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.

  • Research Article
  • Published:

CTLA4Ig delivered by high-capacity adenoviral vector induces stable expression of dystrophin in mdx mouse muscle

Abstract

Adenoviral (Ad) vector-mediated gene delivery of normal, full-length dystrophin to skeletal muscle provides a promising strategy for the treatment of Duchenne muscular dystrophy (DMD), an X-linked recessive, dystrophin-deficient muscle disease. Studies in animal models suggest that successful DMD gene therapy by Ad vector-mediated gene transfer would be precluded by cellular and humoral immune responses induced by vector capsid and transgene proteins. To address the immunity induced by Ad vector-mediated dystrophin gene delivery to dystrophic muscle, we developed high-capacity adenoviral (HC-Ad) vectors expressing mouse dystrophin driven by the muscle creatine kinase promoter (AdmDys) and mCTLA4Ig (AdmCTLA4Ig) individually, or together from one vector (AdmCTLA4Ig/mDys). We found stable expression of dystrophin protein in the tibialis anterior muscles of mdx mice, coinjected with AdmCTLA4Ig and AdmDys, or injected alone with AdmCTLA4Ig/mDys, whereas the expression of dystrophin protein in the control group coinjected with AdmDys and an empty vector decreased by at least 50% between 2 and 8 weeks after administration. Additionally, we observed reductions in Ad vector-induced Th1 and Th2 cytokines, Ad vector-specific cytotoxic T lymphocyte activation and neutralizing anti-Ad antibodies in both experimental groups that received a mCTLA4Ig-expressing vector as compared to the control group. This study demonstrates that the coexpression of mCTLA4Ig and dystrophin in skeletal muscle provided by HC-Ad vector-mediated gene transfer can provide stable expression of dystrophin in immunocompetent, adult mdx mouse muscle and applies a potentially powerful strategy to overcome adaptive immunity induced by Ad vector-mediated dystrophin gene delivery toward the ultimate goal of treatment for DMD.

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
Figure 2
Figure 3
Figure 4
Figure 5
Figure 6
Figure 7

Similar content being viewed by others

References

  1. Morral N et al. High doses of a helper-dependent adenoviral vector yield supraphysiological levels of alpha1-antitrypsin with negligible toxicity. Hum Gene Ther 1998; 9: 2709–2716.

    Article  CAS  PubMed  Google Scholar 

  2. Scheidner G et al. Genomic DNA transfer with a high-capacity adenovirus vector results in improved in vivo gene expression and decreased toxicity. Nat Genet 1998; 18: 180–183.

    Article  Google Scholar 

  3. Clemens PR et al. In vivo muscle gene transfer of full-length dystrophin with an adenoviral vector that lacks all viral genes. Gene Therapy 1996; 3: 965–972.

    CAS  PubMed  Google Scholar 

  4. DelloRusso C et al. Functional correction of adult mdx mouse muscle using gutted adenoviral vectors expressing full-length dystrophin. Proc Natl Acad Sci USA 2002; 99: 12979–12984.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gilchrist SC, Ontell MP, Kochanek S, Clemens PR . Immune response to full-length dystrophin delivered to dmd muscle by a high-capacity adenoviral vector. Mol Ther 2002; 6: 359–368.

    Article  CAS  PubMed  Google Scholar 

  6. Ohtsuka Y et al. Dystrophin acts as a transplantation rejection antigen in dystrophin-deficient mice: implication for gene therapy. J Immunol 1998; 160: 4635–4640.

    CAS  PubMed  Google Scholar 

  7. Xu M, Lepisto AJ, Hendricks RL . Co-stimulatory requirements of effector T cells at inflammatory sites. DNA Cell Biol 2002; 21: 461–465.

    Article  CAS  PubMed  Google Scholar 

  8. Walunas TL, Bakker CY, Bluestone JA . CTLA-4 ligation blocks CD28-dependent T cell activation. J Exp Med 1996; 183: 2541–2550.

    Article  CAS  PubMed  Google Scholar 

  9. Li TS et al. Prolonged survival of xenograft fetal cardiomyocytes by adenovirus-mediated CTLA4-Ig expression. Transplantation 2001; 72: 1983–1985.

    Article  CAS  PubMed  Google Scholar 

  10. Matsui Y et al. Blockade of T cell costimulatory signals using adenovirus vectors prevents both the induction and the progression of experimental autoimmune myocarditis. J Mol Cell Cardiol 2002; 34: 279–295.

    Article  CAS  PubMed  Google Scholar 

  11. Mihara M et al. CTLA4Ig inhibits T cell-dependent B-cell maturation in murine systemic lupus erythematosus. J Clin Invest 2000; 106: 91–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Jooss K, Turka LA, Wilson JM . Blunting of immune responses to adenoviral vectors in mouse liver and lung with CTLA4Ig. Gene Therapy 1998; 5: 309–319.

    Article  CAS  PubMed  Google Scholar 

  13. Kay MA et al. Long-term hepatic adenovirus-mediated gene expression in mice following CTLA4Ig administration. Nat Genet 1995; 11: 191–197.

    Article  CAS  PubMed  Google Scholar 

  14. Kay MA et al. Transient immunomodulation with anti-CD40 ligand antibody and CTLA4Ig enhances persistence and secondary adenovirus-mediated gene transfer into mouse liver. Proc Natl Acad Sci USA 1997; 94: 4686–4691.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Schowalter DB et al. Constitutive expression of murine CTLA4Ig from a recombinant adenovirus vector results in prolonged transgene expression. Gene Therapy 1997; 4: 853–860.

    Article  CAS  PubMed  Google Scholar 

  16. Thummala NR et al. A non-immunogenic adenoviral vector, coexpressing CTLA4Ig and bilirubin-uridine-diphosphoglucuronateglucuronosyltransferase permits long-term, repeatable transgene expression in the Gunn rat model of Crigler–Najjar syndrome. Gene Therapy 2002; 9: 981–990.

    Article  CAS  PubMed  Google Scholar 

  17. Wilson CB et al. Transient inhibition of CD28 and CD40 ligand interactions prolongs adenovirus-mediated transgene expression in the lung and facilitates expression after secondary vector administration. J Virol 1998; 72: 7542–7550.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Guibinga GH et al. Combinatorial blockade of calcineurin and CD28 signaling facilitates primary and secondary therapeutic gene transfer by adenovirus vectors in dystrophic (mdx) mouse muscles. J Virol 1998; 72: 4601–4609.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Guerette B et al. Prevention of immune reactions triggered by first-generation adenoviral vectors by monoclonal antibodies and CTLA4Ig. Hum Gene Ther 1996; 7: 1455–1463.

    Article  CAS  PubMed  Google Scholar 

  20. Jiang ZL et al. Local high-capacity adenovirus-mediated mCTLA4Ig and mCD40Ig expression prolongs recombinant gene expression in skeletal muscle. Mol Ther 2001; 3: 892–900.

    Article  CAS  PubMed  Google Scholar 

  21. Hoffman EP, Morgan JE, Watkins SC, Partridge TA . Somatic reversion/suppression of the mouse mdx phenotype in vivo. J Neurol Sci 1990; 99: 9–25.

    Article  CAS  PubMed  Google Scholar 

  22. Hoffman EP, Brown Jr RH . Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 1987; 51: 919–928.

    Article  CAS  PubMed  Google Scholar 

  23. Sicinski P et al. The molecular basis of muscular dystrophy in the mdx mouse: a point mutation. Science 1989; 244: 1578–1580.

    Article  CAS  PubMed  Google Scholar 

  24. Ibraghimov-Beskrovnaya O et al. Primary structure of dystrophin-associated glycoproteins linking dystrophin to the extracellular matrix. Nature 1992; 355: 696–702.

    Article  CAS  PubMed  Google Scholar 

  25. Acsadi G et al. Dystrophin expression in muscles of mdx mice after adenovirus-mediated in vivo gene transfer. Hum Gene Ther 1996; 7: 129–140.

    Article  CAS  PubMed  Google Scholar 

  26. Alameddine HS et al. Expression of a recombinant dystrophin in mdx mice using adenoviurs vector. Neuromusc Disord 1994; 4: 193–203.

    Article  CAS  PubMed  Google Scholar 

  27. Clemens PR et al. Recombinant truncated dystrophin minigenes: construction, expression, and adenoviral delivery. Hum Gene Ther 1995; 6: 1477–1485.

    Article  CAS  PubMed  Google Scholar 

  28. Gilbert R et al. Dystrophin expression in muscle following gene transfer with a fully deleted (‘gutted’) adenovirus is markedly improved by trans-acting adenoviral gene products. Hum Gene Ther 2001; 12: 1741–1755.

    Article  CAS  PubMed  Google Scholar 

  29. Gilbert R et al. Improved performance of a fully gutted adenovirus vector containing two full-length dystrophin cDNAs regulated by a strong promoter. Mol Ther 2002; 6: 501–509.

    Article  CAS  PubMed  Google Scholar 

  30. Ragot T et al. Efficient adenovirus-mediated transfer of a human minidystrophin gene to skeletal muscle. Nature 1993; 361: 647–650.

    Article  CAS  PubMed  Google Scholar 

  31. Vincent N et al. Long-term correction of mouse dystrophic degeneration by adenovirus-mediated transfer of a minidystrophin gene. Nat Genet 1993; 5: 130–134.

    Article  CAS  PubMed  Google Scholar 

  32. Yang L et al. Adenovirus-mediated dystrophin minigene transfer improves muscle strength in adult dystrophic (MDX) mice. Gene Therapy 1998; 5: 369–379.

    Article  CAS  PubMed  Google Scholar 

  33. Lochmuller H et al. Immunosuppression by FK506 markedly prolongs expression of adenovirus-delivered transgene in skeletal muscles of adult dystrophic (mdx) mice. Biochem Biophys Res Commun 1995; 213: 569–574.

    Article  CAS  PubMed  Google Scholar 

  34. Lochmuller H et al. Transient immunosuppression by FK506 permits a sustained high-level dystrophin expression after adenovirus-mediated dystrophin minigene transfer to skeletal muscles of adult dystrophic (mdx) mice. Gene Therapy 1996; 3: 706–716.

    CAS  PubMed  Google Scholar 

  35. Kreppel F, Biermann V, Kochanek S, Schiedner G . A DNA-based method to assay total and infectious particle contents and helper virus contamination in high-capacity adenoviral vector preparations. Hum Gene Ther 2002; 13: 1151–1156.

    Article  CAS  PubMed  Google Scholar 

  36. Rando TA, Blau HM . Primary mouse myoblast purification, characterization, and transplantation for cell-mediated gene therapy. J Cell Biol 1994; 125: 1275–1287.

    Article  CAS  PubMed  Google Scholar 

  37. Jaynes JB et al. Transcriptional regulation of the muscle creatine kinase gene and regulated expression in transfected mouse myoblasts. Mol Cell Biol 1986; 6: 2855–2864.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Einfeld DA et al. Reducing the native tropism of adenovirus vectors requires removal of both CAR and integrin interactions. J Virol 2001; 75: 11284–11291.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Jiang Z, Feingold E, Kochanek S, Clemens PR . Systemic delivery of a high-capacity adenoviral vector expressing mouse CTLA4Ig improves skeletal muscle gene therapy. Mol Ther 2002; 6: 369–376.

    Article  CAS  PubMed  Google Scholar 

  40. Hoffman EP et al. Cross-reactive protein in Duchenne muscle. Lancet 1989; ii: 1211–1212.

    Article  Google Scholar 

  41. Koenig M, Kunkel LM . Detailed analysis of the repeat domain of dystrophin reveals four potential hinge segments that may confer flexibility. J Biol Chem 1990; 265: 4560–4566.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Merck & Co., Inc. for the helper virus AdLC8cluc and 293Cre4 cells, Dr SD Hauschka for a plasmid carrying the MCK promoter, and Xinyan Gu for technical assistance. This research was supported by a DMDRC fellowship to ZJ and grants from the NIH and Muscular Dystrophy Association to PRC.

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jiang, Z., Schiedner, G., Gilchrist, S. et al. CTLA4Ig delivered by high-capacity adenoviral vector induces stable expression of dystrophin in mdx mouse muscle. Gene Ther 11, 1453–1461 (2004). https://doi.org/10.1038/sj.gt.3302315

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/sj.gt.3302315

Keywords

This article is cited by

Search

Quick links