R. Medzhitov and T. Horng, Transcriptional control of the inflammatory response, Nat Rev Immunol, vol.9, pp.692-703, 2009.

S. T. Smale, Selective transcription in response to an inflammatory stimulus, Cell, vol.140, pp.833-844, 2010.

S. T. Smale, A. Tarakhovsky, and G. Natoli, Chromatin contributions to the regulation of innate immunity, Annu Rev Immunol, vol.32, pp.489-511, 2014.

H. Bierne, M. Hamon, and P. Cossart, Epigenetics and bacterial infections, Cold Spring Harb Perspect Med, vol.2, p.10272, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02651713

A. Pacis, Bacterial infection remodels the DNA methylation landscape of human dendritic cells, Genome Res, vol.25, pp.1801-1811, 2015.

A. K. Marr, Leishmania donovani infection causes distinct epigenetic DNA methylation changes in host macrophages, PLoS Pathog, vol.10, p.1004419, 2014.

D. Bruniquel and R. H. Schwartz, Selective, stable demethylation of the interleukin-2 gene enhances transcription by an active process, Nat Immunol, vol.4, pp.235-240, 2003.

K. Ichiyama, The methylcytosine dioxygenase Tet2 promotes DNA demethylation and activation of cytokine gene expression in T cells, Immunity, vol.42, pp.613-626, 2015.

A. Murayama, A specific CpG site demethylation in the human interleukin 2 gene promoter is an epigenetic memory, EMBO J, vol.25, pp.1081-1092, 2006.

S. H. Sinclair, S. Yegnasubramanian, and J. S. Dumler, Global DNA methylation changes and differential gene expression in Anaplasma phagocytophilum-infected human neutrophils, Clin Epigenetics, vol.7, p.77, 2015.

J. K. Wiencke, The DNA methylation profile of activated human natural killer cells, Epigenetics, vol.11, pp.363-380, 2016.

X. Zhang, DNA methylation dynamics during ex vivo differentiation and maturation of human dendritic cells, Epigenetics Chromatin, vol.7, p.21, 2014.

D. Cizmeci, Mapping epigenetic changes to the host cell genome induced by Burkholderia pseudomallei reveals pathogen-specific and pathogen-generic signatures of infection, Sci Rep, vol.6, p.30861, 2016.

H. A. Schreiber and M. Sandor, The role of dendritic cells in mycobacterium-induced granulomas, Immunol Lett, vol.130, pp.26-31, 2010.

X. Jiao, Dendritic cells are host cells for mycobacteria in vivo that trigger innate and acquired immunity, J Immunol, vol.168, pp.1294-1301, 2002.
URL : https://hal.archives-ouvertes.fr/hal-02682621

T. Tian, J. Woodworth, M. Sköld, and S. M. Behar, In vivo depletion of CD11c+ cells delays the CD4+ T cell response to Mycobacterium tuberculosis and exacerbates the outcome of infection, J Immunol, vol.175, pp.3268-3272, 2005.

A. J. Wolf, Mycobacterium tuberculosis infects dendritic cells with high frequency and impairs their function in vivo, J Immunol, vol.179, pp.2509-2519, 2007.

S. Hambleton, IRF8 mutations and human dendritic-cell immunodeficiency, N Engl J Med, vol.365, pp.127-138, 2011.

A. Pacis, SeqCap Epi data. NCBI Gene Expression Omnibus, 2018.

N. D. Heintzman, Histone modifications at human enhancers reflect global cell-type-specific gene expression, Nature, vol.459, pp.108-112, 2009.

H. Feng, K. N. Conneely, and H. Wu, A Bayesian hierarchical model to detect differentially methylated loci from single nucleotide resolution sequencing data, Nucleic Acids Res, vol.42, p.69, 2014.

X. Wu and Y. Zhang, TET-mediated active DNA demethylation: Mechanism, function and beyond, Nat Rev Genet, vol.18, pp.517-534, 2017.

M. Klug, S. Schmidhofer, C. Gebhard, R. Andreesen, and M. Rehli, -Hydroxymethylcytosine is an essential intermediate of active DNA demethylation processes in primary human monocytes, Genome Biol, vol.14, issue.5, p.46, 2013.

D. Álvarez-errico, R. Vento-tormo, M. Sieweke, and E. Ballestar, Epigenetic control of myeloid cell differentiation, identity and function, Nat Rev Immunol, vol.15, pp.7-17, 2015.

E. A. Mahé, Cytosine modifications modulate the chromatin architecture of transcriptional enhancers, Genome Res, vol.27, pp.947-958, 2017.

M. Yu, Base-resolution analysis of 5-hydroxymethylcytosine in the mammalian genome, Cell, vol.149, pp.1368-1380, 2012.

E. Calo and J. Wysocka, Modification of enhancer chromatin: What, how, and why?, Mol Cell, vol.49, pp.825-837, 2013.

A. Pacis, RNA-seq data. NCBI Gene Expression Omnibus, 2018.

A. Pacis, ChIP-seq data. NCBI Gene Expression Omnibus, 2018.

Y. Nedelec, Genetic ancestry and natural selection drive population differences in immune responses to pathogens, Cell, vol.167, pp.657-669, 2016.

A. Pacis, ATAC-seq data. NCBI Gene Expression Omnibus, 2018.

J. D. Buenrostro, P. G. Giresi, L. C. Zaba, H. Y. Chang, and W. J. Greenleaf, Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNAbinding proteins and nucleosome position, Nat Methods, vol.10, pp.1213-1218, 2013.

Y. Yin, Impact of cytosine methylation on DNA binding specificities of human transcription factors, Science, vol.356, p.2239, 2017.

S. Domcke, Competition between DNA methylation and transcription factors determines binding of NRF1, Nature, vol.528, pp.575-579, 2015.

H. Zhu, G. Wang, and J. Qian, Transcription factors as readers and effectors of DNA methylation, Nat Rev Genet, vol.17, pp.551-565, 2016.

R. Vento-tormo, IL-4 orchestrates STAT6-mediated DNA demethylation leading to dendritic cell differentiation, Genome Biol, vol.17, p.4, 2016.

A. A. Sérandour, Dynamic hydroxymethylation of deoxyribonucleic acid marks differentiation-associated enhancers, Nucleic Acids Res, vol.40, pp.8255-8265, 2012.

G. C. Hon, 5mC oxidation by Tet2 modulates enhancer activity and timing of transcriptome reprogramming during differentiation, Mol Cell, vol.56, pp.286-297, 2014.

M. P. Creyghton, Histone H3K27ac separates active from poised enhancers and predicts developmental state, Proc Natl Acad Sci, vol.107, pp.21931-21936, 2010.

M. U. Kaikkonen, Remodeling of the enhancer landscape during macrophage activation is coupled to enhancer transcription, Mol Cell, vol.51, pp.310-325, 2013.

D. Bhatt and S. Ghosh, Regulation of the NF-?B-mediated transcription of inflammatory genes, Front Immunol, vol.5, p.71, 2014.

Y. W. Zhang, Acetylation enhances TET2 function in protecting against abnormal DNA methylation during oxidative stress, Mol Cell, vol.65, pp.323-335, 2017.

E. E. Ford, Frequent lack of repressive capacity of promoter DNA methylation identified through genome-wide epigenomic manipulation, 2017.

M. B. Stadler, DNA-binding factors shape the mouse methylome at distal regulatory regions, Nature, vol.480, p.550, 2011.

L. Han, I. G. Lin, and C. L. Hsieh, Protein binding protects sites on stable episomes and in the chromosome from de novo methylation, Mol Cell Biol, vol.21, pp.3416-3424, 2001.

C. Kress, H. Thomassin, and T. Grange, Active cytosine demethylation triggered by a nuclear receptor involves DNA strand breaks, Proc Natl Acad Sci, vol.103, pp.11112-11117, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00093585

N. Sato, M. Kondo, and K. Arai, The orphan nuclear receptor GCNF recruits DNA methyltransferase for Oct-3/4 silencing, Biochem Biophys Res Commun, vol.344, pp.845-851, 2006.

D. Schübeler, Function and information content of DNA methylation, Nature, vol.517, pp.321-326, 2015.

R. L. De-la, PU.1 target genes undergo Tet2-coupled demethylation and DNMT3b-mediated methylation in monocyte-to-osteoclast differentiation, Genome Biol, vol.14, p.99, 2013.

R. Rickels, Histone H3K4 monomethylation catalyzed by Trr and mammalian COMPASS-like proteins at enhancers is dispensable for development and viability, Nat Genet, vol.49, pp.1647-1653, 2017.

K. M. Dorighi, Mll3 and Mll4 facilitate enhancer RNA synthesis and transcription from promoters independently of H3K4 monomethylation, Mol Cell, vol.66, pp.568-576, 2017.

A. Vandenbon, Y. Kumagai, M. Lin, Y. Suzuki, and K. Nakai, Waves of chromatin modifications in mouse dendritic cells in response to LPS stimulation, 2017.

J. Quintin, S. C. Cheng, J. W. Van-der-meer, and M. G. Netea, Innate immune memory: Towards a better understanding of host defense mechanisms, Curr Opin Immunol, vol.29, pp.1-7, 2014.

S. Saeed, Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity, Science, vol.345, p.1251086, 2014.

R. Ostuni, Latent enhancers activated by stimulation in differentiated cells, Cell, vol.152, pp.157-171, 2013.

E. Kaufmann, BCG educates hematopoietic stem cells to generate protective innate immunity against tuberculosis, Cell, vol.172, pp.176-190, 2018.

F. Krueger and S. R. Andrews, Bismark: A flexible aligner and methylation caller for bisulfite-seq applications, Bioinformatics, vol.27, pp.1571-1572, 2011.

K. D. Hansen, B. Langmead, and R. A. Irizarry, BSmooth: From whole genome bisulfite sequencing reads to differentially methylated regions, Genome Biol, vol.13, p.83, 2012.

G. Bindea, ClueGO: A cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks, Bioinformatics, vol.25, pp.1091-1093, 2009.

A. Khan, JASPAR 2018: Update of the open-access database of transcription factor binding profiles and its web framework, Nucleic Acids Res, vol.46, p.1284, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01646126