D. Mathis and C. Benoist, Back to Central Tolerance, Immunity, vol.20, pp.509-516, 2004.

C. C. Goodnow, J. Sprent, B. F. De-st-groth, and C. G. Vinuesa, Cellular and genetic mechanisms of self tolerance and autoimmunity, Nature, vol.435, pp.590-597, 2005.

L. Klein, B. Kyewski, P. M. Allen, and K. A. Hogquist, Positive and negative selection of the T cell repertoire: what thymocytes see (and don't see), Nat Rev Immunol, vol.14, pp.377-391, 2014.

J. Gäbler, J. Arnold, and B. Kyewski, Promiscuous gene expression and the developmental dynamics of medullary thymic epithelial cells, Eur. J. Immunol, vol.37, pp.3363-3372, 2007.

Y. Hamazaki, Medullary thymic epithelial cells expressing Aire represent a unique lineage derived from cells expressing claudin, Nat. Immunol, vol.8, pp.304-311, 2007.

J. Gill, Thymic generation and regeneration, Immunological Reviews, vol.195, pp.28-50, 2003.

L. Klein, M. Hinterberger, G. Wirnsberger, and B. Kyewski, Antigen presentation in the thymus for positive selection and central tolerance induction, Nat Rev Immunol, vol.9, pp.833-844, 2009.

S. N. Sansom, Population and single-cell genomics reveal the Aire dependency, relief from Polycomb silencing, and distribution of self-antigen expression in thymic epithelia, Genome Res, vol.24, pp.1918-1931, 2014.

J. Derbinski, A. Schulte, B. Kyewski, and L. Klein, Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self, Nat. Immunol, vol.2, pp.1032-1039, 2001.

M. S. Anderson, Projection of an Immunological Self Shadow Within the Thymus by the Aire Protein, Science, vol.298, pp.1395-1401, 2002.

K. Nagamine, Positional cloning of the APECED gene, Nat. Genet, vol.17, pp.393-398, 1997.

P. Peterson, J. Pitkanen, N. Sillanpaa, and K. Krohn, Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy (APECED): a model disease to study molecular aspects of endocrine autoimmunity, Clin. Exp. Immunol, vol.135, pp.348-357, 2004.

T. Org, The autoimmune regulator PHD finger binds to non-methylated histone H3K4 to activate gene expression, EMBO Rep, vol.9, pp.370-376, 2008.

A. S. Koh, Aire employs a histone-binding module to mediate immunological tolerance, linking chromatin regulation with organ-specific autoimmunity, Proc Natl Acad Sci, vol.105, pp.15878-15883, 2008.

M. Waterfield, The transcriptional regulator Aire coopts the repressive ATF7ip-MBD1 complex for the induction of immunotolerance, Nat. Immunol, vol.15, pp.258-265, 2014.

I. Oven, AIRE Recruits P-TEFb for Transcriptional Elongation of Target Genes in Medullary Thymic Epithelial Cells, Mol Cell Biol, vol.27, pp.8815-8823, 2007.

M. Giraud, Aire unleashes stalled RNA polymerase to induce ectopic gene expression in thymic epithelial cells, Proc Natl Acad Sci, vol.109, pp.535-540, 2012.

M. Giraud, An RNAi screen for Aire cofactors reveals a role for Hnrnpl in, Experimental Medicine, vol.175, pp.1601-1605, 1992.

M. Giraud, An RNAi screen for Aire cofactors reveals a role for Hnrnpl in polymerase release and Aire-activated ectopic transcription, Proc Natl Acad Sci, vol.111, pp.1491-1496, 2014.

M. Giraud, Aire unleashes stalled RNA polymerase to induce ectopic gene expression in thymic epithelial cells, Proc Natl Acad Sci, vol.109, pp.535-540, 2012.

H. Rodewald, Thymus organogenesis, Annu. Rev. Immunol, vol.26, pp.355-388, 2008.

G. Anderson and Y. Takahama, Thymic epithelial cells: working class heroes for T cell development and repertoire selection, Trends Immunol, vol.33, pp.256-263, 2012.

K. Aschenbrenner, Selection of Foxp3 + regulatory T cells specific for self antigen expressed and presented by Aire + medullary thymic epithelial cells, Nat. Immunol, vol.8, pp.351-358, 2007.

J. E. Cowan, The thymic medulla is required for Foxp3 + regulatory but not conventional CD4 + thymocyte development, J. Exp. Med, vol.210, pp.675-681, 2013.

L. Klein, B. Kyewski, P. M. Allen, and K. A. Hogquist, Positive and negative selection of the T cell repertoire: what thymocytes see (and don't see), Nat. Rev. Immunol, vol.14, pp.377-391, 2014.

J. Derbinski, A. Schulte, B. Kyewski, and L. Klein, Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self, Nat. Immunol, vol.2, pp.1032-1039, 2001.

M. S. Anderson, Projection of an immunological self shadow within the thymus by the aire protein, Science, vol.298, pp.1395-1401, 2002.

C. Ramsey, Aire deficient mice develop multiple features of APECED phenotype and show altered immune response, Hum. Mol. Genet, vol.11, pp.397-409, 2002.

K. Nagamine, Positional cloning of the APECED gene, Nat. Genet, vol.17, pp.393-398, 1997.

D. Mathis, C. Benoist, and . Aire, Annu. Rev. Immunol, vol.27, pp.287-312, 2009.

P. Peterson, T. Org, and A. Rebane, Transcriptional regulation by AIRE: molecular mechanisms of central tolerance, Nat. Rev. Immunol, vol.8, pp.948-957, 2008.

T. Org, The autoimmune regulator PHD finger binds to non-methylated histone H3K4 to activate gene expression, EMBO Rep, vol.9, pp.370-376, 2008.

A. S. Koh, Aire employs a histone-binding module to mediate immunological tolerance, linking chromatin regulation with organ-specific autoimmunity, Proc. Natl. Acad. Sci. USA 105, pp.15878-15883, 2008.

T. Org, AIRE activated tissue specific genes have histone modifications associated with inactive chromatin, Hum. Mol. Genet, vol.18, pp.4699-4710, 2009.

M. Giraud, Aire unleashes stalled RNA polymerase to induce ectopic gene expression in thymic epithelial cells, Proc. Natl. Acad. Sci. USA, vol.109, pp.535-540, 2012.

I. Oven, AIRE recruits P-TEFb for transcriptional elongation of target genes in medullary thymic epithelial cells, Mol. Cell. Biol, vol.27, pp.8815-8823, 2007.

J. Abramson, M. Giraud, C. Benoist, and D. Mathis, Aire's partners in the molecular control of immunological tolerance, Cell, vol.140, pp.123-135, 2010.

I. Liiv, DNA-PK contributes to the phosphorylation of AIRE: importance in transcriptional activity, Biochim. Biophys. Acta, vol.1783, pp.74-83, 2008.

K. ?umer, A. K. Low, H. Jiang, K. Saksela, and B. M. Peterlin, Unmodified histone H3K4 and DNA-dependent protein kinase recruit autoimmune regulator to target genes, Mol. Cell. Biol, vol.32, pp.1354-1362, 2012.

M. Waterfield, The transcriptional regulator Aire coopts the repressive ATF7ip-MBD1 complex for the induction of immunotolerance, Nat. Immunol, vol.15, pp.258-265, 2014.

D. Herranz and M. Serrano, SIRT1: recent lessons from mouse models, Nat. Rev. Cancer, vol.10, pp.819-823, 2010.

G. Afshar and J. P. Murnane, Characterization of a human gene with sequence homology to Saccharomyces cerevisiae SIR2, Gene, vol.234, pp.161-168, 1999.

J. M. Gardner, Deletional tolerance mediated by extrathymic Aire-expressing cells, Science, vol.321, pp.843-847, 2008.

H. Li, SirT1 modulates the estrogen-insulin-like growth factor-1 signaling for postnatal development of mammary gland in mice, Breast Cancer Res, vol.9, p.1, 2007.

J. Gordon, Specific expression of lacZ and cre recombinase in fetal thymic epithelial cells by multiplex gene targeting at the Foxn1 locus, BMC Dev. Biol, vol.7, p.69, 2007.

J. N. Feige and J. Auwerx, Transcriptional targets of sirtuins in the coordination of mammalian physiology, Curr. Opin. Cell Biol, vol.20, pp.303-309, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00311100

T. Zhang and W. L. Kraus, SIRT1-dependent regulation of chromatin and transcription: linking NAD + metabolism and signaling to the control of cellular functions, Biochim. Biophys. Acta, vol.1804, pp.1666-1675, 2010.

M. W. Mcburney, The mammalian SIR2 protein has a role in embryogenesis and gametogenesis, Mol. Cell. Biol, vol.23, pp.38-54, 2003.

E. L. Seifert, SirT1 catalytic activity is required for male fertility and metabolic homeostasis in mice, FASEB J, vol.26, pp.555-566, 2012.

M. Saare, A. Rebane, B. Rajashekar, J. Vilo, and P. Peterson, Autoimmune regulator is acetylated by transcription coactivator CBP/p300, Exp. Cell Res, vol.318, pp.1767-1778, 2012.

W. Jiang, M. S. Anderson, R. Bronson, D. Mathis, and C. Benoist, Modifier loci condition autoimmunity provoked by Aire deficiency, J. Exp. Med, vol.202, pp.805-815, 2005.

M. Guerau-de-arellano, M. Martinic, C. Benoist, and D. Mathis, Neonatal tolerance revisited: a perinatal window for Aire control of autoimmunity, J. Exp. Med, vol.206, pp.1245-1252, 2009.

M. S. Anderson, The cellular mechanism of Aire control of T cell tolerance, Immunity, vol.23, pp.227-239, 2005.

J. Sequeira, sirt1-null mice develop an autoimmune-like condition, Exp. Cell Res, vol.314, pp.3069-3074, 2008.

J. Zhang, The type III histone deacetylase Sirt1 is essential for maintenance of T cell tolerance in mice, J. Clin. Invest, vol.119, pp.3048-3058, 2009.

S. Kong, M. W. Mcburney, and D. Fang, Sirtuin 1 in immune regulation and autoimmunity, Immunol. Cell Biol, vol.90, pp.6-13, 2012.

U. H. Beier, Sirtuin-1 targeting promotes Foxp3 + T-regulatory cell function and prolongs allograft survival, Mol. Cell. Biol, vol.31, pp.1022-1029, 2011.

T. Akimova, Targeting sirtuin-1 alleviates experimental autoimmune colitis by induction of Foxp3 + T-regulatory cells, Mucosal Immunol, 2014.

J. Sakamoto, T. Miura, K. Shimamoto, and Y. Horio, Predominant expression of Sir2alpha, an NAD-dependent histone deacetylase, in the embryonic mouse heart and brain, FEBS Lett, vol.556, pp.281-286, 2004.

A. Biason-lauber, Identification of a SIRT1 mutation in a family with type 1 diabetes, Cell Metab, vol.17, pp.448-455, 2013.

E. S. Husebye and M. S. Anderson, Autoimmune polyendocrine syndromes: clues to type 1 diabetes pathogenesis, Immunity, vol.32, pp.479-487, 2010.

S. Amor, N. Groome, C. Linington, M. M. Morris, K. Dornmair et al., Identification of epitopes of myelin oligodendrocyte glycoprotein for the induction of experimental allergic encephalomyelitis in SJL and Biozzi AB/H mice, J. Immunol, vol.153, pp.4349-4356, 1994.

S. Anders and W. Huber, DESeq: Differential expression analysis for sequence count data, Genome Biol, vol.11, p.106, 2010.

G. Anderson and Y. Takahama, Thymic epithelial cells: working class heroes for T cell development and repertoire selection, Trends Immunol, vol.33, pp.256-263, 2012.

A. C. Anderson, L. B. Nicholson, K. L. Legge, V. Turchin, H. Zaghouani et al., High Frequency of Autoreactive Myelin Proteolipid Protein-Specific T Cells in the Periphery of Naive Mice: Mechanisms of Selection of the Self-Reactive Repertoire, J. Exp. Med, vol.191, pp.761-770, 2000.

M. S. Anderson, E. S. Venanzi, L. Klein, Z. Chen, S. P. Berzins et al., Projection of an immunological self shadow within the thymus by the aire protein, Science, vol.298, pp.1395-1401, 2002.

K. Aschenbrenner, L. M. Cruz, E. H. Vollmann, M. Hinterberger, J. Emmerich et al., Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells, Nat. Immunol, vol.8, pp.351-358, 2007.

L. Bazak, A. Haviv, M. Barak, J. Jacob-hirsch, P. Deng et al., A-to-I RNA editing occurs at over a hundred million genomic sites, located in a majority of human genes, Genome Res, vol.24, pp.365-376, 2014.

V. Blanc and N. O. Davidson, APOBEC-1-mediated RNA editing, Wiley Interdiscip. Rev. Syst. Biol. Med, vol.2, pp.594-602

V. Blanc, E. Park, S. Schaefer, M. Miller, Y. Lin et al., Genome-wide identification and functional analysis of Apobec-1-mediated C-to-U RNA editing in mouse small intestine and liver, Genome Biol, vol.15, p.79, 2014.

P. Brennecke, A. Reyes, S. Pinto, K. Rattay, M. Nguyen et al., Single-cell transcriptome analysis reveals coordinated ectopic gene-expression patterns in medullary thymic epithelial cells, Nat. Immunol, vol.16, pp.933-941, 2015.

C. M. Burns, H. Chu, S. M. Rueter, L. K. Hutchinson, H. Canton et al., Regulation of serotonin-2C receptor G-protein coupling by RNA editing, Nature, vol.387, pp.303-308, 1997.

S. Chen, G. Habib, C. Yang, Z. Gu, B. Lee et al., Apolipoprotein B-48 is the product of a messenger RNA with an organ-specific in-frame stop codon, Science, vol.238, pp.363-366, 1987.

A. Chuprin, A. Avin, Y. Goldfarb, Y. Herzig, B. Levi et al., The deacetylase Sirt1 is an essential regulator of Aire-mediated induction of central immunological tolerance, Nat. Immunol, vol.16, pp.737-745, 2015.

S. G. Conticello, The AID/APOBEC family of nucleic acid mutators, Genome Biol, vol.9, p.229, 2008.

M. P. Cooke and R. M. Perlmutter, Expression of a novel form of the fyn protooncogene in hematopoietic cells, New Biol, vol.1, pp.66-74, 1989.

J. E. Cowan, S. M. Parnell, K. Nakamura, J. H. Caamano, P. J. Lane et al., The thymic medulla is required for Foxp3+ regulatory but not conventional CD4+ thymocyte development, J. Exp. Med, vol.210, pp.675-681, 2013.

P. Danecek, C. Nellåker, R. E. Mcintyre, J. E. Buendia-buendia, S. Bumpstead et al., High levels of RNA-editing site conservation amongst 15 laboratory mouse strains, Genome Biol, vol.13, p.26, 2012.

J. Derbinski, A. Schulte, B. Kyewski, and L. Klein, Promiscuous gene expression in medullary thymic epithelial cells mirrors the peripheral self, Nat. Immunol, vol.2, pp.1032-1039, 2001.

J. Derbinski, J. Gäbler, B. Brors, S. Tierling, S. Jonnakuty et al., Promiscuous gene expression in thymic epithelial cells is regulated at multiple levels, J. Exp. Med, vol.202, pp.33-45, 2005.

A. Dobin, C. Davis, F. Schlesinger, J. Drenkow, C. Zaleski et al., STAR: ultrafast universal RNA-seq aligner, Bioinformatics, vol.29, pp.15-21, 2013.

S. Farajollahi and S. Maas, Molecular diversity through RNA editing: a balancing act, Trends Genet, vol.26, pp.221-230, 2010.

M. Fiume, V. Williams, A. Brook, and M. Brudno, Savant: genome browser for high-throughput sequencing data, Bioinformatics, vol.26, pp.1938-1944, 2010.

P. J. Grabowski and D. L. Black, Alternative RNA splicing in the nervous system, 2001.

, Prog. Neurobiol, vol.65, pp.289-308

B. R. Graveley, Alternative splicing: increasing diversity in the proteomic world, Trends Genet, vol.17, pp.100-107, 2001.

A. R. Grosso, A. Q. Gomes, N. L. Barbosa-morais, S. Caldeira, N. P. Thorne et al., Tissue-specific splicing factor gene expression signatures, Nucleic Acids Res, vol.36, pp.4823-4832, 2008.

K. Higuchi, K. Kitagawa, K. Kogishi, and T. Takeda, Developmental and agerelated changes in apolipoprotein B mRNA editing in mice, J. Lipid Res, vol.33, pp.1753-1764, 1992.

M. B. Johnson, Y. I. Kawasawa, C. E. Mason, Z. Krsnik, G. Coppola et al., Functional and evolutionary insights into human brain development through global transcriptome analysis, Neuron, vol.62, pp.494-509, 2009.

P. Keane, R. Ceredig, and C. Seoighe, Promiscuous mRNA splicing under the control of AIRE in medullary thymic epithelial cells, Bioinformatics, vol.31, pp.986-990, 2015.

O. Kelemen, P. Convertini, Z. Zhang, Y. Wen, M. Shen et al., Function of alternative splicing, Gene, vol.514, pp.1-30, 2013.

E. Kim, A. Goren, A. , and G. , Alternative splicing: current perspectives, BioEssays, vol.30, pp.38-47, 2008.

L. Klein, B. Kyewski, P. M. Allen, and K. A. Hogquist, Positive and negative selection of the T cell repertoire: what thymocytes see (and don't see), Nat. Rev. Immunol, vol.14, pp.377-391, 2014.

B. Kyewski and J. Derbinski, Self-representation in the thymus: an extended view, Nat. Rev. Immunol, vol.4, pp.688-698, 2004.

B. Kyewski and L. Klein, A central role for central tolerance, Annu. Rev. Immunol, vol.24, pp.571-606, 2006.

B. Kyewski, J. Derbinski, J. Gotter, and L. Klein, Promiscuous gene expression and central T-cell tolerance: more than meets the eye, Trends Immunol, vol.23, pp.364-371, 2002.

B. Li and C. N. Dewey, RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome, BMC Bioinformatics, vol.12, p.323, 2011.

Y. Liao, G. K. Smyth, and W. Shi, featureCounts: an efficient general purpose program for assigning sequence reads to genomic features, Bioinformatics, vol.30, pp.923-930, 2013.

E. Lkhagvasuren, M. Sakata, I. Ohigashi, and Y. Takahama, Lymphotoxin ? receptor regulates the development of CCL21-expressing subset of postnatal medullary thymic epithelial cells, J. Immunol, vol.190, pp.5110-5117, 2013.

E. Melamud and J. Moult, Stochastic noise in splicing machinery, Nucleic Acids Res, vol.37, pp.4873-4886, 2009.

M. Meredith, D. Zemmour, D. Mathis, and C. Benoist, Aire controls gene expression in the thymic epithelium with ordered stochasticity, Nat. Immunol, vol.16, pp.942-949, 2015.

J. Merkin, C. Russell, P. Chen, and C. B. Burge, Evolutionary dynamics of gene and isoform regulation in Mammalian tissues, Science, vol.338, pp.1593-1599, 2012.

T. C. Metzger, I. S. Khan, J. M. Gardner, M. L. Mouchess, K. P. Johannes et al., Lineage Tracing and Cell Ablation Identify a Post-Aire-Expressing Thymic Epithelial Cell Population. Cell Rep, vol.5, pp.166-179, 2013.

K. Nagamine, P. Peterson, H. S. Scott, J. Kudoh, S. Minoshima et al., Positional cloning of the APECED gene, Nat. Genet, vol.17, pp.393-398, 1997.

T. W. Nilsen and B. R. Graveley, Expansion of the eukaryotic proteome by alternative splicing, Nature, vol.463, pp.457-463, 2010.

K. Nishikura, Functions and regulation of RNA editing by ADAR deaminases, Annu. Rev. Biochem, vol.79, pp.321-349, 2010.

Q. Pan, O. Shai, L. J. Lee, B. J. Frey, and B. J. Blencowe, Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing, Nat. Genet, vol.40, pp.1413-1415, 2008.

M. S. Paul and B. L. Bass, Inosine exists in mRNA at tissue-specific levels and is most abundant in brain mRNA, EMBO J, vol.17, pp.1120-1127, 1998.

P. Peterson, T. Org, R. , and A. , Transcriptional regulation by AIRE: molecular mechanisms of central tolerance, Nat. Rev. Immunol, vol.8, pp.948-957, 2008.

E. Picardi and G. Pesole, REDItools: high-throughput RNA editing detection made easy, Bioinformatics, vol.29, pp.1813-1814, 2013.

J. K. Pickrell, A. A. Pai, Y. Gilad, and J. K. Pritchard, Noisy splicing drives mRNA isoform diversity in human cells, PLoS Genet, vol.6, p.1001236, 2010.

H. T. Porath, S. Carmi, and E. Y. Levanon, A genome-wide map of hyper-edited RNA reveals numerous new sites, Nat. Commun, vol.5, p.4726, 2014.

L. M. Powell, S. C. Wallis, R. J. Pease, Y. H. Edwards, T. J. Knott et al., A novel form of tissue-specific RNA processing produces apolipoprotein-B48 in intestine, Cell, vol.50, pp.831-840, 1987.

G. Ramaswami and J. B. Li, RADAR: a rigorously annotated database of A-to-I RNA editing, Nucleic Acids Res. gkt996, 2013.

D. Ramsköld, E. T. Wang, C. B. Burge, and R. Sandberg, An abundance of ubiquitously expressed genes revealed by tissue transcriptome sequence data, PLoS Comput. Biol, vol.5, 2009.

W. Ritchie, S. Granjeaud, D. Puthier, and D. Gautheret, Entropy measures quantify global splicing disorders in cancer, PLoS Comput. Biol, vol.4, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00286322

B. R. Rosenberg, C. E. Hamilton, M. M. Mwangi, S. Dewell, and F. N. Papavasiliou, , 2011.

, Transcriptome-wide sequencing reveals numerous APOBEC1 mRNA-editing targets in transcript 3? UTRs, Nat. Struct. Mol. Biol, vol.18, pp.230-236

J. J. Rosenthal, The emerging role of RNA editing in plasticity, J. Exp. Biol, vol.218, pp.1812-1821, 2015.

Y. Sakamoto, T. Matsumoto, Y. Mizunoe, M. Haraoka, M. Sakumoto et al., Testicular Injury Induces Cell-Mediated Autoimmune Response to Testis, J. Urol, vol.153, pp.1316-1320, 1995.

S. N. Sansom, N. Shikama-dorn, S. Zhanybekova, G. Nusspaumer, I. C. Macaulay et al., Population and single-cell genomics reveal the Aire dependency, relief from Polycomb silencing, and distribution of self-antigen expression in thymic epithelia, Genome Res, vol.24, pp.1918-1931, 2014.

Y. A. Savva, L. E. Rieder, and R. A. Reenan, The ADAR protein family, Genome Biol, vol.13, p.252, 2012.

E. E. Schmidt, Transcriptional promiscuity in testes, Curr. Biol, vol.6, pp.768-769, 1996.

S. Sharma, S. K. Patnaik, R. T. Taggart, E. D. Kannisto, S. M. Enriquez et al., APOBEC3A cytidine deaminase induces RNA editing in monocytes and macrophages, Nat. Commun, vol.6, p.6881, 2015.

H. C. Smith, R. P. Bennett, A. Kizilyer, W. M. Mcdougall, and K. M. Prohaska, , 2012.

, Functions and regulation of the APOBEC family of proteins, Semin. Cell Dev. Biol, vol.23, pp.258-268

M. Soumillon, A. Necsulea, M. Weier, D. Brawand, X. Zhang et al., Cellular source and mechanisms of high transcriptome complexity in the mammalian testis, Cell Rep, vol.3, pp.2179-2190, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00852777

S. Stamm, S. Ben-ari, I. Rafalska, Y. Tang, Z. Zhang et al., Function of alternative splicing, Gene, vol.344, pp.1-20, 2005.

C. St-pierre, S. Brochu, J. R. Vanegas, M. Dumont-lagacé, S. Lemieux et al., Transcriptome sequencing of neonatal thymic epithelial cells, 2013.

G. L. Stritesky, S. C. Jameson, and K. A. Hogquist, Selection of self-reactive T cells in the thymus, Annu. Rev. Immunol, vol.30, pp.95-114, 2012.

E. T. Wang, R. Sandberg, S. Luo, I. Khrebtukova, L. Zhang et al., Alternative isoform regulation in human tissue transcriptomes, Nature, vol.456, pp.470-476, 2008.

E. T. Wang, N. A. Cody, S. Jog, M. Biancolella, T. T. Wang et al., Transcriptome-wide regulation of pre-mRNA splicing and mRNA localization by muscleblind proteins, Cell, vol.150, pp.710-724, 2012.

K. Wang, M. Li, and H. Hakonarson, ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data, Nucleic Acids Res, vol.38, p.164, 2010.

S. G. Young, Recent progress in understanding apolipoprotein B, Circulation, vol.82, pp.1574-1594, 1990.

B. Kyewski, L. A. Klein, . Central, . For, and . Tolerance, Annu Rev Immunol, vol.24, issue.1, pp.571-606, 2006.

T. K. Starr, J. Sc, K. A. Hogquist, . Positive, . Negative et al., Annu Rev Immunol, vol.21, issue.1, pp.139-76, 2003.

E. F. Lind, S. E. Prockop, H. E. Porritt, and H. T. Petrie, Mapping precursor movement through the postnatal thymus reveals specific microenvironments supporting defined stages of early lymphoid development, J Exp Med, vol.194, issue.2, pp.127-161, 2001.

D. J. Pennington, B. Silva-santos, and A. C. Hayday, ?? T cell development -having the strength to get there, Curr Opin Immunol, vol.17, issue.2, pp.108-123, 2005.

E. J. Adams, S. Gu, and A. M. Luoma, Human gamma delta T cells: Evolution and ligand recognition, Cell Immunol, vol.296, issue.1, pp.31-40, 2015.

P. Borgulya, H. Kishi, Y. Uematsu, V. Boehmer, and H. , Exclusion and inclusion of ? and ? T cell receptor alleles, Cell, vol.69, issue.3, pp.529-566, 1992.

B. Kyewski and L. Klein, A central role for central tolerance, Annu Rev Immunol, vol.24, pp.571-606, 2006.

T. Baba, Y. Nakamoto, and N. Mukaida, Crucial Contribution of Thymic Sirp + Conventional Dendritic Cells to Central Tolerance against Blood-Borne Antigens in a CCR2-Dependent Manner, J Immunol, vol.183, issue.5, pp.3053-63, 2009.

J. Oh and J. Shin, The Role of Dendritic Cells in Central Tolerance, Immune Netw, vol.15, issue.3, p.111, 2015.

H. Von-boehmer, I. Aifantis, F. Gounari, O. Azogui, L. Haughn et al., Thymic selection revisited: how essential is it? Immunol Rev, vol.191, pp.62-78, 2003.

T. W. Mckeithan, Kinetic proofreading in T-cell receptor signal transduction, Proc Natl Acad Sci, vol.92, issue.11, pp.5042-5048, 1995.

J. A. Williams, S. O. Sharrow, A. J. Adams, and R. J. Hodes, CD40 ligand functions non-cell autonomously to promote deletion of self-reactive thymocytes, J Immunol Baltim Md, vol.168, issue.6, pp.2759-65, 1950.

K. Nagamine, P. Peterson, H. S. Scott, J. Kudoh, S. Minoshima et al., Positional cloning of the APECED gene, Nat Genet, vol.17, issue.4, pp.393-401, 1997.

A. Y. Chan and M. S. Anderson, Central tolerance to self revealed by the autoimmune regulator: Central tolerance by autoimmune regulator (Aire), Ann N Y Acad Sci, vol.1356, issue.1, pp.80-89, 2015.

S. Yang, K. Bansal, J. Lopes, C. Benoist, and D. Mathis, Aire's plant homeodomain(PHD)-2 is critical for induction of immunological tolerance, Proc Natl Acad Sci, vol.110, issue.5, pp.1833-1841, 2013.

Q. Cheng, B. Yi, A. Wang, and X. Jiang, Exploring and exploiting the fundamental role of microRNAs in tumor pathogenesis, OncoTargets Ther, vol.6, pp.1675-84, 2013.

D. Mathis, C. Benoist, and . Aire, Annu Rev Immunol, vol.27, issue.1, pp.287-312, 2009.

A. Finnish-german and . Consortium, An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains, Nat Genet, vol.17, issue.4, pp.399-403, 1997.

J. Abramson and E. S. Husebye, Autoimmune regulator and self-tolerance -molecular and clinical aspects, Immunol Rev, vol.271, issue.1, pp.127-167, 2016.

B. J. Ferguson, C. Alexander, S. W. Rossi, I. Liiv, A. Rebane et al., AIRE's CARD Revealed, a New Structure for Central Tolerance Provokes Transcriptional Plasticity, J Biol Chem, vol.283, issue.3, pp.1723-1754, 2008.

F. Buzi, R. Badolato, C. Mazza, S. Giliani, L. D. Notarangelo et al.,

, Autoimmune Polyendocrinopathy-Candidiasis-Ectodermal Dystrophy Syndrome: Time to Review Diagnostic Criteria?, J Clin Endocrinol Metab, vol.88, issue.7, pp.3146-3154, 2003.

M. S. Anderson, Projection of an Immunological Self Shadow Within the Thymus by the Aire Protein. Science, vol.298, pp.1395-401, 2002.

F. Hubert, S. A. Kinkel, K. E. Webster, P. Cannon, P. E. Crewther et al., A Specific Anti-Aire Antibody Reveals Aire Expression Is Restricted to Medullary Thymic Epithelial Cells and Not Expressed in Periphery, J Immunol, vol.180, issue.6, pp.3824-3856, 2008.

J. Lee, M. Epardaud, J. Sun, J. E. Becker, A. C. Cheng et al., Peripheral antigen display by lymph node stroma promotes T cell tolerance to intestinal self, Nat Immunol, vol.8, issue.2, pp.181-90, 2007.

C. Ramsey, O. Winqvist, L. Puhakka, M. Halonen, A. Moro et al., Aire deficient mice develop multiple features of APECED phenotype and show altered immune response, Hum Mol Genet, vol.11, issue.4, pp.397-409, 2002.

C. Y. Wang, J. D. Shi, A. Davoodi-semiromi, and J. X. She, Cloning of Aire, the mouse homologue of the autoimmune regulator (AIRE) gene responsible for autoimmune polyglandular syndrome type 1 (ASP1), Genomics, vol.55, issue.3, pp.322-328, 1999.

J. Derbinski, J. Gäbler, B. Brors, S. Tierling, S. Jonnakuty et al., Promiscuous gene expression in thymic epithelial cells is regulated at multiple levels, J Exp Med, vol.202, issue.1, pp.33-45, 2005.

M. S. Anderson, E. S. Venanzi, Z. Chen, S. P. Berzins, C. Benoist et al., The Cellular Mechanism of Aire Control of T Cell Tolerance, Immunity, vol.23, issue.2, pp.227-266, 2005.

R. T. Taniguchi, J. J. Devoss, J. J. Moon, J. Sidney, A. Sette et al., Detection of an autoreactive T-cell population within the polyclonal repertoire that undergoes distinct autoimmune regulator (Aire)-mediated selection, Proc Natl Acad Sci, vol.109, issue.20, pp.7847-52, 2012.

T. Yamano, J. Nedjic, M. Hinterberger, M. Steinert, S. Koser et al., Thymic B Cells Are Licensed to Present Self Antigens for Central T Cell Tolerance Induction, Immunity, vol.42, issue.6, pp.1048-61, 2015.

S. Sakaguchi, . Naturally-arising-cd4+-regulatory-t-cells-for, . Self-tolerance, . Negative, . Of et al., Annu Rev Immunol, vol.22, issue.1, pp.531-62, 2004.

M. Azimi, S. Aslani, S. Mortezagholi, A. Salek, M. R. Javan et al.,

, Isolation, and Functional Assay of Regulatory T Cells. Immunol Invest, pp.1-19, 2016.

N. Kuroda, T. Mitani, N. Takeda, N. Ishimaru, R. Arakaki et al., Development of Autoimmunity against Transcriptionally Unrepressed Target Antigen in the Thymus of Aire-Deficient Mice, J Immunol, vol.174, issue.4, pp.1862-70, 2005.

Z. Chen, C. Benoist, and D. Mathis, How defects in central tolerance impinge on a deficiency in regulatory T cells, Proc Natl Acad Sci, vol.102, issue.41, pp.14735-14775, 2005.

S. M. Laakso, T. Laurinolli, L. H. Rossi, A. Lehtoviita, H. Sairanen et al.,

, Regulatory T cell defect in APECED patients is associated with loss of naive FOXP3+ precursors and impaired activated population, J Autoimmun, vol.35, issue.4, pp.351-358, 2010.

K. Aschenbrenner, L. M. Cruz, E. H. Vollmann, M. Hinterberger, J. Emmerich et al., Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells, Nat Immunol, vol.8, issue.4, pp.351-359, 2007.

S. Malchow, D. S. Leventhal, S. Nishi, B. I. Fischer, L. Shen et al., AireDependent Thymic Development of Tumor-Associated Regulatory T Cells, Science, vol.339, issue.6124, pp.1219-1243, 2013.

J. Perry, C. Lio, A. L. Kau, K. Nutsch, Z. Yang et al., Distinct Contributions of Aire and Antigen-Presenting-Cell Subsets to the Generation of SelfTolerance in the Thymus, Immunity, vol.41, issue.3, pp.414-440, 2014.

S. Malchow, D. S. Leventhal, V. Lee, S. Nishi, N. D. Socci et al., Aire Enforces Immune Tolerance by Directing Autoreactive T Cells into the Regulatory T Cell Lineage, Immunity, vol.44, issue.5, pp.1102-1115, 2016.

C. Pomie, R. Vicente, Y. Vuddamalay, B. A. Lundgren, M. Van-der-hoek et al., Autoimmune regulator (AIRE)-deficient CD8+CD28low regulatory T lymphocytes fail to control experimental colitis, Proc Natl Acad Sci, vol.108, issue.30, pp.12437-12479, 2011.

N. Thiault, J. Darrigues, V. Adoue, M. Gros, B. Binet et al., Peripheral regulatory T lymphocytes recirculating to the thymus suppress the development of their precursors, Nat Immunol, vol.16, issue.6, pp.628-662, 2015.

C. Ramsey, S. Hässler, P. Marits, O. Kämpe, C. D. Surh et al., Increased antigen presenting cell-mediated T cell activation in mice and patients without the autoimmune regulator, Eur J Immunol, vol.36, issue.2, pp.305-322, 2006.

J. M. Gardner, T. C. Metzger, E. J. Mcmahon, B. B. Au-yeung, A. K. Krawisz et al.,

, Extrathymic Aire-Expressing Cells Are a Distinct Bone Marrow-Derived Population that Induce Functional Inactivation of CD4+ T Cells, Immunity, vol.39, issue.3, pp.560-72, 2013.

J. M. Gardner, J. J. Devoss, R. S. Friedman, D. J. Wong, Y. X. Tan et al., Deletional Tolerance Mediated by Extrathymic Aire-Expressing Cells, Science, vol.321, issue.5890, pp.843-850, 2008.

N. Lopes, P. Ferrier, and M. Irla, Induction de la tolérance centrale dans le thymus par le facteur de transcription Aire. médecine/sciences, vol.31, pp.742-749, 2015.

J. B. Johnnidis, E. S. Venanzi, D. J. Taxman, J. Ting, C. O. Benoist et al.,

, Chromosomal clustering of genes controlled by the aire transcription factor, Proc Natl Acad Sci, vol.102, issue.20, pp.7233-7241, 2005.

E. S. Venanzi, R. Melamed, D. Mathis, and C. Benoist, The variable immunological self: Genetic variation and nongenetic noise in Aire-regulated transcription, Proc Natl Acad Sci, vol.105, issue.41, pp.15860-15865, 2008.

J. Villasenor, W. Besse, C. Benoist, and D. Mathis, Ectopic expression of peripheral-tissue antigens in the thymic epithelium: Probabilistic, monoallelic, misinitiated, Proc Natl Acad Sci, vol.105, issue.41, pp.15854-15863, 2008.

M. Meredith, D. Zemmour, D. Mathis, and C. Benoist, Aire controls gene expression in the thymic epithelium with ordered stochasticity, Nat Immunol, vol.16, issue.9, pp.942-951, 2015.

S. N. Sansom, N. Shikama-dorn, S. Zhanybekova, G. Nusspaumer, and I. C. Macaulay,

M. E. Deadman, Population and single-cell genomics reveal the Aire dependency, relief from Polycomb silencing, and distribution of self-antigen expression in thymic epithelia

, Genome Res, vol.24, issue.12, pp.1918-1949, 2014.

B. Kyewski, J. Derbinski, J. Gotter, and L. Klein, Promiscuous gene expression and central T-cell tolerance: more than meets the eye, Trends Immunol, vol.23, issue.7, pp.364-71, 2002.

J. Derbinski, S. Pinto, S. Rosch, K. Hexel, and B. Kyewski, Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism, Proc Natl Acad Sci, vol.105, issue.2, pp.657-62, 2008.

R. Taubert, J. Schwendemann, and B. Kyewski, Highly variable expression of tissuerestricted self-antigens in human thymus: Implications for self-tolerance and autoimmunity, Eur J Immunol, vol.37, issue.3, pp.838-886, 2007.

G. W. Muse, D. A. Gilchrist, S. Nechaev, R. Shah, J. S. Parker et al., RNA polymerase is poised for activation across the genome, Nat Genet, vol.39, issue.12, pp.1507-1518, 2007.

M. Giraud, H. Yoshida, J. Abramson, P. B. Rahl, R. A. Young et al., Aire unleashes stalled RNA polymerase to induce ectopic gene expression in thymic epithelial cells, Proc Natl Acad Sci, vol.109, issue.2, pp.535-575, 2012.

N. J. Proudfoot, A. Furger, and M. J. Dye, Integrating mRNA Processing with Transcription

, Cell, vol.108, issue.4, pp.501-513, 2002.

J. Abramson, M. Giraud, C. Benoist, and D. Mathis, Aire's Partners in the Molecular Control of Immunological Tolerance, Cell, vol.140, issue.1, pp.123-158, 2010.

I. Liiv, A. Rebane, T. Org, M. Saare, J. Maslovskaja et al., DNA-PK contributes to the phosphorylation of AIRE: Importance in transcriptional activity, Biochim Biophys Acta BBA -Mol Cell Res, vol.1783, issue.1, pp.74-83, 2008.

N. Mondal and J. D. Parvin, DNA topoisomerase II? is required for RNA polymerase II transcription on chromatin templates, Nature, vol.413, issue.6854, pp.435-443, 2001.

B. Ju, A Topoisomerase II -Mediated dsDNA Break Required for Regulated Transcription, Science, vol.312, issue.5781, pp.1798-802, 2006.

R. Krishnakumar, M. J. Gamble, K. M. Frizzell, J. G. Berrocal, M. Kininis et al.,

, Reciprocal Binding of PARP-1 and Histone H1 at Promoters Specifies Transcriptional Outcomes, Science, vol.319, issue.5864, pp.819-840, 2008.

A. S. Koh, A. J. Kuo, S. Y. Park, P. Cheung, J. Abramson et al., Aire employs a histone-binding module to mediate immunological tolerance, linking chromatin regulation with organ-specific autoimmunity, Proc Natl Acad Sci, vol.105, issue.41, pp.15878-83, 2008.

T. Org, F. Chignola, C. Hetényi, M. Gaetani, A. Rebane et al., The autoimmune regulator PHD finger binds to non-methylated histone H3K4 to activate gene expression, EMBO Rep, vol.9, issue.4, pp.370-376, 2008.

B. L. Mahaney, K. Meek, and S. P. Lees-miller, Repair of ionizing radiation-induced DNA double-strand breaks by non-homologous end-joining, Biochem J, vol.417, issue.3, pp.639-50, 2009.

J. Pitkanen, The Autoimmune Regulator Protein Has Transcriptional Transactivating Properties and Interacts with the Common Coactivator CREB-binding Protein, J Biol Chem, vol.275, issue.22, pp.16802-16811, 2000.

M. Saare, A. Rebane, B. Rajashekar, J. Vilo, and P. Peterson, Autoimmune regulator is acetylated by transcription coactivator CBP/p300, Exp Cell Res, vol.318, issue.14, pp.1767-78, 2012.

A. Chuprin, A. Avin, Y. Goldfarb, Y. Herzig, B. Levi et al., The deacetylase Sirt1 is an essential regulator of Aire-mediated induction of central immunological tolerance, Nat Immunol, vol.16, issue.7, pp.737-782, 2015.

D. Uchida, S. Hatakeyama, A. Matsushima, H. Han, S. Ishido et al., AIRE Functions As an E3 Ubiquitin Ligase, J Exp Med, vol.199, issue.2, pp.167-72, 2004.

B. Roy, M. Haupt, L. , R. Griffiths, and L. , Review: Alternative Splicing (AS) of Genes As An Approach for Generating Protein Complexity, Curr Genomics, vol.14, issue.3, pp.182-94, 2013.

M. Chen and J. L. Manley, Mechanisms of alternative splicing regulation: insights from molecular and genomics approaches, Nat Rev Mol Cell Biol, 2009.

,

P. Keane, R. Ceredig, and C. Seoighe, Promiscuous mRNA splicing under the control of AIRE in medullary thymic epithelial cells, Bioinformatics, vol.31, issue.7, pp.986-90, 2015.

C. Mayr, Evolution and Biological Roles of Alternative 3?UTRs, Trends Cell Biol, vol.26, issue.3, pp.227-264, 2016.

B. Tian, A large-scale analysis of mRNA polyadenylation of human and mouse genes, Nucleic Acids Res, vol.33, issue.1, pp.201-213, 2005.

S. Lianoglou, V. Garg, J. L. Yang, C. S. Leslie, and C. Mayr, Ubiquitously transcribed genes use alternative polyadenylation to achieve tissue-specific expression, Genes Dev, vol.27, issue.21, pp.2380-96, 2013.

Z. Ji, J. Y. Lee, Z. Pan, B. Jiang, and B. Tian, Progressive lengthening of 3' untranslated regions of mRNAs by alternative polyadenylation during mouse embryonic development, Proc Natl Acad Sci, vol.106, issue.17, pp.7028-7061, 2009.

C. Mayr and D. P. Bartel, Widespread Shortening of 3?UTRs by Alternative Cleavage and Polyadenylation Activates Oncogenes in Cancer Cells, Cell, vol.138, issue.4, pp.673-84, 2009.

P. Pinto, T. Henriques, M. O. Freitas, T. Martins, R. G. Domingues et al., RNA polymerase II kinetics in polo polyadenylation signal selection: RNA polymerase II kinetics and alternative polyadenylation, EMBO J, vol.30, issue.12, pp.2431-2475, 2011.

A. Meinhart and P. Cramer, Recognition of RNA polymerase II carboxy-terminal domain by 3??-RNA-processing factors, Nature, vol.430, issue.6996, pp.223-229, 2004.

Z. Ji, W. Luo, W. Li, M. Hoque, Z. Pan et al., Transcriptional activity regulates alternative cleavage and polyadenylation, Mol Syst Biol, vol.7, issue.1, pp.534-534, 2014.

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.1643-1650, 2008.

D. P. Bartel, MicroRNAs: Target Recognition and Regulatory Functions, Cell, vol.136, issue.2, pp.215-248, 2009.

C. Chen and A. Shyu, AU-rich elements: characterization and importance in mRNA degradation, Trends Biochem Sci, vol.20, issue.11, pp.465-70, 1995.

I. A. Vlasova, N. M. Tahoe, D. Fan, O. Larsson, B. Rattenbacher et al.,

G. Conserved, Elements Mediate mRNA Decay by Binding to CUG-Binding Protein 1, Mol Cell, vol.29, issue.2, pp.263-70, 2008.

Y. Shi, D. Giammartino, D. C. Taylor, D. Sarkeshik, A. Rice et al.,

, Molecular Architecture of the Human Pre-mRNA 3? Processing Complex, Mol Cell, vol.33, issue.3, pp.365-76, 2009.

J. Deckert, K. Hartmuth, D. Boehringer, N. Behzadnia, C. L. Will et al.,

, Protein Composition and Electron Microscopy Structure of Affinity-Purified Human Spliceosomal B Complexes Isolated under Physiological Conditions, Mol Cell Biol, vol.26, issue.14, pp.5528-5571, 2006.

D. Giammartino, D. C. Nishida, K. Manley, and J. L. , Mechanisms and Consequences of Alternative Polyadenylation, Mol Cell, vol.43, issue.6, pp.853-66, 2011.

B. Mazumder, V. Seshadri, and P. L. Fox, Translational control by the 3?-UTR: the ends specify the means, Trends Biochem Sci, vol.28, issue.2, pp.91-99, 2003.

P. Oikonomou, H. Goodarzi, and S. Tavazoie, Systematic Identification of Regulatory Elements in Conserved 3? UTRs of Human Transcripts, Cell Rep, vol.7, issue.1, pp.281-92, 2014.

A. R. Gruber, G. Martin, P. Müller, A. Schmidt, A. J. Gruber et al., Global 3? UTR shortening has a limited effect on protein abundance in proliferating T cells, Nat Commun, vol.5, p.5465, 2014.

J. Nam, O. S. Rissland, D. Koppstein, C. Abreu-goodger, C. H. Jan et al.,

, Global Analyses of the Effect of Different Cellular Contexts on MicroRNA Targeting, Mol Cell, vol.53, issue.6, pp.1031-1074, 2014.

C. H. Moor, . De, H. Meijer, and S. Lissenden, Mechanisms of translational control by the 3? UTR in development and differentiation, Semin Cell Dev Biol, vol.16, issue.1, pp.49-58, 2005.

A. Jambhekar and J. L. Derisi, Cis-acting determinants of asymmetric, cytoplasmic RNA transport. RNA, vol.13, pp.625-667, 2007.

J. J. An, K. Gharami, G. Liao, N. H. Woo, A. G. Lau et al., Distinct Role of Long 3? UTR BDNF mRNA in Spine Morphology and Synaptic Plasticity in Hippocampal Neurons, Cell, vol.134, issue.1, pp.175-87, 2008.

B. D. Berkovits and C. Mayr, Alternative 3? UTRs act as scaffolds to regulate membrane protein localization, Nature, vol.522, issue.7556, pp.363-370, 2015.

A. G. Baltz, M. Munschauer, B. Schwanhäusser, A. Vasile, Y. Murakawa et al., The mRNA-Bound Proteome and Its Global Occupancy Profile on Protein-Coding Transcripts, Mol Cell, vol.46, issue.5, pp.674-90, 2012.

Y. Lee, M. Kim, J. Han, K. Yeom, S. Lee et al., MicroRNA genes are transcribed by RNA polymerase II, EMBO J, vol.23, issue.20, pp.4051-60, 2004.

W. Filipowicz, S. N. Bhattacharyya, and N. Sonenberg, Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight?, Nat Rev Genet, issue.2, pp.102-116, 2008.

S. Zuklys, C. E. Mayer, S. Zhanybekova, H. E. Stefanski, G. Nusspaumer et al.,

, MicroRNAs Control the Maintenance of Thymic Epithelia and Their Competence for T Lineage Commitment and Thymocyte Selection, J Immunol, vol.189, issue.8, pp.3894-904, 2012.

O. Ucar, L. Tykocinski, J. Dooley, A. Liston, and B. Kyewski, An evolutionarily conserved mutual interdependence between Aire and microRNAs in promiscuous gene expression: Molecular immunology, Eur J Immunol, vol.43, issue.7, pp.1769-78, 2013.

C. Macedo, A. F. Evangelista, M. M. Marques, S. Octacílio-silva, E. A. Donadi et al., Autoimmune regulator (Aire) controls the expression of microRNAs in medullary thymic epithelial cells, Immunobiology, vol.218, issue.4, pp.554-60, 2013.

B. Kyewski and J. Derbinski, Self-representation in the thymus: an extended view, Nat Rev Immunol, vol.4, issue.9, pp.688-98, 2004.

T. C. Metzger, I. S. Khan, J. M. Gardner, M. L. Mouchess, K. P. Johannes et al.,

, Lineage Tracing and Cell Ablation Identify a Post-Aire-Expressing Thymic Epithelial Cell Population. Cell Rep, vol.5, issue.1, pp.166-79, 2013.

A. Ucar, O. Ucar, P. Klug, S. Matt, F. Brunk et al., Adult Thymus Contains FoxN1? Epithelial Stem Cells that Are Bipotent for Medullary and Cortical Thymic Epithelial Lineages, Immunity, vol.41, issue.2, pp.257-69, 2014.

K. Wong, N. L. Lister, M. Barsanti, J. Lim, M. V. Hammett et al.,

, Multilineage Potential and Self-Renewal Define an Epithelial Progenitor Cell Population in the Adult Thymus, Cell Rep, vol.8, issue.4, pp.1198-209, 2014.

N. L. Alves, Y. Takahama, I. Ohigashi, A. R. Ribeiro, S. Baik et al., Serial progression of cortical and medullary thymic epithelial microenvironments: HIGHLIGHTS, Eur J Immunol, vol.44, issue.1, pp.16-22, 2014.

N. A. Roberts, A. J. White, W. E. Jenkinson, G. Turchinovich, K. Nakamura et al., Rank Signaling Links the Development of Invariant ?? T Cell Progenitors and Aire+ Medullary Epithelium, Immunity, vol.36, issue.3, pp.427-464, 2012.

T. Akiyama, Y. Shimo, H. Yanai, J. Qin, D. Ohshima et al., The Tumor Necrosis Factor Family Receptors RANK and CD40 Cooperatively Establish the Thymic Medullary Microenvironment and Self-Tolerance, Immunity, vol.29, issue.3, pp.423-460, 2008.

N. Lopes, A. Sergé, P. Ferrier, and M. Irla, Thymic Crosstalk Coordinates Medulla Organization and T-Cell Tolerance Induction. Front Immunol, vol.6, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01201019

L. Klein, M. Hinterberger, G. Wirnsberger, and B. Kyewski, Antigen presentation in the thymus for positive selection and central tolerance induction, Nat Rev Immunol, vol.9, issue.12, pp.833-877, 2009.

S. W. Rossi, M. Kim, A. Leibbrandt, S. M. Parnell, W. E. Jenkinson et al., RANK signals from CD4 + 3 ? inducer cells regulate development of Aire-expressing epithelial cells in the thymic medulla, J Exp Med, vol.204, issue.6, pp.1267-72, 2007.

I. S. Khan, M. L. Mouchess, M. Zhu, B. Conley, K. J. Fasano et al., Enhancement of an anti-tumor immune response by transient blockade of central T cell tolerance, J Exp Med, vol.211, issue.5, pp.761-769, 2014.

S. Pinto, C. Michel, H. Schmidt-glenewinkel, N. Harder, K. Rohr et al., Overlapping gene coexpression patterns in human medullary thymic epithelial cells generate self-antigen diversity, Proc Natl Acad Sci, vol.110, issue.37, pp.3497-505, 2013.

K. Rattay, H. V. Meyer, C. Herrmann, B. Brors, and B. Kyewski, Evolutionary conserved gene co-expression drives generation of self-antigen diversity in medullary thymic epithelial cells, J Autoimmun, vol.67, pp.65-75, 2016.

P. Brennecke, A. Reyes, S. Pinto, K. Rattay, M. Nguyen et al., Single-cell transcriptome analysis reveals coordinated ectopic gene-expression patterns in medullary thymic epithelial cells, Nat Immunol, vol.16, issue.9, pp.933-974, 2015.

D. Gray, J. Abramson, C. Benoist, and D. Mathis, Proliferative arrest and rapid turnover of thymic epithelial cells expressing Aire, J Exp Med, vol.204, issue.11, pp.2521-2529, 2007.

A. J. White, K. Nakamura, W. E. Jenkinson, M. Saini, C. Sinclair et al., Lymphotoxin Signals from Positively Selected Thymocytes Regulate the Terminal Differentiation of Medullary Thymic Epithelial Cells, J Immunol, vol.185, issue.8, pp.4769-76, 2010.

X. Wang, M. Laan, R. Bichele, K. Kisand, H. S. Scott et al., Post-Aire Maturation of Thymic Medullary Epithelial Cells Involves Selective Expression of Keratinocyte-Specific Autoantigens. Front Immunol, vol.3, 2012.

Y. Nishikawa, H. Nishijima, M. Matsumoto, J. Morimoto, F. Hirota et al.,

, Temporal Lineage Tracing of Aire-Expressing Cells Reveals a Requirement for Aire in Their Maturation Program, J Immunol, vol.192, issue.6, pp.2585-92, 2014.

N. Watanabe, Y. Wang, H. K. Lee, T. Ito, Y. Wang et al., Hassall's corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus, Nature, vol.436, issue.7054, pp.1181-1186, 2005.

N. Spies, C. B. Burge, and D. P. Bartel, UTR-isoform choice has limited influence on the stability and translational efficiency of most mRNAs in mouse fibroblasts, Genome Res, vol.3, issue.12, pp.2078-90, 2013.

A. Husini, N. Kudla, P. Ansari, and A. , A Role for CF1A 3? End Processing Complex in Promoter-Associated Transcription, PLoS Genet, vol.9, issue.8, p.1003722, 2013.

T. Zhang and W. L. Kraus, SIRT1-dependent regulation of chromatin and transcription: Linking NAD+ metabolism and signaling to the control of cellular functions, Biochim Biophys Acta BBA -Proteins Proteomics, issue.8, pp.1666-75, 1804.

D. Herranz and M. Serrano, SIRT1: recent lessons from mouse models, Nat Rev Cancer, vol.10, issue.12, pp.819-842, 2010.

J. Sequeira, G. Boily, S. Bazinet, S. Saliba, X. He et al., sirt1-null mice develop an autoimmune-like condition, Exp Cell Res, vol.314, issue.16, pp.3069-74, 2008.

U. H. Beier, L. Wang, T. R. Bhatti, Y. Liu, R. Han et al., Sirtuin-1 Targeting Promotes Foxp3+ T-Regulatory Cell Function and Prolongs Allograft Survival, Mol Cell Biol, vol.31, issue.5, pp.1022-1031, 2011.

B. Doctorat-en, spécialité Immunologie Ecole Doctorale GC2ID (maintenant BIO Sorbonne Paris Cité), Université Descartes, Paris Bourse du ministère (2012-2015) et gratification de la FRM, 2015.

, Options : Organisation des génomes, macromolécules, intéraction protéine-protéine, régulation de l'expression génique, ontogénèse, Cellules Cibles, Thérapeutiques, cursus Génome et Destin Cellulaire, 2010.

, Cellules Cibles, Thérapeutiques, Paris VII Options : pharmacogénomique, interaction protéines protéines, biotechnologie, pathologie moléculaire, modélisation moléculaire, 2009.

, UHP de Nancy Options : expérimentation animale, stress oxydant, séparation des protéines, anglais, différenciation cellulaire, Licence 3 Science du Vivant, cursus Biologie Moléculaire, 2008.

, Licence 2 Science du Vivant, cursus Biologie Moléculaire, pp.2006-2008

P. Médecine, . De-dijon, and . Reçu, , 2006.

, Baccalauréat Scientifique (Option Science et vie)