P. Majewski, M. Majchrzak-gorecka, B. Grygier, J. Skrzeczynskamoncznik, O. Osiecka et al., Inhibitors of serine proteases in regulating the production and function of neutrophil extracellular traps, Front Immunol, vol.7, 2016.

J. Diana, Y. Simoni, L. Furio, L. Beaudoin, B. Agerberth et al., Crosstalk between neutrophils, B-1a cells and plasmacytoid dendritic cells initiates autoimmune diabetes, Nat Med, vol.19, pp.65-73, 2013.
DOI : 10.1038/nm.3042

B. Desnues, A. B. Macedo, D. Ordonez-rueda, A. Roussel-queval, B. Malissen et al., The transcriptional repressor Gfi1 prevents lupus autoimmunity by restraining TLR7 signaling, Eur J Immunol, vol.46, pp.2801-2811, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02008644

K. Kessenbrock, M. Krumbholz, U. Schonermarck, W. Back, W. L. Gross et al., Netting neutrophils in autoimmune small-vessel vasculitis, Nat Med, vol.15, pp.623-625, 2009.

M. Vono, A. Lin, A. Norrby-teglund, R. A. Koup, F. Liang et al., Neutrophils acquire the capacity for antigen presentation to memory CD4(+) T cells in vitro and ex vivo, Blood, vol.129, pp.1991-2001, 2017.

I. Puga, M. Cols, and C. M. Barra, B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen, Nat Immunol, vol.13, pp.170-180, 2011.

M. A. Lowes, M. Suarez-farinas, and J. G. Krueger, Immunology of psoriasis, Annu Rev Immunol, vol.32, pp.227-255, 2014.

M. Majchrzak-gorecka, P. Majewski, B. Grygier, K. Murzyn, and J. Cichy, Secretory leukocyte protease inhibitor (SLPI), a multifunctional protein in the host defense response, Cytokine Growth Factor Rev, vol.28, pp.79-93, 2016.

J. Skrzeczynska-moncznik, A. Wlodarczyk, K. Zabieglo, M. Kapinskamrowiecka, E. Marewicz et al., , 2012.

, Secretory leukocyte proteinase inhibitor-competent DNA deposits are potent stimulators of plasmacytoid dendritic cells: implication for psoriasis, J Immunol, vol.189, pp.1611-1617

S. Caielli, S. Athale, and B. Domic, Oxidized mitochondrial nucleoids released by neutrophils drive type I interferon production in human lupus, J Exp Med, vol.213, pp.697-713, 2016.

C. Carmona-rivera and M. J. Kaplan, Low-density granulocytes: a distinct class of neutrophils in systemic autoimmunity, Semin Immunopathol, vol.35, pp.455-463, 2013.

C. Rosales, Neutrophil: a cell with many roles in inflammation or several cell types?, Front Physiol, vol.9, p.113, 2018.

A. M. Bruger, A. Dorhoi, and G. Esendagli, How to measure the immunosuppressive activity of MDSC: assays, problems and potential solutions, Cancer Immunol Immunother, 2018.

L. Ziegler-heitbrock, P. Ancuta, and S. Crowe, Nomenclature of monocytes and dendritic cells in blood, Blood, vol.116, pp.74-80, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00611173

F. Ginhoux and M. Guilliams, Tissue-resident macrophage ontogeny and homeostasis, Immunity, vol.44, pp.439-449, 2016.
DOI : 10.1016/j.immuni.2016.02.024

URL : https://doi.org/10.1016/j.immuni.2016.02.024

M. Morell, N. Varela, and C. Maranon, Myeloid populations in systemic autoimmune diseases, Clin Rev Allergy Immunol, vol.53, pp.198-218, 2017.

A. Amoruso, D. Sola, L. Rossi, J. A. Obeng, L. G. Fresu et al., Relation among anti-rheumatic drug therapy, CD14(+)CD16(+) blood monocytes and disease activity markers (DAS28 and US7 scores) in rheumatoid arthritis: a pilot study, Pharmacol Res, vol.107, pp.308-314, 2016.

B. R. Yoon, S. J. Yoo, Y. Choi, Y. H. Chung, J. Kim et al., Functional phenotype of synovial monocytes modulating inflammatory T-cell responses in rheumatoid arthritis (RA), PloS One, vol.9, p.75, 2014.

C. C. Bain, C. L. Scott, and H. Uronen-hansson, Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors, Mucosal Immunol, vol.6, pp.498-510, 2013.

A. Waschbisch, S. Schroder, and D. Schraudner, Pivotal role for CD16+ monocytes in immune surveillance of the central nervous system, J Immunol, vol.196, pp.1558-1567, 2016.

I. B. Mcinnes and G. Schett, The pathogenesis of rheumatoid arthritis, N Engl J Med, vol.365, pp.2205-2219, 2011.

D. Y. Vogel, E. J. Vereyken, and J. E. Glim, Macrophages in inflammatory multiple sclerosis lesions have an intermediate activation status, J Neuroinflammation, vol.10, p.35, 2013.

A. C. Vos, M. E. Wildenberg, and I. Arijs, Regulatory macrophages induced by infliximab are involved in healing in vivo and in vitro, Inflamm Bowel Dis, vol.18, pp.401-408, 2012.

F. Veglia, M. Perego, and D. Gabrilovich, Myeloid-derived suppressor cells coming of age, Nat Immunol, vol.19, pp.108-119, 2018.

V. Bronte, S. Brandau, and S. H. Chen, Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards, Nat Commun, vol.7, p.12150, 2016.

W. Fujii, E. Ashihara, and H. Hirai, Myeloid-derived suppressor cells play crucial roles in the regulation of mouse collagen-induced arthritis, J Immunol, vol.191, pp.1073-1081, 2013.

M. Ioannou, T. Alissafi, and I. Lazaridis, Crucial role of granulocytic myeloid-derived suppressor cells in the regulation of central nervous system autoimmune disease, J Immunol, vol.188, pp.1136-1146, 2012.

J. Kurko, A. Vida, T. T. Glant, C. R. Scanzello, R. S. Katz et al., Identification of myeloidderived suppressor cells in the synovial fluid of patients with rheumatoid arthritis: a pilot study, BMC Musculoskelet Disord, vol.15, p.281, 2014.

V. Moline-velazquez, H. Cuervo, -. Vila, V. Sol, M. C. Ortega et al., Myeloid-derived suppressor cells limit the inflammation by promoting T lymphocyte apoptosis in the spinal cord of a murine model of multiple sclerosis, Brain Pathol, vol.21, pp.678-691, 2011.

G. Amodio and S. Gregori, Dendritic cells: a doubleedge sword in autoimmune responses, 2012.

G. Amodio, M. Comi, D. Tomasoni, M. E. Gianolini, R. Rizzo et al., Hla-g expression levels influence the tolerogenic activity of human DC-10, Haematologica, vol.100, pp.548-557, 2015.

S. Gregori, D. Tomasoni, V. Pacciani, M. Scirpoli, M. Battaglia et al., Differentiation of type 1 T regulatory cells (Tr1) by tolerogenic DC-10 requires the IL-10-dependent ILT4/HLA-G pathway, Blood, vol.116, pp.935-944, 2010.

G. Amodio, A. Mugione, A. M. Sanchez, P. Vigano, M. Candiani et al., HLA-G expressing DC-10 and CD4(+) T cells accumulate in human decidua during pregnancy, Hum Immunol, vol.74, pp.406-411, 2013.

S. Xia, H. Sha, L. Yang, Y. Ji, S. Ostrand-rosenberg et al., Gr-1+ CD11b+ myeloid-derived suppressor cells suppress inflammation and promote insulin sensitivity in obesity, J Biol Chem, vol.286, pp.23591-23599, 2011.
DOI : 10.1074/jbc.m111.237123

URL : http://europepmc.org/articles/pmc3123122?pdf=render

L. Drujont, L. Carretero-iglesia, L. Bouchet-delbos, G. Beriou, E. Merieau et al., Evaluation of the therapeutic potential of bone marrow-derived myeloid suppressor cell (MDSC) adoptive transfer in mouse models of autoimmunity and allograft rejection, PloS One, vol.9, 2014.

M. R. Garcia, L. Ledgerwood, and Y. Yang, Monocytic suppressive cells mediate cardiovascular transplantation tolerance in mice, J Clin Investig, vol.120, pp.2486-2496, 2010.
DOI : 10.1172/jci41628

URL : http://www.jci.org/articles/view/41628/files/pdf

M. A. Boks, J. R. Kager-groenland, M. S. Haasjes, J. J. Zwaginga, S. M. Van-ham et al., IL-10-generated tolerogenic dendritic cells are optimal for functional regulatory T cell inductiona comparative study of human clinical-applicable DC, Clin Immunol, vol.142, pp.332-342, 2012.

N. Giannoukakis, B. Phillips, D. Finegold, J. Harnaha, and M. Trucco, Phase I (safety) study of autologous tolerogenic dendritic cells in type 1 diabetic patients, Diabetes Care, vol.34, pp.2026-2032, 2011.

H. Benham, H. J. Nel, and S. C. Law, Citrullinated peptide dendritic cell immunotherapy in HLA risk genotype-positive rheumatoid arthritis patients, Sci Transl Med, vol.7, 2015.
DOI : 10.1126/scitranslmed.aaa9301

C. M. Hilkens and J. D. Isaacs, Tolerogenic dendritic cell therapy for rheumatoid arthritis: where are we now?, Clin Exp Immunol, vol.172, pp.148-157, 2013.
DOI : 10.1111/cei.12038

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1111/cei.12038

A. Jauregui-amezaga, R. Cabezon, and A. Ramirez-morros, Intraperitoneal administration of autologous tolerogenic dendritic cells for refractory Crohn's disease: a phase I study, J Crohns Colitis, vol.9, pp.1071-1078, 2015.

J. C. Ochando, N. R. Krieger, and J. S. Bromberg, Direct versus indirect allorecognition: visualization of dendritic cell distribution and interactions during rejection and tolerization, Am J Transplant, vol.6, pp.2488-2496, 2006.

S. Celli, M. L. Albert, and P. Bousso, Visualizing the innate and adaptive immune responses underlying allograft rejection by twophoton microscopy, Nat Med, vol.17, pp.744-749, 2011.
DOI : 10.1038/nm.2376

URL : https://hal.archives-ouvertes.fr/pasteur-01402079

H. Liu, W. Gao, and J. Yuan, Exosomes derived from dendritic cells improve cardiac function via activation of CD4(+) T lymphocytes after myocardial infarction, J Mol Cell Cardiol, vol.91, pp.123-133, 2016.

J. R. Espinosa, K. P. Samy, and A. D. Kirk, Memory T cells in organ transplantation: progress and challenges, Nat Rev Nephrol, vol.12, pp.339-347, 2016.

M. H. Oberbarnscheidt, Q. Zeng, Q. Li, H. Dai, A. L. Williams et al., Non-self recognition by monocytes initiates allograft rejection, J Clin Investig, vol.124, pp.3579-3589, 2014.
DOI : 10.1172/jci74370

URL : http://www.jci.org/articles/view/74370/files/pdf

J. Ochando, P. Conde, and V. Bronte, Monocyte-derived suppressor cells in transplantation, Curr Transplant Rep, vol.2, pp.176-183, 2015.
DOI : 10.1007/s40472-015-0054-9

URL : https://link.springer.com/content/pdf/10.1007%2Fs40472-015-0054-9.pdf

F. K. Swirski, M. Wildgruber, and T. Ueno, Myeloperoxidaserich Ly-6C+ myeloid cells infiltrate allografts and contribute to an imaging signature of organ rejection in mice, J Clin Investig, vol.120, pp.2627-2634, 2010.

P. Conde, M. Rodriguez, and W. Van-der-touw, DC-SIGN(+) macrophages control the induction of transplantation tolerance, Immunity, vol.42, pp.1143-1158, 2015.

J. Ochando and M. S. Braza, Nanoparticle-based modulation and monitoring of antigen-presenting cells in organ transplantation, Front Immunol, vol.8, p.1888, 2017.

J. D. Fisher, A. P. Acharya, and S. R. Little, Micro and nanoparticle drug delivery systems for preventing allotransplant rejection, Clin Immunol, vol.160, pp.24-35, 2015.
DOI : 10.1016/j.clim.2015.04.013

URL : http://europepmc.org/articles/pmc4554802?pdf=render

A. Moreau, B. Alliot-licht, M. C. Cuturi, and G. Blancho, Tolerogenic dendritic cell therapy in organ transplantation, Transpl Int, vol.30, pp.754-764, 2017.
DOI : 10.1111/tri.12889

URL : https://hal.archives-ouvertes.fr/inserm-02150923

A. Moreau, E. Chiffoleau, and G. Beriou, Superiority of bone marrow-derived dendritic cells over monocyte-derived ones for the expansion of regulatory T cells in the macaque, Transplantation, vol.85, pp.1351-1356, 2008.

M. Hill, P. Thebault, and M. Segovia, Cell therapy with autologous tolerogenic dendritic cells induces allograft tolerance through interferon-gamma and epstein-barr virus-induced gene 3, Am J Transplant, vol.11, pp.2036-2045, 2011.
DOI : 10.1111/j.1600-6143.2011.03651.x

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1111/j.1600-6143.2011.03651.x

A. Moreau, M. Hill, and T. P. , Tolerogenic dendritic cells actively inhibit T cells through heme oxygenase-1 in rodents and in nonhuman primates, FASEB J, vol.23, pp.3070-3077, 2009.

J. A. Hutchinson, P. Riquelme, and B. Sawitzki, Cutting edge: immunological consequences and trafficking of human regulatory macrophages administered to renal transplant recipients, J Immunol, vol.187, pp.2072-2078, 2011.

A. Moreau, E. Varey, G. Beriou, M. Hill, L. Bouchet-delbos et al., Tolerogenic dendritic cells and negative vaccination in transplantation: from rodents to clinical trials, Front Immunol, vol.3, 2012.
DOI : 10.3389/fimmu.2012.00218

URL : https://www.frontiersin.org/articles/10.3389/fimmu.2012.00218/pdf

F. Fu, N. L. Thai, Y. Li, L. Lu, A. W. Thomson et al., Second-set rejection of mouse liver allografts is dependent on radiation-sensitive nonparenchymal cells of graft bone marrow origin, Transplantation, vol.61, pp.1228-1233, 1996.

Y. Zhou, J. Shan, Y. Guo, S. Li, D. Long et al., Effects of adoptive transfer of tolerogenic dendritic cells on allograft survival in organ transplantation models: an overview of systematic reviews, J Immunol Res, p.5730674, 2016.

H. Peche, B. Trinite, B. Martinet, M. C. Cuturi, M. B. Ezzelarab et al., Eomesodermin(lo) CTLA4(hi) alloreactive CD8+ memory T cells are associated with prolonged renal transplant survival induced by regulatory dendritic cell, Am J Transplant, vol.5, pp.91-102, 2005.