M. Koenig, E. P. Hoffman, C. J. Bertelson, A. P. Monaco, C. Feener et al., Complete cloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals, Cell, vol.50, issue.3, pp.509-526, 1987.

E. P. Hoffman, R. H. Brown, and L. M. Kunkel, Dystrophin: the protein product of the Duchenne muscular dystrophy locus, Cell, vol.51, issue.6, pp.919-947, 1987.

E. Bonilla, C. E. Samitt, A. F. Miranda, A. P. Hays, G. Salviati et al., Duchenne muscular dystrophy: deficiency of dystrophin at the muscle cell surface, Cell, vol.54, issue.4, pp.447-52, 1988.

E. E. Zubrzycka-gaarn, D. E. Bulman, G. Karpati, A. H. Burghes, B. Belfall et al., The Duchenne muscular dystrophy gene product is localized in sarcolemma of human skeletal muscle, Nature, vol.333, issue.6172, pp.466-475, 1988.

I. N. Rybakova, J. R. Patel, and J. M. Ervasti, The dystrophin complex forms a mechanically strong link between the sarcolemma and costameric actin, J Cell Biol, vol.150, issue.5, pp.1209-1223, 2000.

K. Matsumura, F. M. Tomé, V. Ionasescu, J. M. Ervasti, R. D. Anderson et al., Deficiency of dystrophin-associated proteins in Duchenne muscular dystrophy patients lacking COOH-terminal domains of dystrophin, J Clin Invest, vol.92, issue.2, pp.866-71, 1993.

K. S. Ramaswamy, M. L. Palmer, J. H. Van-der-meulen, A. Renoux, T. Y. Kostrominova et al., Lateral transmission of force is impaired in skeletal muscles of dystrophic mice and very old rats, J Physiol, vol.589, pp.1195-208, 2011.

H. D. Geissinger, P. V. Rao, and C. K. Mcdonald-taylor, mdx' mouse myopathy: histopathological, morphometric and histochemical observations on young mice, J Comp Pathol, vol.102, issue.3, pp.249-63, 1990.

N. Mallouk, V. Jacquemond, and B. Allard, Elevated subsarcolemmal Ca2+ in mdx mouse skeletal muscle fibers detected with Ca2+-activated K+ channels, Proc Natl Acad Sci, vol.97, issue.9, pp.4950-4955, 2000.

D. G. Allen, O. L. Gervasio, E. W. Yeung, and N. P. Whitehead, Calcium and the damage pathways in muscular dystrophy, Can J Physiol Pharmacol, vol.88, issue.2, pp.83-91, 2010.

A. Ascah, M. Khairallah, F. Daussin, C. Bourcier-lucas, R. Godin et al., Stress-induced opening of the permeability transition pore in the dystrophin-deficient heart is attenuated by acute treatment with sildenafil, Am J Physiol Heart Circ Physiol, vol.300, issue.1, pp.144-53, 2011.

M. Pauly, F. Daussin, Y. Burelle, T. Li, R. Godin et al., AMPK activation stimulates autophagy and ameliorates muscular dystrophy in the mdx mouse diaphragm, Am J Pathol, vol.181, issue.2, pp.583-92, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02546568

J. M. Percival, M. P. Siegel, G. Knowels, and D. J. Marcinek, Defects in mitochondrial localization and ATP synthesis in the mdx mouse model of Duchenne muscular dystrophy are not alleviated by PDE5 inhibition, Hum Mol Genet, vol.22, issue.1, pp.153-67, 2013.

M. H. Disatnik, J. Dhawan, Y. Yu, M. F. Beal, M. M. Whirl et al., Evidence of oxidative stress in mdx mouse muscle: studies of the pre-necrotic state, J Neurol Sci, vol.161, issue.1, pp.77-84, 1998.

N. P. Whitehead, E. W. Yeung, S. C. Froehner, and D. G. Allen, Skeletal muscle NADPH oxidase is increased and triggers stretch-induced damage in the mdx mouse, PloS One, vol.5, issue.12, p.15354, 2010.

R. J. Khairallah, G. Shi, F. Sbrana, B. L. Prosser, C. Borroto et al., Microtubules underlie dysfunction in duchenne muscular dystrophy, Sci Signal, vol.5, issue.236, p.56, 2012.

S. Hodgetts, H. Radley, M. Davies, and M. D. Grounds, Reduced necrosis of dystrophic muscle by depletion of host neutrophils, or blocking TNFalpha function with Etanercept in mdx mice, Neuromuscul Disord NMD, vol.16, issue.9, pp.591-602, 2006.

H. G. Radley and M. D. Grounds, Cromolyn administration (to block mast cell degranulation) reduces necrosis of dystrophic muscle in mdx mice, Neurobiol Dis, vol.23, issue.2, pp.387-97, 2006.

S. A. Villalta, H. X. Nguyen, B. Deng, T. Gotoh, and J. G. Tidball, Shifts in macrophage phenotypes and macrophage competition for arginine metabolism affect the severity of muscle pathology in muscular dystrophy, Hum Mol Genet, vol.18, issue.3, pp.482-96, 2009.

N. A. Dumont, Y. X. Wang, J. Von-maltzahn, A. Pasut, C. F. Bentzinger et al., Dystrophin expression in muscle stem cells regulates their polarity and asymetric division, Nat Med, vol.21, issue.12, pp.1455-1463, 2015.

C. Webster and H. M. Blau, Accelerated age-related decline in replicative life-span of Duchenne muscular dystrophy myoblasts: implications for cell and gene therapy, Somat Cell Mol Genet, vol.16, issue.6, pp.557-65, 1990.

Z. Yablonka-reuveni and J. E. Anderson, Satellite cells from dystrophic (mdx) mice display accelerated differentiation in primary cultures and in isolated myofibers, Dev Dyn Off Publ Am Assoc Anat, vol.235, issue.1, pp.203-215, 2006.

V. M. Shkryl, A. S. Martins, N. D. Ullrich, M. C. Nowycky, E. Niggli et al., Reciprocal amplification of ROS and Ca(2+) signals in stressed mdx dystrophic skeletal muscle fibers, Pflüg Arch Eur J Physiol, vol.458, issue.5, pp.915-943, 2009.

M. Eagle, J. Bourke, R. Bullock, M. Gibson, J. Mehta et al., Managing Duchenne muscular dystrophy--the additive effect of spinal surgery and home nocturnal ventilation in improving survival, Neuromuscul Disord NMD, vol.17, issue.6, pp.470-475, 2007.

L. Passamano, A. Taglia, A. Palladino, E. Viggiano, D. 'ambrosio et al., Improvement of survival in Duchenne Muscular Dystrophy: retrospective analysis of 835 patients. Acta Myol Myopathies Cardiomyopathies Off J Mediterr Soc Myol Ed Gaetano Conte Acad Study Striated Muscle Dis, vol.31, pp.121-126, 2012.

A. Goyenvalle, A. Vulin, F. Fougerousse, F. Leturcq, J. Kaplan et al., Rescue of dystrophic muscle through U7 snRNAmediated exon skipping, Science, vol.306, issue.5702, pp.1796-1805, 2004.

P. Gregorevic, M. J. Blankinship, J. M. Allen, and J. S. Chamberlain, Systemic microdystrophin gene delivery improves skeletal muscle structure and function in old dystrophic mdx mice, Mol Ther J Am Soc Gene Ther, vol.16, issue.4, pp.657-64, 2008.

J. Shin, X. Pan, C. H. Hakim, H. T. Yang, Y. Yue et al., Microdystrophin ameliorates muscular dystrophy in the canine model of duchenne muscular dystrophy, Mol Ther J Am Soc Gene Ther, vol.21, issue.4, pp.750-757, 2013.

L. Guiner, C. Montus, M. Servais, L. Cherel, Y. Francois et al., Forelimb treatment in a large cohort of dystrophic dogs supports delivery of a recombinant AAV for exon skipping in Duchenne patients, Mol Ther J Am Soc Gene Ther, vol.22, issue.11, pp.1923-1958, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02447482

K. Kawecka, M. Theodoulides, Y. Hasoglu, S. Jarmin, H. Kymalainen et al., Adeno-Associated Virus (AAV) Mediated Dystrophin Gene Transfer Studies and Exon Skipping Strategies for Duchenne Muscular Dystrophy (DMD), Curr Gene Ther, vol.15, issue.4, pp.395-415, 2015.

J. R. Mendell, K. Campbell, L. Rodino-klapac, Z. Sahenk, C. Shilling et al., Dystrophin immunity in Duchenne's muscular dystrophy, N Engl J Med, vol.363, issue.15, pp.1429-1466, 2010.

F. Mingozzi and K. A. High, Immune responses to AAV vectors: overcoming barriers to successful gene therapy, Blood, vol.122, issue.1, pp.23-36, 2013.

R. J. Chandler, M. C. Lafave, G. K. Varshney, N. S. Trivedi, N. Carrillo-carrasco et al., Vector design influences hepatic genotoxicity after adeno-associated virus gene therapy, J Clin Invest, vol.125, issue.2, pp.870-80, 2015.

C. A. Pacak, T. Conlon, C. S. Mah, and B. J. Byrne, Relative persistence of AAV serotype 1 vector genomes in dystrophic muscle, Genet Vaccines Ther, vol.6, p.14, 2008.

L. Hir, M. Goyenvalle, A. Peccate, C. Précigout, G. Davies et al., AAV Genome Loss From Dystrophic Mouse Muscles During AAV-U7 snRNA-mediated Exon-skipping Therapy, Mol Ther J Am Soc Gene Ther, 2013.

J. Dupont, B. Tournaire, C. Georger, B. Marolleau, L. Jeanson-leh et al., Short-lived recombinant adeno-associated virus transgene expression in dystrophic muscle is associated with oxidative damage to transgene mRNA, Mol Ther Methods Clin Dev, vol.2, p.15010, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01799048

K. Yuasa, M. Sakamoto, Y. Miyagoe-suzuki, A. Tanouchi, H. Yamamoto et al., Adeno-associated virus vector-mediated gene transfer into dystrophin-deficient skeletal muscles evokes enhanced immune response against the transgene product, Gene Ther, vol.9, issue.23, pp.1576-88, 2002.

G. Moulay, C. Masurier, P. Bigey, D. Scherman, and A. Kichler, Soluble TNF-? receptor secretion from healthy or dystrophic mice after AAV6-mediated muscle gene transfer, Gene Ther, vol.17, issue.11, pp.1400-1410, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02495337

D. Favre, Y. Cherel, N. Provost, V. Blouin, N. Ferry et al., Hyaluronidase enhances recombinant adeno-associated virus (rAAV)-mediated gene transfer in the rat skeletal muscle, Gene Ther, vol.7, issue.16, pp.1417-1437, 2000.

A. Toromanoff, Y. Chérel, M. Guilbaud, M. Penaud-budloo, R. O. Snyder et al., Safety and efficacy of regional intravenous (r.i.) versus intramuscular (i.m.) delivery of rAAV1 and rAAV8 to nonhuman primate skeletal muscle, Mol Ther J Am Soc Gene Ther, vol.16, issue.7, pp.1291-1300, 2008.

P. Xiao, C. Li, A. Neumann, and R. J. Samulski, Quantitative 3D tracing of gene-delivery viral vectors in human cells and animal tissues, Mol Ther J Am Soc Gene Ther, vol.20, issue.2, pp.317-345, 2012.

J. S. Chamberlain, J. Metzger, M. Reyes, D. Townsend, and J. A. Faulkner, Dystrophin-deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma, FASEB J Off Publ Fed Am Soc Exp Biol, vol.21, issue.9, pp.2195-204, 2007.

C. M. Pearson, Histopathological features of muscle in the preclinical stages of muscular dystrophy, Brain J Neurol, vol.85, pp.109-129, 1962.

W. G. Bradley, P. Hudgson, P. F. Larson, T. A. Papapetropoulos, and J. ,

M. , Structural changes in the early stages of Duchenne muscular dystrophy, J Neurol Neurosurg Psychiatry, vol.35, issue.4, pp.451-456, 1972.

J. Denard, C. Beley, R. Kotin, R. Lai-kuen, S. Blot et al., Human galectin 3 binding protein interacts with recombinant adenoassociated virus type 6, J Virol, vol.86, issue.12, pp.6620-6651, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02881163

J. Denard, B. Marolleau, C. Jenny, T. N. Rao, H. J. Fehling et al., C-reactive protein (CRP) is essential for efficient systemic transduction of recombinant adeno-associated virus vector 1 (rAAV-1) and rAAV-6 in mice, J Virol, vol.87, pp.10784-91, 2013.

Y. Hathout, R. L. Marathi, S. Rayavarapu, A. Zhang, K. J. Brown et al., Discovery of serum protein biomarkers in the mdx mouse model and cross-species comparison to Duchenne muscular dystrophy patients, Hum Mol Genet, vol.23, issue.24, pp.6458-69, 2014.

J. Rouillon, J. Poupiot, A. Zocevic, F. Amor, T. Léger et al., Serum proteomic profiling reveals fragments of MYOM3 as potential biomarkers for monitoring the outcome of therapeutic interventions in muscular dystrophies, Hum Mol Genet, vol.24, issue.17, pp.4916-4948, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01226478

V. Monteilhet, S. Saheb, S. Boutin, C. Leborgne, P. Veron et al., A 10 Patient Case Report on the Impact of Plasmapheresis Upon Neutralizing Factors Against Adeno-associated Virus (AAV) Types 1, 2, 6 and 8, Mol Ther, vol.19, issue.11, pp.2084-2091, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02881133

L. G. Chicoine, C. L. Montgomery, W. G. Bremer, K. M. Shontz, D. A. Griffin et al., Plasmapheresis Eliminates the Negative Impact of AAV Antibodies on Microdystrophin Gene Expression Following Vascular Delivery, Mol Ther, vol.22, issue.2, pp.338-347, 2014.

F. Mingozzi, X. M. Anguela, G. Pavani, Y. Chen, R. J. Davidson et al., Overcoming Preexisting Humoral Immunity to AAV Using Capsid Decoys, Sci Transl Med, vol.5, pp.194-92, 0194.
URL : https://hal.archives-ouvertes.fr/hal-02881152

J. E. Brenman, D. S. Chao, H. Xia, K. Aldape, and D. S. Bredt, Nitric oxide synthase complexed with dystrophin and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy, Cell, vol.82, issue.5, pp.743-52, 1995.

L. Arning, P. Jagiello, U. Schara, M. Vorgerd, N. Dahmen et al., Transcriptional profiles from patients with dystrophinopathies and limb girdle muscular dystrophies as determined by qRT-PCR, J Neurol, vol.251, issue.1, pp.72-80, 2004.

M. Sander, B. Chavoshan, S. A. Harris, S. T. Iannaccone, J. T. Stull et al., Functional muscle ischemia in neuronal nitric oxide synthase-deficient skeletal muscle of children with Duchenne muscular dystrophy, Proc Natl Acad Sci, vol.97, issue.25, pp.13818-13841, 2000.

M. Khairallah, R. J. Khairallah, M. E. Young, B. G. Allen, M. A. Gillis et al., Sildenafil and cardiomyocyte-specific cGMP signaling prevent cardiomyopathic changes associated with dystrophin deficiency, Proc Natl Acad Sci, vol.105, pp.7028-7061, 2008.

C. M. Adamo, D. Dai, J. M. Percival, E. Minami, M. S. Willis et al., Sildenafil reverses cardiac dysfunction in the mdx mouse model of Duchenne muscular dystrophy, Proc Natl Acad Sci, vol.107, issue.44, pp.19079-83, 2010.

J. M. Percival, N. P. Whitehead, M. E. Adams, C. M. Adamo, J. A. Beavo et al., Sildenafil reduces respiratory muscle weakness and fibrosis in the mdx mouse model of Duchenne muscular dystrophy, J Pathol, vol.228, issue.1, pp.77-87, 2012.

M. D. Nelson, F. Rader, X. Tang, J. Tavyev, S. F. Nelson et al., PDE5 inhibition alleviates functional muscle ischemia in boys with Duchenne muscular dystrophy, Neurology, vol.82, issue.23, pp.2085-91, 2014.

A. B. Katwal, P. R. Konkalmatt, B. A. Piras, S. Hazarika, S. S. Li et al., Adeno-associated virus serotype 9 efficiently targets ischemic skeletal muscle following systemic delivery, Gene Ther, vol.20, issue.9, pp.930-938, 2013.

L. Loufrani, K. Matrougui, D. Gorny, M. Duriez, I. Blanc et al., Flow (shear stress)-induced endothelium-dependent dilation is altered in mice lacking the gene encoding for dystrophin, vol.103, pp.864-70, 2001.
URL : https://hal.archives-ouvertes.fr/inserm-00135482

K. Sato, T. Yokota, S. Ichioka, M. Shibata, and S. Takeda, Vasodilation of intramuscular arterioles under shear stress in dystrophin-deficient skeletal muscle is impaired through decreased nNOS expression

, Acta Myol Myopathies Cardiomyopathies Off J Mediterr Soc Myol Ed Gaetano Conte Acad Study Striated Muscle Dis, vol.27, pp.30-36, 2008.

U. Rauch, A. Shami, F. Zhang, V. Carmignac, M. Durbeej et al., Increased neointimal thickening in dystrophin-deficient mdx mice, PloS One, vol.7, issue.1, p.29904, 2012.

S. Straino, A. Germani, D. Carlo, A. Porcelli, D. et al., Enhanced arteriogenesis and wound repair in dystrophindeficient mdx mice, Circulation, vol.110, issue.21, pp.3341-3349, 2004.

C. Latroche, B. Matot, A. Martins-bach, D. Briand, B. Chazaud et al., Structural and Functional Alterations of Skeletal Muscle Microvasculature in Dystrophin-Deficient mdx Mice, Am J Pathol, vol.185, issue.9, pp.2482-94, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01151848

D. Wang, L. Zhong, M. A. Nahid, and G. Gao, The potential of adenoassociated viral vectors for gene delivery to muscle tissue, Expert Opin Drug Deliv, vol.11, issue.3, pp.345-64, 2014.

N. P. Evans, S. A. Misyak, J. L. Robertson, J. Bassaganya-riera, and R. W. Grange, Immune-mediated mechanisms potentially regulate the disease time-course of duchenne muscular dystrophy and provide targets for therapeutic intervention, PM R, vol.1, issue.8, pp.755-68, 2009.

A. G. Engel and K. Arahata, Mononuclear cells in myopathies: quantitation of functionally distinct subsets, recognition of antigen-specific cellmediated cytotoxicity in some diseases, and implications for the pathogenesis of the different inflammatory myopathies, Hum Pathol, vol.17, issue.7, pp.704-725, 1986.

R. M. Mcdouall, M. J. Dunn, and V. Dubowitz, Nature of the mononuclear infiltrate and the mechanism of muscle damage in juvenile dermatomyositis and Duchenne muscular dystrophy, J Neurol Sci, vol.99, issue.2-3, pp.199-217, 1990.

J. D. Porter, S. Khanna, H. J. Kaminski, J. S. Rao, A. P. Merriam et al., A chronic inflammatory response dominates the skeletal muscle molecular signature in dystrophin-deficient mdx mice, Hum Mol Genet, vol.11, issue.3, pp.263-72, 2002.

M. Pescatori, A. Broccolini, C. Minetti, E. Bertini, C. Bruno et al., Gene expression profiling in the early phases of DMD: a constant molecular signature characterizes DMD muscle from early postnatal life throughout disease progression, FASEB J Off Publ Fed Am Soc Exp Biol, vol.21, issue.4, pp.1210-1236, 2007.

S. T. Appleyard, M. J. Dunn, V. Dubowitz, and M. L. Rose, Increased expression of HLA ABC class I antigens by muscle fibres in Duchenne muscular dystrophy, inflammatory myopathy, and other neuromuscular disorders, Lancet Lond Engl, vol.1, issue.8425, pp.361-364, 1985.

R. M. Mcdouall, M. J. Dunn, and V. Dubowitz, Expression of class I and class II MHC antigens in neuromuscular diseases, J Neurol Sci, vol.89, issue.2-3, pp.213-239, 1989.

R. Mantegazza, F. Andreetta, P. Bernasconi, F. Baggi, J. R. Oksenberg et al., Analysis of T cell receptor repertoire of muscleinfiltrating T lymphocytes in polymyositis. Restricted V alpha/beta rearrangements may indicate antigen-driven selection, J Clin Invest, vol.91, issue.6, pp.2880-2886, 1993.

E. Gussoni, G. K. Pavlath, R. G. Miller, M. A. Panzara, M. Powell et al., Specific T cell receptor gene rearrangements at the site of muscle degeneration in Duchenne muscular dystrophy, J Immunol Baltim Md, vol.153, issue.10, pp.4798-805, 1950.

M. J. Spencer, E. Montecino-rodriguez, K. Dorshkind, and J. G. Tidball, Helper (CD4(+)) and cytotoxic (CD8(+)) T cells promote the pathology of dystrophin-deficient muscle, Clin Immunol Orlando Fla, vol.98, issue.2, pp.235-278, 2001.

M. Wehling, M. J. Spencer, and J. G. Tidball, A nitric oxide synthase transgene ameliorates muscular dystrophy in mdx mice, J Cell Biol, vol.155, issue.1, pp.123-154, 2001.

Q. Yang, Y. Tang, K. Imbrogno, A. Lu, J. D. Proto et al., AAVbased shRNA silencing of NF-?B ameliorates muscle pathologies in mdx mice, Gene Ther, vol.19, issue.12, pp.1196-204, 2012.

S. A. Villalta, W. Rosenthal, L. Martinez, A. Kaur, T. Sparwasser et al., Regulatory T cell suppress muscle inflammation and injury in muscular dystrophy, Sci Transl Med, vol.6, issue.258, pp.258-142, 2014.

F. Boisgerault and F. Mingozzi, The Skeletal Muscle Environment and Its Role in Immunity and Tolerance to AAV Vector-Mediated Gene Transfer, Curr Gene Ther, vol.15, issue.4, pp.381-94, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02880969

K. Chandrasekharan, J. H. Yoon, Y. Xu, S. Devries, M. Camboni et al., A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy, Sci Transl Med, vol.2, issue.42, pp.42-54, 2010.

Y. Nitahara-kasahara, H. Hayashita-kinoh, T. Chiyo, A. Nishiyama, H. Okada et al., Dystrophic mdx mice develop severe cardiac and respiratory dysfunction following genetic ablation of the anti-inflammatory cytokine IL-10, Hum Mol Genet, vol.23, issue.15, pp.3990-4000, 2014.

T. Larcher, A. Lafoux, L. Tesson, R. S. Thepenier, V. François et al., Characterization of dystrophin deficient rats: a new model for Duchenne muscular dystrophy, PloS One, vol.9, issue.10, p.110371, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01190124

M. Nonnenmacher and T. Weber, Intracellular transport of recombinant adeno-associated virus vectors, Gene Ther, vol.19, issue.6, pp.649-58, 2012.

C. Summerford and R. J. Samulski, Membrane-associated heparan sulfate proteoglycan is a receptor for adeno-associated virus type 2 virions, J Virol, vol.72, issue.2, pp.1438-1483, 1998.

K. Qing, C. Mah, J. Hansen, S. Zhou, V. Dwarki et al., Human fibroblast growth factor receptor 1 is a co-receptor for infection by adeno-associated virus 2, Nat Med, vol.5, issue.1, pp.71-78, 1999.

C. Summerford, J. S. Bartlett, and R. J. Samulski, AlphaVbeta5 integrin: a coreceptor for adeno-associated virus type 2 infection, Nat Med, vol.5, issue.1, pp.78-82, 1999.

Y. Kashiwakura, K. Tamayose, K. Iwabuchi, Y. Hirai, T. Shimada et al., Hepatocyte growth factor receptor is a coreceptor for adeno-associated virus type 2 infection, J Virol, vol.79, issue.1, pp.609-623, 2005.

B. Akache, D. Grimm, K. Pandey, S. R. Yant, H. Xu et al., The 37/67-kilodalton laminin receptor is a receptor for adeno-associated virus serotypes 8, 2, 3, and 9, J Virol, vol.80, issue.19, pp.9831-9837, 2006.

A. Asokan, J. B. Hamra, L. Govindasamy, M. Agbandje-mckenna, and R. J. Samulski, Adeno-associated virus type 2 contains an integrin alpha5beta1 binding domain essential for viral cell entry, J Virol, vol.80, issue.18, pp.8961-8970, 2006.

C. Chen, R. L. Jensen, B. C. Schnepp, M. J. Connell, R. Shell et al., Molecular characterization of adeno-associated viruses infecting children, J Virol, vol.79, issue.23, pp.14781-92, 2005.

S. Pillay, N. L. Meyer, A. S. Puschnik, O. Davulcu, J. Diep et al., An essential receptor for adeno-associated virus infection, Nature, vol.530, pp.108-120, 2016.

J. Larraín, G. Cizmeci-smith, V. Troncoso, R. C. Stahl, D. J. Carey et al., Syndecan-1 expression is down-regulated during myoblast terminal differentiation. Modulation by growth factors and retinoic acid, J Biol Chem, vol.272, issue.29, pp.18418-18442, 1997.

L. Fuentealba, D. J. Carey, and E. Brandan, Antisense inhibition of syndecan-3 expression during skeletal muscle differentiation accelerates myogenesis through a basic fibroblast growth factordependent mechanism, J Biol Chem, vol.274, issue.53, pp.37876-84, 1999.

E. Brandan, D. J. Carey, J. Larraín, F. Melo, and A. Campos, Synthesis and processing of glypican during differentiation of skeletal muscle cells, Eur J Cell Biol, vol.71, issue.2, pp.170-176, 1996.

S. Cáceres, C. Cuellar, J. C. Casar, J. Garrido, L. Schaefer et al., Synthesis of proteoglycans is augmented in dystrophic mdx mouse skeletal muscle, Eur J Cell Biol, vol.79, issue.3, pp.173-81, 2000.

K. Alvarez, R. Fadic, and E. Brandan, Augmented synthesis and differential localization of heparan sulfate proteoglycans in Duchenne muscular dystrophy, J Cell Biochem, vol.85, issue.4, pp.703-716, 2002.

D. Duan, Q. Li, A. W. Kao, Y. Yue, J. E. Pessin et al., Dynamin is required for recombinant adeno-associated virus type 2 infection, J Virol, vol.73, issue.12, pp.10371-10377, 1999.

J. S. Bartlett, R. Wilcher, and R. J. Samulski, Infectious entry pathway of adeno-associated virus and adeno-associated virus vectors, J Virol, vol.74, issue.6, pp.2777-85, 2000.

S. Sanlioglu, P. K. Benson, J. Yang, E. M. Atkinson, T. Reynolds et al., Endocytosis and nuclear trafficking of adenoassociated virus type 2 are controlled by rac1 and phosphatidylinositol-3 kinase activation, J Virol, 2000.

A. D. Sanlioglu, B. Karacay, P. K. Benson, J. F. Engelhardt, and S. Sanlioglu, Novel approaches to augment adeno-associated virus type-2 endocytosis and transduction, Virus Res, vol.104, issue.1, pp.51-60, 2004.

M. Nonnenmacher and T. Weber, Adeno-associated virus 2 infection requires endocytosis through the CLIC/GEEC pathway, Cell Host Microbe, vol.10, issue.6, pp.563-76, 2011.

F. Sonntag, S. Bleker, B. Leuchs, R. Fischer, and J. A. Kleinschmidt, Adenoassociated virus type 2 capsids with externalized VP1/VP2 trafficking domains are generated prior to passage through the cytoplasm and are maintained until uncoating occurs in the nucleus, J Virol, vol.80, issue.22, pp.11040-54, 2006.

S. Stahnke, K. Lux, S. Uhrig, F. Kreppel, M. Hösel et al., Intrinsic phospholipase A2 activity of adeno-associated virus is involved in endosomal escape of incoming particles, Virology, vol.409, issue.1, pp.77-83, 2011.

A. M. Douar, K. Poulard, D. Stockholm, and O. Danos, Intracellular trafficking of adeno-associated virus vectors: routing to the late endosomal compartment and proteasome degradation, J Virol, vol.75, issue.4, pp.1824-1857, 2001.

M. E. Nonnenmacher, J. Cintrat, D. Gillet, and T. Weber, Syntaxin 5-dependent retrograde transport to the trans-Golgi network is required for adeno-associated virus transduction, J Virol, vol.89, issue.3, pp.1673-87, 2015.

K. Pajusola, M. Gruchala, H. Joch, T. F. Lüscher, S. Ylä-herttuala et al., Cell-type-specific characteristics modulate the transduction efficiency of adeno-associated virus type 2 and restrain infection of endothelial cells, J Virol, vol.76, issue.22, pp.11530-11570, 2002.

W. Ding, L. N. Zhang, C. Yeaman, and J. F. Engelhardt, rAAV2 traffics through both the late and the recycling endosomes in a dosedependent fashion, Mol Ther J Am Soc Gene Ther, vol.13, issue.4, pp.671-82, 2006.

N. W. Keiser, Z. Yan, Y. Zhang, D. Lei-butters, and J. F. Engelhardt, Unique characteristics of AAV1, 2, and 5 viral entry, intracellular trafficking, and nuclear import define transduction efficiency in HeLa cells, Hum Gene Ther, vol.22, issue.11, pp.1433-1477, 2011.

U. Bantel-schaal, B. Hub, and J. Kartenbeck, Endocytosis of adenoassociated virus type 5 leads to accumulation of virus particles in the Golgi compartment, J Virol, vol.76, issue.5, pp.2340-2349, 2002.

B. J. Petrof, J. B. Shrager, H. H. Stedman, A. M. Kelly, and H. L. Sweeney, Dystrophin protects the sarcolemma from stresses developed during muscle contraction, Proc Natl Acad Sci, vol.90, issue.8, pp.3710-3714, 1993.

M. J. Cullen and F. L. Mastaglia, Morphological changes in dystrophic muscle, Br Med Bull, vol.36, issue.2, pp.145-122, 1980.

M. J. Cullen and E. Jaros, Ultrastructure of the skeletal muscle in the X chromosome-linked dystrophic (mdx) mouse. Comparison with Duchenne muscular dystrophy, Acta Neuropathol (Berl), vol.77, issue.1, pp.69-81, 1988.

J. M. Percival, P. Gregorevic, G. L. Odom, G. B. Banks, J. S. Chamberlain et al., rAAV6-microdystrophin rescues aberrant Golgi complex organization in mdx skeletal muscles, Traffic Cph Den, vol.8, issue.10, pp.1424-1463, 2007.

D. Palma, C. Morisi, F. Cheli, S. Pambianco, S. Cappello et al., Autophagy as a new therapeutic target in Duchenne muscular dystrophy, Cell Death Dis, vol.3, p.418, 2012.

P. Spitali, P. Grumati, M. Hiller, M. Chrisam, A. Aartsma-rus et al., Autophagy is Impaired in the Tibialis Anterior of Dystrophin Null Mice, PLoS Curr, vol.5, 2013.

R. Pal, M. Palmieri, J. A. Loehr, S. Li, R. Abo-zahrah et al., Src-dependent impairment of autophagy by oxidative stress in a mouse model of Duchenne muscular dystrophy, Nat Commun, vol.5, p.4425, 2014.

S. Duguez, W. Duddy, H. Johnston, J. Lainé, L. Bihan et al., Dystrophin deficiency leads to disturbance of LAMP1-vesicleassociated protein secretion, Cell Mol Life Sci CMLS, vol.70, issue.12, pp.2159-74, 2013.

C. Ren, A. F. White, and S. Ponnazhagan, Notch1 augments intracellular trafficking of adeno-associated virus type 2, J Virol, vol.81, issue.4, pp.2069-73, 2007.

R. Turk, E. Sterrenburg, E. J. De-meijer, G. Van-ommen, and J. T. Den-dunnen, Muscle regeneration in dystrophin-deficient mdx mice studied by gene expression profiling, BMC Genomics, vol.6, p.98, 2005.

J. E. Church, J. Trieu, A. Chee, T. Naim, S. M. Gehrig et al., Alterations in Notch signalling in skeletal muscles from mdx and dko dystrophic mice and patients with Duchenne muscular dystrophy, Exp Physiol, vol.99, issue.4, pp.675-87, 2014.

C. Jiang, Y. Wen, K. Kuroda, K. Hannon, M. A. Rudnicki et al., Notch signaling deficiency underlies age-dependent depletion of satellite cells in muscular dystrophy, Dis Model Mech, vol.7, issue.8, pp.997-1004, 2014.

N. M. Vieira, I. Elvers, M. S. Alexander, Y. B. Moreira, A. Eran et al., Jagged 1 Rescues the Duchenne Muscular Dystrophy Phenotype, Cell, vol.163, issue.5, pp.1204-1217, 2015.

L. Zhong, W. Zhao, J. Wu, B. Li, S. Zolotukhin et al., A dual role of EGFR protein tyrosine kinase signaling in ubiquitination of AAV2 capsids and viral second-strand DNA synthesis, Mol Ther J Am Soc Gene Ther, vol.15, issue.7, pp.1323-1353, 2007.

L. Zhong, X. Zhou, Y. Li, K. Qing, X. Xiao et al., Singlepolarity recombinant adeno-associated virus 2 vector-mediated transgene expression in vitro and in vivo: mechanism of transduction. Restriction factors against rAAV in DMD muscles, Journal Name, vol.0, issue.0, p.15, 2015.

, Mol Ther J Am Soc Gene Ther, vol.16, issue.2, pp.290-295, 2008.

Y. Kawanabe, N. Hashimoto, and T. Masaki, Characterization of Ca2+ channels involved in ET-1-induced transactivation of EGF receptors, Am J Physiol Heart Circ Physiol, vol.283, issue.6, pp.2671-2676, 2002.

Y. Kawanabe, K. Nozaki, N. Hashimoto, and T. Masaki, Involvement of extracellular Ca2+ influx and epidermal growth factor receptor tyrosine kinase transactivation in endothelin-1-induced arachidonic acid release, Br J Pharmacol, vol.139, issue.8, pp.1516-1538, 2003.

P. Xiao and R. J. Samulski, Cytoplasmic trafficking, endosomal escape, and perinuclear accumulation of adeno-associated virus type 2 particles are facilitated by microtubule network, J Virol, vol.86, pp.10462-73, 2012.

J. J. Belanto, T. L. Mader, M. D. Eckhoff, D. M. Strandjord, G. B. Banks et al., Microtubule binding distinguishes dystrophin from utrophin, Proc Natl Acad Sci, vol.111, issue.15, pp.5723-5731, 2014.

K. W. Prins, J. L. Humston, A. Mehta, V. Tate, E. Ralston et al., Dystrophin is a microtubule-associated protein, J Cell Biol, vol.186, issue.3, pp.363-372, 2009.

W. Xiao, K. H. Warrington, P. Hearing, J. Hughes, and N. Muzyczka, Adenovirus-facilitated nuclear translocation of adeno-associated virus type 2, J Virol, vol.76, issue.22, pp.11505-11522, 2002.

S. C. Nicolson and R. J. Samulski, Recombinant adeno-associated virus utilizes host cell nuclear import machinery to enter the nucleus, J Virol, vol.88, issue.8, pp.4132-4176, 2014.

S. Taranum, E. Vaylann, P. Meinke, S. Abraham, L. Yang et al., LINC complex alterations in DMD and EDMD/CMT fibroblasts, Eur J Cell Biol, vol.91, issue.8, pp.614-642, 2012.

K. Stieger, J. Schroeder, N. Provost, A. Mendes-madeira, B. Belbellaa et al., Detection of intact rAAV particles up to 6 years after successful gene transfer in the retina of dogs and primates, Mol Ther J Am Soc Gene Ther, vol.17, issue.3, pp.516-539, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02659213

K. Nash, W. Chen, W. F. Mcdonald, X. Zhou, and N. Muzyczka, Purification of host cell enzymes involved in adeno-associated virus DNA replication, J Virol, vol.81, issue.11, pp.5777-87, 2007.

K. Qing, J. Hansen, K. A. Weigel-kelley, M. Tan, S. Zhou et al., Adeno-associated virus type 2-mediated gene transfer: role of cellular FKBP52 protein in transgene expression, J Virol, vol.75, issue.19, pp.8968-76, 2001.

K. Qing, X. S. Wang, D. M. Kube, S. Ponnazhagan, A. Bajpai et al., Role of tyrosine phosphorylation of a cellular protein in adenoassociated virus 2-mediated transgene expression, Proc Natl Acad Sci, vol.94, issue.20, pp.10879-84, 1997.

C. Mah, K. Qing, B. Khuntirat, S. Ponnazhagan, X. S. Wang et al., Adeno-associated virus type 2-mediated gene transfer: role of epidermal growth factor receptor protein tyrosine kinase in transgene expression, J Virol, vol.72, issue.12, pp.9835-9878, 1998.

K. Qing, W. Li, L. Zhong, M. Tan, J. Hansen et al., Adeno-associated virus type 2-mediated gene transfer: role of cellular T-cell protein tyrosine phosphatase in transgene expression in established cell lines in vitro and transgenic mice in vivo, J Virol, vol.77, issue.4, pp.2741-2747, 2003.

K. Qing, B. Khuntirat, C. Mah, D. M. Kube, X. S. Wang et al., Adeno-associated virus type 2-mediated gene transfer: correlation of tyrosine phosphorylation of the cellular single-stranded D sequence-binding protein with transgene expression in human cells in vitro and murine tissues in vivo, J Virol, vol.72, issue.2, pp.1593-1602, 1998.

P. R. Turner, T. Westwood, C. M. Regen, and R. A. Steinhardt, Increased protein degradation results from elevated free calcium levels found in muscle from mdx mice, Nature, vol.335, issue.6192, pp.735-743, 1988.

A. Franco and J. B. Lansman, Calcium entry through stretch-inactivated ion channels in mdx myotubes, Nature, vol.344, issue.6267, pp.670-673, 1990.

C. Vandebrouck, D. Martin, M. Colson-van-schoor, H. Debaix, and P. Gailly, Involvement of TRPC in the abnormal calcium influx observed in dystrophic (mdx) mouse skeletal muscle fibers, J Cell Biol, vol.158, issue.6, pp.1089-96, 2002.

B. C. Schnepp, K. R. Clark, D. L. Klemanski, C. A. Pacak, and P. R. Johnson, Genetic fate of recombinant adeno-associated virus vector genomes in muscle, J Virol, vol.77, issue.6, pp.3495-504, 2003.

M. Penaud-budloo, L. Guiner, C. Nowrouzi, A. Toromanoff, A. Chérel et al., Adeno-associated virus vector genomes persist as episomal chromatin in primate muscle, J Virol, vol.82, issue.16, pp.7875-85, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02666015

X. Xiao, J. Li, and R. J. Samulski, Efficient long-term gene transfer into muscle tissue of immunocompetent mice by adeno-associated virus vector, J Virol, vol.70, issue.11, pp.8098-108, 1996.

R. O. Snyder, S. K. Spratt, C. Lagarde, D. Bohl, K. B. Sloan et al., Efficient and stable adeno-associated virus-mediated transduction in the skeletal muscle of adult immunocompetent mice, Hum Gene Ther, vol.8, issue.16, pp.1891-900, 1997.

N. Vincent-lacaze, R. O. Snyder, R. Gluzman, D. Bohl, C. Lagarde et al., Structure of adeno-associated virus vector DNA following transduction of the skeletal muscle, J Virol, vol.73, issue.3, pp.1949-55, 1999.

K. A. Nakai and H. , The Role of DNA Repair Pathways in Adeno-Associated Virus Infection and Viral Genome Replication / Recombination / Integration, 2011.

S. Song, P. J. Laipis, K. I. Berns, and T. R. Flotte, Effect of DNA-dependent protein kinase on the molecular fate of the rAAV2 genome in skeletal muscle, Proc Natl Acad Sci, vol.98, issue.7, pp.4084-4092, 2001.

M. P. Cataldi and D. M. Mccarty, Differential effects of DNA double-strand break repair pathways on single-strand and self-complementary adeno-associated virus vector genomes, J Virol, vol.84, issue.17, pp.8673-82, 2010.

R. A. Schwartz, J. A. Palacios, G. D. Cassell, S. Adam, M. Giacca et al., The Mre11/Rad50/Nbs1 complex limits adenoassociated virus transduction and replication, J Virol, vol.81, issue.23, pp.12936-12981, 2007.

M. P. Cataldi and D. M. Mccarty, Hairpin-end conformation of adenoassociated virus genome determines interactions with DNA-repair pathways, Gene Ther, vol.20, issue.6, pp.686-93, 2013.

J. Lovric, M. Mano, L. Zentilin, A. Eulalio, S. Zacchigna et al., Terminal differentiation of cardiac and skeletal myocytes induces permissivity to AAV transduction by relieving inhibition imposed by DNA damage response proteins, Mol Ther J Am Soc Gene Ther, vol.20, issue.11, pp.2087-97, 2012.

W. M. Schmidt, M. H. Uddin, S. Dysek, K. Moser-thier, C. Pirker et al., DNA damage, somatic aneuploidy, and malignant sarcoma susceptibility in muscular dystrophies, PLoS Genet, vol.7, issue.4, p.1002042, 2011.

K. Fernandez, Y. Serinagaoglu, S. Hammond, L. T. Martin, and P. T. Martin, Mice lacking dystrophin or alpha sarcoglycan spontaneously develop embryonal rhabdomyosarcoma with cancer-associated p53 mutations and alternatively spliced or mutant Mdm2 transcripts, Am J Pathol, vol.176, issue.1, pp.416-450, 2010.

T. B. Lentz and R. J. Samulski, Insight into the mechanism of inhibition of adeno-associated virus by the Mre11/Rad50/Nbs1 complex, J Virol, vol.89, issue.1, pp.181-94, 2015.

W. Y. Chen, E. C. Bailey, S. L. Mccune, J. Y. Dong, and T. M. Townes, Reactivation of silenced, virally transduced genes by inhibitors of histone deacetylase, Proc Natl Acad Sci U S A, vol.94, issue.11, pp.5798-803, 1997.

T. Okada, R. Uchibori, M. Iwata-okada, M. Takahashi, T. Nomoto et al., A histone deacetylase inhibitor enhances recombinant adeno-associated virus-mediated gene expression in tumor cells, Mol Ther J Am Soc Gene Ther, vol.13, issue.4, pp.738-784, 2006.

G. C. Minetti, C. Colussi, R. Adami, C. Serra, C. Mozzetta et al., Functional and morphological recovery of dystrophic muscles in mice treated with deacetylase inhibitors, Nat Med, vol.12, issue.10, pp.1147-50, 2006.

C. Colussi, R. Berni, J. Rosati, S. Straino, S. Vitale et al., The histone deacetylase inhibitor suberoylanilide hydroxamic acid reduces cardiac arrhythmias in dystrophic mice, Cardiovasc Res, vol.87, issue.1, pp.73-82, 2010.

S. Consalvi, C. Mozzetta, P. Bettica, M. Germani, F. Fiorentini et al., Preclinical studies in the mdx mouse model of duchenne muscular dystrophy with the histone deacetylase inhibitor givinostat, Mol Med Camb Mass, vol.19, pp.79-87, 2013.

C. Colussi, C. Mozzetta, A. Gurtner, B. Illi, J. Rosati et al., HDAC2 blockade by nitric oxide and histone deacetylase inhibitors reveals a common target in Duchenne muscular dystrophy treatment, Proc Natl Acad Sci, vol.105, issue.49, pp.19183-19190, 2008.

C. Colussi, A. Gurtner, J. Rosati, B. Illi, G. Ragone et al., Nitric oxide deficiency determines global chromatin changes in Duchenne muscular dystrophy, FASEB J Off Publ Fed Am Soc Exp Biol, vol.23, issue.7, pp.2131-2172, 2009.

C. Mozzetta, S. Consalvi, V. Saccone, M. Tierney, A. Diamantini et al., Fibroadipogenic progenitors mediate the ability of HDAC inhibitors to promote regeneration in dystrophic muscles of young, but not old Mdx mice, EMBO Mol Med, vol.5, issue.4, pp.626-665, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01549090

G. Sohi and F. J. Dilworth, Noncoding RNAs as epigenetic mediators of skeletal muscle regeneration, FEBS J, vol.282, issue.9, pp.1630-1676, 2015.

G. Falcone, A. Perfetti, B. Cardinali, and F. Martelli, Noncoding RNAs: emerging players in muscular dystrophies, BioMed Res Int, p.503634, 2014.

T. Beiter, M. Hoene, F. Prenzler, F. C. Mooren, J. M. Steinacker et al., Exercise, skeletal muscle and inflammation: ARE-binding proteins as key regulators in inflammatory and adaptive networks, Exerc Immunol Rev, vol.21, pp.42-57, 2015.

Y. Saletore, K. Meyer, J. Korlach, I. D. Vilfan, S. Jaffrey et al., The birth of the Epitranscriptome: deciphering the function of RNA modifications, Genome Biol, vol.13, issue.10, p.175, 2012.

M. O'connell, RNA modification and the epitranscriptome; the next frontier, RNA N Y N, vol.21, issue.4, pp.703-707, 2015.

Y. Lai, Y. Yue, M. Liu, and D. Duan, Synthetic intron improves transduction efficiency of trans-splicing adeno-associated viral vectors, Hum Gene Ther, vol.17, issue.10, pp.1036-1078, 2006.

Z. Wu, J. Sun, T. Zhang, C. Yin, F. Yin et al., Optimization of self-complementary AAV vectors for liver-directed expression results in sustained correction of hemophilia B at low vector dose, Mol Ther J Am Soc Gene Ther, vol.16, issue.2, pp.280-289, 2008.

A. Douglas and M. Wood, RNA splicing: disease and therapy, Brief Funct Genomics, vol.10, issue.3, pp.151-64, 2011.

A. G. Matera and Z. Wang, A day in the life of the spliceosome, Nat Rev Mol Cell Biol, vol.15, issue.2, pp.108-129, 2014.

Q. Pan, O. Shai, L. J. Lee, B. J. Frey, and B. J. Blencowe, Deep surveying of alternative splicing complexity in the human transcriptome by highthroughput sequencing, Nat Genet, vol.40, issue.12, pp.1413-1418, 2008.

M. Bakay, P. Zhao, J. Chen, and E. P. Hoffman, A web-accessible complete transcriptome of normal human and DMD muscle, Neuromuscul Disord NMD, vol.12, issue.1, pp.125-166, 2002.

M. Sironi, R. Cagliani, G. P. Comi, U. Pozzoli, A. Bardoni et al., Trans-acting factors may cause dystrophin splicing misregulation in BMD skeletal muscles, FEBS Lett, vol.537, issue.1-3, pp.30-34, 2003.

D. Dominissini, S. Moshitch-moshkovitz, S. Schwartz, M. Salmon-divon, L. Ungar et al., Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq, Nature, vol.485, issue.7397, pp.201-207, 2012.

T. W. Nilsen, Internal mRNA methylation finally finds functions, Science, vol.343, issue.6176, pp.1207-1215, 2014.

S. Geula, S. Moshitch-moshkovitz, D. Dominissini, A. A. Mansour, N. Kol et al., Stem cells. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation, Science, vol.347, issue.6225, pp.1002-1008, 2015.

F. Mechler, S. Imre, and P. Dioszeghy, Lipid peroxidation and superoxide dismutase activity in muscle and erythrocytes in Duchenne muscular dystrophy, J Neurol Sci, vol.63, issue.3, pp.279-83, 1984.

Y. Nakae, P. J. Stoward, T. Kashiyama, M. Shono, A. Akagi et al., Early onset of lipofuscin accumulation in dystrophindeficient skeletal muscles of DMD patients and mdx mice, J Mol Histol, vol.35, issue.5, pp.489-99, 2004.

R. Renjini, N. Gayathri, and A. Nalini, Srinivas Bharath MM. Oxidative damage in muscular dystrophy correlates with the severity of the pathology: role of glutathione metabolism, Neurochem Res, vol.37, issue.4, pp.885-98, 2012.

A. F. El-shafey, A. E. Armstrong, J. R. Terrill, M. D. Grounds, and P. G. Arthur, Screening for increased protein thiol oxidation in oxidatively stressed muscle tissue, Free Radic Res, vol.45, issue.9, pp.991-1000, 2011.

T. Iwasaki, J. Terrill, T. Shavlakadze, M. D. Grounds, and P. G. Arthur, Visualizing and quantifying oxidized protein thiols in tissue sections: a comparison of dystrophic mdx and normal skeletal mouse muscles. Free Radic Biol Med, vol.65, pp.1408-1424, 2013.

M. C. Rodriguez and M. A. Tarnopolsky, Patients with dystrophinopathy show evidence of increased oxidative stress. Free Radic Biol Med, vol.34, pp.1217-1237, 2003.

X. Shan, H. Tashiro, and C. G. Lin, The identification and characterization of oxidized RNAs in Alzheimer's disease, J Neurosci Off J Soc Neurosci, vol.23, issue.12, pp.4913-4934, 2003.

X. Shan and C. Lin, Quantification of oxidized RNAs in Alzheimer's disease, Neurobiol Aging, vol.27, issue.5, pp.657-62, 2006.

Y. Chang, Q. Kong, X. Shan, G. Tian, H. Ilieva et al., Messenger RNA oxidation occurs early in disease pathogenesis and promotes motor neuron degeneration in ALS, PloS One, vol.3, issue.8, p.2849, 2008.

A. Jorgensen, K. Broedbaek, A. Fink-jensen, U. Knorr, G. Soendergaard et al., Increased systemic oxidatively generated DNA and RNA damage in schizophrenia, Psychiatry Res, vol.209, issue.3, pp.417-440, 2013.

M. Tateyama, A. Takeda, Y. Onodera, M. Matsuzaki, T. Hasegawa et al., Oxidative stress and predominant Abeta42(43) deposition in myopathies with rimmed vacuoles, Acta Neuropathol (Berl), vol.105, issue.6, pp.581-586, 2003.

T. Hofer, E. Marzetti, J. Xu, A. Y. Seo, S. Gulec et al., Increased iron content and RNA oxidative damage in skeletal muscle with aging and disuse atrophy, Exp Gerontol, vol.43, issue.6, pp.563-70, 2008.

M. Tanaka, P. B. Chock, and E. R. Stadtman, Oxidized messenger RNA induces translation errors, Proc Natl Acad Sci, vol.104, issue.1, pp.66-71, 2007.

X. Shan, Y. Chang, and C. G. Lin, Messenger RNA oxidation is an early event preceding cell death and causes reduced protein expression

, FASEB J Off Publ Fed Am Soc Exp Biol, vol.21, issue.11, pp.2753-64, 2007.

C. L. Simms, B. H. Hudson, J. W. Mosior, A. S. Rangwala, and H. S. Zaher, An active role for the ribosome in determining the fate of oxidized mRNA, Cell Rep, vol.9, issue.4, pp.1256-64, 2014.

E. De-klerk, I. Fokkema, K. Thiadens, J. J. Goeman, M. Palmblad et al., Assessing the translational landscape of myogenic differentiation by ribosome profiling, Nucleic Acids Res, vol.43, issue.9, pp.4408-4436, 2015.

I. Dalle-donne, G. Aldini, M. Carini, R. Colombo, R. Rossi et al., Protein carbonylation, cellular dysfunction, and disease progression, J Cell Mol Med, vol.10, issue.2, pp.389-406, 2006.

C. K. Jana, N. Das, and R. S. Sohal, Specificity of age-related carbonylation of plasma proteins in the mouse and rat, Arch Biochem Biophys, vol.397, issue.2, pp.433-442, 2002.

J. Choi, H. D. Rees, S. T. Weintraub, A. I. Levey, L. Chin et al., Oxidative modifications and aggregation of Cu,Zn-superoxide dismutase associated with Alzheimer and Parkinson diseases, J Biol Chem, vol.280, issue.12, pp.11648-55, 2005.

M. Kastle and T. Grune, Protein oxidative modification in the aging organism and the role of the ubiquitin proteasomal system, Curr Pharm Des, vol.17, issue.36, pp.4007-4029, 2011.

A. Asokan, J. C. Conway, J. L. Phillips, C. Li, J. Hegge et al., Reengineering a receptor footprint of adeno-associated virus enables selective and systemic gene transfer to muscle, Nat Biotechnol, vol.28, issue.1, pp.79-82, 2010.

D. E. Bowles, S. Mcphee, C. Li, S. J. Gray, J. J. Samulski et al., Phase 1 gene therapy for Duchenne muscular dystrophy using a translational optimized AAV vector, Mol Ther J Am Soc Gene Ther, vol.20, issue.2, pp.443-55, 2012.

W. Li, A. Asokan, Z. Wu, T. Van-dyke, N. Diprimio et al., Engineering and selection of shuffled AAV genomes: a new strategy for producing targeted biological nanoparticles, Mol Ther J Am Soc Gene Ther, vol.16, issue.7, pp.1252-60, 2008.

L. Yang, J. Li, and X. Xiao, Directed evolution of adeno-associated virus (AAV) as vector for muscle gene therapy, Methods Mol Biol Clifton NJ, vol.709, pp.127-166, 2011.

E. Zinn, S. Pacouret, V. Khaychuk, H. T. Turunen, L. S. Carvalho et al., In Silico Reconstruction of the Viral Evolutionary Lineage Yields a Potent Gene Therapy Vector, Cell Rep, vol.12, issue.6, pp.1056-68, 2015.

X. Li, E. M. Eastman, R. J. Schwartz, and R. Draghia-akli, Synthetic muscle promoters: activities exceeding naturally occurring regulatory sequences, Nat Biotechnol, vol.17, issue.3, pp.241-246, 1999.

M. Z. Salva, C. L. Himeda, P. W. Tai, E. Nishiuchi, P. Gregorevic et al., Design of tissue-specific regulatory cassettes for high-level rAAV-mediated expression in skeletal and cardiac muscle, Mol Ther J Am Soc Gene Ther, vol.15, issue.2, pp.320-329, 2007.

H. Foster, P. S. Sharp, T. Athanasopoulos, C. Trollet, I. R. Graham et al., Codon and mRNA sequence optimization of microdystrophin transgenes improves expression and physiological outcome in dystrophic mdx mice following AAV2/8 gene transfer, Mol Ther J Am Soc Gene Ther, vol.16, issue.11, pp.1825-1857, 2008.

T. Athanasopoulos, H. Foster, K. Foster, and G. Dickson, Codon optimization of the microdystrophin gene for Duchene muscular dystrophy gene therapy, Methods Mol Biol Clifton NJ, vol.709, pp.21-37, 2011.

E. Lecomte, B. Tournaire, B. Cogné, J. Dupont, and P. Lindenbaum, Restriction factors against rAAV in DMD muscles Journal Name, vol.0, p.17, 2015.

M. Martin-fontaine, Advanced Characterization of DNA Molecules in rAAV Vector Preparations by Single-stranded Virus Next-generation Sequencing, Mol Ther Nucleic Acids, vol.4, p.260, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01799968

D. Duan, Y. Yue, Z. Yan, J. Yang, and J. F. Engelhardt, Endosomal processing limits gene transfer to polarized airway epithelia by adeno-associated virus, J Clin Invest, vol.105, issue.11, pp.1573-87, 2000.

L. Denby, S. A. Nicklin, and A. H. Baker, Adeno-associated virus (AAV)-7 and -8 poorly transduce vascular endothelial cells and are sensitive to proteasomal degradation, Gene Ther, vol.12, issue.20, pp.1534-1542, 2005.

A. C. Nathwani, M. Cochrane, J. Mcintosh, C. Ng, J. Zhou et al., Enhancing transduction of the liver by adeno-associated viral vectors, Gene Ther, vol.16, issue.1, pp.60-69, 2009.

G. Moulay, S. Boutin, C. Masurier, D. Scherman, and A. Kichler, Polymers for improving the in vivo transduction efficiency of AAV2 vectors, PloS One, vol.5, issue.12, p.15576, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02495339

S. H. Rahman, S. Bobis-wozowicz, D. Chatterjee, K. Gellhaus, K. Pars et al., The nontoxic cell cycle modulator indirubin augments transduction of adeno-associated viral vectors and zincfinger nuclease-mediated gene targeting, Hum Gene Ther, vol.24, issue.1, pp.67-77, 2013.

G. Bonuccelli, F. Sotgia, W. Schubert, D. S. Park, P. G. Frank et al., Proteasome inhibitor (MG-132) treatment of mdx mice rescues the expression and membrane localization of dystrophin and dystrophin-associated proteins, Am J Pathol, vol.163, issue.4, pp.1663-75, 2003.

E. Gazzerro, S. Assereto, A. Bonetto, F. Sotgia, S. Scarfì et al., Therapeutic potential of proteasome inhibition in Duchenne and Becker muscular dystrophies, Am J Pathol, vol.176, issue.4, pp.1863-77, 2010.

S. Abmayr, P. Gregorevic, J. M. Allen, and J. S. Chamberlain, Phenotypic improvement of dystrophic muscles by rAAV/microdystrophin vectors is augmented by Igf1 codelivery, Mol Ther J Am Soc Gene Ther, vol.12, issue.3, pp.441-50, 2005.

W. Hoogaars, E. Mouisel, A. Pasternack, J. J. Hulmi, K. Relizani et al., Combined effect of AAV-U7-induced dystrophin exon skipping and soluble activin Type IIB receptor in mdx mice, Hum Gene Ther, vol.23, issue.12, pp.1269-79, 2012.

P. Gregorevic, M. J. Blankinship, J. M. Allen, R. W. Crawford, L. Meuse et al., Systemic delivery of genes to striated muscles using adeno-associated viral vectors, Nat Med, vol.10, issue.8, pp.828-862, 2004.

P. Gregorevic, J. M. Allen, E. Minami, M. J. Blankinship, M. Haraguchi et al., rAAV6-microdystrophin preserves muscle function and extends lifespan in severely dystrophic mice, Nat Med, vol.12, issue.7, pp.787-796, 2006.