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.

  • Review Article
  • Published:

Targeting RNA to treat neuromuscular disease

Key Points

  • Neuromuscular disorders comprise a heterogeneous group of clinical conditions that primarily affect one or more components of the neuromuscular unit — typically skeletal muscle — but are often also multisystemic. The considerable clinical impact of these diseases is exemplified by the muscular dystrophies and by spinal muscular atrophy (SMA), for which the development for novel molecular therapies is both urgent and challenging.

  • Recent advances in RNA biology have accelerated the progress of a new generation of molecular therapies based on RNA. Single- or double-stranded nucleic acid agents and small-molecule agents are being developed as novel therapies to target the mutant mRNAs that are involved in neuromuscular disease. These approaches modulate pre-mRNA processing or inhibit the deleterious effects of toxic RNAs.

  • In the case of Duchenne muscular dystrophy (DMD), exon-skipping therapies to restore a viable open reading frame in the DMD gene are well advanced. Two recent systemic delivery clinical trials have reported encouraging data on the restoration of dystrophin protein expression, and second-generation compounds are in development. An alternative approach to stimulate the readthrough of premature stop codons, which is applicable to a subset of patients with DMD, is also in development.

  • Myotonic dystrophy arises as a result of an expanded microsatellite repeat mutation that leads to a gain-of-function toxic mRNA, which ultimately causes muscle degeneration and multisystem dysfunction. Various approaches to correct the deleterious toxic effects of toxic RNA using small-molecule-based, oligonucleotide-based or RNA interference-based methods to inhibit or degrade the toxic RNA are being investigated.

  • A viable therapeutic strategy for SMA, which arises as a result of the loss-of-function of the survival of motor neuron 1 (SMN1) gene, is to restore the splicing of exon 7 in the related SMN2 gene, which has the potential to fully compensate for the loss of the SMN1 protein. Recent studies using oligonucleotide-mediated exon-inclusion methods appear to be highly promising.

  • Although the development of RNA-based therapies for neuromuscular disease remains challenging, recent progress in this field is encouraging. However, major barriers remain the poor in vivo delivery of most RNA therapeutic agents and the regulatory hurdles that are associated with the development of novel personalized medicines.

Abstract

The development of effective therapies for neuromuscular disorders such as Duchenne muscular dystrophy (DMD) is hampered by considerable challenges: skeletal muscle is the most abundant tissue in the body, and many neuromuscular disorders are multisystemic conditions. However, despite these barriers there has recently been substantial progress in the search for novel treatments. In particular, the use of antisense oligonucleotides, which are designed to target RNA and modulate pre-mRNA splicing to restore functional protein isoforms or directly inhibit the toxic effects of pathogenic RNAs, offers great promise and these approaches are now being tested in the clinic. Here, we review recent advances in the development of such antisense oligonucleotides and other promising novel approaches, including the induction of readthrough nonsense mutations.

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: DMD gene: effects of mutations and exon skipping.
Figure 2: mdx mice and effects of antisense oligonucleotide exon skipping.
Figure 3: Clinical trials for antisense oligonucleotide exon 51 skipping in DMD.
Figure 4: Myotonic dystrophy type 1: triplet repeat expansion and targeting expanded DMPK alleles.
Figure 5: SMA: SMN1 splicing and SMN2 exon 7 inclusion using antisense oligonucleotides.

Similar content being viewed by others

References

  1. Davies, K. E. & Nowak, K. J. Molecular mechanisms of muscular dystrophies: old and new players. Nature Rev. Mol. Cell Biol. 7, 762–773 (2006).

    Article  CAS  Google Scholar 

  2. Bushby, K. et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care. Lancet Neurol. 9, 177–189 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Bushby, K. et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management. Lancet Neurol. 9, 77–93 (2010).

    Article  PubMed  Google Scholar 

  4. Muntoni, F., Torelli, S. & Ferlini, A. Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol. 2, 731–740 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Manzur, A. Y., Kinali, M. & Muntoni, F. Update on the management of Duchenne muscular dystrophy. Arch. Dis. Child. 93, 986–990 (2008).

    Article  CAS  PubMed  Google Scholar 

  6. Emery, A. & Muntoni, F. (eds) Duchenne Muscular Dystrophy (Oxford University Press, New York, 2003).

    Google Scholar 

  7. Hoffman, E. P., Brown, R. H., Jr & Kunkel, L. M. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 51, 919–928 (1987).

    Article  CAS  PubMed  Google Scholar 

  8. Hoffman, E. P., Monaco, A. P., Feener, C. C. & Kunkel, L. M. Conservation of the Duchenne muscular dystrophy gene in mice and humans. Science 238, 347–350 (1987).

    Article  CAS  PubMed  Google Scholar 

  9. Osborne, R. J. & Thornton, C. A. RNA-dominant diseases. Hum. Mol. Genet. 2006 (Suppl. 2), R162–R169 (2006).

    Article  CAS  Google Scholar 

  10. Ranum, L. P. & Cooper, T. A. RNA-mediated neuromuscular disorders. Annu. Rev. Neurosci. 29, 259–277 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Wheeler, T. M. & Thornton, C. A. Myotonic dystrophy: RNA-mediated muscle disease. Curr. Opin. Neurol. 20, 572–576 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. 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 

  13. Talbot, K. & Davies, K. E. Spinal muscular atrophy. Semin. Neurol. 21, 189–197 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Zhang, Z. et al. SMN deficiency causes tissue-specific perturbations in the repertoire of snRNAs and widespread defects in splicing. Cell 133, 585–600 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stephenson, M. L. & Zamecnik, P. C. Inhibition of Rous sarcoma viral RNA translation by a specific oligodeoxyribonucleotide. Proc. Natl Acad. Sci. USA 75, 285–288 (1978).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Marwick, C. First “antisense” drug will treat CMV retinitis. JAMA 280, 871 (1998).

    Article  CAS  PubMed  Google Scholar 

  17. Kurreck, J. Antisense technologies. Improvement through novel chemical modifications. Eur. J. Biochem. 270, 1628–1644 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Lebedeva, I. & Stein, C. A. Antisense oligonucleotides: promise and reality. Annu. Rev. Pharmacol. Toxicol. 41, 403–419 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Opalinska, J. B. & Gewirtz, A. M. Nucleic-acid therapeutics: basic principles and recent applications. Nature Rev. Drug Discov. 1, 503–514 (2002).

    Article  CAS  Google Scholar 

  20. Bennett, C. F. & Swayze, E. E. RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu. Rev. Pharmacol. Toxicol. 50, 259–293 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Dominski, Z. & Kole, R. Restoration of correct splicing in thalassemic pre-mRNA by antisense oligonucleotides. Proc. Natl Acad. Sci. USA 90, 8673–8677 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Ahmad, A., Brinson, M., Hodges, B. L., Chamberlain, J. S. & Amalfitano, A. Mdx mice inducibly expressing dystrophin provide insights into the potential of gene therapy for duchenne muscular dystrophy. Hum. Mol. Genet. 9, 2507–2515 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Ghahramani Seno, M. M. et al. RNAi-mediated knockdown of dystrophin expression in adult mice does not lead to overt muscular dystrophy pathology. Hum. Mol. Genet. 17, 2622–2632 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Bushby, K. M. & Gardner-Medwin, D. The clinical, genetic and dystrophin characteristics of Becker muscular dystrophy. I. Natural history. J. Neurol. 240, 98–104 (1993).

    Article  CAS  PubMed  Google Scholar 

  25. Khurana, T. S. & Davies, K. E. Pharmacological strategies for muscular dystrophy. Nature Rev. Drug Discov. 2, 379–390 (2003).

    Article  CAS  Google Scholar 

  26. Chamberlain, J. S. Stem-cell biology: a move in the right direction. Nature 444, 552–553 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Wang, Z. et al. Sustained AAV-mediated dystrophin expression in a canine model of Duchenne muscular dystrophy with a brief course of immunosuppression. Mol. Ther. 15, 1160–1166 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Mendell, J. R. et al. Dystrophin immunity in Duchenne's muscular dystrophy. N. Engl. J. Med. 363, 1429–1437 (2010). This study reported the detection of dystrophin-specific T cells targeting truncated dystrophin epitopes following the delivery of a functional dystrophin transgene in patients with DMD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Berry, S. E., Liu, J., Chaney, E. J. & Kaufman, S. J. Multipotential mesoangioblast stem cell therapy in the mdx/utrn−/− mouse model for Duchenne muscular dystrophy. Regen. Med. 2, 275–288 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Galvez, B. G. et al. Complete repair of dystrophic skeletal muscle by mesoangioblasts with enhanced migration ability. J. Cell Biol. 174, 231–243 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Dellavalle, A. et al. Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nature Cell Biol. 9, 255–267 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. van Deutekom, J. C. & van Ommen, G. J. Advances in Duchenne muscular dystrophy gene therapy. Nature Rev. Genet. 4, 774–783 (2003).

    Article  CAS  PubMed  Google Scholar 

  33. Grange, R. W., Gainer, T. G., Marschner, K. M., Talmadge, R. J. & Stull, J. T. Fast-twitch skeletal muscles of dystrophic mouse pups are resistant to injury from acute mechanical stress. Am. J. Physiol. Cell Physiol. 283, C1090–C1101 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Dunckley, M. G., Manoharan, M., Villiet, P., Eperon, I. C. & Dickson, G. Modification of splicing in the dystrophin gene in cultured Mdx muscle cells by antisense oligoribonucleotides. Hum. Mol. Genet. 7, 1083–1090 (1998).

    Article  CAS  PubMed  Google Scholar 

  35. Pramono, Z. A. et al. Induction of exon skipping of the dystrophin transcript in lymphoblastoid cells by transfecting an antisense oligodeoxynucleotide complementary to an exon recognition sequence. Biochem. Biophys. Res. Commun. 226, 445–449 (1996).

    Article  CAS  PubMed  Google Scholar 

  36. Takeshima, Y., Nishio, H., Sakamoto, H., Nakamura, H. & Matsuo, M. Modulation of in vitro splicing of the upstream intron by modifying an intra-exon sequence which is deleted from the dystrophin gene in dystrophin Kobe. J. Clin. Invest. 95, 515–520 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Aartsma-Rus, A. et al. Targeted exon skipping as a potential gene correction therapy for Duchenne muscular dystrophy. Neuromuscul. Disord. 12 (Suppl. 1), 71–77 (2002).

    Article  Google Scholar 

  38. Aartsma-Rus, A. et al. Comparative analysis of antisense oligonucleotide analogs for targeted DMD exon 46 skipping in muscle cells. Gene Ther. 11, 1391–1398 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Popplewell, L. J. et al. Comparative analysis of antisense oligonucleotide sequences targeting exon 53 of the human DMD gene: implications for future clinical trials. Neuromuscul. Disord. 20, 102–110 (2010).

    Article  PubMed  Google Scholar 

  40. Popplewell, L. J., Trollet, C., Dickson, G. & Graham, I. R. Design of phosphorodiamidate morpholino oligomers (PMOs) for the induction of exon skipping of the human DMD gene. Mol. Ther. 17, 554–561 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. van Deutekom, J. C. et al. Antisense-induced exon skipping restores dystrophin expression in DMD patient derived muscle cells. Hum. Mol. Genet. 10, 1547–1554 (2001).

    CAS  PubMed  Google Scholar 

  42. Bulfield, G., Siller, W. G., Wight, P. A. & Moore, K. J. X chromosome-linked muscular dystrophy (mdx) in the mouse. Proc. Natl Acad. Sci. USA 81, 1189–1192 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lu, Q. L. et al. Functional amounts of dystrophin produced by skipping the mutated exon in the mdx dystrophic mouse. Nature Med. 9, 1009–1014 (2003). This was the first in vivo study reporting the restoration of functional dystrophin protein following the intramuscular administration of an antisense oligonucleotide in the mdx mouse.

    Article  CAS  PubMed  Google Scholar 

  44. Mann, C. J. et al. Antisense-induced exon skipping and synthesis of dystrophin in the mdx mouse. Proc. Natl Acad. Sci. USA 98, 42–47 (2001).

    Article  CAS  PubMed  Google Scholar 

  45. Mann, C. J., Honeyman, K., McClorey, G., Fletcher, S. & Wilton, S. D. Improved antisense oligonucleotide induced exon skipping in the mdx mouse model of muscular dystrophy. J. Gene Med. 4, 644–654 (2002).

    Article  CAS  PubMed  Google Scholar 

  46. Sazani, P. et al. Systemically delivered antisense oligomers upregulate gene expression in mouse tissues. Nature Biotech. 20, 1228–1233 (2002).

    Article  CAS  Google Scholar 

  47. Lu, Q. L. et al. Systemic delivery of antisense oligoribonucleotide restores dystrophin expression in body-wide skeletal muscles. Proc. Natl Acad. Sci. USA 102, 198–203 (2005). This was the first in vivo study reporting the successful restoration of dystrophin protein in multiple peripheral muscle groups following the systemic intravenous administration of an antisense oligonucleotide in the mdx mouse.

    Article  CAS  PubMed  Google Scholar 

  48. Heemskerk, H. et al. Preclinical PK and PD studies on 2′-O-methyl-phosphorothioate RNA antisense oligonucleotides in the mdx mouse model. Mol. Ther. 18, 1210–1217 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Alter, J. et al. Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology. Nature Med. 12, 175–177 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Yokota, T. et al. Efficacy of systemic morpholino exon-skipping in duchenne dystrophy dogs. Ann. Neurol. 65, 667–676 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Malerba, A. et al. Chronic systemic therapy with low-dose morpholino oligomers ameliorates the pathology and normalizes locomotor behavior in mdx mice. Mol. Ther. 19, 345–354 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. Malerba, A., Thorogood, F. C., Dickson, G. & Graham, I. R. Dosing regimen has a significant impact on the efficiency of morpholino oligomer-induced exon skipping in mdx mice. Hum. Gene Ther. 20, 955–965 (2009).

    Article  CAS  PubMed  Google Scholar 

  53. Wu, B. et al. Dose-dependent restoration of dystrophin expression in cardiac muscle of dystrophic mice by systemically delivered morpholino. Gene Ther. 17, 132–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  54. Fayssoil, A., Nardi, O., Orlikowski, D. & Annane, D. Cardiomyopathy in Duchenne muscular dystrophy: pathogenesis and therapeutics. Heart Fail. Rev. 15, 103–107 (2010).

    Article  PubMed  Google Scholar 

  55. Heemskerk, H. A. et al. In vivo comparison of 2′-O-methyl phosphorothioate and morpholino antisense oligonucleotides for Duchenne muscular dystrophy exon skipping. J. Gene Med. 11, 257–266 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Levin, A. A. A review of the issues in the pharmacokinetics and toxicology of phosphorothioate antisense oligonucleotides. Biochim. Biophys. Acta 1489, 69–84 (1999).

    Article  CAS  PubMed  Google Scholar 

  57. Henry, S. P. et al. Complement activation is responsible for acute toxicities in rhesus monkeys treated with a phosphorothioate oligodeoxynucleotide. Int. Immunopharmacol. 2, 1657–1666 (2002).

    Article  CAS  PubMed  Google Scholar 

  58. Wang, Q. et al. In vitro evaluation of novel antisense oligonucleotides is predictive of in vivo exon skipping activity for Duchenne muscular dystrophy. J. Gene Med. 12, 354–364 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. Crisp, A. et al. Diaphragm rescue alone prevents heart dysfunction in dystrophic mice. Hum. Mol. Genet. 20, 413–421 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Muntoni, F. et al. Brief report: deletion of the dystrophin muscle-promoter region associated with X-linked dilated cardiomyopathy. N. Engl. J. Med. 329, 921–925 (1993).

    Article  CAS  PubMed  Google Scholar 

  61. Townsend, D., Yasuda, S., Li, S., Chamberlain, J. S. & Metzger, J. M. Emergent dilated cardiomyopathy caused by targeted repair of dystrophic skeletal muscle. Mol. Ther. 16, 832–835 (2008).

    Article  CAS  PubMed  Google Scholar 

  62. McClorey, G., Moulton, H. M., Iversen, P. L., Fletcher, S. & Wilton, S. D. Antisense oligonucleotide-induced exon skipping restores dystrophin expression in vitro in a canine model of DMD. Gene Ther. 13, 1373–1381 (2006).

    Article  CAS  PubMed  Google Scholar 

  63. Nelson, M. H. et al. Arginine-rich peptide conjugation to morpholino oligomers: effects on antisense activity and specificity. Bioconjug. Chem. 16, 959–966 (2005).

    Article  CAS  PubMed  Google Scholar 

  64. Abes, S. et al. Peptide-based delivery of nucleic acids: design, mechanism of uptake and applications to splice-correcting oligonucleotides. Biochem. Soc. Trans. 35, 53–55 (2007).

    Article  CAS  PubMed  Google Scholar 

  65. Abes, S. et al. Vectorization of morpholino oligomers by the (R-Ahx-R)4 peptide allows efficient splicing correction in the absence of endosomolytic agents. J. Control Release 116, 304–313 (2006).

    Article  CAS  PubMed  Google Scholar 

  66. Amantana, A. et al. Pharmacokinetics, biodistribution, stability and toxicity of a cell-penetrating peptide-morpholino oligomer conjugate. Bioconjug. Chem. 18, 1325–1331 (2007).

    Article  CAS  PubMed  Google Scholar 

  67. Fletcher, S. et al. Morpholino oligomer-mediated exon skipping averts the onset of dystrophic pathology in the mdx mouse. Mol. Ther. 15, 1587–1592 (2007).

    Article  CAS  PubMed  Google Scholar 

  68. Jearawiriyapaisarn, N. et al. Sustained dystrophin expression induced by peptide-conjugated morpholino oligomers in the muscles of mdx mice. Mol. Ther. 16, 1624–1629 (2008).

    Article  CAS  PubMed  Google Scholar 

  69. Wu, B. et al. Effective rescue of dystrophin improves cardiac function in dystrophin-deficient mice by a modified morpholino oligomer. Proc. Natl Acad. Sci. USA 105, 14814–14819 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Yin, H., Lu, Q. & Wood, M. Effective exon skipping and restoration of dystrophin expression by peptide nucleic acid antisense oligonucleotides in mdx mice. Mol. Ther. 16, 38–45 (2008). The studies in Refs 68–70 reported a considerably enhanced restoration of dystrophin protein following the systemic administration of a peptide-conjugated antisense oligonucleotide.

    Article  CAS  PubMed  Google Scholar 

  71. Moulton, H. M. et al. Cell-penetrating peptide-morpholino conjugates alter pre-mRNA splicing of DMD (Duchenne muscular dystrophy) and inhibit murine coronavirus replication in vivo. Biochem. Soc. Trans. 35, 826–828 (2007).

    Article  CAS  PubMed  Google Scholar 

  72. Moulton, H. M. et al. Peptide-morpholino conjugate: a promising therapeutic for Duchenne muscular dystrophy. Ann. NY Acad. Sci. 1175, 55–60 (2009).

    Article  CAS  PubMed  Google Scholar 

  73. Samoylova, T. I. & Smith, B. F. Elucidation of muscle-binding peptides by phage display screening. Muscle Nerve 22, 460–466 (1999).

    Article  CAS  PubMed  Google Scholar 

  74. Yin, H., Moulton, H., Betts, C. & Wood, M. CPP-directed oligonucleotide exon skipping in animal models of Duchenne muscular dystrophy. Methods Mol. Biol. 683, 321–338 (2011).

    Article  CAS  PubMed  Google Scholar 

  75. Yin, H. et al. Functional rescue of dystrophin-deficient mdx mice by a chimeric peptide-PMO. Mol. Ther. 18, 1822–1829 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Yin, H. et al. A fusion peptide directs enhanced systemic dystrophin exon skipping and functional restoration in dystrophin-deficient mdx mice. Hum. Mol. Genet. 18, 4405–4414 (2009).

    Article  CAS  PubMed  Google Scholar 

  77. Yin, H. et al. Pip5 transduction peptides direct high efficiency oligonucleotide-mediated dystrophin exon skipping in heart and phenotypic correction in mdx mice. Mol. Ther. 19, 1295–1303 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Ivanova, G. D. et al. Improved cell-penetrating peptide–PNA conjugates for splicing redirection in HeLa cells and exon skipping in mdx mouse muscle. Nucleic Acids Res. 36, 6418–6428 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Ivanova, G. D. et al. PNA-peptide conjugates as intracellular gene control agents. Nucleic Acids Symp. Ser. (Oxf.) 52, 31–32 (2008).

    Article  CAS  Google Scholar 

  80. Amantana, A. & Iversen, P. L. Pharmacokinetics and biodistribution of phosphorodiamidate morpholino antisense oligomers. Curr. Opin. Pharmacol. 5, 550–555 (2005).

    Article  CAS  PubMed  Google Scholar 

  81. Iversen, P. L. Phosphorodiamidate morpholino oligomers: favorable properties for sequence-specific gene inactivation. Curr. Opin. Mol. Ther. 3, 235–238 (2001).

    CAS  PubMed  Google Scholar 

  82. Crooke, S. T. et al. Pharmacokinetic properties of several novel oligonucleotide analogs in mice. J. Pharmacol. Exp. Ther. 277, 923–937 (1996).

    CAS  PubMed  Google Scholar 

  83. Morandi, L. et al. Dystrophin characterization in BMD patients: correlation of abnormal protein with clinical phenotype. J. Neurol. Sci. 132, 146–155 (1995).

    Article  CAS  PubMed  Google Scholar 

  84. Kinali, M. et al. Local restoration of dystrophin expression with the morpholino oligomer AVI-4658 in Duchenne muscular dystrophy: a single-blind, placebo-controlled, dose-escalation, proof-of-concept study. Lancet Neurol. 8, 918–928 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. van Deutekom, J. C. et al. Local dystrophin restoration with antisense oligonucleotide PRO051. N. Engl. J. Med. 357, 2677–2686 (2007). This was the first clinical study to report the successful restoration of dystrophin protein following the intramuscular administration of a 2OMe antisense oligonucleotide in patients with DMD.

    Article  CAS  PubMed  Google Scholar 

  86. Aartsma-Rus, A. Antisense-mediated modulation of splicing: therapeutic implications for Duchenne muscular dystrophy. RNA Biol. 7, 453–461 (2010).

    Article  CAS  PubMed  Google Scholar 

  87. Aartsma-Rus, A. et al. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum. Mutat. 30, 293–299 (2009).

    Article  PubMed  Google Scholar 

  88. Helderman-van den Enden, A. T. et al. Becker muscular dystrophy patients with deletions around exon 51; a promising outlook for exon skipping therapy in Duchenne patients. Neuromuscul. Disord. 20, 251–254 (2010).

    Article  CAS  PubMed  Google Scholar 

  89. Furling, D. et al. Viral vector producing antisense RNA restores myotonic dystrophy myoblast functions. Gene Ther. 10, 795–802 (2003).

    Article  CAS  PubMed  Google Scholar 

  90. Saengpattrachai, M., Ray, P. N., Hawkins, C. E., Berzen, A. & Banwell, B. L. Grandpa and I have dystrophinopathy?: approach to asymptomatic hyperCKemia. Pediatr. Neurol. 35, 145–149 (2006).

    Article  PubMed  Google Scholar 

  91. Goemans, N. M. et al. Systemic administration of PRO051 in duchenne's muscular dystrophy. N. Engl. J. Med. 364, 1513–1522 (2011). This was another clinical study that reported the successful restoration of dystrophin protein following systemic administration of a 2OMe antisense oligonucleotide in patients with DMD.

    Article  CAS  PubMed  Google Scholar 

  92. Wagner, K. R. et al. Gentamicin treatment of Duchenne and Becker muscular dystrophy due to nonsense mutations. Ann. Neurol. 49, 706–711 (2001).

    Article  CAS  PubMed  Google Scholar 

  93. Dunant, P., Walter, M. C., Karpati, G. & Lochmuller, H. Gentamicin fails to increase dystrophin expression in dystrophin-deficient muscle. Muscle Nerve 27, 624–627 (2003).

    Article  CAS  PubMed  Google Scholar 

  94. Barton-Davis, E. R., Cordier, L., Shoturma, D. I., Leland, S. E. & Sweeney, H. L. Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice. J. Clin. Invest. 104, 375–381 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Malik, V. et al. Gentamicin-induced readthrough of stop codons in Duchenne muscular dystrophy. Ann. Neurol. 67, 771–780 (2010).

    CAS  PubMed  Google Scholar 

  96. Welch, E. M. et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature 447, 87–91 (2007). This study identified a chemical entity, PTC124, that selectively induced ribosomal readthrough of premature termination codons and demonstrated efficacy in promoting dystrophin protein production in primary human muscle cells and in mdx mice.

    Article  PubMed  Google Scholar 

  97. Howard, M. T. et al. Readthrough of dystrophin stop codon mutations induced by aminoglycosides. Ann. Neurol. 55, 422–426 (2004).

    Article  CAS  PubMed  Google Scholar 

  98. Day, J. W. et al. Myotonic dystrophy type 2: molecular, diagnostic and clinical spectrum. Neurology 60, 657–664 (2003).

    Article  CAS  PubMed  Google Scholar 

  99. Jiang, H., Mankodi, A., Swanson, M. S., Moxley, R. T. & Thornton, C. A. Myotonic dystrophy type 1 is associated with nuclear foci of mutant RNA, sequestration of muscleblind proteins and deregulated alternative splicing in neurons. Hum. Mol. Genet. 13, 3079–3088 (2004).

    Article  CAS  PubMed  Google Scholar 

  100. Lee, J. E. & Cooper, T. A. Pathogenic mechanisms of myotonic dystrophy. Biochem. Soc. Trans. 37, 1281–1286 (2009).

    Article  CAS  PubMed  Google Scholar 

  101. Mahadevan, M. S. et al. Reversible model of RNA toxicity and cardiac conduction defects in myotonic dystrophy. Nature Genet. 38, 1066–1070 (2006).

    Article  CAS  PubMed  Google Scholar 

  102. Ranum, L. P. & Day, J. W. Myotonic dystrophy: RNA pathogenesis comes into focus. Am. J. Hum. Genet. 74, 793–804 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Orengo, J. P. et al. Expanded CTG repeats within the DMPK 3′ UTR causes severe skeletal muscle wasting in an inducible mouse model for myotonic dystrophy. Proc. Natl Acad. Sci. USA 105, 2646–2651 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  104. Ward, A. J., Rimer, M., Killian, J. M., Dowling, J. J. & Cooper, T. A. CUGBP1 overexpression in mouse skeletal muscle reproduces features of myotonic dystrophy type 1. Hum. Mol. Genet. 19, 3614–3622 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Yadava, R. S. et al. RNA toxicity in myotonic muscular dystrophy induces NKX2-5 expression. Nature Genet. 40, 61–68 (2008).

    Article  CAS  PubMed  Google Scholar 

  106. Yuan, Y. et al. Muscleblind-like 1 interacts with RNA hairpins in splicing target and pathogenic RNAs. Nucleic Acids Res. 35, 5474–5486 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Liquori, C. L. et al. Myotonic dystrophy type 2: human founder haplotype and evolutionary conservation of the repeat tract. Am. J. Hum. Genet. 73, 849–862 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Margolis, J. M., Schoser, B. G., Moseley, M. L., Day, J. W. & Ranum, L. P. DM2 intronic expansions: evidence for CCUG accumulation without flanking sequence or effects on ZNF9 mRNA processing or protein expression. Hum. Mol. Genet. 15, 1808–1815 (2006).

    Article  CAS  PubMed  Google Scholar 

  109. Cooper, T. A. Molecular biology. Neutralizing toxic RNA. Science 325, 272–273 (2009).

    Article  CAS  PubMed  Google Scholar 

  110. Koshelev, M., Sarma, S., Price, R. E., Wehrens, X. H. & Cooper, T. A. Heart-specific overexpression of CUGBP1 reproduces functional and molecular abnormalities of myotonic dystrophy type 1. Hum. Mol. Genet. 19, 1066–1075 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Orengo, J. P., Ward, A. J. & Cooper, T. A. Alternative splicing dysregulation secondary to skeletal muscle regeneration. Ann. Neurol. 69, 681–690 (2010).

    Article  CAS  Google Scholar 

  112. Ho, T. H. et al. Colocalization of muscleblind with RNA foci is separable from mis-regulation of alternative splicing in myotonic dystrophy. J. Cell Sci. 118, 2923–2933 (2005).

    Article  CAS  PubMed  Google Scholar 

  113. Lin, X. et al. Failure of MBNL1-dependent post-natal splicing transitions in myotonic dystrophy. Hum. Mol. Genet. 15, 2087–2097 (2006).

    Article  CAS  PubMed  Google Scholar 

  114. Kuyumcu-Martinez, N. M., Wang, G. S. & Cooper, T. A. Increased steady-state levels of CUGBP1 in myotonic dystrophy 1 are due to PKC-mediated hyperphosphorylation. Mol. Cell 28, 68–78 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Wang, G. S., Kearney, D. L., De Biasi, M., Taffet, G. & Cooper, T. A. Elevation of RNA-binding protein CUGBP1 is an early event in an inducible heart-specific mouse model of myotonic dystrophy. J. Clin. Invest. 117, 2802–2811 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Osborne, R. J. et al. Transcriptional and post-transcriptional impact of toxic RNA in myotonic dystrophy. Hum. Mol. Genet. 18, 1471–1481 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Wang, G. S. et al. PKC inhibition ameliorates the cardiac phenotype in a mouse model of myotonic dystrophy type 1. J. Clin. Invest. 119, 3797–3806 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Ranum, L. P. & Day, J. W. Pathogenic RNA repeats: an expanding role in genetic disease. Trends Genet. 20, 506–512 (2004).

    Article  CAS  PubMed  Google Scholar 

  119. Du, H. et al. Aberrant alternative splicing and extracellular matrix gene expression in mouse models of myotonic dystrophy. Nature Struct. Mol. Biol. 17, 187–193 (2010).

    Article  CAS  Google Scholar 

  120. Ho, T. H., Bundman, D., Armstrong, D. L. & Cooper, T. A. Transgenic mice expressing CUG-BP1 reproduce splicing mis-regulation observed in myotonic dystrophy. Hum. Mol. Genet. 14, 1539–1547 (2005).

    Article  CAS  PubMed  Google Scholar 

  121. Mulders, S. A., van Engelen, B. G., Wieringa, B. & Wansink, D. G. Molecular therapy in myotonic dystrophy: focus on RNA gain-of-function. Hum. Mol. Genet. 19, R90–R97 (2010).

    Article  CAS  PubMed  Google Scholar 

  122. Cooper, T. A. Chemical reversal of the RNA gain of function in myotonic dystrophy. Proc. Natl Acad. Sci. USA 106, 18433–18434 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  123. Warf, M. B., Nakamori, M., Matthys, C. M., Thornton, C. A. & Berglund, J. A. Pentamidine reverses the splicing defects associated with myotonic dystrophy. Proc. Natl Acad. Sci. USA 106, 18551–18556 (2009). This study identified the ability of the small molecule pentamidine to reverse missplicing of pre-mRNAs that are implicated in myotonic dystrophy.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Wheeler, T. M. et al. Reversal of RNA dominance by displacement of protein sequestered on triplet repeat RNA. Science 325, 336–339 (2009). This study showed the reversal of a transgenic myotonic dystrophy phenotype using a PMO antisense oligonucleotide, CAG25, that binds to microsatellite expanded RNA and blocks its interaction with MBNL1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Mulders, S. A. et al. Triplet-repeat oligonucleotide-mediated reversal of RNA toxicity in myotonic dystrophy. Proc. Natl Acad. Sci. USA 106, 13915–13920 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  126. Krol, J. et al. Ribonuclease dicer cleaves triplet repeat hairpins into shorter repeats that silence specific targets. Mol. Cell 25, 575–586 (2007). This was a report on the use of a 2OMe (CAG) 7 antisense oligonucleotide that silences RNA expression of mutated DMPK , reduces the number of ribonuclear aggregates and normalizes aberrant pre-mRNA splicing in vivo in mouse models of DM1.

    Article  CAS  PubMed  Google Scholar 

  127. Langlois, M. A. et al. Cytoplasmic and nuclear retained DMPK mRNAs are targets for RNA interference in myotonic dystrophy cells. J. Biol. Chem. 280, 16949–16954 (2005).

    Article  CAS  PubMed  Google Scholar 

  128. Passini, M. A. & Cheng, S. H. Prospects for the gene therapy of spinal muscular atrophy. Trends Mol. Med. 17, 259–265 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  130. Valori, C. F. et al. Systemic delivery of scAAV9 expressing SMN prolongs survival in a model of spinal muscular atrophy. Sci. Transl. Med. 2, 35ra42 (2010).

    Article  CAS  PubMed  Google Scholar 

  131. Wheeler, T. M., Lueck, J. D., Swanson, M. S., Dirksen, R. T. & Thornton, C. A. Correction of ClC-1 splicing eliminates chloride channelopathy and myotonia in mouse models of myotonic dystrophy. J. Clin. Invest. 117, 3952–3957 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. 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). This study reported the use of methoxyethyl-modified antisense oligonucleotides that were administered directly to the CNS in mice and cynomolgus monkeys, and demonstrated successful splicing redirection of the SMN2 gene.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Politano, L. et al. Gentamicin administration in Duchenne patients with premature stop codon. Preliminary results. Acta Myol. 22, 15–21 (2003).

    CAS  PubMed  Google Scholar 

  134. Baughan, T. et al. Stimulating full-length SMN2 expression by delivering bifunctional RNAs via a viral vector. Mol. Ther. 14, 54–62 (2006).

    Article  CAS  PubMed  Google Scholar 

  135. Baughan, T. D., Dickson, A., Osman, E. Y. & Lorson, C. L. Delivery of bifunctional RNAs that target an intronic repressor and increase SMN levels in an animal model of spinal muscular atrophy. Hum. Mol. Genet. 18, 1600–1611 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Dickson, A., Osman, E. & Lorson, C. L. A negatively acting bifunctional RNA increases survival motor neuron both in vitro and in vivo. Hum. Gene Ther. 19, 1307–1315 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Skordis, L. A., Dunckley, M. G., Yue, B., Eperon, I. C. & Muntoni, F. Bifunctional antisense oligonucleotides provide a trans-acting splicing enhancer that stimulates SMN2 gene expression in patient fibroblasts. Proc. Natl Acad. Sci. USA 100, 4114–4119 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. 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). This was the first study to demonstrate effective in vivo CNS splicing redirection of the SMN2 gene in the CNS in a mouse model of SMA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Hua, Y., Vickers, T. A., Baker, B. F., Bennett, C. F. & Krainer, A. R. Enhancement of SMN2 exon 7 inclusion by antisense oligonucleotides targeting the exon. PLoS Biol. 5, e73 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. 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  PubMed  PubMed Central  Google Scholar 

  141. Owen, N. et al. Design principles for bifunctional targeted oligonucleotide enhancers of splicing. Nucleic Acids Res. 20 May 2011 (doi:10.1093/nar/gkr152).

  142. Burghes, A. H. & McGovern, V. L. Antisense oligonucleotides and spinal muscular atrophy: skipping along. Genes Dev. 24, 1574–1579 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Cartegni, L. & Krainer, A. R. Correction of disease-associated exon skipping by synthetic exon-specific activators. Nature Struct. Biol. 10, 120–125 (2003).

    Article  CAS  PubMed  Google Scholar 

  144. Coady, T. H., Shababi, M., Tullis, G. E. & Lorson, C. L. Restoration of SMN function: delivery of a trans-splicing RNA re-directs SMN2 pre-mRNA splicing. Mol. Ther. 15, 1471–1478 (2007).

    Article  CAS  PubMed  Google Scholar 

  145. Singh, N. N., Shishimorova, M., Cao, L. C., Gangwani, L. & Singh, R. N. A short antisense oligonucleotide masking a unique intronic motif prevents skipping of a critical exon in spinal muscular atrophy. RNA Biol. 6, 341–350 (2009).

    Article  CAS  PubMed  Google Scholar 

  146. Meyer, K. et al. Rescue of a severe mouse model for spinal muscular atrophy by U7 snRNA-mediated splicing modulation. Hum. Mol. Genet. 18, 546–555 (2009).

    Article  CAS  PubMed  Google Scholar 

  147. Alvarez-Erviti, L. et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nature Biotech. 29, 341–345 (2011).

    Article  CAS  Google Scholar 

  148. Mitrpant, C. et al. Rational design of antisense oligomers to induce dystrophin exon skipping. Mol. Ther. 17, 1418–1426 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Muntoni, F. The development of antisense oligonucleotide therapies for Duchenne muscular dystrophy: report on a TREAT-NMD workshop hosted by the European Medicines Agency (EMA), on September 25th 2009. Neuromuscul. Disord. 20, 355–362 (2010).

    Article  PubMed  Google Scholar 

  150. Lee, J. E. & Cooper, T. A. Pathogenic mechanisms of myotonic dystrophy. Biochem. Soc. Trans. 37, 1281–1286 (2009).

    Article  CAS  PubMed  Google Scholar 

  151. Yin, H., Lu, Q. & Wood, M. Effective exon skipping and restoration of dystrophin expression by peptide nucleic acid antisense oligonucleotides in mdx mice. Mol. Ther. 16, 38–45 (2008).

    Article  CAS  PubMed  Google Scholar 

  152. Yin, H. et al. Optimization of peptide nucleic acid antisense oligonucleotides for local and systemic dystrophin splice correction in the mdx mouse. Mol. Ther. 18, 819–827 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Quinlivan, R., Kirschner, J., Comi, G. & Bushby, K. for the Ataluren DBMD Study Group. Safety and efficacy of ataluren 10,10,20 mg/kg TID in patients with nonsense mutation dystrophinopathy. European Paediatric Neurology Society Congress, 9th EPNS Congress [online], (2011).

    Google Scholar 

Download references

Acknowledgements

F.M. is supported by the Great Ormond Street Children's Charity. The financial support of the Medical Research Council (MRC), the Muscular Dystrophy Campaign, the French Muscular Dystrophy Association (AFM), the Wellcome Trust and TREAT-NMD is also gratefully acknowledged. M.J.A.W. is supported by the MRC, the AFM, the Wellcome Trust, Parkinson's UK, Action Duchenne, the Muscular Dystrophy Campaign, Duchenne Ireland and Muscular Dystrophy Ireland. The authors wish to thank Q. Lu, Neuromuscular/ALS Center, Carolinas Medical Center, Charlotte, North Carolina, USA, for providing Figure 2.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Francesco Muntoni.

Ethics declarations

Competing interests

Francesco Muntoni has previously served on the scientific advisory boards for AVI BioPharma, he has a patent pending regarding tailed antisense oligonucleotides to redirect splicing, and his university receives research support from AVI BioPharma and GlaxoSmithKline in relation to clinical trials on antisense oligonucleotides.

Matthew J. A. Wood has a patent pending regarding peptides for the delivery of antisense oligonucleotides.

Related links

Related links

FURTHER INFORMATION

Francesco Muntoni's homepages

Dubowitz Neuromuscular Centre

Centre for Neuromuscular Diseases

The MDEX Consortium

Matthew J. A. Wood's homepage

AVI BioPharma website

ClinicalTrials.gov

Leiden Muscular Dystrophy Pages

PTC Therapeutics website

TREAT-NMD website

Glossary

Neuromuscular disease

A disease affecting one or more constituent components of the neuromuscular unit, which comprise motor neurons, neuromuscular synapses and skeletal muscle.

Duchenne muscular dystrophy

(DMD). A severe, X-linked neuromuscular disease that arises as a result of mutations in the dystrophin gene that abolish the open reading frame, leading to the absence of dystrophin protein.

Myotonic dystrophy

The most common cause of muscular dystrophy in adults, which arises as a result of microsatellite repeat expansion mutations in the myotonin protein kinase gene (leading to muscular dystrophy type 1) or the cellular nucleic acid-binding protein gene (leading to muscular dystrophy type 2). These mutations lead to the formation of directly pathogenic RNAs and widespread dysregulation of splicing.

Spinal muscular atrophy

(SMA). A very common childhood genetic disease that arises as a result of loss-of-function mutations in the survival of motor neuron 1 gene, which lead to motor neuron degeneration and subsequent axial and limb muscle weakness.

Pre-mRNA splicing

The processing of a pre-mRNA transcript to remove specific intronic and some exonic sequences that are not required for the generation of the mature mRNA.

RNA-targeted therapies

Therapeutic modalities that are based on the targeted modification or inhibition of RNAs (for example, pre-mRNA, mature mRNA or non-coding RNA) that are directly implicated in disease.

Antisense oligonucleotide

A short single-stranded nucleic acid, typically 15–25 nucleotides in length, that has the ability to mediate therapeutic effects by directly interacting with pre-mRNA or mRNA in a sequence-specific manner.

2′-O-methyl RNA

An antisense oligonucleotide in which the RNA backbone is modified by the addition of methyl (CH3) groups at the 2′ position in the ribose ring.

Phosphorodiamidate morpholino

(PMO). An extensively modified antisense oligonucleotide that is neutrally charged and comprises a morpholino ring-based RNA backbone.

Open reading frame

(ORF). A stretch of a DNA sequence that does not contain a stop or termination codon, and typically corresponds to the legible DNA sequence of a single gene.

Exon skipping

The processing of a pre-mRNA transcript to result in the exclusion of a specific exon within the mature mRNA transcript.

mdx mouse

A naturally occurring mouse mutant that carries a premature termination codon in exon 23 of the dystrophin (Dmd) gene, which results in the absence of dystrophin protein.

Premature termination codon

A mutation that results in the presence of a de novo termination or stop codon in advance of where one would normally be found, and which serves to disrupt the open reading frame for that gene.

Cell-penetrating peptides

Short peptide fragments that are able to mediate enhanced cellular uptake of a range of cargoes by virtue of their cationic charge and the presence of specific cationic amino acid residues, typically arginine residues.

(RXR)4

An arginine-rich cell-penetrating peptide comprising eight arginine residues and the non-natural amino acid X (6-aminohexanoic acid).

Peptide–PMO

(PPMO). A phosphorodiamidate morpholino (PMO) oligonucleotide to which a short peptide fragment is attached at either the 5′ or 3′ end by direct chemical conjugation.

Stop codon readthrough

A therapeutic method of addressing the deleterious effect of a premature termination codon in which the readthrough of a premature termination codon is stimulated in order to yield a full-length open reading frame.

Microsatellite repeat expansion

The presence of repeat units of a microsatellite sequence in the genome, which expand beyond the number of repeat units that are associated with normal health, thus leading to a mutation that is directly pathogenic.

Spliceopathy

A disease that arises through widespread dysregulation of pre-mRNA splicing.

Exon inclusion

The processing of a pre-mRNA transcript to result in the inclusion of a specific exon within the mature mRNA transcript.

Personalized molecular therapies

The development of therapeutic agents that are directly tailored to specific mutations or to individual patients or subsets of patients.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Muntoni, F., Wood, M. Targeting RNA to treat neuromuscular disease. Nat Rev Drug Discov 10, 621–637 (2011). https://doi.org/10.1038/nrd3459

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd3459

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