Hostname: page-component-8448b6f56d-c47g7 Total loading time: 0 Render date: 2024-04-19T01:32:22.969Z Has data issue: false hasContentIssue false

Emerging strategies for cell and gene therapy of the muscular dystrophies

Published online by Cambridge University Press:  25 June 2009

Lindsey A. Muir
Affiliation:
Program in Molecular and Cellular Biology, University of Washington, Seattle, Washington 98195, USA.
Jeffrey S. Chamberlain*
Affiliation:
Department of Neurology, Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, Department of Biochemistry, and Department of Medicine, University of Washington, Seattle, Washington 98195, USA.
*
*Corresponding author: Jeffrey S. Chamberlain, Department of Neurology, University of Washington School of Medicine, HSB Room K233, Box 357720, Seattle, Washington 98195, USA. Tel: +1 206 616 6645; Fax: +1 206 616 8272; E-mail: jsc5@u.washington.edu

Abstract

The muscular dystrophies are a heterogeneous group of over 40 disorders that are characterised by muscle weakness and wasting. The most common are Duchenne muscular dystrophy and Becker muscular dystrophy, which result from mutations within the gene encoding dystrophin; myotonic dystrophy type 1, which results from an expanded trinucleotide repeat in the myotonic dystrophy protein kinase gene; and facioscapulohumeral dystrophy, which is associated with contractions in the subtelomeric region of human chromosome 1. Currently the only treatments involve clinical management of symptoms, although several promising experimental strategies are emerging. These include gene therapy using adeno-associated viral, lentiviral and adenoviral vectors and nonviral vectors, such as plasmid DNA. Exon-skipping and cell-based therapies have also shown promise in the effective treatment and regeneration of dystrophic muscle. The availability of numerous animal models for Duchenne muscular dystrophy has enabled extensive testing of a wide range of therapeutic approaches for this type of disorder. Consequently, we focus here on the therapeutic developments for Duchenne muscular dystrophy as a model of the types of approaches being considered for various types of dystrophy. We discuss the advantages and limitations of each therapeutic strategy, as well as prospects and recent successes in the context of future clinical applications.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2009

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

1Emery, A.E.H., eds (2001) The Muscular Dystrophies. Oxford University Press, OxfordCrossRefGoogle Scholar
2Emery, A.E. and Muntoni, F. (2003) Duchenne Muscular Dystrophy. (3rd, edn), Oxford University Press, OxfordGoogle Scholar
3Wheeler, T.M. et al. (2007) Correction of ClC-1 splicing eliminates chloride channelopathy and myotonia in mouse models of myotonic dystrophy. Journal of Clinical Investigation 117, 3952-3957Google ScholarPubMed
4Tawil, R. et al. (1998) Facioscapulohumeral dystrophy: A distinct regional myopathy with a novel molecular pathogenesis. Annals of Neurology 43, 279-282CrossRefGoogle ScholarPubMed
5Bansal, D. and Campbell, K.P. (2004) Dysferlin and the plasma membrane repair in muscular dystrophy. Trends in Cell Biology 14, 206-213CrossRefGoogle ScholarPubMed
6Davies, K.E. and Nowak, K.J. (2006) Molecular mechanisms of muscular dystrophies: old and new players. Nature Reviews Molecular Cell Biology 7, 762-773CrossRefGoogle ScholarPubMed
7Monaco, A.P. et al. (1988) An explanation for the phenotypic differences between patients bearing partial deletions of the DMD locus. Genomics 2, 90-95Google Scholar
8Abmayr, S. and Chamberlain, J.S. (2006) The structure and function of dystrophin. In The Molecular Mechanisms in Muscular Dystrophy (Winder, S.J., eds), pp. 14-34, Landes Biosciences, GeorgetownGoogle Scholar
9Huang, X. et al. (2000) Structure of a WW domain containing fragment of dystrophin in complex with beta-dystroglycan. Nature Structural and Molecular Biology 7, 634-638Google Scholar
10Yoshida, M. and Ozawa, E. (1990) Glycoprotein complex anchoring dystrophin to sarcolemma. Journal of Biochemistry 108, 748-752CrossRefGoogle ScholarPubMed
11Ervasti, J.M. and Campbell, K.P. (1993) A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. Journal of Cell Biology 122, 809-823CrossRefGoogle ScholarPubMed
12Petrof, B.J. et al. (1993) Dystrophin protects the sarcolemma from stresses developed during muscle contraction. Proceedings of the National Academy of Sciences of the United States of America 90, 3710-3714CrossRefGoogle Scholar
13Turner, P.R. et al. (1988) Increased protein degradation results from elevated free calcium levels found in muscle from mdx mice. Nature 335, 735-738CrossRefGoogle ScholarPubMed
14Batchelor, C.L. and Winder, S.J. (2006) Sparks, signals and shock absorbers: how dystrophin loss causes muscular dystrophy. Trends in Cell Biology 16, 198-205CrossRefGoogle ScholarPubMed
15Whitehead, N.P. et al. (2006) Streptomycin reduces stretch-induced membrane permeability in muscles from mdx mice. Neuromuscular Disorders 16, 845-854Google Scholar
16Baumbach, L.L. et al. (1989) Molecular and clinical correlations of deletions leading to Duchenne and Becker muscular dystrophies. Neurology 39, 465-474Google Scholar
17Ozawa, E. et al. (1998) From dystrophinopathy to sarcoglycanopathy: Evolution of a concept of muscular dystrophy. Muscle & Nerve 21, 421-4383.0.CO;2-B>CrossRefGoogle ScholarPubMed
18Campbell, K.P. (1995) Three muscular dystrophies: Loss of cytoskeleton-extracellular matrix linkage. Cell 80, 675-679CrossRefGoogle ScholarPubMed
19Manzur, A. et al. (2008) Glucocorticoid corticosteroids for Duchenne muscular dystrophy Cochrane Database of Systematic Reviews 2008 1 (CD003725), DOI: 10.1002/14651858.CD14003725.pub14651853.CrossRefGoogle Scholar
20Chamberlain, J.S. and Rando, T.A., eds (2006) Duchenne Muscular Dystrophy: Advances in Therapeutics. Taylor & Francis Group, New YorkCrossRefGoogle Scholar
21Banks, G.B., Chamberlain, J.S. and Krauss, R.S. (2008) Chapter 9, The value of mammalian models for Duchenne muscular dystrophy in developing therapeutic strategies. Current Topics in Developmental Biology 84, 431-453Google Scholar
22Chamberlain, J. and Caskey, C.T. (1990) Duchenne Muscular Dystrophy. In Current Neurology 10 (Appel, S.H., eds), pp. 65-103, Yearbook Medical Publishers, ChicagoGoogle Scholar
23Chamberlain, J.S. (2002) Gene therapy of muscular dystrophy. Human Molecular Genetics 11, 2355-2362CrossRefGoogle ScholarPubMed
24England, S.B. (1990) Very mild muscular dystrophy associated with the deletion of 46% of dystrophin. Nature 343, 180-182CrossRefGoogle ScholarPubMed
25Matsumura, K. et al. (1994) Immunohistochemical analysis of dystrophin-associated proteins in Becker/Duchenne muscular dystrophy with huge in-frame deletions in the NH2-terminal and rod domains of dystrophin. Journal of Clinical Investigation 93, 99-105CrossRefGoogle ScholarPubMed
26Harper, S.Q. et al. (2002) Modular flexibility of dystrophin: Implications for gene therapy of Duchenne muscular dystrophy. Nature Medicine 8, 253-261CrossRefGoogle ScholarPubMed
27Phelps, S.F. et al. (1995) Expression of full-length and truncated dystrophin mini-genes in transgenic mdx mice. Human Molecular Genetics 4, 1251-1258CrossRefGoogle ScholarPubMed
28Sakamoto, M. (2002) Micro-dystrophin cDNA ameliorates dystrophic phenotypes when introduced into mdx mice as a transgene. Biochemical and Biophysical Research Communications 293, 1265-1272CrossRefGoogle ScholarPubMed
29Wang, B., Li, J. and Xiao, X. (2000) Adeno-associated virus vector carrying human minidystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model. Proceedings of the National Academy of Sciences of the United States of America 97, 13714-13719CrossRefGoogle Scholar
30Ishikawa-Sakurai, M. et al. (2004) ZZ domain is essentially required for the physiological binding of dystrophin and utrophin to {beta}-dystroglycan. Human Molecular Genetics 13, 693-702Google Scholar
31Huard, J. et al. (1992) Human myoblast transplantation between immunohistocompatible donors and recipients produces immune reactions. Transplant Proceedings 24, 3049-3051Google ScholarPubMed
32Ohtsuka, Y. et al. (1998) Dystrophin acts as a transplantation rejection antigen indystrophin-deficient mice: implication for gene therapy. Journal of Immunology 160, 4635-4640Google Scholar
33Gilchrist, S.C. et al. (2002) Immune response to full-length dystrophin delivered to DMD muscle by a high-capacity adenoviral vector. Molecular Therapy 6, 359-368Google Scholar
34Wells, D.J., Ferrer, A. and Wells, K.E. (2002) Immunological hurdles in the path to gene therapy for Duchenne muscular dystrophy. Expert Reviews in Molecular Medicine 4, 1-23CrossRefGoogle ScholarPubMed
35Tinsley, J.M. et al. (1992) Primary structure of dystrophin-related protein. Nature 360, 591-593CrossRefGoogle ScholarPubMed
36Matsumura, K. et al. (1992) Association of dystrophin-related protein with dystrophin-associated proteins in mdx mouse muscle. Nature 360, 588-591CrossRefGoogle ScholarPubMed
37Winder, S.J. et al. (1995) Utrophin actin binding domain: analysis of actin binding and cellular targeting. Journal of Cell Science 108, 63-71CrossRefGoogle ScholarPubMed
38Nguyen, T.M. (1991) Localization of the DMDL gene-encoded dystrophin-related protein using a panel of nineteen monoclonal antibodies: presence at neuromuscular junctions, in the sarcolemma of dystrophic skeletal muscle, in vascular and other smooth muscles, and in proliferating brain cell lines. Journal of Cell Biology 115, 1695-1700CrossRefGoogle ScholarPubMed
39Khurana, T.S. et al. (1991) Immunolocalization and developmental expression of dystrophin related protein in skeletal muscle. Neuromuscular Disorders 1, 185-194CrossRefGoogle ScholarPubMed
40Mizuno, Y. et al. (1993) Reciprocal expression of dystrophin and utrophin in muscles of Duchenne muscular dystrophy patients, female DMD-carriers and control subjects. Journal of the Neurological Sciences 119, 43-52Google Scholar
41Weir, A.P., Morgan, J.E. and Davies, K.E. (2004) A-utrophin up-regulation in mdx skeletal muscle is independent of regeneration. Neuromuscular Disorders 14, 19-23CrossRefGoogle ScholarPubMed
42Deol, J.R. et al. (2007) Successful compensation for dystrophin deficiency by a helper-dependent adenovirus expressing full-length utrophin. Molecular Therapy 15, 1767-1774CrossRefGoogle ScholarPubMed
43Tinsley, J.M. (1996) Amelioration of the dystrophic phenotype of mdx mice using a truncated utrophin transgene. Nature 384, 349-353CrossRefGoogle ScholarPubMed
44Deconinck, A.E. (1997) Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell 90, 717-727CrossRefGoogle Scholar
45Cerletti, M. (2003) Dystrophic phenotype of canine X-linked muscular dystrophy is mitigated by adenovirus-mediated utrophin gene transfer. Gene Therapy 10, 750-757CrossRefGoogle ScholarPubMed
46Odom, G.L. et al. (2008) Microutrophin delivery through rAAV6 increases lifespan and improves muscle function in dystrophic dystrophin/utrophin-deficient mice. Molecular Therapy 16, 1539-1545CrossRefGoogle ScholarPubMed
47Atchison, R., Casto, B. and Hammon, W. (1965) Adenovirus-associated defective virus particles. Science 13, 754-756CrossRefGoogle Scholar
48Muzyczka, N. and Berns, K.I. (2001) Parvoviridae: the viruses and their replication. In Fields Virology (4th edn), (Knipe, D. and Howley, P., eds), pp. 2327-2359, Lippincott Williams & Wilkins, PhiladelphiaGoogle Scholar
49Podsakoff, G., Wong, K.K. Jr. and Chatterjee, S. (1994) Efficient gene transfer into nondividing cells by adeno-associated virus-based vectors. Journal of Virology 68, 5656-5666CrossRefGoogle ScholarPubMed
50Gregorevic, P. et al. (2004) Systemic delivery of genes to striated muscles using adeno-associated viral vectors. Nature Medicine 10, 828-834Google Scholar
51Gao, G. et al. (2004) Clades of adeno-associated viruses are widely disseminated in human tissues. Journal of Virology 78, 6381-6388CrossRefGoogle ScholarPubMed
52Chao, H. et al. (2000) Several log increase in therapeutic transgene delivery by distinct adeno-associated viral serotype vectors. Molecular Therapy 2, 619-623CrossRefGoogle ScholarPubMed
53Duan, D. et al. (2001) Enhancement of muscle gene delivery with pseudotyped adeno-associated virus type 5 correlates with myoblast differentiation. Journal of Virology 75, 7662-7671CrossRefGoogle ScholarPubMed
54Grimm, D. et al. (2003) Preclinical in vivo evaluation of pseudotyped adeno-associated virus vectors for liver gene therapy. Blood 102, 2412-2419CrossRefGoogle ScholarPubMed
55Halbert, C.L., Allen, J.M. and Miller, A.D. (2001) Adeno-associated virus type 6 (AAV6) vectors mediate efficient transduction of airway epithelial cells in mouse lungs compared to that of AAV2 vectors. Journal of Virology 75, 6615-6624CrossRefGoogle ScholarPubMed
56Duan, D. et al. (1998) Circular intermediates of recombinant adeno-associated virus have defined structural characteristics responsible for long-term episomal persistence in muscle tissue. Journal of Virology 72, 8568-8577CrossRefGoogle ScholarPubMed
57Schnepp, B.C. et al. (2009) Infectious molecular clones of adeno-associated virus isolated directly from human tissues. Journal of Virology 83, 1456-1464Google Scholar
58Chamberlain, J.R. et al. (2004) Gene targeting in stem cells from individuals with osteogenesis imperfecta. Science 303, 1198-1201Google Scholar
59Inagaki, K. et al. (2007) DNA palindromes with a modest arm length of greater than or equal to 20 base pairs are a significant target for recombinant adeno-associated virus vector integration in the liver, muscles, and heart in mice. Journal of Virology 81, 11290-11303Google Scholar
60Herzog, R.W. et al. (1999) Long-term correction of canine hemophilia B by gene transfer of blood coagulation factor IX mediated by adeno-associated viral vector. Nature Medicine 5, 56-63Google Scholar
61Manno, C.S. (2003) AAV-mediated factor IX gene transfer to skeletal muscle in patients with severe hemophilia B. Blood 101, 2963-2972CrossRefGoogle ScholarPubMed
62Yuasa, K. (2002) Adeno-associated virus vector-mediated gene transfer into dystrophin-deficient skeletal muscles evokes enhanced immune response against the transgene product. Gene Therapy 9, 1576-1588CrossRefGoogle ScholarPubMed
63Hartigan-O'Connor, D. et al. (2001) Immune evasion by muscle-specific gene expression in dystrophic muscle. Molecular Therapy 4, 525-533CrossRefGoogle ScholarPubMed
64Wang, Z. et al. (2007) Sustained AAV-mediated dystrophin expression in a canine model of Duchenne muscular dystrophy with a brief course of immunosuppression. Molecular Therapy 15, 1160-1166CrossRefGoogle Scholar
65Manno, C.S. et al. (2006) Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response. Nature Medicine 12, 342-347CrossRefGoogle ScholarPubMed
66Mingozzi, F. and High, K. (2007) Immune responses to AAV in clinical trials. Current Gene Therapy 7, 316-324CrossRefGoogle ScholarPubMed
67Mingozzi, F. et al. (2007) CD8+ T-cell responses to adeno-associated virus capsid in humans. Nature Medicine 13, 419-422Google Scholar
68Jiang, H. et al. (2006) Effects of transient immunosuppression on adeno-associated virus-mediated liver-directed gene transfer in rhesus macaques and implications for human gene therapy. Blood 108, 3321-3328CrossRefGoogle ScholarPubMed
69Townsend, D. et al. (2008) Emergent dilated cardiomyopathy caused by targeted repair of dystrophic skeletal muscle. Molecular Therapy 16, 832-835CrossRefGoogle ScholarPubMed
70Mah, C. et al. (2005) Sustained correction of glycogen storage disease type II using adeno-associated virus serotype 1 vectors. Gene Therapy 12, 1405-1409Google Scholar
71Wang, Z. et al. (2005) Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart. Nature Biotechnology 23, 321-328CrossRefGoogle ScholarPubMed
72Inagaki, K. et al. (2006) Robust systemic transduction with AAV9 vectors in mice: efficient global cardiac gene transfer superior to that of AAV8. Molecular Therapy 14, 45-53CrossRefGoogle ScholarPubMed
73Yue, Y. et al. (2008) A single intravenous injection of adeno-associated virus serotype-9 leads to whole body skeletal muscle transduction in dogs. Molecular Therapy 16, 1944-1952Google Scholar
74Schultz, B.R. and Chamberlain, J.S. (2008) Recombinant adeno-associated virus transduction and integration. Molecular Therapy 16, 1189-1199Google Scholar
75Banks, G.B. et al. (2007) Functional capacity of dystrophins carrying deletions in the N-terminal actin-binding domain. Human Molecular Genetics 16, 2105-2113Google Scholar
76Wells, K.E. et al. (2003) Relocalization of neuronal nitric oxide synthase (nNOS) as a marker for complete restoration of the dystrophin associated protein complex in skeletal muscle. Neuromuscular Disorders 13, 21-31CrossRefGoogle ScholarPubMed
77Judge, L.M., Haraguchi, M. and Chamberlain, J.S. (2006) Dissecting the signaling and mechanical functions of the dystrophin-glycoprotein complex. Journal of Cell Science 119, 1537-1546CrossRefGoogle ScholarPubMed
78Lai, Y. et al. (2009) Dystrophins carrying spectrin-like repeats 16 and 17 anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model of muscular dystrophy. Journal of Clinical Investigation 119, 624-635Google Scholar
79Thomas, G.D. et al. (1998) Impaired metabolic modulation of alpha-adrenergic vasoconstriction in dystrophin-deficient skeletal muscle. Proceedings of the National Academy of Sciences of the United States of America 95, 15090-15095CrossRefGoogle Scholar
80Kobayashi, Y.M. et al. (2008) Sarcolemma-localized nNOS is required to maintain activity after mild exercise. Nature 456, 511-515Google Scholar
81Banks, G.B. et al. (2008) Molecular and cellular adaptations to chronic myotendinous strain injury in mdx mice expressing a truncated dystrophin. Human Molecular Genetics 17, 3975-3986Google Scholar
82Banks, G.B., Chamberlain, J.S. and Froehner, S.C. (2009) Truncated dystrophins can influence neuromuscular synapse structure. Molecular and Cellular Neuroscience 40, 433-441Google Scholar
83Sun, L., Li, J. and Xiao, X. (2000) Overcoming adeno-associated virus vector size limitation through viral DNA heterodimerization. Nature Medicine 6, 599-602CrossRefGoogle ScholarPubMed
84Yan, Z. et al. (2000) Trans-splicing vectors expand the utility of adeno-associated virus for gene therapy. Proceedings of the National Academy of Sciences of the United States of America 97, 6716-6721Google Scholar
85Nakai, H., Storm, T.A. and Kay, M.A. (2000) Increasing the size of rAAV-mediated expression cassettes in vivo by intermolecular joining of two complementary vectors. Nature Biotechnology 18, 527-532CrossRefGoogle ScholarPubMed
86Duan, D., Yue, Y. and Engelhardt, J.F. (2001) Expanding AAV packaging capacity with trans-splicing or overlapping vectors: a quantitative comparison. Molecular Therapy 4, 383-391CrossRefGoogle ScholarPubMed
87Lai, Y. et al. (2006) Synthetic intron improves transduction efficiency of trans-splicing adeno-associated viral vectors. Human Gene Therapy 17, 1036-1042CrossRefGoogle ScholarPubMed
88Halbert, C.L., Allen, J.M. and Miller, A.D. (2002) Efficient mouse airway transduction following recombination between AAV vectors carrying parts of a larger gene. Nature Biotechnology 20, 697-701CrossRefGoogle ScholarPubMed
89Ghosh, A. et al. (2007) A hybrid vector system expands adeno-associated viral vector packaging capacity in a transgene-independent manner. Molecular Therapy 16, 124-130CrossRefGoogle Scholar
90Li, S. et al. (2005) Stable transduction of myogenic cells with lentiviral vectors expressing a minidystrophin. Gene Therapy 12, 1099-1108CrossRefGoogle ScholarPubMed
91Hacein-Bey-Abina, S. et al. (2003) A serious adverse event after successful gene therapy for X-linked severe combined immunodeficiency. New England Journal of Medicine 348, 255-256CrossRefGoogle ScholarPubMed
92Ciuffi, A. et al. (2006) Integration site selection by HIV-based vectors in dividing and growth-arrested IMR-90 lung fibroblasts. Molecular Therapy 13, 366-373CrossRefGoogle ScholarPubMed
93Beard, B.C. et al. (2007) Unique integration profiles in a canine model of long-term repopulating cells transduced with gammaretrovirus, lentivirus, or foamy virus. Human Gene Therapy 18, 423-434CrossRefGoogle ScholarPubMed
94Barquinero, J., Eixarch, H. and Perez-Melgosa, M.Retroviral vectors: new applications for an old tool. Gene Therapy 11, S3-S9CrossRefGoogle Scholar
95Kafri, T. et al. (1997) Sustained expression of genes delivered directly into liver and muscle by lentiviral vectors. Nature Genetics 17, 314-317CrossRefGoogle ScholarPubMed
96Bonci, D. et al. (2003) ‘Advanced’ generation lentiviruses as efficient vectors for cardiomyocyte gene transduction in vitro and in vivo. Gene Therapy 10, 630-636CrossRefGoogle ScholarPubMed
97Naldini, L. et al. (1996) Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proceedings of the National Academy of Sciences of the United States of America 93, 11382-11388CrossRefGoogle Scholar
98Annoni, A. et al. (2007) The immune response to lentiviral-delivered transgene is modulated in vivo by transgene-expressing antigen-presenting cells but not by CD4 + CD25+ regulatory T cells. Blood 110, 1788-1796CrossRefGoogle Scholar
99Bachrach, E. et al. (2006) Muscle engraftment of myogenic progenitor cells following intraarterial transplantation. Muscle & Nerve 34, 44-52CrossRefGoogle ScholarPubMed
100Dellavalle, A. et al. (2007) Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nature Cell Biology 9, 255-267Google Scholar
101Kumar-Singh, R. and Chamberlain, J.S. (1996) Encapsidated adenovirus minichromosomes allow delivery and expression of a 14 kb dystrophin cDNA to muscle cells. Human Molecular Genetics 5, 913-921Google Scholar
102Clemens, P. et al. (1996) In vivo muscle gene transfer of full-length dystrophin with an adenoviral vector that lacks all viral genes. Gene Therapy 3, 965-972Google ScholarPubMed
103Chen, H.-H. et al. (1997) Persistence in muscle of an adenoviral vector that lacks all viral genes. Proceedings of the National Academy of Sciences of the United States of America 94, 1645-1650CrossRefGoogle Scholar
104Hartigan-O'Connor, D. et al. (2002) Generation and growth of adenoviral vectors. Methods in Enzymology 346, 224-246Google Scholar
105DelloRusso, C. et al. (2002) Functional correction of adult mdx mouse muscle using gutted adenoviral vectors expressing full-length dystrophin. Proceedings of the National Academy of Sciences of the United States of America 99, 12979-12984CrossRefGoogle Scholar
106Gilbert, R. et al. (2003) Prolonged dystrophin expression and functional correction of mdx mouse muscle following gene transfer with a helper-dependent (gutted) adenovirus-encoding murine dystrophin. Human Molecular Genetics 12, 1287-1299Google Scholar
107Su, L.T. et al. (2005) Uniform scale-independent gene transfer to striated muscle after transvenular extravasation of vector. Circulation 112, 1780-1788Google Scholar
108Gao, G.P., Yang, Y. and Wilson, J.M. (1996) Biology of adenovirus vectors with E1 and E4 deletions for liver- directed gene therapy. Journal of Virology 70, 8934-8943Google Scholar
109Acsadi, G. et al. (1994) A differential efficiency of adenovirus-mediated in vivo gene transfer into skeletal muscle cells of different maturity. Human Molecular Genetics 3, 579-584CrossRefGoogle ScholarPubMed
110Zoltick, P.W. et al. (2001) Biology of E1-deleted adenovirus vectors in nonhuman primate muscle. Journal of Virology 75, 5222-5229Google Scholar
111Muruve, D.A. et al. (1999) Adenoviral gene therapy leads to rapid induction of multiple chemokines and acute neutrophil-dependent hepatic injury in vivo. Human Gene Therapy 10, 965-976CrossRefGoogle ScholarPubMed
112Brunetti-Pierri, N. et al. (2004) Acute toxicity after high-dose systemic injection of helper-dependent adenoviral vectors into nonhuman primates. Human Gene Therapy 15, 35-46CrossRefGoogle ScholarPubMed
113Raper, S.E. et al. (2003) Fatal systemic inflammatory response syndrome in a ornithine transcarbamylase deficient patient following adenoviral gene transfer. Molecular Genetics and Metabolism 80, 148-158Google Scholar
114Morral, N. et al. (1999) Administration of helper-dependent adenoviral vectors and sequential delivery of different vector serotype for long-term liver-directed gene transfer in baboons. Proceedings of the National Academy of Sciences of the United States of America 96, 12816-12821CrossRefGoogle Scholar
115Wolff, J. et al. (1990) Direct gene transfer into mouse muscle in vivo. Science 247, 1465-1468CrossRefGoogle ScholarPubMed
116Zhang, G. et al. (2004) Intraarterial delivery of naked plasmid DNA expressing full-length mouse dystrophin in the mdx mouse model of Duchenne muscular dystrophy. Human Gene Therapy 15, 770-782Google Scholar
117Hagstrom, J.E. et al. (2004) A facile nonviral method for delivering genes and siRNAs to skeletal muscle of mammalian limbs. Molecular Therapy 10, 386-398Google Scholar
118Richard, P. et al. (2005) Amphiphilic block copolymers promote gene delivery in vivo to pathological skeletal muscles. Human Gene Therapy 16, 1318-1324CrossRefGoogle ScholarPubMed
119Chollet, P. et al. (2002) Side-effects of a systemic injection of linear polyethylenimine-DNA complexes. The Journal of Gene Medicine 4, 84-91Google Scholar
120Trubetskoy, V.S. et al. (2003) Recharging cationic DNA complexes with highly charged polyanions for in vitro and in vivo gene delivery. Gene Therapy 10, 261-271CrossRefGoogle ScholarPubMed
121Romero, N.B. et al. (2004) Phase I study of dystrophin plasmid-based gene therapy in Duchenne/Becker muscular dystrophy. Human Gene Therapy 15, 1065-1076Google Scholar
122Sherratt, T.G. et al. (1993) Exon skipping and translation in patients with frameshift deletions in the dystrophin gene. American Journal of Human Genetics 53, 1007-1015Google Scholar
123Partridge, T. and Lu, Q.-L. (2008) The enigma of the ‘dystrophin revertant’ muscle fibre. In Recent advances in skeletal muscle differentiation (Tsuchida, K. and Takeda, S., eds), pp. 93-107, Research Signpost, TrivandrumGoogle Scholar
124Dominski, Z. and Kole, R. (1993) Restoration of correct splicing in thalassemic pre-mRNA by antisense oligonucleotides. Proceedings of the National Academy of Sciences of the United States of America 90, 8673-8677Google Scholar
125Takeshima, Y. et al. (1995) 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. Journal of Clinical Investigation 95, 515-520Google Scholar
126Pramono, Z.A. (1996) Induction of exon skipping of the dystrophin transcript in lymphoblastoid cells by transfecting an antisense oligodeoxynucleotide complementary to an exon recognition sequence. Biochemical and Biophysical Research Communications 226, 445-449Google Scholar
127Shibahara, S. et al. (1989) Inhibition of human immunodeficiency virus (HIV-1) replication by synthetic oligo-RNA derivatives. Nucleic Acids Research 17, 239-252CrossRefGoogle ScholarPubMed
128Dunckley, M.G. et al. (1998) Modification of splicing in the dystrophin gene in cultured Mdx muscle cells by antisense oligoribonucleotides. Human Molecular Genetics 7, 1083-1090Google Scholar
129McClorey, G. et al. (2006) Antisense oligonucleotide-induced exon skipping restores dystrophin expression in vitro in a canine model of DMD. Gene Therapy 13, 1373-1381CrossRefGoogle Scholar
130van Deutekom, J.C. (2001) Antisense-induced exon skipping restores dystrophin expression in DMD patient derived muscle cells. Human Molecular Genetics 10, 1547-1554CrossRefGoogle ScholarPubMed
131Mann, C.J. et al. (2001) Antisense-induced exon skipping and synthesis of dystrophin in the mdx mouse. Proceedings of the National Academy of Sciences of the United States of America 98, 42-47Google Scholar
132Summerton, J. and Weller, D. (1997) Morpholino antisense oligomers: design, preparation, and properties. Antisense and Nucleic Acid Drug Development 7, 187-195CrossRefGoogle ScholarPubMed
133Yokota, T. et al. (2009) A renaissance for antisense oligonucleotide drugs in neurology: exon skipping breaks new ground. Archives of Neurology 66, 32-38Google Scholar
134Sazani, P. et al. (2002) Systemically delivered antisense oligomers upregulate gene expression in mouse tissues. Nature Biotechnology 20, 1228-1233CrossRefGoogle ScholarPubMed
135Lebleu, B. et al. (2008) Cell penetrating peptide conjugates of steric block oligonucleotides. Advanced Drug Delivery Reviews 60, 517-529CrossRefGoogle ScholarPubMed
136Wu, B. et al. (2009) Octa-guanidine morpholino restores dystrophin expression in cardiac and skeletal muscles and ameliorates pathology in dystrophic mdx mice. Molecular Therapy 17, 864-871CrossRefGoogle ScholarPubMed
137Ivanova, G.D. et al. (2008) Improved cell-penetrating peptide-PNA conjugates for splicing redirection in HeLa cells and exon skipping in mdx mouse muscle. Nucleic Acids Research 36, 6418-6428Google Scholar
138Alter, J. et al. (2006) Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology. Nature Medicine 12, 175-177CrossRefGoogle ScholarPubMed
139Fletcher, S. et al. (2007) Morpholino oligomer-mediated exon skipping averts the onset of dystrophic pathology in the mdx mouse. Molecular Therapy 15, 1587-1592Google Scholar
140Lu, Q.L. et al. (2005) Systemic delivery of antisense oligoribonucleotide restores dystrophin expression in body-wide skeletal muscles. Proceedings of the National Academy of Sciences of the United States of America 102, 198-203CrossRefGoogle Scholar
141Jearawiriyapaisarn, N. et al. (2008) Sustained dystrophin expression induced by peptide-conjugated morpholino oligomers in the muscles of mdx mice. Molecular Therapy 16, 1624-1629Google Scholar
142Wu, B. et al. (2008) Effective rescue of dystrophin improves cardiac function in dystrophin-deficient mice by a modified morpholino oligomer. Proceedings of the National Academy of Sciences of the United States of America 105, 14814-14819Google Scholar
143Popplewell, L.J. et al. (2009) Design of phosphorodiamidate morpholino oligomers (PMOs) for the induction of exon skipping of the human DMD gene. Molecular Therapy 17, 554-561Google Scholar
144Mitrpant, C. et al. (2009) By-passing the nonsense mutation in the 4 (CV) mouse model of muscular dystrophy by induced exon skipping. The Journal of Gene Medicine 11, 46-56Google Scholar
145De Angelis, F.G. (2002) Chimeric snRNA molecules carrying antisense sequences against the splice junctions of exon 51 of the dystrophin pre-mRNA induce exon skipping and restoration of a dystrophin synthesis in [delta] 48-50 DMD cells. Proceedings of the National Academy of Sciences of the United States of America 99, 9456-9461Google Scholar
146Goyenvalle, A. et al. (2004) Rescue of Dystrophic Muscle Through U7 snRNA-Mediated Exon Skipping. Science 306, 1796-1799Google Scholar
147Denti, M.A. et al. (2006) Chimeric adeno-associated virus/antisense U1 small nuclear RNA effectively rescues dystrophin synthesis and muscle function by local treatment of mdx mice. Human Gene Therapy 17, 565-574Google Scholar
148Heemskerk, H.A. et al. (2009) In vivo comparison of 2'-O-methyl phosphorothioate and morpholino antisense oligonucleotides for Duchenne muscular dystrophy exon skipping. The Journal of Gene Medicine 11, 257-266Google Scholar
149Skuk, D. and Tremblay, J.P. (2003) Myoblast transplantation: the current status of a potential therapeutic tool for myopathies. Journal of Muscle Research and Cell Motility 24, 285-300CrossRefGoogle ScholarPubMed
150Skuk, D. et al. (2007) First test of a “high-density injection” protocol for myogenic cell transplantation throughout large volumes of muscles in a Duchenne muscular dystrophy patient: eighteen months follow-up. Neuromuscular Disorders 17, 38-46Google Scholar
151Skuk, D. et al. (2007) Ischemic central necrosis in pockets of transplanted myoblasts in nonhuman primates: implications for cell-transplantation strategies. Transplantation 84, 1307-1315Google Scholar
152Montarras, D. et al. (2005) Direct Isolation of Satellite Cells for Skeletal Muscle Regeneration. Science 309, 2064-2067CrossRefGoogle ScholarPubMed
153Peault, B. et al. (2007) Stem and progenitor cells in skeletal muscle development, maintenance, and therapy. Molecular Therapy 15, 867-877Google Scholar
154Webster, C. and Blau, H. (1990) Accelerated age-related decline in replicative life-span of Duchenne muscular dystrophy myoblasts: implications for cell and gene therapy. Somatic Cell and Molecular Genetics 16, 557-565Google Scholar
155Fan, Y. et al. (1996) Rapid death of injected myoblasts in myoblast transfer therapy. Muscle & Nerve 19, 853-860Google Scholar
156Tremblay, J.P. and Skuk, D. (2008) Another new “super muscle stem cell” leaves unaddressed the real problems of cell therapy for Duchenne muscular dystrophy. Molecular Therapy 16, 1907-1909Google Scholar
157Deasy, B.M. et al. (2002) Mechanisms of muscle stem cell expansion with cytokines. Stem Cells 20, 50-60CrossRefGoogle ScholarPubMed
158Collins, C.A. et al. (2005) Stem cell function, self-renewal, and behavioral heterogeneity of cells from the adult muscle satellite cell niche. Cell 122, 289-301Google Scholar
159Cornelison, D.D.W. (2008) Context matters: In vivo and in vitro influences on muscle satellite cell activity. Journal of Cellular Biochemistry 105, 663-669Google Scholar
160Gussoni, E. et al. (1999) Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature 401, 390-394Google Scholar
161Lee, J.Y. et al. (2000) Clonal isolation of muscle-derived cells capable of enhancing muscle regeneration and bone healing. Journal of Cell Biology 150, 1085-1100Google Scholar
162Torrente, Y. et al. (2007) Autologous transplantation of muscle-derived CD133+ stem cells in Duchenne muscle patients. Cell Transplantation 16, 563-577Google Scholar
163Ferrari, G. et al. (1998) Muscle regeneration by bone marrow-derived myogenic progenitors. Science 279, 1528-1530Google Scholar
164Dezawa, M. et al. (2005) Bone marrow stromal cells generate muscle cells and repair muscle degeneration. Science 309, 314-317Google Scholar
165Sampaolesi, M. et al. (2003) Cell therapy of {alpha}-sarcoglycan null dystrophic mice through intra-arterial delivery of mesoangioblasts. Science 301, 487-492Google Scholar
166Sampaolesi, M. et al. (2006) Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs. Nature 444, 574-579Google Scholar
167Barberi, T. et al. (2007) Derivation of engraftable skeletal myoblasts from human embryonic stem cells. Nature Medicine 13, 642-648Google Scholar
168Darabi, R. et al. (2008) Functional skeletal muscle regeneration from differentiating embryonic stem cells. Nature Medicine 14, 134-143Google Scholar
169Kimura, E. et al. (2008) Cell-lineage regulated myogenesis for dystrophin replacement: a novel therapeutic approach for treatment of muscular dystrophy. Human Molecular Genetics 17, 2507-2517CrossRefGoogle ScholarPubMed
170Takahashi, K. et al. (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861-872Google Scholar
171Yu, J. et al. (2007) Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917-1920Google Scholar
172Rebuzzini, P. et al. (2008) Chromosome number variation in three mouse embryonic stem cell lines during culture. Cytotechnology 58, 17-23Google Scholar
173Catalina, P. et al. (2008) Human ESCs predisposition to karyotypic instability: Is a matter of culture adaptation or differential vulnerability among hESC lines due to inherent properties? Molecular Cancer 7, 76Google Scholar
174Hanna, J. et al. (2007) Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science 318, 1920-1923Google Scholar
175Dickson, G. et al. (2002) Recombinant micro-genes and dystrophin viral vectors. Neuromuscular Disorders 12, S40-S44Google Scholar
176Goncalves, M.A.F.V. et al. (2008) Targeted chromosomal insertion of large DNA into the human genome by a fiber-modified high-capacity adenovirus-based vector system. PLoS ONE 3, 3084Google Scholar
177Salva, M.Z. et al. (2007) Design of tissue-specific regulatory cassettes for high-level rAAV-mediated expression in skeletal and cardiac muscle. Molecular Therapy 15, 320-329Google Scholar
178Zaldumbide, A. and Hoeben, R.C. (2007) How not to be seen: immune-evasion strategies in gene therapy. Gene Therapy 15, 239-246Google Scholar
179van Deutekom, J.C. et al. (2007) Local dystrophin restoration with antisense oligonucleotide PRO051. New England Journal of Medicine 357, 2677-2686Google Scholar
180Newey, S.E. et al. (2000) Alternative splicing of dystrobrevin regulates the stoichiometry of syntrophin binding to the dystrophin protein complex. Current Biology 10, 1295-1298Google Scholar
181Adams, M.E., Mueller, H.A. and Froehner, S.C. (2001) In vivo requirement of the {alpha}-syntrophin PDZ domain for the sarcolemmal localization of nNOS and aquaporin-4. Journal of Cell Biology 155, 113-122Google Scholar
182Gee, S.H. et al. (1998) Interaction of muscle and brain sodium channels with multiple members of the syntrophin family of dystrophin-associated proteins. Journal of Neuroscience 18, 128-137Google Scholar

Further reading, resources and contacts

Blake, D.J. et al. (2002) Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiological Reviews 82, 291-329CrossRefGoogle ScholarPubMed
Abmayr, S. and Chamberlain, J.S. (2006) The structure and function of dystrophin. In The Molecular Mechanisms in Muscular Dystrophy (Winder, S.J., eds), Landes Biosciences, Georgetown.Google Scholar
Chamberlain, J.S. and Rando, T.A., eds (2006) Duchenne muscular dystrophy: advances in therapeutics. Taylor & Francis Group, New York.Google Scholar
Odom, G. L., Gregorevic, P. and Chamberlain, J. S. (2007) Viral-mediated gene therapy for the muscular dystrophies: Successes, limitations and recent advances. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1772, 243-262Google Scholar
Online Mendelian Inheritance in Man (OMIM) information on the dystrophin gene (DMD), available from the National Center for Biotechnology Information (NCBI), no. 300377:Google Scholar
Blake, D.J. et al. (2002) Function and genetics of dystrophin and dystrophin-related proteins in muscle. Physiological Reviews 82, 291-329CrossRefGoogle ScholarPubMed
Abmayr, S. and Chamberlain, J.S. (2006) The structure and function of dystrophin. In The Molecular Mechanisms in Muscular Dystrophy (Winder, S.J., eds), Landes Biosciences, Georgetown.Google Scholar
Chamberlain, J.S. and Rando, T.A., eds (2006) Duchenne muscular dystrophy: advances in therapeutics. Taylor & Francis Group, New York.Google Scholar
Odom, G. L., Gregorevic, P. and Chamberlain, J. S. (2007) Viral-mediated gene therapy for the muscular dystrophies: Successes, limitations and recent advances. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1772, 243-262Google Scholar
Online Mendelian Inheritance in Man (OMIM) information on the dystrophin gene (DMD), available from the National Center for Biotechnology Information (NCBI), no. 300377:Google Scholar