Y. A. Medvedeva, A. Lennartsson, R. Ehsani, I. V. Kulakovskiy, I. E. Vorontsov et al., EpiFactors: a comprehensive database of human epigenetic factors and complexes, Database (Oxford), p.4494013, 2015.

T. Itoh, L. Fairall, F. W. Muskett, C. P. Milano, P. J. Watson et al., Structural and functional characterization of a cell cycle associated HDAC1/2 complex reveals the structural basis for complex assembly and nucleosome targeting, Nucleic Acids Res, vol.43, issue.4, p.4344507, 2015.

N. Plaster, C. Sonntag, T. F. Schilling, and M. Hammerschmidt, REREa/Atrophin-2 interacts with histone deacetylase and Fgf8 signaling to regulate multiple processes of zebrafish development. Developmental dynamics: an official publication of the American Association of Anatomists, vol.236, pp.1891-904, 2007.

H. Bierne, T. N. Tham, E. Batsche, A. Dumay, M. Leguillou et al., Human BAHD1 promotes heterochromatic gene silencing, Proc Natl Acad Sci, vol.106, issue.33, p.2728979, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00411478

A. Lebreton, G. Lakisic, V. Job, L. Fritsch, T. N. Tham et al., A bacterial protein targets the BAHD1 chromatin complex to stimulate type III interferon response, Science, vol.331, issue.6022, pp.1319-1340, 2011.
URL : https://hal.archives-ouvertes.fr/cea-00819299

G. Lakisic, A. Lebreton, R. Pourpre, O. Wendling, E. Libertini et al., Role of the BAHD1 Chromatin-Repressive Complex in Placental Development and Regulation of Steroid Metabolism, PLoS Genet, vol.12, issue.3, p.1005898, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01350966

P. Central and P. , , p.4777444

N. Yang and R. M. Xu, Structure and function of the BAH domain in chromatin biology, Crit Rev Biochem Mol Biol, vol.48, issue.3, pp.211-232, 2013.

L. L. Gillespie and G. D. Paterno, MIER1 (mesoderm induction early response 1 homolog (Xenopus laevis))

, Atlas Genet Cytogenet Oncol Haematol, vol.16, issue.2, pp.127-157, 2012.

Z. Ding, L. L. Gillespie, F. C. Mercer, and G. D. Paterno, The SANT domain of human MI-ER1 interacts with Sp1 to interfere with GC box recognition and repress transcription from its own promoter, J Biol Chem, vol.279, issue.27, pp.28009-28025, 2004.

R. Derwish, G. D. Paterno, and L. L. Gillespie, Differential HDAC1 and 2 Recruitment by Members of the MIER Family, PLoS One, vol.12, issue.1, p.5207708, 2017.

M. Escamilla-del-arenal, S. T. Da-rocha, C. G. Spruijt, O. Masui, O. Renaud et al., Cdyl, a new partner of the inactive X chromosome and potential reader of H3K27me3 and H3K9me2, Mol Cell Biol, vol.33, issue.24, p.3889549, 2013.

M. Weimann, A. Grossmann, J. Woodsmith, Z. Ozkan, P. Birth et al., A Y2H-seq approach defines the human protein methyltransferase interactome, Nature methods, vol.10, issue.4, pp.339-381, 2013.

E. Libertini, A. Lebreton, G. Lakisic, M. A. Dillies, S. Beck et al., Overexpression of the Heterochromatinization Factor BAHD1 in HEK293 Cells Differentially Reshapes the DNA Methylome on Autosomes and X Chromosome, Front Genet, vol.6, p.4664705, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01350961

D. Zhao, X. Zhang, H. Guan, X. Xiong, X. Shi et al., The BAH domain of BAHD1 is a histone H3K27me3 reader, Protein Cell, vol.7, issue.3, pp.222-228, 2016.

K. Krause and F. Turck, Plant H3K27me3 has finally found its readers, Nat Genet, vol.50, issue.9, pp.1206-1214, 2018.

T. Nagase, K. Ishikawa, M. Suyama, R. Kikuno, M. Hirosawa et al., Prediction of the coding sequences of unidentified human genes. XIII. The complete sequences of 100 new cDNA clones from brain which code for large proteins in vitro, DNA Res, vol.6, issue.1, pp.63-70, 1999.

A. Lebreton, V. Job, M. Ragon, L. Monnier, A. Dessen et al., Structural basis for the inhibition of the chromatin repressor BAHD1 by the bacterial nucleomodulin LntA, MBio, vol.5, issue.1, p.3903274, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01109386

H. Zhu, X. Wan, J. Li, L. Han, X. Bo et al., Computational Prediction and Validation of BAHD1 as a Novel Molecule for Ulcerative Colitis, Sci Rep, vol.5, p.4505333, 2015.

W. Huang-da, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nat Protoc, vol.4, issue.1, pp.44-57, 2009.

M. Uhlen, L. Fagerberg, B. M. Hallstrom, C. Lindskog, P. Oksvold et al., Proteomics. Tissuebased map of the human proteome, Science, vol.347, issue.6220, p.1260419, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01479709

F. Yue, Y. Cheng, A. Breschi, J. Vierstra, W. Wu et al., A comparative encyclopedia of DNA elements in the mouse genome, Nature, vol.515, issue.7527, p.4266106, 2014.

E. S. Lein, M. J. Hawrylycz, N. Ao, M. Ayres, A. Bensinger et al., Genome-wide atlas of gene expression in the adult mouse brain, Nature, vol.445, issue.7124, pp.168-76, 2007.

M. J. Hawrylycz, E. S. Lein, A. L. Guillozet-bongaarts, E. H. Shen, L. Ng et al., An anatomically comprehensive atlas of the adult human brain transcriptome, Nature, vol.489, issue.7416, p.4243026, 2012.

A. Saunders, E. Z. Macosko, A. Wysoker, M. Goldman, F. M. Krienen et al., Molecular Diversity and Specializations among the Cells of the Adult Mouse Brain, Cell, vol.174, issue.4, p.6447408, 2018.

A. D. Rouillard, G. W. Gundersen, N. F. Fernandez, Z. Wang, C. D. Monteiro et al., The harmonizome: a collection of processed datasets gathered to serve and mine knowledge about genes and proteins, Database (Oxford), 2016.

P. Central and P. , , p.4930834

I. Rusinova, S. Forster, S. Yu, A. Kannan, M. Masse et al., Interferome v2.0: an updated database of annotated interferon-regulated genes, Nucleic Acids Res, vol.41, p.3531205, 2013.

I. A. Qureshi, S. Gokhan, and M. F. Mehler, REST and CoREST are transcriptional and epigenetic regulators of seminal neural fate decisions, Cell Cycle, vol.9, issue.22, p.3048046, 2010.

J. Jaworska, M. Ziemka-nalecz, and T. Zalewska, Histone deacetylases 1 and 2 are required for brain development, Int J Dev Biol, vol.59, issue.4-6, pp.171-178, 2015.

L. Prut and C. Belzung, The open field as a paradigm to measure the effects of drugs on anxiety-like behaviors: a review, Eur J Pharmacol, vol.463, issue.1-3, pp.3-33, 2003.

K. R. Lezak, G. Missig, and C. Wa, Behavioral methods to study anxiety in rodents, Dialogues Clin Neurosci, vol.19, issue.2, p.5573562, 2017.

A. K. Kraeuter, P. C. Guest, and Z. Sarnyai, The Open Field Test for Measuring Locomotor Activity and Anxiety-Like Behavior, Methods Mol Biol, vol.1916, pp.99-103, 2019.

H. Anisman, S. Hayley, O. Kelly, T. Borowski, and Z. Merali, Psychogenic, neurogenic, and systemic stressor effects on plasma corticosterone and behavior: mouse strain-dependent outcomes, Behav Neurosci, vol.115, issue.2, pp.443-54, 2001.

A. Ennaceur, S. Michalikova, A. Bradford, and S. Ahmed, Detailed analysis of the behavior of Lister and Wistar rats in anxiety, object recognition and object location tasks, Behav Brain Res, vol.159, issue.2, pp.247-66, 2005.

P. Simon, R. Dupuis, and J. Costentin, Thigmotaxis as an index of anxiety in mice. Influence of dopaminergic transmissions, Behav Brain Res, vol.61, issue.1, pp.90008-90014, 1994.

S. B. Powell, M. Weber, and M. A. Geyer, Genetic models of sensorimotor gating: relevance to neuropsychiatric disorders, Curr Top Behav Neurosci, vol.12, p.3357439, 2012.

C. Ioannidou, G. Marsicano, and A. Busquets-garcia, Assessing Prepulse Inhibition of Startle in Mice Bio-protocol, vol.8, p.2789, 2018.

M. Pienkowski, V. Kucharczyk, M. Mlynek, M. Szczaluba, K. Rydzanicz et al., Mapping of breakpoints in balanced chromosomal translocations by shallow whole-genome sequencing points to EFNA5, BAHD1 and PPP2R5E as novel candidates for genes causing human Mendelian disorders, J Med Genet, vol.56, issue.2, pp.104-116, 2019.

D. M. Bannerman, R. Sprengel, D. J. Sanderson, S. B. Mchugh, J. N. Rawlins et al., Hippocampal synaptic plasticity, spatial memory and anxiety, Nat Rev Neurosci, vol.15, issue.3, pp.181-92, 2014.

E. A. Phelps, Human emotion and memory: interactions of the amygdala and hippocampal complex, Curr Opin Neurobiol, vol.14, issue.2, pp.198-202, 2004.

M. S. Bienkowski, I. Bowman, M. Y. Song, L. Gou, T. Ard et al., Integration of gene expression and brain-wide connectivity reveals the multiscale organization of mouse hippocampal networks, Nat Neurosci, vol.21, issue.11, p.6398347, 2018.

J. W. Vogel, L. Joie, R. Grothe, M. J. Diaz-papkovich, A. Doyle et al., A molecular gradient along the longitudinal axis of the human hippocampus informs large-scale behavioral systems, Nat Commun, vol.11, issue.1, p.960, 2020.

P. Central and P. , , p.7031290

S. A. Small, S. A. Schobel, R. B. Buxton, M. P. Witter, and C. A. Barnes, A pathophysiological framework of hippocampal dysfunction in ageing and disease, Nat Rev Neurosci, vol.12, issue.10, p.3312472, 2011.

R. D. Rubin, P. D. Watson, M. C. Duff, and N. J. Cohen, The role of the hippocampus in flexible cognition and social behavior, Front Hum Neurosci, vol.8, p.4179699, 2014.

A. Montagrin, C. Saiote, and D. Schiller, The social hippocampus, Hippocampus, vol.28, issue.9, pp.672-681, 2018.

T. Bast, The hippocampal learning-behavior translation and the functional significance of hippocampal dysfunction in schizophrenia, Curr Opin Neurobiol, vol.21, issue.3, pp.492-501, 2011.

S. Nakahara, M. Matsumoto, and T. Van-erp, Hippocampal subregion abnormalities in schizophrenia: A systematic review of structural and physiological imaging studies, Neuropsychopharmacol Rep, vol.38, issue.4, pp.156-66, 2018.

A. Tavitian, W. Song, and H. M. Schipper, Dentate Gyrus Immaturity in Schizophrenia, Neuroscientist, p.1073858418824072, 2019.

N. R. Swerdlow and M. A. Geyer, Using an animal model of deficient sensorimotor gating to study the pathophysiology and new treatments of schizophrenia, Schizophr Bull, vol.24, issue.2, pp.285-301, 1998.

N. R. Swerdlow, M. Weber, Y. Qu, G. A. Light, and D. L. Braff, Realistic expectations of prepulse inhibition in translational models for schizophrenia research, Psychopharmacology (Berl), vol.199, issue.3, p.2771731, 2008.

N. R. Swerdlow, C. H. Benbow, S. Zisook, M. A. Geyer, and D. L. Braff, A preliminary assessment of sensorimotor gating in patients with obsessive compulsive disorder, Biol Psychiatry, vol.33, issue.4, pp.90300-90303, 1993.

W. Perry, A. Minassian, D. Feifel, and D. L. Braff, Sensorimotor gating deficits in bipolar disorder patients with acute psychotic mania, Biol Psychiatry, vol.50, issue.6, pp.1184-1186, 2001.

S. Ludewig, K. Ludewig, M. A. Geyer, D. Hell, and F. X. Vollenweider, Prepulse inhibition deficits in patients with panic disorder, Depress Anxiety, vol.15, issue.2, pp.55-60, 2002.

W. Perry, A. Minassian, B. Lopez, L. Maron, and A. Lincoln, Sensorimotor gating deficits in adults with autism, Biol Psychiatry, vol.61, issue.4, pp.482-488, 2007.

R. Jaenisch and A. Bird, Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals, Nat Genet, vol.33, pp.245-54, 2003.

S. E. Sillivan, T. Vaissiere, and C. A. Miller, Neuroepigenetic Regulation of Pathogenic Memories, Neuroepigenetics, vol.1, p.4310006, 2015.

M. J. Kwon, S. Kim, M. H. Han, and S. B. Lee, Epigenetic Changes in Neurodegenerative Diseases, Mol Cells, vol.39, issue.11, p.5125933, 2016.

M. A. Christopher, S. M. Kyle, and D. J. Katz, Neuroepigenetic mechanisms in disease, Epigenetics Chromatin, vol.10, issue.1, p.5644115, 2017.

C. J. Pena and E. J. Nestler, Progress in Epigenetics of Depression, Prog Mol Biol Transl Sci, vol.157, p.6047749, 2018.

, IMC

S. Bahari-javan, M. A. Kerimoglu, C. Wittnam, J. Held, T. Bahr et al., HDAC1 regulates fear extinction in mice, J Neurosci, vol.32, issue.15, pp.5062-73, 2012.

M. Jakovcevski, R. Bharadwaj, J. Straubhaar, G. Gao, D. P. Gavin et al., Prefrontal cortical dysfunction after overexpression of histone deacetylase 1, Biol Psychiatry, vol.74, issue.9, p.3797203, 2013.

S. Bahari-javan, H. Varbanov, R. Halder, B. E. Kaurani, L. Burkhardt et al., HDAC1 links early life stress to schizophrenia-like phenotypes, Proc Natl Acad Sci, vol.114, issue.23, p.5468618, 2017.

J. S. Guan, S. J. Haggarty, E. Giacometti, J. H. Dannenberg, N. Joseph et al., HDAC2 negatively regulates memory formation and synaptic plasticity, Nature, vol.459, issue.7243, p.3498958, 2009.

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, vol.33, issue.15, p.3773986, 2013.

M. Mahgoub, M. Adachi, K. Suzuki, X. Liu, E. T. Kavalali et al., MeCP2 and histone deacetylases 1 and 2 in dorsal striatum collectively suppress repetitive behaviors, Nat Neurosci, vol.19, issue.11, p.5083208, 2016.

A. Schaefer, S. C. Sampath, A. Intrator, A. Min, T. S. Gertler et al., Control of cognition and adaptive behavior by the GLP/G9a epigenetic suppressor complex, Neuron, vol.64, issue.5, p.2814156, 2009.

G. Iacono, A. Dubos, H. Meziane, M. Benevento, E. Habibi et al., Increased H3K9 methylation and impaired expression of Protocadherins are associated with the cognitive dysfunctions of the Kleefstra syndrome, Nucleic Acids Res, vol.46, issue.10, p.6007260, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02187058

Y. Jiang, M. Jakovcevski, R. Bharadwaj, C. Connor, F. A. Schroeder et al., Setdb1 histone methyltransferase regulates mood-related behaviors and expression of the NMDA receptor subunit NR2B, J Neurosci, vol.30, issue.21, p.2893142, 2010.

Y. Jiang, Y. E. Loh, P. Rajarajan, T. Hirayama, W. Liao et al., The methyltransferase SETDB1 regulates a large neuron-specific topological chromatin domain, Nat Genet, vol.49, issue.8, p.5560095, 2017.

X. Zhao, T. Ueba, B. R. Christie, B. Barkho, M. J. Mcconnell et al., Mice lacking methyl-CpG binding protein 1 have deficits in adult neurogenesis and hippocampal function, Proc Natl Acad Sci U S A, vol.100, issue.11, pp.6777-82, 2003.

P. Central and P. , , p.164523

A. M. Allan, X. Liang, Y. Luo, C. Pak, X. Li et al., The loss of methyl-CpG binding protein 1 leads to autism-like behavioral deficits, Hum Mol Genet, vol.17, issue.13, pp.2047-57, 2008.

Y. Liu, M. Li, M. Fan, Y. Song, H. Yu et al., Chromodomain Y-like Protein-Mediated Histone Crotonylation Regulates Stress-Induced Depressive Behaviors, Biol Psychiatry, vol.11, p.25, 2018.

J. Jakobsson, M. I. Cordero, R. Bisaz, A. C. Groner, V. Busskamp et al., KAP1-mediated epigenetic repression in the forebrain modulates behavioral vulnerability to stress, Neuron, vol.60, issue.5, pp.818-849, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00350884

Y. Qin and P. A. Wade, Crosstalk between the microbiome and epigenome: messages from bugs, J Biochem, vol.163, issue.2, p.5892391, 2018.

A. Schlachetzki, J. Glass, C. K. Mazmanian, and S. K. , Microbiome-microglia connections via the gut-brain axis, J Exp Med, vol.216, issue.1, p.6314531, 2019.

H. Bierne, Cross Talk Between Bacteria and the Host Epigenetic Machinery, Epigenetics of Infectious Diseases, pp.113-58, 2017.

R. M. Mcmanus and M. T. Heneka, Role of neuroinflammation in neurodegeneration: new insights, Alzheimers Res Ther, vol.9, issue.1, p.28259169, 2017.

P. Central and P. , , p.5336609

D. Cossu, K. Yokoyama, and N. Hattori, Bacteria-Host Interactions in Multiple Sclerosis, Front Microbiol, vol.9, p.6288311, 2018.