Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Accelerat ing t he world's research.

Metabolic and molecular basis of


peroxisomal disorders: A review
Ronald Wanders
American Journal of Medical Genetics

Cite this paper Downloaded from Academia.edu 

Get the citation in MLA, APA, or Chicago styles

Related papers Download a PDF Pack of t he best relat ed papers 

Clinical approach t o inherit ed peroxisomal disorders: A series of 27 pat ient s


Marc Espeel, Frank Roels

Laborat ory Diagnosis of Peroxisomal Disorders in t he -Omics Era and t he Cont inued Import ance of Bi…
Ronald Wanders

Diagnost ic work-up of a peroxisomal pat ient


Marc Espeel
American Journal of Medical Genetics 126A:355 – 375 (2004)

Research Review
Metabolic and Molecular Basis of Peroxisomal Disorders:
A Review*
Ronald J.A. Wanders**
University of Amsterdam, Academic Medical Centre, Departments of Clinical Chemistry and Pediatrics, Emma Children’s Hospital,
Laboratory of Genetic Metabolic Diseases, Amsterdam, The Netherlands

The group of peroxisomal disorders now includes consensus to use a classification in which two groups of
17 different disorders with Zellweger syndrome as disorders are distinguished: (1) the disorders of peroxisome
prototype. Thanks to the explosion of new infor- biogenesis and (2) the single peroxisomal enzyme deficiencies.
mation about the functions and biogenesis of It is the purpose of this review to update the reader on the
peroxisomes, the metabolic and molecular basis clinical, metabolic, and molecular aspects of peroxisomal
of most of the peroxisomal disorders has been disorders. The major basic aspects of peroxisome biogenesis
resolved. A review of peroxisomal disorders is and function will be discussed first.
provided in this paper. ß 2004 Wiley-Liss, Inc.
Peroxisome Biogenesis
KEY WORDS: peroxisomes; genetic diseases;
phospholipids; fatty acids; Zell- Identification of genes and proteins involved in peroxisomes
weger syndrome biogenesis has been helped by the conservation of peroxisome
biogenesis among eukaryotic species, from yeasts and plants to
mammals. In fact, studies in yeasts have set the stage followed
by the subsequent identification of corresponding human
INTRODUCTION genes and proteins. To avoid confusion, a unified nomenclature
A characteristic feature of eukaryotic cells is the presence of has been agreed in which genes involved in peroxisome
specialized subcompartments which all play their own distinct biogenesis are called PEX-genes and the encoded proteins
role in cell physiology. In order to fulfil their separate peroxins.
functions, the different subcellular compartments (mitochon- The first principle of peroxisome biogenesis is that perox-
drion, lysosome, nucleus, cytosol, and endoplasmatic reticu- isomal proteins are encoded by nuclear genes, synthesized on
lum) are all equipped with a unique set of enzymes to carry out free cytosolic polyribosomes, and imported posttranslationally.
their different tasks. After long being underrated as relic This applies to both peroxisomal matrix and peroxisomal
organelles of little physiological relevance, the true function of membrane proteins. The implication is that newly synthesized
peroxisomes in cell physiology has become more clear in recent peroxisomal proteins must have specific targeting signals
years. Most of the functions of peroxisomes involve lipid that direct them to and into peroxisomes. It turns out that
metabolism, and defects in one or more peroxisomal functions peroxisomal matrix proteins contain one of two different
are associated usually with severe clinical manifestations. The signals called PTS1 and PTS2. The PTS1 signal is a C-terminal
prototype of the group of peroxisomal disorders is the cerebro- tripeptide with the sequence Ser-Lys-Leu (SKL), or a con-
hepato-renal syndrome or Zellweger syndrome (ZS), first served variant thereof, used by most (>90%) of the human
described in 1964 as a familial syndrome of multiple congenital peroxisomal proteins. A few matrix proteins are targeted by a
defects in two pairs of sibs [Bowen et al., 1964]. distinct signal, PTS2, located near the N-terminus which
The group of peroxisomal disorders now includes 17 different conforms to the motif Arg/Lys-Leu/Val/Ile-X5-Gln/His-Leu in
diseases listed in Table I. Although different classifications which X denotes any amino acid (Fig. 1). The second principle of
have been proposed through the years, there is growing peroxisome biogenesis is that peroxisomal proteins are taken
up into pre-existing peroxisomes thereby expanding the
peroxisomal compartment until a critical size is reached which
results either in fission of peroxisomes into two daughter
peroxisomes or in budding from the peroxisome reticulum
followed by subsequent growth. Conceptually, the complicated
process of peroxisome biogenesis can be subdivided into
Grant sponsor: The Princess Beatrix Fund; Grant sponsor: The different steps including: (1) recognition of the PTS1 and
Dutch Foundation for Scientific Research (NWO); Grant numbers: PTS2 signals by soluble receptors; (2) docking of the receptors
901-03-159, 901-03-133; Grant sponsor: The European Union; loaded with the PTS1 or PTS2 protein to the peroxisomal
Grant numbers: QLG1-CT-2001-1277, QLG3-CT-2002-00696. membrane; (3) dissociation of the receptor–ligand complexes
*After this paper was submitted, the gene defective in the last followed by transport of the ligand across the peroxisomal
complementation group (Table II) was reported [Matsumoto et al. membrane; and (4) recycling of the receptors. Two different
(2003) Nat Cell Biol 5:454–460; Matsumoto et al. (2003) Am J strategies have been productive in identification of human
Hum Genet 73:233–246]. The gene involved, PEX26, codes for a genes involved in peroxisome biogenesis. The first strategy
peroxisomal membrane protein which recruits the Pex1p–Pex6p involved homology probing making use of the information
AAA ATPase complex to the peroxisomal membrane. available from the different genetic studies in yeasts. Com-
**Correspondence to: Prof., Dr. Ronald J.A. Wanders, Lab puter-based searches of mammalian sequence databases
Genetic Metabolic Diseases, Academic Medical Centre, Meiberg- with yeasts peroxin sequences have identified several human
dreef 9, 1105 AZ Amsterdam, The Netherlands. homologs [Gould and Valle, 2000]. The second strategy
E-mail: r.j.wanders@amc.uva.nl involved peroxisome-deficient Chinese Hamster Ovary
Received 30 June 2003; Accepted 9 September 2003 (CHO) cells and mammalian cDNA expression libraries to
DOI 10.1002/ajmg.a.20661 complement the defect. These two strategies have led to

ß 2004 Wiley-Liss, Inc.


356 Wanders

TABLE I. The Peroxisomal Disorders

Disorder Abbreviation Protein involved Gene Chromosome MIM

Disorders of peroxisome
biogenesis
Zellweger syndrome ZS Peroxins PEX-genes Multiple loci 214100
Neonatal adrenoleukodystrophy NALD Peroxins PEX-genes Multiple loci 214110
Infantile Refsum disease IRD Peroxins PEX-genes Multiple loci 202370
Hyperpipecolic acidaemiaa HPA Peroxins PEX-genes Multiple loci 266510
Rhizomelic chondrodysplasia RCDP type 1 Pex7p PEX7 6q21-q22.2 215100
punctata type 1
Single peroxisomal enzyme/
protein deficiencies
X-linked adrenoleukodystrophy X-ALD ALDP ABCD1 Xq28 300100
Acyl-CoA oxidase deficiency ACOX1-deficiency Straight-chain acyl-CoA ACOX1 17q25.1 264470
(pseudo-neonatal ALD) oxidase (SCOX/
ACOX1)
D-bifunctional protein D-BP deficiency D-BP/MF2/MFEII/ HSD17B4 5q2 261515
deficiency D-PBE
2-methylacyl-CoA racemase Racemase AMACR AMACR 5p13.3-p12 604489
deficiency deficiency
Rhizomelic chondrodysplasia RCDP type 2 DHAPAT GNPAT 1q42.1-42.3 222765
punctata type 2 (DHAPAT
deficiency)
Rhizomelic chondrodysplasia RCDP type 3 ADHAPS AGPS 2q33 600121
punctata type 3 (alkylDHAP
synthase deficiency)
Refsum disease (phytanoyl-CoA ARD Phytanoyl-CoA PHYH/PAHX 10p15-p14 266500
hydroxylase deficiency) hydroxylase (PhyH)
Hyperoxaluria type 1 PH1 Alanine glyoxylate AGXT 2q37.3 259900
aminotransferase
(AGT)
Glutaric acidemia type 3 GA3 ? ? ? 231690
Mevalonate kinase deficiency MK-deficiency Mevalonate kinase MVK 12q24.1 251170, 260920
Acatalasaemia — Catalase CAT 11p13 115500
Mulibrey nanism MUL Trim37p TRIM 17q22-23 253250
a
At least the four original HPA-patients described by Gatfield et al. [1968], Burton et al. [1981], and Thomas et al. [1975] are now known to be affected by a
disorder of peroxisome biogenesis. As described in the text, additional HPA-patients have been described in literature in whom peroxisome biogenesis is
normal. The primary defect in these patients remains to be identified.

the identification of many human PEX-genes and allowed the different fatty acids and fatty acid derivatives. Indeed mito-
virtually complete resolution of the molecular basis of the chondria catalyze the b-oxidation of the bulk of fatty acids
peroxisome biogenesis disorders (PBDs). Furthermore, our derived from the diet including palmitate, oleate, linoleate, and
understandings about the functions of peroxins have rapidly linolenate. Peroxisomes play an important role by oxidizing a
grown. Figure 1 is a schematic representation of peroxisome different set of fatty acids and fatty acid derivatives: (1) very-
biogenesis in humans and the proposed function of the long-chain fatty acids (VLCFA), notably hexacosanoic acid
different peroxins [Gould and Valle, 2000]. (C26:0); (2) pristanic acid (2, 6, 10, 14-tetramethylpentadeca-
noic acid) as derived from dietary sources either directly or
indirectly (from phytanic acid) and; (3) di- and trihydroxycho-
FUNCTIONS OF PEROXISOMES
lestanoic acid (DHCA and THCA). The latter two compounds
Peroxisomes play an indispensable role in human metabo- are intermediates in the formation of the primary bile acids,
lism as stressed by the devastating consequences of the cholic acid and chenodeoxycholic acid from cholesterol in the
absence of peroxisomes in Zellweger patients. The main liver (Fig. 2).
functions of peroxisomes in humans are briefly discussed Another major function of the peroxisomal b-oxidation
below. system concerns ‘‘biosynthesis’’ of polyunsaturated fatty acids
including docosahexaenoic acid (C22:6o3). Recent studies have
established that the formation of C22:6o3 from linolenic acid
Fatty Acid Beta-Oxidation
(C18:3o3) involves the active participation of the peroxisomal
Peroxisomes contain a fatty acid b-oxidation machinery just fatty acid oxidation (FAO) system (Fig. 2). It should be noted
as mitochondria do. The mechanism of b-oxidation in the two that the peroxisomal FAO system is only able to chain-shorten
organelles is identical and involves four consecutive reactions: fatty acids and not able to degrade fatty acids to completion.
dehydrogenation, hydration (of the double bond), dehydro- This is most convincingly demonstrated for pristanic acid
genation again, and thiolytic cleavage. Through this four-step which undergoes three cycles of b-oxidation in peroxisomes to
mechanism a two-carbon unit is split from the fatty acid in the produce 4,8-dimethylnonanoyl-CoA which is then shuttled to
form of an acetyl-CoA unit, which can then be degraded in the the mitochondria in the form of its carnitine-ester for full
citric acid (Krebs) cycle to produce CO2 and H2O plus energy oxidation to CO2 and H2O. The same is true for VLCFA which
(ATP). are probably chain shortened to a medium-chain acyl-CoA-
The peroxisomal and mitochondrial b-oxidation systems ester followed by transport to the mitochondria as carnitine-
have different functions as they catalyze the b-oxidation of ester to allow complete oxidation to CO2 and H2O (Fig. 2).
Peroxisomal Disorders 357

Fig. 1. Current model of peroxisome biogenesis. PTS1-proteins synthe- Pex14p and subsequently to Pex13p, the complexes dissociate and the PTS1-
sized on free polyribosomes are recognized and bound by the PTS1-receptor and PTS2-proteins (ligands) translocate across the peroxisomal membrane,
(either Pex5pS or Pex5pL). Pex5pS is capable of targeting PTS1-proteins to a process dependent on Pex2p, Pex10p, and Pex12p. The receptors Pex5pS,
the peroxisomal docking machinery with Pex14p as the first binding Pex5pL, and Pex7p recycle back to the cytosol. Pex1p and Pex6p are
partner, whereas Pex5pL is needed for PTS2-import with Pex7p as the supposed to be involved in the latter process. Not shown are Pex3p, Pex16p,
PTS2-receptor. In the absence of Pex5pL, the Pex7p/PTS2-protein complex and Pex19p which are involved in the correct targeting of peroxisomal
is unable to dock at the peroxisomal membrane. After binding of the Pex5pS/ membrane proteins (PMP).
PTS1-protein or Pex5pL/PTS1-protein/Pex7p/PTS2-protein complex to
358 Wanders

Fig. 2. Schematic representation of the role of the peroxisomal b-oxidation system in the oxidation of VLCFAs, pristanic acid, tetracosahexaenoic acid
(C24:6o3), and di- and trihydroxycholestanoic acid (DHCA and THCA) and the interaction between peroxisomes and mitochondria, the ultimate site of
oxidation of the end products of peroxisomal b-oxidation to CO2 and H2O.
Peroxisomal Disorders 359

Fig. 3. Enzymology of the peroxisomal fatty acid b-oxidation system. of the two acyl-CoA oxidases, i.e., the enoyl-CoA esters of C26:0, pristanic
Human peroxisomes contain two acyl-CoA oxidases, one specific for straight- acid and di- and trihydroxycholestanoic acid, are handled by a single en-
chain fatty acyl-CoAs and called straight-chain acyl-CoA oxidase (SCOX), zyme, i.e. D-bifunctional protein (D-BP) whereas two thiolases are involved
and a second, specifically reactive with 2-methyl branched-chain fatty acyl- in the final cleavage step.
CoAs (pristanoyl-CoA, di- and trihydroxycholestanoyl-CoA). The products

Figure 2 also shows chenodeoxycholoyl-CoA and choloyl-CoA converted into taurine or glycine conjugates which are then
to be the products of b-oxidation of the bile acid intermediates exported from peroxisomes into bile.
dihydroxycholestanoyl-CoA and trihydroxycholestanoyl-CoA. Figure 3 depicts the enzymology of the peroxisomal fatty acid
In peroxisomes, chenodeoxycholoyl-CoA and choloyl-CoA are b-oxidation system. Human peroxisomes contain 2 acyl-CoA
360 Wanders

oxidases, one specific for straight-chain fatty acids like C26:0, Biosynthesis of Cholesterol
therefore called straight-chain acyl-CoA oxidase (SCOX or and Other Isoprenoids
ACOX1) and a second one, catalyzing the dehydrogenation of
Peroxisomes may play a major role in isoprenoid biosynth-
2-methyl branched-chain fatty acids like pristanoyl-CoA and
esis as enzymes like mevalonate kinase, phosphomevalonate
di- and trihydroxycholestanoyl-CoA. The enoyl-CoA-esters of
kinase, and mevalonate pyrophosphate decarboxylase have
C26:0, pristanic acid, DHCA, and THCA are all handled by a
been claimed to be localised in peroxisome. If the first part of
single bifunctional enzyme harboring both enoyl-CoA hydra-
the de novo isoprenoid biosynthesis pathway from acetyl-CoA
tase and 3-hydroxyacyl-CoA dehydrogenase activity. This
to farnesylpyrophosphate is indeed localised in peroxisomes,
bifunctional protein named DBP is the single enzyme involved
mevalonate kinase is a peroxisomal disorder (see footnote1
in both oxidation of C26:0, pristanic acid, DHCA, and THCA.
however).
The role of the originally identified L-bifunctional enzyme
(LBP) remains to be resolved.
Detoxification of Glyoxylate
Ether-Phospholipid Biosynthesis Glyoxylate is a toxic metabolite as it is a substrate for the
ubiquitous enzyme lactate dehydrogenase which interconverts
Ether-phospholipids are a special class of phospholipids
pyruvate and lactate but also converts glyoxylate into oxalate.
which differ from the regular diacylphospholipids in an ether-
Oxalate rapidly precipitates as calcium oxalate with severe
linkage rather than an ester-linkage at the sn-1 position of
consequences for the tissues involved. Under normal condi-
the glycerol backbone. Two groups of ether phospholipids
tions, glyoxylate is rapidly converted into glycine via the
can be distinguished with either an 1-O-alkyl or an 1-O-alk-10 -
peroxisomal enzyme alanine glyoxylate aminotransferase
enyl-linkage. The latter phospholipids (1-O-alk-10 -enyl-2-acyl-
(AGT). In hyperoxaluria type 1, this enzyme is deficient,
phosphoglycerides) with an a, b-unsaturated ether-bond are
glyoxylate accumulates, and is either oxidized to oxalate or
also known as plasmalogens. Platelet-activating factor (PAF; reduced to glycolate (Fig. 6).
1-O-alkyl-2-acetylglycerophosphocholine) is the best known
ether-phospholipid. Figure 4 depicts the enzymology of the
etherphospholipid biosynthetic pathway with part of the L-Pipecolic Acid Oxidation
enzymes localized in peroxisomes and another part in L-lysine is normally degraded via the saccharopine pathway
the endoplasmatic reticulum. Biosynthesis of ether-phospho- involving the sequential action of: (1) L-lysine: 2-oxoglutarate
lipids starts in the peroxisome with production of dihydrox- reductase and (2) saccharopine dehydrogenase. However, L-
yacetonephosphate (DHAP) to acyl-DHAP as catalyzed by the lysine may also be degraded through the L-pipecolic acid
peroxisomal enzyme dihydroxyacetonephosphate-acyltrans- pathway which may especially be important in the brain since
ferase (DHAPAT), followed by the introduction of the typical the activity of the saccharopine pathway is low in the brain. In
ether-bond by the enzyme alkyl-DHAP synthase. Both the L-pipecolic acid pathway, L-pipecolic acid is produced from
enzymes are strictly peroxisomal. The third step is catalyzed L-lysine in 2 steps, and then oxidized by L-pipecolate oxidase, a
by the enzyme alkyl/acyl-DHAP:NAD(P)H oxidoreductase peroxisomal enzyme in man [Mihalik et al., 1989; Wanders
which has a bimodel distribution in both peroxisomes and et al., 1989]. The structure and function of the L-pipecolic acid
endoplasmatic reticulum. The product alkylglycerol-3-phos- pathway remains incompletely understood.
phate (alkyl-G-3P) undergoes subsequent conversion into
plasmalogens.
THE PEROXISOMAL BIOGENESIS
DISORDERS (PBDs)
Fatty Acid a-Oxidation
Zellweger Syndrome (MIM 214100)
3-methyl-branched fatty acids cannot be b-oxidized directly
due to the presence of a methyl group at the 3-position. Instead, ZS was first described by Bowen et al. [1964] in two pairs of
3-methyl fatty acids first undergo a-oxidation thereby remov- siblings. Subsequently, many additional patients have been
ing the terminal carboxyl group as CO2. One of the most described in the literature (for reviews see [Heymans, 1984;
important 3-methyl-branched fatty acids is phytanic acid, long Wilson et al., 1986; Wanders et al., 1988a]). The clinical
known to accumulate in Refsum disease as well as in other presentation of ZS is dominated by the typical craniofacial
peroxisomal disorders (see below). The mechanism involved in features and neurological aberrations. Facial features include
the oxidative decarboxylation of 3-methyl fatty acids has been a large anterior fontanel with widely spaced sutures (96%), a
resolved in recent years. Figure 5 depicts the structure and broad, full forehead (97%), micrognathia (100%), external ear
enzymology of the peroxisomal fatty acid a-oxidation system. deformity (97%), low and broad nasal bridge (100%), shallow
Phytanic acid first undergoes activation to phytanoyl-CoA, via orbital ridges (100%), and redundant skin folds in the neck
the long-chain acyl-CoA synthetase (LACS) present at the (100%). The neurological picture is dominated by profound
outer aspect of the peroxisomal membrane and then enters hypotonia (99%) resulting in poor sucking (97%) requiring
the peroxisomal matrix probably via a carrier protein. Once gavage feeding, depressed neonatal and deep tendon reflexes,
inside peroxisomes, phytanoyl-CoA undergoes hydroxylation and a flat occiput. Other neurological abnormalities include an
followed by cleavage to pristanal and formyl-CoA. Subse- abnormal Moro response (100%), hypo/areflexia (98%), and
quently, pristanal is converted into pristanic acid which is then seizures (92%). Ocular abnormalities including corneal cloud-
activated to pristanoyl-CoA. Pristanoyl-CoA undergoes three ing, congenital cataracts, and glaucoma, whereas Brushfield
cycles of b-oxidation in the peroxisome to produce 4,8- spots and abnormal retinal degeneration are also frequently
dimethylnonanoyl-CoA which leaves the peroxisome as carni- encountered. The electroretinogram was extinguished in vir-
tine-ester to undergo full oxidation to CO2 and H2O in tually all patients.
mitochondria. Evidence holds that the complete pathway
from phytanoyl-CoA to 4,8-dimethylnonanoyl-CoA is intra-
peroxisomal (Fig. 5). Most enzymes involved in peroxisomal 1
A recent report by Hogenboom et al. [2004] provides convincing
fatty acid a-oxidation and their encoding genes have been evidence in favour of a cytosolic localisation of mevalonate kinase,
identified with the exception of pristanal dehydrogenase which implies that mevalonate kinase deficiency can no longer be
[Wanders et al., 2001b,c]. considered a peroxisomal disorder.
Peroxisomal Disorders 361

Fig. 4. Pathway for the synthesis of etherphospholipids. Biosynthesis starts in the peroxisome by acyldihydroxyacetonephosphate (acylDHAP)
formation using dihydroxyacetonephosphate acyltransferase (DHAPAT), and then the ether-bond is introduced by alkyldihydroxyacetonephosphate
synthase (ADHAPS) generating alkylDHAP. Both DHAPAT and ADHAPS are strictly peroxisomal in contrast to the other enzymes.

The liver is enlarged (78%) and fibrotic (78%) with severe The most conspicuous pathological abnormalities are found
micronodular cirrhosis. Renal cysts were observed in 78 of in the brain [Liu et al., 1976; Evrard et al., 1978; Aubourg et al.,
80 patients who were studied pathologically [Heymans, 1984; 1985; Powers et al., 1989]. Macroscopic abnormalities include
Wanders et al., 1988a]. Calcific stippling of the patellae and deviant sulci and gyri such as deep, almost vertical parietal
other bones occurs in 50–60% of patients. clefts and pachymicrogyria. Microscopic abnormalities show
362 Wanders

Fig. 5. Structure and enzymology of the peroxisomal phytanic acid a- the enzyme phytanoyl-CoA hydroxylase. Subsequently, 2-hydroxyphyta-
oxidation system. Phytanic acid first undergoes activation to phytanoyl- noyl-CoA is cleaved by the enzyme 2-hydroxyphytanoyl-CoA lyase (2HPCL)
CoA, a reaction probably carried out by the long-chain acyl-CoA synthase into formyl-CoA and pristanal, which is converted into pristanic acid via
(LACS) present at the outer aspect of the peroxisomal membrane. an ill-defined aldehyde dehydrogenase. Pristanic acid is converted into its
Phytanoyl-CoA then enters the peroxisome via an unknown mechanism CoA-ester, probably by the enzyme very-long-chain acyl-CoA synthetase
after which phytanoyl-CoA is hydroxylated to 2-hydroxyphytanoyl-CoA via (VLACS) followed by b-oxidation.
Peroxisomal Disorders 363

Fig. 6. Metabolism of glyoxylate into glycine, glycolate, and oxalate. Under normal conditions, glyoxylate is converted into glycine within the peroxisome
via the enzyme alanine glyoxylate-aminotransferase (AGT). If AGT is functionally inactive, either through loss of catalytic activity or through
mislocalization to mitochondria, glyoxylate accumulates followed by oxidation to oxalate or reduction to glycolate.

two types of abnormalities: dysontogenetic and degenerative. mia, and dysmorphic features. Ultrastructural analysis of a
The former type affects neuromigration at different levels and liver biopsy specimen revealed lamellar structures that
in different areas. The neocortex migration defect consists of resembled those normally found in plant chloroplasts, which
medium and large sized neurons immediately beneath the are known to contain bound phytol. This prompted the authors
molecular layer, marginal glial heterotopia, simple microgyri to measure plasma phytanic acid, which turned out to be highly
with apparently fused first layers, microgyria of the cloverleaf elevated (50–100 mg/ml: normal: 3 mg/ml). This finding led to
type, and subcortical and deep nodular neuronal heterotopia. the name ‘‘infantile phytanic acid storage disease,’’ or IRD,
Germinolytic cysts in the subependymal areas of the cerebral which name is confusing because of the implied relationship to
hemispheres relate to early loss of cells inhabiting the germinal adult Refsum disease. The patients described by Scotto et al.
matrix. Purkinje cell heterotopia and dysplastic inferior [1982] were later re-described in more detail by Poll-Thé et al.
olivary nuclei are found. The degenerative features are those [1987].
of storage and cell degeneration and cell death: storage as Further studies demonstrated that the accumulation of
neutral fat in astrocytes, glycogen in cortical neurons, and phytanic acid is a phenomenon secondary to a defect in
lamellar-lipid and lamellar membrane inclusions in lateral peroxisome biogenesis. Indeed, hepatic peroxisomes are
cuneate and Clarke’s column neurons, cell degeneration in severely diminished in number [Roels et al., 1986], plasmalo-
cortical neuronal loss, fibrous gliosis, globoid cells, lipid gen synthesis is impaired, very long chain fatty acids, bile acid
phagocytes, and glial nodules. intermediates, and pipecolic acid are elevated and phytanic
acid oxidation is deficient. These data underline the conclusion
Neonatal Adrenoleukodystrophy (MIM 202370) that ZS, NALD, and IRD are phenotypic variants resulting
from a defect in peroxisome biogenesis with partly overlapping,
Another PBD is neonatal adrenoleukodystrophy (NALD),
partly divergent features [Aubourg et al., 1986; Kelley et al.,
first described by Ulrich et al. [1978] as connatal ALD in a boy
1986]. This is exemplified by the autopsy findings in a patient
with hypotonia, convulsions, absent grasp reflexes, and little
with IRD who had, at 11 years of age, severe growth re-
spontaneous movements at birth. The patient had all the signs
tardation, normocephaly, sunken eyes, tapetoretinal degen-
characteristic for adrenoleukodystrophy including demyelina-
eration, high prominent nasal bridge, numerous ectatic veins,
tion of the central nervous system (CNS) white matter, atrophy
redundant skin folds in the neck, and a small mouth [Torvik
of the adrenal cortex, ballooned adrenocortical cells, and
et al., 1988]. The patient did not speak or walk, gave no reaction
splinter-like lamellar elements composed of electron-dense
to visual or sound stimuli, and was spastic with myoclonic fits.
leaflets separated by a clear space. However, there were also a
There were no malformations of the cerebral cortex or
number of CNS abnormalities not described in X-linked
heterotopias, no cortical renal cysts, no skeletal changes, and
adrenoleukodystrophy (XALD) such as cerebral heterotopias,
no liver damage. So this case [Torvik et al., 1988] also differed
micropachygyria, and other gray matter changes reflecting
from NALD in many respects.
generalized cerebral maldevelopment. We know now that
Distinction between Zellweger syndrome, neonatal
XALD and NALD are different disorders, NALD belonging to
adrenoleukodystrophy, and infantile Refsum disease.
the PBDs and XALD to the single peroxisomal dysfunction
Because of the phenotypic overlap among the PBDs it is often
disorders [Kelley et al., 1986].
difficult to classify patients, especially since phenotypic
variants have been described which do not belong to either
Infantile Refsum Disease (MIM 266510)
known PBD [Barth et al., 2001]. ZS, NALD, and IRD all have
Infantile Refsum disease (IRD) was first described in 1982 by liver disease, variable neurodevelopmental delay, retinopathy,
two groups of investigators [Boltshauser et al., 1982; Scotto and perceptive deafness with onset in the first months of life.
et al., 1982]. Scotto et al. [1982] described three patients In addition, patients with ZS are severely hypotonic and
including a 5-year-old boy with an enlarged liver, mental weak from birth and have distinct facial features, periartic-
retardation, sensorineural deafness, pigmented retina, anos- ular calcifications, severe brain dysfunction associated with
364 Wanders

neuronal migration disorder and die before 1 year of age. Increased pipecolic acid levels have been reported [Roesel
Patients with NALD have hypotonia, seizures, may have et al., 1991] in plasma and urine of a patient with Dyggve–
polymicrogyria, have progressive white matter disease, and Melchior–Clausen syndrome, characterized by mental retar-
usually die in late infancy. Patients with IRD may have dation, and spondylometaepiphyseal dysostosis. We have since
external features reminiscent of ZS, no neuronal migration tested more than 10 patients with this syndrome and have not
disorder, and no progressive white matter disease. The found elevated pipecolic acid levels suggesting that the earlier
cognitive and motor development varies between severe global finding may have been fortuitous.
handicaps and moderate learning disabilities with deafness Vallat et al. [1996] reported the serendipitous discovery of
and visual impairment due to retinopathy. Major points of major HPA in a 44-year-old man, his major health problem
difference [Raymond and Moser, 1997] are: (1) survival. In being recurrent embolism possibly related to a moderate
general, the mean age of death of NALD patients is much later fasting hyperhomocysteinemia (20 mmol/L; normal: <14).
compared to classical ZS, while in IRD patients survival to the Plasma pipecolic acid levels were 220–250 mmol/L (normal:
second decade is not uncommon. Several patients who are now <2.5) in the presence of normal lysine levels. The chirality of
over 20 years of age are known to us; (2) NALD and IRD the pipecolic acid accumulating in the patient was determined
patients have hypotonia and some degree of psychomotor to be L-. The authors concluded that L-HPA may be a benign
development. Most achieve some head control, are able to sit trait. However, others [Kerckaert et al., 2000] identified three
up, and may achieve the ability to stand and walk indepen- patients with diverse neurological abnormalities all showing
dently, and to engage in some form of alternative communica- isolated HPA. In two, the pipecolic acid in CSF was also
tion, although only a few develop the capacity to speak; (3) strongly elevated. Hepatic peroxisomes were normally pre-
NALD and IRD patients do not have the profound and sent. The exact consequences of isolated HPA remain to be
characteristic defect of neuronal migration as in ZS [Evrard established.
et al., 1978], although some milder degrees of migration defects Biochemical and molecular basis of ZS, NALD, and
may be present; (4) facial dysmorphism is less severe in NALD IRD. Peroxisomes are strongly reduced in number and size,
and IRD than in classical ZS; (5) neonatal seizures are present or absent in ZS, NALD, and IRD. This causes a generalized loss
in both ZS and NALD, but not in IRD; (6) renal cysts and of peroxisomal functions in peroxisomal disorders, which is
chondrodysplasia punctata are present in most patients with consequently reflected in a series of abnormalities:
ZS, but not in the milder variants. Children with NALD/IRD do
have evidence of a chondrodysplasia in short stature, flat face,
* Elevated plasma very long chain fatty acids: peroxisomes
and delayed eruption of primary teeth; (7) severe hearing
are the site of oxidation of very long chain fatty acids and
impairment and pigmentary degeneration of the retina appear
oxidation of these fatty acids is deficient in fibroblasts of ZS,
to be present in all patients with the milder variants. Retinal
NALD, and IRD-patients, although the extent of the defi-
abnormalities are probably present in all patients with
ciency differs in the order ZS > NALD > IRD [Wanders et al.,
classical ZS, but may not be recognized before the patients’
1995], which explains that on average VLCFAs are highest
early death; (8) leukodystrophy may develop in the children
in plasma from ZS-patients [Lazarow and Moser, 1995].
with the more mild phenotypes: variable age of onset, loss of
* Elevated plasma di- and trihydroxycholestanoic acid
already acquired milestones, development of spasticity, and
(DHCA/THCA): in peroxisomes these acids normally under-
progressive course often leading to death.
go chain-shortening to chenodeoxycholic acid and cholic
acid, respectively (Figs. 2 and 3). In peroxisomal disorders,
Hyperpipecolic Acidemia (pMIM 239400) oxidation of DHCA and THCA is deficient, which leads to the
accumulation of these cholestanoic acids in plasma.
Several reports have appeared on what was first presumed
to be isolated hyperpipecolic acidaemia (HPA), the first one * Elevated plasma pristanic acid and phytanic acid: pristanic
[Gatfield et al., 1968] on a male infant with hepatomegaly, acid b-oxidation and phytanic acid a-oxidation is restricted
retinopathy, and a progressive neurological disorder. Three to peroxisomes, which explains why pristanic acid and
additional patients [Thomas et al., 1975; Burton et al., 1981] phytanic acid accumulate in ZS, NALD, and IRD-patients.
had hepatomegaly, retinopathy, developmental delay, and However, levels may vary markedly and may even be
dysmorphic features. When L-pipecolic acid was later found to normal, which is explained by the dietary origin of phytanic
be elevated in PBD patients too, these cases were re-evaluated acid and pristanic acid (either as pristanic acid itself or as
and in each a deficiency of peroxisomes was found [Wanders phytanic acid).
et al., 1988b, 2001a]. Hepatic peroxisomes were stated to be * Elevated L-pipecolic acid: the deficient activity of L-pipecolate
normal in the two Burton et al. cases [Challa et al., 1983], but oxidase in peroxisome-deficient cells leads to elevated
these results are hard to reconcile with data from several plasma L-pipecolic acid levels. The levels in plasma from ZS,
groups showing a generalized loss of peroxisomal functions in NALD, and IRD patients varies widely for unknown reasons.
fibroblasts from the two Burton et al. cases and the absence of * Deficient docosahexaenoic acid (DHA) levels in plasma and
punctate immunofluorescence indicating a true disorder of erythrocytes: peroxisomes are essential in DHA-formation,
peroxisome biogenesis. Others [Rao and Chang, 1992] reported which explains the deficiency of DHA in Zellweger patients
also that both L-pipecolate oxidase and acyl-CoA oxidase were although in milder affected patients levels may be normal.
deficient in fibroblasts from the first Burton patient. Recently, The deficiency is usually more pronounced in erythrocytes
we have performed more extensive studies in fibroblasts from as compared to plasma.
the Thomas et al. case by complementation studies: this * Deficient erythrocyte plasmalogens: fibroblasts studies
allowed identification of the defective gene (PEX1) (Wanders have shown that biosynthesis of etherphospholipids includ-
et al., unpublished). Based on these results, we conclude that ing plasmalogens, is severely impaired in ZS-patients but
probably all above patients were affected by a peroxisome less so in NALD and IRD patients. Erythrocyte plasmalogen
biogenesis defect. HPA does not represent a distinct pheno- levels are clearly deficient in ZS-patients but may only be
type among the disorders of peroxisome biogenesis and has partially reduced or even normal in milder affected patients
become obsolete in the classification of such disorders, espe- [Wanders et al., 1986].
cially since true, isolated HPA may well exist. Indeed, in the
literature, several patients have been described with isolated The increased knowledge on peroxisome biogenesis and the
pipecolic acidaemia. proteins involved has also led to more insight in the genetics of
Peroxisomal Disorders 365

TABLE II. Complementation Groups in Peroxisome Biogenesis Disorders (PBD)

Complementation groups PTS import


Gene
Gifu KKI Adam PTS1 PTS2 Ghosts involved Phenotypes

A 8 VI   þ PEX 26* ZS/NALD/IRD


B 7 (¼5) VII   þ PEX 10 ZS/NALD/IRD
C 4 (¼6) III   þ PEX 6 ZS/NALD/IRD
D 9 VIII    PEX 16 ZS
E 1 II   þ PEX 1 ZS/NALD/IRD
F 10 V   þ PEX 2 ZS
G 12 IX    PEX 3 NALD
H 13 X   þ PEX 13 ZS/NALD
J 14 XI    PEX 19 ZS
2 IV  ** þ PEX 5 ZS/NALD/IRD
3 XII   þ PEX 12 ZS/NALD/IRD
R 11 I þ  þ PEX 7 RCDP
*See Addendum.
**A single patient has been described in literature in which only the PTS1 import route was blocked [see Gould and
Valle, 2000].

PBDs [Gould and Valle, 2000]. A major step was the application Peroxisomal abnormalities are only found in the rhizomelic
of complementation analysis to the study of the genetic basis of type. This type is clinically characterized by a disproportion-
PBDs. A first study [Brul et al., 1988] performed with ally short stature primarily affecting the proximal parts of the
fibroblasts from seven patients, revealed five different genetic extremities, facial features such as a broad nasal bridge, highly
complementation groups (CGs) indicative of marked genetic arched palate, dysplastic ears, and micrognathia, congenital
heterogeneity. A similar conclusion was reached in subsequent contractures, and severe mental retardation with spasticity.
studies [Roscher et al., 1989; Yajima et al., 1992]. A col- Roentgenological studies have shown symmetrical shortening
laborative study showed that the CGs identified in the three of femur and humerus with irregular and broad metaphysics,
centres could be assigned to nine unique CGs [Shimozawa calcific stippling mainly of the epiphyses, absent femur head
et al., 1993], to which in subsequent years three additional nucleus, coronal clefts of vertebrae, increased intravertebral
groups have been added. In 11 of the 12 GCs, the gene in- disc spaces, cupping of dorsal ribs, and a barrel-formed thorax.
volved has been identified [Gould and Valle, 2000], and the Review of unpublished cases from our own center and patients
PTS1- and PTS2 import routes as well as the phenotypes described in literature shows that calcific stippling of the
associated with each CG are listed in Table II. One group, epiphyses and periarticular areas is present in all patients
number 12, is different from the 11 other groups in terms of its below 18 months of age and disappears in older patients. The
associated cell biology (only the PTS2 pathway is defective). femoral heads remain severely deformed and all patients also
Furthermore, this group is associated with a single peroxiso- lack a clear nucleus of the femur head. No coronal cleft was
mal disorder, i.e., rhizomelic chondro-dysplasia punctata noted in the majority of patients beyond 3 years of age. In most
(RCDP). We have performed complementation studies now in cases the spinal column showed irregular and/or small discs
301 PBD-patients of which all 80 RCDP Type 1 patients with increased distances between the vertebrae. In patients
belonged to group 12 and had a mutation in PEX7. The beyond 10 years of age, radiographs of the chest show signs of
remaining 221 patients, with phenotypes ranging from ZS to chronic infections and a barrel-shaped thorax with severe
IRD, were found to belong to the 11 remaining CGs: major torsion scoliosis.
groups were 139 (63%) in the PEX1 group, 22 (10%) in the Biochemical and molecular basis of RCDP type 1. The
PEX6-group and 12 (5.4%) in the PEX12-group (Gootjes and identification of RCDP as a peroxisomal disorder dates back
Wanders, unpublished). to when Heymans et al. [1985] reported that erythrocyte
Mutation analysis has shown two common mutations in the plasmalogen levels were markedly deficient in RCDP patients.
PEX1 gene in patients of the PEX1 group, including a missense Phytanic acid was also strongly elevated suggesting the
mutation (G843D) [Portsteffen et al., 1997; Reuber et al., 1997] involvement of two different peroxisomal pathways. Later
which causes a partial dysfunction of Pex1p, and a 1 bp in- studies led to the identification of distinct peroxisomal
sertion (C.2097insT) [Maxwell et al., 1999] which causes a abnormalities including deficiencies in: (1) alkyl-DHAP
complete loss of function of Pex1p. Most other mutations were synthase; (2) phytanoyl-CoA hydroxylase; (3) DHAPAT (par-
private. Interestingly, patients homozygous for the G843D tial deficiency); and (4) mature 41 kDa thiolase (only the
mutation invariably show a mild phenotype, usually of the precursor 44 kDa form is present). This perplexing combina-
IRD-type, whereas patients homozygous for C.2097insT show tion of deficiencies was resolved in 1997 when independently
the severe Zellweger-type. three groups found PEX7 as the defective gene. A loss of Pex7p
function blocks the PTS2 import pathway, and explains the
deficiency of the enzymes. The reduced activity of DHAPAT
Rhizomelic Chondrodysplasia Punctata (RCDP)
is the secondary consequence of the absence of alkyl-DHAP
Type 1 (MIM 215100)
synthase [de Vet et al., 1999]. RCDP is genetically hetero-
Chondrodysplasia punctata (CDP) refers to a heterogeneous geneous (see below). However, in all RCDP patients identified
group of bone dysplasias of which stippling of the epiphyses in by us until now, erythrocyte plasmalogen levels have been
infancy is the common feature. Two major types are the found deficient making plasmalogen analysis a highly reliable
Conradi-Hünermann type (McKusick 118650) with autosomal start for diagnosis.
dominant inheritance and the autosomal recessive rhizomelic Molecular studies have identified many different mutations
type (McKusick 215100). In addition, X-linked recessive [Braverman et al., 2002; Motley et al., 2002], although a
(McKusick 302950) and X-linked dominant (lethal in males) single mutation, L292ter, accounts for about half of the
forms of chondrodysplasia punctata have been identified. mutant PEX7 genes in RCDP probands. Haplotype analysis
366 Wanders

has shown in every case that this mutation is on the same Mutation analysis of the ABCD1-gene has shown many
background, strongly suggesting a founder effect in Northern different, often private mutations [Kemp et al., 2001], which
Europeans. can be studied in the mutation database for X-ALD (http://
www.XALD.nl).
SINGLE PEROXISOMAL ENZYME DEFICIENCIES
Acyl-CoA Oxidase Deficiency
The Disorders of Peroxisomal Beta Oxidation
(Pseudo NALD) (MIM 264470)
X-linked adrenoleucodystrophy (MIM 300100). X-
Acyl-CoA oxidase deficiency was first described [Poll-Thé
linked adrenoleucodystrophy (X-ALD) is the most common
et al., 1988] in two siblings with neonatal hypotonia, severe
single peroxisomal disorder with a minimum incidence of
delayed motor development, sensory deafness, and retinopa-
1:21,000 males in the USA [Bezman et al., 2001] to 1:15,000
thy, without dysmorphisms. Both patients showed some initial
males in France (Aubourg, cited in Kemp et al., 2001). The
progress in the first 2 years of life followed by a progressive
phenotype varies widely. At present, at least six phenotypic
neurological regression, including progressive hypodensity of
variants can be distinguished [Moser et al., 2000]. This
cerebral white matter in neuroimaging. This led to the presum-
classification is somewhat arbitrary and based on age of onset
ptive diagnosis neonatal adrenoleucodystrophy (NALD), and
and the organs principally involved. The two most frequent
elevated plasma VLCFA levels were found. However, in
phenotypes are childhood cerebral ALD (CCALD) and adreno-
contrast to NALD [Aubourg et al., 1986] levels of the bile acid
myeloneuropathy (AMN). Onset of CCALD is between 3 and
intermediates, phytanic acid, pristanic acid, and pipecolic acid
10 years of age with progressive behavioral, cognitive,
were normal. Furthermore, peroxisomes were found to be
and neurologic deterioration, often leading to total disability
present in liver biopsy specimens although they were enlarged
within 3 years. The cerebral phenotype is not only observed in
in size and decreased in number. These patients were found to
childhood but may also present later in life in adolescence
be deficient for acyl-CoA oxidase 1 (ACOX1), the first enzyme
(adolescent cerebral ALD) or adulthood (adult cerebral ALD).
involved in the b-oxidation of VLCFA. This explains the nor-
There is a marked difference between the cerebral phenotypes
mal presence of all peroxisomal metabolites except VLCFAs
and AMN since the cerebral phenotypes show an inflammatory
(Fig. 3). In a second family [Wanders et al., 1990], the index
reaction in the cerebral white matter which resembles what
patient showed generalized hypotonia, failure to thrive, deaf-
is observed in multiple sclerosis. In contrast to CCALD, the
ness, hepatomegaly, psychomotor retardation, absent reflexes,
inflammatory response is absent or mild in AMN which has a
frequent convulsions, and demise at 3 11/12 year. Two ad-
much later age of onset (28  9 years) and a much slower rate of
ditional cases [Suzuki et al., 1994] had profound hypotonia,
progression. Nevertheless approximately 40% of AMN
dysmorphic features (hypertelorism, low nasal bridge, low-set
patients do develop some cerebral involvement associated
ears, and polydactyly), and initial moderate developmental
with a more rapid downhill progression. It is important to
delay with later motor regression. Subsequently, three similar
emphasize that approximately 40% of women heterozygous for
cases were identified [Watkins et al., 1995].
XALD develop AMN-like symptoms in middle age or later.
Plasma abnormalities in acyl-CoA deficiency are restricted
Cerebral involvement and adrenocortical insufficiency are
to the accumulation of VLCFAs (Fig. 3). There is only a single
rare, however.
report in literature on the molecular basis of acyl-CoA oxidase
The biochemical hallmark of X-ALD is the accumulation of
deficiency [Fournier et al., 1994] showing a large deletion in the
VLCFAs in plasma and different cell types. Analysis of plasma
acyl-CoA oxidase 1 gene in the original patients described by
VLCFAs has proved to be a powerful diagnostic method with
Poll-Thé et al. [1988].
very few if any false negatives [Moser et al., 1999]. In 15–20%
of obligate heterozygotes, however, test results are falsely
D-Bifunctional Protein Deficiency
negative which makes mutation analysis the only reliable test
for heterozygote identification. Bifunctional protein deficiency was first described in a
The accumulation of VLCFAs is due to the defective male with severe hypotonia and therapy refractory seizures
oxidation of VLCFAs in peroxisomes. Indeed, when C26:0 [Watkins et al., 1989]. No developmental progress was ob-
beta-oxidation is studied in fibroblasts, the rate of oxidation in served. There was no hepatomegaly, fontanelles were large
X-ALD fibroblasts is 20–30% of control compared to a virtually with open metopic sutures, visual evoked responses and
complete deficiency in ZS fibroblasts. The X-ALD-gene was brainstem auditory evoked responses were grossly abnormal,
identified in 1993 using a positional cloning strategy [Mosser and brain biopsy at 6 weeks showed polymicrogyria and
et al., 1993]. The protein proved to belong to the superfamily of heterotopias. The patient died at 5 months of age. Autopsy
ABC transmembrane transporter proteins. Functionally, showed minute glomerular cysts in the kidneys, adrenal
eukaryotic ABC transporters contain two homologous halves, atrophy with ballooned, lipid laden cells, and some portal
each with a transmembrane domain (TMD), and an ATP fibrosis in the liver. Based on the clinical features, neonatal
binding domain (ATP). Most functional ABC transporters have ALD was considered. In plasma, elevated levels of VLCFA
the following topology: TMD–ATP–TMD–ATP. In some, the and bile acid intermediates levels were found, and there were
entire transporter is encoded by a single gene, in others two abundant peroxisomes in a liver biopsy. Immunoblot studies
ABC half-transporters homodimerize to generate a functional subsequently revealed the absence of L-bifunctional protein
ABC transporter. ALDP with one membrane domain and one (LBP) in postmortem liver material. Many additional patients
ATP binding domain has the structure of an ABC half- have been described in the literature: data on 44 patients
transporter. [Wanders et al., 2001a] indicated that children show severe
Three additional mammalian peroxisomal ABC half-trans- central nervous system involvement with profound hypotonia
porters have been identified, closely related by nucleic acid (98%), uncontrolled seizures (95%), and failure to acquire any
and protein sequence: ALDRP; PMP70; and PMP69 or significant development (100%). Children are usually full term
PMP70R. These four transporters may form different hetero- and show no evidence of intrauterine growth retardation.
dimeric combinations in the peroxisomal membrane and are Dysmorphic features include macrocephaly, high forehead,
probably involved in the transport of different metabolites. flat nasal bridge, low-set ears, large open fontanelles, and
Although definitive evidence is lacking, ALDP, as homodimer micrognathia. Abnormal neuronal migration (88%) as poly-
or as heterodimer, seems to transport VLCFA across the microgyria, heterotopia, and other symptoms were found in
peroxisomal membrane either as free acid or as CoA-ester. cerebrum and cerebellum [Kaufmann et al., 1996].
Peroxisomal Disorders 367

Two studies [Suzuki et al., 1997; van Grunsven et al., 1998] heredopathia-atactica-polyneuritiformis. Cardinal manifes-
independently identified the deficiency of D-bifunctional pro- tations include retinitis pigmentosa, cerebellar ataxia, chronic
tein (D-BP). D-BP is involved in the peroxisomal b-oxidation of polyneuropathy, and an elevated CSF protein level. Less
all known peroxisomal substrates including VLCFAs, DHCA, constant features include sensory neural hearing loss, anos-
and THCA, and pristanic acid (Fig. 2) which explains the mia, ichtyosis, skeletal malformations, and cardiac abnormal-
accumulation of all metabolites in most patients. The group ities. The clinical picture of Refsum disease is often that of
of bifunctional protein deficiency is heterogeneous consisting a slowly developing, progressive peripheral neuropathy man-
of three subgroups, explained by the two catalytic activities ifested by severe motor weakness and muscular wasting,
(a hydratase and 3-hydroxyacyl-CoA dehydrogenase activity) especially of the lower extremities. Onset has occasionally
of the protein [Watkins et al., 1989]. Accordingly, in subgroup been detected in early childhood, is observed most before
A D-BP is completely absent causing loss of both activities 20 years, and can be in the 5th decade in others [Wanders et al.,
[van Grunsven et al., 1999a,b]. Subgroup B represents D-BP 2001b].
enoyl-CoA hydratase deficiency and in subgroup C D-3- The accumulation of phytanic acid in Refsum patients was
hydroxyacyl-CoA dehydrogenase is functionally inactive discovered in the early 1960s [Wanders et al., 2001b], but the
[van Grunsven et al., 1998]. Molecular studies have identified enzyme deficiency of phytanoyl-CoA hydroxylase [Jansen et al.,
a variety of mutations including a rather frequent mutation 1997], and mutation analysis [Jansen et al., 2000] were more
found in patients belonging to subgroup C [Wanders et al., recent. Studies have shown that in a subset of patients the
2001a]. defect is not in phytanoyl-CoA hydroxylase but involves PEX7
[Braverman et al., 2002; van den Brink et al., 2003].
Peroxisomal Thiolase Deficiency
(Pseudo Zellweger Syndrome) (MIM 261510)
DISORDERS OF ETHERPHOSPHOLIPID
In 1986, a girl with ‘pseudo-ZS’ was reported [Goldfischer BIOSYNTHESIS
et al., 1986] showing marked facial dysmorphism, muscle
weakness, hypotonia, no psychomotor development, and Rhizomelic Chondrodysplasia Punctata (RCDP)
demise at 11 months. At autopsy, the patient had renal cysts, Type 2 DHAPAT-Deficiency) (MIM 222765)
atrophic adrenals with striated cells, minor or little fibrosis, RCDP was first found to be genetically heterogeneous in
hypomyelination in the cerebral white matter, foci of neuronal 1992 when isolated DHAPAT-deficiency was described in a
heterotopia, and a sudanophilic leucodystrophy. Plasma patient showing symptoms of RCDP [Wanders et al., 1992]. All
analysis showed elevated VLCFAs, but peroxisomes were craniofacial abnormalities were present, he was extremely
found to be abundantly present in the liver. Subsequent hypotonic, had cataracts, and pronounced rhizomelic short-
immunoblot analysis indicated selective deficiency of perox- ening especially of upper limbs. Radiologically all abnormal-
isomal thiolase suggesting peroxisomal thiolase deficiency. ities consistent with RCDP were seen except for stippled
However, re-investigation of this patient [Ferdinandusse et al., calcifications of patellae and acetabulum. Deficient erythro-
2002] disclosed that the true defect in this patient is not at the cyte plasmalogens but normal phytanic acid level were found.
level of peroxisomal thiolase but in the D-BP. As a consequence, Fibroblast studies showed isolated DHAPAT-deficiency with
peroxisomal thiolase deficiency can no longer be considered a normal values for alkyl DHAP synthase, phytanic acid a-
distinct peroxisomal disorder. oxidation, and peroxisomal thiolase 1 [Wanders et al., 1992]. A
second patient [Barr et al., 1993] also had the severe
Peroxisomal 2-Methylacyl-CoA Racemase phenotype, but two sibs [Clayton et al., 1994] who have another
(AMACR) Deficiency affected sib at follow-up [Sztriha et al., 1997] were less severely
affected, and a mild case has been reported [Moser et al., 1995;
A new defect in the peroxisomal fatty acid beta-oxidation
Elias et al., 1998] who lacked rhizomelia and craniofacial
pathway in patients suffering from an adult-onset sensory
dysmorphism.
motor neuropathy was recently identified [Ferdinandusse
In all patients with RCDP type 2, DHAPAT activity is
et al., 2000]. Sensory motor neuropathy may occur in X-linked
severely deficient, which is associated with the inability to
adrenoleucodystrophy and Refsum disease, resulting from
synthesize ether-phospholipids including plasmalogens.
accumulation of VLCFA and phytanic acid, respectively.
This is reflected in deficient plasmalogen levels in all tissues
Analysis of two patients with adult onset sensory motor
including erythrocytes. In all patients studied so far, erythro-
neuropathy and additional signs suggesting Refsum dis-
cyte plasmalogen levels have been clearly deficient. The
ease and X-linked adrenoleucodystrophy showed normal
identification of the human DHAPAT cDNA [Thai et al.,
VLCFAs, marginally elevated phytanic acid, and definitely
1997] allowed molecular studies showing a variety of muta-
increased levels of the 2-methyl branched chain fatty acids,
tions [Ofman et al., 1998]. In severely affected cases [Wanders
pristanic acid and di- and trihydroxycholestanoic acid
et al., 1992; Barr et al., 1993], we have found mutations (848 TT
[Ferdinandusse et al., 2000]. This suggested a specific defect
insertion and 567del204bp deletion, respectively) that cause
in the peroxisomal beta-oxidation of branched-chain fatty
complete absence of DHAPAT activity, in mildly affected pa-
acids. Fibroblast studies showed complete deficiency of
tients [Clayton et al., 1994; Sztriha et al., 1997] a homozygous
racemase activity (AMACR). This enzyme is not directly
632G!A mutation was found, which gave some residual
involved in beta-oxidation itself but is important in beta-
activity in DHAPAT-activity. Therefore, there may be some
oxidation of both pristanic acid and di- and trihydroxychol-
phenotype/genotype correlation.
estanoic acid. Subsequent molecular studies led to the
identification of mutations in the gene coding for AMACR
[Ferdinandusse et al., 2000]. Rhizomelic Chondrodysplasia Punctata
(RCDP) Type 3 (Alkyl DHAP Synthase
Deficiency) (MIM 600121)
DISORDERS OF FATTY ACID a-OXIDATION
A first case of isolated alkyl DHAP synthase deficiency was
Refsum Disease (MIM 266500)
found in a patient with classical RCDP, completely deficient
Refsum disease was first delineated as a distinct entity erythrocyte plasmalogen levels, and normal plasma phytanic
clinically by Sigvald Refsum in the 1940s under the name acid levels [Wanders et al., 1994]. Fibroblast studies indicated
368 Wanders

absent alkyl DHAP synthase and normal DHAPAT, phytanoyl- to this last group, AGT is mistargeted to the mitochondria
CoA hydroxylase, and peroxisomal thiolase. Three unpub- where the enzyme may well be active but incapable to detoxify
lished patients have been identified by Dr. H.W. Moser the glyoxylate produced in the peroxisome, and, thus, patients
(Baltimore), who also show classical RCDP with early demise. with relatively high residual activities still present with a
Another patient [de Vet et al., 1999] is less affected and is still severe form of hyperoxaluria Type 1 [Danpure et al., 1993].
alive at 7 years, showing mild rhizomelia, generalized con- The identification of the cDNA [Takada et al., 1990] and
tractures, inability to sit or crawl, cataract, seizures, and the resolution of the exon/intron structure of the AGT gene
profound developmental delay. [Purdue et al., 1991] has allowed molecular studies which
In the original patient [Wanders et al., 1994], alkyl DHAP have shown a series of different mutations [Danpure and
synthase was fully deficient, but in a subsequent patient [de Purdue, 1995].
Vet et al., 1999] not only alkyl DHAP synthase was deficient
but also DHAPAT activity was low (15%). Complementation
studies indicated that the patient did belong to RCDP group 3. Glutaric Aciduria Type 3 (MIM 231690)
Antibodies against the alkyl DHAP synthase [de Vet et al., A single patient was described at 11 months of age because of
1997] showed that alkyl DHAP synthase and DHAPAT form a failure to thrive and postprandial vomiting [Bennett et al.,
complex within the peroxisome [Biermann et al., 1999a,b]. 1991]. She was found to be homozygous for b-thalassemia but
Detailed further studies [de Vet et al., 1997, 1998, 1999; also had significant glutaric aciduria. Fibroblasts showed
Biermann et al., 1999a,b] have shown that the deficient normal glutaryl-CoA dehydrogenase activity and glutaryl-CoA
activity of DHAPAT in the De Vet case is secondary to the oxidase activity was not detectable. This study has not been
complete absence of the alkyl DHAP synthase protein, and in followed up at the protein or DNA level. No additional cases
the Wanders patient the protein is normally expressed and have been identified so far.
localized in peroxisomes but functionally inactive (due to a
G1256A mutation). As DHAPAT is stable only in a complex
with alkyl-DHAP synthase within the peroxisome (Fig. 4), Mevalonate Kinase Deficiency
DHAPAT activity is normal in the Wanders patient but (MIM 251170 and MIM 260920)
deficient in the De Vet patient [Ofman et al., 1998, 1999]. Mevalonate kinase deficiency is the only disorder involv-
ing the peroxisomal part of isoprenoid biosynthesis. There
Hyperoxaluria Type 1 (Alanine Glyoxylate is the classical form of mevalonate kinase deficiency (MIM
Aminotransferase Deficiency) (MIM 259900) 251170) associated with profound developmental delay, facial
dysmorphism, cataract, hepatosplenomegaly, lymphade-
Primary hyperoxaluria describes two rare disorders char- nopathy, and early death [Hoffmann et al., 1993], and
acterized by recurrent calcium oxalate nephrolithiasis and mevalonate kinase deficiency has been observed in hyperim-
nephrocalcinosis, frequently leading to progressive renal in- munoglobulinaemia D and periodic fever syndrome (HIDS;
sufficiency and death before the age of 20 years. In both forms, MIM 260920) [Drenth et al., 1999; Houten et al., 1999a,b]
there is excessive synthesis and excretion of oxalate. In type 1 showing recurrent episodes of fever associated with lympha-
(glycolic aciduria), excessive amounts of glyoxylic and glycolic denopathy, arthralgia, gastrointestinal dismay, and skin
acids are also found in the urine, in type II (L-glyceric aciduria), rash. In HIDS patients, mevalonate kinase was found to
large amounts of L-glyceric acid but normal amounts of be strongly reduced (1.2–3.4% of normal) but not fully
glyoxylic and glycolic acid are excreted. The enzymatic basis deficient as in classical mevalonate kinase deficiency [Drenth
in the two types involves the peroxisomal enzyme alanine: et al., 1999; Houten et al., 1999a,b]. Laboratory diagnosis
glyoxylate aminotransferase in PH1 and the cytosolic enzyme involves measurement of mevalonic acid in urine, mevalonate
D-glycerate dehydrogenase/glyoxylate reductase in PH2,
kinase activity measurements in leukocytes, platelets, or
respectively. fibroblasts, and eventually molecular analysis [Waterham,
PH1 is clinically variable both in symptoms and timing. In 2002].
most patients, first symptoms start before 5 years of age. There
is an enormous spread, however, in the ages at which the
disease becomes apparent, which can be as early as the first Acatalasemia (MIM 115500)
year of life or may happen much later in life even beyond the
fifth decade. Deposition of calcium oxalate within the kidney Acatalasemia is a rare disease which has mainly been
parenchymatous tissue (nephrocalcinosis) or the renal pelvis/ described in Japan and Switzerland. In Japan, acatalasaemia
urinary tract (urolithiasis) continues until eventually renal is associated with ulcerating, often gangrenous, oral lesions
function is impaired causing subsequent sequelae like uremia whereas these abnormalities were not seen in Swiss patients
and systemic oxalosis. The failure to clear oxalate from the [Eaton and Mouchou, 1995].
body leads to its further deposition in almost all areas of the
body affecting multiple tissues in addition to the kidneys and
Mulibrey Nanism (MIM 253250)
urinary tract including the myocardium with heart block,
myocarditis, and cardioembolic stroke, the nerves (peripheral Muscle-liver-brain-eye (mulibrey) nanism (MUL) is an
neuropathy), bone (bone pain, multiple fractures, osteosclero- autosomal recessive disorder with severe growth failure of
sis), the eyes (retinopathy, optic atrophy), bone marrow, and perinatal onset, characteristic dysmorphic features, pericar-
other organs [Danpure and Purdue, 1995]. dial constriction, and hepatomegaly. Other common features
The primary defect of PH1 is a mutation in the alanine include J-shaped sella turcica, yellowish dots in the ocular
glyoxylate aminotransferase gene. As a consequence, perox- fundi, slight muscular weakness, cutaneous naevi flammei,
isomal glyoxylate accumulates and glyoxylate is either oxidiz- and enlargement of cerebral ventricles. Psychomotor develop-
ed to oxalate, which precipitates as calcium oxalate or is ment of patients with Mulibrey nanism is within normal limits.
reduced to glycolate (Fig. 6). Definitive diagnosis requires a Mulibrey nanism occurs world-wide but is most common in the
liver biopsy since the enzyme is only expressed in liver. In most Finnish population. Mutations in the TRIM37 gene (previously
patients (60%), there is virtually no residual AGT activity but designated MUL), a RING-B-box-coiled-coil subfamily of zinc-
in the remainder, activities are between 2 and 48% of controls finger proteins, on chromosome 17q22-23, underlie Mulibrey
[Danpure and Purdue, 1995]. In most of the patients belonging nanism [Avela et al., 2000]. Recent studies [Kallijarvi et al.,
Peroxisomal Disorders 369

2002] have shown that the TRIM37 protein is localized in III. Rhizomelic chondrodysplasia punctata. This includes all
peroxisomes. The possibility that the disorder may be a types.
peroxisomal disorder was considered earlier [Schutgens et al., III. X-linked adrenoleucodystrophy complex. This includes all
1994], although normal plasma VLCFA and normal peroxiso- types.
mal functions in fibroblasts were found. It seems that TRIM37 IV. The remaining peroxisomal disorders. This includes
is not involved in one of the known peroxisomal functions Refsum disease, 2-methylacyl-CoA racemase (AMACR)
and does not produce any abnormalities similar to other deficiency, hyperoxaluria type 1, glutaryl-CoA oxidase
peroxisomal disorders and its true function remains to be deficiency, mevalonate kinase deficiency, acatalasaemia,
established. and Mulibrey nanism.

LABORATORY DIAGNOSIS OF Laboratory Diagnosis of the Disorders


PEROXISOMAL DISORDERS of Diagnostic Group I
No single laboratory test is capable of identifying all The similarity in clinical presentation of these two groups
peroxisomal disorders. The selection of laboratory analyses of disorders dictate the grouping (Table III). VLCFA are
should be based on the clinical presentation of the patient abnormal in all disorders listed: we have now studied over
involved. We have introduced the concept of diagnostic groups >500 patients with either a defect in peroxisome biogenesis or
[Wanders et al., 1996] in order to develop logical guidelines a peroxisomal beta-oxidation disorder, and VLCFA-levels have
for the laboratory diagnosis of the various peroxisomal always been abnormal. Normal VLCFA-levels rule out a PBD
disorders. Although not further specified here prenatal diag- or peroxisomal beta-oxidation defect. If VLCFAs are abnormal,
nostic laboratory tests can be done in virtually all peroxisomal additional tests have to be performed for further discrimina-
disorders. tion. This includes analysis in erythrocytes (plasmalogens) and
These diagnostic groups are: plasma (bile acid intermediates, pristanic acid, and phytanic
acid). If erythrocytes are available and plasmalogen levels are
III. the disorders of peroxisome biogenesis and of the per- deficient, the patient definitely suffers from a ‘‘disorder of
oxisomal beta-oxidation. This group include ZS, NALD, peroxisome biogenesis’’. This should be followed by detailed
IRD, acyl-CoA oxidase deficiency, and D-bifunction pro- studies in fibroblasts, to establish whether the defect is ex-
tein deficiency, but not X-ALD and AMACR-deficiency. pressed in fibroblasts, for complementation studies, and to

TABLE III. Biochemical Characteristics of Different Peroxisomal Disorders

Diagnostic group

1 2 3

RCDP RCDP RCDP


ZS NALD/IRD AOXD DBPD type 1 type 2 type 3 X-ALD

Plasma
Very-long chain fatty acids " " " " N N N "
Di- and trihydroxy " " N "a N N N N
cholestanoic acid
Phytanic acid N-"b N-"b N N-"b,d N-"b N N N
Pristanic acid N-"c N-"c N N-"c N N N N
Erythrocytes
Plasmalogen level # N N N # # # N
Liver
Peroxisomes Deficient Deficient Present but Present but Present Present Present Present
abnormal abnormal
Fibroblasts
Plasmalogen synthesis # # N N # # # N
DHAPAT # # N N # # #e N
Alkyl DHAP synthase # # N N # N # N
C26:0 b-oxidation # # # # N N N #
Pristanic acid b-oxidation # # N # N N N N
Acyl-CoA oxidase 1 # # # N N N N N
D-bifunctional protein # # N # N N N N
Phytanic acid a-oxidation # # N N # N N N
Phytanoyl CoA hydroxylase # # N N # N N N
Peroxisomes Deficient Deficient Present but Present but Present Present Present Present
abnormal abnormal
Abbreviations: ZS, Zellweger syndrome; NALD, neonatal adrenoleukodystrophy; IRD, infantile Refsum disease; AOXO, acyl-CoA oxidase 1 deficiency;
DBPD, D-bifunctional protein deficiency; RCDP, rhizomelic chondrodysplasia punctata.
a
Di- and trihydroxycholestanoic acid are not elevated in all DBPD-patients (see [van Grunsven et al., 1999a,b]).
b
Phytanic acid is derived from dietary sources only and may, therefore, vary from normal to elevated in patients in whom phytanic acid a-oxidation is
deficient.
c
Pristanic acid is derived from dietary sources only either directly or indirectly from phytanic acid via a-oxidation and may, therefore, vary from normal to
elevated if pristanic acid b-oxidation is deficient.
d
Phytanic acid is often elevated if pristanic acid b-oxidation is impaired even if phytanic acid a-oxidation per se is normal.
e
In all patients with RCDP type 3, except one [Wanders et al., 1994], DHAPAT is deficient as a consequence of its instability in the absence of alkyl DHAP
synthase (see text).
370 Wanders

Fig. 7. Flow chart for the differential diagnosis of patients suffering from a peroxisome biogenesis disorder (PBD) or a peroxisomal b-oxidation disorder
(POD).
Peroxisomal Disorders 371

Fig. 8. Flow chart for the differential diagnosis of rhizomelic chondrodysplasia punctata (RCDP) type 1, 2, and 3.

establish the gene defective in this patient. If plasmalogen the milder disorders of peroxisomal biogenesis (mainly IRD)
levels are normal, a disorder of peroxisomal beta-oxidation can [Wanders et al., 1986]. In such cases, the question whether one
be expected (Fig. 7). However, the erythrocyte plasmalogen is dealing with a disorder of peroxisomal b-oxidation or a
levels may be completely normal in patients affected by one of disorder of peroxisome biogenesis, can only be resolved via
372 Wanders

detailed studies in fibroblasts (Fig. 7). Furthermore, identifica- of all three acids. Definitive diagnosis of PH1 requires a liver
tion of the precise defect in the peroxisomal b-oxidation biopsy for assessment of AGT-activity.
pathway can also be resolved through studies in fibroblasts Glutaric aciduria type 3 (glutaryl-CoA oxidase defi-
only (Fig. 7). ciency). In the single patient with this defect there was
increased urinary excretion of glutaric acid not due to glutaric
Laboratory Diagnosis of the Disorders aciduria type 1 or type 2. Glutaryl-CoA oxidase was subse-
of Diagnostic Group II quently found to be deficient.
Mevalonate kinase deficiency (MK-deficiency). In the
The clinical similarities of the three RCDP types warrants classic form of MK-deficiency, urinary mevalonic acid is
inclusion into a single diagnostic group. The biochemical extremely high in all patients. If abnormal, mevalonate kinase
characteristics of these disorders are listed in Table III. activity should be measured in blood cells (leukocytes, lym-
Erythrocyte plasmalogens are deficient in each type, so this phocytes, thrombocytes) or in fibroblasts, followed by molecu-
is a reliable initial laboratory test. Our experience in >100 lar studies [Houten et al., 1999a,b]. In hyper IgD-periodic fever
RCDP-patients shows that erythrocyte plasmalogen levels syndrome (HITS), mevalonate kinase is also deficient, but
were always deficient even in milder affected cases as in the urinary mevalonic acid may be completely normal, which
patient described by Smeitink et al. [1992]. As shown in the implies that diagnosis should be based on direct enzyme
flowchart of Figure 8, detailed fibroblast studies are required analysis in white blood cells, platelets, and/or fibroblasts.
for further discrimination. Only in Type 1 also other perox- Peroxisomal 2-methyl-CoA racemase (AMACR) defi-
isomal functions including phytanic acid alpha-oxidation are ciency. Patients with a deficiency of AMACR are unable to
impaired. Furthermore, peroxisomal thiolase is in its 44 kDa degrade pristanic acid and the bile acid intermediates. Plasma
precursor form in RCDP type 1 but in its mature 41 kDa form in level studies should, therefore, include analysis of pristanic
types 2 and 3. Molecular analysis of the PEX7 gene has to follow acid by GC-MS or bile acid intermediates preferably by
[Braverman et al., 1997, 2002; Motley et al., 1997, 2002; tandem-MS. If abnormal, fibroblast studies, including mea-
Purdue et al., 1997]. Discrimination between types 2 and 3 surements of racemase activity, and molecular studies should
requires measurement of both DHAPAT and alkylDHAP be performed [Ferdinandusse et al., 2000].
synthase in fibroblasts homogenates, followed by molecular
analysis [Wanders et al., 2001a].
ACKNOWLEDGMENTS
Laboratory Diagnosis Of Diagnostic Group The author gratefully acknowledges Mrs. Maddy Festen, Iet
III (X-Linked Adrenoleucodystrophy) van der Gracht, and Susan Gersen-van Zadel for expert pre-
Studies in hundreds of patients have shown that analysis of paration of the manuscript and Mr. Jos Ruiter for preparation
plasma VLCFA is a reliable initial test to verify whether a of the figures.
patient is affected by X-ALD. If abnormal, one may proceed
with fibroblasts studies and molecular analysis. Fibroblasts REFERENCES
studies, however, are not obligatory and direct molecular
studies in blood cells can be performed too. Aubourg P, Robain O, Rocchiccioli F, Dancea S, Scotto J. 1985. The cerebro-
hepato-renal (Zellweger) syndrome: Lamellar lipid profiles in adreno-
Heterozygote detection is not as straightforward. Plasma cortical, hepatic mesenchymal, astrocyte cells, and increased levels of
VLCFA-levels have been found to be normal in about 15% very long chain fatty acids and phytanic acid in the plasma. J Neurol Sci
of obligate heterozygotes. We advocate to perform direct 69:9–25.
molecular studies and omit VLCFA-analysis in families in Aubourg P, Scotto J, Rocchiccioli F, Feldmann-Pautrat D, Robain O. 1986.
which a molecular defect has been established. In case the Neonatal adrenoleukodystrophy. J Neurol Neurosurg Psych 49:77–86.
family history is negative, first plasma VLCFA-analysis can Avela K, Lipsanen-Nyman M, Idanheimo N, Seemanova E, Rosengren S,
be performed, if needed followed by fibroblast studies includ- Makela TP, Perheentupa J, Chapelle AD, Lehesjoki AE. 2000. Gene
ing immunofluorescence using antibodies against the ALD- encoding a new RING-B-box-coiled-coil protein is mutated in mulibrey
protein. This method may especially be rewarding since the nanism. Nat Genet 25:298–301.
product of the mutant X-ALD allele often produces an unstable Barr DG, Kirk JM, al Howasi M, Wanders RJA, Schutgens RBH. 1993.
protein so that in heterozygotes a mosaic pattern is observed Rhizomelic chondrodysplasia punctata with isolated DHAP-AT defi-
ciency. Arch Dis Child 68:415–417.
[Kemp et al., 2001].
Barth PG, Gootjes J, Bode H, Vreken P, Majoie CB, Wanders RJA. 2001. Late
onset white matter disease in peroxisome biogenesis disorder. Neurology
Laboratory Diagnosis of Disorders of Group IV 57:1949–1955.
Bennett MJ, Pollitt RJ, Goodman SI, Hale DE, Vamecq J. 1991. Atypical
Disorders of group IV are clinically heterogeneous and all riboflavin-responsive glutaric aciduria, and deficient peroxisomal
require separate laboratory test for diagnosis described below. glutaryl-CoA oxidase activity: A new peroxisomal disorder. J Inherit
Refsum disease. In most, but not all Refsum disease Metab Dis 14:165–173.
patients phytanoyl-CoA hydroxylase is deficient leading to the Bezman L, Moser AB, Raymond GV, Rinaldo P, Watkins PA, Smith KD,
impaired degradation of phytanic acid. As phytanic acid is Kass NE, Moser HW. 2001. Adrenoleukodystrophy: incidence, new
derived from dietary sources, plasma phytanic acid levels may mutation rate, and results of extended family screening. Ann Neurol 49:
vary widely. However, normal phytanic acid levels in proven 512–517.
Refsum patients are not reliably reported, and plasma phy- Biardi L, Krisans SK. 1996. Compartmentalization of cholesterol biosynth-
tanic acid analysis appears to be a reliable test for Refsum esis. Conversion of mevalonate to farnesyl diphosphate occurs in the
disease. Definite diagnosis requires measurement of phyta- peroxisomes. J Biol Chem 271:1784–1788.
noyl-CoA hydroxylase in fibroblasts and molecular analyses Biermann J, Gootjes J, Humbel BM, Dansen TB, Wanders RJA, van den
[Jansen et al., 2000]. Bosch H. 1999a. Immunological analyses of alkyl-dihydroxyacetonepho-
sphate synthase in human peroxisomal disorders. Eur J Cell Biol 78:
Primary hyperoxaluria type 1 (PH1). In patients with 339–348.
hyperoxaluria type 1, alanine glyoxylate-aminotransferase is
Biermann J, Just WW, Wanders RJA, van den Bosch H. 1999b. Alkyl-
deficient which leads to a block in glyoxylate detoxification, dihydroxyacetone phosphate synthase and dihydroxyacetone phosphate
and, hence, oxidation to oxalate or reduction to glycolate. In acyltransferase form a protein complex in peroxisomes. Eur J Biochem
most, but not all patients, there is increased urinary excretion 261:492–499.
Peroxisomal Disorders 373

Boltshauser E, Spycher MA, Steinmann B, Briner J, Isler W, Kuster T, deficiency: Identification of the true defect at the level of D-bifunctional
Poulos A, Pollard A. 1982. Infantile phytanic acid storage disease: A protein. Am J Hum Genet 70:1589–1593.
variant of Refsum disease? Eur J Pediatr 139:317–318. Fournier B, Saudubray JM, Benichou B, Lyonnet S, Munnich A, Clevers H,
Bowen P, Lee CSM, Zellweger H, Lindenberg R. 1964. A familial syndrome Poll-Thé BT. 1994. Large deletion of the peroxisomal acyl-CoA oxidase
of multiple congenital defects. Bull Johns Hopkins Hosp 114:402– gene in pseudoneonatal adrenoleukodystrophy. J Clin Invest 94:526–
414. 531.
Braverman N, Steel G, Obie C, Moser AB, Moser HW, Gould SJ, Valle D. Gatfield PD, Taller E, Hinton GG, Wallace AC, Abdelnour GM, Haust MD.
1997. Human PEX7 encodes the peroxisomal PTS2 receptor and is 1968. Hyperpipecolatemia: A new metabolic disorder associated with
responsible for rhizomelic chondrodysplasia punctata. Nat Genet 15: neuropathy and hepatomegaly: A case study. Can Med Assoc J 99:1215–
369–376. 1233.
Braverman N, Chen L, Lin P, Obie C, Steel G, Douglas P, Chakraborty PK, Goldfischer S, Collins J, Rapin I, Neumann P, Neglia W, Spiro AJ, Ishii T,
Clarke JT, Boneh A, Moser A, Moser H, Valle D. 2002. Mutation analysis Roels F, Vamecq J, Van Hoof F. 1986. Pseudo-Zellweger syndrome:
of PEX7 in 60 probands with rhizomelic chondrodysplasia punctata and Deficiencies in several peroxisomal oxidative activities. J Pediatr
functional correlations of genotype with phenotype. Hum Mutat 20:284– 108(1):25–32.
297. Gould SJ, Collins J, Rapin I, Neumann P, Neglia W, Spiro AJ, Ishii T, Roels F,
Brul S, Westerveld A, Strijland A, Wanders RJA, Schram AW, Heymans Vamecq J, Van Hoof F. 2000. Peroxisome biogenesis disorders: Genetics
HSA, Schutgens RBH, van den Bosch H, Tager JM. 1988. Genetic and cell biology. Trends Genet 16:340–345.
heterogeneity in the cerebrohepatorenal (Zellweger) syndrome and Heymans HSA. 1984. Clinical and biochemical consequences of peroxisomal
other inherited disorders with a generalized impairment of peroxisomal dysfunction. PhD thesis
functions. A study using complementation analysis. J Clin Invest 81:
1710–1715. Heymans HSA, Oorthuys JW, Nelck G, Wanders RJA, Schutgens RBH.
1985. Rhizomelic chondrodysplasia punctata: Another peroxisomal
Burton BK, Reed SP, Remy WT. 1981. Hyperpipecolic acidemia: Clinical and disorder. N Engl J Med 313:187–188.
biochemical observations in two male siblings. J Pediatr 99:729–734.
Hoffmann GF, Charpentier C, Mayatepek E, Mancini J, Leichsenring M,
Challa VR, Geisinger KR, Burton BK. 1983. Pathologic alterations in the Gibson KM, Divry P, Hrebicek M, Lehnert W, Sartor K, et al. 1993.
brain and liver in hyperpipecolic acidemia. J Neuropathol Exp Neurol Clinical and biochemical phenotype in 11 patients with mevalonic
42:627–638. aciduria. Pediatrics 91:915–921.
Clayton PT, Eckhardt S, Wilson J, Hall CM, Yousuf Y, Wanders RJA, Hogenboom S, Tuyp JM, Espeel M, Koster J, Wanders RJA, Waterham HR.
Schutgens RBH. 1994. Isolated dihydroxyacetonephosphate acyltrans- 2004. Mevalonate kinase is a cytosolic enzyme in humans. J Cell Sci
ferase deficiency presenting with developmental delay. J Inherit Metab 17:631–639.
Dis 17:533–540.
Houten SM, Kuis W, Duran M, de Koning TJ, van Royen-Kerkhof A, Romeijn
Danpure CJ, Purdue PE. 1995. The metabolic and molecular basis of GJ, Frenkel J, Dorland L, de Barse MM, Huijbers WA, Rijkers GT,
inherited disease. Primary hyperoxaluria. In: Scriver CR, Beaudet AL, Waterham HR, Wanders RJA, Poll-Thé BT. 1999a. Mutations in MVK,
Sly WS, Valle D, editors. McGraw-Hill. pp 2385–2426. encoding mevalonate kinase, cause hyperimmunoglobulinaemia D and
Danpure CJ, Purdue PE, Fryer P, Griffiths S, Allsop J, Lumb MJ, Guttridge periodic fever syndrome. Nat Genet 22:175–177.
KM, Jennings PR, Scheinman JI, Mauer SM. 1993. Enzymological and Houten SM, Romeijn GJ, Koster J, Gray RGF, Darbyshire P, Smit GPA, de
mutational analysis of a complex primary hyperoxaluria type 1 Klerk JBC, Duran M, Gibson KM, Wanders RJA, Waterham HR. 1999b.
phenotype involving alanine:glyoxylate aminotransferase peroxisome- Identification and characterization of three novel missense mutations
to-mitochondrion mistargeting and intraperoxisomal aggregation. Am in mevalonate kinase cDNA causing mevalonic aciduria, a disorder of
J Hum Genet 53:417–432. isoprene biosynthesis. Hum Mol Genet 8:1523–1528.
de Vet EC, Biermann J, van den Bosch H. 1997. Immunological localization Jansen GA, Wanders RJA, Watkins PA, Mihalik SJ. 1997. Phytanoyl-
and tissue distribution of alkyldihydroxyacetonephosphate synthase Coenzyme A hydroxylase deficiency—the enzyme defect in Refsum’s
and deficiency of the enzyme in peroxisomal disorders. Eur J Biochem disease. N Engl J Med 337:133–134.
247:511–517.
Jansen GA, Hogenhout EM, Ferdinandusse S, Waterham HR, Ofman R,
de Vet EC, IJlst L, Oostheim W, Wanders RJA, van den Bosch H. 1998. Alkyl- Jakobs C, Skjeldal OH, Wanders RJA. 2000. Human phytanoyl-CoA
dihydroxyacetonephosphate synthase: Fate in peroxisome biogenesis hydroxylase: resolution of the gene structure and the molecular basis of
disorders and identification of the point mutation underlying a single Refsum’s disease. Hum Mol Genet 9:1195–1200.
enzyme deficiency. J Biol Chem 273:10296–10301.
Kallijarvi J, Avela K, Lipsanen-Nyman M, Ulmanen I, Lehesjoki AE. 2002.
de Vet EC, IJlst L, Oostheim W, Dekker C, Moser HW, van den Bosch H, The TRIM37 gene encodes a peroxisomal RING-B-box-coiled-coil pro-
Wanders RJA. 1999. Ether lipid biosynthesis. Alkyl-dihydroxyacetone- tein: Classification of mulibrey nanism as a new peroxisomal disorder.
phosphate synthase protein deficiency leads to reduced dihydroxyace- Am J Hum Genet 70:1215–1228.
tonephosphate acyltransferase activities. J Lipid Res 40:1998–2003.
Kaufmann WE, Theda C, Naidu S, Watkins PA, Moser AB, Moser HW. 1996.
Drenth JP, Cuisset L, Grateau G, Vasseur C, van de Velde-Visser SD, de Neuronal migration abnormality in peroxisomal bifunctional enzyme
Jong JG, Beckmann JS, van der Meer JW, Delpech M. 1999. Mutations defect. Ann Neurol 39:268–271.
in the gene encoding mevalonate kinase cause hyper-IgD and periodic
fever syndrome. International Hyper-IgD Study Group. Nat Genet 22: Kelley RI, Datta NS, Dobyns WB, Hajra AK, Moser AB, Noetzel MJ, Zackai
178–181. EH, Moser HW. 1986. Neonatal adrenoleukodystrophy: New cases,
biochemical studies, and differentiation from Zellweger and related
Eaton JW, Mouchou Ma. 1995. Acatalasemia. In: Scriver CR, Sly AL, peroxisomal polydystrophy syndromes. Am J Med Genet 23:869–901.
Beaudet WS, Valle D, editors. The metabolic and molecular basis of
inherited disease. McGraw-Hill. pp 2371–2383. Kemp S, Pujol A, Waterham HR, van Geel BM, Boehm CD, Raymond GV,
Cutting GR, Wanders RJA, Moser HW. 2001. ABCD1 mutations and the
Elias ER, Mobassaleh M, Hajra AK, Moser AB. 1998. Developmental delay X-linked adrenoleukodystrophy mutation database: Role in diagnosis
and growth failure caused by a peroxisomal disorder, dihydroxyaceto- and clinical correlations. Hum Mutat 18:499–515.
nephosphate acyltransferase (DHAP-AT) deficiency. Am J Med Genet
80:223–226. Kerckaert I, Poll-Thé BT, Espeel M, Duran M, Roeleveld AB, Wanders RJA,
Roels F. 2000. Hepatic peroxisomes in isolated hyperpipecolic acidae-
Evrard P, Caviness VS Jr, Prats-Vinas J, Lyon G. 1978. The mechanism of mia: Evidence supporting its classification as a single peroxisomal
arrest of neuronal migration in the Zellweger malformation: An
enzyme deficiency. Virchow Arch 436:459–465.
hypothesis bases upon cytoarchitectonic analysis. Acta Neuropathol
41:109–117. Krisans SK. 1996. Cell compartmentalization of cholesterol biosynthesis.
Ann NY Acad Sci 804:142–164.
Ferdinandusse S, Denis S, Clayton PT, Graham A, Rees JE, Allen JT,
Mclean BN, Brown AY, Vreken P, Waterham HR, Wanders RJA. 2000. Lazarow PB, Moser HW. 1995. Disorders of peroxisome biogenesis. In:
Mutations in the gene encoding peroxisomal alpha-methylacyl-CoA Scriver CR, Sly AL, Valle WS, Beaudet D, editors. The metabolic and
racemase cause adult-onset sensory motor neuropathy. Nat Genet molecular bases of inherited disease. 7th edn. New York: McGraw-Hill.
24:188–191. pp 2287–2324.
Ferdinandusse S, van Grunsven EG, Oostheim W, Denis S, Hogenhout EM, Liu HM, Bangaru BS, Kidd J, Boggs J. 1976. Neuropathological considera-
IJlst L, van Roermund CWT, Waterham HR, Goldfischer S, Wanders tions in cerebro-hepato-renal syndrome (Zellweger’s syndrome). Acta
RJA. 2002. Reinvestigation of peroxisomal 3-ketoacyl-CoA thiolase Neuropathol 34:115–123.
374 Wanders

Maxwell MA, Nelson PV, Chin SJ, Paton BC, Carey WF, Crane DI. 1999. A Roels F, Cornelis A, Poll-Thé BT, Aubourg P, Ogier H, Scotto J, Saudubray
common PEX1 frameshift mutation in patients with disorders of JM. 1986. Hepatic peroxisomes are deficient in infantile Refsum disease:
peroxisome biogenesis correlates with the severe Zellweger syndrome A cytochemical study of 4 cases. Am J Med Genet 25:257–271.
phenotype. Hum Genet 105:38–44. Roesel RA, Carroll JE, Rizzo WB, van der Zalm T, Hahn DA. 1991. Dyggve–
Mihalik SJ, Moser HW, Watkins PA, Danks DM, Poulos A, Rhead WJ. 1989. Melchior–Clausen syndrome with increased pipecolic acid in plasma
Peroxisomal L-pipecolic acid oxidation is deficient in liver from Zellweger and urine. J Inherit Metab Dis 14:876–880.
syndrome patients. Pediatr Res 25:548–552. Roscher AA, Hoefler S, Hoefler G, Paschke E, Paltauf F, Moser A, Moser H.
Moser AB, Rasmussen M, Naidu S, Watkins PA, McGuinness M, Hajra AK, 1989. Genetic and phenotypic heterogeneity in disorders of peroxisome
Chen G, Raymond G, Liu A, Gordon D, Garnaas K, Walton DS, Skjeldal biogenesis—a complementation study involving cell lines from 19
OH, Guggenheim MA, Jackson LG, Elias ER, Moser HW. 1995. Pheno- patients. Pediatr Res 26:67–72.
type of patients with peroxisomal disorders subdivided into sixteen Schutgens RBH, Ryynanen M, Wanders RJA. 1994. Peroxisomal functions
complementation groups. J Pediatr 127:13–22. in mulibrey nanism. J Inherit Metab Dis 17:626–627.
Moser AB, Kreiter N, Bezman L, Lu S, Raymond GV, Naidu S, Moser HW. Scotto JM, Hadchouel M, Odievre M, Laudat MH, Saudubray JM, Dulac O,
1999. Plasma very long chain fatty acids in 3,000 peroxisome disease Beucler I, Beaune P. 1982. Infantile phytanic acid storage disease, a
patients and 29,000 controls. Ann Neurol 45:100–110. possible variant of Refsum’s disease: Three cases, including ultrastruc-
Moser HW, Loes DJ, Melhem ER, Raymond GV, Bezman L, Cox CS, Lu SE. tural studies of the liver. J Inherit Metab Dis 5:83–90.
2000. X-linked adrenoleukodystrophy: Overview and prognosis as a Shimozawa N, Suzuki Y, Orii T, Moser A, Moser HW, Wanders RJA. 1993.
function of age and brain magnetic resonance imaging abnormality. A Standardization of complementation grouping of peroxisome-deficient
study involving 372 patients. Neuropediatrics 31:227–239. disorders and the second Zellweger patient with peroxisomal assembly
Mosser J, Douar AM, Sarde CO, Kioschis P, Feil R, Moser H, Poustka AM, factor-1 (PAF-1) defect. Am J Hum Genet 52:843–844.
Mandel JL, Aubourg P. 1993. Putative X-linked adrenoleukodystrophy Smeitink JA, Beemer FA, Espeel M, Donckerwolcke RA, Jakobs C, Wanders
gene shares unexpected homology with ABC transporters. Nature 361: RJA, Schutgens RBH, Roels F, Duran M, Dorland L, et al. 1992. Bone
726–730. dysplasia associated with phytanic acid accumulation and deficient
Motley AM, Hettema EH, Hogenhout EM, Brites P, ten Asbroek AL, Wijburg plasmalogen synthesis: A peroxisomal entity amenable to plasmapher-
FA, Baas F, Heijmans HSA, Tabak HF, Wanders RJA, Distel B. 1997. esis. J Inherit Metab Dis 15:377–380.
Rhizomelic chondrodysplasia punctata is a peroxisomal protein target- Suzuki Y, Shimozawa N, Yajima S, Tomatsu S, Kondo N, Nakada Y,
ing disease caused by a non-functional PTS2 receptor. Nat Genet 15: Akaboshi S, Lai M, Tanabe Y, Hashimoto T, et al. 1994. Novel subtype
377–380. of peroxisomal acyl-CoA oxidase deficiency and bifunctional enzyme
Motley AM, Brites P, Gerez L, Hogenhout E, Haasjes J, Benne R, Tabak HF, deficiency with detectable enzyme protein: Identification by means of
Wanders RJA, Waterham HR. 2002. Mutational spectrum in the PEX7 complementation analysis. Am J Hum Genet 54(1):36–43.
gene and functional analysis of mutant alleles in 78 patients with Suzuki Y, Jiang LL, Souri M, Miyazawa S, Fukuda S, Zhang Z, Une M,
rhizomelic chondrodysplasia punctata type 1. Am J Hum Genet 70:612– Shimozawa N, Kondo N, Orii T, Hashimoto T. 1997. D-3-hydroxyacyl-
624. CoA dehydratase/D-3-hydroxyacyl-CoA dehydrogenase bifunctional
Ofman R, Hettema EH, Hogenhout EM, Caruso U, Muijsers AO, Wanders protein deficiency: A newly identified peroxisomal disorder. Am J Hum
RJA. 1998. Acyl-CoA-dihydroxyacetonephosphate acyltransferase— Genet 61:1153–1162.
cloning of the human cDNA and resolution of the molecular basis in Sztriha LS, Nork MP, Abdulrazzaq YM, al-Gazali LI, Bakalinova DB. 1997.
rhizomelic chondrodysplasia punctata type 2. Hum Mol Genet 7:847– Abnormal myelination in peroxisomal isolated dihydroxyacetonepho-
853. sphate acyltransferase deficiency. Pediatr Neurol 16:232–236.
Ofman R, Hogenhout EM, Wanders RJA. 1999. Identification and Takada Y, Kaneko N, Esumi H, Purdue PE, Danpure C J. 1990. Human
characterization of the mouse cDNA encoding acyl-CoA: Dihydroxyace- peroxisomal L-alanine: Glyoxylate aminotransferase. Evolutionary loss
tone phosphate acyltranferase. Biochim Biophys Acta 1439:89–94. of a mitochondrial targeting signal by point mutation of the initiation
Poll-Thé BT, Saudubray JM, Ogier HA, Odievre M, Scotto JM, Monnens L, codon. Biochem J 268:517–520.
Govaerts LC, Roels F, Cornelis A, Schutgens RBH, et al. 1987. Infantile Thai TP, Heid H, Rackwitz HR, Hunziker A, Gorgas K, Just WW. 1997.
Refsum disease: An inherited peroxisomal disorder. Comparison with Ether lipid biosynthesis-isolation and molecular characterization of
Zellweger syndrome and neonatal adrenoleukodystrophy. Eur J Pediatr human dihydroxyacetonephosphate acyltransferase. FEBS Lett 420:
146:477–483. 205–211.
Poll-Thé BT, Roels F, Ogier H, Scotto J, Vamecq J, Schutgens RBH, Wanders Thomas GH, Haslam RH, Batshaw ML, Capute AJ, Neidengard L, Ransom
RJA, van Roermund CWT, van Wijland MJ, Schram AW, Tager JM, JL. 1975. Hyperpipecolic acidemia associated with hepatomegaly,
Saudubray JM. 1988. A new peroxisomal disorder with enlarged per- mental retardation, optic nerve dysplasia, and progressive neurological
oxisomes and a specific deficiency of acyl-CoA oxidase (pseudo-neonatal disease. Clin Genet 8:376–382.
adrenoleukodystrophy). Am J Hum Genet 42:422–434.
Torvik A, Torp S, Kase BF, Ek J, Skjeldal OH, Stokke O. 1988. Infantile
Portsteffen H, Beyer A, Becker E, Epplen C, Pawlak A, Kunau WH, Dodt G. Refsum’s disease: A generalized peroxisomal disorder. Report of a case
1997. Human PEX1 is mutated in complementation group 1 of the with postmortem examination. J Neurol Sci 85:39–53.
peroxisome biogenesis disorders. Nat Genet 17:449–452.
Ulrich J, Herschkowitz N, Heitz P, Sigrist T, Baerlocher P. 1978. Adreno-
Powers JM, Tummons RC, Caviness VS Jr, Moser AB, Moser HW. 1989. leukodystrophy: Preliminary report of a connatal case. Light and
Structural and chemical alterations in the cerebral maldevelopment of electron microscopical, immunohistochemical, and biochemical find-
fetal cerebro-hepato-renal (Zellweger) syndrome. J Neuropathol Exp ings. Acta Neuropathol 43:77–82.
Neurol 48:270–289.
Vallat C, Denis S, Bellet H, Jakobs C, Wanders RJA, Mion H. 1996. Major
Purdue PE, Lumb MJ, Fox M, Griffo G, Hamon-Benais C, Povey S, Danpure hyperpipecolataemia in a normal adult. J Inherit Metab Dis 19:624–
CJ. 1991. Characterization and chromosomal mapping of a genomic 626.
clone encoding human alanine:glyoxylate aminotransferase. Genomics
10:34–42. van den Brink DM, Brites P, Haasjes J, Wierzbicki AS, Mitchell J, Lambert-
Hamill M, de Belleroche J, Jansen GA, Waterham HR, Wanders RJA.
Purdue PE, Zhang JW, Skoneczny M, Lazarow PB. 1997. Rhizomelic 2003. Identification of PEX7 as the second gene involved in Refsum
chondrodysplasia punctata is caused by deficiency of human PEX7, a disease. Am J Hum Genet 72:471–477.
homologue of the yeast PTS2 receptor. Nat Genet 15:381–384.
van Grunsven EG, van Berkel E, IJlst L, Vreken P, de Klerk JB, Adamski J,
Rao VV, Chang YF. 1992. Assay for L-pipecolate oxidase activity in human Lemonde H, Clayton PT, Cuebas DA, Wanders RJA. 1998. Peroxisomal
liver: Detection of enzyme deficiency in hyperpipecolic acidaemia. D-hydroxyacyl-CoA dehydrogenase deficiency: Resolution of the enzyme
Biochim Biophys Acta 1139:189–195. defect and its molecular basis in bifunctional protein deficiency. Proc
Raymond GV, Moser HW. 1997. Clinical diagnosis and therapy of Natl Acad Sci USA 95:2128–2133.
peroxisomal diseases. In: Applegarth DA, Dimmick JE, Hall JG, editors. van Grunsven EG, Mooijer PAW, Aubourg P, Wanders RJA. 1999a. Enoyl-
Organelle Diseases. London: Chapman & Hall. CoA hydratase deficiency: Identification of an new type of D-bifunctional
Reuber BE, Germain-Lee E, Collins CS, Morrell JC, Ameritunga R, Moser protein deficiency. Hum Mol Genet 8:1509–1516.
HW, Valle D, Gould SJ. 1997. Mutations in PEX1 are the most common van Grunsven EG, van Berkel E, Mooijer PAW, Watkins PA, Moser HW,
cause of peroxisome biogenesis disorders. Nat Genet 17:445–448. Suzuki Y, Jiang LL, Hashimoto T, Hoefler G, Adamski J, Wanders RJA.
Peroxisomal Disorders 375

1999b. Peroxisomal bifunctional protein deficiency revisited: Resolution in cultured human skin fibroblasts. J Inherit Metab Dis 18(Suppl 1):
of its true enzymatic and molecular basis. Am J Hum Genet 64:99– 113–124.
107. Wanders RJA, Barth PG, Schutgens RBH, Heijmans HSA. 1996. Perox-
Wanders RJA, Purvis YR, Heymans HSA, Bakkeren JA, Parmentier GG, isomal disorders: Post- and prenatal diagnosis based on a new classi-
Van Eldere J, Eyssen H, van den Bosch H, Tager JM, Schutgens RBH. fication with flowcharts. Int Pediatr 11:203–214.
1986. Age-related differences in plasmalogen content of erythrocytes Wanders RJA, Barth PG, Heymans HSA. 2001a. Single peroxisomal enzyme
from patients with the cerebro-hepato-renal (Zellweger) syndrome: deficiencies. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. The
Implications for postnatal detection of the disease. J Inherit Metab Dis Metabolic & Molecular Bases of Inherited Disease. 8th edn. New York:
9:335–342. McGraw-Hill. pp 3219–3256.
Wanders RJA, van Roermund CW, van Wijland MJ, Schutgens RB, Tager Wanders RJA, Jakobs C, Skjeldal OH. 2001b. Refsum disease. In: Scriver
JM, van den Bosch H, Thomas GH. 1988a. Peroxisomes and peroxisomal CR, Beaudet AL, Sly WS, Valle D, editors. The metabolic & molecular
functions in hyperpipecolic acidaemia. J Inherit Metab Dis 11(Suppl 2): bases of inherited disease. 8th edn. New York: Mc Graw-Hill. pp 3303–
161–164. 3321.
Wanders RJA, Heymans HSA, Schutgens RBH, Barth PG, van den Bosch H, Wanders RJA, Jansen GA, Skjeldal OH. 2001c. Refsum disease, peroxi-
Tager JM. 1988b. Peroxisomal disorders in neurology. J Neurol Sci 88: somes, and phytanic acid oxidation: A review. J Neuropathol Exp Neurol
1–39. 60:1021–1031.
Wanders RJA, Romeyn GJ, Schutgens RBH, Tager JM. 1989. L-pipecolate Waterham HR. 2002. Inherited disorders of cholesterol biosynthesis. Clin
oxidase: A distinct peroxisomal enzyme in man. Biochem Biophys Res Genet 61:393–403.
Commun 164:550–555.
Watkins PA, Chen WW, Harris CJ, Hoefler G, Hoefler S, Blake DC Jr, Balfe
Wanders RJA, Schelen A, Feller N, Schutgens RBH, Stellaard F, Jakobs C, A, Kelley RI, Moser AB, Beard ME. 1989. Peroxisomal bifunctional
Mitulla B, Seidlitz G. 1990. First prenatal diagnosis of acyl-CoA oxidase enzyme deficiency. J Clin Invest 83:771–777.
deficiency. J Inherit Metab Dis 13:371–374.
Watkins PA, McGuinness MC, Raymond GV, Hicks BA, Sisk JM, Moser AB,
Wanders RJA, Schumacher H, Heikoop J, Schutgens RBH, Tager JM. 1992. Moser HW. 1995. Distinction between peroxisomal bifunctional enzyme
Human dihydroxyacetonephosphate acyltransferase deficiency: A new and acyl-CoA oxidase deficiencies. Ann Neurol 38:472–477.
peroxisomal disorder. J Inherit Metab Dis 15:389–391.
Wilson GN, Holmes RD, Custer J, Lipowitz JL, Stover J, Datta N, Hajra A.
Wanders RJA, Dekker C, Horvath VA, Schutgens RBH, Tager JM, Van Laer 1986. Zellweger syndrome: Diagnostic assays, syndrome delineation,
P, Lecoutere D. 1994. Human alkyldihydroxyacetonephosphate syn- and potential therapy. Am J Med Genet 24:69–82.
thase deficiency: A new peroxisomal disorder. J Inherit Metab Dis 17:
Yajima S, Suzuki Y, Shimozawa N, Yamaguchi S, Orii T, Fujiki Y, Osumi T,
315–318. Hashimoto T, Moser HW. 1992. Complementation study of peroxisome-
Wanders RJA, Denis S, Ruiter JPN, Schutgens RBH, van Roermund CWT, deficient disorders by immunofluorescence staining and characteriza-
Jakobs BS. 1995. Measurement of peroxisomal fatty acid beta-oxidation tion of fused cells. Hum Genet 88:491–499.

You might also like