F. Dreyer and A. Baur, Biogenesis and Functions of Exosomes and Extracellular Vesicles, Lentiviral Vectors and Exosomes as Gene and Protein Delivery Tools, vol.1448, pp.201-216, 2016.

R. J. Simpson, S. S. Jensen, and J. W. Lim, Proteomic profiling of exosomes: Current perspectives, PROTEOMICS, vol.8, issue.19, pp.4083-4099, 2008.

H. Valadi, K. Ekström, A. Bossios, M. Sjöstrand, J. J. Lee et al., Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nature Cell Biology, vol.9, issue.6, pp.654-659, 2007.

D. Fröhlich, W. P. Kuo, C. Frühbeis, J. J. Sun, C. M. Zehendner et al., Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation, Philosophical Transactions of the Royal Society B: Biological Sciences, vol.369, issue.1652, p.20130510, 2014.

C. Frühbeis, D. Fröhlich, W. P. Kuo, and E. M. Krämer-albers, Extracellular vesicles as mediators of neuron-glia communication, Frontiers in Cellular Neuroscience, vol.7, p.182, 2013.

C. Frühbeis, D. Fröhlich, and E. M. Krämer-albers, Emerging Roles of Exosomes in Neuron?Glia Communication, Frontiers in Physiology, vol.3, p.119, 2012.

Y. S. Takeda and Q. Xu, Neuronal Differentiation of Human Mesenchymal Stem Cells Using Exosomes Derived from Differentiating Neuronal Cells, PLOS ONE, vol.10, issue.8, p.e0135111, 2015.

C. Admyre, S. M. Johansson, K. R. Qazi, J. J. Filén, R. Lahesmaa et al., Exosomes with Immune Modulatory Features Are Present in Human Breast Milk, The Journal of Immunology, vol.179, issue.3, pp.1969-1978, 2007.

P. D. Robbins and A. E. Morelli, Regulation of immune responses by extracellular vesicles, Nature Reviews Immunology, vol.14, issue.3, pp.195-208, 2014.

R. H. Staals and G. J. Pruijn, The Human Exosome and Disease, Advances in Experimental Medicine and Biology, vol.702, pp.132-142, 2010.

I. Russo, L. Bubacco, and E. Greggio, Exosomes-associated neurodegeneration and progression of Parkinson's disease, Am. J. Neurodegener. Dis, vol.1, pp.217-225, 2012.

Y. Wang, V. Balaji, S. Kaniyappan, L. Krüger, S. Irsen et al., The release and trans-synaptic transmission of Tau via exosomes, Molecular Neurodegeneration, vol.12, issue.1, p.5, 2017.

J. K. Wang, P. Langfelder, S. Horvath, and M. J. Palazzolo, Exosomes and Homeostatic Synaptic Plasticity Are Linked to Each other and to Huntington's, Parkinson's, and Other Neurodegenerative Diseases by Database-Enabled Analyses of Comprehensively Curated Datasets, Frontiers in Neuroscience, vol.11, p.149, 2017.

L. Alvarez-erviti, Y. Seow, H. Yin, C. Betts, S. Lakhal et al., Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes, Nature Biotechnology, vol.29, issue.4, pp.341-345, 2011.

J. Dashkoff, E. P. Lerner, N. Truong, J. A. Klickstein, Z. Fan et al., Tailored transgene expression to specific cell types in the central nervous system after peripheral injection with AAV9, Molecular Therapy - Methods & Clinical Development, vol.3, p.16081, 2016.

J. S. Johnson and R. J. Samulski, Enhancement of Adeno-Associated Virus Infection by Mobilizing Capsids into and Out of the Nucleolus, Journal of Virology, vol.83, issue.6, pp.2632-2644, 2008.

M. Moskalenko, L. Chen, M. Van-roey, B. A. Donahue, R. O. Snyder et al., Epitope Mapping of Human Anti-Adeno-Associated Virus Type 2 Neutralizing Antibodies: Implications for Gene Therapy and Virus Structure, Journal of Virology, vol.74, issue.4, pp.1761-1766, 2000.

B. György, Z. Fitzpatrick, M. H. Crommentuijn, D. Mu, and C. A. Maguire, Naturally enveloped AAV vectors for shielding neutralizing antibodies and robust gene delivery in vivo, Biomaterials, vol.35, issue.26, pp.7598-7609, 2014.

C. A. Maguire, L. Balaj, S. Sivaraman, M. H. Crommentuijn, M. Ericsson et al., Microvesicle-associated AAV Vector as a Novel Gene Delivery System, Molecular Therapy, vol.20, issue.5, pp.960-971, 2012.

V. Ramakrishnaiah, C. Thumann, I. Fofana, F. Habersetzer, Q. Pan et al., Exosome-mediated transmission of hepatitis C virus between human hepatoma Huh7.5 cells, Proceedings of the National Academy of Sciences, vol.110, issue.32, pp.13109-13113, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02494666

B. György, C. Sage, A. A. Indzhykulian, D. I. Scheffer, A. R. Brisson et al., Rescue of Hearing by Gene Delivery to Inner-Ear Hair Cells Using Exosome-Associated AAV, Molecular Therapy, vol.25, issue.2, pp.379-391, 2017.

S. J. Wassmer, L. S. Carvalho, B. György, L. H. Vandenberghe, and C. A. Maguire, Exosome-associated AAV2 vector mediates robust gene delivery into the murine retina upon intravitreal injection, Scientific Reports, vol.7, issue.1, p.45329, 2017.

E. Laemmel, M. Genet, G. Le-goualher, A. Perchant, J. F. Le-gargasson et al., Fibered Confocal Fluorescence Microscopy (Cell-viZio?) Facilitates Extended Imaging in the Field of Microcirculation, Journal of Vascular Research, vol.41, issue.5, pp.400-411, 2004.

J. A. Lieberman, R. R. Girgis, G. Brucato, H. Moore, F. Provenzano et al., Hippocampal dysfunction in the pathophysiology of schizophrenia: a selective review and hypothesis for early detection and intervention, Molecular Psychiatry, vol.23, issue.8, pp.1764-1772, 2018.

S. J. Gray, K. T. Woodard, and R. J. Samulski, Viral vectors and delivery strategies for CNS gene therapy, Therapeutic Delivery, vol.1, issue.4, pp.517-534, 2010.

K. S. Bankiewicz, J. Forsayeth, J. L. Eberling, R. Sanchez-pernaute, P. Pivirotto et al., Long-Term Clinical Improvement in MPTP-Lesioned Primates after Gene Therapy with AAV-hAADC, Molecular Therapy, vol.14, issue.4, pp.564-570, 2006.

K. S. Bankiewicz, J. Forsayeth, J. L. Eberling, R. Sanchez-pernaute, P. Pivirotto et al., Long-Term Clinical Improvement in MPTP-Lesioned Primates after Gene Therapy with AAV-hAADC, Molecular Therapy, vol.14, issue.4, pp.564-570, 2006.

M. Hocquemiller, L. Giersch, M. Audrain, S. Parker, and N. Cartier, Adeno-Associated Virus-Based Gene Therapy for CNS Diseases, Human Gene Therapy, vol.27, issue.7, pp.478-496, 2016.

M. Zerah, F. Piguet, M. A. Colle, S. Raoul, J. Y. Deschamps et al., Intracerebral Gene Therapy Using AAVrh.10-hARSA Recombinant Vector to Treat Patients with Early-Onset Forms of Metachromatic Leukodystrophy: Preclinical Feasibility and Safety Assessments in Nonhuman Primates, Human Gene Therapy Clinical Development, vol.26, issue.2, pp.113-124, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01164426

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.

N. Nayerossadat, P. Ali, and T. Maedeh, Viral and nonviral delivery systems for gene delivery, Advanced Biomedical Research, vol.1, issue.1, p.27, 2012.

T. B. Lentz, S. J. Gray, and R. J. Samulski, Viral vectors for gene delivery to the central nervous system, Neurobiology of Disease, vol.48, issue.2, pp.179-188, 2012.

K. S. Linterman, D. N. Palmer, G. W. Kay, L. A. Barry, N. L. Mitchell et al., Lentiviral-Mediated Gene Transfer to the Sheep Brain: Implications for Gene Therapy in Batten Disease, Human Gene Therapy, vol.22, issue.8, pp.1011-1020, 2011.

S. Boutin, V. Monteilhet, P. Veron, C. Leborgne, O. Benveniste et al., Prevalence of Serum IgG and Neutralizing Factors Against Adeno-Associated Virus (AAV) Types 1, 2, 5, 6, 8, and 9 in the Healthy Population: Implications for Gene Therapy Using AAV Vectors, Human Gene Therapy, vol.21, issue.6, pp.704-712, 2010.

F. Mingozzi, X. M. Anguela, G. Pavani, Y. Chen, R. J. Davidson et al., Overcoming Preexisting Humoral Immunity to AAV Using Capsid Decoys, Science Translational Medicine, vol.5, issue.194, pp.194ra92-194ra92, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02881152

T. J. Mccown, X. Xiao, J. Li, G. R. Breese, and R. Jude-samulski, Differential and persistent expression patterns of CNS gene transfer by an adeno-associated virus (AAV) vector, Brain Research, vol.713, issue.1-2, pp.99-107, 1996.

R. L. Klein, E. M. Meyer, A. L. Peel, S. Zolotukhin, C. Meyers et al., Neuron-Specific Transduction in the Rat Septohippocampal or Nigrostriatal Pathway by Recombinant Adeno-associated Virus Vectors, Experimental Neurology, vol.150, issue.2, pp.183-194, 1998.

J. C. Paterna, T. Moccetti, A. Mura, J. Feldon, and H. Büeler, Influence of promoter and WHV post-transcriptional regulatory element on AAV-mediated transgene expression in the rat brain, Gene Therapy, vol.7, issue.15, pp.1304-1311, 2000.

S. J. Gray, S. B. Foti, J. W. Schwartz, L. Bachaboina, B. Taylor-blake et al., Optimizing Promoters for Recombinant Adeno-Associated Virus-Mediated Gene Expression in the Peripheral and Central Nervous System Using Self-Complementary Vectors, Human Gene Therapy, vol.22, issue.9, pp.1143-1153, 2011.

J. Mellor, M. J. Dobson, N. A. Roberts, A. J. Kingsman, and S. M. Kingsman, Factors affecting heterologous gene expression in Saccharomyces cerevisiae, Gene, vol.33, issue.2, pp.215-226, 1985.

R. Derynck, A. Singh, and D. V. Goeddel, Expression of the human interferon?? cDNA in yeast, Nucleic Acids Research, vol.11, issue.6, pp.1819-1837, 1983.

J. Mellor, M. J. Dobson, N. A. Roberts, M. F. Tuite, J. S. Emtage et al., Efficient synthesis of enzymatically active calf chymosin in Saccharomyces cerevisiae, Gene, vol.24, issue.1, pp.1-14, 1983.

M. F. Tuite, M. J. Dobson, N. A. Roberts, R. M. King, D. C. Burke et al., Regulated high efficiency expression of human interferon-alpha in Saccharomyces cerevisiae., The EMBO Journal, vol.1, issue.5, pp.603-608, 1982.

B. L. Davidson, C. S. Stein, J. A. Heth, I. Martins, R. M. Kotin et al., Recombinant adeno-associated virus type 2, 4, and 5 vectors: Transduction of variant cell types and regions in the mammalian central nervous system, Proceedings of the National Academy of Sciences, vol.97, issue.7, pp.3428-3432, 2000.

D. F. Aschauer, S. Kreuz, and S. Rumpel, Analysis of Transduction Efficiency, Tropism and Axonal Transport of AAV Serotypes 1, 2, 5, 6, 8 and 9 in the Mouse Brain, PLoS ONE, vol.8, issue.9, p.e76310, 2013.

S. Duque, B. Joussemet, C. Riviere, T. Marais, L. Dubreil et al., Intravenous Administration of Self-complementary AAV9 Enables Transgene Delivery to Adult Motor Neurons, Molecular Therapy, vol.17, issue.7, pp.1187-1196, 2009.
URL : https://hal.archives-ouvertes.fr/hal-02666576

K. D. Foust, E. Nurre, C. L. Montgomery, A. Hernandez, C. M. Chan et al., Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes, Nature Biotechnology, vol.27, issue.1, pp.59-65, 2008.

S. A. Martin, T. M. Demuth, K. N. Miller, T. D. Pugh, M. A. Polewski et al., Regional metabolic heterogeneity of the hippocampus is nonuniformly impacted by age and caloric restriction, Aging Cell, vol.15, issue.1, pp.100-110, 2015.

C. N. Cearley and J. H. Wolfe, Transduction characteristics of adeno-associated virus vectors expressing cap serotypes 7, 8, 9, and Rh10 in the mouse brain, Molecular Therapy, vol.13, issue.3, pp.528-537, 2006.

C. N. Cearley, L. H. Vandenberghe, M. K. Parente, E. R. Carnish, J. M. Wilson et al., Expanded Repertoire of AAV Vector Serotypes Mediate Unique Patterns of Transduction in Mouse Brain, Molecular Therapy, vol.16, issue.10, pp.1710-1718, 2008.

D. Sondhi, N. R. Hackett, D. A. Peterson, J. Stratton, M. Baad et al., Enhanced Survival of the LINCL Mouse Following CLN2 Gene Transfer Using the rh.10 Rhesus Macaque-derived Adeno-associated Virus Vector, Molecular Therapy, vol.15, issue.3, pp.481-491, 2007.

H. Chen, D. M. Mccarty, A. T. Bruce, K. Suzuki, and K. Suzuki, Gene transfer and expression in oligodendrocytes under the control of myelin basic protein transcriptional control region mediated by adeno-associated virus, Gene Therapy, vol.5, issue.1, pp.50-58, 1998.

P. A. Lawlor, R. J. Bland, A. Mouravlev, D. Young, and M. J. During, Efficient Gene Delivery and Selective Transduction of Glial Cells in the Mammalian Brain by AAV Serotypes Isolated From Nonhuman Primates, Molecular Therapy, vol.17, issue.10, pp.1692-1702, 2009.

M. Nonnenmacher and T. Weber, Intracellular transport of recombinant adeno-associated virus vectors, Gene Therapy, vol.19, issue.6, pp.649-658, 2012.

J. S. Bartlett, R. Wilcher, and R. J. Samulski, Infectious Entry Pathway of Adeno-Associated Virus and Adeno-Associated Virus Vectors, Journal of Virology, vol.74, issue.6, pp.2777-2785, 2000.

T. Matsushita, S. Elliger, C. Elliger, G. Podsakoff, L. Villarreal et al., Adeno-associated virus vectors can be efficiently produced without helper virus, Gene Therapy, vol.5, issue.7, pp.938-945, 1998.

E. Ayuso, F. Mingozzi, J. Montane, X. Leon, X. M. Anguela et al., High AAV vector purity results in serotype- and tissue-independent enhancement of transduction efficiency, Gene Therapy, vol.17, issue.4, pp.503-510, 2009.

G. Paxinos and C. Watson, WITHDRAWN: Acknowledgements, The Rat Brain in Stereotaxic Coordinates, p.vii, 1982.