Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Peripheral SMN restoration is essential for long-term rescue of a severe spinal muscular atrophy mouse model

Abstract

Spinal muscular atrophy (SMA) is a motor neuron disease and the leading genetic cause of infant mortality; it results from loss-of-function mutations in the survival motor neuron 1 (SMN1) gene1. Humans have a paralogue, SMN2, whose exon 7 is predominantly skipped2, but the limited amount of functional, full-length SMN protein expressed from SMN2 cannot fully compensate for a lack of SMN1. SMN is important for the biogenesis of spliceosomal small nuclear ribonucleoprotein particles3, but downstream splicing targets involved in pathogenesis remain elusive. There is no effective SMA treatment, but SMN restoration in spinal cord motor neurons is thought to be necessary and sufficient4. Non-central nervous system (CNS) pathologies, including cardiovascular defects, were recently reported in severe SMA mouse models and patients5,6,7,8, reflecting autonomic dysfunction or direct effects in cardiac tissues. Here we compared systemic versus CNS restoration of SMN in a severe mouse model9,10. We used an antisense oligonucleotide (ASO), ASO-10-27, that effectively corrects SMN2 splicing and restores SMN expression in motor neurons after intracerebroventricular injection11,12. Systemic administration of ASO-10-27 to neonates robustly rescued severe SMA mice, much more effectively than intracerebroventricular administration; subcutaneous injections extended the median lifespan by 25 fold. Furthermore, neonatal SMA mice had decreased hepatic Igfals expression, leading to a pronounced reduction in circulating insulin-like growth factor 1 (IGF1), and ASO-10-27 treatment restored IGF1 to normal levels. These results suggest that the liver is important in SMA pathogenesis, underscoring the importance of SMN in peripheral tissues, and demonstrate the efficacy of a promising drug candidate.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Systemic versus ICV ASO-10-27 injections in SMA mice.
Figure 2: SMN2 splicing and protein expression in mouse tissues after SC injection of ASO-10-27.
Figure 3: Evaluation of affected tissues and motor function.
Figure 4: The IGF1 system is disrupted in SMA mice.

Similar content being viewed by others

References

  1. Lefebvre, S. et al. Identification and characterization of a spinal muscular atrophy-determining gene. Cell 80, 155–165 (1995)

    Article  CAS  Google Scholar 

  2. Lorson, C. L., Rindt, H. & Shababi, M. Spinal muscular atrophy: mechanisms and therapeutic strategies. Hum. Mol. Genet. 19, R111–R118 (2010)

    Article  CAS  Google Scholar 

  3. Burghes, A. H. & Beattie, C. E. Spinal muscular atrophy: why do low levels of survival motor neuron protein make motor neurons sick? Nature Rev. Neurosci. 10, 597–609 (2009)

    Article  CAS  Google Scholar 

  4. Gavrilina, T. O. et al. Neuronal SMN expression corrects spinal muscular atrophy in severe SMA mice while muscle-specific SMN expression has no phenotypic effect. Hum. Mol. Genet. 17, 1063–1075 (2008)

    Article  CAS  Google Scholar 

  5. Rudnik-Schoneborn, S. et al. Congenital heart disease is a feature of severe infantile spinal muscular atrophy. J. Med. Genet. 45, 635–638 (2008)

    Article  CAS  Google Scholar 

  6. Bevan, A. K. et al. Early heart failure in the SMNΔ7 model of spinal muscular atrophy and correction by postnatal scAAV9-SMN delivery. Hum. Mol. Genet. 19, 3895–3905 (2010)

    Article  CAS  Google Scholar 

  7. Heier, C. R., Satta, R., Lutz, C. & DiDonato, C. J. Arrhythmia and cardiac defects are a feature of spinal muscular atrophy model mice. Hum. Mol. Genet. 19, 3906–3918 (2010)

    Article  CAS  Google Scholar 

  8. Shababi, M. et al. Cardiac defects contribute to the pathology of spinal muscular atrophy models. Hum. Mol. Genet. 19, 4059–4071 (2010)

    Article  CAS  Google Scholar 

  9. Gogliotti, R. G., Hammond, S. M., Lutz, C. & Didonato, C. J. Molecular and phenotypic reassessment of an infrequently used mouse model for spinal muscular atrophy. Biochem. Biophys. Res. Commun. 391, 517–522 (2010)

    Article  CAS  Google Scholar 

  10. Riessland, M. et al. SAHA ameliorates the SMA phenotype in two mouse models for spinal muscular atrophy. Hum. Mol. Genet. 19, 1492–1506 (2010)

    Article  CAS  Google Scholar 

  11. Hua, Y. et al. Antisense correction of SMN2 splicing in the CNS rescues necrosis in a type III SMA mouse model. Genes Dev. 24, 1634–1644 (2010)

    Article  CAS  Google Scholar 

  12. Passini, M. A. et al. Antisense oligonucleotides delivered to the mouse CNS ameliorate symptoms of severe spinal muscular atrophy. Sci. Transl. Med. 3, 72ra18 (2011)

    Article  Google Scholar 

  13. Hua, Y., Vickers, T. A., Okunola, H. L., Bennett, C. F. & Krainer, A. R. Antisense masking of an hnRNP A1/A2 intronic splicing silencer corrects SMN2 splicing in transgenic mice. Am. J. Hum. Genet. 82, 834–848 (2008)

    Article  CAS  Google Scholar 

  14. Passini, M. A. et al. CNS-targeted gene therapy improves survival and motor function in a mouse model of spinal muscular atrophy. J. Clin. Invest. 120, 1253–1264 (2010)

    Article  CAS  Google Scholar 

  15. Foust, K. D. et al. Rescue of the spinal muscular atrophy phenotype in a mouse model by early postnatal delivery of SMN. Nature Biotechnol. 28, 271–274 (2010)

    Article  CAS  Google Scholar 

  16. Dominguez, E. et al. Intravenous scAAV9 delivery of a codon-optimized SMN1 sequence rescues SMA mice. Hum. Mol. Genet. 20, 681–693 (2011)

    Article  CAS  Google Scholar 

  17. Ek, C. J., Habgood, M. D., Dziegielewska, K. M. & Saunders, N. R. Structural characteristics and barrier properties of the choroid plexuses in developing brain of the opossum (Monodelphis domestica). J. Comp. Neurol. 460, 451–464 (2003)

    Article  Google Scholar 

  18. Steele, A. D., Jackson, W. S., King, O. D. & Lindquist, S. The power of automated high-resolution behavior analysis revealed by its application to mouse models of Huntington’s and prion diseases. Proc. Natl Acad. Sci. USA 104, 1983–1988 (2007)

    Article  CAS  ADS  Google Scholar 

  19. Park, G. H., Kariya, S. & Monani, U. R. Spinal muscular atrophy: new and emerging insights from model mice. Curr. Neurol. Neurosci. Rep. 10, 108–117 (2010)

    Article  Google Scholar 

  20. Wu, Y., Sun, H., Yakar, S. & LeRoith, D. Elevated levels of insulin-like growth factor (IGF)-I in serum rescue the severe growth retardation of IGF-I null mice. Endocrinology 150, 4395–4403 (2009)

    Article  CAS  Google Scholar 

  21. Kaspar, B. K., Llado, J., Sherkat, N., Rothstein, J. D. & Gage, F. H. Retrograde viral delivery of IGF-1 prolongs survival in a mouse ALS model. Science 301, 839–842 (2003)

    Article  CAS  ADS  Google Scholar 

  22. Colao, A. The GH–IGF-I axis and the cardiovascular system: clinical implications. Clin. Endocrinol. 69, 347–358 (2008)

    Article  CAS  Google Scholar 

  23. Domené, H. M. et al. Human acid-labile subunit deficiency: clinical, endocrine and metabolic consequences. Horm. Res. 72, 129–141 (2009)

    Article  Google Scholar 

  24. Kenyon, C. The plasticity of aging: insights from long-lived mutants. Cell 120, 449–460 (2005)

    Article  CAS  Google Scholar 

  25. Shababi, M., Glascock, J. & Lorson, C. L. Combination of SMN trans-splicing and a neurotrophic factor increases the life span and body mass in a severe model of spinal muscular atrophy. Hum. Gene Ther. 22, 135–144 (2011)

    Article  CAS  Google Scholar 

  26. Bosch-Marce, M. et al. Increased IGF-1 in muscle modulates the phenotype of severe SMA mice. Hum. Mol. Genet. 20, 1844–1853 (2011)

    Article  CAS  Google Scholar 

  27. Trejo, J. L., Carro, E., Garcia-Galloway, E. & Torres-Aleman, I. Role of insulin-like growth factor I signaling in neurodegenerative diseases. J. Mol. Med. 82, 156–162 (2004)

    Article  CAS  Google Scholar 

  28. Powell-Braxton, L. et al. IGF-I is required for normal embryonic growth in mice. Genes Dev. 7, 2609–2617 (1993)

    Article  CAS  Google Scholar 

  29. Millino, C. et al. Different atrophy-hypertrophy transcription pathways in muscles affected by severe and mild spinal muscular atrophy. BMC Med. 7, 14 (2009)

    Article  Google Scholar 

  30. Sorenson, E. J. et al. Subcutaneous IGF-1 is not beneficial in 2-year ALS trial. Neurology 71, 1770–1775 (2008)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge support from the Muscular Dystrophy Association, the National Institute of General Medical Sciences and St. Giles Foundation. We thank J. Bu and M. Passini for protocols and advice on NMJ staining, and S. Hearn for assistance with microscope imaging.

Author information

Authors and Affiliations

Authors

Contributions

Y.H., A.R.K. and C.F.B. designed the study and wrote the paper. Y.H., K.S., F.R., G. Hung and G. Horev carried out the experiments and analysed the data. All authors read the manuscript.

Corresponding author

Correspondence to Adrian R. Krainer.

Ethics declarations

Competing interests

F.R., G. Hung, and C.F.B. may materially benefit either directly or indirectly through stock options. Y.H., K.S. and A.R.K., along with their employer, could materially benefit if a therapeutic for SMA results from this work.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-3, Supplementary Figures 1-15 with legends and an additional reference. (PDF 1843 kb)

Supplementary Movie 1

This movie shows two P7 pups, both untreated. The smaller one is an SMA mouse and the other one is its heterozygous littermate. (MOV 18216 kb)

Supplementary Movie 2

This movie shows five 3-week-old newly weaned mice. Three representative SMA mice that had been treated with subcutaneous ASO administration (SC80) between P0-P3 have shorter tails, and two untreated heterozygous mice have normal tails. (MOV 21283 kb)

Supplementary Movie 3

This movie shows two 3-month-old mice on a Rotarod test with a four-phase acceleration profile. The one that passed the test and has a shorter tail is an SMA mouse from group SC160, and the other mouse is a heterozygote. 36% of SC160 rescued SMA mice passed this test; 33% of heterozygous mice did not pass the test. (MOV 8152 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hua, Y., Sahashi, K., Rigo, F. et al. Peripheral SMN restoration is essential for long-term rescue of a severe spinal muscular atrophy mouse model. Nature 478, 123–126 (2011). https://doi.org/10.1038/nature10485

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature10485

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing