E. Eckelhart, W. Warsch, E. Zebedin, O. Simma, D. Stoiber et al., A novel Ncr1-Cre mouse reveals the essential role of STAT5 for NK-cell survival and development, Blood, vol.117, pp.1565-73, 2011.

K. Imada, E. T. Bloom, H. Nakajima, J. A. Horvath-arcidiacono, G. B. Udy et al., Stat5b is essential for natural killer cell-mediated proliferation and cytolytic activity, J Exp Med, vol.188, pp.2067-74, 1998.

H. Guo, A. Samarakoon, B. Vanhaesebroeck, and S. Malarkannan, The p110 of PI3K plays a critical role in NK cell terminal maturation and cytokine/chemokine generation, J Exp Med, vol.205, pp.2419-2454, 2008.

N. Kim, A. Saudemont, L. Webb, M. Camps, T. Ruckle et al., The p110delta catalytic isoform of PI3K is a key player in NK-cell development and cytokine secretion, Blood, vol.110, pp.3202-3210, 2007.

I. Tassi, M. Cella, S. Gilfillan, I. Turnbull, T. G. Diacovo et al., p110gamma and p110delta phosphoinositide 3-kinase signaling pathways synergize to control development and functions of murine NK cells, Immunity, vol.27, pp.214-241, 2007.

R. Polak and M. Buitenhuis, The PI3K/PKB signaling module as key regulator of hematopoiesis: implications for therapeutic strategies in leukemia, Blood, vol.119, pp.911-934, 2012.

A. Saudemont, F. Garçon, H. Yadi, M. Roche-molina, N. Kim et al., p110? and p110? isoforms of phosphoinositide 3-kinase differentially regulate natural killer cell migration in health and disease, Proc Natl Acad Sci U S A, vol.106, pp.5795-800, 2009.

Y. T. Bryceson, M. E. March, H. Ljunggren, and E. O. Long, Activation, coactivation, and costimulation of resting human natural killer cells, Immunol Rev, vol.214, pp.73-91, 2006.

H. Zeng and C. H. Mtor, Curr Opin Immunol, vol.25, pp.347-55, 2013.

T. Baranek, T. Manh, Y. Alexandre, M. A. Maqbool, J. Z. Cabeza et al., Differential responses of immune cells to type I interferon contribute to host resistance to viral infection, Cell Host Microbe, vol.12, pp.571-84, 2012.

P. Brennan, J. W. Babbage, B. M. Burgering, B. Groner, K. Reif et al., Phosphatidylinositol 3-kinase couples the interleukin-2 receptor to the cell cycle regulator E2F, Immunity, vol.7, p.80388, 1997.

E. A. Grimm, R. J. Robb, J. A. Roth, L. M. Neckers, L. B. Lachman et al., Lymphokine-activated killer cell phenomenon. III. Evidence that IL-2 is sufficient for direct activation of peripheral blood lymphocytes into lymphokineactivated killer cells, J Exp Med, vol.158, pp.1356-61, 1983.

J. L. Strausser and S. A. Rosenberg, In vitro growth of cytotoxic human lymphocytes I. Growth of cells sensitized in vitro to alloantigens, J Immunol, vol.121, pp.1491-1496, 1978.

F. Bihl, J. Pecheur, B. Bréart, G. Poupon, J. Cazareth et al., Primed antigen-specific CD4+ T cells are required for NK cell activation in vivo upon Leishmania major infection, J Immunol, issue.185, pp.2174-81, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00724243

F. Granucci, I. Zanoni, N. Pavelka, S. Dommelen, C. E. Van-andoniou et al., A contribution of mouse dendritic cell-derived IL-2 for NK cell activation, J Exp Med, vol.200, pp.287-95, 2004.

G. Gasteiger, S. Hemmers, M. A. Firth, A. L. Floc'h, M. Huse et al., IL-2dependent tuning of NK cell sensitivity for target cells is controlled by regulatory T cells, J Exp Med, 2013.

J. Sitrin, A. Ring, K. C. Garcia, C. Benoist, and D. Mathis, Regulatory T cells control NK cells in an insulitic lesion by depriving them of IL-2, J Exp Med, vol.93, p.2260, 2013.

J. Ni, M. Miller, A. Stojanovic, N. Garbi, and A. Cerwenka, Sustained effector function of IL-12/15/18-preactivated NK cells against established tumors, J Exp Med, vol.209, pp.2351-65, 2012.

G. Gasteiger, S. Hemmers, P. D. Bos, J. C. Sun, and A. Y. Rudensky, IL-2-dependent adaptive control of NK cell homeostasis, J Exp Med, vol.33, p.119, 2013.

S. Lee, M. F. Fragoso, and C. A. Biron, Cutting edge: a novel mechanism bridging innate and adaptive immunity: IL-12 induction of CD25 to form highaffinity IL-2 receptors on NK cells, J Immunol, issue.189, pp.2712-2718, 2012.

K. Ozaki, K. Kikly, D. Michalovich, P. R. Young, and W. J. Leonard, Cloning of a type I cytokine receptor most related to the IL-2 receptor ? chain, Proc Natl Acad Sci U S A, vol.97, pp.11439-11483, 2000.

J. Parrish-novak, S. R. Dillon, A. Nelson, A. Hammond, C. Sprecher et al., Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function, Nature, vol.408, pp.57-63, 2000.

M. T. Kasaian, M. J. Whitters, L. L. Carter, L. D. Lowe, J. M. Jussif et al., IL21 limits NK cell responses and promotes antigen-specific T cell activation: a mediator of the transition from innate to adaptive immunity, Immunity, vol.16, pp.559-69, 2002.

J. Brady, Y. Hayakawa, M. J. Smyth, and S. L. Nutt, IL-21 induces the functional maturation of murine NK cells, J Immunol, vol.172, pp.2048-58, 2004.

J. Brady, S. Carotta, R. Thong, C. J. Chan, Y. Hayakawa et al., The interactions of multiple cytokines control NK cell maturation, J Immunol, vol.185, pp.6679-88, 2010.

M. Strengell, S. Matikainen, J. Sirén, A. Lehtonen, D. Foster et al., IL-21 in synergy with IL-15 or IL-18 enhances IFN-? production in human NK and T cells, J Immunol, vol.170, pp.5464-5473, 2003.

H. Elsaesser, K. Sauer, and D. G. Brooks, IL-21 is required to control chronic viral infection, Science, vol.324, pp.1569-72, 2009.

A. Fröhlich, J. Kisielow, I. Schmitz, S. Freigang, A. T. Shamshiev et al., IL21R on T cells is critical for sustained functionality and control of chronic viral infection, Science, vol.324, pp.1576-80, 2009.

J. S. Yi, M. Du, and A. J. Zajac, A vital role for interleukin-21 in the control of a chronic viral infection, Science, vol.324, pp.1572-1578, 2009.

I. Schmitz, C. Schneider, A. Fröhlich, H. Frebel, D. Christ et al., IL21 restricts virus-driven Treg cell expansion in chronic LCMV infection, PLoS Pathog, vol.9, 2013.

A. Iannello, M. Boulassel, S. Samarani, C. Tremblay, T. E. Routy et al., IL-21 enhances NK cell functions and survival in healthy and HIV-infected patients with minimal stimulation of viral replication, J Leukoc Biol, vol.87, pp.857-67, 2010.

N. Strbo, L. De-armas, H. Liu, M. A. Kolber, M. Lichtenheld et al., IL-21 augments natural killer effector functions in chronically HIV-infected individuals, AIDS, vol.22, pp.1551-60, 2008.
DOI : 10.1097/qad.0b013e3283089367

URL : http://pdfs.journals.lww.com/aidsonline/2008/08200/IL_21_augments_natural_killer_effector_functions.3.pdf?token=method|ExpireAbsolute;source|Journals;ttl|1506127470341;payload|mY8D3u1TCCsNvP5E421JYK6N6XICDamxByyYpaNzk7FKjTaa1Yz22MivkHZqjGP4kdS2v0J76WGAnHACH69s21Csk0OpQi3YbjEMdSoz2UhVybFqQxA7lKwSUlA502zQZr96TQRwhVlocEp/sJ586aVbcBFlltKNKo+tbuMfL73hiPqJliudqs17cHeLcLbV/CqjlP3IO0jGHlHQtJWcICDdAyGJMnpi6RlbEJaRheGeh5z5uvqz3FLHgPKVXJzdN2ziiHhY6wPF9tfA80junvZ7o44/ElLN4EiRYf5ZMfw=;hash|IkNPzKbvBbU2blYdp66bOg==

U. Freeden-jeffry, V. Vieira, P. Lucian, L. A. Mcneil, T. Burdach et al., Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine, J Exp Med, vol.181, pp.1519-1545, 1995.

Y. W. He and T. R. Malek, Interleukin-7 receptor alpha is essential for the development of gamma delta + T cells, but not natural killer cells, J Exp Med, vol.184, pp.289-93, 1996.

Y. Rochman and W. J. Leonard, The role of thymic stromal lymphopoietin in CD8+ T cell homeostasis, J Immunol, vol.181, pp.7699-705, 2008.

C. Vosshenrich, M. E. García-ojeda, S. I. Samson-villéger, V. Pasqualetto, L. Enault et al., A thymic pathway of mouse natural killer cell development characterized by expression of GATA-3 and CD127, Nat Immunol, vol.7, pp.1217-1241, 2006.
URL : https://hal.archives-ouvertes.fr/pasteur-00362040

E. Marcenaro, D. Chiesa, M. Bellora, F. Parolini, S. Millo et al., IL12 or IL-4 prime human NK cells to mediate functionally divergent interactions with dendritic cells or tumors, J Immunol, issue.174, pp.3992-4000, 2005.
DOI : 10.4049/jimmunol.174.7.3992

URL : http://www.jimmunol.org/content/174/7/3992.full.pdf

S. Agaugué, E. Marcenaro, B. Ferranti, L. Moretta, and A. Moretta, Human natural killer cells exposed to IL-2, IL-12, IL-18, or IL-4 differently modulate priming of naive T cells by monocyte-derived dendritic cells, Blood, vol.112, pp.1776-83, 2008.

A. Marçais, M. Tomkowiak, T. Walzer, C. Coupet, A. Ravel-chapuis et al., Maintenance of CCL5 mRNA stores by post-effector and memory CD8 T cells is dependent on transcription and is coupled to increased mRNA stability, Eur J Immunol, vol.36, pp.2745-54, 2006.

A. Marçais, C. Coupet, T. Walzer, M. Tomkowiak, R. Ghittoni et al., Cellautonomous CCL5 transcription by memory CD8 T cells is regulated by IL-4, J Immunol, vol.177, pp.4451-4458, 2006.

E. Ventre, L. Brinza, S. Schicklin, J. Mafille, C. Coupet et al., Negative regulation of NKG2D expression by IL-4 in memory CD8 T cells, J Immunol, vol.189, pp.3480-3489, 2012.

Y. J. Lee, S. C. Jameson, and K. A. Hogquist, Alternative memory in the CD8 T cell lineage, Trends Immunol, vol.32, pp.50-56, 2011.
DOI : 10.1016/j.it.2010.12.004

URL : http://europepmc.org/articles/pmc3039080?pdf=render

M. A. Weinreich, O. A. Odumade, S. C. Jameson, and K. A. Hogquist, T cells expressing the transcription factor PLZF regulate the development of memory-like CD8+ T cells, Nat Immunol, vol.11, pp.709-725, 2010.

S. C. Morris, S. M. Heidorn, D. R. Herbert, C. Perkins, D. A. Hildeman et al., Endogenously produced IL-4 nonredundantly stimulates CD8+ T cell proliferation, J Immunol, vol.182, pp.1429-1467, 2009.
DOI : 10.4049/jimmunol.182.3.1429

URL : http://www.jimmunol.org/content/182/3/1429.full.pdf

D. Vignali and V. K. Kuchroo, IL-12 family cytokines: immunological playmakers, Nat Immunol, vol.13, pp.722-730, 2012.
DOI : 10.1038/ni.2366

URL : http://europepmc.org/articles/pmc4158817?pdf=render

M. Kobayashi, L. Fitz, M. Ryan, R. M. Hewick, S. C. Clark et al., Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes, J Exp Med, vol.170, pp.827-872, 1989.

Y. Hyodo, K. Matsui, N. Hayashi, H. Tsutsui, S. Kashiwamura et al., IL-18 Up-regulates perforin-mediated NK activity without increasing perforin messenger RNA expression by binding to constitutively expressed IL-18 receptor, J Immunol, vol.162, pp.1662-1670, 1999.

M. Cella, D. Scheidegger, K. Palmer-lehmann, P. Lane, A. Lanzavecchia et al., Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation, J Exp Med, vol.184, pp.747-52, 1996.

S. E. Macatonia, N. A. Hosken, M. Litton, P. Vieira, C. S. Hsieh et al., Dendritic cells produce IL-12 and direct the development of Th1 cells from naive CD4+ T cells, J Immunol, vol.154, pp.5071-5080, 1995.

C. S. Hsieh, S. E. Macatonia, C. S. Tripp, S. F. Wolf, O. Garra et al., Development of TH1 CD4+ T cells through IL-12 produced by Listeria-induced macrophages, Science, vol.260, pp.547-556, 1993.

J. S. Orange and C. A. Biron, Characterization of early IL-12, IFN-alphabeta, and TNF effects on antiviral state and NK cell responses during murine cytomegalovirus infection, J Immunol, vol.156, pp.4746-56, 1996.

C. Sousa, S. Hieny, T. Scharton-kersten, D. Jankovic, H. Charest et al., In vivo microbial stimulation induces rapid CD40 ligand-independent production of interleukin 12 by dendritic cells and their redistribution to T cell areas, J Exp Med, vol.186, pp.1819-1848, 1997.

R. S. Goldszmid, P. Caspar, A. Rivollier, S. White, A. Dzutsev et al., NK cell-derived interferon-gamma; orchestrates cellular dynamics and the differentiation of monocytes into dendritic cells at the site of infection, Immunity, vol.36, pp.1047-59, 2012.

X. Ma, J. M. Chow, G. Gri, G. Carra, F. Gerosa et al., The interleukin 12 p40 gene promoter is primed by interferon gamma in monocytic cells, J Exp Med, vol.183, pp.147-57, 1996.

R. B. Mailliard, Y. Son, R. Redlinger, P. T. Coates, A. Giermasz et al., Dendritic cells mediate NK cell help for Th1 and CTL responses: two-signal requirement for the induction of NK cell helper function, J Immunol, vol.171, pp.2366-73, 2003.

S. Kuwajima, T. Sato, K. Ishida, H. Tada, H. Tezuka et al., Interleukin 15dependent crosstalk between conventional and plasmacytoid dendritic cells is essential for CpG-induced immune activation, Nat Immunol, vol.7, pp.740-746, 2006.
DOI : 10.1038/ni1348

C. Borg, A. Jalil, D. Laderach, K. Maruyama, H. Wakasugi et al., NK cell activation by dendritic cells (DCs) requires the formation of a synapse leading to IL-12 polarization in DCs, Blood, vol.104, pp.3267-75, 2004.

C. M. Bacon, D. W. Mcvicar, J. R. Ortaldo, R. C. Rees, O. Shea et al., IL-12) induces tyrosine phosphorylation of JAK2 and TYK2: differential use of Janus family tyrosine kinases by IL-2 and IL-12, Interleukin, vol.12, pp.399-404, 1995.

N. G. Jacobson, S. J. Szabo, R. M. Weber-nordt, Z. Zhong, R. D. Schreiber et al., Interleukin 12 signaling in T helper type 1 (Th1) cells involves tyrosine phosphorylation of signal transducer and activator of transcription (Stat)3 and Stat4, J Exp Med, vol.181, pp.1755-62, 1995.

K. Shimoda, K. Kato, K. Aoki, T. Matsuda, A. Miyamoto et al., Tyk2 plays a restricted role in IFN? signaling, although it is required for IL12-mediated T cell function, Immunity, vol.13, pp.55-63, 2000.

K. M. Page, D. Chaudhary, S. J. Goldman, and M. T. Kasaian, Natural killer cells from protein kinase C ?-/-mice stimulated with interleukin-12 are deficient in production of interferon-?, J Leukoc Biol, vol.83, pp.1267-76, 2008.

J. S. Orange, B. Wang, C. Terhorst, and C. A. Biron, Requirement for natural killer cell-produced interferon gamma in defense against murine cytomegalovirus infection and enhancement of this defense pathway by interleukin 12 administration, J Exp Med, vol.182, pp.1045-56, 1995.

S. Kang, H. Liang, B. Reizis, and R. M. Locksley, Regulation of hierarchical clustering and activation of innate immune cells by dendritic cells, Immunity, vol.29, pp.819-852, 2008.

D. M. Andrews, A. A. Scalzo, W. M. Yokoyama, M. J. Smyth, and M. A. Degli-esposti, Functional interactions between dendritic cells and NK cells during viral infection, Nat Immunol, vol.4, pp.175-81, 2003.
DOI : 10.1038/ni880

J. C. Sun, S. Madera, N. A. Bezman, J. N. Beilke, M. H. Kaplan et al., Proinflammatory cytokine signaling required for the generation of natural killer cell memory, J Exp Med, vol.209, pp.947-54, 2012.

S. Haeberlein, H. Sebald, C. Bogdan, and U. Schleicher, IL-18, but not IL-15, contributes to the IL-12-dependent induction of NK-cell effector functions by Leishmania infantum in vivo, Eur J Immunol, vol.40, pp.1708-1725, 2010.

J. C. Sun, A. Ma, and L. L. Lanier, Cutting edge: IL-15-independent NK cell response to mouse cytomegalovirus infection, J Immunol, vol.183, pp.2911-2915, 2009.
DOI : 10.4049/jimmunol.0901872

URL : http://www.jimmunol.org/content/183/5/2911.full.pdf

A. R. French, H. Sjölin, S. Kim, R. Koka, L. Yang et al., DAP12 signaling directly augments proproliferative cytokine stimulation of NK cells during viral infections, J Immunol, vol.177, pp.4981-90, 2006.
DOI : 10.4049/jimmunol.177.8.4981

URL : https://hal.archives-ouvertes.fr/hal-00165608

S. H. Chan, M. Kobayashi, D. Santoli, B. Perussia, and G. Trinchieri, Mechanisms of IFN-gamma induction by natural killer cell stimulatory factor (NKSF/IL-12)

, Role of transcription and mRNA stability in the synergistic interaction between NKSF and IL-2, J Immunol, vol.148, pp.92-100, 1992.

R. L. Ogilvie, J. R. Sternjohn, B. Rattenbacher, I. A. Vlasova, D. A. Williams et al., Tristetraprolin mediates interferon-? mRNA decay, J Biol Chem, vol.284, pp.11216-11239, 2009.
DOI : 10.1074/jbc.m901229200

URL : http://www.jbc.org/content/284/17/11216.full.pdf

D. F. Steiner, M. F. Thomas, J. K. Hu, Z. Yang, J. E. Babiarz et al., MicroRNA-29 regulates T-box transcription factors and interferon-gamma
DOI : 10.1016/j.immuni.2011.07.009

URL : https://doi.org/10.1016/j.immuni.2011.07.009

, T cells. Immunity, vol.35, pp.169-81, 2011.

A. Mavropoulos, G. Sully, A. P. Cope, and A. R. Clark, Stabilization of IFN-? mRNA by MAPK p38 in IL-12-and IL-18-stimulated human NK cells, Blood, vol.105, pp.282-290, 2005.

D. Robinson, K. Shibuya, A. Mui, F. Zonin, E. Murphy et al., IGIF does not drive Th1 development but synergizes with IL-12 for interferon? production and activates IRAK and NF?B, Immunity, vol.7, pp.571-81, 1997.
DOI : 10.1016/s1074-7613(00)80378-7

URL : https://doi.org/10.1016/s1074-7613(00)80378-7

R. E. Berg, E. Crossley, S. Murray, and J. Forman, Relative contributions of NK and CD8 T cells to IFN-? mediated innate immune protection against Listeria monocytogenes, J Immunol, vol.175, pp.1751-1758, 2005.

M. A. Cooper, J. M. Elliott, P. A. Keyel, L. Yang, J. A. Carrero et al., Cytokineinduced memory-like natural killer cells, Proc Natl Acad Sci U S A, vol.106, pp.1915-1924, 2009.
DOI : 10.1111/j.0105-2896.2010.00891.x

URL : http://europepmc.org/articles/pmc2907109?pdf=render

O. Leary, J. G. Goodarzi, M. Drayton, D. L. Von-andrian, and U. H. , T cell-and B cellindependent adaptive immunity mediated by natural killer cells, Nat Immunol, vol.7, pp.507-523, 2006.

H. Okamura, H. Tsutsui, T. Komatsu, M. Yutsudo, A. Hakura et al., Cloning of a new cytokine that induces IFN-? production by T cells, Nature, vol.378, pp.88-91, 1995.
DOI : 10.1038/378088a0

K. Takeda, H. Tsutsui, T. Yoshimoto, O. Adachi, N. Yoshida et al., Defective NK cell activity and Th1 response in IL-18-deficient mice, Immunity, vol.8, pp.80543-80552, 1998.
DOI : 10.1016/s1074-7613(00)80543-9

URL : https://doi.org/10.1016/s1074-7613(00)80543-9

Y. Wang, G. Chaudhri, R. J. Jackson, and G. Karupiah, IL-12p40 and IL-18 play pivotal roles in orchestrating the cell-mediated immune response to a poxvirus infection, J Immunol, vol.183, pp.3324-3355, 2009.

X. Wei, B. P. Leung, W. Niedbala, D. Piedrafita, G. Feng et al., Altered immune responses and susceptibility to Leishmania major and Staphylococcus aureus infection in IL-18-deficient mice, J Immunol, vol.163, pp.2821-2829, 1999.

J. E. Sims and D. E. Smith, The IL-1 family: regulators of immunity, Nat Rev Immunol, vol.10, pp.89-102, 2010.

J. A. Gracie, S. E. Robertson, and I. B. Mcinnes, Interleukin-18, J Leukoc Biol, vol.73, pp.213-237, 2003.

T. Ghayur, S. Banerjee, M. Hugunin, D. Butler, L. Herzog et al., Caspase-1 processes IFN-?-inducing factor and regulates LPS-induced IFN-? production, Nature, vol.386, pp.619-642, 1997.
DOI : 10.1038/386619a0

Y. Gu, K. Kuida, H. Tsutsui, G. Ku, K. Hsiao et al., Activation of interferon-gamma inducing factor mediated by interleukin-1beta converting enzyme, Science, vol.275, pp.206-215, 1997.
DOI : 10.1126/science.275.5297.206

L. Bossaller, P. Chiang, C. Schmidt-lauber, S. Ganesan, W. J. Kaiser et al., Cutting edge: FAS (CD95) mediates noncanonical IL-1? and IL-18 maturation via caspase-8 in an RIP3-independent manner, J Immunol, issue.189, pp.5508-5520, 2012.

R. Pierini, M. Perret, S. Djebali, C. Juruj, M. Michallet et al., ASC controls IFN-? levels in an IL-18-dependent manner in caspase-1-deficient mice infected with Francisella novicida, J Immunol, pp.3847-57, 0191.
DOI : 10.4049/jimmunol.1203326

URL : https://hal.archives-ouvertes.fr/hal-00965035

S. Sugawara, A. Uehara, T. Nochi, T. Yamaguchi, H. Ueda et al., Neutrophil proteinase 3-mediated induction of bioactive IL-18 secretion by human oral epithelial cells, J Immunol, issue.167, pp.6568-75, 2001.
DOI : 10.4049/jimmunol.167.11.6568

URL : http://www.jimmunol.org/content/167/11/6568.full.pdf

Y. Omoto, K. Yamanaka, K. Tokime, S. Kitano, M. Kakeda et al., Granzyme B is a novel interleukin-18 converting enzyme, J Dermatol Sci, vol.59, pp.129-164, 2010.
DOI : 10.1016/j.jdermsci.2010.05.004

W. Kastenmüller, P. Torabi-parizi, N. Subramanian, and T. Lämmermann, Germain RN. A spatially-organized multicellular innate immune response in lymph nodes limits systemic pathogen spread, Cell, vol.150, pp.1235-1283, 2012.

M. Oertli, M. Sundquist, I. Hitzler, D. B. Engler, I. C. Arnold et al., DCderived IL-18 drives Treg differentiation, murine Helicobacter pylori-specific immune tolerance, and asthma protection, J Clin Invest, vol.122, pp.1082-96, 2012.

R. Spörri, N. Joller, H. Hilbi, and A. Oxenius, A novel role for neutrophils as critical activators of NK cells, J Immunol, vol.181, pp.7121-7151, 2008.

T. T. Pizarro, M. H. Michie, M. Bentz, J. Woraratanadharm, M. F. Smith et al., IL-18, a novel immunoregulatory cytokine, is up-regulated in Crohn's disease: expression and localization in intestinal mucosal cells, J Immunol, issue.162, pp.6829-6864, 1999.

S. Carta, R. Lavieri, and A. Rubartelli, Different members of the IL-1 family come out in different ways: DAMPs vs, cytokines? Front Immunol, vol.4, p.123, 2013.

F. Bellora, R. Castriconi, A. Dondero, G. Reggiardo, L. Moretta et al., The interaction of human natural killer cells with either unpolarized or polarized macrophages results in different functional outcomes, Proc Natl Acad Sci U S A, vol.107, pp.21659-64, 2010.

F. Gerosa, B. Baldani-guerra, C. Nisii, V. Marchesini, G. Carra et al., Reciprocal activating interaction between natural killer cells and dendritic cells, J Exp Med, vol.195, pp.327-360, 2002.

J. Humann and L. L. Lenz, Activation of naive NK cells in response to Listeria monocytogenes requires IL-18 and contact with infected dendritic cells, J Immunol, issue.184, pp.5172-5180, 2010.

N. Lapaque, T. Walzer, S. Méresse, E. Vivier, and J. Trowsdale, Interactions between human NK cells and macrophages in response to Salmonella infection, J Immunol, issue.182, pp.4339-4387, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00380263

D. Piccioli, S. Sbrana, E. Melandri, and N. M. Valiante, Contact-dependent stimulation and inhibition of dendritic cells by natural killer cells, J Exp Med, vol.195, pp.335-376, 2002.

C. Semino, G. Angelini, A. Poggi, and A. Rubartelli, NK/iDC interaction results in IL-18 secretion by DCs at the synaptic cleft followed by NK cell activation and release of the DC maturation factor HMGB1, Blood, vol.106, pp.609-625, 2005.

F. Bellora, R. Castriconi, A. Doni, C. Cantoni, L. Moretta et al., MCSF induces the expression of a membrane-bound form of IL-18 in a subset of human monocytes differentiating in vitro toward macrophages, Eur J Immunol, vol.42, pp.1618-1644, 2012.

D. Novick, S. Kim, G. Fantuzzi, L. L. Reznikov, C. A. Dinarello et al., Interleukin-18 binding protein: a novel modulator of the Th1 cytokine response, Immunity, vol.10, pp.127-163, 1999.

K. Hoshino, H. Tsutsui, T. Kawai, K. Takeda, K. Nakanishi et al., Cutting edge: generation of IL-18 receptor-deficient mice: evidence for IL-1 receptorrelated protein as an essential IL-18 binding receptor, J Immunol, vol.162, pp.5041-5045, 1999.

N. Suzuki, N. Chen, D. G. Millar, S. Suzuki, T. Horacek et al., IL-1 receptor-associated kinase 4 is essential for IL-18-mediated NK and Th1 cell responses, J Immunol, vol.170, pp.4031-4036, 2003.

C. A. Rowland, G. Lertmemongkolchai, A. Bancroft, A. Haque, M. S. Lever et al., Critical role of type 1 cytokines in controlling initial infection with Burkholderia mallei, Infect Immun, vol.74, pp.5333-5373, 2006.

K. Kawakami, Y. Koguchi, M. H. Qureshi, A. Miyazato, S. Yara et al., IL-18 contributes to host resistance against infection with Cryptococcus neoformans in mice with defective IL-12 synthesis through induction of IFN-gamma production by NK cells, J Immunol, issue.165, pp.941-948, 2000.

G. C. Pien, A. R. Satoskar, K. Takeda, S. Akira, and C. A. Biron, Cutting edge: selective IL-18 requirements for induction of compartmental IFN-gamma responses during viral infection, J Immunol, issue.165, pp.4787-91, 2000.

J. Chaix, M. S. Tessmer, K. Hoebe, N. Fuséri, B. Ryffel et al., Cutting edge: priming of NK cells by IL-18, J Immunol, issue.181, pp.1627-1658, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00363717

R. B. Mailliard, S. M. Alber, H. Shen, S. C. Watkins, J. M. Kirkwood et al., IL-18-induced CD83+CCR7+ NK helper cells, J Exp Med, vol.202, pp.941-53, 2005.

J. Schulthess, B. Meresse, E. Ramiro-puig, N. Montcuquet, S. Darche et al., Interleukin-15-dependent NKp46+ innate lymphoid cells control intestinal inflammation by recruiting inflammatory monocytes, Immunity, vol.37, issue.1, pp.108-129, 2012.

M. Terme, E. Ullrich, L. Aymeric, K. Meinhardt, J. D. Coudert et al., Cancer-induced immunosuppression: IL-18-elicited immunoablative NK cells, Cancer Res, vol.72, pp.2757-67, 2012.

I. C. Allen, E. M. Tekippe, R. Woodford, J. M. Uronis, E. K. Holl et al., The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer, J Exp Med, vol.207, pp.1045-56, 2010.

R. Salcedo, A. Worschech, M. Cardone, Y. Jones, Z. Gyulai et al., MyD88-mediated signaling prevents development of adenocarcinomas of the colon: role of interleukin 18, J Exp Med, vol.207, pp.1625-1661, 2010.

J. L. Wong, E. Berk, R. P. Edwards, and P. Kalinski, IL-18-primed helper NK cells collaborate with dendritic cells to promote recruitment of effector CD8+ T cells to the tumor microenvironment, Cancer Res, vol.73, pp.4653-62, 2013.

P. C. Reading, P. G. Whitney, D. P. Barr, M. Wojtasiak, J. D. Mintern et al., IL-18, but not IL-12, regulates NK cell activity following intranasal herpes simplex virus type 1 infection, J Immunol, vol.179, pp.3214-3235, 2007.

M. O. Li, Y. Y. Wan, S. Sanjabi, A. Robertson, and R. A. Flavell, Transforming growth factor-beta regulation of immune responses, Annu Rev Immunol, vol.24, pp.99-146, 2006.

A. Weiss and L. Attisano, The TGFbeta superfamily signaling pathway, Wiley Interdiscip Rev Dev Biol, vol.2, pp.47-63, 2013.

K. Ichiyama, T. Sekiya, N. Inoue, T. Tamiya, I. Kashiwagi et al., Transcription factor Smad-independent T helper 17 cell induction by transforminggrowth factor-? is mediated by suppression of eomesodermin, Immunity, vol.34, pp.741-54, 2011.

T. Espevik, I. S. Figari, G. E. Ranges, and M. A. Palladino, Transforming growth factor-beta 1 (TGF-beta 1) and recombinant human tumor necrosis factoralpha reciprocally regulate the generation of lymphokine-activated killer cell activity. Comparison between natural porcine platelet-derived TGF-beta 1 and TGF-beta 2, and recombinant human TGF-beta 1, J Immunol, issue.140, pp.2312-2318, 1988.

E. A. Grimm, W. L. Crump, . Iii, A. Durett, J. P. Hester et al., TGF-beta inhibits the in vitro induction of lymphokine-activated killing activity, Cancer Immunol Immunother, vol.27, pp.53-61, 1988.

J. J. Mulé, S. L. Schwarz, A. B. Roberts, M. B. Sporn, and S. A. Rosenberg, Transforming growth factor-beta inhibits the in vitro generation of lymphokineactivated killer cells and cytotoxic T cells, Cancer Immunol Immunother, vol.26, pp.95-100, 1988.

A. H. Rook, J. H. Kehrl, L. M. Wakefield, A. B. Roberts, M. B. Sporn et al., Effects of transforming growth factor beta on the functions of natural killer cells: depressed cytolytic activity and blunting of interferon responsiveness, J Immunol, issue.136, pp.3916-3936, 1986.

S. C. Wallick, I. S. Figari, R. E. Morris, A. D. Levinson, and M. A. Palladino, Immunoregulatory role of transforming growth factor beta (TGF-beta) in development of killer cells: comparison of active and latent TGF-beta 1, J Exp Med, vol.172, pp.1777-84, 1990.

H. C. Su, K. A. Leite-morris, L. Braun, and C. A. Biron, A role for transforming growth factor-beta 1 in regulating natural killer cell and T lymphocyte proliferative responses during acute infection with lymphocytic choriomeningitis virus, J Immunol, issue.147, pp.2717-2744, 1991.

C. L. Arteaga, S. D. Hurd, A. R. Winnier, M. D. Johnson, B. M. Fendly et al., Antitransforming growth factor (TGF)-beta antibodies inhibit breast cancer cell tumorigenicity and increase mouse spleen natural killer cell activity. Implications for a possible role of tumor cell/host TGF-beta interactions in human breast cancer progression, J Clin Invest, vol.92, pp.2569-76, 1993.

G. Bellone, M. Aste-amezaga, G. Trinchieri, and U. Rodeck, Regulation of NK cell functions by TGF-beta 1, J Immunol, issue.155, pp.1066-73, 1995.

C. A. Hunter, L. Bermudez, H. Beernink, W. Waegell, and J. S. Remington, Transforming growth factor-beta inhibits interleukin-12-induced production of interferon-gamma by natural killer cells: a role for transforming growth factorbeta in the regulation of T cell-independent resistance to Toxoplasma gondii, Eur J Immunol, vol.25, pp.994-1000, 1995.

S. K. Meadows, M. Eriksson, A. Barber, and C. L. Sentman, Human NK cell IFNgamma production is regulated by endogenous TGF-beta, Int Immunopharmacol, vol.6, pp.1020-1028, 2006.

J. Yu, M. Wei, B. Becknell, R. Trotta, S. Liu et al., Pro-and antiinflammatory cytokine signaling: reciprocal antagonism regulates interferongamma production by human natural killer cells, Immunity, vol.24, pp.575-90, 2006.

R. Trotta, D. Col, J. Yu, J. Ciarlariello, D. Thomas et al., TGFbeta utilizes SMAD3 to inhibit CD16-mediated IFN-gamma production and antibody-dependent cellular cytotoxicity in human NK cells, J Immunol, issue.181, pp.3784-92, 2008.

R. Castriconi, C. Cantoni, D. Chiesa, M. Vitale, M. Marcenaro et al., Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells, Proc Natl Acad Sci U S A, vol.100, pp.4120-4125, 2003.

J. Lee, K. Lee, D. Kim, and D. S. Heo, Elevated TGF-beta1 secretion and downmodulation of NKG2D underlies impaired NK cytotoxicity in cancer patients, J Immunol, issue.172, pp.7335-7375, 2004.

R. Castriconi, A. Dondero, F. Bellora, L. Moretta, A. Castellano et al., Neuroblastoma-derived TGF-?1 modulates the chemokine receptor repertoire of human resting NK cells, J Immunol, pp.5321-5329, 0190.

H. C. Su, R. Ishikawa, and C. A. Biron, Transforming growth factor-beta expression and natural killer cell responses during virus infection of normal, nude, and SCID mice, J Immunol, issue.151, pp.4874-90, 1993.

Y. Laouar, F. S. Sutterwala, L. Gorelik, and R. A. Flavell, Transforming growth factor-beta controls T helper type 1 cell development through regulation of natural killer cell interferon-gamma, Nat Immunol, vol.6, pp.600-607, 2005.

J. P. Marcoe, J. R. Lim, K. L. Schaubert, N. Fodil-cornu, M. Matka et al., TGF-? is responsible for NK cell immaturity during ontogeny and increased susceptibility to infection during mouse infancy, Nat Immunol, vol.13, pp.843-50, 2012.

J. R. Ortaldo, A. T. Mason, O. Shea, J. J. Smyth, M. J. Falk et al., Mechanistic studies of transforming growth factor-beta inhibition of IL-2-dependent activation of CD3-large granular lymphocyte functions. Regulation of IL-2R beta (p75) signal transduction, J Immunol, issue.146, pp.3791-3799, 1991.

D. F. Fiorentino, M. W. Bond, and T. R. Mosmann, Two types of mouse T helper cell. IV. Th2 clones secrete a factor that inhibits cytokine production by Th1 clones, J Exp Med, vol.170, pp.2081-95, 1989.

M. Kamanaka, S. T. Kim, Y. Y. Wan, F. S. Sutterwala, M. Lara-tejero et al., Expression of interleukin-10 in intestinal lymphocytes detected by an interleukin-10 reporter Knockin tiger mouse, Immunity, vol.25, pp.941-52, 2006.

S. Lee, K. Kim, N. Fodil-cornu, S. M. Vidal, and C. A. Biron, Activating receptors promote NK cell expansion for maintenance, IL-10 production, and CD8 T cell regulation during viral infection, J Exp Med, vol.206, pp.2235-51, 2009.

K. W. Moore, R. De-waal-malefyt, R. L. Coffman, O. Garra, and A. , Interleukin-10 and the interleukin-10 receptor, Annu Rev Immunol, vol.19, pp.683-765, 2001.

G. Perona-wright, K. Mohrs, F. M. Szaba, L. W. Kummer, R. Madan et al., Systemic but not local infections elicit immunosuppressive IL-10 production by natural killer cells, Cell Host Microbe, vol.6, pp.503-515, 2009.

G. Cai, R. A. Kastelein, and C. A. Hunter, IL-10 enhances NK cell proliferation, cytotoxicity and production of IFN-? when combined with IL-18, Eur J Immunol, vol.29, pp.2658-65, 1999.

W. E. Carson, M. J. Lindemann, R. Baiocchi, M. Linett, J. C. Tan et al., The functional characterization of interleukin-10 receptor expression on human natural killer cells, Blood, vol.85, pp.3577-85, 1995.

B. Chiu, V. R. Stolberg, and S. W. Chensue, Mononuclear phagocyte-derived IL10 suppresses the innate IL-12/IFN-? axis in lung-challenged aged mice, J Immunol, vol.181, pp.3156-66, 2008.

C. Qian, X. Jiang, H. An, Y. Yu, Z. Guo et al., TLR agonists promote ERK-mediated preferential IL-10 production of regulatory dendritic cells (diffDCs), leading to NK-cell activation, Blood, vol.108, pp.2307-2322, 2006.

Y. Shibata, L. A. Foster, M. Kurimoto, H. Okamura, R. M. Nakamura et al., Immunoregulatory roles of IL-10 in innate immunity: IL-10 inhibits macrophage production of IFN-?-inducing factors but enhances NK cell production of IFN-?, J Immunol, vol.161, pp.4283-4291, 1998.

M. G. Lassen, J. R. Lukens, J. S. Dolina, M. G. Brown, and Y. S. Hahn, Intrahepatic IL-10 maintains NKG2A+Ly49-liver NK cells in a functionally hyporesponsive state, J Immunol, vol.184, pp.2693-701, 2010.

M. A. Stacey, M. Marsden, E. Wang, G. Wilkinson, and I. R. Humphreys, IL10 restricts activation-induced death of NK cells during acute murine cytomegalovirus infection, J Immunol, vol.187, pp.2944-52, 2011.

S. C. Ganal, S. L. Sanos, C. Kallfass, K. Oberle, C. Johner et al., Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota, Immunity, vol.37, pp.171-86, 2012.

M. Gidlund, A. Orn, H. Wigzell, A. Senik, and I. Gresser, Enhanced NK cell activity in mice injected with interferon and interferon inducers, Nature, vol.273, pp.759-61, 1978.

H. Beuneu, J. Deguine, I. Bouvier, J. Santo, M. L. Albert et al., Cutting edge: a dual role for type I IFNs during polyinosinic-polycytidylic acid-induced NK cell activation, J Immunol, vol.187, pp.2084-2092, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-01402066

J. Martinez, X. Huang, and Y. Yang, Direct action of type I IFN on NK cells is required for their activation in response to vaccinia viral infection in vivo, J Immunol, vol.180, pp.1592-1599, 2008.

, Conflict of Interest Statement: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest