Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/8929770

Spinal muscular atrophy: Molecular genetics and diagnostics

Article  in  Expert Review of Molecular Diagnostics · February 2004


DOI: 10.1586/14737159.4.1.15 · Source: PubMed

CITATIONS READS
115 994

2 authors, including:

Shuji Ogino
Brigham and Women's Hospital
674 PUBLICATIONS   34,446 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

NET translation medicine research project View project

Colorectal cancer View project

All content following this page was uploaded by Shuji Ogino on 29 May 2014.

The user has requested enhancement of the downloaded file.


Review

For reprint orders, please contact reprints@future-drugs.com

Spinal muscular atrophy:


molecular genetics and diagnostics
Shuji Ogino† and Robert B Wilson

Spinal muscular atrophy is one of the most common autosomal recessive diseases,
affecting approximately one in 10,000 live births and with a carrier frequency of
approximately one in 50. Spinal muscular atrophy is caused by a deficiency of the
ubiquitous protein survival of motor neuron (SMN), which is encoded by the SMN genes,
SMN1 and SMN2. Due to a single nucleotide polymorphism (840C>T), SMN2 produces less
full-length transcript than SMN1 and cannot entirely prevent neuronal cell death at
CONTENTS physiologic gene dosages. The 38-kDa SMN protein comprises 294 amino acids and is
involved in the biogenesis of uridine-rich small nuclear ribonucleoproteins, facilitating their
Clinical features
cytoplasmic assembly into the spliceosome. Various animal models have been developed
Molecular genetics to study the pathogenesis of spinal muscular atrophy, as well as to test novel therapeutics.
Molecular diagnostics Common PCR-restriction fragment length polymorphism assays can detect the
Genetic counseling & homozygous absence of SMN1 in approximately 94% of patients with clinically typical
risk assessment spinal muscular atrophy. SMN gene dosage analysis can determine the copy number of
Potential therapeutic SMN1 to detect carriers and patients heterozygous for the absence of SMN1. Due to the
approaches genetic complexity and the high carrier frequency, accurate risk assessment and genetic
Conclusion counseling are particularly important. Comprehensive SMA genetic testing, combined with
appropriate genetic counseling and risk assessment, provides the most complete
Expert opinion
evaluation of patients and their families at this time. New technologies, such as monosomal
Five-year view
analysis techniques, may be widely available in the future.
Key issues
Expert Rev. Mol. Diagn. 4(1), 15–29 (2004)
References
Affiliations Clinical features may also be affected in SMA Type I patients
Typical features [4]. Type II SMA (intermediate; OMIM
Spinal muscular atrophy (SMA) is one of the number 253550) is characterized by the onset
most common autosomal recessive diseases, of proximal muscle weakness before
affecting approximately one in 10,000 live 18 months of age, the ability to sit but not to
births and with a carrier frequency of approxi- walk unaided and survival beyond 4 years of
mately one in 50 [1]. SMA is characterized by age. Type III SMA (Kugelberg–Welander dis-
symmetric proximal muscle weakness second- ease; OMIM number 253400) is characterized

ary to degeneration of the anterior horn cells by the onset of proximal muscle weakness
Author for correspondence
Department of Pathology,
of the spinal cord. SMA is clinically sub- after the age of 2 years, the ability to walk
Brigham and Women’s Hospital, divided into three types based on age of onset independently until the disease progresses
75 Francis Street, Amory 3rd Floor, and clinical severity, according to established and survival into adulthood. It was proposed
Boston, MA 02115, USA diagnostic criteria [2,3]. Type I SMA (Werdnig– that Type III SMA be subdivided into
Tel.: +1 617 632 3978 Hoffmann disease; Online Mendelian Inheri- Type IIIa and Type IIIb, depending on age of
Fax: +1 617 632 5762
sogino@partners.org
tance in Man [OMIM] database number onset (Type IIIa before 3 years and Type IIIb
253300) is characterized by the onset of after 3 years) [5]. The probabilities of being
KEYWORDS: severe muscle weakness and hypotonia in the ambulatory at 10, 20 and 40 years after onset
carrier, conversion, deletion,
gene dosage, genetic counseling, first few months of life and the inability to sit were 73, 44 and 34%, respectively, in patients
genetic testing, risk assessment, or walk. Fatal respiratory failure usually occurs with SMA Type IIIa, and 97, 89 and 67%,
SMN, SMN1, SMN2
before the age of 2 years. Sensory neurons respectively, in patients with SMA Type IIIb [5].

www.future-drugs.com © Future Drugs Ltd. All rights reserved. ISSN 1473-7159 15


Ogino & Wilson

Approximately 60% of SMA patients have Type I and the The authors previously reported the case of an infant with
remaining patients have Type II or Type III, with Type II homozygous absence of SMN1 who was suspected to have
being more common [6]. sudden infant death syndrome (SIDS) [16]. Another report
described an infant who nearly suffered sudden death at the age
Atypical features of 7 weeks due to diaphragmatic paralysis and was later diag-
Type IV (or so-called adult-type) SMA has been a controversial nosed to have Type I SMA [17]. Diaphragmatic paralysis and
entity in the literature, especially with regard to the age of respiratory distress were reported as initial manifestations of
onset [7–9]. It has been defined by some as SMA with an age of SMA in six infants [18–20]. However, the diagnosis of SMN1-
onset after 30 years [5]. Although Type III SMA is character- related SMA may not be appropriate in a setting of muscle weak-
ized by a rather variable age of onset spanning three decades, ness that is predominantly distal and the status of SMN1 in these
few individuals with Type III SMA become symptomatic in infants was not determined. The authors speculate that the cause
their twenties [5]. SMN1 was not homozygously absent in four of some clinically unexplained infant deaths may be SMA.
patients with SMA and an age of onset after 30 years, although Strictly speaking, the diagnosis of SIDS is inappropriate if a
they fulfilled the diagnostic criteria of SMA [8]. In another homozygous SMN1 mutation confirms the diagnosis of SMA.
study, five patients with ages of onset of SMA ranging from 20 More studies are necessary to draw a definitive conclusion on the
to 32 years did show homozygous absence of SMN1 [7]. It frequency of undiagnosed SMA being misdiagnosed as SIDS.
remains to be determined whether Type IV patients who do
not lack both copies of SMN1 have small intragenic mutations Molecular genetics
in SMN1 or have an SMN1-unrelated disease. Disease gene SMN1 & its homolog SMN2
Rare examples of SMN1-related SMA with atypical clinical SMA of all types is associated with homozygous mutations in
features have been described. These features include extended the survival of motor neurone 1 gene (SMN1; OMIM
CNS involvement (i.e., cerebral atrophy, abnormal head cir- number 600354) [21]. SMN1 and its centromeric homolog,
cumference, pyramidal tract signs and cerebellar signs), con- SMN2 (OMIM number 601627), lie within the telomeric
genital fiber type disproportion (Type 1 fiber atrophy and and centromeric halves, respectively, of a large inverted repeat
Type 2 fiber hypertrophy), creatine kinase (CK) values greater in chromosome region 5q13 [21]. SMN consists of nine exons
than 1000 U/l, congenital malformations of other organs (i.e., (exons 1, 2a, 2b and 3–8), with the stop codon present near
ventricular septal defects of the heart and urogenital malforma- the end of exon 7 [22]. The coding sequence of SMN2 differs
tions) and complex syndromes [10]. It remains to be determined from that of SMN1 by a single nucleotide (840C>T) in
whether these rare examples include deletions of additional, exon 7 (FIGURE 1) [22]. In contrast to SMN1, of which at least
contiguous genes (i.e., represent contiguous gene syndromes); one copy is indispensable for the normal survival of motor
are part of the spectrum of SMN1-related defects; or represent neurons, SMN2 is dispensable since approximately 5–10% of
coincidental, simultaneous occurrences of conditions unlinked normal individuals lack both copies [21,23,24]. The failure of
to chromosome 5q. SMN2 to compensate fully for the loss of SMN1 is due to the
Arthrogryposis represents a group of heterogeneous syn- sequence difference in exon 7. Although translationally silent,
dromes, all of which have joint contractures present at birth [11]. the 840C>T transition in SMN2 has been reported to
The major clinical form, arthrogryposis multiplex congenita decrease the activity of an exonic splicing enhancer so that less
(AMC), is caused by decreased fetal movements in utero, due to full-length transcript is generated, leading to deficiency of
a large number of conditions, including neuropathies, myo- normal, stable SMN protein [25,26]. More recent evidence sug-
pathies, connective tissue diseases and conditions that limit the gests that the 840C>T transition in SMN2 activates an exonic
space within the uterus [12]. SMA with congenital bone frac- splicing silencer, which functions as a binding site for the
tures (which is chromosome 5 unlinked) is a rare syndrome known repressor protein hnRNP A1 [27].
characterized by floppiness in infancy and by multiple fractures
of the long bones [13]. The features of SMA with congenital SMN protein & the SMN complex
bone fractures overlap with those of SMA with congenital SMN is a ubiquitously expressed 38-kDa protein consisting
arthrogryposis and bone fractures [10]. Six of 12 patients with of 294 amino acids. Most of the SMN protein in a normal
neurogenic AMC showed homozygous absence of SMN1 [14]. individual is derived from SMN1, as indicated in part by the
Two of four patients with arthrogryposis, not otherwise speci- fact that there is only a very small amount of the SMN pro-
fied, showed homozygous absence of SMN1 [15]. By contrast, tein derived from SMN2 in affected individuals with a
two of two patients affected with SMA with congenital arthro- homozygous absence of SMN1. SMA patients with a
gryposis and bone fractures did not show homozygous absence homozygous absence of SMN1 always have at least one copy
of SMN1 [10], as well as only one of seven cases referred for of SMN2 and a homozygous deletion of the mouse SMN
arthrogryposis in another study [16]. However, comparing these gene causes embryonic lethality. Thus, the SMN protein is
data is problematic because of the heterogeneous etiologies for essential for viability. The SMN protein protected neurons
arthrogryposis and the lack of uniform inclusion criteria in in culture and in vivo from virus-induced apoptosis, whereas
these studies. disease-associated mutant forms of the protein were proapoptotic

16 Expert Rev. Mol. Diagn. 4(1), (2004)


Spinal muscular atrophy

in the same systems [28]. It remains unexplained why motor with fresh Sm proteins to regain its activity and this loading
neurons are more susceptible than other cell types to defi- reaction may be regulated by the PRMT5 complex, composed
ciency of such a ubiquitously expressed protein. It is likely of methylosome (plCln, WD45 and PRMT5 [JBP1]) and Sm
that requirements for the SMN protein differ among various proteins [30,31]. Sm proteins B, D1 and D3 are methylated by
cell types. methylosome before loading into the SMN complex [30].
SMN is involved in the biogenesis of uridine-rich small
nuclear ribonucleoproteins (UsnRNPs) and facilitates cytoplas- SMN1 deletion & conversion mutations
mic assembly of UsnRNPs into the spliceosome, a large Approximately 94% of individuals with clinically typical SMA
snRNA–protein complex that catalyzes pre-mRNA splicing lack both copies of SMN1 exon 7 [32]. Loss of SMN1 can occur
[29,30]. UsnRNP biogenesis by the SMN complex is well by deletion (typically a large deletion that includes the whole
described in a recent review [30]. In the nucleus, SMN appears gene [33–35]) or by conversion to SMN2 [36]. Affected individuals
to be directly involved in pre-mRNA splicing. SMN associates lacking both copies of SMN1 exon 7 but retaining at least one
with multiple proteins to form a complex [30,31]. Core SMN- copy of SMN1 exon 8 sequence have been described [21,36]. In
interacting proteins in the complex include Gemin2, Gemin3, some of these individuals, exon 8 of SMN1 was found to be
Gemin4, Gemin5, Gemin6 and Gemin7. Substrates and sub- physically juxtaposed to exon 7 of SMN2, consistent with an
stoichiometric components include Sm and Sm-like (LSm) SMN1-to-SMN2 gene conversion event involving the func-
proteins B, D1, D2, D3, E, F and G; uridine-rich snRNAs tionally critical exon 7 sequence difference [36,37]. A large
(UsnRNAs); fibrillarin; GAR1; coilin; profilin; ZPR1; OSF; majority of genes converted from SMN1 to SMN2 had SMN2
nucleolin; B23; hsc70; RNA helicase A; RNA polymerase II; sequences in all of the polymorphic nucleotides except for the
snurportin; importin β; galectin1/3; and heterogeneous nuclear SMN1 sequence in exon 8 and half of the hybrid
RNP-Q and RNP-R [29–31]. UsnRNP assembly is mediated by SMN1/SMN2 genes had a single Ag1-CA/C212 haplotype,
the SMN complex, which subsequently facilitates hypermethy- suggesting that these hybrid SMN alleles had a common origin
lation of the methyl-G cap of UsnRNAs [30]. The hypermethy- [37]. Minor variants of hybrid SMN genes have been described.
lated cap (m3G-cap) structure is specifically bound by snurpor- These include SMN2 intron 6 and exon 7 juxtaposed to
tin, which then interacts with the import receptor importin β SMN1 intron 7 and exon 8 [37], SMN2 exon 7 flanked by
and, together with an as-yet-unknown import receptor that SMN1 intron 6 and exon 8 [38] and SMN1 sequences in all of
recognizes the Sm core, mediates UsnRNA import [30]. After the polymorphic nucleotides except for the SMN2 sequence in
each round of assembly, the SMN complex must be reloaded exon 8, the last of which was found in healthy individuals [37].

20 kb
Centromeric Telomeric
SMN2 SMN1

500 Kb 500 Kb
Large inverted duplication in 5q13
SMN

1 2a 2b 3 4 5 6 7 8

g C a a G

SMN1

SMN2
7 8
a T g g A

Essential difference
Figure 1. The survival of motor neuron gene (SMN) consists of 9 exons, with the stop codon near the end of exon 7. The coding sequence of SMN2 differs
from that of SMN1 by a single nucleotide (840C>T) in exon 7.

www.future-drugs.com 17
Ogino & Wilson

A number of hypotheses have been proposed for the generation Some of these small intragenic mutations have been placed
of these hybrid genes, including unequal recombination, intra- into expression constructs and the effects on oligomerization
chromosomal deletion and gene conversion [37]. At least one of have been investigated. In vitro experiments showed that
the SMN2 copies from mildly affected patients with SMA was G279V (836G>T) and Y272C (815A>G) caused a severe
located in the telomeric position [39]. Taken together with the impairment of oligomerization, while S262I (785G>T) and
increased number of SMN2 copies generally in mildly affected T274I (821C>T) caused a mild impairment [51]. The degree of
patients with SMA, these data are consistent with gene conver- impairment correlated with the severity of SMA associated with
sion from SMN1 to SMN2. The absence of SMN1 exon 7 the mutations in vivo [51]. A nonsense mutation, W102X
sequence correlates with either a functional or a de facto (305G>A), caused exon 3 skipping and thereby generated a
absence of the SMN1 gene. short form of SMN transcript lacking exon 3 [52]. Western blot
At least one of the factors underlying the variable phenotype analysis showed a band at the size expected for SMN protein
of SMA is the SMN2 copy number. Individuals with Type III lacking the amino acids encoded by exon 3. These mutations
SMA have, on average, more copies of SMN2 than individuals were detected in three patients who had only two copies of
with Type II or Type I SMA and likely express more full-length SMN2 but mild symptoms [52].
SMN transcript derived from the extra copies of SMN2 The frequency of small intragenic SMN1 mutations and/or
[23,36,40,41]. It has been shown that 6.9, 73 and 20% of SMA of SMA unlinked to chromosome 5q13 may be higher in cer-
Type I patients had one, two and three copies of SMN2, respec- tain non-European populations, such as black South African
tively. In contrast, 11, 82 and 7.3% of Type II patients and 3.9, patients [53], although data that do not support this hypothesis
51 and 45% of Type III patients, had two, three and four copies also exist [54].
of SMN2, respectively [41].
SMN polymorphisms
SMN1 small intragenic mutations SMN polymorphisms have been reviewed elsewhere [32]. Most
The SMA in many of the approximate 6% of affected individ- polymorphisms are present in the promoter region and intron 6
uals who do not lack both copies of SMN1 is not likely to be [26]. The authors reported five unrelated individuals who have
related to SMN1 [40]. However, in a substantial number of the two alleles that differ in length due to a single thymidine inser-
SMA patients who do not lack both copies of SMN1, small tion in a pre-existing polythymidine tract (8T) in SMN1
intragenic mutations (also referred to as subtle or nondeletion intron 6 (IVS6-24dupT or IVS6-24_IVS6-23insT) [16]. Two
mutations) in SMN1 have been identified (TABLE 1). These were symptomatic (one with Type II SMA and the other with
intragenic mutations provide solid evidence that SMN1 is Type III SMA) and the other three were asymptomatic. All five
indeed the SMA gene, since intragenic SMN1 mutations are of the samples contained the normal 8T SMN1 peak, in addi-
associated with the SMA phenotype regardless of the status of tion to the one-base-larger 9T SMN1 peak, and each peak
NAIP or other genes. Commonly used nomenclature has not corresponded in dosage to one copy of SMN1 [16]. No indi-
been based on the nucleotide position starting from the initia- viduals were found with only the 9T SMN1 peak without the
tion ATG codon, which has led to some confusion. The use of normal 8T SMN1 peak. The biological significance, if any, of
standard nomenclature based on the published sequence [22,37] this single nucleotide insertion remains unknown.
and published nomenclature guidelines [42,43] has been pro-
posed (TABLE 1) [40]. Use of correct nomenclature in the litera- De novo SMN1 mutations
ture is essential since clinical diagnosis and decision making SMN1 has a high rate of de novo deletion mutations. The high
are based on data in the literature regarding a specific muta- rate of de novo mutations in SMN1 likely underlies the high
tion detected in a consultand or affected family member. The carrier frequency for SMA in the general population despite the
three most frequently reported small intragenic mutations are high genetic lethality of the disease. The large inverted repeat at
the Y272C missense mutation (815A>G) [21,32,40,44], the chromosome 5q13, as well as smaller repeat sequences around
768_778dupTGCTGATGCTT frameshift mutation [32,45–48] and the SMN1 and SMN2 loci, likely predispose to unequal crossing
the 399_402delAGAG frameshift mutation [48–50]. over and other recombination events and therefore likely
Wirth and coworkers found that 18 of 501 patients with underlie the high de novo mutation rate of SMN1. De novo
SMA – six of 265 Type I, six of 122 Type II and six of 114 mutations were present in a total of 12 of 494 affected individ-
Type III – with identifiable mutations in both chromosome 5s uals with a homozygous absence of SMN1 [6,55,56]. Among the
had one small intragenic mutation and one deletion mutation 12 patients, there were nine paternal de novo deletion muta-
[40]. Thus, the proportion of disease alleles with small intragenic tions [6,55,56], one paternal de novo conversion mutation [55],
mutations was highest in Type III SMA and lowest in Type I one maternal de novo deletion [55] and one maternal de novo
SMA [40]. Among 525 patients with clinically typical SMA, 24 conversion mutation [40]. Thus, most reported de novo muta-
lacked identifiable mutations in both chromosome 5s and were tions were paternal in origin. The reason why de novo muta-
considered to have SMN1-unrelated disease [40]. Disease and tions of paternal origin are much more frequent than those of
nondisease allele frequencies have a significant impact on risk maternal origin is unknown. Using the method of Wirth and
assessment in SMA genetic testing [1,6]. coworkers [56], paternal and maternal de novo mutation rates are

18 Expert Rev. Mol. Diagn. 4(1), (2004)


Spinal muscular atrophy

Table 1. List of small intragenic SMN1 mutations (modified from [1]).


Standard nomenclature Single amino Other nomenclature Site of Type of Type of Unrelated Ref.
of nucleotide change acid change used in the literature mutation mutation SMA patients

5C>G A2G 38C>G Exon 1 Missense II, III 3 [46]

22_23insA (or 22dupA) Exon 1 Frameshift I or II 1 [114]

43C>T Q15X 78C>T Exon 1 Nonsense I, III 2 [40]

81_81+1insG (or 81dupG) Q27insG Exon 1 Frameshift II 2 [115]

91_92insT (or 91dupT) 124insT Exon 2a Frameshift I 1 [40]

208_209insGTGT 241-242in4 Exon 2b Frameshift III 1 [40]

305G>A W102X Exon 3 Nonsense II, III 2 [52]

399_402delAGAG 430del4 Exon 3 Frameshift I, II, III 11 [48–50]

400G>A E134K 433G>A Exon 3 Missense I 1 [32,47,52]

439_443delGAAGT 425del5, 472del5 Exon 3 Frameshift I 2 [52,116]

509_510delGT 542delGT Exon 4 Frameshift I, II, III 3 [46,91]

558delA 591delA Exon 4 Frameshift II 1 [40]

585_586insT (or 585dupT) 618insT Exon 4 Frameshift I 1 [32,47]

IVS4_IVS6del Intron 4–6 Deletion I 2 [40]

683T>A L228X Exon 5 Nonsense I or II 1 [114]

734C>T P245L 767C>T Exon 6 Missense III 1 [44]

740_741insC (or 740dupC) 773insC Exon 6 Frameshift III 1 [48]

768_778dupTGCTGATGCTT 813ins/dup11, Exon 6 Frameshift I, II 8 [32,


(or 770_780dupCTGATGCTTTG) 800ins11 45–48]

785G>T S262I 818G>T Exon 6 Missense III 2 [23,117]

815A>G Y272C 848A>G Exon 6 Missense I, II, III 9 [21,32,


40,44]

821C>T T274I 854C>T Exon 6 Missense II, III 4 [40,46,


117]

823G>A G275S Exon 6 Missense III 1 [115]

834+2T>G (or IVS6+2T>G) c.867+2T>G Intron 6 Splice site I 1 [48]

IVS6-18_IVS6-12delCCTTTAT c.868-11del7 Intron 6 Splice site I 1 [21,32]

835G>T G279C 868G>T Exon 7 Missense II, III 2 [118]

836G>T G279V 869G>T Exon 7 Missense I 2 [119]

IVS7+4_IVS7+7delAGTC c.922+3del4 Intron 7 Splice site II 1 [21,32]

IVS7+6T>G c.922+6T>G Intron 7 Splice site III 1 [40]

EX8del§ Exon 8 Deletion II, III 2 [120]

§
Exact extent of deletion not determined. One patient revealed homozygous deletion of NAIP exon 5, and the other patient did not reveal homozygous deletion of NAIP exon 5.
SMA: Spinal muscular atrophy.

www.future-drugs.com 19
Ogino & Wilson

calculated as 2.11 × 10-4 and 4.15 × 10-5, respectively. However, a nonessential splicing factor, was found to modify the alterna-
these figures are based on very small numbers of patients who tive splicing of SMN2 and to regulate the inclusion of SMN
had de novo mutations. Further studies are necessary to calcu- exon 7 to produce full-length transcripts from SMN2 [66].
late de novo mutation rates more accurately. The possibility of a However, phenotypic variations between discordant siblings
de novo mutation is highly relevant to genetic risk assessments did not correlate with polymorphic variants or differences in
in some clinical scenarios [1]. expression of Htra2-β1 [67]. Likewise, phenotypic variations
between siblings with identical 5q13 haplotypes did not corre-
SMN1 mosaicism late with mutations or differences in expression of Gemin2
Campbell and coworkers described possible maternal somatic (SIP1) [68]. It remains a possibility that SMA phenotypes are
or germline mosaicism for an SMN1 mutation [57]. Germline modified by genes responsible for other neuromuscular disorders,
mosaicism may result from a de novo mutation if it occurs early including IGHMBP2, HEXA, SCO2 and others [1]. It is also
in embryonic gonadal development. De novo mutations are possible that some of the other components of the SMN and
usually associated with a very low recurrence risk. However, in PRMT5 complexes and other components of the spliceosome
the setting of germline mosaicism, recurrence risk is unpredict- may be modifiers for SMA.
able unless SMN gene dosage analysis is performed on both
gametes and peripheral blood. The authors have encountered a Animal models & orthologs of SMN
possible case of germline mosaicism for SMN1 in which the SMN gene duplication occurred more than 5 million years ago,
SMN1 signal from a sperm sample was approximately 80% of before the separation of human and chimpanzee lineages; how-
that from the same individual’s peripheral blood, demonstrated ever, the SMN2 gene sequence per se appeared for the first time
repeatedly by SMN gene dosage analysis [58,59]. only in Homo sapiens, whereas the chimpanzee has duplicated
SMN but no SMN2 [69]. A knockout of murine SMN resulted
Candidate modifying genes for SMA in massive apoptosis and embryonic lethality, indicating that
SMN2 is a well-established, disease-modifying gene for SMA the SMN protein is essential for cell survival [70]. To construct
and has been shown to decrease the severity of SMA in a dose- mouse models of SMA, it was necessary either to retain some
dependent manner [23,40,41,58]. The copy number of SMN2 also amount of SMN protein in the mouse embryo by introducing
correlates with longer survival of individuals affected with the human SMN2 gene [71–73], or to delete murine SMN
Type I SMA [41]. However, the SMN2 copy number in Type I, exon 7 in neurons using cre-lox-mediated recombination [74].
II and III SMA overlaps. In addition, rare families have been Human SMN2 rescued mice from embryonic lethality and
reported in which different degrees of disease severity were alleviated symptoms resembling human SMA in a dose-
present in affected individuals with identical SMN1 and SMN2 dependent manner [71,73]. Deletion of murine SMN exon 7 in
copy numbers [40,41]. Thus, although SMN2 copy number skeletal muscle by cre-lox-mediated recombination led to
apparently explains much of the phenotypic variation in severe muscular dystrophy, suggesting that skeletal muscle may
SMA, there must be other disease-modifying factors. Indeed, also be a primary site of degeneration in SMA [75]. Other animal
a small intragenic SMN2 mutation (859G>C; G287R) [41,60] models of human SMA include Drosophila with a point muta-
and other SMN2 mutations or polymorphisms may underlie tion in SMN [76], and zebrafish with SMN protein levels
the discrepancy between SMN2 copy number and SMA type. decreased by the use of an antisense morpholino oligonucleo-
The neuronal apoptosis inhibitory protein (NAIP) gene was a tide [77]. These animal models will be useful for studies on the
candidate disease gene for SMA because it was found to be pathogenesis of SMA, as well as for the development and testing
deleted in approximately two-thirds of Type I SMA chromo- of novel therapeutics.
somes and because of its homology with baculoviral apoptosis
inhibitory proteins [61]. With the confirmation of SMN1 as the Molecular diagnostics
disease gene for SMA [21], NAIP has been hypothesized to be a Linkage analysis
disease-modifying gene. Homozygous deletions of NAIP are Localization of the SMA critical region allowed for linkage
found more often in Type I SMA than in the other two types analysis, the first genetic test for SMA. From centromeric to
[60,62–64]. However, homozygous deletions of NAIP may simply telomeric, informative linkage markers that have been
reflect larger chromosomal deletions involving both SMN1 and reported are as follows: D5S679, D5S680, D5S125, D5S681,
SMN2. Further studies are necessary to confirm or rule out a D5S435, D5S629, D5S823, D5S1556/D5F150 (intra-
role of NAIP in the pathogenesis of SMA. H4F5 (SERF1) is genic/SMN1 promoter region), D5S149 (intragenic/SMN1
closer to SMN1 on chromosome 5 than any other known gene. promoter region), D5S557, D5S610, D5S351, 5´-MAP1B,
By microsatellite analysis, homozygous deletions of H4F5 were 3´-MAP1B, D5S112, D5S127 and D5S539 [78]. Even after
found in approximately 90% of cases of Type I SMA [65]. identification of the disease gene [21], linkage analysis remains
However, as with NAIP, homozygous deletions of H4F5 may an important tool in certain clinical scenarios [16,58]. Multi-
simply reflect larger chromosomal deletions involving both copy markers, such as C212 and C272, have also been used in
SMN1 and SMN2. Further studies are necessary to confirm or linkage analysis [33,79,80], although the interpretation of results
rule out a role of H4F5 in the pathogenesis of SMA. Htra2-β1, requires considerable skill.

20 Expert Rev. Mol. Diagn. 4(1), (2004)


Spinal muscular atrophy

PCR-single-strand conformation polymorphism Rapid PCR-denaturing high performance liquid chromatog-


PCR-single-strand conformation polymorphism (SSCP) analysis raphy (DHPLC) methods to detect the homozygous absence of
has been used to detect the homozygous absence of SMN1 SMN1 exon 7 have been described [85,86].
exon 7 and exon 8 [21,81]. However, although PCR-SSCP analysis
can detect small intragenic mutations in an amplified segment, Quantitative SMN gene dosage analyses
inconsequential polymorphisms are also detected. Using PCR- Since qualitative PCR-RFLP cannot distinguish carriers with
SSCP analysis, Wang and coworkers found that 4% of asymp- one copy of SMN1 from normal individuals with two copies of
tomatic family members of patients with SMA had polymor- SMN1, a quantitative approach (i.e., SMN gene dosage analysis)
phisms in SMN1 exon 7 that mimicked the homozygous is necessary to identify SMA carriers. The rationale for a quan-
absence of SMN1 [82]. With the development of PCR-restriction titative competitive PCR method using a genomic reference
fragment length polymorphism (RFLP) assays [83], PCR-SSCP sequence, along with internal control sequences for both the
has largely been replaced in clinical laboratories. PCR-SSCP target and the reference sequences, has been described [59].
may still be used to screen for small intragenic mutations in McAndrew and coworkers developed the first highly precise
affected individuals who do not lack both copies of SMN1. assay to determine copy numbers of SMN1 and SMN2 using
this method [23]. The assay utilizes quantitative competitive
PCR-RFLP PCR-RFLP analysis, with CFTR exon 4 as a two-copy genomic
The most commonly used method to genetically confirm the reference. To correct for variation in amplification efficiency,
diagnosis of SMA is a qualitative PCR-RFLP assay (also known the assay includes SMN and CFTR internal standard sequences.
as the SMN1 deletion assay) to detect the homozygous absence Each internal standard has the same primer binding sites as its
of SMN1 [83]. The PCR-RFLP assay takes advantage of the base genomic counterpart and a small internal deletion, which
differences in exons 7 and 8 to distinguish SMN1 from SMN2. allows the internal standard PCR products and the genomic
The restriction endonuclease DraI digests only the SMN2 exon 7 PCR products to be separated and quantified independently
PCR products because of a restriction site generated by a mis- [23]. The copy number of SMN1 per cell (or more precisely per
matched reverse primer [83]. Another restriction enzyme that diploid genome) is determined by coamplification of SMN1,
can be utilized is Hinf I [40]. In the Hinf I method, internal con- SMN2, the SMN internal standard, CFTR and the CFTR inter-
trol restriction sites are introduced in both the SMN1 and the nal standard; DraI digestion of SMN2; and quantification of
SMN2 PCR products to ensure complete Hinf I digestion [40]. each PCR product [23,58].
Wirth reviewed the frequencies of SMA patients without a A variety of highly precise SMN gene dosage analyses have
homozygous absence of SMN1 exon 8 among SMA patients with since been developed, including a nonradioactive modification
a homozygous absence of SMN1 exon 7 [32]. The frequencies using fluorescence-labeled primers and an automated sequencer
were 21 of 400 (5.3%) for Type I, 19 of 241 (7.9%) for Type II, [40,58,87], combined quantitative competitive PCR with quanti-
26 of 185 (14%) for Type III and 79 of 1054 (7.5%) for all tative primer extension [24] and real-time quantitative PCR
types combined. These data are consistent with the tendency of [41,88]. Various features of quantitative competitive PCR-RFLP
patients with Type III SMA to have more copies of SMN2 and real-time quantitative PCR have been compared [89].
exon 7 juxtaposed to SMN1 exon 8 due to gene conversion. Heteroduplex formation may affect quantitative PCR-RFLP
Combining the data of Wirth with the data of Mailman and analyses [59]. Heteroduplexes between SMN1 and SMN2 PCR
coworkers (29 of 261 for all types combined) [84] and Ogino products cannot be digested by DraI, increasing the SMN1 signal
and coworkers (16 of 126 for all types combined) [16], a total of and decreasing the SMN2 signal [59]. In addition to heteroduplex
124 of 1441 (8.6%) patients with SMA and a homozygous formation, an unexpected and reproducible PCR bias between
absence of SMN1 exon 7 had at least one copy of SMN1 exon 8. SMN1 and SMN2 sequences has been found (i.e., amplifica-
In clinical practice, patients referred for SMA genetic testing tion efficiency for SMN2 was only approximately 80% that of
do not always have clinically typical features of SMA. In one SMN1) [90]. One should be aware of heteroduplex formation
study, 349 of 610 (57%) patients tested did not have a and/or PCR bias in any quantitative competitive PCR.
homozygous absence of SMN1 exon 7 [84]. In another study, a SMN gene dosage analysis can be used for detection of the
total of 217 of 430 (50%) patients did not have a homozygous heterozygous absence of SMN1 in patients with SMA who do
absence of SMN1 exon 7 [16]. Among those patients in which not lack both copies of SMN1 [40,44,91]. However, SMN gene
the SMA type was specified, this comprised 34 of 94 Type I dosage analysis for this purpose should be used with caution
patients (36%), five of 40 Type II patients (12%) and 42 of 62 and should be limited to patients for whom SMN1-related
Type III patients (68%) [16]. Therefore, the clinical diagnosis of SMA is strongly suspected (i.e., clinically typical cases). This is
SMA can be problematic and genetic testing is often ordered to because the high frequency of SMA carriers with one copy of
exclude SMN1-related SMA because of atypical clinical fea- SMN1 in the general population would otherwise lead to an
tures, or to confirm SMN1-related SMA. The predictive value unacceptably low positive predictive value [6,16].
of a presumptive clinical diagnosis of Type II SMA was consid- It has been shown that occasional individuals have three or
erably higher (35 of 40) than that of the other SMA types [16], four copies of SMN1, implying the presence of occasional
probably due to the fewer conditions that mimic Type II SMA. chromosome 5s with two copies of SMN1 [6,23,40,41]. These

www.future-drugs.com 21
Ogino & Wilson

chromosomes can mask a deletion or conversion mutation of Preimplantation genetic testing in general has been reviewed
SMN1 on the other chromosome 5, causing false-negative [96–98].PCR-RFLP-based preimplantation genetic testing to
results in SMN gene dosage analysis (i.e., carriers with two detect the homozygous absence of SMN1 exon 7 has also been
SMN1 copies on one chromosome 5 and a deletion/conversion described [99–103].
mutation on the other chromosome 5 cannot be distinguished
by dosage analysis alone from normal individuals). Two SMN1 Monosomal analysis
copies on a single chromosome may be due to gene conversion Yan and coworkers developed a monosomal analysis method
from SMN2 to SMN1, or alternatively due to remaining dupli- involving the separation of single human chromosomes of
cated SMN1 genes without conversion to SMN2 [92]. The dis- homologous pairs by fusing human cells with mouse cells and
tinction between two and three copies of SMN1 is sometimes culturing with selection [104]. This method is useful for analyzing
important in families with relatives suspected to have the 2 + 0 oncogenic mutations, which may be masked by the presence of
SMN1 genotype [1]. wild type alleles. It can also be applied to the detection of muta-
tions in hereditary diseases. Mailman and coworkers used this
SMN1 small intragenic mutation analysis method to identify an SMN1 deletion carrier who had two
Analysis for the detection of small intragenic mutations is not copies of SMN1 by dosage analysis [105]. Monosomal analysis
currently offered routinely in the clinical laboratory setting obviates the need to perform extensive linkage and dosage
because it is typically performed by DNA sequencing and is analyses to distinguish carriers with two copies of SMN1 (the
therefore too labor intensive. A multiplex, allele-specific PCR 2 + 0 SMN1 genotype) from noncarriers (with the normal 1 + 1
assay to detect the seven most common small intragenic muta- genotype) who passed a de novo deletion mutation to an
tions has been described [84]. For more information regarding affected child [58].
assays to detect small intragenic mutations, the laboratory of
H Scheffer (Groningen, The Netherlands), or the laboratory of Genetic counseling & risk assessment
B Wirth (Bonn, Germany) may be contacted [78]. Assays to Both genetic counseling and risk assessment are integral
detect small intragenic mutations in SMN1 may become a part components of genetic testing and are particularly impor-
of routine practice in the future if sensitive and cost-effective tant for SMA families because of the high carrier frequency
methods to detect such mutations are developed. (1 in 50) and the complexity of SMA genetics [1]: small
intragenic mutations are undetectable by the standard PCR-
Prenatal & preimplantation genetic testing RFLP and SMN gene dosage analyses; the presence of two
Prenatal testing to detect the homozygous absence of SMN1 normal copies of SMN1 on one chromosome 5 can mask a
can be performed on chorionic villous sampling (CVS) speci- deletion mutation on the other chromosome 5 in SMN gene
mens or amniotic fluids. To avoid false-negatives due to mater- dosage analyses; and the paternal de novo mutation rate is
nal contamination, the amount of maternal DNA relative to relatively high.
fetal DNA should be determined, for example by quantitative It should be noted that genetic risk assessments are esti-
PCR of polymorphic microsatellite or SNP markers. The mates. They are based on published data from a finite number
authors have used a method that was developed to quantify of cases in specific populations. It should also be noted that
engraftment after bone marrow transplantation [93]. Although additional family information, especially additional genetic
CVS specimens are more often contaminated with maternal information, often dramatically improves the accuracy of
DNA than amniotic fluid specimens, such contamination is genetic risk assessments. Thus, genetic risk assessment is often
rarely a problem if CVS specimens are handled properly. In an ongoing process and genetic testing on additional family
each SMN PCR-RFLP assay, a 10%-contamination control is members should be performed whenever indicated by the
also run that contains a mixture of DNA lacking SMN1 (90%) initial results [1].
and DNA that contains SMN1 (10%) [1]. If the SMN1 band of
a fetal sample with less than 5% maternal contamination is SMN1 allele & genotype frequencies
much brighter than that of the 10%-contamination control, For the purpose of SMA risk assessment, normal and disease
the SMN1 band of the fetal sample is much more likely to SMN1 allele frequencies in the general population and for
derive from the fetal DNA than from contaminated maternal each SMA type have been estimated [1]. Most disease SMN1
DNA. However, interpretation of such results is still carried out alleles are deletions/conversions of at least exon 7 of SMN1
with caution since the standard PCR-RFLP assay is at most (0-copy alleles). These alleles are usually detectable by dosage
only semiquantitative. analysis. A normal chromosome 5 usually has one copy of
Prenatal testing can be performed on circulating fetal cells SMN1 (1-copy allele). However, two copies of SMN1 are often
in maternal blood [94]. However, the issue of persistent fetal present on the same chromosome 5, for which the term 2-copy
cells from the prior pregnancy is a cause for concern, although allele is used. Disease alleles with small intragenic mutations in
a recent study found no evidence that there is interference of SMN1 are referred to as 1D alleles (standing for 1-copy-disease);
persistent fetal cells from prior pregnancies with prenatal 1D alleles are indistinguishable from normal 1-copy alleles by
diagnosis [95]. dosage analysis.

22 Expert Rev. Mol. Diagn. 4(1), (2004)


Spinal muscular atrophy

Complete, updated haplotype and genotype frequencies in that upregulate full-length SMN protein is underway [108].
the general population and for each SMA type are shown in Potential gene therapy strategies for SMA are also under inves-
TABLE 2. The frequencies for a given SMA type should be used tigation; for example, adenovirus-mediated gene delivery [112]
only when the substantial risk of a 1D haplotype is present due and cardiotrophin-1 gene transfer [113]. Further elucidation of
to an index patient who is affected with clinically typical SMA the molecular genetics and pathogenesis of SMA should facili-
of the given type [1,6]. tate development of effective treatments and potentially a cure
In-depth and detailed methods for risk calculations have for SMA.
been illustrated utilizing a variety of examples [1].
Conclusion
Potential therapeutic approaches Since the discovery of the SMA disease gene, SMN1, consid-
There is currently no effective treatment or cure for SMA. erable progress in our understanding of the biochemical basis
However, potential therapies are an active area of investiga- for SMA and in genetic testing for SMA have been made. At
tion and new therapeutic approaches will likely emerge in the this time, comprehensive SMA testing comprising qualitative
near future. These include: to increase full-length mRNA PCR-RFLP, SMN gene dosage analysis and linkage analysis, in
from SMN2 by altering the splicing pattern of SMN2 exon 7 combination with appropriate genetic counseling and risk
in vivo, either through the activation of transacting factors or assessment, offers the most complete evaluation of SMA
by using antisense oligonucleotides [106]; to activate the over- patients and their families. The accuracy of risk assessments for
all level of SMN transcription; to stabilize the SMN protein; SMA will increase as more data on the genetics of SMA emerge.
to repair degenerated motor neurons; to replace degenerated In addition, new technologies, such as monosomal analysis,
motor neurons by stem cell therapy [107]; and to target modi- may be widely available in the near future.
fying factors other than SMN2 [108]. To date, aclarubicin
[109], sodium butyrate [110] and valproic acid [111] have been Expert opinion
shown to increase the level of full-length transcripts from Published guidelines state that genetic testing laboratories
SMN2 and thus are promising potential therapeutic agents for should perform SMN gene dosage analysis for SMA carrier
SMA. High-throughput drug screening to identify compounds testing on SMA family members only when the index patient

Table 2. Updated SMN1 allele and genotype frequencies.


SMN1 allele Designation General Type I§ Type II§ Type III§
population
0-copy (disease) a 9.88 × 10-3 9.95 × 10-3 9.86 × 10-3 9.76 × 10-3
1-copy (normal) b 9.50 × 10-1 9.50 × 10-1 9.50 × 10-1 9.50 × 10-1
2-copy (normal) c 4.04 × 10-2 4.04 × 10-2 4.04 × 10-2 4.04 × 10-2
1D-copy (disease) d 1.81 × 10-4 1.14 × 10-4 2.08 × 10-4 3.06 × 10-4
Status Genotype§§ Copy Designation General Type I Type II Type III
number population
Noncarrier 2+2 4 c2 1.63 × 10-3 1.63 × 10-3 1.63 × 10-3 1.63 × 10-3
2+1 3 2bc 7.67 × 10-2 7.67 × 10-2 7.67 × 10-2 7.67 × 10-2
1+1 2 b2 9.02 × 10-1 9.02 × 10-1 9.02 × 10-1 9.02 × 10-1

Carrier 2 + 1D 3 2cd 1.46 × 10-5 9.20 × 10-6 1.68 × 10-5 2.47 × 10-5
2+0 2 2ac 7.98 × 10-4 8.04 × 10-4 7.96 × 10-4 7.88 × 10-4
1 + 1D 2 2bd 3.43 × 10-4 2.16 × 10-4 3.95 × 10-4 5.82 × 10-4
1+0 1 2ab 1.88 × 10-2 1.89 × 10-2 1.87 × 10-2 1.85 × 10-2

Affected 1D + 1D 2 d2 3.27 × 10-8 1.30 × 10-8 4.32 × 10-8 9.38 × 10-8


1D + 0 1 2ad 3.57 × 10-6 2.27 × 10-6 4.10 × 10-6 5.98 × 10-6
0+0 0 a2 9.77 × 10-5 9.90 × 10-5 9.72 × 10-5 9.52 × 10-5
§
Relative allele frequencies for each spinal muscular atrophy (SMA) type, which should be used when the risk of a 1D allele is present due to an index case affected with
clinically typical SMA of a given type.
§§
Genotype is indicated by (SMN1 allele on one chromosome 5) + (SMN1 allele on the other chromosome 5).

www.future-drugs.com 23
Ogino & Wilson

has been found to lack both copies of SMN1 [78]. When the genotype (with a low recurrence risk) from the 2 + 0 genotype
index patient with clinically typical SMA is unavailable for (with a high recurrence risk of 1 in 4) [58]. The monosomal
testing, SMN gene dosage analysis can be performed on the analysis method in combination with dosage analysis, though
parents to confirm the presence of deletion/conversion muta- less extensively evaluated as yet, can also be used to distinguish
tions. In the authors experience, however, a deceased index the 1 + 1 genotype from the 2 + 0 genotype [105].
patient, as well as his or her parents, are occasionally unavailable
for genetic testing when an unaffected relative is referred for Five-year view
genetic testing. After careful genetic counseling and informed Elucidation of SMA pathogenesis and the precise function of
consent, SMN gene dosage analysis can be offered in such the SMN protein are highly active areas of investigation. High-
cases. One reason is that unaffected relatives are more than throughput drug screening for SMA is in progress [108]. There is
occasionally found to have one copy of SMN1, which is of every reason to believe that in 5 years, our understanding of
course readily interpretable. Had testing of such individuals SMA pathogenesis will have advanced considerably and
been denied, important information for family planning would improved therapies may be available. Data on SMA genetics
have been unavailable. Another reason is that when the index will also accumulate, serving to improve genetic risk assessment
patient and his or her parents are unavailable for testing, other and our understanding of the molecular evolution of the SMN1
relatives of the patient are sometimes available and are found to gene and its homolog SMN2. Disease modifying genes and
have only one copy of SMN1. their polymorphisms will likely be further elucidated. Large
As previously shown [1], accurate risk assessment in such population registries and databases for population-based studies
cases is possible even when negative results are obtained. One should aid in testing various hypotheses concerning modifying
of the aims of this previous study was to provide versatile factors for SMA.
methods for calculating genetic risk in complicated clinical Diagnostic testing techniques will likely continue to improve,
scenarios, even when genetic information on the index case or particularly in terms of cost effectiveness. Monosomal analysis
the parents of the index case is limited. SMN gene dosage is a promising technology that obviates the necessity for extensive
analysis and carrier risk assessment are appropriate provided dosage analysis and linkage analysis to distinguish the 2 + 0
that all available information is taken into account and pro- genotype from the 1 + 1 genotype. Assays to detect various
vided that the limitations are explained clearly to those being small intragenic SMN1 mutations may improve and be per-
tested. One of the limitations is the dependence of the risk formed routinely for patients who do not lack both copies of
assessment on whether the disease in the affected family mem- SMN1. As SMA modifiers are identified, molecular testing for
ber is SMN1-related. In general, of course, the more individuals their presence will likely emerge. Lastly, genetic testing for drug
tested, and the more genetic information that is available, the sensitivity and resistance may be needed in the future as new
more accurate the genetic risk assessment becomes. This prin- pharmaceutical agents become available for SMA patients.
ciple holds true even in the absence of genetic information on
the index case or the parents. Acknowledgements
With regard to SMA genetic testing for affected individuals We thank the following individuals for their support of various
who do not lack both copies of SMN1, SMN gene dosage aspects of our work: DGB Leonard, V Van Deerlin, H Rennert,
analysis for the detection of heterozygous SMN1 deletion/ TW Prior, PE McAndrew, C Turino, T Smith, K-L Chen, H-J
conversion should be used with caution and should be Dong, S Gao, WJ Ewens, M Loda and CS Fuchs.
restricted to patients with a high index of clinical suspicion, in
order to ensure a high positive predictive value. This is because Information resources
the frequency of 1-copy carriers in the general population • GeneTests
(~2%) is comparable with the fraction of SMA patients who www.genetests.org
have a small intragenic mutation in one SMN1 allele (~3.5% (Viewed December 2003)
for all SMA types [40]) [1]. Provided that this limitation is fully • GeneCards for SMN1
understood, SMN gene dosage can be offered in such circum- http://bioinfo.weizmann.ac.il/cards-bin/carddisp?SMN1
stances. Although not a routine practice at this time, the SMN1 (Viewed December 2003)
alleles without gene deletion/conversion mutations can be
• NiceProt View of Swiss-Prot: SMN, accession number Q16637
interrogated for small intragenic mutations.
http://us.expasy.org/cgi-bin/niceprot.pl?Q16637
Linkage analysis in combination with SMN gene dosage
(Viewed December 2003)
analysis is recommended when small intragenic mutations are
present in families, or when two copies of SMN1 are found in • Spinal Muscular Atrophy Net
the parent of an affected child who lacks both copies of SMN1 www.affari.com/smanet
[16,58]. In the former case, linkage analysis can be used to track (Viewed December 2003)
the mutation through the family. In the latter case, linkage • Families of Spinal Muscular Atrophy (FSMA)
analysis can detect crossover events associated with de novo www.fsma.org
SMN1 deletions, helping to distinguish the 1 + 1 SMN1 (Viewed December 2003)

24 Expert Rev. Mol. Diagn. 4(1), (2004)


Spinal muscular atrophy

Key issues

• Qualitative PCR-restriction fragment length polymorphism is used to detect the homozygous absence of SMN1 exon 7, which is
observed in approximately 94% of clinically typical spinal muscular atrophy (SMA) patients. It is a reasonably sensitive assay, but
cannot detect patients with small intragenic SMN1 mutations.
• Quantitative SMN gene dosage analysis can determine the copy number of SMN1 and SMN2 and is used for SMA carrier testing.
However, it cannot detect a carrier with two SMN1 copies on one chromosome 5 and a deletion/conversion mutation on the other
chromosome 5 (the 2 + 0 SMN1 genotype) or a carrier with a small intragenic mutation. SMN gene dosage analysis can also detect
SMA patients who do not lack both copies of SMN1 exon 7, although it should be used with caution because of a potentially low
positive predictive value if the index of clinical suspicion is low.
• SMN1 has an exceptionally high paternal de novo mutation rate (ν = 2.11 × 10-4).
• Linkage analysis or monosomal analysis can be used in combination with SMN gene dosage analysis in certain clinical scenarios to
distinguish a carrier with the 2 + 0 genotype from a 1 + 1 normal individual who has passed a de novo mutation to the index case.
• Due to the complexity of SMA genetics and the high carrier frequency (1 in 50), genetic counseling and risk assessment are essential
components of SMA genetic testing. Accurate genetic risk assessment is possible if all available family and genetic information is
taken into account. Testing of additional family members may dramatically improve the accuracy of genetic risk calculations;
therefore, testing of additional family members should be suggested and offered when appropriate.

References 7 Brahe C, Servidei S, Zappata S et al. degeneration associated with deletion of the
Papers of special note have been highlighted as: Genetic homogeneity between childhood- SMNT gene. Neurology 49(3), 848–851
• of interest onset and adult-onset autosomal recessive (1997).
spinal muscular atrophy. Lancet 346(8977), 16 Ogino S, Leonard DG, Rennert H,
•• of considerable interest
741–742 (1995). Wilson RB. Spinal muscular atrophy
1 Ogino S, Wilson RB. Genetic testing and
8 Zerres K, Rudnik-Schoneborn S, Forkert R, genetic testing experience at an academic
risk assessment for spinal muscular atrophy Wirth B. Genetic basis of adult-onset spinal medical center. J. Mol. Diagn. 4(1), 53–58
(SMA). Hum. Genet. 111(6), 477–500 muscular atrophy. Lancet 346(8983), 1162 (2002).
(2002). (1995). 17 Poets C, Heyer R, von der Hardt H,
•• First comprehensive review
demonstrating how to calculate genetic 9 Zerres K, Wirth B, Rudnik-Schoeneborn S. Walter GF. Acute respiratory insufficiency
risks in various spinal muscular atrophy Spinal muscular atrophy – clinical and as the initial clinical manifestations of
(SMA) clinical settings. genetic correlation. Neuromuscul. Disord. 7, spinal muscular atrophy. Monatsschr.
202–207 (1997). Kinderheilkd. 138(3), 157–159 (1990).
2 Munsat TL, Davies KE. International SMA
consortium meeting. Neuromuscul. Disord. 10 Rudnik-Schoneborn S, Forkert R, Hahnen E, 18 Sivan Y, Galvis A. Early diaphragmatic
2(5–6), 423–428 (1992). Wirth B, Zerres K. Clinical spectrum and paralysis in infants with genetic disorders.
diagnostic criteria of infantile spinal Clin. Pediatr. 29(3), 169–171 (1990).
3 Zerres K, Davies KE. 59th ENMC
muscular atrophy: further delineation on 19 Mellins RB, Hays AP, Gold AP, Berdon WE,
International Workshop: spinal muscular
the basis of SMN gene deletion findings. Bowdler JD. Respiratory distress as the
atrophies: recent progress and revised
Neuropediatrics 27(1), 8–15 (1996). initial manifestation of Werdnig–Hoffmann
diagnostic criteria 17–19 April 1998,
Soestduinen, The Netherlands. 11 Goldberg MJ. Syndrome of orthopaedic disease. Pediatrics 53(1), 33–40 (1974).
Neuromuscul. Disord. 9(4), 272–278 importance. In: Lovell and Winter’s Pediatric 20 McWilliam RC, Gardner–Medwin D,
(1999). Orthopaedics. Morrissy RT, Weinstein SL Doyle D, Stephenson JB. Diaphragmatic
(Eds), Lippincott-Raven Publishers, PA, paralysis due to spinal muscular atrophy.
4 Rudnik-Schoneborn S, Goebel HH,
USA, 262–271 (1996). An unrecognised cause of respiratory failure
Schlote W et al. Classical infantile spinal
muscular atrophy with SMN deficiency 12 Gordon N. Arthrogryposis multiplex in infancy? Arch. Dis. Child. 60(2),
causes sensory neuronopathy. Neurology congenita. Brain Dev. 20(7), 507–511 145–149 (1985).
60(6), 983–987 (2003). (1998). 21 Lefebvre S, Burglen L, Reboullet S et al.
5 Zerres K, Rudnik-Schoneborn S. 13 Talbot K, Davies KE. Spinal muscular Identification and characterization of a
Natural history in proximal spinal muscular atrophy. Semin. Neurol. 21(2), 189–197 spinal muscular atrophy-determining gene.
atrophy. Clinical analysis of 445 patients (2001). Cell 80(1), 155–165 (1995).
and suggestions for a modification of 14 Burglen L, Amiel J, Viollet L et al. •• Describes the identification of the disease
existing classifications. Arch. Neurol. 52(5), Survival motor neurone gene deletion in gene for SMA. A complete contig of the
518–523 (1995). the arthrogryposis multiplex disease gene region on chromosome 5 is
congenita–spinal muscular atrophy provided.
6 Ogino S, Leonard DG, Rennert H,
Ewens WJ, Wilson RB. Genetic risk association. J. Clin. Invest. 98(5), 22 Burglen L, Lefebvre S, Clermont O et al.
assessment in carrier testing for spinal 1130–1132 (1996). Structure and organization of the human
muscular atrophy. Am. J. Med. Genet. 15 Bingham PM, Shen N, Rennert H et al. survival motor neurone (SMN) gene.
110(4), 301–307 (2002). Arthrogryposis due to infantile neuronal Genomics 32(3), 479–482 (1996).

www.future-drugs.com 25
Ogino & Wilson

23 McAndrew PE, Parsons DW, Simard LR 32 Wirth B. An update of the mutation reliable carrier testing and prediction of
et al. Identification of proximal spinal spectrum of the survival motor neurone severity of spinal muscular atrophy. Am. J.
muscular atrophy carriers and patients by gene (SMN1) in autosomal recessive spinal Hum. Genet. 70, 358–368 (2002).
analysis of SMNT and SMNC gene copy muscular atrophy (SMA). Hum. Mutat. • First description of methods to quantify
number. Am. J. Hum. Genet. 60(6), 15(3), 228–237 (2000). the copy numbers of SMN1 and SMN2
1411–1422 (1997). • First comprehensive review of SMN1 using real-time PCR.
•• Describes the first robust method to mutations. 42 den Dunnen JT, Antonarakis SE.
quantify the copy numbers of SMN1 and 33 Wirth B, Hahnen E, Morgan K et al. Nomenclature for the description of human
SMN2 using competitive quantitative PCR. Allelic association and deletions in autosomal sequence variations. Hum. Genet. 109(1),
24 Gerard B, Ginet N, Matthijs G et al. recessive proximal spinal muscular atrophy: 121–124 (2001).
Genotype determination at the survival association of marker genotype with disease 43 den Dunnen JT, Antonarakis SE.
motor neurone locus in a normal severity and candidate cDNAs. Hum. Mol. Mutation nomenclature extensions and
population and SMA carriers using Genet. 4(8), 1273–1284 (1995). suggestions to describe complex mutations:
competitive PCR and primer extension. 34 Burlet P, Burglen L, Clermont O et al. a discussion. Hum. Mutat. 15(1), 7–12
Hum. Mutat. 16(3), 253–263 (2000). Large scale deletions of the 5q13 region are (2000).
25 Lorson CL, Hahnen E, Androphy EJ, specific to Werdnig–Hoffmann disease. 44 Rochette CF, Surh LC, Ray PN et al.
Wirth B. A single nucleotide in the SMN J. Med. Genet. 33(4), 281–283 (1996). Molecular diagnosis of non-deletion SMA
gene regulates splicing and is responsible for 35 Simard LR, Rochette C, Semionov A, patients using quantitative PCR of SMN
spinal muscular atrophy. Proc. Natl Acad. Morgan K, Vanasse M. SMN(T) and NAIP exon 7. Neurogenetics 1(2), 141–147
Sci. 96, 6307–6311 (1999). mutations in Canadian families with spinal (1997).
• Demonstrated that the translationally silent muscular atrophy (SMA): 45 Parsons DW, McAndrew PE, Monani UR
single nucleotide polymorphism at position genotype/phenotype correlations with et al. An 11 base pair duplication in exon 6
840 in SMN2 exon 7 is responsible for the disease severity. Am. J. Med. Genet. 72(1), of the SMN gene produces a Type I spinal
inefficient splicing of the SMN2 transcript. 51–58 (1997). muscular atrophy (SMA) phenotype:
26 Monani UR, Lorson CL, Parsons DW et al. 36 Burghes AH. When is a deletion not a further evidence for SMN as the primary
A single nucleotide difference that alters deletion? When it is converted. Am. J. SMA-determining gene. Hum. Mol. Genet.
splicing patterns distinguishes the SMA Hum. Genet. 61(1), 9–15 (1997). 5(11), 1727–1732 (1996).
gene SMN1 from the copy gene SMN2.
37 Hahnen E, Schonling J, Rudnik- 46 Parsons DW, McAndrew PE, Iannaccone ST
Hum. Mol. Genet. 8(7), 1177–1183 (1999).
Schoneborn S, Zerres K, Wirth B. et al. Intragenic telSMN mutations:
• Demonstrated that the translationally silent
Hybrid survival motor neurone genes in frequency, distribution, evidence of a
single nucleotide polymorphism at position
patients with autosomal recessive spinal founder effect and modification of the
840 in SMN2 exon 7 is responsible for the
muscular atrophy: new insights into spinal muscular atrophy phenotype by
inefficient splicing of the SMN2 transcript.
molecular mechanisms responsible for the cenSMN copy number. Am. J. Hum. Genet.
27 Kashima T, Manley JL. A negative element disease. Am. J. Hum. Genet. 59(5), 63(6), 1712–1723 (1998).
in SMN2 exon 7 inhibits splicing in spinal 1057–1065 (1996). 47 Clermont O, Burlet P, Cruaud C et al.
muscular atrophy. Nature Genet. 34(4),
38 Cusco I, Barcelo MJ, del Rio E et al. Mutation analysis of the SMN gene in
460–463 (2003).
Characterisation of SMN hybrid genes in undeleted SMA patients. Annual meeting of
• Demonstrated that the translationally
Spanish SMA patients: de novo, homozygous the American Society of Human Genetics.
silent single nucleotide polymorphism at
and compound heterozygous cases. Am. J. Hum. Genet. 61, A329 (1997).
position 840 in SMN2 exon 7 activates an
Hum. Genet. 108(3), 222–229 (2001). 48 Martin Y, Valero A, del Castillo E, Pascual SI,
exonic splice silencer.
39 Campbell L, Potter A, Ignatius J, Hernandez-Chico C. Genetic study of
28 Kerr DA, Nery JP, Traystman RJ,
Dubowitz V, Davies K. Genomic variation SMA patients without homozygous SMN1
Chau BN, Hardwick JM. Survival motor
and gene conversion in spinal muscular deletions: identification of compound
neurone protein modulates neuron-specific
atrophy: implications for disease process heterozygotes and characterisation of novel
apoptosis. Proc. Natl Acad. Sci. USA
and clinical phenotype. Am. J. Hum. Genet. intragenic SMN1 mutations. Hum. Genet.
97(24), 13312–13317 (2000).
61(1), 40–50 (1997). 110(3), 257–263 (2002).
29 Frugier T, Nicole S, Cifuentes-Diaz C,
40 Wirth B, Herz M, Wetter A et al. 49 Cusco I, Lopez E, Soler-Botija C et al.
Melki J. The molecular bases of spinal
Quantitative analysis of survival motor A genetic and phenotypic analysis in Spanish
muscular atrophy. Curr. Opin. Genet. Dev.
neurone copies: identification of subtle spinal muscular atrophy patients with
12(3), 294–298 (2002).
SMN1 mutations in patients with spinal c.399_402del AGAG, the most frequently
• Excellent overview of SMA.
muscular atrophy, genotype–phenotype found subtle mutation in the SMN1 gene.
30 Paushkin S, Gubitz AK, Massenet S, correlation and implications for genetic Hum. Mutat. 22, 136–143 (2003).
Dreyfuss G. The SMN complex, an counseling. Am. J. Hum. Genet. 64, 50 Bussaglia E, Clermont O, Tizzano E et al.
assemblyosome of ribonucleoproteins. 1340–1356 (1999). A frame-shift deletion in the survival motor
Curr. Opin. Cell Biol. 14(3), 305–312 (2002). •• Only comprehensive study investigating neurone gene in Spanish spinal muscular
• Excellent review of the function of SMN. the frequency of SMN1 small intragenic atrophy patients. Nature Genet. 11(3),
31 Meister G, Eggert C, Fischer U. mutations for each SMA type. 335–337 (1995).
SMN-mediated assembly of RNPs: a 41 Feldkotter M, Schwarzer V, Wirth R, 51 Lorson CL, Strasswimmer J, Yao JM et al.
complex story. Trends Cell Biol. 12(10), Wienker TF, Wirth B. Quantitative SMN oligomerization defect correlates with
472–478 (2002). analyses of SMN1 and SMN2 based on spinal muscular atrophy severity. Nature
• Excellent review of the function of SMN. real-time LightCycler PCR: fast and highly Genet. 19(1), 63–66 (1998).

26 Expert Rev. Mol. Diagn. 4(1), (2004)


Spinal muscular atrophy

52 Sossi V, Giuli A, Vitali T et al. muscular atrophy gene (SMA) region at 72 Monani UR, Coovert DD, Burghes AH.
Premature termination mutations in exon 3 5q13 based on YAC and cosmid contiguous Animal models of spinal muscular atrophy.
of the SMN1 gene are associated with exon arrays. Genomics 26(3), 451–460 (1995). Hum. Mol. Genet. 9(16), 2451–2457
skipping and a relatively mild SMA 63 Rodrigues NR, Owen N, Talbot K et al. (2000).
phenotype. Eur. J. Hum. Genet. 9(2), Deletions in the survival motor neurone 73 Hsieh-Li HM, Chang JG, Jong YJ et al.
113–120 (2001). gene on 5q13 in autosomal recessive spinal A mouse model for spinal muscular atrophy.
53 Stevens G, Yawitch T, Rodda J, Verhaart S, muscular atrophy. Hum. Mol. Genet. 4(4), Nature Genet. 24(1), 66–70 (2000).
Krause A. Different molecular basis for 631–634 (1995). • Established a mouse model of SMA.
spinal muscular atrophy in South African 64 Hahnen E, Forkert R, Marke C et al. 74 Frugier T, Tiziano FD, Cifuentes-Diaz C
black patients. Am. J. Med. Genet. 86(5), Molecular analysis of candidate genes on et al. Nuclear targeting defect of SMN
420–426 (1999). chromosome 5q13 in autosomal recessive lacking the C-terminus in a mouse model
54 Wilmshurst JM, Reynolds L, Van Toorn R, spinal muscular atrophy: evidence of of spinal muscular atrophy. Hum. Mol.
Leisegang F, Henderson HE. Spinal muscular homozygous deletions of the SMN gene in Genet. 9(5), 849–858 (2000).
atrophy in black South Africans: concordance unaffected individuals. Hum. Mol. Genet. 75 Cifuentes-Diaz C, Frugier T, Tiziano FD
with the universal SMN1 genotype. 4(10), 1927–1933 (1995). et al. Deletion of murine SMN exon 7
Clin. Genet. 62(2), 165–168 (2002). 65 Scharf JM, Endrizzi MG, Wetter A et al. directed to skeletal muscle leads to severe
55 Cusin V, Clermont O, Gerard B, Identification of a candidate modifying muscular dystrophy. J. Cell. Biol. 152(5),
Chantereau D, Elion J. Prevalence of gene for spinal muscular atrophy by 1107–1114 (2001).
SMN1 deletion and duplication in carrier comparative genomics. Nature Genet. 76 Chan YB, Miguel-Aliaga I, Franks C et al.
and normal populations: implication for 20(1), 83–86 (1998). Neuromuscular defects in a Drosophila
genetic counselling. J. Med. Genet. 40(4), 66 Hofmann Y, Lorson CL, Stamm S, survival motor neurone gene mutant.
E39 (2003). Androphy EJ, Wirth B. Htra2-beta 1 Hum. Mol. Genet. 12(12), 1367–1376
56 Wirth B, Schmidt T, Hahnen E et al. stimulates an exonic splicing enhancer and (2003).
De novo rearrangements found in 2% of can restore full-length SMN expression to 77 McWhorter ML, Monani UR,
index patients with spinal muscular survival motor neurone 2 (SMN2). Proc. Burghes AH, Beattie CE. Knockdown of
atrophy: mutational mechanisms, parental Natl Acad. Sci. USA 97(17), 9618–9623 the survival motor neurone (Smn) protein
origin, mutation rate and implications for (2000). in zebrafish causes defects in motor axon
genetic counseling. Am. J. Hum. Genet. 67 Helmken C, Wirth B. Exclusion of outgrowth and pathfinding. J. Cell. Biol.
61(5), 1102–1111 (1997). Htra2-beta1, an upregulator of full-length 162(5), 919–932 (2003).
•• Largest study investigating the frequency SMN2 transcript, as a modifying gene for 78 Scheffer H, Cobben JM, Matthijs G, Wirth B.
of SMN1 de novo deletions among SMA spinal muscular atrophy. Hum. Genet. Best practice guidelines for molecular
patients lacking SMN1. 107(6), 554–558 (2000). analysis in spinal muscular atrophy. Eur. J.
57 Campbell L, Daniels RJ, Dubowitz V, 68 Helmken C, Wetter A, Hum. Genet. 9(7), 484–491 (2001).
Davies KE. Maternal mosaicism for a Rudnik-Schoneborn S et al. An essential 79 Melki J, Lefebvre S, Burglen L et al.
second mutational event in a Type I spinal SMN interacting protein (SIP1) is not De novo and inherited deletions of the 5q13
muscular atrophy family. Am. J. Hum. involved in the phenotypic variability of region in spinal muscular atrophies. Science
Genet. 63(1), 37–44 (1998). spinal muscular atrophy (SMA). Eur. J. 264(5164), 1474–1477 (1994).
58 Chen KL, Wang YL, Rennert H et al. Hum. Genet. 8(7), 493–499 (2000).
80 DiDonato CJ, Morgan K, Carpten JD et al.
Duplications and de novo deletions of the 69 Rochette CF, Gilbert N, Simard LR. Association between Ag1-CA alleles and
SMNT gene demonstrated by fluorescence- SMN gene duplication and the emergence severity of autosomal recessive proximal
based carrier testing for spinal muscular of the SMN2 gene occurred in distinct spinal muscular atrophy. Am. J. Hum.
atrophy. Am. J. Med. Genet. 85(5), hominids: SMN2 is unique to Genet. 55(6), 1218–1229 (1994).
463–469 (1999). Homo sapiens. Hum. Genet. 108(3),
81 Matthijs G, Schollen E, Legius E et al.
59 Ogino S, Leonard DG, Rennert H, Gao S, 255–266 (2001).
Unusual molecular findings in autosomal
Wilson RB. Heteroduplex formation in • Investigated the molecular evolution of
recessive spinal muscular atrophy. J. Med.
SMN gene dosage analysis. J. Mol. Diagn. the SMN genes.
Genet. 33(6), 469–474 (1996).
3(4), 150–157 (2001). 70 Schrank B, Gotz R, Gunnersen JM et al.
82 Wang CH, Xu J, Carter TA et al.
60 Velasco E, Valero C, Valero A, Moreno F, Inactivation of the survival motor neurone
Characterization of survival motor neurone
Hernandez-Chico C. Molecular analysis of gene, a candidate gene for human spinal
(SMNT) gene deletions in asymptomatic
the SMN and NAIP genes in Spanish spinal muscular atrophy, leads to massive cell
carriers of spinal muscular atrophy. Hum.
muscular atrophy (SMA) families and death in early mouse embryos. Proc. Natl
Mol. Genet. 5(3), 359–365 (1996).
correlation between number of copies of Acad. Sci. USA 94(18), 9920–9925
cBCD541 and SMA phenotype. Hum. (1997). 83 van der Steege G, Grootscholten P,
Mol. Genet. 5(2), 257–263 (1996). 71 Monani UR, Sendtner M, Coovert DD van der Vlies P et al. PCR-based DNA test
61 Roy N, Mahadevan MS, McLean M et al. et al. The human centromeric survival to confirm clinical diagnosis of autosomal
The gene for neuronal apoptosis inhibitory motor neurone gene (SMN2) rescues recessive spinal muscular atrophy. Lancet
protein is partially deleted in individuals embryonic lethality in Smn(-/-) mice and 345, 985–986 (1995).
with spinal muscular atrophy. Cell 80(1), results in a mouse with spinal muscular 84 Mailman MD, Heinz JW, Papp AC et al.
167–178 (1995). atrophy. Hum. Mol. Genet. 9(3), 333–339 Molecular analysis of spinal muscular
62 Roy N, McLean MD, Besner-Johnston A (2000). atrophy and modification of the phenotype
et al. Refined physical map of the spinal • Established a mouse model of SMA. by SMN2. Genet. Med. 4(1), 20–26 (2002).

www.future-drugs.com 27
Ogino & Wilson

85 Sutomo R, Akutsu T, Takeshima Y et al. 96 Thornhill AR, Snow K. 108 Wirth B. Spinal muscular atrophy:
Rapid SMN1 deletion test using DHPLC Molecular diagnostics in preimplantation state-of-the-art and therapeutic
to screen patients with spinal muscular genetic diagnosis. J. Mol. Diagn. 4(1), 11–29 perspectives. Amyotroph. Lateral Scler. Other
atrophy. Am. J. Med. Genet. 113(2), (2002). Motor Neuron Disord. 3(2), 87–95
225–226 (2002). 97 Verlinsky Y, Kuliev A. Preimplantation (2002).
86 Mazzei R, Conforti FL, Muglia M et al. diagnosis for diseases with genetic • Excellent overview of SMA.
A simple method for diagnosis of predisposition and nondisease testing. 109 Andreassi C, Jarecki J, Zhou J et al.
autosomal recessive spinal muscular Expert Rev. Mol. Diagn. 2(5), 509–513 Aclarubicin treatment restores SMN levels
atrophy by denaturing high-performance (2002). to cells derived from Type I spinal muscular
liquid chromatography. J. Child Neurol. 98 Braude P, Pickering S, Flinter F, atrophy patients. Hum. Mol. Genet. 10(24),
18(4), 269–271 (2003). Ogilvie CM. Preimplantation genetic 2841–2849 (2001).
87 Scheffer H, Cobben JM, Mensink RG et al. diagnosis. Nature Rev. Genet. 3(12), 110 Chang JG, Hsieh-Li HM, Jong YJ et al.
SMA carrier testing: validation of 941–953 (2002). Treatment of spinal muscular atrophy by
hemizygous SMN exon 7 deletion test for 99 Fallon L, Harton GL, Sisson ME et al. sodium butyrate. Proc. Natl Acad. Sci. USA
the identification of proximal spinal Preimplantation genetic diagnosis for spinal 98(17), 9808–9813 (2001).
muscular atrophy carriers and patients with muscular atrophy Type I. Neurology 53(5), 111 Brichta L, Hofmann Y, Hahnen E et al.
a single allele deletion. Eur. J. Hum. Genet. 1087–1090 (1999). Valproic acid increases the SMN2 protein
8(2), 79–86 (2000). level: a well-known drug as a potential
100 Dreesen JC, Bras M, de Die-Smulders C
88 Anhuf D, Eggermann T, et al. Preimplantation genetic diagnosis of therapy for spinal muscular atrophy.
Rudnik-Schoneborn S, Zerres K. spinal muscular atrophy. Mol. Hum. Hum. Mol. Genet. 12(19), 2481–2489
Determination of SMN1 and SMN2 copy Reprod. 4(9), 881–885 (1998). (2003).
number using TaqMan® technology. Hum. 112 DiDonato CJ, Parks RJ, Kothary R.
101 Blake D, Tan SL, Ao A. Assessment of
Mutat. 22(1), 74–78 (2003). Development of a gene therapy strategy for
multiplex fluorescent PCR for screening
• Describes methods to quantify the copy the restoration of survival motor neurone
single cells for trisomy 21 and single gene
number of SMN1 and SMN2 using protein expression: implications for spinal
defects. Mol. Hum. Reprod. 5(12),
TaqMan real-time PCR. muscular atrophy therapy. Hum. Gene Ther.
1166–1175 (1999).
89 Cusco II, Barcelo MJ, Baiget M, 14(2), 179–188 (2003).
102 Daniels G, Pettigrew R, Thornhill A et al.
Tizzano EF. Implementation of SMA 113 Lesbordes JC, Cifuentes-Diaz C,
Six unaffected livebirths following
carrier testing in genetic laboratories: Miroglio A et al. Therapeutic benefits of
preimplantation diagnosis for spinal
comparison of two methods for quantifying cardiotrophin-1 gene transfer in a mouse
muscular atrophy. Mol. Hum. Reprod.
the SMN1 gene. Hum. Mutat. 20(6), model of spinal muscular atrophy.
7(10), 995–1000 (2001).
452–459 (2002). Hum. Mol. Genet. 12(11), 1233–1239
103 Pickering S, Polidoropoulos N, Caller J
90 Ogino S, Wilson RB. Quantification of (2003).
et al. Strategies and outcomes of the first
PCR bias caused by a single nucleotide 114 Tsai CH, Jong YJ, Hu CJ et al.
100 cycles of preimplantation genetic
polymorphism in SMN gene dosage Molecular analysis of SMN, NAIP and
diagnosis at the Guy’s and St. Thomas’
analysis. J. Mol. Diagn. 4(4), 185–190 P44 genes of SMA patients and their
Center. Fertil. Steril. 79(1), 81–90 (2003).
(2002). families. J. Neurol. Sci. 190(1–2), 35–40
104 Yan H, Papadopoulos N, Marra G et al.
91 Parsons DW, McAndrew PE, Allinson PS (2001).
Conversion of diploidy to haploidy. Nature
et al. Diagnosis of spinal muscular atrophy 115 Skordis LA, Dunckley MG, Burglen L et al.
403(6771), 723–724 (2000).
in an SMN non-deletion patient using a Characterisation of novel point mutations
quantitative PCR screen and mutation 105 Mailman MD, Hemingway T, Darsey RL
in the survival motor neurone gene SMN in
analysis. J. Med. Genet. 35(8), 674–676 et al. Hybrids monosomal for human
three patients with SMA. Hum. Genet.
(1998). chromosome 5 reveal the presence of a
108(4), 356–357 (2001).
92 Ogino S, Gao S, Leonard DG, Paessler M, spinal muscular atrophy (SMA) carrier with
two SMN1 copies on one chromosome. 116 Brahe C, Clermont O, Zappata S et al.
Wilson RB. Inverse correlation between Frameshift mutation in the survival motor
SMN1 and SMN2 copy numbers: evidence Hum. Genet. 108(2), 109–115 (2001).
•• Describes methods to detect carriers with neurone gene in a severe case of SMA
for gene conversion from SMN2 to SMN1. Type I. Hum. Mol. Genet. 5(12),
Eur. J. Hum. Genet. 11(3), 275–277 (2003) two SMN1 copies on one chromosome 5
and a deletion/conversion on the other. 1971–1976 (1996).
Addendum in 11(9), 723.
106 Skordis LA, Dunckley MG, Yue B, 117 Hahnen E, Schonling J,
93 Van Deerlin VM, Leonard DG. Rudnik-Schoneborn S et al.
Eperon IC, Muntoni F. Bifunctional
Bone marrow engraftment analysis after Missense mutations in exon 6 of the
antisense oligonucleotides provide a
allogeneic bone marrow transplantation. survival motor neurone gene in patients
trans-acting splicing enhancer that
Clin. Lab. Med. 20(1), 197–225 (2000). with spinal muscular atrophy (SMA).
stimulates SMN2 gene expression in patient
94 Beroud C, Karliova M, Bonnefont JP et al. fibroblasts. Proc. Natl Acad. Sci. USA Hum. Mol. Genet. 6(5), 821–825
Prenatal diagnosis of spinal muscular atrophy 100(7), 4114–4119 (2003). (1997).
by genetic analysis of circulating fetal cells. 118 Wang CH, Papendick BD, Bruinsma P,
107 Kerr DA, Llado J, Shamblott MJ et al.
Lancet 361(9362), 1013–1014 (2003). Day JK. Identification of a novel missense
Human embryonic germ cell derivatives
95 Benachi A, Steffann J, Gautier E et al. facilitate motor recovery of rats with diffuse mutation of the SMN(T) gene in two
Fetal DNA in maternal serum: does it motor neurone injury. J. Neurosci. 23(12), siblings with spinal muscular atrophy.
persist after pregnancy? Hum. Genet. 5131–5140 (2003). Neurogenetics 1(4), 273–276 (1998).
113(1), 76–79 (2003).

28 Expert Rev. Mol. Diagn. 4(1), (2004)


Spinal muscular atrophy

119 Talbot K, Ponting CP, Theodosiou AM Affiliations • Robert B Wilson, MD, PhD
et al. Missense mutation clustering in the • Shuji Ogino, MD, PhD Associate Professor
survival motor neurone gene: a role for a Associate Pathologist Department of Pathology and
conserved tyrosine and glycine rich region Department of Pathology, Laboratory Medicine,
of the protein in RNA metabolism? Brigham and Women’s Hospital University of Pennsylvania Medical Center,
Hum. Mol. Genet. 6(3), 497–500 Philadelphia, PA 19104, USA
Staff Research Scientist
(1997). Tel.: +1 215 898 0606
Department of Medical Oncology,
Dana–Farber Cancer Institute Fax: +1 215 573 8944
120 Gambardella A, Mazzei R, Toscano A et al. wilsonr@mail.med.upenn.edu
Spinal muscular atrophy due to an isolated Harvard Medical School,
deletion of exon 8 of the telomeric survival Boston, MA 02115, USA
motor neurone gene. Ann. Neurol. 44(5), Tel.: +1 617 632 3978
836–839 (1998). Fax: +1 617 632 5762
sogino@partners.org

www.future-drugs.com 29

View publication stats

You might also like