J. Banchereau and R. M. Steinman, Dendritic cells and the control of immunity, Nature, vol.392, pp.245-52, 1998.

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, D. Ye, H. Cui, Y. Zhang et al., 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.

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.411-418, 1997.

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.

E. Gianchecchi, D. V. Delfino, and A. Fierabracci, Recent insights into the role of the PD-1/ PD-L1 pathway in immunological tolerance and autoimmunity, Autoimmun Rev, vol.12, pp.1091-100, 2013.

C. Mok, L. Ho, and C. To, Annual incidence and standardized incidence ratio of cerebrovascular accidents in patients with systemic lupus erythematosus, Scand J Rheumatol, vol.38, pp.362-370, 2009.

S. Bernatsky, J. F. Boivin, L. Joseph, S. Manzi, E. Ginzler et al., 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 lowaffinity Fc? receptors and costimulatory molecules on dendritic cells, Immunology, vol.128, pp.334-375, 2009.

J. C. Crispin, V. C. Kyttaris, C. Terhorst, and G. C. Tsokos, T cells as therapeutic targets in SLE, Nat Rev Rheumatol, vol.6, pp.317-342, 2010.

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

E. Schwarz and C. Ritchlin, Clinical development of anti-RANKL therapy, Arthritis Res Ther, vol.9, p.7, 2007.

F. Houssiau and E. Ginzler, Current treatment of lupus nephritis, Lupus, vol.17, pp.426-456, 2008.

B. H. Hahn, Belimumab for systemic lupus erythematosus, N Engl J Med, vol.368, pp.1528-1563, 2013.

J. Lee, T. Kim, H. E. Park, E. G. Lee, N. Jung et al., Myosin-primed tolerogenic dendritic cells ameliorate experimental autoimmune myocarditis, Cardiovasc Res, vol.101, pp.203-213, 2014.

A. Mukhopadhaya, T. Hanafusa, I. Jarchum, Y. Chen, Y. Iwai et al., Selective delivery of ? cell antigen to dendritic cells in vivo leads to deletion and tolerance of autoreactive CD8+ T cells in NOD mice, Proc Natl Acad Sci, vol.105, pp.6374-6383, 2008.

I. Van-brussel, W. P. Lee, M. Rombouts, A. H. Nuyts, M. Heylen et al., Tolerogenic dendritic cell vaccines to treat autoimmune diseases: can the unattainable dream turn into reality?, Autoimmun Rev, vol.13, pp.138-50, 2014.

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

L. O'neill, Toll-like receptor signal transduction and the tailoring of innate immunity: a role for Mal?, Trends Immunol, vol.23, pp.296-300, 2002.

S. Ioannou and M. Voulgarelis, Toll-like receptors, tissue injury, and tumourigenesis, Mediators Inflamm, 2010.

A. L. Blasius and B. Beutler, Intracellular Toll-like receptors, Immunity, vol.32, pp.305-320, 2010.

S. M. Bueno, C. A. Riedel, L. J. Carreno, and A. M. Kalergis, Virulence mechanisms displayed by Salmonella to impair dendritic cell function, Curr Med Chem, vol.17, pp.1156-66, 2010.

M. I. Iruretagoyena, M. Wiesendanger, and A. M. Kalergis, The dendritic cell-T cell synapse as a determinant of autoimmune pathogenesis, Curr Pharm Des, vol.12, pp.131-178, 2006.

A. M. Kalergis, Modulation of T cell immunity by TCR/pMHC dwell time and activating/inhibitory receptor pairs on the antigen-presenting cell, Curr Pharm Des, vol.9, pp.233-277, 2003.

C. Llanos, L. J. Carreno, M. A. Gutierrez, C. A. Riedel, S. H. Jacobelli et al., Genetic and pharmacological modulation of dendritic cell-T cell interactions as a therapeutic strategy for systemic lupus erythematosus, Curr Gene Ther, vol.11, 2011.

S. S. Diebold, Determination of T-cell fate by dendritic cells, Immunol Cell Biol, vol.86, pp.389-97, 2008.

O. Brain, O. Benjamin, M. J. Pichulik, T. Allan, P. Khatamzas et al., The intracellular sensor NOD2 induces microRNA-29 expression in human dendritic cells to limit IL-23 release, Immunity, vol.39, pp.521-557, 2013.

G. Ferwerda, M. Kramer, D. De-jong, A. Piccini, L. A. Joosten et al., Engagement of NOD2 has a dual effect on proIL-1? mRNA transcription and secretion of bioactive IL-1?, Eur J Immunol, vol.38, pp.184-91, 2008.

A. Brosbøl-ravnborg, C. L. Hvas, J. Agnholt, J. F. Dahlerup, I. Vind et al., Toll-like receptor-induced granulocyte-macrophage colony-stimulating factor secretion is impaired in Crohn's disease by nucleotide oligomerization domain 2-dependent and -independent pathways, Clin Exp Immunol, vol.155, pp.487-95, 2009.

A. M. Piccinini and K. S. Midwood, DAMPening inflammation by modulating TLR signalling, Mediators Inflamm, 2010.

Z. Wen, L. Xu, X. Chen, W. Xu, Z. Yin et al., Autoantibody induction by DNAcontaining 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, M. J. Shlomchik et al., Chromatin-IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors, Nature, vol.416, pp.603-610, 2002.

Y. Doring, H. D. Manthey, M. Drechsler, D. Lievens, R. T. Megens et al., 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.

R. Lande, J. Gregorio, V. Facchinetti, B. Chatterjee, Y. H. Wang et al., Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide, Nature, vol.449, pp.564-573, 2007.

D. Ganguly, G. Chamilos, R. Lande, J. Gregorio, S. Meller et al., Self-RNAantimicrobial peptide complexes activate human dendritic cells through TLR7 and TLR8, J Exp Med, vol.206, pp.1983-94, 2009.

Y. Dombrowski and J. Schauber, Cathelicidin LL-37: a defense molecule with a potential role in psoriasis pathogenesis, Exp Dermatol, vol.21, pp.327-357, 2012.

M. Gilliet and R. Lande, Antimicrobial peptides and self-DNA in autoimmune skin inflammation, Curr Opin Immunol, vol.20, pp.401-408, 2008.

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, N. J. Trendell-smith, A. Wu, L. Tian et al., 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.

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

G. Ruiz-irastorza, M. Ramos-casals, P. Brito-zeron, and M. A. Khamashta, Clinical efficacy and side effects of antimalarials in systemic lupus erythematosus: a systematic review, Ann Rheum Dis, vol.69, pp.20-28, 2010.

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.

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.

E. E. Brown, J. C. Edberg, and R. P. Kimberly, Fc receptor genes and the systemic lupus erythematosus diathesis, Autoimmunity, vol.40, pp.567-81, 2007.

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, FcgammaRIV: a novel FcR with distinct IgG subclass specificity, Immunity, vol.23, pp.41-51, 2005.

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

M. I. Iruretagoyena, C. A. Riedel, E. D. Leiva, M. A. Gutierrez, S. H. Jacobelli et al., Activating and inhibitory Fcgamma receptors can differentially modulate T cellmediated autoimmunity, Eur J Immunol, vol.38, pp.2241-50, 2008.

J. V. Ravetch and S. Bolland, IgG Fc receptors, Annu Rev Immunol, vol.19, pp.275-90, 2001.

A. A. Herrada, F. J. Contreras, J. A. Tobar, R. Pacheco, and A. M. Kalergis, Immune complexinduced enhancement of bacterial antigen presentation requires Fcgamma receptor III expression on dendritic cells, Proc Natl Acad Sci U S A, vol.104, pp.13402-13409, 2007.

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

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

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

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

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

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

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.

J. A. Tobar, P. A. Gonzalez, and A. M. Kalergis, Salmonella escape from antigen presentation can be overcome by targeting bacteria to Fc gamma receptors on dendritic cells, J Immunol, vol.173, pp.4058-65, 2004.

A. A. Herrada, F. J. Contreras, J. A. Tobar, R. Pacheco, and A. M. Kalergis, Immune complexinduced enhancement of bacterial antigen presentation requires Fc? receptor III expression on dendritic cells, Proc Natl Acad Sci, vol.104, pp.13402-13409, 2007.

R. J. Brownlie, K. E. Lawlor, H. A. Niederer, A. J. Cutler, Z. Xiang et al., Distinct cell-specific control of autoimmunity and infection by Fc?RIIb, 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.

E. M. Beckman, S. A. Porcelli, C. T. Morita, S. M. Behar, S. T. Furlong et al., Recognition of a lipid antigen by CD1-restricted alpha beta+ T cells, Nature, vol.372, pp.691-695, 1994.

D. J. Montamat-sicotte, K. A. Millington, C. R. Willcox, S. Hingley-wilson, S. Hackforth et al., A mycolic acid-specific CD1-restricted T cell population contributes to acute and memory immune responses in human tuberculosis infection, J Clin Invest, vol.121, pp.2493-503, 2011.

L. J. Carreno, P. A. Gonzalez, and A. M. Kalergis, Modulation of T cell function by TCR/pMHC binding kinetics, Immunobiology, vol.211, pp.47-64, 2006.

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.

M. Hubo, B. Trinschek, F. Kryczanowsky, A. Tüttenberg, K. Steinbrink et al., Costimulatory molecules on immunogenic versus tolerogenic human dendritic cells, Front Immunol, issue.4, 2013.

J. Romo-tena, D. Gómez-martín, A. Varela, and J. , CTLA-4 and autoimmunity: new insights into the dual regulator of tolerance, Autoimmun Rev, vol.12, pp.1171-1177, 2013.

H. Schneider, J. Downey, A. Smith, B. H. Zinselmeyer, C. Rush et al., Reversal of the TCR stop signal by CTLA-4, Science, vol.313, pp.1972-1977, 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.

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.

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. C. Crispin, M. I. Vargas-rojas, A. Monsivais-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.

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, O. Duramad, T. Hori, G. J. Delespesse et al., TSLPactivated dendritic cells induce an inflammatory T helper type 2 cell response through OX40 ligand, J Exp Med, vol.202, pp.1213-1236, 2005.

F. M. Gaspal, D. Withers, M. Saini, V. Bekiaris, F. M. Mcconnell et al., 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, S. D. Khare, U. Sarmiento, J. Guo et al., T-cell costimulation through B7RP-1 and ICOS, Nature, vol.402, pp.827-859, 1999.

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.

E. J. Witsch, M. Peiser, A. Hutloff, K. Büchner, B. G. Dorner et al., 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, J. L. Yang, Y. Wu, S. Washington et al., 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, S. K. Yoshinaga, A. Wakeham, L. Boucher et al., 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, R. E. Miller, K. Brasel, D. Smedt et al., 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, D. Lukas, K. Kautz-neu, K. Karram et al., 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, H. Iwai, H. Matsuda, M. Aoki et al., Expression of programmed death 1 ligands by murine T cells and APC, J Immunol, vol.169, pp.5538-5583, 2002.

X. Zhong, J. R. Tumang, W. Gao, C. Bai, and T. L. Rothstein, PD-L2 expression extends beyond dendritic cells/macrophages to B1 cells enriched for VH11/VH12 and phosphatidylcholine binding, Eur J Immunol, vol.37, pp.2405-2415, 2007.

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, P. Hulin, F. Coulon, C. Mary et al., 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.
URL : https://hal.archives-ouvertes.fr/inserm-02165637

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.

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 U S A, 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, Immunity, vol.11, pp.141-51, 1999.

S. Chiba, M. Baghdadi, H. Akiba, H. Yoshiyama, I. Kinoshita et al., Tumor-infiltrating DCs suppress nucleic acid-mediated 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.

O. Berggren, N. Hagberg, G. Weber, G. V. Alm, L. Rönnblom et al., B lymphocytes enhance interferon-? production by plasmacytoid dendritic cells, Arthritis Rheum, vol.64, pp.3409-3428, 2012.

A. Karni, M. Abraham, A. Monsonego, G. Cai, G. J. Freeman et al., Innate immunity in multiple sclerosis: myeloid dendritic cells in secondary progressive multiple sclerosis are activated and drive a proinflammatory immune response, J Immunol, vol.177, pp.4196-202, 2006.

M. Tucci, N. Calvani, H. B. Richards, C. Quatraro, and F. Silvestris, The interplay of chemokines and dendritic cells in the pathogenesis of lupus nephritis, Ann N Y Acad Sci, vol.1051, pp.421-453, 2005.

P. Decker, I. Kotter, R. Klein, B. Berner, and H. G. Rammensee, Monocyte-derived dendritic cells over-express CD86 in patients with systemic lupus erythematosus, Rheumatology (Oxford), vol.45, pp.1087-95, 2006.

T. Kaisho, Pathogen sensors and chemokine receptors in dendritic cell subsets, Vaccine, vol.30, pp.7652-7659, 2012.

D. Jarrossay, G. Napolitani, M. Colonna, F. Sallusto, and A. Lanzavecchia, Specialization and complementarity in microbial molecule recognition by human myeloid and plasmacytoid dendritic cells, Eur J Immunol, vol.31, pp.3388-93, 2001.

A. Krug, A. Towarowski, S. Britsch, S. Rothenfusser, V. Hornung et al., 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, J. Oldenburg, J. Hoch, T. Bieber et al., Human plasmacytoid dendritic cells support Th17 cell effector function in response to TLR7 ligation, J Immunol, vol.184, pp.1159-67, 2010.

J. A. Villadangos and L. Young, Antigen-presentation properties of plasmacytoid dendritic cells, Immunity, vol.29, pp.352-61, 2008.

A. Boonstra, C. Asselin-paturel, M. Gilliet, C. Crain, G. Trinchieri et al., Flexibility of mouse classical and plasmacytoid-derived dendritic cells in directing T helper type 1 and 2 cell development: dependency on antigen dose and differential Toll-like receptor ligation, J Exp Med, vol.197, pp.101-110, 2003.

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

Y. J. Liu, IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors, Annu Rev Immunol, vol.23, pp.275-306, 2005.

M. Kuwana, J. Kaburaki, T. M. Wright, Y. Kawakami, and Y. Ikeda, Induction of antigenspecific human CD4+ T cell anergy by peripheral blood DC2 precursors, Eur J Immunol, vol.31, pp.2547-57, 2001.

A. Izaguirre, B. J. Barnes, S. Amrute, W. S. Yeow, N. Megjugorac et al., Comparative analysis of IRF and IFN-alpha expression in human plasmacytoid and monocytederived dendritic cells, J Leukoc Biol, vol.74, pp.1125-1163, 2003.

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.

G. Jego, A. K. Palucka, J. Blanck, C. Chalouni, V. Pascual et al., Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6, Immunity, vol.19, pp.225-259, 2003.

A. Cerutti, X. Qiao, and B. He, Plasmacytoid dendritic cells and the regulation of immunoglobulin heavy chain class switching, Immunol Cell Biol, vol.83, pp.554-62, 2005.

H. Poeck, M. Wagner, J. Battiany, S. Rothenfusser, D. Wellisch et al., Plasmacytoid dendritic cells, antigen, and CpG-C license human B cells for plasma cell differentiation and immunoglobulin production in the absence of T-cell help, Blood, vol.103, pp.3058-64, 2004.

L. E. Ronnblom, G. V. Alm, and K. Oberg, Autoimmune phenomena in patients with malignant carcinoid tumors during interferon-alpha treatment, Acta Oncol, vol.30, pp.537-577, 1991.

L. E. Wilson, D. Widman, S. H. Dikman, and P. D. Gorevic, Autoimmune disease complicating antiviral therapy for hepatitis C virus infection, Semin Arthritis Rheum, vol.32, pp.163-73, 2002.

K. M. Kalkner, L. Ronnblom, K. Parra, A. K. Bengtsson, M. Olsson et al., Antibodies against double-stranded DNA and development of polymyositis during treatment with interferon, QJM, vol.91, pp.393-402, 1998.

R. Maldonado-lopez, D. Smedt, T. Pajak, B. Heirman, C. Thielemans et al., Role of CD8alpha+ and CD8alpha-dendritic cells in the induction of primary immune responses in vivo, J Leukoc Biol, vol.66, pp.242-248, 1999.

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, Y. Cui, L. Sun, D. Ye et al., 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, P. Fragapane, D. Salerno, C. Marcantonio et al., IFN-? regulates Blimp-1 expression via miR-23a and miR-125b in both monocytes-derived DC and pDC, PLoS One, vol.8, p.72833, 2013.

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-alpha, IFN-gamma), 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.

F. Bonnefoy, M. Couturier, A. Clauzon, J. Rémy-martin, B. Gaugler et al., TGF-?-exposed plasmacytoid dendritic cells participate in Th17 commitment, J Immunol, vol.186, pp.6157-64, 2011.

S. Fagarasan and T. Honjo, T-Independent immune response: new aspects of B cell biology, Science, vol.290, pp.89-92, 2000.

J. W. Gregersen and D. Jayne, B-cell depletion in the treatment of lupus nephritis, Nat Rev Nephrol, vol.8, pp.505-519, 2012.

A. Lanzavecchia, Antigen-specific interaction between T and B cells, Nature, vol.314, pp.537-546, 1985.

F. D. Batista and N. E. Harwood, The who, how and where of antigen presentation to B cells, Nat Rev Immunol, vol.9, pp.15-27, 2009.

J. Tian, D. Zekzer, L. Hanssen, Y. Lu, A. Olcott et al., Lipopolysaccharideactivated B cells down-regulate Th1 immunity and prevent autoimmune diabetes in nonobese diabetic mice, J Immunol, vol.167, pp.1081-1090, 2001.

P. A. Blair, L. Y. Norena, F. Flores-borja, D. J. Rawlings, D. A. Isenberg et al., CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients, Immunity, vol.32, pp.129-169, 2010.

M. W. Klinker, T. J. Reed, D. A. Fox, and S. K. Lundy, Interleukin-5 supports the expansion of Fas ligand-expressing killer B cells that induce antigen-specific apoptosis of CD4

, + T cells and secrete interleukin-10, PLoS One, vol.8, p.70131, 2013.

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.

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.

Z. Hua, A. J. Gross, C. Lamagna, N. Ramos-hernández, P. Scapini et al., Requirement for MyD88 signaling in B cells and dendritic cells for germinal center anti-nuclear antibody production in Lyn-deficient mice, J Immunol, vol.192, pp.875-85, 2014.

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.

L. Qian, C. Qian, Y. Chen, Y. Bai, Y. Bao et al., Regulatory dendritic cells program B cells to differentiate into CD19hiFcgammaIIbhi regulatory B cells through IFN-beta and CD40L, Blood, vol.120, pp.581-91, 2012.

S. Sakaguchi, T. Yamaguchi, T. Nomura, and M. Ono, Regulatory T cells and immune tolerance, Cell, vol.133, pp.775-87, 2008.

S. Sakaguchi, M. Ono, R. Setoguchi, H. Yagi, S. Hori et al., Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease, Immunol Rev, vol.212, pp.8-27, 2006.

C. Pasare and R. Medzhitov, Toll pathway-dependent blockade of CD4+CD25+ T cellmediated 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.

A. Mcnally, M. Mcnally, R. Galea, R. Thomas, and R. J. Steptoe, Immunogenic, but not steady-state, antigen presentation permits regulatory T-cells to control CD8 + T-cell effector differentiation by IL-2 modulation, PLoS One, vol.9, p.85455, 2014.

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.

G. S. Dean, J. Tyrrell-price, E. Crawley, and D. A. Isenberg, Cytokines and systemic lupus erythematosus, Ann Rheum Dis, vol.59, pp.243-51, 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, proinflammatory 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.

E. Tackey, P. E. Lipsky, and G. G. Illei, Rationale for interleukin-6 blockade in systemic lupus erythematosus, Lupus, vol.13, pp.339-382, 2004.

R. M. Steinman, D. Hawiger, and M. C. Nussenzweig, Tolerogenic dendritic cells, Annu Rev Immunol, vol.21, pp.685-711, 2003.

S. Guerder, N. Joncker, K. Mahiddine, and L. Serre, Dendritic cells in tolerance and autoimmune diabetes, Curr Opin Immunol, vol.25, pp.670-675, 2013.

D. Raïch-regué, M. Glancy, and A. W. Thomson, Regulatory dendritic cell therapy: From rodents to clinical application, Immunol Lett, vol.161, pp.216-237, 2014.

C. Llanos, J. P. Mackern-oberti, F. Vega, S. H. Jacobelli, and A. M. Kalergis, Tolerogenic dendritic cells as a therapy for treating lupus, Clin Immunol, vol.148, pp.237-282, 2013.

D. Rea, C. Van-kooten, K. E. Van-meijgaarden, T. H. Ottenhoff, C. J. Melief et al., Glucocorticoids transform CD40-triggering of dendritic cells into an alternative activation pathway resulting in antigen-presenting cells that secrete IL-10, Blood, vol.95, pp.3162-3169, 2000.

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.

M. Moser, D. Smedt, T. Sornasse, T. Tielemans, F. Chentoufi et al., Glucocorticoids down-regulate dendritic cell function in vitro and in vivo, Eur J Immunol, vol.25, pp.2818-2842, 1995.

M. I. Iruretagoyena, J. A. Tobar, P. A. Gonzalez, S. E. Sepulveda, C. A. Figueroa et al., Andrographolide interferes with T cell activation and reduces experimental autoimmune encephalomyelitis in the mouse, J Pharmacol Exp Ther, vol.312, pp.366-72, 2005.

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.

H. Hackstein, T. Taner, A. J. Logar, and A. W. Thomson, Rapamycin inhibits macropinocytosis and mannose receptor-mediated endocytosis by bone marrow-derived dendritic cells, Blood, vol.100, pp.1084-1091, 2002.

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.

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.

D. Leung and J. W. Bloom, Update on glucocorticoid action and resistance, J Allergy Clin Immunol, vol.111, pp.3-22, 2003.

H. Ko, B. D. Hambly, J. M. Eris, V. Levidiotis, K. Wyburn et al., Dentritic cell derived IL-18 production is inhibited by rapamycin and sanglifehrin A, but not cyclosporine A, Transpl Immunol, vol.20, pp.99-105, 2008.

T. Simon, S. Pogu, V. Tardif, K. Rigaud, S. Rémy et al., Carbon monoxidetreated dendritic cells decrease ?1-integrin induction on CD8+ T cells and protect from type 1 diabetes, Eur J Immunol, vol.43, pp.209-227, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-02164776

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

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

Y. Ding, D. Chen, A. Tarcsafalvi, R. Su, L. Qin et al., Suppressor of cytokine signaling 1 inhibits IL-10-mediated immune responses, J Immunol, vol.170, pp.1383-91, 2003.

K. Sato, H. Nagayama, K. Tadokoro, T. Juji, and T. A. Takahashi, Extracellular signalregulated kinase, stress-activated protein kinase/c-Jun N-terminal kinase, and p38mapk are involved in il-10-mediated selective repression of TNF-?-induced activation and maturation of human peripheral blood monocyte-derived dendritic cells, J Immunol, vol.162, pp.3865-72, 1999.

A. Knodler, S. M. Schmidt, A. Bringmann, M. M. Weck, K. M. Brauer et al., Post-transcriptional regulation of adapter molecules by IL-10 inhibits TLRmediated activation of antigen-presenting cells, Leukemia, vol.23, pp.535-579, 2008.

P. J. Murray, Understanding and exploiting the endogenous interleukin-10/STAT3-mediated anti-inflammatory response, Curr Opin Pharmacol, vol.6, pp.379-86, 2006.

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.

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.

M. N. Lopez, B. Pesce, M. Kurte, C. Perez, G. Segal et al., A synthetic peptide homologous to IL-10 functional domain induces monocyte differentiation to TGFbeta+ tolerogenic dendritic cells, Immunobiology, vol.216, pp.1117-1143, 2011.

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

D. , A. A. Colucci, M. Pugliese, O. Quintieri, F. Boirivant et al., Cholera toxin B subunit promotes the induction of regulatory T cells by preventing human dendritic cell maturation, J Leukoc Biol, vol.84, pp.661-669, 2008.

F. Carranza, C. R. Falcon, N. Nunez, C. Knubel, S. G. Correa et al., Helminth antigens enable CpG-activated dendritic cells to inhibit the symptoms of collageninduced arthritis through Foxp3+ regulatory T cells, PLoS One, vol.7, p.40356, 2012.

K. V. Vukman, P. N. Adams, M. Metz, M. Maurer, O. Neill et al., Fasciola hepatica tegumental coat impairs mast cells' ability to drive Th1 immune responses, J Immunol, vol.190, pp.2873-2882, 2013.

K. Pauley and S. Cha, RNAi therapeutics in autoimmune disease, Pharmaceuticals, vol.6, pp.287-94, 2013.

F. Apparailly and C. Jorgensen, siRNA-based therapeutic approaches for rheumatic diseases, Nat Rev Rheumatol, vol.9, pp.56-62, 2013.

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

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

X. Zheng, M. Suzuki, X. Zhang, T. Ichim, F. Zhu et al., RNAi-mediated CD40-CD154 interruption promotes tolerance in autoimmune arthritis, Arthritis Res Ther, vol.12, p.13, 2010.

T. Kalantari, M. H. Karimi, B. Ciric, Y. Yan, A. Rostami et al., Tolerogenic dendritic cells produced by Lentiviral-mediated CD40-and IL-23p19-specific shRNA can ameliorate experimental autoimmune encephalomyelitis by suppressing Th17 cells, Clin Exp Immunol, vol.176, pp.180-189, 2014.

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.

M. A. Coleman and R. J. Steptoe, Induction of antigen-specific tolerance through hematopoietic stem cell-mediated gene therapy: the future for therapy of autoimmune disease?, Autoimmun Rev, vol.12, pp.195-203, 2012.

G. L. Mancardi, A. Murialdo, P. Rossi, F. Gualandi, G. Martino et al., Autologous stem cell transplantation as rescue therapy in malignant forms of multiple sclerosis, Mult Scler, vol.11, pp.367-71, 2005.

J. A. Snowden, J. Passweg, J. J. Moore, S. Milliken, P. Cannell et al., Autologous hemopoietic stem cell transplantation in severe rheumatoid arthritis: a report from the EBMT and ABMTR, J Rheumatol, vol.31, pp.482-490, 2004.

R. K. Burt, A. Traynor, L. Statkute, W. G. Barr, R. Rosa et al., Nonmyeloablative hematopoietic stem cell transplantation for systemic lupus erythematosus, JAMA, vol.295, pp.527-562, 2006.

C. E. Couri, M. C. Oliveira, A. B. Stracieri, D. A. Moraes, F. Pieroni et al., C-peptide levels and insulin independence following autologous nonmyeloablative hematopoietic stem cell transplantation in newly diagnosed type 1 diabetes mellitus, JAMA, vol.301, pp.1573-1582, 2009.

J. Chan, E. J. Ban, K. H. Chun, S. Wang, B. T. Bäckström et al., Transplantation of bone marrow transduced to express self-antigen establishes deletional tolerance and permanently remits autoimmune disease, J Immunol, vol.181, pp.7571-80, 2008.

R. J. Steptoe, J. M. Ritchie, and L. C. Harrison, Transfer of hematopoietic stem cells encoding autoantigen prevents autoimmune diabetes, J Clin Invest, vol.111, pp.1357-63, 2003.

R. Fritsch-stork, D. Mullegger, K. Skriner, B. Jahn-schmid, J. Smolen et al., The spliceosomal autoantigen heterogeneous nuclear ribonucleoprotein A2 (hnRNP-A2) is a major T cell autoantigen in patients with systemic lupus erythematosus, Arthritis Res Ther, vol.8, p.118, 2006.

A. M. Kalergis, M. I. Iruretagoyena, M. J. Barrientos, P. A. Gonzalez, A. A. Herrada et al., Modulation of nuclear factor-kappaB activity can influence the susceptibility to systemic lupus erythematosus, Immunology, vol.128, pp.306-320, 2009.

H. K. Kang, M. Liu, and S. K. Datta, Low-dose peptide tolerance therapy of lupus generates plasmacytoid dendritic cells that cause expansion of autoantigen-specific regulatory T cells and contraction of inflammatory Th17 cells, J Immunol, vol.178, pp.7849-58, 2007.

H. Kang, M. Chiang, M. Liu, D. Ecklund, and S. Datta, The histone peptide H471-94 alone is more effective than a cocktail of peptide epitopes in controlling lupus: immunoregulatory mechanisms, J Clin Immunol, vol.31, pp.379-94, 2011.

U. Sela, A. Sharabi, M. Dayan, R. Hershkoviz, and E. Mozes, The role of dendritic cells in the mechanism of action of a peptide that ameliorates lupus in murine models, Immunology, vol.128, pp.395-405, 2009.

Z. Sthoeger, H. Zinger, A. Sharabi, I. Asher, and E. Mozes, The tolerogenic peptide, hCDR1, down-regulates the expression of interferon-? in murine and human systemic lupus erythematosus, PLoS One, vol.8, p.60394, 2013.

E. M. Ginzler, M. A. Dooley, C. Aranow, M. Y. Kim, J. Buyon et al., Mycophenolate mofetil or intravenous cyclophosphamide for lupus nephritis, N Engl J Med, vol.353, pp.2219-2247, 2005.

J. A. Singh, G. A. Wells, R. Christensen, T. Ghogomu, E. Maxwell et al., Adverse effects of biologics: a network meta-analysis and Cochrane overview. Cochrane Database Syst Rev, vol.2, p.8794, 2011.

T. Bongartz, A. J. Sutton, M. J. Sweeting, I. Buchan, E. L. Matteson et al., Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials, JAMA, vol.295, pp.2275-85, 2006.

R. Beyaert, L. Beaugerie, G. Van-assche, L. Brochez, J. Renauld et al., Cancer risk in immune-mediated inflammatory diseases (IMID), Mol Cancer, vol.12, p.98, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00859833

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.

N. Giannoukakis, Interview: immunoregulatory dendritic cells to treat autoimmunity are ready for the clinic, Immunotherapy, vol.5, pp.919-940, 2013.

D. Ra?ch-regué, L. Grau-lópez, M. Naranjo-gómez, C. Ramo-tello, R. Pujol-borrell et al., 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. Chiurchiù, 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.

C. C. Gross, H. Jonuleit, and H. Wiendl, Fulfilling the dream: tolerogenic dendritic cells to treat multiple sclerosis, Eur J Immunol, vol.42, pp.569-72, 2012.

R. Thomas, N. Ramnoruth, H. Pahau, S. Law, and M. Brunck, 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.

H. A. Doyle and M. J. Mamula, Autoantigenesis: the evolution of protein modifications in autoimmune disease, Curr Opin Immunol, vol.24, pp.112-120, 2012.

O. P. Rekvig and J. C. Nossent, Anti-double-stranded DNA antibodies, nucleosomes, and systemic lupus erythematosus: a time for new paradigms?, Arthritis Rheum, vol.48, pp.300-312, 2003.

N. Figueroa-vega, G. Galindo-rodríguez, S. Bajaña, D. Portales-pérez, C. Abud-mendoza et al., Phenotypic analysis of IL-10-treated, monocyte-derived dendritic cells in patients with systemic lupus erythematosus, Scand J Immunol, vol.64, pp.668-76, 2006.

L. J. Carreno, C. A. Riedel, and A. M. Kalergis, Induction of tolerogenic dendritic cells by NFkappaB blockade and Fcgamma receptor modulation, Methods Mol Biol, vol.677, pp.339-53, 2010.