E. Corsiero, A. Nerviani, M. Bombardieri, and C. Pitzalis, Ectopic Lymphoid Structures: Powerhouse of Autoimmunity, Frontiers in Immunology, vol.73, issue.10, 2016.
DOI : 10.1136/annrheumdis-2012-202861

J. Weiss, P. Cufi, J. Bismuth, B. Eymard, E. Fadel et al., SDF-1/CXCL12 recruits B cells and antigen-presenting cells to the thymus of autoimmune myasthenia gravis patients, Immunobiology, vol.218, issue.3, pp.373-81, 2013.
DOI : 10.1016/j.imbio.2012.05.006

M. Mitsdoerffer and A. Peters, Tertiary Lymphoid Organs in Central Nervous System Autoimmunity, Frontiers in Immunology, vol.158, issue.1, 2016.
DOI : 10.1016/j.clim.2015.03.009

URL : http://doi.org/10.3389/fimmu.2016.00451

F. Truffault, V. De-montpreville, B. Eymard, T. Sharshar, L. Panse et al., 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.108-132, 2017.
DOI : 10.1016/S0003-4975(96)00376-1

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

G. Wolfe, H. Kaminski, I. Aban, G. Minisman, H. Kuo et al., Randomized Trial of Thymectomy in Myasthenia Gravis, New England Journal of Medicine, vol.375, issue.6, pp.511-533, 2016.
DOI : 10.1056/NEJMoa1602489

M. Sudres, M. Maurer, R. M. Bismuth, J. Truffault, F. Girard et al., Preconditioned mesenchymal stem cells treat myasthenia gravis in a humanized preclinical model, JCI Insight, vol.2, issue.7, 2017.
DOI : 10.1172/jci.insight.89665DS1

URL : https://hal.archives-ouvertes.fr/hal-01510169

J. Weiss, P. Cufi, L. Panse, R. Berrih-aknin, and S. , The thymus in autoimmune Myasthenia Gravis: Paradigm for a tertiary lymphoid organ, Revue Neurologique, vol.169, issue.8-9, pp.640-649, 2013.
DOI : 10.1016/j.neurol.2013.02.005

S. Berrih-aknin, N. Ruhlmann, J. Bismuth, G. Cizeron-clairac, E. Zelman et al., CCL21 overexpressed on lymphatic vessels drives thymic hyperplasia in myasthenia, Annals of Neurology, vol.6, issue.4, pp.521-552, 2009.
DOI : 10.4049/jimmunol.173.8.4791

N. Ruddle, High Endothelial Venules and Lymphatic Vessels in Tertiary Lymphoid Organs: Characteristics, Functions, and Regulation, Frontiers in Immunology, vol.11, issue.3, 2016.
DOI : 10.1089/lrb.2013.0013

URL : http://doi.org/10.3389/fimmu.2016.00491

A. Méraouna, G. Cizeron-clairac, L. Panse, R. Bismuth, J. Truffault et al., The chemokine CXCL13 is a key molecule in autoimmune myasthenia gravis, Blood, vol.108, issue.2, pp.432-472, 2006.
DOI : 10.1182/blood-2005-06-2383

L. Panse, R. Cizeron-clairac, G. Bismuth, J. Berrih-aknin, and S. , 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.7868-79, 2006.
DOI : 10.4049/jimmunol.177.11.7868

A. Wakkach, T. Guyon, C. Bruand, S. Tzartos, S. Cohen-kaminsky et al., Expression of acetylcholine receptor genes in human thymic epithelial cells: implications for myasthenia gravis, J Immunol, vol.157, issue.8, pp.3752-60, 1996.

X. Zhang, S. Liu, T. Chang, J. Xu, C. Zhang et al., 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.120-151, 2016.
DOI : 10.1016/j.coi.2011.08.007

A. Melms, B. Schalke, T. Kirchner, H. Muller-hermelink, E. Albert et al., 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.902-910, 1988.
DOI : 10.1172/JCI113401

C. Leprince, S. Cohen-kaminsky, S. Berrih-aknin, B. Vernet-der-garabedian, D. Treton et al., Thymic B cells from myasthenia gravis patients are activated B cells phenotypic and functional analysis, J Immunol, issue.7, pp.1452115-1452137, 1990.

K. Vrolix, J. Fraussen, M. Losen, J. Stevens, K. Lazaridis et al., 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

G. Cizeron-clairac, L. Panse, R. Frenkian-cuvelier, M. Méraouna, A. Truffault et al., Thymus and Myasthenia Gravis: What can we learn from DNA microarrays?, Journal of Neuroimmunology, vol.201, issue.202, pp.1-20257, 2008.
DOI : 10.1016/j.jneuroim.2008.06.028

P. Cufi, N. Dragin, J. Weiss, P. Martinez-martinez, D. Baets et al., Implication of double-stranded RNA signaling in the etiology of autoimmune myasthenia gravis, Annals of Neurology, vol.29, issue.suppl, pp.281-93, 2013.
DOI : 10.1016/j.vaccine.2010.08.002

P. Cufi, N. Dragin, N. Ruhlmann, J. Weiss, E. Fadel et al., 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

N. Dragin, J. Bismuth, G. Cizeron-clairac, M. Biferi, C. Berthault et al., Estrogen-mediated downregulation of AIRE influences sexual dimorphism in autoimmune diseases, Journal of Clinical Investigation, vol.126, issue.4, pp.1525-1562, 2016.
DOI : 10.1172/JCI81894DS1

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

P. Cavalcante, P. Cufi, R. Mantegazza, S. Berrih-aknin, P. Bernasconi et al., Etiology of myasthenia gravis: Innate immunity signature in pathological thymus, Autoimmunity Reviews, vol.12, issue.9, pp.863-74, 2013.
DOI : 10.1016/j.autrev.2013.03.010

D. Lucchesi and M. Bombardieri, The role of viruses in autoreactive B cell activation within tertiary lymphoid structures in autoimmune diseases, Journal of Leukocyte Biology, vol.94, issue.6, pp.1191-1200, 2013.
DOI : 10.1189/jlb.0413240

P. Bernasconi, M. Barberis, F. Baggi, L. Passerini, M. Cannone et al., Increased Toll-Like Receptor 4 Expression in Thymus of Myasthenic Patients with Thymitis and Thymic Involution, The American Journal of Pathology, vol.167, issue.1, pp.129-168, 2005.
DOI : 10.1016/S0002-9440(10)62960-4

C. Cordiglieri, R. Marolda, S. Franzi, C. Cappelletti, C. Giardina et al., Innate immunity in myasthenia gravis thymus: Pathogenic effects of Toll-like receptor 4 signaling on autoimmunity, Journal of Autoimmunity, vol.52, pp.74-89, 2014.
DOI : 10.1016/j.jaut.2013.12.013

P. Cavalcante, B. Galbardi, S. Franzi, S. Marcuzzo, C. Barzago et al., Increased expression of Toll-like receptors 7 and 9 in myasthenia gravis thymus characterized by active Epstein???Barr virus infection, Immunobiology, vol.221, issue.4, pp.516-543, 2016.
DOI : 10.1016/j.imbio.2015.12.007

M. Robinet, S. Maillard, M. Cron, S. Berrih-aknin, L. Panse et al., 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, pp.133-180, 2017.
DOI : 10.1002/art.10304

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

E. Tuzun, S. Berrih-aknin, T. Brenner, L. Kusner, L. Panse et al., Guidelines for standard preclinical experiments in the mouse model of myasthenia gravis induced by acetylcholine receptor immunization, Experimental Neurology, vol.270, pp.11-18, 2015.
DOI : 10.1016/j.expneurol.2015.02.009

E. Meinl, W. 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, issue.5, pp.995-1008, 1991.

J. Weiss, R. M. Aricha, R. Cufi, P. Villeret, B. Lantner et al., Novel CXCL13 transgenic mouse: inflammation drives pathogenic effect of CXCL13 in experimental myasthenia gravis, Oncotarget, vol.7, issue.7, pp.7550-626885, 2016.
DOI : 10.18632/oncotarget.6885

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

Y. Graus, P. Van-breda-vriesman, and M. De-baets, Characterization of anti-acetylcholine receptor (AChR) antibodies from mice differing in susceptibility for experimental autoimmune myasthenia gravis (EAMG), Clinical & Experimental Immunology, vol.81, issue.256, pp.506-519, 1993.
DOI : 10.1212/WNL.39.8.1057

A. Krieg and J. Vollmer, Toll-like receptors 7, 8, and 9: linking innate immunity to autoimmunity, Immunological Reviews, vol.160, issue.1, pp.251-69, 2007.
DOI : 10.1073/pnas.231606698

Y. Liu, H. Yin, M. Zhao, and Q. Lu, TLR2 and TLR4 in Autoimmune Diseases: a Comprehensive Review, Clinical Reviews in Allergy & Immunology, vol.146, issue.Suppl 54, pp.136-183, 2014.
DOI : 10.1016/j.jep.2013.01.009

. Pitié-salpêtrière and . Hospital, This work was supported by grants from the, Agence Nationale de la Recherche

J. Kleinnijenhuis, M. Oosting, L. Joosten, M. Netea, and R. Van-crevel, Innate immune recognition of Mycobacterium tuberculosis, Clin Dev Immunol, pp.405310-405320, 2011.

W. Allman, S. Saini, E. Tuzun, and P. Christadoss, Characterization of peripheral blood acetylcholine receptor-binding B cells in experimental myasthenia gravis, Cellular Immunology, vol.271, issue.2, pp.292-300, 2011.
DOI : 10.1016/j.cellimm.2011.07.007

Y. Ma and A. Ross, Toll-Like Receptor 3 Ligand and Retinoic Acid Enhance Germinal Center Formation and Increase the Tetanus Toxoid Vaccine Response, Clinical and Vaccine Immunology, vol.16, issue.10, pp.1476-84, 2009.
DOI : 10.1128/CVI.00282-09

URL : http://cvi.asm.org/content/16/10/1476.full.pdf

L. Genestier, M. Taillardet, P. Mondiere, H. Gheit, C. Bella et al., TLR Agonists Selectively Promote Terminal Plasma Cell Differentiation of B Cell Subsets Specialized in Thymus-Independent Responses, The Journal of Immunology, vol.178, issue.12, pp.7779-86, 2007.
DOI : 10.4049/jimmunol.178.12.7779

B. Hou, P. Saudan, G. Ott, M. Wheeler, J. M. Kuzmich et al., Selective Utilization of Toll-like Receptor and MyD88 Signaling in B Cells for Enhancement of the Antiviral Germinal Center Response, Immunity, vol.34, issue.3, pp.375-84, 2011.
DOI : 10.1016/j.immuni.2011.01.011

H. Guay, T. Andreyeva, R. Garcea, R. Welsh, and E. Szomolanyi-tsuda, MyD88 Is Required for the Formation of Long-Term Humoral Immunity to Virus Infection, The Journal of Immunology, vol.178, issue.8, pp.5124-5155, 2007.
DOI : 10.4049/jimmunol.178.8.5124

E. Walsh, P. Pisitkun, E. Voynova, J. Deane, B. Scott et al., Dual signaling by innate and adaptive immune receptors is required for TLR7-induced B-cell-mediated autoimmunity, Proceedings of the National Academy of Sciences, vol.102, issue.5, pp.16276-81, 2012.
DOI : 10.1016/S0092-8674(00)00078-7

URL : http://www.pnas.org/content/109/40/16276.full.pdf

C. Soni, E. Wong, P. Domeier, T. Khan, T. Satoh et al., B Cell???Intrinsic TLR7 Signaling Is Essential for the Development of Spontaneous Germinal Centers, The Journal of Immunology, vol.193, issue.9, pp.4400-4414, 2014.
DOI : 10.4049/jimmunol.1401720

A. Boneparth, W. Huang, R. Bethunaickan, M. Woods, R. Sahu et al., TLR7 Influences Germinal Center Selection in Murine SLE, PLOS ONE, vol.191, issue.3, 2015.
DOI : 10.1371/journal.pone.0119925.s004

URL : http://doi.org/10.1371/journal.pone.0119925

J. Clingan and M. Matloubian, B Cell-Intrinsic TLR7 Signaling Is Required for Optimal B Cell Responses during Chronic Viral Infection, The Journal of Immunology, vol.191, issue.2, pp.810-818, 2013.
DOI : 10.4049/jimmunol.1300244

URL : http://www.jimmunol.org/content/jimmunol/191/2/810.full.pdf

I. Moisini, W. Huang, R. Bethunaickan, R. Sahu, P. Ricketts et al., Locus and IFN-?? Fine-Tune Germinal Center B Cell Selection in Murine Systemic Lupus Erythematosus, The Journal of Immunology, vol.189, issue.9, pp.4305-4317, 2012.
DOI : 10.4049/jimmunol.1200745

URL : http://www.jimmunol.org/content/jimmunol/189/9/4305.full.pdf

J. Bessa, M. Kopf, and M. Bachmann, Cutting Edge: IL-21 and TLR Signaling Regulate Germinal Center Responses in a B Cell-Intrinsic Manner, The Journal of Immunology, vol.184, issue.9, pp.4615-4624, 2010.
DOI : 10.4049/jimmunol.0903949

URL : http://www.jimmunol.org/content/jimmunol/184/9/4615.full.pdf

A. Das, B. Heesters, A. Bialas, J. O-'flynn, I. Rifkin et al., Follicular Dendritic Cell Activation by TLR Ligands Promotes Autoreactive B Cell Responses, Immunity, vol.46, issue.1, pp.106-125, 2017.
DOI : 10.1016/j.immuni.2016.12.014

A. Krieg, A. Yi, S. Matson, T. Waldschmidt, G. Bishop et al., CpG motifs in bacterial DNA trigger direct B-cell activation, Nature, vol.374, issue.6522, pp.546-555, 1995.
DOI : 10.1038/374546a0

J. Jung, A. Yi, X. Zhang, J. Choe, L. Li et al., Distinct Response of Human B Cell Subpopulations in Recognition of an Innate Immune Signal, CpG DNA, The Journal of Immunology, vol.169, issue.5, pp.2368-73, 2002.
DOI : 10.4049/jimmunol.169.5.2368

D. Rookhuizen and A. Defranco, Toll-like receptor 9 signaling acts on multiple elements of the germinal center to enhance antibody responses, Proceedings of the National Academy of Sciences, vol.204, issue.7, pp.3224-3257, 2014.
DOI : 10.1084/jem.20062648

E. Leadbetter, I. Rifkin, A. Hohlbaum, B. Beaudette, M. Shlomchik et al., Chromatin???IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors, Nature, vol.416, issue.6881, pp.603-610, 2002.
DOI : 10.1038/416603a

B. Desnues, A. Macedo, A. Roussel-queval, J. Bonnardel, S. Henri et al., TLR8 on dendritic cells and TLR9 on B cells restrain TLR7-mediated spontaneous autoimmunity in C57BL/6 mice, Proceedings of the National Academy of Sciences, vol.56, issue.10, pp.1497-502, 2014.
DOI : 10.1002/art.22916

M. Heikenwalder, M. Polymenidou, T. Junt, C. Sigurdson, H. Wagner et al., Lymphoid follicle destruction and immunosuppression after repeated CpG oligodeoxynucleotide administration, Nature Medicine, vol.10, issue.2, pp.187-92, 2004.
DOI : 10.1038/nm987

P. Cavalcante, B. Serafini, B. Rosicarelli, L. Maggi, M. Barberis et al., Epstein-barr virus persistence and reactivation in myasthenia gravis thymus, Annals of Neurology
DOI : 10.1002/ana.21902

M. Billard, A. Gruver, and G. Sempowski, Acute Endotoxin-Induced Thymic Atrophy Is Characterized By Intrathymic Inflammatory and Wound Healing Responses, PLoS ONE, vol.21, issue.3, 2011.
DOI : 10.1371/journal.pone.0017940.s011

URL : http://doi.org/10.1371/journal.pone.0017940

A. Martin, E. Coronel, G. Sano, S. Chen, G. Vassileva et al., A Novel Model for Lymphocytic Infiltration of the Thyroid Gland Generated by Transgenic Expression of the CC Chemokine CCL21, The Journal of Immunology, vol.173, issue.8, pp.4791-4799, 2004.
DOI : 10.4049/jimmunol.173.8.4791

G. Janossy, J. Snajdr, and M. Simak-ellis, Patterns of B-lymphocyte gene expression elicited by lipopolysaccharide mitogen, Immunology, vol.30, issue.6, pp.799-810, 1976.

L. Barrio, J. Saez-de-guinoa, and Y. Carrasco, TLR4 Signaling Shapes B Cell Dynamics via MyD88-Dependent Pathways and Rac GTPases, The Journal of Immunology, vol.191, issue.7, pp.3867-75, 2013.
DOI : 10.4049/jimmunol.1301623

I. Hwang, C. Park, K. Harrison, and J. Kehrl, TLR4 signaling augments B lymphocyte migration and overcomes the restriction that limits access to germinal center dark zones, The Journal of Experimental Medicine, vol.32, issue.12, pp.2641-57, 2009.
DOI : 10.4049/jimmunol.176.4.2142

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2806604

A. Garin, M. Meyer-hermann, M. Contie, M. Figge, V. Buatois et al., Toll-like Receptor 4 Signaling by Follicular Dendritic Cells Is Pivotal for Germinal Center Onset and Affinity Maturation, Immunity, vol.33, issue.1, pp.84-95, 2010.
DOI : 10.1016/j.immuni.2010.07.005

URL : http://doi.org/10.1016/j.immuni.2010.07.005

S. Berrih-aknin, E. Morel, F. Raimond, D. Safar, C. Gaud et al., 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, 1987.
DOI : 10.1016/0022-510X(81)90170-2

S. Nandula, Y. Scindia, P. Dey, H. Bagavant, and U. Deshmukh, Activation of innate immunity accelerates sialoadenitis in a mouse model for Sj??gren???s syndrome-like disease, Oral Diseases, vol.5, issue.8, 2011.
DOI : 10.1038/ni1087

M. Asada, A. Nishio, T. Akamatsu, J. Tanaka, K. Saga et al., Analysis of Humoral Immune Response in Experimental Autoimmune Pancreatitis in Mice, Pancreas, vol.39, issue.2, pp.224-255, 2010.
DOI : 10.1097/MPA.0b013e3181bab5e2

J. Hwang, T. Randall, and A. Silva-sanchez, Inducible Bronchus-Associated Lymphoid Tissue: Taming Inflammation in the Lung, Frontiers in Immunology, vol.40, issue.4, 2016.
DOI : 10.1016/j.bcmd.2007.06.026

T. Lee, J. Huang, C. Liu, H. Chen, Y. Chen et al., Featured Article: Interactions of surface-expressed TLR-4 and endosomal TLR-9 accelerate lupus progression in anti-dsDNA antibody transgenic mice, Experimental Biology and Medicine, vol.183, issue.6, pp.715-2310, 2014.
DOI : 10.4049/jimmunol.0901031