You are on page 1of 20

Handbook of Clinical Neurology, Vol.

113 (3rd series)


Pediatric Neurology Part III
O. Dulac, M. Lassonde, and H.B. Sarnat, Editors
© 2013 Elsevier B.V. All rights reserved

Chapter 146

Hereditary motor-sensory, motor, and sensory


neuropathies in childhood

PIERRE LANDRIEU1*, JONATHAN BAETS2,3, AND PETER DE JONGHE2,3


1
Department of Paediatric Neurology, Universit Paris Sud, Bictre Hospital, Paris, France
2
Neurogenetics Group, VIB-Department of Molecular Genetics and Laboratory of Neurogenetics,
Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
3
Division of Neurology, University Hospital Antwerp, Antwerp, Belgium

INTRODUCTION hereditary diseases are, therefore, considered here in the


first instance.
Hereditary neuropathies are a heterogeneous group of
Categories of neurons mostly involved, according to
disorders that have an overall prevalence of 1 in 2500
clinical and standard EMG examinations, is the most
individuals. There are no good estimations of their prev-
classical element for the initial delineation of genetic
alence in the pediatric population.
neuropathies, in clinical neurology. A subdivision can
There are many ways of categorizing the hereditary be made into three main categories (Dyck, 1984). The
peripheral neuropathies, depending on the starting point
first, and also most prevalent, group is that of hereditary
(clinical presentation, biological mechanism, causal geno-
motor and sensory neuropathy (HMSN). More uncom-
mic rearrangement, and so on), as well as the hierarchy of
mon are the hereditary motor neuropathies (HMN),
the main parameters. If the clinical presentation is logically
characterized by selective involvement of the peripheral
chosen as the starting point in a clinical handbook, the hier-
motor neurons, and their counterparts, the hereditary
archy of the pertinent clinical parameters, both in neuro-
sensory and autonomic neuropathies (HSAN), charac-
logical practice and for classification purposes, must be
terized by a selective involvement of the sensory and/
briefly discussed (Table 146.1). or autonomic neurons. Though each group represents
Primary neuropathies are opposed to the various
a distinct disease spectrum, some areas of overlap exist.
situations in which the neuropathy occurs either as a
In an index case, the neuropathy can initially appear to be
late complication of a chronic hereditary disease, or as
restricted to a specific nerve fiber type, both clinically
a feature relatively secondary in the diagnosis of a multi-
and physiologically, but may extend later to other cate-
systemic congenital syndrome. The present chapter
gories of fiber. In the same family, other cases can pre-
focuses mainly on those entities in which the neuropathy
sent right away as a neuropathy affecting several neuron
is either the only presenting symptom or an early/major
populations.
clinical element. However, for the neurologist facing an
insidious polyneuropathy, the first concern is to ensure
PATHOPHYSIOLOGY OF NERVE
that it is not the unusual presenting symptom of a
FIBER DEGENERATION:
general disease, whether acquired or hereditary. This
ELECTROPHYSIOLOGICAL
rare situation is suspected in two circumstances: (1)
EXAMINATION
the neuropathy exhibits an unusual course, that is, a reg-
ular aggravation, rarely observed in the initial course of Measurement of nerve conduction velocities (NCVs)
primary neuropathies; (2) there are unusual accompany- and needle electromyography (EMG) in children are usu-
ing symptoms or signs in other organs (brain, skin, diges- ally limited to basic measurements, due to restricted
tive tract, heart, etc.). Neuropathies secondary to general tolerance. It is, however, possible to get a reliable

*Correspondence to: Pierre Landrieu, Service de Neurologie Pédiatrique, CHU Paris sud-Hôpital Bicêtre, 94270 F Paris, France.
E-mail: pierre.landrieu@bct.aphp.fr
1414 P. LANDRIEU ET AL.
Table 146.1 of the neuropathy, whether axonal or demyelinating
Most pertinent clinical parameters for categorizing genetic in nature, or to severe wasting of the marker muscle.
peripheral neuropathies ● The clinical experience reveals many neuropathies
with an intermediate electrophysiology (Davis et
Primary versus secondary occurrence al.,1978): in some patients, NCVs fall within a range
Category of peripheral neurons involved of 25–45 m/s and show an overlap between CMT1
Pathophysiology of the nerve fiber degeneration
and CMT. In the same way, in a single family some
Distinct neuropathological markers
patients may present electrophysiologically as CMT1
Apparent mode of inheritance
Isolated versus syndromic forms while others present as CMT2. Some genetic defects
Period of life are known to underlie these phenotypes, and for
many authors this “intermediate CMT” category
remains clinically useful, even if it does not neces-
sarily refer to a precise pathological interpretation.
measurement even in young children which will help in
● In the child, and especially in the infant, many
orienting the genetic diagnosis. Since sensory signs on
genetic neuropathies can be viewed as a develop-
clinical examination can be very mild, especially in
mental rather than a degenerative disorder. Abnor-
young individuals, NCV studies can be the sole tool to
mal development can affect the myelination
clearly prove or exclude the involvement of sensory neu-
without evidence of segmental demyelination, the
rons. Basic neurophysiological parameters in relation to
axonal growth without evidence of dying back
the growth of the peripheral nerve have been reported in
degeneration, or both processes simultaneously,
several studies (Parano et al., 1993; Garcı́a et al., 2000).
making the NCV values more complex to relate to
NCVs and EMG have been major parameters for
the neuropathological findings (Fig. 146.1).
delineating the various forms of hereditary motor and
● Due to the availability of routine molecular testing
sensory neuropathies in adults. When electrophysiologi-
for several of the known genes, sural nerve biopsy
cal and nerve biopsy studies became routinely available
has become obsolete in most patients, making the
in clinical practice, their results showed a coherent pat-
confrontation between neuropathological findings
tern: prominent Schwann cell pathology leads to segmen-
and NCVs increasingly less often needed in the clin-
tal demyelination and results in marked NCV slowing,
ical discussion.
whereas purely axonal degeneration, when respecting
a sufficient proportion of large myelinated nerve fibers, Distinct electrophysiological features can be also
results in near-normal NCVs. found in the two other groups of hereditary neuropa-
Thus, the most universal and simple parameter for thies. HMN is characterized by a pure motor neuropathy
describing HMSN is the measurement of NCVs, allow-
ing subdivision into two categories (Harding and
Thomas, 1980):
1. demyelinating neuropathies with clearly reduced
NCVs, typically defined as slowing of the motor
NCV for the median nerve < 38 m/s (HMSN1)
2. axonal neuropathies characterized by normal or
moderately slowed NCV > 38 m/s for the motor
median nerve (HMSN2).
This cut-off value of 38 m/s for motor NCV in the
upper limbs has mainly been validated for adult patients
with HMSN, but from experience this subdivision can
also be used in children as young as 1 year. However,
the simplistic superposition of the NCV values with Fig. 146.1. Developmental anomalies in a syndromic congen-
the underlying pathological mechanism is only partially ital neuropathy due to a SOX10 neomutation, in a 6-week-old
operative, especially in children’s hereditary neuropa- infant (superficial branch of the lateral popliteal nerve):
thies, for several reasons: atypical partitioning of the nerve trunk in numerous minifasci-
cles; reduced density of large myelinated axons and hypo-
● It is difficult to distinguish the axonal from the myelination of the myelinated fibers (right: control nerve;
demyelinating type when the action potentials can- original magnification  400). Motor and sensory NCVs
not be elicited or are markedly diminished upon were in the range 5–8 m/s in the median nerve (Pingault
stimulation. This could either be due to the severity et al., 2000).
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1415
of the axonal type, with normal or only slightly reduced should be taken when interpreting this information in a
motor NCV. However, minor sensory involvement has given pedigree. Especially in early onset neuropathies,
been reported in some HMN cases (Irobi et al., 2006). there is a substantial degree of de novo dominant muta-
HSAN phenotypes, on the other hand, exhibit a broad tion that in essence presents as an isolated patient
range of electrophysiological features. Often a predom- within the family. In small kinships this can be difficult
inantly sensory axonal neuropathy can be found but to distinguish from recessive forms that are also com-
occasional electrophysiological signs of demyelination mon in early onset Charcot–Marie–Tooth (CMT) dis-
can be seen as well. Electrophysiological abnormalities ease. On the other hand, if the vast majority of gene
in motor nerves are sometimes present, illustrating the defects behave either in a dominant or a recessive
overlap between HSAN and HMSN. way, a few notable exceptions need to be taken into
Distinct neuropathological markers. Since EMG find- account for genes that can support both inheritance
ings can accurately predict the underlying primary patho- patterns.
logical process in most hereditary neuropathies, the need Syndromic versus nonsyndromic forms is another
for a nerve biopsy becomes limited mostly to the rare major element to consider in pediatrics. A syndrome is
cases in which diagnosis of an acquired (inflammatory) the nonrandom association of clinical and paraclinical ele-
neuropathy remains uncertain. However, it is well known ments, not linked to each other by a sequential process,
that the morphological abnormalities in hereditary neu- allowing identification of a clinical entity. A congenital
ropathies are much more diverse and sometimes patho- syndrome is a syndrome in which at least one element is
gnomonic in the very young age group, compared to a developmental (i.e. with fetal onset) anomaly. As the ner-
the more monomorphic abnormalities observed in late- vous system is a complex, multisystemic organ, two major
onset forms (Schr€ oder, 2006). For example, myelin out- types of syndromes can be delineated in neurology:
foldings or redundant myelin loops can sometimes orient
1. Purely neurological syndromes, associating dys-
towards a specific group of genes (Fig. 146.2).
functions in different nerve cell populations, are
Inheritance pattern in the family is an important ele-
encountered both in adult and pediatric neurology.
ment for further subdivision of hereditary neuropathies.
They are numerous and generally classified in ref-
As a rule, inherited peripheral neuropathies are trans-
erence to the main initial symptom.
mitted as monogenic disorders with high penetrance.
2. Syndromes associating both neurological and extra-
Autosomal dominant (AD), X-linked as well as autoso-
neurological anomalies are more specific to the
mal recessive (AR) forms have been described. Caution
pediatric population and very rare for most.
The physician must be aware that no clear-cut frontier
exists between syndromic and non-syndromic disorders,
thus examining and re-examining the patient remains a
basic law in pediatrics. Some neuropathies initially con-
sidered as nonsyndromic can exhibit new findings along
their course, sometimes including extraneurological
ones. For practical reasons, some of them are here main-
tained in the category of primary neuropathies.
Period of life is a clinical category familiar to pedia-
tricians, as each exhibits its specific range of diseases.
As regard the peripheral neuropathies, the most relevant
pediatric category is the congenital neuropathies, i.e.
expressing in the perinatal period or in early infancy.
Many of them are syndromic, very specific to pediatric
practice and linked to genes different from those of clas-
sical neuropathies.

NEUROLOGICAL AND GENETIC


CLASSIFICATIONS OF HEREDITARY
NEUROPATHIES
Fig. 146.2. Myelin outfoldings in a case of demyelinating
CMT4. This finding points to several candidate genes, in the No unifying classification or diagnostic algorithm is capa-
first place those coding for myotubularin-related proteins ble of taking into account at the same time all the relevant
(bar ¼ 1 mm, Ax ¼ axon). clinical elements, permitting the regular introduction of
1416 P. LANDRIEU ET AL.
new genetic entities and staying simple. Historically, neuropathies have been distinguished from spinal muscu-
hereditary neuropathies were first classified by neurolo- lar atrophies clinically by the distal predominance and
gists according to neurological parameters, then were clas- genetically by the usual AD transmission, but unavoidable
sified by geneticists according to succinct neurological overlaps exist between the two categories.
parameters in combination with novel genetic findings.
The classification proposed here (Table 146.2) is a version FROM PHENOTYPE TO GENOTYPE,
of the classification more universally used, adapted for AND VICE VERSA
pediatric needs. CMT, in the genetically based classifica-
tion, is the imperfect counterpart of HMSN in the previ- Though a correct clinical evaluation is capable of orient-
ous, clinically oriented classification, which explains ing the genetic testing, the neurologist needs to take into
several illogical points. For example, HMSN type III, account the extensive genetic heterogeneity that under-
which referred to a severe, early demyelinating neuropa- lies any peripheral neuropathy. Conversely, for a given
thy in a patient with nonaffected parents, has not been gene, he must be aware that some mutations can be
retained in the CMT classification. So its transposition into responsible for distinct clinical presentations, including
the CMT subtypes has to be found either in the recessive some without a neuropathic trait.
forms (CMT4) with slow NCVs, in the severe CMT1 result-
ing from a de novo dominant mutation, or in the eponymic I RULING OUT A NEUROPATHY
Dejerine–Sottas disease that some authors continue to SECONDARY TO A HEREDITARY
use outside of any classification. Hereditary motor GENERAL DISEASE
Monogenic metabolic diseases
Table 146.2
A “metabolic disease”, for the physician, refers to any
General classification of hereditary peripheral disease in which the primary diagnosis is based upon bio-
neuropathies in childhood
chemical markers in body fluids and/or enzymological
I Hereditary neuropathies secondary to general diseases markers in cells amenable to investigation. They are usu-
II Primary neuropathies ally classified according to the deficient biological
IIa Hereditary motor sensory neuropathies function.
(HMSN) ¼ CMT In only a few cases, the peripheral neuropathy is a
CMT1 (slow NCV, AD): a, b, c major or even the first presenting feature. The paradigm
CMT2 (normal/intermediary NCV) is metachromatic leukodystrophy, in which some juve-
CMT4 (AR)
nile forms present as a progressive demyelinating poly-
CMT4 with slow NCV
neuropathy. Actually, almost all metabolic diseases
CMT4 with normal/intermediary NCV
CMT X (various types of X-linked HMSN) resulting in a neurodegenerative disorder can, sooner
IIb Distal hereditary motor neuropathies (HMN) or later, involve the lower motor neuron and the auto-
HMN II nomic or sensory ganglia (see Ch. 148, Metabolic neurop-
HMN IV athies and myopathies).
HMN V/Silver syndrome
HMN VI LYSOSOMAL STORAGE DISEASES (SPECIFIC HYDROLASE
HMN VII DEFICIENCIES) COMPLICATED BY PERIPHERAL
ALS4 NEUROPATHY
(spinal muscular atrophies SMA)
HMN (distal) See Table 146.3 (see also Chs 164, 174–177).
IIc Hereditary sensory (þ/ dysautonomic)
neuropathies (HSN) OTHER LYSOSOMAL DISEASES
HSAN I (AD):
HSAN II (AR, familial dysautonomia, Riley–Day Niemann—Pick disease type C results from an aberrant
syndrome) traffic of exogenous cholesterol. Mutations in NPC1,
HSAN III (AR congenital sensory neuropathy) an integral membrane glycoprotein of the late endo-
HSAN IV (AR, HSN with anhiydrosis) some, or in NPC2, a small endoluminal protein of lyso-
HSAN V somes, result in the lysosomal storage of a complex
III Syndromic neuropathies, congenital neuropathies.
mixture of unesterified cholesterol and glycosphingoli-
Syndromic neuropathies with late onset
pids. Neuronoaxonal peripheral degeneration is a late,
Congenital neuropathies
unconstant complication.
AD, autosomal dominant; AR, autosomal recessive; NCV, nerve con- Salla disease is a free sialic acid storage disorder
duction velocities. resulting from mutations in a presumptive sialic acid
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1417
Table 146.3
Lysosomal storage diseases (specific hydrolase deficiencies) complicated by peripheral neuropathy

Peripheral
neuropathy Biochemical
Disease characteristics Other organs markers (urine) Deficient enzyme

Metachromatic Demyelinating CNS Sulfocerebrosides Arylsulfatase A


leukodystrophy
Krabbe disease Demyelinating CNS Galactocerebroside b-Galactosylceramidase
Niemann–Pick A Demyelinating CNS, liver Sphingomyelinase
Gaucher Axonal CNS, macrophages Glucocerebroside b-Glucocerebrosidase
(type 2)
Tay–Sachs Distal axonal CNS b -Hexosaminidase
(late forms)
Fabry Neuropathic pains Heart, kidney a-Galactosidase A
Gut
b-Mannosidosis Demyelinating CNS, skin, bone Mannose-rich b-Mannosidase
oligosaccharides
a-Mannosidosis Immune, CNS, Mannose-rich a-Mannosidase
hearing, bone oligosaccharides
Mucopolysaccharidoses Carpal tunnel Bone Specific MPS Specific hydrolases
(MPS 1–7) syndrome
Schindler/Kanzaki Neuroaxonal CNS O-linked a-N-acetylgalactosaminidase
dystrophy Skin glycopeptides

transport protein. Sialic aciduria is the main biochemical picture progressively includes retinitis pigmentosa, blind-
marker. Peripheral neuropathy, a secondary manifesta- ness, anosmia, deafness, and ataxia. The peripheral neu-
tion compared with the severe mental retardation, is at ropathy rarely begins in childhood. It is dominated by
least in part due to demyelination (Varho et al., 2000). sensory features and results both from a Schwann cell dis-
order and from axonal degeneration.
PEROXISOMAL DISORDERS In deficiency of peroxisomal D-bifunctional protein,
Disorders of peroxisome biogenesis, of which > 12 the progressive neurodegenerative syndrome begins in
genetic subtypes have been identified by complementa- childhood. A peripheral neuropathy is present which
tion studies, are responsible for a large phenotypic results in a severe reduction of myelinated fibers in
range: Zellweger syndrome, neonatal adrenoleukodys- the nerve biopsy (Schr€oder et al., 2004).
trophy (ALD), and infantile Refsum disease, among
others (see Ch. 163). A peripheral component could be
MITOCHONDRIAL OXPHOS DEFICIENCY
partly responsible for the major hypotonia frequently
observed in these babies (Baumgartner et al., 1998), (see also Ch. 168)
but a peripheral neuropathy has not been clearly demon- A peripheral neuropathy is frequently encountered in
strated in any of the few neuropathological studies. the course of the mitochondrial cytopathies, character-
In X linked ALD, due to mutations in ABCD1, a gene ized by unique or multiple mtDNA deletions, which bear
that encodes an ABC half-transporter involved in the the classical denominations of Kearns–Sayre syndrome,
transmembranous traffic of VLCFA, a peripheral neu- progressive external ophthalmoplegia þ, multiple
ropathy, mixing axonal degeneration and demyelination, mtDNA deletions syndromes, etc. Most of them occur
is sometimes associated with the degeneration of the in a sporadic way, whereas rare cases are due to inherited
long tracts of the CNS. This picture of adrenomyelo- mutations affecting nuclear genes involved in the repli-
neuropathy usually begins beyond adolescence. cation of mtDNA (POLG; Twinkle, ANT1, OPA1). The
Classic Refsum disease shows an accumulation of phy- clinical onset of the neuropathy is rarely before the juve-
tanic acid in plasma- and lipid-containing tissues. Most nile period. Frequently the sensory fibers are more
cases are due to a deficiency in a peroxisomal enzyme, involved clinically than the motor fibers. The pathologi-
phytanoyl-CoA 2-hydroxylase (PAHX). The clinical cal picture is dominated by axonal degeneration, but
1418 P. LANDRIEU ET AL.
segmental demyelination is also present (Santoro et al., dehydrogenase (PDH) deficiency and in diseases causing
2006). In some cases, however, the neuropathy is a major a secondary deficiency of PDH (see Ch. 169) or of the
presenting feature, prompting some authors to delineate mitochondrial respiratory chain. Here the neuropathy
separate entities like SANDO (sensory ataxic neuropa- is usually a mixture of axonal degeneration and Schwann
thy, dysarthria, and ophthalmoparesis) and MIRAS cell dysfunction. (3) Multiple factors are involved in
(mitochondrial recessive ataxia syndrome). more complex situations such as disorders of folate
A few syndromes begin in infancy. Infantile-onset remethylation, the most frequent being methylenetetra-
spinocerebellar ataxia (IOSCA), an autosomal recessive hydrofolate (MTHF) reductase deficiency (see Ch. 184).
disorder originally described in Finland, includes a
peripheral axonal neuropathy with a sensory predomi- DISORDERS OF POSTTRANSLATIONAL GLYCOSYLATION
nance. It has been related to special mutations of Twin-
kle (a helicase specific for mtDNA), offering a good The carbohydrate-deficient glycoprotein (CDG) syn-
example that the same gene can carry both dominant dromes are a family of autosomal recessive disorders
and recessive mutations which result in different pheno- in relation to defects of the protein N glycosylation,
types (Nikali et al., 2005). which can be observed with isoelectric focusing of serum
In single base mutations of mtDNA, transmitted transferrine as the most widely used diagnostic test
through maternal lineage, a peripheral neuropathy does (see Ch. 179). The most frequent form, CDG-Ia or
not occur as an isolated presenting feature. However, it phosphomannomutase-2 deficiency, affects many
can be a part of the clinical picture associated both with organs, including the nervous system, in a not very pre-
the most frequent mtDNA mutations, such as the dictable way. Neurological forms are characterized by
8993 T > C (Leigh /NARP mutation), the 3243 A > G psychomotor retardation, strabismus, cerebellar hypo-
(MELAS), the 8344A > G (MERRF), and with many plasia and atrophy, retinitis pigmentosa, and stroke-like
rarer mutations. episodes. A slow lower-limb sensory-motor axonal neu-
Deficiencies of respiratory chain complexes related ropathy is an inconstant complication, rarely observed
to mutations in nuclear genes also result in various clin- clinically before adolescence. In a series of sural nerve
ical pictures, some of them including a peripheral neu- biopsies, the most striking findings were attenuation
ropathy. No isolated deficiency of a specific complex of myelin sheaths, multivacuolar myelinoid bodies in
seems to be particularly responsible for a peripheral Schwann cells, and axonal degeneration of scattered
nerve degeneration. In multiple respiratory chain com- unmyelinated fibers (Nordborg et al., 1991).
plex deficiency due to a depletion of mtDNA, a periph-
eral neuropathy has been mostly described in those SPECIFIC SYSTEMIC CARRIERS
related to POLG mutations, some of them resembling a-Tocopherol transfer protein. Various recessive muta-
CMT II (Harrower et al., 2008). In other forms linked tions of the TTPA gene are responsible for AVED (ataxia
to unidentified nuclear genes, presumably coding for with vitamin E deficiency), a disease which reveals itself
proteins involved in the ribosomal translation or in other usually in the late infantile or juvenile period with ataxia,
mitochondrial regulations, the peripheral neuropathy is dystonia, and retinopathy (see Ch. 184). In most patients,
rarely but possibly a presenting feature, including in the peripheral neuropathy is clinically absent or moder-
severe neonatal forms (Ferreiro-Barros et al., 2008). ate, with normal NCVs and sensory potentials. In rare
cases, however, the peripheral neuropathy is an overt
INTERMEDIARY METABOLISMS OF AMINO ACIDS AND clinical feature, presenting as a sensory, sensory-motor,
OTHER SIMPLE SUBSTRATES or distal motor axonopathy (Zouari et al., 1998; Fusco
et al., 2008). In the neurological complications secondary
Various hereditary diseases involve the intermediary to diseases resulting in malabsorption or deficient trans-
metabolism of simple molecules and are responsible port of all liposoluble vitamins (cholestasis, mucovisci-
for the diffusion of abnormal substrates in body fluids. dosis, A-b-lipoproteinemia), a severe peripheral axonal
Sometimes peripheral nerve lesions can occur as a major neuropathy was more often observed before the era of
presenting symptom. Various mechanisms can be systematic preventive measures.
involved: (1) Direct toxicity of abnormal metabolites
on the neuron or on its axonal extension, usually result-
Monogenic degenerative diseases
ing in a sensory-motor axonopathy. This is probably the
recognizable by nonbiochemical markers
major phenomenon in hepatic porphyrias (neurotoxicity
of d-aminolevulinic acid). (2) Energetic crisis in the Chediak–Higashi disease is dominated by a disorder of
peripheral nerve cell, resulting directly from the meta- the immune system and recognized by the presence of
bolic defect. This is probably the case in pyruvate special inclusions in leukocytes. Causal mutations are
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1419
to be found in the Beige gene (CHS1/LYST), a 430 kD mutations and many of them will not be transmitted,
cytosolic protein of poorly understood function. A slow due to the lack of reproductive fitness in severely
peripheral neuropathy of the axonal type frequently affected patients. Recessive forms tend to start early
develops during the course of the disease. in childhood and run a more severe course than the dom-
Disorders of DNA repair, whatever their type, are inant forms, and present with delayed motor milestones,
frequently complicated by a neurological degradation loss of ambulation, and variable associated signs
including a peripheral neuropathy (see Ch. 167). In some (Dubourg et al., 2006). In the European population,
disorders (e.g., xeroderma pigmentosum, Cockayne syn- AR forms of HMSN account for less than 10% of fam-
drome), the diagnosis is facilitated by tests measuring ilies, while in communities with a high percentage of con-
the process of excision-repair of single DNA strands sanguineous marriages this proportion may rise to over
on cultured cells submitted to mutagenic stress such 40% (Dubourg et al., 2006). Overall there is a lack of
as UV irradiation. Though the microarray technology large-scale studies looking into the mutation distribution
will be capable of measuring specific DNA repair activ- and frequency in a pediatric population, making diffi-
ities, no generic test is currently available in most cases. cult any prediction regarding the yield of mutation
The diagnosis remains based upon investigation of can- screening.
didate genes in syndromic neuropathies with some The molecular investigation becomes increasingly
orienting features (see below). important not only for a correct diagnosis, but also
for genetic counseling, pre-implantation diagnosis and
II PRIMARY NEUROPATHIES eventually genotype-specific therapies. However, hered-
itary neuropathies show a bewildering genetic heteroge-
IIa Hereditary motor sensory neuropathies,
neity and routine diagnosis is only available for a few
Charcot–Marie–Tooth disease
genes. So far over 40 genes have been implicated and this
The majority of hereditary neuropathies, both in adults number is only expected to grow since more loci have
and in children, present as mixed motor and sensory neu- been identified. Screening of large cohorts of well-
ropathies (HMSN). All these forms have been exten- defined phenotypes usually shows that a considerable
sively described in adults, whereas studies specifically number of cases are not explained by mutations in any
focusing on children and adolescents are scarce or non- of the known relevant genes. Fortunately, easily accessi-
existent. The earliest signs and symptoms do not seem to ble parameters such as mode of inheritance, clinical fea-
differ between children and adults but the pace of pro- tures, and additional characteristics often orient towards
gression is clearly different. Inherited neuropathies in a more restricted subgroup, limiting the number of
the pediatric age group tend to be more severe, some- potential gene defects to be tested.
times resulting in early death and often compromising
the reproductive fitness of the more severely affected
AD DEMYELINATING NEUROPATHIES (CMT1)
patients. The early signs include areflexia, problems in
heel walking, atrophy of intrinsic foot muscles, clawing CMT1A is due to a 1.5 Mb duplication on chromosome
of toes, pes cavus or pes varus, Achilles tendon shorten- 17p12 containing the peripheral myelin protein 22 gene.
ing, peroneal weakness, and stocking hypoesthesia when PMP22 is an integral membrane protein produced by
the sensory axons are involved. Sensory involvement is Schwann cells, where it plays a crucial role in the devel-
often difficult to detect clinically, especially in children, opment and maintenance of compact myelin. CMT1A
but can be documented by electrophysiological studies. accounts for the vast majority of CMT patients: up to
Special clinical attention needs to be given to any addi- 70% of all CMT patients if AD inheritance is present
tional features such as pyramidal tract signs, ulcerations, and close to 50% of all CMT patients (Houlden and
and autonomic symptoms. Reilly, 2006; Szigeti et al., 2006). CMT1A is the only
Occasionally, the expression of a HMSN can be CMT phenotype that has been systematically studied
acutely provoked by exposure to neurotoxic drugs. in a small cohort of 12 young children (Berciano et al.,
Over the years several asymptomatic children have 2003). In these at-risk infants (one parent had confirmed
been reported with different forms of inherited neurop- CMT1A), a molecular diagnosis was made irrespective
athies who progressed to almost complete tetraplegia of the presence of symptoms or signs of neuropathy.
within days or weeks after exposure to vincristine All children developed signs including areflexia,
(Cil et al., 2009). problems in heel walking, atrophy of intrinsic foot mus-
A positive family history is the classical hallmark of cles, clawing of toes, pes cavus or pes varus, Achilles
HMSN, but its absence does not argue against the tendon shortening, peroneal weakness, and stocking
genetic nature of the neuropathy, especially in children. hypoesthesia. Not every sign was present in all children.
Many isolated cases represent de novo dominant Despite these signs, only 3 out of 12 children became
1420 P. LANDRIEU ET AL.
symptomatic at the age of 10 years. Serial electrophysi- CMT1C, related to mutations in LITAF
ological studies, including motor and sensory NCVs, (lipopolysaccharide-induced tumor necrosis factor-a is
motor distal latencies and F-wave latencies, showed a protein of uncertain role that participates in the break-
abnormalities in all children by the age of 2 years. Inter- down of various substances), exhibits a classic CMT1
estingly, NCVs reached their maximum, which are still phenotype. CMT1D is related to dominant mutations
severely reduced compared to unaffected children, at in EGR2 (early growth response 2, a transcription factor
the age of 5 years and remained stable from then on, a with three tandem C2H2-type zinc fingers, that controls
finding that suggests a combination of both dysmyelina- expression of myelin protein genes); CMT1F, related to
tion (a developmental problem) and demyelination (a mutations in NEFL (neurofilament light chain polypep-
degenerative process). Within the second decade most tide), is apparently rare and little is known about its pre-
CMT1A patients develop symptoms. CMT1A is often sentation in children (Houlden and Reilly, 2006). NEFL
mild or moderately severe and progresses only slowly mutations are associated with a fairly broad phenotypic
over many years. spectrum of AD CMT including early onset forms
Given the high prevalence of CMT1A and the com- (Jordanova et al., 2003) and axonal/intermediate CMT
mon occurrence of de novo mutations, molecular variants (CMT2E).
genetic testing should be considered in all children with Despite possible differences in average forms, in clin-
a demyelinating neuropathy without another evident ical practice it is impossible to distinguish between
cause. CMT1A, CMT1B, CMT1C, CMT1D, and CMT1F. Given
No specific treatment is currently available. How- the relative frequencies of these AD forms, it is advis-
ever, in a transgenic mouse model that shows a huge able to start with screening for the CMT1A duplication,
overexpression of PMP22, a study has demonstrated a followed by the analysis of the MPZ gene and the
beneficial effect of ascorbic acid, possibly by lowering PMP22 gene. Performing mutational analysis of the
PMP22 levels (Passage et al., 2004). Several clinical trials rarely involved genes LITAF, EGR2, and NEFL is
of vitamin C in CMT1A are still ongoing (Pareyson et al., restricted to research laboratories.
2006). In children, a recent trial reported no effect over a
period of 1 year (Burns et al., 2009).
CONGENITAL HYPOMYELINATING NEUROPATHY
Hereditary neuropathy with liability to pressure
AND DEJERINE–SOTTAS DISEASE
palsies is caused by the reciprocal deletion of the same
region on chromosome 17 where the CMT1A duplica- Congenital hypomyelinating neuropathy (CHN) corre-
tion lies. This results in the loss of one copy of the sponds to a severe congenital or very early childhood-
PMP22 gene. Symptoms of this episodic disorder are onset neuropathy resulting in pronounced hypotonia
painless, recurrent, focal motor and/or sensory periph- and associated breathing and feeding problems. Electro-
eral neuropathies. Onset of this disorder is usually in physiology shows severely slowed to absent NCVs. Neu-
adolescence but it can be seen occasionally in younger ropathology reveals markedly reduced or even absent
children. Some degree of phenotypic overlap with myelin, suggesting a developmental problem of the mye-
CMT1 exists, obscuring the clinical diagnosis in some lin’s formation. CHN is closely related to Dejerine–
patients (Chance, 2006). Sottas disease (DSN), a severe demyelinating neuropa-
CMT1B, which represents around 5% of CMT1 thy with somewhat later onset in childhood but usually
(Szigeti et al., 2006), is due to mutations in MPZ. Myelin before the age of 5 years (Houlden and Reilly, 2006).
protein zero is a homophilic adhesion molecule and a DSN, for some, is strictly defined by findings including
major structural protein of peripheral myelin. CMT1B early onset, severely slowed NCVs and high protein con-
also starts in the first or second decade of life. Studies tent in the spinal fluid. Others have loosely applied the
specifically aimed at children are not available. DSN designation to any early-onset severe CMT, regard-
A follow-up study in the family that was instrumental less of the underlying pathology and NCV findings.
in localizing and cloning the MPZ gene showed that Whether or not CHN and DSN can be considered as sep-
NCVs were only marginally decreased over many years, arate disease entities, molecular genetics has shown that
despite a clear increase in disease severity (Bird et al., both are in fact the severe end of the CMT1 (or HMSN1)
1997). This again highlights that NCVs are a diagnosis disease spectrum. When identified, the most frequent
marker but a poor parameter for monitoring disease pro- molecular genetic causes are monoallelic mutations in
gression. Occasionally, CMT1B presents as a CMT2 phe- MPZ (myelin protein zero), PMP22 or EGR2 (early
notype and is then often associated with abnormalities of growth response 2) (Smit et al., 2008). Occurring de novo
the pupil reflexes. However, this form almost exclu- in patients without reproductive fitness, these dominant
sively starts in adulthood (Marrosu et al., 1998; De mutations usually appear as isolated cases. On the other
Jonghe et al., 1999). hand, several recessive demyelinating CMT (CMT4)
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1421
tend to be associated with severe early-onset phenotypes specific enough to orient screening in the direction
resembling CHN or DSN as well, sometimes occurring in of MTMRs (Dubourg et al., 2006). Early-onset glau-
several sibs (see next section). coma has been described as an associated feature in
SBF2 mutation carriers (Azzedine et al., 2003).
AR HEREDITARY MOTOR SENSORY NEUROPATHIES ● HMSN-Lom (hereditary motor and sensory neurop-
(CMT4) athy Lom type) is caused by mutations in NDRG1
(N-myc downstream regulated gene 1, a member
Most AR CMT subtypes display an onset in early infancy,
of the a/b hydrolase superfamily, is a cytoplasmic
either congenital or at least resulting in delayed motor
protein involved in cell growth and differentiation).
milestones (Dubourg et al., 2006). With a few notable
This recessive phenotype is almost exclusively
exceptions (Bernard et al., 2006), the majority of recessive
restricted to Gypsies of Eastern European descent,
CMT forms are demyelinating and are thus characterized
the vast majority of patients carrying the same foun-
by slow NCVs. They are grouped together under the cat-
der mutation. HMSN-Lom has an onset in the first
egory CMT4 and are numbered accordingly. Due to the
decade of life and weakness often spreads to the
rarity of these forms it is difficult to make reliable esti-
proximal parts of the limbs. NCVs are in the range
mates of the relative contribution of the several genes.
of 10–20 m/s. A suggestive additional finding is
These forms are more prominent in countries or ethnic
the presence of sensorineural deafness.
groups with a high degree of consanguinity. Their relative
● Mutations in FGD4 (FGD1-related F-actin binding
frequencies may differ from one population to another
protein is a nucleotide exchange factor specific to
due to the occurrence of founder mutations. Although
a RhoGTPase that plays a key role in mediating actin
a broad overlap exists between the different phenotypes,
cytoskeleton changes during morphogenesis) can
certain distinctive characteristics are of importance:
give rise to an early infancy-onset phenotype result-
● Mutations in GDAP1 (ganglioside-induced ing in delayed motor milestones in some patients.
differentiation-associated protein-1, a regulator of Slow progression, however, has been reported. Elec-
the mitochondrial network) are known to cause a trophysiology often shows very slow NCVs < 15 m/s,
wide variety of CMT forms including AR demyelin- and myelin outfoldings can be seen on neuropathol-
ating CMT but also AR axonal, intermediate forms ogy (Stendel et al., 2007; Fabrizi et al., 2009).
and even rare AD forms. The demyelinating form ● Mutations in EGR2 can behave both as dominant
(CMT4A) has an onset before the age of 3 years, (CMT1D, see above) and as recessive traits. In both
with a rapidly progressing and severe weakness instances onset can be congenital with markedly
often resulting in loss of ambulation. NCVs are in reduced NCVs in the range of 5–20 m/s. Involve-
the range from 25 to 35 m/s (Dubourg et al., 2006). ment of cranial nerves has been reported.
● SH3TC2 (SH3 domain and tetratricopeptide repeat ● Mutations in PRX (periaxin is a protein mainly
domain 2 is a protein that interacts in assembling expressed during development that is important in
protein complexes) is probably the most frequently Schwann elongation) typically result in a CMT with
mutated gene in early-onset AR CMT patients (per- severely reduced motor NCVs, as low as < 5 m/s.
sonal data, unpublished). Onset can be in the first In some instances myelin outfoldings can be seen
year of life but progression is often slow. Motor on neuropathology.
NCVs are clearly slowed, but the range is broad.
A particular finding in several patients is a severe
AXONAL FORMS (CMT2)
and early scoliosis, sometimes even preceding man-
ifest distal weakness (Dubourg et al., 2006). AD CMT2. Dominantly inherited axonal CMT (CMT2) is
● Among MTMRs (myotubularin-related proteins are estimated to represent about one third of all CMT cases.
tyrosine phosphatases that are believed to be regula- From a purely clinical point of view no distinction can be
tors of membrane dynamics), MTMR2 and made from CMT1. Electrophysiological evaluation,
MTMR13 or SBF2 (SET binding factor 2) cause however, allows separation, as motor NCVs are normal
AR CMT variants. Onset is in childhood but usually or only mildly slowed (>38 m/s). As for demyelinating
after normal motor milestones have been reached. forms, CMT2 is genetically heterogeneous (Zuchner
There is often a rapid progression resulting in the and Vance, 2006). Most forms have their onset in adult-
loss of ambulation and also in facial, bulbar, and dia- hood with some notable exceptions.
phragmatic weakness in some patients. On neuropa- Mutations in MFN2 (mitofusin 2 regulates both the
thology, irregular foldings and redundant myelin fusion of the mitochondria and the respiratory chain)
loops (or myelin outfoldings) can be seen are found in up to 20% of CMT2 patients and give rise
(Fig. 146.2). This finding is not exclusive but is to various axonal phenotypes (CMT2A). Severity of
1422 P. LANDRIEU ET AL.
CMT2A is more pronounced than for other CMT2 large family from Costa Rica affected with axonal CMT
forms. Age at onset varies widely but can be as early (Leal et al., 2009).
as 5 years (Verhoeven et al., 2006). MFN2 mutations
can also cause an axonal sensorimotor neuropathy with AD INTERMEDIATE FORMS
pyramidal tract signs, usually with an adult onset (also
called HMSN V). Sometimes patients carrying a As mentioned before, “intermediate CMT” refers to
MFN2 mutation develop an acute drop in visual acuity families and individual cases in which NCVs show large
due to an optic neuritis resulting in optic atrophy. This variations and overlap between CMT1 and CMT2. Some
syndrome, also known as HMSN VI, can have an onset genetic defects are especially candidates for these
in the first decade of life (Z€uchner et al., 2006). phenotypes.
A variant SEOAN syndrome (severe early onset axonal Mutations in DNM2 (dynamin 2 is a fission protein
neuropathy) has been described in young children. It is in that participates in endocytic vesicle formation and in
fact a biallelic gene disorder caused by the co-occurrence the dynamics of microtubules) usually give rise to an
of two MFN2 mutations (Nicholson et al., 2008). adult-onset phenotype with intermediate electrophysiol-
KIF1Bbeta is an isoform of KIF1B, a member of the ogy. As for all CMT variants, there is considerable var-
kinesin superfamily involved in mitochondrion trans- iability even within families and onset in early childhood
port. A KIF1B mutation has been described in a single has been described (Claeys et al., 2009). The phenotype is
small AD CMT2 family (Zhao et al., 2001), but its path- a classical CMT but in some families concomitant neu-
ogenicity is still equivocal. tropenia and early onset cataract have been reported.
GARS is a glycyl-tRNA synthetase expressed in Mutations in YARS (tyrosyl-tRNA synthetase is
sprouting neurites. GARS mutations have been shown involved in protein synthesis) result in AD intermediate
in several families with axonal CMT2D (Nangle et al., CMT. This form can start in childhood but so far
2007). RAB7 is a small GTP-ase late endosomal protein. only a few mutations have been reported (Jordanova
Mutations of the gene have been shown to be responsible et al., 2006).
for CMT2B in a few familial cases in which foot ulcers
were a prominent finding. HSPB1 (or HSP27), a 27 kDa X-LINKED CMT
small heat shock protein, and HSPB8 (HSP22), the Mutations in GJB1 (gap junction associated protein B1
22 kDa small heat shock protein, are responsible for a forms gap junctions between myelin layers) cause the
subset of purely motor neuropathies (HMN-II, see non-syndromic CMT1X which represents the second
below). The same genes are mutated in rare CMT2F most common variant of CMT, accounting for around
and CMT2L families presenting with an axonal motor 10% of all patients (Kleopa and Scherer, 2006). CMT1X,
and sensory neuropathy. especially in males, can begin in early childhood. Males
AR CMT2. Mutations in GDAP1, as mentioned are more severely affected but females can develop a
before, can cause a wide variety of phenotypes among clear phenotype as well, presumably by skewed
which the AR axonal phenotype is probably the most X-inactivation. Onset in females is usually later and
severe. In terms of mutation frequencies, GDAP1 is symptoms are milder; however, a broad spectrum has
likely to be an important player in ARCMT. The pheno- been described ranging from true asymptomatic carriers
type is characterized by early onset of weakness with to phenotypes as severe as in male patients (Kleopa and
rapid progression. Scoliosis and breathing difficulties Scherer, 2006). Early involvement of small hand muscles
due to diaphragmatic paralysis and vocal cord paralysis may be a sign pointing to CMT1X. NCVs can still be nor-
are features that orient the genetic screening toward mal in young female mutation carriers, while in young
GDAP1 (Bernard et al., 2006). male children they can fall within the range of CMT2
One single recessive founder mutation in LMNA or “intermediate CMT”. Given the high prevalence of
found in Moroccan and Algerian families gives rise to CMT1X, testing should be considered in any family that
a severe axonal CMT phenotype with an onset between does not reveal a clear father–son transmission (Szigeti
6 and 27 years. Lamins A/C, belonging to type V interme- et al., 2006).
diate filaments superfamily, are major structural con-
stituents of the nuclear lamina that regulate the
IIb Hereditary motor neuropathies
chromatin structure as well as various proteins. This neu-
ropathy forms one of the diverse disorders also called Distal hereditary motor neuropathies (HMN) are esti-
laminopathies (Bernard et al., 2006). mated to represent around 10% of all inherited neurop-
MED25 is a subunit of the activator-recruited cofac- athies (Irobi et al., 2006). Epidemiological data more
tor (ARC), a family of large transcriptional coactivator specific for pediatric populations are not available.
complexes. Homozygous mutation has been found in a HMN cover a spectrum of clinically and genetically
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1423
heterogeneous diseases characterized by the selective (<10 years), distal but also proximal weakness, and rapid
involvement of motor neurons in the peripheral nervous evolution toward tetraplegia and respiratory insuffi-
system (Dierick et al., 2008). In contrast to proximal spi- ciency. Nothing is known about the mutation frequency
nal muscular atrophies (SMA) (see Ch. 145), distal HMN of this gene and the variability of the associated phenotype.
initially and predominantly affects the distal limb mus- HMN V/Silver syndrome (BSCL2, 11q12-q14). Muta-
cles, suggesting a length-dependent mechanism affect- tions in BSCL2 (Bernardinelli-Seip congenital lipody-
ing the longest motor axons first. The disease usually strophy 2 is an endoplasmic reticulum membrane
begins in childhood or adolescence with weakness and protein of unknown function) cause a wide spectrum
wasting of distal muscles of the anterior tibial and pero- of AD disorders ranging from a pure distal motor neu-
neal compartments. Later, weakness and atrophy may ropathy without pyramidal tract signs to spastic paraple-
expand to the proximal muscles of the lower limbs gia (SPG17) (Windpassinger et al., 2004). Silver
and/or to the distal upper limbs. However, in some syndrome is one of the well-known phenotypic variants
patients and families the disease starts or predominates characterized by spasticity of the legs accompanied by
in the hands. Foot deformities (e.g., pes cavus) are some- amyotrophy of the hands and occasionally of the lower
times present. A classification has been proposed in limbs (Irobi et al., 2006).
seven subtypes based on age at onset, mode of inheri- Onset ages can vary widely between, and even within,
tance, distribution of muscle weakness, and additional families, but can be as early as 5 years (Dierick et al.,
features (Harding, 1993). Some features that were not 2008). Only two mutations in a mutational hotspot of
included in the original classification have resulted in BSCL2 have been described so far. In a recent study,
further diversification of the clinical spectrum, includ- mutation yield was 7% in a large HMN cohort, rising
ing pyramidal signs, congenital onset and X-linked to 30% in selected patients with proven AD inheritance
inheritance (Irobi et al., 2006). and clear pyramidal tract signs (Dierick et al., 2008),
So far, eight genes have been identified for distal making the two hotspot mutations good candidates to
HMN, six of which are associated with autosomal dom- screen in such patients.
inant (AD) forms. The combined mutation frequencies HMN VI (IGHMBP2, 11q-q14). The phenotype asso-
for all the known dominant genes in a larger cohort ciated with mutations in IGHMBP2 (immunoglobulin
are relatively low (15%), making the yield of diagnosis m-binding protein 2a) is known as SMARD1 or (distal)
screening poor (Dierick et al., 2008). Only the forms spinal muscular atrophy with respiratory distress
characterized at the gene level are described here. (Grohmann et al., 2001). Age of onset is very early after
HMN II is related to mutations in HSPB8 (12q24.3) or birth but usually after a symptom-free interval of several
HSPB1 (7q11-q21). Heat shock proteins block signals weeks. Despite the misleading abbreviation SMA, the
inducing apoptosis and are involved in stabilization of weakness has a distal distribution. An important addi-
newly produced proteins and repair of damaged pro- tional feature is bilateral diaphragmatic paralysis rapidly
teins, especially from the cytoskeleton (Evgrafov leading to respiratory insufficiency, with a poor long-
et al., 2004; Irobi et al., 2004). The HMN II phenotype term prognosis in the majority of patients (Bertini
is characterized by a pure motor neuropathy with distal et al., 1989).
weakness and muscle atrophy without pyramidal tract ALS4 (SETX, 9q34). Dominant mutations in SETX
signs and typically with the age of onset in adolescence. (senataxin is a DNA/RNA helicase) cause a phenotype
In a recent report, however, the Pro182Leu mutation in with the somewhat misleading name amyotrophic lateral
HSPB1 was found in association with an early onset sclerosis 4 (ALS4) or juvenile ALS (Chen et al., 2004).
age (5 years) in two children also displaying mild pyrami- Age at onset varies widely but usually falls within the
dal tract signs (Dierick et al., 2008). first decade (Irobi et al., 2006). The phenotype is charac-
Overall the combined mutation frequency for HSPB8 terized by progressive distal weakness and atrophy with-
and HSPB1 in a large cohort of HMN patients is only 6% out sensory disturbances and with variable pyramidal
(Dierick et al., 2008). Interestingly, mutations in these tract signs. There is no involvement of the cranial nerves
genes can also result in AD axonal sensorimotor neurop- or respiratory muscles and survival is normal. Mutation
athy (CMT2F and CMT2L, respectively). frequencies for SETX, however, account for less than
HMN IV (PLEKHG5, 1p36). HMN IV is a recessive 2% in a large HMN cohort (Dierick et al., 2008).
disorder caused by mutations in PLEKHG5 (pleckstrin
homology domain-containing, family G member 5 gene;
IIc Hereditary sensory and autonomic
pleckstrin domain is present in a wide range of proteins
neuropathies
involved in intracellular signaling or as constituents of
the cytoskeleton) (Maystadt et al., 2007). A mutation Hereditary sensory and autonomic neuropathies
was identified in one African family with childhood onset (HSAN) probably form the rarest subgroup within the
1424 P. LANDRIEU ET AL.
hereditary neuropathies and are clinically and genetically in the absence of manifest dysautonomic features
heterogenous. A selective degeneration of the sensory (Auer-Grumbach et al., 2006; Coen et al., 2006). The
neurons in the peripheral nervous system gives rise to mutation yield of 3% in a larger cohort of HSAN is
prominent distal sensory loss which can lead to chronic too low to warrant routine diagnostics.
ulceration of the feet and hands, osteomyelitis, and HSAN III (IKBKAP, 9q31). Riley–Day syndrome, or
amputations of toes and fingers. Due to the concomitant familial dysautonomia, is an AR disorder caused by
degeneration of autonomic nerve fibers in some forms, mutations in IKBKAP (inhibitor of k-light polypeptide
features such as anhidrosis, fever, blood pressure fluc- gene enhancer in B cells, kinase complex associated pro-
tuations, and gastrointestinal disturbances can be pre- tein, a protein that plays a role in transcriptional elonga-
sent in some patients (Dyck, 1993). The designation tion and in neuronal migration) (Slaugenhaupt et al.,
HSAN suggests an exclusive sensory and autonomic 2001). This disorder has a high prevalence in individuals
involvement, but in some forms clear weakness may of Ashkenazi or Eastern European Jewish origin. HSAN
be present. III presents as a predominantly autonomic disorder with
A classification into HSAN types I–V was made congenital onset. Symptoms include disturbances of gas-
based on age at onset, inheritance pattern, and additional trointestinal motility resulting in feeding difficulties and
features. HSAN can be transmitted as an AD or AR trait. pneumonia due to aspiration. In addition, patients may
Isolated patients are also known (Dyck, 1993; Auer- display pronounced blood pressure fluctuations. Signs
Grumbach et al., 2006). The AD forms (HSAN1) usually of sensory loss are present but mild compared to the
present in the second or third decade of life with marked autonomic features (Auer-Grumbach et al., 2006;
sensory, minimal autonomic and variable motor involve- Axelrod and Gold-von Simson, 2007).
ment. In contrast, the AR forms usually have an early or HSAN III seems to be genetically homogenous, war-
congenital onset and present with striking sensory and ranting screening in selected patients. Over 99% of
autonomic abnormalities or as almost pure autonomic patients are shown to carry the same mutation (Auer-
disorders (Auer-Grumbach et al., 2006). Additional fea- Grumbach et al., 2006).
tures such as mental retardation can be hallmark fea- HSAN IV (NTRK1, 1q21-22). Congenital insensitivity
tures of specific subtypes. to pain with anhidrosis (CIPA) or HSAN IV is an AR dis-
Overall the contribution of the known genes to the order caused by mutations in NTRK1 (neurotrophic tyro-
genetic spectrum of HSAN is not higher than 20%. sine kinase, receptor type) (Indo et al., 1996). The CIPA
Due to this low screening yield, molecular diagnosis is phenotype has characteristic features: recurrent episodic
not forthcoming. However, several phenotypic charac- fevers due to anhidrosis, absence of reaction to painful
teristics result in strict genotype-phenotype correlations stimuli, self-mutilating behavior and mental retardation
that can help in orienting the molecular screening (Axelrod and Gold-von Simson, 2007). Nerve conduction
(Rotthier et al., 2009). values are usually within the normal range (Shatzky
HSAN I (SPTLC1, 9q22.2). This dominantly inherited et al., 2000; Auer-Grumbach et al., 2003; Axelrod and
phenotype is caused by mutations in SPTLC1 (serine pal- Gold-von Simson, 2007). Nerve biopsy findings are dom-
mitoyltransferase long chain subunit 1 is a key enzyme in inated by almost complete disappearance of unmyelin-
sphingolipid synthesis) (Dawkins et al., 2001). It is a sen- ated fibers.
sory neuropathy without marked autonomic features Evidence so far suggests that CIPA is a genetically
and variable motor involvement in some patients. homogeneous disorder making diagnostic screening
It was only described in adults until recently, when a de worthwhile in selected patients. Mutations in NTRK1
novo mutation was described in a child with a congenital are found in up to 7% of cases in large HSAN cohorts
disease onset characterized by hypotonia, microcephaly, (Indo, 2001; Rotthier et al., 2009).
vocal cord paralysis, and breathing problems (Rotthier HSAN V (NGFB, 1p13.1). The phenotype caused by
et al., 2009). In the same study, mutation frequency in this mutations in NGFB (nerve growth factor b) is closely
gene was reported to be very low (2% of HSAN). related to CIPA but patients do not display prominent
HSAN II (WNK1/HSN2, 12p13.3). Mutations in mental retardation and the disease onset is in childhood.
WNK1/HSN2 (protein kinase with-no-lysine(K)-1/hered- The only mutation described so far may be a private
itary sensory neuropathy type 2 is a signaling molecule mutation since broader screenings have remained nega-
involved in regulation of neurite extension) cause tive (Einarsdottir et al., 2004; Rotthier et al., 2009). In
autosomal recessive HSAN type II, an early onset ulcer- rare instances, NTRK1 mutations can also give rise to
omutilating sensory neuropathy (Lafreniere et al., 2004). a HSAN V phenotype (Houlden et al., 2001).
Onset can be congenital or later in childhood and HSN and RAB7 (small GPTase late endosomal pro-
the phenotype is relatively uniform, with pronounced tein). RAB7 mutations are responsible for CMT2B,
distal sensory loss resulting in severe acromutilations which is a motor and sensory neuropathy with AD
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1425
transmission (see above). However, the profound sen- form, linked to the Spastin gene (SPG4). A sensory
sory loss and ulceromutilations found in these patients motor axonopathy is frequently encountered in others,
led to considering this phenotype as a part of the HSAN such as SPG 10 (KIF5 mutations; Goizet et al., 2009),
spectrum (Rotthier et al., 2009). Age of onset is usually SPG 11 (KIAA1840) and in a form linked to a 12q23-24
in adulthood but in some patients first symptoms can be locus (Sch€ule et al., 2009). In other SPG, the peripheral
noted in their teens (Kwon et al., 1995). neuropathy mainly affects the motor neurons, some-
times with a striking predominance in the hands. Some
III SYNDROMIC NEUROPATHIES are linked to the two known mutations in the Seipin/
BSCL2 gene (SPG 17 or Silver syndrome, see above)
IIIa Neurological syndromes with (Dierick et al., 2008).
postnatal revelation Progressive hearing loss must be checked in any
Two situations, though clinically different, lead to the patient with a peripheral neuropathy. This rare associa-
same diagnostic approach. In the first situation, fre- tion is mainly suggestive of a mitochondrial cytopathy,
quently included among CMT subcategories (see above), such as MIDD (maternally inherited diabetes and
the peripheral neuropathy appears as the initial, prevail- deafness), a syndrome usually encountered beyond ado-
ing feature, but becomes secondarily enriched by other lescence and related to the A3243G mutation of mtDNA.
neurological features. The second situation covers a Hearing loss is mainly encountered in mutations of the
number of multisystemic neurological disorders in connexin gene family. In a large family with a dominant
which the peripheral neuropathy appears as a secondary mutation in the connexin 31 gene (GJB3), some patients
feature. Most of the latter syndromes have been also exhibited a severe axonal neuropathy with a sensory
described in adults and are denominated by reference predominance, resulting in distal ulcerative complica-
to the symptom clinically prevalent, such as spinocere- tions (López-Bigas et al., 2001). In the more frequent
bellar atrophy þ, spastic paraplegia þ, ophthalmoplegia neuropathy due to connexin 32 (GJB1) mutations, hear-
þ, etc. Other ways of promoting a syndrome which ing loss is found in only a minority of cases (Stojkovic
avoid having to select a prevalent symptom have been et al., 1999). It is a variable feature found in a few
eponyms, such as Friedreich disease, or acronyms, such CMT1A patients but also in CMT subtypes caused by
as SANDO (see above). To a large extent, the vagaries of mutations in PMP22, MPZ, and NDRG1 (Pareyson
history explain why the same clinical picture can be et al., 2006).
attributed to different syndromes, in function of the Optic atrophy is associated with a peripheral axonal
chosen denomination, or why the same gene can be neuropathy in a few disorders with AD or mitochondrial
responsible for apparently different syndromes. transmission, including cases with a pediatric onset:
The mode of transmission is a major parameter which LHON þ syndromes (Leber hereditary optic neuropathy
allows separation into two groups: related to mtDNA mutations), OPA1 þ as well as
MFN2-related CMT2.
IIIA1 CLASSICAL FAMILIAL, AUTOSOMAL DOMINANT
SYNDROMES WITH OCCASIONAL ONSET IN CHILDHOOD
IIIA2 SYNDROMES WITH AUTOSOMAL (OR X-LINKED)
In most syndromes, the neuropathy is a distal sensory-
RECESSIVE TRANSMISSION, WITH FREQUENT OR USUAL
motor axonopathy with normal NCVs. The real occur-
ONSET IN CHILDHOOD
rence of the neuropathy is difficult to assess, as it largely
depends on the electrophysiological criteria used and the Friedreich disease (see Ch. 192) is the most frequent dis-
stage of the disease. order of this category (estimated prevalence: 1/50 000
In a large group of AD spinocerebellar atrophy, a people, carrier prevalence 1/110). In addition to the cere-
peripheral neuropathy, when systematically investi- bellar ataxia, other somatic features are cardiomyopathy,
gated, can be elicited in up to 70% of adult cases, espe- retinopathy, and pancreatic B cell dysfunction. The
cially in the forms related to expansions of nucleotide largely prevailing mutation is a biallelic amplification
triplets (SCA1, 2, 3, 7) (van de Warrenburg et al., of the triplet GAA in the first intron of the FXN gene,
2004). The age at clinical onset appears as a function which codes for frataxin, an iron-binding protein acting
of various factors, such as the nature of the protein, as a chaperone for various mitochondrial iron-sulfur pro-
the length of the polyglutamine expansion, and unknown teins. During the juvenile period, the presenting feature is
familial factors. usually ataxia, mostly due to the degeneration of spino-
In a group of AD spastic paraplegia (>16 entities in cerebellar tracts as well as sensory nerves. During the
2009; see Ch. 195), the presence of a neuropathy is highly course of the disease, sensory nerve action potentials
variable. It is usually not found in the most frequent and NCVs become inelicitable. Biopsy of sensory nerves
1426 P. LANDRIEU ET AL.
shows a marked axonal degeneration predominating on neurological features reveal the disease, whereas the
large myelinated fibers. other somatic features will appear later on in childhood.
Other AR spinocerebellar ataxias are frequently mul- Thus, the frontier between purely neurological syn-
tisystemic disorders that include a peripheral nerve com- dromes and multiorgan syndromes is neither absolute
ponent. Many of the recently identified forms have been nor permanent. This holds true for all disorders in which
related to genes involved in repair of single-strand the involved biological function affects any tissue in a
breaks, the most common DNA damage occurring in poorly predictable way, like mitochondrial OXPHOS,
cells. Their clinical denomination depends of the preva- DNA repair, N-glycosylation of proteins, centrosome-
lent clinical feature. Ataxia and oculomotor apraxia are mediated translocation of various substrates, and more
prominent findings in AOA1 (aprataxin or APTX gene) generally, any disorder affecting a protein with ubiqui-
and AOA2 (senataxin or SETX gene) (Koenig, 2003). tous functions. Moreover, rare cases will present with
The neuropathy is highly variable in precocity and sever- a fetal hypotrophy or macrosomia, or even with a true
ity. Its physiopathology is multiple, with a predominance congenital malformation such as polydactyly, blurring
of sensory-motor axonopathy (Ochsner et al., 2005). the frontier with the category of congenital syndromes
A form in which the neuropathy appears a major feature with a regular perinatal expression. Table 146.4 shows
has been denominated spinocerebellar ataxia with neu- a nonexhaustive list of such syndromes.
ropathy 1 and related to the gene TDP1. Giant axonal neuropathy, related to GAN (gigaxonin)
AR spastic paraplegias are frequently complicated mutations, is one of the most recognizable. In addition to
forms, in which the peripheral neuropathy is the most fre- neuropathy, the phenotype includes variable central ner-
quent complication (see Ch. 195). It is usually a sensory- vous system findings such as mental retardation and
motor axonopathy, like in SPG 11 (gene KIAA1840) and ataxia, as well as bone and hair abnormalities. On neuro-
SPG 15 (gene ZFYVE26 or spastizin). pathological examination, the characteristic giant axons
Hereditary sensory neuropathy with spastic paraplegia are due to accumulation of neurofilaments. EMG exam-
is caused by mutations in CCT5 (cytosolic chaperonin- ination can reveal an unusual pattern that mixes features
containing t-complex peptide-1; Bouhouche et al., 2006). of axonal degeneration, marked variation of NCVs from
Mutations seem to be confined to patients displaying the one nerve trunk to another, and large dispersion in motor
typical phenotype of an early childhood onset spastic and sensory potentials, probably reflecting the presence
paraplegia with pronounced distal sensory loss and acro- of irregular demyelination along the dilated axonal seg-
mutilations (Rotthier et al., 2009). ments (Fig. 146.3). The phenotype, however, is variable
Infantile neuroaxonal dystrophy (INAD, Seitelberger (Tazir et al., 2009).
disease) is a progressive encephalopathy with cerebellar Muscular dystrophy and peripheral neuropathy.
atrophy and accumulation of iron in globus pallidus as Since EMG anomalies are more systematically investi-
the main RMI features. Peripheral sensory-motor neu- gated in all muscular disorders resulting from proteins
ropathy can be an early and major feature. Nerve biopsy expressed both in muscle and in nervous system, the
shows striking axonal dilatations with accumulation of association of the two disorders is a growing chapter.
mitochondria, vesiculotubular structures and neurofila- Merosinopathies and disorders of related sarcolem-
ments. Most cases are due to mutations in PLA2G6, mal complexes. One of the most frequent congenital
encoding a calcium-independent group VI phospholi- muscular dystrophies, type 1A, is due to mutations in
pase A2 (Morgan et al., 2006) (see Ch. 194). the gene encoding merosin (laminin 2), a protein of
CMTX type 5 PRPS1 (phosphoribosyl pyrophosphate the extracellular matrix (see Ch. 143). A peripheral neu-
synthetase) is the mutated gene in this X-linked recessive ropathy is frequently observed, which results in a pro-
syndromic disorder characterized by early-onset neurop- gressive, moderate slowing of motor and sensory
athy (first decade), sensorineural deafness, and optic NCVs along the course of the disease. In a few cases
atrophy. Electrophysiology in these patients has both sural nerve biopsies have shown abnormal variation of
demyelinating and axonal features (Kim et al., 2007). myelin sheath and internodal length, without evidence
of segmental demyelination (Deodato et al., 2002; Di
Muzio et al., 2003).
IIIb Multiorgan syndromes with usually
Formation of myelin and internodal segments along
postnatal revelation
the nerve fibers is driven by complex cascades involving
In many syndromes with a protracted onset, the elements axonal proteins like periaxin, fences of actin and tubulin
that mostly contribute to the clinical diagnosis are the in Schwann cell as well as their connections to the
extraneurological features, whereas the peripheral neu- a-dystroglycan complex, to integrin a6b4 and to pro-
ropathy or other neurological complications will appear teins of the extracellular matrix. Mutations in proteins
as secondary events. In others, on the contrary, the of the a-dystroglycan complex and in enzymes of their
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1427
Table 146.4
Multiorgan genetic syndromes with usually postnatal revelation, including a peripheral neuropathy

Protein biological
Syndrome/disorder Neuropathy type Other organs Gene(s) function

Bardet–Biedl Sensory motor, Obesity, retinopathy, >11 BBS Centrosome-mediated


syndrome axonal MR, kidney, genes intracell transport
hypogenitalism
Ataxia telangiectasia Sensory motor, axonal Immune disorder, ATM DNA double strand
telangiectasia break repair
Xeroderma Axonal/demyelinating Premature aging, skin, >11 genes Nucleotide excision repair
pigmentosum/ Sensory motor eye, brain, liver
trichothiodystrophy/
Cockayne syndrome
Giant axonal Axonal/demyelinating Kinky hair GAN Cytoskeleton dynamics
neuropathy Intermediate filament Brain
accumulation
AAA syndrome Autonomic Achalasia, alacrimia, AAAS ALADIN:
neuropathy, distal adrenal gland nuclear pore complex
motor neuropathy

dominant and recessive transmission (Selcen et al.,


2004). A peripheral neuropathy is elicitable in about one
third of cases, with a mixture of axonal and Schwann cell
degeneration. The nerve biopsy occasionally shows an
aspect of giant axonal neuropathy (Sabatelli et al., 1992).
Laminopathies (see above) are responsible for vari-
ous phenotypes including Emery–Dreifuss muscular
dystrophy, lipodystrophy, restrictive dermopathy, pre-
mature aging syndromes and axonal neuropathy. Some
cases can exhibit both myopathic and neuropathic find-
ings (Benedetti et al., 2005).

IIIc Congenital neuropathies with


Fig. 146.3. Single fiber from giant axonal neuropathy, iso- presentation in the perinatal/early
lated by teasing technique. Some internodal segments keep a infancy period
relatively normal myelination (top), whereas others exhibit
more or less complete demyelination, especially in the most When the diagnosis of polyneuropathy is made in the
dilated axonal portions. (bottom, arrows) (bar ¼ 100 mm). neonatal period, or is suggested in late gestation by
abnormal ultrasound findings, it usually implicates spe-
O-glycosylation are responsible for a category of cial features, which are not exclusive: (1) the polyneuro-
syndromes that include muscular dystrophy, brain and pathy is severe, thus vital functions are frequently
ocular malformations, and CNS dysmyelination threatened in the first days of life; (2) the fetal onset
(Walker–Warburg syndrome, Fukuyama congenital of motor disturbances can result in joint and skeleton
muscular dystrophy, muscle-eye-brain syndrome; see deformations (arthrogryposis), that will raise serious
Ch. 143). Peripheral nerve dysmyelination, which has orthopedic difficulties; (3) syndromic forms are fre-
been shown in animal models of these a-dystroglycano- quent, thus malformative elements must be looked for
pathies, remains to investigate in the human syndromes. by multidirectional investigations; (4) given the particu-
Myofibrillar myopathies (MFMs) are related to disin- lar sociological status of the newborn, special ethical
tegration of Z-disks and then of myofibrils, followed by considerations are put forward in the most severe cases.
ectopic accumulation of multiple proteins. Mutations For the physician facing a baby with a diffuse motor
have been found in various genes (alphaB-crystallin, insufficiency together with other elements suggestive of
desmin, myotilin, Zasp, filamin-C, Bag3), both with a neuromuscular disease, the most relevant parameters are:
1428 P. LANDRIEU ET AL.
● autonomy of vital functions (ventilation, sucking/ mutations in the genes involved in other CMT1 forms,
swallowing) whereas homozygosity or compound heterozygosity
● evidence of an associated encephalopathy for usually dominant CMT1 mutations remain the excep-
● evidence of dysautonomic features tion (see supra).
● presence of orthopedic complications (arthrogrypo- Early-onset axonal neuropathy is a heterogeneous
sis, luxations, bone hypoplasia) phenotype, with clinical onset in early infancy for some
● evidence of associated malformations. cases (Ouvrier et al., 1981). Most forms are sporadic and
unresolved at the molecular level. A growing number of
Whether or not a specific clinical score will totalize the cases, however, have been related to homozygous or
grade of severity for each item, three schematic situa- compound heterozygous mutations in a gene involved
tions can be encountered: in a classical form of AD axonal neuropathy, such as
Grade 1: the baby has autonomy for all vital functions, MFN2 (see supra).
motor activity is not severely deficient, and no malfor- Disorders originally described as motor neuron dis-
mative context is evident. The first imperative is to rule ease are in fact sensory-motor neuropathies, in which
out a general disease, liable to secondary worsening if the sensory component is easily unrecognized behind
the opportunity for an appropriate treatment is missed. the severe motor presentation. One example is the neu-
Once this has been done using the appropriate clinical ropathy called SMARD1 or HMN type VI (see above)
and metabolic investigations, a diagnostic approach will (Fig. 146.4). In the same way, detailed pathological stud-
be embarked on taking all the time necessary, depending ies of the peripheral nerve remain to be performed in
on the parental choices. many other motor neuron disorders with a perinatal
Grade 2: the neonate presents with no sign of vital dis- expression. This is the case with lethal congenital con-
tress but with one or several preoccupying findings: tractures syndromes (LCCS 1, 2, 3), recently ascribed
severe motor deficiency, poor social contact, minor mal- to genes involved in mRNA processing and generation
formations, limited arthrogryposis, dysautonomic fea- of precursors of Schwann cells (Narkis et al., 2007;
tures. A complete neurological and genetic work-up is Nousiainen et al., 2008).
usually demanded and will be done in an ordered way.
Grade 3: the neonate presents with a severe malfor- EARLY SYNDROMES WITH A MARKED NEUROLOGICAL
mative context and/or with no autonomy of vital func- EXPRESSION
tions. Some forms are regularly lethal in the fetal or The possible association with an encephalopathy is a cen-
neonatal period, whereas others will survive beyond tral preoccupation in any neonate with a neuromuscular
the neonatal period with major handicaps. Ethical discus- disorder. Several cases have been reported associating a
sions will be initiated within a reasonable period of time, congenital axonal neuropathy and an encephalopathy.
usually less than 2 weeks, that aim towards reaching as Nerve biopsy is usually poorly specific, showing only a
accurate a diagnosis as possible as soon as possible, if reduction of large diameter fibers. Central nervous
necessary by using invasive procedures (neuromuscular
biopsy). In case of death occurring without a clear diag-
nosis, an autopsy should be proposed, including brain,
spinal cord, peripheral nerve and muscle, as well as stor-
ing of DNA and fibroblast culture.

ISOLATED NEONATAL POLYNEUROPATHY


In an akinetic but alert newborn, once the early forms of
spinal muscular atrophies, congenital myopathies,
and myasthenic syndromes (see Chs 138, 150, 151) have
been ruled out, a polyneuropathic origin is rare but
heterogeneous.
Early forms of hypomyelinating neuropathies usually
show themselves with hypotonia in early infancy rather
than real difficulties in the neonatal period. Many will
Fig. 146.4. Severe axonal reduction in the superficial (sen-
exhibit a severe handicap, though a stable course and a sory) branch of the lateral popliteal nerve in a 4-month-old
favorable mental outcome are frequent (Levy et al., baby with a typical phenotype related to a SMARD1 mutation
1997; Phillips et al., 1999). The CMT1A duplication has (original magnification  200) Sensory NCVs were unobtain-
not been reported as the causal mutation in severe neo- able in the sural nerve, 30 m/s in the median nerve. (Unpub-
natal forms. Rare cases have been related to uniallelic lished case; courtesy of C. Lacroix.)
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1429
system manifestations and MRI study are also poorly neuromuscular degeneration. The neuropathy,
specific, including microcephaly, hypoplasia of corpus mainly a sensory-motor axonopathy (Zimmer
callosum, seizures, and developmental delay. In one et al., 1992), is clinically blurred by the presence of
case, absence or marked decrease of microtubule- a muscular dystrophy. The causative gene, SIL1, is
associated proteins was shown by western blot in cortex a nucleotide exchange factor for the Hsp70 chaper-
samples (Chau et al., 2008), a finding probably second- one BiP, a key regulator of the endoplasmic reticu-
ary to a disorder of the cytoskeleton’s organization. lum functions (Senderek et al., 2005).
Association with a Hirchsprung or pseudo- ● Congenital cataract-facial dysmorphism-neuropa-
Hirchsprung disease together with congenital deafness thy (CCFDN) is a rare AR syndrome, specific to
is very suggestive of mutations in SOX10, a transcription patients of Vlax Roma ethnicity, that resembles
factor including a homeobox domain. In the example the Marinesco–Sj€ogren syndrome. Peripheral neu-
shown Figure 146.1, NCVs were clearly in the range of ropathy is dominated by hypomyelination of nerve
a dysmyelinating neuropathy, whereas the pathological fibers. A myopathic component is responsible for
findings included both developmental anomalies (micro- a risk of postinfectious rhabdomyolysis. A unique
fasciculation of the nerve trunks) and severe hypomye- ancestral mutation is found in the CTDP1 gene,
lination of myelinated fibers (Pingault et al., 2000). encoding a phosphatase specific for the phosphory-
Association with a partial gonadal dysgenesis has lated serine residues of the carboxy-terminal domain
been described in a male patient carrying a homozygous of the largest subunit of RNA polymerase (Varon
missense in exon 1 of the desert hedgehog (DHH) gene. et al., 2003).
Sural nerve pathology revealed extensive minifascicular
Congenital deafness must be looked for in any neo-
organization of the endoneurium and a decreased den-
nate or infant exhibiting a peripheral neuropathy,
sity of myelinated fibers (Umehara et al., 2000).
whether or not other neurological or extraneurological
Association with a congenital cataract directs investi-
anomaly is present. Until now, this association has not
gations toward a few recognizable entities:
been reported in mutations of the connexin gene family,
● Deficiency of Hyccin, a membrane protein involved but principally in multisystemic metabolic diseases (per-
in myelination of both the central and the peripheral oxisome, mitochondrion disorders) and in SOX10
nervous system, is responsible for a picture of neona- mutants (see above).
tal hypotonia followed by the progressive appearance
of spasticity, cerebellar ataxia, and mental retarda-
CONGENITAL SYNDROMES WITH PERIPHERAL
tion. MRI shows a diffuse supratentorial hypomye-
NEUROPATHY AS A MARGINAL FEATURE
lination. The peripheral neuropathy is characterized
by slowed NCVs and, on sural nerve biopsy, by a The peripheral nerve has not been thoroughly studied in
reduction in myelinated fiber density and in thickness most congenital syndromes affecting many organs,
of myelin sheath (Biancheri et al., 2007). including the nervous system, in a severe way.
● Marinesco syndrome is an autosomal recessive Table 146.5 gives a (not exhaustive) list of examples.
disorder characterized by cerebellar atrophy, cata- The expression of the peripheral neuropathy is very var-
racts, developmental delay, dysmorphism, and iable, both in precocity and in severity.
Table 146.5
Multiorgan genetic syndromes with a peripheral nervous system PNS component and usual presentation in neonatal
period/early infancy period.

Syndrome Neuropathic findings Main organs involved Gene Protein function

ARC syndrome Arthrogryposis Skin, liver, kidney, VPS33B Vesicle fusion


brain
CEDNIK syndrome Poorly defined Brain, skin SNAP29 Vesicle fusion
Trichomegaly-chorioretinopathy Motor–sensory Retinae, hair, brain ?
(Oliver–McFarlane syndrome) axonopathy
St€
uve–Wiedemann Dysautonomia Skeleton, brain LIFR Embryonic
differentiation
MICRO syndrome Motor neuropathy Brain, eye, face RAB3GAP Exocytose
Noonan/ Axonal motor- sensory Skin, heart, face RAS/MAPK Cell cycle
Cardiofaciocutaneous neuropathy pathway
syndrome
1430 P. LANDRIEU ET AL.
REFERENCES Chen YZ, Bennett CL, Huynh HM et al. (2004). DNA/RNA
helicase gene mutations in a form of juvenile amyo-
Auer-Grumbach M (2004). Hereditary sensory neuropathies. trophic lateral sclerosis (ALS4). Am J Hum Genet 74:
Drugs Today (Barc) 40: 385–394. 1128–1135.
Auer-Grumbach M, De Jonghe P, Verhoeven K et al. (2003). Cil T, Altintas A, Tamam Y et al. (2009). Low dose vincristine-
Autosomal dominant inherited neuropathies with promi- induced severe polyneuropathy in a Hodgkin lymphoma
nent sensory loss and mutilations: a review. Arch Neurol patient: a case report (vincristine-induced severe poly-
60: 329–334. neuropathy). J Pediatr Hematol Oncol 31: 787–789.
Auer-Grumbach M, Mauko B, Auer-Grumbach P et al. (2006). Claeys KG, Zuchner S, Kennerson M et al. (2009). Phenotypic
Molecular genetics of hereditary sensory neuropathies. spectrum of dynamin 2 mutations in Charcot–Marie–Tooth
Neuromolecular Med 8: 147–158. neuropathy. Brain 132: 1741–1752.
Axelrod FB, Gold-von Simson G (2007). Hereditary sensory Coen K, Pareyson D, Auer-Grumbach M et al. (2006). Novel
and autonomic neuropathies: types II, III, and IV. mutations in the HSN2 gene causing hereditary sensory and
Orphanet J Rare Dis 2: 39. autonomic neuropathy type II. Neurology 66: 748–751.
Azzedine H, Bolino A, Taieb T et al. (2003). Mutations in Davis CJF, Bradley W, Madrid R (1978). The peroneal mus-
MTMR13, a new pseudophosphatase homologue of cular atrophy syndrome: clinical, genetic, electrophysio-
MTMR2 and Sbf1, in two families with an autosomal reces- logical and nerve biopsy studies. J Genet Hum 26: 311–349.
sive demyelinating form of Charcot–Marie–Tooth disease Dawkins JL, Hulme DJ, Brahmbhatt SB et al. (2001).
associated with early-onset glaucoma. Am J Hum Genet Mutations in SPTLC1, encoding serine palmitoyltransfer-
72: 1141–1153. ase, long chain base subunit-1, cause hereditary sensory
Baumgartner MR, Verhoeven NM, Jakobs C et al. (1998). neuropathy type I. Nat Genet 27: 309–312.
Defective peroxisome biogenesis with a neuromuscular De Jonghe P, Timmerman V, Ceuterick C et al. (1999). The
disorder resembling Werdnig–Hoffmann disease. Thr124Met mutation in the peripheral myelin protein zero
Neurology 51: 1427–1432. (MPZ) gene is associated with a clinically distinct Charcot–
Benedetti S, Bertini E, Iannaccone S et al. (2005). Dominant Marie–Tooth phenotype. Brain 122: 281–290.
LMNA mutations can cause combined muscular dystrophy Deodato F, Sabatelli M, Ricci E et al. (2002).
and peripheral neuropathy. J Neurol Neurosurg Psychiat Hypermyelinating neuropathy, mental retardation and epi-
25: 1019–1021. lepsy in a case of merosin deficiency. Neuromuscul Disord
Berciano J, Garcia A, Combarros O (2003). Initial semeiology 12: 392–398.
in children with Charcot–Marie–Tooth disease 1A duplica- Dierick I, Baets J, Irobi J et al. (2008). Relative contribution of
tion. Muscle Nerve 27: 34–39. mutations in genes for autosomal dominant distal heredi-
Bernard R, De Sandre-Giovannoli A, Delague et al. (2006). tary motor neuropathies: a genotype-phenotype correlation
Molecular genetics of autosomal-recessive axonal Charcot– study. Brain 131: 1217–1227.
Marie–Tooth neuropathies. Neuromolecular Med 8: 87–106. Di Muzio A, De Angelis MV, Di Fulvio P et al. (2003).
Bertini E, Gadisseux JL, Palmieri G et al. (1989). Distal infan- Dysmyelinating sensory-motor neuropathy in merosin-
tile spinal muscular atrophy associated with paralysis of the deficient congenital muscular dystrophy. Muscle Nerve
diaphragm: a variant of infantile spinal muscular atrophy. 27: 500–506.
Am J Med Genet 33: 328–335. Dubourg O, Azzedine H, Verny C et al. (2006). Autosomal-
Biancheri R, Zara F, Bruno C et al. (2007). Phenotypic char- recessive forms of demyelinating Charcot–Marie–Tooth
acterization of hypomyelination and congenital cataract. disease. Neuromolecular Med 8: 75–86.
Ann Neurol 62: 121–127. Dyck PJ (1984). Inherited neuronal degeneration and atrophy
Bird TD, Kraft GH, Lipe HP et al. (1997). Clinical and patholog- affecting peripheral motor, sensory and autonomic neu-
ical phenotype of the original family with Charcot–Marie– rons. In: PJ Dyck, PK Thomas, EH Lambert et al. (Eds.),
Tooth type 1B: a 20-year study. Ann Neurol 41: 463–469. Peripheral Neuropathy. WB Sanders, Philadelphia,
Bouhouche A, Benomar A, Bouslam N et al. (2006). Mutation pp. 1600–1642.
in the epsilon subunit of the cytosolic chaperonin- Dyck PJ (1993). Neuronal atrophy and degeneration predom-
containing t-complex peptide-1 (Cct5) gene causes autoso- inantly affecting peripheral sensory and autonomic neu-
mal recessive mutilating sensory neuropathy with spastic rons. In: PJ Dyck, PK Thomas, JW Griffin et al. (Eds.),
paraplegia. J Med Genet 43: 441–443. Peripheral Neuropathy. 3rd edn. WB Saunders,
Burns J, Ouvrier RA, Yiu EM et al. (2009). Ascorbic acid for Philadelphia, pp. 1065–1093.
Charcot–Marie–Tooth disease type 1A in children: a ran- Einarsdottir E, Carlsson A, Minde J et al. (2004). A mutation in
domised, double-blind, placebo-controlled, safety and effi- the nerve growth factor beta gene (NGFB) causes loss of
cacy trial. Lancet Neurol 8: 537–544. pain perception. Hum Mol Genet 13: 799–805.
Chance PF (2006). Inherited focal, episodic neuropathies: Evgrafov OV, Mersiyanova I, Irobi J et al. (2004). Mutant
hereditary neuropathy with liability to pressure palsies small heat-shock protein 27 causes axonal Charcot–
and hereditary neuralgic amyotrophy. Neuromolecular Marie–Tooth disease and distal hereditary motor neuropa-
Med 8: 159–174. thy. Nat Genet 36: 602–606.
Chau V, Clément JF, Robitaille Y et al. (2008). Congenital Fabrizi GM, Taioli F, Cavallaro T et al. (2009). Further evidence
axonal neuropathy and encephalopathy. Pediatr Neurol that mutations in FGD4/frabin cause Charcot–Marie–Tooth
38: 261–266. disease type 4H. Neurology 72: 1160–1164.
HEREDITARY MOTOR-SENSORY, MOTOR, AND SENSORY NEUROPATHIES IN CHILDHOOD 1431
Ferreiro-Barros CC, Tengan CH, Barros MH et al. (2008). Kleopa KA, Scherer SS (2006). Molecular genetics of
Neonatal mitochondrial encephaloneuromyopathy due to X-linked Charcot–Marie–Tooth disease. Neuromolecular
a defect of mitochondrial protein synthesis. J Neurol Sci Med 8: 107–122.
275: 128–132. Koenig M (2003). Rare forms of autosomal recessive neurode-
Fusco C, Frattini D, Pisani C et al. (2008). Isolated vitamin E generative ataxia. Review. Semin Pediatr Neurol 10:
deficiency mimicking distal hereditary motor neuropathy 183–192.
in a 13-year-old boy. J Child Neurol 23: 1328–1330. Kwon JM, Elliott JL, Yee WC et al. (1995). Assignment of a
Garcı́a A, Calleja J, Antolı́n FM et al. (2000). Peripheral motor second Charcot–Marie–Tooth type II locus to chromosome
and sensory nerve conduction studies in normal infants and 3q. Am J Hum Genet 57: 853–858.
children. Clin Neurophysiol 111: 513–520. Lafreniere RG, MacDonald ML, Dube MP et al. (2004).
Goizet C, Boukhris A, Mundwiller E et al. (2009). Complicated Identification of a novel gene (HSN2) causing hereditary
forms of autosomal dominant hereditary spastic paraplegia sensory and autonomic neuropathy type II through the
are frequent in SPG10. Hum Mutat 30: 376–385. study of Canadian genetic isolates. Am J Hum Genet 74:
Grohmann K, Schuelke M, Diers A et al. (2001). Mutations in 1064–1073.
the gene encoding immunoglobulin mu-binding protein 2 Leal A, Huehne K, Bauer F et al. (2009). Identification of the
cause spinal muscular atrophy with respiratory distress variant Ala335Val of MED25 as responsible for CMT2B2:
type 1. Nat Genet 29: 75–77. molecular data, functional studies of the SH3 recognition
Harding AE, Thomas PK (1980). The clinical features of motif and correlation between wild-type MED25 and
hereditary motor and sensory neuropathy types I and II. PMP22 RNA levels in CMT1A animal models.
Brain 103: 259–280. Neurogenetics 10: 275–287.
Harrower T, Stewart JD, Hudson G et al. (2008). POLG1 muta- Levy BK, Fenton GA, Loaiza S et al. (1997). Unexpected
tions manifesting as autosomal recessive axonal Charcot– recovery in a newborn with severe hypomyelinating neu-
Marie–Tooth disease. Arch Neurol 65: 133–136. ropathy. Pediatr Neurol 16: 245–248.
Harding AE (1993). Inherited neuronal atrophy and degener- López-Bigas N, Olivé M, Rabionet R et al. (2001). Connexin
ation predominantly of lower motor neurons. In: PJ 31 (GJB3) is expressed in the peripheral and auditory
Dyck, PK Thomas, JW Griffin et al. (Eds.), Peripheral nerves and causes neuropathy and hearing impairment.
Neuropathy. WB Saunders. p. 1051–1064. Hum Mol Genet 10: 947–952.
Houlden H, Reilly MM (2006). Molecular genetics of Marrosu MG, Vaccargiu S, Marrosu G et al. (1998). Charcot–
autosomal-dominant demyelinating Charcot–Marie– Marie–Tooth disease type 2 associated with mutation of the
Tooth disease. Neuromolecular Med 8: 43–62. myelin protein zero gene. Neurology 50: 1397–1401.
Houlden H, King RH, Hashemi-Nejad A et al. (2001). A novel Maystadt I, Rezs€ohazy R, Barkats M et al. (2007). The nuclear
TRK A (NTRK1) mutation associated with hereditary sensory factor kappaB-activator gene PLEKHG5 is mutated in a
and autonomic neuropathy type V. Ann Neurol 49: 521–525. form of autosomal recessive lower motor neuron disease
Indo Y (2001). Molecular basis of congenital insensitivity to with childhood onset. Am J Hum Genet 81: 67–76.
pain with anhidrosis (CIPA): mutations and polymorphisms Morgan NV, Westaway SK, Morton JE et al. (2006).
in TRKA (NTRK1) gene encoding the receptor tyrosine PLA2G6, encoding a phospholipase A2, is mutated in
kinase for nerve growth factor. Hum Mutat 18: 462–471. neurodegenerative disorders with high brain iron. Nat
Indo Y, Tsuruta M, Hayashida Y et al. (1996). Mutations in the Genet 38: 752–754.
TRKA/NGF receptor gene in patients with congenital Nangle LA, Zhang W, Xie W et al. (2007). Charcot–Marie–
insensitivity to pain with anhidrosis. Nat Genet 13: 485–488. Tooth disease-associated mutant tRNA synthetases linked
Irobi J, Van Impe K, Seeman P et al. (2004). Hot-spot residue to altered dimer interface and neurite distribution defect.
in small heat-shock protein 22 causes distal motor neurop- Proc Natl Acad Sci U S A 104: 11239–11244.
athy. Nat Genet 36: 597–601. Narkis G, Ofir R, Manor E et al. (2007). Lethal congenital con-
Irobi J, Dierick I, Jordanova A et al. (2006). Unraveling the tractural syndrome type 2 (LCCS2) is caused by a mutation
genetics of distal hereditary motor neuronopathies. in ERBB3 (Her3), a modulator of the phosphatidylinositol-
Neuromolecular Med 8: 131–146. 3-kinase/Akt pathway. Am J Hum Genet 8: 589–595.
Jordanova A, De Jonghe P, Boerkoel CF et al. (2003). Nicholson GA, Magdelaine C, Zhu D et al. (2008). Severe
Mutations in the neurofilament light chain gene (NEFL) early-onset axonal neuropathy with homozygous and com-
cause early onset severe Charcot–Marie–Tooth disease. pound heterozygous MFN2 mutations. Neurology 70:
Brain 126: 590–597. 1678–1681.
Jordanova A, Irobi J, Thomas FP et al. (2006). Disrupted func- Nikali K, Suomalainen A, Saharinen J et al. (2005). Infantile
tion and axonal distribution of mutant tyrosyl-tRNA syn- onset spinocerebellar ataxia is caused by recessive muta-
thetase in dominant intermediate Charcot–Marie–Tooth tions in mitochondrial proteins Twinkle and Twinky.
neuropathy. Nat Genet 38: 197–202. Hum Mol Genet 14: 2981–2990.
Kim HJ, Sohn KM, Shy ME et al. (2007). Mutations in Nordborg C, Hagberg B, Kristiansson B (1991). Sural nerve
PRPS1, which encodes the phosphoribosyl pyrophosphate pathology in the carbonhydrate-deficient glycoprotein syn-
synthetase enzyme critical for nucleotide biosynthesis, drome. Acta Paediatr Scand Suppl 375: 39–49.
cause hereditary peripheral neuropathy with hearing loss Nousiainen HO, Kestilä M, Pakkasjärvi N et al. (2008).
and optic neuropathy (cmtx5). Am J Hum Genet 81: Mutations in mRNA export mediator GLE1 result in a fetal
552–558. motoneuron disease. Nat Genet 40: 155–157.
1432 P. LANDRIEU ET AL.
Ochsner F, Le Ber I, Said G et al. (2005). Mutation of the apra- Smit LS, Roofthooft D, van Ruissen F et al. (2008). Congenital
taxin gene presenting with Charcot–Marie–Tooth-like neu- hypomyelinating neuropathy, a long term follow-up
ropathy and cerebellar ataxia. Rev Neurol (Paris) 61: 331–336. study in an affected family. Neuromuscul Disord 18:
Ouvrier RA, McLeod JG, Morgan G et al. (1981). Hereditary 59–62.
motor and sensory neuropathy of neuronal type with onset Stendel C, Roos A, Deconinck T et al. (2007). Peripheral nerve
in early childhood. Neurol Sci 51: 181–197. demyelination caused by a mutant Rho GTPase guanine
Parano E, Uncini A, De Vivo DC et al. (1993). Electrophysiologic nucleotide exchange factor, frabin/FGD4. Am J Hum
correlates of peripheral nervous system maturation in infancy Genet 81: 158–164.
and childhood. J Child Neurol 8: 336–338. Stojkovic T, Latour P, Vandenberghe A et al. (1999).
Passage E, Norreel JC, Noack-Fraissignes P et al. (2004). Sensorineural deafness in X-linked Charcot–Marie–
Ascorbic acid treatment corrects the phenotype of a mouse Tooth disease with connexin 32 mutation (R142Q).
model of Charcot–Marie–Tooth disease. Nat Med 10: Neurology 52: 1010–1014.
396–401. Szigeti K, Nelis E, Lupski JR (2006). Molecular diagnostics of
Pareyson D, Scaioli V, Laura M (2006). Clinical and electro- Charcot–Marie–Tooth disease and related peripheral neu-
physiological aspects of Charcot–Marie–Tooth disease. ropathies. Neuromolecular Med 8: 243–254.
Neuromolecular Medecine 8: 3–22. Tazir M, Nouioua S, Magy L et al. (2009). Phenotypic variabil-
Phillips JP, Warner LE, Lupski JR et al. (1999). Congenital ity in giant axonal neuropathy. Neuromuscul Disord 19:
hypomyelinating neuropathy: two patients with long-term 270–274.
follow-up. Pediatr Neurol 20: 226–232. Umehara F, Tate G, Itoh K et al. (2000). A novel mutation of
Pingault V, Guiochon-Mantel A, Bondurand N et al. (2000). desert hedgehog in a patient with 46, XY partial gonadal
Peripheral neuropathy with hypomyelination, chronic dysgenesis accompanied by minifascicular neuropathy.
intestinal pseudo-obstruction and deafness: a developmen- Am J Hum Genet 67: 1302–1305.
tal “neural crest syndrome” related to a SOX10 mutation. van de Warrenburg BP, Notermans NC, Schelhaas HJ et al.
Ann Neurol 48: 671–676. (2004). Peripheral nerve involvement in spinocerebellar
Rotthier A, Baets J, Vriendt ED et al. (2009). Genes for hered- ataxias. Arch Neurol 61: 257–261.
itary sensory and autonomic neuropathies: a genotype- Varho T, Jääskeläinen S, Tolonen U et al. (2000). Central and
phenotype correlation. Brain 132: 2699–2711. peripheral nervous system dysfunction in the clinical vari-
Sabatelli M, Bertini E, Ricci E et al. (1992). Peripheral neurop- ation of Salla disease. Neurology 55: 99–104.
athy with giant axons and cardiomyopathy associated with Varon R, Gooding R, Steglich C et al. (2003). Partial defi-
desmin type intermediate filaments in skeletal muscle. ciency of the C-terminal-domain phosphatase of RNA
J Neurol Sci 109: 1–10. polymerase II is associated with congenital cataracts facial
Santoro L, Manganelli F, Lanzillo R et al. (2006). A new dysmorphism neuropathy syndrome. Nat Genet 35:
POLG1 mutation with peo and severe axonal and demye- 185–189.
linating sensory-motor neuropathy. J Neurol 253: 869–874. Verhoeven K, Claeys KG, Z€ uchner S et al. (2006). MFN2
Schr€oder JM, Hackel V, Wanders RJ et al. (2004). Optico- mutation distribution and genotype/phenotype correlation
cochleo-dentate degeneration associated with severe periph- in Charcot-Marie-Tooth type 2. Brain. 129: 2093–2102.
eral neuropathy and caused by peroxisomal D-bifunctional Windpassinger C, Auer-Grumbach M, Irobi J et al. (2004).
protein deficiency. Acta Neuropathol 108: 154–167. Heterozygous missense mutations in BSCL2 are associated
Schr€oder JM (2006). Neuropathology of Charcot-Marie-Tooth with distal hereditary motor neuropathy and Silver syn-
and related disorders. Neuromolecular Med 8: 23–42. drome. Nat Genet 36: 271–276.
Sch€ule R, Bonin M, D€ urr A et al. (2009). Autosomal dominant Zhao C, Takita J, Tanaka Y et al. (2001). Charcot-Marie-Tooth
spastic paraplegia with peripheral neuropathy maps to disease type 2A caused by mutation in a microtubule motor
chr12q23-24. Neurology 72: 1893–1898. KIF1Bbeta. Cell 105: 587–597.
Selcen D, Ohno K, Engel AG (2004). Myofibrillar myopathy: Zimmer C, Gosztonyi G, Cervos-Navarro J et al. (1992).
clinical, morphological and genetic studies in 63 patients. Neuropathy with lysosomal changes in Marinesco–
Brain 127: 439–451. Sj€ogren syndrome: fine structural findings in skeletal mus-
Senderek J, Krieger M, Stendel C et al. (2005). Mutations in cle and conjunctiva. Neuropediatrics 23: 329–335.
SIL1 cause Marinesco–Sj€ogren syndrome, a cerebellar Zouari M, Feki M, Ben Hamida C et al. (1998).
ataxia with cataract and myopathy. Nat Genet 37: 1312–1314. Electrophysiology and nerve biopsy: comparative study
Shatzky S, Moses S, Levy J et al. (2000). Congenital insensi- in Friedreich’s ataxia and Friedreich’s ataxia phenotype
tivity to pain with anhidrosis (CIPA) in Israeli-Bedouins: with vitamin E deficiency. Neuromuscul Disord 8:
genetic heterogeneity, novel mutations in the TRKA/ 416–425.
NGF receptor gene, clinical findings, and results of nerve Zuchner S, Vance JM (2006). Molecular genetics of
conduction studies. Am J Med Genet 92: 353–360. autosomal-dominant axonal Charcot–Marie–Tooth dis-
Slaugenhaupt SA, Blumenfeld A, Gill SP et al. (2001). Tissue- ease. Neuromolecular Med 8: 63–74.
specific expression of a splicing mutation in the IKBKAP uchner S, De Jonghe P, Jordanova A et al. (2006). Axonal
Z€
gene causes familial dysautonomia. Am J Hum Genet 68: neuropathy with optic atrophy is caused by mutations in
598–605. mitofusin 2. Ann Neurol 59: 276–281.

View publication stats

You might also like