I. K. Gratz, M. D. Rosenblum, M. M. Maurano, J. S. Paw, H. A. Truong et al., Cutting edge: self-antigen controls the balance between effector and regulatory T cells in peripheral tissues, J Immunol, vol.192, pp.1351-1356, 2014.

X. Luo, W. Yang, and D. Ye, A functional variant in microRNA-146a promoter modulates its expression and confers disease risk for systemic lupus erythematosus, PLoS Genet, vol.7, p.1002128, 2011.

S. N. Kariuki, Y. Ghodke-puranik, and J. M. Dorschner, Genetic analysis of the pathogenic molecular sub-phenotype interferon-a identifies multiple novel loci involved in systemic lupus erythematosus, Genes Immun, vol.16, pp.15-23, 2015.

V. L. Perez, L. Van-parijs, A. Biuckians, X. X. Zheng, T. B. Strom et al., Induction of peripheral T cell tolerance in vivo requires CTLA-4 engagement, Immunity, vol.6, pp.411-428, 1997.

S. Bernatsky, J. F. Boivin, and L. Joseph, Mortality in systemic lupus erythematosus, Arthritis Rheum, vol.54, pp.2550-2557, 2006.

L. J. Carreño, R. Pacheco, M. A. Gutierrez, S. Jacobelli, and A. M. Kalergis, Disease activity in systemic lupus erythematosus is associated with an altered expression of low-affinity Fcc receptors and costimulatory molecules on dendritic cells, Immunology, vol.128, pp.334-375, 2009.

U. S. Gaipl, L. E. Munoz, and G. Grossmayer, Clearance deficiency and systemic lupus erythematosus (SLE), J Autoimmun, vol.28, pp.114-135, 2007.

A. A. Herrada, C. Llanos, and J. P. Mackern-oberti, Haem oxygenase 1 expression is altered in monocytes from patients with systemic lupus erythematosus, Immunology, vol.136, pp.414-438, 2012.

A. Khan, H. Fu, L. A. Tan, J. E. Harper, S. C. Beutelspacher et al., Dendritic cell modification as a route to inhibiting corneal graft rejection by the indirect pathway of allorecognition, Eur J Immunol, vol.43, pp.734-780, 2013.

W. Unger, S. Laban, F. S. Kleijwegt, A. R. Van-der-slik, and B. O. Roep, Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: differential role for PD-L1, Eur J Immunol, vol.39, pp.3147-59, 2009.

L. L. Teichmann, M. L. Ols, M. Kashgarian, B. Reizis, D. H. Kaplan et al., Dendritic cells in lupus are not required for activation of T and B cells but promote their expansion, resulting in tissue damage, Immunity, vol.33, pp.967-78, 2010.

K. Shortman and Y. Liu, Mouse and human dendritic cell subtypes, Nat Rev Immunol, vol.2, pp.151-61, 2002.

K. Santana-de-anda, D. Mart-in, A. E. Monsivais-urenda, M. Salgado-bustamante, R. Gonz-alez-amaro et al., Interferon regulatory factor 3 as key element of the interferon signature in plasmacytoid dendritic cells from systemic lupus erythematosus patients: novel genetic associations in the Mexican mestizo population, Clin Exp Immunol, vol.178, pp.428-465, 2014.

J. J. Hooks, H. M. Moutsopoulos, S. A. Geis, N. I. Stahl, J. L. Decker et al., Immune interferon in the circulation of patients with autoimmune disease, N Engl J Med, vol.301, pp.5-8, 1979.

M. C. Dall'era, P. M. Cardarelli, B. T. Preston, A. Witte, and J. C. Davis, Type I interferon correlates with serological and clinical manifestations of SLE, Ann Rheum Dis, vol.64, pp.1692-1699, 2005.

E. C. Baechler, P. K. Gregersen, and T. W. Behrens, The emerging role of interferon in human systemic lupus erythematosus, Curr Opin Immunol, vol.16, pp.801-808, 2004.

B. P. Tsao, R. M. Cantor, and K. C. Kalunian, Evidence for linkage of a candidate chromosome 1 region to human systemic lupus erythematosus, J Clin Invest, vol.99, pp.725-756, 1997.

B. R-ethi, P. Szatmari, I. Veres, A. Erdôs, E. Nagy et al., SLAM/SLAM interactions inhibit CD40-induced production of inflammatory cytokines in monocyte-derived dendritic cells, Blood, vol.107, pp.2821-2830, 2006.

N. Hagberg, J. Theorell, H. Schlums, M. Eloranta, and Y. T. Bryceson, Systemic lupus erythematosus immune complexes increase the expression of SLAM family members CD319 (CRACC) and CD229 (LY-9) on plasmacytoid dendritic cells and CD319 on CD56dim NK cells, J Immunol, vol.191, pp.2989-98, 2013.

N. Mozaffarian, A. E. Wiedeman, and A. M. Stevens, Active systemic lupus erythematosus is associated with failure of antigen-presenting cells to express programmed death ligand-1, Rheumatology, vol.47, pp.1335-1376, 2008.

H. M. Seitz, T. D. Camenisch, G. Lemke, H. S. Earp, and G. K. Matsushima, Macrophages and dendritic cells use different Axl/Mertk/Tyro3 receptors in clearance of apoptotic cells, J Immunol, vol.178, pp.5635-5677, 2007.

B. Hilliard, G. Zizzo, M. Ulas, M. Linan, J. Schreiter et al., Increased expression of Mer tyrosine kinase in circulating dendritic cells and monocytes of lupus patients: correlations with plasma interferon activity and steroid therapy, Arthritis Res Ther, vol.16, p.76, 2014.

A. Krug, A. Towarowski, and S. Britsch, Toll-like receptor expression reveals CpG DNA as a unique microbial stimulus for plasmacytoid dendritic cells which synergizes with CD40 ligand to induce high amounts of IL-12, Eur J Immunol, vol.31, pp.3026-3063, 2001.

C. F. Yu, W. M. Peng, and J. Oldenburg, Human plasmacytoid dendritic cells support Th17 cell effector function in response to TLR7 ligation, J Immunol, vol.184, pp.1159-67, 2010.

T. Ito, H. Kanzler, O. Duramad, W. Cao, and Y. J. Liu, Specialization, kinetics, and repertoire of type 1 interferon responses by human plasmacytoid predendritic cells, Blood, vol.107, pp.2423-2454, 2006.

C. Guiducci, C. Ghirelli, M. Marloie-provost, T. Matray, R. L. Coffman et al., PI3K is critical for the nuclear translocation of IRF-7 and type I IFN production by human plasmacytoid predendritic cells in response to TLR activation, J Exp Med, vol.205, pp.315-337, 2008.

R. A. Zeuner, D. M. Klinman, G. Illei, C. Yarboro, K. J. Ishii et al., Response of peripheral blood mononuclear cells from lupus patients to stimulation by CpG oligodeoxynucleotides, Rheumatology, vol.42, pp.563-572, 2003.

A. Rodriguez-pla, P. Patel, and H. T. Maecker, IFN priming is necessary but not sufficient to turn on a migratory dendritic cell program in lupus monocytes, J Immunol, vol.192, pp.5586-98, 2014.

L. Morel, C. Mohan, Y. Yu, B. P. Croker, N. Tian et al., Functional dissection of systemic lupus erythematosus using congenic mouse strains, J Immunol, vol.158, pp.6019-6047, 1997.

M. J. Shlomchik, M. P. Madaio, D. Ni, M. Trounstein, and D. Huszar, The role of B cells in lpr/lpr-induced autoimmunity, J Exp Med, vol.180, pp.1295-306, 1994.

P. Pisitkun, J. A. Deane, M. J. Difilippantonio, T. Tarasenko, A. B. Satterthwaite et al.,

, Autoreactive B cell responses to RNA-related antigens due to TLR7 gene duplication, Science, vol.312, pp.1669-72, 2006.

S. Bolland and J. V. Ravetch, Spontaneous autoimmune disease in Fc(c)RIIB-deficient mice results from strain-specific epistasis, Immunity, vol.13, pp.277-85, 2000.

S. J. Kim, Y. R. Zou, J. Goldstein, B. Reizis, and B. Diamond, Tolerogenic function of Blimp-1 in dendritic cells, J Exp Med, vol.208, pp.2193-2202, 2011.

J. Han, H. Zheng, and Y. Cui, Genome-wide association study in a Chinese Han population identifies nine new susceptibility loci for systemic lupus erythematosus, Nat Genet, vol.41, pp.1234-1241, 2009.

S. Parlato, R. Bruni, and P. Fragapane, IFN-a regulates blimp-1 expression via miR-23a and miR-125b in both monocytes-derived DC and pDC, PLoS ONE, vol.8, p.72833, 2013.

A. Bondanza, V. S. Zimmermann, G. Dell'antonio, E. Dal-cin, A. Capobianco et al., Cutting edge: dissociation between autoimmune response and clinical disease after vaccination with dendritic cells, J Immunol, vol.170, pp.24-31, 2003.

L. Ma, K. W. Chan, and T. Nj, Systemic autoimmune disease induced by dendritic cells that have captured necrotic but not apoptotic cells in susceptible mouse strains, Eur J Immunol, vol.35, pp.3364-75, 2005.

M. Georgiev, L. Agle, J. L. Chu, K. B. Elkon, and D. Ashany, Mature dendritic cells readily break tolerance in normal mice but do not lead to disease expression, Arthritis Rheum, vol.52, pp.225-263, 2005.

L. Abeler-d?-orner, C. C. Rieger, and B. Berger, Interferon-a abrogates the suppressive effect of apoptotic cells on dendritic cells in an in vitro model of systemic lupus erythematosus pathogenesis, J Rheumatol, vol.40, pp.1683-96, 2013.

Z. Liu, R. Bethunaickan, W. Huang, U. Lodhi, I. Solano et al., Interferon-a accelerates murine systemic lupus erythematosus in a T cell-dependent manner, Arthritis Rheum, vol.63, pp.219-248, 2011.

S. L. Rowland, J. M. Riggs, S. Gilfillan, M. Bugatti, W. Vermi et al., Early, transient depletion of plasmacytoid dendritic cells ameliorates autoimmunity in a lupus model, J Exp Med, vol.211, pp.1977-91, 2014.

Z. Lian, K. Kikuchi, G. Yang, A. A. Ansari, S. Ikehara et al., Expansion of bone marrow IFN-a-producing dendritic cells in New Zealand black (NZB) mice: high level expression of TLR9 and secretion of IFN-a in NZB bone marrow, J Immunol, vol.173, pp.5283-5292, 2004.

U. Sriram, L. Varghese, H. L. Bennett, N. R. Jog, D. K. Shivers et al., Myeloid dendritic cells from B6.NZM Sle1/Sle2/Sle3 lupus-prone mice express an IFN signature that precedes disease onset, J Immunol, vol.189, pp.80-91, 2012.

R. Baccala, R. Gonzalez-quintial, R. D. Schreiber, B. R. Lawson, D. H. Kono et al., Anti-IFN-a/b receptor antibody treatment ameliorates disease in lupus-predisposed mice, J Immunol, vol.189, pp.5976-84, 2012.

C. Wang, H. Wang, D. Chang, J. Hao, M. Zhao et al., High mobility group box 1 contributes to anti-neutrophil cytoplasmic antibody-induced neutrophils activation through receptor for advanced glycation end products (RAGE) and Toll-like receptor 4, Arthritis Res Ther, vol.17, p.64, 2015.

Z. Wen, L. Xu, X. Chen, W. Xu, Z. Yin et al., Autoantibody induction by DNA-containing immune complexes requires HMGB1 with the TLR2/microRNA-155 pathway, J Immunol, vol.190, pp.5411-5433, 2013.

E. A. Leadbetter, I. R. Rifkin, A. M. Hohlbaum, B. C. Beaudette, and M. J. Shlomchik, MarshakRothstein A. Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors, Nature, vol.416, pp.603-610, 2002.

D. Y. Chen, Y. M. Chen, M. C. Wen, T. Y. Hsieh, W. T. Hung et al., The potential role of Th17 cells and Th17-related cytokines in the pathogenesis of lupus nephritis, Lupus, vol.21, pp.1385-96, 2012.

A. Rana, R. W. Minz, R. Aggarwal, S. Anand, N. Pasricha et al., Gene expression of cytokines (TNF-a, IFN-c), serum profiles of IL-17 and IL-23 in paediatric systemic lupus erythematosus, Lupus, vol.21, pp.1105-1117, 2012.

C. K. Wong, L. C. Lit, L. S. Tam, E. K. Li, P. T. Wong et al., Hyperproduction of IL-23 and IL-17 in patients with systemic lupus erythematosus: implications for Th17-mediated inflammation in auto-immunity, Clin Immunol, vol.127, pp.385-93, 2008.

W. C. Pan, R. M. Chen, Y. C. Shen, C. C. Chen, and Y. F. Ueng, Suppressive effect of tobacco smoke extracts on oral P-glycoprotein function and its impact in smoke-induced insult to oral epidermal cells, Toxicol Lett, vol.185, pp.116-139, 2009.

L. Xu, C. Wang, Y. Zhou, T. Ren, and Z. Wen, CpG oligonucleotides induce the differentiation of CD4+ Th17 cells by triggering plasmacytoid dendritic cells in adoptively cell transfer immunotherapy, Immunol Lett, vol.142, pp.55-63, 2012.

A. Ouabed, F. X. Hubert, D. Chabannes, L. Gautreau, M. Heslan et al., Differential control of T regulatory cell proliferation and suppressive activity by mature plasmacytoid versus conventional spleen dendritic cells, J Immunol, vol.180, pp.5862-70, 2008.
DOI : 10.4049/jimmunol.180.9.5862

URL : http://www.jimmunol.org/content/180/9/5862.full.pdf

M. Lu, S. Yu, W. Xu, B. Gao, and S. Xiong, HMGB1 promotes systemic lupus erythematosus by enhancing macrophage inflammatory response, J Immunol Res, p.12, 2015.
DOI : 10.1155/2015/946748

URL : https://doi.org/10.1155/2015/946748

V. Urbonaviciute, . Bg, and S. Meister, Induction of inflammatory and immune responses by HMGB1-nucleosome complexes: implications for the pathogenesis of SLE, J Exp Med, vol.205, pp.3007-3025, 2008.

P. Scaffidi, T. Misteli, and M. E. Bianchi, Release of chromatin protein HMGB1 by necrotic cells triggers inflammation, Nature, vol.418, pp.191-196, 2002.

V. M. R?-onnefarth, A. Erbacher, T. Lamkemeyer, J. Madlung, A. Nordheim et al., TLR2/TLR4-independent neutrophil activation and recruitment upon endocytosis of nucleosomes reveals a new pathway of innate immunity in systemic lupus erythematosus, J Immunol, vol.177, pp.7740-7749, 2006.

T. K. Means, E. Latz, F. Hayashi, M. R. Murali, D. T. Golenbock et al., Human lupus autoantibody-DNA complexes activate DCs through cooperation of CD32 and TLR9, J Clin Invest, vol.115, pp.407-424, 2005.
DOI : 10.1172/jci200523025

URL : http://www.jci.org/articles/view/23025/files/pdf

S. R. Christensen, M. Kashgarian, L. Alexopoulou, R. A. Flavell, S. Akira et al., Toll-like receptor 9 controls anti-DNA autoantibody production in murine lupus, J Exp Med, vol.202, pp.321-352, 2005.
DOI : 10.1084/jem.20050338

URL : https://hal.archives-ouvertes.fr/hal-00079137

S. R. Christensen, J. Shupe, K. Nickerson, M. Kashgarian, F. Richard et al., Toll-like receptor 7 and TLR9 dictate autoantibody specificity and have opposing inflammatory and regulatory roles in a murine model of lupus, Immunity, vol.25, pp.417-445, 2006.

M. Ehlers, H. Fukuyama, T. L. Mcgaha, A. Aderem, and J. V. Ravetch, TLR9/MyD88 signaling is required for class switching to pathogenic IgG2a and 2b autoantibodies in SLE, J Exp Med, vol.203, pp.553-61, 2006.
DOI : 10.1084/jem.20052438

URL : http://europepmc.org/articles/pmc2118244?pdf=render

A. Lartigue, P. Courville, I. Auquit, A. Franc-ßois, C. Arnoult et al., Role of TLR9 in anti-nucleosome and anti-DNA antibody production in lpr mutation-induced murine lupus, J Immunol, vol.177, pp.1349-54, 2006.

P. Alba, L. Bento, M. J. Cuadrado, Y. Karim, M. F. Tungekar et al., Anti-dsDNA, anti-Sm antibodies, and the lupus anticoagulant: significant factors associated with lupus nephritis, Ann Rheum Dis, vol.62, pp.556-60, 2003.
DOI : 10.1136/ard.62.6.556

URL : https://ard.bmj.com/content/62/6/556.full.pdf

C. M. Lau, C. Broughton, and A. S. Tabor, RNA-associated autoantigens activate B cells by combined B cell antigen receptor/Toll-like receptor 7 engagement, J Exp Med, vol.202, pp.1171-1178, 2005.
DOI : 10.1084/jem.20050630

URL : http://jem.rupress.org/content/202/9/1171.full.pdf

A. Sadanaga, H. Nakashima, M. Akahoshi, K. Masutani, K. Miyake et al., Protection against autoimmune nephritis in MyD88-deficient MRL/lpr mice, Arthritis Rheum, vol.56, pp.1618-1646, 2007.
DOI : 10.1002/art.22571

R. Lande, J. Gregorio, and V. Facchinetti, Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide, Nature, vol.449, pp.564-573, 2007.
DOI : 10.1038/nature06116

Y. Doring, H. D. Manthey, and M. Drechsler, Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis, Circulation, vol.125, pp.1673-83, 2012.

S. Sandgren, A. Wittrup, F. Cheng, M. Jonsson, E. Eklund et al., The human antimicrobial peptide LL-37 transfers extracellular DNA plasmid to the nuclear compartment of mammalian cells via lipid rafts and proteoglycan-dependent endocytosis, J Biol Chem, vol.279, pp.17951-17957, 2004.

Y. J. Hwang, H. J. Jung, M. J. Kim, N. K. Roh, J. W. Jung et al., Serum levels of LL-37 and inflammatory cytokines in plaque and guttate psoriasis, Mediators Inflamm, p.6, 2014.

K. Sacre, L. A. Criswell, and J. M. Mccune, Hydroxychloroquine is associated with impaired interferon-a and tumor necrosis factor-a production by plasmacytoid dendritic cells in systemic lupus erythematosus, Arthritis Res Ther, vol.14, p.155, 2012.

, Immunology, vol.146, pp.497-507

A. Ku-znik, M. Ben-cina, U. Svajger, M. Jeras, B. Rozman et al., Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines, J Immunol, vol.186, pp.4794-804, 2011.

M. Lamphier, W. Zheng, and E. Latz, Novel small molecule inhibitors of TLR7 and TLR9: mechanism of action and efficacy in vivo, Mol Pharmacol, vol.85, pp.429-469, 2014.

Z. M. Bamboat, V. P. Balachandran, L. M. Ocuin, H. Obaid, G. Plitas et al., Tolllike receptor 9 inhibition confers protection from liver ischemia-reperfusion injury, Hepatology, vol.51, pp.621-653, 2010.
DOI : 10.1002/hep.23365

URL : http://europepmc.org/articles/pmc3164814?pdf=render

F. Nimmerjahn and J. V. Ravetch, Fc-receptors as regulators of immunity, Adv Immunol, vol.96, pp.179-204, 2007.

F. Nimmerjahn, P. Bruhns, K. Horiuchi, and J. V. Ravetch, FccRIV: a novel FcR with distinct IgG subclass specificity, Immunity, vol.23, pp.41-51, 2005.
DOI : 10.1016/j.immuni.2005.05.010

URL : https://doi.org/10.1016/j.immuni.2005.05.010

K. G. Smith and M. R. Clatworthy, FccRIIB in autoimmunity and infection: evolutionary and therapeutic implications, Nat Rev Immunol, vol.10, pp.328-371, 2010.
DOI : 10.1038/nri2762

URL : http://europepmc.org/articles/pmc4148599?pdf=render

M. R. Clatworthy, C. Aronin, R. J. Mathews, N. Y. Morgan, K. Smith et al., Immune complexes stimulate CCR7-dependent dendritic cell migration to lymph nodes, Nat Med, vol.20, pp.1458-63, 2014.
DOI : 10.1038/nm.3709

URL : http://europepmc.org/articles/pmc4283039?pdf=render

M. I. Iruretagoyena, C. A. Riedel, E. D. Leiva, M. A. Gutierrez, S. H. Jacobelli et al., Activating and inhibitory Fcc receptors can differentially modulate T cell-mediated autoimmunity, Eur J Immunol, vol.38, pp.2241-50, 2008.
DOI : 10.1002/eji.200838197

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/eji.200838197

R. Clynes, C. Dumitru, and J. V. Ravetch, Uncoupling of immune complex formation and kidney damage in autoimmune glomerulonephritis, Science, vol.279, pp.1052-1056, 1998.

T. Lovgren, M. L. Eloranta, B. Kastner, M. Wahren-herlenius, G. V. Alm et al., Induction of interferon-a by immune complexes or liposomes containing systemic lupus erythematosus autoantigen-and Sjogren's syndrome autoantigen-associated RNA, Arthritis Rheum, vol.54, pp.1917-1944, 2006.

U. Bave, M. Magnusson, M. L. Eloranta, A. Perers, G. V. Alm et al., FccRIIa is expressed on natural IFN-a-producing cells (plasmacytoid dendritic cells) and is required for the IFN-a production induced by apoptotic cells combined with lupus IgG, J Immunol, vol.171, pp.3296-302, 2003.

A. Regnault, D. Lankar, and V. Lacabanne, Fcc receptor-mediated induction of dendritic cell maturation and major histocompatibility complex class I-restricted antigen presentation after immune complex internalization, J Exp Med, vol.189, pp.371-80, 1999.
DOI : 10.1084/jem.189.2.371

URL : http://jem.rupress.org/content/189/2/371.full.pdf

A. M. Boruchov, G. Heller, M. Veri, E. Bonvini, J. V. Ravetch et al., Activating and inhibitory IgG Fc receptors on human DCs mediate opposing functions, J Clin Invest, vol.115, pp.2914-2937, 2005.
DOI : 10.1172/jci24772

URL : http://www.jci.org/articles/view/24772/files/pdf

A. M. Kalergis and J. V. Ravetch, Inducing tumor immunity through the selective engagement of activating Fcc receptors on dendritic cells, J Exp Med, vol.195, pp.1653-1662, 2002.

M. R. Clatworthy, S. K. Harford, R. J. Mathews, and K. Smith, FccRIIb inhibits immune complex-induced VEGF-A production and intranodal lymphangiogenesis, Proc Natl Acad Sci, vol.111, pp.17971-17977, 2014.
DOI : 10.1073/pnas.1413915111

URL : http://www.pnas.org/content/111/50/17971.full.pdf

T. Yuasa, S. Kubo, T. Yoshino, A. Ujike, K. Matsumura et al., Deletion of fcc receptor IIB renders H-2(b) mice susceptible to collagen-induced arthritis, J Exp Med, vol.189, pp.187-94, 1999.
DOI : 10.1084/jem.189.1.187

URL : http://jem.rupress.org/content/jem/189/1/187.full.pdf

M. Chang, C. J. Binder, M. Torzewski, and J. L. Witztum, C-reactive protein binds to both oxidized LDL and apoptotic cells through recognition of a common ligand: phosphorylcholine of oxidized phospholipids, Proc Natl Acad Sci, vol.99, pp.13043-13051, 2002.

C. Mold and T. Clos, C-reactive protein inhibits plasmacytoid dendritic cell interferon responses to autoantibody immune complexes, Arthritis Rheum, vol.65, pp.1891-901, 2013.
DOI : 10.1002/art.37968

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/art.37968

G. S. Garcia-romo, S. Caielli, and B. Vega, Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus, Sci Transl Med, vol.3, issue.73, 2011.

R. Lande, D. Ganguly, and V. Facchinetti, Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus, Sci Transl Med, vol.3, pp.73-92, 2011.
DOI : 10.1126/scitranslmed.3001180

URL : http://europepmc.org/articles/pmc3399524?pdf=render

R. J. Brownlie, K. E. Lawlor, and H. A. Niederer, Distinct cell-specific control of autoimmunity and infection by FccRIIb, J Exp Med, vol.205, pp.883-95, 2008.

N. Bhardwaj, J. W. Young, A. J. Nisanian, J. Baggers, and R. M. Steinman, Small amounts of superantigen, when presented on dendritic cells, are sufficient to initiate T cell responses, J Exp Med, vol.178, pp.633-675, 1993.

C. Koorella, J. R. Nair, M. E. Murray, L. M. Carlson, S. K. Watkins et al., Novel regulation of CD80/CD86-induced phosphatidylinositol 3-kinase signaling by NOTCH1 protein in interleukin-6 and indoleamine 2,3-dioxygenase production by dendritic cells, J Biol Chem, vol.289, pp.7747-62, 2014.

P. S. Linsley, W. Brady, L. Grosmaire, A. Aruffo, N. K. Damle et al., Binding of the B cell activation antigen B7 to CD28 costimulates T cell proliferation and interleukin 2 mRNA accumulation, J Exp Med, vol.173, pp.721-751, 1991.

S. Tseng, J. C. Waite, M. Liu, S. Vardhana, and M. L. Dustin, T cell-dendritic cell immunological synapses contain TCR-dependent CD28-CD80 clusters that recruit protein kinase Ch, J Immunol, vol.181, pp.4852-63, 2008.
DOI : 10.4049/jimmunol.181.7.4852

URL : http://www.jimmunol.org/content/jimmunol/181/7/4852.full.pdf

J. C. Crisp-in, M. I. Vargas-rojas, A. Monsiv-ais-urenda, and J. Alcocer-varela, Phenotype and function of dendritic cells of patients with systemic lupus erythematosus, Clin Immunol, vol.143, pp.45-50, 2012.

D. Ding, H. Mehta, W. J. Mccune, and M. J. Kaplan, Aberrant phenotype and function of myeloid dendritic cells in systemic lupus erythematosus, J Immunol, vol.177, pp.5878-89, 2006.

I. S. Grewal, H. G. Foellmer, K. D. Grewal, J. Xu, F. Hardardottir et al., Requirement for CD40 ligand in costimulation induction, T cell activation, and experimental allergic encephalomyelitis, Science, vol.273, pp.1864-1871, 1996.
DOI : 10.1126/science.273.5283.1864

K. Gerritse, J. D. Laman, R. J. Noelle, A. Aruffo, J. A. Ledbetter et al., CD40-CD40 ligand interactions in experimental allergic encephalomyelitis and multiple sclerosis, Proc Natl Acad Sci, vol.93, pp.2499-504, 1996.
DOI : 10.1073/pnas.93.6.2499

URL : http://www.pnas.org/content/93/6/2499.full.pdf

L. M. Bagenstose, R. K. Agarwal, P. B. Silver, D. M. Harlan, S. C. Hoffmann et al., Disruption of CD40/CD40-ligand interactions in a retinal autoimmunity model results in protection without tolerance, J Immunol, vol.175, pp.124-154, 2005.

J. K. Burgess, S. Carlin, R. A. Pack, G. M. Arndt, W. W. Au et al., Detection and characterization of OX40 ligand expression in human airway smooth muscle cells: a possible role in asthma?, J Allergy Clin Immunol, vol.113, pp.683-692, 2004.

T. Ito, Y. Wang, and O. Duramad, TSLP-activated dendritic cells induce an inflammatory T helper type 2 cell response through OX40 ligand, J Exp Med, vol.202, pp.1213-1236, 2005.
DOI : 10.1084/jem.20051135

URL : http://jem.rupress.org/content/202/9/1213.full.pdf

F. Gaspal, D. Withers, and M. Saini, Abrogation of CD30 and OX40 signals prevents autoimmune disease in FoxP3-deficient mice, J Exp Med, vol.208, pp.1579-84, 2011.

S. K. Yoshinaga, J. S. Whoriskey, and S. D. Khare, T-cell co-stimulation through B7RP-1 and ICOS, Nature, vol.402, pp.827-859, 1999.
DOI : 10.1038/45582

M. Kopf, A. J. Coyle, N. Schmitz, M. Barner, A. Oxenius et al., Inducible costimulator protein (Icos) controls T helper cell subset polarization after virus and parasite infection, J Exp Med, vol.192, pp.53-62, 2000.
DOI : 10.1084/jem.192.1.53

URL : http://jem.rupress.org/content/192/1/53.full.pdf

E. J. Witsch, M. Peiser, A. Hutloff, K. B?-uchner, B. G. Dorner et al., Kroczek RA. ICOS and CD28 reversely regulate IL-10 on re-activation of human effector T cells with mature dendritic cells, Eur J Immunol, vol.32, pp.2680-2686, 2002.

G. C. Sim, N. Martin-orozco, and J. L. , IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients, J Clin Invest, vol.124, pp.99-110, 2014.

X. Gao, L. Zhao, S. Wang, J. Yang, and X. Yang, Enhanced inducible costimulator ligand (ICOS-L) expression on dendritic cells in interleukin-10 deficiency and its impact on T-cell subsets in respiratory tract infection, Mol Med, vol.19, pp.346-56, 2013.

T. W. Mak, A. Shahinian, and S. K. Yoshinaga, Costimulation through the inducible costimulator ligand is essential for both T helper and B cell functions in T cell-dependent B cell responses, Nat Immunol, vol.4, pp.765-72, 2003.

M. I. Ramos, P. P. Tak, and M. C. Lebre, Fms-like tyrosine kinase 3 ligand-dependent dendritic cells in autoimmune inflammation, Autoimmun Rev, vol.13, pp.117-141, 2014.

H. J. Mckenna, K. L. Stocking, and R. E. Miller, Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells, Blood, vol.95, pp.3489-97, 2000.

M. Dehlin, M. Bokarewa, R. Rottapel, S. J. Foster, M. Magnusson et al., Intra-articular Fms-like tyrosine kinase 3 ligand expression is a driving force in induction and progression of arthritis, PLoS ONE, vol.3, p.3633, 2008.

M. C. Erlandsson, K. Forslind, S. Andersson, A. Lund, and M. I. Bokarewa, Metastasin S100A4 is increased in proportion to radiographic damage in patients with RA, Rheumatology, vol.51, pp.932-972, 2012.

N. Yogev, F. Frommer, and D. Lukas, Dendritic cells ameliorate autoimmunity in the CNS by controlling the homeostasis of PD-1 receptor+ regulatory T cells, Immunity, vol.37, pp.264-75, 2012.

T. Yamazaki, H. Akiba, and H. Iwai, Expression of programmed death 1 ligands by murine T cells and APC, J Immunol, vol.169, pp.5538-5583, 2002.

R. C. Mcpherson, J. E. Konkel, and C. T. Prendergast, Epigenetic modification of the PD-1 (Pdcd1) promoter in effector CD4

, + T cells tolerized by peptide immunotherapy, eLife, vol.3, p.3416, 2014.

M. E. Keir, L. M. Francisco, and A. H. Sharpe, PD-1 and its ligands in T-cell immunity, Curr Opin Immunol, vol.19, pp.309-323, 2007.

N. Dilek, N. Poirier, and P. Hulin, CTLA-4 and PD-L1 costimulation differentially controls immune synapses and function of human regulatory and conventional T-cells, PLoS ONE, vol.28, p.83139, 2013.

L. M. Francisco, V. H. Salinas, K. E. Brown, V. K. Vanguri, G. J. Freeman et al., PD-L1 regulates the development, maintenance, and function of induced regulatory T cells, J Exp Med, vol.206, pp.3015-3044, 2009.

B. Schreiner, S. L. Bailey, T. Shin, L. Chen, and S. D. Miller, PD-1 ligands expressed on myeloid-derived APC in the CNS regulate T-cell responses in EAE, Eur J Immunol, vol.38, pp.2706-2723, 2008.

Y. E. Latchman, S. C. Liang, Y. Wu, T. Chernova, R. A. Sobel et al., PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells, Proc Natl Acad Sci, vol.101, pp.10691-10697, 2004.

H. Nishimura, M. Nose, H. Hiai, N. Minato, and T. Honjo, Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor, Immunology, vol.11, pp.497-507, 1999.

S. Chiba, M. Baghdadi, and H. Akiba, Tumor-infiltrating DCs suppress nucleic acidmediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1, Nat Immunol, vol.13, pp.832-874, 2012.

N. Maurya, R. Gujar, M. Gupta, V. Yadav, S. Verma et al., Immunoregulation of dendritic cells by the receptor T cell Ig and mucin protein-3 via Bruton's tyrosine kinase and c-Src, J Immunol, vol.193, pp.3417-3442, 2014.

T. G. Phan, I. Grigorova, T. Okada, and J. G. Cyster, Subcapsular encounter and complementdependent transport of immune complexes by lymph node B cells, Nat Immunol, vol.8, pp.992-1000, 2007.

J. Fayette, B. Dubois, S. Vandenabeele, J. Bridon, B. Vanbervliet et al., Human dendritic cells skew isotype switching of CD40-activated Naive B cells towards IgA1 and IgA2, J Exp Med, vol.185, pp.1909-1927, 1997.

S. Wan, Z. Zhou, B. Duan, and L. Morel, Direct B cell stimulation by dendritic cells in a mouse model of lupus, Arthritis Rheum, vol.58, pp.1741-50, 2008.

A. Sang, Y. Zheng, Y. Yin, I. Dozmorov, H. Li et al., Dysregulated cytokine production by dendritic cells modulates B cell responses in the NZM2410 mouse model of lupus, PLoS ONE, vol.9, p.102151, 2014.

J. Shaw, Y. H. Wang, T. Ito, K. Arima, and Y. J. Liu, Plasmacytoid dendritic cells regulate B-cell growth and differentiation via CD70, Blood, vol.115, pp.3051-3058, 2010.

M. B. Litinskiy, B. Nardelli, D. M. Hilbert, B. He, A. Schaffer et al., DCs induce CD40-independent immunoglobulin class switching through BLyS and APRIL, Nat Immunol, vol.3, pp.822-831, 2002.
DOI : 10.1038/ni829

URL : http://europepmc.org/articles/pmc4621779?pdf=render

L. Qian, C. Qian, Y. Chen, Y. Bai, Y. Bao et al., Regulatory dendritic cells program B cells to differentiate into CD19 hi FccIIb hi regulatory B cells through IFN-b and CD40L, Blood, vol.120, pp.581-91, 2012.
DOI : 10.1182/blood-2011-08-377242

URL : http://www.bloodjournal.org/content/120/3/581.full.pdf

C. Pasare and R. Medzhitov, Toll pathway-dependent blockade of CD4+ CD25+ T cell-mediated suppression by dendritic cells, Science, vol.299, pp.1033-1039, 2003.

S. Wan, C. Xia, and L. Morel, IL-6 produced by dendritic cells from lupus-prone mice inhibits CD4+ CD25+ T cell regulatory functions, J Immunol, vol.178, pp.271-280, 2007.
DOI : 10.4049/jimmunol.178.1.271

URL : http://www.jimmunol.org/content/178/1/271.full.pdf

G. Grondal, I. Gunnarsson, J. Ronnelid, S. Rogberg, L. Klareskog et al., Cytokine production, serum levels and disease activity in systemic lupus erythematosus, Clin Exp Rheumatol, vol.18, pp.565-70, 2000.

P. Amerio, A. Frezzolini, D. Abeni, P. Teofoli, C. R. Girardelli et al., Increased IL-18 in patients with systemic lupus erythematosus: relations with Th-1, Th-2, pro-inflammatory cytokines and disease activity. IL-18 is a marker of disease activity but does not correlate with pro-inflammatory cytokines, Clin Exp Rheumatol, vol.20, pp.535-543, 2002.

A. Galil, S. M. Ezzeldin, N. El-boshy, and M. E. , The role of serum IL-17 and IL-6 as biomarkers of disease activity and predictors of remission in patients with lupus nephritis, Cytokine, issue.15, pp.187-195, 2015.

B. Liang, D. B. Gardner, D. E. Griswold, P. J. Bugelski, and X. Song, Anti-interleukin-6 monoclonal antibody inhibits autoimmune responses in a murine model of systemic lupus erythematosus, Immunology, vol.119, pp.296-305, 2006.

P. Terness, T. Oelert, and S. Ehser, Mitomycin C-treated dendritic cells inactivate autoreactive T cells: toward the development of a tolerogenic vaccine in autoimmune diseases, Proc Natl Acad Sci, vol.105, pp.18442-18449, 2008.

D. Ra?-ich-regu-e, M. Glancy, and A. W. Thomson, Regulatory dendritic cell therapy: from rodents to clinical application, Immunol Lett, vol.161, pp.216-237, 2014.

N. Xing, M. L. Lm, L. A. Bachman, D. J. Mckean, R. Kumar et al., Distinctive dendritic cell modulation by vitamin D(3) and glucocorticoid pathways, Biochem Biophys Res Commun, vol.297, pp.645-52, 2002.
DOI : 10.1016/s0006-291x(02)02262-3

S. Kubsch, E. Graulich, J. Knop, and K. Steinbrink, Suppressor activity of anergic T-cells induced by IL-10-treated human dendritic cells: association with IL-2-and CTLA-4-dependent G1 arrest of the cell cycle regulated by p27Kip1, Eur J Immunol, vol.33, pp.1988-97, 2003.

R. Volchenkov, J. Brun, R. Jonsson, and S. Appel, In vitro suppression of immune responses using monocyte-derived tolerogenic dendritic cells from patients with primary Sjogren's syndrome, Arthritis Res Ther, vol.15, p.114, 2013.

H. Hackstein, A. E. Morelli, A. T. Larregina, R. W. Ganster, G. D. Papworth et al., Aspirin inhibits in vitro maturation and in vivo immunostimulatory function of murine myeloid dendritic cells, J Immunol, vol.166, pp.7053-62, 2001.

S. Huang and J. A. Cidlowski, Phosphorylation status modulates Bcl-2 function during glucocorticoid-induced apoptosis in T lymphocytes, FASEB J, vol.16, pp.825-857, 2002.

. Sergeev, 125-Dihydroxyvitamin D3 induces Ca 2+ -mediated apoptosis in adipocytes via activation of calpain and caspase-12, Biochem Biophys Res Commun, vol.384, pp.18-21, 2009.

K. O. Dixon, J. O'flynn, S. W. Van-der-kooij, and C. Van-kooten, Phagocytosis of apoptotic or necrotic cells differentially regulates the transcriptional expression of IL-12 family members in dendritic cells, J Leukoc Biol, vol.96, pp.313-337, 2014.

H. Hackstein, T. Taner, A. F. Zahorchak, A. E. Morelli, A. J. Logar et al., Rapamycin inhibits IL-4-induced dendritic cell maturation in vitro and dendritic cell mobilization and function in vivo, Blood, vol.101, pp.4457-63, 2003.

R. Volchenkov, M. Karlsen, R. Jonsson, and S. Appel, Type 1 regulatory T cells and regulatory B cells induced by tolerogenic dendritic cells, Scand J Immunol, vol.77, pp.246-54, 2013.

E. Martin, C. Capini, E. Duggan, V. P. Lutzky, P. Stumbles et al., Antigen-specific suppression of established arthritis in mice by dendritic cells deficient in NF-jB, Arthritis Rheum, vol.56, pp.2255-66, 2007.

A. M. Kalergis, M. I. Iruretagoyena, and M. J. Barrientos, Modulation of nuclear factorkappaB activity can influence the susceptibility to systemic lupus erythematosus, Immunology, vol.128, pp.306-320, 2009.

C. Chauveau, S. Remy, and P. J. Royer, Heme oxygenase-1 expression inhibits dendritic cell maturation and proinflammatory function but conserves IL-10 expression, Blood, vol.106, pp.1694-702, 2005.

K. Steinbrink, E. Graulich, S. Kubsch, J. Knop, and A. H. Enk, CD4+ and CD8+ anergic T cells induced by interleukin-10-treated human dendritic cells display antigen-specific suppressor activity, Blood, vol.99, pp.2468-76, 2002.

X. Li, A. Yang, H. Huang, X. Zhang, J. Town et al., Induction of Type 2 T helper cell allergen tolerance by IL-10-differentiated regulatory dendritic cells, Am J Respir Cell Mol Biol, vol.42, pp.190-199, 2010.

F. W. Velten, K. Duperrier, J. Bohlender, P. Metharom, and S. Goerdt, A gene signature of inhibitory MHC receptors identifies a BDCA3+ subset of IL-10-induced dendritic cells with reduced allostimulatory capacity in vitro, Eur J Immunol, vol.34, pp.2800-2811, 2004.

D. C. Thomas, F. S. Wong, P. Zaccone, and E. A. Green, Protection of islet grafts through transforming growth factor-b-induced tolerogenic dendritic cells, Diabetes, vol.62, pp.3132-3174, 2013.

R. Li, X. Zheng, and I. Popov, Gene silencing of IL-12 in dendritic cells inhibits autoimmune arthritis, J Transl Med, vol.10, p.19, 2012.

X. Zheng, M. Suzuki, and T. E. Ichim, Treatment of autoimmune arthritis using RNA interference-modulated dendritic cells, J Immunol, vol.184, pp.6457-64, 2010.

X. Zheng, M. Suzuki, and X. Zhang, RNAi-mediated CD40-CD154 interruption promotes tolerance in autoimmune arthritis, Arthritis Res Ther, vol.12, p.13, 2010.

D. A. Ferenbach, V. Ramdas, N. Spencer, L. Marson, I. Anegon et al., Macrophages expressing heme oxygenase-1 improve renal function in ischemia/reperfusion injury, Mol Ther, vol.18, pp.1706-1719, 2010.

I. Dufait, T. Liechtenstein, A. Lanna, C. Bricogne, R. Laranga et al., Escors D. Retroviral and lentiviral vectors for the induction of immunological tolerance, Scientifica, p.14, 2012.

D. Ra?ch-regu-e, L. Grau-l-opez, M. Naranjo-g-omez, C. Ramo-tello, R. Pujol-borrell et al., Borr as FE. Stable antigen-specific T-cell hyporesponsiveness induced by tolerogenic dendritic cells from multiple sclerosis patients, Eur J Immunol, vol.42, pp.771-82, 2012.

V. Chiurchi-u, M. T. Cencioni, E. Bisicchia, D. Bardi, M. Gasperini et al., Distinct modulation of human myeloid and plasmacytoid dendritic cells by anandamide in multiple sclerosis, Ann Neurol, vol.73, pp.626-662, 2013.

M. J. Mansilla, C. Sell-es-moreno, S. Puig, J. Amoedo, J. Navarro-barriuso et al., Beneficial effect of tolerogenic dendritic cells pulsed with MOG autoantigen in experimental autoimmune encephalomyelitis, CNS Neurosci Ther, vol.21, pp.222-252, 2015.

E. M. Ginzler, M. A. Dooley, and C. Aranow, Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis, New Engl J Med, vol.353, pp.2219-2247, 2005.

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.2026-2058, 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, pp.290-87, 2015.

R. Thomas, N. Ramnoruth, and H. Pahau, Feasibility, safety and clinical effects of Single intradermal administration of autologous tolerising dendritic cells exposed to citrullinated peptides in patients with rheumatoid arthritis, Arthritis Rheum, vol.63, p.2430, 2011.

, Immunology, vol.146, pp.497-507