S. Berrih-aknin, L. Panse, and R. , Myasthenia gravis: A comprehensive review of immune dysregulation and etiological mechanisms, Journal of Autoimmunity, vol.52, pp.90-100, 2014.
DOI : 10.1016/j.jaut.2013.12.011

A. Marx, F. Pfister, B. Schalke, G. Saruhan-direskeneli, A. Melms et al., The different roles of the thymus in the pathogenesis of the various myasthenia gravis subtypes, Autoimmunity Reviews, vol.12, issue.9, pp.875-84, 2013.
DOI : 10.1016/j.autrev.2013.03.007

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

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

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

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

J. Chen, G. Papp, P. Szodoray, and M. Zeher, The role of microRNAs in the pathogenesis of autoimmune diseases, Autoimmunity Reviews, vol.15, issue.12, pp.1171-80, 2016.
DOI : 10.1016/j.autrev.2016.09.003

J. Winter, S. Jung, S. Keller, R. Gregory, and S. Diederichs, Many roads to maturity: microRNA biogenesis pathways and their regulation, Nature Cell Biology, vol.11, issue.3, pp.228-262, 2009.
DOI : 10.1126/science.1138050

M. Ebert and P. Sharp, Roles for MicroRNAs in Conferring Robustness to Biological Processes, Cell, vol.149, issue.3, pp.515-539, 2012.
DOI : 10.1016/j.cell.2012.04.005

L. Gulyaeva and N. Kushlinskiy, Regulatory mechanisms of microRNA expression, Journal of Translational Medicine, vol.5, issue.6, p.143, 2016.
DOI : 10.1016/j.apsb.2015.01.002

A. Papadopoulou, J. Dooley, M. Linterman, W. Pierson, O. Ucar et al., 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.181-188, 2012.
DOI : 10.1006/meth.2001.1262

I. Khan, R. Taniguchi, K. Fasano, M. Anderson, and L. Jeker, Canonical microRNAs in thymic epithelial cells promote central tolerance, European Journal of Immunology, vol.321, issue.5, pp.1313-1322, 2014.
DOI : 10.1126/science.1159407

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/eji.201344079

N. Chunjie, N. Huijuan, Y. Zhao, W. Jianzhao, and Z. Xiaojian, 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.61-67, 2015.

Y. Xin, H. Cai, T. Lu, Y. Zhang, Y. Yang et al., miR-20b Inhibits T Cell Proliferation and Activation via NFAT Signaling Pathway in Thymoma-Associated Myasthenia Gravis, BioMed Research International, vol.7, issue.5, p.9595718, 2016.
DOI : 10.1074/jbc.M113.460287

URL : http://downloads.hindawi.com/journals/bmri/2016/9595718.pdf

E. Zhu, X. Wang, B. Zheng, Q. Wang, J. Hao et al., miR-20b Suppresses Th17 Differentiation and the Pathogenesis of Experimental Autoimmune Encephalomyelitis by Targeting ROR??t and STAT3, The Journal of Immunology, vol.192, issue.12, pp.5599-609, 2014.
DOI : 10.4049/jimmunol.1303488

J. Lou, Y. Wang, Z. Zhang, and W. Qiu, MiR-20b inhibits mycobacterium tuberculosis induced inflammation in the lung of mice through targeting NLRP3, Experimental Cell Research, vol.358, issue.2, pp.120-128, 2017.
DOI : 10.1016/j.yexcr.2017.06.007

J. Ingwersen, T. Menge, B. Wingerath, D. Kaya, J. Graf et al., Natalizumab restores aberrant miRNA expression profile in multiple sclerosis and reveals a critical role for miR-20b, Annals of Clinical and Translational Neurology, vol.11, issue.2, pp.43-55, 2015.
DOI : 10.1016/j.it.2005.08.014

J. Li, D. Qiu, Z. Chen, W. Du, J. Liu et al., Altered expression of miR-125a-5p in thymoma-associated myasthenia gravis and its down-regulation of foxp3 expression in Jurkat cells, Immunology Letters, vol.172, pp.47-55, 2016.
DOI : 10.1016/j.imlet.2016.02.005

T. Nishi, S. Yokoyama, S. Takamori, T. Matsuo, D. Murakami et al., Thymoma in Patient with Myasthenia Gravis Has Significantly Fewer Forkhead Box P3 Positive Lymphocytes than that without One, The Kurume Medical Journal, vol.61, issue.3.4, pp.65-71, 2015.
DOI : 10.2739/kurumemedj.MS65002

A. Balandina, S. Lecart, P. Dartevelle, A. Saoudi, and S. Berrih-aknin, Functional defect of regulatory CD4+CD25+ T cells in the thymus of patients with autoimmune myasthenia gravis, Blood, vol.105, issue.2, pp.735-776, 2005.
DOI : 10.1182/blood-2003-11-3900

S. Jander and G. Stoll, Increased serum levels of the interferon-?????inducing cytokine interleukin-18 in myasthenia gravis, Neurology, vol.59, issue.2, pp.287-296, 2002.
DOI : 10.1212/WNL.59.2.287

M. Souroujon, P. Maiti, T. Feferman, S. Im, L. Raveh et al., Suppression of Myasthenia Gravis by Antigen-Specific Mucosal Tolerance and Modulation of Cytokines and Costimulatory Factors, Annals of the New York Academy of Sciences, vol.15, issue.1, pp.533-539, 2003.
DOI : 10.1096/fj.01-0072com

M. Zhang, L. Peng, Y. Wang, J. Wang, J. Liu et al., Roles of A20 in autoimmune diseases, Immunologic Research, vol.9, issue.2, pp.337-381, 2016.
DOI : 10.1371/journal.pgen.1003750

S. Kim, K. Ramasamy, H. Bouamar, A. Lin, D. Jiang et al., MicroRNAs miR-125a and miR-125b constitutively activate the NF-kappaB pathway by targeting the tumor necrosis 24

, factor alpha-induced protein 3 (TNFAIP3, A20), Proc Natl Acad Sci U S A, vol.109, pp.7865-70, 2012.

J. Hamerman, J. Pottle, M. Ni, Y. He, Z. Zhang et al., Negative regulation of TLR signaling in myeloid cells-implications for autoimmune diseases, Immunological Reviews, vol.21, issue.1, pp.212-239, 2016.
DOI : 10.1158/1078-0432.CCR-14-2522

N. Avidan, L. Panse, R. Harbo, H. Bernasconi, P. Poulas et al., VAV1 and BAFF, via NFkB pathway, are genetic risk factors for myasthenia gravis, Annals of Clinical and Translational Neurology, 2014.

Y. Oshikawa, M. Jinnin, T. Makino, I. Kajihara, K. Makino et al., Decreased miR-7 Expression in the Skin and Sera of Patients with Dermatomyositis, Acta Dermato Venereologica, vol.93, issue.3, pp.273-279, 2013.
DOI : 10.2340/00015555-1459

A. Vaknin-dembinsky, H. Charbit, L. Brill, O. Abramsky, D. Gur-wahnon et al., Circulating microRNAs as biomarkers for rituximab therapy, in neuromyelitis optica (NMO), Journal of Neuroinflammation, vol.72, issue.9, p.179, 2016.
DOI : 10.1001/jamaneurol.2015.1276

URL : https://jneuroinflammation.biomedcentral.com/track/pdf/10.1186/s12974-016-0648-x

H. Nguyen, G. Dalmasso, Y. Yan, H. Laroui, S. Dahan et al., MicroRNA-7 Modulates CD98 Expression during Intestinal Epithelial Cell Differentiation, Journal of Biological Chemistry, vol.97, issue.Pt 2, pp.1479-89, 2010.
DOI : 10.1016/j.cyto.2008.07.016

URL : http://www.jbc.org/content/285/2/1479.full.pdf

H. Kurobe, C. Liu, T. Ueno, F. Saito, I. Ohigashi et al., CCR7-Dependent Cortex-to-Medulla Migration of Positively Selected Thymocytes Is Essential for Establishing Central Tolerance, Immunity, vol.24, issue.2, pp.165-77, 2006.
DOI : 10.1016/j.immuni.2005.12.011

D. Larizza, V. Calcaterra, and M. Martinetti, Autoimmune stigmata in Turner syndrome: When lacks an X chromosome, Journal of Autoimmunity, vol.33, issue.1, pp.25-30, 2009.
DOI : 10.1016/j.jaut.2009.03.002

T. Winarni, W. Chonchaiya, T. Sumekar, P. Ashwood, G. Morales et al.,

, Immune-mediated disorders among women carriers of fragile X premutation alleles

, American journal of medical genetics Part A, vol.158, pp.2473-81, 2012.

D. Pretto, J. Eid, C. Yrigollen, H. Tang, E. Loomis et al., mRNA isoforms in premutation carriers, Journal of Medical Genetics, vol.5, issue.1, pp.42-52, 2015.
DOI : 10.1186/gb-2004-5-2-r12

A. Naumann, N. Hochstein, S. Weber, E. Fanning, and W. Doerfler, A Distinct DNA-Methylation Boundary in the 5???- Upstream Sequence of the FMR1 Promoter Binds Nuclear Proteins and Is Lost in Fragile X Syndrome, The American Journal of Human Genetics, vol.85, issue.5, pp.606-622, 2009.
DOI : 10.1016/j.ajhg.2009.09.018

S. Lanni, M. Goracci, L. Borrelli, G. Mancano, P. Chiurazzi et al., Role of CTCF Protein in Regulating FMR1 Locus Transcription, PLoS Genetics, vol.98, issue.7, pp.1003601-1003626, 2013.
DOI : 10.1371/journal.pgen.1003601.s006

C. He, X. Zhao, H. Jiang, Z. Zhong, and R. Xu, Demethylation of miR-10b plays a suppressive role in ccRCC cells, International journal of clinical and experimental pathology, vol.8, pp.10595-604, 2015.