E. Abordo-adesida, A. Follenzi, C. Barcia, S. Sciascia, M. G. Castro et al., Stability of lentiviral vector-mediated transgene expression in the brain in the presence of systemic antivector immune responses. Hum, Gene Ther, vol.16, pp.741-751, 2005.

W. Adriani, F. Boyer, D. Leo, R. Canese, F. Podo et al., Social withdrawal and gambling-like profile after lentiviral manipulation of DAT expression in the rat accumbens, Int. J. Neuropsychopharmacol, vol.13, pp.1329-1342, 2010.

K. N. Agwuh and A. Macgowan, Pharmacokinetics and pharmacodynamics of the tetracyclines including glycylcyclines, J. Antimicrob. Chemother, vol.58, pp.256-265, 2006.

M. Akerblom, R. Sachdeva, L. Quintino, E. E. Wettergren, K. Z. Chapman et al., Visualization and genetic modification of resident brain microglia using lentiviral vectors regulated by microRNA-9, Nat. Commun, vol.4, p.1770, 2013.

A. Annoni, M. Battaglia, A. Follenzi, A. Lombardo, L. Sergi-sergi et al., 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, vol.110, pp.1788-1796, 2007.

B. N. Armbruster, X. Li, M. H. Pausch, S. Herlitze, and B. L. Roth, Evolving the lock to fit the key to create a family of G protein-coupled receptors potently activated by an inert ligand, Proc. Natl. Acad. Sci. U.S.A, vol.104, pp.5163-5168, 2007.

G. Aston-jones and K. Deisseroth, Recent advances in optogenetics and pharmacogenetics, Brain Res, vol.1511, pp.1-5, 2013.

D. Atasoy, J. N. Betley, H. H. Su, and S. M. Sternson, Deconstruction of a neural circuit for hunger, Nature, vol.488, pp.172-177, 2012.

C. C. Bartholomae, A. Arens, K. S. Balaggan, R. J. Yáñez-muñoz, E. Montini et al., Lentiviral vector integration profiles differ in rodent postmitotic tissues, Mol. Ther, vol.19, pp.703-710, 2011.

R. D. Beech, M. A. Cleary, H. B. Treloar, A. J. Eisch, A. V. Harrist et al., Nestin promoter/enhancer directs transgene expression to precursors of adult generated periglomerular neurons, J. Comp. Neurol, vol.475, pp.128-141, 2004.

K. Benzekhroufa, B. H. Liu, A. G. Teschemacher, and S. Kasparov, Targeting central serotonergic neurons with lentiviral vectors based on a transcriptional amplification strategy, Gene Ther, vol.16, pp.681-688, 2009.

U. Blomer, L. Naldini, T. Kafri, D. Trono, I. M. Verma et al., Highly efficient and sustained gene transfer in adult neurons with a lentivirus vector, J. Virol, vol.71, pp.6641-6649, 1997.

C. Bosch, B. Degos, J. M. Deniau, and L. Venance, Subthalamic nucleus high-frequency stimulation generates a concomitant synaptic excitationinhibition in substantia nigra pars reticulata, J. Physiol, vol.589, pp.4189-4207, 2011.

E. S. Boyden, F. Zhang, E. Bamberg, G. Nagel, and K. Deisseroth, Millisecond-timescale, genetically targeted optical control of neural activity, Nat. Neurosci, vol.8, pp.1263-1268, 2005.

A. R. Brooks, R. N. Harkins, P. Wang, H. S. Qian, P. Liu et al., Transcriptional silencing is associated with extensive methylation of the CMV promoter following adenoviral gene delivery to muscle, J. Gene Med, vol.6, pp.395-404, 2004.

D. Cai, K. B. Cohen, T. Luo, J. W. Lichtman, and J. R. Sanes, Improved tools for the Brainbow toolbox, Nat. Methods, vol.10, pp.540-547, 2013.

E. M. Callaway, Transneuronal circuit tracing with neurotropic viruses, Curr. Opin. Neurobiol, vol.18, pp.617-623, 2008.

D. C. Carpentier, K. Vevis, A. Trabalza, C. Georgiadis, S. M. Ellison et al., Enhanced pseudotyping efficiency of HIV-1 lentiviral vectors by a rabies/vesicular stomatitis virus chimeric envelope glycoprotein, Gene Ther, vol.19, pp.761-774, 2012.

A. Cetin, S. Komai, M. Eliava, P. H. Seeburg, and P. Osten, , 2006.

, Stereotaxic gene delivery in the rodent brain, Nat. Protoc, vol.1, pp.3166-3173

D. Chaudhury, J. J. Walsh, A. K. Friedman, B. Juarez, S. M. Ku et al., Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons, Nature, vol.493, pp.532-536, 2013.

J. Chen, C. H. Kwon, L. Lin, Y. Li, and P. L. , Inducible sitespecific recombination in neural stem/progenitor cells, Genesis, vol.47, pp.122-131, 2009.

L. Cheng, Z. Jin, L. Liu, Y. Yan, T. Li et al., Characterization and promoter analysis of the mouse nestin gene, FEBS Lett, vol.565, pp.195-202, 2004.

S. Y. Cho, L. Ravasi, L. P. Szajek, J. Seidel, M. V. Green et al., Evaluation of 76 Br-FBAU as a PET reporter probe for HSV1-tk gene expression imaging using mouse models of human glioma, J. Nucl. Med, vol.46, 1923.

L. M. Chow, J. Zhang, and S. J. Baker, Inducible Cre recombinase activity in mouse mature astrocytes and adult neural precursor cells, Transgenic Res, vol.17, pp.919-928, 2008.

K. Chung, J. Wallace, S. Kim, S. Kalyanasundaram, A. S. Andalman et al., Structural and molecular interrogation of intact biological systems, Nature, vol.497, pp.332-337, 2013.

A. Colin, M. Faideau, N. Dufour, G. Auregan, R. Hassig et al., Engineered lentiviral vector targeting astrocytes in vivo, Glia, vol.57, pp.667-679, 2009.
URL : https://hal.archives-ouvertes.fr/tel-00348988

C. H. Contag and M. H. Bachmann, Advances in in vivo bioluminescence imaging of gene expression, Annu. Rev. Biomed. Eng, vol.4, pp.235-260, 2002.

J. Cronin, X. Y. Zhang, and J. Reiser, Altering the tropism of lentiviral vectors through pseudotyping, Curr. Gene Ther, vol.5, pp.387-398, 2005.

D. Dautan, I. Huerta-ocampo, I. B. Witten, K. Deisseroth, J. P. Bolam et al., A major external source of cholinergic innervation of the striatum and nucleus accumbens originates in the brainstem, J. Neurosci, vol.34, pp.4509-4518, 2014.

K. Deisseroth and M. J. Schnitzer, Engineering approaches to illuminating brain structure and dynamics, Neuron, vol.80, pp.568-577, 2013.

A. Delzor, N. Dufour, F. Petit, M. Guillermier, D. Houitte et al., Restricted transgene expression in the brain with cell-type specific neuronal promoters. Hum, Gene Ther. Methods, vol.23, pp.242-254, 2012.

D. Marco, R. J. Groneberg, A. H. Yeh, C. M. Castillo-ramírez, L. A. Ryu et al., Optogenetic elevation of endogenous glucocorticoid level in larval zebrafish, Front. Neural Circuits, vol.7, p.82, 2013.

C. M. Deroose, V. Reumers, R. Gijsbers, G. Bormans, Z. Debyser et al., Noninvasive monitoring of long-term lentiviral vector-mediated gene expression in rodent brain with bioluminescence imaging, Mol. Ther, vol.14, pp.423-431, 2006.

M. Desai, I. Kahn, U. Knoblich, J. Bernstein, H. Atallah et al., Mapping brain networks in awake mice using combined optical neural control and fMRI, J. Neurophysiol, vol.105, pp.1393-1405, 2011.

N. Desmaris, A. Bosch, C. Salaün, C. Petit, M. C. Prévost et al., Production and neurotropism of lentivirus vectors pseudotyped with lyssavirus envelope glycoproteins, Mol. Ther, vol.4, pp.149-156, 2001.
URL : https://hal.archives-ouvertes.fr/pasteur-01372718

I. Diester, M. T. Kaufman, M. Mogri, R. Pashaie, W. Goo et al., An optogenetic toolbox designed for primates, Nat. Neurosci, vol.14, pp.387-397, 2011.

T. Dittgen, A. Nimmerjahn, S. Komai, P. Licznerski, J. Waters et al., Lentivirus-based genetic manipulations of cortical neurons and their optical and electrophysiological monitoring in vivo, Proc. Natl. Acad. Sci. U.S.A, vol.101, pp.18206-18211, 2004.

H. Duale, S. Kasparov, J. F. Paton, and A. G. Teschemacher, Differences in transductional tropism of adenoviral and lentiviral vectors in the rat brainstem, Exp. Physiol, vol.90, pp.71-78, 2005.

T. Dull, R. Zufferey, M. Kelly, R. J. Mandel, M. Nguyen et al., A third-generation lentivirus vector with a conditional packaging system, J. Virol, vol.72, pp.8463-8471, 1998.

E. E. Fanselow and B. W. Connors, Navigating a sensorimotor loop, Neuron, vol.45, pp.329-330, 2005.

L. Fenno, O. Yizhar, and K. Deisseroth, The development and application of optogenetics, Annu. Rev. Neurosci, vol.34, pp.389-412, 2011.

M. Figueiredo, S. Lane, F. Tang, B. H. Liu, J. Hewinson et al., Optogenetic experimentation on astrocytes, Exp. Physiol, vol.96, pp.40-50, 2011.

D. Finkelshtein, A. Werman, D. Novick, S. Barak, and M. Rubinstein, LDL receptor and its family members serve as the cellular receptors for vesicular stomatitis virus, Proc. Natl. Acad. Sci. U.S.A, vol.110, pp.7306-7311, 2013.

M. Folcher, S. Oesterle, K. Zwicky, T. Thekkottil, J. Heymoz et al., Mind-controlled transgene expression by a wireless-powered optogenetic designer cell implant, Nat. Commun, vol.5, p.5392, 2014.

A. Galvan, X. Hu, Y. Smith, and T. Wichmann, In vivo optogenetic control of striatal and thalamic neurons in non-human primates, PLoS ONE, vol.7, p.50808, 2012.

M. Gossen and H. Bujard, Tight control of gene expression in mammalian cells by tetracycline-responsive promoters, Proc. Natl. Acad. Sci. U.S.A, vol.89, pp.5547-5551, 1992.

M. Gossen and H. Bujard, Efficacy of tetracycline-controlled gene expression is influenced by cell type: commentary, BioTechniques, vol.19, pp.213-216, 1995.

M. Gossen, S. Freundlieb, G. Bender, G. Müller, W. Hillen et al., Transcriptional activation by tetracyclines in mammalian cells, Science, vol.268, pp.1766-1769, 1995.

V. Gradinaru, F. Zhang, C. Ramakrishnan, J. Mattis, R. Prakash et al., Molecular and cellular approaches for diversifying and extending optogenetics, Cell, vol.141, pp.154-165, 2010.

V. Grinevich, M. Brecht, and P. Osten, Monosynaptic pathway from rat vibrissa motor cortex to facial motor neurons revealed by lentivirus-based axonal tracing, J. Neurosci, vol.25, pp.8250-8258, 2005.

S. Hacein-bey-abina, C. Von-kalle, M. Schmidt, M. P. Mccormack, N. Wulffraat et al., LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1, Science, vol.302, pp.415-419, 2003.

H. Hama, H. Kurokawa, M. Schmidt, M. P. Mccormack, N. Wulffraat et al., Scale: a chemical approach for fluorescence imaging and reconstruction of transparent mouse brain, Nat. Neurosci, vol.14, pp.1481-1488, 2011.

X. Han, X. Qian, J. G. Bernstein, H. H. Zhou, G. T. Franzesi et al., Millisecond-timescale optical control of neural dynamics in the nonhuman primate brain, Neuron, vol.62, pp.191-198, 2009.

E. Hasegawa, M. Yanagisawa, T. Sakurai, and M. Mieda, Orexin neurons suppress narcolepsy via 2 distinct efferent pathways, J. Clin. Invest, vol.124, pp.604-616, 2014.

M. Hausser, Optogenetics: the age of light, Nat. Methods, vol.11, pp.1012-1014, 2014.

F. Heitz, T. Johansson, K. Baumgärtel, R. Gecaj, P. Pelczar et al., Heritable and inducible gene knockdown in astrocytes or neurons in vivo by a combined lentiviral and RNAi approach, Front. Cell Neurosci, vol.8, p.62, 2014.

H. Hioki, H. Kameda, H. Nakamura, T. Okunomiya, K. Ohira et al., Efficient gene transduction of neurons by lentivirus with enhanced neuron-specific promoters, Gene Ther, vol.14, pp.872-882, 2007.

Y. Huh, M. S. Oh, P. Leblanc, and K. S. Kim, Gene transfer in the nervous system and implications for transsynaptic neuronal tracing, Expert Opin. Biol. Ther, vol.10, pp.763-772, 2010.

S. Indraccolo, S. Minuzzo, F. Feroli, F. Mammano, F. Calderazzo et al., Pseudotyping of Moloney leukemia virus-based retroviral vectors with simian immunodeficiency virus envelope leads to targeted infection of human CD4+ lymphoid cells, Gene Ther, vol.5, pp.209-217, 1998.

J. Jakobsson, C. Ericson, M. Jansson, E. Björk, and C. Lundberg, Targeted transgene expression in rat brain using lentiviral vectors, J. Neurosci. Res, vol.73, pp.876-885, 2003.

S. Kato, K. Kobayashi, K. Inoue, M. Kuramochi, T. Okada et al., A lentiviral strategy for highly efficient retrograde gene transfer by pseudotyping with fusion envelope glycoprotein, Hum. Gene Ther, vol.22, pp.197-206, 2011.

S. Kato, M. Kuramochi, K. Takasumi, K. Kobayashi, K. Inoue et al., Neuron-specific gene transfer through retrograde transport of lentiviral vector pseudotyped with a novel type of fusion envelope glycoprotein, Hum. Gene Ther, vol.22, pp.1511-1523, 2011.

S. Kato, K. Kobayashi, and K. Kobayashi, Improved transduction efficiency of a lentiviral vector for neuron-specific retrograde gene transfer by optimizing the junction of fusion envelope glycoprotein, J. Neurosci. Methods, vol.227, pp.151-158, 2014.

M. T. Ke, S. Fujimoto, and T. Imai, SeeDB: a simple and morphologypreserving optical clearing agent for neuronal circuit reconstruction, Nat. Neurosci, vol.16, pp.1154-1161, 2013.

M. C. Klaw, C. Xu, and V. J. Tom, Intraspinal AAV injections immediately rostral to a thoracic spinal cord injury site efficiently transduces neurons in spinal cord and brain, Mol. Ther. Nucleic Acids, vol.2, p.108, 2013.

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, Exp. Neurol, vol.150, pp.183-194, 1998.

T. Knöpfel, Genetically encoded optical indicators for the analysis of neuronal circuits, Nat. Rev. Neurosci, vol.13, pp.687-700, 2012.

S. Konermann, M. D. Brigham, A. E. Trevino, P. D. Hsu, M. Heidenreich et al., Optical control of mammalian endogenous transcription and epigenetic states, Nature, vol.500, pp.472-476, 2013.

R. Kumar-singh, Barriers for retinal gene therapy: separating fact from fiction, Vision Res, vol.48, 2008.

J. H. Lee, R. Durand, V. Gradinaru, F. Zhang, I. Goshen et al., Global and local fMRI signals driven by neurons defined optogenetically by type and wiring, Nature, vol.465, pp.788-792, 2010.

W. Lerchner, B. Corgiat, V. Der-minassian, R. C. Saunders, R. et al., Injection parameters and virus dependent choice of promoters to improve neuron targeting in the nonhuman primate brain, Gene Ther, vol.21, pp.233-241, 2014.

P. A. Lewitt, A. R. Rezai, M. A. Leehey, S. G. Ojemann, A. W. Flaherty et al., AAV2-GAD gene therapy for advanced Parkinson's disease: a double-blind, sham-surgery controlled, randomised trial, Lancet Neurol, vol.10, pp.309-319, 2011.

M. Li, N. Husic, Y. Lin, H. Christensen, I. Malik et al., Optimal promoter usage for lentiviral vector-mediated transduction of cultured central nervous system cells, J. Neurosci. Methods, vol.189, pp.56-64, 2010.

Y. Li, X. F. Du, and J. L. Du, Resting microglia respond to and regulate neuronal activity in vivo, Commun. Integr. Biol, vol.6, p.24493, 2013.

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, Hum. Gene Ther, vol.22, pp.1011-1020, 2011.

K. C. Liu, B. S. Lin, A. D. Gao, H. Y. Ma, M. Zhao et al., Integrase-deficient lentivirus: opportunities and challenges for human gene therapy, Curr. Gene Ther, vol.14, pp.352-364, 2014.

Y. J. Liu, M. U. Ehrengruber, M. Negwer, H. J. Shao, A. H. Cetin et al., Tracing inputs to inhibitory or excitatory neurons of mouse and cat visual cortex with a targeted rabies virus, Curr. Biol, vol.23, pp.1746-1755, 2013.

E. Lobbestael, V. Reumers, A. Ibrahimi, K. Paesen, I. Thiry et al., Immunohistochemical detection of transgene expression in the brain using small epitope tags, BMC Biotechnol, vol.10, p.16, 2010.

T. F. Massoud, A. Singh, and S. S. Gambhir, Noninvasive molecular neuroimaging using reporter genes: part I, principles revisited, AJNR Am. J. Neuroradiol, vol.29, pp.229-234, 2008.

N. D. Mazarakis, M. Azzouz, J. B. Rohll, F. M. Ellard, F. J. Wilkes et al., Rabies virus glycoprotein pseudotyping of lentiviral vectors enables retrograde axonal transport and access to the nervous system after peripheral delivery, Hum. Mol. Genet, vol.10, pp.2109-2121, 2001.

S. R. Mciver, C. S. Lee, J. M. Lee, S. H. Green, M. S. Sands et al., Lentiviral transduction of murine oligodendrocytes in vivo, J. Neurosci. Res, vol.82, pp.397-403, 2005.

S. R. Mciver, M. Muccigrosso, E. R. Gonzales, J. M. Lee, M. S. Roberts et al., Oligodendrocyte degeneration and recovery after focal cerebral ischemia, Neuroscience, vol.169, pp.1364-1375, 2010.

W. Mittmann, D. J. Wallace, U. Czubayko, J. T. Herb, A. T. Schaefer et al., Two-photon calcium imaging of evoked activity from L5 somatosensory neurons in vivo, Nat. Neurosci, vol.14, pp.1089-1093, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00658166

T. Miyashita, Y. R. Shao, J. Chung, O. Pourzia, and D. E. Feldman, Longterm channelrhodopsin-2 (ChR2) expression can induce abnormal axonal morphology and targeting in cerebral cortex, Front. Neural Circuits, vol.7, issue.8, 2013.

H. Miyoshi, M. Takahashi, F. H. Gage, and I. M. Verma, Stable and efficient gene transfer into the retina using an HIV-based lentiviral vector, Proc. Natl. Acad. Sci. U.S.A, vol.94, pp.10319-10323, 1997.

L. Naldini, Lentiviruses as gene transfer agents for delivery to nondividing cells, Curr. Opin. Biotechnol, vol.9, pp.80029-80032, 1998.

J. L. Nathanson, Y. Yanagawa, K. Obata, and E. M. Callaway, Preferential labeling of inhibitory and excitatory cortical neurons by endogenous tropism of adeno-associated virus and lentivirus vectors, Neuroscience, vol.161, pp.441-450, 2009.

T. T. Nielsen, I. Marion, L. Hasholt, and C. Lundberg, Neuron-specific RNA interference using lentiviral vectors, J. Gene Med, vol.11, pp.559-569, 2009.

A. M. Packer, B. Roska, and M. Häusser, Targeting neurons and photons for optogenetics, Nat. Neurosci, vol.16, pp.805-815, 2013.

S. Palfi, J. M. Gurruchaga, G. S. Ralph, H. Lepetit, S. Lavisse et al., Long-term safety and tolerability of ProSavin, a lentiviral vector-based gene therapy for Parkinson's disease: a dose escalation, open-label, phase 1/2 trial, Nat. Methods, vol.383, p.447, 2011.

G. P. Pathak, J. D. Vrana, and C. L. Tucker, Optogenetic control of cell function using engineered photoreceptors, Biol. Cell, vol.105, pp.59-72, 2013.

K. Pluta, M. J. Luce, L. Bao, S. Agha-mohammadi, and J. Reiser, Tight control of transgene expression by lentivirus vectors containing secondgeneration tetracycline-responsive promoters, J. Gene Med, vol.7, pp.803-817, 2005.

J. D. Pollock, D. Y. Wu, and J. S. Satterlee, Molecular neuroanatomy: a generation of progress, Trends Neurosci, vol.37, pp.106-123, 2014.

J. Y. Qin, L. Zhang, K. L. Clift, I. Hulur, A. P. Xiang et al., Systematic comparison of constitutive promoters and the doxycyclineinducible promoter, PLoS ONE, vol.5, p.10611, 2010.

S. C. Rogan and B. L. Roth, Remote control of neuronal signaling, Pharmacol. Rev, vol.63, pp.291-315, 2011.

B. Sauer and H. N. , Site-specific DNA recombination in mammalian cells by the Cre recombinase of bacteriophage P1, Proc. Natl. Acad. Sci. U.S.A, vol.85, pp.5166-5170, 1988.

L. Schoderboeck, S. Riad, A. M. Bokor, H. E. Wicky, M. Strauss et al., Chimeric rabies SADB19-VSVg pseudotyped lentiviral vectors mediate long-range retrograde transduction from the mouse spinal cord, Gene Ther, 2015.

S. Scotto-lomassese, A. Nissant, T. Mota, M. Néant-féry, B. A. Oostra et al., Fragile X mental retardation protein regulates new neuron differentiation in the adult olfactory bulb, J. Neurosci, vol.31, pp.2205-2215, 2011.

S. Seeger-armbruster, C. Bosch-bouju, S. T. Little, R. A. Smither, S. M. Hughes et al., Patterned, but not tonic, optogenetic stimulation in motor thalamus improves reaching in acute drug-induced parkinsonian rats, J. Neurosci, vol.35, pp.1211-1216, 2015.

K. Shah, S. Hingtgen, R. Kasmieh, J. L. Figueiredo, E. Garcia-garcia et al., Bimodal viral vectors and in vivo imaging reveal the fate of human neural stem cells in experimental glioma model, J. Neurosci, vol.28, pp.4406-4413, 2008.

X. Shu, V. Lev-ram, T. J. Deerinck, Y. Qi, E. B. Ramko et al., A genetically encoded tag for correlated light and electron microscopy of intact cells, tissues, and organisms, PLoS Biol, vol.9, p.1001041, 2011.

V. S. Sohal, F. Zhang, O. Yizhar, and K. Deisseroth, Parvalbumin neurons and gamma rhythms enhance cortical circuit performance, Nature, vol.459, pp.698-702, 2009.

G. E. Sosinsky, B. N. Giepmans, T. J. Deerinck, G. M. Gaietta, and M. H. Ellisman, Markers for correlated light and electron microscopy, Methods Cell Biol, vol.79, pp.575-591, 2007.

P. Stern, S. Astrof, S. J. Erkeland, J. Schustak, P. A. Sharp et al., A system for Cre-regulated RNA interference in vivo, Proc. Natl. Acad. Sci. U.S.A, vol.105, pp.13895-13900, 2008.

M. Y. Sun, M. J. Yetman, T. Lee, Y. Chen, and J. L. Jankowsky, Specificity and efficiency of reporter expression in adult neural progenitors vary substantially among nestin-CreER(T2) lines, J. Comp. Neurol, vol.522, pp.1191-1208, 2014.

M. Takada, K. Inoue, D. Koketsu, S. Kato, K. Kobayashi et al., Elucidating information processing in primate basal ganglia circuitry: a novel technique for pathway-selective ablation mediated by immunotoxin, Front. Neural Circuits, vol.7, p.140, 2013.

P. K. Thanos, L. Robison, E. J. Nestler, R. Kim, M. Michaelides et al., Mapping brain metabolic connectivity in awake rats with muPET and optogenetic stimulation, J. Neurosci, vol.33, pp.6343-6349, 2013.

R. Tomer, L. Ye, B. Hsueh, and K. Deisseroth, Advanced CLARITY for rapid and high-resolution imaging of intact tissues, Nat. Protoc, vol.9, pp.1682-1697, 2014.

A. Trabalza, C. Georgiadis, I. Eleftheriadou, J. N. Hislop, S. M. Ellison et al., Venezuelan equine encephalitis virus glycoprotein pseudotyping confers neurotropism to lentiviral vectors, Gene Ther, vol.20, pp.723-732, 2013.

D. Trono, Lentiviral vectors: turning a deadly foe into a therapeutic agent, Gene Ther, vol.7, pp.20-23, 2000.

K. M. Tye and K. Deisseroth, Optogenetic investigation of neural circuits underlying brain disease in animal models, Nat. Rev. Neurosci, vol.13, pp.251-266, 2012.

L. W. Van-hooijdonk, M. Ichwan, T. F. Dijkmans, T. G. Schouten, M. W. De-backer et al., Lentivirus-mediated transgene delivery to the hippocampus reveals sub-field specific differences in expression, BMC Neurosci, vol.10, issue.2, 2009.

I. M. Verma and N. Somia, Gene therapy -promises, problems and prospects, Nature, vol.389, pp.239-242, 1997.

E. Vigna and L. Naldini, Lentiviral vectors: excellent tools for experimental gene transfer and promising candidates for gene therapy, J. Gene Med, vol.2, pp.308-316, 2000.

M. A. Vodicka, Determinants for lentiviral infection of non-dividing cells, Somat. Cell Mol. Genet, vol.26, pp.35-49, 2001.

X. Wang, X. Chen, Y. , and Y. , Spatiotemporal control of gene expression by a light-switchable transgene system, Nat. Methods, vol.9, pp.266-269, 2012.

S. Watanabe, M. W. Davis, and E. M. Jorgenson, Flash-and-freeze electron microscopy: coupling optogenetics with high-pressure freezing, Nanoscale Imag. Synap, vol.84, pp.43-57, 2014.

D. J. Watson, G. P. Kobinger, M. A. Passini, J. M. Wilson, and W. J. , Targeted transduction patterns in the mouse brain by lentivirus vectors pseudotyped with VSV, Ebola, Mokola, LCMV, or MuLV envelope proteins, Mol. Ther, vol.5, pp.528-537, 2002.

B. Weber, Neuroscience: the case for brain imaging technology, Nature, vol.483, p.541, 2012.

T. E. Willnow, The low-density lipoprotein receptor gene family: multiple roles in lipid metabolism, J. Mol. Med. (Berl.), vol.77, pp.306-315, 1999.

M. Wiznerowicz and D. Trono, Conditional suppression of cellular genes: lentivirus vector-mediated drug-inducible RNA interference, J. Virol, vol.77, pp.8957-8961, 2003.

M. Yaguchi, Y. Ohashi, T. Tsubota, A. Sato, K. W. Koyano et al., Characterization of the properties of seven promoters in the motor cortex of rats and monkeys after lentiviral vector-mediated gene transfer, Hum. Gene Ther. Methods, vol.24, pp.333-344, 2013.

B. Yang, J. B. Treweek, R. P. Kulkarni, B. E. Deverman, C. Chen et al., Single-cell phenotyping within transparent intact tissue through whole-body clearing, Cell, vol.158, pp.945-958, 2014.

F. Zhang, A. M. Aravanis, A. Adamantidis, L. De-lecea, and K. Deisseroth, Circuit-breakers: optical technologies for probing neural signals and systems, Nat. Rev. Neurosci, vol.8, pp.577-581, 2007.

F. Zhang, V. Gradinaru, A. R. Adamantidis, R. Durand, R. D. Airan et al., Optogenetic interrogation of neural circuits: technology for probing mammalian brain structures, Nat. Protoc, vol.5, pp.439-456, 2010.

S. Zhou, D. Mody, S. S. Deravin, J. Hauer, T. Lu et al., A selfinactivating lentiviral vector for SCID-X1 gene therapy that does not activate LMO2 expression in human T cells, Blood, vol.116, pp.900-908, 2010.