F. Truffault, V. De-montpreville, and B. Eymard, Thymic Germinal Centers and Corticosteroids in Myasthenia Gravis: an Immunopathological Study in 1035 Cases and a Critical Review, Clinical Reviews in Allergy & Immunology, vol.62, issue.202, pp.108-124, 2017.
DOI : 10.1016/S0003-4975(96)00376-1

URL : https://hal.archives-ouvertes.fr/hal-01338400

P. Zisimopoulou, P. Evangelakou, and J. Tzartos, A comprehensive analysis of the epidemiology and clinical characteristics of anti-LRP4 in myasthenia gravis, Journal of Autoimmunity, vol.52, 2013.
DOI : 10.1016/j.jaut.2013.12.004

K. Kondo, K. Takada, and &. Y. Takahama, Antigen processing and presentation in the thymus: implications for T cell repertoire selection, Current Opinion in Immunology, vol.46, pp.53-57, 2017.
DOI : 10.1016/j.coi.2017.03.014

L. Panse and R. S. Berrih-aknin, Thymic myoid cells protect thymocytes from apoptosis and modulate their differentiation: implication of the ERK and Akt signaling pathways, Cell Death and Differentiation, vol.2, issue.5, pp.463-472, 2005.
DOI : 10.4049/jimmunol.171.3.1285

M. S. Anderson, E. S. Venanzi, and L. Klein, Projection of an Immunological Self Shadow Within the Thymus by the Aire Protein, Science, vol.298, issue.5597, pp.1395-1401, 2002.
DOI : 10.1126/science.1075958

H. Takaba, Y. Morishita, and Y. Tomofuji, Fezf2 Orchestrates a Thymic Program of Self-Antigen Expression for Immune Tolerance, Cell, vol.163, issue.4, pp.975-987, 2015.
DOI : 10.1016/j.cell.2015.10.013

L. Mesnard-rouiller, J. Bismuth, and A. Wakkach, Thymic myoid cells express high levels of muscle genes, Journal of Neuroimmunology, vol.148, issue.1-2, pp.97-105, 2004.
DOI : 10.1016/j.jneuroim.2003.11.013

M. Giraud, R. Taubert, and C. Vandiedonck, An IRF8-binding promoter variant and AIRE control CHRNA1 promiscuous expression in thymus, Nature, vol.199, issue.7156, pp.934-937, 2007.
DOI : 10.1101/gr.8.3.175

N. Dragin, J. Bismuth, and G. Cizeron-clairac, Estrogen-mediated downregulation of AIRE influences sexual dimorphism in autoimmune diseases, Journal of Clinical Investigation, vol.126, issue.4, pp.1525-1537, 2016.
DOI : 10.1172/JCI81894DS1

URL : https://hal.archives-ouvertes.fr/hal-01310502

A. Wakkach, S. Poea, and E. Chastre, Establishment of a Human Thymic Myoid Cell Line, The American Journal of Pathology, vol.155, issue.4, pp.1229-1240, 1999.
DOI : 10.1016/S0002-9440(10)65225-X

J. E. Cowan, S. M. Parnell, and K. Nakamura, thymocyte development, The Journal of Experimental Medicine, vol.210, issue.4, pp.675-681, 2013.
DOI : 10.1038/23287

D. Nazzal, A. Gradolatto, and F. Truffault, Human thymus medullary epithelial cells promote regulatory T-cell generation by stimulating interleukin-2 production via ICOS ligand. Cell death & disease, p.1420, 2014.
DOI : 10.1038/cddis.2014.377

URL : https://hal.archives-ouvertes.fr/hal-01311608

N. Watanabe, Y. H. Wang, and H. K. Lee, Hassall's corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus, Nature, vol.107, issue.7054, pp.1181-1185, 2005.
DOI : 10.1172/JCI11734

S. Berrih-aknin, E. Morel, and F. Raimond, The Role of the Thymus in Myasthenia Gravis: Immunohistological and Immunological Studies in 115 Cases, Annals of the New York Academy of Sciences, vol.633, issue.1 Myasthenia Gr, pp.50-70, 1987.
DOI : 10.1016/0022-510X(81)90170-2

A. Balandina, S. Lecart, and P. Dartevelle, Functional defect of regulatory CD4+CD25+ T cells in the thymus of patients with autoimmune myasthenia gravis, Blood, vol.105, issue.2, pp.735-741, 2005.
DOI : 10.1182/blood-2003-11-3900

M. Thiruppathi, J. Rowin, and B. Ganesh, Impaired regulatory function in circulating CD4+CD25highCD127low/??? T cells in patients with myasthenia gravis, Clinical Immunology, vol.145, issue.3, pp.209-223, 2012.
DOI : 10.1016/j.clim.2012.09.012

A. Gradolatto, D. Nazzal, and F. Truffault, Both Treg cells and Tconv cells are defective in the Myasthenia gravis thymus: Roles of IL-17 and TNF-??, Journal of Autoimmunity, vol.52, pp.53-63, 2014.
DOI : 10.1016/j.jaut.2013.12.015

P. G. Isaacson, A. J. Norton, and &. J. Addis, THE HUMAN THYMUS CONTAINS A NOVEL POPULATION OF B LYMPHOCYTES, The Lancet, vol.330, issue.8574, pp.1488-1491, 1987.
DOI : 10.1016/S0140-6736(87)92622-5

T. Yamano, J. Nedjic, and M. Hinterberger, Thymic B Cells Are Licensed to Present Self Antigens for Central T Cell Tolerance Induction, Immunity, vol.42, issue.6, pp.1048-1061, 2015.
DOI : 10.1016/j.immuni.2015.05.013

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

S. Nunez, C. Moore, and B. Gao, The human thymus perivascular space is a functional niche for viral-specific plasma cells, Science immunology, vol.1, 2016.

J. W. Bradfield, Altered venules in the stimulated human thymus as evidence of lymphocyte recirculation, Clin. Exp. Immunol, vol.13, pp.243-252, 1973.

K. G. Flores, J. Li, and &. P. Hale, B cells in epithelial and perivascular compartments of human adult thymus, Human Pathology, vol.32, issue.9, pp.926-934, 2001.
DOI : 10.1053/hupa.2001.27106

A. Méraouna, G. Cizeron-clairac, and R. Le-panse, The chemokine CXCL13 is a key molecule in autoimmune myasthenia gravis, Blood, vol.108, issue.2, pp.432-440, 2006.
DOI : 10.1182/blood-2005-06-2383

X. Zhang, S. Liu, and T. Chang, Intrathymic Tfh/B Cells Interaction Leads to Ectopic GCs Formation and Anti-AChR Antibody Production: Central Role in Triggering MG Occurrence, Molecular Neurobiology, vol.23, issue.6, pp.120-131, 2016.
DOI : 10.1016/j.coi.2011.08.007

D. Safar, S. Berrih-aknin, and &. E. Morel, In vitro anti-acetylcholine receptor antibody synthesis by myasthenia gravis patient lymphocytes: Correlations with thymic histology and thymic epithelial-cell interactions, Journal of Clinical Immunology, vol.16, issue.3, pp.225-234, 1987.
DOI : 10.1007/BF00915728

C. Leprince, S. Cohen-kaminsky, and S. Berrih-aknin, Thymic B cells from myasthenia gravis patients are activated B cells. Phenotypic and functional analysis, J. Immunol, vol.145, pp.2115-2122, 1990.

A. Melms, B. C. Schalke, and T. Kirchner, Thymus in myasthenia gravis. Isolation of T-lymphocyte lines specific for the nicotinic acetylcholine receptor from thymuses of myasthenic patients., Journal of Clinical Investigation, vol.81, issue.3, pp.902-908, 1988.
DOI : 10.1172/JCI113401

S. M. Mclachlan, L. V. Nicholson, and G. Venables, Acetylcholine receptor antibody synthesis in lymphocyte cultures, J. Clin. Lab. Immunol, vol.5, pp.137-142, 1981.

K. Vrolix, J. Fraussen, and M. Losen, Clonal heterogeneity of thymic B cells from early-onset myasthenia gravis patients with antibodies against the acetylcholine receptor, Journal of Autoimmunity, vol.52, 2014.
DOI : 10.1016/j.jaut.2013.12.008

V. Guigou, D. Emilie, and S. Berrih-aknin, Individual germinal centres of myasthenia gravis human thymuses contain polyclonal activated B cells that express all the VH and VK families, Clinical & Experimental Immunology, vol.164, issue.2, pp.262-266, 1991.
DOI : 10.1212/WNL.26.11.1054

L. Panse, R. , G. Cizeron-clairac, and J. Bismuth, Microarrays Reveal Distinct Gene Signatures in the Thymus of Seropositive and Seronegative Myasthenia Gravis Patients and the Role of CC Chemokine Ligand 21 in Thymic Hyperplasia, The Journal of Immunology, vol.177, issue.11, pp.7868-7879, 2006.
DOI : 10.4049/jimmunol.177.11.7868

M. Sudres, M. Maurer, and M. Robinet, Preconditioned mesenchymal stem cells treat myasthenia gravis in a humanized preclinical model. JCI insight, p.89665, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01510169

G. I. Wolfe, H. J. Kaminski, and I. B. Aban, Randomized Trial of Thymectomy in Myasthenia Gravis, New England Journal of Medicine, vol.375, issue.6, pp.511-522, 2016.
DOI : 10.1056/NEJMoa1602489

U. H. Von-andrian, Homing and cellular traffic in lymph nodes, Nature Reviews Immunology, vol.99, issue.11, pp.867-878, 2003.
DOI : 10.1073/pnas.172368799

J. M. Weiss, P. Cufi, and J. Bismuth, SDF-1/CXCL12 recruits B cells and antigen-presenting cells to the thymus of autoimmune myasthenia gravis patients, Immunobiology, vol.218, issue.3, pp.373-381, 2013.
DOI : 10.1016/j.imbio.2012.05.006

S. Berrih-aknin, N. Ruhlmann, and J. Bismuth, CCL21 overexpressed on lymphatic vessels drives thymic hyperplasia in myasthenia, Annals of Neurology, vol.6, issue.4, pp.521-531, 2009.
DOI : 10.4049/jimmunol.173.8.4791

V. Angeli, F. Ginhoux, and J. Llodra, B Cell-Driven Lymphangiogenesis in Inflamed Lymph Nodes Enhances Dendritic Cell Mobilization, Immunity, vol.24, issue.2, pp.203-215, 2006.
DOI : 10.1016/j.immuni.2006.01.003

URL : https://doi.org/10.1016/j.immuni.2006.10.002

M. &. Miyasaka and . Tanaka, Lymphocyte trafficking across high endothelial venules: dogmas and enigmas, Nature Reviews Immunology, vol.168, issue.5, pp.360-370, 2004.
DOI : 10.4049/jimmunol.168.11.5441

J. G. Cyster, Chemokines and Cell Migration in Secondary Lymphoid Organs, Science, vol.286, issue.5447, pp.2098-2102, 1999.
DOI : 10.1126/science.286.5447.2098

T. Feferman, P. K. Maiti, and S. Berrih-aknin, Overexpression of IFN-Induced Protein 10 and Its Receptor CXCR3 in Myasthenia Gravis, The Journal of Immunology, vol.174, issue.9, pp.5324-5331, 2005.
DOI : 10.4049/jimmunol.174.9.5324

M. Colombara, V. Antonini, and A. P. Riviera, Constitutive Activation of p38 and ERK1/2 MAPKs in Epithelial Cells of Myasthenic Thymus Leads to IL-6 and RANTES Overexpression: Effects on Survival and Migration of Peripheral T and B Cells, The Journal of Immunology, vol.175, issue.10, pp.7021-7028, 2005.
DOI : 10.4049/jimmunol.175.10.7021

C. Cordiglieri, R. Marolda, and S. Franzi, Innate immunity in myasthenia gravis thymus: Pathogenic effects of Toll-like receptor 4 signaling on autoimmunity, Journal of Autoimmunity, vol.52, 2014.
DOI : 10.1016/j.jaut.2013.12.013

F. Annunziato, P. Romagnani, and L. Cosmi, Chemokines and lymphopoiesis in human thymus, Trends in Immunology, vol.22, issue.5, pp.277-281, 2001.
DOI : 10.1016/S1471-4906(01)01889-0

Y. Nagane, K. Utsugisawa, and D. Obara, Dendritic cells in hyperplastic thymuses from patients with myasthenia gravis, Muscle & Nerve, vol.93, issue.5, pp.582-589, 2003.
DOI : 10.1073/pnas.93.6.2588

D. F. Legler, M. Loetscher, and R. S. Roos, B Cell???attracting Chemokine 1, a Human CXC Chemokine Expressed in Lymphoid Tissues, Selectively Attracts B Lymphocytes via BLR1/CXCR5, The Journal of Experimental Medicine, vol.55, issue.4, pp.655-660, 1998.
DOI : 10.1084/jem.185.1.111

J. M. Weiss, M. Robinet, and R. Aricha, Novel CXCL13 transgenic mouse: inflammation drives pathogenic effect of CXCL13 in experimental myasthenia gravis, Oncotarget, vol.7, issue.7, pp.7550-7562, 2016.
DOI : 10.18632/oncotarget.6885

URL : https://hal.archives-ouvertes.fr/hal-01293674

E. Meinl, W. E. Klinkert, and &. H. Wekerle, The thymus in myasthenia gravis. Changes typical for the human disease are absent in experimental autoimmune myasthenia gravis of the Lewis rat, Am. J. Pathol, vol.139, pp.995-1008, 1991.

S. D. Katzman, Pathogen-imposed skewing of mouse chemokine and cytokine expression at the infected tissue site, Journal of Clinical Investigation, vol.118, pp.801-811, 2008.
DOI : 10.1172/JCI33174

D. L. Hodge, D. Reynolds, and F. M. Cerban, MCP-1/CCR2 interactions direct migration of peripheral B and T lymphocytes to the thymus during acute infectious/inflammatory processes, European Journal of Immunology, vol.150, issue.10, pp.2644-2654, 2012.
DOI : 10.4049/jimmunol.180.5.2886

G. R. Foster, S. H. Masri, and R. David, IFN-?? Subtypes Differentially Affect Human T Cell Motility, The Journal of Immunology, vol.173, issue.3, pp.1663-1670, 2004.
DOI : 10.4049/jimmunol.173.3.1663

URL : http://www.jimmunol.org/content/jimmunol/173/3/1663.full.pdf

P. Cavalcante, M. Barberis, and M. Cannone, Detection of poliovirus-infected macrophages in thymus of patients with myasthenia gravis, Neurology, vol.74, issue.14, pp.1118-1126, 2010.
DOI : 10.1212/WNL.0b013e3181d7d884

P. Cavalcante, B. Serafini, and B. Rosicarelli, Epstein-barr virus persistence and reactivation in myasthenia gravis thymus, Annals of Neurology, vol.67, pp.726-738, 2010.
DOI : 10.1002/ana.21902

T. S. Kawai and . Akira, The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors, Nature Immunology, vol.1799, issue.5, pp.373-384, 2010.
DOI : 10.1126/science.1179050

M. Robinet, S. Maillard, and M. A. Cron, Review on Toll-Like Receptor Activation in Myasthenia Gravis: Application to the Development of New Experimental Models, Clinical Reviews in Allergy & Immunology, vol.46, issue.1, 2017.
DOI : 10.1002/art.10304

URL : https://hal.archives-ouvertes.fr/hal-01321397

P. Cufi, N. Dragin, and J. M. Weiss, Implication of double-stranded RNA signaling in the etiology of autoimmune myasthenia gravis, Annals of Neurology, vol.29, issue.suppl, pp.281-293, 2013.
DOI : 10.1016/j.vaccine.2010.08.002

D. Iwakiri, L. Zhou, and M. Samanta, Epstein-Barr virus (EBV)???encoded small RNA is released from EBV-infected cells and activates signaling from toll-like receptor 3, The Journal of Experimental Medicine, vol.13, issue.10, 2009.
DOI : 10.4049/jimmunol.178.7.4548

URL : http://jem.rupress.org/content/jem/206/10/2091.full.pdf

L. H. Kasper and . Reder, Immunomodulatory activity of interferon-beta, Annals of Clinical and Translational Neurology, vol.348, issue.Pt 2, pp.622-631, 2014.
DOI : 10.1056/NEJMoa020696

L. Bennett, A. K. Palucka, and E. Arce, Interferon and Granulopoiesis Signatures in Systemic Lupus Erythematosus Blood, The Journal of Experimental Medicine, vol.149, issue.6, pp.711-723, 2003.
DOI : 10.1136/ard.58.5.309

URL : http://jem.rupress.org/content/jem/197/6/711.full.pdf

I. Rauch, M. Muller, and &. T. Decker, The regulation of inflammation by interferons and their STATs, JAK-STAT, vol.69, issue.1, p.23820, 2013.
DOI : 10.1016/j.jneuroim.2004.07.016

J. P. Stubgen, Interferon alpha and neuromuscular disorders, Journal of Neuroimmunology, vol.207, issue.1-2, pp.3-17, 2009.
DOI : 10.1016/j.jneuroim.2008.12.008

A. Meager, M. Wadhwa, and P. Dilger, Anti-cytokine autoantibodies in autoimmunity: preponderance of neutralizing autoantibodies against interferon-alpha, interferon-omega and interleukin-12 in patients with thymoma and/or myasthenia gravis, Clinical and Experimental Immunology, vol.161, issue.1, 2003.
DOI : 10.1084/jem.192.12.1775

S. Poea-guyon, P. Christadoss, and R. Le-panse, Effects of Cytokines on Acetylcholine Receptor Expression: Implications for Myasthenia Gravis, The Journal of Immunology, vol.174, issue.10, pp.5941-5949, 2005.
DOI : 10.4049/jimmunol.174.10.5941

P. Cufi, N. Dragin, and N. Ruhlmann, Central role of interferon-beta in thymic events leading to myasthenia gravis, Journal of Autoimmunity, vol.52, pp.44-52, 2014.
DOI : 10.1016/j.jaut.2013.12.016

URL : https://hal.archives-ouvertes.fr/hal-01514459

P. Cufi, P. Soussan, and F. Truffault, Thymoma-associated myasthenia gravis: On the search for a pathogen signature, Journal of Autoimmunity, vol.52, pp.29-35, 2014.
DOI : 10.1016/j.jaut.2013.12.018

T. Punga, R. Le-panse, and M. Andersson, Circulating miRNAs in myasthenia gravis: miR-150-5p as a new potential biomarker, Annals of Clinical and Translational Neurology, vol.17, issue.1, pp.49-58, 2014.
DOI : 10.1517/14728222.2013.773311

G. Nogales-gadea, A. Ramos-fransi, and X. Suarez-calvet, Analysis of serum miRNA profiles of myasthenia gravis patients. PloS one, p.91927, 2014.

N. Chunjie, N. Huijuan, and Y. Zhao, Disease-specific signature of serum miR-20b and its targets IL-8 and IL-25, in myasthenia gravis patients, Eur. Cytokine Netw, vol.26, pp.61-66, 2015.

Z. Cheng, S. Qiu, and L. Jiang, MiR-320a is Downregulated in Patients with Myasthenia Gravis and Modulates Inflammatory Cytokines Production by Targeting Mitogen-activated Protein Kinase 1, Journal of Clinical Immunology, vol.118, issue.3, pp.567-576, 2013.
DOI : 10.1111/j.1365-2567.2006.02398.x

C. Barzago, J. Lum, and P. Cavalcante, A novel infection- and inflammation-associated molecular signature in peripheral blood of myasthenia gravis patients, Immunobiology, vol.221, issue.11, pp.1227-1236, 2016.
DOI : 10.1016/j.imbio.2016.06.012

G. Skogberg, J. Gudmundsdottir, S. Van-der, and . Post, Characterization of Human Thymic Exosomes, PLoS ONE, vol.40, issue.7, p.67554, 2013.
DOI : 10.1371/journal.pone.0067554.s005

L. Linhares-lacerda, C. C. Palu, and M. Ribeiro-alves, Differential Expression of microRNAs in Thymic Epithelial Cells from Trypanosoma cruzi Acutely Infected Mice: Putative Role in Thymic Atrophy, Frontiers in Immunology, vol.63, p.428, 2015.
DOI : 10.1002/art.30115

A. S. Papadopoulou, J. Dooley, and M. A. Linterman, The thymic epithelial microRNA network elevates the threshold for infection-associated thymic involution via miR-29a mediated suppression of the IFN-?? receptor, Nature Immunology, vol.175, issue.2, pp.181-187, 2012.
DOI : 10.1006/meth.2001.1262