Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Neuromuscular Diseases of the Newborn

Alex J. Fay, MD, PhD

The peripheral nervous system (PNS) is composed of motor neurons, nerve roots, plexuses,
peripheral nerves (motor, sensory and autonomic), neuromuscular junction, and skeletal
muscles. Disorders of the PNS in neonates most frequently cause weakness, hypotonia,
and contractures, which may be generalized or focal. Since these findings may also occur
with brain and spinal cord lesions, key features of the history and neurologic exam, together
with diagnostic testing, are helpful in reaching a diagnosis. This review covers the diagnos-
tic approach to PNS disorders in the neonate and includes a discussion of representative
diseases of the motor neuron, brachial plexus, peripheral nerves, neuromuscular junction,
and muscles. The importance of reaching a precise genetic diagnosis is highlighted with a
discussion of current and emerging treatments for neonatal PNS diseases, particularly spi-
nal muscular atrophy.
Semin Pediatr Neurol 32:100771 © 2019 Elsevier Inc. All rights reserved.

Introduction Muscle Tone in the Newborn

D isorders of the peripheral nervous system (PNS) affect


the motor neurons, peripheral nerves, neuromuscular
junction, or muscle, and in some diseases, may involve the
Muscle tone in a newborn is assessed by a combination of
observation, provocative maneuvers and positioning, and
determination of velocity-dependent resistance to passive
central nervous system (CNS) and other organs. Typical range of motion. Normal tone depends on gestational age for
manifestations of neuromuscular disorders in the newborn premature infants and can change with the state of the child
include hypotonia, weakness (generalized or focal), multiple (sleeping, awake and quiet, or agitated).1 If only a single
contractures (arthrogryposis), respiratory failure, and feeding examination is possible, as in the case of an outpatient visit,
difficulties. Weakness may be fixed, progressive, or episodic. assessment of the infant in the quiet, awake state is most
As these symptoms are not specific to dysfunction of the informative.
PNS, the history, neurologic examination, and diagnostic The examination of muscle tone should begin with an
testing are all helpful in localizing to the peripheral nerves, understanding of the normal evolution of muscle tone with
neuromuscular junction, and muscle. Electrodiagnostic test- gestational age (GA). Prior to 28 weeks’ GA, low axial and
ing, such as nerve conduction studies (NCS) and electromy- appendicular tone is normal, without flexion of the arms or
ography (EMG), muscle biopsy, magnetic resonance legs in the quiet, awake state. By 32 weeks’ GA, flexor tone
imaging, and genetic testing can be useful as extensions of begins to appear in the lower extremities, with full flexion of
the neurologic exam and as tools for establishing a precise the hips and knees appearing around 36 weeks’ GA. At 36
diagnosis. Current and emerging therapies for early onset weeks’ GA, flexor tone starts to appear in the upper extremi-
neuromuscular diseases, most notably spinal muscular atro- ties, with full flexor tone at the elbows by 40 weeks’ GA.2
phy, have placed inherited PNS disorders at the forefront of
targeted treatments.

Neuromuscular Examination
Department of Neurology, University of California, San Francisco, San Dysmorphic facial features with “myopathic facies,” where
Francisco, CA. the face is elongated, with dolichocephaly and sometimes
Financial Disclosures: The author has no commercial, proprietary, or ptosis, may be seen in congenital myopathies, muscular dys-
financial interest in any products or companies described in this article.
Address reprint requests to Alex J. Fay, MD, PhD, Department of Neurology,
trophies, and myasthenic syndromes. The examiner should
University of California, San Francisco, 1550 4th St., RH546, MB2922, also note whether the palate is high-arched, a common find-
San Francisco, CA 94143. E-mail: alexander.fay@ucsf.edu ing in congenital neuromuscular disorders.

https://doi.org/10.1016/j.spen.2019.08.007 1
1071-9091/11/© 2019 Elsevier Inc. All rights reserved.
2 A.J. Fay

The cranial nerve examination should focus on eye move- chronic/developmental CNS disorders, systemic illness, med-
ments, crying facies, and the vigor of crying and the sucking ication effects, and PNS disorders. It may occur together with
and rooting reflexes. Limitation of extraocular movements arthrogryposis or episodic weakness, which will be discussed
and/or ptosis may be seen in some congenital myopathies and below.3 Hypotonia may be localized to the PNS by:
myasthenic syndromes. There may be reduced change in facial
expression with crying in children with the myopathic facies  Exclusion of CNS etiologies
mentioned above, asymmetric crying facies with absent  Positive exam findings:
depressor anguli ori, or complete lack of facial movement in ○ symmetric, proximal or distal weakness
M€ obius syndrome. A weak, hypophonic cry can be a sign of ○ prominent facial or bulbar weakness
bulbar weakness (cranial nerves IX and X) from a CNS or PNS ○ hyporeflexia/areflexia
cause. The rooting and sucking reflexes are helpful to assess  Diagnostic testing:
the function of cranial nerves V, VII, and XII, and a weak suck ○ elevated creatine kinase level (muscular dystro-
may correlate with feeding difficulties, though such difficulties phies)
are common in CNS and systemic disease. The tongue should ○ EMG/NCS
be observed for fasciculations, as may occur in motor neuron ○ muscle biopsy
diseases such as spinal muscular atrophy. ○ imaging of muscle or nerves
Low axial and appendicular tone is characteristic of neuro-
muscular disorders, typically in a symmetric pattern. Marked Table 1 provides general patterns for disorders of the
hypotonia and weakness in a single limb should raise con- motor neuron, peripheral nerves, neuromuscular junction,
cern for a traumatic injury, such as brachial plexus palsy, and muscle. These categories are not rigid, as there is overlap
while congenital hemiplegia and diplegia are more likely due and there are exceptions to the rules (eg, distal myopathies).
to brain or spinal cord lesions. Muscle strength can be graded Below, I discuss several representative disorders of each of
on the Medical Research Council (MRC) scale if one carefully these parts of the PNS, and the reader should explore the
isolates muscle groups, but it can be more helpful to define a references for in-depth information about specific causes of
pattern of movement and weakness: limited proximal muscle neonatal hypotonia. One should always consider reversible
movement with relatively preserved movement of the fingers causes of weakness and hypotonia, such as electrolyte and
and toes might be seen in an infant with spinal muscular endocrine abnormalities (hypokalemia, hypermagnesemia,
atrophy or a congenital myopathy, while more severe limita- hypoglycemia, hypo-/hyperthyroidism) in the acute setting.4
tion in distal muscle movement would be characteristic of
peripheral nerve dysfunction (Dejerine-Sottas) or distal
arthrogryposis. While some contractures, such as club foot, Spinal Muscular Atrophy
may be obvious, others are apparent only with testing range
of motion.  SMA is a motor neuron disease that causes proximal
The primitive reflexes may be incomplete or absent in a weakness, initially sparing the eyes and face, and fasci-
child with low muscle tone, or asymmetric with focal CNS or culations.
peripheral injuries. Asymmetry of the Moro reflex should be  Disease onset and severity depend on the number of
apparent in a brachial plexus injury or unilateral CNS lesion, copies of SMN2.
while a muscle or motor neuron disease may result in a  Treatment with nursinersen is now available, with
response that is absent or incomplete. The plantar (Babinski) additional treatments pending.
reflex has little value in localization, unless there is marked
asymmetry. Combined with evaluation of response to gentle Spinal Muscular Atrophy5 is a disorder of the motor neu-
pinprick, light touch, and cold temperature as a part of the rons, most commonly caused by recessive loss of SMN1 pro-
sensory examination, marked abnormalities may localize to tein expression, with disease severity determined by the
spinal cord or sensory nerves. Deep tendon reflexes are often number of copies of the highly homologous SMN2 gene.
reduced proportionally to weakness and hypotonia in PNS Thus, an individual with 4 or more copies of SMN2 may not
disorders. Reflexes may be reduced or absent following an show symptoms until adulthood, while an individual with 1
acute CNS injury, but there is typically a transition to hyper- or 2 copies of SMN2 will typically shows symptoms in utero
reflexia over days or weeks. Hypotonia, limb weakness, or in the first month of life. The age of onset and SMN2 copy
hyporeflexia, and normal mental status in a newborn without number allow for description of SMA subtypes: type 0 (1
history suggestive of a CNS anomaly (such as perinatal hyp- copy SMN2, onset prior to birth), type 1 (2 copies SMN2,
oxia, prolonged resuscitation, or CNS malformation) are con- onset prior to 6 months), type 2 (2-3 copies SMN2, onset
cerning for a PNS cause. 6-18 months), type 3 (3-4 copies SMN2, onset 18 months-
21 years), and type 4 (4-8 copies SMN2, onset >21 years).
Infants with type 0 typically die in the first month of life
from respiratory failure, while infants with type 1 never sit
Hypotonia independently, infants with type 2 never stand indepen-
Low muscle tone in the neonate can present a diagnostic dently, and children with type 3 exhibit a wide range of
challenge, with a broad differential that includes acute and motor ability, from standing only to walking independently.
Neuromuscular Disorders of the Newborn 3

Table 1 Characteristics Features of Disorders of the Peripheral Nervous System


Weakness Sensory
Localization Pattern Symptoms Reflexes Other Disease Examples

Motor neuron Proximal No Reduced/absent Atrophy, cramps, Spinal muscular atro-


fasciculations phy, acute flaccid
myelitis
Plexus Upper trunk, Distribution of Focally reduced Shoulder dystocia, Erb-Duchenne,
lower affected nerves trauma, Horner’s if Klumpke palsies,
trunk, or both T1 root injured hereditary neuralgic
amyotrophy
Peripheral nerve Distal Yes Reduced/absent Pes Cavus, hammer Charcot-Marie-Tooth,
toes, +/- autonomic Guillain-Barr e
symptoms syndrome
Neuromuscular Ocular, proximal, No Normal or Fatigability; improve Congenital myasthenic
junction bulbar reduced w/ ice, acetylcho- syndrome, botulism
linesterase
inhibitors
Muscle Proximal, No Normal or Myotonia, Congenital myopathy,
+/-facial reduced pseudohypertrophy muscular dystrophy,
myotonia congenita

SMA type 1 is the most common subtype, and in natural his- autonomic neuropathies).12 Early onset hypomyelinating
tory studies,6 is fatal in the first 2 years of life without inva- neuropathies are also referred to as Dejerine-Sottas syn-
sive ventilatory support. Initial manifestations of the disease drome,13 with marked slowing of nerve conduction veloc-
include axial and appendicular hypotonia, weakness in the ities and elevated protein in the cerebrospinal fluid. Genes
proximal more than the distal muscles with early sparing of with dominant mutations most commonly associated with
the muscles of facial expression, fasciculations in the tongue these hypomyelinating neuropathies include PMP22, MPZ,
and other muscles, low muscle bulk, chest circumference and EGR2; several recessive forms exist, as well (EGR2,
smaller than head circumference, absent deep tendon periaxin mutations). Characteristic exam findings include
reflexes, and intact sensation. hypotonia, distal more than proximal weakness, club feet,
In one of the most dramatic medical advances of the 21st absent sensation, areflexia, and sometimes cranial neurop-
century, the development of nusinersen,7 a synthetic, stabi- athies, including sensorineural hearing loss. Nerve con-
lized antisense oligonucleotide that targets splicing of SMN2 duction velocities (CV) may be in the range of 3-8 m/s
and increases formation of a more stable protein product, (normal neonatal CV are >20 m/s), often with loss of
has transformed SMA type I, from a fatal disease to a treatable compound motor action potentials (CMAP) due to sec-
condition. Earlier treatment leads to better outcomes,8 with ondary axon loss. Nerve biopsy typically shows absent,
initiation prior to symptom onset being associated with thin, or abnormally folded myelin surrounding nerve
nearly normal early motor development in some infants.9 fibers. Recessive congenital hypomyelinating neuropathies
Gene therapy, with a single intravenous infusion of an have also been reported (as part of the CMT4 category),
adeno-associated virus vector carrying the full length SMN1 with mutations in genes such as GDAP1, SH3TC2, FGD4,
gene, showed promising results in early trials in infants with and EGR2. While in most cases, arthrogryposis is attrib-
SMA type I, and is now approved in the United States for uted to defects in motor neurons and Schwann cells, in
children under two years of age.10 some cases, contractures appear to be due to sensory
neuron dysfunction (PIEZO2, NALCN).14 Giant axonal
neuropathy is a multisystem disease with global develop-
Congenital Neuropathies mental delay, ataxia, short stature, and kinked hair, with
gene therapy treatment currently in clinical trials after
 Congenital neuropathies present with distal-predominant promising preclinical studies.15 Many lysosomal storage
weakness and contractures, sensory loss, and areflexia. disorders also affect the peripheral nerves, and the reader
 NCS can distinguish between demyelinating and axo- should explore references for more information on this
nal forms. topic.16-18 Acquired, immune, and postinfectious neuropa-
thies have rarely been reported in the neonatal period, so
Peripheral neuropathies can be either dysmyelinating or Guillain-Barre syndrome,19 chronic immune demyelinat-
axonal, and most commonly affect both the sensory and ing polyneuropathy,20 and enterovirus-associated acute
motor neurons, though there are forms that specifically flaccid myelitis21 should be considered in a previously
affect motor neurons (hereditary motor neuropathies)11 or normal child who develops signs and symptoms of a dif-
sensory and autonomic neurons (hereditary sensory and fuse, peripheral neuropathy.
4 A.J. Fay

Congenital Myopathies and Muscular Examples include alpha-2-laminin,24 alpha-dystroglycanopa-


Dystrophies thies (proteins that affect glycosylation of alpha-dystrogly-
can),25 and collagen VI.26 Compared to the congenital
 Congenital muscular dystrophies: progressive, limb- myopathies, the congenital muscular dystrophies are more
girdle weakness, early respiratory involvement, ele- likely to affect other organs, particularly the brain27 and
vated CK, muscle biopsy shows replacement of muscle heart.28 Walker-Warburg syndrome (a severe disorder of O-
fibers with connective tissue and fat. glycosylation), for example, is associated with cobblestone
 Congenital myopathies: static/slowly progressive, lissencephaly, microcephaly, microphthalmia, and severe
prominent facial/ocular and limb-girdle weakness, developmental delays.29 Alpha-2-laminin deficiency (also
early respiratory involvement, normal CK, muscle known as merosin deficiency) may also manifest with epi-
biopsy shows small fibers with distinct histological fea- lepsy and diffuse, symmetric white matter abnormalities on
tures. brain MRI (typically present by 6 months of age), though the
neuromuscular manifestations of contractures, hypotonia,
There are 2 broad categories of congenital muscle dis- and elevated creatine kinase are typically present at birth. Ull-
eases,22 which can be distinguished by examination findings, rich and Bethlem myopathies (collagen VI mutations) typi-
creatine kinase level, EMG features, and muscle biopsy, and cally involve the skin (hyperkeratosis pilari, velvety palms/
which have distinct clinical courses and pattern of muscle soles) and may have a normal or only mildly elevated creatine
involvement over time (Table 2). kinase level.
Dystrophies23 are progressive disorders that cause ongoing Congenital myopathies, by contrast, are typically non-
degeneration and regeneration of muscle fibers, with gradual progressive or slowly progressive, and tend to spare
accumulation of fibrotic tissue and fatty replacement of mus- organs other than the skeletal muscle. Genes implicated
cle fibers over time. These pathologic features are best appre- in congenital myopathies are primarily involved in excita-
ciated with muscle biopsy, though a dystrophy may be tion or contraction, and include ryanodine receptor 1
suspected if creatine kinase is markedly elevated and EMG is (RYR1), actin (ACTA1), myotubularin-1 (MTM1), nebu-
consistent with an “irritable myopathy” (fibrillations and pos- lin, titin, and myosin.30 Creatine kinase is normal or only
itive sharp waves with small, polyphasic motor unit poten- slightly elevated, and muscle biopsy does not typically
tials). Weakness tends to be in a limb-girdle pattern, with show prominent degeneration-regeneration or fibrous-
less facial/bulbar involvement than the congenital myopa- fatty replacement; rather, there are characteristic muscle
thies and congenital myasthenic syndromes. Most of the con- histologic changes such as central nuclei, nemaline rods,
genital muscular dystrophies are associated with mutations central cores, or fiber type disproportion. Muscle fibers
in genes that encode extracellular matrix proteins or sarco- are small, which corresponds with the reduced muscle
lemmal proteins associated with the extracellular matrix. bulk seen in patients with congenital myopathies. Facial

Table 2 Distinguishing Features of Congenital Muscle and Neuromuscular Junction Diseases


Congenital Muscular Congenital Myasthenic
Clinical Features Congenital Myopathy Dystrophy Syndromes

Weakness pattern Static/slow progression facial/ Progressive weakness, limb- Episodic, static/slow progres-
ocular/bulbar, limb-girdle girdle sion, ocular, facial/bulbar, limb-
girdle
Creatine kinase (CK) Normal/mildly elevated Marked elevation (exception: can Normal/mildly elevated
be normal in collagen VI
disorders)
Eye Ptosis, ophthalmoplegia common Eye structural changes (alpha- Ptosis, ophthalmoplegia common
dystroglycanopathies)
Heart Unaffected May be affected Unaffected
EMG/NCS Small, polyphasic motor unit Fibrillations, positive sharp Decremental response with
potentials waves, small, polyphasic motor repetitive nerve stimulation
unit potentials
Muscle biopsy Nemaline rods, central cores Degeneration/regeneration, Normal gross (EM may show
central nuclei, fiber type fatty/fibrous replacement of NMJ abnormalities)
muscle fibers
Proteins Excitation, contraction Extracellular matrix, ECM-inter- Neuromuscular junction ion
acting proteins, channels, receptors, ACh
O-glycosylation release
Treatment Gene therapy for MTM1 (in trial) Steroids? gene therapy? Acetylcholinesterase inhibitors,
albuterol, ephedrine
Ach, acetylcholine; ECM, extracellular matrix; EM, electron microscopy; NMJ, neuromuscular junction
Neuromuscular Disorders of the Newborn 5

and bulbar muscles are often weak, resulting in elon- Special Cases: Pompe
gated, “myopathic” facies, high-arched palate, and early
feeding difficulties, sometimes with ptosis or ophthalmo-
 Pompe disease: Hypotonia, macroglossia, early cardio-
plegia. Early respiratory support31 may be necessary to
myopathy, elevated CK; treatable with enzyme
compensate for weakness of the diaphragm and accessory
replacement
muscles. The brain and heart are not typically affected.
With the increasing availability of next-generation
sequencing, muscle biopsy may no longer be necessary in Pompe disease, also known as acid maltase deficiency or
many cases for diagnosis of congenital myopathies and mus- glycogen storage disease type II, can present from the neona-
cular dystrophies, if the history and exam findings are consis- tal period to late adulthood, but the neonatal phenotype has
tent with genetic testing results. Creatine kinase level and some distinguishing features from later onset forms and from
EMG/NCS can be helpful in narrowing the differential in a other forms of congenital muscular dystrophies. The severe
child with hypotonia, and brain and muscle MRI can neonatal onset Pompe disease is associated with cardiomyop-
aid diagnosis in some cases. Muscle pathology may be athy in almost all cases (>90%), and affects not only skeletal
helpful when a clear genetic etiology cannot be identified. and cardiac muscles, but also brain development.42 Affected
Caution should be exercised when sedating children with infants are typically hypotonic, with macroglossia, and crea-
suspected congenital muscle weakness, as mutations in tine kinase is elevated. Diagnosis is typically made by sending
RYR1, CACNA1S, STAC3, and several other genes have been acid maltase enzyme activity testing, and confirming with tar-
associated with malignant hyperthermia upon exposure to geted genetic testing. Muscle biopsy is rarely needed with the
inhaled anesthetics.32 rapid return of results from these tests, but in cases where it
Management of the congenital muscular dystrophies is performed, shows glycogen accumulation on Periodic
and myopathies is largely supportive,33,34 with close Acid-Schiff staining. The course of Pompe disease is progres-
attention to respiratory and feeding complications. Physi- sive, and the neonatal form typically leads to death from car-
cal and occupational therapy, orthotics, mobility equip- diac disease or respiratory failure in infancy without
ment, and corrective surgeries for contractures and treatment.41 With the advent of enzyme replacement ther-
scoliosis may be necessary during childhood. Many of the apy, however, the course of disease can be slowed signifi-
myopathies and muscular dystrophies affect neuromuscu- cantly.43,44 Trials are underway to enhance uptake of
lar junction transmission, so if there is fatigable weakness recombinant GAA enzyme using chaperones, and gene ther-
or electrodecremental response to repetitive nerve stimu- apy trials are also on the horizon, so the treatment landscape
lation, a trial of acetylcholinesterase inhibitor and/or albu- may change significantly in the coming years.45
terol is reasonable.35 Small trials suggest that some
patients with congenital muscular dystrophies may show
improved strength with corticosteroids,36-38 similar to the Special Cases: FSHD
response seen in Duchenne muscular dystrophy, but at
this point, there are not enough data to support routine
 FSHD: Early facial involvement; requires specific test-
use of steroids in newborns or infants with muscular dys-
ing to make diagnosis
trophy. Finally, there are emerging genetically targeted
therapies, including a gene therapy trial for centronuclear
myopathy due to MTM1 mutation.39 The rapidly chang- Facioscapulohumeral muscular dystrophy typically begins
ing therapeutic landscape makes reaching a genetic diag- to manifest in later childhood, adolescence, or early adult-
nosis increasingly important, as patients may be eligible hood, but in its most severe form may present in the neonatal
for trials of therapies that could change the course of period.46 Facial weakness is prominent at onset, and weak-
their disease. ness may be asymmetric. As noted above, FSHD1 cannot be
While gene panels and whole exome sequencing may have diagnosed by gene panels or whole exome sequencing;
diagnostic yields approaching or exceeding 50%40,41 in some rather, testing involves identification of a permissive allele on
neuromuscular disorders, it is important to recognize that the terminal part of chromosome 4q, with a reduced number
these tests may miss several conditions. The most common of D4Z4 repeats.47 Greater contraction of the number of
of these is myotonic dystrophy, type I (DM1), while faciosca- D4Z4 repeats is associated with earlier onset of disease, and
pulohumeral muscular dystrophy (FSHD) may also occur in the shortened number of repeats is thought to weaken sup-
the neonatal period in its most severe manifestation. Each of pression of a toxic downstream gene, DUX4.48 Although
these requires specific testing: trinucleotide repeat sequenc- muscle biopsy shows inflammatory changes, corticosteroids
ing in the case of DM1, and chromosome 4q D4Z4 repeat have not been found to be effective in slowing the course of
contraction in FSHD. In addition, next-generation sequenc- disease. Several approaches to treat FSHD are under investi-
ing may take weeks to months to return results, so in treat- gation, aiming to suppress DUX4 expression.46 FSHD2 is
able disorders such as infantile-onset Pompe disease (acid caused by dominant mutations in the SMCHD1 gene, and
maltase deficiency), enzyme testing should be sent early to may be diagnosed with individual gene testing, panel testing,
expedite diagnosis. or whole exome sequencing.
6 A.J. Fay

Special Cases: Myotonic Dystrophy, Type I blood, urine, or CSF testing. Fatty acid oxidation defects and
(DM1) disorders of glycogen storage or metabolism should be con-
sidered in a newborn with hypoglycemia, while encephalopa-
 DM1: Profound hypotonia, distinct facies, early respi- thy, and hypotonia, often with metabolic acidosis after
initiation of feeding, should prompt consideration of amino
ratory failure and feeding difficulties; examine mother,
acidopathies and organic acidurias. Mitochondrial disorders
send specific trinucleotide repeat testing to diagnose
may manifest with severe CNS abnormalities, such as
encephalopathy, seizures, and stroke-like episodes, or may
As with other trinucleotide repeat diseases, DM1 may have affect skeletal or cardiac muscle, peripheral nerves, eyes,
a variable age of onset, depending on the number of repeats. liver, or some combination of these tissues. Suggested initial
DM1, especially the congenital form, should be considered a testing includes serum amino acids, urine organic acids, total
multisystem, degenerative disorder. The neonatal form is the and free carnitine, acylcarnitine profile, ammonia, lactate and
most severe and may lead to death from respiratory failure pyruvate, uric acid, urine creatine metabolites, and urine
during infancy.49 The presenting features include severe purines and pyrimidines. CSF analysis of lactate/pyruvate
hypotonia, respiratory and feeding difficulties, and a charac- ratio, amino acids, and MR spectroscopy should be consid-
teristic facial weakness with a “fish-shaped” mouth. Hypoto- ered in the setting of CNS symptoms. Muscle biopsy may be
nia is thought to be from CNS pathology in the neonatal helpful, and in the case of suspected mitochondrial disease,
period, as creatine kinase is usually normal and muscle it is preferable to perform sequencing of mitochondrial DNA
biopsy does not show the dystrophic features that are typical from muscle tissue instead of blood, due to differences in
of more advanced disease. Myotonia, clinical and electro- heteroplasmy.53
graphic, is not present until late childhood.50 Thus, examin-
ing the parents, particularly the mother, may be the most
helpful part of the neurologic examination: instability of
the repeat length is greater in women than in men, such that Arthrogryposis
the neonatal form of DM1 is typically inherited from the
mother.51 The mother of a child with DM1 may manifest
 Contractures at 2 or more joints (excluding club feet
with grip or percussion myotonia (elicited by striking the
thenar eminence with a reflex hammer and watching for and hips)
 Causes can include CNS, PNS, connective tissue;
delayed relaxation), cataracts, distal weakness (particularly
foot drop), or mild intellectual disability. Although prognosis acquired or inherited
is poor for infants with congenital myotonic dystrophy, the
mother may benefit from diagnosis and referral to a cardiolo- Arthrogryposis is defined as the presence of contractures at
gist, as cardiac conduction defects cause early mortality in 2 or more joints but not limited to club feet or hip dysplasia.
later onset DM1. It is a syndrome, rather than a disease, and has diverse
causes, from CNS injuries and malformations, to maternal
and uterine factors such as oligohydramnios and uterine
Inborn Errors of Metabolism and the PNS bands, to connective tissue disorders and diseases of the
PNS.54 This section focuses on PNS causes but touches upon
 Metabolic disorders may present with acute hypotonia, CNS causes in identifying some of the distinguishing features
encephalopathy, cardiomyopathy, with or without of neuromuscular disorders.
metabolic acidosis, hypoglycemia. Hypotonia and arthrogryposis are more frequently due to
 Work-up includes blood and urine metabolic studies, CNS causes, most commonly following perinatal hypoxic-
brain MRI with spectroscopy. ischemic brain injury, than to PNS causes. The presence of
encephalopathy at birth, seizures, and characteristic findings
A variety of defects in lipid, amino acid, glycogen, and on brain MRI points to a CNS cause. In addition, contrac-
mitochondrial metabolism may lead to neonatal myopathies, tures following perinatal CNS injury are not typically present
often with CNS and other organ involvement, including car- until weeks or months after birth. Reduced fetal movements,
diomyopathy and liver dysfunction.52 A detailed description contractures identified by antenatal ultrasound or present at
of each of these disorders is beyond the scope of this review birth, and reduced muscle bulk at birth are more characteris-
but some general principles are helpful in considering this tic of PNS disorders, though these findings can also be seen
class of disorders. The importance of making diagnoses of in cases of CNS malformations or injury from TORCH infec-
metabolic disorders is that many can be treated, or at least tions. Club foot and hip dysplasia are relatively common and
mitigated, by dietary restrictions or by supplementing cofac- are typically not associated with abnormal muscle or
tors. While newborn screening captures many metabolic peripheral nerve dysfunction, so they are not covered specifi-
abnormalities on blood spots, especially among the amino cally in this section. Rather, I will review several PNS disor-
acidopathies, organic acidurias, and fatty acid oxidation dis- ders which present with prominent contractures at birth.
orders, it must be recognized that many metabolic diseases Any disorder that causes severely reduced movement in
are missed by newborn screening, and may require specific utero, however, can lead to arthrogryposis. Pathologically,
Neuromuscular Disorders of the Newborn 7

arthrogryposis may be associated with increased connective Contractures may improve after birth, but recur progressively
tissue at joints, and with muscle atrophy due to disuse.55 later in childhood. While some children may achieve ambu-
lation, most do not, and progressive respiratory insufficiency
due to scoliosis typically occurs in childhood. Collagen XII
Amyoplasia mutations have also been reported to cause a similar pheno-
The most common cause of arthrogryposis multiplex conge- type. Bethlem muscular dystrophy is a milder form of colla-
nita, accounting for approximately one-third of cases, is gen VI dysfunction, which may cause neonatal hypotonia,
amyoplasia,56 which is thought to be an acquired disorder but typically causes weakness and contractures in the infan-
that occurs sporadically because of vascular injury of the tile period or later childhood. As collagen VI is also expressed
motor neurons early in development. It is nonprogressive in skin, dermatologic features may include hyperkeratosis
but leaves affected individuals with severely restricted move- pilari, keloid formation, velvety skin over the palms, and
ment as they grow up. Amyoplasia most commonly affects atrophic scarring.
all 4 limbs, but 3 limbs, upper extremity only and lower
extremity only involvement have been reported in case series.
Clubfoot deformities are seen in almost all cases, with other
common features including symmetric elbow extension con-
Episodic Weakness: Disorders of
tractures, wrist flexion contractures, short limbs with the Neuromuscular Junction
reduced muscle bulk, and midline forehead nevus flammeus.
Additional findings that may be associated with amyoplasia  Congenital myasthenic syndromes: episodic weakness
include gastroschisis, bowel atresia, and partial absence of (ptosis, ophthalmoplegia, and facial weakness), respi-
fingers or toes. Muscle biopsy typically shows replacement of ratory failure, feeding difficulties; NCS with repetitive
muscle tissue with fat and fibrous connective tissue. Manage- nerve stimulation can aid diagnosis; treatment with
ment consists of casting, surgical tendon releases, and physi- pyridostigmine, albuterol.
cal therapy. Since the injury is nonprogressive, and  Botulism and maternal myasthenia gravis are acquired
amyoplasia does not typically involve any CNS injury, the causes of neonatal weakness.
prognosis is good with diligent management of contractures.

Distal Arthrogryposis Congenital Myasthenic Syndromes


The distal arthrogryposes cause club feet, ulnar deviation of Fluctuating weakness in the neonate, particularly when it
the hands with overlapping fingers and finger flexion con- affects facial and/or bulbar muscles, is most likely to be
tractures, and may involve characteristic facial features, such caused by congenital myasthenic syndrome,63 hereditary dis-
as the “whistling face” in Sheldon-Freeman syndrome.57 orders of neuromuscular transmission. Some rare causes of
Most are dominant and caused by mutations in either muscle acquired neuromuscular junction disease, including botulism
or peripheral nerve genes.58,59 Club feet may be initially and maternal myasthenia gravis, may also cause facial and
treated with serial casting, though the response is typically bulbar weakness that fluctuates. Episodic disorders of skele-
less than would be seen in idiopathic clubfoot, where there is tal muscle, such as periodic paralyses and myotonias, do not
no underlying weakness. typically manifest until later in childhood, though the
sodium channel myotonias have been reported to present as
neonatal apnea64 or stridor65 and even sudden infant death
Escobar Congenital Myasthenic Syndrome syndrome.66
Mutations in acetylcholine receptor subunits, particularly the In older children, a history of fatigability can often be eli-
fetally expressed gamma subunit, may lead to multiple pte- cited, but this may be more challenging for parents to iden-
rygium syndrome, where reduced movement in utero leads tify in neonates, who are less active. Progressive ptosis over
to webbing of skin (pterygia) across joints.60 Diffuse contrac- the course of waking hours and fatigue during feeding may
tures may also be seen with recessive rapsyn mutations.61 be present. The weakness pattern of ptosis with or without
When mutations are present in the gamma AChR subunit, ophthalmoplegia, facial weakness, and bulbar weakness,
there is often improvement in strength in infancy and child- superimposed on diffuse axial and appendicular hypotonia,
hood, as the gamma subunit expression is reduced and the is typical of both congenital myopathies and congenital
delta subunit is expressed in mature muscle postsynaptic sar- myasthenic syndromes. With more dramatic symptoms,
colemma. such as episodic apneas, CNS causes (seizure, nonaccidental
trauma) and infectious syndromes like seizures, sepsis, or
meningitis, should be excluded first.
Ullrich Congenital Muscular Dystrophy Diagnostic work-up for disorders of the neuromuscular
Mutations in collagen VI genes (COL6A1, COL6A2, junction includes NCS with repetitive nerve stimulation and
COL6A3), both dominant and recessive, lead to a unique EMG. CMAP amplitudes may be normal or slightly reduced,
syndrome of proximal contractures and distal joint hyperlax- while EMG is often normal or consistent with a nonirritable
ity, called Ullrich congenital muscular dystrophy.62 myopathic process. Repetitive nerve stimulation at 2 Hz,
8 A.J. Fay

recording from a weak muscle, is usually sufficient to dem- Botulism typically affects infants in the first 6 months of life
onstrate the diagnostic >10% electrodecrement, even in a and may occasionally occur in newborns.71 It is a disease of
fussy newborn. Some presynaptic disorders, such as choline bacterial overgrowth by the anaerobe Clostridium botulinum (or
acetyltransferase (ChAT) deficiency and botulism, require more rarely, other subtypes of clostridia), which releases a
higher frequency (10 Hz or more) stimulation.67 Repetitive toxin that enters the motor neuron presynaptic membrane,
CMAPs, a unique feature of the slow channel myasthenic where it cleaves proteins that are critical for calcium-depen-
syndromes (dominant acetylcholine receptor mutations) and dent release of acetylcholine into the neuromuscular junction.
endplate cholinesterase deficiency (recessive ColQ muta- Ingestion of C. botulinum spores from soil is the most common
tions), can be seen with standard NCS. exposure (more common than unpasteurized honey), and is
Several of the congenital myasthenic syndromes have char- followed by bacterial replication and toxin release among the
acteristic features that may raise suspicion before any diag- infant’s immature gut flora. Clinical manifestations begin with
nostic tests are performed. Escobar syndrome (multiple constipation, followed by bulbar weakness with poor feeding
pterygia) is discussed above. Recessive mutations in ColQ and a weak cry, and progressing to diffuse hypotonia and
can cause sluggish pupillary reactivity,68 which is not typi- weakness, often with respiratory failure. A definitive diagnosis
cally seen in other congenital myasthenic syndromes. Epi- is made by detection of toxin in the stool by ELISA, but, if
sodic apneas should raise suspicion for choline available, electrodiagnostic studies may help support the diag-
acetyltransferase deficiency, sodium channel (SCN4A), or nosis more quickly than the stool toxin results. NCS show
rapsyn mutations. While most congenital myasthenic syn- reduced CMAP amplitudes, with an increase in CMAP ampli-
dromes selectively affect skeletal muscle, there are several tude upon high frequency (10 Hz or greater) repetitive nerve
forms (SNAP25, DPAGT1, ALG2, ALG14, GMPPB, and stimulation. EMG may be consistent with acute denervation,
GFPT1) that may affect the CNS.69 with fibrillations, positive sharp waves, and a reduced recruit-
The importance of diagnosing a congenital myasthenic syn- ment pattern, or may have more typically myopathic features
drome is that these are treatable conditions.63 Most subtypes of small, polyphasic motor unit potentials and an early recruit-
are responsive to acetylcholinesterase inhibitors such as pyri- ment pattern. Treatment, however, should not be delayed
dostigmine, with some exceptions: individuals with DOK7 when awaiting diagnostic testing results: human botulinum
mutations may show worsening weakness with pyridostig- antitoxin (BabyBIG) should be requested from the California
mine, while those with slow channel syndrome and endplate Department of Public Health, and given to the child as soon as
cholinesterase deficiency (ColQ mutations) may worsen or not the diagnosis is suspected. BabyBIG treats only subtypes A
respond to acetylcholinesterase inhibitors. Response to pyri- and B, by far the most common, but occasionally equine hep-
dostigmine should be immediate with appropriate dosing. tavalent antitoxin (covers all subtypes, A-G) is necessary once
Similarly, the potassium channel blocker 3,4-diaminopyridine, stool toxin results return.72 BabyBIG shortens hospitalization
used also to treat Lambert-Eaton myasthenia, can improve and time on mechanical ventilation, and is cost effective.73
symptoms rapidly, but should be avoided in DOK7 myas- Prognosis for affected infants is excellent with prompt sup-
thenic syndrome and slow channel syndrome. Adrenergic ago- portive care and treatment.
nists, including albuterol and ephedrine, have also been
widely reported to improve strength and reduce fatigability in
myasthenic syndromes, though benefit is usually evident after
1-3 months of treatment. Other treatments include sodium Focal Weakness
channel blocking medications (fluoxetine, quinidine, quinine)
for slow channel syndrome. A trial of one or more of these Brachial Plexus Injury
treatments should be considered in children with congenital The most common acquired brachial plexus injury is the Erg-
myopathies and congenital muscular dystrophies,35 especially Duchenne palsy,74 often resulting from shoulder dystocia.
if there is fatigable weakness and/or a decremental response This upper brachial plexus injury may be due to stretching
on repetitive nerve stimulation. Even with treatment, there is the parts of the plexus deriving from the C5 and C6 roots
typically residual weakness, which may gradually worsen over when the infant’s head and neck are stretched away from the
many years. shoulder. The common pattern of weakness with this injury
manifests as arm adduction (deltoid and supraspinatus weak-
ness), internal rotation (infraspinatus weakness), extension at
Acquired Neuromuscular Junction Disorders the elbow (biceps weakness), often with a “waiter’s tip” posi-
Mothers with untreated or undertreated myasthenia gravis tioning of the wrist in flexion. In most cases, the injury recov-
and elevated anti-AChR antibodies may pass these antibodies ers spontaneously in the first 3 months with physical
transplacentally to the fetus, resulting in neonatal arthrogry- therapy, but with more severe injuries, such as nerve root
posis and/or hypotonia with weakness and respiratory avulsion, surgical intervention may be necessary. Still, even
depression.70 If there is uncertainty about the diagnosis, it in cases that appear severe at birth, diagnostic testing (EMG/
should be confirmed in the mother with anti-AChR antibod- NCS and MRI) is typically deferred until the child is 3
ies and electrodiagnostic studies. Symptoms resolve over sev- months of age, as spontaneous recovery may still occur.75
eral weeks with supportive care and cholinesterase Lower plexus injury, leading to a Klumpke palsy,76 is
inhibitors, such as pyridostigmine. much less common that the Erb-Duchenne palsy, and may
Neuromuscular Disorders of the Newborn 9

result from forced abduction of the arm that puts tension Table 3 Disorders Causing Both CNS and PNS Involvement
especially on the portions of the plexus derived from C8 and
Motor neuron
T1. The resulting palsy leads to elbow flexion (triceps weak-  Acquired: Enterovirus encephalitis/acute flaccid myelitis
ness) with wrist and finger flexion and a “claw hand.” If the  Hereditary: Spinal muscular atrophy with pontocerebellar
sympathetic neurons that emerge from the T1 nerve root are hypoplasia, SMA with progressive myoclonic epilepsy
injured, a Horner’s syndrome may be seen on the affected (ASAH1 deficiency)
side. In the most severe cases of birth injury, the entire bra- Peripheral nerve
chial plexus may be injured, resulting in a flaccid arm and  Lysosomal storage diseases (eg, Krabbe, metachromatic
Horner’s syndrome. leukodystrophy, Farber’s)
 Congenital disorder of glycosylation, type 1a
 Peroxisomal diseases (Infantile Refsum’s, Zellweger’s dis-
ease)
Mononeuropathies  Hypomyelinating neuropathy (SOX10)
Mononeuropathies are relatively uncommon at birth, but  Giant axonal neuropathy, infantile neuroaxonal dystrophy,
may include radial nerve palsy and facial nerve palsy. Injury cerebral dysgenesis, neuropathy, ichthyosis, and palmo-
to the facial nerve77 can occur with forceps delivery or in plantar keratoderma (CEDNIK), congenital disorder of
utero compression, causing mechanical trauma to the facial deglycosylation (NGLY1)
nerve, and leading to an asymmetric crying facies, often  Andermann syndrome
 Hereditary sensory and autonomic neuropathy, type 4;
involving the upper and lower face. This is not to be con-
Navajo sensory and autonomic neuropathy with
fused with absence of the depressor anguli oris,78 which
arthropathy
causes asymmetry in displacement of the lateral lips with cry-
Neuromuscular junction
ing, but does not affect the nasolabial fold or upper face.  Congenital myasthenic syndromes: SNAP25, DPAGT1,
Recovery from facial nerve injury may lead to Frey’s syn- ALG2, ALG14, GMPPB, GFPT1
drome,79 where there is unilateral facial flushing and hyper- Myopathy
hidrosis with eating, due to aberrant reinnervation of blood  Congenital myotonic dystrophy
vessels and sweat glands as the facial nerve recovers from  Congenital Pompe disease (acid maltase deficiency)
injury. M€ obius syndrome80 involves hypoplasia or absence  Congenital muscular dystrophy (alpha-dystroglycanop-
of the facial nerve, unilaterally or bilaterally, typically associ- athy, LAMA2 deficiency)
ated with ipsilateral abducens nerve hypoplasia, and some-  Inborn errors of metabolism (mitochondrial, fatty acid oxi-
times impairment of other cranial nerves or limb dation disorders)
 Marinesco-Sjogren syndrome
malformations.
 22q11.2 deletion with absent depressor anguli oris
Radial nerve injury has also been reported perinatally,81,82
associated with intrauterine constriction, prolonged pressure
on the arm during delivery, or rarely, fracture of the humerus.
Clinical manifestations include wrist drop and inability to disorders and mitochondrial disorders, which often affect
extend the thumb and metacarpophalangeal joints. Prognosis both brain and muscle. Congenital muscular dystrophies,
is excellent, with complete recovery in most children. particularly alpha-2-laminin (merosin) deficiency and the
alpha-dystroglycanopathies, affect both the brain and mus-
cle, as do myotonic dystrophy and Pompe disease. The lyso-
somal storage and peroxisomal disorders, such as Krabbe
Disorders Affecting the Central disease and infantile-onset Refsum disease, can be associated
with CNS white matter abnormalities and demyelinating
and PNSs peripheral neuropathy. Table 3 lists examples of diseases that
A newborn may have symptoms and signs indicative of both cause both CNS and PNS dysfunction. Brain MRI is recom-
CNS and PNS dysfunction, which can make the localization mended as part of the work-up for most children with con-
of weakness and hypotonia a challenge. In the case of a child genital hypotonia to evaluate for CNS pathology, and
with encephalopathy, seizures, or spasticity, which point to hypotonic infants should also be tested for inborn errors of
CNS pathology, concurrent PNS disease should be consid- metabolism. Further work-up for suspected PNS disease is
ered especially when there is low muscle bulk, a proximal or discussed in the next section.
distal-predominant weakness pattern, hyporeflexia/areflexia,
or elevated creatine kinase. In some cases, a child may have a
rare genetic syndrome affecting the PNS, and coincidentally
develop hypoxic-ischemic encephalopathy from labor com-
Diagnostic Approaches to
plications. In others, a PNS disorder may lead to secondary Neuromuscular Disorders
brain injury, as may occur from a prolonged apnea related to
neuromuscular weakness (congenital myopathies and myas-  Check CK: Marked elevation is indicative of a muscular
thenic syndromes). Finally, there are conditions that impair dystrophy
function of both CNS and PNS tissues.83 These include  EMG/NCS helps to localize, especially to motor neu-
inborn errors of metabolism, such as fatty acid oxidation ron, peripheral nerve, NMJ
10 A.J. Fay

 Genetic testing by single gene (SMA), panel (most Current and Emerging
muscle, NMJ, and nerve diseases), or whole exome/ Therapeutics
genome is replacing muscle biopsy, which is neverthe-
less still helpful in challenging cases The developments of nusinersen and gene therapy for spinal
muscular atrophy mark the forefront of therapeutics for neu-
romuscular diseases, but targeted therapies are under devel-
The PNS differs from the CNS in that it can be directly opment for many other conditions. Examples include
and readily studied with electrophysiological techniques antisense oligonucleotide-mediated exon skipping for Duch-
and tissue biopsy. Electrophysiology for myasthenia and enne muscular dystrophy (currently with conditional FDA
muscle biopsy for immune myopathies, for example, is approval for exon 51 skipping), gene therapy for centronu-
still mainstays in diagnosis. EMG/NCS is an uncomfort- clear myopathy (MTM1) and giant axonal neuropathy, and
able test at any age and is often challenging in newborns CRISPR/Cas9-based gene editing87 studies for many disor-
because the signal amplitudes are smaller than those of ders, currently in the preclinical phase. Rapid developments
older children (a problem that is compounded by electri- in therapeutics highlight the importance of achieving a pre-
cal noise in intensive care unit settings) and the difficulty cise genetic diagnosis for children with suspected inherited
in coaxing a young child to activate muscles during neuromuscular disorders, as this will determine eligibility for
EMG. Still, in certain settings, such as congenital myas- current and future clinical trials and treatments. This is a
thenic syndromes, demyelinating and axonal neuropa- hopeful and exciting time, with the expectation that the next
thies, and motor neuron diseases, even a limited study decade will bring more therapeutics that will transform life-
may provide critical diagnostic information. In the setting limiting and disabling disorders of the PNS into treatable
of a marked creatine kinase elevation (>5£ upper limit conditions.
of normal), however, EMG may be unnecessary, as the
laboratory value is already suggestive of an irritable
myopathy. Although muscle biopsy was once the main-
stay of diagnosing congenital myopathies and congenital References
muscular dystrophies, this invasive procedure is no lon- 1. Volpe JJ: Neonatal hypotonia. In: Darras BT, et al (ed): Neuromuscular
ger essential in all cases, with the availability of genetic Disorders of Infancy, Childhood, and Adolescence, ed 2, New York:
Academic Press, 85-95, 2015
testing. With the growing use of ultrasound and MRI in
2. S. Saint-Anne Dargassies: Neurological Development in the Full-term
the diagnosis of inherited myopathies, imaging may pro- and Premature Neonate. New York: Excerpta Medica, 1977
vide enough information to make muscle biopsy unneces- 3. Bodensteiner JB: The evaluation of the hypotonic infant. Semin Pediatr
sary in some instances. Neurol 15:10-20, 2008
Newborn screening for spinal muscular atrophy is 4. Darras BT, Jones HR Jr., et al: Neuromuscular problems of the critically
already available in some states, and is being considered ill neonate and child. In: Darras BT, et al (ed): Neuromuscular Disorders
of Infancy, Childhood, and Adolescence, ed 2, New York: Academic
for Pompe disease,84 as well. Gene panels are available Press, 885-903, 2015
from many companies that cover most of the genes 5. Darras BT: Spinal muscular atrophies. Pediatr Clin North Am 62:743-
known to be associated with inherited muscle and 766, 2015
peripheral nerve diseases.85 Comparative genomic hybrid- 6. Kolb SJ, Coffey CS, Yankey JW, et al: Natural history of infantile-onset
ization/single nucleotide polymorphism arrays should be spinal muscular atrophy. Ann Neurol 82:883-891, 2017
7. Finkel RS, Mercuri E, Darras BT, et al: Nusinersen versus sham control
considered if panel testing is negative, or if multiorgan in infantile-onset spinal muscular atrophy. N Engl J Med 377:1723-
involvement or dysmorphic features (beyond those 1732, 2017
explained by muscle weakness) are present. If gene panel 8. Mercuri E, Darras BT, Chiriboga CA, et al: Nusinersen versus sham con-
testing is negative, or if the child’s phenotype does not trol in later-onset spinal muscular atrophy. N Engl J Med 378:625-635,
2018
seem to fit known phenotypes, whole exome sequencing
9. Kraszewski JN, Kay DM, Stevens CF, et al: Pilot study of population-
should be obtained, ideally along with at least 2 other based newborn screening for spinal muscular atrophy in New York
first degree relatives (a “trio,” to aid in filtering out non- state. Genet Med 20:608-613, 2018
pathogenic polymorphisms). It may be reasonable to rec- 10. Mendell JR, Al-Zaidy S, Shell R, et al: Single-dose gene-replacement
ommend muscle biopsy in the case of a negative gene therapy for spinal muscular atrophy. N Engl J Med 377:1713-1722,
panel or whole exome sequencing, in order to establish a 2017
11. Rossor AM, Kalmar B, Greensmith L, Reilly MM. The distal hereditary
diagnosis. Whole genome sequencing is currently per- motor
formed mostly on a research basis, though its availability 12. Auer-Grumbach M. Hereditary sensory and autonomic neuropathies.
and yield can be expected to increase as costs come Handb Clin
down and more labs become comfortable with the inten- 13. Landrieu P, Baets J: Early onset (childhood) monogenic neuropathies.
sive data analysis. There have been some notable suc- Handb Clin Neurol 115:863-891, 2013
14. Delle Vedove A, Storbeck M, Heller R, et al: Biallelic loss of propriocep-
cesses in using whole genome and transcriptome tion-related PIEZO2 causes muscular atrophy with perinatal respiratory
sequencing to identify intronic mutations in collagen VI, distress, arthrogryposis, and scoliosis. Am J Hum Genet 99:1206-1216,
titin, and other muscular dystrophy-associated genes.86 2016
Neuromuscular Disorders of the Newborn 11

15. Bailey RM, Armao D, Nagabhushan Kalburgi S, Gray SJ: Development of 38. Komaki H, Hayashi YK, Tsuburaya R, et al: Inflammatory changes in
intrathecal AAV9 gene therapy for giant axonal neuropathy. Mol Ther infantile-onset LMNA-associated myopathy. Neuromuscul Disord
Methods Clin Dev 9:160-171, 2018 21:563-568, 2011
16. D'Amico A, Bertini E: Metabolic neuropathies and myopathies. Handb 39. Mack DL, Poulard K, Goddard MA, et al: Systemic AAV8-mediated gene
Clin Neurol 113:1437-1455, 2013 therapy drives whole-body correction of myotubular myopathy in dogs.
17. Siddiqi ZA, Sanders DB, Massey JM: Peripheral neuropathy in Krabbe Mol Ther 25:839-854, 2017
disease: Electrodiagnostic findings. Neurology 67:263-267, 2006 40. Ghaoui R, Cooper ST, Lek M, et al: Use of whole-exome sequencing for
18. Haberlandt E, Scholl-B€ urgi S, Neuberger J, et al: Peripheral neuropathy diagnosis of limb-girdle muscular dystrophy: Outcomes and lessons
as the sole initial finding in three children with infantile metachromatic learned. JAMA Neurol 72:1424-1432, 2015
leukodystrophy. Eur J Paediatr Neurol 13:257-260, 2009 41. Schofield D, Alam K, Douglas L, et al: Cost-effectiveness of massively
19. Anastasopoulou S, Lindefeldt M, Bartocci M, Wickstr€om R: Acute parallel sequencing for diagnosis of paediatric muscle diseases. NPJ
inflammatory demyelinating polyradiculoneuropathy in a newborn Genom Med 2, 2017. pii: 4
infant. Eur J Paediatr Neurol 20:754-757, 2016 42. van den Hout HM, Hop W, van Diggelen OP, et al: The natural course
20. Majumdar A, Hartley L, Manzur AY, et al: A case of severe congenital of infantile Pompe’s disease: 20 original cases compared with 133 cases
chronic inflammatory demyelinating polyneuropathy with complete from the literature. Pediatrics 112:332-340, 2003
spontaneous remission. Neuromuscul Disord 14:818-821, 2004 43. Kishnani PS, Corzo D, Nicolino M, et al: Recombinant human acid
21. Greninger AL, Naccache SN, Messacar K, et al: A novel outbreak enterovi- [alpha]-glucosidase: Major clinical benefits in infantile-onset Pompe dis-
rus D68 strain associated with acute flaccid myelitis cases in the USA ease. Neurology 68:99-109, 2007
(2012-14): A retrospective cohort study. Lancet Infect Dis 15:671-682, 2015 44. Kishnani PS, Corzo D, Leslie ND, et al: Early treatment with alglucosi-
22. Schorling DC, Kirschner J, B€onnemann CG: Congenital muscular dys- dase alpha prolongs long-term survival of infants with Pompe disease.
trophies and myopathies: An overview and update. Neuropediatrics Pediatr Res 66:329-335, 2009
48:247-261, 2017 45. Kishnani PS, Beckemeyer AA: New therapeutic approaches for Pompe
23. Kirschner J: Congenital muscular dystrophies. Handb Clin Neurol disease: Enzyme replacement therapy and beyond. Pediatr Endocrinol
113:1377-1385, 2013 Rev 12(Suppl 1):114-124, 2014
24. Durbeej M: Laminin-a2 chain-deficient congenital muscular dystrophy: 46. Klinge L, Eagle M, Haggerty ID, et al: Severe phenotype in infantile
Pathophysiology and development of treatment. Curr Top Membr facioscapulohumeral muscular dystrophy. Neuromuscul Disord
76:31-60, 2015 16:553-558, 2006
25. Lim BC, Lee S, Shin JY, et al: Molecular diagnosis of congenital muscu- 47. Statland JM, Tawil R: Facioscapulohumeral muscular dystrophy. Con-
lar dystrophies with defective glycosylation of alpha-dystroglycan using tinuum (Minneap Minn) 22:1916-1931, 2016. (6, Muscle and Neuro-
next-generation sequencing technology. Neuromuscul Disord 23:337- muscular Junction Disorders)
344, 2013 48. Lemmers RJ, van der Vliet PJ, Klooster R, et al: A unifying genetic model
26. Lampe AK, Flanigan KM, Bushby KM, Hicks D. Collagen type VI- for facioscapulohumeral muscular dystrophy. Science 329:1650-1653,
related disorders. 2004 Jun 25 [updated 2012 Aug 9]. In: Adam MP, 2010
Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens K, Amemiya 49. Reardon W, Newcombe R, Fenton I, et al: The natural history of con-
A, (eds). GeneReviewsÒ [Internet]. Seattle WA: University of Washing- genital myotonic dystrophy: Mortality and long term clinical aspects.
ton, Seattle; 1993-2018. Available from http://www.ncbi.nlm.nih.gov/ Arch Dis Child 68:177-181, 1993
books/NBK1503/PubMed PMID: 20301676. 50. Echenne B, Bassez G: Congenital and infantile myotonic dystrophy.
27. Yamamoto T, Hiroi A, Osawa M, Shibata N: The muscular dystrophies Handb Clin Neurol 113:1387-1393, 2013
associated with central nervous system lesions: A brief review from a 51. Yum K, Wang ET, Kalsotra A: Myotonic dystrophy: Disease repeat
standpoint of the localization and function of causative genes. Curr range, penetrance, age of onset, and relationship between repeat size
Pediatr Rev 10:282-291, 2014 and phenotypes. Curr Opin Genet Dev 44:30-37, 2017
28. Finsterer J, Ramaciotti C, Wang CH, et al: Cardiac findings in congenital 52. Tarnopolsky MA: Metabolic myopathies. Continuum (Minneap
muscular dystrophies. Pediatrics 126:538-545, 2010 Minn) 22:1829-1851, 2016. (6, Muscle and Neuromuscular Junction
29. Falsaperla R, Pratico AD, Ruggieri M, et al: Congenital muscular dystro- Disorders)
phy: From muscle to brain. Ital J Pediatr 42:78, 2016 53. Parikh S, Goldstein A, Koenig MK, et al: Diagnosis and management of
30. Jungbluth H, Treves S, Zorzato F, et al: Congenital myopathies: Disor- mitochondrial disease: A consensus statement from the Mitochondrial
ders of excitation-contraction coupling and muscle contraction. Nat Rev Medicine Society. Genet Med 17:689-701, 2015
Neurol 14:151-167, 2018 54. Hall JG, Vincent A, et al: Arthrogryposis. In: Darras BT, et al (ed): Neu-
31. Colombo I, Scoto M, Manzur AY, et al: Congenital myopathies: Natural romuscular Disorders of Infancy, Childhood, and Adolescence, ed 2,
history of a large pediatric cohort. Neurology 84:28-35, 2015 New York: Academic Press, 96-114, 2015
32. Litman RS, Griggs SM, Dowling JJ, Riazi S: Malignant hyperthermia sus- 55. Kowalczyk B, Felus J: Arthrogryposis: An update on clinical aspects, eti-
ceptibility and related diseases. Anesthesiology 128:159-167, 2018 ology, and treatment strategies. Arch Med Sci 12:10-24, 2016
33. Kang PB, Morrison L, Iannaccone ST, et al: Evidence-based guideline 56. Hall JG, Aldinger KA, Tanaka KI: Amyoplasia revisited. Am J Med Genet
summary: Evaluation, diagnosis, and management of congenital muscu- A 164A:700-730, 2014
lar dystrophy: Report of the Guideline Development Subcommittee of 57. Bamshad M, Jorde LB, Carey JC: A revised and extended classification of
the American Academy of Neurology and the Practice Issues Review the distal arthrogryposes. Am J Med Genet 65:277-281, 1996
Panel of the American Association of Neuromuscular & Electrodiagnos- 58. Sung SS, Brassington AM, Grannatt K, et al: Mutations in genes encod-
tic Medicine. Neurology 84:1369-1378, 2015 ing fast-twitch contractile proteins cause distal arthrogryposis syn-
34. North KN, Wang CH, Clarke N, et al: Approach to the diagnosis of con- dromes. Am J Hum Genet 72:681-690, 2003
genital myopathies. Neuromuscul Disord 24:97-116, 2014 59. Dieterich K, Quijano-Roy S, Monnier N, et al: The neuronal endopepti-
35. Rodríguez Cruz PM, Sewry C, Beeson D, et al: Congenital myopathies dase ECEL1 is associated with a distinct form of recessive distal arthrog-
with secondary neuromuscular transmission defects: A case report and ryposis. Hum Mol Genet 22:1483-1492, 2013
review of the literature. Neuromuscul Disord 24:1103-1110, 2014 60. Hoffmann K, Muller JS, Stricker S, et al: Escobar syndrome is a prenatal
36. Connolly AM, Pestronk A, Mehta S, Al Lozi M: Primary alpha myasthenia caused by disruption of the acetylcholine receptor fetal
sarcoglycan deficiency responsive to immunosuppression over three gamma subunit. Am J Hum Genet 79:303-312, 2006
years. Muscle and Nerve 21:1549-1553, 1998 61. Vogt J, Harrison BJ, Spearman H, et al: Mutation analysis of
37. Godfrey C, Escolar D, Brockington M, et al: Fukutin gene mutations in CHRNA1, CHRNB1, CHRND, and RAPSN genes in multiple pteryg-
steroid-responsive limb girdle muscular dystrophy. Ann Neurol ium syndrome/fetal akinesia patients. Am J Hum Genet 82:222-227,
60:603-610, 2006 2008
12 A.J. Fay

62. Yonekawa T, Nishino I: Ullrich congenital muscular dystrophy: Clinico- 74. Pham CB, Kratz JR, Jelin AC, Gelfand AA: Child neurology: Brachial
pathological features, natural history and pathomechanism(s). J Neurol plexus birth injury: What every neurologist needs to know. Neurology
Neurosurg Psychiatry 86:280-287, 2015 77:695-697, 2011
63. Engel AG, Shen XM, Selcen D, Sine SM: Congenital myasthenic syn- 75. Buterbaugh KL, Shah AS: The natural history and management of bra-
dromes: Pathogenesis, diagnosis, and treatment. Lancet Neurol 14:420- chial plexus birth palsy. Curr Rev Musculoskelet Med 9:418-426, 2016
434, 2015 76. Buchanan EP, Richardson R, Tse R: Isolated lower brachial plexus
64. Gay S, Dupuis D, Faivre L, et al: Severe neonatal non-dystrophic myoto- (Klumpke) palsy with compound arm presentation: Case report. J Hand
nia secondary to a novel mutation of the voltage-gated sodium channel Surg Am 38:1567-1570, 2013
(SCN4A) gene. Am J Med Genet A 146A:380-383, 2008 77. Falco NA, Eriksson E: Facial nerve palsy in the newborn: Incidence and
65. Matthews E, Manzur AY, Sud R, et al: Stridor as a neonatal presentation of outcome. Plast Reconstr Surg 85:1-4, 1990
skeletal muscle sodium channelopathy. Arch Neurol 68:127-129, 2011 78. Lahat E, Heyman E, Barkay A, Goldberg M: Asymmetric crying facies
66. M€annikk€ o R, Wong L, Tester DJ, et al: Dysfunction of NaV1.4, a skeletal and associated congenital anomalies: Prospective study and review of
muscle voltage-gated sodium channel, in sudden infant death syn- the literature. J Child Neurol 15:808-810, 2000
drome: A case-control study. Lancet 391:1483-1492, 2018 79. Tillman BN, Lesperance MM, Brinkmeier JV: Infantile Frey's syndrome.
67. Ohno K, Tsujino A, Brengman JM, et al: Choline acetyltransferase muta- Int J Pediatr Otorhinolaryngol 79:929-931, 2015
tions cause myasthenic syndrome associated with episodic apnea in 80. McKay VH, Touil LL, Jenkins D, Fattah AY: Managing the child with a
humans. Proc Natl Acad Sci USA 98:2017-2022, 2001 diagnosis of Moebius syndrome: More than meets the eye. Arch Dis
68. Mihaylova V, M€ uller JS, Vilchez JJ, et al: Clinical and molecular genetic Child 101:843-846, 2016
findings in COLQ-mutant congenital myasthenic syndromes. Brain 131 81. Carsi MB, Clarke AM, Clarke NP: Transient neonatal radial nerve palsy.
(Pt 3):747-759, 2008 A case series and review of the literature. J Hand Ther 28:212-215, 2015
69. Belaya K, Rodríguez Cruz PM, Liu WW, et al: Mutations in GMPPB 82. Alsubhi FS, Althunyan AM, Curtis CG, Clarke HM: Radial nerve palsy
cause congenital myasthenic syndrome and bridge myasthenic disorders in the newborn: A case series. CMAJ 183:1367-1370, 2011
with dystroglycanopathies. Brain 138(Pt 9):2493-2504, 2015 83. www.neuromuscular.edu. Accessed 7/3/2018.
70. Jovandaric MZ, Despotovic DJ, Jesic MM, Jesic MD: Neonatal outcome 84. Bodamer OA, Scott CR, Giugliani R, Pompe Disease Newborn Screening
in pregnancies with autoimmune myasthenia gravis. Fetal Pediatr Pathol Working Group: Newborn screening for Pompe disease. Pediatrics 140
35:167-172, 2016 (Suppl 1):S4-S13, 2017. https://doi.org/10.1542/peds.2016-0280C
71. Jackson L, Madan-Khetarpal S, Naik M, et al: Infant botulism in the very 85. Ankala A, da Silva C, Gualandi F, et al: A comprehensive genomic
young neonate: A case series. AJP Rep 7:e163-e166, 2017 approach for neuromuscular diseases gives a high diagnostic yield. Ann
72. Yu PA, Lin NH, Mahon BE, et al: Safety and improved clinical outcomes Neurol 77:206-214, 2015
in patients treated with new equine-derived heptavalent botulinum anti- 86. Cummings BB, Marshall JL, Tukiainen T, et al: Improving genetic diag-
toxin. Clin Infect Dis 66(Suppl_1):S57-S64, 2017 nosis in Mendelian disease with transcriptome sequencing. Sci Transl
73. Arnon SS, Schechter R, Maslanka SE, et al: Human botulism immune Med 9, 2017. https://doi.org/10.1126/scitranslmed.aal5209
globulin for the treatment of infant botulism. N Engl J Med 354:462- 87. Mendell JR, Rodino-Klapac LR: Duchenne muscular dystrophy:
471, 2006 CRISPR/Cas9 treatment. Cell Res 26:513-514, 2016

You might also like