A. Iwasaki and R. Medzhitov, Regulation of Adaptive Immunity by the Innate Immune System, Science, vol.327, issue.5963, pp.291-296, 2010.
DOI : 10.1126/science.1183021

S. Akira, S. Uematsu, and O. Takeuchi, Pathogen Recognition and Innate Immunity, Cell, vol.124, issue.4, pp.783-801, 2006.
DOI : 10.1016/j.cell.2006.02.015

H. Kumar, T. Kawai, and S. Akira, Pathogen recognition in the innate immune response, Biochemical Journal, vol.45, issue.1, pp.1-16, 2009.
DOI : 10.1016/j.cell.2007.01.045

Y. Ogura, F. Sutterwala, and R. Flavell, The Inflammasome: First Line of the Immune Response to Cell Stress, Cell, vol.126, issue.4, pp.659-62, 2006.
DOI : 10.1016/j.cell.2006.08.002

V. Hornung and E. Latz, Intracellular DNA recognition, Nature Reviews Immunology, vol.24, issue.2, pp.123-153, 2010.
DOI : 10.1038/nri2690

L. Unterholzner, S. Keating, M. Baran, K. Horan, S. Jensen et al., IFI16 is an innate immune sensor for intracellular DNA, Nature Immunology, vol.175, issue.11, pp.997-1004, 2010.
DOI : 10.1084/jem.20061845

URL : https://hal.archives-ouvertes.fr/hal-00582635

O. Takeuchi and S. Akira, Pattern Recognition Receptors and Inflammation, Cell, vol.140, issue.6, pp.805-825, 2010.
DOI : 10.1016/j.cell.2010.01.022

L. Franchi, T. Eigenbrod, R. Munoz-planillo, and G. Nunez, The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis, Nature Immunology, vol.164, issue.3, pp.241-248, 2009.
DOI : 10.1038/nature07416

F. Martinon, K. Burns, and J. Tschopp, The Inflammasome, Molecular Cell, vol.10, issue.2, pp.417-443, 2002.
DOI : 10.1016/S1097-2765(02)00599-3

K. Schroder and J. Tschopp, The Inflammasomes, Cell, vol.140, issue.6, pp.821-853, 2010.
DOI : 10.1016/j.cell.2010.01.040

J. Ting, R. Lovering, E. Alnemri, J. Bertin, J. Boss et al., The NLR Gene Family: A Standard Nomenclature, Immunity, vol.28, issue.3, pp.285-292, 2008.
DOI : 10.1016/j.immuni.2008.02.005

F. Sutterwala, Y. Ogura, M. Szczepanik, M. Lara-tejero, G. Lichtenberger et al., Critical Role for NALP3/CIAS1/Cryopyrin in Innate and Adaptive Immunity through Its Regulation of Caspase-1, Immunity, vol.24, issue.3, pp.317-344, 2006.
DOI : 10.1016/j.immuni.2006.02.004

J. Anderson, J. Mueller, S. Rosengren, D. Boyle, P. Schaner et al., Structural, expression, and evolutionary analysis of mouse CIAS1, Gene, vol.338, issue.1, pp.25-34, 2004.
DOI : 10.1016/j.gene.2004.05.002

F. Bauernfeind, G. Horvath, A. Stutz, E. Alnemri, K. Macdonald et al., Cutting Edge: NF-??B Activating Pattern Recognition and Cytokine Receptors License NLRP3 Inflammasome Activation by Regulating NLRP3 Expression, The Journal of Immunology, vol.183, issue.2
DOI : 10.4049/jimmunol.0901363

S. Mccall, M. Sahraei, A. Young, C. Worley, J. Duncan et al., Osteoblasts Express NLRP3, a Nucleotide-Binding Domain and Leucine-Rich Repeat Region Containing Receptor Implicated in Bacterially Induced Cell Death, Journal of Bone and Mineral Research, vol.277, issue.Suppl 26, pp.30-40, 2008.
DOI : 10.1359/jbmr.071002

Y. Nakamura, N. Kambe, M. Saito, R. Nishikomiri, Y. Kim et al., Mast cells mediate neutrophil recruitment and vascular leakage through the NLRP3 inflammasome in histamine-independent urticaria, J Exp Med, 2009.

H. Watanabe, O. Gaide, V. Petrilli, F. Martinon, E. Contassot et al., Activation of the IL-1??-Processing Inflammasome Is Involved in Contact Hypersensitivity, Journal of Investigative Dermatology, vol.127, issue.8, pp.1956-63, 2007.
DOI : 10.1038/sj.jid.5700819

M. Gattorno, S. Tassi, S. Carta, L. Delfino, F. Ferlito et al., Pattern of interleukin-1?? secretion in response to lipopolysaccharide and ATP before and after interleukin-1 blockade in patients withCIAS1 mutations, Arthritis & Rheumatism, vol.23, issue.9, pp.3138-3186, 2007.
DOI : 10.1002/art.22842

J. Kummer, R. Broekhuizen, H. Everett, L. Agostini, L. Kuijk et al., Inflammasome Components NALP 1 and 3 Show Distinct but Separate Expression Profiles in Human Tissues Suggesting a Site-specific Role in the Inflammatory Response, Journal of Histochemistry & Cytochemistry, vol.591, issue.5, pp.443-52, 2007.
DOI : 10.1038/nrm1019

G. Meng and W. Strober, mutations, European Journal of Immunology, vol.2, issue.3, pp.649-53, 2010.
DOI : 10.1002/eji.200940191

I. Allen, M. Scull, C. Moore, E. Holl, E. Mcelvania-tekippe et al., The NLRP3 Inflammasome Mediates In Vivo Innate Immunity to Influenza A Virus through Recognition of Viral RNA, Immunity, vol.30, issue.4, pp.556-65, 2009.
DOI : 10.1016/j.immuni.2009.02.005

G. Meng, F. Zhang, I. Fuss, A. Kitani, and W. Strober, A Mutation in the Nlrp3 Gene Causing Inflammasome Hyperactivation Potentiates Th17 Cell-Dominant Immune Responses, Immunity, vol.30, issue.6, pp.860-74, 2009.
DOI : 10.1016/j.immuni.2009.04.012

Y. Chung, S. Chang, G. Martinez, X. Yang, R. Nurieva et al., Critical Regulation of Early Th17 Cell Differentiation by Interleukin-1 Signaling, Immunity, vol.30, issue.4, pp.576-87, 2009.
DOI : 10.1016/j.immuni.2009.02.007

C. Sutton, C. Brereton, B. Keogh, K. Mills, and E. Lavelle, A crucial role for interleukin (IL)-1 in the induction of IL-17???producing T cells that mediate autoimmune encephalomyelitis, The Journal of Experimental Medicine, vol.203, issue.7, pp.1685-91, 2006.
DOI : 10.1074/jbc.M308809200

S. Eisenbarth, O. Colegio, O. Connor, W. Sutterwala, F. Flavell et al., Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants, Nature, vol.103, issue.7198, pp.1122-1128, 2008.
DOI : 10.1038/nature06939

R. Spreafico, P. Ricciardi-castagnoli, and A. Mortellaro, The controversial relationship between NLRP3, alum, danger signals and the next-generation adjuvants, European Journal of Immunology, vol.296, issue.3, pp.638-680, 2010.
DOI : 10.1002/eji.200940039

H. Hoffman, J. Mueller, D. Broide, A. Wanderer, and R. Kolodner, Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome, Nature Genetics, vol.29, issue.3, pp.301-306, 2001.
DOI : 10.1038/ng756

J. Ting, D. Kastner, and H. Hoffman, CATERPILLERs, pyrin and hereditary immunological disorders, Nature Reviews Immunology, vol.52, issue.3, pp.183-95, 2006.
DOI : 10.1016/S0140-6736(04)17401-1

D. Littman and A. Rudensky, Th17 and Regulatory T Cells in Mediating and Restraining Inflammation, Cell, vol.140, issue.6, pp.845-58, 2010.
DOI : 10.1016/j.cell.2010.02.021

O. Shea, J. Paul, and W. , Mechanisms Underlying Lineage Commitment and Plasticity of Helper CD4+ T Cells, Science, vol.327, issue.5969, pp.1098-102, 2010.
DOI : 10.1126/science.1178334

J. Zhu and W. Paul, Heterogeneity and plasticity of T helper cells, Cell Research, vol.180, issue.1, pp.4-12, 2010.
DOI : 10.1038/ni.1730

K. Rautajoki, M. Kylaniemi, S. Raghav, K. Rao, and R. Lahesmaa, An insight into molecular mechanisms of human T helper cell differentiation, Annals of Medicine, vol.3, issue.5, pp.322-357, 2008.
DOI : 10.1038/nature01063

C. Dinarello, IL-18: A TH1 -inducing, proinflammatory cytokine and new member of the IL-1 family, Journal of Allergy and Clinical Immunology, vol.103, issue.1, pp.11-24, 1999.
DOI : 10.1016/S0091-6749(99)70518-X

K. Nakanishi, T. Yoshimoto, H. Tsutsui, and H. Okamura, Interleukin-18 is a unique cytokine that stimulates both Th1 and Th2 responses depending on its cytokine milieu, Cytokine & Growth Factor Reviews, vol.12, issue.1, pp.53-72, 2001.
DOI : 10.1016/S1359-6101(00)00015-0

D. Gris, Z. Ye, H. Iocca, H. Wen, R. Craven et al., NLRP3 Plays a Critical Role in the Development of Experimental Autoimmune Encephalomyelitis by Mediating Th1 and Th17 Responses, The Journal of Immunology, vol.185, issue.2, pp.974-81, 2010.
DOI : 10.4049/jimmunol.0904145

P. Shaw, J. Lukens, S. Burns, C. H. Mcgargill, M. Kanneganti et al., Cutting Edge: Critical Role for PYCARD/ASC in the Development of Experimental Autoimmune Encephalomyelitis, The Journal of Immunology, vol.184, issue.9, pp.4610-4614, 2010.
DOI : 10.4049/jimmunol.1000217

M. Zaki, P. Vogel, M. Body-malapel, M. Lamkanfi, and T. Kanneganti, IL-18 Production Downstream of the Nlrp3 Inflammasome Confers Protection against Colorectal Tumor Formation, The Journal of Immunology, vol.185, issue.8, pp.4912-4932, 2010.
DOI : 10.4049/jimmunol.1002046

B. Siegmund, Interleukin-18 in Intestinal Inflammation: Friend and Foe?, Immunity, vol.32, issue.3, pp.300-302, 2010.
DOI : 10.1016/j.immuni.2010.03.010

M. Zaki, K. Boyd, P. Vogel, M. Kastan, M. Lamkanfi et al., The NLRP3 Inflammasome Protects against Loss of Epithelial Integrity and Mortality during Experimental Colitis, Immunity, vol.32, issue.3, pp.379-91, 2010.
DOI : 10.1016/j.immuni.2010.03.003

J. Dupaul-chicoine, G. Yeretssian, K. Doiron, K. Bergstrom, C. Mcintire et al., Control of Intestinal Homeostasis, Colitis, and Colitis-Associated Colorectal Cancer by the Inflammatory Caspases, Immunity, vol.32, issue.3, pp.367-78, 2010.
DOI : 10.1016/j.immuni.2010.02.012

I. Allen, E. Tekippe, R. Woodford, J. Uronis, E. Holl et al., The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer, The Journal of Experimental Medicine, vol.34, issue.5, pp.1045-56, 2010.
DOI : 10.1038/nri1936

S. Hirota, J. Ng, A. Lueng, M. Khajah, K. Parhar et al., NLRP3 inflammasome plays a key role in the regulation of intestinal homeostasis, Inflammatory Bowel Diseases, vol.17, issue.6, 2010.
DOI : 10.1002/ibd.21478

C. Bauer, P. Duewell, C. Mayer, H. Lehr, K. Fitzgerald et al., Colitis induced in mice with dextran sulfate sodium (DSS) is mediated by the NLRP3 inflammasome, Gut, 2010.

F. Ghiringhelli, L. Apetoh, A. Tesniere, L. Aymeric, Y. Ma et al., Activation of the NLRP3 inflammasome in dendritic cells induces IL-1?????dependent adaptive immunity against tumors, Nature Medicine, vol.7, issue.10, pp.1170-1178, 2009.
DOI : 10.1038/nm.2028

URL : https://hal.archives-ouvertes.fr/hal-00419823

W. Paul and J. Zhu, How are TH2-type immune responses initiated and amplified?, Nature Reviews Immunology, vol.183, issue.4, pp.225-260, 2010.
DOI : 10.1038/nri2735

H. Li, S. Willingham, J. Ting, and F. Re, Cutting Edge: Inflammasome Activation by Alum and Alum's Adjuvant Effect Are Mediated by NLRP3, The Journal of Immunology, vol.181, issue.1, pp.17-21, 2008.
DOI : 10.4049/jimmunol.181.1.17

W. Arend, G. Palmer, and C. Gabay, IL-1, IL-18, and IL-33 families of cytokines, Immunological Reviews, vol.173, issue.1, pp.20-38, 2008.
DOI : 10.1111/j.1600-065X.2008.00624.x

M. Komai-koma, D. Xu, Y. Li, A. Mckenzie, I. Mcinnes et al., IL-33 is a chemoattractant for human Th2 cells, European Journal of Immunology, vol.182, issue.10, pp.2779-86, 2007.
DOI : 10.1002/eji.200737547

J. Schmitz, A. Owyang, E. Oldham, Y. Song, E. Murphy et al., IL-33, an Interleukin-1-like Cytokine that Signals via the IL-1 Receptor-Related Protein ST2 and Induces T Helper Type 2-Associated Cytokines, Immunity, vol.23, issue.5, pp.479-90, 2005.
DOI : 10.1016/j.immuni.2005.09.015

T. Ohno, K. Oboki, N. Kajiwara, E. Morii, K. Aozasa et al., Caspase-1, Caspase-8, and Calpain Are Dispensable for IL-33 Release by Macrophages, The Journal of Immunology, vol.183, issue.12
DOI : 10.4049/jimmunol.0802449

C. Cayrol and J. Girard, The IL-1-like cytokine IL-33 is inactivated after maturation by caspase-1, Proceedings of the National Academy of Sciences, vol.106, issue.22, pp.9021-9027, 2009.
DOI : 10.1073/pnas.0812690106

D. Talabot-ayer, C. Lamacchia, C. Gabay, and G. Palmer, Interleukin-33 Is Biologically Active Independently of Caspase-1 Cleavage, Journal of Biological Chemistry, vol.284, issue.29, pp.19420-19426, 2009.
DOI : 10.1074/jbc.M901744200

W. Zhao and Z. Hu, The enigmatic processing and secretion of interleukin-33, Cellular and Molecular Immunology, vol.181, issue.4, pp.260-262, 2010.
DOI : 10.1016/j.bbrc.2009.04.081

G. Palmer, D. Talabot-ayer, C. Lamacchia, D. Toy, C. Seemayer et al., Inhibition of interleukin-33 signaling attenuates the severity of experimental arthritis, Arthritis & Rheumatism, vol.5, issue.3, pp.738-787, 2009.
DOI : 10.1002/art.24305

L. Kolly, M. Karababa, L. Joosten, S. Narayan, R. Salvi et al., Inflammatory Role of ASC in Antigen-Induced Arthritis Is Independent of Caspase-1, NALP-3, and IPAF, The Journal of Immunology, vol.183, issue.6, pp.4003-4015, 2009.
DOI : 10.4049/jimmunol.0802173

S. Ippagunta, D. Brand, J. Luo, K. Boyd, C. Calabrese et al., Inflammasome-independent Role of Apoptosis-associated Speck-like Protein Containing a CARD (ASC) in T Cell Priming Is Critical for Collagen-induced Arthritis, Journal of Biological Chemistry, vol.285, issue.16, pp.12454-62, 2010.
DOI : 10.1074/jbc.M109.093252

M. Kool, V. Petrilli, D. Smedt, T. Rolaz, A. Hammad et al., Cutting Edge: Alum Adjuvant Stimulates Inflammatory Dendritic Cells through Activation of the NALP3 Inflammasome, The Journal of Immunology, vol.181, issue.6, pp.3755-3764, 2008.
DOI : 10.4049/jimmunol.181.6.3755

A. Mckee, M. Munks, M. Macleod, C. Fleenor, N. Van-rooijen et al., Alum Induces Innate Immune Responses through Macrophage and Mast Cell Sensors, But These Sensors Are Not Required for Alum to Act As an Adjuvant for Specific Immunity, The Journal of Immunology, vol.183, issue.7, pp.4403-4417, 2009.
DOI : 10.4049/jimmunol.0900164

B. Stockinger and M. Veldhoen, Differentiation and function of Th17 T cells, Current Opinion in Immunology, vol.19, issue.3, pp.281-287, 2007.
DOI : 10.1016/j.coi.2007.04.005

C. Dong, Differentiation and function of pro-inflammatory Th17 cells. Microbes and infection, Inst Pasteur, vol.11, pp.584-592, 2009.

W. Ouyang, J. Kolls, and Y. Zheng, The Biological Functions of T Helper 17 Cell Effector Cytokines in Inflammation, Immunity, vol.28, issue.4, pp.454-67, 2008.
DOI : 10.1016/j.immuni.2008.03.004

L. Steinman, A brief history of TH17, the first major revision in the TH1/TH2 hypothesis of T cell???mediated tissue damage, Nature Medicine, vol.59, issue.2, pp.139-184, 2007.
DOI : 10.1038/nm1551

C. Dinarello, The biological properties of interleukin-1, Eur Cytokine Netw, vol.5, pp.517-548, 1994.

I. Kryczek, S. Wei, L. Vatan, J. Escara-wilke, W. Szeliga et al., Cutting Edge: Opposite Effects of IL-1 and IL-2 on the Regulation of IL-17+ T Cell Pool IL-1 Subverts IL-2-Mediated Suppression, The Journal of Immunology, vol.179, issue.3, pp.1423-1429, 2007.
DOI : 10.4049/jimmunol.179.3.1423

M. Veldhoen, R. Hocking, C. Atkins, R. 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, issue.2, pp.179-89, 2006.
DOI : 10.1016/j.immuni.2006.01.001

A. Brustle, S. Heink, M. Huber, C. Rosenplanter, C. Stadelmann et al., The development of inflammatory TH-17 cells requires interferon-regulatory factor 4, Nature Immunology, vol.163, issue.9, pp.958-66, 2007.
DOI : 10.1016/j.jneuroim.2006.06.026

K. Ghoreschi, A. Laurence, X. Yang, C. Tato, M. Mcgeachy et al., Generation of pathogenic TH17 cells in the absence of TGF-?? signalling, Nature, vol.282, issue.7318, pp.967-71, 2010.
DOI : 10.1038/nature09447

B. Neven, I. Callebaut, A. Prieur, J. Feldmann, C. Bodemer et al., Molecular basis of the spectral expression of CIAS1 mutations associated with phagocytic cell-mediated autoinflammatory disorders CINCA/NOMID, MWS, and FCU, Blood, vol.103, issue.7, pp.2809-2824, 2004.
DOI : 10.1182/blood-2003-07-2531

URL : https://hal.archives-ouvertes.fr/hal-00086300

S. Brydges, J. Mueller, M. Mcgeough, C. Pena, A. Misaghi et al., Inflammasome-Mediated Disease Animal Models Reveal Roles for Innate but Not Adaptive Immunity, Immunity, vol.30, issue.6, pp.875-87, 2009.
DOI : 10.1016/j.immuni.2009.05.005

A. Dunne, P. Ross, E. Pospisilova, J. Masin, A. Meaney et al., Inflammasome Activation by Adenylate Cyclase Toxin Directs Th17 Responses and Protection against Bordetella pertussis, The Journal of Immunology, vol.185, issue.3, pp.1711-1720, 2010.
DOI : 10.4049/jimmunol.1000105

URL : http://www.tara.tcd.ie/bitstream/2262/57469/1/Inflammasome%20Activation%20by%20Adenylate%20Cyclase%20Toxin%20Directs%20Th17%20Responses%20and%20Protection%20against%20Bordetella%20pertussis.pdf

H. Watanabe, S. Gehrke, E. Contassot, S. Roques, J. Tschopp et al., Danger Signaling through the Inflammasome Acts as a Master Switch between Tolerance and Sensitization, The Journal of Immunology, vol.180, issue.9, pp.5826-5858, 2008.
DOI : 10.4049/jimmunol.180.9.5826

G. Guarda, C. Dostert, F. Staehli, K. Cabalzar, R. Castillo et al., T cells dampen innate immune responses through inhibition of NLRP1 and NLRP3 inflammasomes, Nature, vol.5, issue.7252, pp.269-73, 2009.
DOI : 10.1038/nature08100

A. Zaiss, A. Vilaysane, M. Cotter, S. Clark, H. Meijndert et al., Antiviral Antibodies Target Adenovirus to Phagolysosomes and Amplify the Innate Immune Response, The Journal of Immunology, vol.182, issue.11, pp.7058-68, 2009.
DOI : 10.4049/jimmunol.0804269

D. Gregorio, E. , D. Oro, U. Wack, and A. , Immunology of TLR-independent vaccine adjuvants, Current Opinion in Immunology, vol.21, issue.3, pp.339-384, 2009.
DOI : 10.1016/j.coi.2009.05.003

J. Harris, F. Sharp, and E. Lavelle, The role of inflammasomes in the immunostimulatory effects of particulate vaccine adjuvants, European Journal of Immunology, vol.30, issue.3, pp.634-642, 2010.
DOI : 10.1002/eji.200940172

V. Hornung, F. Bauernfeind, A. Halle, E. Samstad, H. Kono et al., Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization, Nature Immunology, vol.109, issue.8, pp.847-56, 2008.
DOI : 10.1016/0008-8749(88)90236-5

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2834784

L. Franchi and G. Nunez, The Nlrp3 inflammasome is critical for aluminium hydroxide-mediated IL-1?? secretion but dispensable for adjuvant activity, European Journal of Immunology, vol.281, issue.8, pp.2085-2094, 2008.
DOI : 10.1002/eji.200838549

M. Kool, T. Soullie, M. Van-nimwegen, M. Willart, F. Muskens et al., Alum adjuvant boosts adaptive immunity by inducing uric acid and activating inflammatory dendritic cells, The Journal of Experimental Medicine, vol.25, issue.4, pp.869-82, 2008.
DOI : 10.1084/jem.20040035

H. Li, S. Nookala, and F. Re, Aluminum Hydroxide Adjuvants Activate Caspase-1 and Induce IL-1?? and IL-18 Release, The Journal of Immunology, vol.178, issue.8, pp.5271-5277, 2007.
DOI : 10.4049/jimmunol.178.8.5271

F. Sharp, D. Ruane, B. Claass, E. Creagh, J. Harris et al., Uptake of particulate vaccine adjuvants by dendritic cells activates the NALP3 inflammasome, Proceedings of the National Academy of Sciences, vol.106, issue.3, pp.870-875, 2009.
DOI : 10.1073/pnas.0804897106

S. Demento, S. Eisenbarth, H. Foellmer, C. Platt, M. Caplan et al., Inflammasome-activating nanoparticles as modular systems for optimizing vaccine efficacy, Vaccine, vol.27, issue.23, pp.3013-3034, 2009.
DOI : 10.1016/j.vaccine.2009.03.034

T. Morishige, Y. Yoshioka, H. Inakura, A. Tanabe, X. Yao et al., The effect of surface modification of amorphous silica particles on NLRP3 inflammasome mediated IL-1?? production, ROS production and endosomal rupture, Biomaterials, vol.31, issue.26, pp.6833-6875, 2010.
DOI : 10.1016/j.biomaterials.2010.05.036

H. Kumar, Y. Kumagai, T. Tsuchida, P. Koenig, T. Satoh et al., Involvement of the NLRP3 Inflammasome in Innate and Humoral Adaptive Immune Responses to Fungal ??-Glucan, The Journal of Immunology, vol.183, issue.12, pp.8061-8068, 2009.
DOI : 10.4049/jimmunol.0902477

J. Curtsinger, C. Schmidt, A. Mondino, D. Lins, R. Kedl et al., Inflammatory cytokines provide a third signal for activation of naive CD4(+) and CD8(+) T cells, J Immunol, vol.162, pp.3256-62, 1999.

M. Huber, H. Beuscher, P. Rohwer, R. Kurrle, M. Rollinghoff et al., Costimulation via TCR and IL-1 receptor reveals a novel IL-1alpha-mediated autocrine pathway of Th2 cell proliferation, J Immunol, vol.160, pp.4242-4249, 1998.

K. Pape, A. Khoruts, A. Mondino, and M. Jenkins, Inflammatory cytokines enhance the in vivo clonal expansion and differentiation of antigen-activated CD4(+) T cells, J Immunol, vol.159, pp.591-599, 1997.

T. Yoshimoto, H. Mizutani, H. Tsutsui, N. Noben-trauth, K. Yamanaka et al., IL-18 induction of IgE: dependence on CD4+ T cells, IL-4 and STAT6, Nature Immunology, vol.1, issue.2, pp.132-139, 2000.
DOI : 10.1038/77811