P. Dean and D. Pinkel, High resolution dual laser flow cytometry . The journal of histochemistry and cytochemistry: official journal of the, Histochemistry Society, vol.26, issue.8, pp.622-627, 1978.
DOI : 10.1177/26.8.357646

R. Drapkin, M. Adreeff, B. Koziner, A. Strife, D. Wisniewski et al., Subpopulations of human peripheral blood cells: Analysis of granulocytic progenitor cells by flow cytometry and immunologic surface markers, American Journal of Hematology, vol.36, issue.2, pp.163-172, 1979.
DOI : 10.1002/ajh.2830070209

E. Matutes, K. Owusu-ankomah, R. Morilla, G. Marco, J. Houlihan et al., The immunological profile of B-cell disorders and proposal of a scoring system for the diagnosis of CLL, Leukemia, vol.8, issue.10, pp.1640-1645, 1994.

C. Goujard, M. Bonarek, L. Meyer, F. Bonnet, M. Chaix et al., CD4 Cell Count and HIV DNA Level Are Independent Predictors of Disease Progression after Primary HIV Type 1 Infection in Untreated Patients, Clinical Infectious Diseases, vol.42, issue.5, pp.709-71510500213, 1086.
DOI : 10.1086/500213

U. Sack, A. Boldt, N. Mallouk, R. Gruber, V. Krenn et al., Cellular analyses in the monitoring of autoimmune diseases, Autoimmunity Reviews, vol.15, issue.9, pp.883-889, 2016.
DOI : 10.1016/j.autrev.2016.07.010

L. Simonin, E. Pasquier, C. Leroyer, D. Cornec, J. Lemerle et al., Lymphocyte disturbances in primary antiphospholipid syndrome and application to venous thromboembolism followup, Clin Rev Allergy Immunol, pp.1-14, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01352093

M. Milin, D. Cornec, M. Chastaing, V. Griner, S. Berrouiguet et al., Sicca symptoms are associated with similar fatigue, anxiety, depression, and quality-of-life impairments in patients with and without primary Sj??gren's syndrome, Joint Bone Spine, vol.83, issue.6, pp.681-685
DOI : 10.1016/j.jbspin.2015.10.005

D. Cornec, V. Devauchelle-pensec, X. Mariette, S. Jousse-joulin, J. Berthelot et al., Severe health-related quality-of-life impairment in active primary Sjogren's syndrome is driven by patient-reported outcomes: Data from a large therapeutic trial, Arthritis Care Res, 2016.

G. Carvajal-alegria, D. Guellec, V. Devauchelle-pensec, and A. Saraux, Is there specific neurological disorders of primary Sj??gren's syndrome?, Joint Bone Spine, vol.82, issue.2, pp.86-89, 2015.
DOI : 10.1016/j.jbspin.2014.04.002

G. Carvajal-alegria, D. Guellec, M. X. Gottenberg, J. Dernis, E. Dubost et al., Epidemiology of neurological manifestations in Sj??gren's syndrome: data from the French ASSESS Cohort, RMD Open, vol.2, issue.1, pp.179-189, 2016.
DOI : 10.1136/rmdopen-2015-000179

C. Darrieutort-laffite, V. Andre, G. Hayem, A. Saraux, L. Guern et al., Sj??gren's syndrome complicated by interstitial cystitis: A case series and literature review, Joint Bone Spine, vol.82, issue.4, pp.245-250, 2015.
DOI : 10.1016/j.jbspin.2014.12.007

C. Vitali, S. Bombardieri, R. Jonsson, H. Moutsopoulos, E. Alexander et al., Classification criteria for Sjogren's syndrome: a revised version of the European criteria proposed by the American-European Consensus Group, Annals of the Rheumatic Diseases, vol.61, issue.6, pp.554-558
DOI : 10.1136/ard.61.6.554

S. Shiboski, C. Shiboski, L. Criswell, A. Baer, S. Challacombe et al., American College of Rheumatology classification criteria for Sj??gren's syndrome: A data-driven, expert consensus approach in the Sj??gren's International Collaborative Clinical Alliance Cohort, Arthritis Care & Research, vol.66, issue.Suppl, pp.475-487
DOI : 10.1002/acr.21591

D. Cornec, A. Saraux, B. Cochener, J. Pers, S. Jousse-joulin et al., Level of agreement between 2002 American???European Consensus Group and 2012 American College of Rheumatology classification criteria for Sj??gren???s syndrome and reasons for discrepancies, Arthritis Research & Therapy, vol.16, issue.2, pp.74-84
DOI : 10.1136/annrheumdis-2013-203953

D. Guellec, D. Cornec, S. Jousse-joulin, T. Marhadour, P. Marcorelles et al., Diagnostic value of labial minor salivary gland biopsy for Sj??gren's syndrome: A systematic review, Autoimmunity Reviews, vol.12, issue.3, pp.416-420
DOI : 10.1016/j.autrev.2012.08.001

C. Capaldo, C. Alegria, G. Cornec, D. Jousse-joulin, S. Devauchelle-pensec et al., The active immunological profile in patients with primary Sjogren's syndrome is restricted to typically encountered autoantibodies. Clinical and experimental rheumatology 17 A comprehensive review of autoantibodies in primary Sjogren's syndrome: clinical phenotypes and regulatory mechanisms, J Autoimmun, vol.51, pp.67-74, 2014.

D. Cornec, C. Jamin, and J. Pers, Sj??gren's syndrome: Where do we stand, and where shall we go?, Journal of Autoimmunity, vol.51, 2014.
DOI : 10.1016/j.jaut.2014.02.006

J. Gottenberg, F. Lavie, K. Abbed, J. Gasnault, L. Nevot et al., CD4 CD25 regulatory T cells are not impaired in patients with primary Sj??gren's syndrome, Journal of Autoimmunity, vol.24, issue.3, pp.235-242, 2005.
DOI : 10.1016/j.jaut.2005.01.015

X. Li, X. Li, L. Qian, G. Wang, H. Zhang et al., T regulatory cells are markedly Clinic Rev Allerg Immunol Author's personal copy diminished in diseased salivary glands of patients with primary Sjogren's syndrome, J Rheumatol, vol.34, issue.12, pp.2438-2445, 2007.

M. Liu, L. Lin, C. Weng, and M. Weng, Decreased CD4+CD25+bright T cells in peripheral blood of patients with primary Sj??gren's syndrome, Lupus, vol.24, issue.1, pp.34-39, 2008.
DOI : 10.1177/0961203307085248

P. Szodoray, G. Papp, I. Horvath, S. Barath, S. Sipka et al., Cells with regulatory function of the innate and adaptive immune system in primary Sj??gren's syndrome, Clinical & Experimental Immunology, vol.13, issue.3, pp.343-349, 2009.
DOI : 10.1111/j.1365-2249.2009.03966.x

X. Li, Z. Wu, J. Ding, Z. Zheng, X. Li et al., Role of the frequency of blood CD4+ CXCR5+ CCR6+ T cells in autoimmunity in patients with Sj??gren???s syndrome, Biochemical and Biophysical Research Communications, vol.422, issue.2, pp.238-244, 2012.
DOI : 10.1016/j.bbrc.2012.04.133

S. Rusakiewicz, G. Nocturne, T. Lazure, M. Semeraro, C. Flament et al., NCR3/NKp30 Contributes to Pathogenesis in Primary Sjogren's Syndrome, Science Translational Medicine, vol.110, issue.1, 0195.
DOI : 10.1034/j.1600-0722.2002.00152.x

A. Alunno, O. Bistoni, E. Bartoloni, S. Caterbi, B. Bigerna et al., T cells are expanded in the peripheral blood, infiltrate salivary glands and are resistant to corticosteroids in patients with primary Sj??gren's syndrome, Annals of the Rheumatic Diseases, vol.72, issue.2, pp.286-292, 2013.
DOI : 10.1136/annrheumdis-2012-201511

K. Szabo, G. Papp, A. Szanto, T. Tarr, and M. Zeher, A comprehensive investigation on the distribution of circulating follicular T helper cells and B cell subsets in primary Sj??gren's syndrome and systemic lupus erythematosus, Clinical & Experimental Immunology, vol.26, issue.1, pp.76-89, 2016.
DOI : 10.1111/cei.12703

M. Mingueneau, S. Boudaoud, S. Haskett, T. Reynolds, G. Nocturne et al., Cytometry by time-of-flight immunophenotyping identifies a blood Sj??gren's signature correlating with disease activity and glandular inflammation, Journal of Allergy and Clinical Immunology, vol.137, issue.6, pp.1809-1821, 2016.
DOI : 10.1016/j.jaci.2016.01.024

N. Singh and P. Cohen, The T cell in Sjogren's syndrome: Force majeure, not spectateur, Journal of Autoimmunity, vol.39, issue.3, pp.229-233, 2012.
DOI : 10.1016/j.jaut.2012.05.019

A. Bakhshi, N. Miyasaka, P. Kavathas, T. Daniels, C. Strand et al., Lymphocyte subsets in Sjogren's syndrome: a quantitative analysis using monoclonal antibodies and the fluorescence-activated cell sorter, J Clin Lab Immunol, vol.10, issue.2, pp.63-69, 1983.

K. Aziz, P. Mccluskey, and D. Wakefield, Phenotypic and functional abnormalities in the peripheral blood T-cells of patients with primary Sjogren's syndrome, Cytometry, vol.13, issue.1, pp.35-41, 1994.
DOI : 10.1002/cyto.990180108

G. Ferraccioli, E. Tonutti, L. Casatta, I. Pegoraro, D. Vita et al., CD4 cytopenia and occasional expansion of CD4+CD8+lymphocytes in Sjogren's syndrome, Clin Exp Rheumatol, vol.14, issue.2, pp.125-130, 1996.

T. Mandl, A. Bredberg, L. Jacobsson, R. Manthorpe, and G. Henriksson, CD4+ T-lymphocytopenia?a frequent finding in anti-SSA antibody seropositive patients with primary Sjogren's syndrome, J Rheumatol, vol.31, issue.4, pp.726-728, 2004.

Y. Ichikawa, H. Shimizu, M. Yoshida, and S. Arimori, Activation of t cell subsets in the peripheral blood of patients with Sj??gren's syndrome multicolor flow cytometric analysis, Arthritis & Rheumatism, vol.10, issue.11, pp.1674-1681, 1990.
DOI : 10.1002/art.1780331110

M. Moriyama, A. Tanaka, T. Maehara, S. Furukawa, H. Nakashima et al., T helper subsets in Sj??gren's syndrome and IgG4-related dacryoadenitis and sialoadenitis: A critical review, Journal of Autoimmunity, vol.51, pp.81-88, 2014.
DOI : 10.1016/j.jaut.2013.07.007

D. Mitsias, A. Tzioufas, C. Veiopoulou, E. Zintzaras, I. Tassios et al., The Th1/Th2 cytokine balance changes with the progress of the immunopathological lesion of Sjogren???s syndrome, Clinical & Experimental Immunology, vol.72, issue.(Suppl. 1), pp.562-568, 2002.
DOI : 10.1046/j.1365-2249.2002.01869.x

A. Alunno, F. Carubbi, E. Bartoloni, O. Bistoni, S. Caterbi et al., Unmasking the pathogenic role of IL-17 axis in primary Sj??gren's syndrome: A new era for therapeutic targeting?, Autoimmunity Reviews, vol.13, issue.12, pp.1167-1173, 2014.
DOI : 10.1016/j.autrev.2014.08.022

A. Alunno, F. Carubbi, S. Caterbi, O. Bistoni, E. Bartoloni et al., The role of T helper 17 cell subsets in Sj??gren's syndrome: similarities and differences between mouse model and humans, Annals of the Rheumatic Diseases, vol.284, issue.7, pp.42-52, 2014.
DOI : 10.1136/annrheumdis-2014-205517

X. Lin, K. Rui, J. Deng, J. Tian, X. Wang et al., Th17 cells play a critical role in the development of experimental Sj??gren's syndrome, Annals of the Rheumatic Diseases, vol.64, issue.6, pp.1302-1310, 2015.
DOI : 10.1136/annrheumdis-2013-204584

A. Alunno, F. Carubbi, O. Bistoni, S. Caterbi, E. Bartoloni et al., CD4???CD8??? T-cells in primary Sj??gren's syndrome: Association with the extent of glandular involvement, Journal of Autoimmunity, vol.51, pp.38-43, 2014.
DOI : 10.1016/j.jaut.2014.01.030

S. Crotty, ), Annual Review of Immunology, vol.29, issue.1, pp.621-663, 2011.
DOI : 10.1146/annurev-immunol-031210-101400

M. Akiyama, K. Suzuki, K. Yamaoka, H. Yasuoka, M. Takeshita et al., Brief Report: Number of Circulating Follicular Helper 2 T Cells Correlates With IgG4 and Interleukin-4 Levels and Plasmablast Numbers in IgG4-Related Disease, Arthritis & Rheumatology, vol.58, issue.Suppl 1, pp.2476-2481, 2015.
DOI : 10.1002/art.39209

Y. Gong, J. Nititham, K. Taylor, C. Miceli-richard, C. Sordet et al., Differentiation of follicular helper T cells by salivary gland epithelial cells in primary Sj??gren's syndrome, Journal of Autoimmunity, vol.51, pp.57-66, 2014.
DOI : 10.1016/j.jaut.2013.11.003

S. Pesce, L. Moretta, A. Moretta, and E. Marcenaro, Human NK Cell Subsets Redistribution in Pathological Conditions: A Role for CCR7 Receptor, Frontiers in Immunology, vol.101, issue.5, 2016.
DOI : 10.3324/haematol.2015.135301

J. Bohnhorst, M. Bjorgan, J. Thoen, J. Natvig, and K. Thompson, Bm1-Bm5 Classification of Peripheral Blood B Cells Reveals Circulating Germinal Center Founder Cells in Healthy Individuals and Disturbance in the B Cell Subpopulations in Patients with Primary Sjogren's Syndrome, The Journal of Immunology, vol.167, issue.7, pp.3610-3618, 2001.
DOI : 10.4049/jimmunol.167.7.3610

A. Hansen, M. Odendahl, K. Reiter, A. Jacobi, E. Feist et al., Diminished peripheral blood memory B cells and accumulation of memory B cells in the salivary glands of patients with Sj??gren's syndrome, Arthritis & Rheumatism, vol.42, issue.Suppl, pp.2160-2171, 2002.
DOI : 10.1002/art.10445

A. Binard, L. Pottier, L. Devauchelle-pensec, V. Saraux, A. Youinou et al., Is the blood B-cell subset profile diagnostic for Sjogren syndrome?, Annals of the Rheumatic Diseases, vol.68, issue.9, pp.1447-1452, 2008.
DOI : 10.1136/ard.2008.096172

URL : https://hal.archives-ouvertes.fr/hal-01103762

D. Cornec, A. Saraux, J. Pers, S. Jousse-joulin, T. Marhadour et al., Clinic Rev Allerg Immunol Author's personal copy

V. Devauchelle-pensec, Diagnostic accuracy of blood Bcell subset profiling and autoimmunity markers in Sjogren's syndrome, Arthritis Res Ther, vol.16, issue.1, pp.10-1186, 2014.

W. Lin, J. L. Chen, H. Wu, Q. Fei, Y. Zheng et al., B cell subsets and dysfunction of regulatory B cells in IgG4-related diseases and primary Sjogren's syndrome: the similarities and differences Primary Sjogren's syndrome is characterized by distinct phenotypic and transcriptional profiles of IgD+ unswitched memory B cells, Arthritis Res Ther Arthritis & Rheumatology, vol.16, issue.669, pp.2558-2569, 2014.

Q. Simon, J. Pers, D. Cornec, L. Pottier, L. Mageed et al., In-depth characterization of CD24 high CD38 high transitional human B??cells reveals different regulatory profiles, Journal of Allergy and Clinical Immunology, vol.137, issue.5, pp.1577-1584, 2016.
DOI : 10.1016/j.jaci.2015.09.014

URL : https://hal.archives-ouvertes.fr/hal-01353377

D. Cornec, V. Devauchelle-pensec, G. Tobon, J. Pers, S. Jousse-joulin et al., B cells in Sj??gren's syndrome: From pathophysiology to diagnosis and treatment, Journal of Autoimmunity, vol.39, issue.3, pp.161-167, 2012.
DOI : 10.1016/j.jaut.2012.05.014

P. Youinou and J. Pers, Primary Sj??gren's syndrome at a glance today, Joint Bone Spine, vol.82, issue.2, pp.75-76, 2015.
DOI : 10.1016/j.jbspin.2014.10.018

V. Pascual, Y. Liu, A. Magalski, O. De-bouteiller, J. Banchereau et al., Analysis of somatic mutation in five B cell subsets of human tonsil, Journal of Experimental Medicine, vol.180, issue.1, pp.329-339, 1994.
DOI : 10.1084/jem.180.1.329

L. Boiardi, C. Salvarani, P. Macchioni, C. Maldini, M. Mancini et al., CD8 LYMPHOCYTE SUBSETS IN ACTIVE POLYMYALGIA RHEUMATICA: COMPARISON WITH ELDERLY-ONSET AND ADULT RHEUMATOID ARTHRITIS AND INFLUENCE OF PREDNISONE THERAPY, Rheumatology, vol.35, issue.7, pp.642-648, 1996.
DOI : 10.1093/rheumatology/35.7.642

A. Skapenko, J. Wendler, P. Lipsky, J. Kalden, and H. Schulze-koops, Altered memory T cell differentiation in patients with early rheumatoid arthritis, J Immunol, vol.163, issue.1, pp.491-499, 1999.

C. Lawson, A. Brown, V. Bejarano, S. Douglas, C. Burgoyne et al., Early rheumatoid arthritis is associated with a deficit in the CD4+CD25high regulatory T cell population in peripheral blood, Rheumatology, vol.45, issue.10, pp.1210-1217, 2006.
DOI : 10.1093/rheumatology/kel089

H. Kokkonen, I. Soderstrom, J. Rocklov, G. Hallmans, K. Lejon et al., Up-regulation of cytokines and chemokines predates the onset of rheumatoid arthritis, Arthritis & Rheumatism, vol.62, issue.2, pp.383-391, 2010.
DOI : 10.1002/art.27186

S. Kawashiri, A. Kawakami, A. Okada, T. Koga, M. Tamai et al., CD4+CD25highCD127low/- Treg Cell Frequency from Peripheral Blood Correlates with Disease Activity in Patients with Rheumatoid Arthritis, The Journal of Rheumatology, vol.38, issue.12, pp.2517-2521, 2011.
DOI : 10.3899/jrheum.110283

L. Ma, B. Liu, Z. Jiang, and Y. Jiang, Reduced numbers of regulatory B cells are negatively correlated with disease activity in patients with new-onset rheumatoid arthritis, Clinical Rheumatology, vol.6, issue.5, pp.187-195, 2014.
DOI : 10.1007/s10067-013-2359-3

J. Lubbers, M. Van-beers-tas, S. Vosslamber, S. Turk, S. De-ridder et al., Changes in peripheral blood lymphocyte subsets during arthritis development in arthralgia patients, Arthritis Research & Therapy, vol.8, issue.1, pp.205-215, 2016.
DOI : 10.1186/s13075-016-1102-2

P. Chalan, J. Bijzet, B. Kroesen, A. Boots, and E. Brouwer, Altered Natural Killer Cell Subsets in Seropositive Arthralgia and Early Rheumatoid Arthritis Are Associated with Autoantibody Status, The Journal of Rheumatology, vol.43, issue.6, pp.1008-1016, 2016.
DOI : 10.3899/jrheum.150644

D. Pensec, V. Saraux, A. Berthelot, J. Alapetite, S. Chales et al., Ability of hand radiographs to predict a further diagnosis of rheumatoid arthritis in patients with early arthritis, J Rheumatol, vol.28, issue.12, pp.2603-2607, 2001.

Z. Yunt and J. Solomon, Lung Disease in Rheumatoid Arthritis, Rheumatic Disease Clinics of North America, vol.41, issue.2, pp.225-236, 2015.
DOI : 10.1016/j.rdc.2014.12.004

G. Carvajal-alegria, A. Uguen, S. Genestet, P. Marcorelles, A. Saraux et al., New onset of rheumatoid vasculitis during abatacept therapy and subsequent improvement after rituximab, Joint Bone Spine, vol.83, issue.5, pp.605-606, 2016.
DOI : 10.1016/j.jbspin.2015.08.016

URL : https://hal.archives-ouvertes.fr/hal-01307022

G. Goddard, A. Soriano, B. Gilburd, M. Lidar, S. Kivity et al., A novel bedside test for ACPA: the CCPoint test is moving the laboratory to the rheumatologist???s office, Immunologic Research, vol.3, issue.7, pp.10-1007, 2016.
DOI : 10.4155/bio.11.200

D. Winter, L. Hansen, W. Van-steenbergen, H. Geusens, P. Lenaerts et al., Autoantibodies to two novel peptides in seronegative and early rheumatoid arthritis, Rheumatology, vol.55, issue.8, pp.1431-1436, 2016.
DOI : 10.1093/rheumatology/kew198

J. Mccomish, J. Mundy, T. Sullivan, S. Proudman, and P. Hissaria, Changes in peripheral blood B cell subsets at diagnosis and after treatment with disease-modifying anti-rheumatic drugs in patients with rheumatoid arthritis: correlation with clinical and laboratory parameters, International Journal of Rheumatic Diseases, vol.31, issue.Suppl 1, pp.421-432, 2015.
DOI : 10.1111/1756-185X.12325

C. Daien, S. Gailhac, T. Mura, R. Audo, B. Combe et al., Regulatory B10 Cells Are Decreased in Patients With Rheumatoid Arthritis and Are Inversely Correlated With Disease Activity, Arthritis & Rheumatology, vol.33, issue.Suppl 39, pp.2037-2046, 2014.
DOI : 10.1002/art.38666

R. Jimeno, J. Leceta, M. Garin, A. Ortiz, M. Mellado et al., Th17 polarization of memory Th cells in early arthritis: the vasoactive intestinal peptide effect, Journal of Leukocyte Biology, vol.98, issue.2, pp.257-2693, 2015.
DOI : 10.1189/jlb.3A0714-327R

G. Yaniv, G. Twig, D. Shor, A. Furer, Y. Sherer et al., A volcanic explosion of autoantibodies in systemic lupus erythematosus: A diversity of 180 different antibodies found in SLE patients, Autoimmunity Reviews, vol.14, issue.1, pp.75-79, 2015.
DOI : 10.1016/j.autrev.2014.10.003

M. Petri, A. Orbai, G. Alarcon, C. Gordon, J. Merrill et al., Derivation and validation of the Systemic Lupus International Collaborating Clinics classification criteria for systemic lupus erythematosus, Arthritis & Rheumatism, vol.65, issue.Suppl, pp.2677-2686, 2012.
DOI : 10.1002/art.34473

L. Amezcua-guerra, V. Higuera-ortiz, U. Arteaga-garcia, S. Gallegos-nava, and C. Hubbe-tena, Performance of the 2012 Systemic Lupus International Collaborating Clinics and the 1997 American College of Rheumatology classification Clinic Rev Allerg Immunol Author's personal copy criteria for systemic lupus erythematosus in a real-life scenario, 2015.

L. Ines, C. Silva, M. Galindo, F. Lopez-longo, G. Terroso et al., Classification of Systemic Lupus Erythematosus: Systemic Lupus International Collaborating Clinics Versus American College of Rheumatology Criteria. A Comparative Study of 2,055 Patients From a Real-Life, International Systemic Lupus Erythematosus Cohort, Arthritis Care & Research, vol.64, issue.8, pp.1180-1185
DOI : 10.1002/acr.22539

C. Yu, M. Gershwin, and C. Chang, Diagnostic criteria for systemic lupus erythematosus: A critical review, Journal of Autoimmunity, vol.48, issue.49, 2014.
DOI : 10.1016/j.jaut.2014.01.004

M. Scheinberg and E. Cathcart, B cell and T cell lymphopenia in systemic lupus erythematosus, Cellular Immunology, vol.12, issue.2, pp.309-314, 1974.
DOI : 10.1016/0008-8749(74)90083-5

F. Erkeller-yusel, F. Hulstaart, I. Hannet, D. Isenberg, and P. Lydyard, Lymphocyte Subsets in a Large Cohort of Patients with Systemic Lupus Erythematosus, Lupus, vol.94, issue.4, pp.227-231, 1993.
DOI : 10.1177/096120339300200404

F. Erkeller-yuksel, P. Lydyard, and D. Isenberg, Lack of NK cells in lupus patients with renal involvement, Lupus, vol.145, issue.9, pp.708-712, 1997.
DOI : 10.1177/096120339700600905

S. Mortezagholi, Z. Babaloo, P. Rahimzadeh, M. Ghaedi, H. Namdari et al., Evaluation of PBMC distribution and TLR9 expression in patients with systemic lupus erythematosus, Iranian journal of allergy, asthma, and immunology, vol.15, issue.3, pp.229-236, 2016.

C. Wouters, C. Diegenant, J. Ceuppens, H. Degreef, and E. Stevens, The circulating lymphocyte profiles in patients with discoid lupus erythematosus and systemic lupus erythematosus suggest a pathogenetic relationship, British Journal of Dermatology, vol.6, issue.4, pp.693-700, 2004.
DOI : 10.1016/S0145-2126(97)00152-5

M. Odendahl, A. Jacobi, A. Hansen, E. Feist, F. Hiepe et al., Disturbed Peripheral B Lymphocyte Homeostasis in Systemic Lupus Erythematosus, The Journal of Immunology, vol.165, issue.10, pp.5970-5979, 2000.
DOI : 10.4049/jimmunol.165.10.5970

C. Wehr, H. Eibel, M. Masilamani, H. Illges, M. Schlesier et al., A new CD21low B cell population in the peripheral blood of patients with SLE, Clinical Immunology, vol.113, issue.2, pp.161-171, 2004.
DOI : 10.1016/j.clim.2004.05.010

C. Wei, J. Anolik, A. Cappione, B. Zheng, A. Pugh-bernard et al., A New Population of Cells Lacking Expression of CD27 Represents a Notable Component of the B Cell Memory Compartment in Systemic Lupus Erythematosus, The Journal of Immunology, vol.178, issue.10, pp.6624-6633, 2007.
DOI : 10.4049/jimmunol.178.10.6624

B. Rodriguez-bayona, A. Ramos-amaya, J. Perez-venegas, C. Rodriguez, and J. Brieva, Decreased frequency and activated phenotype of blood CD27 IgD IgM B lymphocytes is a permanent abnormality in systemic lupus erythematosus patients, Arthritis Research & Therapy, vol.12, issue.3, pp.10-3042, 1186.
DOI : 10.1186/ar3042

S. Malkiel, V. Jeganathan, S. Wolfson, M. Orduno, N. Marasco et al., Checkpoints for Autoreactive B Cells in the Peripheral Blood of Lupus Patients Assessed by Flow Cytometry, Arthritis & Rheumatology, vol.211, issue.9, pp.2210-2220, 2016.
DOI : 10.1002/art.39710

Y. Renaudineau, S. Croquefer, S. Jousse, E. Renaudineau, V. Devauchelle et al., Association of ??-actinin???binding anti???double-stranded DNA antibodies with lupus nephritis, Arthritis & Rheumatism, vol.100, issue.8, pp.2523-2532, 2006.
DOI : 10.1002/art.22015

Y. Renaudineau, J. Pers, B. Bendaoud, C. Jamin, and P. Youinou, Dysfunctional B cells in systemic lupus erythematosus, Autoimmunity Reviews, vol.3, issue.7-8, pp.516-523, 2004.
DOI : 10.1016/j.autrev.2004.07.035

W. Brooks and Y. Renaudineau, Epigenetics and autoimmune diseases: the X chromosome-nucleolus nexus, Frontiers in Genetics, vol.63, issue.e67689, 2015.
DOI : 10.1002/art.30196

URL : https://hal.archives-ouvertes.fr/hal-01132409

T. Fali, L. Dantec, C. Thabet, Y. Jousse, S. Hanrotel et al., DNA methylation modulates HRES1/p28 expression in B cells from patients with Lupus, Autoimmunity, vol.103, issue.4, pp.265-271, 2014.
DOI : 10.4161/epi.20523

URL : https://hal.archives-ouvertes.fr/hal-00996373

S. Garaud, L. Dantec, C. Jousse-joulin, S. Hanrotel-saliou, C. Saraux et al., IL-6 Modulates CD5 Expression in B Cells from Patients with Lupus by Regulating DNA Methylation, The Journal of Immunology, vol.182, issue.9, pp.5623-5632, 2009.
DOI : 10.4049/jimmunol.0802412

T. Taher, H. Muhammad, A. Rahim, F. Flores-borja, Y. Renaudineau et al., Aberrant B-lymphocyte responses in lupus: inherent or induced and potential therapeutic targets, European Journal of Clinical Investigation, vol.11, issue.Suppl 1, pp.866-880, 2013.
DOI : 10.1111/eci.12111

URL : https://hal.archives-ouvertes.fr/hal-01068367

E. Lazaro, M. Scherlinger, M. Truchetet, L. Chiche, T. Schaeverbeke et al., Biotherapies in systemic lupus erythematosus: New targets, Joint Bone Spine, vol.84, issue.3, 2016.
DOI : 10.1016/j.jbspin.2016.07.004

L. Chiche, N. Jourde, G. Thomas, N. Bardin, C. Bornet et al., New treatment options for lupus – a focus on belimumab, Therapeutics and Clinical Risk Management, vol.8, pp.33-43, 2012.
DOI : 10.2147/TCRM.S19819

Z. Vadasz, R. Peri, N. Eiza, G. Slobodin, A. Balbir-gurman et al., The expansion of CD25 high IL-10 high FoxP3 high B regulatory cells is in association with SLE disease activity, J Immunol Res, pp.254245-254255, 2015.

S. Lin, K. Chen, C. Lin, C. Kuo, Q. Ling et al., The quantitative analysis of peripheral blood FOXP3-expressing T cells in systemic lupus erythematosus and rheumatoid arthritis patients, European Journal of Clinical Investigation, vol.176, issue.12, pp.987-996, 2007.
DOI : 10.1172/JCI200320274

B. Zhang, X. Zhang, F. Tang, L. Zhu, Y. Liu et al., Clinical significance of increased CD4+CD25-Foxp3+ T cells in patients with new-onset systemic lupus erythematosus, Annals of the Rheumatic Diseases, vol.67, issue.7, pp.1037-1040083543, 2007.
DOI : 10.1136/ard.2007.083543

J. Suen, H. Li, Y. Jong, B. Chiang, and J. Yen, regulatory T-cell subpopulations in systemic lupus erythematosus, Immunology, vol.148, issue.2, pp.196-205, 2009.
DOI : 10.1111/j.1365-2567.2008.02937.x

Y. Park, S. Kee, Y. Cho, E. Lee, H. Lee et al., Impaired differentiation and cytotoxicity of natural killer cells in systemic lupus erythematosus, Arthritis & Rheumatism, vol.103, issue.6, pp.1753-1763, 2009.
DOI : 10.1002/art.24556

W. Li, H. Pan, J. Hu, C. Wang, N. Zhang et al., Assay of T- and NK-cell subsets and the expression of NKG2A and NKG2D in patients with new-onset systemic lupus erythematosus, Clinical Rheumatology, vol.105, issue.426???8, pp.315-323, 2010.
DOI : 10.1007/s10067-009-1322-9

Y. Cho, S. Kee, S. Lee, S. Seo, T. Kim et al., Numerical and functional deficiencies of natural killer T cells in systemic lupus erythematosus: their deficiency related to disease activity, Rheumatology, vol.50, issue.6, pp.1054-1063, 2011.
DOI : 10.1093/rheumatology/keq457

X. Pan, X. Yuan, Y. Zheng, W. Wang, J. Shan et al., Increased CD45RA+ Clinic Rev Allerg Immunol Author's personal copy, 2012.

A. Henriques, L. Teixeira, L. Ines, T. Carvalheiro, A. Goncalves et al., NK cells dysfunction in systemic lupus erythematosus: relation to disease activity, Clinical Rheumatology, vol.32, issue.6, pp.805-813, 2013.
DOI : 10.1007/s10067-013-2176-8

H. Ma, L. Zhao, Z. Jiang, Y. Jiang, L. Feng et al., Dynamic changes in the numbers of different subsets of peripheral blood NK cells in patients with systemic lupus erythematosus following classic therapy, Clinical Rheumatology, vol.236, issue.4804, pp.1603-1610, 2014.
DOI : 10.1007/s10067-014-2712-1

L. Song, X. Wang, X. Wang, D. Li, F. Ding et al., Increased Tim-3 expression on peripheral T lymphocyte subsets and association with higher disease activity in systemic lupus erythematosus, Diagnostic Pathology, vol.183, issue.Suppl 1, p.71, 2015.
DOI : 10.1186/s13000-015-0306-0

J. Chen, M. Wu, J. Wang, and X. Li, Immunoregulation of NKT Cells in Systemic Lupus Erythematosus, Journal of Immunology Research, vol.8, issue.12, pp.206731-206741, 2015.
DOI : 10.1046/j.1365-2567.2000.00001.x

J. Chen, L. Ding, W. Meng, Y. J. Yan, C. Xie et al., Vincristine-cyclophosphamide combination therapy positively affects T-cell subset distribution in systemic lupus erythematosus patients. Medical science monitor : international medical journal of experimental and clinical research, pp.505-510, 2015.

A. Bakke, P. Kirkland, R. Kitridou, F. Quismorio, . Jr et al., T Lymphocyte Subsets in Systemic Lupus Erythematosus, Arthritis & Rheumatism, vol.24, issue.6, pp.745-750, 1983.
DOI : 10.1002/art.1780260607

E. Robak, H. Niewiadomska, T. Robak, J. Bartkowiak, J. Blonski et al., Lymphocytes T???? in clinically normal skin and peripheral blood of patients with systemic lupus erythematosus and their correlation with disease activity, Mediators of Inflammation, vol.10, issue.4, pp.179-1891009629350124724, 1080.
DOI : 10.1080/09629350124724

R. Spada, J. Rojas, and D. Barber, Recent findings on the role of natural killer cells in the pathogenesis of systemic lupus erythematosus, Journal of Leukocyte Biology, vol.98, issue.4, pp.479-487, 2015.
DOI : 10.1189/jlb.4RU0315-081RR

J. Jennette, R. Falk, P. Bacon, N. Basu, M. Cid et al., 2012 Revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides, revised International Chapel Hill Consensus Conference nomenclature of vasculitides, pp.1-11, 2012.
DOI : 10.1002/art.37715

J. Damoiseaux, E. Csernok, N. Rasmussen, F. Moosig, P. Van-paassen et al., Detection of antineutrophil cytoplasmic antibodies (ANCAs): a multicentre European Vasculitis Study Group (EUVAS) evaluation of the value of indirect immunofluorescence (IIF) versus antigen-specific immunoassays, Annals of the Rheumatic Diseases, vol.76, issue.4, pp.10-1136, 2016.
DOI : 10.1136/annrheumdis-2016-209507

V. Cottin, E. Bel, P. Bottero, K. Dalhoff, M. Humbert et al., Revisiting the systemic vasculitis in eosinophilic granulomatosis with polyangiitis (Churg-Strauss), Autoimmunity Reviews, vol.16, issue.1, pp.1-9
DOI : 10.1016/j.autrev.2016.09.018

URL : https://hal.archives-ouvertes.fr/hal-01467544

M. Groh, C. Pagnoux, C. Baldini, E. Bel, P. Bottero et al., Eosinophilic granulomatosis with polyangiitis (Churg???Strauss) (EGPA) Consensus Task Force recommendations for evaluation and management, European Journal of Internal Medicine, vol.26, issue.7, pp.545-553, 2015.
DOI : 10.1016/j.ejim.2015.04.022

A. Greco, D. Virgilio, A. Rizzo, M. Gallo, A. Magliulo et al., Microscopic polyangiitis: Advances in diagnostic and therapeutic approaches, Autoimmunity Reviews, vol.14, issue.9, pp.837-844, 2015.
DOI : 10.1016/j.autrev.2015.05.005

M. Weiner and M. Segelmark, The clinical presentation and therapy of diseases related to anti-neutrophil cytoplasmic antibodies (ANCA), Autoimmunity Reviews, vol.15, issue.10, pp.978-982, 2016.
DOI : 10.1016/j.autrev.2016.07.016

A. Greco, M. Rizzo, D. Virgilio, A. Gallo, A. Fusconi et al., Churg???Strauss syndrome, Autoimmunity Reviews, vol.14, issue.4, pp.341-348, 2015.
DOI : 10.1016/j.autrev.2014.12.004

D. Cornec, C. Gall, E. Fervenza, F. Specks, and U. , ANCA-associated vasculitis ??? clinical utility of using ANCA specificity to classify patients, Nature Reviews Rheumatology, vol.60, issue.4, pp.570-579, 2016.
DOI : 10.1038/nrrheum.2016.123

URL : https://hal.archives-ouvertes.fr/hal-01352043

M. Yates, R. Watts, I. Bajema, M. Cid, B. Crestani et al., EULAR/ERA-EDTA recommendations for the management of ANCA-associated vasculitis, Annals of the Rheumatic Diseases, vol.73, issue.36, pp.1583-1594, 2016.
DOI : 10.1136/annrheumdis-2016-209133

C. Kallenberg, Usefulness of antineutrophil cytoplasmic autoantibodies in diagnosing and managing systemic vasculitis., Current Opinion in Rheumatology, vol.28, issue.1, pp.8-14, 2016.
DOI : 10.1097/BOR.0000000000000233

J. Land, A. Rutgers, and C. Kallenberg, Anti-neutrophil cytoplasmic autoantibody pathogenicity revisited: pathogenic versus non-pathogenic anti-neutrophil cytoplasmic autoantibody, Nephrology Dialysis Transplantation, vol.29, issue.4, 2014.
DOI : 10.1093/ndt/gft416

P. Jarrot and G. Kaplanski, Pathogenesis of ANCA-associated vasculitis: An update, Autoimmunity Reviews, vol.15, issue.7, pp.704-713, 2016.
DOI : 10.1016/j.autrev.2016.03.007

URL : https://hal.archives-ouvertes.fr/hal-01459842

N. Dumoitier, B. Terrier, J. London, S. Lofek, and L. Mouthon, Implication of B lymphocytes in the pathogenesis of ANCA-associated vasculitides, Autoimmunity Reviews, vol.14, issue.11, pp.996-1004, 2015.
DOI : 10.1016/j.autrev.2015.06.008

J. Thiel, U. Salzer, F. Hassler, N. Effelsberg, C. Hentze et al., B cell homeostasis is disturbed by immunosuppressive therapies in patients with ANCA-associated vasculitides, Autoimmunity, vol.115, issue.7, pp.429-438
DOI : 10.1136/ard.2008.096495

M. Schlesier, T. Kaspar, J. Gutfleisch, G. Wolff-vorbeck, and H. Peter, Activated CD4+ and CD8+ T-cell subsets in Wegener's granulomatosis, Rheumatology International, vol.36, issue.5, pp.213-219, 1995.
DOI : 10.1007/BF00262300

E. Popa, C. Stegeman, N. Bos, C. Kallenberg, and J. Tervaert, Differential B- and T-cell activation in Wegener???s granulomatosis, Journal of Allergy and Clinical Immunology, vol.103, issue.5, pp.885-894, 1999.
DOI : 10.1016/S0091-6749(99)70434-3

F. Hua, B. Wilde, S. Dolff, and O. Witzke, T-Lymphocytes and Disease Mechanisms in Wegener???s Granulomatosis, Kidney and Blood Pressure Research, vol.32, issue.6, pp.389-398, 2009.
DOI : 10.1159/000256409

A. Berden, C. Kallenberg, C. Savage, B. Yard, W. Abdulahad et al., Cellular immunity in Wegener's granulomatosis: Characterizing T lymphocytes, Arthritis & Rheumatism, vol.28, issue.6, pp.1578-1587, 2009.
DOI : 10.1002/art.24576

W. Abdulahad, C. Stegeman, Y. Van-der-geld, B. Doornbos-van-der-meer, P. Limburg et al., Functional defect of circulating regulatory CD4+ T cells in patients with Wegener's granulomatosis in remission, Arthritis & Rheumatism, vol.125, issue.6, pp.2080-2091, 2007.
DOI : 10.1002/art.22692

S. Marinaki, A. Kalsch, P. Grimminger, A. Breedijk, R. Birck et al., Persistent T-cell activation and clinical correlations in patients with ANCA-associated systemic vasculitis. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association, European Renal Association, vol.21, issue.7, pp.1825-1832, 2006.

W. Abdulahad, Y. Van-der-geld, C. Stegeman, and C. Kallenberg, Persistent expansion of CD4+ effector memory T cells in Wegener's granulomatosis, Kidney International, vol.70, issue.5, pp.938-947, 2006.
DOI : 10.1038/sj.ki.5001670

B. Wilde, S. Dolff, X. Cai, C. Specker, J. Becker et al., CD4+CD25+ T-cell populations expressing CD134 and GITR are associated with disease activity in patients with Wegener's granulomatosis. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association, European Renal Association, vol.24, issue.1, pp.161-171, 2009.

P. Lamprecht, A. Mueller, and W. Gross, CD28- T cells display features of effector memory T cells in Wegener's granulomatosis, Kidney International, vol.65, issue.3, 2004.
DOI : 10.1111/j.1523-1755.2004.501_5.x

W. Abdulahad, C. Stegeman, P. Limburg, and C. Kallenberg, Skewed distribution of Th17 lymphocytes in patients with Wegener's granulomatosis in remission, Arthritis & Rheumatism, vol.153, issue.7, pp.2196-2205, 2008.
DOI : 10.1002/art.23557

M. De-menthon, M. Lambert, E. Guiard, S. Tognarelli, B. Bienvenu et al., Excessive interleukin-15 transpresentation endows NKG2D+CD4+ T cells with innate-like capacity to lyse vascular endothelium in granulomatosis with polyangiitis (Wegener's), Arthritis & Rheumatism, vol.114, issue.7, pp.2116-2126, 2011.
DOI : 10.1002/art.30355

C. Braudeau, K. Amouriaux, A. Neel, G. Herbreteau, N. Salabert et al., Persistent deficiency of circulating mucosal-associated invariant T (MAIT) cells in ANCA-associated vasculitis, Journal of Autoimmunity, vol.70, pp.73-79, 2016.
DOI : 10.1016/j.jaut.2016.03.015

W. Merkt, P. Sturm, F. Lasitschka, T. Tretter, C. Watzl et al., Peripheral blood natural killer cell percentages in granulomatosis with polyangiitis correlate with disease inactivity and stage, Arthritis Research & Therapy, vol.16, issue.10, p.337, 2015.
DOI : 10.1186/s13075-015-0851-7

W. Merkt, C. M. Blank, N. Hundemer, M. Cerwenka, A. Lorenz et al., Active but not inactive granulomatosis with polyangiitis is associated with decreased and phenotypically and functionally altered CD56dim natural killer cells, Arthritis Research & Therapy, vol.63, issue.6, pp.204-214, 2016.
DOI : 10.1186/s13075-016-1098-7

URL : http://doi.org/10.1186/s13075-016-1098-7

F. Moosig, E. Csernok, G. Wang, and W. Gross, Costimulatory molecules in Wegener's granulomatosis (WG): lack of expression of CD28 and preferential up-regulation of its ligands B7-1 (CD80) and B7-2 (CD86) on T cells, Clinical and Experimental Immunology, vol.24, issue.1, pp.113-118, 1998.
DOI : 10.1084/jem.177.3.845

J. Land, W. Abdulahad, J. Sanders, C. Stegeman, P. Heeringa et al., Regulatory and effector B cell cytokine production in patients with relapsing granulomatosis with polyangiitis, Arthritis Research & Therapy, vol.25, issue.65, pp.84-94, 2016.
DOI : 10.1186/s13075-016-0978-1

B. Wilde, M. Thewissen, J. Damoiseaux, S. Knippenberg, M. Hilhorst et al., Regulatory B cells in ANCA-associated vasculitis, Annals of the Rheumatic Diseases, vol.180, issue.8, pp.1416-1419, 2013.
DOI : 10.1136/annrheumdis-2012-202986

N. Lepse, W. Abdulahad, A. Rutgers, C. Kallenberg, C. Stegeman et al., Altered B cell balance, but unaffected B cell capacity to limit monocyte activation in anti-neutrophil cytoplasmic antibody-associated vasculitis in remission, Rheumatology, vol.53, issue.9, pp.1683-1692, 2014.
DOI : 10.1093/rheumatology/keu149

S. Todd, R. Pepper, J. Draibe, A. Tanna, C. Pusey et al., Regulatory B cells are numerically but not functionally deficient in anti-neutrophil cytoplasm antibody-associated vasculitis, Rheumatology, vol.53, issue.9, pp.1693-1703, 2014.
DOI : 10.1093/rheumatology/keu136

S. Garaud, T. Taher, M. Debant, M. Burgos, S. Melayah et al., CD5 expression promotes IL-10 production through activation of the MAPK/Erk pathway and upregulation of TRPC1 channels in B lymphocytes, Cellular & Molecular Immunology, vol.87, 2016.
DOI : 10.1038/nrrheum.2013.7

URL : https://hal.archives-ouvertes.fr/hal-01353347

D. Bunch, J. Mcgregor, N. Khandoobhai, L. Aybar, M. Burkart et al., Decreased CD5+ B Cells in Active ANCA Vasculitis and Relapse after Rituximab, Clinical Journal of the American Society of Nephrology, vol.8, issue.3, pp.382-391, 2013.
DOI : 10.2215/CJN.03950412

L. Aybar, J. Mcgregor, S. Hogan, Y. Hu, C. Mendoza et al., regulatory B cells in active anti-neutrophil cytoplasmic autoantibody-associated vasculitis permit increased circulating autoantibodies, Clinical & Experimental Immunology, vol.182, issue.Suppl. 1, pp.178-188, 2015.
DOI : 10.1111/cei.12483

D. Bunch, C. Mendoza, L. Aybar, E. Kotzen, K. Colby et al., B cells portend a shorter time to relapse after B cell depletion in patients with ANCA-associated vasculitis, Annals of the Rheumatic Diseases, vol.50, issue.9, pp.1784-1786, 2015.
DOI : 10.1136/annrheumdis-2014-206756

S. Unizony, N. Lim, D. Phippard, V. Carey, E. Miloslavsky et al., Peripheral CD5+ B Cells in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis, Arthritis & Rheumatology, vol.53, issue.Suppl 1, pp.535-544, 2015.
DOI : 10.1002/art.38916

C. Therene, E. Brenaut, H. Sonbol, E. Pasquier, A. Saraux et al., Itch and systemic sclerosis: frequency, clinical characteristics and consequences, Br J Dermatol, 2016.

A. Lescoat, G. Coiffier, A. Rouil, C. Droitcourt, C. Cazalets et al., Vascular evaluation of the hand by power Doppler ultrasonography provides new predictive markers of ischemic digital ulcers in systemic sclerosis, Arthritis Care Res, 2016.

A. Morales-cardenas, C. Perez-madrid, L. Arias, P. Ojeda, M. Mahecha et al., Pulmonary involvement in systemic sclerosis, Autoimmunity Reviews, vol.15, issue.11, pp.1094-1108, 2016.
DOI : 10.1016/j.autrev.2016.07.025

Y. Braun-moscovici, R. Brun, and M. Braun, Systemic Sclerosis and the Gastrointestinal Tract???Clinical Approach, Rambam Maimonides Medical Journal, vol.7, issue.4, 2016.
DOI : 10.5041/RMMJ.10258

Y. Renaudineau, E. Grunebaum, I. Krause, S. Praprotnik, R. Revelen et al., Anti-Endothelial Cell Antibodies (AECA) in Systemic Sclerosis - Increased Sensitivity using Different Endothelial Cell Substrates and Association with Other Autoantibodies, Autoimmunity, vol.175, issue.3, pp.171-179, 2001.
DOI : 10.1002/path.1711660307

E. Dalkilic, K. Dilek, M. Gullulu, M. Yavuz, Y. Karakoc et al., Lymphocyte phenotypes in systemic sclerosis, Annals of the Rheumatic Diseases, vol.58, issue.11, pp.719-720, 1999.
DOI : 10.1136/ard.58.11.719a

T. Gambichler, C. Tigges, B. Burkert, S. Hoxtermann, P. Altmeyer et al., Absolute count of T and B lymphocyte subsets is decreased in systemic sclerosis, European Journal of Medical Research, vol.15, issue.1, pp.44-46, 2010.
DOI : 10.1186/2047-783X-15-1-44

L. Ercole, M. Malvezzi, A. Boaretti, S. Utiyama, and A. Rachid, Analysis of lymphocyte subpopulations in systemic sclerosis, J Investig Allergol Clin Immunol, vol.13, issue.2, pp.87-93, 2003.

F. Ingegnoli, D. Trabattoni, M. Saresella, F. Fantini, and M. Clerici, Distinct immune profiles characterize patients with diffuse or limited systemic sclerosis, Clinical Immunology, vol.108, issue.1, pp.21-28, 2003.
DOI : 10.1016/S1521-6616(03)00062-7

C. Artlett, L. Cox, R. Ramos, T. Dennis, R. Fortunato et al., Increased Microchimeric CD4+ T Lymphocytes in Peripheral Blood from Women with Systemic Sclerosis, Clinical Immunology, vol.103, issue.3, pp.303-308, 2002.
DOI : 10.1006/clim.2002.5222

D. Fenoglio, F. Battaglia, A. Parodi, S. Stringara, S. Negrini et al., Alteration of Th17 and Treg cell subpopulations co-exist in patients affected with systemic sclerosis, Clinical Immunology, vol.139, issue.3, pp.249-257, 2011.
DOI : 10.1016/j.clim.2011.01.013

G. Papp, I. Horvath, S. Barath, E. Gyimesi, S. Sipka et al., Altered T-cell and regulatory cell repertoire in patients with diffuse cutaneous systemic sclerosis, Scandinavian Journal of Rheumatology, vol.34, issue.3, pp.205-210, 2010.
DOI : 10.1002/art.1780350111

A. Giovannetti, E. Rosato, C. Renzi, A. Maselli, L. Gambardella et al., Analyses of T cell phenotype and function reveal an altered T cell homeostasis in systemic sclerosis, Clinical Immunology, vol.137, issue.1, pp.122-133, 2010.
DOI : 10.1016/j.clim.2010.06.004

N. Brembilla and C. Chizzolini, T cell abnormalities in systemic sclerosis with a focus on Th17 cells, Eur Cytokine Netw, vol.23, issue.4, pp.128-139, 2012.

T. Rodriguez-reyna, J. Furuzawa-carballeda, J. Cabiedes, L. Fajardo-hermosillo, C. Martinez-reyes et al., Th17 peripheral cells are increased in diffuse cutaneous systemic sclerosis compared with limited illness: a cross-sectional study, Rheumatology International, vol.17, issue.Suppl.1, pp.2653-2660, 2012.
DOI : 10.1007/s00296-011-2056-y

X. Yang, J. Yang, X. Xing, L. Wan, and M. Li, Increased frequency of Th17 cells in systemic sclerosis is related to disease activity and collagen overproduction, Arthritis Research & Therapy, vol.16, issue.1, pp.10-1186, 2014.
DOI : 10.1371/journal.pone.0005981

A. Mavropoulos, T. Simopoulou, A. Varna, C. Liaskos, C. Katsiari et al., Breg Cells Are Numerically Decreased and Functionally Impaired in Patients With Systemic Sclerosis, Arthritis & Rheumatology, vol.12, issue.Suppl 1, pp.494-504, 2016.
DOI : 10.1002/art.39437

I. Lundberg, F. Miller, A. Tjarnlund, and M. Bottai, Diagnosis and classification of idiopathic inflammatory myopathies, Journal of Internal Medicine, vol.15, issue.1, pp.39-51, 2016.
DOI : 10.1002/art.38914

A. Reed and F. Ernste, The inflammatory milieu in idiopathic inflammatory myositis, Current Rheumatology Reports, vol.56, issue.Pt8, pp.295-301, 2009.
DOI : 10.1007/s11926-009-0041-1

M. Viguier, S. Fouere, P. De-la-salmoniere, C. Rabian, C. Lebbe et al., Peripheral Blood Lymphocyte Subset Counts in Patients with Dermatomyositis, Medicine, vol.82, issue.2, pp.82-86, 2003.
DOI : 10.1097/00005792-200303000-00002

D. Wang, X. Lu, N. Zu, B. Lin, L. Wang et al., Clinical significance of peripheral blood lymphocyte subsets in patients with polymyositis and dermatomyositis, Clinical Rheumatology, vol.20, issue.1, pp.1691-1697, 2012.
DOI : 10.1007/s10067-012-2075-4

F. Espinosa-ortega, D. Gomez-martin, S. Anda, K. Romo-tena, J. Villasenor-ovies et al., Quantitative T cell subsets profile in peripheral blood from patients with idiopathic inflammatory myopathies: tilting the balance towards proinflammatory and pro-apoptotic subsets, Clinical & Experimental Immunology, vol.8, issue.3, pp.520-528, 2015.
DOI : 10.1016/j.nmd.2014.06.432

X. Tang, X. Tian, Y. Zhang, W. Wu, J. Tian et al., Correlation between the Frequency of Th17 Cell and the Expression of MicroRNA-206 in Patients with Dermatomyositis, Clinical and Developmental Immunology, vol.2013, pp.345347-345357, 2013.
DOI : 10.1007/s10067-010-1465-8

P. Ungprasert, C. Crowson, V. Chowdhary, F. Ernste, K. Moder et al., Epidemiology of Mixed Connective Tissue Disease, 1985-2014: A Population-Based Study, Arthritis Care & Research, vol.40, issue.12, pp.1843-1848, 1985.
DOI : 10.1002/acr.22872

S. Cappelli, S. Bellando-randone, D. Martinovic, M. Tamas, K. Pasalic et al., ???To Be or Not To Be,??? Ten Years After: Evidence for Mixed Connective Tissue Disease as a Distinct Entity, Seminars in Arthritis and Rheumatism, vol.41, issue.4, pp.589-598, 2012.
DOI : 10.1016/j.semarthrit.2011.07.010

E. Bodolay, M. Aleksza, P. Antal-szalmas, J. Vegh, P. Szodoray et al., Serum cytokine levels and type 1 and type 2 intracellular T cell cytokine profiles in mixed connective tissue disease, J Rheumatol, vol.29, issue.10, pp.2136-2142, 2002.

M. Longhi, Y. Ma, C. Grant, M. Samyn, P. Gordon et al., T-regs in autoimmune hepatitis-systemic lupus erythematosus/mixed connective tissue disease overlap syndrome are functionally defective and display a Th1 cytokine profile, Journal of Autoimmunity, vol.41, pp.146-151, 2013.
DOI : 10.1016/j.jaut.2012.12.003

S. Barath, S. Sipka, M. Aleksza, A. Szegedi, P. Szodoray et al., Regulatory T cells in peripheral blood of patients with mixed connective tissue disease, Scandinavian Journal of Rheumatology, vol.26, issue.4, pp.300-304, 2006.
DOI : 10.1016/0002-9343(72)90064-2

A. Hajas, S. Barath, P. Szodoray, B. Nakken, P. Gogolak et al., FRI0398???Derailed b cell homeostasis in patients with mixed connective tissue disease, Annals of the Rheumatic Diseases, vol.72, issue.Suppl 3, pp.833-841, 2013.
DOI : 10.1136/annrheumdis-2013-eular.1525

W. Robinson and R. Mao, Decade in review???technology: Technological advances transforming rheumatology, Nature Reviews Rheumatology, vol.148, issue.11, pp.626-628, 2015.
DOI : 10.1038/nrrheum.2015.137

C. Jamin, L. Lann, L. Alvarez-errico, D. Barbarroja, N. Cantaert et al., Multi-center harmonization of flow cytometers in the context of the European ???PRECISESADS??? project, Autoimmunity Reviews, vol.15, issue.11, pp.1038-1045, 2016.
DOI : 10.1016/j.autrev.2016.07.034

URL : https://hal.archives-ouvertes.fr/hal-01352724