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.

  • Original Article
  • Published:

Hydrostatic isolated limb perfusion with adeno-associated virus vectors enhances correction of skeletal muscle in Pompe disease

Abstract

Glycogen storage disease type II (Pompe disease; MIM 232300) stems from the inherited deficiency of acid-α-glucosidase (GAA; acid maltase; EC 3.2.1.20), which primarily involves cardiac and skeletal muscles. We hypothesized that hydrostatic isolated limb perfusion (ILP) administration of an adeno-associated virus (AAV) vector containing a muscle-specific promoter could achieve relatively higher transgene expression in the hindlimb muscles of GAA-knockout (GAA-KO) mice, in comparison with intravenous (IV) administration. ILP administration of AAV2/8 vectors encoding alkaline phosphatase or human GAA-transduced skeletal muscles of the hindlimb widely, despite the relatively low number of vector particles administered (1 × 1011), and IV administration of an equivalent vector dose failed to transduce skeletal muscle detectably. Similarly, ILP administration of fewer vector particles of the AAV2/9 vector encoding human GAA (3 × 1010) transduced skeletal muscles of the hindlimb widely and significantly reduced glycogen content to, in comparison with IV administration. The only advantage for IV administration was moderately high-level transduction of cardiac muscle, which demonstrated compellingly that ILP administration sequestered vector particles within the perfused limb. Reduction of glycogen storage in the extensor digitorum longus demonstrated the potential advantage of ILP-mediated delivery of AAV vectors in Pompe disease, because type II myofibers are resistant to enzyme replacement therapy. Thus, ILP will enhance AAV transduction of multiple skeletal muscles while reducing the required dosages in terms of vector particle numbers.

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

Similar content being viewed by others

References

  1. Hirschhorn R, Reuser AJJ . Glycogen storage disease type II: acid a-glucosidase (acid maltase) deficiency. In: Scriver CR, Beaudet AL, Sly WS, Valle D (eds). The Metabolic and Molecular Basis for Inherited Disease. McGraw-Hill: New York, 2001, pp 3389–3420.

    Google Scholar 

  2. Hoefsloot LH, Hoogeveen-Westerveld M, Reuser AJ, Oostra BA . Characterization of the human lysosomal alpha-glucosidase gene. Biochem J 1990; 1: 493–497.

    Article  Google Scholar 

  3. Wisselaar HA, Kroos MA, Hermans MMP, Vanbeeumen J, Reuser AJJ . Structural and functional-changes of lysosomal acid alpha-glucosidase during intracellular-transport and maturation. J Biol Chem 1993; 268: 2223–2231.

    CAS  PubMed  Google Scholar 

  4. Bijvoet AG, Van Hirtum H, Kroos MA, Van de Kamp EH, Schoneveld O, Visser P et al. Human acid alpha-glucosidase from rabbit milk has therapeutic effect in mice with glycogen storage disease type II. Hum Mol Genet 1999; 8: 2145–2153.

    Article  CAS  Google Scholar 

  5. Raben N, Danon M, Gilbert AL, Dwivedi S, Collins B, Thurberg BL et al. Enzyme replacement therapy in the mouse model of Pompe disease. Mol Genet Metab 2003; 80: 159–169.

    Article  CAS  Google Scholar 

  6. Amalfitano A, Bengur AR, Morse RP, Majure JM, Case LE, Veerling DL et al. Recombinant human acid alpha-glucosidase enzyme therapy for infantile glycogen storage disease type II: results of a phase I/II clinical trial. Genet Med 2001; 3: 132–138.

    CAS  PubMed  Google Scholar 

  7. Van den Hout JMP, Kamphoven JHJ, Winkel LPF, Arts WFM, De Klerk JBC, Loonen MCB et al. Long term intravenous treatment of Pompe disease with recombinant human alpha-glucosidase from milk. Pediatrics 2004; 113: E448–E457.

    Article  Google Scholar 

  8. Kishnani PS, Corzo D, Nicolino M, Byrne B, Mandel H, Hwu WL et al. Recombinant human acid {alpha}-glucosidase: major clinical benefits in infantile-onset Pompe disease. Neurology 2007; 68: 99–109.

    Article  CAS  Google Scholar 

  9. Kishnani PS, Goldenberg PC, Dearmey SL, Heller J, Benjamin D, Young S et al. Cross-reactive immunologic material status affects treatment outcomes in Pompe disease infants. Mol Genet Metab 2010; 99: 26–36.

    Article  CAS  Google Scholar 

  10. Franco LM, Sun B, Yang X, Bird A, Zhang H, Schneider A et al. Evasion of immune responses to introduced human acid alpha-glucosidase by liver-restricted expression in glycogen storage disease type II. Mol Ther 2005; 12: 876–884.

    Article  CAS  Google Scholar 

  11. Ziegler RJ, Bercury SD, Fidler J, Zhao MA, Foley J, Taksir TV et al. Ability of adeno-associated virus serotype 8-mediated hepatic expression of acid alpha-glucosidase to correct the biochemical and motor function deficits of presymptomatic and symptomatic Pompe mice. Hum Gene Ther 2008; 19: 609–621.

    Article  CAS  Google Scholar 

  12. Sun B, Zhang H, Benjamin Jr DK, Brown T, Bird A, Young SP et al. Enhanced efficacy of an AAV vector encoding chimeric, highly secreted acid alpha-glucosidase in glycogen storage disease type II. Mol Ther 2006; 14: 822–830.

    Article  CAS  Google Scholar 

  13. Sun B, Zhang H, Franco LM, Brown T, Bird A, Schneider A et al. Correction of glycogen storage disease type II by an adeno-associated virus vector containing a muscle-specific promoter. Mol Ther 2005; 11: 889–898.

    Article  CAS  Google Scholar 

  14. Sun B, Young SP, Li P, Di C, Brown T, Salva MZ et al. Correction of multiple striated muscles in murine Pompe disease through adeno-associated virus-mediated gene therapy. Mol Ther 2008; 16: 1366–1371.

    Article  CAS  Google Scholar 

  15. 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  CAS  Google Scholar 

  16. Manno CS, Pierce GF, Arruda VR, Glader B, Ragni M, Rasko JJ et al. Successful transduction of liver in hemophilia by AAV-factor IX and limitations imposed by the host immune response. Nat Med 2006; 12: 342–347.

    Article  CAS  Google Scholar 

  17. 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  CAS  Google Scholar 

  18. Su LT, Gopal K, Wang Z, Yin X, Nelson A, Kozyak BW et al. Uniform scale-independent gene transfer to striated muscle after transvenular extravasation of vector. Circulation 2005; 112: 1780–1788.

    Article  CAS  Google Scholar 

  19. Qiao C, Li J, Zheng H, Bogan J, Li J, Yuan Z et al. Hydrodynamic limb vein injection of AAV8 canine myostatin propeptide gene in normal dogs enhances muscle growth. Hum Gene Ther 2009; 20: 1–10.

    Article  CAS  Google Scholar 

  20. Salva MZ, Himeda CL, Tai PW, Nishiuchi E, Gregorevic P, Allen JM et al. Design of tissue-specific regulatory cassettes for high-level rAAV-mediated expression in skeletal and cardiac muscle. Mol Ther 2007; 15: 320–329.

    Article  CAS  Google Scholar 

  21. Gregorevic P, Schultz BR, Allen JM, Halldorson JB, Blankinship MJ, Meznarich NA et al. Evaluation of vascular delivery methodologies to enhance rAAV6-mediated gene transfer to canine striated musculature. Mol Ther 2009; 17: 1427–1433.

    Article  CAS  Google Scholar 

  22. Toumi H, Hegge J, Subbotin V, Noble M, Herweijer H, Best TM et al. Rapid intravascular injection into limb skeletal muscle: a damage assessment study. Mol Ther 2006; 13: 229–236.

    Article  CAS  Google Scholar 

  23. Vigen KK, Hegge JO, Zhang G, Mukherjee R, Braun S, Grist TM et al. Magnetic resonance imaging-monitored plasmid DNA delivery in primate limb muscle. Hum Gene Ther 2007; 18: 257–268.

    Article  CAS  Google Scholar 

  24. Hegge JO, Wooddell CI, Zhang G, Hagstrom JE, Braun S, Huss T et al. Evaluation of hydrodynamic limb vein injections in non-human primates. Hum Gene Ther 2010; 21: 829–842.

    Article  CAS  Google Scholar 

  25. Fukuda T, Roberts A, Ahearn M, Zaal K, Ralston E, Plotz PH et al. Autophagy and lysosomes in Pompe disease. Autophagy 2006; 2: 318–320.

    Article  CAS  Google Scholar 

  26. Haida N, Fowler Jr WM, Abresch RT, Larson DB, Sharman RB, Taylor RG et al. Effect of hind-limb suspension on young and adult skeletal muscle. I. Normal mice. Exp Neurol 1989; 103: 68–76.

    Article  CAS  Google Scholar 

  27. Wortmann RL, DiMauro S . Differentiating idiopathic inflammatory myopathies from metabolic myopathies. Rheum Dis Clin N Am 2002; 28: 759–778.

    Article  Google Scholar 

  28. Gao GP, Alvira MR, Wang L, Calcedo R, Johnston J, Wilson JM . Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci USA 2002; 99: 11854–11859.

    Article  CAS  Google Scholar 

  29. Raben N, Nagaraju K, Lee E, Kessler P, Byrne B, Lee L et al. Targeted disruption of the acid alpha-glucosidase gene in mice causes an illness with critical features of both infantile and adult human glycogen storage disease type II. J Biol Chem 1998; 273: 19086–19092.

    Article  CAS  Google Scholar 

  30. Amalfitano A, McVie-Wylie AJ, Hu H, Dawson TL, Raben N, Plotz P et al. Systemic correction of the muscle disorder glycogen storage disease type II after hepatic targeting of a modified adenovirus vector encoding human acid-alpha-glucosidase. Proc Natl Acad Sci USA 1999; 96: 8861–8866.

    Article  CAS  Google Scholar 

  31. Sun B, Zhang H, Franco LM, Young SP, Schneider A, Bird A et al. Efficacy of an adeno-associated virus 8-pseudotyped vector in glycogen storage disease type II. Mol Ther 2005; 11: 57–65.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH Grant R01 HL081122 from the National Heart, Lung and Blood Institute. BS was supported by the Muscular Dystrophy Association. GAA-KO mice were provided courtesy of Dr Nina Raben at the National Institutes of Health (Bethesda, MD, USA). The AAV8 and AAV9 packaging plasmids were provided courtesy of Dr James M Wilson at the University of Pennsylvania (Philadelphia, PA, USA).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to D D Koeberl.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sun, B., Li, S., Bird, A. et al. Hydrostatic isolated limb perfusion with adeno-associated virus vectors enhances correction of skeletal muscle in Pompe disease. Gene Ther 17, 1500–1505 (2010). https://doi.org/10.1038/gt.2010.109

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/gt.2010.109

Keywords

Search

Quick links