Open access

Spinal Muscular Atrophy: Classification, Diagnosis, Background, Molecular Mechanism and Development of Therapeutics

Written By

Faraz Tariq Farooq, Martin Holcik and Alex MacKenzie

Submitted: 05 June 2012 Published: 15 May 2013

DOI: 10.5772/53800

From the Edited Volume

Neurodegenerative Diseases

Edited by Uday Kishore

Chapter metrics overview

4,036 Chapter Downloads

View Full Metrics

1. Introduction

Spinal muscular atrophy (SMA) is an autosomal recessive neurodegenerative disease and one of the most common genetic causes of infant death. The loss or mutation of the SMN1 gene results in reduced SMN protein level leading to motor neuron death and progressive muscle atrophy. Although recent progress has been made in our understanding of the molecular mechanisms underlying the pathogenesis of the disease, there is currently no cure for SMA. In this review, we summarize the clinical manifestations, molecular pathogenesis, diagnostic strategy and development of therapeutic regimes for the better understanding and treatment of SMA.

Advertisement

2. Epidemiology

Spinal muscular atrophy (SMA) is an autosomal recessive neuromuscular disorder characterized by the loss of motor neurons from the anterior horn of the spinal cord which leads to muscle weakness, hypotonia and ultimately muscle atrophy [1]. With a pan ethnic incidence of 1:11,000 live births and a carrier frequency of 1:50, SMA is one of the leading genetic causes of infant death globally [1-5].

Advertisement

3. Clinical classification

Due to the range of clinical severity, SMA is broadly classified into four major categories characterized by the age of onset as well as severity of the disease [6-9]. SMA type I, which was originally described by Werdnig and Hoffmann in the late 18th century is the most severe and prevalent form of the disease and accounts for more than 50% of the known diagnosed cases of SMA. Type I SMA presents within the first six months after birth and although historically patients succumbed within the first 2 years of life, with better ventilatory and nutritional support, the life expectancy of children with type I SMA can be increased beyond the 5th birthday. Infants with type I SMA experience a rapid loss of skeletal muscle mass with profound hypotonia and general muscle weakness characterized by poor head control, difficulty with suckling, swallowing and an inability to sit without support. These children develop problems with breathing over time due to impaired bulbar function and respiratory muscle weakness leading to respiratory insufficiency. Respiratory failure due to aspiration pneumonia is an important cause of SMA mortality [6, 10, 11]. The intermediate form of SMA, known as type II, has an onset between 6 and 18 months of age. Patients with type II SMA can sit unaided but still develop progressive muscle weakness and can never stand or walk on their own. Other symptoms and physical signs include respiratory insufficiency due to reduced bulbar function, poor weight gain, fine hand tremors and joint contractures [6]. SMA type III has an onset between 18 months to 30 years of age. Patients are able to stand and walk unaided, however they develop variable degree of muscle weakness which leads to a broad spectrum of physical signs and symptoms. While most walk independently, some lose ambulation during early adulthood and require wheelchair assistance. Others develop cramps and joint overuse problems; some develop scoliosis [6, 12, 13]. Type IV SMA is the mildest form of the condition and is characterized by adult onset with normal mobility. They have mild muscle weakness in adulthood with normal longevity [6] (Table 1).

SMA Type Other Names Age of Onset Life
Span
Highest Motor Activity
Type I
years
(Severe)
Werdnig- Hoffmann disease 0-6 months 2-5 Never sit
Type II
(Intermediate)
SMA, Dubowitz type 7-18 months >2 years Sit, Never stand
Type III
(Mild)
Kugelberg-Welander
disease
>18 months Adult Stand and walk
(may require assistance)
Type IV
(Adult)
------------- Adulthood Normal Walk during
adulthood-unassisted
(some muscle weakness)

Table 1.

Classification of SMA disease

Advertisement

4. Diagnosis and treatment

The diagnosis of SMA is made by a thorough patient history and physical examination followed by genetic testing. The survival of motor neuron (SMN) -1 genotyping has to a large degree replaced electromyography (EMG) and muscle biopsies (Fig 1) [2, 14]. There is in 2012 no cure for SMA; current treatment is symptomatic and supportive. This includes clinical management through family education and counselling along with attention to pulmonary, gastrointestinal/nutrition and orthopedics/rehabilitation in an effort to managing symptoms of the patients [9].

Figure 1.

SMA diagnosis

Advertisement

5. Genetics of the disease

The SMA disease causing SMN1 gene maps to a complex genomic region of chromosome 5q13.1. This region is characterized by an inverted duplication of the element with 4 genes (SMN, neuronal apoptosis inhibitor protein {NAIP}, SERF and GTFH2) present in telomeric and centromeric copies (Fig 2a) [15, 16]. In 1995, it was reported that homozygous deletions of the SMN1 gene were observed in and thus likely the cause of 95% of SMA patients [15]. All SMA patients have one or more copies of a nearly identical gene, SMN2. These two genes are distinguished by five nucleotide changes in exon 7 and 8. The critical nucleotide difference which makes SMN2 only partially functional is a C to T transition at position 6 of exon 7. This change leads to the exclusion of exon 7 in the majority of transcripts. This mRNA is subsequently translated to form an unstable truncated non-oligomerizing isoform of SMN protein. However, SMN2 gene still produces 5-10% functional full length SMN transcripts (Fig 1.2b) [15, 17, 18]. The SMN2 gene is present in variable copy numbers in the population; all SMA patients have one or more copy of the SMN2 gene which, due to its partial functionality, acts as a positive disease modifier. There is thus an inverse correlation between the number of SMN2 gene (which can produce between 10-50% of SMN protein depending on copy number) and the severity of the disease [2]. Low levels of SMN protein allows embryonic development but is not enough, in the long term, to allow motor neurons to survive in the spinal cord [19, 20]. Type I patients usually have 2 copies whereas Type II have 3 copies of SMN2. Type III and IV have 3-4 copies of the SMN2 gene. Individuals with 5 or more copies of the SMN2 gene, despite having no functional SMN1 gene are completely asymptomatic and are protected against the disease manifestation.

Figure 2.

(a) Human SMN locus and (b) genetics of SMA patients

Advertisement

6. Pathology

The pathological hallmark of all forms of SMA is the loss of motor neurons from the lower brainstem and the anterior horn of the spinal cord [21]. Anterograde axonal degeneration results in denervation of the myocytes within the motor unit. This sometimes leads to reinnervation of muscle, where adjacent uninjured motor neurons sprout leads to fiber type grouping of myocytes. Histopathologic assessment of SMA muscle tissues reveals a large number of rounded atrophic fibers resulting from denervation. The widely held notion had been that SMA is primarily a neuronopathy (involving the cell body) with secondary degeneration of the axons. However, more recent observations in the field have shifted the focus of SMA pathology from the motor neuron cell body to the distal axon [22, 23] and the possibility of a synaptopathic defect [20, 24]. Specifically it has been suggested that the presynaptic transcriptome may be in some manner dysregulated; the direct inference is that SMN plays a role in the peripheral transport of critical mRNA, among which is that species encoding beta-actin. Regardless of the subcellular location of SMN mediated pathology, SMA is primarily considered as a motor neuron disease and consequently treatment strategies focus on drugs which can cross the blood brain barrier (BBB) to target the central nervous system (CNS). However, motor neuron autonomy of SMA pathogenesis has recently been called into question as multi-system involvement (including cardiovascular, peripheral necrosis and liver defects) have been reported recently in both SMA patients and SMA mice models [25-33]. In addition, one report has outlined the superiority of systemic SMN antisense oligonucleotide (ASO) therapy compared with intrathecal delivery in severe murine SMA calling into question the exclusive role of the motor neuron in disease causation [33].

Advertisement

7. Function of the SMN protein

SMN is a 294 amino acid long ubiquitously expressed protein with a molecular weight of 38 kilodaltons (kD). SMN is found in both the nucleus and cytoplasm. Within the nucleus, it is localized both throughout the nucleoplasm as well as in nuclear structures called Gems and Cajal bodies [34]. It is also found in abundance within the growth cones of the motor neurons [35]. SMN has been implicated in ribonucleoprotein biogenesis (e.g. assembly, metabolism and transport of various ribonucleoproteins), as well as playing a major role in the splicing machinery. It is part of a multiprotein complex comprised of Gemins [2-8], spliceosomal U-snRNPs, Sm proteins and profilins called the SMN complex. This complex is essential for the biogenesis of snRNPs [36-45]. Given the variety of roles that SMN has been implicated in, not surprisingly, the complete absence of SMN genes is embryonically lethal in virtually all metazoan life forms tested, indeed even cell cultures cannot survive without SMN [19, 20, 46].

Advertisement

8. Molecular mechanism: splicing defect in SMA

Splicing is mediated by a complex called the spliceosome, the activity of which depends on a number of factors. In particular, various cis- and trans-acting elements regulate the splicing of both SMN1 and SMN2. The C-T transition at position 6 of exon 7 in SMN2 gene disrupts the function of an exonic splice enhancer (ESE; recognized by SF2/ASF to promote exon 7 inclusion) and/or creates an exonic splice suppressor (ESS; recognized by hnRNP A1/A2) which results in exon 7 skipping (Fig 3) [47-53].

Figure 3.

Splicing in SMA

Advertisement

9. Therapeutic strategies

Although there is no cure for SMA, the SMN2 gene locus serves as a target for SMA treatment. The general treatment strategies for SMA are to compensate fully or in part for the absence of SMN1 gene by increasing the levels of functional SMN protein levels though three distinct approaches: i) to induce the expression of SMN2, ii) to modulate splicing of SMN2 transcript, and iii) to stabilize the full length SMN mRNA and/or protein. In addition, gene and stem cell therapies are also under development for the treatment of SMA. These and other strategies are discussed below.

  1. SMN dependent therapies: As outlined above, there is an inverse correlation between the SMN2 gene copy number and disease severity [54, 55] which implies that directly targeting the SMN2 gene in SMA patients through different pathways could be one key for the development of a SMA drug treatment. Alternatively, SMN protein can also be produced through gene replacement therapy.

  1. Activation of SMN2 promoter: Histone deacetylases (HDACs) repress transcription of genes including SMN2 by chromatin condensation. Thus, HDAC inhibitors can increase transcription of the SMN2 gene and can produce more full length SMN transcripts and protein which may have a beneficial effect in patients. Various HDAC inhibitors have been analyzed in cell culture, mouse models and in clinical trials as potential therapeutic for SMA. Sodium butyrate, Valproic acid (VPA) and phenylbutyrate showed promise in cell culture and mouse models and were also well tolerated by the patients [56-63]. However, no clinical improvement was observed in SMA patients with HDAC inhibitors [61-63].

    Recent studies with other HDAC inhibitors, LBH589, Trichostatin A (TSA) and Suberoylanilide hydroxamic acid (SAHA) showed SMN2 gene induction in culture as well as in a number of animal models of neurodegeneration [62, 64-66]. In addition to these compounds, we have shown that the lactation hormone prolactin (PRL) which can both cross the blood brain barrier and, through binding to its receptor, activate the JAK2/STAT5 pathway also upregulates SMN2 gene transcription [68]. Interestingly the degree of induction in SMN seen with the prolactin in the genetically engineered ∆7 SMA mouse model (where SMN2 gene is the only source of SMN protein) is significantly greater than that seen in cell culture and wild type mice. We have determined that this is because of the difference between the promoter regions in SMN1 and SMN2 genes, the latter uniquely having STAT5a transcription binding motifs. This might prove beneficial as all SMA patients have SMN2 as the only source of SMN protein. Since PRL has been successfully tested and proven safe in humans for the treatment of lactation deficient mothers [67], it may bypass other compounds which are yet to be tested for clinical safety and join the short list of drugs which may have immediate potential SMA therapeutic potential [68].

  2. Correction of splicing: The suppression of exon 7 skipping to produce more full length transcript from the SMN2 gene is another treatment strategy being explored for SMA. HDAC inhibitors such as VPA, TSA and sodium butyrate appear to have a dual effect on SMN mRNA expression; they not only open chromatin structure and therefore increase the rate of transcription but also appear to affect the splicing process [56-58, 64]. The antibiotic aclarubicin has been shown to increase full length SMN transcript by altering the splicing process in vitro [69]. The most promising compounds which correct splicing by preventing SMN2 exon 7 skipping are antisense oligos (ASOs). An ASO complementary to SMN2 exon 7 pre-mRNA sequences has been shown to inhibit binding of negative splicing factors and increase full length SMN transcript and protein production [30, 33, 70, 71]. The major hurdle in using ASOs for SMA therapeutics, however, is their inability to cross the blood brain barrier. However, Hua et al. 2011 documented a marked improvement in motor function along with an increase in survival in SMA mice with systemic delivery of ASO which results into increase in SMN levels mostly in peripheral tissues especially in liver. Interestingly, they documented only a slight increase in SMN levels in CNS tissues [33]. However, there are a number of issues which need to be addressed before clinical introduction of ASOs for SMA treatment (clinical safety, quantity of ASO, cost, immune response etc) [72].

  3. Full length SMN transcript stabilization: In this relatively new approach by by Singh et al., decapping enzyme DcpS, an integral part of the RNA degradation machinery, was targeted by C5-substituted quinazolines which interact and open the enzyme into a catalytically inactivated conformation. Full length SMN mRNA decay is in this fashion blocked, ultimately increasing SMN protein in cell culture [73].

    In a different approach, SMN mRNA has been shown to have a specific AU rich element (ARE) region in its 3’ UTR which marks the mRNA for degradation. Our laboratory has shown that activation of the p38 pathway results in the accumulation of RNA binding protein HuR in the cytoplasm which then binds to the ARE in 3’UTR region of SMN mRNA and stabilizes the transcript. Importantly, transcript stabilization is not associated with any discernible inhibition of SMN protein translation. This study provided a novel mechanism through which SMN mRNA could be stabilized using p38 activating compounds which can cross the blood brain barrier to develop new therapeutics for SMA treatment [74].

  4. Full length SMN protein stabilization: Aminoglycosides are class of antibiotics which have been shown to mask premature stop codon mutations in some genes, allowing read through translation to occur. This moderates translation termination through an alteration in the conformation of the ribosomal reading site. Various aminoglycosides including tobramycin and amikacin have been used successfully in patient fibroblasts to increase SMN protein levels. However, their in vivo efficacy and safety has yet to be demonstrated [75-77].

    An alternative potential therapeutic approach involves targeting the ubiquitin-proteasome pathway which mediates intracellular protein turnover. Proteins are marked with poly ubiquitin (Ub) molecules by the action of the enzymes E1 (Ub activating enzyme), E2 (Ub conjugating enzyme) and E3 (Ub ligase). The polyubiquitin modification marks the protein for destruction by the proteasome complex. SMN is one of the many proteins degraded by the ubiquitin proteasome pathway. It has been shown that FDA approved proteasome inhibitor bortezomib increases SMN both in vitro and in vivo by blocking proteolysis of SMN protein. However, it should be noted that bortezomib cannot cross the BBB; thus, it must be used in combination with other drugs which can cross the BBB for the treatment of SMA [78].

  5. Gene therapy: One of the most encouraging SMA therapeutic advances is the use of gene therapy which shows significant promise. In the last three years several groups have used self complementary adeno-associated virus (scAAV) 8 and 9 vectors carrying the SMN1 cDNA to treat mice models of SMA, resulting in the most dramatic extension in the life span of mice yet observed combined with an overall amelioration of disease phenotype [79-82]. However, early pre-symptomatic intervention is necessary for the success of this therapy as is seen with other treatment strategies as well. Moreover, several challenges must be addressed for this mode of SMA treatment before bringing it to clinical application successfully. The most pressing issues are clinical safety, dealing with the cross-species barriers, the cost of virus production along with the possibility of an immune response to AAV which can neutralize its impact [83].

  1. SMN-independent strategies: There have been some recent advances in SMN-independent strategies for the treatment of SMA. These include:

  1. Stem cell therapy: Stem cell therapy has generated much attention as a treatment for motor neuron diseases, including SMA, through replacement of the lost motor neurons and, more realistically perhaps, supporting the existing neuron population. Primary murine neuronal stem cells as well as embryonic stem cell-derived neural stem cells injected into the spinal cord of animal models of SMA have been shown to ameliorate disease phenotype and increase survival [84, 85]. It is unclear whether this is through motor neuron and other cell replacement and/or through neuroprotection of host motor neurons by the numerous factors released from the donor cells. Although induced pluripotent stem (iPS) cells from an SMA patient have been differentiated into motor neurons [86, 87], there are several obstacles which hinder their use as a therapeutic for SMA treatment. These challenges include the production of the large number of stem cells and their successful transplantation into the patients, which could populate and cover the entire nervous system. Also, lentivirus vectors are used to deliver the cocktail of factors, required to produce iPS cells in vitro; these would be unsuitable for use in patients as they have the potential for insertional mutagenesis which could result into oncogenesis. Finally, even if motor neurons could develop in situ, the prospect that they would at a meaningful level connect with the host CNS must be viewed as highly unlikely at this time.

  2. Modifying neuromuscular junctions through actin dynamics: The pharmacological Rho-kinase inhibitor (downstream effector of RhoA-GTP which plays role in actin dynamics) dramatically increases the life span of a mild SMA mouse model and improves disease phenotype. This improvement in the disease phenotype is independent of SMN increase, mainly through making neuromuscular junctions (NMJ) better, larger and more mature [88]. This suggests that there are novel SMN independent avenues for the development of therapeutics for SMA.

Figure 4.

Development of therapeutics for SMA

Advertisement

10. Future directions

Combination therapy: The impressive results seen so far with gene therapy and ASO's in the field of SMA will be difficult to equal with a monotherapy approach. However, unless and until gene therapy and ASO treatments are cleared for clinical safety as a therapeutic option for SMA treatment, combinatorial approaches for SMA shall likely be necessary to target not only CNS but also other tissues which are affected because of a lack of SMN. As outlined above, SMA can be targeted through different approaches, we can in a safe combination use compounds which are already FDA approved and can increase SMN levels through SMN2 gene activation (such as PRL) along with SMN2 transcript stabilizers (p38 pathway activators such as celecoxib) and/or SMN protein stabilizer (proteasome inhibitor bortezomib) [18] and/or neuroprotective compounds (Rho kinase inhibitor) [19], or a cocktail of the best suitable combination of these compounds (Fig 5.1). I believe that this approach will speed up the process of finding the best possible and safest treatment of SMA. This approach is currently being assayed in our laboratory and others, showing some positive and promising results in the severe mouse model of the disease. More work is required to assess the potential drug interactions and their side effects in the animal models of the disease before pushing this approach for human clinical trials.

Designing clinical trials for SMA: In the last 5 years, a tremendous amount of promising translational work has been done using animal models of the SMA which is progressing rapidly towards the pre-clinical stage. However there are major challenges for designing a perfect clinical trial for SMA which includes 1) Variability of the disease phenotype, 2) lack of molecular biomarkers, 3) Accessibility of treatment centers and 4) lack of agreement for standard of care and disease management. However these issues are likely to be resolved as recently there has been a remarkable cooperation and collaboration between researchers, clinicians, industry, government and volunteer organizations which is bringing everyone on the same page to address these issues and reach a consensus for designing standard human clinical trials for SMA internationally.

Early intervention: New born screening: We and others have seen, irrespective of the modality, that early timing of the treatment is critical for maximum benefit in the mouse model of the disease. Presymptomatic identification of infants with SMA through new-born screening represents an important step in the effective treatment of SMA. In essence we shall need to intervene before the damage is done; to do so we need to rapidly identify infants with SMA, cases who will also serve as the best candidates to show the efficacy of promising therapeutic treatments in the near future. Children in which the disease has already progressed may also benefit with the use of best combinational approach, however the aim will be more towards ameliorating the disease progress and preserving the function of remaining motor neurons and other tissues rather than a complete reversal of the disease phenotype.

Figure 5.

Proposed model of combination therapy for SMA treatment.

References

  1. 1. PearnJ1978Incidence, prevalence, and gene frequency studies of chronic childhood spinal muscular atrophy. J. Med. Genet. 15409413
  2. 2. FeldkotterMSchwarzerVWirthRWienkerT. Fand WirthB2002Quantitative analyses of SMN1 and SMN2 based on real-time lightCycler PCR: fast and highly reliable carrier testing and prediction of severity of spinal muscular atrophy. Am. J. Hum. Genet. 70358368
  3. 3. OginoSLeonardD. GRennertHEwensW. Jand WilsonR. B2002Genetic risk assessment in carrier testing for spinal muscular atrophy. Am. J. Med. Genet. 110301307
  4. 4. OginoSWilsonR. Band GoldB2004New insights on the evolution of the SMN1 and SMN2 region: simulation and meta-analysis for allele and haplotype frequency calculations. Eur. J. Hum. Genet. 1210151023
  5. 5. Sugarman, E. A., Nagan, N., Zhu, H., Akmaev, V. R., Zhou, Z., Rohlfs, E. M., Flynn, K., Hendrickson, B. C., Scholl, T., Sirko-Osadsa, D. A., Allitto, B.A. 2012. Pan-ethnic carrier screening and prenatal diagnosis for spinal muscular atrophy: clinical laboratory analysis of >72,400 specimens. Eur. J. Hum. Genet. 20(1):27-32.
  6. 6. ZerresKand DaviesK. E1999th ENMC International Workshop: Spinal Muscular Atrophies: recent progress and revised diagnostic criteria 17-19 April 1998, Soestduinen, The Netherlands. Neuromuscul. Disord. 9272278
  7. 7. DubowitzV1995Chaos in the classification of SMA: a possible resolution. Neuromuscul. Disord. 535
  8. 8. IannacconeS. TRussmanB. SBrowneR. HBuncherC. RWhiteMand SamahaF. J2000Prospective analysis of strength in spinal muscular atrophy. DCN/Spinal Muscular Atrophy Group. J. Child Neurol. 1597101
  9. 9. WangC. HFinkelR. SBertiniE. SSchrothMSimondsAWongBAloysiusAMorrisonLMainMCrawfordT. Oet al2007Consensus statement for standard of care in spinal muscular atrophy. J. Child Neurol. 2210271049
  10. 10. OHagenJ. MGlanzmanA. MMcdermottM. PRyanP. AFlickingerJQuigleyJRileySSanbornEIrvineCMartensW.B. et al2007An expanded version of the Hammersmith Functional Motor Scale for SMA II and III patients. Neuromuscul. Disord. 17693697
  11. 11. ThomasN. Hand DubowitzV1994The natural history of type I (severe) spinal muscular atrophy. Neuromuscul. Disord. 4497502
  12. 12. ZerresKRudnik-schonebornSForrestELusakowskaABorkowskaJand Hausmanowa-petrusewiczI1997A collaborative study on the natural history of childhood and juvenile onset proximal spinal muscular atrophy (type II and III SMA): 569 patients. J. Neurol. Sci. 1466772
  13. 13. ZerresKWirthBand Rudnik-schonebornS1997Spinal muscular atrophy--clinical and genetic correlations. Neuromuscul. Disord. 7202207
  14. 14. PriorT. Wand RussmanB. S1993Spinal Muscular Atrophy. In GeneReviews. R.A. Pagon, T.D. Bird, C.R. Dolan, and K. Stephens, editors. University of Washington, Seattle. Seattle (WA).
  15. 15. LefebvreSBurglenLReboulletSClermontOBurletPViolletLBenichouBCruaudCMillasseauPand ZevianiM1995Identification and characterization of a spinal muscular atrophy-determining gene. Cell 80155165
  16. 16. MahadevanM. SKornelukR. GRoyNMacKenzie, A., and Ikeda, J. 1995SMA genes: deleted and duplicated. Nat. Genet. 9112113
  17. 17. LorsonC. LHahnenEAndrophyE. Jand WirthB1999A single nucleotide in the SMN gene regulates splicing and is responsible for spinal muscular atrophy. Proc. Natl. Acad. Sci. U. S. A. 9663076311
  18. 18. MonaniU. RLorsonC. LParsonsD. WPriorT. WAndrophyE. JBurghesA. Hand McphersonJ. D1999A single nucleotide difference that alters splicing patterns distinguishes the SMA gene SMN1 from the copy gene SMN2. Hum. Mol. Genet. 811771183
  19. 19. SchrankBGotzRGunnersenJ. MUreJ. MToykaK. VSmithA. Gand SendtnerM1997Inactivation of the survival motor neuron gene, a candidate gene for human spinal muscular atrophy, leads to massive cell death in early mouse embryos. Proc. Natl. Acad. Sci. U. S. A. 9499209925
  20. 20. SimicG2008Pathogenesis of proximal autosomal recessive spinal muscular atrophy. Acta Neuropathol. 116223234
  21. 21. MurayamaSBouldinT. Wand SuzukiK1991Immunocytochemical and ultrastructural studies of Werdnig-Hoffmann disease. Acta Neuropathol. 81408417
  22. 22. PinterM. JWaldeckR. FWallaceNand CorkL. C1995Motor unit behavior in canine motor neuron disease. J. Neurosci. 1534473457
  23. 23. CorkL. CStrubleR. GGoldB. GDiCarlo, C., Fahnestock, K.E., Griffin, J.W., and Price, D.L. 1989Changes in size of motor axons in hereditary canine spinal muscular atrophy. Lab. Invest. 61333342
  24. 24. KariyaSParkG. HMaeno-hikichiYLeykekhmanOLutzCArkovitzM. SLandmesserL. Tand MonaniU. R2008Reduced SMN protein impairs maturation of the neuromuscular junctions in mouse models of spinal muscular atrophy. Hum. Mol. Genet. 1725522569
  25. 25. MenkeL. APoll-theB. TClurS. ABilardoC. MVan Der WalA. CLemminkH. Hand CobbenJ. M2008Congenital heart defects in spinal muscular atrophy type I: a clinical report of two siblings and a review of the literature. Am. J. Med. Genet. A. 146A:740744
  26. 26. Rudnik-schonebornSHellerRBergCBetzlerCGrimmTEggermannTEggermannKWirthRWirthBand ZerresK2008Congenital heart disease is a feature of severe infantile spinal muscular atrophy. J. Med. Genet. 45635638
  27. 27. BevanA. KHutchinsonK. RFoustK. DBraunLMcgovernV. LSchmelzerLWardJ. GPetruskaJ. CLucchesiP. ABurghesA. Het al2010Early heart failure in the SMN∆7 model of spinal muscular atrophy and correction by postnatal scAAV9-SMN delivery. Hum. Mol. Genet. 1938953905
  28. 28. HeierC. RSattaRLutzCand DiDonato, C.J. 2010Arrhythmia and cardiac defects are a feature of spinal muscular atrophy model mice. Hum. Mol. Genet. 1939063918
  29. 29. ShababiMHabibiJYangH. TValeS. MSewellW. Aand LorsonC. L2010Cardiac defects contribute to the pathology of spinal muscular atrophy models. Hum. Mol. Genet. 1940594071
  30. 30. WilliamsJ. HSchrayR. CPattersonC. AAyiteyS. OTallentM. Kand LutzG. J2009Oligonucleotide-mediated survival of motor neuron protein expression in CNS improves phenotype in a mouse model of spinal muscular atrophy. J. Neurosci. 2976337638
  31. 31. Rudnik-schonebornSVogelgesangSArmbrustSGraul-neumannLFuschCand ZerresK2010Digital necroses and vascular thrombosis in severe spinal muscular atrophy. Muscle Nerve 42144147
  32. 32. Araujo AdeQ., Araujo, M., and Swoboda, K.J. 2009Vascular perfusion abnormalities in infants with spinal muscular atrophy. J. Pediatr. 155292294
  33. 33. HuaYSahashiKRigoFHungGHorevGBennettC. Fand KrainerA. R2011Peripheral SMN restoration is essential for long-term rescue of a severe spinal muscular atrophy mouse model. Nature 478123126
  34. 34. LiuQand DreyfussG1996A novel nuclear structure containing the survival of motor neurons protein. EMBO J. 1535553565
  35. 35. ZhangHXingLRossollWWichterleHSingerR. Hand BassellG. J2006Multiprotein complexes of the survival of motor neuron protein SMN with Gemins traffic to neuronal processes and growth cones of motor neurons. J. Neurosci. 2686228632
  36. 36. FriesenW. Jand DreyfussG2000Specific sequences of the Sm and Sm-like (Lsm) proteins mediate their interaction with the spinal muscular atrophy disease gene product (SMN). J. Biol. Chem. 2752637026375
  37. 37. BuhlerDRakerVLuhrmannRand FischerU1999Essential role for the tudor domain of SMN in spliceosomal U snRNP assembly: implications for spinal muscular atrophy. Hum. Mol. Genet. 823512357
  38. 38. JonesK. WGorzynskiKHalesC. MFischerUBadbanchiFTernsR. Mand TernsM. P2001Direct interaction of the spinal muscular atrophy disease protein SMN with the small nucleolar RNA-associated protein fibrillarin. J. Biol. Chem. 2763864538651
  39. 39. MeisterGBuhlerDLaggerbauerBZobawaMLottspeichFand FischerU2000Characterization of a nuclear 20S complex containing the survival of motor neurons (SMN) protein and a specific subset of spliceosomal Sm proteins. Hum. Mol. Genet. 919771986
  40. 40. MourelatosZAbelLYongJKataokaNand DreyfussG2001SMN interacts with a novel family of hnRNP and spliceosomal proteins. EMBO J. 2054435452
  41. 41. NarayananUAchselTLuhrmannRand MateraA. G2004Coupled in vitro import of U snRNPs and SMN, the spinal muscular atrophy protein. Mol. Cell 16223234
  42. 42. PellizzoniLBacconJCharrouxBand DreyfussG2001The survival of motor neurons (SMN) protein interacts with the snoRNP proteins fibrillarin and GAR1. Curr. Biol. 1110791088
  43. 43. PellizzoniLCharrouxBand DreyfussG1999SMN mutants of spinal muscular atrophy patients are defective in binding to snRNP proteins. Proc. Natl. Acad. Sci. U. S. A. 961116711172
  44. 44. PellizzoniLCharrouxBRappsilberJMannMand DreyfussG2001A functional interaction between the survival motor neuron complex and RNA polymerase II. J. Cell Biol. 1527585
  45. 45. FischerULiuQand DreyfussG1997The SMN-SIP1 complex has an essential role in spliceosomal snRNP biogenesis. Cell 9010231029
  46. 46. KariyaSMauricioRDaiYand MonaniU. R2009The neuroprotective factor Wld(s) fails to mitigate distal axonal and neuromuscular junction (NMJ) defects in mouse models of spinal muscular atrophy. Neurosci. Lett. 449246251
  47. 47. HofmannYLorsonC. LStammSAndrophyE. Jand WirthB2000Htra2-beta 1 stimulates an exonic splicing enhancer and can restore full-length SMN expression to survival motor neuron 2 (SMN2). Proc. Natl. Acad. Sci. U. S. A. 9796189623
  48. 48. ChenH. HChangJ. GLuR. MPengT. Yand TarnW. Y2008The RNA binding protein hnRNP Q modulates the utilization of exon 7 in the survival motor neuron 2 (SMN2) gene. Mol. Cell. Biol. 2869296938
  49. 49. HofmannYand WirthB2002hnRNP-G promotes exon 7 inclusion of survival motor neuron (SMN) via direct interaction with Htra2-beta1. Hum. Mol. Genet. 1120372049
  50. 50. YoungP. JDiDonato, C.J., Hu, D., Kothary, R., Androphy, E.J., and Lorson, C.L. 2002SRp30c-dependent stimulation of survival motor neuron (SMN) exon 7 inclusion is facilitated by a direct interaction with hTra2 beta 1. Hum. Mol. Genet. 11577587
  51. 51. CartegniLand KrainerA. R2002Disruption of an SF2/ASF-dependent exonic splicing enhancer in SMN2 causes spinal muscular atrophy in the absence of SMN1. Nat. Genet. 30377384
  52. 52. CartegniLand KrainerA. R2003Correction of disease-associated exon skipping by synthetic exon-specific activators. Nat. Struct. Biol. 10120125
  53. 53. KashimaTand ManleyJ. L2003A negative element in SMN2 exon 7 inhibits splicing in spinal muscular atrophy. Nat. Genet. 34460463
  54. 54. McandrewP. EParsonsD. WSimardL. RRochetteCRayP. NMendellJ. RPriorT. Wand BurghesA. H1997Identification of proximal spinal muscular atrophy carriers and patients by analysis of SMNT and SMNC gene copy number. Am. J. Hum. Genet. 6014111422
  55. 55. CampbellLPotterAIgnatiusJDubowitzVand DaviesK1997Genomic variation and gene conversion in spinal muscular atrophy: implications for disease process and clinical phenotype. Am. J. Hum. Genet. 614050
  56. 56. ChangJ. GHsieh-liH. MJongY. JWangN. MTsaiC. Hand LiH2001Treatment of spinal muscular atrophy by sodium butyrate. Proc. Natl. Acad. Sci. U. S. A. 9898089813
  57. 57. SumnerC. JHuynhT. NMarkowitzJ. APerhacJ. SHillBCoovertD. DSchusslerKChenXJareckiJBurghesA. Het al2003Valproic acid increases SMN levels in spinal muscular atrophy patient cells. Ann. Neurol. 54647654
  58. 58. BrichtaLHofmannYHahnenESiebzehnrublF. ARaschkeHBlumckeIEyupogluI. Yand WirthB2003Valproic acid increases the SMN2 protein level: a well-known drug as a potential therapy for spinal muscular atrophy. Hum. Mol. Genet. 1224812489
  59. 59. TsaiL. KTsaiM. STingC. Hand LiH2008Multiple therapeutic effects of valproic acid in spinal muscular atrophy model mice. J. Mol. Med. (Berl) 8612431254
  60. 60. AndreassiCAngelozziCTizianoF. DVitaliTDe VincenziEBoninsegnaAVillanovaMBertiniEPiniANeriGet al2004Phenylbutyrate increases SMN expression in vitro: relevance for treatment of spinal muscular atrophy. Eur. J. Hum. Genet. 125965
  61. 61. SwobodaK. JScottC. BReynaS. PPriorT. WLaSalle, B., Sorenson, S.L., Wood, J., Acsadi, G., Crawford, T.O., Kissel, J.T. et al. 2009Phase II open label study of valproic acid in spinal muscular atrophy. PLoS One 4:e5268.
  62. 62. GarbesLRiesslandMHolkerIHellerRHaukeJTrankleCCorasRBlumckeIHahnenEand WirthB2009LBH589 induces up to 10-fold SMN protein levels by several independent mechanisms and is effective even in cells from SMA patients non-responsive to valproate. Hum. Mol. Genet. 1836453658
  63. 63. MercuriEBertiniEMessinaSSolariADAmicoAAngelozziCBattiniRBerardinelliABoffiPBrunoC. et al2007Randomized, double-blind, placebo-controlled trial of phenylbutyrate in spinal muscular atrophy. Neurology 685155
  64. 64. AvilaA. MBurnettB. GTayeA. AGabanellaFKnightM. AHartensteinPCizmanZDi Prospero, N.A., Pellizzoni, L., Fischbeck, K.H. et al. 2007Trichostatin A increases SMN expression and survival in a mouse model of spinal muscular atrophy. J. Clin. Invest. 117659671
  65. 65. NarverH. LKongLBurnettB. GChoeD. WBosch-marceMTayeA. AEckhausM. Aand SumnerC. J2008Sustained improvement of spinal muscular atrophy mice treated with trichostatin A plus nutrition. Ann. Neurol. 64465470
  66. 66. RiesslandMAckermannBForsterAJakubikMHaukeJGarbesLFritzscheIMendeYBlumckeIHahnenEet al2010SAHA ameliorates the SMA phenotype in two mouse models for spinal muscular atrophy. Hum. Mol. Genet. 1914921506
  67. 67. PoweC. EAllenMPuopoloK. MMerewoodAWordenSJohnsonL. CFleischmanAand WeltC. K2010Recombinant human prolactin for the treatment of lactation insufficiency. Clin. Endocrinol. (Oxf) 73645653
  68. 68. FarooqFMolinaF. AHadwenJMacKenzie, D., Witherspoon, L., Osmond, M., Holcik, M., and MacKenzie, A. 2011Prolactin increases SMN expression and survival in a mouse model of severe spinal muscular atrophy via the STAT5 pathway. J. Clin. Invest. 12130423050
  69. 69. AndreassiCJareckiJZhouJCoovertD. DMonaniU. RChenXWhitneyMPollokBZhangMAndrophyEet al2001Aclarubicin treatment restores SMN levels to cells derived from type I spinal muscular atrophy patients. Hum. Mol. Genet. 1028412849
  70. 70. SinghN. NSinghR. Nand AndrophyE. J2007Modulating role of RNA structure in alternative splicing of a critical exon in the spinal muscular atrophy genes. Nucleic Acids Res. 35371389
  71. 71. SinghN. NShishimorovaMCaoL. CGangwaniLand SinghR. N2009A short antisense oligonucleotide masking a unique intronic motif prevents skipping of a critical exon in spinal muscular atrophy. RNA Biol. 6341350
  72. 72. MacKenzieA. 2012Sense in antisense therapy for spinal muscular atrophy. N. Engl. J. Med. 366761763
  73. 73. SinghJSalciusMLiuS. WStakerB. LMishraRThurmondJMichaudGMattoonD. RPrintenJChristensenJet al2008DcpS as a therapeutic target for spinal muscular atrophy. ACS Chem. Biol. 3711722
  74. 74. FarooqFBalabanianSLiuXHolcikMand MacKenzie, A. 200938Mitogen-activated protein kinase stabilizes SMN mRNA through RNA binding protein HuR. Hum. Mol. Genet. 18:4035-4045.
  75. 75. HeierC. Rand DiDonato, C.J. 2009Translational readthrough by the aminoglycoside geneticin (G418) modulates SMN stability in vitro and improves motor function in SMA mice in vivo. Hum. Mol. Genet. 1813101322
  76. 76. MattisV. BRaiRWangJChangC. WCoadyTand LorsonC. L2006Novel aminoglycosides increase SMN levels in spinal muscular atrophy fibroblasts. Hum. Genet. 120589601
  77. 77. WolstencroftE. CMattisVBajerA. AYoungP. Jand LorsonC. L2005ANon-sequencespecific requirement for SMN protein activity: the role of aminoglycosides in inducing elevated SMN protein levels. Hum. Mol. Genet. 1411991210
  78. 78. KwonD. YMotleyW. WFischbeckK. Hand BurnettB. G2011Increasing expression and decreasing degradation of SMN ameliorate the spinal muscular atrophy phenotype in mice. Hum. Mol. Genet. 2036673677
  79. 79. FoustK. DWangXMcgovernV. LBraunLBevanA. KHaidetA. MLeT. TMoralesP. RRichM. MBurghesA. Het al2010Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nat. Biotechnol. 28271274
  80. 80. PassiniM. ABuJRoskelleyE. MRichardsA. MSardiS. PORiordanC. RKlingerK. WShihabuddinL. Sand ChengS. H2010Cns-targetedgene therapy improves survival and motor function in a mouse model of spinal muscular atrophy. J. Clin. Invest. 12012531264
  81. 81. ValoriC. FNingKWylesMMeadR. JGriersonA. JShawP. Jand AzzouzM2010Systemic delivery of scAAV9 expressing SMN prolongs survival in a model of spinal muscular atrophy. Sci. Transl. Med. 2:35ra42.
  82. 82. DominguezEMaraisTChatauretNBenkhelifa-ziyyatSDuqueSRavassardPCarcenacRAstordSDe MouraA. PVoitTet al2011Intravenous scAAV9 delivery of a codon-optimized SMN1 sequence rescues SMA mice. Hum. Mol. Genet. 20681693
  83. 83. MacKenzieA. 2010Genetic therapy for spinal muscular atrophy. Nat. Biotechnol. 28235237
  84. 84. CortiSNizzardoMNardiniMDonadoniCSalaniSDel Bo, R., Papadimitriou, D., Locatelli, F., Mezzina, N., Gianni, F. et al. 2009Motoneuron transplantation rescues the phenotype of SMARD1 (spinal muscular atrophy with respiratory distress type 1). J. Neurosci. 291176111771
  85. 85. CortiSNizzardoMNardiniMDonadoniCSalaniSRonchiDSimoneCFalconeMPapadimitriouDLocatelliFet al2010Embryonic stem cell-derived neural stem cells improve spinal muscular atrophy phenotype in mice. Brain 133465481
  86. 86. DimosJ. TRodolfaK. TNiakanK. KWeisenthalL. MMitsumotoHChungWCroftG. FSaphierGLeibelRGolandRet al2008Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 32112181221
  87. 87. EbertA. DYuJRoseF. FJr, Mattis, V.B., Lorson, C.L., Thomson, J.A., and Svendsen, C.N. 2009Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature 457277280
  88. 88. BowermanMBeauvaisAAndersonC. Land KotharyR2010Rho-kinase inactivation prolongs survival of an intermediate SMA mouse model. Hum. Mol. Genet. 1914681478

Written By

Faraz Tariq Farooq, Martin Holcik and Alex MacKenzie

Submitted: 05 June 2012 Published: 15 May 2013