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.

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.

S. Aggarwal, N. Ghilardi, M. H. Xie, F. J. De-sauvage, and A. L. Gurney, , 2003.

, Promotes a Distinct CD4 T Cell Activation State Characterized by the Production of Interleukin-17, J Biol Chem, vol.278, pp.1910-1914

P. Agnihotri, N. M. Robertson, S. E. Umetsu, K. Arakcheeva, and S. Winandy, Lack of Ikaros cripples expression of Foxo1 and its targets in naive T cells, Immunology, vol.152, pp.494-506, 2017.

D. V. Agoston, M. Szemes, A. Dobi, M. Palkovits, K. Georgopoulos et al., Ikaros is expressed in developing striatal neurons and involved in enkephalinergic differentiation: Ikaros in striatal development, J Neurochem, vol.102, pp.1805-1816, 2007.

D. Allman, A. Sambandam, S. Kim, J. P. Miller, A. Pagan et al., Thymopoiesis independent of common lymphoid progenitors, Nat Immunol, vol.4, pp.168-174, 2003.

D. Allman, M. Dalod, C. Asselin-paturel, T. Delale, S. H. Robbins et al., Ikaros is required for plasmacytoid dendritic cell differentiation, Blood, vol.108, pp.4025-4034, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00165590

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.

A. Apostolov, I. Litim-mecheri, A. Oravecz, M. Goepp, P. Kirstetter et al., Sumoylation Inhibits the Growth Suppressive Properties of Ikaros, PLoS One, vol.11, 2016.

O. Arican, M. Aral, S. Sasmaz, and P. Ciragil, 12, IL-17, and IL-18 in Patients With Active Psoriasis and Correlation With Disease Severity, Serum Levels of TNF-? , IFN-?, pp.273-279, 2005.

N. Avitahl, S. Winandy, C. Friedrich, B. Jones, Y. Ge et al., Ikaros sets thresholds for T cell activation and regulates chromosome propagation, Immunity, vol.10, pp.333-343, 1999.

T. C. Badea, Y. Wang, and J. Nathans, A noninvasive genetic/pharmacologic strategy for visualizing cell morphology and clonal relationships in the mouse, J Neurosci, vol.23, pp.2314-2322, 2003.

G. Bain, I. Engel, E. C. Robanus-maandag, H. P. Te-riele, J. R. Voland et al., E2A deficiency leads to abnormalities in alphabeta T-cell development and to rapid development of T-cell lymphomas, Mol Cell Biol, vol.17, pp.4782-4791, 1997.

J. C. Barrett, S. Hansoul, D. L. Nicolae, J. H. Cho, R. H. Duerr et al., Genome-wide association defines more than 30 distinct susceptibility loci for Crohn's disease, Nat Genet, vol.40, pp.955-962, 2008.

K. Benlagha, T. Kyin, A. Beavis, L. Teyton, and A. Bendelac, A thymic precursor to the NK T cell lineage, Science, vol.296, pp.553-555, 2002.

K. Benlagha, D. G. Wei, J. Veiga, L. Teyton, and A. Bendelac, Characterization of the early stages of thymic NKT cell development, J Exp Med, vol.202, pp.485-492, 2005.

J. Bentham, D. L. Morris, D. S. Graham, C. L. Pinder, P. Tombleson et al., Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus, Nat Genet, vol.47, pp.1457-1464, 2015.

E. Bettelli, B. Sullivan, S. J. Szabo, R. A. Sobel, L. H. Glimcher et al., Loss of T-bet, But Not STAT1, Prevents the Development of Experimental Autoimmune Encephalomyelitis, J Exp Med, vol.200, pp.79-87, 2004.

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.

S. S. Boggs, M. Trevisan, K. Patrene, G. , and K. , Lack of natural killer cell precursors in fetal liver of Ikaros knockout mutant mice, Nat Immun, vol.16, pp.137-145, 1998.

G. Boschetti, S. Nancey, R. Kanjarawi, T. Almeras, K. Ruel et al., , 2012.

, Réponses immunitaires au cours des MICI : implication de l'axe IL-23/Th17, Hépato Gastro, vol.19, pp.446-454

S. Bottardi, L. Mavoungou, V. Bourgoin, N. Mashtalir, B. Affar-el et al., Direct Protein Interactions Are Responsible for Ikaros-GATA and Ikaros-Cdk9 Cooperativeness in Hematopoietic Cells, Mol Cell Biol, vol.33, pp.3064-3076, 2013.

F. Broere, S. G. Apasov, M. V. Sitkovsky, and W. Van-eden, A2 T cell subsets and T cellmediated immunity, Principles of Immunopharmacology, pp.15-27, 2011.

K. E. Brown, S. S. Guest, S. T. Smale, K. Hahm, M. Merkenschlager et al., , 1997.

, Association of transcriptionally silent genes with Ikaros complexes at centromeric heterochromatin, Cell, vol.91, pp.845-854

A. Brüstle, S. Heink, M. Huber, C. Rosenplänter, C. Stadelmann et al., The development of inflammatory TH-17 cells requires interferonregulatory factor 4, Nat Immunol, vol.8, pp.958-966, 2007.

A. Carbo, R. Hontecillas, T. Andrew, K. Eden, Y. Mei et al., Computational modeling of heterogeneity and function of CD4+ T cells, Front Cell Dev Biol, vol.2, p.31, 2014.

T. Caza and S. Landas, Functional and Phenotypic Plasticity of CD4(+) T Cell Subsets, Biomed Res Int, p.521957, 2015.

M. Cella, A. Fuchs, W. Vermi, F. Facchetti, K. Otero et al., A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity, Nature, vol.457, pp.722-725, 2009.

F. Chalmin, G. Mignot, M. Bruchard, A. Chevriaux, F. Végran et al., Stat3 and Gfi-1 Transcription Factors Control Th17 Cell Immunosuppressive Activity via the Regulation of Ectonucleotidase Expression, Immunity, vol.36, pp.362-373, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00821485

J. M. Coquet, S. Chakravarti, K. Kyparissoudis, F. W. Mcnab, L. A. Pitt et al., Diverse cytokine production by NKT cell subsets and identification of an IL-17-producing CD4-NK1.1-NKT cell population, Proc Natl Acad Sci U S A, vol.105, pp.11287-11292, 2008.

L. Cosmi, F. Liotta, E. Maggi, S. Romagnani, A. et al., Th17 cells: new players in asthma pathogenesis: T lymphocytes and asthma phenotypes, vol.66, pp.989-998, 2011.

S. Crotty, Follicular helper CD4 T cells (TFH), Annu Rev Immunol, vol.29, pp.621-663, 2011.

D. J. Cua, J. Sherlock, Y. Chen, C. A. Murphy, B. Joyce et al., Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain, Nature, vol.421, pp.744-748, 2003.

V. Dardalhon, A. Awasthi, H. Kwon, G. Galileos, W. Gao et al., IL-4 inhibits TGF-?induced Foxp3+ T cells and, together with TGF-?, generates IL-9+ IL-10+ Foxp3? effector T cells, Nat Immunol, vol.9, pp.1347-1355, 2008.

S. Deaglio, K. M. Dwyer, W. Gao, D. Friedman, A. Usheva et al., Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression, J Exp Med, vol.204, pp.1257-1265, 2007.

J. Desbarats, T. Wade, W. F. Wade, and M. K. Newell, Dichotomy between naïve and memory CD4(+) T cell responses to Fas engagement, Proc Natl Acad Sci U S A, vol.96, pp.8104-8109, 1999.

S. Dovat, C. Song, K. J. Payne, L. , and Z. , Ikaros, CK2 kinase, and the road to leukemia, Mol Cell Biochem, vol.356, pp.201-207, 2011.

J. Du, Z. Li, J. Shi, and L. Bi, Associations between serum interleukin-23 levels and clinical characteristics in patients with systemic lupus erythematosus, J Int Med Res, vol.42, pp.1123-1130, 2014.

R. Du, H. Zhao, F. Yan, L. , and H. , IL-17+Foxp3+ T cells: an intermediate differentiation stage between Th17 cells and regulatory T cells, J Leukoc Biol, vol.96, pp.39-48, 2014.

A. Dumortier, R. Jeannet, P. Kirstetter, E. Kleinmann, M. Sellars et al., Notch Activation Is an Early and Critical Event during T-Cell Leukemogenesis in Ikaros-Deficient Mice, Mol Cell Biol, vol.26, pp.209-220, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00187901

A. Dupuis, M. P. Gaub, M. Legrain, B. Drenou, L. Mauvieux et al., Biclonal and biallelic deletions occur in 20% of B-ALL cases with IKZF1 mutations, Leukemia, vol.27, pp.503-507, 2013.

K. M. Dwyer, S. Deaglio, W. Gao, D. Friedman, T. B. Strom et al., CD39 and control of cellular immune responses, Purinergic Signal, vol.3, pp.171-180, 2007.

G. Eberl, S. Marmon, M. J. Sunshine, P. D. Rennert, Y. Choi et al., An essential function for the nuclear receptor ROR?t in the generation of fetal lymphoid tissue inducer cells, Nat Immunol, vol.5, pp.64-73, 2004.

M. El-behi, B. Ciric, H. Dai, Y. Yan, M. Cullimore et al., The encephalitogenicity of TH17 cells is dependent on IL-1-and IL-23-induced production of the cytokine GM-CSF, Nat Immunol, vol.12, pp.568-575, 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.

S. Ezzat, R. Mader, S. Fischer, S. Yu, C. Ackerley et al., An essential role for the hematopoietic transcription factor Ikaros in hypothalamic-pituitary-mediated somatic growth, Proc Natl Acad Sci U S A, vol.103, pp.2214-2219, 2006.

A. Ferraro, C. Socci, A. Stabilini, A. Valle, P. Monti et al., Expansion of Th17 Cells and Functional Defects in T Regulatory Cells Are Key Features of the Pancreatic Lymph Nodes in Patients With Type 1 Diabetes, Diabetes, vol.60, pp.2903-2913, 2011.

D. Fina, M. Sarra, M. C. Fantini, A. Rizzo, R. Caruso et al., Regulation of Gut Inflammation and Th17 Cell Response by Interleukin-21, Gastroenterology, vol.134, pp.1038-1048, 2008.

G. J. Freeman, A. J. Long, Y. Iwai, K. Bourque, T. Chernova et al., Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation, J Exp Med, vol.192, pp.1027-1034, 2000.

S. Fujino, A. Andoh, S. Bamba, A. Ogawa, K. Hata et al., Increased expression of interleukin 17 in inflammatory bowel disease, Gut, vol.52, pp.65-70, 2003.

N. Gagliani, M. C. Vesely, A. Iseppon, L. Brockmann, H. Xu et al., Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation, Nature, vol.523, pp.221-225, 2015.

V. Gateva, J. K. Sandling, G. Hom, K. E. Taylor, S. A. Chung et al., A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus, Nat Genet, vol.41, pp.1228-1233, 2009.

J. Geginat, M. Paroni, S. Maglie, J. S. Alfen, I. Kastirr et al., Plasticity of Human CD4 T Cell Subsets. Front Immunol 5, p.630, 2014.

A. S. Geimer-le-lay, A. Oravecz, J. Mastio, C. Jung, P. Marchal et al., The Tumor Suppressor Ikaros Shapes the Repertoire of Notch Target Genes in T Cells, Sci Signal, vol.7, p.28, 2014.

K. Georgopoulos, D. D. Moore, and B. Derfler, Ikaros, an early lymphoid-specific transcription factor and a putative mediator for T cell commitment, Science, vol.258, pp.808-812, 1992.

K. Georgopoulos, M. Bigby, J. H. Wang, A. Molnar, P. Wu et al., , 1994.

, The Ikaros gene is required for the development of all lymphoid lineages, Cell, vol.79, pp.143-156

K. Ghoreschi, A. Laurence, X. P. Yang, C. M. Tato, M. J. Mcgeachy et al., Generation of pathogenic TH17 cells in the absence of TGF-? signaling, Nature, vol.467, pp.967-971, 2010.

A. Gladiator, N. Wangler, K. Trautwein-weidner, and S. Leibundgut-landmann, Cutting Edge: IL-17-Secreting Innate Lymphoid Cells Are Essential for Host Defense against Fungal Infection, J Immunol, vol.190, pp.521-525, 2013.

J. Glas, J. Seiderer, M. Nagy, C. Fries, F. Beigel et al., Evidence for STAT4 as a Common Autoimmune Gene: rs7574865 Is Associated with Colonic Crohn's Disease and Early Disease Onset, PLoS One, vol.5, 2010.

D. I. Godfrey, J. Kennedy, T. Suda, and A. Zlotnik, A developmental pathway involving four phenotypically and functionally distinct subsets of CD3-CD4-CD8-triple-negative adult mouse thymocytes defined by CD44 and CD25 expression, J Immunol, vol.150, pp.4244-4252, 1993.

P. Gómez-del-arco, K. Maki, G. , and K. , Phosphorylation controls Ikaros's ability to negatively regulate the G(1)-S transition, Mol Cell Biol, vol.24, p.2807, 2004.

A. Henriques, L. Inês, M. Couto, S. Pedreiro, C. Santos et al., Frequency and functional activity of Th17, Tc17 and other T-cell subsets in Systemic Lupus Erythematosus, Cell Immunol, vol.264, pp.97-103, 2010.

K. Hirota, H. Yoshitomi, M. Hashimoto, S. Maeda, S. Teradaira et al., Preferential recruitment of CCR6-expressing Th17 cells to inflamed joints via CCL20 in rheumatoid arthritis and its animal model, J Exp Med, vol.204, pp.2803-2812, 2007.

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.

K. Hirota, J. E. Turner, M. Villa, J. H. Duarte, J. Demengeot et al., Plasticity of TH17 cells in Peyer's patches is responsible for the induction of T cell-dependent IgA responses, Nat Immunol, vol.14, pp.372-379, 2013.

J. Honkanen, J. K. Nieminen, R. Gao, K. Luopajarvi, H. M. Salo et al., IL-17 Immunity in Human Type 1 Diabetes, J Immunol, vol.185, pp.1959-1967, 2010.

A. Hoshino, S. Okada, K. Yoshida, N. Nishida, Y. Okuno et al., Abnormal hematopoiesis and autoimmunity in human subjects with germline IKZF1 mutations, J Allergy Clin Immunol, vol.140, pp.223-231, 2017.

Y. Hu, Z. Zhang, M. Kashiwagi, T. Yoshida, I. Joshi et al., Superenhancer reprogramming drives a B-cell-epithelial transition and high-risk leukemia, Genes Dev, vol.30, pp.1971-1990, 2016.

M. Huber, S. Heink, H. Grothe, A. Guralnik, K. Reinhard et al., A Th17-like developmental process leads to CD8(+) Tc17 cells with reduced cytotoxic activity, Eur J Immunol, vol.39, pp.1716-1725, 2009.

H. Igarashi, S. C. Gregory, T. Yokota, N. Sakaguchi, and P. W. Kincade, Transcription from the RAG1 locus marks the earliest lymphocyte progenitors in bone marrow, Immunity, vol.17, pp.117-130, 2002.

K. Ikuta and I. L. Weissman, Evidence that hematopoietic stem cells express mouse c-kit but do not depend on steel factor for their generation, Proc Natl Acad Sci U S A, vol.89, pp.1502-1506, 1992.

I. I. Ivanov, B. S. Mckenzie, L. Zhou, C. E. Tadokoro, A. Lepelley et al., The Orphan Nuclear Receptor ROR?t Directs the Differentiation Program of Proinflammatory IL-17+ T Helper Cells, Cell, vol.126, pp.1121-1133, 2006.

I. I. Ivanov, K. Atarashi, N. Manel, E. L. Brodie, T. Shima et al., Induction of Intestinal Th17 Cells by Segmented Filamentous Bacteria, Cell, vol.139, pp.485-498, 2009.

A. Jäger, V. Dardalhon, R. A. Sobel, E. Bettelli, and V. K. Kuchroo, Th1, Th17, and Th9 Effector Cells Induce Experimental Autoimmune Encephalomyelitis with Different Pathological Phenotypes, J Immunol, vol.183, pp.7169-7177, 2009.

R. Jeannet, J. Mastio, A. Macias-garcia, A. Oravecz, T. Ashworth et al., , 2010.

, Oncogenic activation of the Notch1 gene by deletion of its promoter in Ikaros-deficient T-ALL, Blood, vol.116, pp.5443-5454

M. K. Jenkins and R. H. Schwartz, Antigen presentation by chemically modified splenocytes induces antigen-specific T cell unresponsiveness in vitro and in vivo, J Exp Med, vol.165, pp.302-319, 1987.

Y. Jin, S. A. Birlea, P. R. Fain, T. M. Ferrara, S. Ben et al., Genome-wide association analyses identify 13 new susceptibility loci for generalized vitiligo, Nat Genet, vol.44, pp.676-680

L. Jostins, S. Ripke, R. K. Weersma, R. H. Duerr, D. P. Mcgovern et al., Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease, Nature, vol.491, pp.119-124, 2012.

C. H. June, J. A. Ledbetter, M. M. Gillespie, T. Lindsten, and C. B. Thompson, T-cell proliferation involving the CD28 pathway is associated with cyclosporine-resistant interleukin 2 gene expression, Mol Cell Biol, vol.7, pp.4472-4481, 1987.

J. Koipally, G. , and K. , A Molecular Dissection of the Repression Circuitry of Ikaros, J Biol Chem, vol.277, pp.27697-27705, 2002.

M. Kondo, I. L. Weissman, and K. Akashi, Identification of clonogenic common lymphoid progenitors in mouse bone marrow, Cell, vol.91, pp.661-672, 1997.

T. Korn, M. Oukka, V. Kuchroo, and E. Bettelli, Th17 cells: Effector T cells with inflammatory properties, Semin Immunol, vol.19, pp.362-371, 2007.

I. D. Krantz, Alagille syndrome: chipping away at the tip of the iceberg, Am J Med Genet, vol.112, pp.160-162, 2002.

T. Krausgruber, C. Schiering, K. Adelmann, O. J. Harrison, A. Chomka et al., T-bet is a key modulator of IL-23-driven pathogenic CD4+ T cell responses in the intestine, Nat Commun, vol.7, p.11627, 2016.

I. Kryczek, S. Wei, W. Szeliga, L. Vatan, and W. Zou, Endogenous IL-17 contributes to reduced tumor growth and metastasis, Blood, vol.114, pp.357-359, 2009.

H. S. Kuehn, B. Boisson, C. Cunningham-rundles, J. Reichenbach, A. Stray-pedersen et al., Loss of B Cells in Patients with Heterozygous Mutations in IKAROS, N Engl J Med, vol.374, pp.1032-1043, 2016.

V. C. Kyttaris, Z. Zhang, V. K. Kuchroo, M. Oukka, and G. C. Tsokos, Cutting edge: IL-23 receptor deficiency prevents the development of lupus nephritis in C57BL/6-lpr/lpr mice, J Immunol, vol.184, pp.4605-4609, 2010.

K. J. Lafferty and A. J. Cunningham, A new analysis of allogeneic interactions, Aust J Exp Biol Med Sci, vol.53, pp.27-42, 1975.

A. Lainé, B. Martin, M. Luka, L. Mir, C. Auffray et al., , 2015.

, Foxo1 Is a T Cell-Intrinsic Inhibitor of the ROR?t-Th17 Program, J Immunol, vol.195, pp.1791-1803

C. L. Langrish, Y. Chen, W. M. Blumenschein, J. Mattson, B. Basham et al., IL-23 drives a pathogenic T cell population that induces autoimmune inflammation, J Exp Med, vol.201, pp.233-240, 2005.

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.

Y. Lee, A. Awasthi, N. Yosef, F. J. Quintana, S. Xiao et al., Induction and molecular signature of pathogenic TH17 cells, Nat Immunol, vol.13, pp.991-999, 2012.

Y. K. Lee, R. Mukasa, R. D. Hatton, and C. T. Weaver, Developmental plasticity of Th17 and Treg cells, Curr Opin Immunol, vol.21, pp.274-280, 2009.

M. O. Li, Y. Y. Wan, and R. A. Flavell, T Cell-Produced Transforming Growth Factor-?1, 2007.

, Controls T Cell Tolerance and Regulates Th1-and Th17-Cell Differentiation, vol.26, pp.579-591

Z. Li, L. A. Perez-casellas, A. Savic, C. Song, and S. Dovat, Ikaros isoforms: The saga continues, World J Biol Chem, vol.2, pp.140-145, 2011.

Z. Liang, K. E. Brown, T. Carroll, B. Taylor, I. F. Vidal et al., A high-resolution map of transcriptional repression, ELife, vol.6, p.22767, 2017.

P. S. Linsley, E. A. Clark, and J. A. Ledbetter, T-cell antigen CD28 mediates adhesion with B cells by interacting with activation antigen B7/BB-1, Proc Natl Acad Sci U S A, vol.87, pp.5031-5035, 1990.

R. Liu, Q. Wu, D. Su, N. Che, H. Chen et al., A regulatory effect of IL-21 on T follicular helper-like cell and B cell in rheumatoid arthritis, Arthritis Res Ther, vol.14, p.255, 2012.

K. Lo, N. R. Landeau, and S. T. Smale, LyF-1, a transcriptional regulator that interacts with a novel class of promoters for lymphocyte-specific genes, Mol Cell Biol, vol.11, pp.5229-5243, 1991.

R. V. Luckheeram, R. Zhou, A. D. Verma, and B. Xia, CD4?T cells: differentiation and functions, Clin Dev Immunol, 2012.

B. Malissen, C. Grégoire, M. Malissen, and R. Roncagalli, Integrative biology of T cell activation, Nat Immunol, vol.15, pp.790-797, 2014.

P. R. Mangan, L. E. Harrington, D. B. O'quinn, W. S. Helms, D. C. Bullard et al., Transforming growth factor-? induces development of the TH17 lineage, Nature, vol.441, pp.231-234, 2006.

A. Marçais, R. Jeannet, L. Hernandez, J. Soulier, F. Sigaux et al., Genetic inactivation of Ikaros is a rare event in human T-ALL, Leuk Res, vol.34, pp.426-429, 2010.

I. Marenholz, J. Esparza-gordillo, F. Rüschendorf, A. Bauerfeind, D. P. Strachan et al., Meta-analysis identifies seven susceptibility loci involved in the atopic march, Nat Commun, vol.6, p.8804, 2015.

B. R. Marks, H. N. Nowyhed, J. Y. Choi, A. C. Poholek, J. M. Odegard et al., Thymic self-reactivity selects natural interleukin 17-producing T cells that can regulate peripheral inflammation, Nat Immunol, vol.10, pp.1125-1132, 2009.

F. Martin, L. Apetoh, and F. Ghiringhelli, Controversies on the role of Th17 in cancer: a TGF?-dependent immunosuppressive activity?, Trends Mol Med, vol.18, pp.742-749, 2012.

N. Martin-orozco, P. Muranski, Y. Chung, X. O. Yang, T. Yamazaki et al., T Helper 17 Cells Promote Cytotoxic T Cell Activation in Tumor Immunity, Immunity, vol.31, pp.787-798, 2009.

K. Masuda, K. Kakugawa, T. Nakayama, N. Minato, Y. Katsura et al., T cell lineage determination precedes the initiation of TCR beta gene rearrangement, J Immunol, vol.179, pp.3699-3706, 2007.

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.

I. L. Mcwilliams, R. Rajbhandari, S. Nozell, E. Benveniste, and L. E. Harrington, STAT4 controls GM-CSF production by both Th1 and Th17 cells during EAE, J Neuroinflammation, vol.12, p.128, 2015.

M. L. Michel, D. Mendes-da-cruz, A. C. Keller, M. Lochner, E. Schneider et al., Critical role of ROR-t in a new thymic pathway leading to IL-17-producing invariant NKT cell differentiation, Proc Natl Acad Sci U S A, vol.105, 2008.

A. Molnár, G. , and K. , The Ikaros gene encodes a family of functionally diverse zinc finger DNA-binding proteins, Mol Cell Biol, vol.14, pp.8292-8303, 1994.

B. Morgan, L. Sun, N. Avitahl, K. Andrikopoulos, T. Ikeda et al., Aiolos, a lymphoid restricted transcription factor that interacts with Ikaros to regulate lymphocyte differentiation, EMBO J, vol.16, pp.2004-2013, 1997.

T. R. Mosmann, H. Cherwinski, M. W. Bond, M. A. Giedlin, and R. L. Coffman, Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins, J Immunol, vol.136, pp.2348-2357, 1986.

D. Mucida, Y. Park, G. Kim, O. Turovskaya, I. Scott et al., , 2007.

T. Reciprocal and T. Regulatory, Cell Differentiation Mediated by Retinoic Acid. Science, vol.317, pp.256-260

C. G. Mullighan, X. Su, J. Zhang, I. Radtke, L. A. Phillips et al., Deletion of IKZF1 and Prognosis in Acute Lymphoblastic Leukemia, N Engl J Med, vol.360, pp.470-480, 2009.

C. A. Murphy, C. L. Langrish, Y. Chen, T. Mcclanahan, R. A. Kastelein et al., Divergent Pro-and Antiinflammatory Roles for IL-23 and IL-12 in Joint Autoimmune Inflammation, J Exp Med, vol.198, pp.1951-1957, 2003.

S. Y. Ng, T. Yoshida, J. Zhang, G. , and K. , Genome-wide Lineage-Specific Transcriptional Networks Underscore Ikaros-Dependent Lymphoid Priming in Hematopoietic Stem Cells, vol.30, pp.493-507, 2009.

A. Nichogiannopoulou, M. Trevisan, S. Neben, C. Friedrich, G. et al., Defects in hemopoietic stem cell activity in Ikaros mutant mice, J Exp Med, vol.190, pp.1201-1214, 1999.

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.

C. A. Notley, J. J. Inglis, S. Alzabin, F. E. Mccann, K. E. Mcnamee et al., Blockade of tumor necrosis factor in collagen-induced arthritis reveals a novel immunoregulatory pathway for Th1 and Th17 cells, J Exp Med, vol.205, pp.2491-2497, 2008.

M. Numasaki, J. Fukushi, M. Ono, S. K. Narula, P. J. Zavodny et al., Interleukin-17 promotes angiogenesis and tumor growth, Blood, vol.101, pp.2620-2627, 2003.

M. Numasaki, M. Watanabe, T. Suzuki, H. Takahashi, A. Nakamura et al., IL-17 enhances the net angiogenic activity and in vivo growth of human non-small cell lung cancer in SCID mice through promoting CXCR-2-dependent angiogenesis, J Immunol, vol.175, pp.6177-6189, 2005.

S. O'brien, R. M. Thomas, G. B. Wertheim, F. Zhang, H. Shen et al., Ikaros Imposes a Barrier to CD8+ T Cell Differentiation by Restricting Autocrine IL-2 Production, J Immunol, vol.192, pp.5118-5129, 2014.

T. Ohsugi, A transgenic mouse model of human T cell leukemia virus type 1-associated diseases, Front Microbiol, vol.4, p.49, 2013.

T. Okazaki, Y. Iwai, and T. Honjo, New regulatory co-receptors: inducible co-stimulator and PD-1, Curr Opin Immunol, vol.14, pp.779-782, 2002.

B. Oppmann, R. Lesley, B. Blom, J. C. Timans, Y. Xu et al., Novel p19 protein engages IL-12p40 to form a cytokine, Immunity, vol.13, issue.23, pp.715-725, 2000.

A. Oravecz, A. Apostolov, K. Polak, B. Jost, S. Le-gras et al., Ikaros mediates gene silencing in T cells through Polycomb repressive complex 2, Nat Commun, vol.6, p.8823, 2015.

P. Papathanasiou, A. C. Perkins, B. S. Cobb, R. Ferrini, R. Sridharan et al., Widespread failure of hematolymphoid differentiation caused by a recessive niche-filling allele of the Ikaros transcription factor, Immunity, vol.19, pp.131-144, 2003.

P. H. Papotto, J. C. Ribot, and B. Silva-santos, IL-17+ ?? T cells as kick-starters of inflammation, Nat Immunol, vol.18, pp.604-611, 2017.

E. Pardali, M. J. Goumans, and P. Dijke, Signaling by members of the TGF-? family in vascular morphogenesis and disease, Trends Cell Biol, vol.20, pp.556-567, 2010.

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.

J. S. Park, M. K. Park, S. Y. Lee, H. J. Oh, M. A. Lim et al., TWEAK promotes the production of Interleukin-17 in rheumatoid arthritis, Cytokine, vol.60, pp.143-149, 2012.

K. J. Payne, G. Huang, E. Sahakian, J. Y. Zhu, N. S. Barteneva et al., Ikaros isoform x is selectively expressed in myeloid differentiation, J Immunol, vol.170, pp.3091-3098, 2003.

E. A. Perotti, K. Georgopoulos, Y. , and T. , An Ikaros Promoter Element with Dual Epigenetic and Transcriptional Activities, PLoS One, vol.10, 2015.

H. E. Porritt, L. L. Rumfelt, S. Tabrizifard, T. M. Schmitt, J. C. Zuniga-pflucker et al., Heterogeneity among DN1 prothymocytes reveals multiple progenitors with different capacities to generate T cell and non-T cell lineages, Immunity, vol.20, pp.735-745, 2004.

J. C. Pui, D. Allman, L. Xu, S. Derocco, F. G. Karnell et al., Notch1 expression in early lymphopoiesis influences B versus T lineage determination, Immunity, vol.11, pp.299-308, 1999.

S. Punt, G. J. Fleuren, E. Kritikou, E. Lubberts, J. B. Trimbos et al., Angels and demons: Th17 cells represent a beneficial response, while neutrophil IL-17 is associated with poor prognosis in squamous cervical cancer, Oncoimmunology, vol.4, p.984539, 2015.

F. J. Quintana, H. Jin, E. J. Burns, M. Nadeau, A. Yeste et al., Aiolos promotes TH17 differentiation by directly silencing Il2 expression, Nat Immunol, vol.13, pp.770-777, 2012.

M. R. Quirion, G. D. Gregory, S. E. Umetsu, S. Winandy, and M. A. Brown, Cutting edge: Ikaros is a regulator of Th2 cell differentiation, J Immunol, vol.182, pp.741-745, 2009.

F. Radtke, A. Wilson, G. Stark, M. Bauer, J. Van-meerwijk et al.,

, Deficient T cell fate specification in mice with an induced inactivation of Notch1, Immunity, vol.10, pp.547-558

K. A. Read, M. D. Powell, C. E. Baker, B. K. Sreekumar, V. M. Ringel-scaia et al., Integrated STAT3 and Ikaros Zinc Finger Transcription Factor Activities Regulate Bcl-6 Expression in CD4 + Th Cells, J Immunol, vol.199, pp.2377-2387, 2017.

K. Renner, S. Hellerbrand, F. Hermann, C. Riedhammer, Y. Talke et al., IL-3 promotes the development of experimental autoimmune encephalitis, JCI Insight, vol.1, p.87157, 2016.

C. W. Roberts and S. H. Orkin, The SWI/SNF complex -chromatin and cancer, Nat Rev Cancer, vol.4, pp.133-142, 2004.

R. Roncagalli, S. Hauri, F. Fiore, Y. Liang, Z. Chen et al., Quantitative proteomics analysis of signalosome dynamics in primary T cells identifies the surface receptor CD6 as a Lat adaptor-independent TCR signaling hub, Nat Immunol, vol.15, pp.384-392, 2014.

L. Rovedatti, T. Kudo, P. Biancheri, M. Sarra, C. H. Knowles et al., Differential regulation of interleukin 17 and interferon production in inflammatory bowel disease, Gut, vol.58, pp.1629-1636, 2009.

S. Sakagushi, T. Yamaguchi, T. Nomura, and M. Ono, Regulatory T Cells and Immune Tolerance, Cell, vol.133, pp.775-787, 2008.

S. Sawa, M. Cherrier, M. Mochner, N. Satoh-takayama, H. J. Fehling et al., Lineage Relationship Analysis of ROR t+ Innate Lymphoid Cells, Science, vol.330, pp.665-669, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-01402753

L. Schirmer, D. Merkler, F. B. König, W. Brück, and C. Stadelmann, Neuroaxonal Regeneration is More Pronounced in Early Multiple Sclerosis than in Traumatic Brain Injury Lesions: Neuroaxonal De-and Regeneration in MS and TBI, Brain Pathol, vol.23, pp.2-12, 2013.

H. Schjerven, J. Mclaughlin, T. L. Arenzana, S. Frietze, D. Cheng et al., Selective regulation of lymphopoiesis and leukemogenesis by individual zinc fingers of Ikaros, Nat Immunol, vol.14, pp.1073-1083, 2013.

N. Schuster and K. Krieglstein, Mechanisms of TGF-?-mediated apoptosis, Cell Tissue Res, vol.307, pp.1-14, 2002.

M. Sellars, B. Reina-san-martin, P. Kastner, and S. Chan, Ikaros controls isotype selection during immunoglobulin class switch recombination, J Exp Med, vol.206, pp.1073-1087, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00387911

K. Shah, W. W. Lee, S. H. Lee, S. H. Kim, S. W. Kang et al., Dysregulated balance of Th17 and Th1 cells in systemic lupus erythematosus, Arthritis Res Ther, vol.12, p.53, 2010.

W. Sheng, F. Yang, Y. Zhou, H. Yang, P. Y. Low et al., STAT5 programs a distinct subset of GM-CSF-producing T helper cells that is essential for autoimmune neuroinflammation, Cell Res, vol.24, pp.1387-1402, 2014.

D. S. Shin, A. Jordan, S. Basu, R. M. Thomas, S. Bandyopadhyay et al., Regulatory T cells suppress CD4+ T cells through NFAT-dependent transcriptional mechanisms, EMBO Rep, vol.15, pp.991-999, 2014.

P. A. Shrikant, R. Rao, Q. Li, J. Kesterson, C. Eppolito et al., Regulating functional cell fates in CD8 T cells, Immunol Res, vol.46, pp.12-22, 2010.

M. R. Shurin, G. V. Shurin, A. Lokshin, Z. R. Yurkovetsky, D. W. Gutkin et al., Intratumoral cytokines/chemokines/growth factors and tumor infiltrating dendritic cells: friends or enemies?, Cancer Metastasis Rev, vol.25, pp.333-356, 2006.

E. Sitnicka, C. Brakebusch, I. L. Martensson, M. Svensson, W. W. Agace et al., Complementary signaling through flt3 and Interleukin-7 Receptor ? Is Indispensable for Fetal and Adult B Cell Genesis, J Exp Med, vol.198, pp.1495-1506, 2003.

E. Soldaini, S. John, S. Moro, J. Bollenbacher, U. Schindler et al., DNA binding site selection of dimeric and tetrameric Stat5 proteins reveals a large repertoire of divergent tetrameric Stat5a binding sites, Mol Cell Biol, vol.20, pp.389-401, 2000.

G. J. Spangrude, S. Heimfeld, and I. L. Weissman, Purification and characterization of mouse hematopoietic stem cells, Science, vol.241, pp.58-62, 1988.

H. Spits, D. Santo, and J. P. , The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling, Nat Immunol, vol.12, pp.21-27, 2011.

C. Stienne, M. F. Michieletto, M. Benamar, N. Carrié, I. Bernard et al., Foxo3 Transcription Factor Drives Pathogenic T Helper 1 Differentiation by Inducing the Expression of Eomes, Immunity, vol.45, pp.774-787, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02631699

B. Stockinger and S. Omenetti, The dichotomous nature of T helper 17 cells, Nat Rev Immunol, 2017.

L. Sun, A. Liu, G. , and K. , Zinc finger-mediated protein interactions modulate Ikaros activity, a molecular control of lymphocyte development, EMBO J, vol.15, pp.5358-5369, 1996.

M. S. Sundrud and A. Rao, Regulation of T Helper 17 Differentiation by Orphan Nuclear Receptors: It's Not Just ROR?t Anymore, Immunity, vol.28, pp.5-7, 2008.

A. D. Swafford, J. M. Howson, L. J. Davison, C. Wallace, D. J. Smyth et al., An Allele of IKZF1 (Ikaros) Conferring Susceptibility to Childhood Acute Lymphoblastic Leukemia Protects Against Type 1 Diabetes, Diabetes, vol.60, pp.1041-1044, 2011.

S. J. Szabo, S. T. Kim, G. L. Costa, X. Zhang, C. G. Fathman et al., A novel transcription factor, T-bet, directs Th1 lineage commitment, Cell, vol.100, pp.655-669, 2000.

J. Tabarkiewicz, K. Pogoda, A. Karczmarczyk, P. Pozarowski, G. et al., The Role of IL-17 and Th17 Lymphocytes in Autoimmune Diseases, Arch Immunol Ther Exp (Warsz), vol.63, pp.435-449, 2015.

T. Taghon, M. A. Yui, R. Pant, R. A. Diamond, and E. V. Rothenberg, Developmental and molecular characterization of emerging beta-and gammadelta-selected pre-T cells in the adult mouse thymus, Immunity, vol.24, pp.53-64, 2006.

D. Tautz, R. Lehmann, H. Schnürch, R. Schuh, E. Seifert et al., Finger protein of novel structure encoded by hunchback, a second member of the gap class of Drosophila segmentation genes, Nature, vol.327, pp.383-389, 1987.

R. M. Thomas, C. Chen, N. Chunder, L. Ma, J. Taylor et al., Ikaros Silences T-bet Expression and Interferon-? Production during T Helper 2 Differentiation, J Biol Chem, vol.285, pp.2545-2553, 2010.

S. E. Umetsu and S. Winandy, Ikaros Is a Regulator of Il10 Expression in CD4+ T Cells, J Immunol, vol.183, pp.5518-5525, 2009.

P. Urbanek, Z. Q. Wang, I. Fetka, E. F. Wagner, and M. Busslinger, Complete block of early B cell differentiation and altered patterning of the posterior midbrain in mice lacking Pax5/BSAP, Cell, vol.79, pp.901-912, 1994.

A. J. Van-beelen, M. B. Teunissen, M. L. Kapsenberg, and E. C. Jong, Interleukin-17 in inflammatory skin disorders, Curr Opin Allergy Clin Immunol, vol.7, pp.374-381, 2007.

J. P. Van-hamburg, O. B. Corneth, S. M. Paulissen, N. Davelaar, P. S. Asmawidjaja et al., IL-17/Th17 mediated synovial inflammation is IL-22 independent, Ann Rheum Dis, vol.72, pp.1700-1707, 2013.

N. Van-panhuys, TCR Signal Strength Alters T-DC Activation and Interaction Times and Directs the Outcome of Differentiation, Front Immunol, vol.7, p.6, 2016.

F. Végran, L. Apetoh, and F. Ghiringhelli, Th9 Cells: A Novel CD4 T-cell Subset in the Immune War against Cancer, Cancer Res, vol.75, pp.475-479, 2015.

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.

M. Veldhoen, C. Uyttenhove, J. Van-snick, H. Helmby, A. Westendorf et al., Transforming growth factor-beta 'reprograms' the differentiation of T helper 2 cells and promotes an interleukin-9 producing subset, Nat Immunol, vol.9, pp.1341-1346, 2008.

D. Vremec, J. Pooley, H. Hochrein, L. Wu, and K. Shortman, CD4 and CD8 expression by dendritic cell subtypes in mouse thymus and spleen, J Immunol, vol.164, pp.2978-2986, 2000.

H. H. Wang, Y. Q. Dai, W. Qiu, Z. Q. Lu, F. H. Peng et al., Interleukin-17-secreting T cells in neuromyelitis optica and multiple sclerosis during relapse, J Clin Neurosci, vol.18, pp.1313-1317, 2011.

J. H. Wang, A. Nichogiannopoulou, L. Wu, L. Sun, A. H. Sharpe et al., Selective defects in the development of the fetal and adult lymphoid system in mice with an Ikaros null mutation, Immunity, vol.5, pp.537-549, 1996.

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

S. Winandy, P. Wu, G. , and K. , A dominant mutation in the Ikaros gene leads to rapid development of leukemia and lymphoma, Cell, vol.83, pp.289-299, 1995.

L. Y. Wong, J. K. Hatfield, and M. A. Brown, Ikaros Sets the Potential for Th17 Lineage Gene Expression through Effects on Chromatin State in Early T Cell Development, J Biol Chem, vol.288, pp.35170-35179, 2013.

L. Wu, C. L. Li, and K. Shortman, Thymic dendritic cell precursors: relationship to the T lymphocyte lineage and phenotype of the dendritic cell progeny, J Exp Med, vol.184, pp.903-911, 1996.

L. Wu, A. Nichogiannopoulou, K. Shortman, G. , and K. , Cell-autonomous defects in dendritic cell populations of Ikaros mutant mice point to a developmental relationship with the lymphoid lineage, Immunity, vol.7, pp.483-492, 1997.

M. Yadav, C. Louvet, D. Davini, J. M. Gardner, M. Martinez-llordella et al., Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo, J Exp Med, vol.209, pp.1713-1722, 2012.

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 TH17 cells, Nature, vol.454, pp.350-352, 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.

J. Ye, R. S. Livergood, and G. Peng, The Role and Regulation of Human Th17 Cells in Tumor Immunity, Am J Pathol, vol.182, pp.10-20, 2013.

H. R. Yen, T. J. Harris, S. Wada, J. F. Grosso, D. Getnet et al., Tc17 CD8 T Cells: Functional Plasticity and Subset Diversity, vol.183, pp.7161-7168, 2009.

N. Yosef, A. K. Shalek, J. T. Gaublomme, H. Jin, Y. Lee et al., Dynamic regulatory network controlling TH17 cell differentiation, Nature, vol.496, pp.461-468, 2013.

T. Yoshida, S. Y. Ng, G. , and K. , Awakening lineage potential by Ikaros-mediated transcriptional priming, Curr Opin Immunol, vol.22, pp.154-160, 2010.

T. Yoshida, E. Landhuis, M. Dose, I. Hazan, J. Zhang et al., Transcriptional regulation of the Ikzf1 locus, Blood, vol.122, pp.3149-3159, 2013.

T. Yoshida, G. , and K. , Ikaros fingers on lymphocyte differentiation, Int J Hematol, vol.100, pp.220-229, 2014.

L. Zhang, Y. G. Li, Y. H. Li, L. Qi, X. G. Liu et al., Increased Frequencies of Th22 Cells as well as Th17 Cells in the Peripheral Blood of Patients with Ankylosing Spondylitis and Rheumatoid Arthritis, PLoS One, vol.7, p.31000, 2012.

W. Zheng and R. A. Flavell, The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells, Cell, vol.89, pp.587-596, 1997.

L. Zhou, I. I. Ivanov, R. Spolski, R. Min, K. Shenderov et al., IL-6 programs TH-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways, Nat Immunol, vol.8, pp.967-974, 2007.

L. Zhou, M. M. Chong, and D. R. Littman, Plasticity of CD4+ T Cell Lineage Differentiation, Immunity, vol.30, pp.646-655, 2009.