J Clin Neurol.  2010 Sep;6(3):111-116. 10.3988/jcn.2010.6.3.111.

Gene Therapy for Muscular Dystrophies: Progress and Challenges

Affiliations
  • 1Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea. pks1126@chol.com
  • 2Department of Neurology, Incheon Medical Center, Incheon, Korea.

Abstract

Muscular dystrophies are groups of inherited progressive diseases of the muscle caused by mutations of diverse genes related to normal muscle function. Although there is no current effective treatment for these devastating diseases, various molecular strategies have been developed to restore the expressions of the associated defective proteins. In preclinical animal models, both viral and nonviral vectors have been shown to deliver recombinant versions of defective genes. Antisense oligonucleotides have been shown to modify the splicing mechanism of mesenger ribonucleic acid to produce an internally deleted but partially functional dystrophin in an experimental model of Duchenne muscular dystrophy. In addition, chemicals can induce readthrough of the premature stop codon in nonsense mutations of the dystrophin gene. On the basis of these preclinical data, several experimental clinical trials are underway that aim to demonstrate efficacy in treating these de-vastating diseases.

Keyword

gene therapy; muscular dystrophies; Duchenne

MeSH Terms

Codon, Nonsense
Dystrophin
Genetic Therapy
Models, Animal
Models, Theoretical
Muscles
Muscular Dystrophies
Muscular Dystrophy, Duchenne
Oligonucleotides, Antisense
Proteins
RNA
Codon, Nonsense
Dystrophin
Oligonucleotides, Antisense
Proteins
RNA

Figure

  • Fig. 1 Full-length and truncated dystrophin genes. A: Full-length dystrophin gene. B: Minidystrophin gene. C: Microdystrophin gene. ABD: N-terminal actin-binding domain, H1-H4: hinge regions, 1-24: spectrin-like repeats, iABD: internal actin-binding domain, CR: cysteine-rich region, CT: carboxy terminus.

  • Fig. 2 Exon skipping using antisense oligonucleotides. A: An out-of-frame deletion of exons 45-54 causes a stop codon in exon 55 that arrests dystrophin pro-duction in Duchenne muscular dystrophy. B: Skipping of exon 44 using antisense oligonucleotides restores the reading frame back "in-frame", producing a partially functional dystrophin protein. AON: antisense oligonucleotide.


Reference

1. Acsadi G, Dickson G, Love DR, Jani A, Walsh FS, Gurusinghe A, et al. Human dystrophin expression in mdx mice after intramuscular injection of DNA constructs. Nature. 1991. 352:815–818.
Article
2. Aihara H, Miyazaki J. Gene transfer into muscle by electroporation in vivo. Nat Biotechnol. 1998. 16:867–870.
Article
3. Mir LM, Bureau MF, Gehl J, Rangara R, Rouy D, Caillaud JM, et al. High-efficiency gene transfer into skeletal muscle mediated by electric pulses. Proc Natl Acad Sci U S A. 1999. 96:4262–4267.
Article
4. Durieux AC, Bonnefoy R, Busso T, Freyssenet D. In vivo gene electrotransfer into skeletal muscle: effects of plasmid DNA on the occurrence and extent of muscle damage. J Gene Med. 2004. 6:809–816.
Article
5. Molnar MJ, Gilbert R, Lu Y, Liu AB, Guo A, Larochelle N, et al. Factors influencing the efficacy, longevity, and safety of electroporation-assisted plasmid-based gene transfer into mouse muscles. Mol Ther. 2004. 10:447–455.
Article
6. Vilquin JT, Kennel PF, Paturneau-Jouas M, Chapdelaine P, Boissel N, Delaère P, et al. Electrotransfer of naked DNA in the skeletal muscles of animal models of muscular dystrophies. Gene Ther. 2001. 8:1097–1107.
Article
7. Murakami T, Nishi T, Kimura E, Goto T, Maeda Y, Ushio Y, et al. Full-length dystrophin cDNA transfer into skeletal muscle of adult mdx mice by electroporation. Muscle Nerve. 2003. 27:237–241.
Article
8. McMahon JM, Signori E, Wells KE, Fazio VM, Wells DJ. Optimisation of electrotransfer of plasmid into skeletal muscle by pretreatment with hyaluronidase--increased expression with reduced muscle damage. Gene Ther. 2001. 8:1264–1270.
Article
9. Mennuni C, Calvaruso F, Zampaglione I, Rizzuto G, Rinaudo D, Dammassa E, et al. Hyaluronidase increases electrogene transfer efficiency in skeletal muscle. Hum Gene Ther. 2002. 13:355–365.
Article
10. Budker V, Zhang G, Danko I, Williams P, Wolff J. The efficient expression of intravascularly delivered DNA in rat muscle. Gene Ther. 1998. 5:272–276.
Article
11. Zhang G, Ludtke JJ, Thioudellet C, Kleinpeter P, Antoniou M, Herweijer H, et al. Intraarterial delivery of naked plasmid DNA expressing full-length mouse dystrophin in the mdx mouse model of duchenne muscular dystrophy. Hum Gene Ther. 2004. 15:770–782.
Article
12. Hagstrom JE, Hegge J, Zhang G, Noble M, Budker V, Lewis DL, et al. A facile nonviral method for delivering genes and siRNAs to skeletal muscle of mammalian limbs. Mol Ther. 2004. 10:386–398.
Article
13. Romero NB, Braun S, Benveniste O, Leturcq F, Hogrel JY, Morris GE, et al. Phase I study of dystrophin plasmid-based gene therapy in Duchenne/Becker muscular dystrophy. Hum Gene Ther. 2004. 15:1065–1076.
Article
14. DelloRusso C, Scott JM, Hartigan-O'Connor D, Salvatori G, Barjot C, Robinson AS, et al. Functional correction of adult mdx mouse muscle using gutted adenoviral vectors expressing fulll-ength dystrophin. Proc Natl Acad Sci U S A. 2002. 99:12979–12984.
Article
15. Nunes FA, Furth EE, Wilson JM, Raper SE. Gene transfer into the liver of nonhuman primates with E1-deleted recombinant adenoviral vectors: safety of readministration. Hum Gene Ther. 1999. 10:2515–2526.
Article
16. Jooss K, Yang Y, Fisher KJ, Wilson JM. Transduction of dendritic cells by DNA viral vectors directs the immune response to transgene products in muscle fibers. J Virol. 1998. 72:4212–4223.
Article
17. Wang B, Li J, Xiao X. Adeno-associated virus vector carrying human minidystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model. Proc Natl Acad Sci U S A. 2000. 97:13714–13719.
Article
18. Yoshimura M, Sakamoto M, Ikemoto M, Mochizuki Y, Yuasa K, Miyagoe-Suzuki Y, et al. AAV vector-mediated microdystrophin expression in a relatively small percentage of mdx myofibers improved the mdx phenotype. Mol Ther. 2004. 10:821–828.
Article
19. Gregorevic P, Blankinship MJ, Allen JM, Crawford RW, Meuse L, Miller DG, et al. Systemic delivery of genes to striated muscles using adeno-associated viral vectors. Nat Med. 2004. 10:828–834.
Article
20. Rodino-Klapac LR, Lee JS, Mulligan RC, Clark KR, Mendell JR. Lack of toxicity of alpha-sarcoglycan overexpression supports clinical gene transfer trial in LGMD2D. Neurology. 2008. 71:240–247.
Article
21. Yuasa K, Sakamoto M, Miyagoe-Suzuki Y, Tanouchi A, Yamamoto H, Li J, et al. Adeno-associated virus vector-mediated gene transfer into dystrophin-deficient skeletal muscles evokes enhanced immune response against the transgene product. Gene Ther. 2002. 9:1576–1588.
Article
22. Rivière C, Danos O, Douar AM. Long-term expression and repeated administration of AAV type 1, 2 and 5 vectors in skeletal muscle of immunocompetent adult mice. Gene Ther. 2006. 13:1300–1308.
Article
23. Gregorevic P, Allen JM, Minami E, Blankinship MJ, Haraguchi M, Meuse L, et al. rAAV6-microdystrophin preserves muscle function and extends lifespan in severely dystrophic mice. Nat Med. 2006. 12:787–789.
Article
24. Inagaki K, Fuess S, Storm TA, Gibson GA, Mctiernan CF, Kay MA, et al. Robust systemic transduction with AAV9 vectors in mice: efficient global cardiac gene transfer superior to that of AAV8. Mol Ther. 2006. 14:45–53.
Article
25. Mendell JR, Rodino-Klapac LR, Rosales-Quintero X, Kota J, Coley BD, Galloway G, et al. Limb-girdle muscular dystrophy type 2D gene therapy restores alpha-sarcoglycan and associated proteins. Ann Neurol. 2009. 66:290–297.
Article
26. Li J, Dressman D, Tsao YP, Sakamoto A, Hoffman EP, Xiao X. rAAV vector-mediated sarcogylcan gene transfer in a hamster model for limb girdle muscular dystrophy. Gene Ther. 1999. 6:74–82.
Article
27. Monaco AP, Bertelson CJ, Liechti-Gallati S, Moser H, Kunkel LM. An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics. 1988. 2:90–95.
Article
28. Dunckley MG, Manoharan M, Villiet P, Eperon IC, Dickson G. Modification of splicing in the dystrophin gene in cultured Mdx muscle cells by antisense oligoribonucleotides. Hum Mol Genet. 1998. 7:1083–1090.
Article
29. Mann CJ, Honeyman K, Cheng AJ, Ly T, Lloyd F, Fletcher S, et al. Antisense-induced exon skipping and synthesis of dystrophin in the mdx mouse. Proc Natl Acad Sci U S A. 2001. 98:42–47.
Article
30. Alter J, Lou F, Rabinowitz A, Yin H, Rosenfeld J, Wilton SD, et al. Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology. Nat Med. 2006. 12:175–177.
Article
31. Lu QL, Mann CJ, Lou F, Bou-Gharios G, Morris GE, Xue SA, et al. Functional amounts of dystrophin produced by skipping the mutated exon in the mdx dystrophic mouse. Nat Med. 2003. 9:1009–1014.
Article
32. Wells KE, Fletcher S, Mann CJ, Wilton SD, Wells DJ. Enhanced in vivo delivery of antisense oligonucleotides to restore dystrophin expression in adult mdx mouse muscle. FEBS Lett. 2003. 552:145–149.
Article
33. Bremmer-Bout M, Aartsma-Rus A, de Meijer EJ, Kaman WE, Janson AA, Vossen RH, et al. Targeted exon skipping in transgenic hDMD mice: a model for direct preclinical screening of human-specific antisense oligonucleotides. Mol Ther. 2004. 10:232–240.
Article
34. Takeshima Y, Yagi M, Wada H, Ishibashi K, Nishiyama A, Kakumoto M, et al. Intravenous infusion of an antisense oligonucleotide results in exon skipping in muscle dystrophin mRNA of Duchenne muscular dystrophy. Pediatr Res. 2006. 59:690–694.
Article
35. van Deutekom JC, Janson AA, Ginjaar IB, Frankhuizen WS, Aartsma-Rus A, Bremmer-Bout M, et al. Local dystrophin restoration with antisense oligonucleotide PRO051. N Engl J Med. 2007. 357:2677–2686.
Article
36. Barton-Davis ER, Cordier L, Shoturma DI, Leland SE, Sweeney HL. Aminoglycoside antibiotics restore dystrophin function to skeletal muscles of mdx mice. J Clin Invest. 1999. 104:375–381.
Article
37. Wagner KR, Hamed S, Hadley DW, Gropman AL, Burstein AH, Escolar DM, et al. Gentamicin treatment of Duchenne and Becker muscular dystrophy due to nonsense mutations. Ann Neurol. 2001. 49:706–711.
Article
38. Politano L, Nigro G, Nigro V, Piluso G, Papparella S, Paciello O, et al. Gentamicin administration in Duchenne patients with premature stop codon. Preliminary results. Acta Myol. 2003. 22:15–21.
39. Howard MT, Shirts BH, Petros LM, Flanigan KM, Gesteland RF, Atkins JF. Sequence specificity of aminoglycoside-induced stop condon readthrough: potential implications for treatment of Duchenne muscular dystrophy. Ann Neurol. 2000. 48:164–169.
Article
40. Welch EM, Barton ER, Zhuo J, Tomizawa Y, Friesen WJ, Trifillis P, et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature. 2007. 447:87–91.
Article
41. Hirawat S, Welch EM, Elfring GL, Northcutt VJ, Paushkin S, Hwang S, et al. Safety, tolerability, and pharmacokinetics of PTC124, a nonaminoglycoside nonsense mutation suppressor, following single- and multiple-dose administration to healthy male and female adult volunteers. J Clin Pharmacol. 2007. 47:430–444.
Article
42. Quenneville SP, Chapdelaine P, Rousseau J, Tremblay JP. Dystrophin expression in host muscle following transplantation of muscle precursor cells modified with the phiC31 integrase. Gene Ther. 2007. 14:514–522.
Article
43. Ikemoto M, Fukada S, Uezumi A, Masuda S, Miyoshi H, Yamamoto H, et al. Autologous transplantation of SM/C-2.6(+) satellite cells transduced with micro-dystrophin CS1 cDNA by lentiviral vector into mdx mice. Mol Ther. 2007. 15:2178–2185.
Article
44. Benchaouir R, Meregalli M, Farini A, D'Antona G, Belicchi M, Goyenvalle A, et al. Restoration of human dystrophin following transplantation of exon-skipping-engineered DMD patient stem cells into dystrophic mice. Cell Stem Cell. 2007. 1:646–657.
Article
45. Sampaolesi M, Blot S, D'Antona G, Granger N, Tonlorenzi R, Innocenzi A, et al. Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature. 2006. 444:574–579.
Article
46. Torrente Y, Belicchi M, Marchesi C, Dantona G, Cogiamanian F, Pisati F, et al. Autologous transplantation of muscle-derived CD133+ stem cells in Duchenne muscle patients. Cell Transplant. 2007. 16:563–577.
Article
Full Text Links
  • JCN
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr