J. R. Forbes and P. Gros, Divalent-metal transport by NRAMP proteins at the interface of host?pathogen interactions, Trends in Microbiology, vol.9, issue.8, pp.397-403, 2001.
DOI : 10.1016/S0966-842X(01)02098-4

A. Cesar-razquin, B. Snijder, T. Frappier-brinton, R. Isserlin, G. Gyimesi et al., A Call for Systematic Research on Solute Carriers, CrossRef] [PubMed] 3. Cellier, M.F. Nutritional immunity: Homology modeling of Nramp metal import, pp.478-487, 2012.
DOI : 10.1016/j.cell.2015.07.022

S. Soe-lin, S. S. Apte, M. R. Mikhael, L. K. Kayembe, G. Nie et al., Both Nramp1 and DMT1 are necessary for efficient macrophage iron recycling, Experimental Hematology, vol.38, issue.8, pp.609-617, 2010.
DOI : 10.1016/j.exphem.2010.04.003

B. Peracino, C. Wagner, A. Balest, A. Balbo, B. Pergolizzi et al., Function and mechanism of action of Dictyostelium Nramp1 (Slc11a1) in bacterial infection [CrossRef] [PubMed] 7. Peracino, B.; Buracco, S.; Bozzaro, S. The Nramp (Slc11) proteins regulate development, resistance to pathogenic bacteria and iron homeostasis in Dictyostelium discoideum Dictyostelium Nramp1, which is structurally and functionally similar to mammalian DMT1 transporter, mediates phagosomal iron efflux An integrated encyclopedia of DNA elements in the human genome, Traffic J. Cell Sci. J. Cell Sci Nature, vol.7, issue.489, pp.22-38, 2006.

R. E. Thurman, E. Rynes, R. Humbert, J. Vierstra, M. T. Maurano et al., The accessible chromatin landscape of the human genome, Nature, vol.27, issue.7414, pp.75-82, 2012.
DOI : 10.1038/nature11232

M. F. Cellier, Cell-Type Specific Determinants of NRAMP1 Expression in Professional Phagocytes, Biology, vol.337, issue.1, pp.233-283, 2013.
DOI : 10.1016/j.cell.2011.12.006

URL : https://hal.archives-ouvertes.fr/pasteur-01130396

E. Richer, C. G. Campion, B. Dabbas, J. H. White, and M. F. Cellier, Transcription factors Sp1 and C/EBP regulate NRAMP1 gene expression, FEBS Journal, vol.31, issue.20, pp.5074-5089, 2008.
DOI : 10.1111/j.1742-4658.2008.06640.x

T. H. Pham, C. Benner, M. Lichtinger, L. Schwarzfischer, Y. Hu et al., Dynamic epigenetic enhancer signatures reveal key transcription factors associated with monocytic differentiation states, Blood, vol.119, issue.24, pp.161-171, 2012.
DOI : 10.1182/blood-2012-01-402453

A. Kundaje, W. Meuleman, J. Ernst, M. Bilenky, A. Yen et al., Integrative analysis of 111 reference human epigenomes, Nature, vol.32, issue.7539, pp.317-330, 2015.
DOI : 10.1038/nature14248

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4530010

S. Saeed, J. Quintin, H. H. Kerstens, N. A. Rao, A. Aghajanirefah et al., Epigenetic programming of monocyte-to-macrophage differentiation and trained innate immunity, Science, vol.5, issue.12, p.1251086, 2014.
DOI : 10.1038/nri1733

R. Andersson, C. Gebhard, I. Miguel-escalada, I. Hoof, J. Bornholdt et al., An atlas of active enhancers across human cell types and tissues, Nature, vol.45, issue.7493, pp.455-461, 2014.
DOI : 10.1093/bioinformatics/btq033

A. R. Forrest, H. Kawaji, M. Rehli, J. K. Baillie, M. J. De-hoon et al., A promoter-level mammalian expression atlas, Nature, vol.22, issue.7493, pp.462-470, 2014.
DOI : 10.1038/nature13182

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4529748

D. Vernimmen and W. A. Bickmore, The Hierarchy of Transcriptional Activation: From Enhancer to Promoter, Trends in Genetics, vol.31, issue.12, pp.696-708
DOI : 10.1016/j.tig.2015.10.004

S. Pott and J. Lieb, What are super-enhancers?, Nature Genetics, vol.8, issue.1, pp.8-12
DOI : 10.1126/science.1247005

S. Witte, J. J. Shea, and G. Vahedi, Super-enhancers: Asset management in immune cell genomes, Trends in Immunology, vol.36, issue.9, pp.519-526, 2015.
DOI : 10.1016/j.it.2015.07.005

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4679291

A. Soufi, M. F. Garcia, A. Jaroszewicz, N. Osman, M. Pellegrini et al., Pioneer Transcription Factors Target Partial DNA Motifs on Nucleosomes to Initiate Reprogramming, Cell, vol.161, issue.3, pp.555-568, 2015.
DOI : 10.1016/j.cell.2015.03.017

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4409934

C. E. Romanoski, V. M. Link, S. Heinz, and C. K. Glass, Exploiting genomics and natural genetic variation to decode macrophage enhancers, Trends in Immunology, vol.36, issue.9, pp.507-518, 2015.
DOI : 10.1016/j.it.2015.07.006

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4548828

K. S. Zaret and S. E. Mango, Pioneer transcription factors, chromatin dynamics, and cell fate control, Current Opinion in Genetics & Development, vol.37, pp.76-81, 2016.
DOI : 10.1016/j.gde.2015.12.003

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4914445

D. Schubeler, Function and information content of DNA methylation, Nature, vol.62, issue.7534, pp.321-326, 2015.
DOI : 10.1038/ng.2442

T. K. Kim, M. Hemberg, J. M. Gray, A. M. Costa, D. M. Bear et al., Widespread transcription at neuronal activity-regulated enhancers, Nature, vol.57, issue.7295, pp.182-187, 2010.
DOI : 10.1186/gb-2003-4-5-p3

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3020079

D. Wang, I. Garcia-bassets, C. Benner, W. Li, X. Su et al., Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA, Nature, vol.18, issue.7351, pp.390-394, 2011.
DOI : 10.1038/nature10006

C. A. Melo, J. Drost, P. J. Wijchers, H. Van-de-werken, E. De-wit et al., eRNAs Are Required for p53-Dependent Enhancer Activity and Gene Transcription, Molecular Cell, vol.49, issue.3, pp.524-535, 2013.
DOI : 10.1016/j.molcel.2012.11.021

URL : http://doi.org/10.1016/j.molcel.2012.11.021

A. L. Jiao and F. J. Slack, RNA-mediated gene activation, Epigenetics, vol.486, issue.1, pp.27-36
DOI : 10.1016/j.cell.2011.07.014

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3928182

M. T. Lam, W. Li, M. G. Rosenfeld, and C. K. Glass, Enhancer RNAs and regulated transcriptional programs, Trends in Biochemical Sciences, vol.39, issue.4, pp.170-182
DOI : 10.1016/j.tibs.2014.02.007

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4266492

Y. Murakawa, M. Yoshihara, H. Kawaji, M. Nishikawa, H. Zayed et al., Enhanced Identification of Transcriptional Enhancers Provides Mechanistic Insights into Diseases, Trends in Genetics, vol.32, issue.2, pp.76-88, 2016.
DOI : 10.1016/j.tig.2015.11.004

K. Mousavi, H. Zare, M. Koulnis, and V. Sartorelli, The emerging roles of eRNAs in transcriptional regulatory networks, RNA Biology, vol.13, issue.2, pp.106-110
DOI : 10.4161/trns.2.5.17712

M. U. Kaikkonen, N. J. Spann, S. Heinz, C. E. Romanoski, K. A. Allison et al., Remodeling of the Enhancer Landscape during Macrophage Activation Is Coupled to Enhancer Transcription, Molecular Cell, vol.51, issue.3, pp.310-325, 2013.
DOI : 10.1016/j.molcel.2013.07.010

S. Heinz, C. E. Romanoski, C. Benner, and C. K. Glass, The selection and function of cell type-specific enhancers, Nature Reviews Molecular Cell Biology, vol.14, issue.3, pp.144-154, 2015.
DOI : 10.1016/j.cell.2014.05.010

A. A. Sigova, B. J. Abraham, X. Ji, B. Molinie, N. M. Hannett et al., Transcription factor trapping by RNA in gene regulatory elements, Science, vol.141, issue.1, pp.978-981, 2015.
DOI : 10.1242/dev.103341

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4720525

H. Wu, A. S. Nord, J. A. Akiyama, M. Shoukry, V. Afzal et al., Tissue-Specific RNA Expression Marks Distant-Acting Developmental Enhancers, PLoS Genetics, vol.302, issue.9, p.1004610, 2014.
DOI : 10.1371/journal.pgen.1004610.s012

URL : http://doi.org/10.1371/journal.pgen.1004610

N. E. Iiott, J. A. Heward, B. Roux, E. Tsitsiou, P. S. Fenwick et al., et al. Long non-coding RNAs and enhancer RNAs regulate the lipopolysaccharide-induced inflammatory response in human monocytes, Nat. Commun, vol.5, pp.2014-3979

D. Gosselin and C. K. Glass, Epigenomics of macrophages, Immunological Reviews, vol.470, issue.1, pp.96-112
DOI : 10.1111/imr.12213

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4203424

W. A. Whyte, D. A. Orlando, D. Hnisz, B. J. Abraham, C. Y. Lin et al., Master Transcription Factors and Mediator Establish Super-Enhancers at Key Cell Identity Genes, Cell, vol.153, issue.2, pp.307-319, 2013.
DOI : 10.1016/j.cell.2013.03.035

URL : http://doi.org/10.1016/j.cell.2013.03.035

J. Loven, H. A. Hoke, C. Y. Lin, A. Lau, D. A. Orlando et al., Selective Inhibition of Tumor Oncogenes by Disruption of Super-Enhancers, Cell, vol.153, issue.2, pp.320-334, 2013.
DOI : 10.1016/j.cell.2013.03.036

A. Asare and D. Zheng, Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice, Nature, vol.521, pp.366-370, 2015.

N. Hah, C. Benner, L. W. Chong, R. T. Yu, M. Downes et al., Inflammation-sensitive super enhancers form domains of coordinately regulated enhancer RNAs, Proc. Natl. Acad. Sci. USA 2015, pp.297-302
DOI : 10.1038/nature10006

T. J. Stasevich, Y. Hayashi-takanaka, Y. Sato, K. Maehara, Y. Ohkawa et al., Regulation of RNA polymerase II activation by histone acetylation in single living cells, Nature, vol.322, issue.7530, pp.272-275, 2014.
DOI : 10.1038/nature13714

Q. Jin, L. R. Yu, L. Wang, Z. Zhang, L. H. Kasper et al., Distinct roles of GCN5/PCAF-mediated H3K9ac and CBP/p300-mediated H3K18/27ac in nuclear receptor transactivation, The EMBO Journal, vol.29, issue.2, pp.249-262, 2011.
DOI : 10.1038/emboj.2010.318

Y. Zhu, L. Sun, Z. Chen, J. W. Whitaker, T. Wang et al., Predicting enhancer transcription and activity from chromatin modifications, Nucleic Acids Research, vol.41, issue.22, pp.10032-10043, 2013.
DOI : 10.1093/nar/gkt826

URL : http://doi.org/10.1093/nar/gkt826

B. Schwalb, M. Michel, B. Zacher, K. Fruhauf, C. Demel et al., TT-seq maps the human transient transcriptome, Science, vol.51, issue.6290, pp.1225-1228, 2016.
DOI : 10.1073/pnas.95.4.1460

K. R. Rosenbloom, J. Armstrong, G. P. Barber, J. Casper, H. Clawson et al., The UCSC Genome Browser database: 2015 update, Nucleic Acids Research, vol.43, issue.D1, pp.670-681, 2015.
DOI : 10.1093/nar/gku1177

URL : http://doi.org/10.1093/nar/gku1177

C. G. Danko, S. L. Hyland, L. J. Core, A. L. Martins, C. T. Waters et al., Identification of active transcriptional regulatory elements from GRO-seq data, Nature Methods, vol.460, issue.5, pp.433-438, 2015.
DOI : 10.1016/j.celrep.2013.01.004

G. Consortium, The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue gene regulation in humans, Science, vol.55, issue.4, pp.648-660, 2015.
DOI : 10.1016/j.biopsych.2003.10.013

M. J. Fullwood, Y. Han, C. L. Wei, X. Ruan, and Y. Ruan, Chromatin Interaction Analysis Using Paired-End Tag Sequencing, Curr. Protoc. Mol. Biol, vol.38, 2010.
DOI : 10.1002/0471142727.mb2115s89

G. Li, M. J. Fullwood, H. Xu, F. H. Mulawadi, S. Velkov et al., ChIA-PET tool for comprehensive chromatin interaction analysis with paired-end tag sequencing, R22. [CrossRef], 2010.
DOI : 10.1186/gb-2010-11-2-r22

G. Li, X. Ruan, R. K. Auerbach, K. S. Sandhu, M. Zheng et al., Extensive Promoter-Centered Chromatin Interactions Provide a Topological Basis for Transcription Regulation, Cell, vol.148, issue.1-2, pp.84-98, 2012.
DOI : 10.1016/j.cell.2011.12.014

Y. Nedelec, J. Sanz, G. Baharian, Z. A. Szpiech, A. Pacis et al., Genetic Ancestry and Natural Selection Drive Population Differences in Immune Responses to Pathogens, Genetic Ancestry and Natural Selection Drive Population Differences in Immune Responses to Pathogens, pp.657-669, 2016.
DOI : 10.1016/j.cell.2016.09.025

M. V. Rudan, S. Hadjur, and T. Sexton, Detecting Spatial Chromatin Organization by Chromosome Conformation Capture II: Genome-Wide Profiling by Hi-C, Methods Mol. Biol, pp.1589-1636, 2017.

Y. Guo, Q. Xu, D. Canzio, J. Shou, J. Li et al., CRISPR Inversion of CTCF Sites Alters Genome Topology and Enhancer/Promoter Function, et al. CRISPR Inversion of CTCF Sites Alters Genome Topology and Enhancer/Promoter Function, pp.900-910, 2015.
DOI : 10.1016/j.cell.2015.07.038

G. Fudenberg, M. Imakaev, C. Lu, A. Goloborodko, N. Abdennur et al., Formation of Chromosomal Domains by Loop Extrusion. Cell Rep, pp.2038-2049, 2016.

J. Wang, J. Zhuang, S. Iyer, X. Y. Lin, M. C. Greven et al., Factorbook.org: a Wiki-based database for transcription factor-binding data generated by the ENCODE consortium, Nucleic Acids Research, vol.41, issue.D1, pp.171-176, 2013.
DOI : 10.1093/nar/gks1221

Y. Guo, K. Monahan, H. Wu, J. Gertz, K. E. Varley et al., CTCF/cohesin-mediated DNA looping is required for protocadherin alpha promoter choice, Proc. Natl
DOI : 10.1073/pnas.1219280110

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3529044

A. Mathelier, O. Fornes, D. J. Arenillas, C. Y. Chen, G. Denay et al., JASPAR 2016: a major expansion and update of the open-access database of transcription factor binding profiles, Nucleic Acids Research, vol.44, issue.D1, pp.110-115, 2016.
DOI : 10.1093/nar/gkv1176

URL : https://hal.archives-ouvertes.fr/hal-01281181

T. Shiraki, S. Kondo, S. Katayama, K. Waki, T. Kasukawa et al., Cap analysis gene expression for high-throughput analysis of transcriptional starting point and identification of promoter usage, Proc. Natl. Acad. Sci. USA 2003, pp.15776-15781
DOI : 10.1093/nar/26.1.362

R. Andersson, Y. Chen, L. Core, J. T. Lis, A. Sandelin et al., Human Gene Promoters Are Intrinsically Bidirectional, Molecular Cell, vol.60, issue.3, pp.346-347, 2015.
DOI : 10.1016/j.molcel.2015.10.015

S. H. Duttke, S. A. Lacadie, M. M. Ibrahim, C. K. Glass, D. L. Corcoran et al., Human Promoters Are Intrinsically Directional, Molecular Cell, vol.57, issue.4, pp.674-684, 2015.
DOI : 10.1016/j.molcel.2014.12.029

URL : http://doi.org/10.1016/j.molcel.2014.12.029

A. Joshi, C. Pooley, T. C. Freeman, A. Lennartsson, M. Babina et al., Technical Advance: Transcription factor, promoter, and enhancer utilization in human myeloid cells, Journal of Leukocyte Biology, vol.97, issue.5, pp.985-995, 2015.
DOI : 10.1189/jlb.6TA1014-477RR

Y. Zhang, Y. Kim, N. Genoud, J. Gao, J. W. Kelly et al., Determinants for Dephosphorylation of the RNA Polymerase II C-Terminal Domain by Scp1, Molecular Cell, vol.24, issue.5, pp.759-770, 2006.
DOI : 10.1016/j.molcel.2006.10.027

M. Yeo, P. S. Lin, M. E. Dahmus, and G. N. Gill, A Novel RNA Polymerase II C-terminal Domain Phosphatase That Preferentially Dephosphorylates Serine 5, Journal of Biological Chemistry, vol.278, issue.28, pp.26078-26085, 2003.
DOI : 10.1074/jbc.M301791200

S. Marquet, P. Lepage, T. J. Hudson, J. M. Musser, and E. Schurr, Complete nucleotide sequence and genomic structure of the human NRAMP1 gene region on Chromosome region 2q35, Mammalian Genome, vol.11, issue.9, pp.755-762, 2000.
DOI : 10.1007/s003350010151

A. J. Gonzalez, M. Setty, and C. S. Leslie, Early enhancer establishment and regulatory locus complexity shape transcriptional programs in hematopoietic differentiation, Nature Genetics, vol.11, issue.11, pp.1249-1259, 2015.
DOI : 10.1093/nar/gkn923

F. Bontems, R. J. Fish, I. Borlat, F. Lembo, S. Chocu et al., Neerman-Arbez, M.; Bairoch, A.; et al. C2orf62 and TTC17 are involved in actin organization and ciliogenesis in zebrafish and human, PLoS ONE, vol.2014, issue.9, p.86476

M. Cellier, C. Shustik, W. Dalton, E. Rich, J. Hu et al., Expression of the human NRAMP1 gene in professional primary phagocytes: Studies in blood cells and in HL-60 promyelocytic leukemia, J. Leukoc. Biol, pp.61-96, 1997.

G. M. De, M. Hosseini, and D. Vernimmen, Intragenic enhancers act as alternative promoters, Mol. Cell, vol.45, pp.447-458, 2012.

E. A. Roig, E. Richer, F. Canonne-hergaux, P. Gros, and M. F. Cellier, Regulation of NRAMP1 gene expression by 1alpha,25-dihydroxy-vitamin D(3) in HL-60 phagocytes, J. Leukoc. Biol, vol.71, pp.890-904, 2002.

F. Canonne-hergaux, J. Calafat, E. Richer, M. Cellier, S. Grinstein et al., Expression and subcellular localization of NRAMP1 in human neutrophil granules, Blood, vol.100, issue.1, pp.268-275, 2002.
DOI : 10.1182/blood.V100.1.268

S. Neph, J. Vierstra, A. B. Stergachis, A. P. Reynolds, E. Haugen et al., An expansive human regulatory lexicon encoded in transcription factor footprints, Nature, vol.3, issue.7414, pp.83-90, 2012.
DOI : 10.1038/nature11212

Y. Sugi, K. Takahashi, K. Nakano, A. Hosono, and S. Kaminogawa, Transcription of the Tollip gene is elevated in intestinal epithelial cells through impaired O-GlcNAcylation-dependent nuclear translocation of the negative regulator Elf-1, Biochemical and Biophysical Research Communications, vol.412, issue.4, pp.704-709, 2011.
DOI : 10.1016/j.bbrc.2011.08.035

Y. Okada, H. Nobori, M. Shimizu, M. Watanabe, M. Yonekura et al., Multiple ETS Family Proteins Regulate PF4 Gene Expression by Binding to the Same ETS Binding Site, PLoS ONE, vol.30, issue.9, p.24837, 2011.
DOI : 10.1371/journal.pone.0024837.s006

P. Xiang, C. Lo, B. Argiropoulos, C. B. Lai, A. Rouhi et al., Identification of E74-like factor 1 (ELF1) as a transcriptional regulator of the Hox cofactor MEIS1, Experimental Hematology, vol.38, issue.9, p.38, 2010.
DOI : 10.1016/j.exphem.2010.06.006

Q. H. Wang, C. Nishiyama, N. Nakano, N. Shimokawa, M. Hara et al., Suppressive effect of Elf-1 on Fc?RI ?-chain expression in primary mast cells, Immunogenetics, vol.14, issue.10, pp.557-563, 2008.
DOI : 10.1007/s00251-008-0318-y

G. Jalagadugula, D. N. Dhanasekaran, S. Kim, S. P. Kunapuli, and A. K. Rao, q gene in megakaryocytic cells, Journal of Thrombosis and Haemostasis, vol.104, issue.12, pp.2678-2686, 2006.
DOI : 10.1111/j.1538-7836.2006.02229.x

M. D. Bjerregaard, J. Jurlander, P. Klausen, N. Borregaard, and J. B. Cowland, The in vivo profile of transcription factors during neutrophil differentiation in human bone marrow, Blood, vol.101, issue.11, pp.4322-4332, 2003.
DOI : 10.1182/blood-2002-03-0835

M. Rolli-derkinderen, F. Machavoine, J. M. Baraban, A. Grolleau, L. Beretta et al., ERK and p38 Inhibit the Expression of 4E-BP1 Repressor of Translation through Induction of Egr-1, Journal of Biological Chemistry, vol.278, issue.21, pp.18859-18867, 2003.
DOI : 10.1074/jbc.M211696200

O. Reilly, D. Quinn, C. M. El-shanawany, T. Gordon, S. Greaves et al., Multiple Ets Factors and Interferon Regulatory Factor-4 Modulate CD68 Expression in a Cell Type-specific Manner, Journal of Biological Chemistry, vol.278, issue.24, pp.21909-21919, 2003.
DOI : 10.1074/jbc.M212150200

M. Guha and N. Mackman, The Phosphatidylinositol 3-Kinase-Akt Pathway Limits Lipopolysaccharide Activation of Signaling Pathways and Expression of Inflammatory Mediators in Human Monocytic Cells, Journal of Biological Chemistry, vol.277, issue.35, pp.32124-32132, 2002.
DOI : 10.1074/jbc.M203298200

Z. Xu, R. Dziarski, Q. Wang, K. Swartz, K. M. Sakamoto et al., Bacterial Peptidoglycan-Induced tnf-? Transcription Is Mediated Through the Transcription Factors Egr-1, Elk-1, and NF-?B, The Journal of Immunology, vol.167, issue.12, pp.6975-6982, 2001.
DOI : 10.4049/jimmunol.167.12.6975

K. S. Voo and D. G. Skalnik, Elf-1 and PU.1 induce expression of gp91(phox) via a promoter element mutated in a subset of chronic granulomatous disease patients, Blood, vol.93, pp.3512-3520, 1999.

D. Battista, J. A. Martel-pelletier, J. Pelletier, and J. , Suppression of tumor necrosis factor (TNF-??) gene expression by prostaglandin E2. Role of early growth response protein-1 (Egr-1), Osteoarthritis and Cartilage, vol.7, issue.4, pp.395-398, 1999.
DOI : 10.1053/joca.1998.0222

S. Nimer, J. Zhang, H. Avraham, and Y. Miyazaki, Transcriptional regulation of interleukin-3 expression in megakaryocytes, Blood, vol.88, pp.66-74, 1996.

T. Affymetrix and . Project, Cold Spring Harbor Laboratory ENCODE Transcriptome Project. Post-transcriptional processing generates a diversity of 5 -modified long and short RNAs, Nature, vol.457, pp.1028-1032, 2009.

H. Kawaji, J. Severin, M. Lizio, A. Waterhouse, S. Katayama et al., The FANTOM web resource: from mammalian transcriptional landscape to its dynamic regulation, Genome Biology, vol.10, issue.4, p.40, 2009.
DOI : 10.1186/gb-2009-10-4-r40

H. Kawaji, T. Kasukawa, S. Fukuda, S. Katayama, C. Kai et al., CAGE Basic/Analysis Databases: the CAGE resource for comprehensive promoter analysis, Nucleic Acids Research, vol.34, issue.90001, pp.632-636, 2006.
DOI : 10.1093/nar/gkj034

W. R. Mccombie, Directional DNA methylation changes and complex intermediate states accompany lineage specificity in the adult hematopoietic compartment, Mol. Cell, vol.44, pp.17-28, 2011.

Y. Li, J. Zhu, G. Tian, N. Li, Q. Li et al., The DNA Methylome of Human Peripheral Blood Mononuclear Cells, PLoS Biology, vol.24, issue.11, p.1000533, 2010.
DOI : 10.1371/journal.pbio.1000533.s020

C. T. Ong and V. G. Corces, CTCF: an architectural protein bridging genome topology and function, Nature Reviews Genetics, vol.460, issue.4, pp.234-246
DOI : 10.1126/science.1236083

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4610363

Y. Qiao, E. G. Giannopoulou, C. H. Chan, S. H. Park, S. Gong et al., Synergistic Activation of Inflammatory Cytokine Genes by Interferon-??-Induced Chromatin Remodeling and Toll-like Receptor Signaling, Immunity, vol.39, issue.3, pp.454-469, 2013.
DOI : 10.1016/j.immuni.2013.08.009

D. Alvarez-errico, R. Vento-tormo, M. Sieweke, and E. Ballestar, Epigenetic control of myeloid cell differentiation, identity and function, Nature Reviews Immunology, vol.447, issue.1, pp.7-17, 2015.
DOI : 10.1093/nar/gkr1015

A. Khan and X. Zhang, dbSUPER: A database of super-enhancers in mouse and human genome, Nucleic Acids Res, vol.2016, issue.44, pp.164-171

H. E. Pelish, B. B. Liau, I. I. Nitulescu, A. Tangpeerachaikul, Z. C. Poss et al., Mediator kinase inhibition further activates super-enhancer-associated genes in AML, Nature, vol.57, issue.7572, pp.273-276, 2015.
DOI : 10.1016/j.cell.2013.04.048

M. Tausendschon, M. Rehli, N. Dehne, C. Schmidl, C. Doring et al., Genome-wide identification of hypoxia-inducible factor-1 and -2 binding sites in hypoxic human macrophages alternatively activated by IL-10, Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms, vol.1849, issue.1, pp.10-22, 2015.
DOI : 10.1016/j.bbagrm.2014.10.006

S. Searle and J. M. Blackwell, Evidence for a functional repeat polymorphism in the promoter of the human NRAMP1 gene that correlates with autoimmune versus infectious disease susceptibility, J. Med. Genet, vol.36, pp.295-299, 1999.

H. K. Bayele, C. Peyssonnaux, A. Giatromanolaki, W. W. Arrais-silva, H. S. Mohamed et al., HIF-1 regulates heritable variation and allele expression phenotypes of the macrophage immune response gene SLC11A1 from a Z-DNA forming microsatellite, Blood, vol.110, issue.8, pp.3039-3048, 2007.
DOI : 10.1182/blood-2006-12-063289

R. Galeev, A. Baudet, P. Kumar, N. A. Rundberg, B. Nilsson et al., Genome-wide RNAi Screen Identifies Cohesin Genes as Modifiers of Renewal and Differentiation in Human HSCs, Cell Reports, vol.14, issue.12, pp.2988-3000, 2016.
DOI : 10.1016/j.celrep.2016.02.082

H. Quach, M. Rotival, J. Pothlichet, Y. E. Loh, M. Dannemann et al., Genetic Adaptation and Neandertal Admixture Shaped the Immune System of Human Populations, Genetic Adaptation and Neandertal Admixture Shaped the Immune System of Human Populations, pp.643-656, 2016.
DOI : 10.1016/j.cell.2016.09.024

URL : https://hal.archives-ouvertes.fr/pasteur-01385620

J. R. Dixon, D. U. Gorkin, and B. Ren, Chromatin Domains: The Unit of Chromosome Organization, Molecular Cell, vol.62, issue.5, pp.668-680, 2016.
DOI : 10.1016/j.molcel.2016.05.018

S. P. Yip, K. H. Leung, and C. K. Lin, Extent and distribution of linkage disequilibrium around the SLC11A1 locus, Genes and Immunity, vol.4, issue.3, pp.212-221, 2003.
DOI : 10.1038/sj.gene.6363944

N. S. Archer, N. T. Nassif, and B. A. O-'brien, Genetic variants of SLC11A1 are associated with both autoimmune and infectious diseases: systematic review and meta-analysis, Genes and Immunity, vol.23, issue.4, pp.275-283, 2015.
DOI : 10.1038/gene.2015.8

M. Cellier, G. Govoni, S. Vidal, T. Kwan, N. Groulx et al., Human natural resistance-associated macrophage protein: cDNA cloning, chromosomal mapping, genomic organization, and tissue-specific expression, Journal of Experimental Medicine, vol.180, issue.5, pp.1741-1752, 1994.
DOI : 10.1084/jem.180.5.1741

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2191750/pdf

Y. Z. Xu, M. S. Di, I. Gallouzi, M. Rola-pleszczynski, and D. Radzioch, RNA-Binding Protein HuR Is Required for Stabilization of SLC11A1 mRNA and SLC11A1 Protein Expression, Molecular and Cellular Biology, vol.25, issue.18, pp.8139-8149, 2005.
DOI : 10.1128/MCB.25.18.8139-8149.2005

J. Moisan, T. Thuraisingam, J. Henault, J. De-sanctis, and D. Radzioch, Role of SLC11A1 (formerly NRAMP1) in regulation of signal transduction induced by Toll-like receptor 7 ligands, FEMS Immunology & Medical Microbiology, vol.47, issue.1, pp.138-147, 2006.
DOI : 10.1111/j.1574-695X.2006.00077.x

Y. Z. Xu, T. Thuraisingam, R. Marino, and D. Radzioch, Recruitment of SWI/SNF Complex Is Required for Transcriptional Activation of the SLC11A1 Gene during Macrophage Differentiation of HL-60 Cells, Journal of Biological Chemistry, vol.286, issue.15
DOI : 10.1074/jbc.M110.185637

M. G. Zaahl, K. J. Robson, L. Warnich, and M. J. Kotze, Expression of the SLC11A1 (NRAMP1) 5?-(GT)n repeat: Opposite effect in the presence of ?237C?T, Blood Cells, Molecules, and Diseases, vol.33, issue.1, pp.45-50, 2004.
DOI : 10.1016/j.bcmd.2004.04.003

S. Taka, M. Gazouli, P. K. Politis, K. I. Pappa, and N. P. Anagnou, Transcription factor ATF-3 regulates allele variation phenotypes of the human SLC11A1 gene, Molecular Biology Reports, vol.5, issue.7, pp.2263-2271, 2012.
DOI : 10.1007/s11033-012-2289-1

E. Mass, I. Ballesteros, M. Farlik, F. Halbritter, P. Gunther et al., Specification of tissue-resident macrophages during organogenesis, Science, vol.57, issue.6172, p.4238, 2016.
DOI : 10.1126/science.1247651

D. Gosselin, V. M. Link, C. E. Romanoski, G. J. Fonseca, D. Z. Eichenfield et al., Environment Drives Selection and Function of Enhancers Controlling Tissue-Specific Macrophage Identities, Cell, vol.159, issue.6, pp.1327-1340, 2014.
DOI : 10.1016/j.cell.2014.11.023

Y. Lavin, D. Winter, R. Blecher-gonen, E. David, H. Keren-shaul et al., Tissue-Resident Macrophage Enhancer Landscapes Are Shaped by the Local Microenvironment, Cell, vol.159, issue.6, pp.1312-1326, 2014.
DOI : 10.1016/j.cell.2014.11.018

A. K. Jha, S. C. Huang, A. Sergushichev, V. Lampropoulou, Y. Ivanova et al., Network Integration of Parallel Metabolic and Transcriptional Data Reveals Metabolic Modules that Regulate Macrophage Polarization, Immunity, vol.42, issue.3, pp.419-430, 2015.
DOI : 10.1016/j.immuni.2015.02.005

B. Kelly and L. A. Neill, Metabolic reprogramming in macrophages and dendritic cells in innate immunity, Cell Research, vol.131, issue.7, pp.771-784
DOI : 10.1016/j.cmet.2014.03.014

M. F. Cellier and P. Courville, Campion, C. Nramp1 phagocyte intracellular metal withdrawal defense. Microbes Infect, pp.1662-1670, 2007.

J. D. Brown, C. Y. Lin, Q. Duan, G. Griffin, A. J. Federation et al., NF-?B Directs Dynamic Super Enhancer Formation in Inflammation and Atherogenesis, Molecular Cell, vol.56, issue.2, pp.219-231, 2014.
DOI : 10.1016/j.molcel.2014.08.024

J. G. Peeters, S. J. Vervoort, S. C. Tan, G. Mijnheer, S. De-roock et al., Inhibition of Super-Enhancer Activity in Autoinflammatory Site-Derived T Cells Reduces Disease-Associated Gene Expression. Cell Rep, pp.1986-1996, 2015.

J. S. Roe, F. Mercan, K. Rivera, D. J. Pappin, and C. R. Vakoc, BET Bromodomain Inhibition Suppresses the Function of Hematopoietic Transcription Factors in Acute Myeloid Leukemia, Molecular Cell, vol.58, issue.6, pp.1028-1039, 2015.
DOI : 10.1016/j.molcel.2015.04.011

D. X. Quang, M. R. Erdos, S. C. Parker, and F. S. Collins, Motif signatures in stretch enhancers are enriched for disease-associated genetic variants, Epigenetics & Chromatin, vol.8, issue.2, p.23, 2015.
DOI : 10.1186/s13072-015-0015-7

B. Novakovic, E. Habibi, S. Y. Wang, R. J. Arts, R. Davar et al., ?-Glucan Reverses the Epigenetic State of LPS-Induced Immunological Tolerance, Cell, vol.167, issue.5, pp.1354-1368, 2016.
DOI : 10.1016/j.cell.2016.09.034

V. D. Marinescu, I. S. Kohane, and A. Riva, The MAPPER database: a multi-genome catalog of putative transcription factor binding sites, Nucleic Acids Research, vol.33, issue.Database issue, pp.91-97, 2005.
DOI : 10.1093/nar/gki103

S. Roy, S. Schmeier, E. Arner, T. Alam, S. P. Parihar et al., Redefining the transcriptional regulatory dynamics of classically and alternatively activated macrophages by deepCAGE transcriptomics, Nucleic Acids Research, vol.43, issue.14, pp.6969-6982, 2015.
DOI : 10.1093/nar/gkv646

H. Suzuki, A. R. Forrest, E. Van-nimwegen, C. O. Daub, P. J. Balwierz et al., The transcriptional network that controls growth arrest and differentiation in a human myeloid leukemia cell line, Nature Genetics, vol.178, issue.5, pp.553-562, 2009.
DOI : 10.1006/excr.2000.5044

J. Xue, S. V. Schmidt, J. Sander, A. Draffehn, W. Krebs et al., Transcriptome-Based Network Analysis Reveals a Spectrum Model of Human Macrophage Activation, Immunity, vol.40, issue.2, pp.274-288, 2014.
DOI : 10.1016/j.immuni.2014.01.006

J. Drouin and . Minireview, Minireview: Pioneer Transcription Factors in Cell Fate Specification, Molecular Endocrinology, vol.28, issue.7, pp.989-998
DOI : 10.1210/me.2014-1084

M. Iwafuchi-doi and K. S. Zaret, Pioneer transcription factors in cell reprogramming, Genes & Development, vol.28, issue.24, pp.2679-2692
DOI : 10.1101/gad.253443.114

K. R. Salci, B. A. Mcintyre, and M. Bhatia, Foundational concepts of cell fate conversion to the hematopoietic lineage, Current Opinion in Genetics & Development, vol.23, issue.5, pp.585-590, 2013.
DOI : 10.1016/j.gde.2013.04.007

W. Ebina and D. J. Rossi, Transcription factor-mediated reprogramming toward hematopoietic stem cells, The EMBO Journal, vol.34, issue.6, pp.694-709
DOI : 10.15252/embj.201490804

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4369309

F. Ginhoux and M. Guilliams, Tissue-Resident Macrophage Ontogeny and Homeostasis, Immunity, vol.44, issue.3, pp.439-449, 2016.
DOI : 10.1016/j.immuni.2016.02.024

K. E. Mcgrath, J. M. Frame, K. H. Fegan, J. R. Bowen, S. J. Conway et al., Distinct Sources of Hematopoietic Progenitors Emerge before HSCs and Provide Functional Blood Cells in the Mammalian Embryo, Cell Reports, vol.11, issue.12, pp.1892-1904, 2015.
DOI : 10.1016/j.celrep.2015.05.036

P. Cauchy, S. R. James, J. Zacarias-cabeza, A. Ptasinska, M. R. Imperato et al., Chronic FLT3-ITD Signaling in Acute Myeloid Leukemia Is Connected to a Specific Chromatin Signature, Cell Reports, vol.12, issue.5, pp.821-836, 2015.
DOI : 10.1016/j.celrep.2015.06.069

A. Eriksson, A. Lennartsson, and S. Lehmann, Epigenetic aberrations in acute myeloid leukemia: Early key events during leukemogenesis, Experimental Hematology, vol.43, issue.8, pp.609-624, 2015.
DOI : 10.1016/j.exphem.2015.05.009

A. M. Sotoca, K. H. Prange, B. Reijnders, A. Mandoli, L. N. Nguyen et al., The oncofusion protein FUS?ERG targets key hematopoietic regulators and modulates the all-trans retinoic acid signaling pathway in t(16;21) acute myeloid leukemia, Oncogene, vol.83, issue.15, pp.1965-1976, 2016.
DOI : 10.1093/bioinformatics/btp352

Y. Li, H. Wang, X. Wang, W. Jin, Y. Tan et al., Genome-wide studies identify a novel interplay between AML1 and AML1/ETO in t(8;21) acute myeloid leukemia, Blood, vol.127, issue.2, pp.233-242, 2016.
DOI : 10.1182/blood-2015-03-626671

D. J. Trombly, T. W. Whitfield, S. Padmanabhan, J. A. Gordon, J. B. Lian et al., Genome-wide co-occupancy of AML1-ETO and N-CoR defines the t(8;21) AML signature in leukemic cells, BMC Genomics, vol.102, issue.43, pp.16-309, 2015.
DOI : 10.1186/s12864-015-1445-0

A. Mandoli, A. A. Singh, P. W. Jansen, A. T. Wierenga, H. Riahi et al., CBFB?MYH11/RUNX1 together with a compendium of hematopoietic regulators, chromatin modifiers and basal transcription factors occupies self-renewal genes in inv(16) acute myeloid leukemia, Leukemia, vol.18, issue.4, pp.770-778, 2014.
DOI : 10.1038/leu.2012.49

J. Tober, A. D. Yzaguirre, E. Piwarzyk, and N. A. Speck, Distinct temporal requirements for Runx1 in hematopoietic progenitors and stem cells, Development, vol.140, issue.18, pp.3765-3776, 2013.
DOI : 10.1242/dev.094961

M. Lanotte, V. Martin-thouvenin, S. Najman, P. Balerini, F. Valensi et al., NB4, a maturation inducible cell line with t(15;17) marker isolated from a human acute promyelocytic leukemia (M3), Blood, vol.77, pp.1080-1086, 1991.

M. R. Brent, Past Roadblocks and New Opportunities in Transcription Factor Network Mapping, Trends in Genetics, vol.32, issue.11, pp.736-750
DOI : 10.1016/j.tig.2016.08.009

C. L. Frank, D. Manandhar, R. Gordan, and G. E. Crawford, HDAC inhibitors cause site-specific chromatin remodeling at PU.1-bound enhancers in K562 cells, Epigenetics & Chromatin, vol.42, issue.Web Server issu, p.15
DOI : 10.1186/s13072-016-0065-5

URL : http://doi.org/10.1186/s13072-016-0065-5

L. M. Carlin, E. G. Stamatiades, C. Auffray, R. N. Hanna, L. Glover et al., Nr4a1-Dependent Ly6Clow Monocytes Monitor Endothelial Cells and Orchestrate Their Disposal, Cell, vol.153, issue.2, pp.362-375, 2013.
DOI : 10.1016/j.cell.2013.03.010

URL : http://doi.org/10.1016/j.cell.2013.03.010

G. Thomas, R. Tacke, C. C. Hedrick, and R. N. Hanna, Nonclassical Patrolling Monocyte Function in the Vasculature, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.35, issue.6, pp.1306-1316, 2015.
DOI : 10.1161/ATVBAHA.114.304650

G. D. Thomas, R. N. Hanna, N. T. Vasudevan, A. A. Hamers, C. E. Romanoski et al., Deleting an Nr4a1 Super-Enhancer Subdomain Ablates Ly6Clow Monocytes while Preserving Macrophage Gene Function, Immunity, vol.45, issue.5, pp.975-987, 2016.
DOI : 10.1016/j.immuni.2016.10.011

C. Schmidl, K. Renner, K. Peter, R. Eder, T. Lassmann et al., Transcription and enhancer profiling in human monocyte subsets, Blood, vol.123, issue.17, pp.90-99, 2014.
DOI : 10.1182/blood-2013-02-484188

E. Gammella, P. Buratti, G. Cairo, S. Recalcati, and . Macrophages, Macrophages: central regulators of iron balance, Metallomics, vol.339, issue.8, pp.1336-1345, 2014.
DOI : 10.1002/mrm.25267

T. Ganz, Macrophages and Iron Metabolism

P. Rawat, S. Kumar, N. Sheokand, C. I. Raje, and M. Raje, This article is part of Special Issue entitled Lactoferrin and has undergone the Journal's usual peer review process., Biochemistry and Cell Biology, vol.90, issue.3, pp.329-338, 2012.
DOI : 10.1139/o11-058

F. Zhang, Y. Tao, Z. Zhang, X. Guo, P. An et al., Metalloreductase Steap3 coordinates the regulation of iron homeostasis and inflammatory responses, Haematologica, vol.97, issue.12, pp.1826-1835, 2012.
DOI : 10.3324/haematol.2012.063974

S. Jacolot, Y. Yang, P. Paitry, C. Ferec, and C. Mura, Iron metabolism in macrophages from HFE hemochromatosis patients, Molecular Genetics and Metabolism, vol.101, issue.2-3, pp.258-267, 2010.
DOI : 10.1016/j.ymgme.2010.07.010

K. Venkatachalam, C. O. Wong, and M. X. Zhu, The role of TRPMLs in endolysosomal trafficking and function, Cell Calcium, vol.58, issue.1, pp.48-56, 2015.
DOI : 10.1016/j.ceca.2014.10.008

R. M. Dayam, A. Saric, R. E. Shilliday, and R. J. Botelho, Channel, TRPML1, Is Required for Phagosome Maturation, Traffic, vol.382, issue.9, pp.1010-1026, 2015.
DOI : 10.1111/tra.12303

C. White, X. Yuan, P. J. Schmidt, E. Bresciani, T. K. Samuel et al., HRG1 Is Essential for Heme Transport from the Phagolysosome of Macrophages during Erythrophagocytosis, Cell Metabolism, vol.17, issue.2, pp.261-270, 2013.
DOI : 10.1016/j.cmet.2013.01.005

C. A. Schaer, F. Vallelian, A. Imhof, G. Schoedon, and D. J. Schaer, Heme carrier protein (HCP-1) spatially interacts with the CD163 hemoglobin uptake pathway and is a target of inflammatory macrophage activation, Journal of Leukocyte Biology, vol.83, issue.2, pp.325-333, 2008.
DOI : 10.1189/jlb.0407226

S. P. Duffy, J. Shing, P. Saraon, L. C. Berger, M. V. Eiden et al., The Fowler Syndrome-Associated Protein FLVCR2 Is an Importer of Heme, Molecular and Cellular Biology, vol.30, issue.22, pp.5318-5324, 2010.
DOI : 10.1128/MCB.00690-10

A. G. Frey, A. Nandal, J. H. Park, P. M. Smith, T. Yabe et al., Iron chaperones PCBP1 and PCBP2 mediate the metallation of the dinuclear iron enzyme deoxyhypusine hydroxylase, Proc. Natl. Acad. Sci. USA 2014, pp.8031-8036
DOI : 10.1038/nprot.2006.478

W. E. Dowdle, B. Nyfeler, J. Nagel, R. A. Elling, S. Liu et al., Selective VPS34 inhibitor blocks autophagy and uncovers a role for NCOA4 in ferritin degradation and iron homeostasis in vivo, Nature Cell Biology, vol.245, issue.11, pp.1069-1079, 2014.
DOI : 10.1021/pr101065j

J. D. Mancias, X. Wang, S. P. Gygi, J. W. Harper, and A. C. Kimmelman, Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy, Nature, vol.9, issue.7498, pp.105-109, 2014.
DOI : 10.1038/nature13148

S. B. Keel, R. T. Doty, Z. Yang, J. G. Quigley, J. Chen et al., A Heme Export Protein Is Required for Red Blood Cell Differentiation and Iron Homeostasis, Science, vol.73, issue.8, pp.825-828, 2008.
DOI : 10.1038/nm1629

T. G. Nakashige, B. Zhang, C. Krebs, and E. M. Nolan, Human calprotectin is an iron-sequestering host-defense protein, Nature Chemical Biology, vol.145, issue.10, pp.765-771, 2015.
DOI : 10.1016/0003-2697(71)90405-2

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4575267

O. Neill, L. A. Kishton, R. J. Rathmell, and J. , A guide to immunometabolism for immunologists, Nature Reviews Immunology, vol.226, issue.9, pp.553-565, 2016.
DOI : 10.1016/j.celrep.2015.09.036

J. Rozowsky, G. Euskirchen, R. K. Auerbach, Z. D. Zhang, T. Gibson et al., PeakSeq enables systematic scoring of ChIP-seq experiments relative to controls, Nature Biotechnology, vol.6, issue.1, pp.66-75, 2009.
DOI : 10.1038/nbt.1518

P. H. Krijger, B. Di-stefano, E. De-wit, F. Limone, C. Van-oevelen et al., Cell-of-Origin-Specific 3D Genome Structure Acquired during Somatic Cell Reprogramming, Cell Stem Cell, vol.18, issue.5, pp.597-610, 2016.
DOI : 10.1016/j.stem.2016.01.007

D. K. Goode, N. Obier, M. S. Vijayabaskar, A. L. Lie, A. J. Lilly et al., Dynamic Gene Regulatory Networks Drive Hematopoietic Specification and Differentiation, Developmental Cell, vol.36, issue.5, pp.572-587, 2016.
DOI : 10.1016/j.devcel.2016.01.024

URL : http://doi.org/10.1016/j.devcel.2016.01.024

F. Yue, Y. Cheng, A. Breschi, J. Vierstra, W. Wu et al., A comparative encyclopedia of DNA elements in the mouse genome, Nature, vol.46, issue.7527, pp.355-364, 2014.
DOI : 10.1038/nature13992

A. Luyten, C. Zang, X. S. Liu, and R. A. Shivdasani, Active enhancers are delineated de novo during hematopoiesis, with limited lineage fidelity among specified primary blood cells, Genes & Development, vol.28, issue.16, pp.1827-1839, 2014.
DOI : 10.1101/gad.240101.114

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4197967

F. Paul, Y. Arkin, A. Giladi, D. A. Jaitin, E. Kenigsberg et al., Transcriptional Heterogeneity and Lineage Commitment in Myeloid Progenitors, Transcriptional Heterogeneity and Lineage Commitment in Myeloid Progenitors, pp.1663-1677, 2015.
DOI : 10.1016/j.cell.2015.11.013

F. Ferri, A. Parcelier, V. Petit, A. S. Gallouet, D. Lewandowski et al., et al. TRIM33 switches off Ifnb1 gene transcription during the late phase of macrophage activation, Nat. Commun, vol.6, pp.2015-8900

D. Langlais, L. B. Barreiro, and P. Gros, The macrophage IRF8/IRF1 regulome is required for protection against infections and is associated with chronic inflammation, The Journal of Experimental Medicine, vol.15, issue.4, pp.585-603, 2016.
DOI : 10.1038/nm.2007

O. Matcovitch-natan, D. R. Winter, A. Giladi, A. S. Vargas, A. Spinrad et al., Microglia development follows a stepwise program to regulate brain homeostasis, Science, vol.10, issue.3, p.8670, 2016.
DOI : 10.1186/gb-2009-10-3-r25

URL : https://hal.archives-ouvertes.fr/hal-01438174

G. Govoni, S. Gauthier, F. Billia, N. N. Iscove, and P. Gros, Cell-specific and inducible Nramp1 gene expression in mouse macrophages in vitro and in vivo, J. Leukoc. Biol, vol.62, pp.277-286, 1997.

A. Weiner, D. Lara-astiaso, V. Krupalnik, O. Gafni, E. David et al., Co-ChIP enables genome-wide mapping of histone mark co-occurrence at single-molecule resolution, Nature Biotechnology, vol.543, issue.9, pp.953-961, 2016.
DOI : 10.1038/nsmb.1972

V. Kumar, N. A. Rayan, M. Muratani, S. Lim, B. Elanggovan et al., Comprehensive benchmarking reveals H2BK20 acetylation as a distinctive signature of cell-state-specific enhancers and promoters, Genome Research, vol.26, issue.5, pp.612-623, 2016.
DOI : 10.1101/gr.201038.115

S. T. Smale, A Stringent Systems Approach Uncovers Gene-Specific Mechanisms Regulating Inflammation, Cell, vol.165, pp.165-179, 2016.

A. S. Bhagwat, J. S. Roe, B. Y. Mok, A. F. Hohmann, J. Shi et al., BET Bromodomain Inhibition Releases the Mediator Complex from Select cis-Regulatory Elements. Cell Rep, pp.519-530, 2016.

M. S. Hasemann, F. K. Lauridsen, J. Waage, J. S. Jakobsen, A. K. Frank et al., C/EBP?? Is Required for Long-Term Self-Renewal and Lineage Priming of Hematopoietic Stem Cells and for the Maintenance of Epigenetic Configurations in Multipotent Progenitors, PLoS Genetics, vol.20, issue.4, p.1004079, 2014.
DOI : 10.1371/journal.pgen.1004079.s016

H. Zhang, M. Alberich-jorda, G. Amabile, H. Yang, P. B. Staber et al., Sox4 Is a Key Oncogenic Target in C/EBP? Mutant Acute Myeloid Leukemia, Cancer Cell, vol.24, issue.5, pp.575-588, 2013.
DOI : 10.1016/j.ccr.2013.09.018

R. Kumar, V. Fossati, M. Israel, and H. W. Snoeck, Lin-Sca1+Kit- Bone Marrow Cells Contain Early Lymphoid-Committed Precursors That Are Distinct from Common Lymphoid Progenitors, The Journal of Immunology, vol.181, issue.11, pp.7507-7513, 2008.
DOI : 10.4049/jimmunol.181.11.7507

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2596664/pdf

N. Miyake and N. Matsumoto, Essential role of the IRF8-KLF4 transcription factor cascade in murine monocyte differentiation, Blood, vol.121, pp.1839-1849, 2013.

K. , W. Satpathy, A. T. Rapaport, A. S. Briseno, C. G. Wu et al., Myc expression by dendritic cells is required for optimal T-cell priming, Nature, vol.507, pp.243-247, 2014.

T. Tamura, T. Nagamura-inoue, Z. Shmeltzer, T. Kuwata, and K. Ozato, ICSBP Directs Bipotential Myeloid Progenitor Cells to Differentiate into Mature Macrophages, Immunity, vol.13, issue.2, pp.155-165, 2000.
DOI : 10.1016/S1074-7613(00)00016-9

URL : http://doi.org/10.1016/s1074-7613(00)00016-9

A. Yanez and H. S. Goodridge, Interferon regulatory factor 8 and the regulation of neutrophil, monocyte, and dendritic cell production, Current Opinion in Hematology, vol.23, issue.1, pp.11-17, 2016.
DOI : 10.1097/MOH.0000000000000196

Y. Oishi, N. J. Spann, V. M. Link, E. D. Muse, T. Strid et al., SREBP1 Contributes to Resolution of Pro-inflammatory TLR4 Signaling by Reprogramming Fatty Acid Metabolism, Cell Metabolism, vol.25, issue.2, pp.412-427, 2017.
DOI : 10.1016/j.cmet.2016.11.009

D. Villar, C. Berthelot, S. Aldridge, T. F. Rayner, M. Lukk et al., Enhancer Evolution across 20 Mammalian Species, Cell, vol.160, issue.3, pp.554-566, 2015.
DOI : 10.1016/j.cell.2015.01.006

URL : http://doi.org/10.1016/j.cell.2015.01.006

T. Van-den-bosch, A. Boichenko, N. G. Leus, M. E. Ourailidou, H. Wapenaar et al., The histone acetyltransferase p300 inhibitor C646 reduces pro-inflammatory gene expression and inhibits histone deacetylases, Biochemical Pharmacology, vol.102, pp.130-140, 2016.
DOI : 10.1016/j.bcp.2015.12.010

M. G. Netea, L. A. Joosten, E. Latz, K. H. Mills, G. Natoli et al., Trained immunity: A program of innate immune memory in health and disease, Science, vol.7, issue.7, p.1098, 2016.
DOI : 10.1371/journal.pone.0041399

L. Tacchini, E. Gammella, C. De-ponti, S. Recalcati, and G. Cairo, Role of HIF-1 and NF-?B Transcription Factors in the Modulation of Transferrin Receptor by Inflammatory and Anti-inflammatory Signals, Journal of Biological Chemistry, vol.283, issue.30, pp.20674-20686, 2008.
DOI : 10.1074/jbc.M800365200

C. K. Glass, Genetic and Genomic Approaches to Understanding Macrophage Identity and FunctionATVB Named Lecture Reviews?Insight Into Author, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.35, issue.4, pp.755-762
DOI : 10.1161/ATVBAHA.114.304051

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4376616

M. Fattahi-dolatabadi, T. Mousavi, H. Mohammadi-barzelighi, S. Irian, B. Bakhshi et al., Akbari, N. NRAMP1 gene polymorphisms and cutaneous leishmaniasis: An evaluation on host susceptibility and treatment outcome, J. Vector Borne Dis, vol.53, pp.257-263, 2016.

M. Kadkhodazadeh, A. R. Ebadian, R. Amid, P. Zarnegarnia, F. Mollaverdi et al., Natural Resistance Associated Macrophage Protein 1 Gene Polymorphism is Associated with Chronic Periodontitis Not Peri-Implantitis in an Iranian Population: A Cross Sectional Study, Acta Med. Iran, vol.54, pp.323-329, 2016.

M. J. Brochado, M. F. Gatti, M. A. Zago, and A. M. Roselino, Association of the solute carrier family 11 member 1

C. Negrete-gonzález, B. Oliva-ramírez, and L. Baranda, González-Amaro, R. The Role of NRAMP1/SLC11A1 Gene Variant D543N (1730G/A) in the Genetic Susceptibility to Develop Rheumatoid Arthritis in the Mexican Mestizo population, Rev. Investig. Clin, vol.69, pp.2017-2022

R. A. Albrecht, A. Garcia-sastre, and S. H. Kleinstein, Interactive Big Data Resource to Elucidate Human Immune Pathways and Diseases, Immunity, vol.43, pp.605-614, 2015.

G. Dey, A. Jaimovich, S. R. Collins, A. Seki, and T. Meyer, Systematic Discovery of Human Gene Function and Principles of Modular Organization through Phylogenetic Profiling. Cell Rep, pp.993-1006, 2015.