H. Park, Z. Li, X. O. Yang, S. H. Chang, R. Nurieva et al., A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17, Nat Immunol, vol.6, pp.1133-1141, 2005.

M. Veldhoen, R. J. Hocking, C. J. Atkins, R. M. Locksley, and B. Stockinger, TGF-? in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells, Immunity, vol.24, pp.179-189, 2006.

E. Bettelli, Y. Carrier, W. Gao, T. Korn, T. B. Strom et al., Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells, Nature, vol.441, pp.235-238, 2006.

P. R. Mangan, L. E. Harrington, D. B. O'quinn, W. S. Helms, D. C. Bullard et al., Transforming growth factor-beta induces development of the T(H)17 lineage, Nature, vol.441, pp.231-234, 2006.

E. V. Acosta-rodriguez, L. Rivino, J. Geginat, D. Jarrossay, M. Gattorno et al., Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells, Nat Immunol, vol.8, pp.639-646, 2007.

F. Annunziato, L. Cosmi, V. Santarlasci, L. Maggi, F. Liotta et al., Phenotypic and functional features of human Th17 cells, J Exp Med, vol.204, pp.1849-1861, 2007.

W. Sato, T. Aranami, and T. Yamamura, Cutting edge: Human Th17 cells are identified as bearing CCR2+CCR5-phenotype, J Immunol, vol.178, pp.7525-7529, 2007.

L. Maggi, V. Santarlasci, M. Capone, A. Peired, F. Frosali et al., CD161 is a marker of all human IL-17-producing T-cell subsets and is induced by RORC, Eur J Immunol, vol.40, pp.2174-2181, 2010.

M. Veldhoen, R. J. Hocking, R. A. Flavell, and B. Stockinger, Signals mediated by transforming growth factor-beta initiate autoimmune encephalomyelitis, but chronic inflammation is needed to sustain disease, Nat Immunol, vol.7, pp.1151-1156, 2006.

I. Gutcher, M. K. Donkor, Q. Ma, A. Y. Rudensky, R. A. Flavell et al., Autocrine transforming growth factor-beta1 promotes in vivo Th17 cell differentiation, Immunity, vol.34, pp.396-408, 2011.

T. Korn, E. Bettelli, W. Gao, A. Awasthi, A. Jager et al., IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells, Nature, vol.448, pp.484-487, 2007.

R. Nurieva, X. O. Yang, G. Martinez, Y. Zhang, A. D. Panopoulos et al., Essential autocrine regulation by IL-21 in the generation of inflammatory T cells, Nature, vol.448, pp.480-483, 2007.

L. Zhou, . Ivanov, . Ii, R. Spolski, R. Min et al., IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways, Nat Immunol, vol.8, pp.967-974, 2007.

T. Korn, E. Bettelli, M. Oukka, and V. K. Kuchroo, IL-17 and Th17 Cells, vol.27, pp.485-517, 2009.

M. J. Mcgeachy, Y. Chen, C. M. Tato, A. Laurence, B. Joyce-shaikh et al., The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo, Nat Immunol, vol.10, pp.314-324, 2009.

H. Qin, L. Wang, T. Feng, C. O. Elson, S. A. Niyongere et al., TGF-? promotes Th17 cell development through inhibition of SOCS3, J Immunol, vol.183, pp.97-105, 2009.

K. Ghoreschi, A. Laurence, X. P. Yang, C. M. Tato, M. J. Mcgeachy et al., Generation of pathogenic T(H)17 cells in the absence of TGF-? signalling, vol.467, pp.967-971, 2010.

J. Das, G. Ren, L. Zhang, A. I. Roberts, X. Zhao et al., Transforming growth factor ? is dispensable for the molecular orchestration of Th17 cell differentiation, J Exp Med, vol.206, pp.2407-2416, 2009.

E. V. Acosta-rodriguez, G. Napolitani, A. Lanzavecchia, and F. Sallusto, Interleukins 1? and 6 but not transforming growth factor-? are essential for the differentiation of interleukin 17-producing human T helper cells, Nat Immunol, vol.8, pp.942-949, 2007.

N. J. Wilson, K. Boniface, J. R. Chan, B. S. Mckenzie, W. M. Blumenschein et al., Malefyt R: Development, cytokine profile and function of human interleukin 17-producing helper T cells, Nat Immunol, vol.8, pp.950-957, 2007.

L. Yang, D. E. Anderson, C. Baecher-allan, W. D. Hastings, E. Bettelli et al., IL-21 and TGF-? are required for differentiation of human T(H)17 cells, Nature, vol.454, pp.350-352, 2008.

N. Manel, D. Unutmaz, and D. R. Littman, The differentiation of human T(H)-17 cells requires transforming growth factor-? and induction of the nuclear receptor ROR?t, Nat Immunol, vol.9, pp.641-649, 2008.

E. Volpe, N. Servant, R. Zollinger, S. I. Bogiatzi, P. Hupe et al., Soumelis V: A critical function for transforming growth factor-?, interleukin 23 and proinflammatory cytokines in driving and modulating human T(H)-17 responses, Nat Immunol, vol.9, pp.650-657, 2008.

L. Cosmi, D. Palma, R. Santarlasci, V. Maggi, L. Capone et al., Human interleukin 17-producing cells originate from a CD161+CD4+ T cell precursor, J Exp Med, vol.205, pp.1903-1916, 2008.

W. W. Lee, S. W. Kang, J. Choi, S. H. Lee, K. Shah et al., Regulating human Th17 cells via differential expression of IL-1 receptor, Blood, vol.115, pp.530-540, 2010.

V. Santarlasci, L. Maggi, M. Capone, F. Frosali, V. Querci et al., TGF-? indirectly favors the development of human Th17 cells by inhibiting Th1 cells, Eur J Immunol, vol.39, pp.207-215, 2009.

G. J. Martinez, Z. Zhang, J. M. Reynolds, S. Tanaka, Y. Chung et al., Smad2 positively regulates the generation of Th17 cells, J Biol Chem, vol.285, pp.29039-29043, 2010.

K. Ichiyama, T. Sekiya, N. Inoue, T. Tamiya, I. Kashiwagi et al., Transcription factor Smad-independent T helper 17 cell induction by transforming-growth factor-? is mediated by suppression of eomesodermin, Immunity, vol.34, pp.741-754, 2011.

L. Wei, A. Laurence, K. M. Elias, O. Shea, and J. J. , IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner, J Biol Chem, vol.282, pp.34605-34610, 2007.

Z. Chen, A. Laurence, Y. Kanno, M. Pacher-zavisin, B. M. Zhu et al., Selective regulatory function of Socs3 in the formation of IL-17-secreting T cells, Proc Natl Acad Sci, vol.103, pp.8137-8142, 2006.

Y. Chung, S. H. Chang, G. J. Martinez, X. O. Yang, R. Nurieva et al., Critical regulation of early Th17 cell differentiation by interleukin-1 signaling, Immunity, vol.30, pp.576-587, 2009.

A. Brustle, S. Heink, M. Huber, C. Rosenplanter, C. Stadelmann et al., The development of inflammatory T(H)-17 cells requires interferon-regulatory factor, vol.4, pp.958-966, 2007.

K. Okamoto, Y. Iwai, M. Oh-hora, M. Yamamoto, T. Morio et al., I?B? regulates T(H)17 development by cooperating with ROR nuclear receptors, Nature, vol.464, pp.1381-1385, 2010.

B. U. Schraml, K. Hildner, W. Ise, W. L. Lee, W. A. Smith et al., The AP-1 transcription factor Batf controls T(H)17 differentiation, Nature, vol.460, pp.405-409, 2009.

F. Zhang, G. Meng, and W. Strober, Interactions among the transcription factors Runx1, ROR?t and Foxp3 regulate the differentiation of interleukin 17-producing T cells, Nat Immunol, vol.9, pp.1297-1306, 2008.

L. Zhou, J. E. Lopes, M. M. Chong, . Ivanov, M. R. Ii et al., TGF-?-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing ROR?t function, Nature, vol.453, pp.236-240, 2008.

X. O. Yang, A. D. Panopoulos, R. Nurieva, S. H. Chang, D. Wang et al., STAT3 regulates cytokine-mediated generation of inflammatory helper T cells, J Biol Chem, vol.282, pp.9358-9363, 2007.

A. Laurence, C. M. Tato, T. S. Davidson, Y. Kanno, Z. Chen et al., Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation, Immunity, vol.26, pp.371-381, 2007.

J. Moisan, R. Grenningloh, E. Bettelli, M. Oukka, and I. C. Ho, Ets-1 is a negative regulator of Th17 differentiation, J Exp Med, vol.204, pp.2825-2835, 2007.

W. Liao, J. X. Lin, L. Wang, P. Li, and W. J. Leonard, Modulation of cytokine receptors by IL-2 broadly regulates differentiation into helper T cell lineages, Nat Immunol, vol.12, pp.551-559, 2011.

K. M. Elias, A. Laurence, T. S. Davidson, G. Stephens, Y. Kanno et al., Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat-3/Stat-5 independent signaling pathway, Blood, vol.111, pp.1013-1020, 2008.

D. Mucida, Y. Park, G. Kim, O. Turovskaya, I. Scott et al., Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid, Science, vol.317, pp.256-260, 2007.

F. J. Quintana, A. S. Basso, A. H. Iglesias, T. Korn, M. F. Farez et al., Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor, Nature, vol.453, pp.65-71, 2008.

M. Veldhoen, K. Hirota, A. M. Westendorf, J. Buer, L. Dumoutier et al., The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins, Nature, vol.453, pp.106-109, 2008.

F. J. Quintana, G. Murugaiyan, M. F. Farez, M. Mitsdoerffer, A. M. Tukpah et al., An endogenous aryl hydrocarbon receptor ligand acts on dendritic cells and T cells to suppress experimental autoimmune encephalomyelitis, Proc Natl Acad Sci U S A, vol.107, pp.20768-20773, 2010.

R. Gandhi, D. Kumar, E. J. Burns, M. Nadeau, B. Dake et al., Activation of the aryl hydrocarbon receptor induces human type 1 regulatory T cell-like and Foxp3(+) regulatory T cells, Nat Immunol, vol.11, pp.846-853, 2010.

Y. Chen, C. J. Haines, I. Gutcher, K. Hochweller, W. M. Blumenschein et al., Foxp3(+) Regulatory T Cells Promote T Helper 17 Cell Development In Vivo through Regulation of Interleukin-2, Immunity, vol.34, pp.409-421, 2011.

P. Pandiyan, H. R. Conti, L. Zheng, A. C. Peterson, D. R. Mathern et al., CD4(+)CD25(+)Foxp3(+) Regulatory T Cells Promote Th17 Cells In Vitro and Enhance Host Resistance in Mouse Candida albicans Th17 Cell Infection Model, vol.34, pp.422-434, 2011.

V. Lazarevic, X. Chen, J. H. Shim, E. S. Hwang, E. Jang et al., T-bet represses T(H)17 differentiation by preventing Runx1-mediated activation of the gene encoding ROR?t, Nat Immunol, vol.12, pp.96-104, 2011.

M. Batten, J. Li, S. Yi, N. M. Kljavin, D. M. Danilenko et al., Interleukin 27 limits autoimmune encephalomyelitis by suppressing the development of interleukin 17-producing T cells, Nat Immunol, vol.7, pp.929-936, 2006.

J. S. Stumhofer, J. S. Silver, A. Laurence, P. M. Porrett, T. H. Harris et al., Interleukins 27 and 6 induce STAT3-mediated T cell production of interleukin 10, Nat Immunol, vol.8, pp.1363-1371, 2007.

D. C. Fitzgerald, G. X. Zhang, M. El-behi, Z. Fonseca-kelly, H. Li et al., Suppression of autoimmune inflammation of the central nervous system by interleukin 10 secreted by interleukin 27-stimulated T cells, Nat Immunol, vol.8, pp.1372-1379, 2007.

J. P. Van-hamburg, M. J. De-bruijn, R. De-almeida, C. Van-zwam, M. Van-meurs et al., Enforced expression of GATA3 allows differentiation of IL-17-producing cells, but constrains Th17-mediated pathology, Eur J Immunol, vol.38, pp.2573-2586, 2008.

K. S. Voo, Y. H. Wang, F. R. Santori, C. Boggiano, K. Arima et al., Identification of IL-17-producing FOXP3+ regulatory T cells in humans, Proc Natl Acad Sci U S A, vol.106, pp.4793-4798, 2009.

L. Cosmi, L. Maggi, V. Santarlasci, M. Capone, E. Cardilicchia et al., Identification of a novel subset of human circulating memory CD4(+) T cells that produce both IL-17A and IL-4, J Allergy Clin Immunol, vol.125, pp.222-230, 2010.

K. Hirota, J. H. Duarte, M. Veldhoen, E. Hornsby, Y. Li et al., Fate mapping of IL-17-producing T cells in inflammatory responses, Nat Immunol, vol.12, pp.255-263, 2011.

M. H. Lexberg, A. Taubner, I. Albrecht, I. Lepenies, A. Richter et al., IFN-? and IL-12 synergize to convert in vivo generated Th17 into Th1/Th17 cells, Eur J Immunol, vol.40, pp.3017-3027, 2010.

E. Esplugues, S. Huber, N. Gagliani, A. E. Hauser, T. Town et al., Control of TH17 cells occurs in the small intestine, vol.475, pp.514-518, 2011.

G. Wei, L. Wei, J. Zhu, C. Zang, J. Hu-li et al., Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells, vol.30, pp.155-167, 2009.

P. Miossec, T. Korn, and V. K. Kuchroo, Interleukin-17 and type 17 helper T cells, N Engl J Med, vol.361, pp.888-898, 2009.

C. M. Wilke, K. Bishop, D. Fox, and W. Zou, Deciphering the role of Th17 cells in human disease, Trends Immunol, vol.32, pp.603-611, 2011.

H. Shen, J. C. Goodall, H. Gaston, and J. S. , Frequency and phenotype of peripheral blood Th17 cells in ankylosing spondylitis and rheumatoid arthritis, Arthritis Rheum, vol.60, pp.1647-1656, 2009.

E. Lubberts, M. I. Koenders, and W. B. Van-den-berg, The role of T-cell interleukin-17 in conducting destructive arthritis: lessons from animal models, Arthritis Res Ther, vol.7, pp.29-37, 2005.

S. K. Kwok, M. L. Cho, M. K. Park, H. J. Oh, J. S. Park et al., Interleukin-21 promotes osteoclastogenesis in rheumatoid arthritis in humans and mice, Arthritis Rheum, 2011.

D. A. Young, M. Hegen, H. L. Ma, M. J. Whitters, L. M. Albert et al., Blockade of the interleukin-21/interleukin-21 receptor pathway ameliorates disease in animal models of rheumatoid arthritis, Arthritis Rheum, vol.56, pp.1152-1163, 2007.

J. Leipe, M. A. Schramm, M. Grunke, M. Baeuerle, C. Dechant et al., Interleukin 22 serum levels are associated with radiographic progression in rheumatoid arthritis, Ann Rheum Dis, vol.70, pp.1453-1457, 2011.

R. J. Marijnissen, M. I. Koenders, R. L. Smeets, M. H. Stappers, C. Nickerson-nutter et al., Increased expression of interleukin-22 by synovial Th17 cells during late stages of murine experimental arthritis is controlled by interleukin-1 and enhances bone degradation, Arthritis Rheum, vol.63, pp.2939-2948, 2011.

L. Cosmi, R. Cimaz, L. Maggi, V. Santarlasci, M. Capone et al., Evidence of the transient nature of the Th17 phenotype of CD4+CD161+ T cells in the synovial fluid of patients with juvenile idiopathic arthritis, Arthritis Rheum, vol.63, pp.2504-2515, 2011.

K. Nistala, S. Adams, H. Cambrook, S. Ursu, B. Olivito et al., Th17 plasticity in human autoimmune arthritis is driven by the inflammatory environment, Proc Natl Acad Sci U S A, vol.107, pp.14751-14756, 2010.

J. Yang, Y. Chu, X. Yang, D. Gao, L. Zhu et al., Th17 and natural Treg cell population dynamics in systemic lupus erythematosus, Arthritis Rheum, vol.60, pp.1472-1483, 2009.

H. C. Hsu, P. Yang, J. Wang, Q. Wu, R. Myers et al., Interleukin 17-producing T helper cells and interleukin 17 orchestrate autoreactive germinal center development in autoimmune BXD2 mice, Nat Immunol, vol.9, pp.166-175, 2008.

A. Doreau, A. Belot, J. Bastid, B. Riche, M. C. Trescol-biemont et al., Interleukin 17 acts in synergy with B cellactivating factor to influence B cell biology and the pathophysiology of systemic lupus erythematosus, Nat Immunol, vol.10, pp.778-785, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00539348

A. H. Sawalha, K. M. Kaufman, J. A. Kelly, A. J. Adler, T. Aberle et al., Genetic association of interleukin-21 polymorphisms with systemic lupus erythematosus, Ann Rheum Dis, vol.67, pp.458-461, 2008.

R. Webb, J. T. Merrill, J. A. Kelly, A. Sestak, K. M. Kaufman et al., Sawalha AH: A polymorphism within IL21R confers risk for systemic lupus erythematosus, vol.60, pp.2402-2407, 2009.

R. X. Leng, H. F. Pan, G. M. Chen, C. C. Feng, Y. G. Fan et al., The dual nature of Ets-1: focus to the pathogenesis of systemic lupus erythematosus, Autoimmun Rev, vol.10, pp.439-443, 2011.

D. Herber, T. P. Brown, S. Liang, D. A. Young, M. Collins et al., IL-21 has a pathogenic role in a lupus-prone mouse model and its blockade with IL-21R.Fc reduces disease progression, J Immunol, vol.178, pp.3822-3830, 2007.

C. Lock, G. Hermans, R. Pedotti, A. Brendolan, E. Schadt et al., Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis, Nat Med, vol.8, pp.500-508, 2002.

J. S. Tzartos, M. A. Friese, M. J. Craner, J. Palace, J. Newcombe et al., Interleukin-17 production in central nervous system-infiltrating T cells and glial cells is associated with active disease in multiple sclerosis, Am J Pathol, vol.172, pp.146-155, 2008.

T. Carlson, M. Kroenke, P. Rao, T. E. Lane, and B. Segal, The Th17-ELR+ CXC chemokine pathway is essential for the development of central nervous system autoimmune disease, J Exp Med, vol.205, pp.811-823, 2008.

Y. Komiyama, S. Nakae, T. Matsuki, A. Nambu, H. Ishigame et al., IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis, J Immunol, vol.177, pp.566-573, 2006.

V. Siffrin, H. Radbruch, R. Glumm, R. Niesner, M. Paterka et al., In vivo imaging of partially reversible th17 cell-induced neuronal dysfunction in the course of encephalomyelitis, Immunity, vol.33, pp.424-436, 2010.

L. Codarri, G. Gyulveszi, V. Tosevski, L. Hesske, A. Fontana et al., ROR?t drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation, Nat Immunol, vol.12, pp.560-567, 2011.

M. El-behi, B. Ciric, H. Dai, Y. Yan, M. Cullimore et al., The encephalitogenicity of T(H)17 cells is dependent on IL-1-and IL-23-induced production of the cytokine GM-CSF, Nat Immunol, vol.12, pp.568-575, 2011.

I. L. King, M. A. Kroenke, and B. M. Segal, GM-CSF-dependent, CD103+ dermal dendritic cells play a critical role in Th effector cell differentiation after subcutaneous immunization, J Exp Med, vol.207, pp.953-961, 2010.

M. A. Lowes, T. Kikuchi, J. Fuentes-duculan, I. Cardinale, L. C. Zaba et al., Psoriasis vulgaris lesions contain discrete populations of Th1 and Th17 T cells, J Invest Dermatol, vol.128, pp.1207-1211, 2008.

H. L. Ma, S. Liang, J. Li, L. Napierata, T. Brown et al., IL-22 is required for Th17 cell-mediated pathology in a mouse model of psoriasis-like skin inflammation, J Clin Invest, vol.118, pp.597-607, 2008.

L. A. Zenewicz, A. Antov, and R. A. Flavell, CD4 T-cell differentiation and inflammatory bowel disease, Trends Mol Med, vol.15, pp.199-207, 2009.

L. A. Zenewicz, G. D. Yancopoulos, D. M. Valenzuela, A. J. Murphy, S. Stevens et al., Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease, 89. van den Berg WB, Miossec P: IL-17 as a future therapeutic target for rheumatoid arthritis, vol.29, pp.549-553, 2008.

M. Milovanovic, G. Drozdenko, C. Weise, M. Babina, and M. Worm, Interleukin-17A promotes IgE production in human B cells, J Invest Dermatol, vol.130, pp.2621-2628, 2010.

J. Bayry, M. Thirion, S. Delignat, N. Misra, S. Lacroix-desmazes et al., Dendritic cells and autoimmunity, Autoimmun Rev, vol.3, pp.183-187, 2004.
URL : https://hal.archives-ouvertes.fr/inserm-02455548

J. Bayry, S. Lacroix-desmazes, M. D. Kazatchkine, and S. V. Kaveri, Monoclonal antibody and intravenous immunoglobulin therapy for rheumatic diseases: rationale and mechanisms of action, Nat Clin Pract Rheumatol, vol.3, pp.262-272, 2007.

S. Andre, D. F. Tough, S. Lacroix-desmazes, S. V. Kaveri, and J. Bayry, Surveillance of antigen-presenting cells by CD4+CD25+ regulatory T cells in autoimmunity: immunopathogenesis and therapeutic implications, Am J Pathol, vol.174, pp.1575-1587, 2009.

J. Bayry, S. Siberil, F. Triebel, D. F. Tough, and S. V. Kaveri, Rescuing CD4+CD25+ regulatory T-cell functions in rheumatoid arthritis by cytokine-targeted monoclonal antibody therapy, Drug Discov Today, vol.12, pp.548-552, 2007.

C. Klemann, B. J. Raveney, A. K. Klemann, T. Ozawa, S. Von-horsten et al., Synthetic retinoid AM80 inhibits Th17 cells and ameliorates experimental autoimmune encephalomyelitis, Am J Pathol, vol.174, pp.2234-2245, 2009.

M. S. Sundrud, S. B. Koralov, M. Feuerer, D. P. Calado, A. E. Kozhaya et al., Halofuginone inhibits TH17 cell differentiation by activating the amino acid starvation response, Science, vol.324, pp.1334-1338, 2009.

G. G. Krueger, R. G. Langley, C. Leonardi, N. Yeilding, C. Guzzo et al., A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis, N Engl J Med, vol.356, pp.580-592, 2007.

W. J. Sandborn, B. G. Feagan, R. N. Fedorak, E. Scherl, M. R. Fleisher et al., A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn's disease, Gastroenterology, vol.135, pp.1130-1141, 2008.

S. Joshi, L. C. Pantalena, X. K. Liu, S. L. Gaffen, H. Liu et al., Youssef S: 1,25-dihydroxyvitamin D(3) ameliorates Th17 autoimmunity via transcriptional modulation of interleukin-17A, Mol Cell Biol, vol.31, pp.3653-3669, 2011.

A. Grigorian, L. Araujo, N. N. Naidu, D. Place, B. Choudhury et al., Nacetylglucosamine inhibits T-helper 1 (Th1) / T-helper 17 (Th17) responses and treats experimental autoimmune encephalomyelitis, J Biol Chem, vol.286, pp.40133-40141, 2011.

W. Hueber, D. D. Patel, T. Dryja, A. M. Wright, I. Koroleva et al., Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis, vol.2, pp.52-72, 2010.

M. C. Genovese, F. Van-den-bosch, S. A. Roberson, S. Bojin, I. M. Biagini et al., LY2439821, a humanized anti-interleukin-17 monoclonal antibody, in the treatment of patients with rheumatoid arthritis: A phase I randomized, double-blind, placebo-controlled, proof-of-concept study, Arthritis Rheum, vol.62, pp.929-939, 2010.

L. Klotz, S. Burgdorf, D. I. Saijo, K. Flossdorf, J. Hucke et al., The nuclear receptor PPAR? selectively inhibits Th17 differentiation in a T cell-intrinsic fashion and suppresses CNS autoimmunity, J Exp Med, vol.206, pp.2079-2089, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00509630

X. Zhang, J. J. Peng, X. Ramgolam, V. S. Markovic-plese, and S. , Simvastatin inhibits IL-17 secretion by targeting multiple IL-17-regulatory cytokines and by inhibiting the expression of IL-17 transcription factor RORC in CD4+ lymphocytes, J Immunol, vol.180, pp.6988-6996, 2008.

J. R. Huh, M. W. Leung, P. Huang, D. A. Ryan, M. R. Krout et al., Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing ROR?t activity, Nature, vol.472, pp.486-490, 2011.

C. Kitabayashi, T. Fukada, M. Kanamoto, W. Ohashi, S. Hojyo et al., Zinc suppresses Th17 development via inhibition of STAT3 activation, Int Immunol, vol.22, pp.375-386, 2010.

T. P. Singh, B. Huettner, H. Koefeler, G. Mayer, I. Bambach et al., Platelet-activating factor blockade inhibits the T-helper type 17 cell pathway and suppresses psoriasis-like skin disease in K5.hTGF-?1 transgenic mice, Am J Pathol, vol.178, pp.699-708, 2011.

W. Cao, Y. Yang, Z. Wang, A. Liu, L. Fang et al., Leukemia inhibitory factor inhibits T helper 17 cell differentiation and confers treatment effects of neural progenitor cell therapy in autoimmune disease, Immunity, vol.35, pp.273-284, 2011.

M. S. Maddur, J. Vani, P. Hegde, S. Lacroix-desmazes, S. V. Kaveri et al., Inhibition of differentiation, amplification, and function of human TH17 cells by intravenous immunoglobulin, J Allergy Clin Immunol, vol.127, pp.823-830, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02455567

M. S. Maddur, S. V. Kaveri, and J. Bayry, Comparison of different IVIg preparations on IL-17 production by human Th17 cells, Autoimmun Rev, vol.10, pp.809-810, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02455563

J. Bayry, V. S. Negi, and S. V. Kaveri, Intravenous immunoglobulin therapy in rheumatic diseases, Nat Rev Rheumatol, vol.7, pp.349-359, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-02455562

A. Pashov, S. Delignat, J. Bayry, and S. V. Kaveri, Enhancement of the affinity of glucocorticoid receptors as a mechanism underlying the steroid-sparing effect of intravenous immunoglobulin, J Rheumatol, vol.38, p.2275, 2011.

H. Kopf, G. M. De-la-rosa, O. M. Howard, and X. Chen, Rapamycin inhibits differentiation of Th17 cells and promotes generation of FoxP3+ T regulatory cells, Int Immunopharmacol, vol.7, pp.1819-1824, 2007.