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.

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.

P. Ahonen, Autoimmune polyendocrinopathy-candidosis-ectodermal dystrophy (APECED): autosomal recessive inheritance, Clin. Genet, vol.27, pp.535-542, 1985.
DOI : 10.1111/j.1399-0004.1985.tb02037.x

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.

J. Aaltonen, N. Horelli-kuitunen, J. B. Fan, P. Björses, J. Perheentupa et al., High-resolution physical and transcriptional mapping of the autoimmune polyendocrinopathy-candidiasisectodermal dystrophy locus on chromosome 21q22.3 by FISH, Genome Res, vol.7, pp.820-829, 1997.

A. G. Myhre, M. Halonen, P. Eskelin, O. Ekwall, H. Hedstrand et al., Autoimmune polyendocrine syndrome type 1 (APS I) in Norway, Clin. Endocrinol. (Oxf.), vol.54, pp.211-217, 2001.
DOI : 10.1046/j.1365-2265.2001.01201.x

P. Ahonen, S. Myllärniemi, I. Sipilä, and J. Perheentupa, Clinical variation of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) in a series of 68 patients, N. Engl. J. Med, vol.322, pp.1829-1836, 1990.

K. Cunnusamy, E. J. Baughman, J. Franco, S. B. Ortega, S. Sinha et al., Disease exacerbation of multiple sclerosis is characterized by loss of terminally differentiated autoregulatory CD8+ T cells, Clin. Immunol, vol.152, pp.115-126, 2014.

C. Betterle, C. Pra, F. Mantero, and R. Zanchetta, Autoimmune adrenal insufficiency and autoimmune polyendocrine syndromes: autoantibodies, autoantigens, and their applicability in diagnosis and disease prediction, Endocr. Rev, vol.23, pp.327-364, 2002.

E. S. Husebye, J. Perheentupa, R. Rautemaa, and O. Kämpe, Clinical manifestations and management of patients with autoimmune polyendocrine syndrome type I, J. Intern. Med, vol.265, pp.514-529, 2009.

E. M. Orlova, A. M. Bukina, E. S. Kuznetsova, M. A. Kareva, E. U. Zakharova et al., Autoimmune polyglandular syndrome type 1 in Russian patients: clinical variants and autoimmune regulator mutations, Horm. Res. Paediatr, vol.73, pp.449-457, 2010.

M. Alimohammadi, P. Björklund, A. Hallgren, N. Pöntynen, G. Szinnai et al., Autoimmune polyendocrine syndrome type 1 and NALP5, a parathyroid autoantigen, N. Engl. J. Med, vol.358, pp.1018-1028, 2008.

A. K. Shum, M. Alimohammadi, C. L. Tan, M. H. Cheng, T. C. Metzger et al., BPIFB1 is a lung-specific autoantigen associated with interstitial lung disease, Sci. Transl. Med, vol.5, pp.206-139, 2013.

J. Aaltonen, P. Björses, J. Perheentupa, N. Horelli-kuitunen, A. Palotie et al., An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains, Nat. Genet, vol.17, pp.399-403, 1997.

F. X. Hubert, S. A. Kinkel, P. E. Crewther, P. Z. Cannon, K. E. Webster et al., Airedeficient C57BL/6 mice mimicking the common human 13-base pair deletion mutation present with only a mild autoimmune phenotype, J. Immunol, vol.182, pp.3902-3918, 2009.

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, pp.397-409, 2002.

T. R. Smith and V. Kumar, Revival of CD8+ Treg-mediated suppression, Trends Immunol, vol.29, pp.337-342, 2008.

A. M. Geurts, G. J. Cost, S. Rémy, X. Cui, L. Tesson et al., Generation of gene-specific mutated rats using zinc-finger nucleases, Methods Mol. Biol, vol.597, pp.211-225, 2010.

E. Picarda, S. Bézie, L. Boucault, E. Autrusseau, S. Kilens et al., Transient antibody targeting of CD45RC induces transplant tolerance and potent antigen-specific regulatory T cells, JCI Insight, vol.2, p.90088, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01833115

M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, p.550, 2014.

M. Danan-gotthold, C. Guyon, M. Giraud, E. Y. Levanon, and J. Abramson, Extensive RNA editing and splicing increase immune self-representation diversity in medullary thymic epithelial cells, Genome Biol, vol.17, p.219, 2016.
DOI : 10.1186/s13059-016-1079-9

URL : https://hal.archives-ouvertes.fr/inserm-01386893

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. 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.

J. Kärner, A. Meager, M. Laan, J. Maslovskaja, M. Pihlap et al., Anticytokine autoantibodies suggest pathogenetic links with autoimmune regulator deficiency in humans and mice, Clin. Exp. Immunol, vol.171, pp.263-272, 2013.

M. Laan and P. Peterson, The many faces of aire in central tolerance, Front. Immunol, vol.4, p.326, 2013.

F. X. Hubert, C. Voisine, C. Louvet, M. Heslan, and R. Josien, Rat plasmacytoid dendritic cells are an abundant subset of MHC class II+ CD4+CD11b-OX62-and type I IFN-producing cells that exhibit selective expression of Toll-like receptors 7 and 9 and strong responsiveness to CpG, J. Immunol, vol.172, pp.7485-7494, 2004.

C. Voisine, F. X. Hubert, B. Trinité, M. Heslan, and R. Josien, Two phenotypically distinct subsets of spleen dendritic cells in rats exhibit different cytokine production and T cell stimulatory activity, J. Immunol, vol.169, pp.2284-2291, 2002.

M. C. Rosatelli, A. Meloni, A. Meloni, M. Devoto, A. Cao et al., A common mutation in Sardinian autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy patients, Hum. Genet, vol.103, pp.428-434, 1998.
DOI : 10.1007/s004390050846

A. M. Geurts, G. J. Cost, Y. Freyvert, B. Zeitler, J. C. Miller et al., Knockout rats via embryo microinjection of zinc-finger nucleases, Science, vol.325, p.433, 2009.
DOI : 10.1126/science.1172447

URL : http://europepmc.org/articles/pmc2831805?pdf=render

J. C. Martin, G. Bériou, M. Heslan, C. Bossard, A. Jarry et al., IL-22BP is produced by eosinophils in human gut and blocks IL-22 protective actions during colitis, Mucosal Immunol, vol.9, pp.539-549, 2016.
DOI : 10.1038/mi.2015.83

URL : https://hal.archives-ouvertes.fr/inserm-02150949

S. Rémy, L. Tesson, S. Ménoret, C. Usal, A. M. Scharenberg et al., Zinc-finger nucleases: a powerful tool for genetic engineering of animals, Transgenic Res, vol.19, pp.363-371, 2010.

V. Chenouard, L. Brusselle, J. M. Heslan, S. Remy, S. Menoret et al., A rapid and costeffective method for genotyping genome-edited animals: a heteroduplex mobility assay using microfluidic capillary electrophoresis, J. Genet. Genomics, vol.43, pp.341-348, 2016.
DOI : 10.1016/j.jgg.2016.04.005

URL : https://hal.archives-ouvertes.fr/inserm-02150677

S. Ménoret, S. Fontanière, D. Jantz, L. Tesson, R. Thinard et al., Generation of Rag1-knockout immunodeficient rats and mice using engineered meganucleases, FASEB J, vol.27, pp.703-711, 2013.

P. D. Burbelo, E. E. Lebovitz, and A. L. Notkins, Luciferase immunoprecipitation systems for measuring antibodies in autoimmune and infectious diseases, Transl. Res, vol.165, pp.325-335, 2015.
DOI : 10.1016/j.trsl.2014.08.006

URL : http://europepmc.org/articles/pmc4306608?pdf=render

N. M. Danzl, S. Jeong, Y. Choi, and K. Alexandropoulos, Identification of novel thymic epithelial cell subsets whose differentiation is regulated by RANKL and Traf6, PLoS One, vol.9, p.86129, 2014.
DOI : 10.1371/journal.pone.0086129

URL : https://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0086129&type=printable

L. Sun, H. Li, H. Luo, and Y. Zhao, Thymic epithelial cell development and its dysfunction in human diseases, BioMed Res. Int, p.206929, 2014.

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, pp.1048-1061, 2015.
DOI : 10.1016/j.immuni.2015.05.013

URL : https://doi.org/10.1016/j.immuni.2015.05.013

L. Fagerberg, B. M. Hallström, P. Oksvold, C. Kampf, D. Djureinovic et al., Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics, Mol. Cell. Proteomics, vol.13, pp.397-406, 2014.

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.
DOI : 10.1038/nri2450

URL : http://europepmc.org/articles/pmc2785478?pdf=render

P. L. Poliani, K. Kisand, V. Marrella, M. Ravanini, L. D. Notarangelo et al., Human peripheral lymphoid tissues contain autoimmune regulator-expressing dendritic cells, Am. J. Pathol, vol.176, pp.1104-1112, 2010.
DOI : 10.2353/ajpath.2010.090956

URL : http://europepmc.org/articles/pmc2832133?pdf=render

O. Leavy, Regulatory T cells: CD8 + T Reg cells join the fold, Nat. Rev. Immunol, vol.10, pp.680-681, 2010.

A. Meager, K. Visvalingam, P. Peterson, K. Möll, A. Murumägi et al., Anti-interferon autoantibodies in autoimmune polyendocrinopathy syndrome type 1, PLoS Med, vol.3, p.289, 2006.
DOI : 10.1371/journal.pmed.0030289

URL : https://journals.plos.org/plosmedicine/article/file?id=10.1371/journal.pmed.0030289&type=printable

K. Kisand, A. S. Wolff, K. T. Podkrajsek, L. Tserel, M. Link et al., Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines, J. Exp. Med, vol.207, pp.299-308, 2010.
DOI : 10.1084/jem.20091669

URL : http://jem.rupress.org/content/207/2/299.full.pdf

T. Ito, S. Hanabuchi, Y. H. Wang, W. R. Park, K. Arima et al., Two functional subsets of FOXP3+ regulatory T cells in human thymus and periphery, Immunity, vol.28, pp.870-880, 2008.

S. Meyer, M. Woodward, C. Hertel, P. Vlaicu, Y. Haque et al., AIRE-deficient patients harbor unique high-affinity disease-ameliorating autoantibodies, Cell, vol.166, pp.582-595, 2016.
DOI : 10.1016/j.cell.2016.06.024

URL : https://doi.org/10.1016/j.cell.2016.06.024

Q. Li, B. Xu, S. A. Michie, K. H. Rubins, R. D. Schreriber et al., Interferon-alpha initiates type 1 diabetes in nonobese diabetic mice, Proc. Natl. Acad. Sci. USA, vol.105, pp.12439-12444, 2008.
DOI : 10.1073/pnas.0806439105

URL : http://www.pnas.org/content/105/34/12439.full.pdf

C. J. Jung, S. Ménoret, L. Brusselle, L. Tesson, C. Usal et al., Comparative analysis of piggyBac, CRISPR/Cas9 and TALEN mediated BAC transgenesis in the zygote for the generation of humanized SIRPA rats, Sci. Rep, vol.6, p.31455, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02150699