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:

Silencing mutant SOD1 using RNAi protects against neurodegeneration and extends survival in an ALS model

Abstract

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease resulting in the selective death of motor neurons in the brain and spinal cord1. Some familial cases of ALS are caused by dominant mutations in the gene encoding superoxide dismutase (SOD1)2,3,4. The emergence of interfering RNA (RNAi) for specific gene silencing could be therapeutically beneficial for the treatment of such dominantly inherited diseases5,6,7. We generated a lentiviral vector to mediate expression of RNAi molecules specifically targeting the human SOD1 gene (SOD1). Injection of this vector into various muscle groups of mice engineered to overexpress a mutated form of human SOD1 (SOD1G93A) resulted in an efficient and specific reduction of SOD1 expression and improved survival of vulnerable motor neurons in the brainstem and spinal cord. Furthermore, SOD1 silencing mediated an improved motor performance in these animals, resulting in a considerable delay in the onset of ALS symptoms by more than 100% and an extension in survival by nearly 80% of their normal life span. These data are the first to show a substantial extension of survival in an animal model of a fatal, dominantly inherited neurodegenerative condition using RNAi and provide the highest therapeutic efficacy observed in this field to date.

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

Access options

Buy this article

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

Figure 1: Silencing of SOD1 expression in vitro using EIAV-mediated expression of shRNAs.
Figure 2: Silencing of mutant SOD1 expression in vivo delays onset of ALS and extends survival of SOD1G93A mice.
Figure 3: Silencing of SOD1 expression using shRNA mediates improved motor performance in SOD1G93A transgenic mice.
Figure 4: Motor neuron survival is improved by RNAi-mediated silencing of SOD1.

Similar content being viewed by others

References

  1. Mulder, D.W. Clinical limits of amyotrophic lateral sclerosis. Adv. Neurol. 36, 15–22 (1982).

    CAS  PubMed  Google Scholar 

  2. Rosen, D.R. et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 362, 59–62 (1993).

    Article  CAS  Google Scholar 

  3. Deng, H.X. et al. Amyotrophic lateral sclerosis and structural defects in Cu,Zn superoxide dismutase. Science 261, 1047–1051 (1993).

    Article  CAS  Google Scholar 

  4. Gurney, M.E. et al. Motor neuron degeneration in mice that express a human Cu,Zn superoxide dismutase mutation. Science 264, 1772–1775 (1994).

    Article  CAS  Google Scholar 

  5. Maxwell, M.M., Pasinelli, P., Kazantsev, A.G. & Brown, R.H., Jr. RNA interference-mediated silencing of mutant superoxide dismutase rescues cyclosporin A-induced death in cultured neuroblastoma cells. Proc. Natl. Acad. Sci. USA 101, 3178–3183 (2004).

    Article  CAS  Google Scholar 

  6. Ding, H. et al. Selective silencing by RNAi of a dominant allele that causes amyotrophic lateral sclerosis. Aging Cell 2, 209–217 (2003).

    Article  CAS  Google Scholar 

  7. Xia, H. et al. RNAi suppresses polyglutamine-induced neurodegeneration in a model of spinocerebellar ataxia. Nat. Med. 8, 816–820 (2004).

    Article  Google Scholar 

  8. Xia, H., Mao, Q., Paulson, H.L. & Davidson, B.L. siRNA-mediated gene silencing in vitro and in vivo. Nat. Biotechnol. 20, 1006–1010 (2002).

    Article  CAS  Google Scholar 

  9. Hommel, J.D., Sears, R.M., Georgescu, D., Simmons, D.L. & DiLeone, R.J. Local gene knockdown in the brain using viral-mediated RNA interference. Nat. Med. 9, 1539–1544 (2003).

    Article  CAS  Google Scholar 

  10. Mazarakis, N.D. et al. Rabies virus glycoprotein pseudotyping of lentiviral vectors enables retrograde axonal transport and access to the nervous system after peripheral delivery. Hum. Mol. Genet. 10, 2109–2121 (2001).

    Article  CAS  Google Scholar 

  11. Wong, L.F. et al. Transduction patterns of pseudotyped lentiviral vectors in the nervous system. Mol. Ther. 9, 101–111 (2004).

    Article  CAS  Google Scholar 

  12. Azzouz, M. et al. VEGF delivery with retrogradely transported lentivector prolongs survival in a mouse ALS model. Nature 429, 413–417 (2004).

    Article  CAS  Google Scholar 

  13. Wong, P.C. et al. An adverse property of a familial ALS-linked SOD1 mutation causes motor neuron disease characterized by vacuolar degeneration of mitochondria. Neuron 14, 1105–1116 (1995).

    Article  CAS  Google Scholar 

  14. Bruijn, L.I. et al. ALS-linked SOD1 mutant G85R mediates damage to astrocytes and promotes rapidly progressive disease with SOD1-containing inclusions. Neuron 18, 327–338 (1997).

    Article  CAS  Google Scholar 

  15. Cleveland, D.W. & Rothstein, J.D. From Charcot to Lou Gehrig: deciphering selective motor neuron death in ALS. Nat. Rev. Neurosci. 2, 806–819 (2001).

    Article  CAS  Google Scholar 

  16. Brummelkamp, T.R., Bernards, R. & Agami, R. A system for stable expression of short interfering RNAs in mammalian cells. Science 296, 550–553 (2002).

    Article  CAS  Google Scholar 

  17. Katsuno, M. et al. Testosterone reduction prevents phenotypic expression in a transgenic mouse model of spinal and bulbar muscular atrophy. Neuron 35, 843–854 (2002).

    Article  CAS  Google Scholar 

  18. Katsuno, M. et al. Leuprorelin rescues polyglutamine-dependent phenotypes in a transgenic mouse model of spinal and bulbar muscular atrophy. Nat. Med. 9, 768–773 (2003).

    Article  CAS  Google Scholar 

  19. Adachi, H. et al. Transgenic mice with an expanded CAG repeat controlled by the human AR promoter show polyglutamine nuclear inclusions and neuronal dysfunction without neuronal cell death. Hum Mol. Genet. 10, 1039–1048 (2001).

    Article  CAS  Google Scholar 

  20. 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  Google Scholar 

  21. Clement, A.M. et al. Wild-type nonneuronal cells extend survival of SOD1 mutant motor neurons in ALS mice. Science 302, 113–117 (2003).

    Article  CAS  Google Scholar 

  22. Mitrophanous, K. et al. Stable gene transfer to the nervous system using a non-primate lentiviral vector. Gene Ther. 6, 1808–1818 (1999).

    Article  CAS  Google Scholar 

  23. Soneoka, Y. et al. A transient three-plasmid expression system for the production of high titer retroviral vectors. Nucleic Acids Res. 23, 628–633 (1995).

    Article  CAS  Google Scholar 

  24. Martin-Rendon, E., White, L.J., Olsen, A., Mitrophanous, K.A. & Mazarakis, N.D. New Methods to Titrate EIAV-Based Lentiviral Vectors. Mol. Ther. 5, 566–570 (2002).

    Article  CAS  Google Scholar 

  25. Abercrombie, M. Estimation of nuclear population from microtome sections. Anatomical Record 94, 239–247 (1948).

    Article  Google Scholar 

Download references

Acknowledgements

Many thanks to the Virology and Neurobiology teams at Oxford BioMedica for assistance with design and preparation of viral vectors. L.G.B. is a Wellcome Trust Prize student. L.G. is the Graham Watts Senior Research Fellow funded by the Brain Research Trust. This work was funded by Oxford Biomedica Ltd.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to G Scott Ralph or Mimoun Azzouz.

Ethics declarations

Competing interests

The authors GSR, PAR, DMD, JMM, MAL, LFW, SMK, KAM, NDM, and MA are share holders in Oxford Biomedica (UK) Ltd and as such have competing financial interests in the publication of this work.

Supplementary information

Supplementary Fig. 1

Analysis of transgene copy number in EIAV-injected SOD1G93A mice used for this study. (PDF 60 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ralph, G., Radcliffe, P., Day, D. et al. Silencing mutant SOD1 using RNAi protects against neurodegeneration and extends survival in an ALS model. Nat Med 11, 429–433 (2005). https://doi.org/10.1038/nm1205

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm1205

This article is cited by

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