Weller JM. Dynamic susceptibility con- ment of cerebral Langerhans cell histiocytosis. J Neurol Sci
trast enhanced MRI in reversible posterior leukoen- 1999; 171: 145–152
cephalopathy syndrome associated with haemolytic Iwuagwu FC, Rigby HS, Payne F, Reid CD. Juvenile xanthogran-
uraemic syndrome. Br J Radiol 2000; 73: 438–442 uloma variant: a clinicopathological case report and review
Thomas SV. Neurological aspects of eclampsia. J Neurol Sci of the literature. Br J Plast Surg 1999; 52: 591–596
1998; 155; 37–43 Kim IT, Lee SM. Choroidal Langerhans’ cell histiocytosis. Acta
Utz N, Kinkel B, Hedde JP, Bewermeyer H. MR imaging of acute Ophthalmol Scand 2000; 78: 97–100
intermittent porphyria mimicking reversible posterior Malpas JS. Langerhans cell histiocytosis in adults. Hematol
leukoencephalopathy syndrome. Neuroradiology 2001; 43: Oncol Clin North Am 1998; 12: 259–268
1059–1062 Miaux Y, Keime F, Martin-Duverneuil N, Cognard C, Savin D,
Wanatabe Y, Mitomo M, Tokuda Y, Yoshida K, Choi S, Hosoki T, Chiras J. Hyperintense basal ganglia on T1-weighted MR in
Ban C. Eclamptic encephalopathy: MRI, including diffusion- a patient with Langerhans cell histiocytosis. AJNR Am J
weighted images. Neuroradiology 2002; 44: 981–958 Neuroradiol 1996; 17: 1193
Weidauer S, Gaa J, Sitzer M, Hefner R, Lanfermann H, Zanella FE. Modan-Moses D, Weintraub M, Meyerovitch J, Segal-Lieber-
Posterior encephalopathy with vasospasm: MRI and angio- man G. Bielorai B, Shimon I. Hypopituitarism in Langerhans
graphy. Neuroradiology 2003; 45: 869–876 cell histiocytosis: seven cases and literature review.
J Endocrinol Invest 2001; 24: 612–617
Pautas E, Chérin P, Pelletier S, Vidailhet M, Herson S. Cerebral
Erdheim-Chester disease: report of two cases with progres-
93 Langerhans Cell Histiocytosis sive cerebellar syndrome with dentate abnormalities on
magnetic resonance imaging. J Neurol Neurosurg Psychia-
Adle-Biassette H, Chetritt J, Bergemer-Fouquet AM,Wechsler J, try 1998; 65: 597–599
Mussini JM, Gray F. Pathology of the central nervous system Poe LB, Dubowy RL, Hochhauser L, Collins GH, Crosley CJ, Kanz-
in Chester-Erdheim disease: report of three cases. J Neu- er MD, Oliphant M, Hodge CJ Jr. Demyelinating and gliotic
ropathol Exp Neurol 1997; 56: 1207–1216 cerebellar lesions in Langerhans cell histiocytosis. AJNR Am
Aricò M, Haupt R, Russotto VS, Bossi G, Scappaticci S, Denasino J Neuroradiol 1994; 15: 1921–1928
C. Langerhans cell histiocytosis in two generations: a new Polizzi A, Coghill S, McShane MA, Squier W. Acute ataxia com-
family and review of the literature. Med Pediatr Oncol 2001; plicating Langerhans cell histiocytosis. Arch Dis Child 2002;
36: 314–316 86: 130–131
Bohlega S, Alwatban J, Tulbah A, Bakheet SM, Powe J. Cerebral Prayer D, Grois N, Prosch H, Gadner H, Barkovich AJ. MR imag-
manifestation of Erdheim-Chester disease:clinical and radi- ing presentation of intracranial disease associated with
ologic findings. Neurology 1997; 49: 1702–1705 Langerhans cell histiocytosis. AJNR Am J Neuroradiol 2004;
Broadbent V, Gadner H. Current therapy for Langerhans cell 25: 880–891
histiocytosis. Hematol Oncol Clin North Am 1998; 12: 327– Pritchard J. Acute ataxia complicating Langerhans cell histio-
338 cytosis. Arch Dis Child 2003; 88: 178–179
Cervera A, García Penas JJ, Díaz MA, Gutiërres-Solana LG, Beni- Rosenfield NS, Abrahams J, Komp D. Brain MR in patients with
to A, Ruiz-Falcó M-L. Villa M. CNS sequellae in Langerhans Langerhans cell histiocytosis: findings and enhancement
cell histiocytosis: progressive spinocerebellar degenera- with Gd-DTPA. Pediatr Radiol 1990; 20: 433–436
tion as a late manifestation of the disease. Pediatr Hematol Saatci L, Baskan O, Haliloglu M, Aydingoz U. Cerebellar and
Oncol 1997; 14: 577–584 basal ganglion involvement in Langerhans cell histiocyto-
Chu T. Langerhans cell histiocytosis. Aus J Dermatol 2001; 42: sis. Neuroradiology 1999; 41: 443–446
237–242 Schmitz L, Favara BE. Nosology and pathology of Langerhans
Evidente VGH, Adler CH, Giannini C, Conley CR, Parisi JE, Fletch- cell histiocytosis. Hematol Oncol Clin North Am 1998; 12:
er GP. Erdheim-Chester disease with extensive intraaxial 221–246
brain stem lesions presenting as a progressive cerebellar Shuper A, Stark B,Yaniv Y, Zaizov R, Carel C, Sadeh M, Steinmetz
syndrome. Mov Disord 1998; 13: 576–581 A. Cerebellar involvement in Langerhans’ cell histiocytosis:
Fuzakawa T, Tsukishima E, Sasaki H, Hamada K, Hamada T, a progressive neuropsychiatric disease.J Child Neurol 2000;
Tashiro K. Erdheim-Chester disease and slowly progressive 15: 824–826
cerebellar dysfunction. J Neurol Neurosurg Psychiatry Strottmann JM, Ginsberg LE, Stanton C. Langerhans cell histio-
1995; 58: 238–240 cytosis involving the corpus callosum and cerebellum:
Gaetani P,Tancioni F, di Rocco M, Rodriguez y Baena R. Isolated gadolinium-enhanced MRI. Neuroradiology 1995; 37: 289–
cerebellar involvement in Rosai-Dorfman disease: case 292
report. Neurosurgery 2000; 46: 479–481 Thorpy MJ, Crosley CJ. Multifocal eosinophilic granuloma and
Goldberg-Stern H, Weitz R, Zaizov R, Gornish M, Gadoth N. Pro- the cerebellum. Ann Neurol 1980; 8: 454
gressive spinocerebellar degeneration “plus” associated Varona L, Sagasta A, Bárcena J, Zarranz J-J. Neurological mani-
with Langerhans cell histiocytosis: a new paraneoplastic festations following treatment of Langerhans cell histio-
syndrome? J Neurol Neurosurg Psychiatry 1995; 58: 180–183 cytosis. J Neurol 1997; 244: 58–60
References and Further Reading 1041

Vassallo R, Ryu JH, Colby TV, Hartman T, Limper AH. Pulmonary Hosler BA,Brown RH Jr.Superoxide dismutase and oxygen rad-
Langerhans’-cell histiocytosis. N Engl J Med 2000; 342: ical neurotoxicity. Curr Opin Neurol 1996; 9: 486–491
1969–1978 Ikeda J, Nagashima G, Saito N, Nowak TS, Joo F, Mies G, Lohr JM,
Weindauer S, von Stuckrad-Barre S, Dettmann E, Zanella FE, Ruetzler CA, Klatzo I. Putative neuroexcitation in cerebral is-
Lanfermann H. Cerebral Erdheim-Chester disease: case re- chemia and brain injury. Stroke 1990; 21 (suppl III): 65–70
port and review of the literature. Neuroradiology 2003; 45: Ikeda Y, Long DM.The molecular basis of brain injury and brain
241–245 edema: the role of oxygen free radicals.Neurosurgery 1990;
Whitsett SF, Kneppers K, Coppes MJ, Egeler RM. Neuropsycho- 27: 1–11
logic deficits in children with Langerhans cell histiocytosis. Jarvis MF, Wagner GC. 1-methyl-4-phenyl-1,2,3,6-tetrahy-
Med Pediatr Oncol 1999; 33: 486–492 dropyridine-induced neurotoxicity in the rat: characteriza-
Wright RA, Hermann RC, Parisi JE. Neurological manifestations tion and age-dependent effects. Synapse 1990; 5: 104–112
of Erdheim-Chester disease. J Neurol Neurosurg Psychiatry Johnston MV. Neurotransmitters and vulnerability of the de-
1999; 66: 72–75 veloping brain. Brain Dev 1995; 17: 301–306
Koroshetz WJ, Freese A, DiFiglia M.The correlation between ex-
citatory amino acid-induced current responses and excito-
toxicity in striatal cultures. Brain Res 1990; 521: 265–272
94 Posthypoxic-ischemic Damage Kucharczyk J, Mintorovitch J, Moseley ME, Asgari HS, Sevick RJ,
Derugin N,Norman D.Ischemic brain damage:reduction by
Albin RL, Greenamyre JT. Alternative excitotoxic hypothesis. sodium-calcium ion channel modulator RS-87476. Radio-
Neurology 1992; 42: 733–738 logy 1991; 179: 221–227
Andersen DL, Tannenberg AEG, Burke CJ, Dodd PR. Regional Levin SD. Mechanisms of damage in the developing brain. Curr
development of glutamate-N-methyl-D-aspartate receptor Opin Neurol Neurosurg 1991; 4: 371–376
sites in asphyxiated newborn infants. J Child Neurol 1998; Lockerbie RO. Neurotrophic factors and development. Curr
13: 149–157 Opin Neurol Neurosurg 1990; 3: 955–958
Chan PH, Fishman RA. Transient formation of superoxide radi- Lundgren J, Zhang H, Agardh CD, Smith ML, Evans PJ, Halliwell
cals in polyunsaturated fatty acid-induced brain swelling. B, Siesjö BK. Acidosis-induced ischemic brain damage: are
J Neurochem 1980; 35: 1004–1007 free radicals involved? J Cereb Blood Flow Metab 1991; 11:
Chan PH,Yurko M, Fishman RA. Phospholipid degradation and 587–596
cellular edema induced by free radicals in brain cortical Marret S, Gressens P, Gadisseux J-F, Evrard P. Prevention by
slices. J Neurochem 1982; 38: 525–531 magnesium of excitotoxic neuronal death in the develop-
Chan PH, Schmidley JW, Fishman RA, Longar SM. Brain injury, ing brain: an animal model for clinical intervention studies.
edema,and vascular permeability changes induced by oxy- Dev Med Child Neurol 1995; 37: 473–484
gen-derived free radicals. Neurology 1984; 34: 315–320 Mattson MP. Cellular signaling mechanisms common to the
Chen M, Bullock R, Graham DI, Frey P, Lowe D, McCulloch J. Eval- development and degeneration of neuroarchitecture.
uation of a competitive NMDA antagonist (D-CPPene) in fe- A review. Mech Ageing Dev 1989; 50: 103–157
line focal cerebral ischemia. Ann Neurol 1991; 30: 62–70 Mattson MP. Excitatory amino acids, growth factors, and calci-
Clark GD. Role of excitatory amino acids in brain injury caused um: a teeter-totter model for neural plasticity and degener-
by hypoxia-ischemia, status epilepticus, and hypoglycemia. ation. ln: Ben-Ari Y, ed. Excitatory amino acids and neuronal
Clin Perinatol 1989; 16: 459–474 plasticity. New York: Plenum Press, 1990: 211–220
Cotman CW, Monaghan DT, Ottersen OP, Storm-Mathisen J. McCord JM. Oxygen-derived free radicals in postischemic tis-
Anatomical organization of excitatory amino acid recep- sue injury. N Engl J Med 1985; 312: 159–163
tors and their pathways.Trends Neurosci 1987; 10: 273–280 Meldrum BS. Excitatory amino acid receptors and disease. Curr
Doble A. Excitatory amino acid receptors and neurodegenera- Opin Neurol Neurosurg 1992; 5: 508–513
tion.Thérapie 1995; 50: 319–337 Mishra OP, Delivoria-Papadopoulos M. Cellular mechanisms of
Foster AC, Donald AE, Willis CL, Tridgett R, Kemp JA, Priestley T. hypoxic injury in the developing brain. Brain Res Bull 1999;
The glycine site on the NMDA receptor: pharmacology and 48: 233–237
involvement in NMDA receptor-mediated neurodegenera- Mitrovic B, Ignarro LJ, Montestruque S, Smoll A, Merrill JE. Nitric
tion. In: Ben-Ari Y, ed. Excitatory Amino Acids and Neuronal oxide as a potential pathological mechanism in demyelina-
Plasticity. New York: Plenum Press, 1990: 93–100 tion: its differential effects on primary glial cells in vitro.
Freund TF, Buzsaki G, Leon A, Baimbridge KG, Somogyi P. Rela- Neuroscience 1994; 61: 575–585
tionship of neuronal vulnerability and calcium binding Nedelcu J, Klein MA, Aguzzi A, Boesiger P, Martin E. Biphasic
protein immunoreactivity in ischemia. Exp Brain Res 1990; edema after hypoxic-ischemic brain injury in neonatal rats
83: 55–66 reflects early neuronal and late glial damage. Pediatr Res
Goldberg MP, Choi DW. Intracellular free calcium increases in 1999; 46: 297–304
cultured cortical neurons deprived of oxygen and glucose. Odeh M. The role of reperfusion-induced injury in the patho-
Stroke 1990; 21 (suppl III): 75–77 genesis of the Crush syndrome. N Engl J Med 1991; 324:
Groenendaal F, Mishra OP, McGowan JE, Hoffman DJ, Delivoria- 1417–1422
Papadopoulos M. Function of cell membranes in cerebral Omar R, Pappolla M. Oxygen free radicals as inducers of heat
cortical tissue of newborn piglets after hypoxia and inhibi- shock protein synthesis in cultured human neuroblastoma
tion of nitric oxide synthase. Pediatr Res 1997; 42: 174–179 cells: relevance to neurodegenerative disease.Eur Arch Psy-
Hasegawa K, Yoshioka H, Sawada T, Nishikawa H. Lipid peroxi- chiatry Clin Neurosci 1993; 242: 262–267
dation in neonatal mouse brain subjected to two different Riikonen RS, Kero PO,Simell OG.Excitatory amino acids in cere-
types of hypoxia. Brain Dev 1991; 13: 101–103 brospinal fluid in neonatal asphyxia. Pediatr Neurol 1992; 8:
Hattori H, Wasterlain CG. Excitatory amino acids in the devel- 37–40
oping brain: ontogeny, plasticity, and excitotoxicity. Pediatr
Neurol 1990; 6: 219–228
1042 References and Further Reading

Rothman S. Synaptic release of excitatory amino acid neuro- Amess PN,Wylezinska M, Lorek A,Townsend J,Wyatt JS, Amiel-
transmitter mediates anoxic neuronal death. J Neurosci Tison C, Cady EB, Stewart A. Early brain proton magnetic
1984; 4: 1884–1891 resonance spectroscopy and neonatal neurology related
Rothman SM.Synaptic activity mediates death of hypoxic neu- to neurodevelopmental outcome at 1 year in term infants
rons. Science 1983; 220: 536–537 after presumed hypoxic-ischaemic brain injury. Dev Med
Rothman SM, Olney JW. Glutamate and the pathophysiology Child Neurol 1999; 41: 436–445
of hypoxic-ischemic brain damage. Ann Neurol 1986; 19: Arzoumanian Y, Mirmiran M, Barnes PD, Woolley K, Ariagno RL,
105–111 Moseley ME, Fleisher BE, Atlas SW. Diffusion tensor brain
Rothman SM, Olney JW. Excitotoxicity and the NMDA receptor. imaging findings at term-equivalent age may predict neu-
Trends Neurosci 1987; 10: 299–302 rologic abnormalities in low birth weight preterm infants.
Rothman SM, Thurston JH, Hauhart RE. Delayed neurotoxicity AJNR Am J Neuroradiol 2003; 24: 1646–1653
of excitatory amino acids in vitro. Neurosci 1987; 22: Azzarelli B, Caldemyer KS, Philipps JP, deMyer WE. Hypoxic-is-
471–480 chemic encephalopathy in areas of primary myelination. A
Sauer D, Allegrini PR,Thedinga KH, Massieu L, Amacker H, Fagg neuroimaging and PET study. Pediatr Neurol 1996; 14:
GE. Evaluation of quinolinic acid induced excitotoxic neu- 108–116
rodegeneration in rat striatum by quantitative magnetic Back SA, Gan X, Li Y, Rosenberg PA,Volpe JJ. Maturation-depen-
resonance imaging in vivo. J Neurosci Methods 1992; 42: dent vulnerability of oligodendrocytes to oxidative stress-
69–74 induced death caused by glutathione depletion. J Neurosci
Schwarcz R,Meldrum B.Excitatory amino acid antagonists pro- 1998; 18: 6241–6253
vide a therapeutic approach to neurological disorders. Back SA, Luo NL, Borenstein NS, Levine JM,Volpe JJ, Kinney HC.
Lancet 1985; II: 140–143 Late oligodendrocyte progenitors coincide with the devel-
Shaw PJ. Excitatory amino acid neurotransmission, excitotoxi- opmental window of vulnerability for human perinatal
city and excitotoxins. Curr Opin Neurol Neurosurg 1992; 5: white matter injury. J Neurosci 2001; 21: 1302–1312
383–390 Baenziger O, Martin E, Steinlin M, Good M, Largo R, Burger R,
Siesjö BK. Cerebral circulation and metabolism. J Neurosurg Fanconi S, Duc G, Buchli R, Rumpel H, Boltshauser F. Early
1984; 60: 883–908 pattern recognition in severe perinatal asphyxia:a prospec-
Siesjö BK. Pathophysiology and treatment of focal cerebral is- tive MRI study. Neuroradiology 1993; 35: 437–442
chemia. Part I: pathophysiology. J Neurosurg 1992; 77: Barkovich AJ. MR and CT evaluation of profound neonatal and
169–184 infantile asphyxia. AJNR Am J Neuroradiol 1992; 13: 959–
Siesjö BK. Pathophysiology and treatment of focal cerebral 972
ischemia. Part II: mechanisms of damage and treatment. Barkovich AJ, Sargent SK. Profound asphyxia in the premature
J Neurosurg 1992; 77: 337–354 infant: imaging findings. AJNR Am J Neuroradiol 1995; 16:
Simon RP, Swan JH, Griffiths T, Meldrum BS. Blockade of N- 1837–1846
methyl-D-aspartate receptors may protect against is- Barkovich AJ,Truwit CL. Brain damage from perinatal asphyxia:
chemic damage in the brain. Science 1984; 226: 850–852 correlation of MR findings with gestational age. AJNR Am J
Sims NR. Energy metabolism and selective neuronal vulnera- Neuroradiol 1990; 11: 1087–1096
bility following global cerebral ischemia. Neurochem Res Barkovich AJ,Westmark K, Partridge C, Sola A, Ferriero DM.Peri-
1992; 17: 923–931 natal asphyxia: MR findings in the first 10 days. AJNR Am J
Stephenson FA. Neurotransmitter receptors. Curr Opin Neurol Neuroradiol 1995; 16: 427–438
Neurosurg 1990; 3: 951–954 Barkovich AJ, Hajnal BL, Vigneron D, Sola A, Partridge JC, Allen
Stys PK,Waxman SG,Ransom BR.Na+-Ca2+ exchanger mediates F, Ferriero DM. Prediction of neuromotor outcome in peri-
Ca2+ influx during anoxia in mammalian central nervous natal asphyxia: evaluation of MR scoring systems. AJNR Am
system white matter. Ann Neurol 1991; 30: 375–380 J Neuroradiol 1998; 19: 143–149
Turski L, Turski WA. Towards an understanding of the role of Barth PG. Prenatal clastic encephalopathies. Clin Neurol Neu-
glutamate in neurodegenerative disorders: energy metab- rosurg 1984; 86: 65–75
olism and neuropathology. Experientia 1993; 49: 1064– Barth PG, Valk J, Olislagers-de Slegte R. Central cortico-subcor-
1072 tical pattern on CT in cerebral palsy.Its relevance to asphyx-
Wieloch T. Neuronal injury and cerebrovascular disorders. Curr ia. J Neuroradiol 1984; 111: 65–71
Opin Neurol Neurosurg 1990; 3: 944–950 Bozzao A,Di Paolo A,Mazzoleni C,Fasoli F,Simonetti A,Fantozzi
Yoshida S, Abe K, Busto R, Watson BD, Kogure K, Ginsberg MD. LM, Floris R. Diffusion-weighted MR imaging in the early di-
Influence of transient ischemia on lipid-soluble antioxi- agnosis of periventricular leukomalacia. Eur Radiol 2003;
dants, free fatty acids and energy metabolites in rat brain. 13: 1571–1576
Brain Res 1982; 245: 307–316 Bydder GM, Rutherford MA, Cowan FM. Diffusion-weighted
Young RSK,Petroff OAC,Aquila WJ,Yates J.Effects of glutamate, imaging in neonates. Childs Nerv Syst 2001; 17: 190–194
quisqualate, and N-methyl-D-aspartate in neonatal brain. Christophe C, Clercx A, Blum D, Hasearts D, Segebarth C, Perl-
Exp Neurol 1991; 111: 362–368 mutter N. Early MR deletion of cortical and subcortical hy-
poxic-ischemic encephalopathy in full-term infants. Pediatr
Radiol 1994; 24: 581–584
Counsell SJ, Allsop JM, Harrison MC, Larkman DJ, Kennea NL,
95 Posthypoxic-ischemic Encephalopathy Kapellou O, Cowan FM, Hanjal JV, Edwards AD, Rutherford
of Neonates MA. Diffusion-weighted imaging of the brain in preterm in-
fants with focal and diffuse white matter abnormalities.
Amato M, Donati F. Update on perinatal hypoxic insult: mecha- Pediatrics 2003; 112: 1–7
nism, diagnosis and interventions. Eur J Paediatr Neurol
2000; 4: 203–209
References and Further Reading 1043

Cowan F, Rutherford M, Groenendaal F, Eken P, Mercuri E, Byd- Hüppi PS, Barnes PD. Magnetic resonance techniques in the
der GM, Meiners LC, Dubowitz LM, de Vries LS. Origin and evaluation of the newborn brain. Clin Perinatol 1997; 24:
timing of brain lesions in term infants with neonatal en- 693–723
cephalopathy. Lancet 2003; 361: 736–742 Inage YW, Itoh M, Takashima S. Correlation between cere-
Cowan FM, Pennock JM, Hanrahan JD, Manji KP, Edwards AD. brovascular maturity and periventricular leukomalacia.
Early detection of cerebral infarction and hypoxic ischemic Pediatr Neurol 2000; 22: 204–208
encephalopathy in neonates using diffusion-weighted Inder TE,Huppi PS,Warfield S,Kikinis FR,Zientara GP,Barnes PD,
magnet resonance imaging.Neuropediatrics 1994; 25:172– Jolesz F, Volpe JJ. Periventricular white matter injury in the
175 premature infant is followed by reduced cerebral cortical
Dammann O, Hagberg H, Leviton A.Is periventricular leukoma- gray matter volume at term. Ann Neurol 1999; 46: 755–760
lacia an axonopathy as well as an oligopathy. Pediatr Res Inder TE, Mocatta T, Darlow B, Spencer C,Volpe JJ,Winterbourn
2001; 49: 453–457 C. Elevated free radical products in the cerebrospinal fluids
De Reuck J. The human periventricular arterial blood supply of VLBW infants with cerebral white matter injury. Pediatr
and the anatomy of cerebral infarctions. Eur Neurol 1971; 5: Res 2002; 52: 213–218
321–334 Inder TE, Wells SJ, Mogridge NB, Spencer C, Volpe JJ. Defining
De Reuck J, Chattha AS, Richardson EP Jr. Pathogenesis and the nature of the cerebral abnormalities in the premature
evaluation of periventricular leukomalacia in infancy. Arch infant: a quantitative magnetic resonance imaging study.
Neurol 1972; 27: 27: 229–236 J Pediatr 2003; 143: 171–179
De Reuck JL. Cerebral angioarchitecture and perinatal brain le- Johnston MV. Selective vulnerability in the neonatal brain. Ann
sions in premature and full-term infants. Acta Neurol Scand Neurol 1998; 44: 155–156
1984; 70: 391–395 Jouvet P, Cowan FM, Cox PH, Lazda E, Rutherford MA, Wig-
De Vries LS, Wigglesworth JS, Regev R, Dubowitz LMS. Evolu- glesworth J, Mehmet H, Edwards AD. Reproducibility and
tion of periventricular leukomalacia during the neonatal accuracy of MR imaging of the brain after severe birth as-
period and infancy: correlation of imaging and post- phyxia. Am J Neuroaradiol 1999; 20: 1343–1348
mortem findings. Early Hum Dev 1988; 17: 205–219 Krägeloh-Mann I, Hagberg B, Petersen D, Riethmüller J, Gut E,
De Vries LS, Eken P. Groenendaal Fl, van Haastert IC, Meiners LC. Michaelis R. Bilateral spastic cerebral palsy – pathogenetic
Correlation between the degree of periventricular leuko- aspects from MRI. Neuropediatrics 1991; 23: 46–48
malacia diagnosed using cranial ultrasound and MRI later Krägeloh-Mann I, Petersen D, Hagberg G,Vollmer B, Hagberg B,
in infancy in children with cerebral palsy. Neuropediatrics Michaelis R. Bilateral spastic cerebral palsy – MRI pathology
1993; 24: 263–268 and origin. Analysis from a representative series of 56 cas-
Du Plessis AJ, Johnston MV. Hypoxic-ischemic brain injury in es. Dev Med Child Neurol 1995; 37: 379–397
the newborn. Cellular mechanisms and potential strategies Kuban K, Sanocka U, Leviton A, Allred EN, Pagano M, Dammann
for neuroprotection. Clin Perinatol 1997; 24: 627–654 O,Share J,Rosenfeld D,Abiri M,diSalvo D,Doubilet P,Kairam
Felderhoff-Mueser U, Rutherford MA, Squier WV, Cox P, Maalouf R, Kazam E, Kirpekar M, Schonfeld S.White matter disorders
EF, Counsell SJ, Bydder GM, Edwards AD. Relationship be- of prematurity: association with intraventricular hemor-
tween MR imaging and histopathologic findings of the rhage and ventriculomegaly. J Pediatr 1999; 134: 539–546
brain in extremely sick preterm infants. AJNR Am J Neuro- Leth H,Toft PB, Pryds O, Peitersen B, Lou HC, Henriksen O. Brain
radiol 1999; 20: 1349–1357 lactate in preterm and growth-retarded neonates. Acta
Fellman V, Raivio KO. Reperfusion injury as the mechanism of Paediatr 1995; 84: 495–499
brain damage after perinatal asphyxia.Pediatr Res 1997; 41: Maalouf EF, Duggan PH, Rutherford MA, Counsell SJ, Fletcher
599–606 AM, Battin M, Cowan F, Edwards AD. Magnetic resonance
Filippi CG, Lin DDM, Tsiouris AJ, Watts R, Packard AM, Heier LA, imaging of the brain in a cohort of extremely preterm in-
Uluğ AM. Diffusion-tensor MR imaging in children with de- fants. J Pediatr 1999; 135: 351–357
velopmental delay: preliminary findings. Radiology 2003; Mader I, Schöning M, Klose U, Küker W. Neonatal cerebral in-
229: 44–50 farction diagnosed by diffusion-weighted MRI. Pseudonor-
Forbes KPN, Pipe JG, Bird R. Neonatal hypoxic-ischemic en- malization occurs early. Stroke 2002; 33: 1142–1145
cephalopathy: detection with diffusion-weighted MR Marín-Padilla M. Developmental neuropathology and impact
imaging. AJNR Am J Neuroradiol 2000; 21: 1490–1496 of perinatal brain damage. II: white matter lesions of the
Grow J, Barks JDE. Pathogenesis of hypoxic-ischemic cerebral neocortex. J Neuropathol Exp Neurol 1997; 56: 219–235
injury in the term infant: current concepts. Clin Perinatol Martin E, Barkovich AJ. Magnetic resonance imaging in perina-
2002; 29: 585–602 tal asphyxia. Arch Dis Child 1995; 72: F62-F70
Gücüyener K, Atalay Y, Aral YZ, Hasanoğlu A, Türkyilmaz C, Mathew OP, Bland H, Boxerman SB, James E. CSF lactate levels
Biberoğlu G. Excitatory amino acids and taurine levels in in high risk neonates with and without asphyxia. Pediatrics
cerebrospinal fluid of hypoxic ischemic encephalopathy in 1980; 66: 224–227
newborn. Clin Neurol Neurosurg 1999; 101: 171–174 Mayer PL, Kier EL. The controversy of the periventricular white
Guzzetta F, Deodato F, Randò T. Brain ischemic lesions of the matter circulation: a review of the anatomic literature.AJNR
newborn. Childs Nerv Syst 2000; 16: 633–637 Am J Neuroradiol 1991; 12: 223–228
Haynes RL, Folkerth RD, Keefe RJ, Sung I, Swzeda LI, Rosenberg McArdle CB, Richardson CJ, Hayden CK, Nicholas DA, Crofford
PA, Volpe JJ, Kiney HC. Nitrosative and oxidative injury to MJ, Amparo EG. Abnormalities of the neonatal brain: MR
premyelinating oligodendrocytes in periventricular leuko- imaging. I. Intracranial hemorrhage. Radiology 1987; 163:
malacia. J Neuropathol Exp Neurol 2003; 62: 441–450 387–394
Huang C-C,Wang S-T, Chang Y-C, Lin K-P,Wu P-L. Measurement Mercuri E, Atkinson J, Braddick O, Anker S, Cowan F, Pennock J,
of the urinary lactate: creatinine ratio for the early identifi- Rutherford MA, Dubowitz LMS.The aetiology of delayed vi-
cation of newborn infants at risk for hypoxic-ischemic en- sual maturation: short review and personal findings in rela-
cephalopathy. N Engl J Med 1999 341: 328–335 tion to magnetic resonance imaging. Eur J Paediatr Neurol
1997; 1: 31–34
1044 References and Further Reading

Mercuri E, Atkinson J, Braddick O, Anker S, Cowan F, Rutherford Shevell MI, Majnemer A, Miller SP. Neonatal neurologic prog-
M, Pennock J, Dubowitz L. Basal ganglia damage and im- nostication: the asphyxiated term newborn. Pediatr Neurol
paired visual function in the newborn infant. Arch Dis Child 1999; 21: 776–784
1997; 77: F111-F114 Sie LTL, van der Knaap MS, Oosting J, de Vries LS, Lafeber HN,
Müller AM, Morales C, Briner J, Baenziger O, Duc G, Bucher HU. Valk J. MR patterns of hypoxic-ischemic brain damage after
Loss of CO2 reactivity of cerebral blood flow is associated prenatal, perinatal or postnatal asphyxia. Neuropediatrics
with severe brain damage in mechanically ventilated very 2000; 31: 128–136
low birth weight infants. Eur J Paediatr Neurol 1997; 5/6: Sie LTL, van der Knaap MS, van Wezel-Meijler G, Taets van
157–163 Amerongen AHM, Lafeber HN, Valk J. Early MR features of
Nakai T, Rhine WD, Enzmann DR, Stevenson DK, Spielman DM. hypoxic–ischemic brain injury in neonates with periven-
A model for detecting early metabolic changes in neonatal tricular densities on sonograms. AJNR Am J Neuroradiol
asphyxia by 1H-MRS. J Magn Reson Imaging 1996; 6: 445– 2000; 21: 852–861
452 Simonati A, Laverda AM, Rizzuto N. Mulicystic encephalomala-
Natsume J,Wanatabe K,Kuno K,Hayakawa F,Hashizume Y.Clin- cia associated with symmetrical necrotizing brain stem le-
ical, neuropsychologic, and neuropathological features of sions in an infant: a case report. Clin Neuropathol 1986; 5:
an infant with brain damage of total asphyxia type (Myers). 139–145
Pediatr Neurol 1995; 61–64 Skranes SJ, Vik T, Nilsen G, Smevik O, Andersson HW, Brubakk
Nelson MD, Gonzalez-Gomez I, Gilles FH.The search for human AM. Cerebral magnetic resonance imaging and motor
telencephalic ventriculofugal arteries. AJNR Am J Neuro- function of a very low birth weight children at six years of
radiol 1991 12: 215–222 age. Neuropediatrics 1997; 28: 149–154
Pasternak JF, Predey TA, Mikhael MA. Neonatal asphyxia: Skullerud K, Westre B. Frequency and prognostic significance
vulnerability of basal ganglia, thalamus, and brainstem. of germinal matrix hemorrhage, periventricular leukomala-
Pediatr Neurol 1991; 7: 147–149 cia, and pontosubicular necrosis in preterm neonates. Acta
Penrice J, Lorek A, Cady EB, Amess PN, Wylezinska M, Cooper Neuropathol (Berl) 1986; 70: 257–261
CE, d’Souza P, Brown CG, Kirbride V, Edwards AD, Wyatt JS, Sohma O, Mito T, Mizuguchi M, Takashima S. The prenatal age
Reynolds EOR. Proton magnetic resonance spectroscopy of critical for the development of the pontosubicular necrosis.
the brain during acute hypoxia-ischemia and delayed cere- Acta Neuropathol (Berl) 1995; 90: 7–10
bral energy failure in the newborn piglet. Pediatr Res 1997; Takashima S,Tanaka K. Development of cerebrovascular archi-
41: 795–802 tecture and its relationship to periventricular leukomalacia.
Perlman JM. Markers of asphyxia and neonatal brain injury. Arch Neurol 1978; 35: 11–16
N Engl J Med 1999; 341: 364–365 Truwit CL, Barkovich AJ, Koch TK, Ferriero DM. Cerebral palsy:
Ramakers RP, van der Knaap MS, Verbeeten B Jr, Barth PG, Valk MR findings in 40 patients.AJNR Am J Neuroradiol 1992; 13:
J. Central cortico-subcortical involvement: a distinct pat- 67–78
tern of brain damage caused by perinatal and postnatal Volpe JJ. Current concepts of brain injury in the premature
asphyxia in term infants. J Comput Assist Tomogr 1995; 19: infant. AJR Am J Roentgenol 1989; 153: 243–251
256–263 Uluğ AM. Monitoring brain development with quantitative dif-
Robertson RL, Ben-Sira L, Barnes PD, Mulkern RV, Robson CD, fusion tensor imaging. Dev Sci 2002; 5: 286–292
Maier SE, Rivkin MJ, du Plessis AJ. MR line-scan diffusion- Valk J, van der Knaap MS, de Grauw T, Taets van Amerongen
weighted imaging of term neonates with perinatal brain AHM. The role of imaging modalities in the diagnosis of
ischemia. AJNR Am J Neuroradiol 1999; 20: 1658–1670 posthypoxic-ischemic and hemorrhagic conditions of in-
Roelants-van Rijn AM, Groenendaal F, Beek FJA, Eken P, van fants (part I of II). Klin Neuroradiol 1991; 1: 72–79
Haastert IC, de Vries LS. Parenchymal brain injury in the Valk J, van der Knaap MS, de Grauw T, Taets van Amerongen
preterm infant: comparison of cranial ultrasound, MRI and AHM. The role of imaging modalities in the diagnosis of
neurodevelopmental outcome. Neuropediatrics 2001; 32: posthypoxic-ischemic and hemorrhagic conditions of in-
80–89 fants (part II of II). Klin Neuroradiol 1991; 1: 127–138
Roland EH, Poskitt K, Rodriguez E, Lupton BA, Hill A. Perinatal Valkama AM, Pääkkö ELE, Vainionpää LK, Lanning FP, Ilkko EA,
hypoxic-ischemic thalamic injury: clinical features and neu- Koivisto ME. Magnetic resonance imaging at term and neu-
roimaging. Ann Neurol 1998; 44: 161–166 romotor outcome in preterm infants. Acta Paediatr 2000;
Rorke LB, Zimmerman RA. Prematurity, postmaturity, and de- 89: 348–355
structive lesions in utero. AJNR Am J Neuroradiol 1992; 13: Vermeulen RJ, Fetter WPF, Hendrikx L, van Schie PEM, van der
517–536 Knaap MS, Barkhof F. Diffusion-weighted MRI in severe
Rutherford MA, Pennock JM, Schwieso JE, Cowan FM, neonatal hypoxic ischeamia: the white cerebrum. Neurope-
Dubowitz LMS. Hypoxic ischaemic encephalopathy: early diatrics 2003; 34: 72–76
magnetic resonance imaging findings and their evolution. Victory R, Penava D, da Silva O, Natale R, Richardson B. Umbili-
Neuropediatrics 1995; 26: 183–191 cal cord pH and base excess values in relation to neonatal
Rutherford M, Pennock J, Schwieso J, Cowan F, Dubowitz L. Hy- morbidity for infants delivered preterm. Am J Obstet Gy-
poxic-ischaemic encephalopathy: early and late magnetic necol 2003; 189: 803–807
resonance imaging findings in relation to outcome. Arch Vintzileos AM,Gaffney SE,Salinger LM,Kontopoulos VG,Camp-
Dis Child 1996; 75: F145-F151 bell WA, Nochimson DJ. The relationships among the fetal
Sarnat HB, Sarnat MS. Neonatal encephalopathy following fe- biophysical profile, umbilical cord pH, and Apgar scores.
tal distress. Arch Neurol 1976; 33: 696–705 Am J Obstet Gynecol 1987; 157: 627–631
Sawada H, Udaka F, Seriu N, Shindou K, Kameyama M, Tsujimu- Voit T, Lemburg P. Damage of thalamus and basal ganglia in
ra M. MRI demonstration of cortical laminar necrosis and asphyxiated full-term neonates. Neuropediatrics 1987; 18:
delayed white matter injury in anoxic encephalopathy. 176–181
Neuroradiology 1990; 32: 319–321 Volpe JJ. Bain injury in the premature infant. Clin Perinatol
1997; 24: 567–587
References and Further Reading 1045

Volpe JJ. Brain injury in the premature infant – from pathogen- Murakami Y,Yamashita Y,Matsuishi T,Utsunomiya H,Okudera T,
esis to prevention. Brain Dev 1997; 19: 519–534 Hashimoto T. Cranial MRI of neurologically impaired chil-
Volpe JJ. Neurobiology of periventricular leukomalacia in the dren suffering from neonatal hypoglycaemia. Pediatr Radi-
premature infant. Pediatr Res 2001; 50: 553–562 ol 1999; 29: 23–27
Volpe JJ. Perinatal brain injury: from pathogenesis to neuro- Pildes RS, Cornblath M,Warren I, et al., A prospective controlled
protection. Mental Retard Dev Disabil Res Rev 2001; 7: study of neonatal hypoglycaemia. Pediatrics 1974; 54: 5–14
56–64 Pryds O, Greisen G, Friis-Hansen B. Compensatory increase
Westmark KD, Barkovich AJ, Sola A, Ferriero D, Partridge JC. Pat- of CBF in preterm infants during hypoglycaemia. Acta
terns and implications of MR contrast enhancement in Paediatr Scand 1988; 77: 632–637
perinatal asphyxia: a preliminary report. AJNR Am J Neuro- Spar JA, Lewine JD, Orrison WW. Neonatal hypoglycaemia:
radiol 1995; 16: 685–692 CT and MR findings. AJNR Am J Neuroradiol 1994; 15:
Wood NS, Marlow N, Costeloe K, Gibson AT, Wilkinson AR. Neu- 1477–1478
rologic and developmental disability after extremely Suhonen-Polvi H, Ruotsalainen U, Kinnala A, Bergman J, Haa-
preterm birth. N Engl J Med 2000; 343: 378–384 paranta MM, Makel AP, Solin O,Wegelius P. FDG-PET in early
Yoshioka H, Kadomoto Y, Mino M, Morikawa Y, Kasubuchi Y, infancy: simplified quantification methods to measure
Kusunoki T. Multicystic encephalomania in liveborn twin cerebral glucose utilization. J Nucl Med 1995; 13: 1249–
with stillborn macerated co-twin. J Pediatr 1979; 95: 1254
798–800 Traill Z, Squier M, Anslow P. Brain imaging in neonatal hypo-
glycaemia. Arch Dis Child Fetal Neonatal Ed 1998; 79: F145–
F147
Van der Knaap MS, Bakker HD, Valk J. MR imaging and proton
96 Neonatal Hypoglycemia spectroscopy in 3-hydroxy-3-methylglutaryl coenzyme A
lyase deficiency. AJNR Am J Neuroradiol 1998; 19: 378–382
Abdul-Rahman A, Agarth A, Siesjo BK. Local cerebral blood Vannucci SJ. Developmental expression of GLUT1 and GLUT3
flow in the rat during severe hypoglycaemia and the recov- glucose transporters. J Neurochem 1994; 62: 240–246
ery period following glucose injection. Acta Physiol Scand Vannucci SJ, Maher F, Simpson IA. Glucose transporter proteins
1980; 109: 307–314. in brain: delivery of glucose to neurons and glia. Glia 1997;
Anderson JM, Milner RDG, Strich SJ. Effects of neonatal hypo- 21: 2–21
glycaemia on the central nervous system: a pathological Volpe JJ. Neurology of the newborn, 4th ed. Philadelphia: WB
study. J Neurol Neurosurg Psychiatry 1967; 30: 295–310 Saunders Co, 2002
Antunes NL, Small TN, George D, Boulad F, Lis E. Posterior Zammarchi E, Filippi L, Fonda C, Benedetti PA, Pistone D, Donati
leukoencephalopathy syndrome may not be reversible. MA. Different neurologic outcomes in two patients with
Pediatr Neurol 1999; 20: 241–3 neonatal hyperinsulinemic hypoglycaemia. Childs Nerv
Anwar M, Vannucci RC. Autoradiographic determination of re- Syst 1996; 12: 413–416
gional cerebral blood flow during hypoglycaemia in new-
born dogs. Pediatr Res 1988; 24: 41–45
Auer RN, Wieloch T, Olsson Y, Siesjö BK. The distribution of hy-
poglycaemic brain damage. Acta Neuropathol (Berl) 1984; 97 Delayed Posthypoxic Leuko-
64: 177–191 encephalopathy
Auer RN, Kalimo H, Olsson Y, Siesjö BK. The temporal evolution
of hypoglycaemic brain damage. Acta Neuropathol (Berl) Chalela JA, Wolf RL, Maldijian JA, Kasner SE. MRI identification
1985;67:13–24 of early white matter injury in anoxic-ischemic encephalo-
Barkovich AJ, Ali FA, Rowley HA, Bass N. Imaging patterns of pathy. Neurology 2001; 56: 481–485
neonatal hypoglycaemia. AJNR Am J Neuroradiol 1998; 19: Chang KH, Han MH, Kim HS, Wie BA, Han MC. Delayed en-
523–528 cephalopathy after acute carbon monoxide intoxication:
Chase HP, Marlow RA, Dabiere CS et al. Hypoglycaemia and MR imaging features and distribution of cerebral white
brain development. Pediatrics 1973; 52: 513–520 matter lesions. Radiology 1992; 184: 117–122
Chugani HT, Phelps ME, Mazziotta JC. Positron emission to- De Reuck J. Arterial vascularisation and angioarchitecture of
mography study of the human brain functional develop- the nucleus caudatus in human brain. Eur Neurol 1971; 5:
ment. Ann Neurol 1987; 22: 487–497 130–136
Collins JE, Leonard JV. Hyperinsulinism in asphyxiated and De Reuck J, van der Eecken H. Periventricular leukoencephalo-
small-for dates infants with hypoglycaemia. Lancet 1984; 2; pathy and meningo-cortical arterio-venous malformation.
311–313 Acta Neurol Belg 1974; 74: 276–283
Fahnehjelm KT, Jacobson L, Hellstrom A, Lewensohn-Fuchs I, Devine SA, Kirkley SM, Palumbo CL, White RF. MRI and neu-
Ygge J. Visually impaired children with posterior ocular ropsychological correlates of carbon monoxide exposure: a
malformations: pre- and neonatal data and visual func- case report. Environ Health Perspect 2002; 110: 1051–1055
tions. Acta Ophthalmol Scand 2003; 81: 361–72. Dragonesco ST, Nereantiu FL, Vuia O. Encéphalopathy hypox-
Hawdon JM. Hypoglycemia and the neonatal brain. Eur J Pedi- ique prolongée. Acta Neuropathol (Berl) 1964; 3: 387–391
atr 1999; 158: S9-S12 Elovaara E, Rantanen J. Carbon monoxide-induced brain in-
Ichord RN, Helfaer MA, Kirsch JR, Wilson D, Traystman RJ. Nitric jury: neurochemical studies after single and repeated ex-
oxide synthetase inhibition attenuates hypoglycaemic hy- posures. J Appl Toxicol 1983; 3: 154–160
peremia in piglets. Am J Physiol 1994; 266; 1062–1068 Feigin I, Budzilovich G, Weinberg S, Ogata J. Degeneration of
Lucas A, Morley R, Cole TJ. Adverse neurodevelopmental out- white matter in hypoxia, acidosis and edema. J Neu-
come in moderate neonatal hypoglycaemia.BMJ 1988; 297: ropathol Exp Neurol 1973; 32: 125–143
1304–1308 Garland H, Pearce J. Neurological complications of carbon
monoxide poisoning. Q J Med 1967; 36: 445–455
1046 References and Further Reading

Ginsberg MD, Myers RE, McDonagh BF. Experimental carbon Okeda R, Funata N,Takano T, Miyazaki Y, Higashino F,Yokoyama
monoxide. Eencephalopathy in the primate. I. Physiologic K, Manabe M. The pathogenesis of carbon monoxide en-
and metabolic aspects. Arch Neurol 1974; 30: 202–208 cephalopathy in the acute phase – physiological and mor-
Ginsberg MD, Myers RE, McDonagh BF. Experimental carbon phological correlation. Acta Neuropathol (Berl) 1981; 54:
monoxide. Encephalopathy in the primate. II. Clinical as- 1–10
pects, neuropathology, and physiologic correlation. Arch Okeda R, Funata N, Song S-J, Higashino F, Takano T, Yokoyama
Neurol 1974; 30: 209–216 K. Comparative study on pathogenesis of selective cerebral
Ginsberg MD, Hedley-Whyte ET, Richardson EP Jr. Hypoxic-is- lesions in carbon monoxide poisoning and nitrogen hy-
chemic leukoencephalopathy in man. Arch Neurol 1976; poxia in cats. Acta Neuropathol (Berl) 1982; 56: 265–272
33: 5–14 Okeda R, Song SY, Funta N, Higashino F. An experimental study
Gorman D,Huang YL,Williams C.A lignocaine infusion worsens of the pathogenesis of Grinker’s myelinopathy in carbon
the leukoencephalopathy due to a carbon monoxide expo- monoxide intoxication. Acta Neuropathol (Berl) 1983; 59:
sure in sheep.Toxicology 2003; 186: 143–150 200–206
Gottfried JA, Mayer SA, Shungu DC, Chang Y, Duyn JH. Delayed Parkinson RB, Hopkins RO, Cleavinger HB, Weaver LK, Victoroff
posthypoxic demyelination. Association with arylsulfatase J,Foley JF,Bigler ED.White matter hyperintensities and neu-
A deficiency and lactic acidosis on MR spectroscopy. Neu- ropsychological outcome following carbon monoxide poi-
rology 1997; 49: 1400–1404 soning. Neurology 2002; 58: 1525–1532
Grinker RR. Ueber einen Fall von Luchtgasvergiftung mit dop- Plum F, Posner JB, Hain RF. Delayed neurological deterioration
pelseitiger Pallidumerweichung und schwerer Degenera- after anoxia. Arch Intern Med 1962; 110: 18–25
tion des tiefen Grosshirnmarklagers. Z Ges Neurol Psychia- Prockop LD, Naidu KA. Brain CT and MRI findings after carbon
tr 1925; 98: 433–456 monoxide toxicity. J Neuroim 1999; 9: 175–181
Inagaki T, Ishino H, Seno H, Umegae N, Aoyama T. A long-term Roohi F, Kula RW, Metha N. Twenty-nine years after carbon
follow-up study of serial magnetic resonance image in pa- monoxide intoxication. Clin Neurol Neurosurg 2001; 103:
tients with delayed encephalopathy after acute carbon 92–95
monoxide poisoning. Psychiatr Clin Neurosci 1997; 51: Sawa GM, Watson CPN, Terbrugge K, Chiu M. Delayed en-
421–423 cephalopathy following carbon monoxide intoxication.
Kamada K, Houkin K, Aoki T, Koiwa M, Kashiwaba T, Iwasaki Y, J Can Sci Neurol 1981; 8: 77–79
Abe H. Cerebral metabolic changes in delayed carbon Sawada Y, Takahashi M, Ohashi N, Fusamoto H, Maemura K,
monoxide sequelae studied by proton MR spectroscopy. Kobayashi H, Yoshioka T, Sugimoto T. Computerised tomo-
Neuroradiology 1994; 36: 104–106 graphy as an indication of long-term outcome after acute
Kelafant GA. Encephalopathy and peripheral neuropathy fol- carbon monoxide poisoning. Lancet 1980; I: 783–784
lowing carbon monoxide poisening from a propane-fueled Sawada Y, Sakamoto T, Nishide K, Sadamitsu D, Fusamoto H,
vehicle. Am J Ind Med 1996; 30: 765–768 Yoshioka T,Sugimoto T,Onishi S.Correlation of pathological
Kim JH, Chang KH, Song IC, Kim KH, Kwon BJ, Kim HC, Kim JH, findings with computed tomographic findings after acute
Han MH. Delayed encephalopathy of acute carbon monox- carbon monoxide poisoning. N Engl J Med 1983; 26: 1296
ide intoxication: diffusivity of cerebral white matter lesions. Sesay M, Bidabe AM, Guyot M, Bedry R, Caille JM, Maurette P.
AJNR Am J Neuroradiol 2003; 24: 1592–1597 Regional cerebral blood flow measurements with Xenon-
Klawans HL, Stein RW, Tanner CM, Goetz CG. A pure Parkinson- CT in the prediction of delayed encephalopathy after car-
ian syndrome following acute carbon monoxide intoxica- bon monoxide intoxication. Acta Neurol Scand Suppl 1996;
tion. Arch Neurol 1982; 39: 302–304 166: 22–27
Kono E, Kono R, Shida K. Computerized tomographies of 34 pa- Silverman CS, Brenner J, Murtagh FR. Hemorrhagic necrosis
tients at the chronic stage of acute carbon monoxide poi- and vascular injury in carbon monoxide poisoning: MR
soning. Arch Psychiatr Nervenkr 1983; 233: 271–278 demonstration. AJNR Am J Neuroradiol 1993; 14: 168–170
Koehler RC, Jones MD, Traystman RJ. Cerebral circulatory re- Taylor R, Holgate RC. Carbon monoxide poisoning: asymmetric
sponse to carbon monoxide and hypoxic hypoxia in the and unilateral changes on CT. AJNR Am J Neuroradiol 1988;
lamb. Am J Physiol 1982; 243: H27-H32 9: 975–977
Ku MC, Huang CC, Kuo HC, Yen TC, Chen CJ, Shih TS, Chang HY. Teksam M, Casey SO, Michel E, Liu H, Truwit CL. Diffusion-
Diffuse white matter lesions in carbon disulfide intoxica- weighted MR images findings in carbon monoxide poison-
tion: microangiopathy or demyelination. Eur Neurol 2003; ing. Neuroradiology 2002; 44: 109–113
50: 220–224 Uchino A, Hasuo K, Shida K, Matsumoto S, Yasumori K, Masuda
Lee MS, Marsden CD. Neurological sequelae following carbon K. MRI of the brain in chronic carbon monoxide poisoning.
monoxide poisoning clinical course and outcome accord- Neuroradiology 1994; 36: 399–401
ing to the clinical types and brain computed tomography Valenzuela R, Court J, Godoy J.Delayed cyanide induced dysto-
scan findings. Mov Disord 1994; 9: 550–558 nia. J Neurol Neurosurg Psychiatry 1992; 55: 198–199
Levine S. Anoxic-ischemic encephalopathy in rats. Am J Pathol Vieregge P, Klostermann W, Blümm RG, Borgis KJ. Carbon
1960; 36: 1–18 monoxide poisoning: clinical, neurophysiological, and
Levine S, Stypulowski W. Experimental cyanide encephalopa- brain imaging observations in acute disease and follow-up.
thy. Arch Pathol 1959; 67: 306–323 J Neurol 1989; 236: 478–481
Mascalchi M, Petruzzi P, Zampa V. MRI of cerebral white matter Vion-Dury J, Jiddane M, van Bunnen Y, Rumeau C, Lavielle J.
damage due to carbon monoxide poisoning: case report. Etude IRM des séquelles d’intoxication au monoxide de
Neuroradiology 1996; 38: S73-S74 carbone: à propos de deux cas. J Neuroradiol 1987; 14:
Murata T, Itoh S, Koshino Y, Sakamoto K, Nishio M, Maeda M,Ya- 60–65
mada H, Ishii Y, Isaki K. Serial cerebral MRI with FLAIR se- Zagami AS, Lethlean AK, Mellick R. Delayed neurological dete-
quences in acute carbon monoxide poisoning. J Comput rioration following carbon monoxide poisoning: MRI find-
Assist Tomogr 1995; 19: 631–634 ings. J Neurol 1993; 240: 113–116
References and Further Reading 1047

98 White Matter Lesions of the Elderly De Reuck J. The human periventricular blood supply and the
anatomy of cerebral infarctions. Eur Neurol 1971; 5:
Awad IA, Spetzler RF, Hodak JA, Awad CA, Carey R. Incidental 321–334
subcortical lesions identified on magnetic resonance De Reuck J. Periventricular leukomalacia in adults. Clinico-
imaging. I. Correlation with age and cerebrovascular risk pathological study of four cases. Arch Neurol 1978; 35:
factors. Stroke 1986; 17: 1084–1089 517–521
Awad IA, Spetzler RF, Hodak JA, Awad CA,Williams F, Carey R. In- Desmond DW. Cognition and white matter lesions. Cere-
cidental lesions noted on magnetic resonance imaging of brovasc Dis 2002; 13: 53–57
the brain: prevalence and clinical significance in various Drayer BP. Imaging of the brain. I. Normal findings. Radiology
age groups. Neurosurgery 1987; 20: 222–227 1988; 166: 785–796
Barber R, Scheltens P, Gholkar A, Ballard C, McKeith I, Ince P, Per- Drayer BP. Imaging of the brain. II. Pathologic conditions. Radi-
ry R, O’Brien J.White matter lesions on magnetic resonance ology 1988; 166: 797–806
imaging in dementia with Lewy bodies, Alzheimer’s dis- Dufouil C, de Kersaint-Gilly A, Besançon V, Levy C, Auffray E,
ease, vascular dementia, and normal aging. J Neurol Neuro- Brunnereau L, Alpérovitch A, Tzourio C. Longitudinal study
surg Psychiatry 1999; 67: 66–72 of blood pressure and white matter hyperintensities. The
Bowen BC, Barker WW, Loewenstein DA, Scheldon J, Duara R. EVA MRI cohort. Neurology 2001; 56: 921–926
MR signal abnormalities in memory disorder and demen- Engelter ST, Provenzale JM, Petrella JR, DeLong DM, MacFall JR.
tia. AJNR Am J Neuroradiol 1990; 11: 283–290 The effect of aging on the apparent diffusion coefficient of
Bradley WG Jr, Waluch V, Brant-Zawadski M, Yadley RA, Wycoff normal-appearing white matter. AJR Am J Roentgenol
RR. Patchy, periventricular white matter lesions in the elder- 2000; 175: 425–430
ly: a common observation during NMR imaging. Noninvas Fazekas F, Kleinert R, Offenbacher H, Schmidt R, Kleinert G,
Med Imaging 1984; 1: 5–41 Payer F, Radner H, Lechner H. Pathological correlates of inci-
Braffman BH, Zimmerman RA, Trojanowski JQ, Gonatas NK, dental MRI white matter signal hyperintensities. Neurology
Hickey WF, Schlaepfer WW. Brain MR: pathologic correlation 1993; 43: 1683–1689
with gross and histopathology. 2. Hyperintense white-mat- Fazekas F, Schmidt R, Scheltens P. Pathophysiologic mecha-
ter foci in the elderly. AJNR Am J Neuroradiol 1988; 9: nisms in the development of age-related white matter
629–636 changes of the brain. Dement Geriatr Cogn Disord 1998; 9:
Brant-Zawadzki M, Fein G, van Dyke C, Kiernan R, Davenport L, 2–5
de Groot J. MR imaging of the aging brain: patchy white- Freeborough PA, Woods RP, Fox NC. Accurate registration of
matter lesions and dementia. AJNR Am J Neuroradiol 1985; serial 3D MR brain images and its application to visualizing
6: 675–682 changes in neurodegenerative disorders. J Comput Assist
Brown WR, Moody DM, Thore CR, Challa VR. Apoptosis in Tomogr 1996; 20: 1012–1022
leukoaraiosis. AJNR Am J Neuroradiol 2000; 21: 79–82 Fukuda H, Kitani M. Differences between treated and untreat-
Charletta D, Gorelick PB, Dollear TJ, Freels S, Harris Y. CT and MRI ed hypertensive subjects in the extent of periventricular
findings among Africa-Americans with Alzheimer’s disease, hyperintensities observed on brain MRI. Stroke 1995; 26:
vascular dementia, and stroke without dementia. Neurolo- 1593–1697
gy 1995; 45: 1456–1461 George AE, de Leon MJ, Gentes CY, Miller J, London E,
Chun T, Filippi CG, Zimmerman RD, Uluğ AM. Diffusion changes Budzilovich GN, Ferris S, Chase N. Leukoencephalopathy in
in the aging human brain. AJNR Am J Neuroradiol 2000; 21: normal and pathologic aging: 1. CT of brain lucencies. AJNR
1078–1083 Am J Neuroradiol 1986; 7: 561–566
Courchesne E, Chisum HJ, Townsend J, Cowles A, Covington J, Gerard G, Weisberg LA. MRI periventricular lesions in adults.
Egaas B, Harwood M, Hinds S, Press GA. Normal brain devel- Neurology 1986; 36: 998–1001
opment and aging: quantitative analysis at in vivo MR Golomb J, Kluger A, Gianutsos J, Ferris SH, de Leon MJ, George
imaging in healthy volunteers. Radiology 2000; 216: AE. Nonspecific leukoencephalopathy associated with
672–682 aging. Neuroimaging Clin North Am 1995; 5: 33–44
Damian MS, Schilling G, Bachmann G, Simon C, Stöppler S, Gunning-Dixon FM, Raz N. The cognitive correlates of white
Dorndorf W. White matter lesions and cognitive deficits: matter abnormalities in normal aging: a quantitative re-
relevance of lesion pattern. Acta Neurol Scand 1994; 90: view. Neuropsychology 2000; 14: 224–232
430–436 Gupta SR, Neheedy MH, Young JC, Ghobrial M, Rubino FA,
De Groot JC, de Leeuw F-E, Oudkerk M, van Gijn J, Hofman A, Hindo W.Periventricular white matter changes and demen-
Jolles J, Breteler MMB. Cerebral white matter lesions and tia. Clinical, neuropsychological, radiological, and patho-
cognitive function: the Rotterdam scan study. Ann Neurol logical correlation. Arch Neurol 1988; 45: 637–641
2000; 47: 145–151 Guttmann CRG, Jolesz FA, Kikinis R, Killiany RJ, Moss MB, Sandor
De Groot JC, de Leeuw F-E, Oudkerk M van Gijn J, Hofman A, T,Albert MS.White matter changes with normal aging.Neu-
Jolles J, Breteler MMB. Periventricular cerebral white matter rology 1998; 50: 972–978
lesions predict rate of cognitive decline. Ann Neurol 2002; Hachinski VC, Potter P, Merskey H. Leuko-araiosis. Arch Neurol
52: 335–341 1987; 44; 21–23
De Leeuw F-E, de Groot JC, Achten E, Oudkerk M, Ramos LMP, Helenius J, Sionne L, Salonen O, Kaste M, Tatlisumak T.
Heijboer R, Hofman A, Jolles J, van Gijn J, Breteler MMB. Leukoaraiosis, ischemic stroke, and normal white matter on
Prevalence of cerebral white matter lesions in elderly peo- diffusion-weighted MRI. Stroke 2002; 33; 45–50
ple: a population based magnetic resonance imaging Hunt AL, Orrison WW, Yeo RA, Haaland KY, Rhyne RL, Garry PJ,
study. The Rotterdam scan study. J Neurol Neurosurg Psy- Rosenberg GA. Clinical significance of MRI white matter
chiatry 2001; 70: 9–14 lesions in the elderly. Neurology 1989; 39: 1470–1474
Inzitari D, Diaz F, Fox A, Hachinski VC, Steingart A, Lau C, Donald
A, Wade J, Mulic H, Merskey H. Vascular risk factors and
leuko-araiosis. Arch Neurol 1987; 44: 42–47
1048 References and Further Reading

Janota I, Mirsen TR, Hachinski VC, Lee DH, Merskey H. Neu- Pullicino P, Benedict RHB, Capruso DX,Vella N,Withaim-Leith S,
ropathologic correlates of leuko-araiosis. Arch Neurol 1989; Kwen PL.Neuroimaging criteria for vascular dementia.Arch
46: 1124–1128 Neurol 1996; 53: 723–728
Ketonen LM. Neuroimaging of the aging brain. Neurol Clin Qiwen M, Xie J, Wen Z, Weng Q, Shuyun Z. A quantitative MR
North Am 1998; 16: 581–598 study of the hippocampal formation, the amygdala, and
Kertesz A, Black SE, Tokar G, Benke T, Carr T, Nicholson L. the temporal horn of the lateral ventricle in healthy sub-
Periventricular and subcortical hyperintensities on mag- jects 40–90 years of age. AJNR Am J Neuroradiol 1999; 20:
netic resonance imaging. ‘Rims, caps, and unidentified 207–211
bright objects’. Arch Neurol 1988; 45: 404–408 Salonen O,Autti T,Raininko R,Ylikoski A,Erkinjuntti T.MRI of the
Kirkpatrick JB,Hayman LA.White-matter lesions in MR imaging brain in neurologically healthy middle-aged and elderly
of clinically healthy brains of elderly subjects: possible individuals. Neuroradiology 1997; 39: 537–545
pathologic basis. Radiology 1987; 162; 509–511 Scheltens P,Barkhof F,Leys D,Wolters EC,Ravid R,Kamphorst W.
Knopman DS, Parisi JE, Salviati A, Floriach-Robert M, Boeve BF, Histopathologic correlates of white matter changes on MRI
Ivnik RJ, Smith GE, Dickson DW, Johnson KA, Petersen LE, in Alzheimer’s disease and normal aging. Neurology 1995;
McDonald WC, Braak H, Petersen RC. Neuropathology of 45: 883–888
cognitively normal elderly. J Neuropathol Exp Neurol 2003; Scheltens P, Erkinjunti T, Leys D,Wahlund LO, Inzitari D, de Ser T,
62: 1087–1095 Pasquier F, Barkhof F, Mantyla R, Bowler J, Wallin A, Ghika J,
Kobari M, Stirling Meyer J, Ichijo M. Leuko-araiosis, cerebral at- Fazekas F, Pantoni L. White matter changes on CT and MRI:
rophy, and cerebral perfusion in normal aging. Arch Neurol an overview of visual rating scales. European task force on
1990; 47: 161–165 age-related white matter changes. Eur Neurol 1998; 39;
Lee D, Fox A,Vinuela F, Pelz D, Lau C, Donald A, Merskey H. Inter- 80–89
observer variation in computed tomography of the brain. Schmidt H,Schmidt R,Fazekas F,Semmler J,Kapeller P,Reinhart
Arch Neurol 1987; 44: 30–31 B, Kostner GM. Apolipoprotein E e4 allele in the normal
Leifer D, Buonanno FS, Richardson EP Jr. Clinicopathologic elderly; neuropsychologic and brain MRI correlates. Clin
correlations of cranial magnetic resonance imaging of Genet 1996; 50: 293–299
periventricular white matter. Neurology 1990; 40: 911–918 Schmidt R, Fazekas F, Kleinert G, Offenbacher H, Grindl K, Payer
Leys D, Soetaert G, Petit H, Fauquette A, Pruvo J-P, Steinling M. F, Freidl W, Niederkorn K, Lechner H. Magnetic resonance
Periventricular and white matter magnetic resonance imaging signal hyperintensities in the deep and subcorti-
imaging hyperintensities do not differ between Alz- cal white matter. A comparative study between stroke pa-
heimer’s disease and normal aging. Arch Neurol 1990; 47: tients and normal volunteers. Arch Neurol 1992; 49:
525–527 825–827
Longstreth WT Jr, Manolino TA, Arnold A, Burke GL, Bryan N, Shintani S, Shiigai T, Arinami T. Subclinical cerebral lesions ac-
Jungreis CA,Enright PL,O’Leary D,Fried L.Clinical correlates cumulation on serial magnetic resonance imaging (MRI) in
of white matter findings on cranial magnetic resonance patients with hypertension: risk factors. Acta Neurol Scand
imaging of 3301 elderly people.The cerebrovascular health 1998; 97: 251–256
study. Stroke 1996; 27: 1274–1282 Skoog I. A review on blood pressure and ischaemic white mat-
Malone MJ, Szoke MC. Neurochemical changes in white mat- ter lesions. Dement Geriatr Cogn Disord 1998; 9: 13–19
ter. Aged human brain and Alzheimer’s disease. Arch Neu- Smith CD, Snowdon DA,Wang H, Markesbery WR.White matter
rol 1985; 42: 1063–1066 volumes and periventricular white matter hyperintensities
Marchall VG, Bradley WG, Marchall CE, Bhoopat T, Rhodes RH. in aging and dementia. Neurology 2000; 54: 538–842
Deep white matter infarction: correlation of MR imaging Steingart A, Hachinski VC, Lau C, Fox AJ, Diaz F, Cape R, Inzitari
and histopathological findings. Radiology 1988; 167: 517– D, Merskey H. Cognitive and neurology findings in subjects
522 with diffuse white matter lucencies on computed tomo-
Mineura K, Sasajima H, Kikuchi K, Kowads M,Tomura N, Monma graphic scan (leuko-araiosis). Arch Neurol 1987; 44: 32–35
K, Segawa Y. White matter hyperintensity in neurologically Sullivan P, Pary R,Telang F, Rifai AH, Zubenko GS. Risk factors for
asymptomatic subjects. Acta Neurol Scand 1995; 92: white matter changes detected by magnetic resonance
151–156 imaging in elderly. Stroke 1990; 21: 1424–1428
Moody DM, Brown WR, Challa VR, Anderson RL. Periventricular Tarvonen-Schröder S,Röyttä M,Räihä I,Kurki T,Rajala T,Souran-
venous collagenosis: association with leukoaraiosis. Radio- der L. Clinical features of leuko-araiosis. J Neurol Neurosurg
logy 1995; 194: 469–476 Psychiatry 1996; 60: 431–436
Murdoch G.Staining for apoptosis:now neuropathologists can Van Gijn J. Leukoaraiosis and vascular dementia. Neurology
“see” leukoaraiosis. AJNR Am J Neuroradiol 2000; 2: 42–43 1998; 51: S3-S8
O’Sullivan M, Lythgoe DJ, Pereira AC, Summers PE, Jarosz JM, van Straaten EC, Scheltens P, Knol DL, van Buchem MA, van Dijk
Williams SCR, Merkus HS. Patterns of cerebral blood flow EJ, Hofman PAM, Karas G, Kjartansson O, de Leeuw FE, Prins
reduction in patients with ischemic leukoaraiosis. Neuro- ND, Schmidt R, Visser MC, Weinstein HC, Barkhof F. Opera-
logy 2002; 59: 321–326 tional definitions for the NINDS-AIREN criteria for vascular
Pantoni L, Garcia JH. Pathogenesis of leukoaraiosis. A review. dementia: an interobserver study. Stroke 2003; 34: 1907–
Stroke 1997; 28: 652–659 1912
Pohjasvaara T, Mäntylä R, Salonen O, Aronen HJ, Ylikoski R, Van Zwieten JC, Staal S, Kapelle LJ, Derix MMA, van Gijn J. Are
Hietanen M, Kaste M, Erkinjuntti T. How complex interac- white matter lesions directly associated with cognitive im-
tions of ischemic brain infarcts, white matter lesions, and pairment in patients with lacunar infarcts. J Neurol 1996;
atrophy relate to poststroke dementia. Arch Neurol 2000; 243: 196–200
57: 1295–1300 Vinters HV, Ellis WG, Zarow C, Zaias BW, Jagust WJ, Mack WJ,
Prencipe M, Marini C.Leuko-araiosis: definition and clinical cor- Chui HC. Neuropathologic substrates of ischemic vascular
relates – an overview. Eur Neurol 1989; 29: 27–29 dementia. J Neuropathol Exp Neurol 2000; 59: 931–945
References and Further Reading 1049

Wahlund L-O, Agartz I, Almqvist O, Basun H, Foressell L, Sääf J, Jellinger K, Danielczyk W, Fischer P, Gabriel E. Clinicopathologi-
Wetterberg L. The brain in healthy aged individuals: MR cal analysis of dementia disorders in the elderly.J Neurol Sci
imaging. Radiology 1990; 174: 675–679 1990: 95: 239–258
Wahlund LO, Barkhof F, Fazekas F, Bronge L, Augustin M, Sjör- Kinkel WR, Jacobs L, Polachini I, Bates V, Heffner RR Jr. Subcorti-
gen M, Wallin A, Ader H, Leys D, Pantoni L, Pasquier F, cal arteriosclerotic encephalopathy (Binswanger’s disease).
Erikjuntti T, Scheltens P. A new rating scale for age-related Computed tomographic nuclear magnetic resonance, and
white matter changes applicable to MRI and CT. Stroke clinical correlations. Arch Neurol 1985; 42: 951–959
2001; 32: 1318–1322 Knopman DS, Rocca WA, Cha RA, Edland SD, Kokmen E.Survival
Wong KT, Grossman RI, Boorstein JM, Lexa FJ, McGowan J. Mag- study of vascular dementia in Rochester, Minnesota. Arch
netization transfer imaging of periventricular hyperintense Neurol 2003; 60: 85–90
white matter in elderly. AJNR Am J Neuroradiol 1995; 16: Kuwabara Y,Ichiya Y,Sasaki M,Yoshida T,Fukumura T,Masuda K,
252–258 Ibayashi S, Fujishima M. Cerebral blood flow and vascular
Yamamauchi H, Fukuyama H, Shio H. Corpus callosum atrophy response to hypercapnia in hypertensive patients with
in patients with leukoaraiosis may indicate global cognitive leukoaraiosis. Ann Nucl Med 1996; 10: 293–298
impairment. Stroke 2000; 31: 1515–1520 Lee A, Yu YL, Tsoi M, Woo E, Chang CM. Subcortical arterioscle-
Ylikoski A, Erkinjuntti T, Raininko R, Sarna S, Sulkava R, Tilvis R. rotic encephalopathy – a controlled psychometric study.
White matter hyperintensities on MRI in the neurologically Clin Neurol Neurosurg 1989; 91: 235–241
nondiseased elderly. Analysis of cohorts of consecutive Liao D, Cooper L, Cai J,Toole JF, Bryan NR, Hutchinson RG,Tyrol-
subjects aged 55–85 years living at home. Stroke 1995; 26; er HA. Presence and severity of cerebral white matter le-
1171–1177 sions and hypertensions, its treatment and its control. The
ARIC study. Stroke 1996; 27: 2262–2270
Loeb C, Meyer JS. Vascular dementia: still a debatable entity?
J Neurol Sci 1996; 143: 31–40
99 Subcortical Arteriosclerotic Loizou LA, Kendall BE, Marshall J. Subcortical arteriosclerotic
Encephalopathy encephalopathy: a clinical and radiological investigation.
J Neurol Neurosurg Psychiatry 1981; 44: 294–304
Aharon-Peretz J, Cummings JL, Hill MA.Vascular dementia and Lotz PR, Ballinger WE Jr, Quisling RG. Subcortical arteriosclerot-
dementia of the Alzheimer type. Arch Neurol 1988; 45: ic encephalopathy: CT spectrum and pathologic correla-
719–721 tion. AJNR Am J Neuroradiol 1986; 7: 817–822
Araki Y, Nomura M, Tanaka H, Yamamoto H, Yamamoto T, Tsuk- McQuinn BA, O’Leary DH. White matter lucencies on comput-
aguchi I, Nakamura H. MRI of the brain in diabetes mellitus. ed tomography, subacute arteriosclerotic encephalopathy
Neuroradiology 1994; 36: 101–103 (Binswanger’s disease), and blood pressure. Stroke 1987;
Caplan LR. Binswanger’s disease revisited. Neurology 1995; 45: 18: 900–905
626–633 Merino JG. Leukoaraiosis. Reifying rarefaction. Arch Neurol
Dantoine TF, Debord J, Merle L, Lacroix-Ramiandrisoa H, 2000; 57: 925–926
Bourzeix L,Charmes JP.Paraoxonase 1 activity:a new vascu- Padovani A, di Piero V, Bragoni M, Iacoboni M, Gualdi GF, Lenzi
lar marker of dementia? Ann N Y Acad Sci 2002; 977: 96–101 GL. Patterns of neuropsychological impairment in mild de-
Del Ser T, Bermejo F, Portera A, Arredondo JM, Bouras C, Con- mentia: a comparison between Alzheimer’s disease and
stantindis J.Vascular dementia. A clinicopathological study. multi-infarct dementia. Acta Neurol Scand 1995; 92: 433–
J Neurol Sci 1990; 96: 1–17 442
De Reuck J, Schaumburg HH. Periventricular atherosclerotic Pantoni L, Garcia JH. The significance of cerebral white matter
leukoencephalopathy. Neurology 1972; 22: 1094–1097 abnormalities 100 years after Binswanger’s report. A re-
De Reuck J, Crevits L, de Coster W, Sieben G, vander Eecken H. view. Stroke 1995; 26: 1293–1301
Pathogenesis of Binswanger chronic progressive subcorti- Pantoni L, Garcia JH. Cognitive impairment and cellular/vascu-
cal encephalopathy. Neurology 1980; 30: 920–928 lar changes in the cerebral white matter. Ann N Y Acad Sci
Drayer BP.Imaging of the aging brain.I.Normal findings.Radio- 1997; 826: 92–102
logy 1988; 166: 785–796 Pullicino P, Ostrow P, Miller L, Snyder W, Munschauer F. Pontine
Drayer BP. Imaging of the aging brain. II. Pathologic conditions. ischemic rarefaction. Ann Neurol 1995; 37: 460–466
Radiology 1988; 166: 797–806 Román GC.Senile dementia of the Binswanger type.A vascular
Erkinjuntti T. Treatment options: the latest evidence with form of dementia in the elderly. J Am Med Assoc 1987; 258:
galantamine (Reminyl). J Neurol Sci 2002; 203–204: 125– 1782–1788
130 Román GC. From UBOs to Binswanger’s disease. Impact of
Ferrer I, Bella R, Serrano MT, Martí E, Guionnet N. Arterioloscle- magnetic resonance imaging on vascular dementia re-
rotic leucoencephalopathy in the elderly and its relation to search. Stroke 1996; 27: 1269–1273
white matter lesions in Binswanger’s disease, multi-infarct Román GC. New insight into Binswanger’s disease.Arch Neurol
encephalopathy and Alzheimer’s disease.J Neurol Sci 1990; 1999; 56: 1061–1062
98: 37–50 Sacquegna T, Guttmann S, Giuliani S, Agati R, Daidone R, Mor-
Fisher CM. Binswanger’s encephalopathy: a review. J Neurol reale A, Ambrosetto G, Gallassi R. Binswanger’s disease: a
1989; 236: 65–79 review of the literature and a personal contribution. Eur
Gupta SR, Naheedy MH, Young JC, Ghobrial M, Rubino FA, Neurol 1989; 29: 20–22
Hindo W.Periventricular white matter changes and demen- Sandijk R. Subcortical arteriosclerotic encephalopathy (Bin-
tia. Arch Neurol 1988; 45: 637–641 swanger’s disease). S Afr Med J 1983; 63: 204–205
Hachinski V. Binswanger’s disease: neither Binswanger’s nor a Steingart A, Hachinski VC, Lau C, Fox AJ, Fox H, Lee D, Inzitari D,
disease. J Neurol Sci 1991; 103: 1 Merskey H. Cognitive and neurologic findings in subjects
with diffuse white matter lucencies on computed tomo-
graphic scan (leuko-araiosis). Arch Neurol 1987; 44: 32–35
1050 References and Further Reading

Tanoi Y, Okeda R, Budka H. Binswanger’s encephalopathy: seri- Lee S-K, Kim DI, Jeong E-K, Kim S-Y, Kim SH, In YK, Kim D-S, Choi
al sections and morphometry of the cerebral arteries. Acta J-U. Postoperative evaluation of Moyamoya disease with
Neuropathol (Berl) 2000; 100: 347–355 perfusion-weighted MR imaging. AJNR Am J Neuroradiol
Tomimoto H,Akiguchi I,Wakita H,Osaki A,Hayashi M.Yamamo- 2003; 24: 741–747
to Y. Coagulation activation in patients with Binswanger Miller DH, Haas LF, Teague C, Neale TJ. Small vessel vasculitis
disease. Arch Neurol 1999; 56: 1104–1108 presenting as neurological disorder. J Neurol Neurosurg
Tomonage M, Yamanouchi H, Tohgi H, Kameyama M. Clinico- Psychiatry 1984; 47: 791–794
pathologic study of progressive subcortical vascular en- Miller DH, Ormerod IEC, Gibson A, du Boulay EPGH, Rudge P,
cephalopathy (Binswanger type) in the elderly.J Am Geriatr McDonald WI.MR brain scanning in patients with vasculitis:
Soc 1982; 30: 524–529 differentiation from multiple sclerosis. Neuroradiology
Vermeer SE,Koudstaal PJ,Oudkerk M,Hofman A,Breteler MMB. 1987; 29: 226–231
Prevalence and risk factors of silent brain infarcts in the Moore PM. Diagnosis and management of isolated angiitis of
population-based Rotterdam scan study. Stroke 2002; 33: the central nervous system. Neurology 1989; 39:167–173
21–25 Moore PM. Neurological manifestations of vasculitis: update
Wiszniewska M, Devuyst G, Bogousslavsky J, Ghica J, van Melle on immunopathogenic mechanisms, diagnostic and thera-
G. What is the significance of leukoaraiosis in patients with peutic options. Ann Neurol 1995; 37: 131–141
acute ischemic stroke? Arch Neurol 2000; 57: 967–973 Moore PM. The vasculitides. Curr Opin Neurol 1999; 12: 383–
Zeumer H, Schonsky B, Sturm KW. Predominant white matter 388
involvement in subcortical arteriosclerotic encephalo- Perez C, Olier J, Rivero M, Montes M. MR imaging of acute en-
pathy (Binswanger disease). J Comput Assist Tomogr 1980; cephalopathy associated with polyarteritis nodosa.AJR Am
4: 14–19 J Roentgenol 1997; 169: 1995–1196
Provenzale JM, Allen NB. Neuroradiologic findings in poly-
arteritis nodosa. AJNR Am J Neuroradiol 1996; 17: 1119–
1126
100 Vasculitis Reichart MD, Bogousslavsky J, Janzer RC. Early lacunar strokes
complicating polyarteritis nodosa: thrombotic mircro-
Primary and Secondary Vasculitis of the CNS
angiopathy. Neurology 2000; 54: 883–889
Rosen CL, DePalma L, Morita A. Primary angiitis of the central
Ay H, Sahin G, Saatci I, Söylemezğlu F, Saribas O. Primary angi- nervous system as a first presentation in Hodgkin’s disease:
itis of the central nervous system and silent cortical hemor- a case report and review of the literature. Neurosurgery
rhages. AJNR Am J Neuroradiol 2002; 23:1561–1563 2000; 46: 1504–1508
Campi A, Benndorf G, Filippi M, Reganati P, Martinelli V, Terreni Scharre D, Petri M, Engman E, DeArmond S, Large intracraniel
MR. Primary angiitis of the central nervous system: serial arteritis with giant cells in systemic lupus erythematosus.
MRI of brain and spinal cord. Neuroradiology 2001; 43: Ann Intern Med 1986; 104: 661–662
599–607 Scully RE, Mark EJ, McNelly WF, McNelly BU. Case 5–1995. Pre-
Ehsan T, Hasan S, Powers JM, Heiserman JE. Serial magnetic res- sentation of case. N Engl J Med 1995; 333: 452–459
onance imaging in isolated angiitis of the central nervous Scully RE, Mark EJ, McNelly WF, McNelly BU. Case 33–1995. Pre-
system. Neurology 1995; 45: 1462–1465 sentation of case. N Engl J Med 1995; 333: 1135–1143
Ferro JM. Vasculitis of the central nervous system. J Neurol Sener RN. Diffusion MRI findings in isolated intracranial angi-
1998; 245: 766–776 itis. Comput Med Imaging Graph 2002; 26: 265–269
Finelli PF, Onyiuke HC, Uphoff DF. Idiopathic granulamatous Sharma BK, Jain S, Radotra BD. An autopsy study of Takayasu
angiitis of the CNS manifesting as diffuse white matter dis- arteritis in India. Int J Cardiol 1998; 66: 85–90
ease. Neurology 1997; 49: 1696–1699 Yuh WTC, Ueda T, Maley JE, Quets JP,White M, Hahn PY, Otake S.
Greenan TJ, Grossman RI, Goldberg HI. Cerebral vasculitis: MR Diagnosis of microvasculopathy in CNS vasculitis: value of
imaging and angiographic correlation. Radiology 1992; perfusion and diffusion imaging. J Magn Reson Imaging
182: 65–72 1999; 10: 310–313
Harada S, Mitsunobu F, Kodama F, Hosaki Y, Mifune T, Tsugeno
H, Okamoto M,Yamamura M, Makino H,Tanizaki Y. Giant cell
arteritis associated with rheumatoid arthritis monitored by Auto-immune Mediated Disorders
magnetic resonance imaging. Intern Med 1999; 38: 675–
678 Akman-Demir G, Serdaroglu P, Tasçi B. Clinical patterns of neu-
Hoffmann M, Corr P, Robbs J. Cerebrovascular findings in rological involvement in Behçet’s disease; evaluation of 200
Takayasu disease. J Neuroimaging 2000; 10: 84–90 patients. Brain 1999; 122: 2171–2181
Jennette CJ, Falk RJ. Small-vessel vasculitis. N Engl J Med 1997; Akman-Demir G, Bahar S, Coban O,Tasci B, Serdaroglu P.Cranial
337; 1512–1523 MRI in Behcet’s disease: 134 examinations of 98 patients.
Joelson E, Ruthrauff B, Ali F, Lindeman N, Sharp FR. Mutifocal Neuroradiology 2003; 45: 851–859
dural enhancement associated with temporal arteritis. Alexander EL, Malinow K, Lejewski JE, Jerdan MS. Provost TT,
Arch Neurol 2000; 57: 119–122 Alexander GF.Primary Sjörgen’s syndrome with central syn-
Komiyama M. Moyamoya disease a progressive occlusive arte- drome with central nervous system disease mimicking
riopathy of the primitive internal carotid artery. Intervent multiple sclerosis. Ann Intern Med 1986; 104: 323–330
Neuroradiol 2003; 9: 39–45 Al-Fahad SA,Al-Araji AH.Neuro-Behçet’s disease in Iraq:a study
Lanthier S, Lortie A, Michaud J, Laxer R, Jay V, deVeber G. Isolat- of 40 patients. J Neurol Sci 1999; 170: 105–111
ed angiitis of the CNS in children. Neurology 2001; 56: Al-Watban J, Patay Z, Bohlega S, Larsson S. Magnetic resonance
837–842 imaging of central nervous system involvement in primary
Lee AG, Brazis PW. Temporal arteritis: a clinical approach. J Am Sjörgen’s syndrome. Riv Neuroradiol 1998; 11: 51–54
Geriatr Soc 1999; 47: 1364–1370
References and Further Reading 1051

Anderson JR. Intracerebral calcification in a case of systemic Çoban O, Bahar S, Akman-Demir G,Talpci B,Yurdakul S,Yazici H,
lupus erythematosus with neurological manifestations. Serdaroğlu P. Masked assessment of MRI findings: is it pos-
Neuropathol Appl Neurobiol 1981; 7: 161–166 sible to differentiate neuro-Behçets disease from other
Ando Y, Kai S, Uyama E, Iyonaga K, Hashimoto Y, Uchino M, An- central nervous system. Neuroradiology 1999; 41: 255–260
do M. Involvement of the central nervous system in Colamussi P, Giganti M, Cittaniti C, Dovigo L, Trotta F, Tola MR,
rheumatoid arthritis: its clinical manifestations and analysis Tamarozzi R, Lucignani G, Piffanelli A. Brain single-photon
by magnetic resonance imaging. Intern Med 1995; 34: emission tomography with 99mTc-HMPAO in neuropsychi-
188–191 atric systemic lupus erythematosus: relations with EEG and
Baraczka K, Lakos G, Sipka S. Immunoserological changes in MRI findings and clinical manifestations. Eur J Nucl Med
the cerebro-spinal fluid and cerebrum in systemic lupus 1995; 22: 17–24
erythematosus patients with demyelinating syndrome and Cox PD, Hales RE. CNS Sjögren’s syndrome: an underrecog-
multiple sclerosis. Acta Neurol Scand 2002; 105: 378–383 nized and underappreciated neuropsychiatric disorder.
Baum KA, Hopf U, Nehrig C, Stöver M, Schörner W. Systemic lu- J Neuropsychiatry Clin Neurosci 1999; 11: 241–247
pus erythematosus: neuropsychiatric signs and symptoms Cristophe C, Azzi N, Bouche B, Dan B, Levivier M, Ferster A. Mag-
related to cerebral MRI findings. Clin Neurol Neurosurg netic resonance imaging in cerebral fungal vasculitis. Neu-
1993; 95: 29–34 ropediatrics 1999; 30: 218–220
Bell CL, Partington C, Robbins M, Graziano F, Turski P, Kornguth Cutolo M, Nobili F, Sulli A, Pizzorni C, Briata M, Faelli F, Viatali P,
S. Magnetic resonance imaging of central nervous system Mariani G, Copello F, Seriolo B, Barone C, Rodriguez G. Evi-
lesions in patients with lupus erythematosus. Correlation dence of cerebral hypoperfusion in scleroderma patients.
with clinical remission and antineurofilament and anticar- Rheumatology 2000; 39: 1366–1373
diolipin antibody titers. Arthritis Rheum 1991; 34: 432–441 Dierckx RA,Aichner F,Gerstenbrand F,Fritsch P.Progressive sys-
Berlis A, Petschner F, Bötefür IC, Spreer J.Wegener ganuloma in temic sclerosis and nervous system involvement: A review
the fourth ventricle. AJNR Am J Neuroradiol 2003; 24: of 14 cases. Eur Neurol 1987; 26: 134–140
523–525 Friedman SD, Stidley CA, Brooks WM, Hart BL, Sibbitt WL Jr.
Besana C, Comi G, del Maschio A, Praderio L, Vergani A, Brain injury and neurometabolic abnormalities in systemic
Medaglini S, Martinelli V, Triulzi F, Locatelli T. Electrophysio- lupus erythematosus. Radiology 1998; 209: 79–84
logical and MRI evaluation of neurological involvement in Fields RA, Sibbitt WL, Toubbeh H, Bankhurst AD. Neuropsychi-
Behçet’s disease. J Neurol Neurosurg Psychiatry 1089; 52: atric lupus erythematosus, cerebral infarctions, and anticar-
749–754 diolipin antibodies. Ann Rheum Dis 1990; 49: 114–117
Bianchi DW, Zickwolf GH, Weil GJ, Sylvester S, DeMaria MA. Futrell N, Schultz LR, Milikan C. Central nervous system disease
Male fetal progenitor cells persist in maternal blood for as in patients with systemic lupus erythematosus. Neurology
long as 27 years postpartum. Proc Nat Acad Sci 1996; 93: 1992; 42: 1649–1657
705–708 Futrell N. Connective tissue disease and sarcoidosis of the cen-
Blaszcyk M, Krolicki L, Krasu M, Glinska O, Jablonska S. Progres- tral nervous system. Curr Opin Neurol 1994; 7: 201–208
sive facial hemiatrophy: central nervous system involve- Gerber S, Biondi A, Dormont D, Wechsler B, Marsault C. Long-
ment and relationship with scleroderma en coup de sabre. term MR follow-up of cerebral lesions in neuro-Behçet’s
J Rheumatol 2003; 30: 1997–2004 disease. Neuroradialogy 1996; 38: 761–768
Bosma GPT, Rood MJ, Zwinderman AH, Huizinga TWJ, van Gieron MA, Khoromi S, Campos A. MRI changes in the central
Buchem MA. Evidence of central nervous system damage nervous system in a child with lupus erythematosus. Pedi-
in patients with neuropsychiatric systemic lupus erythe- atr Radiol 1995; 25: 184–185
matosus, demonstrated by magnetic transfer imaging. Gobernado JM, Leiva C, Rabano J, Alvarez-Cermeno JC, Fernan-
Arthritis Rheum 2000; 43: 48–54 dez-Molina A. Recovery from rheumatoid cerebral vasculi-
Bosma GPT, Huizinga TWJ, Mooijaart SP, van Buchem MA. Ab- tis. J Neurol Neurosurg Psychiatry 1984; 47: 410–413
normal brain diffusivity in patients with neuropsychiatric Gonzalez-Crespo MR, Blanco FJ, Ramos A, Circuelo E, Mateo I,
systemic lupus erythematosus. AJNR Am J Neuroradiol Lopez Pino MA, Gomez-Reino JJ. Magnetic resonance
2003; 24: 850–854 imaging of the brain in systemic lupus erythematosus. Br J
Brooks WM, Jung RE, Ford CC, Greinel EJ, Sibbitt WL Jr. Relation- Rheumatol 1995; 34: 1055–1060
ship between neurometabolite derangement and neu- Griffey RH, Brown MS, Bankhurst AD, Sibbitt RR, Sibbit WL Jr.
rocognitive dysfunction in systemic lupus erythematosis. Depletion of high-energy phosphates in the central ner-
J Rheumatol 1999; 26: 81–85 vous system of patients with systemic lupus erythemato-
Bruyn RPM, van der Veen JPW, Donker AJM, Valk J, Wolters EC. sus, as determined by phosphorus-31 nuclear magnetic
Sneddon’s syndrome. Case report and literature review. resonance spectroscopy. Arthritis Rheum 1990; 33: 827–
J Neurol Sci, 1987; 79: 243–253 833
Cauli A, Montaldo C, Peltz MT, Nurchis P, Sanna G, Garau P, Pala Grosso S, Fioravanti A, Biasi G, Conversano E, Marcolongo R,
R, Passiu G, Mathieu A. Abnormalities of magnetic reso- Morgese G, Balestri P. Linear scleroderma associated with
nance imaging of the central nervous system in patients progressive brain atrophy. Brain Dev 2003; 25: 57–61
with systemic lupus erythematosus correlate with disease Hammad A,Tsukada Y,Torre N.Cerebral occlusive vasculopathy
severity. Clin Rheumatol 1994; 13: 615–618 in systemic lupus erythematosis and speculation on the
Chung MH, Sum J, Morrell MJ, Horoupian DS. Intracerebral part played by complement. Ann Rheum Dis 1992; 51:
involvement in scleroderma en coup de sabre: report of a 550–552
case with neuropathologic findings. Ann Neurol 1995; 37: Hietaharju A, Jaaskeleinen S, Hietarinta M, Frey H. Central ner-
679–681 vous system involvement and psychiatric manifestations in
Çoban O,Bahar S,Akman-Demir G,Serdaroğlu P,Baykan-Kurt B, systemic sclerosis (scleroderma): clinical and neurophysio-
Tolun R,Yurdakul S,Yazici H. A controlled study of reliability logical evaluation. Acta Neurol Scand 1993; 87: 382–387
and validity of MRI findings in neuro-Behçet’s disease. Neu-
roradiology 1996; 38: 312–316
1052 References and Further Reading

Hiraiwa M, Nonaka C, Abe T, Iio M. Positron emission tomogra- Miller DH, Buchanan N, Barker G, Morrissey SP, Kendall BE,
phy in systemic lupus erythematosus: relation of cerebral Rudge P, Khamashta M, Hughes GRV, McDonald WI.
vasculitis to PET findings. AJNR Am J Neuroradiol 1983; 4: Gadolinium-enhanced magnetic resonance imaging of the
541–543 central nervous system in systemic lupus erythematosus.
Ishikawa O, Ohnishi K, Miyachi Y, Ishizaka H. Cerebral lesions in J Neurol 1992; 239: 460–464
systemic lupus erythematosus detected by magnetic reso- Mills JA. Systemic lupus erythematosus. N Engl J Med 1994;
nance imaging. Relationship to anticardiolipin antibody. 330: 1871–1879
J Rheumatol 1994; 21: 87–90 Nagaoka S, Matsunaga K, Chiba J, Ishigatsubo Y, Tani K. Five
Jennings JE, Sundgren PC, Attwood J, McCune J, Maly P. Value cases of systemic lupus erythematosus with intracranial
of MRI of the brain in patients with systemic lupus erythe- calcification. Clin Neurol 1982: 22: 635–643
matosus and neurologic disturbance. Neuroradiology Nelson JL. Maternal-fetal immunology and autoimmune dis-
2004; 46: 15–21 ease. Arthritis Rheum 1996; 39: 191–194
Jung SM, Lee BG, Joh GY, Cha JK, Chung WT, Kim KH. Primary Nelson JL, Furst DE, Malony S, Gooley T, Evans P, Smith A, Bean
Sjorgen’s syndrome manifested as multiple sclerosis and MR, Ober C, Bianchi DW. Microchimerism and HLA-compat-
cutaneous erythematous lesions: a case report. J Korean ible relationships of pregnancy in scleroderma. Lancet
Med Sci 2000; 15: 115–118 1998; 351: 559–562
Keall AT, Shetty M, Lee BCP, Lockshin MD. The diversity of neu- Newman LS, Rose CS, Maier LA. Sarcoidosis. N Engl J Med 1997;
rologic events in systemic lupus erythematosus. Prospec- 336: 1224–1234
tive clinical and computed tomographic classification of 82 Niemelä RK, Hakela M. Primary Sjörgen’s syndrome with severe
events in 71 patients. Arch Neurol 1986; 43: 273–276 central nervous system disease. Semin Arthritis Rheum
Kashihara K, Nakashima S, Kohira I, Shohmori T, Fujiwara Y, 1999; 29: 4–13
Kuroda S. Hyperintense basal ganglia of T1-weighted MR Nobili F, Cutolo M, Sulli A, Castaldi A, Sardanelli F, Accardo S,
images in a patient with central nervous system lupus and Rosadini G, Rodriguez G. Impaired quantitative cerebral
chorea. AJNR Am J Neuroradiol 1998; 19: 284–286 blood flow in scleroderma patients. J Neurol Sci 1997; 152:
Kashihara K, Fukase S, Kohira I, Abe K. Laminar cortical necrosis 63–71
in central nervous system lupus: sequential changes in MR Nobili F, Cutolo M, Sulli A, Vitali P, Vignola S, Rodriguez G. Brain
changes. Clin Neurol Neurosurg 1999; 101: 145–147 functional involvement by perfusion SPECT in systemic
Kidd D, Steuer A, Denman AM, Rudge P. Neurological complica- sclerosis and Behcet’s disease. Ann N Y Acad Sci 2002; 966:
tions in Behçet’s syndrome. Brain 1999; 122: 2183–2194 409–414
Koçer N, Islak C, Siva A, Saip S, Akman C, Kantarci O, Hamuryu- Nowak DA, Widenka DC. Neurosarcoidosis: a review of its
dan V. CNS involvement in neuro-Behçet syndrome: an MR intracranial manifestation. J Neurol 2001; 248: 363–372
study. AJNR Am J Neuroradiol 1999; 20: 1015–1024 Ohtani H, Imai H, Yasuda T, Wakui H, Komatsuda A, Hamia K,
Kozora E,West SG, Kotzin BL, Julian L, Porter S, Bigler E. Magnet- Miura AB. A combination of livedo racemosa, occlusion of
ic resonance imaging abnormalities and cognitive deficits cerebral blood vessels, and nephropathy: kidney involve-
in systemic lupus erythematosus patients without overt ment in Sneddon’s syndrome. Am J Kidney Dis 1995; 26:
central nervous system disease. Arthritis Rheum 1998; 41: 511–515
41–47 Padovan CS, Bise K, Hahn J, Sostak P, Holler E, Kolb H-J, Straube
Kwon SU, Koh JY, Kim JS.Vertebrobasilar artery territory infarc- A. Angiitis of the central nervous system after allogeneic
tion as an initial manifestation of systemic lupus erythe- bone marrow transplantation? Stroke 1999; 30: 1651–1656
matosus. Clin Neurol Neurosurg 1999; 101: 62–67 Patel DV, Neuman MJ, Hier DB. Reversibility of CT and MR find-
Kwong KL, Chu R, Wong SN. Parkinsonism as unusual neuro- ings in neuro-Behçet disease. J Comput Assist Tomogr
logical complication in childhood systemic lupus erythe- 1989; 13: 669–673
matosus. Lupus 2000; 9: 474–477 Pelz MT, Casciu L, Manconi FM, Sanna G, Schiffini P, Mathieu A.
Lambert NC, Evans PC, Hashizumi TL, Maloney S, Gooley T, Furst Distribution of white matter lesions in multiple sclerosis
DE, Nelson JL. Cutting edge: persistent fetal micro- and systemic lupus erythematosus. Differentiation using
chimerism in T lymphocytes is associated with HLA- MRI. Riv Neuroradiol 1998; 11: 17–19
DQA1*0501: implications in autoimmunity. J Immunol Pickuth D, Spielmann RP, Heywang-Köhbrunner SH. Role of
2000; 164: 5545–5548 radiology in the diagnosis of neurosarcoidosis. Eur Radiol
Lim MK, Suh CH, Kim HJ, Cho YK, Choi SH, Kang JH, Park W, Lee 2000; 10: 941–944
JH. Systemic lupus erythematosus: brain MR imaging and Pickuth D, Heywang-Köbrunner SH. Neurosarcoidosis: evalua-
single-voxel hydrogen 1 MR spectroscopy. Radiology 2000; tion with MRI. J Neuroradiol 2000; 27: 185–188
217: 43–49 Pierot L, Sauve C, Leger JM, Martin N, Koeger AC, Wechsler B,
Mascalchi M, Cosottini M, Cellerini M, Paganini M, Arnetoli G. Chiras J. Asymptomatic cerebral involvement in Sjögren’s
MRI of spinal cord involvement in Behcet’s disease. Neuro- syndrome: MRI findings of 15 cases. Neuroradiology 1993;
radiology 1998; 40: 255–257 35: 378–380
Matsumoto R,Shintaku M,Suzuki S,Kato T.Cerebral perivenous Pomper MG,Miller TJ,Stone JH,Tidmore WC,Hellmann DB.CNS
calcification in neuropsychiatric lupus erythematosus: a vasculitis in autoimmune disease: MR imaging findings and
case report. Neuroradiology 1998; 40: 583–586 correlation with angiography.AJNR Am J Neuroradiol 1999;
McAbee GN, Barasch ES. Resolving MRI lesions in lupus erythe- 20: 75–85
matosus selectively involving the brainstem.Pediatr Neurol Provenzale JM, Heinz ER, Ortel TL, Macik BG, Charles LA, Alberts
1990; 6: 186–189 MJ. Antiphospholipid antibodies in patients without sys-
Miller DH, Kendall BE, Barter S, Johnson G, MacManus DG, temic lupus erythematosus: neuroradiogic findings. Radio-
Logsdail SJ, Ormerod IEC, McDonald WI. Magnetic reso- logy 1994; 192: 531–537
nance imaging in central nervous system sarcoidosis. Neu- Provenzale JM, Allen NB. Wegener granulomatosis: CT and MR
rology 1988; 38: 378–383 findings. AJNR Am J Neuroradiol 1996; 17: 785–792
References and Further Reading 1053

Provenzale JM, Allen MB. Neuroradiologic findings in poly- Tah ET, Atilla S, Keskin T, Simonson T, I’ik S,Yuh WTC. MRI in neu-
arteritis nodosa. AJNR Am J Neuroradiol 1996; 17: 1119– ro-Behçet’s disease. Neuroradiology 1997; 39: 2–6
1126 Tanabe J, Weiner MW. MRI-MRS of the brain in systemic lupus
Reichart MD, Bogousslavsky J, Janzer RC. Early lacunar strokes erythematosus. How do we use it to understand causes of
complicating polyarteritis nodosa: thrombic microangio- clinical signs. Ann N Y Acad Sci 1997; 823: 169–184
pathy. Neurology 2000; 54: 883–889 Tokumaru AM, Obata T, Kohyama S, Kaji T, Okizuka H, Suzuki K,
Rovaris M, Viti B, Ciboddo G, Gerevini S, Capra R, Ianucci G, Co- Kusano S. Intracranial meningeal involvement in Churg-
mi G, Filippi M. Brain involvement in systemic mediated dis- Strauss syndrome. AJNR Am J Neuroradiol 2002; 23:
eases: magnetic resonance and magnetization transfer 221–224
imaging study. J Neurol Neurosurg Psychiatry 2000; 68: Trysberg E, Lindgren I,Tarkowski A. Autologous stem cell trans-
170–177 plantation in a case of treatment resistant central nervous
Rovaris M, Inglesi M,Viti B, Ciboddo G, Gerevini S, Capra R, Filip- system lupus. Ann Rheum Dis 2000; 59: 236–238
pi M.The contribution of fast-FLAIR MRI for lesion detection Van der Kaaden AJ, Kamphuis DJ, Nossent JC, Rico RE. Long-
in the brain of patients with systemic immune diseases. standing isolated cerebral systemic lupus erythematosus
J Neurol 2000; 247: 29–33 in an 8-year-old Black girl. Clin Neurol Neurosurg 1993; 95:
Sailer M, Burchert W, Ehrenheim C, Smid HGOM, Haas J,Wildha- 241–244
gen K,Wurster U, Deicher H. Positron emission tomography Wechsler B, Dell’Isola B,Vidaihet M, Dormont D, Piette JC, Bletry
and magnetic resonance imaging for cerebral involvement O, Godeau P. MRI in 31 patients with Behçet’s disease and
in patients with systemic lupus erythematosus. J Neurol neurological involvement: prospective study with clinical
1997; 244: 186–193 correlation. J Neurol Neurosurg Psychiatry 1993; 56: 793–
Sanna G, Piga M, Terberry JW, Peltz MT, Giagheddu S, Satta L, 798
Ahmed A, Cauli A, Montaldo C, Passiu G, Peter JB, Shoenfeld Welsh KMA. Scleroderma: chimerism, the blind man and the
Y, Matthieu A. Central nervous system involvement in sys- scientist. Lancet 1998; 351: 540–541
temic lupus erythematosus: cerebral imaging and sero- Welsh KMA, Nagesh J, Boska M, Moore PM. Detection of cere-
logical profile in patients with and without overt neuropsy- bral ischemia in systemic lupus erythematosus by magnet-
chiatric manifestations. Lupus 2000; 9: 573–583 ic resonance techniques. Ann N Y Acad Sci 1997; 823:
Scully RE, Mark EJ, McNelly WF, Ebeling SH. Case 28–1998. Pre- 120–131
sentation of a case. N Engl J Med 1998; 339: 755–763 Yamamoto K, Nogaki H,Takase Y, Morimatsu M. Systemic lupus
Serdaroğlu P. Behçet’s disease and the nervous system. J Neu- erythematosus associated with marked intracranial calcifi-
rol 1998; 245: 197–205 cation. AJNR Am J Neuroradiol 1992; 13: 1340–1342
Serdaroğlu P, Yazici H, Özdemir C, Yurdakul S, Bahar S, Aktin E. Yuh WTC, Ueda T, Maley JE, Quets JP,White M, Hahn PY, Otake S.
Neurologic involvement in Behçet’s syndrome. A prospec- Diagnosis of microvasculopathy in CNS vasculitis: value of
tive study. Arch Neurol 1989; 46: 265–269 perfusion and diffusion imaging. J Magn Reson 1999; 10:
Sherman JL, Stern BJ. Sarcoidosis of the CNS: comparison of 310–313
unenhanced and enhanced MR images. AJNR Am J Neuro- Zajicek JP. Neurosarcoidosis. Curr Opin Neurol 2000; 13:
radiol 1990; 11: 915–923 323–325
Shibata M, Kibe T, Fujimoto S, Ishikawa T, Murakami M, Ichi T, Zuheir Al Kawi M, Bohlega S, Banna M. MRI findings in neuro-
Wada Y. Diffuse central nervous system lupus involving Behçet’s disease. Neurology 1991; 41: 405–408
white matter, basal ganglia, thalami and brainstem. Brain
Dev 1999; 21: 337–340
Shiotani A, Mukobayashi C, Oohata H, Yamanishi T, Hara T, Itoh Infectious Vasculitis
H,Nishioka S.Wegener’s granulamatosis with dural involve-
ment as the initial clinical manifestation. Intern Med 1997;
Aasley J, Nilsen G. Cerebral atrophy in Lyme disease. Neuro-
36: 514–518
radiology 1990; 32: 253–254
Sigal LH. The neurologic presentation of vasculitic and
Ahmad NM,Boruchoff SE.Multiple cerebral infarcts due to vari-
rheumatologic syndromes. A review. Medicine 1987; 66:
cella-zoster virus large-vessel vasculopathy in an immuno-
157–180
competent adult without skin involvement. Clin Infect Dis
Siva A, Kantarci OH, Saip S, Altintas A, Hamuryudan V, Islak C,
2003; 13: e16–18
Koçer N,Yazici H.Behçet’s disease: diagnositic and prognos-
Arvin AM. Varicella-Zoster virus: pathogenesis, immunity, and
tic aspects of neurological involvement. J Neurol 2001: 248:
clinical management in hematopoietic cell transplant re-
95–103
cipients. Biol Blood Marrow Transplant 2000; 6: 219–230
Smith AS, Meisler DM, Weinstein MA, Tomsak RL, Hanson MR,
Belman AL, Coyle PK, Roque C, Cantos E. MRI findings in chil-
Rudick RA, Farris BK, Ransohoff RM. High-signal periventric-
dren infected bgy Borrelia burgdorferi. Pediatr Neurol 1992;
ular lesions in patients with sarcoidosis: neurosarcoidosis
8: 428–431
or multiple sclerosis? AJNR Am J Neuroradiol 1989; 10:
Bodensteiner JB, Mille MR, Riggs JE.Clinical features of vascular
485–490
thrombosis following varicella. Arch Pediatr Adolesc Med
Stern BJ, Krumholz A, Johns C, Scott P, Nissim J. Sarcoidosis and
1992; 146: 100–106
its neurological manifestations. Arch Neurol 1985; 42:
Cintron R, Pachner AR. Spirochetal diseases of the central ner-
909–917
vous system. Curr Opin Neurol 1994;7: 217–222
Sneddon JB. Cerebrovasular lesions and livedo reticularis. Br J
Coyle PK. Neurologic complications of Lyme disease. Rheum
Dermatol 1965; 77: 180–185
Dis Clin North Am 1993; 19: 993–1009
Szer IS, Miller JH, Rawlings D, Shaham B, Bernstein B. Cerebral
Cristophe C, Azzi N, Bouche B, Dan B, Levivier M, Ferster A. Mag-
perfusion abnormalities in children with central nervous
netic resonance imaging in cerebral fungal vasculitis. Neu-
system manifestations of lupus detected by single proton
ropediatrics 1999; 30: 218–220
emission computed tomography. J Rheumatol 1993; 20:
2143–2148
1054 References and Further Reading

Daniel RT, Henry PT, Rajshekhar V. Unusual MR presentation of Drug-related Vasculitis


cerebral parenchymal tuberculosis. Neurol India 2002; 50:
210–211 Aggerwal SK, Williams V, Levine SR, Cassin BJ, Garcia JH.
Defer G, Levy R, Burgiéres P, Postic D, Degos JD. Lyme disease Cocaine-associated intracranial hemorrhage: absence of
presenting as a stroke in the vertebrobasilar territory: MRI. vasculitis in 14 cases. Neurology 1996; 46: 1741–1743
Neuroradiology 1993; 35: 529–531 Ashchi M, Wiedermann HP, James KB. Cardiac complication
Demaerel P, Wilms G, Casteels K, Silberstein J, Baert AL. Child- form use of cocaine and phenylephirine in nasal septoplas-
hood neuroborreliosos: clinicoradiological correlation. ty. Arch Ontolaryngol 1995; 121: 681–684
Neuroradiology 1995; 37: 578–581 Barbieri EJ, Ferko AP, DiGregoria GJ, Ruch EK. The presence of
Demaerel P, Crevits I, Casteels-Van Daele M, Baert AL. Meningo- cocaine and benzoylecgoninen in rat cerebrospinal fluid
radiculitis due to borreliosis presenting as low back pain after the intravenous administration of cocaine. Life Sci
only. Neuroradiology 1998; 40: 126–127 1992; 51: 1739–1746
Fernandez RE, Rothberg M, Ferencz G, Wujack D. Lyme disease Bartzokis G, Goldstein IB, Hance DB, Beckson M, Shapiro D, Lu
of the CNS: MR imaging findings in 14 cases. AJNR Am J PH, Edwards N, Mintz J, Bridge P. The incidence of T2-
Neuroradiol 1990; 11: 479–481 weighted MR imaging signal abnormalities in the brain of
Finkel MJ, Halperin JJ. Nervous system Lyme borreliosis – revis- cocaine-dependent patients is age-related and region-spe-
ited. Arch Neurol 1992; 49: 102–107 cific. AJNR Am J Neuroradiol 1999; 20: 1628–1635
Garg RK. Tuberculosis of the central nervous system. Postgrad Benders MJ, Dorrepaal CA, van de Bor M, van Bel F. Acute ef-
Med J 1999; 75; 133–140 fects of indometacin on cerebral hemodynamics and oxy-
Gilden DH, Mahalingam R, Cohrs RJ, Kleinschmidt-DeMasters genation. Biol Neonate 1995; 68; 91–99
BK, Forghani B. The protean manifestations of varicella- Buttner A, Mall G, Penning R, Weis S. The neuroapathology of
zoster virus vasculopathy. J Neurovirol 2002; 8 (Suppl 2): heroin abuse. Forensci Sci Int 2000; 113: 435–442
75–79 Cone EJ. Pharmacokinetics and pharmacodynamics of co-
Gilden DH, Kleinschmidt-DeMasters BK, Laguardia JJ, Ma- caine. J Anal Toxicol 1995; 19: 459–478
halingam R, Cohrs RJ. Neurologic complications of the reac- Cuellar ML. Drug-induced vasculitis. Curr Rheumatol Rep 2002;
tivation of varicella-zoster virus. N Engl J Med 2000; 342; 4: 55–59
635–645 De Smet PA. A multidisciplinary overview of intoxicating snuff
Hausler MG, Ramaekers VT, Reul J, Meilicke R, Heimann G. Early rituals in the western hemisphere. J Ethnopharmacol 1985;
and late onset manifestations of cerebral vasculitis related 13: 3–49
to varicella zoster. Neuropediatrics 1998; 29: 202–207 Fowler JS,Volkow ND,Wang GJ, Gatley SJ, Logan J. Cocaine: PET
Krüger H, Heim E, Schuknecht B, Scholtz S. Acute and chronic studies of cocaine pharmacokinetics, dopamine trans-
neuroborreliosis with and without CNS involvement; a clin- porter availability and dopamine transporter occupancy.
ical, MRI, and HLA study of 27 cases. J Neurol 1991; 238: Nucl Med Biol 2001; 28: 561–572
271–280 Gradon JD, Wityk R. Diagnosis of probable cocaine-induced
Lorenzl S, Pfister HW, Padovan C,Yoursy T. MRI abnormalities in cerebral vasculitis by magnetic resonance angiography.
tick-borne encephalitis. Lancet 1996; 347: 698–699 South Med J 1995; 88: 1264–1266
Nelson JA, Wolf MD, Yuh WTC, Peeples ME. Cranial nerve in- He GQ, Zhang A, Altura BT, Altura BM. Cocaine-induced cere-
volvement with Lyme borreliosis demonstrated by mag- brovasospasm and its possible mechanisms of action.
netic resonance imaging. Neurology 1992; 42: 671–673 J Pharmacol Exp Ther 1994; 268: 1532–1539
Oksi J, Kalimo H, Marttila RJ, Marjamäki M, Sonninen P, Nikoske- Heesch CM,Wilhelm CR, Ristch J, Adnane J, Bontempo FA,Wag-
lainen J, Viljanen MK. Inflammatory brain changes in Lyme ner WR. Cocaine activates platelets and increases the for-
borreliosis. A report on three patients and review of the lit- mation of circulating platelet containing microagregates in
erature. Brain 1996; 119: 2143–2154 humans. Heart 2000; 83: 688–695
Pfister H-W, Wilske B, Weber K. Lyme borreliosis: basic science Kaye BR, Fainstat M. Cerebral vasculitis associated with cocaine
and clinical aspects. Lancet 1994; 343: 1013–1016 abuse. JAMA 1987; 258: 2104–2106
Rafto SE, Milton WJ, Galetta SL, Grossman RI. Biopsy-confirmed Lau CE, Iman A, Ma F, Falk JL. Acute effects of cocaine on spon-
CNS Lyme disease: MR appearance at 1.5 T. AJNR Am J Neu- taneous and discriminative motor functions: relation to
roradiol 1990; 11: 482–484 route of administration and pharmacokinetics.J Pharmacol
Sartoretti-Schefer S, Kollias S, Valavanis A. Ramsay Hunt syn- Exp Ther 1991; 257: 444–246
drome associated with brain stem enhancement. AJNR Am Margolis MT, Newton TH. Methamphetamine (“speed”) arteri-
J Neuroradiol 1999; 20: 278–280 tis. Neuroradiology 1971; 2: 179–182
Sencer S, Sencer A, Aydin K, Hepgül K, Payanli A, Minareci O. Martin K, Rogers T, Kavanaugh A. Central nervous system an-
Imaging in tuberculosis of the skull and skull-base: case re- giopathy associated with cocaine abuse.J Rheumatol 1995;
port. Neuroradiology 2003; 45: 160–163 22: 780–782
Shaw DR, La Brooy JT. Non-viral infections of the nervous sys- McEvoy AW, Kitchen ND, Thomas DG. Intracerebral haemor-
tem. Curr Opin Neurol Neurosurg 1991; 4: 227–231 rhage and drug abuse in young adults.Br J Neurosurg 2000;
Steere AC. Lyme disease. N Engl J Med 2001; 345: 115–125 14: 449–454
Ueno M, Oka A, Koeda T, Okamoto R, Takeshita R. Unilateral oc- Merkel PA, Koroshetz WJ, Irizarry MC, Cudkowicz ME. Cocaine-
clusion of the middle cerebral artery after varicella zoster associated cerebral vasculitis. Semin Arthritis Rheum 1995;
virus infection. Brain Dev 2002; 24: 106–108 25: 172
Mockel M, Kampf D, Lobeck H, Frei U. Severe panarteritis asso-
ciated with drug abuse. Intensive Care Med 1999; 25:
113–117
References and Further Reading 1055

Neiman J, Haapaniemi HM, Hillbom M. Neurological complica- Strickland-Marmol LB, Fessler RG, Rojiani AM. Necrotizing sar-
tions of drug abuse: pathophysiological mechanisms. Eur J coid granulomatosis mimicking an intracranial neoplasm:
Neurol 2000; 7: 595 clinicopathologic features and review of the literature.Mod
Niehaus L, Meyer BU. Bilateral borderzone brain infarctions Pathol 2000; 13: 909–913
in association with heroin abuse. J Neurol Sci 1998; 160; Tateishi U, Terae S, Ogata A, Sawamura Y, Suzuki Y, Abe S,
180–182 Miyasaka K. MR imaging of the brain in lymphomatoid
Petitti DB, Sidney S, Quesenberry C, Bernstein A. Stroke and co- granulomatosis.AJNR Am J Neuroradiol 201; 22: 1283–1290
caine or amphetamine use. Epidemiology 1998; 9; 596–600 Wang WC, Langstem JW, Steen RG, Wynn LW, Mulhern RK,
Qureshi AI, Suri MF, Guterman LR, Hopkins LN. Cocaine use and Williams JA, Kim FM, Figueroa RE. Abnormalities of the cen-
the likelihood of nonfetal myocardial infarction and stroke: tral nervous system in very young children with sickle cell
date from the Third National Health and Nutrition Examina- anemia. J Pediatr 1998; 132: 994–998
tion Study. Circulation 2001; 103: 502–506
Samuels N,Shemesh O,Yinnon Am,Fisher D,Abraham AS.Poly-
arteritis nodosa and drug abuse; is there a connection?
Postgrad Med J 1996; 72: 684–685
101 Dural Arteriovenous Fistula
Awad IA, Little JR, Akarawi WP, Ahl J. Intracranial dural arteri-
ovenous malformations: factors predisposing to an aggres-
Disorders Obstructing the Vascular Lumen sive neurological course. J Neurosurg 1990; 72: 839–850
Borden JA, Wu JK, Shucart WA. A proposed classification for
Glauser TA, Siegel MJ, Lee BCP, DeBaun MR. Accuracy of neuro- spinal and cranial dural arteriovenous fistulous malforma-
logic examination and history in detecting evidence of tions and implications for treatment. J Neurosurg 1995; 82:
MRI-diagnosed cerebral infarctions in children with sickle 166–179
cell hemoglobinopathy. J Child Neurol 1995; 10: 88–92 Brown RD Jr, Wiebers DO, Nichols DA. Intracranial dural arteri-
Kenéz J, Barsi P, Majtèniy K, Molnár B, Kocher I, Stangi E, Kolomy ovenous fistulae: angiographic predictors of intracranial
S. Can intravascular lymphomatosis mimic thrombosis? A hemorrhage and clinical outcome in nonsurgical patients.
case report with 8 months’ follow-up and fatal outcome. J Neurosurg 1994; 81: 531–538
Neuroradiology 2000; 42: 436–440 Cognard C, Gobin YP, Pierot L, Bailly AL, Houdart E, Casasco A,
Martin-Duverneuil N, Mokhtari K, Behin A, Lafitte F, Hoang- Chiras J, Merland JJ. Cerebral dural arteriovenous fistulas:
Xuan K, Chiras J. Intravascular malignant lymphomatosis. clinical and angiographic correlation with a revised classifi-
Neuroradiology 2002; 44: 749–754 cation of venous drainage. Radiology 1995; 194: 671–680
Miller D, Hochberg F, Harris N, Gruber M, Louis D, Cohen H. Cognard C, Houdart E, Casasco A, Gabrillargues J, Chiras J, Mer-
Pathology with clinical correlations of primary central ner- land JJ. Long-term changes in intracranial dural arteriove-
vous system non-Hodgkin lymphoma: the Massachusetts nous fistulae leading to worsening in the type of venous
General Hospital experience. Cancer 1994; 74: 1338–1397 drainage. Neuroradiology 1997; 39: 59–66
Moran CJ, Siegel MJ, DeBaun MR. Sickle cell disease: imaging of Collice M, D’Aliberti G,Talamonti G, Branca V, Boccardi E, Scialfa
cerebrovascular complications. Radiology 1998; 206; 311– G et al. Surgical interruption of leptomeningeal drainage as
332 treatment for intracranial dural arteriovenous fistulas with-
Moser FG, Miller ST, Bello JA, Pegelow CH, Zimmerman RA, out dural sinus drainage. J Neurosurg 1996; 84: 810–817
Wang WC, Ohene-Frempong K, Schwartz A, Vichainsky EP, Davies MA, TerBrugge K, Willinsky R, Coyne T, Saleh J, Wallace
Gallagher D, Kinney TR. The spectrum of brain MR abnor- MC. The validity of classification for the clinical presenta-
malities in sickle-cell disease: a report from the cooperative tion of intracranial dural arteriovenous fistulas. J Neurosurg
study of sickle cell disease.AJNR Am J Neuroradiol 1996; 17: 1996; 85: 830–837
965–972 Davies MA, Saleh J, TerBrugge K, Willinsky R, Wallace MC. The
Moussouttas M. Intravascular lymphomatosis presenting as natural history and management of intracranial dural arte-
posterior leukoencephalopathy. Arch Neurol 2002; 59: riovenous fistulae. Part 1: Benign lesions. Interventional
640–641 Neuroradiology 1997; 3: 295–302
Otrakji C,Voight W, Amador A, Gregorius J. Malignant angioen- Davies MA, TerBrugge K, Willinsky R, Wallace MC. The natural
dotheliomatosis: a true lymphoma. A case of intravascular history and management of intracranial dural arteriove-
lymphomatosis studied by Southern blot hybridization nous fistulae. Part 2: Aggressive lesions. Interventional Neu-
analysis. Hum Pathol 1988; 19: 475–478 roradiology 1997; 3: 303–311
Perniciaro C, Winkelmann RK, Daoud MS, Su WP. Malignant an- Duffau H, Lopes M, Janosevic V, Sichez JP, Faillot T, Capelle L,
gioendotheliomatosis is an angiotropic intravascular lym- Ismail M,Bitar A,Arthuis F,Fohanno D.Early rebleeding from
phoma. Immunohistological, ultrastructural and molecular intracranial dural arteriovenous fistulas: report of 20 cases
genetic study. Am J Dermatopathol 1995; 17: 242–248 and review of the literature. J Neurosurg 1999; 90: 78–84
Steen RG, Emudianughe T, Hankins GM, Wynn LW, Wang WC, Greenough GP, Mamourian A, Harbaugh RE. Venous hyperten-
Xiong X, Helton KJ. Brain imaging findings in pediatric pa- sion asociated with a posterior fossa dural arteriovenous
tients with sickle cell disease. Radiology 2003; 228: 216–225 fistula: another cause of bithalamic lesions on MR images.
Steen RG, Miles MA, Helton KJ, Strawn S, Wang W, Xiong XZ, AJNR Am J Neuroradiol 1999; 20: 145–147
Mulherns RK. Cognitive impairment in children with hemo- Houser OW, Campbell JK, Campbell RJ, Sundt TM. Arteriove-
globin SS sickle cell disease: relationship to MR imaging nous malformation affecting the transverse dural venous
findings and hematocrit. AJNR Am J Neuroradiol 2003; 24: sinus–an acquired lesion. Mayo Clin Proc 1979; 54: 651–661
382–389
1056 References and Further Reading

Hurst RW, Bagley LJ, Galetta S, Glosser G, Lieberman AP, Tro- Willinsky R, Goyal M, TerBrugge K, Montanera W. Tortuous, en-
janowski J, Sinson G, Stecker M, Zager E, Raps EC, Flamm ES. gorged pial veins in intracranial dural arteriovenous fistu-
Dementia resulting from dural arteriovenous fistulas: the las: correlations with presentation, location, and MR find-
pathologic findings of venous hypertensive encephalo- ings in 122 patients. AJNR Am J Neuroradiol 1999; 20:
pathy. AJNR Am J Neuroradiol 1998; 19: 1267–1273 1031–1036
Ito M, Sonokawa T, Mishina H, Sato K. Reversible dural arte-
riovenous malformation-induced venous ischemia as a
cause of dementia: treatment by surgical occlusion of
draining dural sinus: case report. Neurosurgery 1995; 3:
102 Leukoencephalopathy After
1187–1191 Chemotherapy and Radiotherapy
Jaillard AS, Peres B, Hommel M. Neuropsychological features of
dementia due to dural arteriovenous malformation. Cere- Antunes NL, Souweidane MM, Lis E, Rosenblum MK, Steinherz
brovasc Dis 1999; 9: 91–97 PG. Methotrexate leukoencephalopathy presenting as
Lasjaunias P,Chiu M,ter Brugge K,Tolia A,Hurth M,Bernstein M. Klüver-Bucy syndrome and uncinate seizures. Pediatr Neu-
Neurological manifestations of intracranial dural arteriove- rol 2002; 26: 305–308
nous malformations. J Neurosurg 1986; 64: 724–730 Bleyer WA. Leukoencephalopathy detectable by magnetic res-
Lawton MT, Jacobowitz R, Spetzler RF. Redefined role of angio- onance imaging: much ado about nothing? Int J Radiat
genesis in the pathogenesis of dural arteriovenous malfor- Oncol Biol Phys 1995; 32: 1251–1552
mations. J Neurosurg 1997; 8: 267–274 Brown MS,Stemmer SM,Simon JH,Stears JC,Jones RB,Cagnoni
Malik GM, Pearce JE, Ausman JI, Mehta B. Dural arteriovenous PJ, Sheeder JL. White matter disease induced by high-dose
malformations and intracranial hemorrhage. Neurosurgery chemotherapy: longitudinal study with MR imaging and
1984; 15: 332–339 proton spectroscopy. AJNR Am J Neuroradiol 1998; 19:
Matsuda S, Waragai M, Shinotoh H, Takahashi N, Takagi K, Hat- 217–221
tori T. Intracranial dural arteriovenous fistula presenting Cain MS, Burton GV, Holcombe RF. Fatal leukoencephalopathy
progressive dementia and parkinsonism. J Neurol Sci 1999; in a patient with non-Hodgkin’s lymphoma treated with
165: 43–47 CHOP chemotherapy and high-dose steroids. Am J Med Sci
Mironov A. Selective transvenous embolization of dural fistu- 1998; 315: 202–207
las without occlusion of the dural sinus. AJNR Am J Neuro- Cantini R, Giorgetti W, Vallandriani AM, Burchianti M, Amodeo
radiol 1998; 19: 389–391 C. Radiation-induced cerebral lesions in childhood. Childs
Newton TH, Cronqvist S. Involvement of dural arteries in in- Nerv Syst 1989; 5: 135–139
tracranial arteriovenous malformations. Radiology 1969; Chen C-Y, Zimmerman RA, Bilaniuk LT, Chou T-W, Molloy PT.
93: 1071–1078 Childhood leukemia: central nervous system abnormalities
Noguchi K, Melhem ER, Kanazawa T, Kubo M, Kuwayama N, Se- during and after treatment. AJNR Am J Neuroradiol 1996;
to H. Intracranial dural arteriovenous fistulas: Evaluation 17: 295–310
with combined 3D Time-of -Flight MR angiography and MR DeAngelis LM, Delattre J-Y, Posner JB. Radiation-induced de-
digital subtraction angiography. AJR Am J Roentgenol mentia in patients cured of brain metastases. Neurology
2004; 182: 183–190 1989; 39: 789–796
Rothbart D, Awad IA, Lee J, Kim J, Harbaugh R, Criscuolo GR. Dietrich U, Wanke I, Mueller T, Wieland R, Moellers M, Forsting
Expression of angiogenic factors and structural proteins in M,Stolke D.White matter disease in children treated for ma-
central nervous system vascular malformations. Neuro- lignant brain tumors. Childs Nerv Syst 2001: 17: 731–738
surgery 1996; 38: 915–924 Gowan GM, Herrington JD, Simonetta AB. Methotrexate-in-
Satomi J, Van Dijk JM, TerBrugge KG, Willinsky RA, Wallace MC. duced toxic leukoencephalopathy. Pharmacotherapy 2002;
Benign cranial dural arteriovenous fistulas: outcome of 22: 1183–1187
conservative management based on the natural history of Hertzberg H, Huk WJ, Ueberall MA, Langer T, Meier W, Dopfer R,
the lesion. J Neurosurg 2002; 97: 767–770 Skalej M, Lackner H, Bode U, Janßen G, Zintl F, Beck JD. CNS
Uranishi R, Nakase H, Sakaki T. Expression of angiogenic late effects after all therapy in childhood. Part I: neuroradio-
growth factors in dural arteriovenous fistula. J Neurosurg logical findings in long-term survivors of childhood. ALL –
1999; 91: 781–786 An evaluation of the interferences between morphology
Van Dijk JM, Willinsky RA. Venous congestive encephalopathy and neurophysiological performances. Med Pediatr Oncol
related to cranial dural arteriovenous fistulas. Neuroimag- 1997; 28: 387–400
ing Clin N Am 2003; 13: 55–72 Kishi T, Tanaka Y, Ueda K. Evidence for hypomethylation in two
Van Dijk JM, TerBrugge KG, Willinsky RA, Wallace MC. Multiplic- children with acute lymphoblastic leukemia and leukoen-
ity of dural arteriovenous fistulas. J Neurosurg 2002; 96: cephalopathy. Cancer 2000; 89: 925–931
76–78 Lai R, Abrey LE, Rosenblum MK, DeAngelis LM. Treatment-in-
Van Dijk JM, TerBrugge KG, Willinsky RA, Wallace MC. Clinical duced leukoencephalopathy in primary CNS lymphoma: a
course of cranial dural arteriovenous fistulas with long- clinical and autopsy study. Neurology 2004; 62: 451–456
term persistent cortical venous reflux. Stroke 2002; 33: Laitt RD, Chambers EJ, Goddard PR, Wakeley CJ, Duncan AW,
1233–1236 Foreman NK. Magnetic resonance imaging and magnetic
Willinsky R, TerBrugge K, Montanera W, Mikulis D, Wallace MC. resonance angiography in long term survivors of acute
Venous congestion: an MR finding in dural arteriovenous lymphoblastic leukemia treated with cranial irradiation.
malformations with cortical venous drainage. AJNR Am J Cancer 1995; 76: 18461852
Neuroradiol 1994; 15: 1501–1507 Laxmi SN, Takahashi S, Matsumoto K, Higano S, Kurihara N,
Imaizumi M, Abe K, Itoyama Y, Sakamoto K.Treatment-relat-
ed disseminated necrotizing leukoencephalopathy with
characteristic contrast enhancement of the white matter.
Radiat Med 1996; 14: 303–307
References and Further Reading 1057

Lövblad K-O, Kelkear P, Ozdoba C, Ramelli C, Remonda L, Di Trapani G, Sabatelli M, Carnevale A, Colosimo C, Mignogna T,
Schroth G. Pure methotrexate encephalopathy presenting Tonali P. Gliomatosis cerebri: report of an atypical case. Clin
with seizures: CT and MRI features. Pediatr Radiol 1998; 28: Neuropathol 1995; 14: 13–18
86–91 Enterline DS, Davey NC, Tien RD. Case 3: gliomatosis cerebri.
Matsumoto K, Takahashi S, Sato A, Imaizumi M, Higano S, AJRAm J Roentgenol 1995; 165: 214–215
Sakamoto K, Asakawa H, Tada K. Leukoencephalopathy in Essig M, Schlemmer H-P, Tronnier V, Hawighorst H, Wirtz R, van
childhood hematopoietic neoplasm caused by moderate- Kaick G. Fluid-attenuated inversion-recovery MR imaging
dose methotrexate and prophylactic cranial radiotherapy – of gliomatosis cerebri. Eur Radiol 2001; 11: 303–308
an MR analysis. Int J Radiat Oncol Biol Phys 1995; 32: Felsberg GJ, Silver SA, Brown MT, Tien RD. Radiologic-patho-
913–918 logic correlation. Gliomatosis cerebri. AJNR Am J Neuro-
Ohmoto Y, Kajiwara K, Kato S, Nisziaki T, Ito H, Tamura S. Atypi- radiol 1994; 15: 1745–1753
cal MRI findings in treatment-related leukoencephalopa- Freund M,Hähnel S,Sommer C,Martmann M,Kiessling M,Tron-
thy: case report. Neuroradiology 1996; 38: 128–133 nier V, Sartor K. CT and MRI findings in gliomatosis cerebri: a
Oka M, Terae S, Kobayashi R, Sawamura Y, Kudoh K, Tha KK, neuroradiologica and neuropathologic review of diffuse
Yoshida M, Kaneda M, Suzuki Y, Miyasaka K. MRI in infiltrating brain neoplasms. Eur Radiol 2001; 11: 309–316
methotrexate-related leukoencephalopathy:Disseminated Galatioto S, Marafioti T, Cavallari V, Batolo D. Gliomatosis cere-
necrotising leukoencephalopathy in comparison with mild bri: clinical, neuropathological, immunohistochemical and
leukoencephalopathy. Neuroradiology 2003; 45: 493–497 morphometric studies. Zentralbl Pathol 1993; 139: 251–267
Peterson K, Clark HB, Hall WA, Truwit CL. Multifocal enhancing Hara A, Sakai N,Yamada H,Tanaka T, Mori H. Assessment of pro-
magnetic resonance imaging lesions following cranial irra- liferative potential in gliomatosis cerebri. J Neurol 1991;
diation. Ann Neurol 1995; 38: 237–244 238: 80–82
Price RA, Birdwell DA.The central nervous system in childhood Hayek J, Valavanis A. Computed tomography of gliomatosis
leukemia. III. Mineralizing angiopathy and dystrophic calci- cerebri. Comput Radiol 1982; 6: 93–98
fication. Cancer 1978; 42: 717–728 Hecht BK Turc-Carel C, Chatel M, Lonjon M, Roche JL, Gioanni J,
Seidl H, Nygaard R, Haave I, Moe PJ. Magnetic resonance imag- Hecht Fl Gaudray P. Chromosomes in gliomatosis cerebri.
ing and neuroradiological evalutation after treatment with Genes Chromosomes Cancer 1995; 14: 149–153
high-dose methotrexate for acute lymphocytic leukaemia Herrlinger U, Felsberg J, Küker W, Bornemann A, Plasswilm L,
in young children. Acta Paediatr 1996:85:450–453 Knobbe CB, Strik H,Wick W, Meyermann R, Dichgans J, Bam-
Shanley DJ. Mineralizing microangiopathy: CT and MRI. Neuro- berg M, Feinberger G, Weller M. Gliomatosis cerebri: molec-
radiology 1995:37:331–333 ular pathology and clinical course. Ann Neurol 2002; 52:
Tahjeb P. Progressive late delayed postirradiation encephalo- 390–399
pathy with Klüver-Bucy syndrome. Serial MRI and clinico- Jennings MT, Frenchman M, Shhab T, Johnson MD, Creasy J,
pathological studies. Clin Neurol Neurosurg 1995; 97: LaPorte K, Dettbarn WD. Gliomatosis cerebri presenting as
264–268 intractable epilepsy during early childhood. J Child Neurol
Tekkök IH, Carter DA, Robinson MG, Brinker R. Reverals of CNS- 1995; 10: 37–45
prophylaxia-related leukoencephalopathy after CSF shunt- Kandler RH, Smith CML, Broome JC, Davies-Jones GAB.
ing: case histories of identical twins. Childs Nerv Syst 1996; Gliomatosis cerebri: a clinical, radiological and pathological
12: 309–314 report of four cases. Br J Neurosurg 1991; 5: 187–193
Valk PE, Dillon WP. Radiation injury of the brain. AJNR Am J Keene DL, Jimenez C, Hsu E. MRI diagnosis of gliomatosis cere-
Neuroradiol 1991; 12: 45–62 bri. Pediatr Neurol 1999; 20: 148–151
Vasquez E, Lucaya J, Castelotte A, Pigueras J, Sainz P, Olive T, Koslow SA, Claassen D, Hirsch WL, Jungreis CA. Gliomatosis
Sanchez-Toledo J, Ortega JJ. Neuroimaging in pediatric cerebri: a case report with autopsy correlation. Neuro-
leukemia and lymphoma: differential diagnosis. Radio- radiology 1992; 34: 331–333
graphics 2002; 22: 1411–1428 Lev MH. Gliomatosis cerebri has normal relative blood volume:
Wong ET. Recurrent cystic radiation necrosis of the brain. really? Who cares? Should you? AJNR Am J Neuroradiol
Oncol Rep 1998; 5: 685–687 2002; 23: 345–346
Levin N, Gomori JM, Siegal T. Chemotherapy as initial treat-
ment in gliomatosis cerebri: results with temozolomide.
Neurology 2004; 63: 354–356
103 Gliomatosis Cerebri Mawrin C, Kirches E, Schneider-Stock R, Scherlach C,Vorwerk C,
von Deimling A, van Landeghem F, Meyermann R, Borne-
Balko MG, Blisard KS, Samaha FJ. Oligodendroglial gliomatosis mann A, Müller A, Romeike B, Stoltenbirg-Didinger G, Wick-
cerebri. Hum Pathol 1992; 23: 706–707 boldt J, Pilz P, Dietzmann K. Analysis of TP53 and PTEN in
Burger PC. Gliomatosis cerebri: molecular pathology and clini- gliomatosis cerebri. Acta Neuropathol (Berl) 2003; 105:
cal course. Ann Neurol 2002; 52: 389 529–536
Bertrand E, Kryst-Widzgowska T, Baranska M. Gliomatosis cere- Mohana-Borges AVR,Imbesi SG,Dietrich R,Alksne J,Amjadi DK.
bri at the 17-year-old girl correlations of histological, CT Role of proton magnetic resonance spectroscopy in the di-
and MRI appearance. Folia Neuropathol 1994; 32: 245–249 agnosis of gliomatosis cerebri. A unique pattern of normal
Del Caprio-O’Donnovan, Korah I, Salazar A, Melancon D. choline but elevated myo-inositol metabolite levels. J Com-
Gliomatosis cerebri. Radiology 1996; 198: 831–835 put Assist Tomogr 2004; 28: 103–105
Dexter MA, Parker GD, Besser M, Ell J, Fulham MJ. MR and Nishioka H, Ito H, Miki T. Difficulties in the antemortem diagno-
positron emission tomography with fludeoxyglucose F 18 sis of gliomatosis cerebri: report of a case with diffuse in-
in gliomatosis cerebri. AJNR Am J Neuroradiol 1995; 16: crease of gemictocyte-like cells, mimicking reactive gliosis.
1507–1510 Br J Neurosurg 1996; 10: 103–107
Pyhtinen J. Proton MR spectroscopy in gliomatosis cerebri.
Neuroradiology 2000; 42: 612–615
1058 References and Further Reading

Pyhtinen J, Pääkkö E. A difficult diagnosis of gliomatosis cere- Medana IM, Esiri MM. Axonal damage: a key predictor of out-
bri. Neuroradiology 1996; 38: 444–448 come in human CNS diseases. Brain 2003; 126: 515–530
Rippe DJ, Boyko OB, Fuller NF, Friedman HS, Oakes WJ, Schold Meythaler JM, Brunner RC, Johnson A, Novack TA. Amantadine
SC. Gadopentetate-dimeglumine – enhanced MR imaging to improve neurorecovery in traumatic brain injury-associ-
of gliomatosis cerebri: appearance mimicking lep- ated diffuse axonal injury: a pilot double-blind randomized
tomeningeal tumor dissemination. AJNR Am J Neuroradiol trial. J Head Trauma Rehabil 2002; 17: 300–313
1990; 11: 800–801 Onaya M. Neuropathological investigation of cerebral white
Ross IB, Robitaille Y, Villemure J-G, Tampieri D. Diagnosis and matter lesions caused by closed head injury. Neuropatho-
management of gliomatosis cerebri: recent trends. Surg logy 2002; 22: 243–251
Neurol 1991; 36: 431–340 Parizel PM, Özsarlak Ö, van Goethem JW, van den Hauwe L,
Sanson M, Cartalat-Carel S, Taillibert S, Napolitano M, Djafari L, Dillen C, Verlooy J, Cosyns P. Imaging findings in diffuse
Cougnard J, Gervais H, Laigle F, Carpentier A, Mokhtari K, axonal injury after closed head trauma. Eur Radiol 1998; 8:
Taillandier L, Chinot O, Duffau H, Honnorat J, Hoang-Xuan K, 960–965
Delattre JY for the ANOCEF Group. Initial chemotherapy in Reichard RR, White CL, Hladik CL, Dolinak D. Beta-amyloid pre-
gliomatosis cerebri. Neurology 2004; 63: 270–275 cursor protein staining of nonaccidental central nervous
Saraf-Lavi E, Bowen BC, Pattany PM, Sklar EML, Murdoch JB, system injury in pediatric autopsies.J Neurotrauma 2003; 4:
Petito CK. Proton MR spectroscopy of gliomatosis cerebri: 347–355
case report of elevated myoinositol with normal choline Scheid R, Preul C, Gruber O, Wiggins C, von Cramon Y. Diffuse
levels. AJNR Am J Neuroradiol 2003; 24: 946–951 axonal injury associated with chronic traumatic brain in-
Schober R,Mai JK,Volk B,Wechsler W.Gliomatosis cerebri; biop- jury: evidence from T2*-weighted gradient-echo imaging
tical approach and neuropathological verification. Acta at 3 T. AJNR Am J Neuroradiol 2003; 24: 1049–1056
Neurochir (Wien) 1991; 113: 131–137 Strich SJ, Oxon DM. Shearing of nerve fibres as a cause of brain
Shin YM, Chang KH, Han MH, Myung NH, Chi JE, Cha SH, Han damage due to head injury. Lancet 1961; II: 443–449
MC. Gliomatosis cerebri: comparison of MR and CT features. Takaoka M,Tabuse H, Kamura E, Nakajima S,Tsuzuki T, Nakamu-
AJR Am J Roentgenol 1993; 161: 859–862 ra K, Okada A, Sugimoto H.Semiquantitative analysis of cor-
Spagnoli MV, Grossman RI, Packer RJ, Hackney DB, Goldberg HI, pus callosum injury using magnetic resonance imaging
Zimmerman RA, Bilaniuk LT. Magnetic resonance imaging indicates clinical severity in patients with diffuse axonal
determination of gliomatosis cerebri. Neuroradiology injury. J Neurol Neurosurg Psychiatry 2002; 73: 289–293
1987; 29: 15–18
Vates GE. Gliomatosis cerebri: a review of 22 cases. Neuro-
surgery 2003; 53: 261–271
Yanaka K, Kamezaki T, Kobayashi E, Matsueda K,Yoshii Y, Nose T.
105 Wallerian Degeneration
MR imaging of diffuse glioma.AJNR Am J Neuroradiol 1992; and Myelin Loss Secondary
13: 349–351 to Neuronal and Axonal Degeneration
Yang S, Wetzel S, Cha S. Dynamic contrast-enhanced T2*-
weighted MR imaging of gliomatosis cerbri. AJNR Am J Basak M, Erturk M, Oflazoğlu B, Ozel A, Yildiz GB, Forta H. Mag-
Neuroradiol 2002; 23: 350–355 netic resonance imaging in amyotrophic lateral sclerosis.
Yip M, Fisch C, Lamarche JB. Gliomatosis cerebri affecting the Acta Neurol Scand 2002; 105: 395–399
entire neuraxis. Radiographics 2003; 23: 247–253 Becerra JL, Puckett WR, Hiester ED, Quencer RM, Marcillo AE,
Donovan Post MJ, Bunge RP. MR-Pathologic comparisons of
wallerian degeneration in spinal cord injury. AJNR Am J
104 Diffuse Axonal Injury Neuroradiol 1995; 16: 125–133
Bignami A,Ralston HJ.The cellular reaction to wallerian degen-
eration in the central nervous system of the cat. Brain Res
Bešenski N. Traumatic injuries: imaging of head injuries. Eur
1969; 13: 444–461
Radiol 2002; 12: 1237–1252
Bignami A, Eng LF. Biochemical studies of myelin in wallerian
Chan JHM, Tsui EYK, Peh WCG, Fong D, Fok KF, Leung KM, Yuen
degeneration of rat optic nerve. J Neurochem 1973; 20:
MK,Fung KKL.Diffuse axonal injury:detection of changes in
165–173
anisotropy of water diffusion by diffusion-weighted imag-
Bozzali M, Falini A, Franceschi M, Cercignani M, Zuffi M, Scotti G,
ing. Neurology 2003; 45: 34–38
Comi G, Filippi M. White matter damage in Alzheimer’s dis-
Graham DI, Raghupathi R, Saatman KE, Meaney D, McIntosh TK.
ease assessed in vivo using diffusion tensor magnetic reso-
Tissue tears in the white matter after lateral fluid percus-
nance imaging. J Neurol Neurosurg Psychiatry 2002; 72:
sion brain injury in the rat: relevance to human brain injury.
742–746
Acta Neuropathol (Berl) 2000; 99: 117–124
Castillo M, Mukherji SK. Early abnormalities related to postin-
Hammoud DA, Wasserman BA. Diffuse axonal injuries: patho-
farction Wallerian degeration: evaluation with MR diffu-
physiology and imaging. Neuroimaging Clin N Am 2002;
sion-weighted imaging. J Comp Assist Tomogr 1999; 23:
12: 205–216
1004–1007
Huisman TAGM, Sorensen AG, Hergan K, Gonzalez RG, Schaefer
Coleman MP, Hugh Perry V. Axon pathology in neurological
PW. Diffusion-Weighted imaging for the evaluation of dif-
disease: a neglected therapeutic target. Trends Neurosci
fuse axonal injury in closed head injury. J Comput Assist
2002; 25: 532–537
Tomogr 2003; 27: 5–11
Comi G, Rovaris M, Leocani L. Neuroimaging in amyotrophic
LaPlaca MC, Thibault LE. Dynamic mechanical deformation of
lateral sclerosis. Eur J Neurol 1999; 6: 629–637
neurons triggers an acute calcium response and cell injury
Cook RD, Wisniewski HM. The role of oligodendroglia and as-
involving the N-methyl-D-aspartate glutamate receptor.
troglia in wallerian degeneration of the optic nerve. Brain
J Neurosci Res 1998; 52: 220–229
Res 1973; 61: 191–206
References and Further Reading 1059

Fukui K, Iguchi I, Kito A,Watanabe Y, Sugita K. Extent of pontine Reigner J, Matthieu JM, Kraus-Ruppert R, Lassmann H, Poduslo
pyramidal tract wallerian degeneration and outcome J. Myelin proteins, glycoproteins, and myelin-related en-
after supratentorial hemorrhagic stroke. Stroke 1994; 25: zymes in experimental demyelination of the rabbit optic
1207–1210 nerve: sequence of events. J Neurochem 1981; 36: 1986–
Griffin JW, George R, Lobato C, Tyor WR, Yan LC, Glass JD. 1995
Macrophage responses and myelin clearance during wal- Savoidardo M, Parevson D, Grisoli M, Forester M, D’Incerti L, Fa-
lerian degeneration: relevance to immune-mediated de- rina L.The effects of wallerian degeneration of the optic ra-
myelination. J Neuroimmunol 1992; 40: 153–166 diations demonstrated by MRI. Neuroradiology 1992; 34:
Igarashi H, Katayama Y,Tsuganezawa T,Yamamuro M.Terashi A, 323–325
Owan C. Three-dimensional anisotropy contrast (3DAC) Seitz RJ, Reiners K, Himmelmann F, Heininger K, Hartung HP,
magnetic resonance imaging of the human brain: applica- Toyka KV. The blood–nerve barrier in wallerian degenera-
tion to assess wallerian degeneration. Int Med 1998; 37: tion: a sequential long-term study. Muscle Nerve 1989:
662–668 627–635
Inoue Y, Matsumura Y, Fukuda T, Nemoto Y, Shirahata N, Suzuki Sonoda S, Tsubahara A, Saito M, Chino N. Extent of pyramidal
T, Shakudo M, Yawata S, Tanaka S, Takemoto K, Onoyama Y. tract wallerian degeneration in the brain stem on MRI and
MR imaging of wallerian degeneration in the brainstem: degree of motor impairment after supratentorial stroke.
temporal relationships. AJNR Am J Neuroradiol 1990; 11: Disabil Rehabil 1992; 14: 89–92
897–903 Tanabe JL, Vermathen M, Miller R, Gelinas D, Weiner MW,
Jacob S, Finsterbusch J,Weishaupt JH, Khorram-Sefat D, Frahm Rooney WD. Reduced MTR in the corticospinal tract and
J, Ehrenreich H. Diffusion tensor imaging for long-term fol- normal T2 in amyotrophic lateral sclerosis. Magn Reson
low-up of corticospinal tract degeneration in amyotrophic Imaging 1998; 16: 1163–1169
lateral sclerosis. Neuroradiology 2003; 45: 598–600 Terae S, Taneichi H, Abumi K. MRI of wallerian degeneration of
Kang DW,Chu K,Yoon BW,Song IC,Chang KH,Roh JK.Diffusion- the injured spinal cord. J Comput Assist Tomogr 1993; 17:
weighted imaging in wallerian degeneration. J Neurol Sci 700–703
2000; 178: 167–169 Türkdoğan-Sözüer D, Özek MM, Sav A, Dinçer A, Pamir MN.
Khurana DS,Strawsburg RH,Robertson RL,Madsen JR,Helmers Serial MRI and MRS studies with unusual findings in Ras-
SL. MRI signal changes in the white matter after corpus cal- mussen’s encephalitis. Eur Radiol 2000; 10: 962–966
losotomy. Pediatr Neurol 1999; 21: 691–695 Udaka F, Sawada H, Seriu N, Shindou K, Nishitani N, Kameyama
Kuhn JM, Johnson KA, Davis KR.Wallerian degeneration: evalu- M. MRI and SPECT findings in amyotrophic lateral sclerosis.
ation with MR imaging. Radiology 1988; 168: 199–202 Demonstration of upper motor neurone involvement by
Kuhn MJ, Mikulis DJ, Ayoub DM, Kosofsky BE, Davis KR, Taveras clinical neuroimaging. Neuroradiology 1992; 34: 389–393
JM. Wallerian degeneration after cerebral infarction: evalu- Uchino A, Imada H, Ohno M. MR imaging of wallerian degener-
ation with sequential MR imaging. Radiology 1989; 172: ation in the human brain stem after ictus. Neuroradiology
179–182 1990; 32: 191–195
Lassmann H, Ammerer HP, Kulnig W. Ultrastructural sequence Uchino A, Sawada A, Takase Y, Egashira R, Kudo S.Transient de-
of myelin degradation. Acta Neuropathol (Berl) 1978; 44: tection of early wallerian degeneration on diffusion-
91–102 weighted MRI after an acute cerebrovascular accident.
Lassmann H, Ammerer HP, Jurecka W, Kulnig W. Ultrastructural Neuroradiology 2004; 46: 183–188
sequence of myelin degradation. Acta Neuropathol (Berl) Werring DJ,Toosy AT,Clark CA, Parker GJM, Barker GJ, Miller DH,
1978; 44: 103–109 Thompson AJ. Diffusion tensor imaging can detect and
Lexa FJ, Grossman RI, Rosenquist AC. MR of wallerian degener- quantify corticospinal tract degeneration after stroke. J
ation in the feline visual system: characterization by mag- Neurol Neurosurg Psychiatry 2000; 269–272
netization transfer rate with histopathologic correlation. Wieshmann UC, Symms MR, Clark CA, Lemieux L, Franconi F,
AJNR Am J Neuroradiol 1994; 15: 201–212 Parker GJM, Barker GJ, Shorvon SD. Wallerian degeneration
Mazumdar A, Mukherjee P, Miller JH, Malde H, McKinstry RC. in the optic radiation after temporal lobectomy demon-
Diffusion-weighted imaging of acute corticospinal tract in- strated in vivo with diffusion tensor imaging. Epilepsia
jury preceding Wallerian degeneration in the maturing hu- 1999; 40: 1155–1158
man brain. AJNR Am J Neuroradiol 2003; 24: 1057–1066 Zhang L, Uluğ AM, Zimmerman RD, Lin MT, Rubin M, Beal MF.
Miyai I, Suzuki T, Kii K, Kang J, Kubota K.Wallerian degeneration The diagnostic utility of FLAIR imaging in clinically verified
of the pyramidal tract does not affect stroke rehabilitation amyotrophic lateral sclerosis. J Magn Reson Imaging 2003;
outcome. Neurology 1998; 51: 1613–1616 17: 521–527
Orita T, Tsurutani T, Izumihar A, Kajiwara K. Early, evolving wal-
lerian degeneration of the pyramidal tract in cerebrovascu-
lar diseases: MR study. J Comput Assist Tomogr 1994; 18:
943–946
106 Diffusion-weighted Imaging
Peretti-Viton P, Azulay JP, Trefouret S, Brunel H, Daniel C, Viton
JM, Flori A, Salazard B, Pouget J, Serratrice G, Salamon G.MRI Arfanakis K, Haughton VM, Carew JD, Rogers BP, Dempsey RJ,
of the intracranial corticospinal tracts in amyotrophic and Meyerand ME.Diffusion tensor MR imaging in diffuse axon-
primary lateral sclerosis. Neuroradiology 1999; 41: 744–749 al injury. AJNR Am J Neuroradiol 2002; 23: 794–802
Pierpaoli C, Barnett A, Pajevic S, Chen R, Penix L, Virta A, Basser Assaf BA, Mohamed FB, Abou-Khaled KJ, Williams M,Yazeji MS,
P. Water diffusion changes in wallerian degeneration and Haselgrove J, Faro SH. Diffusion tensor imaging of the hip-
their dependence on white matter architecture. Neu- pocampal formation in temporal lobe epilepsy. AJNR Am J
roimaging 2001; 13: 1174–1185 Neuroradiol 2003; 24: 1857–1862
Basser PJ, Mattielo J, LeBihan D. Estimation of the effective self-
diffusion tensor from the NMR spin echo. J Magn Reson
1994; 103: 247–254
1060 References and Further Reading

Basser PJ, Mattielo J, LeBihan D. MR diffusion tensor spec- Filippi M, Iannucci G, Gercignani M, Rocca MA, Pratesi A, Comi
troscopy and imaging. Biophys J 1994; 66: 259–267 G.A quantitative study of water diffusion in multiple sclero-
Basser PJ, Pierpaoli C. A simplified method to measure the dif- sis lesions and normal-appearing white matter using echo-
fusion tensor from seven MR images. Magn Reson Med planar imaging. Arch Neurol 2000; 57: 1017–1021
1998; 39: 928–934 Filippi M, Cercignani M, Inglese M, Horsfield MA, Comi G. Diffu-
Bozzao A,Di Paolo A,Mazzoleni C,Fasoli F,Simonetti A,Fantozzi sion tensor magnetic resonance imaging in multiple scle-
LM, Floris R. Diffusion-weighted MR imaging in the early di- rosis. Neurology 2001; 56: 304–311
agnosis of periventricular leukomalacia. Eur Radiol 2003; Fitzek C, Weissmann M, Specter H, Fitzek S, Hopf HC, Schulte E,
13: 1571–1576 Stoeter P. Anatomy of brain-stem white-matter tracts
Bydder GM, Rutherford MA, Hajnal JV. How to perform diffu- shown by diffusion-weighted imaging. Neuroradiology
sion-weighted imaging. Childs Nerv Syst 2001; 17: 195–201 2001; 43: 953–960
Cavalleri F, Berardi A, Burlina AB, Ferrari F, Mavilla L. Diffusion- Forbes KPN, Pipe JG, Heiserman JE. Evidence for cytotoxic ede-
weighed MRI of maple syrup urine disease encephalo- ma in the pathogenesis of cerebral venous infarction. AJNR
pathy. Neuroradiology 2002; 44: 499–502 Am J Neuroradiol 2001; 22: 450–455
Cercignani M, Bozzali M, Iannucci G, Comi G, Filippi M. Magneti- Forbes KPN, Pipe JG, Bird CR. Changes in brain water diffusion
sation transfer ratio and mean diffusivity of normal appear- during the 1st year of life. Radiology 2002; 222: 405–409
ing white and gray matter from patients with multiple scle- Ford BJ. Brownian movement in Clarkia pollen: a reprise of first
rosis. J Neurol Neurosurg Psychiatry 2001; 70: 311–317 observations. Microscope 1992; 40: 235–241
Cercignani M, Inglese M, Pagani E, Comi G, Filippi M. Mean dif- Gelal F, Calli C, Apaydin M, Erdem G. Van der Knaap’s leukoen-
fusivity and fractional anisotrophy histograms of patients cephalopathy: report of five new cases with emphasis on
with multiple sclerosis. AJNR Am J Neuroradiol 2001; 22: diffusion-weighted MRI findings. Neuroradiology 2002; 44:
952–958 625–630
Cercignani M, Bozzali M, Iannucci G, Comi G, Filippi M. Intra- Gray L, MacFall J. Overview of diffusion imaging. MRI Clin N Am
voxel and inter-voxel coherence in patients with multiple 1998; 6: 125–138
sclerosis assessed using diffusion tensor MRI.J Neurol 2002; Guo A,Jewells VL,Provenzale JM.Analysis of normal-appearing
249: 875–883 white matter in multiple sclerosis: comparison of diffusion
Chan JHM, Tsui EYK, Peh WCG, Fong D, Kok KF, Leung KM, Yeun tensor MR imaging and magnetization transfer imaging.
MK, Fung KKl. Diffuse axonal injury: detection of changes in AJNR Am J Neuroradiol 2001; 22: 1893–1900
anisotrophy of water diffusion by diffusion-weighted Guo AC, Petrella JR, Kurzberg J, Provenzale JM. Evaluation of
imaging. Neuroradiology 2003; 45: 34–38 white matter anisotrophy in Krabbe disease with diffusion
Chen C-Y, Lee K-W, Lee C-C, Chin S-C, Chung H-W, Zimmerman tensor MR imaging: initial experience. Radiology 2001; 218:
RA. Heroin-induced spongiform leukoencephalopathy: val- 809–815
ue of diffusion MR imaging. J Comput Assist Tomogr 2000; Guo AC, MacFall JR, Provenzale JM. Multiple sclerosis: diffusion
24: 735–737 tensor MR imaging for evaluation of normal-appearing
Clark CA, Le Bihan D. Water diffusion compartmentation and white matter. Radiology 2002; 222: 729–736
anisotrophy at high b values in the human brain. Magn Heide AC, Richards TL, Alvord EC Jr, Peterson J, Rose LM. Diffu-
Reson Med 2000; 44: 852–859 sion imaging of experimental allergic encephalomyelitis.
Clark CA, Hedehus M, Moseley ME. In vivo mapping of the fast Magn Reson Med 1993; 29: 478–484
and slow diffusion tensors in human brain. Magn Reson Helenius J,Soinne L,Perkiö J,Salonen O,Kamasmäki A,Kaste M,
Med 2002; 47: 623–628 Carano RAD, Aronen HJ,Tatlismak T. Diffusion-weighted MR
Doran M, Bydder GM. Magnetic resonance: perfusion and dif- imaging in normal human brains in various age groups.
fusion imaging. Neuroradiology 1990; 32: 392–398 AJNR Am J Neuroradiol 2002; 23: 194–199
Doran M, Hajnal JV, van Bruggen N, King MD, Young IR, Bydder Horsfield MA. Using diffusion-weighted MRI in multicenter
GM. Normal and abnormal white matter tracts shown clinical trials for multiple sclerosis. J Neurol Sci 2001; 186:
by MR imaging using directional diffusion weighted se- S51-S54
quences. J Comput Assist Tomogr 1990; 14: 865–873 Horsfield MA, Jones DK. Applications of diffusion-weighted
Ducreux D, Oppenheim C,Vandamme X, Dormont D, Samson Y, and diffusion tensor MRI to white matter diseases.A review.
Rancurel G, Cosnard G, Marsault C. Diffusion-weighted NMR Biomed 2002; 15: 570–577
imaging patterns of brain damage associated with cerebral Horsfield MA, Lai M, Webb SL, Barker GJ, Tofts PS, Turner R,
venous thrombosis. AJNR Am J Neuroradiol 2001; 22: Rudge P, Miller DH. Apparent diffusion coefficients in be-
261–268 nign and secondary progressive multiple sclerosis by nu-
Eastwood JD, Engelter ST, MacFall JF, Delong DM, Provenzale clear magnetic resonance. Magn Reson Med 1996; 36:
JM. Quantitative assessment of the time course of infarct 393–400
intensity on diffusion-weighted images. AJNR Am J Neuro- Hüppi PS, Murphy B, Maier SE, Zientara GP, Inder TE, Barnes PD,
radiol 2003; 24: 680–687 Kikinis R, Jolensz FA, Volpe JJ. Microstructural brain devel-
Engelbrecht V, Scherer A, Rassek M, Witsack HJ, Mödder U. Dif- opment after perinatal cerebral white matter injury as-
fusion-weighted MR imaging in the brain in children: find- sessed by diffusion tensor magnetic resonance imaging.
ings in the normal brain and in the brain with white matter Pediatrics 2001; 107: 455–460
diseases. Radiology 2002; 222: 410–418 Huisman TAGM. Diffusion-weighted imaging: basic concepts
Engelter ST, Provenzale JM, Petrella JR. Assessment of vaso- and application in cerebral stroke and head trauma. Eur Ra-
genic edema in eclampsia using diffusion imaging. Neuro- diol 2003; 13: 2283–2297
radiology 2000; 42: 818–820 Igarashi H, Katayama Y,Tsuganezawa T,Yamamuro M,Terashi A,
Fiehler J, Foth M, Kucinski T, Knab R, von Bezold M, Weiller C, Owan C. Three-dimensional anisotrophy contract (3DAC)
Zeumer H, Röther J. Severe ADC decreases do not predict magnetic resonance imaging of the human brain: applica-
irreversible tissue damage in humans. Stroke 2002; 33: tion to assess wallerian degeneration. Intern Med 1998; 37:
79–86 662–668
References and Further Reading 1061

Iizuka T, Sakai F, Kan S, Suzuki N. Slowly progressive spread of Ferriero DM, Barkovich AJ. Serial quantitative diffusion tensor
the stroke-like lesions in MELAS. Neurology 2003; 61: MRI of the premature brain: development in newborn with
1238–1244 and without injury. J Magn Reson Imaging 2002; 16: 621–
Inder T, Huppi PS, Zientara GP, Maier SE, Jolensz FA, di Salvo D, 632
Robertson R,Barnes PD,Volpe JJ.Early detection of periven- Mori S, Crain BJ, Chacko VP, van Zijl PCM. Three-dimensional
tricular leukomalacia by diffusion-weighted magnetic res- tracking of axonal projections in the brain by magnetic res-
onance imaging techniques. J Pediatr 1999; 134: 631–634 onance imaging. Ann Neurol 1999; 45: 265–269
Ito R, Melhem ER, Mori S, Eichler FS, Raymond GV, Moser HW. Mori S, Itoh R, Zhang J, Kaufmann WE, van Zijl PCM, Solaiyap-
Diffusion tensor brain MR imaging in X-linked cerebral pan M,Yarowsky P.Diffusion tensor imaging of the develop-
adrenoleukodystrophy. Neurology 2001; 56: 544–547 ing mouse brain. Magn Reson Med 2001; 46: 18–23
Ito R, Mori S, Melhem ER. Diffusion tensor brain imaging and Mori S, van Zijl PCM. Fiber tracking: principles and strategies –
tractography. Neuroimaging Clin N Am 2002; 12: 1–19 a technical review. NMR Biomed 2002; 15: 468–480
Jan W, Zimmerman RA, Wang ZJ, Berry GT, Kaplan PB, Kaye EM. Moseley ME, Cohem Y, Kucharczyk J, Mintotovitch J, Asgari HS,
MR diffusion imaging and MR spectroscopy of maple syrup Wendland MF,Tsuruda J, Norman D. Diffusion weighted MR
urine disease during acute metabolic decompensation. imaging of anisotropic water diffusion in cat central ner-
Neuroradiology 2003; 45: 393–399 vous system. Radiology 1990; 176: 439–445
Jones DK, Simmons A, Williams SCR, Horsfield MA. Non-inva- Mukherjee P, Miller JH, Shimony JS, Conturo TE, Lee PCP, Almli
sive assessment of axonal fiber connectivity in the human CR, McKinstry RC. Normal brain maturation during child-
brain via diffusion tensor MRI. Magn Reson Med 1999; 42: hood: developmental trends characterized with diffusion-
37–41 tensor MR imaging. Radiology 2001; 221: 349–358
Khong P-L, Lam BCC, Chung BHY, Wong CK-Y, Ooi G-C. Diffu- Mukherjee P, Miller JH, Shimony JS, Philip JV, Nehra D, Snyder
sion-weighted MR imaging in neonatal nonketotic hyper- AZ, Conturo TE, Neil JJ, McKinstry RC. Diffusion-tensor MR
glycemia. AJNR Am J Neuroradiol 2003; 24: 1181–1183 imaging of gray and white matter development during
Kim H-S, Kim D-I, Lee B-I, Jeong E-K, Choi C, Lee JD, Yoon P-H, normal human brain maturation. AJNR Am J Neuroradiol
Kim E-J, Kim S-H, Yoon YK. Diffusion weighted image and 2002; 23: 1445–1456
MR spectroscopic analysis of a case of MELAS with repeat- Neil JJ, Shiran SI, McKinstry RC, Schefft GL, Snyder AZ, Almli CR,
ed attacks.Yonsei Med J 2001; 42: 128–133 Akbudak E, Aronovitz JA, Miller JP, Lee BCP, Conturo TE. Nor-
Le Bihan D. Diffusion, perfusion MR imaging of the brain: from mal brain in human newborns: apparent diffusion coeffi-
structure to function. Radiology 1990; 177: 328–329 cient and diffusion anisotrophy measured by using diffu-
Le Bihan D, Breton E, Lallamand D, Desbleds MT, Aubin ML,Vig- sion tensor MR imaging. Radiology 1998; 209: 57–66
naud J, Roger B. Contribution of intravoxel incoherent mo- Nusbaum AO, Tang CK, Bushbaum MS, Wei TC, Atlas SW. Re-
tion (IVIM) imaging to neuroradiology. J Neuroradiol 1987; gional and global changes in cerebral diffusion with nor-
14: 295–312 mal aging. AJNR Am J Neuroradiol 2001; 22: 136–142
Le Bihan D, Breton E, Lallemand D, Aubin M-L,Vignaud J, Laval- Ohshita T, Oka M, Imon Y, Wanatabe C, Katayama C,Yamaguchi
Jeantet M. Separation of diffusion and perfusion in in- S, Kajima T, Mimori Y, Nakamura S. Serial diffusion-weighted
travoxel incoherent motion MR imaging. Radiology 1988; imaging in MELAS. Neuroradiology 2000; 42: 651–656
168: 497–505 Ono J, Harada K, Sakurai K, Kodaka R, Shimidzu N, Tanaka J,
Le Bihan D, Mangin J-F, Poupon C, Clark CA, Pappata S, Molko N, Nagai T, Okada S. MR diffusion imaging in Pelizaeus-
Chabriat H. Diffusion tensor imaging: concepts and appli- Merzbacher disease. Brain Dev 1994; 16: 219–223
cations. J Magn Reson Imaging 2001; 13: 534–546 Ono J, Harada K, Mano T, Sakurai K, Okada S. Differentiation of
Makris N, Worth AJ, Sorensen AG, Papadimitriou GM, Wu O, dys- and demyelination using diffusional anisotrophy.
Reese TG, Wedeen VJ, Davis TL, Stakes JW, Caviness VS, Ka- Pediatr Neurol 1997; 16: 63–66
plan E, Rosen BR, Pandya DN, Kennedy DN. Morphometry of Oppenheim C, Galanoud D, Samson Y, Sahel M, Dormont D,
in vivo human white matter association pathways with dif- Wechsler B, Marsault C. Can diffusion weighted magnetic
fusion-weighted magnetic resonance imaging. Ann Neurol resonance imaging help differentiate stroke from stroke-
1997; 42: 951–962 like events in MELAS? J Neurol Neurosurg Psychiatry 2000;
Mazumdar A, Mukherjee P, Miller JH, Malde H, McKinstry RC. 69: 248–250
Diffusion-weighted imaging of acute corticospinal tract in- Prayer D, Barkovich J, Kirschner DA, Prayer LM, Roberts TPL,
jury preceding Wallerian degeneration in the maturing hu- Kucharczyk J, Mosely ME. Visualization of nonstructural
man brain. AJNR Am J Neuroradiol 2003; 24: 1057–1066 changes in early white matter development on diffusion-
McGraw P, Liang L, Provenzale JM. Evaluation of normal age-re- weighted MR images: evidence supporting premyelination
lated changes in anisotrophy during infancy and child- anisotrophy. AJNR Am J Neuroradiol 2001; 22: 1572–1576
hood as shown by diffusion tensor imaging. AJR Am J Rovaris M, Bozzali M, Iannucci G, Ghezzi A, Caputo D, Montanari
Roentgenol 2002; 179: 1515–1522 E, Bertolotto A, Bergamaschi R, Capra R, Mancardi GL, Mar-
Miller DH,Thompson AJ, Filippi M. Magnetic resonance studies tinelli V, Comi G, Filippi M. Assessment of normal-appearing
of abnormalities in the normal appearing white matter and white and gray matter in patients with primary progressive
gray matter in multiple sclerosis. J Neurol 2003; 250: 1407– multiple sclerosis. A diffusion-tensor magnetic resonance
1419 imaging study. Arch Neurol 2002; 59: 1406–1412
Miller JH, McKinstry RC, Philip JV,Mukherjee P,Neil JJ.Diffusion- Roychowdhury S, Maldjian JA, Grossman RI. Multiple sclerosis:
tensor MR imaging of normal brain maturation: a guide comparison of trace apparent diffusion coefficients with
to structural development and myelination. AJR Am J MR enhancement patterns of lesions. AJNR Am J Neurora-
Roentgenol 2003; 180: 851–859 diol 2000; 21: 869–874
Miller SP,Vigneron DB, Henry RG, Bohland MA, Ceppi-Cozzio C,
Hoffman C, Newton N, Patridge JC,
1062 References and Further Reading

Scanderberg AC, Tomaiuoo F, Sabatini U, Nocentini U, Grasso Berry I, Barker GJ, Barkhof F, Campi A, Dousset V, Franconi J-M,
MG, Caltagirone C. Demyelination plaques in relapsing- Gass A, Schreiber W, Miller DH, Tofts PS. A multicenter mea-
remitting and secondary-progressive multiple-sclerosis: surement of magnetization transfer ratio in normal white
assessment with diffusion MR imaging. AJNR Am J Neuro- matter. J Magn Reson Imaging 1999; 9: 441–446
radiol 2000; 21: 862–868 Dehmeshki J, Silver NC, Leary SM,Tofts PS,Thompson AJ, Miller
Schaefer PW, Grant PE, Gonzalez RG. Diffusion-weighted MR DH. Magnetization transfer ratio histogram analysis of pri-
imaging of the brain. Radiology 2000; 217: 331–345 mary progressive and other multiple sclerosis subgroups.
Schneider JFL, Il’yasov KA, Boltshauser E, Hennig J, Martin E. J Neurol Sci 2001; 185: 11–17
Diffusion tensor imaging in cases of adrenoleukodystro- Dousset V, Grossman RI, Ramer KN, Schnall MD,Young LH, Gon-
phy: preliminary experience as a marker for early demyeli- zalez-Scarano F, Lavi E, Cohen JA. Experimental allergic en-
nation? AJNR Am J Neuroradiol 2003; 24: 819–824 cephalomyelitis and multiple sclerosis: lesion characteriza-
Schneider JFL, Il’yasov KA, Hennig J, Martin E. Fast quantitative tion with magnetization transfer imaging. Radiology 1992;
diffusion-tensor imaging of cerebral white matter from the 182: 483–491
neonatal period to adolescence. Neuroradiology 2004; 46: Dousset V, Armand J-P, Lacoste D, Mièze S, Letenneur L, Dar-
258–266 tigues J-F, Caillé J-M. Magnetization transfer study of HIV
Sundgren PC, Evardsson B, Holtås S. Serial investigation of per- encephalitis and progressive multifocal leukoencephalo-
fusion disturbances and vasogenic oedema in hyperten- pathy. AJNR Am J Neuroradiol 1997; 18: 895–901
sive encephalopathy by diffusion and perfusion weighted Eng J, Ceckler TL, Balaban RS. Quantitative 1H magnetization
imaging. Neuroradiology 2002; 44: 299–304 transfer imaging in vivo. Magn Reson Med 1991; 17: 304–
Uluğ AM, Moore DF, Bojko AS, Zimmerman RD. Clinical use of 314
diffusion-tensor imaging for diseases causing neuronal Engelbrecht V, Rassek M, Preiss S, Wald C, Mödder U. Age-de-
and axonal damage. AJNR Am J Neuroradiol 1999; 20: pendent changes in magnetization transfer contrast of
1044–1048 white matter in the pediatric brain. AJNR Am J Neuroradiol
Virta A, Barnett A, Pierpaoli C. Visualizing and characterizing 1998; 19: 1923–1929
white matter fiber structure and architecture in the human Ernst T, Chang L, Witt M, Walot I, Aronow H, Leonido-Yee M,
pyramidal tract using diffusion tensor MRI. Magn Reson Singer E. Progressive multifocal leukoencephalopathy and
Imaging 1999; 17: 1121–1133 human immunodeficiency virus-associated white matter
Wakana S, Jiang H, Nagae-Poetscher LM, van Zijl PCM, Mori S. lesions in AIDS: magnetization transfer MR imaging. Radio-
Fiber tract-based atlas of human white matter anatomy. logy 1999; 210: 539–543
Radiology 2004; 230: 77–87 Filippi M. Magnetization transfer imaging to monitor the evo-
Wang XY, Noguchi K, Takashima S, Hayashi N, Ogawa S, Seto H. lution of individual multiple sclerosis lesions. Neurology
Serial diffusion-weighted imaging in a patient with MELAS 1999; 53: S18–S22
and presumed cytotoxic oedema.Neuroradiology 2003; 45: Filippi M, Rovaris M. Magnetization transfer imaging in multi-
640–643 ple sclerosis. J Neurovirol 2000; 6: s115–s120
Westin C-F, Maier SE, Mamata H, Nabavi A, Jolensz FA, Kikinis R. Filippi M, Iannucci G, Tortorella C, Minicucci L, Horsfield MA,
Processing and visualization for diffusion tensor MRI. Med Colombo B, Sormani MP, Comi G. Comparison of MS clinical
Imaging Anal 2002; 6: 93–108 phenotypes using conventional and magnetization trans-
Wiegel MR, Larsson HBW, Wedeen VJ. Fiber crossing in human fer MRI. Neurology 1999; 52: 588–594
brain depicted with diffusion tensor MR imaging. Radiolo- Filippi M, Rocca MA, Minicucci L, Martinelli V, Ghezzi A, Bergam-
gy 2000; 217: 897–903 aschi R, Comi G. Magnetization transfer imaging of patients
Wilson M, Morgan PS, Lin X,Turner BP, Blumhardt LD. Quantita- with definite MS and negative conventional MRI. Neuro-
tive diffusion weighted magnetic resonance imaging, cere- logy 1999; 52: 845–848
bral atrophy, and disability in multiple sclerosis. J Neurol Finelli DA. Magnetization transfer in neuroimaging. MRI Clin
Neurosurg Psychiatry 2001; 70: 318–322 North Am 1998; 6: 31–52
Yoneda M, Maeda M, Kimura H, Fujii A, Katayama K, Kiruyama Fralix TA, Ceckler TL,Wolff SD, Simon SA, Balaban RS.Lipid bilay-
M. Vasogenic edema on MELAS: a serial study with diffu- er and water proton magnetization transfer: effect of cho-
sion-weighted MR imaging. Neurology 1999; 53: 2182– lesterol. Magn Reson Med 1991; 18: 214–223
2184 Ge Y, Grossman RI, Udupa JK, Babb JS, Kolson DL, McGowan JC.
Yonemura K, Hasegawa Y, Kimura K, Minematsu K,Yamaguchi T. Magnetization transfer ratio histogram analysis of gray
Diffusion-weighted MR imaging in a case of mitochondrial matter in relapsing-remitting multiple sclerosis. AJNR Am J
myopathy, encephalopathy, lactic acidosis, and stroke-like Neuroradiol 2001; 22: 470–475
episodes. AJNR Am J Neuroradiol 2001; 22: 269–272 Graham SJ, Henkelman RM. Understanding pulsed magnetiza-
Zhai G, Lin W,Wilber KP, Gerig G, Gilmore JH.Comparisons of re- tion transfer. J Magn Reson Imaging 1997; 7: 903–912
gional white matter diffusion in healthy neonates and Grossman RI, McGowan JC. Perspectives on multiple sclerosis.
adults performed with 3.0-T head-only MR imaging unit. AJNR Am J Neuroradiol 1998; 19: 1251–1265
Radiology 2003; 229: 673–681 Hähnel S, Münkel K, Jansen O, Heiland S, Reidel M, Freund M,
Aschoff A, Sartor K. Magnetization transfer measurements
in normal-appearing cerebral white matter in patients
with chronic obstructive hydrocephalus. J Comput Assist
107 Magnetization Transfer Tomogr 1999; 23: 516–520
Henkelman RM, Stanisz GJ, Graham SJ. Magnetization transfer
Balaban RS, Ceckler TL. Magnetization transfer contrast in in MRI: a review. NMR Biomed 2001; 14: 57–64
magnetic resonance imaging. Magn Res Q 1992; 8: 116–137 Kaiser JS, Grossman RI, Udupa JK, Miki Y, Galetta SL. Magnetiza-
tion transfer histogram analysis of monosymptomatic
episodes of neurologic dysfunction: preliminary findings.
AJNR Am J Neuroradiol 2000; 21; 1043–1047
References and Further Reading 1063

Kalkers NF, Hintzen RQ, van Waesberghe JHTM, Lazeron RHC, Richert ND, Ostuni JL, Bash CN, Duyn JH, McFarland HF, Frank
van Schijndel RA, Adèr HJ, Polman CH, Barkhof F. Magneti- JA. Serial whole-brain magnetization transfer imaging in
zation transfer histogram parameters reflect all dimensions patients with relapsing-remitting multiple sclerosis at
of MS pathology, including atrophy. J Neurol Sci 2001; 184: baseline and during treatment with interferon beta-1b.
155–162 AJNR Am J Neuroradiol 1998; 19: 1705–1713
Kato Y, Matsumura K, Kinosada Y, Narita Y, Kuzuhara S, Naka- Ropele S, Strasser-Fuchs S, Augustin M, Stollberger R, Enzinger
gawa T. Detection of pyramidal tract lesions in amyotroph- C, Hartung H-P, Fazekas F. A comparison of magnetization
ic lateral sclerosis with magnetization-transfer measure- transfer ratio, magnetization transfer rate, and the native
ments. AJNR Am J Neuroradiol 1997; 18: 1541–1547 relaxation time of water protons related to relapsing–re-
Kita M, Goodkin DE, Bacchetti P, Waubant E, Nelson SJ, Majum- mitting multiple sclerosis. AJNR Am J Neuroradiol 2000; 21:
dar S. Magnetization transfer ratio in new MS lesions before 1885–1891
and during therapy with IFNb-1a. Neurology 2000; 54: Rovaris M, Horsfield MA, Filippi M. Correlations between mag-
1741–1745 netization transfer metrics and other magnetic resonance
Koenig SH. Cholesterol of myelin is the determinant of gray- abnormalities in multiple sclerosis. Neurology 1999; 53:
white contrast in MRI of brain. Magn Reson Med 1991; 20: S40–S45
285–291 Rovaris M, Iannucci G, Cercignani M, Sormani MP, De Stefano N,
Koenig SH, Brown RD III, Spiller M, Lundbom N. Relaxometry of Gerevini S, Comi G, Filippi M. Age-related changes in con-
brain: why white matter appears bright in MRI. Magn Reson ventional, magnetization transfer, and diffusion-tensor MR
Med 1990; 14: 482–495 imaging findings: study with whole-brain tissue histogram
Kucharczyk W, Macdonald PM, Stanisz GJ, Henkelman RM. Re- analysis. Radiology 2003; 227: 731–738
laxivity and magnetization transfer of white matter lipids at Sailer M, O’Riordan JI, Thompson AJ, Kingsley DP, MacManus
MR imaging: importance of cerebrosides and pH. Radiolo- DG, McDonald WI, Millder DH. Quantitative MRI in patients
gy 1994; 192: 521–529 with clinically isolated syndromes suggestive of demyeli-
Lexa FJ, Grossman RI, Rosenquist AC. MR of wallerian degener- nation. Neurology 1999; 52: 599–606
ation in the feline visual system: characterization by mag- Santos AC, Narayanan S, de Stefano N, Tartaglia MC, Francis SJ,
netization transfer rate with histopathologic correlation. Aranoutelis R, Caramanos Z, Antel JP, Pike GB, Arnold DL.
AJNR Am J Neuroradiol 1994; 15: 201–212 Magnetization transfer can predict clinical evolutation in
MacKay A, Whittall K, Adler J, Li D, Paty D, Graeb D. In vivo visu- patients with multiple sclerosis. J Neurol 2002; 249:
alization of myelin water in brain by magnetic resonance. 662–668
Magn Reson Med 1994; 31: 673–677 Silver NC, Barker GJ, Miller DH. Standardization of magnetiza-
McGowan JC. Magnetic resonance imaging and magnetiza- tion transfer imaging for multicenter studies. Neurology
tion transfer. Adv Imaging Electron Phys 2001; 118: 1–83 1999; 53: S33-S39
McGowan JC, Filippi M, Campi A, Grossman RI. Magnetization Tomiak MM, Rosenblum JD, Prager JM, Metz CE. Magnetization
transfer imaging: theory and application to multiple sclero- transfer: a potential method to determine the age of multi-
sis. J Neurol Neurosurg Psychiatry 1998; 64: s66–s69 ple sclerosis lesions. AJNR Am J Neuroradiol 1994; 15:
Mehta RC, Pike B, Enzmann DR. Magnetization transfer MR of 1569–1574
the normal adult brain. AJNR Am J Neuroradiol 1995; 16: Van Buchem MA, Udupa JK, McGowan JC, Miki Y, Heyning FH,
2085–2091 Boncoeur-Martel M-P, Kolson DL, Polansky M, Grossman RI.
Melhem ER, Breiter SN, Ulug AM, Raymond GV, Moser HW. Im- Global volumetric estimation of disease burden in multiple
proved tissue characterization in adrenoleukodystrophy sclerosis based on magnetization transfer imaging. AJNR
using magnetization transfer imaging. AJR Am J Roent- Am J Neuroradiol 1997; 18: 1287–1290
genol. 1996; 166: 689–695 Van Buchem MA, Steens SCA, Vrooman HA, Zwinderman AH,
Meyer JR, Androux RW, Salamon N, Rabin B, Callahan C, Parrish McGowan JC, Rassek M, Engelbrecht V. Global estimation of
TB, Prager J, Russell EJ. Contrast-enhanced magnetization myelination in the developing brain on the basis of magne-
transfer MR of the brain: importance of precontrast images. tization transfer imaging: a preliminary study. AJNR Am J
AJNR Am J Neuroradiol 1997; 18: 1515–1521 Neuroradiol 2001; 22: 762–766
Ostuni JL, Richert ND, Lewis BK, Frank JA. Characterization of Van Waesberghe JHTM, van Walderveen MAA, Castelijns JA,
differences between multiple sclerosis and normal brain: a Scheltens P, Lycklama à Nijeholt GJ, Polman CH, Barkhof F.
global magnetization transfer application. AJNR Am J Neu- Patterns of lesion development in multiple sclerosis: longi-
roradiol 1999; 20: 501–507 tudinal observations with T1-weighted spin-echo and
Phillips MD, Grossman RI, Miki Y, Wei L, Kolson DL, van Buchem magnetization transfer MR. AJNR Am J Neuroradiol 1998;
MA, Polansky M, McGowan JC, Udupa JK. Comparison of T2 19: 675–683
lesion volume and magnetization transfer ratio histogram Van Waesberghe JHTM, Kamphorst W, de Groot, CJA, van
analysis and of atrophy and measures of lesion burden in Walderveen MAA,Castelijns JA,Ravid R,Lycklama à Nijeholt
patients with multiple sclerosis. AJNR Am J Neuroradiol GJ, van der Valk P, Polman CH, Thompson AJ, Barkhof F. Ax-
1998; 19: 1055–1060 onal loss in multiple sclerosis lesions: magnetic resonance
Pike GB, de Stephano N, Narayanan S, Worsely KJ, Pelletier D, imaging insights into substrates of disability. Ann Neurol
Francis GS, Antel JP, Arnold DL. Multiple sclerosis: magneti- 1999; 46: 747–754
zation transfer MR imaging of white matter before lesion Wolff SD, Balaban RS. Magnetization transfer contrast (MTC)
appearance on T2-weighted images. Radiology 2000; 215: and tissue water relaxation in vivo. Magn Res Med 1989; 10:
824–830 135–144
Rademacher J, Engelbrecht V, Bürgel U, Freund H-J, Zilles K. Wong KT, Grossman RI, Boorstein JM, Lexa FJ, McGowan JC.
Measuring in vivo myelination of human white matter Magnetization transfer imaging of periventricular hyperin-
giber tracts with magnetization transfer MR. Neuroimage tense white matter in the elderly. AJNR Am J Neuroradiol
1999; 9: 393–406 1995; 16: 253–258
1064 References and Further Reading

108 Magnetic Resonance Spectroscopy Horská A, Kaufmann WE, Brant LJ, Naidu S, Harris JC, Barker PB.
In vivo quantitative proton MRSI study of brain develop-
ment from childhood to adolescence. J Magn Reson Imag-
General
ing 2002; 15: 137–143
Hüppi PS, Fusch C, Boesch C, Burri R, Bossi E, Amato M, Her-
Bottomley PA. Human in vivo NMR spectroscopy in diagnostic schkowitz N. Regional metabolic assessment of human
medicine: clinical tool or research probe? Radiology 1989; brain during development by proton magnetic resonance
170: 1–15 spectroscopy in vivo and high-performance liquid chro-
Brooks WM, Friedman SD, Stidley CA. Reproducibility of 1H- matography/gas chromatography in autopsy tissue. Pedi-
MRS in vivo. Magn Reson Med 1999; 41: 193–197 atr Res 1995; 37: 145–150
Castillo M, Kwock L, Mukherji SK.Clinical applications of proton Jacobs MA, Horská A, van Zijl PCM, Barker PB. Quantitative pro-
MR spectroscopy. AJNR Am J Neuroradiol 1996; 17: 1–15 ton MR spectroscopy imaging of normal human cerebel-
Cox IJ. Development and applications of in vivo clinical mag- lum and brain stem. Magn Reson Med 2001; 46: 699–705
netic resonance spectroscopy. Prog Biophys Mol Biol 1996; Kok RD, van den Berg PP, van den Bergh AJ, Nijland R, Heer-
65: 45–81 schap A. Maturation of the human fetal brain as observed
Kreis R. Quantitative localized 1H MR spectroscopy for clinical by 1H MR spectroscopy. Magn Reson Med 2002; 48:
use. J Progr Nucl Magn Reson Spectr 1997; 31: 155–195 611–616
Novotny E, Ashwal S, Shevell M. Proton magnetic resonance Kreis R, Ernst T, Ross BD.Development of the human brain: in vi-
spectroscopy: an emerging technology in pediatric neurol- vo quantification of metabolite and water content with
ogy research. Pediatr Res 1998; 44: 1–10 proton magnetic resonance spectroscopy. Magn Reson
Provencher SW. Estimation of metabolite concentrations from Med 1993; 30: 424–437
localized in vivo proton NMR spectra. Magn Reson Med Kreis R, Hofmann L, Kuhlmann B, Boesch C, Bossi E, Hüppi PS.
1993; 30: 672–679 Brain metabolite composition during early human brain
Ross B, Bluml, S. Magnetic resonance spectroscopy of the hu- development as measured by quantitative in vivo 1H mag-
man brain. Anat Rec [New Anat] 2001; 265: 54–84 netic resonance spectroscopy. Magn Reson Med 2002; 48:
Simmons A, Smail M, Moore E, Williams SCR. Serial precision of 949–958
the metabolic peak area rations and water referenced Lam WWM, Wang ZJ, Zhao H, Berry GT, Kaplan P, Gibson J,
metabolite peak areas in proton MR spectroscopy of the Kaplan BS, Bilaniuk LT, Hunter JV, Haselgrove JC, Zimmer-
human brain. Magn Reson Imaging 1998; 16: 319–330 mann RA. 1H MR Spectroscopy of the basal ganglia in child-
Tedeschi G,Bertolino A,Campbell G,Barnett AS,Duyn JH,Jacob hood:a semiquantitative analysis.Neuroradiology 1998; 40:
PK, Moonen CTW, Alger JR, di Chiro G. Reproducibility of 315–323
proton MR spectroscopy imaging findings. AJNR Am J Neu- Mader I, Seeger U, Karitzky J, Erb M, Schick F, Klose U. Proton
roradiol 1996; 17: 1871–1879 magnetic resonance spectroscopy with metabolite nulling
Van Zijl PCM, Barker PB. Magnetic resonance spectroscopy and reveals regional differences of macromolecules in normal
spectroscopic imaging for study of brain metabolism. Ann human brain. J Magn Reson Imaging 2002; 16: 538–546
N Y Acad Sci 1997; 820: 75–96 Mascalchi M, Burgnoli R, Guerrini L, Belli G, Nistri M, Politi LS,
Gavazzi C, Lolli F, Argenti G, Villari N. Single-voxel long TE
Normal Age-dependent and Regional Changes 1
H-MR spectroscopy of the normal brainstem and cerebel-
lum. J Magn Reson Imaging 2002; 16: 532–537
Azzopardi D, Wyatt JS, Hamilton PA, Cady EB, Delpy DT, Hope McLean MA,Woermann FG, Barker GJ, Duncan JS. Quantitative
PL, Reynolds EOR. Phosphorus metabolites and intracellu- analysis of short echo time 1H-MRSI of cerebellar gray and
lar pH in the brains of normal and small for gestational age white matter. Magn Reson Med 2000; 44: 401–411
infants investigated by magnetic resonance spectroscopy. Nakada T, Kwee IL. 31P localized spectroscopy of fetal brain in
Pediatr Res 1989; 25: 440–444 utero. Magn Reson Med 1993; 29: 122–124
Blüml S, Seymour KJ, Ross BD. Developmental changes in Pouwels PJW, Frahm J. Regional metabolite concentrations in
choline- and ethanolamine-containing compounds mea- human brain as determined by quantitative localized pro-
sured with proton-decoupled 31P MRS in vivo human brain. ton MRS. Magn Reson Med 1998; 39: 53–60
Magn Reson Med 1999; 42: 643–654 Pouwels PJW,Brockmann K,Kruse B,Wilken B,Wick M,Hanefeld
Buchli R, Boesiger MP, Rumpel H. Developmental changes of F, Frahm J. Regional age dependence of human brain
phosphorus metabolite concentrations in the human metabolites from infancy to adulthood as detected by
brain: a 31P magnetic resonance spectroscopy study in vivo. quantitative localized proton MRS. Pediatr Res 1999; 46:
Pediatr Res 1994; 35: 431–435 474–485
Buchli R, Duc CO, Martin E, Boesiger P. Assessment of absolute Provencher SW. Estimation of metabolite concentrations from
metabolite concentrations in human tissue by 31P MRS in localized in vivo proton NMR spectra. Magn Reson Med
vivo. 1. Cerebrum, cerebellum, cerebellar gray and white 1993; 30: 672–679
matter. Magn Reson Med 1994; 32: 447–452 Tedeschi G, Bertolino A, Righini A, Campbell G, Raman R, Duyn
Filippi CG, Uluğ AM, Deck MDF, Zimmerman RD, Heier LA. De- JH, Moonen CTW, Alger JR, di Chiro G. Brain regional distrib-
velopmental delay in children: assessment with proton MR ution pattern of metabolite signal intensities in young
spectroscopy. AJNR Am J Neuroradiol 2002; 23: 882–888 adults by proton magnetic resonance spectroscopy imag-
Hanaoka S,Takashima S,Morooka K.Study of the maturation of ing. Neurology 1995; 45: 1384–1391
the child’s brain using 31P-MRS. Pediatr Neurol 1998; 18: Toft PB, Leth H, Lou HC, Pryds O, Henriksen O. Metabolite con-
305–310 centrations in the developing brain estimated with proton
MR spectroscopy. J Magn Reson Imaging 1994; 4: 674–680
References and Further Reading 1065

van der Knaap MS,van der Grond J,van Rijen PC,Faber JAJ,Valk pH
J, Willemse K. Age-dependent changes in localized proton
and phosphorus MR spectroscopy of the brain. Radiology Madden A, Leach MO, Sharp JC, Collins DJ, Easton D. A quanti-
1990; 176: 509–515 tative analysis of the accuracy of in vivo pH measurements
Vigneron DB,Barkovich AJ, Noworolski SM, von dem Busche M, with 31P NMR spectroscopy: assessment of pH measure-
Henry RG, Lu Y, Partridge JC, Gregory G, Ferriero DM. Three- ment methodology. NMR Biomed 1991; 4: 1–11
dimensional proton MR spectroscopic imaging of prema- Moon RB, Richards JH. Determination of intracellular pH by 31P
ture and term neonates. AJNR Am J Neuroradiol 2001; 22: magnetic resonance. J Biol Chem 1973; 248: 7276–7278
1424–1433 Petroff OAC, Prichard JW, Behar KL, Alger JR, den Hollander JA,
Shulman RG. Cerebral intracellular pH by 31P nuclear mag-
Metabolites: General netic resonance spectroscopy. Neurology 1985; 35: 781–
788
Bhakoo KK, Williams IT, Williams SR, Gadian DG, Noble MD. Pro- Pettegrew JW, Withers G, Panchalingham K, Post JFM. Consid-
ton nuclear magnetic resonance spectroscopy of primary erations for brain pH assessment by 31P NMR. Magn Reson
cells derived from nervous tissue. J Neurochem 1996; 66: Imaging 1988; 6: 135–142
1254–1263
Govindaraju V, Young K, Mandsley AA. Proton NMR chemical
shifts and coupling constants for brain metabolites. NMR N-Acetylaspartate
Biomed 2000; 13: 129–153
Michaelis T, Merboldt KD, Hänicke W, Gyngell ML, Bruhn H, Baslow MH. Functions of N-acetyl-l-aspartate and N-acetyl-l-
Frahm J. On the identification of cerebral metabolites in aspartylglutamate in the vertebrate brain: role in glial cell-
localized 1H NMR spectra of human brain in vivo. NMR Bio- specific signaling. J Neurochem 2000; 75: 453–459
med 1991; 4: 90–98 Baslow MH. N-Acetylaspartate in the vertebrate brain: metab-
Urenjak J, Williams SR, Gadian DG, Noble M. Proton nuclear olism and function. Neurochem Res 2003; 28: 941–953
magnetic resonance spectroscopy unambiguously identi- Bates TE, Strangward M, Keelan J, Davey GP, Munro PMG, Clark
fies different neural cell types. J Neurosci 1993; 13: 981–989 JB. Inhibition of N-acetylaspartate production: implications
for 1H MRS studies in vivo. Neuroreport 1996; 7: 1397–1400
Bhakoo KK, Pearce D. In vitro expression of N-acetyl aspartate
Phosphomonoester / Phosphodiester by oligodendrocytes: implications for proton magnetic res-
onance spectroscopy signal in vivo. J Neurochem 2000; 74:
254–262
Daly PF, Lyon RC, Faustino PJ, Cohen JS. Phospholipid metabo-
Birken DL, Oldendorf WH. N-acetyl-L-aspartic acid: a literature
lism in cancer cells monitored by 31P NMR spectroscopy.
review of a compound prominent in 1H-NMR spectroscop-
J Biol Chem 1987; 262: 14875–14878
ic studies of brain. Neurosci Biobehav Rev 1989; 13: 23–31
Dawson RMC. Enzymic pathways of phospholipid metabolism
Bjartmar C, Battistuta J, Terada N, Dupree E, Trapp BD. N-Acety-
in the nervous system. In: Eichberg J, ed. Phospholipids in
laspartate is an axon-specific marker of mature white mat-
nervous tissues. New York: Wiley, 1985, pp 45–78
ter in vivo: a biochemical and immunohistochemical study
Gyulai L, Bolinger L, Leigh JS Jr, Barlow C, Chance B. Phospho-
on the rat optic nerve. Ann Neurol 2002; 51: 51–58
rylethanolamine – the major constituent of the phospho-
Block W, Träber, Flacke S, Jessen F, Pohl C, Schild H. In-vivo pro-
monoester peak observed by 31P-NMR on developing dog
ton MR-spectroscopy of the human brain: assessment of N-
brain. FEBS Lett 1984; 178: 137–142
acetylaspartate (NAA) reduction as a marker for neurode-
Kilby PM, Bolas NM, Radda GK. 31P-NMR study of brain phos-
generation. Amino Acids 2002; 23: 317–323
pholipid structures in vivo. Biochim Biophys Acta 1991;
Chakrabourty G, Mekala P,Yahya D,Wu G, Ledeen RW. Intraneu-
1085: 257–264
ronal N-acetylaspartate supplies acetyl groups for myelin
McNamara R, Arias-Mendoza F, Brown TR. Investigation of
lipid synthesis: evidence for myelin-associated aspartoacy-
broad resonances in 31P NMR spectra of the human brain in
lase. J Neurochem 2001; 78: 736–745
vivo. NMR Biomed 1994; 7: 237–242
De Stefano N,Matthews PM,Arnold DL.Reversible decreases in
Murphy EJ, Rajagopalan B, Brindle KM, Radda GK. Phospholipid
N-acetylaspartate after acute brain injury.Magn Reson Med
bilayer contribution to 31P NMR spectra in vivo. Magn
1995; 34: 721–727
Reson Med 1989; 12: 282–289
Gasparovic C, Arfai N, Smid N, Feeney DM. Decrease and recov-
Porcellati G, Arienti G. Metabolism of phosphoglycerides. In:
ery of N-acetylaspartate/creatine in rat brain remote from
Lajtha A, ed. Handbook of neurochemistry, vol 3: Metabo-
focal injury. J Neurotrauma 2001; 18: 241–246
lism in the nervous system. New York, Plenum Press: 1983,
Patel TB, Clark JB. Synthesis of N-acetyl-L-aspartate by rat brain
pp 133–161
mitochondria and its involvement in mitochondrial/cy-
Stanley JA, Pettegrew JW. Postprocessing method to segre-
tosolic carbon transport. Biochem J 1979; 184: 539–546
gate and quantify the broad components underlying the
Pouwels PJW, Frahm J. Differential distribution of NAA and
phosphodiester spectral region of in vivo 31 P brain spec-
NAAG in human brain as determined by quantitative local-
tra. Magn Reson Med 2001; 45: 390–396
ized proton MRS. NMR Biomed 1997; 10: 73–78
Sun GY, Foudin LL. Phospholipid composition and metabolism
Tallan HH, Moore S, Stein WH. N-Acetyl-l aspartic acid in brain.
in the developing and aging nervous system. In: Eichberg J,
J Biol Chem 1956; 219: 257–264
ed. Phospholipids in nervous tissues. New York:Wiley, 1985,
Taylor DL,Davies SEC,Obrenovitch TP,Doheny MH,Patsalos PN,
pp 79–134
Clark JB, Symon L. Investigation into the role of N-acetylas-
Van der Grond J, Dijkstra G, Roelofsen B, Mali WPTM. 31P-NMR
partate in cerebral osmoregulation. J Neurochem 1995; 65:
determination of phosphomonoesters in relation to phos-
275–281
pholipid biosynthesis in testis of the rat at different ages.
Biochim Biophys Acta 1991; 1074: 189–194
1066 References and Further Reading

Urenjak J,Williams SR, Gadian DG, Noble M. Specific expression Shonk T, Ross BD. Role of increased cerebral myo-inositol in the
of N-acetylaspartate in neurons, oligodendrocyte-type-2 dementia of Down syndrome. Magn Reson Med 1995; 33:
astrocyte progenitors, and immature oligodendrocytes in 858–861
vitro. J Neurochem 1992; 59: 55–61 Videen JS, Michaelis T, Pinto P, Ross BD. Human cerebral os-
molytes during chronic hyponatremia. A proton magnetic
resonance spectroscopy study. J Clin Invest 1995; 95:
Creatine 788–793

Chance B, Leigh JS Jr, Clark BJ, Maris J, Kent J, Nioka S, Smith D.


Control of oxidative metabolism and oxygen delivery in Lactate
human skeletal muscle: a steady-state analysis of the
work/energy cost transfer function. Proc Natl Acad Sci
López-Villegas D, Lenkinski RE, Wehrli SL, Ho W-Z, Douglas SD.
1985; 82: 8384–8388
Lactate production by human monocytes / macrophages
Dringen R, Verleysdonk S, Hamprecht B, Willker W, Leibfritz D,
determined by proton MR spectroscopy. Magn Reson Med
Brand A. Metabolism of glycine in primary astroglial cells:
1995; 34: 32–38
synthesis of creatine, serine and glutathione. J Neurochem
Petroff OAC, Graham GD, Blamire AM, Al-Rayess M, Rothman
1998; 70: 835–840
DL, Fayad PB, Brass LM, Shulman RG, Prichard JW. Spectro-
Wyss M, Kaddurah-Daouk R. Creatine and creatinine metabo-
scopic imaging of stroke in humans: histopathology corre-
lism. Physiol Rev 2000; 80: 1107–1213
lates of spectral changes. Neurology 1992; 42: 1349–1354
Prichard JW. What the clinician can learn from MRS lactate
Choline measurements. NMR Biomed 1991; 4: 99–102
Veech RL. The metabolism of lactate. NMR Biomed 1991; 4:
53–58
Gupta RK, Cloughesy TF, Sinha U, Garakian J, Lazareff J, Rubino
G, Rubino L, Becker DP, Vinters HV, Alger JR. Relationships
between choline magnetic resonance spectroscopy,appar-
Glucose
ent diffusion coefficient an quantitative histopathology in
human glioma. J Neurooncol 2000; 50: 215–226
Miller BL. A review of chemical issues in 1H NMR spectroscopy: Gruetter R, Rothman DL, Novotny EJ, Shulman GI, Prichard JW,
N-acetyl-L-aspartate, creatine and choline. NMR Biomed Shulman RG. Detection and assignment of the glucose sig-
1991; 4: 47–52 nal in 1H NMR difference spectra of the human brain. Magn
Miller BL, Chang L, Booth R, Ernst T, Conford M, Nikas D, McBride Reson Med 1992; 27: 183–188
D, Jenden DJ. In vivo 1H MRS choline: correlation with in vi- Gruetter R, Novotny EJ, Boulware SD, Rothman DL, Shulman
vo chemistry/histiology. Life Sci 1996; 58:1929–1935 RG. 1H NMR Studies of glucose transport in the human
brain. J Cereb Blood Flow Metab 1996; 16: 427–438
Rothman DL. Studies of metabolic compartmention and glu-
Myo-inositol cose transport using in vivo MRS. NMR Biomed 2001; 14:
149–160
Brand A, Richter-Landsberg C, Leibfritz D. Multinuclear NMR
studies on the energy metabolism of glial and neuronal Glutamine / Glutamate / GABA
cells. Dev Neurosci 1993; 15: 289–298
Glanville NT, Byers DM. Differences in the metabolism of inosi-
Chamuleau RAFM, Bosman DK, Bové WMMJ, Luyten PR, den
tol. Biochim Biophys Acta 1989; 104: 169–179
Hollander JA. What the clinician can learn from MR gluta-
Häussinger D, Laubenberger J, vom Dahl S, Ernst T, Bayer S,
mine/glutamate assays. NMR Biomed 1991; 4: 103–108
Langer M, Gerok W, Hennig J. Proton magnetic resonance
Govindaraju V, Basus VJ, Matson GB, Maudsley AA. Measure-
spectroscopy studies on human brain myo-inositol in hy-
ment of chemical shifts and coupling constants for gluta-
po-osmolarity and hepatic encephalopathy. Gastroenterol-
mate and glutamine. Magn Reson Med 1998; 39: 1011–
ogy 1994; 107: 1475–1480
1013
Lee JH, Arginue E, Ross BD. Brief report: organic osmolytes in
Keltner JR,Wald LL, Frederick BB, Renshaw PF. In vivo detection
the brain of an infant with hypernatremia. N Engl J Med
of GABA in human brain using a localized double-quantum
1994; 331: 439–442
filter technique. Magn Reson Med 1997; 37: 366–371
Michaelis T, Helms G, Merboldt KD, Hänicke W, Bruhn H, Frahm
Ross BD.Biochemical considerations in 1H spectroscopy.Gluta-
J. Identification of scyllo-inositol in proton NMR spectra of
mate and glutamine; myo-inositol and related metabolites.
human brain in vivo. NMR Biomed 1993; 6: 105–109
NMR Biomed 1991; 4: 59–63
Ross BD.Biochemical considerations in 1H spectroscopy.Gluta-
Rothman DL, Hanstock CC, Petroff OAC, Novotny EJ, Prichard
mate and glutamine; myo-inositol and related metabolites.
JW, Shulman RG. Localized 1H NMR spectra of glutamate in
NMR Biomed 1991; 4: 59–63
the human brain. Magn Reson Med 1992; 25: 94–106
Rumpel H, Lim WEH, Chang HM, Chan LL, Ho GL,Wong MC,Tan
Sonnewald U, Westergaard N, Schousboe A, Svendsen JS, Un-
KP. Is myo-inositol a measure of glial swelling after stroke?
sgard G, Peterson SB. Direct demonstration by [13C]NMR
A magnetic resonance study. J Magn Reson Imaging 2003;
spectroscopy that glutamine from astrocytes is a precursor
17: 11–19
for GABA synthesis in neurons. Neurochem Int 1993;
Seaquist ER, Gruetter R. Identification of a high concentration
22:19–29
of scyllo-inositol in the brain of a healthy human subject us-
Sonnewald U, Westergaard N, Shousboe A. Glutamate trans-
ing 1H- and 13C-NMR. Magn Reson Med 1998; 39: 313–316
port and metabolites in astrocytes. Glia 1997; 21: 56–63
References and Further Reading 1067

Taurine Degaonkar MN, Khubchandihani M, Dhawan JK, Jayasundar R,


Jagannathan NR. Sequential proton MRS study of brain
Hardy DL, Norwood TJ. Spectral editing technique for the in metabolite changes monitored during a complete patho-
vitro and in vivo detection of taurine. Nucl Magn Reson logical cycle of demyelination and remyelination in a
1998; 133: 70–78 lysophosphatidyl choline (LPC)-induced experimental de-
Trenkner E, Sturman JA. The role of taurine in the survival and myelinating lesion model. MNR Biomed 2002; 15: 293–300
function of cerebral cells in cultures of early postnatal cat. Farina L, Bizzi A, Finocchiaro G, Pareyson D, Sghirlanzoni A,
Int J Dev Neurosci 1991; 9: 77–88 Bertagnolio B, Savaoirdo M, Naidu SB, Singhal BS, Wenger
DA. MR imaging and proton MR spectroscopy in adult
Krabbe disease. AJNR Am J Neuroradiol 2000; 21: 1478–
1482
Neuronal Degeneration
Kruse B, Hanefeld F, Christen HJ, Bruhn H, Michaelis T, Hänicke
W, Frahm J. Alterations of brain metabolites in metachro-
Block W, Karitzky J, Träber F, Pohl C, Keller E, Mundegar R, matic leukodystrophy as detected by localized proton
Lamerichs R, Rink H, Ries F, Schild HH, Jerusalem F. Proton magnetic resonance spectroscopy in vivo. J Neurol 1993;
magnetic resonance spectroscopy of the primary motor 241: 68–74
cortex in patients with motor neuron disease. Arch Neurol Silver NC, Barker RA, MacManus DG, Barker GJ, Thom M,
1998; 55: 931–936 Thomas DGT, McDonald WI, Miller DH. Proton magnetic res-
Brockmann K, Pouwels PJW, Christen H-J, Frahm J, Hanefeld F. onance spectroscopy in a pathologically confirmed acute
Localized proton magnetic resonance spectroscopy of demyelinating lesion. J Neurol 1997; 244: 204–207
cerebral metabolic disturbances in children with neuronal van der Knaap MS, van der Grond J, Luyten PR, den Hollander
ceroid lipofuscinosis. Neuropediatrics 1996; 27: 242–248 JA, Nauta JJP, Valk J. 1H and 31P magnetic resonance spec-
Chan S, Shungu DC, Douglas-Akinwande A, Lange DJ, Rowland troscopy of the brain in degenerative cerebral disorders.
LP. Motor neuron diseases: comparison of single-voxel pro- Ann Neurol 1992; 31: 202–211
ton MR spectroscopy of the motor cortex with MR imaging
of the brain. Radiology 1999; 212: 763–769
Ellis CM, Simmons A, Andrews C, Dawson JM, Williams SCR, Hypomyelination
Leigh PN. A proton magnetic resonance spectroscopy
study in ALS. Correlation with clinical findings. Neurology Bonavita S, Schiffmann R, Moore DF, Free K, Choi B, Patronas N,
1998; 51: 1104–1109 Virta A, Boespflüg-Tanguy O, Tedeschi G. Evidence for neu-
Frederico F, Simone IL, Lucivero V, de Mari M, Gianinni P, Iliceto roaxonal injury in patients with proteolipid protein gene
G, Mezzapesa DM, Lamberti P. Proton magnetic resonance mutations. Neurology 2001; 56: 785–788
spectroscopy in Parkinson’s disease and progressive Gebern JY,Yook DA,Moore GJ,Wilds IB,Faulk MW,Klugmann M,
supranuclear palsy. J Neurol Neurosurg Psychiatry 1997; 62: Nave K-A, Sistermans ES, van der Knaap MS, Bird TD, Shy ME,
239–242 Kamholz JA, Griffiths IR. Patients lacking the major CNS
Miyazaki M, Hashimoto T, Yonada Y, Tayama M, Harada M, myelin protein, proteolipid protein 1, develop length-de-
Miyoshi H, Kawano N, Murayama N, Kondo I, Kuroda Y. Pro- pendent axonal degeneration in the absence of demyeli-
ton magnetic resonance spectroscopy on childhood-onset nation and inflammation. Brain 2002; 125: 551–561
dentatorubral-pallidoluysian atrophy (DRPLA). Brain Dev Pizzini F, Fatemi AS, Barker PB, Nagae-Poetscher LM, Horská A,
1996; 18: 142–146 Zimmerman AW, Moser HW, Bibat G, Naidu S. Proton MR
Rooney WD, Miller RG, Gelinas D, Schuff N, Maudsley AA, Wein- spectroscopic imaging in Pelizaeus-Merzbacher disease.
er MW. Decreased N-acetylaspartate in motor cortex and AJNR Am J Neuroradiol 2003; 24: 1683–1689
corticospinal tract in ALS. Neurology 1998; 50: 1800–1805 Plecko B, Stöckler-Ipsirolglu S, Gruber S, Mlynarik V, Moser E,
Tedeschi G,Bertolini A,Massaquoi SG,Campbell G,Patronas NJ, Simbrunner J, Ebner F, Bernert G, Harrer G, Gal A, Prayer D.
Bonavita S, Barnett AS, Alger JR, Hallett M. Proton magnetic Degree of hypomyelination and magnetic resonance spec-
resonance spectroscopic imaging in patients with cerebel- troscopy findings in patient with Pelizaeus Merzbacher
lar degeneration. Ann Neurol 1996; 39: 71–78 phenotype. Neuropediatrics 2003; 34: 127–136
Tedeschi G, Litvan I, Bonavita S, Bertolini A, Lundbom N, Pa- Spalice A, Popolizio T, Parisi P, Scarabino T, Iannetti P. Proton MR
tronas NJ, Hallett M. Proton magnetic resonance spectro- spectroscopy in connatal Pelizaeus-Merzbacher disease.
scopic imaging in progressive supranuclear palsy, Parkin- Pediatr Radiol 2000; 30: 171–175
son’s disease and corticobasal degeneration. Brain 1997; Takanashi J, Inoue K, Tomita M, Kurihara A, Morita F, Ikehira H,
120: 1541–1552 Tanada S, Yoshitome E, Kohno Y. Brain N-acetylaspartate is
Tedeschi G, Bonavita S, Barton NW, Bertolino A, Frank JA, Pa- elevated in Pelizaeus-Merzbacher disease with PLP1 dupli-
tronas NJ, Alger JR, Shiffmann R. Proton magnetic reso- cation. Neurology 2002; 58: 237–241
nance spectroscopic imaging in the clinical evaluation of van der Knaap MS, Naidu S, Pouwels PJ, Bonavita S, van Coster
patients with Niemann-Pick type C disease. J Neurol Neuro- R, Lagae L, Sperner J, Surtees R, Schiffmann R, Valk J. New
surg Psychiatry 1998; 65: 72–79 syndrome characterized by hypomyelination with atrophy
of the basal ganglia and cerebellum. AJNR Am J Neuroradi-
ol 2002; 23: 1466–1474
Demyelinination

Brockmann K, Dechent P, Wilken B, Rusch O, Frahm J, Hanefeld


F.Proton MRS profile of cerebral metabolic abnormalities in
Krabbe disease. Neurology 2003; 60: 819–825
1068 References and Further Reading

Cystic White Matter Degeneration Naegele T,Grodd W,Viebahn R,Seeger U,Klose U,Seitz D,Kaiser
S, Mader I, Mayer J, Lauchart W, Gregor M, Voight K. MR
Hanefeld F, Holzbach U, Kruse B, Wilichowski E, Christen H-J, imaging and 1H spectroscopy of brain metabolites in he-
Frahm J. Diffuse white matter disease in three children: an patic encephalopathy: time course of renormalization after
encephalopathy with unique features on magnetic reso- liver transplantation. Radiology 2000; 216: 683–691
nance imaging and proton magnetic resonance spec- Ross BD, Jacobson S,Villamil F, Korula J, Kreis R, Ernst T, Shonk T,
troscopy. Neuropediatrics 1993; 24: 244–248 Moats RA. Subclinical hepatic encephalopathy proton MR
Salvan A-M, Chabrol B, Lamoureux S, Confort-Gouny S, Coz- spectroscopic abnormalities. Radiology 1994; 193: 457–463
zone PJ, Vion-Dury J. In vivo brain proton MR spectroscopy Van den Heuvel AG, van der Grond J, van Rooij LG, van Wasse-
in a case of molybdenum cofactor deficiency. Pediatr Radi- naer-van Hall HN, Hoogenraad TU, Mali WPTM. Differential
ol 1999; 29: 846–848 between portal-systemic encephalopathy and neuro-
Schiffmann R, Moller JR, Trapp BD, Shih HH-L, Farrer RG, Katz degenerative disorders in patients in Wilson disease: H-1
DA, Alger JR, Parker CC, Hauer PE, Kaneski CR, Heiss JD, Kaye MR spectroscopy. Radiology 1997; 203: 539–543
EM, Quarles RH, Brady RO, Barton NW. Childhood ataxia
with diffuse central nervous system hypomyelination. Ann
Neurol 1994; 35: 331–340 Osmolytes
Tedeschi G, Schiffmann R, Barton NW, Shih HH-L, Gospe SM Jr,
Brady RO, Alger JR, di Chiro G. Proton magnetic resonance Davies SEC, Gotoh M, Richards DA, Obrenovitch TP. Hypoosmo-
spectroscopic imaging in childhood ataxia with diffuse larity induces an increase of extracellular N-acetylaspartate
central nervous system hypomyelination. Neurology 1995; concentration in the rat striatum. Neurchem Res 1998; 23:
45: 1526–1532 1021–1025
van der Knaap MS, Kamphorst W, Barth PG, Kraaijeveld CL, Gut Heilig CW, Stromski ME, Blumenfeld JD, Lee JP, Gullans SR. Char-
E,Valk J. Phenotypic variation in leukoencephalopathy with acterization of the major brain osmolytes that accumulate
vanishing white matter. Neurology 1998; 51: 540–547 in salt-loaded rats. Am J Physiol 1989; 257: F1108–F1116
van der Knaap MS, Barth PG, Gabreëls FJM, Franzoni E, Begeer Lee JH, Arcinue E, Ross BD. Organic osmolytes in the brain of an
JH, Stroink H, Rotteveel JJ, Valk J. A new leukoencephalo- infant with hypernatremia. N Engl J Med 1994; 331:
pathy with vanishing white matter. Neurology 1997; 48: 439–442
845–855 Lien Y-H H, Shapiro JI, Chan L. Study of brain electrolytes and
organic osmolites during correction of chronic hypona-
tremia. Implications for the pathogenesis of central pon-
Hyperammonemia tine myelinolysis. J Clin Invest 1991; 88: 303–309
Strange K, Emma F, Paredes A, Morrison R. Osmoregulatory
Chol C-G,Yoo HW.Localized proton MR spectroscopy in infants changes in myo-inositol content and Na+ / myo-inositol co-
with urea cycle defect. AJNR Am J Neuroradiol 2001; 22: transport in rat cortical astrocytes. Glia 1994; 12: 35–43
834–837 Videen JS, Michaelis T, Pinto R, Ross BD. Human cerebral os-
Connelly A, Cross JH, Gadian DG, Hunter JV, Kirkham FJ, molytes during chronic hyponatremia. A proton magnetic
Leonard JV. Magnetic resonance spectroscopy shows in- resonance spectroscopy study. J Clin Invest 1995; 95:
creased brain glutamine in ornithine carbamoyl trans- 788–793
ferase deficiency. Pediatr Res 1993; 33: 77–81
Haseler LJ, Sibbitt WL Jr, Mojtahezadeh HN, Reddy S, Aragwal
VP, McCarthy DM. Proton MR spectroscopic measurement Perinatal Hypoxia–Ischemia
of neurometabolites in hepatic encephalopathy during
oral lactulose therapy. AJNR Am J Neuroradiol 1998; 19: Azzopardi D, Wyatt JS, Cady EB, Delphy DT, Baudin J, Stewart
1681–1686 AL, Hope PL, Hamilton PA, Reynolds EOR. Prognosis of new-
Kraft E, Trenkwalder C, Bergh FT, Auer DP. Magnetic resonance born infants with hypoxic-ischemic brain injury assessed
proton spectroscopy of the brain in Wilson’s disease. J Neu- by phosphorus magnetic resonance spectroscopy. Pediatr
rol 1999; 246: 693–699 Res 1989; 25: 445–451
Kreis R, Farrow N, Ross BD. Localized 1H NMR spectroscopy in Groenendaal F, Veenhoven RH, van der Grond J, Jansen GH,
patients with chronic hepatic encephalopathy. Analysis of Witkamp TD, de Vries LS. Cerebral lactate and N-acetyl-as-
changes in cerebral glutamine, choline and inositols. NMR partate/choline ratios in asphyxiated full-term neonates
Biomed 1991; 4: 109–116 demonstrated in vivo using proton magnetic resonance
Kreis R,Ross BD,Farrow NA,Ackerman Z.Metabolic disorders of spectroscopy. Pediatr Res 1994; 35: 148–151
the brain in chronic hepatic encephalopathy detected with Groenendaal F, van der Grond J, Witkamp TD, de Vries LS. Pro-
H-1 MR spectroscopy. Radiology 1992; 182: 19–27 ton magnetic resonance spectroscopic imaging in neona-
Laubenberger J, Häussinger D, Bayer S, Gufler H, Hennig J, tal stroke. Neuropediatrics 1995; 26: 243–248
Langer M. Proton magnetic resonance spectroscopy of the Groenendaal F, van der Grond J, Eken P, van Haastert IC, Rade-
brain in symptomatic and asymptomatic patients with liv- maker KJ, Toet MC, de Vries LS. Early cerebral proton MRS
er cirrhosis. Gastroenterology 1997; 112; 1610–1616 and neurodevelopmental outcome in infants with cystic
Lien YHH, Michaelis T, Moats RA, Ross BD. Scyllo-inositol deple- leukomalacia. Dev Med Child Neurol 1997; 39: 373–379
tion in hepatic encephalopathy. Life Sci 1994; 54: 1507– Hanrahan JD, Sargentoni J, Azzopardi D, Manji K, Cowan FM,
1512 Rutherford MA, Cox IJ, Bell JD, Bryant DJ, Edwards AD. Cere-
bral metabolism within 18 hours of birth asphyxia: a proton
magnetic resonance spectroscopic study. Pediatr Res 1996;
39: 584–590
References and Further Reading 1069

Hanrahan JD, Cox IJ, Edwards AD, Cowan FM, Sargentoni J, Bell Capizzano AA, Schuff N, Armend DL, Tanabe JL, Norman D,
JD, Bryant DJ, Rutherford MA. Persistent increases in cerbral Maudsley AA, Jagust W, Chui HC, Fein G, Segal MR, Weiner
lactate concentration after birth asphyxia.Pediatr Res 1998; MW. Subcortical ischemic vascular dementia: assessment
44: 304–311 with quantitative MR imaging and 1H MR spectroscopy.
Hanrahan JD, Cox IJ, Azzopardi D, Ocwan FM, Sargentoni J, Bell AJNR Am J Neuroradiol 2000; 21: 621–630
JD, Bryant DJ, Edwards AD. Relation between proton mag- Oppenheimer SM, Bryan RN, Conturo TE, Soher BJ, Preziosi TJ,
netic resonance spectroscopy within 18 hours of birth as- Barker PB. Proton magnetic resonance spectroscopy and
phyxia and neurodevelopment at 1 year of age. Dev Med gadolinium-DTPA perfusion imaging of asymptomatic MRI
Child Neurol 1999; 41: 76–82 white matter lesions. Magn Reson Med 1995; 33: 61–68
Lorek A, Takei Y, Cady EB, Wyatt JS, Penrice J, Edwards AD, Pee- Van der Grond J, Ramos LMP, Eikelboom BC, Mali WPTM. Cere-
bles D,Weylezinska M, Owen-Reece H, Kirkbridge V, Cooper bral metabolic differences between the severe and critical
CE, Aldridge RF, Roth SC, Brown G, Delpy DT, Reynolds EOR. hypoperfused brain. Neurology 1996; 47: 399–404
Delayed (“secondary”) cerebral energy failure after acute
hypoxia–ischemia in the newborn piglet: continuous 48-
hour studies by phosphorus magnetic resonance spec- Mitochondrial Defects
troscopy. Pediatr Res 1994; 36: 699–706
Martin E, Buchli R, Ritter S, Schmid R, Largo RH, Boltshauser E, Barbiroli B, Montagna P, Martinelli P, Lodi R, Iotti S, Cortelli P, Fu-
Fanconi S, Duc G, Rumpel H. Diagnostic and prognostic val- nicello R, Zaniol P. Defective brain energy metabolism
ue of cerebral 31P magnetic resonance spectroscopy in shown by in vivo 31P MR spectroscopy in 28 patients with
neonates with perinatal asphyxia. Pediatr Res 1996; 40: mitochondrial cytopathies. J Cereb Blood Flow Metab
749–758 1993; 13: 469–474
Moorcraft J, Bolas NM, Ives NK, Ouwerkerk R, Smyth J, Ra- Barbiroli B, Montagna P, Cortelli P, Lotti S, Lod R, Barboni P,
jagopalan B, Hope PL, Radda GK. Global and depth resolved Monari L, Laguresi E, Frassineti C, Zaniol P. Defective brain
phosphorus magnetic resonance spectroscopy to predict and muscle energy metabolism shown by in vivo 31P mag-
outcome after birth asphyxia. Arch Dis Child 1991; 66: netic resonance spectroscopy in nonaffected carriers of
1119–1123 11778 mtDNA mutation. Neurology 1995; 45: 1364–1369
Moorcraft J, Bolas NM, Ives NK, Sutton P, Blackledge MJ, Ra- Bianchi MC,Tosetti M, Battini R, Manca ML, Mancuso M, Cioni G,
jagopalan B, Hope PL, Radda GK. Spatially localized mag- Canapicchi R, Siciliano G. Proton MR spectroscopy of mito-
netic resonance spectroscopy of the brains of normal and chondrial diseases: analysis of brain metabolic abnormali-
asphyxiated newborns. Pediatrics 1991; 87: 273–282 ties and their possible diagnostic relevance. AJNR Am J
Penrice J, Lorek A, Cady EB, Amess PN,Wylezinski M, Cooper CE, Neuroradiol 2003; 24: 1958–1966
D’Souza P, Brown GC, Kirkbride V, Edwards AD, Wyatt JS, Cross JH, Gadian DG, Connelly A, Leonard JV. Proton magnetic
Reynolds EOR. Proton magnetic resonance spectroscopy of resonance spectroscopy studies in lactic acidosis and mito-
the brain during acute hypoxia-ischemia and delayed cere- chondrial disorders. J Inherit Metab Dis 1993; 16: 800–811
bral energy failure in the newborn piglet. Pediatr Res 1997; Detre JA, Wang Z, Bogdan AR, Gusnard DA, Bay CA, Bingham
41: 795–802 PM, Zimmerman RA. Regional variation in brain lactate in
Robertson NJ, Lewis RH, Cowan FM, Allsop JM, Counsell SJ, Ed- Leigh syndrome by localized 1H magnetic resonance spec-
wards AD, Cox IJ. Early increases in brain myo-inositol mea- troscopy. Ann Neurol 1991; 29: 218–221
sured by proton magnetic resonance spectroscopy in term Eleff SM, Barker PB, Blackband SJ, Chatham JC, Lutz NW, Johns
infants with neonatal encephalopathy.Pediatr Res 2001;50: DR, Bryan RN, Hurko O. Phosphorus magnetic resonance
692–700 spectroscopy of patients with mitochondrial cytopathies
Roth SC, Azzopardi D, Edwards AD, Baudin J, Cady EB, demonstrates decreased levels of brain phosphocreatine.
Townsend J, Delpy DT, Stewart AL, Wyatt JS, Osmund E, Ann Neurol 1990; 27: 626–630
Reynolds R. Relation between cerebral oxidative metabo- Herzberg NH, van Schooneveld MJ, Bleeker-Wagemakers E,
lism following birth asphyxia, and neurodevelopmental Zwart R, Cremers FPM, van der Knaap MS, Bolhuis PA, de
outcome and brain growth at one year.Dev Med Child Neu- Visser M. Kearns-Sayre syndrome with a phenocopy of
rol 1992; 34: 285–295 choroideremia instead of pigmentary retinopathy. Neuro-
Roth SC, Baudin J, Cady E, Johal K, Towsend JP, Wyatt JS, logy 1993; 43: 218–221
Reynolds EOR, Stewart AL. Relation of deranged neonatal Kamada K, Takeuchi F, Houkin K, Kitagawa M, Kuriki S, Ogata A,
cerebral oxidative metabolism with neurodevelopmental Tashiro K, Koyanagi I, Mitsumori K, Iwasaki Y. Reversible
outcome and head circumference at 4 years.Dev Med Child brain dysfunction in MELAS: MEG and 1H MRS analysis.
Neurol 1997; 39: 708–725 J Neurol Neurosurg Psychiatry 2001; 70: 675–678
Krägeloh-Mann I, Grodd W, Niemann G, Haas G, Ruitenbeek W.
Assessment and therapy monitoring of Leigh disease by
Chronic Hypoxia–Ischemia MRI and proton spectroscopy.Pediatr Neurol 1992; 8:60–64
Krägeloh-Mann I, Grodd W, Schöning M, Marquard K, Nägele T,
Auer DP, Schirmer T, Heidenreich JO, Herzog J, Pütz B, Dichans Ruitenbeek W. Proton spectroscopy in five patients with
M. Altered white and gray matter metabolism in CADASIL. Leigh’s disease and mitochondrial enzyme deficiency. Dev
A proton MR spectroscopy and 1H-MRSI study. Neurology Med Child Neurol 1993; 35: 769–776
2001; 56: 635–642 Lin DDM, Crawford TO, Barker PB. Proton MR spectroscopy in
Brooks WM, Wesley MH, Kodituwakku PW, Garry PJ, Rosenberg the diagnostic evaluation of suspected mitochondrial dis-
GA. 1H-MRS Differentiates white matter hyperintensities in ease. AJNR Am J Neuroradiol 2003; 24: 33–41
subcortical arteriosclerotic encephalopathy from those in
normal elderly. Stroke 1997; 28: 1940–1943
1070 References and Further Reading

Lodi R, Montagna Pk Iotti S, Zaniol P, Barboni P, Puddu P, Barbi- Tracey I, Carr CA, Guimareas AR, Worth JL, Navia BA, González
oli B. Brain and muscle energy metabolism studied in vivo RG. Brain choline-containing compounds are elevated in
by 31P-magnetic resonance spectroscopy in NARP syn- HIV-positive patients before the onset of AIDS dementia
drome. J Neurol Neurosurg Psychiatry 1994; 57: 1492–1496 complex: a proton magnetic resonance spectroscopic
Matthews PM, Andermann F, Silver K, Karpati G, Arnold DL. Pro- study. Neurology 1996; 46: 783–788
ton MR spectroscopic characterization of differences in re-
gional brain metabolic abnormalities in mitochondrial en-
cephalomyopathies. Neurology 1993; 43: 2484–2490 Canavan Disease
Pavlakis SG, Kingsley PB, Kaplan GP, Stacpoole PW, O’Shea M,
Lustbader D. Magnetic resonance spectroscopy. Use in Austin SJ,Connelly A,Gadian DG,Benton JS,Brett EM.Localized
monitoring MELAS treatment. Arch Neurol 1998; 55: 1H NMR spectroscopy in Canavan’s disease: a report of two
849–852 cases. Magn Reson Med 1991; 19: 439–445
Rubio-Gozalbo ME, Heerschap A, Trijbels JMF, de Meirleir L, Barker PB, Bryan RN, Kumar AJ, Naidu S. Proton NMR spec-
Thijssen HOM, Smeitink JAM. Proton MR spectroscopy in a troscopy of Canavan’s disease. Neuropediatrics 1992; 23:
child with pyruvate dehydrogenase complex deficiency. 263–267
Magn Reson Imaging 1999; 17: 939–944 Marks HG, Caro PA, Wang Z, Detre JA, Bogdan AR, Gusnard DA,
Shevell MI,Matthews PM,Scriver CR,Brown RM,Otero LJ,Legris Zimmerman RA. Use of computed tomography, magnetic
ML, Brown GK, Arnold DL. Cerebral dysgenesis and lactate resonance imaging, and localized 1H magnetic resonance
acidemia: an MRI/MRS phenotype associated with pyru- spectroscopy in Canavan’s disease: a case report. Ann Neu-
vate dehydrogenase deficiency. Pediatr Neurol 1994; 11: rol 1991; 30: 106–110
224–229 Wittsack H-J,Kugel H,Roth B,Heindel W.Quantitative measure-
Takahashi S, Oki J, Miyamoto A, Okuno A. Proton magnetic res- ments with localized 1H MR spectroscopy in children with
onance spectroscopy to study the metabolic changes in Canavan’s disease. J Magn Reson Imaging 1996; 6: 889–893
the brain of a patient with Leigh syndrome. Brain Dev 1999;
21: 200–204
Wilichowski E, Pouwels PJW, Frahm J, Hanefeld F. Quantitative
NAA Synthesis Defect
proton magnetic resonance spectroscopy in patients with
MELAS. Neuropediatrics 1999; 30: 256–263
Zand DJ, Simon EM, Pulitzer SB, Wang DJ, Wang ZJ, Rorke LB, Barker PB. N-Acetyl aspartate – a neuronal marker? Ann Neurol
Palmieri M, Berry GT. In vivo pyruvate detected by MR spec- 2001; 49: 423–424
troscopy in neonatal pyruvate dehydrogenase deficiency. Martin E, Capone A, Schneider J, Hennig J,Thiel T.Absence of N-
AJNR Am J Neuroradiol 2003; 24: 1471–1474 acetylaspartate in the human brain: impact on neurospec-
troscopy? Ann Neurol 2001; 49: 518–521

Encephalitis
Creatine Deficiency Syndromes
Chang L, Ernst T, Tornatore C, Aronow H, Melchor R, Walot I,
Singer E, Conford M. Metabolite abnormalities in progres- Cecil KM, Salomons GS, Ball WS Jr,Wong B, Chuck G,Verhoeven
sive multifocal leukoencephalopathy by proton magnetic NM, Jakobs C, DeGrauw TJ. Irreversible brain creatine defi-
resonance spectroscopy. Neurology 1997; 48: 836–845 ciency with elevated serum and urine creatine: a creatine
Iranzo A, Moreno A, Pujol J, Marti-Fàbregas J, Domingo P. Molet transporter defect. Ann Neurol 2001; 49: 401–404
J, Ris J, Cadafalch J. Proton magnetic resonance spec- DeGrauw TJ, Salomons GS, Cecil KM, Chuck G, Newmeyer A,
troscopy pattern of progressive multifocal leukoence- Shapiro MB, Jakobs C. Congenital creatine transporter defi-
phalopathy in AIDS. J Neurol Neurosurg Psychiatry 1999; ciency. Neuropediatrics 2002; 33: 232–238
66: 520–523 Salomons GJ, van Dooren SJM, Verhoeven NM, Cecil KM, Ball
Laubenberger J, Häussinger D, Bayer S, Thielemann S, Schnei- WS, DeGrauw TJ, Jakobs C. X-linked creatine-transporter
der B, Mundinger A, Hennig J, Langer M. HIV-related meta- gene (SLC6A8) defect: a new creatine-deficiency syndrome.
bolic abnormalities in the brain: depiction with short echo Am J Hum Genet 2001; 68: 1497–1500
times. Radiology 1996; 199: 805–810 Schultze A. Creatine deficiency syndromes. Mol Cell Biochem
Meyerhoff DJ, MacKay S, Bachman L, Poole N, Dillon WP, Wein- 2003; 244: 143–150
er MW, Fein G. Reduced brain N-acetylaspartate suggests Stöckler S, Holzbach U, Hanefeld F, Marquart I, Helms G, Re-
neuronal loss in cognitive impaired neuronal loss in cogni- quart M, Hänicke W, Frahm J. Creatine deficiency in the
tively impaired human immunodeficiency virus-seroposi- brain: a new, treatable inborn error of metabolism. Pediatr
tive individuals. In vivo 1H magnetic resonance spectro- Res 1994; 36: 409–413
scopic imaging. Neurology 1993; 43: 509–515 Stöckler S, Isbrandt D, Hanefeld F, Schmidt B, von Figura. Guani-
Sämann PG, Schlegel J, Müller G, Prantl F, Emminger C, Auer DP. dinoacetate methyltransferase deficiency: the first inborn
Serial proton MR spectroscopy and diffusion imaging find- error of creatine metabolism in man. Am J Hum Genet
ings in HIV-related herpes simplex encephalitis. AJNR Am J 1996; 58: 914–922
Neuroradiol 2003; 24: 2015–2019 Stromberger C, Bodamer OA, Stöckler-Ipsiroglu S.Clinical char-
Takanashi J-I, Sugita K, Ishii M, Aoyagi M, Niimi H. Longitudinal acteristics and diagnostic clues in inborn errors of creatine
MR imaging and proton MR spectroscopy in herplex sim- metabolism. J Inherit Metab Dis 2003; 26: 299–308
plex encephalitis. J Neurol Sci 1997; 149: 99–102 van der Knaap MS, Verhoeven NM, Maaswinkel-Mooij P,
Pouwels PJW, Onkenhout W, Peeters EAJ, Stöckler-Ipsiroglu
S, Jakobs C. Mental retardation and behavioral problems as
presenting signs of a creatine synthesis defect. Ann Neurol
2000; 47: 450–543
References and Further Reading 1071

Nonketotic Hyperglycinemia Salla Disease

Gabis L, Parton P, Roche P, Lenn N, Tudorica A, Huang W. In vivo Sewell AC, Murphy HC, Iles RA. Proton magnetic resonance
1
H magnetic resonance spectroscopic measurement of spectroscopic detection of sialic acid storage disease. Clin
brain glycine levels in nonketotic hyperglycinemia. J Neu- Chem 2002; 48: 35–359
roimaging 2001; 11: 209–211 Varho T, Komhu M, Sonninen P, Holopainen I, Nyman S, Manner
Heindel W, Kugel H, Roth B. Noninvasive detection of increased T, Sillapää M, Aula P, Lundbom N. A new metabolite con-
glycine content by proton MR spectroscopy in the brains of tributing to N-acetyl signal in 1H MRS of the brain in Salla
two infants with nonketotic hyperglycinemia. AJNR Am J disease. Neurology 1999; 52: 1668–1672
Neuroradiol 1993; 14: 629–635
Huisman TAGM, Thiel T, Steinman B, Zeislinger G, Martin E. Pro-
ton magnetic resonance spectroscopy of the brain of a Maple Syrup Urine Disease
neonate with nonketotic hyperglycinemia: in vivo-in vitro
(ex vivo) correlation. Eur Radiol 2002; 12: 858–861
Felber SR, Sperl W, Chemelli A, Murr C, Wendel U. Maple syrup
Viola A, Chabrol B, Nicoli F, Confort-Gouny S, Viout P, Cozzone
urine disease: metabolic decompensation monitored by
PJ. Magnetic resonance spectroscopy study of glycine in
proton magnetic resonance imaging and spectroscopy.
nonketotic hyperglycinemia. Pediatr Res 2002; 52: 292–300
Ann Neurol 1993; 33: 396–401

Phenylketonuria Sjö
gren-Larsson Syndrome

Kreis R. Comments on in vivo proton magnetic resonance Mano T, Ono J, Kaminaga T, Imai K, Sakura K, Harada K, Nagai T,
spectroscopy in phenylketonuria. Eur J Pediatr 2000; 159: Rizzo WB, Okada S. Proton MR spectroscopy of Sjögren-
S126-S128 Larsson’s syndrome. AJNR Am J Neuroradiol 1999; 20:
Kreis R, Pietz J, Penzien J, Herschkowitz N, Boesch C. Identifica- 1671–1673
tion and quantitation of phenylalanine in the brain of pa- Van Domburg PHMF, Willemsen MAAP, Rotteveel JJ, de Jong
tients with phenylalanine by means of localized in vivo 1H JGN, Thijssen HOM Heerschap A, Cruysberg JRM, Wanders
magnetic-resonance spectroscopy. J Magn Reson B 1995; RJA, Gabreëls FJM, Steijlen PM. Sjögren-Larsson syndrome.
107: 242–251 Clinical and MRI/MRS findings in FALDH-deficient patients.
Leuzzi V, Bianchi MC, Tosetti M, Carducci Cl, Carducci Ca, Neurology 1999; 52: 1345–1352
Antonozzi I. Clinical significance of brain phenylalanine Willemsen MAAP, van der Graaf M, van der Knaap MS, Heer-
concentration assessed by in vivo proton magnetic reso- schap A, van Domburg PHMF, Gabreëls FJM, Thijssen HOM,
nance spectroscopy in phenylketonuria.J Inherit Metab Dis Rotteveel JJ. MR imaging and proton MR spectroscopy
2000; 23: 563–570 studies in Sjögren-Larsson syndrome: characterization of
Moats RA, Scadeng M, Nelson MD Jr. MR imaging and spec- the leukoencephalopathy. AJNR Am J Neuroradiol 2004;25:
troscopy in PKU. Ment Retard Dev Disab Res Rev 1999; 5: 649–657
132–135
Moats RA, Mosely KD, Koch R, Nelson M Jr. Brain phenylalanine
concentrations in phenylketonuria:research and treatment Galactosemia
of adults. Pediatrics 2003; 112: 1575–1579
Möller HE,Vermathen, Ullrich K,Weglage J, Koch H-G, Peters PE.
Berry GT, Hunter JV,Wang Z, Dreha S, Mazur A, Brooks DG, Ning
In-vivo NMR spectroscopy in patients with phenylke-
C, Zimmerman RA, Segal S. In vivo evidence of brain galac-
tonuria: changes of cerbral phenylalanine levels under di-
titol accumulation in an infant with galactosemia and en-
etary treatment. Neuropediatrics 1995; 26: 199–202
cephalopathy. J Pediatr 2001; 138: 260–262
Möller HE, Ullrich K, Weglage J. In vivo proton magnetic reso-
Möller HE, Ullrich K,Vermathen P, Schuierer G, Koch H-G. In vivo
nance spectroscopy in phenylketonuria. Eur J Pediatr 2000;
study of brain metabolism in galactosemia by 1H and 31P
159: S121-S125
magnetic resonance spectroscopy. Eur J Pediatr 1995; 154:
Möller HE, Weglage J, Bick U, Wiedermann D, Feldmann R, Ull-
S8-S13
rich K. Brain imaging and proton magnetic resonance spec-
Wang ZJ, Berry GT, Dreha SF, Zhao G, Segal S, Zimmerman RA.
troscopy in patients with phenylketonuria. Pediatrics 2003;
Proton magnetic resonance spectroscopy of brain metabo-
112: 1580–1583
lites in galactosemia. Ann Neurol 2001; 50: 266–269
Novotny EJ Jr, Avison MJ, Herschkowitz N, Petroff OAC, Prichard
JW, Seashore MR, Rothman DL. In vivo measurement of
phenylalanine in human brain by proton nuclear magnetic Defect in Polyol Metabolism
resonance spectroscopy. Pediatr Res 1995; 37: 244–249
Pietz J, Lutz T, Zwygart K, Hoffman GF. Phenylalanine can be
Huck JHJ, Verhoeven NM, Struys EA, Salomons GS, Jakobs C,
detected in brain tissue of healthy subjects by 1H magnet-
van der Knaap MS. Ribose 5-phosphate isomerase deficien-
ic resonance spectroscopy. J Inherit Metab Dis 2003; 26:
cy: new inborn error in the pentose phosphate pathway as-
683–691
sociated with a slowly progressive leukoencephalopathy.
Am J Hum Genet 2004; 74: 745–751
van der Knaap MS, Wevers RA, Struys EA, Verhoeven NM,
Pouwels PJW, Engelke UFH, Feikema W, Valk J, Jakobs C.
Leukoencephalopathy associated with a disturbance in the
metabolism of polyols. Ann Neurol 1999; 46: 925–928
1072 References and Further Reading

Diabetes Mellitus Ross BD, Ernst T, Kreis R, Haseler LJ, Bayer S, Danielsen E, Blüml
S, Shonk T, Mandigo JC, Caton W, Clark C, Jensen SW,
Kreis R, Ross BD. Cerebral metabolic disturbance in patients Lehman NL, Arcinue E, Pudenz R, Shelden CH. 1H MRS in
with subacute and chronic diabetes mellitus: detection acute traumatic brain injury.J Magn Reson Imaging 1998; 8:
with proton MR spectroscopy. Radiology 1992; 184: 829–840
123–130 Signoretti S, Marmarou A, Tavazzi B, Lazzarino G, Beaumont A,
Pan JW, Telang FW, Lee JH, de Graaf RA, Rothman DL, Stein DT, Vagnozzi R. N-Acetylaspartate reduction as a measure of
Hetherington HP. Measurement of b-hydorxybutyrate in injury severity and mitochondrial dysfunction following
acute hyperketonemia in human brain. J Neurochem 2001; diffuse traumatic brain injury. J Neurotrauma 2001; 18:
79: 539–544 977–991

X-Linked Adrenoleukodystrophy Multiple Sclerosis

Eichler FS, Itoh R, Barker PB, Mori S, Garrett ES, van Zijl PCM, Arnold DL, Wolinksky JS, Matthews PM, Falini A. The use of
Moser HW, Raymond GV, Melhem ER. Proton MR spec- magnetic resonance spectroscopy in the evaluation of the
troscopy and diffusion tensor brain MR imaging in X-linked natural history of multiple sclerosis. J Neurol Neurosurg
adrenoleukodystrophy: initial experience. Radiology 2002; Psychiatry 1998; 64: S94–S101
225: 245–252 Bitsch A, Bruhn H, Vougioukas V, Stringaris A, Lassmann H,
Eichler FS, Barker PB, Cox C, Edwin D, Ulug AM, Moser HW, Ray- Frahm J, Brück W. Inflammatory CNS demyelination:
mond GV.Proton MR spectroscopic imaging predicts lesion histopathologic correlation with in vivo quantitative pro-
progression on MRI in X-linked adrenoleukodystrophy. ton MR spectroscopy. AJNR Am J Neuroradiol 1999; 20:
Neurology 2002; 58: 901–907 1619–1627
Korenke GC, Pouwels PJW, Frahm J, Hunneman DH, Stoeckler S, Bjartmar C,Kidd G,Mörk S,Rudick R,Trapp BD.Neurological dis-
Krasemann E, Jost W, Hanefeld F. Arrested cerebral ability correlates with spinal cord axonal loss and reduced
adrenoleukodystrophy: a clinical and proton magnetic res- N-acetyl asparate in chronic multiple sclerosis patients.Ann
onance spectroscopy study in three patients. Pediatr Neu- Neurol 2000; 48: 893–901
rol 1996; 15: 103–107 Bonneville F, Moriarty DM, Li BSY, Babb JS, Grossmann RI, Go-
Pouwels PJW, Kruse B, Korenke GC, Mao X, Hanefeld FA, Frahm nen O.Whole-brain N-acetylaspartate concentration: corre-
J.Quantitative proton magnetic resonance spectroscopy of lation with T2-weighted lesion volume and expanding dis-
childhood adrenoleukodystrophy. Neuropediatrics 1998; 2: ability status scale score in cases of relapsing-remitting
254–264 multiple sclerosis. AJNR Am J Neuroradiol 2002; 23: 371–
Wilken B, Dechent P, Brockmann K, Finsterbusch J, Baumann M, 375
Ebell W, Korenke GC, Pouwels PJW, Hanefeld FA, Frahm J. Chard DT, Griffin CM, McLean MA, Kapeller P, Kapoor R,Thomp-
Quantitative proton magnetic resonance spectroscopy of son AJ, Miller DH. Brian metabolite changes in cortical gray
children with adrenoleukodystrophy before and after and normal-appearing white matter in clinically early re-
hematopoietic stem cell transplantation. Neuropediatric lapsing-remitting multiple sclerosis. Brain 2002; 125:
2003; 34: 237–246 2342–2352
Davie CA, Barker GJ, Thompson AJ, Tofts PS, McDonald WI,
Miller DH. 1H magnetic resonance spectroscopy of chronic
Diffuse Axonal Injury
cerebral white matter lesions and normal appearing white
matter in multiple sclerosis. J Neurol Neurosurg Psychiatry
Auld KL, Ashwal S, Holshouser BA, Tomasi LG, Perkin RM, Ross 1997; 63: 736–742
BD, Hinshaw BD Jr. Proton magnetic resonance spec- De Stefano N, Matthews PM, Antel JP, Preul M, Francis G, Arnold
troscopy in children with acute central nervous system in- DL. Chemical pathology of acute demyelinating lesions
jury. Pediatr Neurol 1995; 12: 323–334 and its correlation with disability. Ann Neurol 1995; 38:
Friedman SD, Brooks WM, Jung RE, Hart BL, Yeo RA. Proton MR 901–909
spectroscopic findings correspond to neuropsychological De Stefano N, Mattews PM, Fu L, Narayanan S, Stanley J, Francis
function in traumatic brain injury. AJNR Am J Neuroradiol GS, Antel JP, Arnold DL. Axonal damage correlates with dis-
1998; 19: 1879–1885 ability in patients with relapsing-remitting multiple sclero-
Friedman SD, Brooks WM, Jung RE, Chiulli SJ, Sloan JH, Montoya sis. Results of a longitudinal magnetic resonance spec-
BT, Hart BL, Yeo RA. Quantitative proton MRS predicts out- troscopy study. Brain 1998; 121; 1469–1477
come after traumatic brain injury. Neurology 1999; 52: De Stefano N, Narayanan S, Francis GS, Arnaoutelis R, Tartaglia
1384–1391 MC, Antel JP, Matthews PM, Arnold DL. Evidence of axonal
Garnett MR, Blamire AM, Corkill RG, Cadoux-Hudson TAD, Ra- damage in the early stages of multiple sclerosis and its rel-
jagopalan B, Styles P. Early proton magnetic resonance evance to disability. Arch Neurol 2001; 58: 65–70
spectroscopy in normal-appearing brain correlates with Gonen O, Moriarty DM, Li BSY, Babb JS, He J, Listerud J, Jacobs
outcome in patients following traumatic brain injury. Brain D, Markowitz CE, Grossman RI. Relapsing–remitting multi-
2000; 123: 2046–2054 ple sclerosis and whole-brain N-acetylaspartate measure-
Haseler LJ, Arcinue E, Danielsen ER, Bluml S, Ross BD. Evidence ment: evidence for different clinical cohorts – initial obser-
from proton magnetic resonance spectroscopy for a meta- vations. Radiology 2002; 225: 261–268
bolic cascade of neuronal damage in shaken baby syn- Husted CA, Goodin DS, Hugg JW, Maudsley AA, Tsuruda JS, de
drome. Pediatrics 1997; 99: 4–14 Bie SH, Fein G, Matson GB, Weiner MW. Biochemical alter-
ations in multiple sclerosis lesions and normal-appearing
white matter detected by in vivo 31P and 1H spectroscopic
imaging. Ann Neurol 1994; 36: 157–165
References and Further Reading 1073

Kapeller P, McLean MA, Griffin CM, Chard D, Parker GJM, Barker Blaser SI, Clarke JTR, Becker LE. Neuroradiology of lysosomal
GJ, Thompson AJ, Miller DH. Preliminary evidence for neu- disorders. Neuroimaging Clin N Am 1994; 4: 283–298
ronal damage in cortical grey matter and normal appear- Blois MS. Clinical judgment and computers. N Engl J Med 1980;
ing white matter in short duration relapsing-remitting mul- 303: 192–197
tiple sclerosis: a quantitative MR spectroscopy imaging Boltshauser E, Martin E, Wichmann W, Valavanis A. Grenzen
study. J Neurol 2001; 248: 131–138 der kernspintomographie aus neuropädiatrischer Sicht.
Leary SM, Davie CA, Parker GJM, Stevenson WL, Wang L, Barker Klin Neuroradiol 1993; 3: 79–82
GJ, Miller DH, Thompson AJ. 1H Magnetic resonance spec- Brismar J. CT and MRI of the brain in inherited metabolic disor-
troscopy of normal appearing white matter in primary pro- ders. J Child Neurol 1992; 7: S122–S131
gressive multiple sclerosis. J Neurol 1999; 246: 1023–1026 Cassedy KJ, Edwards MK. Metabolic and degenerative diseases
Mader I, Roser W, Kappos L, Hagberg G, Seeling J, Radue EW, of childhood.Top Magn Reson Imaging 1993; 5: 73–95
Steinbrich W. Serial proton MR spectroscopy of contrast- Cheon JE, Kim IO, Hwang YS, Kim KJ, Wang KC, Cho BK, Chi JG,
enhancing multiple sclerosis plaques: absolute metabolic Kim CJ, Kim WS,Yeon KM. Leukodystrophy in children: a pic-
values over 2 years during clinical pharmacological study. torial review of MR imaging features. Radiographics 2002;
AJNR Am J Neuroradiol 2000; 21: 1220–1227 22: 461–476
Mader I, Seeger U, Weissert R, Klose U, Naegele T, Melms A, Edwards MK. Imaging of metabolic diseases of the brain. Curr
Grodd W. Proton MR spectroscopy with metabolite-nulling Opin Radiol 1991; 3: 25–30
reveals elevated macromolecules in acute multiple sclero- Ford CC, Ceckler TL, Karp J, Herndon RM. Magnetic resonance
sis. Brain 2001; 124: 953–961 imaging of experimental demyelination lesions. Magn
Narayana PA,Doyle TJ,Lai D,Wolinsky JS.Serial proton magnet- Reson Med 1990; 14: 461–481
ic resonance spectroscopic imaging, contrast-enhanced Forsting M.MR imaging of the brain: metabolic and toxic white
magnetic resonance imaging, and quantitative lesion vol- matter diseases. Eur Radiol 1999; 9: 1061–1065
umetry in multiple sclerosis. Ann Neurol 1998; 43: 56–71 Getty DJ, Pickett RM, D’Orsi CJ, Swets JA. Enhanced interpreta-
Sarchielli P, Presciutti O, Pellicioli GP,Tarducci R, Gobbi G, Chiari- tion of diagnostic images. Invest Radiol 1988; 23: 240–252
ni P, Alberti A, Vicinanza F, Gallai V. Absolute quantification Kendall BE. Disorders of lysosomes, perosixomes and mito-
of brain metabolites by proton magnetic resonance spec- chondria. AJNR Am J Neuroradiol 1992; 13: 621–653
troscopy in normal appearing white matter of multiple Kendall BE.Inborn errors and demyelination: MRI and the diag-
sclerosis patients. Brain 1999; 122: 513–521 nosis of white matter disease. J Inherit Metab Dis 1993;
Sarchielli P, Presciutti O, Tarducci R, Gobbi G, Alberti A, Pellicci- 16:771–786
oli GP, Chiarini P, Gallai V. Localized 1H magnetic resonance Kristjánsdóttir R, Uvebrant P, Hagberg B, Kyllerman M, Wiklund
spectroscopy in mainly cortical gray matter of patients L-M, Blennow G, Flodmark O, Gustavsson L, Ekholm S, Måns-
with multiple sclerosis. J Neurol 2002; 249: 902–910 son J-E. Disorders of the cerebral white matter in children.
Suhy J, Rooney WD, Goodkin DE, Capizzano AA, Soher BJ, The spectrum of the lesions. Neuropediatrics 1996; 27:
Maudsley AA,Waubant E, Andersson PB,Weiner MW. 1H MR- 295–298
SI Comparison of white matter and lesions in primary pro- Miller DH, Robb SA, Ormerod IEC, Pohl KRE, MacManus DG,
gressive and relapsing-remitting MS. Mult Scler 2000; 6: Kendall BE, Moseley IF, McDonald WI. Magnetic resonance
148–155 imaging of inflammatory and demyelinating white matter
Tartaglia MS, Narayanan S, de Stefano N, Arnaoutelis R, Antel diseases of childhood. Dev Med Child Neurol 1990; 32:
SB, Francis SJ, Santos AC, Lapierre Y, Arnold DL. Choline is in- 97–107
creased in pre-lesional normal appearing white matter in Sackett DL, Haynes RB,Tugwell P. Clinical epidemiology: a basic
multiple sclerosis. J Neurol 2002; 249: 1382–1390 science for clinical medicine. Boston: Little, Brown & Co,
Tourbah A, Stievenart JL, Gout O, Fontaine B, Liblau R, Lubetzki 1985
C, Cabanis EA, Lyon-Cean O. Localized proton magnetic res- Valk J, van der Knaap MS. Selective vulnerability in toxic en-
onance spectroscopy in relapsing remitting versus sec- cephalopathies and metabolic disorders. Riv Neuroradiol
ondary progressive multiple sclerosis. Neurology 1999; 53: 1996; 9: 749–760
1091–1097 Valk J, van der Knaap MS. Patterns of myelin breakdown. Eur
Van Walderveen MAA, Barkhof F, Pouwels PJW, van Schijndel Radiol 1999; 9: S3–S14
RA, Polman CH, Castelijns JA. Neuronal damage in T1-hy- van der Knaap MS. Magnetic resonance in childhood white
pointense multiple sclerosis lesions demonstrated in vivo matter disorders. Dev Med Child Neurol 2001; 43: 715–712
using proton magnetic resonance spectroscopy. Ann Neu- van der Knaap MS, Valk J. Non-leukodystrophic white matter
rol 1999; 46: 79–87 changes in inherited disorders. Int J Neuroradiol 1995; 1:
56–66
van der Knaap MS, Valk J, de Neeling N, Nauta JJP. Pattern
recognition in magnetic resonance imaging. Neuroradio-
109 Pattern Recognition logy 1991; 33: 478–493
in White Matter Disorders van der Knaap MS, Breiter SN, Naidu S, Hart AAM, Valk J. Defin-
ing and categorizing leukoencephalopathies of unknown
Barkhof F, van der Knaap MS. Differential diagnosis with origin: MR imaging approach. Radiology 1999; 213: 121–
leukodystrophies and non-vascular acquired white matter 133
disorders. In: Erkinjuntti T, Gauthier S, eds. Vascular cogni-
tive impairment. London: Martin Dunitz, 2002, pp 485–500
Subject Index

A – glycogen storage disease Binswanger disease 767–772


acquired immunodeficiency syndrome type IV 148 biotin metabolism, defects 248
616–627 – magnetic resonance imaging biotinidase deficiency 248–251
– acute HIV-1 encephalopathy 616 149–151 Bloch-Sulzberger syndrome 412–415
– AIDS in infants and children 617 – polyglucosan bodies 147 Borrelia burgdorferi 792
– AIDS-related dementia 616 – – intra-axonal and astrocytic brain maturation
– cellular immunodeficiency 616 location 147 – ADC values 847
– diagnostic criteria Centers – sural nerve biopsy 147 – DWI-DTI changes 847
of Disease Control 616 adult-onset autosomal dominant – MTR values 42, 857
– diagnostic procedures 617 leukoencephalopathies 560–561 brucellosis 635–639
– high risk groups 616 – clinical variants 560 – Brucella species 635
– highly aggressive antiretroviral Aicardi-Goutières syndrome 496–504 – clinical variability 635
therapy (HAART) 622 – atypical manifestations 496, 497 – coccobacilli 635
– HIV-1 virions in brain tissue 617 – calcium deposits 497, 498, 501 – geographic distribution 635
– human immunodeficiency – clinical variants 496 – magnetic resonance imaging
virus type 1 (HIV-1) 616 – Cree encephalitis 497 636–639
– lentovirinae subfamily – CSF lymphocytosis 497 – therapeutic options 636
of retroviruses 620 – interferonopathy 498 – zoonosis 635
– magnetic resonance imaging – interferon-a 497
606–627 – magnetic resonance C
– major envelope proteins 620 imaging 498–504 CADASIL 541–548
– mode of infection 620 – pseudo-TORCH syndrome 497 Canavan disease 326–333
– MS-like encephalopathy 619 Alexander disease 416–435 – aspartoacylase 327
– opportunistic infections 616 – aB-crystallin 416 – clinical variants 326
– pneumocystis carinii – clinical variants 416 – genetic defect 327
pneumonia 616 – genetic defect 418 – magnetic resonance imaging
– spinal cord involvement 619 – glial fibrillary astrocyte protein 418 328–333
– subacute HIV-1 encephalo- – magnetic resonance – N-acetyl aspartate 327
pathy 616 imaging 419–435 – spongy degeneration 326
– therapeutic options 621–623 – megalencephaly 416 – vacuolating myelinopathy 326, 327
activator protein deficiency 74, 98, – Rosenthal fibers 416, 417, 418 – Van Bogaert-Bertrand disease 326
104 amyloid angiopathy 535–540 CARASIL 549–551
acute disseminated encephalo- angiokeratomas (Fabry disease) 112 carbon monoxide intoxication 755
myelitis and acute hemorrhagic anti-phospholipid antibody carboxyl phosphate synthetase
encephalomyelitis 604–615 syndrome 785 deficiency 360, 362
– auto-immune reaction 605 apparent diffusion coefficient cataracts, microcephaly, failure to
– clinical variants 604 (ADC) 840 thrive, kyphoscoliosis syndrome
– Guillain-Barré syndrome 604 arginase deficiency 360, 362, 367 (CAMFAK) 260
– magnetic resonance imaging argininosuccinate central cortical and subcortical
606–615 – lyase deficiency 360, 362, 364 pattern 718, 733, 737–740
– postvaccinal encephalopathy 604 – synthase deficiency 360, 362 central pontine and extrapontine
– therapeutic options 605 astrocyte 5 myelinolysis 684–689
– viral infections 604 ATP synthase deficiency 224, 225, 227 – alcohol abuse 685
acute intermittent porphyria 700 Ayala syndrome 709 – hyponatremia 685
acute profound hypoxic-ischemic – iatrogenic factor 685
insults 718 B – magnetic resonance imaging
adult polyglucosan body Baló concentric sclerosis 567, 600 686–689
disease 147–151 basal ganglia damage after perinatal – myelinolysis 684–685
– corpora amylacea 148 asphysica 718, 738, 739 – prevention 686
– cystic lesions 147 Batten disease 137 – rapid correction of hyponatremia
– genetic defect 148 Behçet disease 789, 791, 792 685
– glucose polymers 148 benign primary angiitis 776 – therapeutic options 686
– glycogen branching enzyme 148 big panda sign 397
1076 Subject Index

cerebral amyloid angiopathy Charcot-Marie-Tooth disease, – dystrophin-associated glycoprotein


535–540 X-Linked 476–478 complex 454
– amyloid – clinical variants 476 – Fukuyama congenital muscular
– – cascade 537 – connexin 32 476 dystrophy 451
– – precursor proteins 536 – genetic defect 476 – genetic defects 454, 455
– – Bri fragment 537 – magnetic resonance imaging 477, – laminin-a2 453–455
– Ab protein subunits 536 478 – limb girdle muscular dystrophy 21
– British variant (Worster-Drought chemotherapy-related encephalo- 452
syndrome) 535 pathy 808–817 – magnetic resonance imaging
– cystatin C 536 childhood ataxia with CNS 457–468
– Dutch variant 535 hypomyelination 481–494 – MDC1C 451
– Flemish variant 535 cholestanol 253 – MDC1D 451
– genetic defects 536, 537 choline phosphoglycerides 7 – merosin-deficient congenital
– Iceland variant 535 Churg-Strauss syndrome 789 muscular dystrophy (MDC1A) 451
– lobar hemorrhage 535 classification – muscle-eye-brain disease 451
– magnetic resonance imaging – of lysosomal storage disorders 69 – therapeutic options 457
537–540 – of mitochondrial disorders 200 – Walker-Warburg syndrome 451
– oculoleptomeningeal – of mucopolysaccharidoses 123 cree leukoencephalopathy 481
amyloidosis 535 – of neuronal ceroid lipofuscinoses CREST 786
– b-sheet content 536 (eponyms) 137 Curschmann-Steinert disease
– sporadic forms 535 – of peroxisomal disorders 153 469–472
– therapeutic options 537 – of toxic encephalopathies 664–678 cyanide intoxication 755
– transthyretin 536 – of white matter disorders 20–24 cystathionine b-synthase deficiency
cerebral autosomal dominant arteri- cobalamin 342, 347
opathy with subcortical infarcts – deficiency 343, 346, 354, 356 cytochrome c oxidase deficiency 225,
and leukoencephalopathy – defects in metabolism 343, 347 227, 238
(CADASIL) 541–548 Cockayne syndrome 259–267 cytomegalovirus infection
– absence of vascular risk factors 541 – cachectic dwarfism 259 – congenital or perinatal 645–657
– diagnostic criteria 541 – CAMFAK syndrome 260 – in AIDS 624
– epidermal growth factor motif 542 – clinical variants 259
– familial Binswanger disease 541 – COFS syndrome 260 D
– genetic defect 542 – complementation groups 261 D-2-hydroxyglutaric aciduria
– granular osmiophilic material 541 – DNA-repair 261–262 338–341
– hereditary multi-infarct dementia – genetic defects 261 – clinical variants 338
541 – magnetic resonance imaging – D-2-hydroxyglutarate dehydro-
– magnetic resonance imaging 263–267 genase 338
542–548 – therapeutic options 262 – genetic defect 338
– migrainous headache 541 – trichothiodystrophy 261 – magnetic resonance imaging
– therapeutic options 542 – UV hypersensitivity 261 339–341
cerebral autosomal recessive arterio- complex I deficiency 224, 226, 229 – secondary respiratory chain
pathy with subcortical infarcts complex IV deficiency 224, 226, 232 deficiency 338
and leukoencephalopathy conduction velocity 15 delayed posthypoxic leuko-
(CARASIL) 549–551 congenital and perinatal cytomegalo- encephalopathy 755–758
– alopecia 549 virus infection 645–657 – carbon monoxide 755
– clinical variants 549 – ganciclovir 647 – cyanide 755
– lumbago 549 – herpes viridae family 645 – magnetic resonance imaging
– magnetic resonance – magnetic resonance imaging 756–758
imaging 549–551 650–657 – therapeutic options 756
cerebro-oculo-facio-skeletal syndrome – maternal infection 645 demyelination 15, 17
(COFS) 260 – mental deficiency 646 – biochemical changes 17
cerebrotendinous xanthomatosis – modes of infection 645 – loss of function 18
252–259 – retrospective diagnosis 646 dentatorubropallidoluysian
– Achilles tendon 252 – sensorineural hearing loss 646 atrophy 530–534
– bile acid metabolism 252, 253, 254 – therapeutic options 648, 649 – anticipation 531
– cholestanol 253 – TORCH infections 646 – atrophin-1 530
– clinical variants 252 – viral gene products 648 – CAG trinucleotide repeat 530
– genetic defect 253 congenital muscular dystrophy – clinical variants 530
– 27-hydroxylase 253 451–468 – genetic defect 530
– magnetic resonance imaging – a-dystroglycan 453 – magnetic resonance imaging
253–258 – – hypoglycosylation 454, 455 531–534
– xanthomas 253 – polyglutamine repeat 531
Subject Index 1077

diabetes insipidus 709 fatty acid b-oxidation, giant axonal neuropathy 436–441
diffuse axonal injury 823–831 peroxisomal 152 – aberrant neurofilaments 437
– acceleration-deceleration folate – genetic defect 437
trauma 823 – defects in metabolism 347 – giant axonal swellings 437
– axonal damage 824 – deficiency 343, 344 – gigaxonin 437
– Genneralli classification 824, 831 fragile X premutation 406–408 – hair abnormalities 436
– magnetic resonance imaging – clinical variants 406 – magnetic resonance imaging
824–831 – genetic defect 406 437–441
– outcome 824 – magnetic resonance imaging – Rosenthal fibers 436
– therapeutic options 824 406–408 giant cell arteriitis 775
diffusion sensitivity 840 – mental retardation 406 gliomatosis cerebri 818–822
diffusion weighted imaging 839–855 – trinucleotide expansion 406 – clinical variants 818
– apparent diffusion coefficient 840 free sialic acid storage disorder – delayed gadolinium enhance-
– Brownian movement 839 133–136 ment 821
– developmental changes 847 – bone abnormalities 134 – genetic patterns 818
– diffusion sensitivity 840 – clinical types 133 – magnetic resonance imaging
– fractional anisotropy 843 – free sialic acid 818–822
– hypoxia-ischemia in neonates 733, – – excretion in urine 133 – therapeutic options 820
736, 738, 745, 747, 847, 848 – – transporter 134 globoid cell leukodystrophy
– intravoxel coherent and incoherent – genetic defect 134 (Krabbe disease) 87–95
motion 840 – magnetic resonance imaging – cerebrosides 89
– relative anisotropy 843 135–136 – clinical variants 87
– Stejskal-Tanner sequence 840 – Salla disease 133 – galactocerebroside b-galactosi-
– trace diffusion-weighted images – vacuolar inclusions in sural nerve dase 88
843 biopsy 134 – genetic defect 89
dihydropteridine reductase deficiency fucosidosis 119–122 – magnetic resonance imaging
286, 291, 292, 293 – clinical variants 119 91–95
DNA – fucose 119 – psychosine 89
– content of brain 13 – genetic defect 120 – therapeutic options 90
– repair disorders 259–267, 268–272 – internal organs enlargement 119 – white matter involvement 89
drug-induced vasculitis 797 – a-L-fucosidase 117 glutaric aciduria type 1 294–300
dysmyelination 15, 17 – magnetic resonance imaging – clinical variants 294, 295
120–122 – genetic defect 295
E – neuronal ballooning 119 – glutaryl-CoA-dehydrogenase 295
eclampsia 699 – therapeutic options 117 – magnetic resonance imaging
elderly with mild cognitive Fukuyama congenital muscular 296–299
impairment 759 dystrophy 451, 454, 459, 461 – myelin splitting 295, 296
eosinophilic granuloma 709 – therapeutic options 295, 296
Erdheim-Chester disease 709, 710, G glycosaminoglucans in urine 126
711, 713 Gagel granuloma 709 GM1 gangliosidosis 96–102
etat galactosemia 377–382 – activator protein 97
– criblé 767 – clinical variants 377 – bone deformities 96
– lacunaire 767 – Duarte variants 378 – clinical variants 96
ethanolamine phosphoglycerides 6 – epimerase 379 – b-galactosidase 98
extrapontine myelinolysis 684–689 – galacticol 378 – gangliosides 97
– galactokinase 377 – genetic defect 98
F – galactosed-1-phosphate uridyltrans- – magnetic resonance imaging
Fabry disease 112–118 ferase 377 100–102
– atypical variants 112 – genetic defects 378, 379 – membranous cytoplasmic
– early and late manifestations 112 – incidence 377 bodies 97
– a-galactosidase 113 – magnetic resonance imaging GM2 gangliosidosis 103–111
– – recombinant 113 380–382 – bone deformities 104
– genetic defect 114 – therapeutic options 379 – clinical variants 103
– glycosphingolipids accumula- – uridine diphosphate – gangliosides 106
tion 114 – – galactose 379 – genetic defect 106
– heterozygous female patients 112 – – galactose-4-epimerase 377 – glycolipid biosynthesis 107
– magnetic resonance imaging gangliosides 7, 81, 95, 102, 104 – hexosaminidase A and B 104
115–118 Genneralli classification 824, 831 – magnetic resonance imaging
– pain 112 germinal layer related 108–111
– therapeutic options 114 hemorrhage 720 – motor neuron disease 104
– vascular changes 113 granulomatous angiitis 774, 775, 776
1078 Subject Index

gyration 45 – regulation of fluid balance 691 – major histocompatibility complex


– iconography: preterm-term – toxicity 690 (MHC) 561
neonates 45–50 hypertensive encephalopathy 699 – molecular and soluble components
hypomelanosis of Ito 409–411 561
H – Blaschklo lines 409 – myelin involvement in inflammatory
Hachinski scale 760 – chromosomal or genetic mosaicism and infectious disorders 565
Hand-Schüller-Christian disease 709 409 – natural killer cells 562
hemolytic-uremic syndrome 700 – clinical phenotypes 409 – oxygen radicals 562
hepatolenticular syndrome 392–399 – enlarged perivascular spaces 410 – T-cell lymphocytes 561
hereditary diffuse leukoencephalo- – incontinentia pigmenti achromians intraperiod line 3
pathy with neuroaxonal 409 intravascular lymphomatosis 798
spheroids 526–529 – magnetic resonance imaging 410,
– axonal spheroids 526 411 J
– clinical variants 526 hypomyelination with atrophy of the Jansky Bielschowski disease 137
– dermatoleukodystrophy with basal ganglia and cerebellum
neuroaxonal spheroids 526 519–525 K
– magnetic resonance imaging – clinical variants 519 Kearns-Sayre syndrome 215–220
527–529 – magnetic resonance imaging – endocrine dysfunction 215
– polycystic lipomembranous 519–525 – genetic defects 216
osteodysplasia with sclerosing hyponatremia 685 – magnetic resonance imaging
leukoencephalopathy (Nasu-Hakola hypoxia-ischemia in neonates, DWI 217–220
disease) 526, 552–556 733, 736, 738, 745, 747, 847, 848 – overlapping syndromes 216
herpes zoster ophthalmica 797 – Pearson syndrome 215
histiocytosis X 709 I – red ragged fibers 215
holocarboxylase synthase iatrogenic toxic encephalopathies – therapeutic options 216
deficiency 248–251 679–683 kinky hair disease 400
Hunter syndrome 83, 124, 130 – host-versus-graft disease 679 Krabbe disease 87–95
Hurler-Scheie syndrome 123, 124, 130 – magnetic resonance imaging Kufs disease 137
Hurler syndrome 123, 124 680–683
3-hydroxy-3-methylglutaryl-CoA lyase – multifocal inflammatory leukoen- L
deficiency 321–324 cephalopathy 679 L-2-hydroxyglutaric aciduria 334–337
– clinical variants 321 – posterior reversible encephalopathy – clinical variants 334
– genetic defect 321 syndrome 681 – genetic defect 334
– HMG-CoA lyase 321 incontinentia pigmenti 412–415 – L-2-hydroxyglutarate dehydroge-
– hypoketotic hypoglycemia 322 – Bloch-Sulzberger syndrome 412 nase 334
– ketone bodies 321 – genetic defect 412 – magnetic resonance imaging
– magnetic resonance imaging – magnetic resonance imaging 334–337
321–325 413–415 Langerhans cell histiocytosis
– therapeutic options 322 – NF-k-B activation 412 709–714
hyperhomocysteinemia 342–359 – proapoptotic signals 412 – diabetes insipidus 709
– clinical variants 343, 344, 345 – skin lesions 412 – eosinophilic granuloma 709
– cobalamin metabolism 342 – type II 412–415 – Erdheim-Chester disease 709, 710
– cystathione b-synthase 342 infantile Refsum syndrome 154–166 – Hand-Schüller-Christian disease
– folate metabolism 342 inflammatory and infectious disorders 709
– genetic defects 347, 348 561–565 – histiocytosis X 709
– homocysteine metabolism 342 – acquired adaptive immune system – Letterer Siwe disease 709
– magnetic resonance imaging 561 – magnetic resonance imaging
348–359 – acute phase proteins 561 710–714
– methyl-folate trap 345 – B-cell lymphocytes 561 Leber hereditary optic atrophy
– methylmalonic acidemia 343 – complement 561 212–214
– mild form 342 – cytokines 563 – genetic defects 213
– pyridoxine (vitamin B6) 348 – exotoxins 563 – magnetic resonance imaging
– subacute combined degeneration – human leukocyte antigen (HLA) 213–214
of the cord 345 581 – multiple sclerosis association 212
– therapeutic options 348 – immune system 561 – Uhthoff symptom 212
– transsulfuration pathway 342 – inflammatory mediators 561 – visual dysfunction 212
hypernatremia 690–694 – inflammatory process 561 Leigh syndrome and mitochondrial
– magnetic resonance imaging – innate non-adaptive immune system leukoencephalopathies 224–244
691–694 561 – ATP synthase deficiency 224, 227
– non-accidental intake of sodium – interferons 561 – causes 224, 226
690 – Langerhans and dendritic cells 563 – clinical variants 224
Subject Index 1079

– complex I deficiency 224, 226 – ovarioleukodystrophy 481 – Larmor


– cytochrome c oxidase deficiency – regulation of protein synthesis 482 – – frequency 859
224, 227 – stress conditions 482 – – precession 859
– magnetic resonance imaging – sububits of eIF2B 482 – myo-inositol 864
228–244 – therapeutic options 482 – N-acetylaspartate 863
– mitochondrial leukoen- – translation-initiation factor eIF2B – normal values 864
cephalopathies 224–244 482 – parts per million (ppm) 860
– pyruvate dehydrogenese complex Lhermitte symptom 566 – phosphocreatine 861
deficiency 224, 226 lipofuscine 137 – phosphodiesters 861
– therapeutic options 227 Loes score 186 – phosphomonoesters 861
Letterer Siwe disease 709 Lorentzo oil 181 – 31
P-MRS
leukoaraiosis 759 Lowe syndrome 387–391 – – peak assignment 861
leukodystrophy 15 – genetic defect 388 – – pH estimation 865
leukoencephalopathy 15 – magnetic resonance imaging – process-specific abnormalities
leukoencephalopathy with brain stem 388–391 873–880
and spinal cord involvement – oculocerebrorenal syndrome 387 – time domain signal 860
and elevated white matter – phosphoinositides 388 magnetization transfer imaging
lactate 510–518 – skeletal abnormalities 387 854–858
– clinical variants 510 – therapeutic options 388 – developmental changes 42, 857
– magnetic resonance imaging – type II phosphatases 388 – magnetization transfer ratio (MTR)
510–518 Lyme disease 792, 793, 794 855
leukoencephalopathy with calcifica- lysosomal storage disorders 68–73 – – histogram analysis 855
tions and cysts 505–509 – classification 69 – – axonal density 856
– angiomatous changes 505 – defects major dense line 3
– calcifications 505, 507, 509 – – in activator proteins 71 maple syrup urine disease 311–320
– clinical variants 505 – – in postsynthetic modification of – branched-chain
– magnetic resonance imaging lysosomal proteins 71 – – a-keto acid decarboxylase (E1)
505–509 – – in protective protein 72 312
– Rosenthal fibers 505 – – in structural lysosomal – – a-keto acid dehydrogenase
leukoencephalopathy and dural proteins 71 complex 312
arteriovenous fistulas 801–808 – – in transport systems 72 – – amino acids 311
– basic fibroblast growth factor 801 – lysosomal lipid metabolism 70 – clinical variants 311
– classification of cranial AV lysosomes 66 – dihydrolipoyl
fistulas 802 – biogenesis 66 – – acyltransferase (E2) 312
– magnetic resonance imaging – defects in individual hydrolases 68 – – dehydrogenase (E3) 312
803–807 – definition 66 – genetic defects 312
– therapeutic options 802 – disorders 66–73 – magnetic resonance imaging
– vascular endothelial growth – functions 66 312–320
factor 801 – pathochemistry 68 – therapeutic options 313
leukoencephalopathy after radiothera- Marchiafava-Bignami disease
py and chemotherapy 808–817 M 695–699
– types of damage 808 magnetic resonance spectroscopy – alcohol 695
– multifocal inflammatory leuko- 859–881 – clinical variants 695
encephalopathy 808 – ATP 861 – corpus callosum splitting 696
– posterior reversible encephalopathy – basic principles 859 – magnetic resonance imaging
syndrome 808 – chemical shift imaging (CSI) 859, 697–698
– Magnetic Resonance Imaging 861 – toxic factors 695
810–817 – choline 863 Maroteaux-Lamy syndrome 83, 125,
leukoencephalopathy with vanishing – creatine 863 132
white matter 481–494 – disease-specific abnormalities measles 640
– childhood ataxia with CNS 865–873 megalencephalic leukoencephalopathy
hypomyelination (CACH) 481 – editing techniques 860 with subcortical cysts 442–450
– clinical variants 487–494 – Fourier transformation 860 – ethnic distribution 442
– Cree leukoencephalopathy 481 – frequency domain signal 860 – genetic defect 443
– guanosine diphosphate- – GABA 864 – macrocephaly 442
guanosine triphosphate – glutamate 864 – magnetic resonance imaging
conversion 482 – 1H-MRS peak assignment 862 443–450
– guanosine exchange factor activity – inorganic phosphate 861 – MLC1 protein 443
482 – J-coupling 860 – phenotypic variation 443
– magnetic resonance imaging – lactate 863 – vacuolating myelinopathy 443
483–494
1080 Subject Index

Menkes disease 400–405 mitochondrial encephalopathy with multiple sclerosis 566–613


– ceruloplasmin levels 401 lactic acidosis and stroke-like – Baló concentric sclerosis 567
– clinical variants 400 episodes (MELAS) 204–211 – Barkhof criteria 568
– copper – calcium deposits 205 – benign MS 566
– – -dependent enzyme processes – clinical variants 204 – CD 4+ and CD 8+ T cell lymphocytes
402 – genetic defects 205 572
– – -transporting P-type – magnetic resonance imaging – Charcot type 566
ATPase 402 206–211 – clinical variants 566
– – levels 401 – MELAS-MERFF overlap 204 – clinically isolated symptom (CIS)
– cranial exostoses 400 – red ragged fibers 204 566
– genetic defect 402 – therapeutic options 205 – cluster of differentiation markers
– kinky hair disease 400 mitochondrial leukoencephalopathies (CD) 569
– magnetic resonance imaging 224–244 – diagnostic criteria 567
403–405 mitochondrial neurogastrointestinal – environmental factors 574
– occipital horn syndrome 400 encephalomyopathy 221–223 – experimental allergic en-
– skeletal abnormalities 400 – genetic defect 222 cephalomyelitis (EAE) 572
– therapeutic options 403 – magnetic resonance imaging – FA and ADC 849, 592, 593
– tortuosity of cerebral vessels 403 222–223 – gadolinium enhancement 569
– trichopoliodystrophy 400 – synonyms 221 – genetic factors 573
– type IX Ehlers-Danlos syndrome molybdenum cofactor deficiency – incidence 566
400 372–376 – Lhermitte symptom 566
– X-linked cutis laxa 400 – isolated sulfite oxidase deficiency – magnetic resonance imaging
merosin-deficient congenital muscular 372–376 577–613
dystrophy 451, 455, 466 – clinical variants 372 – Marburg type 566
metachromatic leukodystrophy – sulphite oxidase 372 – McDonald criteria 568
74–81 – genetic defects 372, 373 – neuromyelitis optica 567
– arylsulfatase A 75 – molybdenum cofactor synthesis 372 – oligoclonal banding 568
– clinical variants 74 – therapeutic options 373 – perivenular inflammation 569
– genetic defect 76 – magnetic resonance imaging – primary progressive MS 566
– magnetic resonance imaging 373–376 – relapsing-remitting MS 566
78–81 Morquio syndrome 83, 125, 132 – remyelination 570
– metachromasia 75 Moyamoya syndrome 777, 778, 779, – Schilder diffuse sclerosis 567
– pseudodeficiency of arylsulfatase A 780 – secondary progressive MS 566
75 mucopolysaccharidoses 123–132 – spinal cord involvement 579,
– sulfatide 75 – classification 123 587–589
– therapeutic options 78 – degradation of mucoplysaccharides – therapeutic options 575, 576
5,10-methylenetetrahydrofolate 126 – tumefactive MS 566
– reductase deficiency 342, 347, 351 – enzyme deficiencies (table) 123 – viral factors 573
– thermolabile variant 342, 347 – enzymes involved 123 multiple sulfatase deficiency 82–86
microscopic polyarteritis 789 – genetic defects 127 – clinical variants 82
mitochondria 195–204 – glycosaminoglycans 127 – genetic defect 83
– citric acid cycle 197 – Magnetic Resonance Imaging – magnetic resonance imaging
– mitochondrial DNA 198 129–132 83–86
– nuclear DNA 198 – mental deficiency 126 – mucopolysaccharidoses 82
– oxidative phosphorylation 197 – proteoglycans 127 – peripheral nerve involvement 83
– respiratory chain 197 – therapeutic options 127 – sulfatases involved 83
– structure and function 195 multicystic encephalopathy 720, 742, – zebra bodies 82
mitochondrial disorders 195–204 743 muscle-eye-brain disease 451, 454,
– classification 200 multifocal inflammatory leuken- 462
– maternal inheritance 199 cephalopathy 679, 680, 808 myelin
– mitochondrial DNA defects multiple carboxylase deficiency – aging 14
200–201 248–251 – basic protein 7, 281
– – deletions, large single 201 – biotinidase deficiency 248 – biochemical composition 6
– – duplications 201 – clinical variants 248 – compositional changes during
– – point mutations 201 – genetic defect 249 development 13
– genetics 198 – holocarboxylase synthase deficiency – function 14
– leukoencephalopathies 224–244 248 – introduction 1
– nuclear DNA defects 201–202 – magnetic resonance imaging 250, – glycoproteins 7
– – types of genes 201 251 – levels of involvement 16
– – assembly factors 202 – skin abnormalities 248 – molecular architecture 9
– – DNA maintenance 202 – therapeutic options 250
Subject Index 1081

– morphology 1 neuronal ceroid lipofuscinoses Pelizaeus-Merzbacher disease and X


– disorders 137–146 linked spastic paraplegia type II
– – classification 20–24 – age pigment 141 272–284
– – definitions 15 – Batten disease 137 – clinical variants 272
– – levels of myelin involvement 16 – classification (table) 137 – DM20 274
– turnover 14 – genes involved 141 – genetic defect 275
– myelination 10, 11, 12 – incidence 137 – magnetic resonance imaging
– myelinogenesis 10 – lysosomal location 140 276–284
– regulation of myelinogenesis 10 – magnetic resonance imaging – proteolipid protein 274
– retarded myelination 18 143–146 – X-inactivation 275
myelination 3, 4, 10, 11, 12, 37–65 – myelin reduction 141 periventricular leukomalacia 719,
– arrest 51 – neuronal lipofuscin storage 140 720, 722, 726, 727
– delayed myelination 51 – protein defects 141 peroxisomal acyl-CoA oxidase
– diffusion tensor imaging 41 – storage material in subtypes (table) deficiency 172–175
– diffusion weighted imaging 41 137 – cranio-facial dysmorphism 172
– Flechsig 37 neurosarcoidosis 787, 788 – fatty acyl-CoA oxidase 172
– hypomyelination 51 node of Ranvier 4, 15 – gene defect 172
– iconography: preterm-adult pattern nonketotic hyperglycinemia 306–311 – magnetic resonance imaging
51–65 – clinical variants 306 174–175
– irregular myelination 51 – genetic defect 307 peroxisomal biochemical functions
– magnetization transfer imaging 42 – glycine cleavage system 306 151
– marker sites 43 – magnetic resonance imaging 309, peroxisomal D-bifunctional protein
– MRI pulse sequences 39 310 deficiency 167–171
– myelination: time tables 42 – P-, T-, H- and L-proteins 307 – clinical phenotype 167
myelinogenesis 10 – therapeutic options 308 – gene defect 168
myoneurogastrointestinal Northern epilepsy 137 – magnetic resonance imaging
encephalopathy (MNGIE) 221–223 168–171
myotonic dystrophy, type 1 469–472 O peroxisomal single enzyme defects
– anticipation 469, 470 occipital horn syndrome 400 153
– clinical variants 469 oculo-cerebro-renal syndrome peroxisome biogenesis 151
– Curschmann-Steinert disease 469 387–391 – disorders 153, 154–166
– genetic defect 470 oculo-dento-digital dysplasia – – clinical variants 154
– magnetic resonance imaging 479–480 – – Zellweger syndrome 154
470–472 – clinical variants 479 – – neonatal adrenoleukodystrophy
– therapeutic options 470 – connexin 43 479 154
– unstable CTG repeat 469 – genetic defect 479 – – infantile refsum syndrome 154
myotonic dystrophy, type 2 473–475 – magnetic resonance imaging 480 – – pathology 155
– clinical variants 473 oculogastrointestinal muscular dystro- – – pathogenesis 157
– genetic defect 474 phy (OGIMD) 221–223 – – genetic defects 157, 158
– magnetic resonance imaging 474, oligodendrocyte 4 – – therapy 159
475 – specific protein 7 – – magnetic resonance imaging
– proximal myotonic dystrophy 473 ornithine transcarbamylase deficiency 160–166
– therapeutic options 474 360, 362, 365 peroxisomes and peroxisomal disor-
– unstable CCTG repeat 474 ovarioleukodystrophy 481 ders 151–153
phenylalanine metabolism 284
N P phenylketonuria 284–293
NARP syndrome 227, 244 pattern recognition – clinical variants 284
Nasu-Hakola disease 552–556 – examples 891–904 – genetic defect 287
neonatal adrenoleukodystrophy – in unclassified white matter disor- – magnetic resonance imaging
154–166 ders 889 289–294
neonatal hypoglycemia 749–754 – in white matter disorders 881–904 – maternal phenylketonuria 285
– causes 749 – – computer-aided pattern recogni- – neonatal screening 285
– glucose transporters 749 tion 886 – phenylalanine
– magnetic resonance imaging – – general characteristics 884 – – hydroxylase 284
752–754 – – non-computerized pattern recog- – – metabolism 284
– selective vulnerability in hypo- nition 882 – tetrahydrobiopterin 284
glycemia 749, 750 – – practical applications 886 – – synthesis 285
– therapeutic options 750, 751 – – sensitivity and specificity 881 – therapeutic options 288
neuromyelitis optica 567, 597 – – special characteristics 884 3-phosphoglygerate dehydrogenase
– – structural image elements 884 deficiency 369–371
PIBIDS 268–272
1082 Subject Index

pigmentary orthochromatic leukodys- – patterns of white matter damage in remyelination 18


trophy 557–558 oxygen deprivation 716, 717 retarded myelination 18, 51, 42
– magnetic resonance imaging 558 – reperfusion damage 714 reversible posterior leukoencephalopa-
Pollit syndrome 268–272 – superoxide dismutase 714 thy syndrome 699–709
polyarteritis nodosa 777, progressive multifocal leukoen- rheumatoid disease 781
polycystic lipomembranous osteodys- cephalopathy 628–634
plasia with sclerosing leuken- – B-cell infection 629 S
cephalopathy 526, 552–556 – JC virus infection 628 Salla disease 133–136
– clinical variants 552 – magnetic resonance imaging saltatory conduction 15
– genetic defects 553 630–634 Sandhoff disease 103–111
– magnetic resonance imaging – oligodendrocyte infection 629 Sanfilippo syndrome 83
553–556 – Papova-Polyoma virus 628 Santavuori disease 137, 144, 145
– Nasu-Hakola disease 552 – simian virus 40 (SV40) 629 Sarnat classification 719
– skeletal abnormalities 552 – therapeutic options 630 Scheie syndrome 123, 124
polyneuropathy, ophthalmoplegia, – tumor induction 630 Schilder diffuse sclerosis 567, 598, 599
leukoencephalopathy, intestinal propionic acidemia 300–306 Schmidt-Lantermann cleft 4
pseudo-obstruction (POLIP) – biotin cofactor 300 Schwann cell 4
221–223 – clinical variants 300 selective vulnerability 25–36
posterior reversible encephalopathy – genetic defect 301 – causes of selective involvement
syndrome 679, 680, 699–709, 808 – ketotic hyperglycinemia 300 30–35
– complication of medical treatment – magnetic resonance imaging – topistic areas 25
699 303–305 serine synthesis defect 369–371
– eclampsia, pre-eclampsia 699 – propionyl-CoA carboxylase 300 – genetic defect 369
– hypertensive encephalopathy 699 – therapeutic options 301 – magnetic resonance imaging 370,
– magnetic resonance imaging proteolipid protein 7, 274 371
701–709 psychosine (in Krabbe disease) 89 – 3-phosphoglycerate dehydrogenase
– related conditions 699 pyruvate carboxylase deficiency 369
– reversible posterior leukoen- 245–247 – treatment options 369
cephalopathy syndrome 699 – biotin dependent carboxylases 246 sialic acid 134
– therapeutic options 700 – clinical variants 245 – transporter 134
– uremic encephalopathies 700 – genetic defect 245 sickle cell disease 798
posthypoxic-ischemic encephalopathy – magnetic resonance imaging Sjögren syndrome 786
in neonates 718–749 246–247 Sjögren-Larsson Syndrome 383–386
– basal ganglia damage 718 – pyruvate-oxalate conversion 246 – clinical triad 383
– central cortical and subcortical dam- – therapeutic options 246 – fatty alcohol-NAD+ oxidoreductase
age 718 pyruvate dehydrogenase complex defi- 383
– germinal layer hemorrhage 720 ciency 226, 229, 224 – fatty aldehyde dehydrogenase 384
– magnetic resonance imaging – genetic defect 383
723–748 R – leukotriene B4 384
– multicystic encephalopathy 720 Radiotherapy 808–817 – magnetic resonance imaging
– periventricular leukomalacia 718, – acute postradiotherapy syndrome 384–386
719 809 – therapeutic options 384
– prognostic factors 719 – early delayed postradiotherapy Sly syndrome 125
– Sarnat classification 719 syndrome 809 Sneddon syndrome 785
– subcortical leukomalacia 718 – late postradiotherapy syndrome Spielmeijer-Vogt disease 137
– therapeutic options 723 809 Stejskal-Tanner sequence 840
– venous infarction 720 recombinant a-galactosidase 113 subacute combined degeneration
posthypoxic-ischemic damage Refsum disease (heredopathia atactica of the cord 350
714–719 polyneuritiformis) 91–194 subacute sclerosing panencephalitis
– anti-oxidance capacity 715 – cardiomyopathy 191 640–644
– arachidonic acid 715 – clinical variants 191 – absence of M protein 642
– calcium-entry blockers 715 – genetic defect 193 – anti-measles virus antibody 640,
– excitatory amino acids 716 – hereditary sensory and motor neu- 641
– free radicals 714, 715 ropathy type 4 191 – M protein gene mutations 641
– – scavengers 714 – ichthyosis 191 – magnetic resonance imaging
– glutamate 716 – magnetic resonance imaging 194 642–644
– Haber-Weiss reaction 714 – phytanic acid levels 191, 192 – measles infection 640
– nitric oxide 715 – phytanoyl-CoA hydroxylase 193 – paramyxoviridae family 641
– N-methyl-D-aspartate (NMDA) 716 – pipecolic acidemia 193 – slow virus infection 640
– oxygen paradox 714 – skeletal deformities 191 – stages of disease 640, 641
– therapeutic options 194
Subject Index 1083

subcortical arteriosclerotic – Pollit syndrome 268 – sickle cell disease 798


encephalopathy 767–772 – Tay syndrome 268 – Sjögren syndrome 786
– Binswanger disease 767–772 – xeroderma pigmentosum 269 – Sneddon syndrome 785
– criteria 767 tuberculosis 795, 796 – syphilitic arteritis 795
– état – systemic lupus erythematosus 783
– – criblé 767 U – Takayasu arteritis 776
– – lacunaire 767 Uhthoff symptom 212 – temporal arteritis 775
– lacunar infarctions 767 urea cycle 360 – tuberculosis 795
– magnetic resonance imaging – disorders 360–369 – varicella-zoster vasculitis 796
768–772 – – arginase deficiency 360 – Wegener disease 789
– therapeutic options 768 – – argininosuccinate lyase deficiency venous infarction 720, 724
– Virchow-Robin spaces 767 360 vitamin B12 deficiency (see also cobal-
subcortical leukomalacia 718 – – argininosuccinate synthetase amin deficiency) 343, 346, 354, 356
sulfite oxidase deficiency 372–376 deficiency 360
18q– syndrome 281–283 – – carbamyl phosphate synthetase W
– clinical variants 281 deficiency 360 Walker-Warburg syndrome 451, 455,
– contiguous gene syndrome 281 – – female heterozygotes of ornithine 466
– magnetic resonance imaging 282, transcarbamylase deficiency 360 Wallerian degeneration 832–837
283 – – genetic defects 362 – magnetic resonance imaging
– myelin basic protein 281 – – hyperargininemia 360 834–838
syphilitic arteritis 795 – – magnetic resonance imaging – stages of myelin degradation 832,
systemic lupus erythematosus 363–369 833
781–784 – – ornithine transcarbamylase Wegener disease 789, 790
deficiency 360 Whipple disease 658–663
T – – therapeutic options 362 – clinical triad 658
Takayasu arteritis 776, 777 – – urea cycle defects 360 – CNS involvement 658
Tay syndrome 268–272 – lipofuscin granules 658
Tay-Sachs disease 103–111 V – magnetic resonance imaging
temporal arteritis 775 Van Bogaert-Bertrand disease 659–663
tetrahydrobiopterin deficiency 285 326–333 – therapeutic options 659
– synthesis 285 vanishing white matter 481–494 – Tropheryma Whippelii 658
thrombotic thrombocytopenic purpura varicella-zoster vasculitis 796 white matter lesions in the elderly
699 vasculitis 773–800 759–766
toxic encephalopathies 664–678 – ANCA test 789 – age-related changes 759
– Bonhoeffer reaction types 664 – angiotensin converting enzyme – cerebral perfusion 760
– chemical affinity 665 787 – leukoaraiosis 759
– classification 667 – anti-phospholipid antibody syn- – magnetic resonance imaging
– drug abuse 670 drome 785 761–766
– endogenous intoxications 667 – Behçet disease 789 – mild cognitive impairment 759
– exogenous-external intoxications – benign primary angiitis 776 – MR grading of lesions 762
667 – Borrelia burgdorferi 792 – quantitative MR techniques 759
– fetal intoxications 677 – Churg-Strauss syndrome 789 – risk factors 760
– historic examples 664 – classification 774 Wilson disease 392–399
– iatrogenic intoxications 669 – CREST 786 – ceruloplasmin 393
– lipophilic substances 665 – drug-induced vasculitis 797 – clinical variants 392, 393
– magnetic resonance imaging – erythema migrans 793 – copper
(examples) 665–678 – giant cell arteritis 775 – – concentration 393
– mechanisms of selective vulnera- – granulomatous angiitis 774 – – homeostasis 394
bility 665 – herpes zoster ophthalmica 797 – – toxicity 395
– organic solvents 670 – hydroxyurea 799 – genetic defect 394
– topistic areas 664 – hypercoagulative states 798 – hepatolenticular degeneration 392
trichothiodystrophy with photo- – intravascular lymphomatosis 798 – Kayser-Fleischer rings 392
sensitivity 268–272 – Lyme disease 792 – magnetic resonance imaging
– basal transcription factor II H 269 – magnetic resonance imaging 396–399
– clinical variants 268 799–800 – metallothionein 394
– DNA-repair 269 – microscopic polyarteritis 789 – therapeutic options 395
– genetic defects 269 – Moyamoya syndrome 777
– hair abnormalities 268 – neurosarcoidosis 787 X
– magnetic resonance imaging – polyarteritis nodosa 777 Xeroderma pigmentosum 260, 269
270–271 – rheumatoid disease 781 X-linked adrenoleukodystrophy
176–190
1084 Subject Index

– ABCD2 gene 182 – magnetic resonance imaging – VLCFA b-oxidation 179


– adrenal insufficiency 176 182–190 – VLCFA-CoA synthase 179
– clinical variants 176 – MTR 187, 857, 858 X-linked ichthyosis 83
– fatty acid chain elongation 180 – pathogenesis 180
– female carriers 177 – therapeutic options 181 Z
– genetic defect 179 – unusual forms of X-ALD 177 Zellweger syndrome 154–166
– Loes score 186 – very-long-chain fatty acids (VLCFA)
– Lorenzo oil 181 177

You might also like