A. Tauber, Timeline: Metchnikoff and the phagocytosis theory, Nature Reviews Molecular Cell Biology, vol.4, issue.11, pp.897-901, 2003.
DOI : 10.1038/nrm1244

M. Sieweke and J. Allen, Beyond Stem Cells: Self-Renewal of Differentiated Macrophages, Science, vol.342, issue.6161, 2013.
DOI : 10.1126/science.1242974

C. Huber and G. Stingl, Macrophages in the Regulation of Immunity, Haematol Blood Transfus, vol.27, pp.31-38, 1981.
DOI : 10.1007/978-3-642-81696-3_4

E. Unanue, D. Beller, J. Calderon, J. Kiely, and M. Stadecker, Regulation of immunity and inflammation by mediators from macrophages, Am J Pathol, vol.85, issue.2, pp.465-78, 1976.

C. Dinarello, Historical insights into cytokines, European Journal of Immunology, vol.15, issue.S1, pp.34-45, 2007.
DOI : 10.1002/eji.200737772

H. Bernheim and M. Kluger, Fever and antipyresis in the lizard Dipsosaurus dorsalis, Am J Physiol, vol.231, issue.1, pp.198-203, 1976.

K. Huynh, J. Kay, J. Stow, and S. Grinstein, Fusion, Fission, and Secretion During Phagocytosis, Physiology, vol.22, issue.6, pp.366-72, 2007.
DOI : 10.1152/physiol.00028.2007

S. Biswas and A. Mantovani, Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm, Nature Immunology, vol.313, issue.10, pp.889-96, 1937.
DOI : 10.1016/j.ccr.2009.06.017

J. Stow and R. Murray, Intracellular trafficking and secretion of inflammatory cytokines, Cytokine & Growth Factor Reviews, vol.24, issue.3, pp.227-266, 2013.
DOI : 10.1016/j.cytogfr.2013.04.001

J. Stow, A. Manderson, R. Murray, M. Fukuda, and A. Descoteaux, SNAREing immunity: the role of SNAREs in the immune system, Nature Reviews Immunology, vol.175, issue.12, pp.919-291737, 2006.
DOI : 10.1038/nri1980

B. Beutler, The role of tumor necrosis factor in health and disease, J Rheumatol, vol.57, pp.16-21, 1999.

E. Carswell, L. Old, R. Kassel, S. Green, N. Fiore et al., An endotoxin-induced serum factor that causes necrosis of tumors., Proceedings of the National Academy of Sciences, vol.72, issue.9, pp.3666-70, 1975.
DOI : 10.1073/pnas.72.9.3666

G. Griffin, G. Newton, M. Tarrio, D. Bu, E. Maganto-garcia et al., IL-17 and TNF-?? Sustain Neutrophil Recruitment during Inflammation through Synergistic Effects on Endothelial Activation, The Journal of Immunology, vol.188, issue.12, pp.6287-99, 2012.
DOI : 10.4049/jimmunol.1200385

S. Vieira, H. Lemos, R. Grespan, M. Napimoga, D. Dal-secco et al., A crucial role for TNF-?? in mediating neutrophil influx induced by endogenously generated or exogenous chemokines, KC/CXCL1 and LIX/CXCL5, British Journal of Pharmacology, vol.8, issue.3, pp.779-89, 2009.
DOI : 10.1111/j.1476-5381.2009.00367.x

S. Sozzani, M. Abbracchio, V. Annese, S. Danese, D. Pita et al., Chronic inflammatory diseases: Do immunological patterns drive the choice of biotechnology drugs? A critical review, Autoimmunity, vol.29, issue.5, pp.287-306, 2014.
DOI : 10.1111/bjd.12331

R. Murray, J. Kay, D. Sangermani, and J. Stow, A Role for the Phagosome in Cytokine Secretion, Science, vol.310, issue.5753, pp.3101492-3101497, 2005.
DOI : 10.1126/science.1120225

J. Pagan, F. Wylie, S. Joseph, C. Widberg, N. Bryant et al., The t-SNARE Syntaxin 4 Is Regulated during Macrophage Activation to Function in Membrane Traffic and Cytokine Secretion, Current Biology, vol.13, issue.2, pp.156-60, 2003.
DOI : 10.1016/S0960-9822(03)00006-X

R. Murray, F. Wylie, T. Khromykh, D. Hume, and J. Stow, Syntaxin 6 and Vti1b Form a Novel SNARE Complex, Which Is Up-regulated in Activated Macrophages to Facilitate Exocytosis of Tumor Necrosis Factor-??, Journal of Biological Chemistry, vol.280, issue.11, pp.10478-83, 2005.
DOI : 10.1074/jbc.M414420200

A. Stanley, C. Wong, M. Micaroni, J. Venturato, T. Khromykh et al., The Rho GTPase Rac1 is required for recycling endosome-mediated secretion of TNF in macrophages, Immunology and Cell Biology, vol.439, issue.3, pp.275-86, 2014.
DOI : 10.1016/S0960-9822(03)00006-X

R. Black, C. Rauch, C. Kozlosky, J. Peschon, J. Slack et al., A metalloproteinase disintegrin that releases tumour-necrosis factor-?? from cells, Nature, vol.385, issue.6618, pp.729-3310385729, 1038.
DOI : 10.1038/385729a0

A. Duque, G. Fukuda, M. Turco, S. Stäger, S. Descoteaux et al., Promastigotes Induce Cytokine Secretion in Macrophages through the Degradation of Synaptotagmin XI, The Journal of Immunology, vol.193, issue.5, pp.2363-72, 2014.
DOI : 10.4049/jimmunol.1303043

URL : https://hal.archives-ouvertes.fr/hal-01178774

D. Baram, R. Adachi, O. Medalia, M. Tuvim, B. Dickey et al., -triggered Exocytosis of Lysosomes in Mast Cells, The Journal of Experimental Medicine, vol.94, issue.10, pp.1649-58, 1999.
DOI : 10.1006/cimm.1994.1272

URL : https://hal.archives-ouvertes.fr/hal-00603255

C. Czibener, N. Sherer, S. Becker, M. Pypaert, E. Hui et al., and synaptotagmin VII???dependent delivery of lysosomal membrane to nascent phagosomes, The Journal of Cell Biology, vol.247, issue.7, pp.997-1007, 2006.
DOI : 10.1073/pnas.81.17.5430

A. Vinet, M. Fukuda, and A. Descoteaux, The Exocytosis Regulator Synaptotagmin V Controls Phagocytosis in Macrophages, The Journal of Immunology, vol.181, issue.8, pp.5289-95, 2008.
DOI : 10.4049/jimmunol.181.8.5289

T. Südhof, Calcium Control of Neurotransmitter Release, Cold Spring Harbor Perspectives in Biology, vol.4, issue.1, 2012.
DOI : 10.1101/cshperspect.a011353

Z. Wang and E. Chapman, Rat and Drosophila Synaptotagmin 4 Have Opposite Effects during SNARE-catalyzed Membrane Fusion, Journal of Biological Chemistry, vol.285, issue.40, pp.30759-66, 2010.
DOI : 10.1074/jbc.M110.137745

A. Milochau, V. Lagree, M. Benassy, S. Chaignepain, J. Papin et al., Synaptotagmin 11 interacts with components of the RNA-induced silencing complex RISC in clonal pancreatic ??-cells, FEBS Letters, vol.32, issue.14, pp.2217-2239, 2014.
DOI : 10.1016/j.febslet.2014.05.031

S. Ben-sasson, J. Hu-li, J. Quiel, S. Cauchetaux, M. Ratner et al., IL-1 acts directly on CD4 T cells to enhance their antigen-driven expansion and differentiation, Proceedings of the National Academy of Sciences, vol.106, issue.17, pp.7119-7143, 2009.
DOI : 10.1073/pnas.0902745106

Y. Carmi, E. Voronov, S. Dotan, N. Lahat, M. Rahat et al., The Role of Macrophage-Derived IL-1 in Induction and Maintenance of Angiogenesis, The Journal of Immunology, vol.183, issue.7, pp.4705-4719, 2009.
DOI : 10.4049/jimmunol.0901511

A. Yazdi, G. Guarda, N. Riteau, S. Drexler, A. Tardivel et al., Nanoparticles activate the NLR pyrin domain containing 3 (Nlrp3) inflammasome and cause pulmonary inflammation through release of IL-1?? and IL-1??, Proceedings of the National Academy of Sciences, vol.107, issue.45, pp.19449-54, 2010.
DOI : 10.1073/pnas.1008155107

C. Chen, H. Kono, D. Golenbock, G. Reed, S. Akira et al., Identification of a key pathway required for the sterile inflammatory response triggered by dying cells, Nature Medicine, vol.116, issue.7, pp.851-857, 1603.
DOI : 10.1038/nm1603

B. Ravikumar, M. Futter, L. Jahreiss, V. Korolchuk, M. Lichtenberg et al., Mammalian macroautophagy at a glance, Journal of Cell Science, vol.122, issue.11, pp.1707-1718, 2009.
DOI : 10.1242/jcs.031773

J. Duran, C. Anjard, C. Stefan, W. Loomis, and V. Malhotra, Unconventional secretion of Acb1 is mediated by autophagosomes, The Journal of Cell Biology, vol.106, issue.4, pp.527-563, 2010.
DOI : 10.1091/mbc.E07-12-1292

N. Dupont, S. Jiang, M. Pilli, W. Ornatowski, D. Bhattacharya et al., Autophagy-based unconventional secretory pathway for extracellular delivery of IL-1??, The EMBO Journal, vol.469, issue.23, pp.4701-4712, 2011.
DOI : 10.1038/emboj.2011.398

S. Jiang, N. Dupont, E. Castillo, and V. Deretic, Secretory versus Degradative Autophagy: Unconventional Secretion of Inflammatory Mediators, Journal of Innate Immunity, vol.5, issue.5, pp.471-480, 2013.
DOI : 10.1159/000346707

D. Brough and N. Rothwell, Caspase-1-dependent processing of pro-interleukin-1beta is cytosolic and precedes cell death, Journal of Cell Science, vol.120, issue.5, pp.772-81, 2007.
DOI : 10.1242/jcs.03377

V. Marty, C. Medina, C. Combe, P. Parnet, and T. Amedee, -stimulated and lipopolysaccharide-primed mouse Schwann cells, Glia, vol.451, issue.4, pp.511-520, 2005.
DOI : 10.1002/glia.20138

URL : https://hal.archives-ouvertes.fr/hal-00857496

Y. Qu, L. Franchi, G. Nunez, and G. Dubyak, Nonclassical IL-1?? Secretion Stimulated by P2X7 Receptors Is Dependent on Inflammasome Activation and Correlated with Exosome Release in Murine Macrophages, The Journal of Immunology, vol.179, issue.3, pp.1913-1938, 1913.
DOI : 10.4049/jimmunol.179.3.1913

N. Nishimoto and T. Kishimoto, Inhibition of IL-6 for the treatment of inflammatory diseases, Current Opinion in Pharmacology, vol.4, issue.4, pp.386-91, 2004.
DOI : 10.1016/j.coph.2004.03.005

T. Barkhausen, T. Tschernig, P. Rosenstiel, M. Van-griensven, R. Vonberg et al., Selective blockade of interleukin-6 trans-signaling improves survival in a murine polymicrobial sepsis model*, Critical Care Medicine, vol.39, issue.6, pp.1407-1420, 2011.
DOI : 10.1097/CCM.0b013e318211ff56

S. Hurst, T. Wilkinson, R. Mcloughlin, S. Jones, S. Horiuchi et al., IL-6 and Its Soluble Receptor Orchestrate a Temporal Switch in the Pattern of Leukocyte Recruitment Seen during Acute Inflammation, Immunity, vol.14, issue.6, pp.705-719, 2001.
DOI : 10.1016/S1074-7613(01)00151-0

J. Scheller, A. Chalaris, D. Schmidt-arras, R. , and S. , The pro-and antiinflammatory properties of the cytokine interleukin-6, Biochim Biophys Acta, vol.1813, issue.5, 2011.

V. Matthews, T. Allen, S. Risis, M. Chan, D. Henstridge et al., Interleukin-6-deficient mice develop hepatic inflammation and systemic insulin resistance, Diabetologia, vol.46, issue.Suppl 2, pp.2431-2472, 2010.
DOI : 10.1007/s00125-010-1865-y

A. Manderson, J. Kay, L. Hammond, D. Brown, and J. Stow, Subcompartments of the macrophage recycling endosome direct the differential secretion of IL-6 and TNF??, The Journal of Cell Biology, vol.276, issue.1, pp.57-69, 2007.
DOI : 10.1242/jcs.00893

S. Dorman and S. Holland, Interferon-?? and interleukin-12 pathway defects and human disease, Cytokine & Growth Factor Reviews, vol.11, issue.4, pp.321-3310, 2000.
DOI : 10.1016/S1359-6101(00)00010-1

A. Croxford, P. Kulig, and B. Becher, IL-12-and IL-23 in health and disease, Cytokine & Growth Factor Reviews, vol.25, issue.4, pp.415-436, 2014.
DOI : 10.1016/j.cytogfr.2014.07.017

K. Wang, D. Frank, and J. Ritz, Interleukin-2 enhances the response of natural killer cells to interleukin-12 through up-regulation of the interleukin-12 receptor and STAT4, Blood, issue.10, pp.953183-90, 2000.

E. Duitman, Z. Orinska, and S. Bulfone-paus, Mechanisms of cytokine secretion: A portfolio of distinct pathways allows flexibility in cytokine activity, European Journal of Cell Biology, vol.90, issue.6-7, pp.90-96, 2011.
DOI : 10.1016/j.ejcb.2011.01.010

M. Logan, P. Lacy, S. Odemuyiwa, M. Steward, F. Davoine et al., A critical role for vesicle-associated membrane protein-7 in exocytosis from human eosinophils and neutrophils, Allergy, vol.155, issue.6, pp.777-84, 2006.
DOI : 10.1038/sj.emboj.7600427

F. Mollinedo, J. Calafat, H. Janssen, B. Martín-martín, J. Canchado et al., Combinatorial SNARE Complexes Modulate the Secretion of Cytoplasmic Granules in Human Neutrophils, The Journal of Immunology, vol.177, issue.5, pp.2831-2872, 2006.
DOI : 10.4049/jimmunol.177.5.2831

J. Pulecio, J. Petrovic, F. Prete, G. Chiaruttini, A. Lennon-dumenil et al., Cdc42-mediated MTOC polarization in dendritic cells controls targeted delivery of cytokines at the immune synapse, The Journal of Experimental Medicine, vol.2, issue.12, pp.2719-2751, 2010.
DOI : 10.1126/science.1099153

S. Gatti, J. Beck, G. Fantuzzi, T. Bartfai, and C. Dinarello, Effect of interleukin-18 on mouse core body temperature, American Journal of Physiology - Regulatory, Integrative and Comparative Physiology, vol.282, issue.3, pp.702-711, 2001.
DOI : 10.1152/ajpregu.00393.2001

J. Lee, S. Kim, E. Lewis, T. Azam, L. Reznikov et al., Differences in signaling pathways by IL-1?? and IL-18, Proceedings of the National Academy of Sciences, vol.101, issue.23, pp.8815-8835, 2004.
DOI : 10.1073/pnas.0402800101

S. Vanden-eijnden, S. Goriely, D. Wit, D. Willems, F. Goldman et al., IL-23 up-regulates IL-10 and induces IL-17 synthesis by polyclonally activated naive T?cells in human, European Journal of Immunology, vol.166, issue.2, pp.469-75, 2005.
DOI : 10.1002/eji.200425677

H. Yoshida, S. Hamano, G. Senaldi, T. Covey, R. Faggioni et al., WSX-1 Is Required for the Initiation of Th1 Responses and Resistance to L. major Infection, Immunity, vol.15, issue.4, pp.569-78, 2001.
DOI : 10.1016/S1074-7613(01)00206-0

J. Stumhofer, A. Laurence, E. Wilson, E. Huang, C. Tato et al., Interleukin 27 negatively regulates the development of interleukin 17???producing T helper cells during chronic inflammation of the central nervous system, Nature Immunology, vol.68, issue.9, pp.937-4510, 1038.
DOI : 10.1038/ni1376

D. Mosser and X. Zhang, Interleukin-10: new perspectives on an old cytokine, Immunological Reviews, vol.226, issue.1, pp.205-223, 2008.
DOI : 10.1111/j.1600-065X.2008.00706.x

D. Fiorentino, A. Zlotnik, T. Mosmann, M. Howard, O. Garra et al., IL-10 inhibits cytokine production by activated macrophages, J Immunol, vol.147, issue.11, pp.3815-3837, 1991.

S. Chadban, G. Tesch, R. Foti, H. Lan, R. Atkins et al., Interleukin-10 differentially modulates MHC class II expression by mesangial cells and macrophages in vitro and in vivo, Immunology, vol.154, issue.1, pp.72-80, 1998.
DOI : 10.1046/j.1365-2567.1998.00487.x

T. Defrance, B. Vanbervliet, F. Briere, I. Durand, F. Rousset et al., Interleukin 10 and transforming growth factor beta cooperate to induce anti-CD40-activated naive human B cells to secrete immunoglobulin A, Journal of Experimental Medicine, vol.175, issue.3, pp.671-82, 1992.
DOI : 10.1084/jem.175.3.671

F. Rousset, E. Garcia, T. Defrance, C. Peronne, N. Vezzio et al., Interleukin 10 is a potent growth and differentiation factor for activated human B lymphocytes., Proceedings of the National Academy of Sciences, vol.89, issue.5, pp.1890-1893, 1992.
DOI : 10.1073/pnas.89.5.1890

I. Oswald, T. Wynn, A. Sher, and S. James, Interleukin 10 inhibits macrophage microbicidal activity by blocking the endogenous production of tumor necrosis factor alpha required as a costimulatory factor for interferon gamma-induced activation., Proceedings of the National Academy of Sciences, vol.89, issue.18, pp.8676-80, 1992.
DOI : 10.1073/pnas.89.18.8676

F. Cunha, S. Moncada, and F. Liew, Interleukin-10 (IL-10) inhibits the induction of nitric oxide synthase by interferon-gamma in murine macrophages, Biochem Biophys Res Commun, vol.18292, issue.3, pp.1155-1164, 1992.

F. Varzaneh, B. Keller, S. Unger, A. Aghamohammadi, K. Warnatz et al., Cytokines in Common Variable Immunodeficiency as Signs of Immune Dysregulation and Potential Therapeutic Targets ??? A Review of the Current Knowledge, Journal of Clinical Immunology, vol.174, issue.1, pp.524-567, 2014.
DOI : 10.1007/s10875-014-0053-0

A. Stanley, Z. Lieu, A. Wall, J. Venturato, T. Khromykh et al., Recycling endosome-dependent and -independent mechanisms for IL-10 secretion in LPS-activated macrophages, Journal of Leukocyte Biology, vol.92, issue.6, pp.1227-1266, 2012.
DOI : 10.1189/jlb.0412191

M. Travis and D. Sheppard, TGF-?? Activation and Function in Immunity, Annual Review of Immunology, vol.32, issue.1, pp.51-8210, 2014.
DOI : 10.1146/annurev-immunol-032713-120257

C. Becker, M. Fantini, C. Schramm, H. Lehr, S. Wirtz et al., TGF-?? Suppresses Tumor Progression in Colon Cancer by Inhibition of IL-6 trans-Signaling, Immunity, vol.21, issue.4, pp.491-501, 2004.
DOI : 10.1016/j.immuni.2004.07.020

S. Josefowicz, L. Lu, and A. Rudensky, Regulatory T Cells: Mechanisms of Differentiation and Function, Annual Review of Immunology, vol.30, issue.1, pp.531-64, 2012.
DOI : 10.1146/annurev.immunol.25.022106.141623

P. Gleizes, J. Munger, I. Nunes, J. Harpel, R. Mazzieri et al., TGF‐β Latency: Biological Significance and Mechanisms of Activation, Stem Cells, vol.133, issue.3, pp.190-197, 1997.
DOI : 10.1002/stem.150190

I. Comerford and S. Mccoll, Mini-review series: focus on chemokines, Immunology and Cell Biology, vol.89, issue.2, pp.183-187, 2011.
DOI : 10.1038/icb.2010.164

B. Moser, I. Clark-lewis, R. Zwahlen, and M. Baggiolini, Neutrophil-activating properties of the melanoma growth-stimulatory activity, Journal of Experimental Medicine, vol.171, issue.5, pp.1797-802, 1990.
DOI : 10.1084/jem.171.5.1797

L. Pelus and S. Fukuda, Peripheral blood stem cell mobilization, Critical Reviews in Oncology/Hematology, vol.43, issue.3, 2006.
DOI : 10.1016/S1040-8428(01)00202-5

C. Addison, D. Arenberg, S. Morris, Y. Xue, M. Burdick et al., The CXC Chemokine, Monokine Induced by Interferon-gamma, Inhibits Non-Small Cell Lung Carcinoma Tumor Growth and Metastasis, Human Gene Therapy, vol.11, issue.2, pp.247-6110, 2000.
DOI : 10.1089/10430340050015996

T. Donlon, A. Krensky, M. Wallace, F. Collins, M. Lovett et al., Localization of a human T-cell-specific gene, RANTES (D17S136E), to chromosome 17q11.2-q12, Genomics, vol.6, issue.3, pp.548-53, 1990.
DOI : 10.1016/0888-7543(90)90485-D

A. Maghazachi, A. Aoukaty, and T. Schall, CC chemokines induce the generation of killer cells from CD56+ cells, European Journal of Immunology, vol.24, issue.2, pp.315-324, 1996.
DOI : 10.1002/eji.1830260207

C. Han, T. Chen, M. Yang, N. Li, H. Liu et al., Human SCAMP5, a Novel Secretory Carrier Membrane Protein, Facilitates Calcium-Triggered Cytokine Secretion by Interaction with SNARE Machinery, The Journal of Immunology, vol.182, issue.5, pp.2986-96, 2009.
DOI : 10.4049/jimmunol.0802002

M. Gouwy, S. Struyf, J. Catusse, P. Proost, and J. Van-damme, Synergy between proinflammatory ligands of G protein-coupled receptors in neutrophil activation and migration, Journal of Leukocyte Biology, vol.76, issue.1, pp.185-94, 2004.
DOI : 10.1189/jlb.1003479

S. Starckx, P. Van-den-steen, A. Wuyts, J. Van-damme, and G. Opdenakker, Neutrophil Gelatinase B and Chemokines in Leukocytosis and Stem Cell Mobilization, Leukemia & Lymphoma, vol.88, issue.2, pp.233-4110, 1080.
DOI : 10.1084/jem.188.3.475

J. Van-damme, S. Struyf, and G. Opdenakker, Chemokine???protease interactions in cancer, Seminars in Cancer Biology, vol.14, issue.3, pp.201-209, 2004.
DOI : 10.1016/j.semcancer.2003.10.007

K. Gijsbers, G. Van-assche, S. Joossens, S. Struyf, P. Proost et al., CXCR1-binding chemokines in inflammatory bowel diseases: down-regulated IL-8/CXCL8 production by leukocytes in Crohn's disease and selective GCP-2/CXCL6 expression in inflamed intestinal tissue, European Journal of Immunology, vol.34, issue.7, pp.1992-2000, 2004.
DOI : 10.1002/eji.200324807

J. Rosenblum, N. Shimoda, A. Schenk, H. Zhang, D. Kish et al., CXC Chemokine Ligand (CXCL) 9 and CXCL10 Are Antagonistic Costimulation Molecules during the Priming of Alloreactive T Cell Effectors, The Journal of Immunology, vol.184, issue.7, pp.3450-60, 2010.
DOI : 10.4049/jimmunol.0903831

E. Keeley, B. Mehrad, and R. Strieter, Chemokines as Mediators of Neovascularization, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.28, issue.11, pp.1928-1964, 2008.
DOI : 10.1161/ATVBAHA.108.162925

J. Dufour, M. Dziejman, M. Liu, J. Leung, T. Lane et al., IFN-??-Inducible Protein 10 (IP-10; CXCL10)-Deficient Mice Reveal a Role for IP-10 in Effector T Cell Generation and Trafficking, The Journal of Immunology, vol.168, issue.7, pp.3195-204, 2002.
DOI : 10.4049/jimmunol.168.7.3195

K. Cole, C. Strick, T. Paradis, K. Ogborne, M. Loetscher et al., Interferon???inducible T Cell Alpha Chemoattractant (I-TAC): A Novel Non-ELR CXC Chemokine with Potent Activity on Activated T Cells through Selective High Affinity Binding to CXCR3, The Journal of Experimental Medicine, vol.159, issue.12, pp.2009-2030, 1998.
DOI : 10.1016/0165-5728(95)00108-E

U. Gundra, N. Girgis, D. Ruckerl, S. Jenkins, L. Ward et al., Alternatively activated macrophages derived from monocytes and tissue macrophages are phenotypically and functionally distinct, Blood, vol.123, issue.20, pp.110-132, 2014.
DOI : 10.1182/blood-2013-08-520619

W. Zhang, W. Xu, and S. Xiong, Blockade of Notch1 Signaling Alleviates Murine Lupus via Blunting Macrophage Activation and M2b Polarization, The Journal of Immunology, vol.184, issue.11, pp.6465-78, 2010.
DOI : 10.4049/jimmunol.0904016

J. Lu, Q. Cao, D. Zheng, Y. Sun, C. Wang et al., Discrete functions of M2a and M2c macrophage subsets determine their relative efficacy in treating chronic kidney disease, Kidney International, vol.84, issue.4, pp.745-55, 2013.
DOI : 10.1038/ki.2013.135

M. Weng, D. Huntley, I. Huang, O. Foye-jackson, L. Wang et al., Alternatively Activated Macrophages in Intestinal Helminth Infection: Effects on Concurrent Bacterial Colitis, The Journal of Immunology, vol.179, issue.7, pp.4721-4752, 2007.
DOI : 10.4049/jimmunol.179.7.4721

N. Hao, M. Lü, Y. Fan, Y. Cao, Z. Zhang et al., Macrophages in Tumor Microenvironments and the Progression of Tumors, Clinical and Developmental Immunology, vol.63, issue.22, p.948098, 2012.
DOI : 10.1158/1078-0432.CCR-08-1608

S. Ong, Y. Tan, O. Beretta, D. Jiang, W. Yeap et al., Macrophages in human colorectal cancer are pro-inflammatory and prime T cells towards an anti-tumour type-1 inflammatory response, European Journal of Immunology, vol.34, issue.1, pp.89-100, 2012.
DOI : 10.1002/eji.201141825

S. Schoppmann, P. Birner, J. Stockl, R. Kalt, R. Ullrich et al., Tumor-Associated Macrophages Express Lymphatic Endothelial Growth Factors and Are Related to Peritumoral Lymphangiogenesis, The American Journal of Pathology, vol.161, issue.3, pp.947-56, 2002.
DOI : 10.1016/S0002-9440(10)64255-1

L. Sibley, Invasion and intracellular survival by protozoan parasites, Immunological Reviews, vol.78, issue.Suppl, pp.72-91, 2011.
DOI : 10.1111/j.1600-065X.2010.00990.x

R. Flannagan, G. Cosio, and S. Grinstein, Antimicrobial mechanisms of phagocytes and bacterial evasion strategies, Nature Reviews Microbiology, vol.69, issue.5, pp.355-66, 2009.
DOI : 10.1038/nrmicro2128

D. Walsh, F. Portaels, and W. Meyers, Buruli Ulcer: Advances in Understanding Mycobacterium ulcerans Infection, Dermatologic Clinics, vol.29, issue.1, 2011.
DOI : 10.1016/j.det.2010.09.006

K. George, L. Pascopella, D. Welty, and P. Small, A Mycobacterium ulcerans Toxin, Mycolactone, Causes Apoptosis in Guinea Pig Ulcers and Tissue Culture Cells, Infection and Immunity, vol.68, issue.2, pp.877-83, 2000.
DOI : 10.1128/IAI.68.2.877-883.2000

K. George, D. Chatterjee, G. Gunawardana, D. Welty, J. Hayman et al., Mycolactone: A Polyketide Toxin from Mycobacterium ulcerans Required for Virulence, Science, vol.283, issue.5403, pp.854-861, 1999.
DOI : 10.1126/science.283.5403.854

C. Demangel, T. Stinear, and S. Cole, Buruli ulcer: reductive evolution enhances pathogenicity of Mycobacterium ulcerans, Nature Reviews Microbiology, vol.4, issue.1, pp.50-60, 2009.
DOI : 10.1093/molbev/msm092

B. Hall and R. Simmonds, virulence factor mycolactone underlying the cell death and immunosuppression seen in Buruli ulcer, Biochemical Society Transactions, vol.11, issue.1, pp.177-83, 2014.
DOI : 10.1002/cbic.200400339

R. Simmonds, F. Lali, T. Smallie, P. Small, and B. Foxwell, Mycolactone Inhibits Monocyte Cytokine Production by a Posttranscriptional Mechanism, The Journal of Immunology, vol.182, issue.4, pp.2194-202, 2009.
DOI : 10.4049/jimmunol.0802294

E. Torrado, S. Adusumilli, A. Fraga, P. Small, A. Castro et al., Mycolactone-Mediated Inhibition of Tumor Necrosis Factor Production by Macrophages Infected with Mycobacterium ulcerans Has Implications for the Control of Infection, Infection and Immunity, vol.75, issue.8, pp.3979-88, 2007.
DOI : 10.1128/IAI.00290-07

B. Hall, K. Hill, M. Mckenna, J. Ogbechi, S. High et al., The Pathogenic Mechanism of the Mycobacterium ulcerans Virulence Factor, Mycolactone, Depends on Blockade of Protein Translocation into the ER, PLoS Pathogens, vol.103, issue.4, 2014.
DOI : 10.1371/journal.ppat.1004061.s004

J. Alvar, I. Velez, C. Bern, M. Herrero, P. Desjeux et al., Leishmaniasis Worldwide and Global Estimates of Its Incidence, PLoS ONE, vol.83, issue.30, 2012.
DOI : 10.1371/journal.pone.0035671.s101

L. Kedzierski, Leishmaniasis vaccine: Where are we today?, Journal of Global Infectious Diseases, vol.2, issue.2, pp.177-85, 2010.
DOI : 10.4103/0974-777X.62881

N. Moradin and A. Descoteaux, Leishmania promastigotes: building a safe niche within macrophages, Frontiers in Cellular and Infection Microbiology, vol.2, 2012.
DOI : 10.3389/fcimb.2012.00121

R. Lodge and A. Descoteaux, Leishmania Invasion and Phagosome Biogenesis, Subcell Biochem, vol.47, pp.174-81, 2008.
DOI : 10.1007/978-0-387-78267-6_14

F. Real, P. Florentino, L. Reis, E. Ramos-sanchez, P. Veras et al., amastigotes is mediated by immunomodulatory LAMP-rich parasitophorous extrusions, Cellular Microbiology, vol.10, issue.Part 9, pp.1549-64, 2014.
DOI : 10.1111/cmi.12311

C. Yao, Major Surface Protease of Trypanosomatids: One Size Fits All?, Infection and Immunity, vol.78, issue.1, pp.22-31, 2010.
DOI : 10.1128/IAI.00776-09

P. Joshi, B. Kelly, S. Kamhawi, D. Sacks, and W. Mcmaster, Targeted gene deletion in Leishmania major identifies leishmanolysin (GP63) as a virulence factor, Molecular and Biochemical Parasitology, vol.120, issue.1, pp.33-40, 2002.
DOI : 10.1016/S0166-6851(01)00432-7

D. Matheoud, N. Moradin, A. Bellemare-pelletier, M. Shio, W. Hong et al., Leishmania Evades Host Immunity by Inhibiting Antigen Cross-Presentation through Direct Cleavage of the SNARE VAMP8, Cell Host & Microbe, vol.14, issue.1, pp.15-25, 2013.
DOI : 10.1016/j.chom.2013.06.003

URL : https://hal.archives-ouvertes.fr/pasteur-01131970

M. Gómez, I. Contreras, M. Halle, M. Tremblay, R. Mcmaster et al., Leishmania GP63 Alters Host Signaling Through Cleavage-Activated Protein Tyrosine Phosphatases, Science Signaling, vol.2, issue.90, 2009.
DOI : 10.1126/scisignal.2000213

M. Jaramillo, M. Gómez, O. Larsson, M. Shio, I. Topisirovic et al., Leishmania Repression of Host Translation through mTOR Cleavage Is Required for Parasite Survival and Infection, Cell Host & Microbe, vol.9, issue.4, pp.331-372, 2011.
DOI : 10.1016/j.chom.2011.03.008

M. Hallé, M. Gómez, M. Stuible, H. Shimizu, W. Mcmaster et al., The Leishmania Surface Protease GP63 Cleaves Multiple Intracellular Proteins and Actively Participates in p38 Mitogen-activated Protein Kinase Inactivation, Journal of Biological Chemistry, vol.284, issue.11, pp.6893-908, 2009.
DOI : 10.1074/jbc.M805861200

I. Contreras, M. Gómez, O. Nguyen, M. Shio, R. Mcmaster et al., Leishmania-Induced Inactivation of the Macrophage Transcription Factor AP-1 Is Mediated by the Parasite Metalloprotease GP63, PLoS Pathogens, vol.1253, issue.10, 2010.
DOI : 10.1371/journal.ppat.1001148.s008

J. Ghosh, M. Bose, S. Roy, and S. Bhattacharyya, Leishmania donovani Targets Dicer1 to Downregulate miR-122, Lower Serum Cholesterol, and Facilitate Murine Liver Infection, Cell Host & Microbe, vol.13, issue.3, pp.277-88, 2013.
DOI : 10.1016/j.chom.2013.02.005

A. Descoteaux, N. Moradin, A. Duque, and G. , Leishmania Dices Away Cholesterol for Survival, Cell Host & Microbe, vol.13, issue.3, pp.245-252, 2013.
DOI : 10.1016/j.chom.2013.02.018

URL : https://hal.archives-ouvertes.fr/pasteur-01130855

A. Arena, A. Capozza, D. Delfino, and D. Iannello, Production of TNF? and interleukin 6 by differentiated U937 cells infected with Leishmania major, New Microbiol, vol.20, issue.3, pp.233-273, 1997.

C. Matte and M. Olivier, Induced Cellular Recruitment during the Early Inflammatory Response: Modulation of Proinflammatory Mediators, The Journal of Infectious Diseases, vol.185, issue.5, pp.673-81, 2002.
DOI : 10.1086/339260

N. Lapara, . Iii, and B. Kelly, Suppression of LPS-induced inflammatory responses in macrophages infected with Leishmania, Journal of Inflammation, vol.7, issue.1, pp.8-10, 2010.
DOI : 10.1186/1476-9255-7-8

U. Wenzel, E. Bank, C. Florian, S. Forster, N. Zimara et al., Leishmania major parasite stage-dependent host cell invasion and immune evasion, The FASEB Journal, vol.26, issue.1, pp.29-39, 2012.
DOI : 10.1096/fj.11-184895

M. Karam, H. Hamdan, A. Chedid, N. Bodman-smith, K. Eales-reynolds et al., Leishmania major: Low infection dose causes short-lived hyperalgesia and cytokines upregulation in mice, Experimental Parasitology, vol.113, issue.3, pp.168-73, 2006.
DOI : 10.1016/j.exppara.2006.01.003

M. Jones, L. Quinton, B. Simms, M. Lupa, M. Kogan et al., Pneumonia, The Journal of Infectious Diseases, vol.193, issue.3, pp.360-369, 2006.
DOI : 10.1086/499312

P. Biswas, F. Delfanti, S. Bernasconi, M. Mengozzi, M. Cota et al., Interleukin-6 induces monocyte chemotactic protein-1 in peripheral blood mononuclear cells and in the U937 cell line, Blood, vol.91, issue.1, pp.258-65, 1998.

Y. Belkaid, K. Hoffmann, S. Mendez, S. Kamhawi, M. Udey et al., in the Skin after Healing and the Therapeutic Potential of Anti???IL-10 Receptor Antibody for Sterile Cure, The Journal of Experimental Medicine, vol.62, issue.10, pp.1497-506, 2001.
DOI : 10.1046/j.1365-3083.2000.00724.x

M. Kane and D. Mosser, The Role of IL-10 in Promoting Disease Progression in Leishmaniasis, The Journal of Immunology, vol.166, issue.2, pp.1141-1148, 2001.
DOI : 10.4049/jimmunol.166.2.1141

F. Ribeiro-gomes, E. Roma, M. Carneiro, N. Doria, D. Sacks et al., Site-Dependent Recruitment of Inflammatory Cells Determines the Effective Dose of Leishmania major, Infection and Immunity, vol.82, issue.7, pp.2713-2740, 2014.
DOI : 10.1128/IAI.01600-13

S. Tiwananthagorn, K. Iwabuchi, M. Ato, T. Sakurai, H. Kato et al., Involvement of CD4+ Foxp3+ Regulatory T Cells in Persistence of Leishmania donovani in the Liver of Alymphoplastic aly/aly Mice, PLoS Neglected Tropical Diseases, vol.6, issue.8, 2012.
DOI : 10.1371/journal.pntd.0001798.s004

K. Silva, M. De-andrade, L. Melo, J. Perosso, R. Vasconcelos et al., CD4+FOXP3+ cells produce IL-10 in the spleens of dogs with visceral leishmaniasis, Veterinary Parasitology, vol.202, issue.3-4, pp.313-321, 2014.
DOI : 10.1016/j.vetpar.2014.03.010