S. Amin, R. Neijts, S. Simmini, C. Van-rooijen, S. C. Tan et al., Cdx and T brachyury co-activate growth signaling in the embryonic axial progenitor niche, Cell Rep, vol.17, pp.3165-3177, 2016.

S. Anders and W. Huber, Differential expression analysis for sequence count data, Genome Biol, vol.11, p.106, 2010.

S. Anders, P. T. Pyl, and W. Huber, HTSeq-a Python framework to work with high-throughput sequencing data, Bioinformatics, vol.31, pp.166-169, 2015.

K. I. Andreasson and W. E. Kaufmann, Role of immediate early gene expression in cortical morphogenesis and plasticity, Results Probl Cell Differ, vol.39, pp.113-137, 2002.

G. Barreto, A. Schäfer, J. Marhold, D. Stach, S. K. Swaminathan et al., Gadd45a promotes epigenetic gene activation by repairmediated DNA demethylation, Nature, vol.445, pp.671-675, 2007.

J. Benoit, A. E. Ayoub, and P. Rakic, Transcriptomics of critical period of visual cortical plasticity in mice, Proc Natl Acad Sci, vol.112, pp.8094-8099, 2015.

C. Bernard and A. Prochiantz, Otx2-PNN interaction to regulate cortical plasticity, Neural Plast, vol.2016, pp.1-7, 2016.

C. Bernard, C. Vincent, D. Testa, E. Bertini, J. Ribot et al., A mouse model for conditional secretion of specific single-chain antibodies provides genetic evidence for regulation of cortical plasticity by a non-cell autonomous homeoprotein transcription factor, PLoS Genet, vol.12, p.1006035, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02159992

M. Beurdeley, J. Spatazza, H. Lee, S. Sugiyama, C. Bernard et al., Otx2 binding to perineuronal nets persistently regulates plasticity in the mature visual cortex, J Neurosci, vol.32, pp.9429-9437, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02380082

J. Cang, V. A. Kalatsky, S. Löwel, and M. P. Stryker, Optical imaging of the intrinsic signal as a measure of cortical plasticity in the mouse, Vis Neurosci, vol.22, pp.685-691, 2005.

G. Chatelain, N. Fossat, G. Brun, and T. Lamonerie, Molecular dissection reveals decreased activity and not dominant negative effect in human OTX2 mutants, J Mol Med, vol.84, pp.604-615, 2006.

W. G. Chen, Q. Chang, Y. Lin, A. Meissner, A. E. West et al., Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2, Science, vol.302, pp.885-889, 2003.

L. Chen, K. Chen, L. A. Lavery, S. A. Baker, C. A. Shaw et al., MeCP2 binds to non-CG methylated DNA as neurons mature, influencing transcription and the timing of onset for Rett syndrome, Proc Natl Acad Sci, vol.112, pp.5509-5514, 2015.

D. Lullo, E. Haton, C. , L. Poupon, C. Volovitch et al., Paracrine Pax6 activity regulates oligodendrocyte precursor cell migration in the chick embryonic neural tube, Development, vol.138, pp.4991-5001, 2011.

D. Nardo, A. A. Nedelec, S. Trembleau, A. Volovitch, M. Prochiantz et al., Dendritic localization and activitydependent translation of Engrailed1 transcription factor, Mol Cell Neurosci, vol.35, pp.230-236, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00184043

S. Durand, A. Patrizi, K. B. Quast, L. Hachigian, R. Pavlyuk et al., NMDA receptor regulation prevents regression of visual cortical function in the absence of Mecp2, Neuron, vol.76, pp.1078-1090, 2012.

M. Fagiolini, C. L. Jensen, and F. A. Champagne, Epigenetic influences on brain development and plasticity, Curr Opin Neurobiol, vol.19, pp.207-212, 2009.

Y. Fu, J. M. Tucciarone, J. S. Espinosa, N. Sheng, D. P. Darcy et al., A cortical circuit for gain control by behavioral state, Cell, vol.156, pp.1139-1152, 2014.

H. W. Gabel, B. Kinde, H. Stroud, C. S. Gilbert, D. A. Harmin et al., Disruption of DNA-methylation-dependent long gene repression in Rett syndrome, Nature, vol.522, pp.89-93, 2015.

D. P. Gavin, R. P. Sharma, K. A. Chase, F. Matrisciano, E. Dong et al., Growth arrest and DNA-damage-inducible, beta (GADD45b)-mediated DNA demethylation in major psychosis, Neuropsychopharmacology, vol.37, pp.531-542, 2012.

N. Gogolla, A. E. Takesian, G. Feng, M. Fagiolini, and T. K. Hensch, Sensory integration in mouse insular cortex reflects GABA circuit maturation, Neuron, vol.83, pp.894-905, 2014.

J. A. Gordon and M. P. Stryker, Experience-dependent plasticity of binocular responses in the primary visual cortex of the mouse, J Neurosci, vol.16, pp.3274-3286, 1996.

J. U. Guo, Y. Su, J. H. Shin, J. Shin, H. Li et al., Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain, Nat Neurosci, vol.17, pp.215-222, 2014.

C. E. Hannon, S. A. Blythe, and E. F. Wieschaus, Concentration dependent chromatin states induced by the bicoid morphogen gradient, Elife, vol.6, p.3165, 2017.

R. Hattori, K. V. Kuchibhotla, R. C. Froemke, and T. Komiyama, Functions and dysfunctions of neocortical inhibitory neuron subtypes, Nat Neurosci, vol.20, pp.1199-1208, 2017.

L. He, N. Liu, T. Cheng, X. Chen, Y. Li et al., Conditional deletion of Mecp2 in parvalbuminexpressing GABAergic cells results in the absence of critical period plasticity, Nat Commun, vol.5, p.5036, 2014.

R. V. Hoch, S. Lindtner, J. D. Price, and J. Rubenstein, OTX2 transcription factor controls regional patterning within the medial ganglionic eminence and regional identity of the septum, Cell Rep, vol.12, pp.482-494, 2015.

M. C. Hollander, M. S. Sheikh, D. V. Bulavin, K. Lundgren, L. Augeri-henmueller et al., Genomic instability in Gadd45a-deficient mice, Nat Genet, vol.23, pp.176-184, 1999.

J. M. Holmes and M. P. Clarke, Amblyopia. Lancet, vol.367, pp.1343-1351, 2006.

M. Hübener and T. Bonhoeffer, Neuronal plasticity: beyond the critical period, Cell, vol.159, pp.727-737, 2014.

A. H. Joliot, A. Triller, M. Volovitch, C. Pernelle, and A. Prochiantz, Alpha-2,8-polysialic acid is the neuronal surface receptor of antennapedia homeobox peptide, New Biol, vol.3, pp.1121-1134, 1991.

L. Jourdren, M. Bernard, M. Dillies, L. Crom, and S. , Eoulsan: a cloud computing-based framework facilitating high throughput sequencing analyses, Bioinformatics, vol.28, pp.1542-1543, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02879328

L. Kaczmarek and A. Chaudhuri, Sensory regulation of immediateearly gene expression in mammalian visual cortex: implications for functional mapping and neural plasticity, Brain Res Brain Res Rev, vol.23, pp.237-256, 1997.

V. A. Kalatsky and M. P. Stryker, New paradigm for optical imaging: temporally encoded maps of intrinsic signal, Neuron, vol.38, pp.529-545, 2003.

N. Kim, D. Acampora, F. Dingli, D. Loew, A. Simeone et al., Immunoprecipitation and mass spectrometry identify non-cell autonomous Otx2 homeoprotein in the granular and supragranular layers of mouse visual cortex, vol.3, p.178, 1000.
URL : https://hal.archives-ouvertes.fr/hal-02380426

K. Krishnan, B. Lau, G. Ewall, Z. J. Huang, and S. D. Shea, MECP2 regulates cortical plasticity underlying a learned behaviour in adult female mice, Nat Commun, vol.8, p.14077, 2017.

K. Krishnan, B. Wang, J. Lu, L. Wang, A. Maffei et al., MeCP2 regulates the timing of critical period plasticity that shapes functional connectivity in primary visual cortex, Proc Natl Acad Sci, vol.112, pp.4782-4791, 2015.

I. Kruglikov and B. Rudy, Perisomatic GABA release and thalamocortical integration onto neocortical excitatory cells are regulated by neuromodulators, Neuron, vol.58, pp.911-924, 2008.

S. J. Kuhlman, N. D. Olivas, E. Tring, T. Ikrar, X. Xu et al., A disinhibitory microcircuit initiates critical-period plasticity in the visual cortex, Nature, vol.501, pp.543-546, 2013.

Y. Kumaki, M. Oda, and M. Okano, QUMA: quantification tool for methylation analysis, Nucleic Acids Res, vol.36, pp.170-175, 2008.

B. Langmead, C. Trapnell, M. Pop, and S. L. Salzberg, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome, Genome Biol, vol.10, p.25, 2009.

P. T. Leach, S. G. Poplawski, J. W. Kenney, B. Hoffman, D. A. Liebermann et al., Gadd45b knockout mice exhibit selective deficits in hippocampus-dependent long-term memory, Learn Mem, vol.19, pp.319-324, 2012.

H. Lee, C. Bernard, Z. Ye, D. Acampora, A. Simeone et al., Genetic Otx2 mislocalization delays critical period plasticity across brain regions, Mol Psychiatry, vol.22, pp.680-688, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02380572

A. Lennartsson, E. Arner, M. Fagiolini, A. Saxena, R. Andersson et al., Remodeling of retrotransposon elements during epigenetic induction of adult visual cortical plasticity by HDAC inhibitors, Epigenetics Chromatin, vol.8, p.55, 2015.

L. Li, J. Carter, X. Gao, J. Whitehead, and W. G. Tourtellotte, The neuroplasticity-associated arc gene is a direct transcriptional target of early growth response (Egr) transcription factors, Mol Cell Biol, vol.25, pp.10286-10300, 2005.

L. Li and R. Dahiya, MethPrimer: designing primers for methylation PCRs, Bioinformatics, vol.18, pp.1427-1431, 2002.

H. Li, B. Handsaker, A. Wysoker, T. Fennell, J. Ruan et al., Genome Project Data Cortical OTX2 Targets Gadd45b/g Apulei et al. | 11, 1000.

, The sequence alignment/map format and SAMtools, Bioinformatics, vol.25, pp.2078-2079, 2009.

D. K. Ma, M. Jang, J. U. Guo, Y. Kitabatake, M. Chang et al., Neuronal activityinduced Gadd45b promotes epigenetic DNA demethylation and adult neurogenesis, Science, vol.323, pp.1074-1077, 2009.

M. Majdan and C. J. Shatz, Effects of visual experience on activity-dependent gene regulation in cortex, Nat Neurosci, vol.9, pp.650-659, 2006.

E. Matsunaga, S. Nambu, M. Oka, and A. Iriki, Comparative analysis of developmentally regulated expressions of Gadd45a, Gadd45b, and Gadd45g in the mouse and marmoset cerebral cortex, Neuroscience, vol.284, pp.566-580, 2015.

M. Mellén, P. Ayata, S. Dewell, S. Kriaucionis, and N. Heintz, MeCP2 binds to 5hmC enriched within active genes and accessible chromatin in the nervous system, Cell, vol.151, pp.1417-1430, 2012.

X. Meng, W. Wang, H. Lu, L. He, W. Chen et al., Manipulations of MeCP2 in glutamatergic neurons highlight their contributions to Rett and other neurological disorders, p.185, 2016.

A. Mo, E. A. Mukamel, F. P. Davis, C. Luo, G. L. Henry et al., Epigenomic signatures of neuronal diversity in the mammalian brain, Neuron, vol.86, pp.1369-1384, 2015.

C. W. Mount and M. Monje, Wrapped to adapt: experiencedependent myelination, Neuron, vol.95, pp.743-756, 2017.

C. Niehrs and A. Schäfer, Active DNA demethylation by Gadd45 and DNA repair, Trends Cell Biol, vol.22, pp.220-227, 2012.

A. Nott, S. Cho, J. Seo, and L. Tsai, HDAC2 expression in parvalbumin interneurons regulates synaptic plasticity in the mouse visual cortex, Neuroepigenetics, vol.1, pp.34-40, 2015.

G. T. Ooi, D. R. Brown, D. S. Suh, L. Y. Tseng, and M. M. Rechler, Cycloheximide stabilizes insulin-like growth factor-binding protein-1 (IGFBP-1) mRNA and inhibits IGFBP-1 transcription in H4-II-E rat hepatoma cells, J Biol Chem, vol.268, pp.16664-16672, 1993.

H. Pi, B. Hangya, D. Kvitsiani, J. I. Sanders, Z. J. Huang et al., Cortical interneurons that specialize in disinhibitory control, Nature, vol.503, pp.521-524, 2013.

A. Prochiantz, D. Nardo, and A. A. , Homeoprotein signaling in the developing and adult nervous system, Neuron, vol.85, pp.911-925, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02380506

E. Putignano, G. Lonetti, L. Cancedda, G. Ratto, M. Costa et al., Developmental downregulation of histone posttranslational modifications regulates visual cortical plasticity, Neuron, vol.53, pp.747-759, 2007.

K. Rai, I. J. Huggins, S. R. James, A. R. Karpf, D. A. Jones et al., DNA demethylation in zebrafish involves the coupling of a deaminase, a glycosylase, and gadd45, Cell, vol.135, pp.1201-1212, 2008.

A. Ranson, C. Cheetham, K. Fox, and F. Sengpiel, Homeostatic plasticity mechanisms are required for juvenile, but not adult, ocular dominance plasticity, Proc Natl Acad Sci, vol.109, pp.1311-1316, 2012.

H. Rekaik, F. Blaudin-de-thé, J. Fuchs, O. Massiani-beaudoin, A. Prochiantz et al., Engrailed homeoprotein protects mesencephalic dopaminergic neurons from oxidative stress, Cell Rep, vol.13, pp.242-250, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02156811

M. L. Rietman and J. Sommeijer, Neuro-Bsik Mouse Phenomics Consortium. Levelt CN, Heimel JA. 2012. Candidate genes in ocular dominance plasticity, Front Neurosci, vol.6, p.11

C. Sampathkumar, Y. Wu, M. Vadhvani, T. Trimbuch, B. Eickholt et al., Loss of MeCP2 disrupts cell autonomous and autocrine BDNF signaling in mouse glutamatergic neurons, Elife, vol.5, p.214, 2016.

A. Samuel, M. Housset, B. Fant, and T. Lamonerie, Otx2 ChIP-seq reveals unique and redundant functions in the mature mouse retina, PLoS One, vol.9, p.89110, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00968745

M. Sato and M. P. Stryker, Distinctive features of adult ocular dominance plasticity, J Neurosci, vol.28, pp.10278-10286, 2008.

A. V. Sharma, F. E. Nargang, and C. T. Dickson, Neurosilence: profound suppression of neural activity following intracerebral administration of the protein synthesis inhibitor anisomycin, J Neurosci, vol.32, pp.2377-2387, 2012.

J. D. Shepherd and M. F. Bear, New views of Arc, a master regulator of synaptic plasticity, Nat Neurosci, vol.14, pp.279-284, 2011.

J. Spatazza, H. Lee, D. Nardo, A. A. Tibaldi, L. Joliot et al., Choroid-plexus-derived Otx2 homeoprotein constrains adult cortical plasticity, Cell Rep, vol.3, pp.1815-1823, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02380061

O. Stettler, R. L. Joshi, A. Wizenmann, J. Reingruber, D. Holcman et al., Engrailed homeoprotein recruits the adenosine A1 receptor to potentiate ephrin A5 function in retinal growth cones, Development, vol.139, pp.215-224, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02160114

S. Sugiyama, D. Nardo, A. A. Aizawa, S. Matsuo, I. Volovitch et al., Experience-dependent transfer of Otx2 homeoprotein into the visual cortex activates postnatal plasticity, Cell, vol.134, pp.508-520, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02380510

F. A. Sultan, J. Wang, J. Tront, D. A. Liebermann, and J. D. Sweatt, Genetic deletion of Gadd45b, a regulator of active DNA demethylation, enhances long-term memory and synaptic plasticity, J Neurosci, vol.32, pp.17059-17066, 2012.

A. E. Takesian and T. K. Hensch, Balancing plasticity/stability across brain development, Prog Brain Res, vol.207, pp.3-34, 2013.

E. Tiraboschi, R. Guirado, D. Greco, P. Auvinen, M. Vetencourt et al., Gene expression patterns underlying the reinstatement of plasticity in the adult visual system, Neural Plast, p.605079, 2013.

P. Tognini, D. Napoli, J. Tola, D. Silingardi, R. Della et al., Experience-dependent DNA methylation regulates plasticity in the developing visual cortex, Nat Neurosci, vol.18, pp.956-958, 2015.

G. S. Tomassy, N. Morello, E. Calcagno, and M. Giustetto, Developmental abnormalities of cortical interneurons precede symptoms onset in a mouse model of Rett syndrome, J Neurochem, vol.131, pp.115-127, 2014.

A. Vallès, A. J. Boender, S. Gijsbers, R. Haast, G. Martens et al., Genomewide analysis of rat barrel cortex reveals time-and layer-specific mRNA expression changes related to experience-dependent plasticity, J Neurosci, vol.31, pp.374-383, 2011.

A. Wizenmann, I. Brunet, J. Lam, L. Sonnier, M. Beurdeley et al., Extracellular Engrailed participates in the topographic guidance of retinal axons in vivo, Neuron, vol.64, pp.355-366, 2009.

T. Yokoo, B. W. Knight, and L. Sirovich, An optimization approach to signal extraction from noisy multivariate data, Neuroimage, vol.14, pp.1309-1326, 2001.

Y. Yotsumoto, L. Chang, R. Ni, R. Pierce, G. J. Andersen et al., White matter in the older brain is more plastic than in the younger brain, Nat Commun, vol.5, p.5504, 2014.

D. Alvarez-fischer, J. Fuchs, F. Castagner, O. Stettler, O. Massiani-beaudoin et al., Engrailed protects mouse midbrain dopaminergic neurons against mitochondrial complex I insults, Nat. Neurosci, vol.14, pp.1260-1266, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00676206

J. Apulei, N. Kim, D. Testa, J. Ribot, D. Morizet et al., Non-cell Autonomous OTX2 Homeoprotein Regulates Visual Cortex Plasticity Through Gadd45b/g. Cereb. Cortex, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02379324

C. Bernard, H. Kim, R. Torero-ibad, E. J. Lee, M. Simonutti et al., Graded Otx2 activities demonstrate dosesensitive eye and retina phenotypes, Hum. Mol. Genet, vol.23, pp.1742-1753, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02380495

C. Bernard, C. Vincent, D. Testa, E. Bertini, J. Ribot et al., A Mouse Model for Conditional Secretion of Specific Single-Chain Antibodies Provides Genetic Evidence for Regulation of Cortical Plasticity by a Non-cell Autonomous Homeoprotein Transcription Factor, PLOS Genet, vol.12, p.1006035, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02159992

M. Beurdeley, J. Spatazza, H. H. Lee, S. Sugiyama, C. Bernard et al., Otx2 Binding to Perineuronal Nets Persistently Regulates Plasticity in the Mature Visual Cortex, J. Neurosci, vol.32, pp.9429-9437, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02380082

F. Blaudin-de-thé, H. Rekaik, E. Peze-heidsieck, O. Massiani-beaudoin, R. L. Joshi et al., Engrailed homeoprotein blocks degeneration in adult dopaminergic neurons through LINE-1 repression, EMBO J. e97374, 2018.

G. Bourque, B. Leong, V. B. Vega, X. Chen, Y. L. Lee et al., Evolution of the mammalian transcription factor binding repertoire via transposable elements, Genome Res, vol.18, pp.1752-1762, 2008.

A. Bulut-karslioglu, V. Perrera, M. Scaranaro, I. A. De-la-rosa-velazquez, S. Van-de-nobelen et al., A transcription factor-based mechanism for mouse heterochromatin formation, Nat. Struct. Mol. Biol, vol.19, pp.1023-1030, 2012.

J. Cholewa-waclaw, A. Bird, M. Von-schimmelmann, A. Schaefer, H. Yu et al., The Role of Epigenetic Mechanisms in the Regulation of Gene Expression in the Nervous System, J. Neurosci, vol.36, pp.11427-11434, 2016.

E. J. Duncan, P. D. Gluckman, and P. K. Dearden, Epigenetics, plasticity, and evolution: How do we link epigenetic change to phenotype?, J. Exp. Zoolog. B Mol. Dev. Evol, vol.322, pp.208-220, 2014.

M. Fagiolini, C. L. Jensen, and F. A. Champagne, Epigenetic influences on brain development and plasticity, Curr. Opin. Neurobiol, vol.19, pp.207-212, 2009.

J. L. Garcia-perez, M. C. Marchetto, A. R. Muotri, N. G. Coufal, F. H. Gage et al., LINE-1 retrotransposition in human embryonic stem cells, Hum. Mol. Genet, vol.16, pp.1569-1577, 2007.

J. L. Goodier, A potential role for the nucleolus in L1 retrotransposition, Hum. Mol. Genet, vol.13, pp.1041-1048, 2004.

W. Härtig, K. Brauer, and G. Brückner, Wisteria floribunda agglutinin-labelled nets surround parvalbumin-containing neurons, NeuroReport, vol.3, pp.869-872, 1992.

J. He, X. Fu, M. Zhang, F. He, W. Li et al., Transposable elements are regulated by context-specific patterns of chromatin marks in mouse embryonic stem cells, Nat. Commun, vol.10, p.34, 2019.

T. K. Hensch, Critical period plasticity in local cortical circuits, Nat. Rev. Neurosci, vol.6, pp.877-888, 2005.

C. Holmes, Amblyopia. Lancet, vol.367, pp.1343-51, 2006.

R. G. Hunter, G. Murakami, S. Dewell, M. Seligsohn, M. E. Baker et al., Acute stress and hippocampal histone H3 lysine 9 trimethylation, a retrotransposon silencing response, Proc. Natl. Acad. Sci, vol.109, pp.17657-17662, 2012.

R. T. Ibad, J. Rheey, S. Mrejen, V. Forster, S. Picaud et al., Otx2 Promotes the Survival of Damaged Adult Retinal Ganglion Cells and Protects against Excitotoxic Loss of Visual Acuity In Vivo, J. Neurosci, vol.31, pp.5495-5503, 2011.

R. B. Jones, K. E. Garrison, J. C. Wong, E. H. Duan, D. F. Nixon et al., , 2008.

, Nucleoside Analogue Reverse Transcriptase Inhibitors Differentially Inhibit Human LINE, issue.1

, PLoS ONE, vol.3, 1547.

G. Köppe, G. Brückner, K. Brauer, W. Härtig, and V. Bigl, Developmental patterns of proteoglycan-containing extracellular matrix in perineuronal nets and neuropil of the postnatal rat brain, Cell Tissue Res, vol.288, pp.33-41, 1997.

H. H. Lee, C. Bernard, Z. Ye, D. Acampora, A. Simeone et al., Genetic Otx2 mis-localization delays critical period plasticity across brain regions, Mol. Psychiatry, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02380572

R. Madabhushi, F. Gao, A. R. Pfenning, L. Pan, S. Yamakawa et al., Activity-Induced DNA Breaks Govern the Expression of Neuronal Early-Response Genes, Cell, vol.161, pp.1592-1605, 2015.

I. Maze, J. Feng, M. B. Wilkinson, H. Sun, L. Shen et al., Cocaine dynamically regulates heterochromatin and repetitive element unsilencing in nucleus accumbens, Proc. Natl. Acad. Sci, vol.108, pp.3035-3040, 2011.

S. Miyata, Y. Komatsu, Y. Yoshimura, C. Taya, and H. Kitagawa, Persistent cortical plasticity by upregulation of chondroitin 6-sulfation, Nat. Neurosci, vol.15, pp.414-422, 2012.

A. R. Muotri, V. T. Chu, M. C. Marchetto, W. Deng, J. V. Moran et al., Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition, Nature, vol.435, pp.903-910, 2005.

A. R. Muotri, C. Zhao, M. C. Marchetto, and F. H. Gage, Environmental Influence on L1 Retrotransposons in the Adult Hippocampus, Hippocampus, vol.19, pp.1002-1007, 2009.

D. Pezic, S. A. Manakov, R. Sachidanandam, and A. A. Aravin, piRNA pathway targets active LINE1 elements to establish the repressive H3K9me3 mark in germ cells, Genes Dev, vol.28, pp.1410-1428, 2014.

A. Prochiantz, D. Nardo, and A. A. , Homeoprotein Signaling in the Developing and Adult Nervous System, Neuron, vol.85, pp.911-925, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02380506

H. Rekaik, F. Blaudin-de-thé, J. Fuchs, O. Massiani-beaudoin, A. Prochiantz et al., Engrailed Homeoprotein Protects Mesencephalic Dopaminergic Neurons from Oxidative Stress, Cell Rep, vol.13, pp.242-250, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02156811

N. Saksouk, E. Simboeck, and J. Déjardin, Constitutive heterochromatin formation and transcription in mammals, Epigenetics Chromatin, vol.8, p.3, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01144005

T. Singer, M. J. Mcconnell, M. C. Marchetto, N. G. Coufal, and F. H. Gage, LINE-1 retrotransposons: mediators of somatic variation in neuronal genomes?, Trends Neurosci, vol.33, pp.345-354, 2010.

L. Sonnier, G. Le-pen, A. Hartmann, J. Bizot, F. Trovero et al., Progressive Loss of Dopaminergic Neurons in the Ventral Midbrain of Adult Mice Heterozygote for Engrailed1, J. Neurosci, vol.27, pp.1063-1071, 2007.

J. Spatazza, H. H. Lee, A. A. Di-nardo, L. Tibaldi, A. Joliot et al., Choroid-Plexus-Derived Otx2 Homeoprotein Constrains Adult Cortical Plasticity, Cell Rep, vol.3, pp.1815-1823, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02380061

J. Spatazza, E. Di-lullo, A. Joliot, E. Dupont, K. L. Moya et al., , 2013.

, Homeoprotein Signaling in Development, Health, and Disease: A Shaking of Dogmas Offers Challenges and Promises from Bench to Bed, Pharmacol. Rev, vol.65, pp.90-104

S. Sugiyama, A. A. Di-nardo, S. Aizawa, I. Matsuo, M. Volovitch et al., Experience-Dependent Transfer of Otx2 Homeoprotein into the Visual Cortex Activates Postnatal Plasticity, Cell, vol.134, pp.508-520, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02380510

C. A. Thomas, A. C. Paquola, and A. R. Muotri, LINE-1 Retrotransposition in the Nervous System, Annu. Rev. Cell Dev. Biol, vol.28, pp.555-573, 2012.

M. Van-meter, M. Kashyap, S. Rezazadeh, A. J. Geneva, T. D. Morello et al., SIRT6 represses LINE1 retrotransposons by ribosylating KAP1 but this repression fails with stress and age, Nat. Commun, vol.5, p.5011, 2014.

T. Wang, J. Zeng, C. B. Lowe, R. G. Sellers, S. R. Salama et al., Species-specific endogenous retroviruses shape the transcriptional network of the human tumor suppressor protein p53, Proc. Natl. Acad. Sci, vol.104, pp.18613-18618, 2007.

T. N. Wiesel and D. H. Hubel, SINGLE-CELL RESPONSES IN STRIATE CORTEX OF KITTENS DEPRIVED OF VISION IN ONE EYE, J. Neurophysiol, vol.26, pp.1003-1017, 1963.

A. Wylie, A. E. Jones, A. Brot, W. Lu, P. Kurtz et al., , 2016.

D. Acampora, M. Gulisano, V. Broccoli, and A. Simeone, Otx genes in brain morphogenesis, Prog. Neurobiol, vol.64, pp.69-95, 2001.

D. Acampora, S. Mazan, Y. Lallemand, V. Avantaggiato, M. Maury et al.,

Z. Agoston and D. Schulte, Meis2 competes with the Groucho co-repressor Tle4 for binding to Otx2 and specifies tectal fate without induction of a secondary midbrain-hindbrain boundary organizer, Development, vol.136, pp.3311-3322, 2009.

S. Akbarian, Gene Expression for Glutamic Acid Decarboxylase Is Reduced Without Loss of Neurons in Prefrontal Cortex of Schizophrenics, Arch. Gen. Psychiatry, vol.52, p.258, 1995.

R. E. Amir, I. B. Van-den-veyver, M. Wan, C. Q. Tran, U. Francke et al., Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2, 1999.

. Genet, , vol.23, pp.185-188

A. Antonini, M. Fagiolini, and M. P. Stryker, Anatomical Correlates of Functional Plasticity in Mouse Visual Cortex, J. Neurosci, vol.19, pp.4388-4406, 1999.

J. Apulei, N. Kim, D. Testa, J. Ribot, D. Morizet et al., Non-cell Autonomous OTX2 Homeoprotein Regulates Visual Cortex Plasticity Through Gadd45b/g. Cereb. Cortex, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02379324

A. A. Aravin, R. Sachidanandam, D. Bourc'his, C. Schaefer, D. Pezic et al., A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice, Mol. Cell, vol.31, pp.785-799, 2008.

J. K. Atwal, J. Pinkston-gosse, J. Syken, S. Stawicki, Y. Wu et al., PirB is a Functional Receptor for Myelin Inhibitors of Axonal Regeneration, Science, vol.322, pp.967-970, 2008.

J. K. Baillie, M. W. Barnett, K. R. Upton, D. J. Gerhardt, T. A. Richmond et al., Somatic retrotransposition alters the genetic landscape of the human brain, Nature, vol.479, pp.534-537, 2011.

D. Baker-andresen, V. S. Ratnu, and T. W. Bredy, Dynamic DNA methylation: a prime candidate for genomic metaplasticity and behavioral adaptation, Trends Neurosci, vol.36, pp.3-13, 2013.

M. P. Ball, J. B. Li, Y. Gao, J. Lee, E. Leproust et al., Targeted and genome-scale methylomics reveals gene body signatures in human cell lines, Nat. Biotechnol, vol.27, pp.361-368, 2009.

G. Barreto, A. Schäfer, J. Marhold, D. Stach, S. K. Swaminathan et al., Gadd45a promotes epigenetic gene activation by repairmediated DNA demethylation, Nature, vol.445, pp.671-675, 2007.

E. Bártová, J. Krej?í, A. Harni?arová, G. Galiová, and S. Kozubek, Histone Modifications and Nuclear Architecture: A Review, J. Histochem. Cytochem, vol.56, pp.711-721, 2008.

C. R. Beck, J. L. Garcia-perez, R. M. Badge, and J. V. Moran, LINE-1 elements in structural variation and disease, Annu. Rev. Genomics Hum. Genet, vol.12, pp.187-215, 2011.

V. P. Belancio, A. M. Roy-engel, and P. Deininger, The impact of multiple splice sites in human L1 elements, Gene, vol.411, pp.38-45, 2008.

J. E. Belforte, V. Zsiros, E. R. Sklar, Z. Jiang, G. Yu et al.,

, Postnatal NMDA receptor ablation in corticolimbic interneurons confers schizophrenia-like phenotypes, Nat. Neurosci, vol.13, pp.76-83

J. Ben-hattar, J. , and J. , Methylation of single CpG dinucleotides within a promoter element of the Herpes simplex virus tk gene reduces its transcription in vivo, Gene, vol.65, pp.219-227, 1988.

N. Berardi, T. Pizzorusso, G. M. Ratto, and L. Maffei, Molecular basis of plasticity in the visual cortex, Trends Neurosci, vol.26, pp.369-378, 2003.

J. Berlose, O. Convert, D. Derossi, A. Brunissen, and G. Chassaing, Conformational and Associative Behaviours of the Third Helix of Antennapedia Homeodomain in Membrane-Mimetic Environments, Eur. J. Biochem, vol.242, pp.372-386, 1996.

C. Bernard, C. Vincent, D. Testa, E. Bertini, J. Ribot et al., A Mouse Model for Conditional Secretion of Specific Single-Chain Antibodies Provides Genetic Evidence for Regulation of Cortical Plasticity by a Non-cell, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02159992

, Autonomous Homeoprotein Transcription Factor, PLOS Genet, vol.12, 1006035.

M. Beurdeley, J. Spatazza, H. H. Lee, S. Sugiyama, C. Bernard et al., Otx2 Binding to Perineuronal Nets Persistently Regulates Plasticity in the Mature Visual Cortex, J. Neurosci, vol.32, pp.9429-9437, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02380082

M. Billeter, Y. Qian, G. Otting, M. Müller, W. J. Gehring et al., Determination of the three-dimensional structure of the Antennapedia homeodomain from Drosophila in solution by 1H nuclear magnetic resonance spectroscopy, J. Mol. Biol, vol.214, pp.83-197, 1990.

M. Billeter, Y. Q. Qian, G. Otting, M. Müller, W. Gehring et al., Determination of the nuclear magnetic resonance solution structure of an Antennapedia homeodomain-DNA complex, J. Mol. Biol, vol.234, pp.1084-1093, 1993.

A. P. Bird, CpG-rich islands and the function of DNA methylation, Nature, vol.321, pp.209-213, 1986.

F. Blaudin-de-thé, H. Rekaik, E. Peze-heidsieck, O. Massiani-beaudoin, R. L. Joshi et al., , 2018.

I. Brunet, C. Weinl, M. Piper, A. Trembleau, M. Volovitch et al., The transcription factor Engrailed-2 guides retinal axons, Nature, vol.438, pp.94-98, 2005.

M. Bundo, M. Toyoshima, Y. Okada, W. Akamatsu, J. Ueda et al., Increased L1 Retrotransposition in the Neuronal Genome in Schizophrenia, Neuron, vol.81, pp.306-313, 2014.

A. Caballero, E. Flores-barrera, D. K. Cass, and K. Y. Tseng, Differential regulation of parvalbumin and calretinin interneurons in the prefrontal cortex during adolescence, Brain Struct, 2014.

. Funct, , vol.219, pp.395-406

R. J. Cabelli, D. L. Shelton, R. A. Segal, and C. J. Shatz, Blockade of Endogenous Ligands of TrkB Inhibits Formation of Ocular Dominance Columns, Neuron, vol.19, pp.63-76, 1997.

A. D. Cardin, H. J. Weintraub, A. Thromb, V. Biol, A. D. Cardin et al., J.R. Protein-Glycosaminoglycan Interactions

M. A. Carmell, A. Girard, H. J. Van-de-kant, D. Bourc'his, T. H. Bestor et al.,

G. J. Hannon, MIWI2 Is Essential for Spermatogenesis and Repression of Transposons in the Mouse Male Germline, Dev. Cell, vol.12, pp.503-514, 2007.

A. E. Carrasco, W. Mcginnis, W. J. Gehring, and E. M. De-robertis, , 1984.

M. C. Chang, J. M. Park, K. A. Pelkey, H. L. Grabenstatter, D. Xu et al., Narp regulates homeostatic scaling of excitatory synapses on parvalbumin-expressing interneurons, Nat. Neurosci, vol.13, pp.1090-1097, 2010.

B. Christiaens, S. Symoens, S. Vanderheyden, Y. Engelborghs, A. Joliot et al.,

J. Vandekerckhove, M. Rosseneu, and B. Vanloo, Tryptophan fluorescence study of the interaction of penetratin peptides with model membranes, Eur. J. Biochem, vol.269, pp.2918-2926, 2002.

D. N. Cooper, M. H. Taggart, and A. P. Bird, Unmethylated domains in vertebrate DNA, Nucleic Acids Res, vol.11, pp.647-658, 1983.

N. G. Coufal, J. L. Garcia-perez, G. E. Peng, G. W. Yeo, Y. Mu et al., L1 retrotransposition in human neural progenitor cells, Nature, vol.460, pp.1127-1131, 2009.

N. G. Coufal, J. L. Garcia-perez, G. E. Peng, M. C. Marchetto, A. R. Muotri et al., Ataxia telangiectasia mutated (ATM) modulates long interspersed element-1 (L1) retrotransposition in human neural stem cells, Proc. Natl. Acad. Sci. U, 2011.

S. , , vol.108, pp.20382-20387

G. D. Cristo, Requirement of ERK Activation for Visual Cortical Plasticity, Science, vol.292, pp.2337-2340, 2001.

D. Derossi, A. Joliot, G. Chassaing, and A. Prochiantz, The third helix of the Antennapedia homeodomain translocates through biological membranes, J. Biol. Chem, vol.269, pp.10444-10450, 1994.

D. Derossi, S. Calvet, A. Trembleau, A. Brunissen, G. Chassaing et al., Cell Internalization of the Third Helix of the Antennapedia Homeodomain Is Receptor-independent, J. Biol. Chem, vol.271, pp.18188-18193, 1996.

E. Di-lullo, C. Haton, C. Le-poupon, M. Volovitch, A. Joliot et al.,

D. Dobbs, Schizophrenia appears during adolescence, 2010.

J. Dubnau and G. Struhl, RNA recognition and translational regulation, 1996.

E. Dupont, A. Prochiantz, J. , and A. , Identification of a Signal Peptide for Unconventional Secretion*?S, vol.282, 2007.

J. A. Erwin, M. C. Marchetto, and F. H. Gage, Mobile DNA elements in the generation of diversity and complexity in the brain, Nat. Rev. Neurosci, vol.15, pp.497-506, 2014.

M. Fagiolini, Specific GABAA Circuits for Visual Cortical Plasticity, Science, vol.303, pp.1681-1683, 2004.

M. Fagiolini and T. K. Hensch, Inhibitory threshold for critical-period activation in primary visual cortex, Nature, vol.404, pp.183-186, 2000.

M. Fagiolini, C. L. Jensen, and F. A. Champagne, Epigenetic influences on brain development and plasticity, Curr. Opin. Neurobiol, vol.19, pp.207-212, 2009.

M. Fasolino and Z. Zhou, The Crucial Role of DNA Methylation and MeCP2 in Neuronal Function, Genes, vol.8, 2017.

J. Feng, Y. Zhou, S. L. Campbell, T. Le, E. Li et al., , p.1, 2010.

, and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons

, Nat. Neurosci, vol.13, pp.423-430

R. Fine, J. Zhang, and H. E. Stevens, Prenatal stress and inhibitory neuron systems: implications for neuropsychiatric disorders, Mol. Psychiatry, vol.19, pp.641-651, 2014.

D. Frantz, M. Weimann, E. Levin, and K. Mcconnell, Otxl and Otx2 Define Layers and Regions in Developing Cerebral Cortex and Cerebellum, vol.16

M. J. Gandal, A. M. Nesbitt, R. M. Mccurdy, and M. D. Alter, Measuring the Maturity of the Fast-Spiking Interneuron Transcriptional Program in Autism, Schizophrenia, and Bipolar Disorder, 2012.

, PLoS ONE, vol.7, 41215.

J. L. Garcia-perez, M. Morell, J. O. Scheys, D. A. Kulpa, S. Morell et al.,

K. L. Collins, K. S. Shea, and P. Menendez, Epigenetic silencing of engineered L1 retrotransposition events in human embryonic carcinoma cells, Nature, vol.466, pp.769-773, 2010.

S. L. Gasior, T. P. Wakeman, B. Xu, and P. L. Deininger, The Human LINE-1, 2006.

, Retrotransposon Creates DNA Double-strand Breaks, J. Mol. Biol, vol.357, pp.1383-1393

D. P. Gavin, R. P. Sharma, K. A. Chase, F. Matrisciano, E. Dong et al., Growth arrest and DNA-damage-inducible, beta (GADD45b)-mediated DNA demethylation in major psychosis, Neuropsychopharmacology, vol.37, pp.531-542, 2012.

W. Gehring, Clonal analysis of determination dynamics in cultures of imaginal disks in Drosophila melanogaster, Dev. Biol, vol.16, pp.438-456, 1967.

J. Gervain, B. W. Vines, L. M. Chen, R. J. Seo, T. K. Hensch et al., , 2013.

, Valproate reopens critical-period learning of absolute pitch, Front. Syst. Neurosci, vol.7

N. Gilbert, S. Lutz, T. A. Morrish, and J. V. Moran, Multiple Fates of L1 Retrotransposition Intermediates in Cultured Human Cells, Mol. Cell. Biol, vol.25, pp.7780-7795, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00016573

N. Gogolla, P. Caroni, A. Lüthi, and C. Herry, Perineuronal Nets Protect Fear Memories from Erasure, Science, vol.325, pp.1258-1261, 2009.

A. Gordon and P. Sttyker, Plasticity of Binocular Responses in the, vol.13

D. Grassi, H. Franz, R. Vezzali, P. Bovio, S. Heidrich et al., Neuronal Activity, TGF?-Signaling and Unpredictable Chronic Stress Modulate Transcription of Gadd45 Family Members and DNA Methylation in the Hippocampus, Cereb. Cortex, pp.1-16, 2017.

Y. Gu, S. Huang, M. Chang, P. Worley, A. Kirkwood et al., Obligatory role for the immediate early gene NARP in critical period plasticity, Neuron, vol.79, pp.335-346, 2013.

J. Guan, S. J. Haggarty, E. Giacometti, J. Dannenberg, N. Joseph et al., HDAC2 negatively regulates memory formation and synaptic plasticity, Nature, vol.459, pp.55-60, 2009.

J. U. Guo, D. K. Ma, H. Mo, M. P. Ball, M. Jang et al.,

B. Xie and E. Ford, Neuronal activity modifies the DNA methylation landscape in the adult brain, Nat. Neurosci, vol.14, pp.1345-1351, 2011.

C. E. Hannon, S. A. Blythe, and E. F. Wieschaus, , 2017.

W. Härtig, K. Brauer, and G. Brückner, Wisteria floribunda agglutinin-labelled nets surround parvalbumin-containing neurons, NeuroReport, vol.3, pp.869-872, 1992.

T. Hashimoto, D. W. Volk, S. M. Eggan, K. Mirnics, J. N. Pierri et al.,

D. A. Lewis, Gene Expression Deficits in a Subclass of GABA Neurons in the Prefrontal Cortex of Subjects with Schizophrenia, J. Neurosci, vol.23, pp.6315-6326, 2003.

C. A. Hassig, T. C. Fleischer, A. N. Billin, S. L. Schreiber, and D. E. Ayer, , 1997.

, Deacetylase Activity Is Required for Full Transcriptional Repression by mSin3A, Cell, vol.89, pp.341-347

H. He, B. Ray, K. Dennis, and E. M. Quinlan, Experience-dependent recovery of vision following chronic deprivation amblyopia, Nat. Neurosci, vol.10, pp.1134-1136, 2007.

L. He, N. Liu, T. Cheng, X. Chen, Y. Li et al., Conditional deletion of Mecp2 in parvalbumin-expressing GABAergic cells results in the absence of critical period plasticity, Nat. Commun, vol.5, p.5036, 2014.

T. K. Hensch, Local GABA Circuit Control of Experience-Dependent Plasticity in Developing Visual Cortex, Science, vol.282, pp.1504-1508, 1998.

T. K. Hensch, CRITICAL PERIOD REGULATION, Annu. Rev. Neurosci, vol.27, pp.549-579, 2004.

T. K. Hensch, Critical period plasticity in local cortical circuits, Nat. Rev. Neurosci, vol.6, pp.877-888, 2005.

C. Holmes, Amblyopia. Lancet, vol.367, pp.1343-51, 2006.

G. P. Holmquist, Role of replication time in the control of tissue-specific gene expression, 1987.

, Am. J. Hum. Genet, vol.40, pp.151-173

J. C. Horton and D. R. Hocking, Timing of the Critical Period for Plasticity of Ocular Dominance Columns in Macaque Striate Cortex, J. Neurosci, vol.17, pp.3684-3709, 1997.

Z. J. Huang, A. Kirkwood, T. Pizzorusso, V. Porciatti, B. Morales et al., BDNF Regulates the Maturation of Inhibition and the Critical Period of Plasticity in Mouse Visual Cortex, Cell, vol.98, pp.739-755, 1999.

S. M. Iguchi-ariga and W. Schaffner, CpG methylation of the cAMP-responsive enhancer/promoter sequence TGACGTCA abolishes specific factor binding as well as transcriptional activation, Genes Dev, vol.3, pp.612-619, 1989.

T. R. Insel, Rethinking schizophrenia, Nature, vol.468, pp.187-193, 2010.

N. P. Issa, J. T. Trachtenberg, B. Chapman, K. R. Zahs, and M. P. Stryker, The Critical Period for Ocular Dominance Plasticity in the Ferret's Visual Cortex, J. Neurosci. Off. J. Soc. Neurosci, vol.19, pp.6965-6978, 1999.

S. Ito, L. Shen, Q. Dai, S. C. Wu, L. B. Collins et al., Tet Proteins Can Convert 5-Methylcytosine to 5-Formylcytosine and 5-Carboxylcytosine, Science, vol.333, pp.1300-1303, 2011.

B. Jiang, S. Huang, R. De-pasquale, D. Millman, L. Song et al., The Maturation of GABAergic Transmission in Visual Cortex Requires Endocannabinoid-Mediated LTD of Inhibitory Inputs during a Critical Period, Neuron, vol.66, pp.248-259, 2010.

P. A. Johansson, M. Irmler, D. Acampora, J. Beckers, A. Simeone et al., The transcription factor Otx2 regulates choroid plexus development and function, Development, vol.140, pp.1055-1066, 2013.

A. Joliot and A. Prochiantz, Transduction peptides: from technology to physiology, Nat. Cell Biol, vol.6, pp.189-196, 2004.

A. Joliot, C. Pernelle, H. Deagostini-bazin, and A. Prochiantz, Antennapedia homeobox peptide regulates neural morphogenesis, Proc. Natl. Acad. Sci, vol.88, pp.1864-1868, 1991.

A. Joliot, A. Trembleau, G. Raposo, S. Calvet, M. Volovitch et al., Association of Engrailed homeoproteins with vesicles presenting caveolae-like properties, p.11

H. K. Jr, C. Wong, H. Youssoufian, A. F. Scottt, and D. G. Phillips, , 1988.

B. P. Jung, D. G. Jugloff, G. Zhang, R. Logan, S. Brown et al., The expression of methyl CpG binding factor MeCP2 correlates with cellular differentiation in the developing rat brain and in cultured cells, J. Neurobiol, vol.55, pp.86-96, 2003.

G. A. Kaas, C. Zhong, D. E. Eason, D. L. Ross, R. V. Vachhani et al., TET1 Controls CNS 5-Methylcytosine Hydroxylation, vol.79, pp.1086-1093, 2013.

H. Kaddour, E. Coppola, A. Di-nardo, A. Wizenmann, M. Volovitch et al., Extracellular Pax6 regulates tangential Cajal-Retzius cell migration in the developing mouse neocortex, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02380330

M. Kaneko, D. Stellwagen, R. C. Malenka, and M. P. Stryker, Tumor Necrosis Factor-? Mediates One Component of Competitive, Experience-Dependent Plasticity in Developing Visual Cortex, Neuron, vol.58, pp.673-680, 2008.

A. Kirkwood, H. Lee, and M. F. Bear, Co-regulation of long-term potentiation and experience-dependent synaptic plasticity in visual cortex by age and experience, Nature, vol.375, pp.328-331, 1995.

N. Kishi, J. D. Macklis, and D. Hst, Dissecting MECP2 Function in the Central Nervous System, vol.7

C. Koike, A. Nishida, S. Ueno, H. Saito, R. Sanuki et al., Functional Roles of Otx2 Transcription Factor in Postnatal Mouse Retinal Development, Mol. Cell. Biol, vol.27, pp.8318-8329, 2007.

G. Köppe, G. Brückner, K. Brauer, W. Härtig, and V. Bigl, Developmental patterns of proteoglycan-containing extracellular matrix in perineuronal nets and neuropil of the postnatal rat brain, Cell Tissue Res, vol.288, pp.33-41, 1997.

T. Kouzarides, Chromatin Modifications and Their Function, Cell, vol.128, pp.693-705, 2007.

S. Kriaucionis and N. Heintz, The nuclear DNA base, 5-hydroxymethylcytosine is present in brain and enriched in Purkinje neurons, Science, vol.324, pp.929-930, 2009.

K. Krishnan, B. Wang, J. Lu, L. Wang, A. Maffei et al., MeCP2 regulates the timing of critical period plasticity that shapes functional connectivity in primary visual cortex, Proc. Natl. Acad. Sci, vol.112, pp.4782-4791, 2015.

K. Kuo, H. Sheu, Y. Huang, and W. Leung, Expression of Transposon LINE, 1998.

, Is Relatively Human-Specific and Function of the Transcripts May Be Proliferation-Essential

, Biochem. Biophys. Res. Commun, vol.253, pp.566-570

C. D. Laherty, W. Yang, J. Sun, J. R. Davie, E. Seto et al., Histone Deacetylases Associated with the mSin3 Corepressor Mediate Mad Transcriptional Repression, Cell, vol.89, pp.349-356, 1997.

B. Larsen and B. Luna, Adolescence as a neurobiological critical period for the development of higher-order cognition, Neurosci. Biobehav. Rev, vol.94, pp.179-195, 2018.

A. Laughon and M. P. Scott, Sequence of a Drosophila segmentation gene: protein structure homology with DNA-binding proteins, Nature, vol.310, pp.25-31, 1984.

S. Layalle, M. Volovitch, B. Mugat, N. Bonneaud, M. Parmentier et al., Engrailed homeoprotein acts as a signaling molecule in the developing fly, Development, vol.138, pp.2315-2323, 2011.

I. Le-roux, A. Joliot, E. Bloch-gallego, A. Prochiantz, and M. Volovitch, Neurotrophic activity of the Antennapedia homeodomain depends on its specific DNA-binding properties, Proc. Natl. Acad. Sci. U. S. A, vol.90, pp.9120-9124, 1993.

P. T. Leach, S. G. Poplawski, J. W. Kenney, B. Hoffman, D. A. Liebermann et al., Gadd45b knockout mice exhibit selective deficits in hippocampus-dependent long-term memory, Learn. Mem, vol.19, pp.319-324, 2012.

H. H. Lee, C. Bernard, Z. Ye, D. Acampora, A. Simeone et al., Genetic Otx2 mis-localization delays critical period plasticity across brain regions, Mol. Psychiatry, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02380572

A. Lennartsson, E. Arner, M. Fagiolini, A. Saxena, R. Andersson et al., Remodeling of retrotransposon elements during epigenetic induction of adult visual cortical plasticity by HDAC inhibitors, Epigenetics Chromatin, vol.8, 2015.

B. Lesaffre, A. Joliot, A. Prochiantz, and M. Volovitch, Direct non-cell autonomous Pax6 activity regulates eye development in the zebrafish, 2007.

J. M. Levenson, T. L. Roth, F. D. Lubin, C. A. Miller, I. Huang et al., Evidence That DNA (Cytosine-5) Methyltransferase Regulates Synaptic Plasticity in the Hippocampus, J. Biol. Chem, vol.281, pp.15763-15773, 2006.

W. Li, W. Ratnu, V. S. Bredy, and T. W. , On the potential role of active DNA demethylation in establishing epigenetic states associated with neural plasticity and memory, Neurobiol. Learn. Mem, vol.105, pp.125-132, 2013.

W. Li, W. Zhao, Q. Widagdo, J. Baker-andresen, D. Flavell et al., Neocortical Tet3-mediated accumulation of 5-hydroxymethylcytosine promotes rapid behavioral adaptation, Proc. Natl. Acad. Sci, vol.111, pp.7120-7125, 2014.

W. Li, L. Prazak, N. Chatterjee, S. Grüninger, L. Krug et al., , 2013.

, Activation of transposable elements during aging and neuronal decline in Drosophila, Nat. Neurosci, vol.16, pp.529-531

X. Li, P. R. Marshall, L. J. Leighton, E. L. Zajaczkowski, Z. Wang et al., The DNA Repair-Associated Protein Gadd45? Regulates the Temporal Coding of Immediate Early Gene Expression within the Prelimbic Prefrontal Cortex and Is Required for the Consolidation of Associative Fear Memory, J. Neurosci, vol.39, pp.970-983, 2019.

C. Lin, L. Yang, B. Tanasa, K. Hutt, B. Ju et al., Nuclear Receptor-Induced Chromosomal Proximity and DNA Breaks Underlie Specific Translocations in Cancer, Cell, vol.139, pp.1069-1083, 2009.

R. Lister, M. Pelizzola, R. H. Dowen, R. D. Hawkins, G. Hon et al., Human DNA methylomes at base resolution show widespread epigenomic differences, Nature, vol.462, pp.315-322, 2009.

R. Lister, E. A. Mukamel, J. R. Nery, M. Urich, C. A. Puddifoot et al., Global Epigenomic Reconfiguration During Mammalian Brain Development, Science, vol.341, p.1237905, 2013.

L. M. Lombardi, S. A. Baker, and H. Y. Zoghbi, MECP2 disorders: from the clinic to mice and back, J. Clin. Invest, vol.125, pp.2914-2923, 2015.

M. J. Lyst and A. Bird, Rett syndrome: a complex disorder with simple roots, Nat. Rev, 2015.

. Genet, , vol.16, pp.261-275

D. K. Ma, M. Jang, J. U. Guo, Y. Kitabatake, M. Chang et al.,

B. Lu, G. Ming, and H. Song, Neuronal Activity-Induced Gadd45b Promotes Epigenetic DNA Demethylation and Adult Neurogenesis, Science, vol.323, pp.1074-1077, 2009.

R. Madabhushi, F. Gao, A. R. Pfenning, L. Pan, S. Yamakawa et al., Activity-Induced DNA Breaks Govern the Expression of Neuronal Early-Response Genes, Cell, vol.161, pp.1592-1605, 2015.

M. Majdan and C. J. Shatz, Effects of visual experience on activity-dependent gene regulation in cortex, Nat. Neurosci, vol.9, pp.650-659, 2006.

C. D. Malone, J. Brennecke, M. Dus, A. Stark, W. R. Mccombie et al., Specialized piRNA Pathways Act in Germline and Somatic Tissues of the Drosophila Ovary, Cell, vol.137, pp.522-535, 2009.

R. Mangiacasale, C. Pittoggi, I. Sciamanna, A. Careddu, E. Mattei et al., Exposure of normal and transformed cells to nevirapine, a reverse transcriptase inhibitor, reduces cell growth and promotes differentiation, Oncogene, vol.22, pp.2750-2761, 2003.

O. Marín, Interneuron dysfunction in psychiatric disorders, Nat. Rev. Neurosci, 2012.

J. R. Martinez-morales, Otx and eye development, vol.12

K. Martinowich, DNA Methylation-Related Chromatin Remodeling in Activity-Dependent Bdnf Gene Regulation, Science, vol.302, pp.890-893, 2003.

R. Massart, R. Barnea, Y. Dikshtein, M. Suderman, O. Meir et al., Role of DNA Methylation in the Nucleus Accumbens in Incubation of Cocaine Craving, J. Neurosci, vol.35, pp.8042-8058, 2015.

N. Mataga, N. Nagai, and T. K. Hensch, Permissive proteolytic activity for visual cortical plasticity, Proc. Natl. Acad. Sci, vol.99, pp.7717-7721, 2002.

N. Mataga, Y. Mizuguchi, and T. K. Hensch, Experience-Dependent Pruning of Dendritic Spines in Visual Cortex by Tissue Plasminogen Activator, Neuron, vol.44, pp.1031-1041, 2004.

P. H. Maxwell, W. C. Burhans, and M. J. Curcio, Retrotransposition is associated with genome instability during chronological aging, Proc. Natl. Acad. Sci, vol.108, pp.20376-20381, 2011.

B. Mcclintock, The Origin and Behavior of Mutable Loci in Maize, Proc. Natl. Acad. Sci, 1950.

U. S. , , vol.36, pp.344-355

W. Mcginnis, R. L. Garber, J. Wirz, A. Kuroiwa, and W. J. Gehring, A homologous proteincoding sequence in Drosophila homeotic genes and its conservation in other metazoans, Cell, vol.37, pp.403-408, 1984.

P. A. Mcrae, M. M. Rocco, G. Kelly, J. C. Brumberg, and R. T. Matthews, Sensory Deprivation Alters Aggrecan and Perineuronal Net Expression in the Mouse Barrel Cortex, J. Neurosci, vol.27, pp.5405-5413, 2007.

M. Mellen, P. Ayata, S. Dewell, S. Kriaucionis, and N. Heintz, MeCP2 binds to 5hmc enriched within active genes and accessible chromatin in the nervous system, Cell, vol.151, pp.1417-1430, 2012.

C. A. Miller, C. F. Gavin, J. A. White, R. R. Parrish, A. Honasoge et al., Cortical DNA methylation maintains remote memory, Nat. Neurosci, vol.13, pp.664-666, 2010.

P. Mita and J. D. Boeke, How retrotransposons shape genome regulation, Curr. Opin. Genet, 2016.

. Dev, , vol.37, pp.90-100

S. Miyata, Y. Komatsu, Y. Yoshimura, C. Taya, and H. Kitagawa, Persistent cortical plasticity by upregulation of chondroitin 6-sulfation, Nat. Neurosci, vol.15, pp.414-422, 2012.

L. D. Moore, T. Le, F. , and G. , DNA Methylation and Its Basic Function, Neuropsychopharmacology, vol.38, pp.23-38, 2013.

A. Morano, T. Angrisano, G. Russo, R. Landi, A. Pezone et al., Targeted DNA methylation by homology-directed repair in mammalian cells. Transcription reshapes methylation on the repaired gene, Nucleic Acids Res, vol.42, pp.804-821, 2014.

M. J. Morris, M. Mahgoub, E. S. Na, H. Pranav, and L. M. Monteggia, Loss of histone deacetylase 2 improves working memory and accelerates extinction learning, J. Neurosci. Off. J. Soc. Neurosci, vol.33, pp.6401-6411, 2013.

A. R. Muotri, V. T. Chu, M. C. Marchetto, W. Deng, J. V. Moran et al., Somatic mosaicism in neuronal precursor cells mediated by L1 retrotransposition, Nature, vol.435, pp.903-910, 2005.

A. R. Muotri, C. Zhao, M. C. Marchetto, and F. H. Gage, Environmental Influence on L1 Retrotransposons in the Adult Hippocampus, Hippocampus, vol.19, pp.1002-1007, 2009.

A. R. Muotri, M. C. Marchetto, N. G. Coufal, R. Oefner, G. Yeo et al., L1 retrotransposition in neurons is modulated by MeCP2, Nature, vol.468, pp.443-446, 2010.

C. Nagy and G. Turecki, Sensitive Periods in Epigenetics: bringing us closer to complex behavioral phenotypes, Epigenomics, vol.4, pp.445-457, 2012.

L. Nagy, H. Kao, D. Chakravarti, R. J. Lin, C. A. Hassig et al., Nuclear Receptor Repression Mediated by a Complex Containing SMRT, mSin3A, and Histone Deacetylase, Cell, vol.89, pp.373-380, 1997.

X. Nan, H. Ng, C. A. Johnson, C. D. Laherty, B. M. Turner et al., , 1998.

, Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex, Nature, vol.393, pp.386-389

S. Nedelec, I. Foucher, I. Brunet, C. Bouillot, A. Prochiantz et al., , p.2, 2004.

, homeodomain transcription factor interacts with eukaryotic translation initiation factor 4E (eIF4E) in the axons of olfactory sensory neurons, Proc. Natl. Acad. Sci, vol.101, pp.10815-10820

F. Nothias, G. Fishell, and A. R. Altaba, Cooperation of intrinsic and extrinsic signals in the elaboration of regional identity in the posterior cerebral cortex, Curr. Biol, vol.8, pp.459-463, 1998.

J. Noutel, Y. K. Hong, B. Leu, E. Kang, C. et al., Experience-Dependent, 2011.

, Retinogeniculate Synapse Remodeling Is Abnormal in MeCP2-Deficient Mice, Neuron, vol.70, pp.35-42

H. M. O'hagan, H. P. Mohammad, and S. B. Baylin, Double Strand Breaks Can Initiate Gene Silencing and SIRT1-Dependent Onset of DNA Methylation in an Exogenous Promoter CpG Island, 2008.

, PLoS Genet, vol.4, 1000155.

S. Oray, A. Majewska, and M. Sur, Dendritic Spine Dynamics Are Regulated by Monocular Deprivation and Extracellular Matrix Degradation, Neuron, vol.44, pp.1021-1030, 2004.

E. M. Ostertag and H. H. Kazazian, Biology of Mammalian L1 Retrotransposons, Annu. Rev. Genet, vol.35, pp.501-538, 2001.

J. Oswald, S. Engemann, N. Lane, W. Mayer, A. Olek et al.,

J. Walter, Active demethylation of the paternal genome in the mouse zygote, Curr. Biol, vol.10, pp.475-478, 2000.

G. Otting, Y. Q. Qian, M. Billeter, M. Müller, M. Affolter et al., , 1990.

, DNA contacts in the structure of a homeodomain--DNA complex determined by nuclear magnetic resonance spectroscopy in solution, EMBO J, vol.9, pp.3085-3092

A. E. Papale and B. M. Hooks, Circuit Changes in Motor Cortex During Motor Skill Learning, Neuroscience, vol.368, pp.283-297, 2018.

A. Patrizi, P. N. Awad, B. Chattopadhyaya, C. Li, G. Di-cristo et al., , 2019.

, Accelerated Hyper-Maturation of Parvalbumin Circuits in the Absence of MeCP2, Cereb. Cortex

F. M. Pauler, M. A. Sloane, R. Huang, K. Regha, M. V. Koerner et al., H3K27me3 forms BLOCs over silent genes and intergenic regions and specifies a histone banding pattern on a mouse autosomal chromosome, Genome Res, vol.19, pp.221-233, 2008.

J. W. Paylor, B. R. Lins, Q. Greba, N. Moen, R. S. De-moraes et al., Developmental disruption of perineuronal nets in the medial prefrontal cortex after maternal immune activation, Sci. Rep, vol.6, p.37580, 2016.

C. J. Peña, H. G. Kronman, D. M. Walker, H. M. Cates, R. C. Bagot et al., Early life stress confers lifelong stress susceptibility in mice via ventral tegmental area OTX2, Science, vol.356, pp.1185-1188, 2017.

C. Pittoggi, I. Sciamanna, E. Mattei, R. Beraldi, A. M. Lobascio et al., Role of endogenous reverse transcriptase in murine early embryo development, Mol. Reprod. Dev, vol.66, pp.225-236, 2003.

T. Pizzorusso, Reactivation of Ocular Dominance Plasticity in the Adult Visual Cortex, Science, vol.298, pp.1248-1251, 2002.

T. Pizzorusso, P. Medini, S. Landi, S. Baldini, N. Berardi et al., Structural and functional recovery from early monocular deprivation in adult rats, Proc. Natl. Acad. Sci. U. S. A, vol.103, pp.8517-8522, 2006.

A. E. Pohodich and H. Y. Zoghbi, Rett syndrome: disruption of epigenetic control of postnatal neurological functions, Hum. Mol. Genet, vol.24, pp.10-16, 2015.

V. Prabhakar, I. Capila, C. J. Bosques, K. Pojasek, and R. Sasisekharan, , 2005.

. Abc-i-from, Proteus vulgaris: cloning, recombinant expression and active site identification, Biochem. J, vol.386, pp.103-112

E. Putignano, G. Lonetti, L. Cancedda, G. Ratto, M. Costa et al., , 2007.

, Developmental Downregulation of Histone Posttranslational Modifications Regulates Visual Cortical Plasticity, Neuron, vol.53, pp.747-759

Y. Q. Qian, M. Billeter, G. Otting, M. Müller, W. J. Gehring et al., The structure of the Antennapedia homeodomain determined by NMR spectroscopy in solution: comparison with prokaryotic repressors, Nmr In Structural Biology: A Collection of Papers by Kurt Wüthrich, pp.477-484, 1995.

K. Rai, I. J. Huggins, S. R. James, A. R. Karpf, D. A. Jones et al., DNA Demethylation in Zebrafish Involves the Coupling of a Deaminase, a Glycosylase, and Gadd45, Cell, vol.135, pp.1201-1212, 2008.

V. S. Ratnu, W. Wei, and T. W. Bredy, Activation-induced cytidine deaminase regulates activity-dependent BDNF expression in post-mitotic cortical neurons, Eur. J. Neurosci, vol.40, pp.3032-3039, 2014.

H. Rekaik, F. Blaudin-de-thé, J. Fuchs, O. Massiani-beaudoin, A. Prochiantz et al., Engrailed Homeoprotein Protects Mesencephalic Dopaminergic Neurons from Oxidative Stress, Cell Rep, vol.13, pp.242-250, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02156811

S. R. Richardson, S. Morell, and G. J. Faulkner, L1 Retrotransposons and Somatic Mosaicism in the Brain, Annu. Rev. Genet, vol.48, pp.1-27, 2014.

J. Del-rio, L. De-lecea, I. Ferrer, and E. Soriano, The development of parvalbuminimmunoreactivity in the neocortex of the mouse, Dev. Brain Res, vol.81, pp.247-259, 1994.

H. C. Roh, L. T. Tsai, .. Lyubetskaya, A. Tenen, D. Kumari et al., , 2017.

, Simultaneous Transcriptional and Epigenomic Profiling from Specific Cell Types within Heterogeneous Tissues In Vivo, Cell Rep, vol.18, pp.1048-1061

E. D. Roth, T. L. Roth, K. M. Money, S. Sengupta, D. E. Eason et al., DNA methylation regulates neurophysiological spatial representation in memory formation, Neuroepigenetics, vol.2, pp.1-8, 2015.

J. L. Rubenstein, Three hypotheses for developmental defects that may underlie some forms of autism spectrum disorder, Curr. Opin. Neurol, vol.23, pp.118-123, 2010.

A. Rudenko, M. M. Dawlaty, J. Seo, A. W. Cheng, J. Meng et al.,

L. Tsai, Tet1 is critical for neuronal activity-regulated gene expression and memory extinction, Neuron, vol.79, pp.1109-1122, 2013.

A. Sale, M. Vetencourt, J. F. Medini, P. Cenni, M. C. Baroncelli et al., Environmental enrichment in adulthood promotes amblyopia recovery through a reduction of intracortical inhibition, Nat. Neurosci, vol.10, pp.679-681, 2007.

R. C. Samaco, Multiple pathways regulate MeCP2 expression in normal brain development and exhibit defects in autism-spectrum disorders, Hum. Mol. Genet, vol.13, pp.629-639, 2004.

K. E. Savell, N. V. Gallus, R. C. Simon, J. A. Brown, J. S. Revanna et al., Extra-coding RNAs regulate neuronal DNA methylation dynamics, Nat. Commun, vol.7, p.12091, 2016.

. Scheiman, H. Birch, and . Cotter, Randomized Trial of Treatment of Amblyopia in Children Aged 7 to 17 Years, Arch. Ophthalmol, vol.123, p.437, 2005.

B. Schüle, D. D. Armstrong, H. Vogel, A. Oviedo, and U. Francke, Severe congenital encephalopathy caused by MECP2 null mutations in males: central hypoxia and reduced neuronal dendritic structure, Clin. Genet, vol.74, pp.116-126, 2008.

D. C. Schultz, SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zincfinger proteins, Genes Dev, vol.16, pp.919-932, 2002.

T. Sexton and G. Cavalli, The Role of Chromosome Domains in Shaping the Functional Genome, Cell, vol.160, pp.1049-1059, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01153591

J. C. Shepherd, W. Mcginnis, A. E. Carrasco, E. M. De-robertis, and W. J. Gehring, Fly and frog homoeo domains show homologies with yeast mating type regulatory proteins, Nature, vol.310, pp.70-71, 1984.

Y. M. Sigal, H. Bae, L. J. Bogart, T. K. Hensch, and X. Zhuang, Structural maturation of cortical perineuronal nets and their perforating synapses revealed by superresolution imaging, Proc. Natl. Acad. Sci, vol.116, pp.7071-7076, 2019.

D. Silingardi, M. Scali, G. Belluomini, and T. Pizzorusso, Epigenetic treatments of adult rats promote recovery from visual acuity deficits induced by long-term monocular deprivation, 2010.

, J. Neurosci, vol.31, pp.2185-2192

A. Simeone, D. Mallamaci, A. Stornaiuolo, A. D'apice, M. R. Nigro et al., A vertebrate gene related to orthodenticle contains a homeodomain of the bicoid class and demarcates anterior neuroectoderm in the gastrulating mouse embryo, EMBO J, vol.12, pp.2735-2747, 1993.

A. Simeone, D. Acampora, M. Gulisano, A. Stornaiuolo, and E. Boncinelli, Nested expression domains of four homeobox genes in developing rostral brain, Nature, vol.358, pp.687-690, 1992.

M. C. Siomi, K. Sato, D. Pezic, and A. A. Aravin, PIWI-interacting small RNAs: the vanguard of genome defence, Nat. Rev. Mol. Cell Biol, vol.12, pp.246-258, 2011.

A. Sookdeo, C. M. Hepp, M. A. Mcclure, and S. Boissinot, , 2013.

D. G. Southwell, R. C. Froemke, A. Alvarez-buylla, M. P. Stryker, and S. P. Gandhi, Cortical Plasticity Induced by Inhibitory Neuron Transplantation, vol.327, pp.1145-1148, 2010.

J. Spatazza, H. H. Lee, A. A. Di-nardo, L. Tibaldi, A. Joliot et al., Choroid-Plexus-Derived Otx2 Homeoprotein Constrains Adult Cortical Plasticity, Cell Rep, vol.3, pp.1815-1823, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02380061

S. Sugiyama, A. A. Di-nardo, S. Aizawa, I. Matsuo, M. Volovitch et al., Experience-Dependent Transfer of Otx2 Homeoprotein into the Visual Cortex Activates Postnatal Plasticity, Cell, vol.134, pp.508-520, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02380510

F. A. Sultan, J. Wang, J. Tront, D. A. Liebermann, and J. D. Sweatt, Genetic Deletion of gadd45b, a Regulator of Active DNA Demethylation, Enhances Long-Term Memory and Synaptic Plasticity, J. Neurosci, vol.32, pp.17059-17066, 2012.

M. Sur, I. Nagakura, N. Chen, and H. Sugihara, Mechanisms of Plasticity in the Developing and Adult Visual Cortex, Progress in Brain Research, pp.243-254, 2013.

J. Syken, PirB Restricts Ocular-Dominance Plasticity in Visual Cortex, Science, vol.313, pp.1795-1800, 2006.

K. E. Szulwach, X. Li, Y. Li, C. Song, H. Wu et al.,

A. I. Levey, 5-hmC-mediated epigenetic dynamics during postnatal neurodevelopment and aging, Nat. Neurosci, vol.14, pp.1607-1616, 2011.

S. Taha and M. P. Stryker, Rapid Ocular Dominance Plasticity Requires Cortical but Not Geniculate Protein Synthesis, Neuron, vol.34, pp.425-436, 2002.

M. Tahiliani, K. P. Koh, Y. Shen, W. A. Pastor, H. Bandukwala et al., Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in Mammalian DNA by MLL Partner TET1, Science, vol.324, pp.930-935, 2009.

A. E. Takesian and T. K. Hensch, Balancing Plasticity/Stability Across Brain Development, Progress in Brain Research, pp.3-34, 2013.

H. Tan, A. Qurashi, M. Poidevin, D. L. Nelson, H. Li et al., Retrotransposon activation contributes to fragile X premutation rCGG-mediated neurodegeneration, 2012.

, Genet, vol.21, pp.57-65

C. A. Thomas, A. C. Paquola, and A. R. Muotri, LINE-1 Retrotransposition in the Nervous System, Annu. Rev. Cell Dev. Biol, vol.28, pp.555-573, 2012.

P. Tognini, I. Manno, J. Bonaccorsi, M. C. Cenni, A. Sale et al., Environmental Enrichment Promotes Plasticity and Visual Acuity Recovery in Adult Monocular Amblyopic Rats, 2012.

, PLoS ONE, vol.7

P. Tognini, D. Napoli, J. Tola, D. Silingardi, F. Della-ragione et al., Experience-dependent DNA methylation regulates plasticity in the developing visual cortex, Nat. Neurosci, vol.18, pp.956-958, 2015.

I. Topisirovic and K. L. Borden, Homeodomain proteins and eukaryotic translation initiation factor 4E (eIF4E): an unexpected relationship, Histol. Histopathol, vol.20, pp.1275-1284, 2005.

J. T. Trachtenberg and M. P. Stryker, Rapid Anatomical Plasticity of Horizontal Connections in the Developing Visual Cortex, J. Neurosci, vol.21, pp.3476-3482, 2001.

E. Tronick and R. G. Hunter, Waddington, Dynamic Systems, and Epigenetics, Front. Behav. Neurosci, vol.10, 2016.

P. J. Uhlhaas and W. Singer, Abnormal neural oscillations and synchrony in schizophrenia, 2010.

, Nat. Rev. Neurosci, vol.11, pp.100-113

P. J. Uhlhaas and W. Singer, Neuronal Dynamics and Neuropsychiatric Disorders: Toward a Translational Paradigm for Dysfunctional Large-Scale Networks, Neuron, vol.75, pp.963-980, 2012.

A. Valles, A. J. Boender, S. Gijsbers, R. A. Haast, G. J. Martens et al., , 2011.

, Genomewide Analysis of Rat Barrel Cortex Reveals Time-and Layer-Specific mRNA Expression Changes Related to Experience-Dependent Plasticity, J. Neurosci, vol.31, pp.6140-6158

J. F. Vetencourt, A. Sale, A. Viegi, L. Baroncelli, R. De-pasquale et al., The Antidepressant Fluoxetine Restores Plasticity in the Adult Visual Cortex, vol.320, pp.385-388, 2008.

D. W. Volk, T. Matsubara, S. Li, E. J. Sengupta, D. Georgiev et al., Deficits in Transcriptional Regulators of Cortical Parvalbumin Neurons in Schizophrenia, Am. J. Psychiatry, vol.169, pp.1082-1091, 2012.

J. Walter, G. Gehring, and . Otting, Homeodomain-DNA Recognition, 1994.

F. Watt and P. L. Molloy, , 1988.

, Dev, vol.2, pp.1136-1143

H. Weintraub and M. Groudine, Chromosomal subunits in active genes have an altered conformation, Science, vol.193, pp.848-856, 1976.

A. Wizenmann, I. Brunet, J. Lam, L. Sonnier, M. Beurdeley et al., Extracellular Engrailed participates in the topographic guidance of retinal axons in vivo, Neuron, vol.64, pp.355-366, 2009.

C. Wolberger, B. Liu, J. , and A. D. , Crystal Structure of a MATa Homeodomain-Operator Complex Suggests a General Model for Homeodomain-DNA Interactions, p.12

M. Wossidlo, J. Arand, V. Sebastiano, K. Lepikhov, M. Boiani et al.,

J. Walter, Dynamic link of DNA demethylation, DNA strand breaks and repair in mouse zygotes, EMBO J, vol.29, pp.1877-1888, 2010.

K. N. Wright, F. Hollis, F. Duclot, A. M. Dossat, C. E. Strong et al., Methyl Supplementation Attenuates Cocaine-Seeking Behaviors and Cocaine-Induced c-Fos Activation in a DNA Methylation-Dependent Manner, J. Neurosci, vol.35, pp.8948-8958, 2015.

Y. Yagasaki, G. Miyoshi, and M. Miyata, Experience-dependent MeCP2 expression in the excitatory cells of mouse visual thalamus, PLOS ONE, vol.13, 2018.

E. Yang, E. W. Lin, and T. K. Hensch, Critical period for acoustic preference in mice, Proc. Natl. Acad. Sci. U. S. A, vol.109, pp.17213-17220, 2012.

S. Yang, T. Kalkan, C. Morissroe, H. Marks, H. Stunnenberg et al., Otx2 and Oct4 Drive Early Enhancer Activation during Embryonic Stem Cell Transition from Naive Pluripotency, Cell Rep, vol.7, pp.1968-1981, 2014.

Y. Yasuoka, Y. Suzuki, S. Takahashi, H. Someya, N. Sudou et al., Occupancy of Tissue-Specific cis-Regulatory Modules by Otx2 and TLE/Groucho for Embryonic Head Specification, Nat. Commun, vol.5, p.4322, 2014.

J. A. Yoder, C. P. Walsh, and T. H. Bestor, Cytosine methylation and the ecology of intragenomic parasites, p.13, 1997.

Z. Zhou, E. J. Hong, S. Cohen, W. Zhao, H. H. Ho et al.,

E. C. Griffith, Brain-Specific Phosphorylation of MeCP2 Regulates Activity-Dependent Bdnf Transcription, Dendritic Growth, and Spine Maturation, Neuron, vol.52, pp.255-269, 2006.