I. Inui, M. , G. Martello, and S. Piccolo, MicroRNA control of signal transduction, Nature Reviews Molecular Cell Biology, vol.9, issue.3, pp.255-265252, 2010.
DOI : 10.1038/nrm2868

R. G. Jones and C. B. Thompson, Revving the Engine: Signal Transduction Fuels T Cell Activation, Immunity, vol.27, issue.2, pp.173-178, 2007.
DOI : 10.1016/j.immuni.2007.07.008

URL : http://doi.org/10.1016/j.immuni.2007.07.008

L. P. Kane, V. S. Shapiro, D. Stokoe, and A. Weiss, Induction of NF-??B by the Akt/PKB kinase, Current Biology, vol.9, issue.11, pp.601-604, 1999.
DOI : 10.1016/S0960-9822(99)80265-6

L. P. Kane, P. G. Andres, K. C. Howland, A. K. Abbas, and A. Weiss, Akt provides the CD28 costimulatory signal for up-regulation of IL-2 and IFN-? but not TH2 cytokines, Nature Immunology, vol.70, issue.1, pp.37-44, 2001.
DOI : 10.1038/83144

S. G. Kang, W. H. Liu, P. Lu, H. Y. Jin, H. W. Lim et al., MicroRNAs of the miR-17???92 family are critical regulators of TFH differentiation, Nature Immunology, vol.14, issue.8, pp.849-857, 2013.
DOI : 10.1016/j.immuni.2011.07.015

J. Kaye, M. L. Hsu, M. E. Sauron, S. C. Jameson, N. R. Gascoigne et al., Selective development of CD4+ T cells in transgenic mice expressing a class II MHC-restricted antigen receptor, Nature, vol.341, issue.6244, pp.746-749, 1989.
DOI : 10.1038/341746a0

A. A. Khan, L. A. Penny, Y. Yuzefpolskiy, S. Sarkar, and V. Kalia, MicroRNA-17??92 regulates effector and memory CD8 T-cell fates by modulating proliferation in response to infections, Blood, vol.121, issue.22, pp.4473-4483, 2013.
DOI : 10.1182/blood-2012-06-435412

K. Lee, P. Gudapati, S. Dragovic, C. Spencer, S. Joyce et al., Mammalian Target of Rapamycin Protein Complex 2 Regulates Differentiation of Th1 and Th2 Cell Subsets via Distinct Signaling Pathways, Immunity, vol.32, issue.6, pp.743-753, 2010.
DOI : 10.1016/j.immuni.2010.06.002

Q. J. Li, J. Chau, P. J. Ebert, G. Sylvester, H. Min et al., miR-181a Is an Intrinsic Modulator of T Cell Sensitivity and Selection, Cell, vol.129, issue.1, pp.147-161, 2007.
DOI : 10.1016/j.cell.2007.03.008

P. S. Linsley, W. Brady, L. Grosmaire, A. Aruffo, N. K. Damle et al., Binding of the B cell activation antigen B7 to CD28 costimulates T cell proliferation and interleukin 2 mRNA accumulation, Journal of Experimental Medicine, vol.173, issue.3, pp.721-730, 1991.
DOI : 10.1084/jem.173.3.721

H. C. Liou and K. A. Smith, The roles of c-rel and interleukin-2 in tolerance: a molecular explanation of self???nonself discrimination, Immunology and Cell Biology, vol.177, issue.1, pp.27-32, 2011.
DOI : 10.1074/jbc.M109.052712

A. Liston, L. F. Lu, D. O-'carroll, A. Tarakhovsky, and A. Y. Rudensky, Dicer-dependent microRNA pathway safeguards regulatory T cell function, The Journal of Experimental Medicine, vol.205, issue.9, pp.1993-2004, 2008.
DOI : 10.1038/ng1744

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2526195

G. B. Loeb, A. A. Khan, D. Canner, J. B. Hiatt, J. Shendure et al., Transcriptome-wide miR-155 Binding Map Reveals Widespread Noncanonical MicroRNA Targeting, Molecular Cell, vol.48, issue.5, pp.760-770, 2012.
DOI : 10.1016/j.molcel.2012.10.002

URL : http://doi.org/10.1016/j.molcel.2012.10.002

L. F. Lu, M. P. Boldin, A. Chaudhry, L. L. Lin, K. D. Taganov et al., Function of miR-146a in Controlling Treg Cell-Mediated Regulation of Th1 Responses, Cell, vol.142, issue.6, pp.914-929, 2010.
DOI : 10.1016/j.cell.2010.08.012

A. Mondino and D. L. Mueller, mTOR at the crossroads of T cell proliferation and tolerance, Seminars in Immunology, vol.19, issue.3, pp.162-172, 2007.
DOI : 10.1016/j.smim.2007.02.008

S. A. Muljo, K. M. Ansel, C. Kanellopoulou, D. M. Livingston, A. Rao et al., Aberrant T cell differentiation in the absence of Dicer, The Journal of Experimental Medicine, vol.202, issue.2, pp.261-269, 2005.
DOI : 10.1038/75973

A. K. Nagaraja, C. J. Creighton, Z. Yu, H. Zhu, P. H. Gunaratne et al., A Link between mir-100 and FRAP1/mTOR in Clear Cell Ovarian Cancer, Molecular Endocrinology, vol.24, issue.2, pp.447-463, 2010.
DOI : 10.1210/me.2009-0295

S. Obad, C. O. Santos, A. Petri, M. Heidenblad, O. Broom et al., Silencing of microRNA families by seed-targeting tiny LNAs, Nature Genetics, vol.442, issue.4, pp.371-378, 2011.
DOI : 10.1074/mcp.M800029-MCP200

. We-thank-dr and . Kozma-for-mtor-lox, Loeb for sharing information on Ago2 binding to the 3? UTR of Mtor and Rictor before publication, Drs P. Hexley and J. Elliott for cell sorting, Dr J. Marvel for critical reading of the manuscript, and the members of the Lymphocyte Development Group for discussion and advice The authors have no conflicting financial interests, This work was supported by the Medical Research Council UK and a postdoctoral fellowship from Fondation Pour la Recherche Médicale (AM)

K. Araki, A. H. Ellebedy, and R. Ahmed, TOR in the immune system, Current Opinion in Cell Biology, vol.23, issue.6, pp.707-715, 2011.
DOI : 10.1016/j.ceb.2011.08.006

D. Baumjohann, R. Kageyama, J. M. Clingan, M. M. Morar, S. Patel et al., The microRNA cluster miR-17???92 promotes TFH cell differentiation and represses subset-inappropriate gene expression, Nature Immunology, vol.177, issue.8, pp.840-848, 2013.
DOI : 10.1084/jem.20040139

Y. Bronevetsky, A. V. Villarino, C. J. Eisley, R. Barbeau, A. J. Barczak et al., T cell activation induces proteasomal degradation of Argonaute and rapid remodeling of the microRNA repertoire, The Journal of Experimental Medicine, vol.15, issue.2, pp.417-432, 2013.
DOI : 10.1111/j.1600-065X.2010.00951.x

M. M. Chong, J. P. Rasmussen, A. Y. Rudensky, and D. R. Littman, The RNAseIII enzyme Drosha is critical in T cells for preventing lethal inflammatory disease, The Journal of Experimental Medicine, vol.205, issue.9, pp.2005-2017, 2008.
DOI : 10.1126/science.1065062

B. S. Cobb, T. B. Nesterova, E. Thompson, A. Hertweck, E. O-'connor et al., T cell lineage choice and differentiation in the absence of the RNase III enzyme Dicer, The Journal of Experimental Medicine, vol.19, issue.9, pp.1367-1373, 2005.
DOI : 10.1038/ng1351

B. S. Cobb, A. Hertweck, J. Smith, E. O-'connor, D. Graf et al., A role for Dicer in immune regulation, The Journal of Experimental Medicine, vol.27, issue.11, pp.2519-2527, 2006.
DOI : 10.1101/gad.816400

J. Cox and M. Mann, MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification, Nature Biotechnology, vol.7, issue.12, pp.1367-1372, 2008.
DOI : 10.1038/nprot.2007.261

G. M. Delgoffe, K. N. Pollizzi, A. T. Waickman, E. Heikamp, D. J. Meyers et al., The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2, Nature Immunology, vol.46, issue.4, pp.295-303, 2005.
DOI : 10.1038/ni.2005

D. R. Desilva, K. B. Urdahl, and M. K. Jenkins, Clonal anergy is induced in vitro by T cell receptor occupancy in the absence of proliferation, J. Immunol, vol.147, pp.3261-3267, 1991.

F. Fornari, M. Milazzo, P. Chieco, M. Negrini, G. A. Calin et al., MiR-199a-3p Regulates mTOR and c-Met to Influence the Doxorubicin Sensitivity of Human Hepatocarcinoma Cells, Cancer Research, vol.70, issue.12, pp.5184-5193, 2010.
DOI : 10.1158/0008-5472.CAN-10-0145

J. Graumann, N. C. Hubner, J. B. Kim, K. Ko, M. Moser et al., Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) and Proteome Quantitation of Mouse Embryonic Stem Cells to a Depth of 5,111 Proteins, Molecular & Cellular Proteomics, vol.7, issue.4, pp.672-683, 2008.
DOI : 10.1074/mcp.M700460-MCP200

F. A. Harding, J. G. Mcarthur, J. A. Gross, D. H. Raulet, J. P. Allison et al., CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones, Nature, vol.356, issue.6370, pp.607-609, 1992.
DOI : 10.1038/356607a0

C. B. Thompson, T. Lindsten, J. A. Ledbetter, S. L. Kunkel, H. A. Young et al., CD28 activation pathway regulates the production of multiple T-cell-derived lymphokines/cytokines., Proc. Natl, 1989.
DOI : 10.1073/pnas.86.4.1333

J. Tsang, J. Zhu, and A. Van-oudenaarden, MicroRNA-Mediated Feedback and Feedforward Loops Are Recurrent Network Motifs in Mammals, Molecular Cell, vol.26, issue.5, 2007.
DOI : 10.1016/j.molcel.2007.05.018

URL : http://doi.org/10.1016/j.molcel.2007.05.018

T. Tsuruta, K. Kozaki, A. Uesugi, M. Furuta, A. Hirasawa et al., miR-152 Is a Tumor Suppressor microRNA That Is Silenced by DNA Hypermethylation in Endometrial Cancer, Cancer Research, vol.71, issue.20, pp.6450-6462, 2011.
DOI : 10.1158/0008-5472.CAN-11-0364

A. Uesugi, K. Kozaki, T. Tsuruta, M. Furuta, K. Morita et al., The Tumor Suppressive MicroRNA miR-218 Targets the mTOR Component Rictor and Inhibits AKT Phosphorylation in Oral Cancer, Cancer Research, vol.71, issue.17, pp.5765-5778, 2011.
DOI : 10.1158/0008-5472.CAN-11-0368

A. D. Wells, New Insights into the Molecular Basis of T Cell Anergy: Anergy Factors, Avoidance Sensors, and Epigenetic Imprinting, The Journal of Immunology, vol.182, issue.12, pp.7331-7341, 2009.
DOI : 10.4049/jimmunol.0803917

C. Xiao, D. P. Calado, G. Galler, T. H. Thai, H. C. Patterson et al., MiR-150 Controls B Cell Differentiation by Targeting the Transcription Factor c-Myb, Cell, vol.131, issue.1, pp.146-159, 2007.
DOI : 10.1016/j.cell.2007.07.021

F. Xiao, Z. Zuo, G. Cai, S. Kang, X. Gao et al., miRecords: an integrated resource for microRNA-target interactions, Nucleic Acids Research, vol.37, issue.Database, pp.105-110, 2009.
DOI : 10.1093/nar/gkn851

L. Yang, M. P. Boldin, Y. Yu, C. S. Liu, C. K. Ea et al., controls the resolution of T cell responses in mice, The Journal of Experimental Medicine, vol.16, issue.9, pp.1655-1670, 20112218.
DOI : 10.1073/pnas.1105398108

H. Zeng, K. Yang, C. Cloer, G. Neale, P. Vogel et al., mTORC1 couples immune signals and metabolic programming to establish Treg-cell function, Proc. Natl. Acad. Sci. USA, pp.485-49021629, 2010.
DOI : 10.1073/pnas.0506580102

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3759242

Y. Zheng, S. L. Collins, M. A. Lutz, A. N. Allen, T. P. Kole et al., A Role for Mammalian Target of Rapamycin in Regulating T Cell Activation versus Anergy, The Journal of Immunology, vol.178, issue.4, pp.2163-2170, 2007.
DOI : 10.4049/jimmunol.178.4.2163

X. Zhou, L. T. Jeker, B. T. Fife, S. Zhu, M. S. Anderson et al., Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity, The Journal of Experimental Medicine, vol.27, issue.9, pp.1983-1991, 2008.
DOI : 10.1006/geno.2000.6451

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2526194

O. N. Ozes, L. D. Mayo, J. A. Gustin, S. R. Pfeffer, L. M. Pfeffer et al., NF-kappaB activation by tumour necrosis factor requires the Akt serine-threonine kinase, Nature, vol.401, issue.6748, pp.82-85, 1999.
DOI : 10.1038/43466

M. Paolino and J. M. Penninger, Cbl-b in T-cell activation, Seminars in Immunopathology, vol.5, issue.2, pp.137-148, 2010.
DOI : 10.1007/s00281-010-0197-9

J. D. Powell and G. M. Delgoffe, The Mammalian Target of Rapamycin: Linking T Cell Differentiation, Function, and Metabolism, Immunity, vol.33, issue.3, pp.301-311, 2010.
DOI : 10.1016/j.immuni.2010.09.002

J. D. Powell, C. G. Lerner, and R. H. Schwartz, Inhibition of cell cycle progression by rapamycin induces T cell clonal anergy even in the presence of costimulation, J. Immunol, vol.162, pp.2775-2784, 1999.

C. Procaccini, V. De-rosa, M. Galgani, L. Abanni, G. Calì et al., An Oscillatory Switch in mTOR Kinase Activity Sets Regulatory T Cell Responsiveness, Immunity, vol.33, issue.6, pp.929-941, 2010.
DOI : 10.1016/j.immuni.2010.11.024

I. Puga, A. Rao, and F. Macian, Targeted Cleavage of Signaling Proteins by Caspase 3 Inhibits T Cell Receptor Signaling in Anergic T??Cells, Immunity, vol.29, issue.2, pp.193-204, 2008.
DOI : 10.1016/j.immuni.2008.06.010

V. Risson, L. Mazelin, M. Roceri, H. Sanchez, V. Moncollin et al., Muscle inactivation of mTOR causes metabolic and dystrophin defects leading to severe myopathy, The Journal of Cell Biology, vol.75, issue.6, pp.859-874, 2009.
DOI : 10.1083/jcb.200903131.dv

R. Sandberg, J. R. Neilson, A. Sarma, P. A. Sharp, and C. B. Burge, Proliferating Cells Express mRNAs with Shortened 3' Untranslated Regions and Fewer MicroRNA Target Sites, Science, vol.320, issue.5883, pp.1643-1647, 2008.
DOI : 10.1126/science.1155390

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2587246

D. D. Sarbassov, S. M. Ali, S. Sengupta, J. H. Sheen, P. P. Hsu et al., Prolonged Rapamycin Treatment Inhibits mTORC2 Assembly and Akt/PKB, Molecular Cell, vol.22, issue.2, pp.159-168, 2006.
DOI : 10.1016/j.molcel.2006.03.029

URL : http://doi.org/10.1016/j.molcel.2006.03.029

M. Selbach, B. Schwanhäusser, N. Thierfelder, Z. Fang, R. Khanin et al., Widespread changes in protein synthesis induced by microRNAs http://dx.doi.org/10 MicroRNA-29 regulates T-box transcription factors and interferon-? production in helper T cells, Nature. Immunity, vol.455, issue.35, pp.58-63169, 1038.

K. D. Taganov, M. P. Boldin, K. J. Chang, and D. Baltimore, NF-kappaBdependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses, Proc. Natl. Acad. Sci. USA, pp.12481-12486, 2006.