S. J. Walsh and L. M. Rau, Autoimmune diseases: a leading cause of death among young and middle-aged women in the United States, Am J Public Health, vol.90, pp.1463-1469, 2000.

D. Mcgonagle and M. F. Mcdermott, A proposed classification of the immunological diseases, PLoS Med, vol.3, p.297, 2006.

A. Doria, M. Zen, and S. Bettio, Autoinflammation and autoimmunity: bridging the divide, Autoimmun Rev, vol.12, pp.22-30, 2012.

F. Ciccarelli, M. Martinis, and L. Ginaldi, An update on autoinflammatory diseases, Curr Med Chem, vol.21, pp.261-270, 2013.

L. Magyari, D. Varszegi, and E. Kovesdi, Interleukins and interleukin receptors in rheumatoid arthritis: Research, diagnostics and clinical implications, World J Orthop, vol.5, pp.516-552, 2014.

E. M. Moran and F. L. Mastaglia, Cytokines in immune-mediated inflammatory myopathies: cellular sources, multiple actions and therapeutic implications, Clin Exp Immunol, vol.178, pp.405-420, 2014.

T. D. Querec, R. S. Akondy, and E. K. Lee, Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans, Nat Immunol, vol.10, pp.116-141, 2009.

G. A. Poland, H. Quill, and A. Togias, Understanding the human immune system in the 21st century: the Human Immunology Project Consortium, Vaccine, vol.31, pp.2911-2913, 2013.

S. Thomas, V. Rouilly, and P. E. , The Milieu Intérieur study-an integrative approach for study of human immunological variance, Clin Immunol, vol.157, pp.277-93, 2015.

A. Urrutia, D. Duffy, and V. Rouilly, Standardized whole-blood transcriptional profiling enables the deconvolution of complex induced immune responses, Cell Rep, vol.16, pp.2777-91, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01367555

, WHO. WHO | WHO guidelines on drawing blood: best practices in phlebotomy, 2017.

, International Human Microbiome Standards (IHMS) project

M. Piram, I. Koné-paut, and H. J. Lachmann, Validation of the autoinflammatory diseases activity index (AIDAI) for hereditary recurrent fever syndromes, Ann Rheum Dis, vol.73, pp.2168-73, 2014.

J. D. Lewis, S. Chuai, and L. Nessel, Use of the noninvasive components of the Mayo score to assess clinical response in ulcerative colitis, Inflamm Bowel Dis, vol.14, pp.1660-1666, 2008.

R. F. Harvey and J. M. Bradshaw, A simple index of Crohn's-disease activity, Lancet Lond Engl, vol.1, p.514, 1980.

S. Garrett, T. Jenkinson, and L. G. Kennedy, A new approach to defining disease status in ankylosing spondylitis: the Bath Ankylosing Spondylitis Disease Activity Index, J Rheumatol, vol.21, pp.2286-91, 1994.

J. H. Stone, G. S. Hoffman, and P. A. Merkel, A disease-specific activity index for Wegener's granulomatosis: modification of the Birmingham Vasculitis Activity Score. International Network for the Study of the Systemic Vasculitides (INSSYS), Arthritis Rheum, vol.44, p.912, 2001.

G. S. Kerr, C. W. Hallahan, and J. Giordano, Takayasu arteritis, Ann Intern Med, vol.120, pp.919-948, 1994.

D. M. Van-der-heijde, M. A. Van-'t-hof, and P. L. Van-riel, Judging disease activity in clinical practice in rheumatoid arthritis: first step on 14 January 2019 by guest, Ann Rheum Dis, vol.49, pp.916-936, 1990.

H. B. Mortensen, P. Hougaard, and P. Swift, New definition for the partial remission period in children and adolescents with type 1 diabetes, Diabetes Care, vol.32, pp.1384-90, 2009.

C. Bombardier, D. D. Gladman, and M. B. Urowitz, Derivation of the SLEDAI. A disease activity index for lupus patients. The Committee on Prognosis Studies in SLE, Arthritis Rheum, vol.35, pp.630-670, 1992.

R. Lorenzon, I. Drakos, and C. Ribet, Clinical data specification and coding for cross-analyses with omics data in autoimmune disease trials. bioRxiv, 2018.

F. Pitoiset, M. Barbié, and G. Monneret, A standardized flow cytometry procedure for the monitoring of regulatory T cells in clinical trials, Cytometry B Clin Cytom, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01680983

D. Saadoun, M. Rosenzwajg, and F. Joly, Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis, N Engl J Med, vol.365, pp.2067-77, 2011.

A. Hartemann, G. Bensimon, and C. A. Payan, Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind, placebo-controlled trial, Lancet Diabetes Endocrinol, vol.1, pp.295-305, 2013.

Y. Allenbach, W. Chaara, and M. Rosenzwajg, Th1 response and systemic treg deficiency in inclusion body myositis, PLoS One, vol.9, p.88788, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01344508

B. Terrier, W. Chaara, and L. Dufat, Serum biomarker signature identifies patients with B-cell non-Hodgkin lymphoma associated with cryoglobulinemia vasculitis in chronic HCV infection, Autoimmun Rev, vol.13, pp.319-345, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01619585

B. Terrier, G. Geri, and W. Chaara, Interleukin-21 modulates Th1 and Th17 responses in giant cell arteritis, Arthritis Rheum, vol.64, pp.2001-2012, 2012.

H. P. Pham, N. Dérian, and W. Chaara, A novel strategy for molecular signature discovery based on independent component analysis, Int J Data Min Bioinform, vol.9, pp.277-304, 2014.

M. Rosenzwajg, G. Churlaud, and R. Mallone, Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients, J Autoimmun, vol.58, pp.48-58, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01112156

A. S. Bergot, W. Chaara, and E. Ruggiero, TCR sequences and tissue distribution discriminate the subsets of naïve and activated/memory Treg cells in mice, Eur J Immunol, vol.45, pp.1524-1558, 2015.

D. Nehar-belaid, T. Courau, and N. Dérian, Regulatory T cells orchestrate similar immune evasion of fetuses and tumors in mice, J Immunol, vol.196, pp.678-90, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01617582

C. Plessy, E. Mariotti-ferrandiz, and R. Manabe, High throughput analysis of T cell antigen receptor sequences, Biorxiv, 2015.

D. Chaussabel, C. Quinn, and J. Shen, A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus, Immunity, vol.29, pp.150-64, 2008.

A. Tenenhaus and M. Tenenhaus, Regularized generalized canonical correlation analysis, Psychometrika, vol.76, pp.257-84, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00609220

K. Rohart, F. Gonzalez, and I. , Omics Data Integration Project, 2017.

A. Tenenhaus and M. Tenenhaus, Regularized generalized canonical correlation analysis for multiblock or multigroup data analysis, Eur J Oper Res, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01071421

A. Tenenhaus, C. Philippe, and V. Guillemot, Variable selection for generalized canonical correlation analysis, Biostatistics, vol.15, pp.569-83, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01071432

A. Tenenhaus, C. Philippe, and V. Frouin, Kernel generalized canonical correlation analysis, Comput Stat Data Anal, vol.90, pp.114-145, 2015.
URL : https://hal.archives-ouvertes.fr/hal-00553602

L. V. Hooper, D. R. Littman, and A. J. Macpherson, Interactions between the microbiota and the immune system, Science, vol.336, pp.1268-73, 2012.

Y. Belkaid and T. W. Hand, Role of the microbiota in immunity and inflammation, Cell, vol.157, pp.121-162, 2014.

H. Zeng and H. Chi, Metabolic control of regulatory T cell development and function, Trends Immunol, vol.36, pp.3-12, 2015.

K. Atarashi, T. Tanoue, and K. Oshima, Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota, Nature, vol.500, pp.232-238, 2013.

L. M. Kato, S. Kawamoto, and M. Maruya, The role of the adaptive immune system in regulation of gut microbiota, Immunol Rev, vol.260, pp.67-75, 2014.

B. Terrier, I. Bièche, and T. Maisonobe, Interleukin-25: a cytokine linking eosinophils and adaptive immunity in Churg-Strauss syndrome, Blood, vol.116, pp.4523-4554, 2010.

F. C. Brosius and W. Ju, The promise of systems biology for diabetic kidney disease, Adv Chronic Kidney Dis, vol.25, pp.202-215, 2018.

J. G. Burel, S. H. Apte, and D. L. Doolan, Systems approaches towards molecular profiling of human immunity, Trends Immunol, vol.37, pp.53-67, 2016.

T. Huan, B. Zhang, and Z. Wang, A systems biology framework identifies molecular underpinnings of coronary heart disease, Arterioscler Thromb Vasc Biol, vol.33, pp.1427-1461, 2013.

K. Cervantes-gracia and H. Husi, Integrative analysis of multiple sclerosis using a systems biology approach, Sci Rep, vol.8, p.5633, 2018.

H. Zhao and H. Li, Network-based meta-analysis in the identification of biomarkers for papillary thyroid cancer, Gene, vol.661, pp.160-168, 2018.

A. Irigoyen, C. Jimenez-luna, and M. Benavides, Integrative multiplatform meta-analysis of gene expression profiles in pancreatic ductal adenocarcinoma patients for identifying novel diagnostic biomarkers, PLoS One, vol.13, p.194844, 2018.

M. Moon and K. Nakai, Integrative analysis of gene expression and DNA methylation using unsupervised feature extraction for detecting candidate cancer biomarkers, J Bioinform Comput Biol, vol.16, p.1850006, 2018.

S. A. Greenberg, Biomarkers of inclusion body myositis, Curr Opin Rheumatol, vol.25, pp.753-62, 2013.

L. Chiche, N. Jourde-chiche, and E. Whalen, Modular repertoire analysis identifies complex coordinated type I-type, Ann Rheum Dis, vol.73, pp.98-107, 2014.

N. Aghaeepour, E. A. Ganio, and D. Mcilwain, An immune clock of human pregnancy, Sci Immunol, vol.2, p.2946, 2017.