Sunteți pe pagina 1din 16

®

NEUROLOGY BOARD REVIEW MANUAL


STATEMENT OF
EDITORIAL PURPOSE Neurogenetics Review
The Hospital Physician Neurology Board Review
Manual is a study guide for residents and Editor and Contributor:
practicing physicians preparing for board
examinations in neurology. Each quarterly Catherine L. Gallagher, MD
manual reviews a topic essential to the cur-
rent practice of neurology.
Assistant Professor of Neurology
University of Wisconsin Movement Disorders Program
PUBLISHING STAFF Staff Physician
PRESIDENT, GROUP PUBLISHER Middleton VA Hospital
Bruce M. White
Madison, WI
EDITORIAL DIRECTOR
Debra Dreger
ASSISTANT EDITOR
Rita E. Gould
EXECUTIVE VICE PRESIDENT
Barbara T. White
EXECUTIVE DIRECTOR
OF OPERATIONS
Jean M. Gaul
PRODUCTION DIRECTOR
Suzanne S. Banish
PRODUCTION ASSISTANT
Kathryn K. Johnson
ADVERTISING/PROJECT MANAGER
Patricia Payne Castle
Table of Contents
SALES & MARKETING MANAGER
Deborah D. Chavis Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . 2

NOTE FROM THE PUBLISHER: Neurologic Disorders with Genetic


This publication has been developed without
involvement of or review by the American Associations . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
Board of Psychiatry and Neurology.
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Endorsed by the
Association for Hospital
Medical Education Cover Illustration by Kathryn K. Johnson
Copyright 2005, Turner White Communications, Inc., Strafford Avenue, Suite 220, Wayne, PA 19087-3391, www.turner-white.com. All rights reserved. No part of this
publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, mechanical, electronic, photocopying, recording, or oth-
erwise, without the prior written permission of Turner White Communications. The preparation and distribution of this publication are supported by sponsorship
subject to written agreements that stipulate and ensure the editorial independence of Turner White Communications. Turner White Communications retains full
control over the design and production of all published materials, including selection of appropriate topics and preparation of editorial content. The authors are
solely responsible for substantive content. Statements expressed reflect the views of the authors and not necessarily the opinions or policies of Turner White
Communications. Turner White Communications accepts no responsibility for statements made by authors and will not be liable for any errors of omission or inac-
curacies. Information contained within this publication should not be used as a substitute for clinical judgment.

www.turner - white.com Neurology Volume 9, Part 1 1


NEUROLOGY BOARD REVIEW MANUAL

Neurogenetics Review
Catherine L. Gallagher, MD

be identified with a genetic disorder. Founder effect is


INTRODUCTION when a particular (especially recessive) mutation is
overrepresented in a population due to a small genetic
In the past half century, an explosion of knowledge pool. An example would be the propagation of
in the field of molecular genetics has revolutionized our X-linked hemophilia in European royalty in the nine-
understanding of human diseases. One third of known teenth and twentieth centuries. A polymorphism is an
single gene defects cause diseases that affect the ner- allele (DNA sequence variation) that occurs in at least
vous system, so knowledge of the clinical approach to 1% of the normal population.
genetic disorders is essential for the practicing neurolo- Genomic imprinting is the preferential inactivation
gist. This manual provides a survey of single gene de- (by methylation) of some regions of a chromosome
fects that affect the nervous system, based on the most based on its parental origin. The most striking exam-
prominently affected neuroanatomic region. ples of imprinting are Prader-Willi syndrome and
Angelman’s syndrome, both caused by deletion of
BASIC CONCEPTS genes on the proximal long arm of chromosome 15.
Genetic Basis of Inheritance Prader-Willi syndrome (obesity, hypotonia, mild mental
Humans have 22 pairs of autosomes and 1 pair of sex retardation, small hands and feet) is caused by a defi-
chromosomes. Each chromosome is made up of 2 com- ciency of paternal gene expression within this region,
plementary strands of DNA consisting of a double helical whereas Angelman’s syndrome (severe mental retarda-
structure surrounding matched nucleotide base pairs tion, epilepsy, facial abnormalities, jerky movements,
(guanine with cytosine, adenine with thymine). Each set hypopigmentation, frequent laughter) is caused by a
of 3 DNA base pairs, or codon, codes for 1 amino acid. deficiency of maternal gene expression.1
DNA is transcribed into messenger RNA (mRNA) by RNA
polymerase and then translated into protein. Introns and Patterns of Inheritance
untranslated regions are portions of the DNA sequence Single gene disorders are traits produced by the
that are transcribed but not translated. effects of a single gene or gene pair. Such traits are
A gene is a portion of the DNA sequence that is the inherited in patterns originally described by Mendel as
basic unit of inheritance. Genotype is an individual’s either dominant (transmitted virtually unchanged by
genetic makeup; phenotype is an individual’s physical hybridization) or recessive (masked in the process). Four
traits or morphology. Expressivity of a gene is the degree inheritance patterns are seen in genetic disorders of the
to which a trait attributable to the gene is evident in the nervous system: autosomal dominant, autosomal recessive,
phenotype. Gene expression is more frequently used to X-linked, and mitochondrial. A patient presenting with a
describe the quantity and location of mRNA and pro- genetic disease without a family history is said to be a
tein transcribed and translated from the gene. Tran- sporadic or isolated case; explanations that should be con-
scription factors are proteins that bind to DNA to regulate sidered in such cases include new mutations (common
gene expression. in certain conditions), false paternity, variable pene-
Alleles are variations in the sequence of nucleotides trance, and recessive inheritance.
that make up a gene. An individual inherits 2 alleles of Autosomal dominant. A dominant trait will manifest
each gene, 1 from each parent. If the alleles are identi- in both the heterozygote and homozygote, meaning that
cal, the genotype is homozygous; if they are different, the a single copy of the mutant allele is sufficient to produce
genotype is heterozygous. The penetrance of an allele is the the trait. Characteristics of an autosomal dominant dis-
proportion of individuals that express its phenotypic order include: multiple successive generations are affect-
manifestations (ie, disease). Penetrance is frequently ed, males and females are affected in similar proportions,
variable, particularly in autosomal dominant disorders. both males and females transmit disease, and at least 1
A proband is the first person within a family or kinship to instance of male-to-male transmission is seen.

2 Hospital Physician Board Review Manual www.turner - white.com


Neurogenetics Review

Autosomal recessive. A recessive trait will manifest sense mutations (truncation of the protein product
only in the homozygote, meaning that both copies of due to introduction of a stop codon)
the allele must be mutant to produce the trait. Charac- • Frame shifts, which disrupt the reading frame of RNA
teristics of an autosomal recessive disorder include: both polymerase due to the excision or addition of base
males and females are affected, the disorder frequently pairs, resulting in the production of an incorrect
occurs in only 1 generation (usually among siblings), amino acid sequence
and the parents may be consanguineous. An individual • Dynamic mutations (repeat sequences that can change
with 1 recessive allele is referred to as a carrier. size on transmission [eg, trinucleotide repeat disor-
X-linked. Because men have only 1 X chromosome, ders])
loss of function mutations in genes carried on this chro- Although the consequences of mutations vary widely
mosome will result in disease. Characteristics of depending on how cellular proteins are altered, in gener-
X-linked recessive inheritance include: males are affect- al, mutations produce their effect through a loss or gain
ed almost exclusively, transmission occurs through un- of protein function. The majority of mutations are loss of
affected carrier females to their sons, daughters of function mutations. In most cases, protein synthesized
affected males become carriers, and male-to-male from the normal (wild-type) allele is sufficient to main-
transmission does not occur. Some X-linked disorders, tain cellular processes, so only individuals with 2 mutant
such as Duchenne muscular dystrophy, do produce a alleles will express disease (ie, recessive inheritance pat-
mild phenotype in carrier females. X-linked dominant tern). In some cases, a reduction in protein quantity or
disorders are rare and imply that females who harbor activity due to heterozygous mutation will result in less
the mutation are affected.2 severe disease than does the homozygous state; this con-
Mitochondrial. Mitochondria are cytoplasmic orga- dition is termed haploinsuffiency. One generalization is that
nelles that are a critical source of ATP for cell function. autosomal recessive disorders often have a more severely
Each mitochondrion contains 2 to 15 copies of mito- affected phenotype than do autosomal dominant disor-
chondrial DNA (mtDNA), which contains 37 genes that ders because recessive disorders involve a complete rather
code for some of the polypeptides involved in oxidative than partial alteration in protein function.
phosphorylation (OXPHOS), 22 mitochondrial trans- Gain of function mutations result in critical changes
fer RNAs (tRNAs), and 2 ribosomal RNAs. Aspects of in protein dosage; a simple example of this would be tri-
mitochondrial OXPHOS important for disease patho- somy 21 (Down syndrome). Gain of function mutations
genesis include energy production, generation of reac- usually produce dominant phenotypes.
tive oxygen species, and regulation of programmed cell
death (apoptosis).3 Because mitochondria are inherit- Molecular Genetics Testing
ed from the oocyte cytoplasm, diseases caused by de- Discoveries in molecular genetics are making possi-
fects in mtDNA follow a maternal inheritance pattern. ble the development of tools for precise DNA-based
Thus, only mothers can transmit disease to male or diagnosis, which have the potential to provide valuable
female offspring. Mutations in nuclear genes that code prognostic and genetic counseling information. Al-
for mitochondrial proteins may be inherited in an auto- though a discussion of genetic testing for heritable neu-
somal dominant, autosomal recessive, or X-linked fash- rologic disorders is beyond the scope of this review, a
ion. few essential points are noted.
Techniques. Beginning with a family in which some
Types of Mutations individuals are affected by a genetic trait or disease, mo-
The normal DNA sequence is referred to as wild- lecular biologic techniques can be used to find the
type. A mutation is an alteration in the DNA sequence chromosomal location of the gene that causes the trait.
that has the potential to cause disease. Mutations range Positional cloning uses linkage analysis (degree of associ-
from changes in a single base pair to large alterations in ation during meiotic segregation) between the disease
a part of or an entire chromosome (eg, trisomy 21). trait and other traits of known location with the
The main types of single gene mutations are: genome to determine the chromosomal location of the
trait of interest. During this process, restriction endonucle-
• Deletions (removal of DNA material) or expansions ases can be used to cut DNA where specific sequences
(insertion or duplication of DNA material) occur; gel electrophoresis is used to separate fragments of
• Single base pair substitutions (also called point muta- DNA, RNA, or protein by their size (molecular weight);
tions), which include missense mutations (alteration and polymerase chain reaction is invariably used to selec-
in an amino acid in the protein product) and non- tively increase the copy number of segments of DNA for

www.turner - white.com Neurology Volume 9, Part 1 3


Neurogenetics Review

Table 1. Examples of Mitochondrial Disorders aged unless treatment can be offered. Particularly in
Mitochondrial genomic mutations
asymptomatic individuals, genetic counseling is recom-
Kearns-Sayre syndrome/progressive external ophthalmoplegia
mended for the patient and family prior to DNA testing.
Leigh syndrome
Online resources. Extensive neurogenetic informa-
Mitochondrial myopathy
tion to support the clinical neurologist is available in pub-
Leber’s hereditary optic neuropathy
lic databases. One of the most useful online databases of
Myoclonic epilepsy with ragged red fibers
information for genetic diagnosis and testing is Online
Mitochondrial myopathy, encephalopathy, lactic acidosis, and
Mendelian Inheritance in Man (OMIM), which can be
stroke-like episodes searched by disease name, symptom/sign, gene symbol,
Neurogenic weakness, ataxia, and retinitis pigmentosa or protein name. OMIM searches can be entered using
Diabetes mellitus the Entrez system on PubMed (www.pubmed.gov).
Cardiomyopathy
Pearson syndrome
NEUROLOGIC DISORDERS WITH GENETIC
Nuclear DNA mutations ASSOCIATIONS
Multiple mitochondrial DNA deletions (Leigh phenotype)
Progressive external ophthalmoplegia
Mitochondrial neurogastrointestinal encephalopathy syndrome MITOCHONDRIAL, TRINUCLEOTIDE REPEAT, AND ION
X-linked hereditary spastic paraplegia (paraplegin deficiency) CHANNEL DISORDERS
Friedreich’s ataxia
Genetic disorders that are frequently discussed
together due to thematic similarities include the mito-
chondrial disorders, trinucleotide repeat disorders, and
further analysis. DNA sequencing is used to determine channelopathies.
the specific sequence of nucleotides that make up a seg-
ment of DNA. Recognition of elements with typical Mitochondrial Disorders
functions within a DNA sequence and comparison to Manifestations of mitochondrial disorders can vary
known genes in other organisms also helps determine significantly between individuals; different mtDNA muta-
which portion of the sequence constitutes the gene. tions can cause similar phenotypes, and different pheno-
Commercially available genetic tests may use various types can be seen in patients with identical mtDNA
techniques for mutation analysis (not always DNA defects. This variation has been explained partially by het-
sequencing). Therefore, these tests may not detect all eroplasmy (presence of wild-type and mutant mitochon-
possible mutations that give rise to a particular disease. drial genomes in the same cell), tissue mosaicism (dispro-
Ethical issues. Genetic tests are one of the few pre- portionate distribution of mutant mtDNA to progenitor
dictive tests available in medicine, and several ethical cells during cell division), and threshold effects (a certain
issues accompany this fact. The disease that the test pre- proportion of mtDNA must carry the mutation to pro-
dicts may be untreatable and a positive result, therefore, duce disease). Implications of mosaicism for the clinician
seen as a “death sentence.” Patients with a positive test are that it is best to select affected tissues for genetic test-
result may be at risk for suicide, whereas those with a ing because the mutation can sometimes be missed in
negative result may be at risk for “survivor guilt.” When serologic tests. Long-lived tissues with high metabolic
ordering a genetic test for a patient, the neurologist demand have a lower threshold for the expression of
must also consider implications of a positive result for mitochondrial disease. In particular, brain, heart, skeletal
family members who are at risk for the disease. For muscle, retina, renal tubules, and endocrine glands tend
example, a positive test for an autosomal dominant mu- to be affected.4 The classic manifestation in skeletal mus-
tation in the patient and the patient’s maternal grand- cle is the ragged red fiber, a contracted irregular muscle
mother may unmask the patient’s mother as an “obli- fiber with a rim that is red on Gomori trichrome stain
gate carrier” of the mutation. due to proliferation and accumulation of mitochondria
Patients at risk for genetic conditions may be sympto- inside the cell membrane.
matic or asymptomatic, adults or children, or cognitively General neurologists should be familiar with the clas-
competent or cognitively impaired. Special considera- sic mtDNA disorders that have primarily neurologic
tions must taken when testing vulnerable groups, par- manifestations (Table 1). Leigh syndrome (familial bilat-
ticularly children, for presymptomatic disease. Test of eral striatal necrosis) presents in infancy or early child-
presymptomatic children prior to age 18 years is discour- hood with encephalopathy, cranial nerve abnormalities,

4 Hospital Physician Board Review Manual www.turner - white.com


Neurogenetics Review

Table 2. Trinucleotide Repeat Disorders

Disorder Inheritance Gene Chromosome Repeat Type


Fragile X syndrome XLD FMR1 X CGG
Kennedy’s disease XLD Androgen receptor X CAG
Huntington’s disease AD Huntingtin 04 CAG
Myotonic dystrophy 1 AD DMPK 19 CTG
Myotonic dystrophy 2/PROMM AD ZNF9 03 CCTG
Friedreich’s ataxia AR Frataxin 09 GAA
DRPLA AD DRPLA/Atrophin 1 12 CAG
SCA1 AD Ataxin 1 06 CAG
SCA2 AD Ataxin 2 12 CAG
SCA3 (MJD) AD Ataxin 3 (josephin) 14 CAG
SCA6 AD CACNA1A 19 CAG
SCA7 AD Ataxin 7 03 CAG
SCA8 AD Unknown 13 CTG
SCA10 AD Ataxin 10 22 ATTCT
SCA12 AD PP2A 05 CAG
SCA17 AD TATA box-binding protein 06 CAG
OPMD AD PABP2 14 GCG

AD = autosomal dominant; AR = autosomal recessive; CACNA1A = voltage-dependent calcium channel alpha-1A subunit; DMPK = dystrophia
myotonica protein kinase; DRPLA = dentatorubropallidoluysian atrophy; FMR1 = fragile X mental retardation 1; MJD = Machado-Joseph disease;
OPMD = oculopharyngeal muscular dystrophy; PABP2 = polyadenylate-binding protein 2; PME = progressive myoclonic epilepsy; PP2A = protein
phosphatase 2A; PROMM = proximal myotonic myopathy; SCA = spinocerebellar ataxia; XLD = X-linked dominant; ZNF9 = zinc finger protein 9.

and ataxia associated with hyperintensity or necrosis in painless loss of central visual acuity in teenage or young
the basal nuclei, cerebellum, and brainstem on T2- adult life, with higher penetrance in men. Neuropathy,
weighted magnetic resonance imaging (MRI). Muta- ataxia, and retinitis pigmentosa (NARP) syndrome is
tions in both nuclear and mitochondrial genes, most also caused by a mutation in ATPase 6. Finally, mito-
notably nuclear-encoded OXPHOS subunits and the chondrial dysfunction is thought to play a role in sever-
gene encoding subunit 6 of mitochondrial ATPase al neurodegenerative syndromes, including Alzheimer’s
(ATPase 6), have been associated with this phenotype. disease and Parkinson’s disease.
Kearns-Sayre syndrome (KSS) and chronic progressive
external ophthalmoplegia (CPEO) are characterized by Trinucleotide Repeat Disorders
ptosis, ophthalmoplegia, and ragged red fiber myopathy Human genomic DNA contains numerous spans of
and are variably associated with other organ system in- repeated trinucleotide elements. When these sequences
volvement and ataxia. KSS is a more severe syndrome, expand beyond the normal length, they can alter gene
whereas CPEO is less severe, with potentially an adult or expression such that disease results (Table 2). In most
adolescent onset. Myoclonic epilepsy with ragged red cases, the disorders are autosomal dominant (toxic gain
fibers (MERRF) and mitochondrial myopathy, enceph- of function). CAG expansions code for polyglutamine
alopathy, and lactic acidosis with stroke-like episodes tracts that accumulate in neuronal nuclear inclusions
(MELAS) are caused by mutations in mitochondrial thought to be the result of altered protein compartmen-
tRNA genes; both conditions have a variable age of on- talization and degradation. In some conditions (eg, frag-
set. MERRF is characterized by myoclonic epilepsy, pro- ile X syndrome, myotonic dystrophy, Friedreich’s ataxia,
gressive ataxia, and mitochondrial myopathy; midline spinocerebellar ataxia type 8 [SCA8], SCA12), the trinu-
lipomas may be associated. MELAS is a cause of evanes- cleotide element involves untranslated regions of DNA.5
cent stroke-like events in children and young adults Generally, the phenotypes of trinucleotide repeat disor-
and is frequently associated with migraine, ataxia, and ders correlate with the repeat length; larger expansions
seizures. Leber’s hereditary optic neuropathy (LHON) cause more severe disease. The repeat length can change
is caused by point mutations in 1 of 7 subunits of com- (frequently expanding) during gametogenesis, leading
plex I (the first step in the electron transport chain) to earlier disease onset and more severe disease in suc-
encoded by mitochondrial genes. LHON presents as cessive generations (anticipation). In some cases, parents

www.turner - white.com Neurology Volume 9, Part 1 5


Neurogenetics Review

Table 3. Channelopathies with Neurologic Manifestations gene families that probably evolved from a common
Ligand-gated channels
ancestral gene by gene duplication and divergence.
Nicotinic acetylcholine receptors
Disorders caused by mutations in ion channel genes are
Myasthenia congenita
typically autosomal dominant (cystic fibrosis is an
Autosomal dominant nocturnal frontal lobe epilepsy
exception) and are characterized by episodic attacks
GABAA receptors
that can be precipitated by particular stressors such as
Generalized epilepsy with febrile seizures
exercise, dietary factors, or neuromuscular blocking
Glycine receptors
agents. Classic examples of channelopathies are long
Familial hyperplexia
QT syndrome, periodic paralyses, paramyotonia con-
genita, and malignant hyperthermia. Several disorders
Calcium channels are caused by allelic defects in the P/Q type voltage-
Sarcoplasmic reticulum calcium release channel (ryanodine receptor 1) dependent calcium channel, including SCA6, episodic
Central core disease ataxia type 2, and familial hemiplegic migraine.6 – 8
Malignant hyperthermia susceptibility type 1
Neuronal P/Q type voltage-dependent calcium channel CEREBRAL CORTICAL DISORDERS
Spinocerebellar ataxia type 6 Developmental Delay
Familial hemiplegic migraine Encephalopathy and developmental delay can be
Episodic ataxia type 2 associated with one of many inborn errors of metabo-
Skeletal muscle L-type voltage-dependent calcium channel lism. These disorders will not be discussed in detail.
Hypokalemic periodic paralysis Most single gene defects that affect development are
Sodium channels
associated with dysmorphic features. Two common dis-
Skeletal muscle
orders are trisomy 21 (Down syndrome) and fragile X
Hyperkalemic periodic paralysis
syndrome. About half of live births with trisomy 21 sur-
Paramyotonia congenita
vive until age 50 years. Some morphologic features of
Brain
trisomy 21 include a third fontanelle, large tongue, sin-
Generalized epilepsy with febrile seizures
gle palmar crease, clinodactyly, epicanthic folds, and
Brushfield’s spots of the iris. Obesity, short stature, and
Potassium channels congenital heart defects are common.
Episodic ataxia with myokymia Fragile X syndrome is caused by an expansion of
Benign familial neonatal convulsions more than 200 CGG repeats in an untranslated region
Chloride channels
of the FMR1 gene on the distal long arm of the X chro-
Myotonia congenita
mosome. The expansion leads to transcriptional silenc-
ing of the FMR1 gene due to abnormal methylation.
Gap junction channels The expansion is visualized as a constriction in the chro-
X-linked Charcot-Marie-Tooth neuropathy mosome in folate-deficient media. Boys with fragile X
GABA = γ-aminobutyric acid.
syndrome have hyperextensible joints, a high-arched
palate and high-pitched voice, pectus excavatum, mac-
roorchidism, developmental delay, and frequently autis-
of patients affected by these disorders carry a premutation, tic features. Men who carry a premutation of about 80
a trinucleotide repeat expansion of insufficient length to to 100 repeats may develop action tremor with parkin-
cause disease in the parent. In fragile X syndrome, pre- sonism in the sixth decade.9
mutations are more likely to expand with maternal trans-
mission. In Huntington’s disease, allele expansion is Neurocutaneous Syndromes
more likely to occur with paternal transmission. Congenital syndromes with skin and nervous system
manifestations are listed in Table 4; tuberous sclerosis
Channelopathies and neurofibromatosis are most common. Although
Mutations in genes that code for protein subunits of genetically unrelated, many neurocutaneous syndromes
ion channels and for the elements of ligand-gated are caused by defects in tumor suppressor genes.
channels (eg, the nicotinic acetylcholine receptor) have Tuberous sclerosis is caused by a mutation in the
been termed channelopathies (Table 3). Although their TSC1 gene on chromosome 9, which codes for
gating characteristics and selectivity vary, many ion hamartin, or a mutation in the TSC2 gene on chromo-
channels have a similar structure and are grouped into some 16, which codes for tuberin; half to three quarters

6 Hospital Physician Board Review Manual www.turner - white.com


Neurogenetics Review

Table 4. Neurocutaneous Disorders


Inheri-
Disorder tance Cutaneous Features Neurologic Features Gene Function
Ataxia-telangiectasia AR Conjunctival telangiectasia, skin Ataxia, intention tremor, ATM DNA repair
telangiectasis (late) decreased tendon reflex-
es, abnormal saccades
Fabry’s disease XLR Angiokeratomas Painful neuropathy, stroke GLA α-Galactosidase A
deficiency
Hypomelanosis of Ito AD Linear or swirling hypopigment- Mental retardation, epilepsy Mosaic of —
ed areas incontentia
pigmenti
Incontentia pigmenti XLD Neonatal: erythematous, macu- Mental retardation, epilepsy, X/autosome Transcription factor
(90% of lar, papular, vesicular lesions; microcephaly, spasticity translocation activator
females) later: pigmented macules,
whorls
Lesch-Nyhan XLR Self-mutilation, especially of Mental retardation, dystonia HPRT1 Defective purine
syndrome digits and lips metabolism
Linear sebaceous Sporadic Linear verrucous lesion near Epilepsy, mental retarda- Unknown Unknown
nevus syndrome midline of face or scalp tion, focal deficits
Menkes (kinky hair) XL Colorless, friable hair, pili torti Seizures, hypotonia, devel- MNK Gut copper-binding
disease (kinky hair), trichlorrhexis opmental delay and protein
nodosa (hair fractures) regression
Neurofibromatosis AD Café-au-lait spots, axillary Learning disability, cogni- NF1 Tumor suppressor
type 1 freckling, Lisch nodules of iris, tive impairment, neuro-
cutaneous neurofibromas, axis tumors
plexiform neurofibromas
Neurofibromatosis AD Skin tumors Neuro-axis tumors NF2 Tumor suppressor
type 2
Osler-Weber-Rendu AD Cutaneous and membrane Abscess, meningitis, embo- Endoglin TGF binding protein
disease telangiectasias lism, cerebral vascular (ENG)
abnormalities
Sturge-Weber Sporadic Facial port-wine stain, glaucoma Epilepsy, mental retarda- ?Somatic 4q Altered fibronectin
syndrome tion, focal deficits inversion expression
Tuberous sclerosis AD Hypomelanotic macules, ungual Epilepsy, mental retarda- TSC1, TSC2 Tumor suppressor
fibromas, shagreen patches, tion, autism, giant cell
facial angiofibromas astrocytoma
von Hippel-Lindau AD Retinal hemangioblastoma Cerebellar hemangioblas- VHL Tumor suppressor
disease toma

AD = autosomal dominant; AR = autosomal recessive; HPRT1 = hypoxanthine guanine phosphoribosyltransferase 1; TGF = transforming growth
factor; XL = X-linked; XLD = X-linked dominant; XLR = X-linked recessive. (Modified from Conneally M, Bird T, Engel AG, et al. Neurogenetics.
Continuum 2000;6[6]:36.)

of cases are sporadic. Both tuberin and hamartin have cortical hamartomas, heterotopic grey matter, and sub-
tumor suppressor activity. Cutaneous manifestations of ependymal giant cell astrocytomas (which may cause
tuberous sclerosis include hypomelanotic leaf-shaped acute hydrocephalus). Although cognitive and behav-
macules (ash-leaf spots), facial angiofibromas, periun- ioral problems are common, only about 50% of patients
gual fibromas, and shagreen patches. Ash-leaf spots are have mental retardation.11
the most frequent lesion present at the earliest age; Neurofibromatosis. Neurofibromatosis type 1 (NF1;
patients typically have 3 or more.10 Systemic manifesta- von Recklinghausen’s disease) is caused by a mutation in
tions include retinal lesions, cardiac rhabdomyomas, a gene on chromosome 17 that codes for neurofibromin;
renal angiomyolipomas, and pulmonary lymphangio- NF2 is caused by a mutation in a gene on chromosome
matosis. Seizures, especially infantile spasms, occur in at 22 that codes for neurofibromin-2 (also called merlin or
least 80% of diagnosed cases. Central nervous system shwannomin). Both genes have tumor suppressor activi-
(CNS) lesions include calcified subependymal nodules, ty. NF1 is a predominantly a peripheral nervous system

www.turner - white.com Neurology Volume 9, Part 1 7


Neurogenetics Review

disease, although it does have some CNS manifestations males and milder mental retardation and seizures (sub-
including high signal lesions in the basal nuclei on T2- cortical band heterotopia) in heterozygous females. It is
weighted MRI. Mild cognitive abnormalities and psychi- hypothesized that in females, X chromosome inacti-
atric symptoms may occur. NF1 is a cause of moyamoya vation leads to 2 distinct populations of migrating
disease. Diagnosis of NF1 requires the presence of 2 or neurons—normal neurons that form the cortex and
more of the following criteria: 6 or more café-au-lait neurons in which the normal DC allele has been inacti-
spots (> 5 mm in diameter before puberty or 15 mm in vated, which fail to migrate normally, forming the sub-
diameter after), axillary or inguinal freckling, optic glio- cortical band heterotopia.14
ma, 2 or more neurofibromas or 1 plexiform neurofi-
broma, a first-degree relative with NF1, and a charac- Epilepsy
teristic bone lesion (ie, sphenoid dysplasia). NF2 has Several genes responsible for familial epilepsy syn-
primarily CNS and cranial nerve manifestations, espe- dromes have been mapped. Many of these are muta-
cially bilateral vestibular shwannomas.11 Both NF1 and tions in genes that code for ion channels (see Table 3).
NF2 are associated with CNS neoplasms (eg, ependymo- Autosomal dominant nocturnal frontal lobe epilepsy is
mas, meningiomas, astrocytomas). caused by mutations that cause defects in the neuronal
Sturge-Weber syndrome typically presents as a facial nicotinic acetylcholine receptor. Patients experience
port-wine stain, most commonly involving the oph- clusters of brief motor seizures (usually with conscious-
thalmic distribution of the trigeminal nerve. The facial ness preserved) during non–rapid eye movement sleep.
angioma is frequently associated with an ipsilateral lep- Ictal findings on electroencephalogram (EEG) are
tomeningeal angioma that is calcified on computed characteristically inconclusive, potentially leading to a
tomography and enhances on MRI. Affected individu- diagnosis of psychogenic seizures. Benign familial neo-
als often develop progressive neurologic problems, in- natal convulsions is a rare autosomal dominant disorder
cluding difficult to control seizures, migraines, stroke- caused by a mutation in a voltage-gated potassium
like episodes, mental retardation, and hemiparesis. The channel that is one of the major regulators of neuronal
syndrome is postulated to result from a somatic muta- excitability in the brain. Brief, multifocal or generalized
tion resulting in mosaic alteration in fibronectin gene tonic clonic seizures typically begin at about 3 days of
expression.12 life. The seizures are associated with suppression of
von Hippel-Lindau disease consists of angiogenic EEG background. Seizures resolve within the first few
tumors in a variety of organs, especially hemangioblas- months of life but may recur later in life. Generalized
toma of the CNS and retina, renal cell carcinoma, epilepsy with febrile seizures is probably autosomal
pheochromocytoma, and pancreatic neuroendocrine dominant with variable penetrance and is the cause of
tumors. A variety of defects in the VHL gene (chromo- a prolonged course of febrile seizures (lasting beyond
some 11) impair its tumor suppressor activity.13 age 6 years) and some more grave phenotypes. It is
caused by mutations in voltage-gated sodium channel
Neuronal Migration Disorders subunits. Alexander disease—a rare cause of childhood
During brain development, neurons migrate along epilepsy, macrocephaly, and progressive neurodegener-
radial glial fibers from the ependymal zone (along the ation with Rosenthal fibers—has recently been linked
cerebral ventricles) to the cerebral cortex. Disruption of to mutations in a gene encoding glial fibrillary acidic
this process results in cerebral cortical malformations and protein (GFAP).15
heterotopias. One such disorder is classic lissencephaly
(4-layered cortex lacking gyri or sulci), which is caused by Dementia
a mutation in the LIS1 gene on chromosome 17. LIS1 Alzheimer’s disease (AD), the most common late-
codes for a subunit of platelet-activating factor that, onset dementia, is characterized by early deficits in for-
when inhibited, suppresses neuronal migration. Classic mation of new memories. Having a first-degree relative
lissencephaly is characterized by profound mental retar- with AD doubles the risk of developing late-onset AD.
dation and epilepsy; it may occur alone or in association The apolipoprotein E4 polymorphism is a risk factor for
with facial dysmorphism as part of the Miller-Dieker late-onset disease and a dose-dependent modifier of
syndrome. age of onset.16 Autosomal dominant mutations in amy-
Another gene responsible for lissencephaly, the dou- loid beta protein precursor (APP; chromosome 21),
ble cortex (DC) gene, was identified on the X chromo- presenilin 1 (PS1; chromosome 14), and presenilin 2
some. DC mutations cause an X-linked dominant disor- (PS2; chromosome 1) account for a significant propor-
der characterized by classic lissencephaly in hemizygous tion of early-onset AD (onset before age 60 years).

8 Hospital Physician Board Review Manual www.turner - white.com


Neurogenetics Review

Pathologically, brains of patients with AD bear neuro- new variant CJD were homozygous for methionine at
fibrillary tangles consisting of paired helical filaments codon 129, and a high percentage of patients with spo-
of hyperphosphorylated tau protein (microtubule- radic and iatrogenic CJD are homozygous for either
associated protein tau [MAPT]; chromosome 17) and methionine or valine at codon 129.20
senile plaques containing insoluble aggregates of
amyloid-beta (A-beta) protein. The protein product of CEREBROVASCULAR DISEASE
APP is cleaved by proteases (β secretase, γ secretase) to Cerebral autosomal dominant arteriopathy with sub-
form A-beta proteins of various lengths. Longer frag- cortical infarcts and leukoencephalopathy (CADASIL)
ments 42 to 42 amino acids in length (A-beta-42) are is a nonamyloid form of small-vessel disease caused by
prone to forming amyloid plaques. Mutations in PS1 mutations in the NOTCH3 gene on chromosome 19.
and PS2 are thought to alter γ secretase activity such NOTCH3 is one of a family of transmembrane receptors
that a higher proportion of amyloidogenic A-beta-42 is that have many roles in organismal development. Clin-
produced.17 ical manifestations include migraine with aura and re-
Frontotemporal dementia. Several neurodegenera- current deep white matter infarcts, leading to cognitive
tive disorders including frontotemporal dementia decline and pseudobulbar symptoms (worse in men);
(FTD) have been categorized as tauopathies on the average age of onset is 45 years.21,22
basis of neurofibrillary tangles pathology. Tau isoforms Cerebral cavernous malformations (CCMs) are dom-
present in the tangles differ somewhat among disor- inantly inherited vascular malformations that cause
ders. FTD typically causes a presenile dementia charac- seizures and cerebral hemorrhages, with associated focal
terized by early executive and language dysfunction. neurologic deficits. Three CCM loci have been mapped,
Autosomal dominant FTD with parkinsonism is associ- two to chromosome 7 (CCM1, CCM2) and one to chro-
ated with missense and splice site mutations in the tau mosome 3 (CCM3). Loss of function mutations have
protein sequence on chromosome 17.18 been identified in the KRIT1 (CCM1) and malcavernin
Prion disease. A small proportion of diseases linked (CCM2) genes. CCM3 is caused by a mutation in a pro-
to proteinaceous infectious particles (prions) are inher- grammed cell death gene (PDCD10).23
ited as autosomal dominant traits. All of these condi- The vascular type of Ehlers-Danlos syndrome (EDS
tions are caused by mutations in the human prion pro- IV), an autosomal dominant cause of structural defects
tein (PRNP) gene on chromosome 20. PRNP mutations in type III collagen, is associated with dissection of intra-
are associated with Creutzfeldt-Jakob disease (CJD), cranial arteries, aneurysm, carotid-cavernous fistula,
Gerstmann-Sträussler disease (GSD), and familial fatal and spontaneous rupture of bowel. Skin is thin, translu-
insomnia (FFI). CJD is characterized by dementia, atax- cent, and mildly extensible. The collagen defect can be
ia, and stimulus-sensitive myoclonus with spongiform identified in cultured skin fibroblasts.
change in the neocortex; GSDS is characterized by atax- Marfan syndrome, caused by a defect in the connec-
ia and dementia associated with the accumulation of tive tissue protein fibrillin 1, causes asthenic body and
multicentric prion protein–containing plaques in affect- facial type, mild to moderate joint laxity, vertebral col-
ed CNS regions; and FFI presents as insomnia and auto- umn abnormalities, subluxation of the lenses (ectopia
nomic symptoms followed by progressive dementia, lentis), dural ectasia, and spontaneous dissection of the
ataxia, dysarthria, myoclonus, and pyramidal signs asso- aortic and cerebral vessels.
ciated with degeneration of the thalamus.
Although several PRNP mutations have been associat- BASAL GANGLIA DISORDERS
ed with human disease, a glutamic acid to lysine change Huntington’s Disease
at codon 200 (E200K mutation) is the most frequent Huntington’s disease (HD) is an autosomal domi-
cause of inherited CJD, a proline to leucine substitution nant disorder characterized by chorea, cognitive de-
at codon 102 (P102L mutation) is most commonly asso- cline, and early degeneration of the caudate nucleus; it
ciated with GSD, and a mutation at codon 178 (D178N is caused by expansion of a polymorphic trinucleotide
mutation) accounts for FFI. In the presence of the repeat (CAG) in the 5′ coding region of the huntingtin
D178N mutation, disease phenotype is determined by gene on chromosome 4. Repeats range in number from
the type of polymorphism present at codon 129; valine at 9 to 37 in normal individuals and from 37 to 86 in HD
codon 129 plus the D178N mutation causes familial CJD, patients; genes with 42 or more CAG repeats are 100%
while methionine at codon 129 plus D178N produces penetrant. In the disease state, intranuclear inclusions
FFI.19 The valine-methionine polymorphism at codon consisting of polyglutamine fragments and truncated
129 also influences susceptibility to CJD; 100% of cases of huntingtin protein are found. The mean age of onset is

www.turner - white.com Neurology Volume 9, Part 1 9


Neurogenetics Review

Table 5. Loci and Genes Linked to Familial Parkinson’s tonia, DYT3 dystonia, and the autosomal recessive form
Disease
of dopa-responsive dystonia, the disorders are autoso-
Chromosomal Mode of mal dominant with variable penetrance.26 The follow-
Locus Location Gene Inheritance ing dystonias are the most clinically relevant to the neu-
PARK1 4q21.3 α-Synuclein AD rologist.
PARK2 6q25.2-27 Parkin AR DYT1 dystonia is strongly represented in Ashkenazi
PARK3 2p13 Unknown AD Jewish populations and typically causes early-onset
PARK4 4p15 Unknown AD generalized dystonia that initially involves 1 foot (also
PARK5 4p14 UCH-L1 AD known as primary or idiopathic torsion dystonia). The
PARK6 1p35-p36 PINK1 AR genetic defect is deletion of 1 of a pair of CAG repeats
PARK7 1p36 DJ1 AR in the gene encoding torsinA, leading to aggregation of
PARK8 12p11.2-q13.1 LRRK2 AD the protein in perinuclear inclusions.27
PARK9 1p36 Unknown AR* Dopa-responsive dystonia (DRD; Segawa syndrome)
PARK10 1p32 Unknown Late-onset sus- can result from any of several genetic defects that im-
ceptibility gene pair dopamine synthesis. DRD is an early-onset gener-
alized dystonia associated with diurnal fluctuations,
AD = autosomal dominant; AR = autosomal recessive; LRRK2 =
leucine-rich repeat kinase 2; PINK1 = PTEN-induced putative kinase 1;
parkinsonism, dramatic response to levodopa, and low
UCH-L1 = ubiquitin carboxyl-terminal esterase L1. (Adapted with per- levels of the dopamine metabolite homovanillic acid in
mission from Dawson TM, Dawson VL. Rare genetic mutations shed cerebrospinal fluid. The condition may be confused
light on the pathogenesis of Parkinson disease. J Clin Invest 2003; with AR-JP and cerebral palsy. A dominant form of DRD
111:146.) is caused by a mutation in the gene encoding GTP
*Kufor-Rakeb syndrome. cyclohydrolase 1 (GCH1; chromosome 14), which im-
pairs biosynthesis of tetrahydrobiopterin, a cofactor for
tyrosine hydroxylase. A defect in the tyrosine hydroxy-
between 35 and 44 years, leading to death after about lase gene (chromosome 11), which codes for the rate-
15 years. Onset earlier than age 20 years (Westphal vari- limiting enzyme in dopamine synthesis, causes a reces-
ant) is frequently associated with akinetic/rigid clinical sive form of DRD.28
signs and 60 or more CAG repeats.24 Dentatorubropal-
lidoluysian atrophy (DRPLA) is in the differential diag- Wilson’s Disease
nosis of these cases. Wilson’s disease is a recessively inherited form of
hepatolenticular degeneration that can present with an
Parkinson’s Disease ataxic, extrapyramidal, psychiatric, or mixed pheno-
Although Parkinson’s disease (PD) usually is sporadic, type. In Wilson’s disease, inadequate function of a cop-
10 genetic loci (PARK1–10) have been linked to PD per transporting ATPase (ATP7B) on chromosome 13
(Table 5). Several of these mutations affect proteins results in excessive free serum copper that accumulates
involved in or degraded by the ubiquitin-proteasomal in tissues, especially the liver, brain, and cornea (Kayser-
pathway. The most extensively studied and most common Fleischer ring seen on slit-lamp examination).
mutation in cases of early-onset PD (autosomal recessive
juvenile parkinsonism [AR-JP]) are mutations in the Pantothenate Kinase-associated Neurodegeneration
parkin gene (PARK2) on chromosome 6, which codes for Since it was described by Hallervorden and Spatz in
a ubiquitin-protein ligase. Features of this disease include 1922, this diagnosis has been applied to children and
early psychiatric symptoms, dystonia, excellent response young adults who develop progressive and inevitably
to levodopa, susceptibility to medication-induced dyski- fatal dystonia, dysarthria, and rigidity associated with
nesias, and relatively slow progression. PARK2 mutations iron deposition in the basal nuclei (“eye of the tiger”
are found in as many as 77% of sporadic cases when onset sign on T2-weighted MRI). Classic cases and one third
occurs earlier than age 20 but in only 3% when onset of atypical cases are correlated with mutations in the
occurs later than age 30 years.25 Most cases of genetic PANK2 gene, which encodes a pantothenatekinase that
parkinsonism do not have Lewy bodies. regulates coenzyme A synthesis.29

Dystonia SPINOCEREBELLAR ATAXIA


At least 14 genetic loci (DYT1–14) have been linked Genetic causes of SCA come in autosomal recessive,
to dystonia (Table 6). With the exception of DYT2 dys- autosomal dominant, and X-linked varieties. The

10 Hospital Physician Board Review Manual www.turner - white.com


Neurogenetics Review

Table 6. Dystonia Genetic Loci

Gene Locus Inheritance Phenotype Gene Product


DYT1 9q34 AD Early limb onset TorsinA
DYT2 Not mapped AR Early onset —
DYT3 Xq13.1 XR Filipino dystonia/parkinsonism Not identified
DYT4 Not mapped AD Whispering dysphonia —
DYT5 14q22.1 AD DRD/parkinsonism GCH1
TH 11p15.5 AR DRD TH
DYT6 8p AD Mixed Not identified
DYT7 18p AD Adult cervical Not identified
DYT8 2q33-35 AD PDC/PNKD Not identified
DYT9 1p21 AD Episodic choreoathetosis/ataxia/spasticity Not identified
DYT10 16 AD PKC/PKD Not identified
DYT11 7q21 AD Myoclonus dystonia Epsilon-sarcoglycan
DYT12 19q AD Rapid-onset dystonia/parkinsonism Not identified
DYT13 1p36.13-p36.32 AD Cervical/cranial/brachial Not identified
DYT14 14q13 AD DRD Not identified

AD = autosomal dominant; AR = autosomal recessive; DRD = dopa-responsive dystonia; GCH1 = GTP cyclohydrolase 1; PDC = paroxysmal
dystonic choreoathetosis; PKC = paroxysmal kinesigenic choreoathetosis or dyskinesia; PKD = paroxysmal kinesogenic dystonia; PNKD = parox-
ysmal non-kinesigenic dyskinesia; TH = tyrosine hydroxylase; XR = X-linked recessive. (Adapted with permission from Bressman SB. Dystonia
genotypes, phenotypes, and classification. In: Fahn S, Hallett M, DeLong MR, editors. Dystonia 4: advances in neurology. Vol. 94. Philadelphia:
Lippincott Williams & Wilkins; 2004:103.)

autosomal recessive forms are typically of earlier onset Although it may be of limited utility to memorize these
(< 20 years of age), whereas autosomal dominant conditions, it is important to recognize particular phe-
SCAs generally are of later onset (> 25 years of age), notypes. These disorders were initially classified by
although some have onset in childhood. X-linked Harding31 according to 3 clinical phenotypes: ADCA
SCAs are rare. type I (associated with varying degrees of dementia and
pyramidal, extrapyramidal, peripheral neuropathic, and
Autosomal Recessive Forms ophthalmologic signs), ADCA type II (cerebellar and
Friedreich’s ataxia is the most common hereditary retinal degeneration), and ADCA type III (pure cere-
form of spinocerebellar degeneration, characterized by bellar syndrome). The prototype of ADCA type I is
progressive limb and gait ataxia, neuropathy, areflexia, Machado-Joseph disease (SCA3), the most common
dysarthria, and pyramidal signs. A trinucleotide repeat form of SCA. This condition was initially described in
expansion on chromosome 9 causes diminished expres- Azorean Greek families but is now known to affect pa-
sion of frataxin, a mitochondrial protein encoded in tients in many ethnic groups; expanded alleles may be
the nucleus, which plays a role in iron homeostasis.30 quite large (up to 200 CAG repeats). SCA1 and SCA2
Although rare, ataxia caused by vitamin E deficiency are phenotypically similar to SCA3, although SCA2 is
(due to a defect in the α-tocopherol transfer protein) characterized by early slow saccades. The prototype for
has a similar phenotype to Friedreich’s ataxia and ADCA type II is SCA7, which causes optic atrophy and
responds to vitamin E. Ataxia-telangiectasia presents in pigmented retinopathy. The ADCA type III phenotype is
childhood with progressive ataxia, ocular and cuta- characteristic of SCAs 5, 6, 10, 11, 14, 15, 16, and 22.
neous telangiectasia, extrapyramidal signs, and cogni- Many ADCAs have onset in the third or fourth decade.
tive decline; it is caused by a defect in DNA repair (ATM The earliest is SCA13, which presents in early childhood
gene). The disease is associated with a high risk of with ataxia associated with motor and cognitive delay. The
hematologic malignancies and low serum IgA levels. latest is typically SCA6, a pure cerebellar late-onset syn-
drome associated with ophthalmoplegia. Episodic ataxia
Autosomal Dominant Forms type 1 (EA1) and episodic ataxia type 2 (EA2) typically
As medical genetics advances, the autosomal domi- have onset in childhood to adolescence. EA1 causes facial
nant cerebellar ataxias (ADCAs) continue to grow into a myokymia and very brief episodes of ataxia that are
bewildering number of genetically defined conditions. responsive to acetazolamide. EA2 causes attacks of ataxia

www.turner - white.com Neurology Volume 9, Part 1 11


Neurogenetics Review

Table 7. Selected Examples of Hereditary Spastic Paraplegia Syndromes


Locus Protein/Function Inheritance Clinical Syndrome
SPG1 L1 adhesion molecule XL Mental retardation, congenital abnormalities
SPG2 Proteolipid protein XL CNS white matter abnormality
SPG4 Spastin/binds microtubules AD Uncomplicated
SPG7 Paraplegin/mitochondrial protein AR Mitochondrial myopathy
SPG10 Kinesin heavy chain/axonal transport AD Uncomplicated
SPG20 Spartin AR Distal muscle wasting (Troyer syndrome)
SPG21 Unknown AR Childhood onset

AD = autosomal dominant; AR = autosomal recessive; CNS = central nervous system; SPG = spastic gait; XL = X= linked.

and signs of vestibulocerebellar dysfunction lasting min- X-linked recessively inherited disease caused by a CAG
utes to days, which are acetazolamide-unresponsive. repeat expansion in the androgen receptor gene. The
SCA12 presents with upper extremity and head tremor. androgen receptor is a ligand-dependent transcription
DRPLA, which is most common in Japanese populations, factor that may play a role in apoptosis. This disease,
is associated with myoclonus, chorea, dementia, parkin- like many polyglutamine repeat disorders, is associated
sonism, and epilepsy; the genetic defect is a CAG repeat with intracellular aggregates.
expansion in atrophin 1 on chromosome 12. DRPLA, Amyotrophic lateral sclerosis (ALS) is the most com-
SCA10, and SCA7 may be associated with epilepsy.5,32,33 mon sporadic cause of motor neuron degeneration. In
a minority of patients with familial ALS, a mutation in
SPINAL CORD DISORDERS the gene that codes for superoxide dismutase 1 (SOD1;
The hereditary spastic paraplegias (HSPs) are disor- copper-zinc superoxide dismutase) causes an autoso-
ders characterized by insidiously progressive lower limb mal dominant form of the disease. SOD1 is a cytoplas-
weakness and spasticity that produce progressive diffi- mic protein that catalyzes detoxification of oxygen free
culty walking.34 At least 21 loci have been linked to HSP. radicals generated by mitochondrial electron transport
Some HSPs are uncomplicated syndromes character- and the cytochrome P-450 system.35
ized only by progressive paraparesis; others are associat-
ed with additional neurologic abnormalities. The most PERIPHERAL NERVE DISORDERS
notable forms for which genes have been mapped are A clinical classification was created in 1975 for the
listed in Table 7. Some of the mutations that cause HSP hereditary motor and sensory neuropathies (HMSN),
code for proteins that may be involved in axonal trans- designating these disorders as HMSN I–VII. Once the
port (spastin, kinesin heavy chain, proteolipid protein). genetic defects responsible for some of these disorders
Some are mitochondrial proteins (paraplegin, chaper- were mapped, this classification made less sense from a
onin [heat shock protein] 60). Some are X-linked (pro- genetic point of view. HMSN I (demyelinating type) is
teolipid protein, L1 cell adhesion molecule). Pelizaeus- the same as Charcot-Marie-Tooth disease type 1 (CMT1),
Merzbacher disease, a progressive leukodystrophy and HSM II (axonal type) is the same as CMT2, but
associated with “tigroid” dysmyelination, is allelic with HMSN III (Dejerine-Sottas disease) can be caused by
an X-linked form of HSP. point mutations in the same genes that contain deletions
in CMT1. A more current classification of CMT neu-
ANTERIOR HORN CELL DISORDERS ropathies is shown in Table 9.36
The spinal muscular atrophies (SMAs) cause geneti-
cally predetermined degeneration of spinal and bulbar CMT Type 1 (CMT1)
motor neurons. SMAs have been classified on the basis CMT1A–D are autosomal dominant disorders (spo-
of age of onset and severity into 5 types (Table 8). Most radic in 20%) with onset in childhood or early adult-
are the result of recessive (loss of function) mutations. hood. These disorders are characterized by demyelinat-
The most important mutation associated with SMA is in ing and re-myelinating neuropathy leading to nerve
the gene encoding the survival motor neuron (SMN) hypertrophy (onion bulbs on pathologic nerve sec-
protein. Several other genetic factors are phenotypic tion), distal weakness, pes cavus, loss of stretch reflexes,
modifiers of SMN deletions, thought to produce dis- and postural (Roussy-Levy) tremor in one third of pa-
ease phenotypes of varying severity. Kennedy’s disease tients. Median nerve conduction velocities are less than
(X-linked spinal-bulbar muscular atrophy) is an 38 m/sec. CMT1A is caused by a duplication or point

12 Hospital Physician Board Review Manual www.turner - white.com


Neurogenetics Review

Table 8. Clinical Classification of Spinal Muscular Atrophies (SMAs)


Main Mode of Age of Pathologic
Type Inheritance Onset Features Hallmark Prognosis
SMA type I AR or XL (rare) In utero to Hypotonia and weakness; Never able to sit Avg life expectancy is
(Werdnig-Hoffmann 6 mo problems with sucking, swal- without sup- 8 mo; 95% dead before
disease) lowing, and breathing port age 1 y
SMA type II AR 3–15 mo Proximal leg weakness, fasci- Death usually occurs
(Dubowiz disease, culations, fine hand tremor after age 2 y
intermediate form)
SMA type III AD, AR 15 mo to teen Proximal leg weakness, Can sit but never Varies depending on
(Wohlfart-Kugelberg- years delayed motor milestones able to stand; extent and timing of
Welander disease, facial muscles respiratory complica-
chronic SMA) spared tions
SMA type IV (adult- AD, AR 37 y (avg) Proximal weakness variable Life expectancy not
onset SMA) and more severe in AD form markedly reduced
Distal SMA type V AD, AR AR: birth or Distal weakness Very slow clinical pro-
(progressive SMA, infancy gression; does not alter
Charcot-Marie- AD: adulthood life span
Tooth type-SMA)

AD = autosomal dominant; AR = autosomal recessive; XL = X-linked. (Adapted with permission from Cole N. Genetic disorders of motor neu-
rons. Semin Neurol 1999;19:408.)

mutation in the peripheral myelin protein-22 (PMP22) Table 9. Charcot-Marie-Tooth (CMT) Neuropathies and
gene on chromosome 17. The protein product is local- Related Disorders
ized to the compact portion of peripheral myelin, Chromosome
contains 4 transmembrane domains, and is highly con- Disorder Locus Gene
served in evolution. The interesting aspect of PMP22 CMT type 1 (CMT1)*
mutations is that a duplication of this genetic material CMT1A 17p11.2-12 PMP22
causes early-onset demyelinating neuropathy, but a de- CMT1B 1q22-23 Po
letion in this gene causes the less severe phenotype, CMT1C Unknown Unknown
hereditary neuropathy with liability to pressure palsies CMT type 2 (CMT2)
(HNPP). CMT1B is caused by point mutations or small CMT2A 1p36 Unknown
deletions in the myelin protein zero (P0) gene, linked CMT2B 3q13-22 Unknown
to the Duffy blood group on chromosome 1q. CMT1D CMT2C Unknown Unknown
has been linked to mutations in early growth response CMT2D 7p14 Unknown
gene 2 (EGR2), a zinc finger protein that has homolo- Dejerine-Sottas disease (DSD)*
gy to a mouse transcription factor. DSDA 17p11.2-12 PMP22
DSDB 1q22-23 Po
CMT Type 2 (CMT2) CMT type 4 (CMT4)
CMT2 is an axonal neuropathy (median nerve con- CMT4A 8q13-21.1 Unknown
duction velocities > 38 m/sec) that is inherited as an CMT4B 11q23 MTMR2
autosomal dominant, autosomal recessive, or X-linked CMT4C 5q23-33 Unknown
trait. The recessive and X-linked forms are rare. The CMTX Xq13.1 Connexin 32
disorder typically presents later than CMT1 with distal HNPPA 17p11.2-12 PMP22
sensory loss and muscle weakness and atrophy. CMT2B HNPPB Unknown Unknown
is associated with poorly healing ulcers. CMT2C is asso-
CMTX = X-linked dominant Charcot-Marie-Tooth disease; HNPP =
ciated with distal spinal muscular atrophy and vocal hereditary neuropathy with liability to pressure palsies; MTMR2 =
cord paralysis. CMT2A and CMT2B have now been myotubularin-related protein-2. (Adapted with permission from
linked to more than 1 gene each, and CMT2D–K have Mendell JR, Kissel JT, Cornblath DR. Diagnosis and management of
been described based on genetic linkage.37 peripheral nerve disorders. New York: Oxford University Press;
2001:430. Copyright © 2001.)
Dejerine-Sottas Disease *Early growth response gene mutations on chromosome 10q21.1-22.2
Dejerine-Sottas disease (DSD) is a severe (usually cause CMT1 and DSD.

www.turner - white.com Neurology Volume 9, Part 1 13


Neurogenetics Review

sporadic but also autosomal dominant) hypertrophic, take or exercise, and are improved by oral glucose
demyelinating neuropathy with severe slowing of intake. In this condition, single amino acid substitutions
conduction velocities and onset in infancy or early in the alpha subunit of the skeletal muscle voltage-
childhood. DSD is frequently caused by point muta- gated sodium channel cause defective inactivation of
tions in PMP22, P0, and others. the open channel. Different point mutations in the
same channel cause paramyotonia congenita or overlap
CMT Type 4 syndromes. Paramyotonia congenita causes muscle stiff-
CMT4 neuropathies are autosomal recessive demyeli- ness, particularly involving the neck, face, and hands
nating disorders. CMT4A and CMT4B have very early and provoked by cold exposure of exercise. Some forms
onset (before age 3 years) and delayed motor mile- are responsive to acetazolamide.38
stones. CMT4C is characterized by severe scoliosis and
mild distal neuropathy, presenting prior to age 10 years. Malignant Hyperthermia
In skeletal muscle, excitation-contraction coupling
X-Linked Dominant CMT is accomplished by an interaction between voltage-
X-linked dominant CMT (CMTX) has a more dependent L-type calcium channels in the T tubule
severe phenotype and earlier onset in males. The phe- membrane and calcium release channels along the sar-
notype is similar to CMT1, although in contrast to coplasmic reticulum.39 Both L-type channel and sar-
CMT1, affected males may have delayed central con- coplasmic reticulum calcium release channel complex-
duction on brain stem auditory evoked responses. es are named for their ligands, dihydropyridine (a
CMTX is the result of a point mutation in a gap junc- calcium channel blocker) and ryanodine (a plant alka-
tion protein (connexin 32) and, thus, is also a chan- loid), respectively. Malignant hyperthermia is an auto-
nelopathy, but it does not have episodic features.36 somal dominant condition caused by mutations in the
ryanodine receptor, which result in excessive calcium
NEUROMUSCULAR JUNCTION DISORDERS release from the sarcoplasmic reticulum in response to
The congenital myasthenic syndromes (CMS) are dis- halothane and succinylcholine. Affected individuals
orders of the neuromuscular junction classified by the who have been given these agents develop masseter
site of the transmission defect (ie, presynaptic, synaptic, spasm, rigidity, hyperpyrexia, hemodynamic instability,
postsynaptic). The majority of these syndromes are and rhabdomyolysis.40
caused by recessive mutations in genes encoding sub-
units of the muscle nicotinic acetylcholine receptor.11 Muscular Dystrophies
Slow channel postsynaptic syndromes are caused by Dystrophin deficiency disorders. Duchenne muscu-
autosomal dominant gain-of-function mutations. CMS lar dystrophy is an X-linked disorder characterized by
is characterized by significant fatigable weakness with early onset progressive proximal muscle weakness, calf
onset in childhood, decremental response to 2-Hz stim- pseudohypertrophy, cardiomyopathy, massively elevat-
ulation on electromyelography, and negative acetylcho- ed creatine kinase, and limited life span. In Duchenne
line receptor antibodies. muscular dystrophy, muscle immunocytochemistry
reveals almost complete loss of dystrophin protein. Dys-
MUSCLE DISORDERS trophin, syntrophins, sarcoglycans, and dystroglycans
Periodic Paralyses are structural elements of the dystrophin-glycoprotein
Hypokalemic periodic paralysis is in most cases caused complex that links the muscle cell cytoskeleton with the
by point mutations in the alpha 1 subunit of the L-type extracellular matrix.41 A milder variant, Becker muscu-
(dihydropyridine-sensitive) calcium channel. Onset is lar dystrophy, is associated with a milder reduction in
most common in the second decade and consists of dystrophin staining. About one third of cases of
attacks of generalized weakness lasting hours to 1 day. Res- Duchenne and Becker muscular dystrophy result from
piration is not compromised. Attacks are precipitated by new mutations, probably in part due to the dystrophin
exercise, heavy carbohydrate load, or other conditions gene (locus, Xp21) being one of the largest known
that increase insulin (thus lowering serum potassium). genes. Most of the mutations that produce complete
Hyperkalemic periodic paralysis is characterized by loss of functional dystrophin cause a frame shift. About
attacks of muscle stiffness and ache followed by diffuse 8% of female carriers are affected. In about 60% of
muscle weakness lasting 15 minutes to 1 hour, begin- cases the diagnosis can be made by DNA analysis. In the
ning in the first decade of life. The attacks tend to occur remainder (point mutations), diagnosis must be made
in the morning, can be precipitated by potassium in- by muscle immunocytochemistry.

14 Hospital Physician Board Review Manual www.turner - white.com


Neurogenetics Review

Limb-girdle muscular dystrophy (LGMD) refers to principles and practice of medical genetics. 3rd ed. New
genetic conditions that cause gradually progressive York: Churchill Livingstone; 2002:104–24.
proximal weakness, often beginning in the hip girdle, 3. Wallace DC. Mitochondrial diseases in man and mouse.
then shoulder girdle, and then distal muscles. Trapezius Science 1999;283:1482–8.
atrophy may be striking. Age of onset varies from child- 4. DiMauro S, Schon EA. Mitochondrial respiratory-chain
hood to adulthood, depending on the genetic defect. diseases. N Engl J Med 2003;348:2656–68.
LGMD occurs in both autosomal dominant and auto- 5. Everett CM, Wood NW. Trinucleotide repeats and neu-
somal recessive forms, with the latter more common. rodegenerative disease. Brain 2004;127(Pt 11):2385–405.
Many of the recessive LGMDs are caused by mutations 6. Hubner CA, Jentsch TJ. Ion channel diseases. Hum Mol
in genes that code for sarcoglycans of the dystrophin- Genet 2002;11:2435–45.
glycoprotein complex. 7. Kullmann DM. The neuronal channelopathies. Brain
Myotonic dystrophy is an autosomal dominant disor- 2002;125(Pt 6):1177–95.
der caused by a CTG repeat in an untranslated region 8. Ptacek LJ, Fu YH. Channels and disease: past, present,
of chromosome 19. Clinical findings include myotonia, and future. Arch Neurol 2004;61:1665–8.
progressive proximal muscle weakness, cataracts, hypo- 9. Hagerman RJ, Leehey M, Heinrichs W, et al. Intention
gonadism, frontal balding, cardiac conduction abnor- tremor, parkinsonism, and generalized brain atrophy in
malities, respiratory insufficiency, insulin resistance, male carriers of fragile X. Neurology 2001;57:127–30.
mild cognitive disorder, and other systemic symptoms. 10. Jozwiak S, Schwartz RA, Janniger CK, et al. Skin lesions in
In classic myotonic dystrophy (type 1), the number of children with tuberous sclerosis complex: their preva-
CTG repeats in leukocytes may be 100 to 750, but the lence, natural course, and diagnostic significance. Int J
number of CTG repeats in skeletal muscle may be as Dermatol 1998;37:911–7.
many as 5000, suggesting that the repeat expands dur- 11. Conneally M, Bird T, Engel AG, et al. Neurogenetics.
ing mitosis or even in postmitotic tissues. Maternal in- Continuum 2000;6:9–154.
heritance of an allele that has expanded beyond 750 12. Comi AM, Hunt P, Vawter MP, et al. Increased fibro-
CTG repeats may result in a severe congenital form of nectin expression in Sturge-Weber syndrome fibroblasts
myotonic dystrophy associated with cardiomyopathy and brain tissue. Pediatr Res 2003;53:762–9.
and mental retardation. 13. Shuin T, Yamazaki I, Tamura K, et al. Recent advances in
Proximal myotonic myopathy (PROMM; myotonic ideas on the molecular pathology and clinical aspects of
dystrophy type 2) is an autosomal dominant disorder von Hippel-Lindau disease. Int J Clin Oncol 2004;9:
with clinical features similar to but milder than those of 283–7.
myotonic dystrophy; it is caused by expansion of a 14. Gleeson JG, Minnerath SR, Fox JW, et al. Characterization
CCTG repeat in intron 1 of the zinc finger protein-9 of mutations in the gene doublecortin in patients with
gene.42 In contrast to myotonic dystrophy, patients with double cortex syndrome. Ann Neurol 1999;45:146–53.
PROMM present with predominant weakness and atro- 15. Brenner M, Johnson AB, Boespflug-Tanguy O, et al.
phy in proximal muscles. Mutations in GFAP, encoding glial fibrillary acidic pro-
Oculopharyngeal muscular dystrophy, which pre- tein, are associated with Alexander disease. Nat Genet
sents with progressive dysphagia, ptosis, and proximal 2001;27:117–20.
weakness, usually in the fifth or sixth decade, is caused 16. Corder EH, Saunders AM, Strittmatter WJ, et al. Gene
by small GCG expansions in the polyadenylate-binding dose of apolipoprotein E type 4 allele and the risk of
protein 2 (PABP2) gene.43 Alzheimer’s disease in late onset families. Science 1993;
261:921–3.
17. Selkoe DJ. Presenilin, Notch, and the genesis and treat-
REFERENCES ment of Alzheimer’s disease. Proc Natl Acad Sci U S A
2001;98:11039–41.
1. Barsh G, Epstein C, et al. Genome structure and gene 18. Hutton M, Lendon CL, Rizzu P, et al. Association of mis-
expression. In: Rimoin DL, Connor JM, Pyeritz RE, edi- sense and 5′-splice-site mutations in tau with the inherit-
tors. Emery and Rimoin’s principles and practice of med- ed dementia FTDP-17. Nature 1998;393:702–5.
ical genetics. 3rd ed. New York: Churchill Livingstone; 19. Wild O, Kretzschmar H. Prion diseases. In: Pulst S, edi-
2002:60–82. tor. Neurogenetics. New York: Oxford University Press;
2. Cook J, Lam W, et al. Mendelian inheritance. In: Rimoin 2000:191–218.
DL, Connor JM, Pyeritz RE, editors. Emery and Rimoin’s 20. Johnson RT, Gibbs CJ Jr. Creutzfeldt-Jakob disease and

www.turner - white.com Neurology Volume 9, Part 1 15


Neurogenetics Review

related transmissible spongiform encephalopathies. 31. Harding AE. The clinical features and classification of
N Engl J Med 1998;339:1994–2004. the late onset autosomal dominant cerebellar ataxias. A
21. Opherk C, Peters N, Herzog J, et al. Long-term progno- study of 11 families including descendants of the ‘the
sis and causes of death in CADASIL: a retrospective study Drew family of Walworth.’ Brain 1982;105(Pt 1):1–28.
in 411 patients. Brain 2004;127(Pt 11):2533–9. 32. Evidente VG, Gwinn-Hardy KA, Caviness JN, Gilman S.
22. Rafalowska J, Dziewulska D, Fidzianska A, et al. CADASIL: Hereditary ataxias. Mayo Clin Proc 2000;75:475–90.
what component of the vessel wall is really a target for 33. Blindauer K. Cerebellar disorders and spinocerebellar
Notch 3 gene mutations? Neurol Res 2004;26:558–62. ataxia. Continuum 2004;10:154–73.
23. Bergametti F, Denier C, Labauge P, et al. Mutations with- 34. Fink JK. The hereditary spastic paraplegias: nine genes
in the programmed cell death 10 gene cause cerebral cav- and counting. Arch Neurol 2003;60:1045–9.
ernous malformations. Am J Hum Genet 2005;76:42–51.
35. Cole N, Siddique T. Genetic disorders of motor neurons.
24. Leavitt BR, Wellington CL, Hayden MR. Recent insights Semin Neurol 1999;19:407–18.
into the molecular pathogenesis of Huntington disease
36. Mendell JR. Charcot-Marie-Tooth neuropathies and re-
[published erratum appears in Semin Neurol 2000;20:
lated disorders. Semin Neurol 1998;18:41–7.
269]. Semin Neurol 1999;19:385–95.
37. Neuromuscular disorders: gene location. Neuromuscul
25. Lucking CB, Durr A, Bonifati V, et al. Association be-
Disord 2005;15:89–114.
tween early-onset Parkinson’s disease and mutations in
the parkin gene. French Parkinson’s Disease Genetics 38. Bulman DE. Phenotype variation and newcomers in ion
Study Group. N Engl J Med 2000;342:1560–7. channel disorders. Hum Mol Genet 1997;6:1679–85.
26. Comella C. Dystonia and related diseases. Continuum 39. Loke J, MacLennan DH. Malignant hyperthermia and
2004;10:89–113. central core disease: disorders of Ca2+ release channels.
Am J Med 1998;104:470–86.
27. Torres GE, Sweeney AL, Beaulieu JM, et al. Effect of
torsinA on membrane proteins reveals a loss of function 40. Denborough M. Malignant hyperthermia. Lancet 1998;
and a dominant-negative phenotype of the dystonia- 352:1131–6.
associated DeltaE-torsinA mutant. Proc Natl Acad Sci 41. Tsao CY, Mendell JR. The childhood muscular dystro-
U S A 2004;101:15650–5. phies: making order out of chaos. Semin Neurol 1999;
28. Knappskog PM, Flatmark T, Mallet J, et al. Recessively 19:9–23.
inherited L-DOPA-responsive dystonia caused by a point 42. Bachinski LL, Udd B, Meola G, et al. Confirmation of the
mutation (Q381K) in the tyrosine hydroxylase gene. type 2 myotonic dystrophy (CCTG)n expansion mutation
Hum Mol Genet 1995;4:1209–12. in patients with proximal myotonic myopathy/proximal
29. Hayflick SJ, Westaway SK, Levinson B, et al. Genetic, clin- myotonic dystrophy of different European origins: a sin-
ical, and radiographic delineation of Hallervorden-Spatz gle shared haplotype indicates an ancestral founder
syndrome. N Engl J Med 2003;348:33–40. effect. Am J Hum Genet 2003;73:835–48.
30. Pandolfo M. Molecular pathogenesis of Friedreich atax- 43. Brais B, Rouleau GA, Bouchard JP, et al. Oculopharyn-
ia. Arch Neurol 1999;56:1201–8. geal muscular dystrophy. Semin Neurol 1999;19:59–66.

Copyright 2005 by Turner White Communications Inc., Wayne, PA. All rights reserved.

16 Hospital Physician Board Review Manual www.turner - white.com

S-ar putea să vă placă și