, Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990-2013: a systematic analysis for the Global Burden of Disease Study, GBD 2013 Mortality and Causes of Death Collaborators, vol.385, pp.117-171, 2013.

M. F. Tosi, Innate immune responses to infection, J. Allergy Clin. Immunol, vol.116, issue.2, pp.241-249, 2005.

J. M. Zhang and J. An, Cytokines, inflammation, and pain, Int. Anesthesiol. Clin, vol.45, issue.2, pp.27-37, 2007.

X. Zhang, L. Majlessi, E. Deriaud, C. Leclerc, and R. Lo-man, Coactivation of Syk kinase and MyD88 adaptor protein pathways by bacteria promotes regulatory properties of neutrophils, Immunity, vol.31, issue.5, pp.761-771, 2009.

K. Lim, Y. M. Hyun, K. Lambert-emo, T. Capece, S. Bae et al., Neutrophil trails guide influenza-specific CD8(+) T cells in the airways, Science, vol.349, issue.6252, p.4352, 2015.

A. W. Stadnyk, Intestinal epithelial cells as a source of inflammatory cytokines and chemokines, Can. J. Gastroenterol, vol.16, issue.4, pp.241-246, 2002.

L. Eckmann, M. F. Kagnoff, and J. Fierer, Epithelial cells secrete the chemokine interleukin-8 in response to bacterial entry, Infect. Immun, vol.61, issue.11, pp.4569-4574, 1993.

P. A. Gonzalez, L. J. Carreno, C. A. Figueroa, and A. M. Kalergis, Modulation of immunological synapse by membrane-bound and soluble ligands, Cytokine Growth Factor Rev, vol.18, issue.1-2, pp.19-31, 2007.

C. A. Dinarello, Historical insights into cytokines, Eur. J. Immunol, vol.37, pp.34-45, 2007.

H. F. Penaloza, P. A. Nieto, N. Munoz-durango, F. J. Salazar-echegarai, J. Torres et al., Interleukin-10 plays a key role in the modulation of neutrophils recruitment and lung inflammation during infection by Streptococcus pneumoniae, Immunology, vol.146, issue.1, pp.100-112, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-02146494

C. T. Ng and M. B. Oldstone, Infected CD8alpha-dendritic cells are the predominant source of IL-10 during establishment of persistent viral infection, Proc. Natl. Acad. Sci. U. S. A, vol.109, issue.35, pp.14116-14121, 2012.

S. Naundorf, M. Schroder, C. Hoflich, N. Suman, H. D. Volk et al., IL-10 interferes directly with TCR-induced IFN-gamma but not IL-17 production in memory T cells, Eur. J. Immunol, vol.39, issue.4, pp.1066-1077, 2009.

A. M. O'farrell, Y. Liu, K. W. Moore, and A. L. Mui, IL-10 inhibits macrophage activation and proliferation by distinct signaling mechanisms: evidence for Stat3-dependent and -independent pathways, EMBO J, vol.17, issue.4, pp.1006-1018, 1998.

P. M. Dang, C. Elbim, J. C. Marie, M. Chiandotto, M. A. Gougerot-pocidalo et al., Anti-inflammatory effect of interleukin-10 on human neutrophil respiratory burst involves inhibition of GM-CSF-induced p47PHOX phosphorylation through a decrease in ERK1/2 activity, FASEB J, vol.20, issue.9, pp.1504-1506, 2006.

K. Itoh and S. Hirohata, The role of IL-10 in human B cell activation, proliferation, and differentiation, J. Immunol, vol.154, issue.9, pp.4341-4350, 1995.

K. Taga and G. Tosato, IL-10 inhibits human T cell proliferation and IL-2 production, J. Immunol, vol.148, issue.4, pp.1143-1148, 1992.

N. K. Petty, N. L. Ben-zakour, M. Stanton-cook, E. Skippington, M. Totsika et al., Global dissemination of a multidrug resistant Escherichia coliclone, Proc. Natl. Acad. Sci. U. S. A, vol.111, issue.15, pp.5694-5699, 2014.

L. S. Munoz-price, L. Poirel, R. A. Bonomo, M. J. Schwaber, G. L. Daikos et al., Clinical epidemiology of the global expansion of Klebsiella pneumoniae carbapenemases, Lancet Infect. Dis, vol.13, issue.9, pp.785-796, 2013.

B. Rowe, L. R. Ward, and E. J. Threlfall, Multidrug-resistant Salmonella typhi: a worldwide epidemic, Clin. Infect. Dis, vol.24, pp.106-115, 1997.

S. M. Bueno, S. Riquelme, C. A. Riedel, and A. M. Kalergis, Mechanisms used by virulent Salmonella to impair dendritic cell function and evade adaptive immunity, Immunology, vol.137, issue.1, pp.28-36, 2012.

A. Gupta, A. Kaul, A. G. Tsolaki, U. Kishore, and S. Bhakta, Mycobacterium tuberculosis: immune evasion, latency and reactivation, Immunobiology, vol.217, issue.3, pp.363-374, 2012.

A. Kadioglu, J. N. Weiser, J. C. Paton, and P. W. Andrew, The role of Streptococcus pneumoniae virulence factors in host respiratory colonization and disease, Nat. Rev. Microbiol, vol.6, issue.4, pp.288-301, 2008.

T. J. Foster, Immune evasion by staphylococci, Nat. Rev. Microbiol, vol.3, issue.12, pp.948-958, 2005.

M. J. Greenberger, R. M. Strieter, S. L. Kunkel, J. M. Danforth, R. E. Goodman et al., Neutralization of IL-10 increases survival in a murine model of Klebsiella pneumonia, J. Immunol, vol.155, issue.2, pp.722-729, 1995.

T. Van-der-poll, A. Marchant, C. V. Keogh, M. Goldman, and S. F. Lowry, Interleukin-10 impairs host defense in murine pneumococcal pneumonia, J. Infect. Dis, vol.174, issue.5, pp.994-1000, 1996.

R. Kuhn, J. Lohler, D. Rennick, K. Rajewsky, and W. Muller, Interleukin-10-deficient mice develop chronic enterocolitis, Cell, vol.75, issue.2, pp.263-274, 1993.

W. J. Dai, G. Kohler, and F. Brombacher, Both innate and acquired immunity to Listeria monocytogenes infection are increased in IL-10-deficient mice, J. Immunol, vol.158, issue.5, pp.2259-2267, 1997.

J. F. Chmiel, M. W. Konstan, J. E. Knesebeck, J. B. Hilliard, T. L. Bonfield et al., IL-10 attenuates excessive inflammation in chronic Pseudomonas infection in mice, Am. J. Respir. Crit. Care Med, vol.160, issue.6, pp.2040-2047, 1999.

M. E. Sewnath, D. P. Olszyna, R. Birjmohun, F. J. Kate, D. J. Gouma et al., IL-10-deficient mice demonstrate multiple organ failure and increased mortality during Escherichia coli peritonitis despite an accelerated bacterial clearance, J. Immunol, vol.166, issue.10, pp.6323-6331, 2001.

D. W. Metzger, S. L. Salmon, and G. Kirimanjeswara, Differing effects of interleukin-10 on cutaneous and pulmonary Francisella tularensis live vaccine strain infection, Infect. Immun, vol.81, issue.6, pp.2022-2027, 2013.

S. L. Poe, M. Arora, T. B. Oriss, M. Yarlagadda, K. Isse et al., STAT1-regulated lung MDSC-like cells produce IL-10 and efferocytose apoptotic neutrophils with relevance in resolution of bacterial pneumonia, Mucosal Immunol, vol.6, issue.1, pp.189-199, 2013.

H. Bouabe, Cytokine reporter mice: the special case of IL-10, Scand. J. Immunol, vol.75, issue.6, pp.553-567, 2012.

K. Josephson, N. J. Logsdon, and M. R. Walter, Crystal structure of the IL-10/IL-10R1 complex reveals a shared receptor binding site, Immunity, vol.15, issue.1, pp.35-46, 2001.

C. M. Hedrich and J. H. Bream, Cell type-specific regulation of IL-10 expression in inflammation and disease, Immunol. Res, vol.47, issue.1-3, pp.185-206, 2010.

A. Boonstra, R. Rajsbaum, M. Holman, R. Marques, C. Asselin-paturel et al., Macrophages and myeloid dendritic cells, but not plasmacytoid dendritic cells, produce IL-10 in response to MyD88-and TRIF-dependent TLR signals, and TLR-independent signals, J. Immunol, vol.177, issue.11, pp.7551-7558, 2006.
URL : https://hal.archives-ouvertes.fr/pasteur-00362493

N. T. Funderburg, J. K. Jadlowsky, M. M. Lederman, Z. Feng, A. Weinberg et al., The Toll-like receptor 1/2 agonists Pam(3) CSK(4) and human betadefensin-3 differentially induce interleukin-10 and nuclear factor-kappaB signalling patterns in human monocytes, Immunology, vol.134, issue.2, pp.151-160, 2011.

N. C. Rogers, E. C. Slack, A. D. Edwards, M. A. Nolte, O. Schulz et al., Sykdependent cytokine induction by Dectin-1 reveals a novel pattern recognition pathway for C type lectins, Immunity, vol.22, issue.4, pp.507-517, 2005.

M. Matsumoto, T. Tanaka, T. Kaisho, H. Sanjo, N. G. Copeland et al., A novel LPS-inducible C-type lectin is a transcriptional target of NF-IL6 in macrophages, J. Immunol, vol.163, issue.9, pp.5039-5048, 1999.

T. B. Geijtenbeek, S. J. Van-vliet, E. A. Koppel, M. Sanchez-hernandez, C. M. Vandenbroucke-grauls et al., Mycobacteria target DC-SIGN to suppress dendritic cell function, J. Exp. Med, vol.197, issue.1, pp.7-17, 2003.

S. I. Gringhuis, J. Dunnen, M. Litjens, B. Van-het, Y. Hof et al., C-type lectin DC-SIGN modulates Toll-like receptor signaling via Raf-1 kinase-dependent acetylation of transcription factor NF-kappaB, Immunity, vol.26, issue.5, pp.605-616, 2007.

E. Y. Chang, B. Guo, S. E. Doyle, and G. Cheng, Cutting edge: involvement of the type I IFN production and signaling pathway in lipopolysaccharide-induced IL-10 production, J. Immunol, vol.178, issue.11, pp.6705-6709, 2007.

F. W. Mcnab, J. Ewbank, A. Howes, L. Moreira-teixeira, A. Martirosyan et al., Type I IFN induces IL-10 production in an IL-27-independent manner and blocks responsiveness to IFN-gamma for production of IL-12 and bacterial killing in Mycobacterium tuberculosisinfected macrophages, J. Immunol, vol.193, issue.7, pp.3600-3612, 2014.

S. S. Iyer, A. A. Ghaffari, and G. Cheng, Lipopolysaccharide-mediated IL-10 transcriptional regulation requires sequential induction of type I IFNs and IL-27 in macrophages, J. Immunol, vol.185, issue.11, pp.6599-6607, 2010.

H. Harizi and N. Gualde, Pivotal role of PGE2 and IL-10 in the cross-regulation of dendritic cell-derived inflammatory mediators, Cell Mol. Immunol, vol.3, issue.4, pp.271-277, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00115608

E. Ricciotti and G. A. Fitzgerald, Prostaglandins and inflammation, Arterioscler. Thromb. Vasc. Biol, vol.31, issue.5, pp.986-1000, 2011.

K. F. Mackenzie, K. Clark, S. Naqvi, V. A. Mcguire, G. Noehren et al., PGE(2) induces macrophage IL-10 production and a regulatory-like phenotype via a protein kinase A-SIK-CRTC3 pathway, J. Immunol, vol.190, issue.2, pp.565-577, 2013.

G. Heine, U. Niesner, H. D. Chang, A. Steinmeyer, U. Zugel et al., Worm, 1,25-dihydroxyvitamin D(3) promotes IL-10 production in human B cells, Eur. J. Immunol, vol.38, issue.8, pp.2210-2218, 2008.

G. Bakdash, T. M. Van-capel, L. M. Mason, M. L. Kapsenberg, E. C. De et al., Vitamin D3 metabolite calcidiol primes human dendritic cells to promote the development of immunomodulatory IL-10-producing T cells, Vaccine, vol.32, issue.47, pp.6294-6302, 2014.

L. Adorini, G. Penna, N. Giarratana, A. Roncari, S. Amuchastegui et al., Dendritic cells as key targets for immunomodulation by Vitamin D receptor ligands, J. Steroid Biochem. Mol. Biol, vol.89, issue.90, pp.437-441, 2004.

A. Brosbol-ravnborg, B. Bundgaard, and P. Hollsberg, Synergy between vitamin D (3) and Toll-like receptor agonists regulates human dendritic cell response during maturation, Clin. Dev. Immunol, vol.2013, p.807971, 2013.

G. Noel, Q. Wang, S. Schwemberger, C. Hanson, N. Giacalone et al., Neutrophils, not monocyte/macrophages, are the major splenic source of postburn IL-10, Shock, vol.36, issue.2, pp.149-155, 2011.

A. M. Piccinini and K. S. Midwood, DAMPening inflammation by modulating TLR signalling, Mediators Inflamm, 2010.
DOI : 10.1155/2010/672395

URL : http://downloads.hindawi.com/journals/mi/2010/672395.pdf

S. H. Im, A. Hueber, S. Monticelli, K. H. Kang, and A. Rao, Chromatin-level regulation of the IL10 gene in T cells, J. Biol. Chem, vol.279, issue.45, pp.46818-46825, 2004.

M. Saraiva, J. R. Christensen, A. V. Tsytsykova, A. E. Goldfeld, S. C. Ley et al., Identification of a macrophage-specific chromatin signature in the IL-10 locus, J. Immunol, vol.175, issue.2, pp.1041-1046, 2005.

L. Larsson, S. Thorbert-mros, L. Rymo, and T. Berglundh, Influence of epigenetic modifications of the interleukin-10 promoter on IL10 gene expression, Eur. J. Oral Sci, vol.120, issue.1, pp.14-20, 2012.

N. Tamassia, M. Zimmermann, M. Castellucci, R. Ostuni, K. Bruderek et al., Cutting edge: an inactive chromatin configuration at the IL-10 locus in human neutrophils, J. Immunol, vol.190, issue.5, pp.1921-1925, 2013.

M. Tone, M. J. Powell, Y. Tone, S. A. Thompson, and H. Waldmann, IL-10 gene expression is controlled by the transcription factors Sp1 and Sp3, J. Immunol, vol.165, issue.1, pp.286-291, 2000.
DOI : 10.4049/jimmunol.165.1.286

URL : http://www.jimmunol.org/content/jimmunol/165/1/286.full.pdf

S. Brenner, S. Prosch, K. Schenke-layland, U. Riese, U. Gausmann et al., cAMP-induced Interleukin-10 promoter activation depends on CCAAT/ enhancer-binding protein expression and monocytic differentiation, J. Biol. Chem, vol.278, issue.8, pp.5597-5604, 2003.
DOI : 10.1074/jbc.m207448200

URL : http://www.jbc.org/content/278/8/5597.full.pdf

F. B. Hickey, C. F. Brereton, and K. H. Mills, Adenylate cycalse toxin of Bordetella pertussis inhibits TLR-induced IRF-1 and IRF-8 activation and IL-12 production and enhances IL-10 through MAPK activation in dendritic cells, J. Leukoc. Biol, vol.84, issue.1, pp.234-243, 2008.

M. Samanta, D. Iwakiri, and K. Takada, Epstein-barr virus-encoded small RNA induces IL-10 through RIG-I-mediated IRF-3 signaling, Oncogene, vol.27, issue.30, pp.4150-4160, 2008.
DOI : 10.1038/onc.2008.75

URL : https://www.nature.com/articles/onc200875.pdf

X. Hu, P. K. Paik, J. Chen, A. Yarilina, L. Kockeritz et al., IFN-gamma suppresses IL-10 production and synergizes with TLR2 by regulating GSK3 and CREB/AP-1 proteins, Immunity, vol.24, issue.5, pp.563-574, 2006.
DOI : 10.1016/j.immuni.2006.02.014

URL : https://doi.org/10.1016/j.immuni.2006.02.014

Y. Alvarez, C. Municio, S. Alonso, M. Sanchez, N. Crespo et al., The induction of IL-10 by zymosan in dendritic cells depends on CREB activation by the coactivators CREB-binding protein and TORC2 and autocrine PGE2, J. Immunol, vol.183, issue.2, pp.1471-1479, 2009.

R. Samarasinghe, P. Tailor, T. Tamura, T. Kaisho, S. Akira et al., Induction of an anti-inflammatory cytokine, IL-10, in dendritic cells after toll-like receptor signaling, J. Interferon Cytokine Res, vol.26, issue.12, pp.893-900, 2006.

J. Shoemaker, M. Saraiva, and A. O'garra, GATA-3 directly remodels the IL-10 locus independently of IL-4 in CD4+ T cells, J. Immunol, vol.176, issue.6, pp.3470-3479, 2006.

J. Xu, Y. Yang, G. Qiu, G. Lal, Z. Wu et al., Ding, c-Maf regulates IL-10 expression during Th17 polarization, J. Immunol, vol.182, issue.10, pp.6226-6236, 2009.
DOI : 10.4049/jimmunol.0900123

URL : http://www.jimmunol.org/content/182/10/6226.full.pdf

L. Apetoh, F. J. Quintana, C. Pot, N. Joller, S. Xiao et al., The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27, Nat. Immunol, vol.11, issue.9, pp.854-861, 2010.

E. Y. Chung, J. Liu, Y. Homma, Y. Zhang, A. Brendolan et al., Interleukin-10 expression in macrophages during phagocytosis of apoptotic cells is mediated by homeodomain proteins Pbx1 and Prep-1, Immunity, vol.27, issue.6, pp.952-964, 2007.

S. Yang, L. Gao, F. Lu, B. Wang, F. Gao et al., Transcription factor myocyte enhancer factor 2D regulates interleukin-10 production in microglia to protect neuronal cells from inflammation-induced death, J. Neuroinflammation, vol.12, p.33, 2015.
DOI : 10.1186/s12974-015-0258-z

URL : https://jneuroinflammation.biomedcentral.com/track/pdf/10.1186/s12974-015-0258-z

M. Riemann, R. Endres, S. Liptay, K. Pfeffer, and R. M. Schmid, The IkappaB protein Bcl-3 negatively regulates transcription of the IL-10 gene in macrophages, J. Immunol, vol.175, issue.6, pp.3560-3568, 2005.

A. Sharma, M. Kumar, J. Aich, M. Hariharan, S. K. Brahmachari et al., Posttranscriptional regulation of interleukin-10 expression by hsamiR-106a, Proc. Natl. Acad. Sci. U. S. A, vol.106, issue.14, pp.5761-5766, 2009.

Y. Liu, Q. Chen, Y. Song, L. Lai, J. Wang et al., MicroRNA-98 negatively regulates IL-10 production and endotoxin tolerance in macrophages after LPS stimulation, FEBS Lett, vol.585, issue.12, pp.1963-1968, 2011.

N. Xie, H. Cui, S. Banerjee, Z. Tan, R. Salomao et al., miR-27a regulates inflammatory response of macrophages by targeting IL-10, J. Immunol, vol.193, issue.1, pp.327-334, 2014.

F. Ma, X. Liu, D. Li, P. Wang, N. Li et al., MicroRNA-466l upregulates IL-10 expression in TLR-triggered macrophages by antagonizing RNA-binding protein tristetraprolin-mediated IL-10 mRNA degradation, J. Immunol, vol.184, issue.11, pp.6053-6059, 2010.

G. Curtale, M. Mirolo, T. A. Renzi, M. Rossato, F. Bazzoni et al., Negative regulation of Toll-like receptor 4 signaling by IL-10-dependent microRNA146b, Proc. Natl. Acad. Sci. U. S. A, vol.110, issue.28, pp.11499-11504, 2013.

C. Tudor, F. P. Marchese, E. Hitti, A. Aubareda, L. Rawlinson et al., The p38 MAPK pathway inhibits tristetraprolin-directed decay of interleukin-10 and pro-inflammatory mediator mRNAs in murine macrophages, FEBS Lett, vol.583, issue.12, pp.1933-1938, 2009.

A. Gaba, S. I. Grivennikov, M. V. Do, D. J. Stumpo, P. J. Blackshear et al., Cutting edge: IL-10-mediated tristetraprolin induction is part of a feedback loop that controls macrophage STAT3 activation and cytokine production, J. Immunol, vol.189, issue.5, pp.2089-2093, 2012.

M. Kubo and Y. Motomura, Transcriptional regulation of the anti-inflammatory cytokine IL-10 in acquired immune cells, Front. Immunol, vol.3, p.275, 2012.

S. V. Kotenko, C. D. Krause, L. S. Izotova, B. P. Pollack, W. Wu et al., Identification and functional characterization of a second chain of the interleukin-10 receptor complex, EMBO J, vol.16, issue.19, pp.5894-5903, 1997.

S. I. Yoon, N. J. Logsdon, F. Sheikh, R. P. Donnelly, and M. R. Walter, Conformational changes mediate interleukin-10 receptor 2 (IL-10R2) binding to IL-10 and assembly of the signaling complex, J. Biol. Chem, vol.281, issue.46, pp.35088-35096, 2006.

K. Wolk, E. Witte, U. Reineke, K. Witte, M. Friedrich et al., Is there an interaction between interleukin-10 and interleukin-22?, Genes Immun, vol.6, issue.1, pp.8-18, 2005.

L. Crepaldi, S. Gasperini, J. A. Lapinet, F. Calzetti, C. Pinardi et al., Up-regulation of IL-10R1 expression is required to render human neutrophils fully responsive to IL-10, J. Immunol, vol.167, issue.4, pp.2312-2322, 2001.

S. Haehling, K. Wolk, C. Hoflich, S. Kunz, B. H. Grunberg et al., Interleukin-10 receptor-1 expression in monocyte-derived antigen-presenting cell populations: dendritic cells partially escape from IL-10's inhibitory mechanisms, Genes Immun, vol.16, issue.1, pp.8-14, 2015.

R. M. Weber-nordt, M. A. Meraz, and R. D. Schreiber, Lipopolysaccharidedependent induction of IL-10 receptor expression on murine fibroblasts, J. Immunol, vol.153, issue.8, pp.3734-3744, 1994.

T. L. Denning, N. A. Campbell, F. Song, R. P. Garofalo, G. R. Klimpel et al., Expression of IL-10 receptors on epithelial cells from the murine small and large intestine, Int. Immunol, vol.12, issue.2, pp.133-139, 2000.

K. Wolk, S. Kunz, E. Witte, M. Friedrich, K. Asadullah et al., IL-22 increases the innate immunity of tissues, Immunity, vol.21, issue.2, pp.241-254, 2004.

F. Sheikh, V. V. Baurin, A. Lewis-antes, N. K. Shah, S. V. Smirnov et al., Cutting edge: IL-26 signals through a novel receptor complex composed of IL-20 receptor 1 and IL-10 receptor 2, J. Immunol, vol.172, issue.4, 2004.

D. S. Finbloom and K. D. Winestock, IL-10 induces the tyrosine phosphorylation of tyk2 and Jak1 and the differential assembly of STAT1 alpha and STAT3 complexes in human T cells and monocytes, J. Immunol, vol.155, issue.3, pp.1079-1090, 1995.

J. Wehinger, F. Gouilleux, B. Groner, J. Finke, R. Mertelsmann et al., IL-10 induces DNA binding activity of three STAT proteins (Stat1, Stat3, and Stat5) and their distinct combinatorial assembly in the promoters of selected genes, FEBS Lett, vol.394, issue.3, pp.365-370, 1996.

A. N. Yilma, S. R. Singh, S. J. Fairley, M. A. Taha, and V. A. Dennis, The antiinflammatory cytokine, interleukin-10, inhibits inflammatory mediators in human epithelial cells and mouse macrophages exposed to live and UVinactivated Chlamydia trachomatis, Mediators Inflamm, p.520174, 2012.

T. D. Smedt, M. Van-mechelen, G. Becker, J. Urbain, O. Leo et al., Effect of interleukin-10 on dendritic cell maturation and function, Eur. J. Immunol, vol.27, issue.5, pp.1229-1235, 1997.

J. Tryzmel, V. Miskolci, S. Castro-alcaraz, I. Vancurova, and D. Davidson, Interleukin-10 inhibits proinflammatory chemokine release by neutrophils of the newborn without suppression of nuclear factor-kappa B, Pediatr. Res, vol.54, issue.3, pp.382-386, 2003.

L. L. Laichalk, J. M. Danforth, and T. J. Standiford, Interleukin-10 inhibits neutrophil phagocytic and bactericidal activity, FEMS Immunol. Med. Microbiol, vol.15, issue.4, pp.181-187, 1996.

M. J. Scott, J. J. Hoth, M. Turina, D. R. Woods, and W. G. Cheadle, Interleukin-10 suppresses natural killer cell but not natural killer T cell activation during bacterial infection, Cytokine, vol.33, issue.2, pp.79-86, 2006.

M. Lingnau, C. Hoflich, H. D. Volk, R. Sabat, and W. D. Docke, Interleukin-10 enhances the CD14-dependent phagocytosis of bacteria and apoptotic cells by human monocytes, Hum. Immunol, vol.68, issue.9, pp.730-738, 2007.

F. Capsoni, F. Minonzio, A. M. Ongari, V. Carbonelli, A. Galli et al., IL-10 up-regulates human monocyte phagocytosis in the presence of IL-4 and IFNgamma, J. Leukoc. Biol, vol.58, issue.3, pp.351-358, 1995.

D. F. Fiorentino, A. Zlotnik, T. R. Mosmann, M. Howard, and A. O'garra, IL-10 inhibits cytokine production by activated macrophages, J. Immunol, vol.147, issue.11, pp.3815-3822, 1991.

E. Mtairag, S. Chollet-martin, M. Oudghiri, N. Laquay, M. P. Jacob et al., Effects of interleukin-10 on monocyte/endothelial cell adhesion and MMP-9/TIMP-1 secretion, Cardiovasc. Res, vol.49, issue.4, pp.882-890, 2001.

N. Makita, Y. Hizukuri, K. Yamashiro, M. Murakawa, and Y. Hayashi, IL-10 enhances the phenotype of M2 macrophages induced by IL-4 and confers the ability to increase eosinophil migration, Int. Immunol, vol.27, issue.3, pp.131-141, 2015.

S. Corinti, C. Albanesi, A. Sala, S. Pastore, and G. Girolomoni, Regulatory activity of autocrine IL-10 on dendritic cell functions, J. Immunol, vol.166, issue.7, pp.4312-4318, 2001.

S. Bhattacharyya, P. Sen, M. Wallet, B. Long, A. S. Tisch et al., Immunoregulation of dendritic cells by IL-10 is mediated through suppression of the PI3K/Akt pathway and of IkappaB kinase activity, Blood, vol.104, issue.4, pp.1100-1109, 2004.

H. Gary-gouy, J. Harriague, G. Bismuth, C. Platzer, C. Schmitt et al., Human CD5 promotes B-cell survival through stimulation of autocrine IL-10 production, Blood, vol.100, issue.13, pp.4537-4543, 2002.

N. Shparago, P. Zelazowski, L. Jin, T. M. Mcintyre, E. Stuber et al., IL-10 selectively regulates murine Ig isotype switching, Int. Immunol, vol.8, issue.5, pp.781-790, 1996.

F. Malisan, F. Briere, J. M. Bridon, N. Harindranath, F. C. Mills et al., Interleukin-10 induces immunoglobulin G isotype switch recombination in human CD40-activated naive B lymphocytes, J. Exp. Med, vol.183, issue.3, pp.937-947, 1996.

S. G. Tangye, A. Ferguson, D. T. Avery, C. S. Ma, and P. D. Hodgkin, Isotype switching by human B cells is division-associated and regulated by cytokines, J. Immunol, vol.169, issue.8, pp.4298-4306, 2002.

D. Londono, J. Carvajal, K. Strle, K. S. Kim, and D. Cadavid, IL-10 prevents apoptosis of brain endothelium during bacteremia, J. Immunol, vol.186, issue.12, pp.7176-7186, 2011.

M. S. Bharhani, R. Borojevic, S. Basak, E. Ho, P. Zhou et al., IL-10 protects mouse intestinal epithelial cells from Fas-induced apoptosis via modulating Fas expression and altering caspase-8 and FLIP expression, Am. J. Physiol. Gastrointest. Liver Physiol, vol.291, issue.5, pp.820-829, 2006.

P. F. Laterre, G. Garber, H. Levy, R. Wunderink, G. T. Kinasewitz et al., Severe communityacquired pneumonia as a cause of severe sepsis: data from the PROWESS study, Crit. Care Med, vol.33, issue.5, pp.952-961, 2005.

K. L. O'brien, L. J. Wolfson, J. P. Watt, E. Henkle, M. Deloria-knoll et al., Pneumococcal Global Burden of Disease Study, Burden of disease caused by Streptococcus pneumoniae in children younger than 5 years: global estimates, Lancet, vol.374, issue.9693, pp.893-902, 2009.

J. J. Drijkoningen and G. G. Rohde, Pneumococcal infection in adults: burden of disease, Clin. Microbiol. Infect, vol.20, pp.45-51, 2014.

A. E. Williams, R. J. Jose, J. S. Brown, and R. C. Chambers, Enhanced inflammation in aged mice following infection with Streptococcus pneumoniae is associated with decreased IL-10 and augmented chemokine production, Am. J. Physiol. Lung Cell. Mol. Physiol, vol.308, issue.6, pp.539-588, 2015.

S. R. Slight, L. Monin, R. Gopal, L. Avery, M. Davis et al., IL-10 restrains IL-17 to limit lung pathology characteristics following pulmonary infection with Francisella tularensis live vaccine strain, Am. J. Pathol, vol.183, issue.5, pp.1397-1404, 2013.

C. I. Kang, S. H. Kim, H. B. Kim, S. W. Park, Y. J. Choe et al., Pseudomonas aeruginosa bacteremia: risk factors for mortality and influence of delayed receipt of effective antimicrobial therapy on clinical outcome, Clin. Infect. Dis, vol.37, issue.6, pp.745-751, 2003.

T. Sawa, D. B. Corry, M. A. Gropper, M. Ohara, K. Kurahashi et al., IL-10 improves lung injury and survival in Pseudomonas aeruginosa pneumonia, J. Immunol, vol.159, issue.6, pp.2858-2866, 1997.

J. F. Chmiel, M. W. Konstan, A. Saadane, J. E. Krenicky, H. L. Kirchner et al., Prolonged inflammatory response to acute Pseudomonas challenge in interleukin-10 knockout mice, Am. J. Respir. Crit. Care Med, vol.165, issue.8, pp.1176-1181, 2002.

L. Sun, R. F. Guo, M. W. Newstead, T. J. Standiford, D. R. Macariola et al., Effect of IL-10 on neutrophil recruitment and survival after Pseudomonas aeruginosa challenge, Am. J. Respir. Cell Mol. Biol, vol.41, issue.1, pp.76-84, 2009.

I. Smith, Mycobacterium tuberculosis pathogenesis and molecular determinants of virulence, Clin. Microbiol. Rev, vol.16, issue.3, pp.463-496, 2003.

G. L. Beamer, D. K. Flaherty, B. D. Assogba, P. Stromberg, M. Gonzalez-juarrero et al., Interleukin-10 promotes Mycobacterium tuberculosis disease progression in CBA/J mice, J. Immunol, vol.181, issue.8, pp.5545-5550, 2008.

E. Doz, R. Lombard, F. Carreras, D. Buzoni-gatel, and N. Winter, Mycobacteriainfected dendritic cells attract neutrophils that produce IL-10 and specifically shut down Th17 CD4T cells through their IL-10 receptor, J. Immunol, vol.191, issue.7, pp.3818-3826, 2013.

D. M. Higgins, J. Sanchez-campillo, A. G. Rosas-taraco, E. J. Lee, I. M. Orme et al., Lack of IL-10 alters inflammatory and immune responses during pulmonary Mycobacterium tuberculosis infection, Tuberculosis (Edinb.), vol.89, issue.2, pp.149-157, 2009.

P. S. Redford, A. Boonstra, S. Read, J. Pitt, C. Graham et al., Enhanced protection to Mycobacterium tuberculosisinfection in IL-10-deficient mice is accompanied by early and enhanced Th1 responses in the lung, Eur. J. Immunol, vol.40, issue.8, pp.2200-2210, 2010.

C. A. Broberg, M. Palacios, and V. L. Miller, Klebsiella: a long way to go towards understanding this enigmatic jet-setter, vol.6, p.64, 2014.

L. S. Tzouvelekis, A. Markogiannakis, M. Psichogiou, P. T. Tassios, and G. L. Daikos, Carbapenemases in Klebsiella pneumoniae and other Enterobacteriaceae: an evolving crisis of global dimensions, Clin. Microbiol. Rev, vol.25, issue.4, pp.682-707, 2012.

P. Nordmann, G. Cuzon, and T. Naas, The real threat of Klebsiella pneumoniae carbapenemase-producing bacteria, Lancet Infect. Dis, vol.9, issue.4, pp.228-236, 2009.

M. S. Lawlor, S. A. Handley, and V. L. Miller, Comparison of the host responses to wild-type and cpsB mutant Klebsiella pneumoniae infections, Infect. Immun, vol.74, issue.9, pp.5402-5407, 2006.

D. Moranta, V. Regueiro, C. March, E. Llobet, J. Margareto et al., Klebsiella pneumoniae capsule polysaccharide impedes the expression of beta-defensins by airway epithelial cells, Infect. Immun, vol.78, issue.3, pp.1135-1146, 2010.

C. March, D. Moranta, V. Regueiro, E. Llobet, A. Tomas et al., Klebsiella pneumoniae outer membrane protein A is required to prevent the activation of airway epithelial cells, J. Biol. Chem, vol.286, issue.12, pp.9956-9967, 2011.

V. Regueiro, D. Moranta, C. G. Frank, E. Larrarte, J. Margareto et al., Klebsiella pneumoniaesubverts the activation of inflammatory responses in a NOD1-dependent manner, Cell. Microbiol, vol.13, issue.1, pp.135-153, 2011.

L. M. Lery, L. Frangeul, A. Tomas, V. Passet, A. S. Almeida et al., Comparative analysis of Klebsiella pneumoniae genomes identifies a phospholipase D family protein as a novel virulence factor, BMC Biol, vol.12, p.41, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01011838

K. Yoshida, T. Matsumoto, K. Tateda, K. Uchida, S. Tsujimoto et al., Induction of interleukin-10 and down-regulation of cytokine production by Klebsiella pneumoniae capsule in mice with pulmonary infection, J. Med. Microbiol, vol.50, issue.5, pp.456-461, 2001.

J. P. Lavigne, G. Cuzon, C. Combescure, G. Bourg, A. Sotto et al., Virulence of Klebsiella pneumoniae isolates harboring bla KPC-2 carbapenemase gene in a Caenorhabditis elegans model, PLoS One, vol.8, issue.7, p.67847, 2013.

L. S. Tzouvelekis, V. Miriagou, S. D. Kotsakis, K. Spyridopoulou, E. Athanasiou et al., KPC-producing, multidrug-resistant Klebsiella pneumoniae sequence type 258 as a typical opportunistic pathogen, Antimicrob. Agents Chemother, vol.57, issue.10, pp.5144-5146, 2013.

C. D. Paddock, G. N. Sanden, J. D. Cherry, A. A. Gal, C. Langston et al., Pathology and pathogenesis of fatal Bordetella pertussis infection in infants, Clin. Infect. Dis, vol.47, issue.3, pp.328-338, 2008.

M. Ryan, G. Murphy, L. Gothefors, L. Nilsson, J. Storsaeter et al., Bordetella pertussis respiratory infection in children is associated with preferential activation of type 1T helper cells, J. Infect. Dis, vol.175, issue.5, pp.1246-1250, 1997.

S. Gilberg, E. Njamkepo, I. P. Chatelet, H. Partouche, P. Gueirard et al., Evidence of Bordetella pertussis infection in adults presenting with persistent cough in a french area with very high whole-cell vaccine coverage, J. Infect. Dis, vol.186, issue.3, pp.415-418, 2002.

R. Higgs, S. C. Higgins, P. J. Ross, and K. H. Mills, Immunity to the respiratory pathogen Bordetella pertussis, Mucosal Immunol, vol.5, issue.5, pp.485-500, 2012.

S. C. Higgins, E. C. Lavelle, C. Mccann, B. Keogh, E. Mcneela et al., Toll-like receptor 4-mediated innate IL-10 activates antigen-specific regulatory T cells and confers resistance to Bordetella pertussis by inhibiting inflammatory pathology, J. Immunol, vol.171, issue.6, pp.3119-3127, 2003.

V. Dirix, V. Verscheure, T. Goetghebuer, M. Hainaut, A. S. Debrie et al., Monocyte-derived interleukin-10 depresses the Bordetella pertussisspecific gamma interferon response in vaccinated infants, Clin. Vaccine Immunol, vol.16, issue.12, pp.1816-1821, 2009.

D. N. Wolfe, A. T. Karanikas, S. E. Hester, M. J. Kennett, and E. T. Harvill, IL-10 induction by Bordetella parapertussis limits a protective IFN-gamma response, J. Immunol, vol.184, issue.3, pp.1392-1400, 2010.

K. Nagamatsu, A. Kuwae, T. Konaka, S. Nagai, S. Yoshida et al., Bordetella evades the host immune system by inducing IL-10 through a type III effector, J. Exp. Med, vol.206, issue.13, pp.3073-3088, 2009.

M. Perez-fontan and F. Lueiro, Escherichia coli peritonitis in patients undergoing peritoneal dialysis: a serious problem that may get worse, Perit. Dial. Int, vol.26, issue.2, pp.174-177, 2006.

F. Cereto, X. Herranz, E. Moreno, A. Andreu, M. Vergara et al., Role of host and bacterial virulence factors in Escherichia colispontaneous bacterial peritonitis, Eur. J. Gastroenterol. Hepatol, vol.20, issue.9, pp.924-929, 2008.

R. Mittal, I. Gonzalez-gomez, A. Panigrahy, K. Goth, R. Bonnet et al., IL-10 administration reduces PGE-2 levels and promotes CR3-mediated clearance of Escherichia coli K1 by phagocytes in meningitis, J. Exp. Med, vol.207, issue.6, pp.1307-1319, 2010.

H. Naruse, T. Hisamatsu, Y. Yamauchi, J. E. Chang, K. Matsuoka et al., Intracellular bacteria recognition contributes to maximal interleukin (IL)-12 production by IL-10-deficient macrophages, Clin. Exp. Immunol, vol.164, issue.1, pp.137-144, 2011.

V. L. Atluri, M. N. Xavier, M. F. Jong, A. B. Hartigh, and R. M. Tsolis, Interactions of the human pathogenic Brucella species with their hosts, Annu. Rev. Microbiol, vol.65, pp.523-541, 2011.

E. Barquero-calvo, E. Chaves-olarte, D. S. Weiss, C. Guzman-verri, C. Chacondiaz et al., Brucella abortus uses a stealthy strategy to avoid activation of the innate immune system during the onset of infection, PLoS One, vol.2, issue.7, p.631, 2007.

A. Svetic, Y. C. Jian, P. Lu, F. D. Finkelman, and W. C. Gause, Brucella abortus induces a novel cytokine gene expression pattern characterized by elevated IL-10 and IFN-gamma in CD4+ T cells, Int. Immunol, vol.5, issue.8, pp.877-883, 1993.

E. A. Murphy, J. Sathiyaseelan, M. A. Parent, B. Zou, and C. L. Baldwin, Interferongamma is crucial for surviving a Brucella abortus infection in both resistant C57BL/6 and susceptible BALB/c mice, Immunology, vol.103, issue.4, pp.511-518, 2001.

D. M. Fernandes and C. L. Baldwin, Interleukin-10 downregulates protective immunity to Brucella abortus, Infect. Immun, vol.63, issue.3, pp.1130-1133, 1995.

P. P. Corsetti, L. A. De-almeida, N. B. Carvalho, V. Azevedo, T. M. Silva et al., Lack of endogenous IL-10 enhances production of proinflammatory cytokines and leads to Brucella abortus clearance in mice, PLoS One, vol.8, issue.9, p.74729, 2013.

M. N. Xavier, M. G. Winter, A. M. Spees, K. Nguyen, V. L. Atluri et al., CD4+ T cell-derived IL-10 promotes Brucella abortus persistence via modulation of macrophage function, PLoS Pathog, vol.9, issue.6, p.1003454, 2013.

E. Barquero-calvo, A. Martirosyan, D. Ordonez-rueda, V. Arce-gorvel, A. Alfaro-alarcon et al., Neutrophils exert a suppressive effect on Th1 responses to intracellular pathogen Brucella abortus, vol.9, p.1003167, 2013.

M. A. Gordon, Invasive nontyphoidal Salmonella disease: epidemiology, pathogenesis and diagnosis, Curr. Opin. Infect. Dis, vol.24, issue.5, pp.484-489, 2011.

R. L. Santos, M. Raffatellu, C. L. Bevins, L. G. Adams, C. Tukel et al., Life in the inflamed intestine, Salmonella style, Trends Microbiol, vol.17, issue.11, pp.498-506, 2009.

C. R. Braden, Salmonella enterica serotype Enteritidis and eggs: a national epidemic in the United States, Clin. Infect. Dis, vol.43, issue.4, pp.512-517, 2006.

G. Dougan and S. Baker, Salmonella enterica serovar Typhi and the pathogenesis of typhoid fever, Annu. Rev. Microbiol, vol.68, pp.317-336, 2014.

T. Arai, K. Hiromatsu, H. Nishimura, Y. Kimura, N. Kobayashi et al., Effects of in vivo administration of anti-IL-10 monoclonal antibody on the host defence mechanism against murine Salmonella infection, Immunology, vol.85, issue.3, pp.381-388, 1995.

K. S. Lee, E. S. Jeong, S. H. Heo, J. H. Seo, D. G. Jeong et al., IL-10 suppresses bactericidal response of macrophages against Salmonella Typhimurium, J. Microbiol, vol.49, issue.6, pp.1050-1053, 2011.

K. Uchiya, E. A. Groisman, and T. Nikai, Involvement of Salmonella pathogenicity island 2 in the up-regulation of interleukin-10 expression in macrophages: role of protein kinase A signal pathway, Infect. Immun, vol.72, issue.4, pp.1964-1973, 2004.

K. Uchiya and T. Nikai, Salmonella pathogenicity island 2-dependent expression of suppressor of cytokine signaling 3 in macrophages, Infect. Immun, vol.73, issue.9, pp.5587-5594, 2005.

T. Nguyen, N. Robinson, S. E. Allison, B. K. Coombes, S. Sad et al., IL-10 produced by trophoblast cells inhibits phagosome maturation leading to profound intracellular proliferation of Salmonella enterica Typhimurium, Placenta, vol.34, issue.9, pp.765-774, 2013.

K. L. Lokken, J. P. Mooney, B. P. Butler, M. N. Xavier, J. Y. Chau et al., Malaria parasite infection compromises control of concurrent systemic non-typhoidal Salmonella infection via IL-10-mediated alteration of myeloid cell function, PLoS Pathog, vol.10, issue.5, p.1004049, 2014.

J. A. Vazquez-boland, M. Kuhn, P. Berche, T. Chakraborty, G. Dominguezbernal et al., Listeria pathogenesis and molecular virulence determinants, Clin. Microbiol. Rev, vol.14, issue.3, pp.584-640, 2001.

K. E. Foulds, M. J. Rotte, and R. A. Seder, IL-10 is required for optimal CD8T cell memory following Listeria monocytogenes infection, J. Immunol, vol.177, issue.4, pp.2565-2574, 2006.

B. Pasche, S. Kalaydjiev, T. J. Franz, E. Kremmer, V. Gailus-durner et al., Sex-dependent susceptibility to Listeria monocytogenes infection is mediated by differential interleukin-10 production, Infect. Immun, vol.73, issue.9, pp.5952-5960, 2005.

K. A. Rhodes, E. M. Andrew, D. J. Newton, D. Tramonti, and S. R. Carding, A subset of IL-10-producing gammadelta T cells protect the liver from Listeria-elicited, CD8(+) T cell-mediated injury, Eur. J. Immunol, vol.38, issue.8, pp.2274-2283, 2008.

C. C. Lee and J. T. Kung, Marginal zone B cell is a major source of Il-10 inListeria monocytogenes susceptibility, J. Immunol, vol.189, issue.7, pp.3319-3327, 2012.

C. A. Forestal, M. Malik, S. V. Catlett, A. G. Savitt, J. L. Benach et al., Francisella tularensishas a significant extracellular phase in infected mice, J. Infect. Dis, vol.196, issue.1, pp.134-137, 2007.

V. Cano, C. March, J. L. Insua, N. Aguilo, E. Llobet et al., Klebsiella pneumoniae survives within macrophages by avoiding delivery to lysosomes, Cell. Microbiol, vol.17, issue.11, pp.1537-1560, 2015.

R. L. Friedman, K. Nordensson, L. Wilson, E. T. Akporiaye, and D. E. Yocum, Uptake and intracellular survival of Bordetella pertussis in human macrophages, Infect. Immun, vol.60, issue.11, pp.4578-4585, 1992.

Y. Lamberti, J. Gorgojo, C. Massillo, and M. E. Rodriguez, Bordetella pertussis entry into respiratory epithelial cells and intracellular survival, Pathog. Dis, vol.69, issue.3, pp.194-204, 2013.

J. Zhang, Y. Chen, X. B. Nie, W. H. Wu, H. Zhang et al., Interleukin-10 polymorphisms and tuberculosis susceptibility: a metaanalysis, Int. J. Tuberc. Lung Dis, vol.15, issue.5, pp.594-601, 2011.

P. M. Gallagher, G. Lowe, T. Fitzgerald, A. Bella, C. M. Greene et al., Association of IL-10 polymorphism with severity of illness in community acquired pneumonia, Thorax, vol.58, issue.2, pp.154-156, 2003.

H. Ohman, A. Tiitinen, M. Halttunen, S. Birkelund, G. Christiansen et al., IL-10 polymorphism and cell-mediated immune response to Chlamydia trachomatis, Genes Immun, vol.7, issue.3, pp.243-249, 2006.

X. Gao, J. Chen, Z. Tong, G. Yang, Y. Yao et al., Interleukin-10 promoter gene polymorphisms and susceptibility to tuberculosis: a meta-analysis, PLoS One, vol.10, issue.6, p.127496, 2015.

A. A. Awomoyi, A. Marchant, J. M. Howson, K. P. Mcadam, J. M. Blackwell et al., Interleukin-10, polymorphism in SLC11A1 (formerly NRAMP1), and susceptibility to tuberculosis, J. Infect. Dis, vol.186, issue.12, pp.1808-1814, 2002.

R. G. Westendorp, J. A. Langermans, T. W. Huizinga, A. H. Elouali, C. L. Verweij et al., Genetic influence on cytokine production and fatal meningococcal disease, Lancet, vol.349, issue.9046, pp.170-173, 1997.

C. J. Broers, R. J. Gemke, S. A. Morre, M. E. Weijerman, and A. M. Van-furth, Increased production of interleukin-10 in children with Down syndrome upon ex vivo stimulation with Streptococcus pneumoniae, Pediatr. Res, vol.75, issue.1-1, pp.109-113, 2014.

B. S. Liu, Y. Cao, T. W. Huizinga, D. A. Hafler, and R. E. Toes, TLR-mediated STAT3 and ERK activation controls IL-10 secretion by human B cells, Eur. J. Immunol, vol.44, issue.7, pp.2121-2129, 2014.

L. Durant, W. T. Watford, H. L. Ramos, A. Laurence, G. Vahedi et al., Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis, Immunity, vol.32, issue.5, pp.605-615, 2010.

L. Ysebrant-de-lendonck, S. Tonon, M. Nguyen, P. Vandevenne, I. Welsby et al., Interferon regulatory factor 3 controls interleukin-17 expression in CD8T lymphocytes, Proc. Natl. Acad. Sci. U. S. A, vol.110, issue.34, pp.3189-97, 2013.

A. Fragale, L. Gabriele, E. Stellacci, P. Borghi, E. Perrotti et al., IFN regulatory factor-1 negatively regulates CD4+ CD25+ regulatory T cell differentiation by repressing Foxp3 expression, J. Immunol, vol.181, issue.3, pp.1673-1682, 2008.

X. Ouyang, R. Zhang, J. Yang, Q. Li, L. Qin et al., Transcription factor IRF8 directs a silencing programme for TH17 cell differentiation, Nat. Commun, vol.2, p.314, 2011.

M. Yang, L. Sun, S. Wang, K. H. Ko, H. Xu et al., Novel function of B cell-activating factor in the induction of IL-10-producing regulatory B cells, J. Immunol, vol.184, issue.7, pp.3321-3325, 2010.

Z. Y. Wang, H. Sato, S. Kusam, S. Sehra, L. M. Toney et al., Regulation of IL-10 gene expression in Th2 cells by Jun proteins, J. Immunol, vol.174, issue.4, pp.2098-2105, 2005.

A. Y. Wen, K. M. Sakamoto, and L. S. Miller, The role of the transcription factor CREB in immune function, J. Immunol, vol.185, issue.11, pp.6413-6419, 2010.

S. Cao, J. Liu, M. Chesi, P. L. Bergsagel, I. C. Ho et al., Differential regulation of IL-12 and IL-10 gene expression in macrophages by the basic leucine zipper transcription factor c-Maf fibrosarcoma, J. Immunol, vol.169, issue.10, pp.5715-5725, 2002.

Q. Zhou and S. Amar, Identification of proteins differentially expressed in human monocytes exposed to Porphyromonas gingivalis and its purified components by high-throughput immunoblotting, Infect. Immun, vol.74, issue.2, pp.1204-1214, 2006.

K. Liopeta, S. Boubali, L. Virgilio, G. Thyphronitis, G. Mavrothalassitis et al., cAMP regulates IL-10 production by normal human T lymphocytes at multiple levels: a potential role for MEF2, Mol. Immunol, vol.46, issue.3, pp.345-354, 2009.

J. Herglotz, L. Unrau, F. Hauschildt, M. Fischer, N. Kriebitzsch et al., Essential control of early B-cell development by Mef2 transcription factors, Blood, vol.127, issue.5, pp.572-581, 2016.

D. Kreisel, S. Sugimoto, J. Tietjens, J. Zhu, S. Yamamoto et al., Bcl3 prevents acute inflammatory lung injury in mice by restraining emergency granulopoiesis, J. Clin. Invest, vol.121, issue.1, pp.265-276, 2011.

K. Brocke-heidrich, B. Ge, H. Cvijic, G. Pfeifer, D. Loffler et al., BCL3 is induced by IL-6 via Stat3 binding to intronic enhancer HS4 and represses its own transcription, vol.25, pp.7297-7304, 2006.

H. D. Brightbill, S. E. Plevy, R. L. Modlin, and S. T. Smale, A prominent role for Sp1 during lipopolysaccharide-mediated induction of the IL-10 promoter in macrophages, J. Immunol, vol.164, issue.4, pp.1940-1951, 2000.

L. Larsson, L. Rymo, and T. Berglundh, Sp1 binds to the G allele of the-1087 polymorphism in the IL-10 promoter and promotes IL-10 mRNA transcription and protein production, Genes Immun, vol.11, issue.2, pp.181-187, 2010.

T. R. Hughes, T. S. Tengku-muhammad, S. A. Irvine, and D. P. Ramji, A novel role of Sp1 and Sp3 in the interferon-gamma-mediated suppression of macrophage lipoprotein lipase gene transcription, J. Biol. Chem, vol.277, issue.13, pp.11097-11106, 2002.

J. R. Ward, P. R. Heath, J. W. Catto, M. K. Whyte, M. Milo et al., Regulation of neutrophil senescence by microRNAs, PLoS One, vol.6, issue.1, p.15810, 2011.

T. E. Davis, K. Kis-toth, A. Szanto, and G. C. Tsokos, Glucocorticoids suppress T cell function by up-regulating microRNA-98, Arthritis Rheum, vol.65, issue.7, pp.1882-1890, 2013.

H. H. Pua, D. F. Steiner, S. Patel, J. R. Gonzalez, J. F. Ortiz-carpena et al., MicroRNAs 24 and 27 suppress allergic inflammation and target a network of regulators of t helper 2 cell-associated cytokine production, Immunity, vol.44, issue.4, pp.821-832, 2016.

F. Allantaz, D. T. Cheng, T. Bergauer, P. Ravindran, M. F. Rossier et al., Expression profiling of human immune cell subsets identifies miRNA-mRNA regulatory relationships correlated with cell type specific expression, PLoS One, vol.7, issue.1, p.29979, 2012.

C. Urbich, D. Kaluza, T. Fromel, A. Knau, K. Bennewitz et al., MicroRNA-27a/b controls endothelial cell repulsion and angiogenesis by targeting semaphorin 6A, Blood, vol.119, issue.6, pp.1607-1616, 2012.

L. P. Tan, M. Wang, J. L. Robertus, R. N. Schakel, J. H. Gibcus et al., A. van den Berg, miRNA profiling of B-cell subsets: specific miRNA profile for germinal center B cells with variation between centroblasts and centrocytes, Lab. Invest, vol.89, issue.6, pp.708-716, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01702994