A. and H. Pitié-salpétrière, Laboratoire d'Hématologie Biologique

H. Laboratoire-d'anatomopathologie and . Pompidou,

, Laboratoire d'anatomopathologie

G. Hospitalier-pitié-salpétrière, 11 Banque des tissus Humains, Hôpital saint Louis, 1149.

A. and H. Pompidou, Laboratoire d'anatomopathologie

A. and H. Pitié-salpétrière, Hôpital Pitié-Salpétrière, 1149.

, Banque des tissus Humains, Hôpital saint Louis

A. , G. Hospitalier-pitié-salpétrière, ;. , and G. Hospitalier-pitié-salpétrière, Service d'imagerie vasculaire

(. and H. Pitié-salpétrière, , pp.47-83

. Paris, , p.42

, Coordinated T follicular helper cells and B cells in tertiary lymphoid structures in Takayasu arteritis

, INSERM, UMR S 959

F. ,

, Hôpital Pitié-Salpêtrière, AP-HP, F-75651

A. and G. Hospitalier-pitié-salpêtrière, références Maladies Autoimmunes et systémiques rares et Maladies Autoinflammatoires rares

H. Laboratoire-d'anatomopathologie and . Pompidou,

, Laboratoire d'anatomopathologie

, Groupe Hospitalier Pitié-Salpétrière

, I3 « Immunology, immunopathology, immunotherapy » UMR 7211 (CNRS/UPMC) INSERM U959, Hôpital Pitié-Salpétrière, 47-83 boulevard de l'Hôpital, vol.17, pp.42-59

S. G. Tangye, C. S. Ma, R. Brink, and E. K. Deenick, The good, the bad and the ugly-TFH cells in human health and disease, Nat Rev Immunol, vol.13, pp.412-438, 2013.

S. L. Gaffen, R. Jain, A. V. Garg, and D. J. Cua, The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing, Nat Rev Immunol, vol.14, pp.585-600, 2014.

P. J. Mease, Inhibition of interleukin-17, interleukin-23 and the TH17 cell pathway in the treatment of psoriatic arthritis and psoriasis, Curr Opin Rheumatol, vol.27, pp.127-160, 2015.

M. H. Kaplan, M. M. Hufford, and M. R. Olson, The development and in vivo function of T helper 9 cells, Nat Rev Immunol, vol.15, pp.295-307, 2015.

F. Ciccia, G. Guggino, A. Ferrante, P. Cipriani, R. Giacomelli et al., Interleukin-9 and T helper type 9 cells in rheumatic diseases: IL-9 in rheumatic diseases, Clin Exp Immunol, vol.185, pp.125-157, 2016.

E. Witte, K. Witte, K. Warszawska, R. Sabat, and K. Wolk, Interleukin-22: a cytokine produced by T, NK and NKT cell subsets, with importance in the innate immune defense and tissue protection, Cytokine Growth Factor Rev, vol.21, pp.365-79, 2010.

A. Mirshafiey, A. Simhag, E. Rouby, N. Azizi, and G. , T-helper 22 cells as a new player in chronic inflammatory skin disorders, Int J Dermatol, vol.54, pp.880-888, 2015.

H. Ueno, Human Circulating T Follicular Helper Cell Subsets in Health and Disease, J Clin Immunol, vol.36, issue.1, pp.34-43, 2016.

N. Schmitt, J. Bustamante, L. Bourdery, S. E. Bentebibel, S. Boisson-dupuis et al., IL-12 receptor ?1 deficiency alters in vivo T follicular helper cell response in humans, Blood, vol.121, pp.3375-85, 2013.

N. Schmitt, Y. Liu, S. Bentebibel, I. Munagala, L. Bourdery et al., The cytokine TGF-? co-opts signaling via STAT3-STAT4 to promote the differentiation of human TFH cells, Nat Immunol, vol.15, pp.856-65, 2014.

R. Morita, N. Schmitt, S. Bentebibel, R. Ranganathan, L. Bourdery et al., Human blood CXCR5(+)CD4(+) T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion, Immunity, vol.34, pp.108-129, 2011.

N. Schmitt, S. Bentebibel, and H. Ueno, Phenotype and functions of memory Tfh cells in human blood, Trends Immunol, vol.35, pp.436-478, 2014.

H. Ueno, J. Banchereau, and C. G. Vinuesa, Pathophysiology of T follicular helper cells in humans and mice, Nat Immunol, vol.16, pp.142-52, 2015.

C. Luo, Y. Li, W. Liu, H. Feng, H. Wang et al., Expansion of circulating counterparts of follicular helper T cells in patients with myasthenia gravis, J Neuroimmunol, vol.256, pp.55-61, 2013.

N. Simpson, P. A. Gatenby, A. Wilson, S. Malik, D. A. Fulcher et al., Expansion of circulating T cells resembling follicular helper T cells is a fixed phenotype that identifies a subset of severe systemic lupus erythematosus, Arthritis Rheum, vol.62, pp.234-278, 2010.

J. Wang, Y. Shan, Z. Jiang, J. Feng, C. Li et al., High frequencies of activated B cells and T follicular helper cells are correlated with disease activity in patients with new-onset rheumatoid arthritis, Clin Exp Immunol, vol.174, pp.212-232, 2013.

L. Coz, C. Joublin, A. Pasquali, J. Korganow, A. Dumortier et al., Circulating TFH subset distribution is strongly affected in lupus patients with an active disease, PloS One, vol.8, p.75319, 2013.

S. Pallikkuth, P. Kanthikeel, S. Silva, S. Y. Fischl, M. Pahwa et al., Upregulation of IL-21 receptor on B cells and IL-21 secretion distinguishes novel 2009 H1N1 vaccine responders from nonresponders among HIV-infected persons on combination antiretroviral therapy, J Immunol Baltim Md, vol.186, pp.6173-81, 1950.

S. Pallikkuth, A. Parmigiani, S. Y. Silva, V. K. George, M. Fischl et al., Impaired peripheral blood T-follicular helper cell function in HIV-infected nonresponders to the 2009 H1N1/09 vaccine, Blood, vol.120, pp.985-93, 2012.

M. A. Linterman and D. L. Hill, Can follicular helper T cells be targeted to improve vaccine efficacy?, vol.5, 2016.

M. Lindqvist, J. Van-lunzen, D. Z. Soghoian, B. D. Kuhl, S. Ranasinghe et al., Expansion of HIV-specific T follicular helper cells in chronic HIV infection, J Clin Invest, vol.122, pp.3271-80, 2012.

C. Petrovas, T. Yamamoto, M. Y. Gerner, K. L. Boswell, K. Wloka et al., CD4 T follicular helper cell dynamics during SIV infection, J Clin Invest, vol.122, pp.3281-94, 2012.

C. L. Maynard, L. E. Harrington, K. M. Janowski, J. R. Oliver, C. L. Zindl et al., Regulatory T cells expressing interleukin 10 develop from Foxp3+ and Foxp3-precursor cells in the absence of interleukin 10, Nat Immunol, vol.8, pp.931-972, 2007.

A. M. Thornton, P. E. Korty, D. Q. Tran, E. A. Wohlfert, P. E. Murray et al., Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells, J Immunol Baltim Md, vol.184, pp.3433-3474, 1950.

M. E. Himmel, K. G. Macdonald, R. V. Garcia, T. S. Steiner, and M. K. Levings, Helios+ and Helios-cells coexist within the natural FOXP3+ T regulatory cell subset in humans, J Immunol Baltim Md, vol.190, pp.2001-2009, 1950.

L. Lu, J. Barbi, and F. Pan, The regulation of immune tolerance by FOXP3, Nat Rev Immunol, 2017.

M. Miyara, Y. Yoshioka, A. Kitoh, T. Shima, K. Wing et al., Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor, Immunity, vol.30, pp.899-911, 2009.

L. Lu, J. Barbi, and F. Pan, The regulation of immune tolerance by FOXP3, Nat Rev Immunol, 2017.

H. Kim, R. A. Barnitz, T. Kreslavsky, F. D. Brown, H. Moffett et al., Stable inhibitory activity of regulatory T cells requires the transcription factor Helios, Science, vol.350, pp.334-343, 2015.

E. A. Wohlfert, J. R. Grainger, N. Bouladoux, J. E. Konkel, G. Oldenhove et al., GATA3 controls Foxp3 + regulatory T cell fate during inflammation in mice, J Clin Invest, vol.121, pp.4503-4518, 2011.
DOI : 10.1172/jci57456

URL : http://www.jci.org/articles/view/57456/files/pdf

Y. Wang, M. A. Su, and Y. Y. Wan, An essential role of the transcription factor GATA-3 for the function of regulatory T cells, Immunity, vol.35, pp.337-385, 2011.

L. Charbonnier, S. Wang, P. Georgiev, E. Sefik, and T. A. Chatila, Control of peripheral tolerance by regulatory T cell-intrinsic Notch signaling, Nat Immunol, vol.16, pp.1162-73, 2015.

N. Komatsu, K. Okamoto, S. Sawa, T. Nakashima, M. Oh-hora et al., Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis, Nat Med, vol.20, pp.62-70, 2014.
DOI : 10.1038/nm.3432

URL : https://cloudfront.escholarship.org/dist/prd/content/qt5944829g/qt5944829g.pdf?t=oijgst

A. G. Levine, A. Medoza, S. Hemmers, B. Moltedo, R. E. Niec et al., Stability and function of regulatory T cells expressing the transcription factor T-bet, Nature, vol.546, pp.421-426, 2017.

K. Minton, Regulatory T cells: Subset-specific suppression, Nat Rev Immunol, vol.17, p.401, 2017.

W. Zou, Regulatory T cells, tumour immunity and immunotherapy, Nat Rev Immunol, vol.6, pp.295-307, 2006.

C. Garlanda, C. A. Dinarello, and A. Mantovani, The Interleukin-1 Family: Back to the Future, Immunity, vol.39, pp.1003-1021, 2013.

C. Garlanda, C. A. Dinarello, and A. Mantovani, The Interleukin-1 Family: Back to the Future, Immunity, vol.39, pp.1003-1021, 2013.

F. Y. Liew, J. Girard, and H. R. Turnquist, Interleukin-33 in health and disease, Nat Rev Immunol, vol.16, pp.676-689, 2016.

M. Peine, R. M. Marek, and M. Löhning, IL-33 in T Cell Differentiation, Function, and Immune Homeostasis, Trends Immunol, vol.37, pp.321-354, 2016.

B. Griesenauer and S. Paczesny, The ST2/iL-33 Axis in immune Cells during inflammatory Diseases. Front Immunol, vol.8, 2017.

C. Cayrol and J. Girard, IL-33: an alarmin cytokine with crucial roles in innate immunity, inflammation and allergy, Curr Opin Immunol, vol.31, pp.31-38, 2014.

Y. Choi, H. Choi, J. Min, B. Pyun, Y. Maeng et al., Interleukin-33 induces angiogenesis and vascular permeability through ST2/TRAF6-mediated endothelial nitric oxide production, Blood, vol.114, pp.3117-3126, 2009.

S. Demyanets, V. Konya, S. P. Kastl, C. Kaun, S. Rauscher et al., Interleukin-33 induces expression of adhesion molecules and inflammatory activation in human endothelial cells and in human atherosclerotic plaques, Arterioscler Thromb Vasc Biol, vol.31, pp.2080-2089, 2011.

C. Homey, B. Soumelis, and V. , A modular view of cytokine networks in atopic dermatitis, Clin Rev Allergy Immunol, vol.41, pp.245-53, 2011.

H. Morita, K. Arae, H. Unno, K. Miyauchi, S. Toyama et al., An Interleukin-33-Mast CellInterleukin-2 Axis Suppresses Papain-Induced Allergic Inflammation by Promoting Regulatory T Cell Numbers, Immunity, vol.43, pp.175-86, 2015.

R. Saluja, M. Khan, M. K. Church, and M. Maurer, The role of IL-33 and mast cells in allergy and inflammation, Clin Transl Allergy, vol.5, p.33, 2015.

A. M. Gilfillan and M. A. Beaven, Regulation of mast cell responses in health and disease, Crit Rev Immunol, vol.31, 2011.

G. Palmer, D. Talabot-ayer, C. Lamacchia, D. Toy, C. A. Seemayer et al., Inhibition of interleukin-33 signaling attenuates the severity of experimental arthritis, Arthritis Rheum, vol.60, pp.738-787, 2009.

D. Xu, H. Jiang, P. Kewin, Y. Li, R. Mu et al., IL-33 exacerbates antigen-induced arthritis by activating mast cells, Proc Natl Acad Sci, vol.105, pp.10913-10918, 2008.

J. Biton, K. Athari, S. Thiolat, A. Santinon, F. Lemeiter et al., In Vivo Expansion of Activated Foxp3 + Regulatory T Cells and Establishment of a Type 2 Immune Response upon IL-33 Treatment Protect against Experimental Arthritis, J Immunol, vol.197, pp.1708-1727, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01933351

Y. Okayama and T. Kawakami, Development, migration, and survival of mast cells, Immunol Res, vol.34, pp.97-115, 2006.

C. Tkaczyk, V. Horejsi, S. Iwaki, P. Draber, L. E. Samelson et al., NTAL phosphorylation is a pivotal link between the signaling cascades leading to human mast cell degranulation following Kit activation and Fc epsilon RI aggregation, Blood, vol.104, pp.207-221, 2004.

Z. Allakhverdi, D. E. Smith, M. R. Comeau, and G. Delespesse, Cutting edge: The ST2 ligand IL-33 potently activates and drives maturation of human mast cells, J Immunol Baltim Md, vol.179, pp.2051-2055, 1950.

M. Iikura, H. Suto, N. Kajiwara, K. Oboki, T. Ohno et al., IL-33 can promote survival, adhesion and cytokine production in human mast cells, Lab Investig J Tech Methods Pathol, vol.87, pp.971-979, 2007.

L. H. Ho, T. Ohno, K. Oboki, N. Kajiwara, H. Suto et al., IL-33 induces IL-13 production by mouse mast cells independently of IgE-FcepsilonRI signals, J Leukoc Biol, vol.82, pp.1481-90, 2007.

M. V. Andrade, S. Iwaki, C. Ropert, R. T. Gazzinelli, J. R. Cunha-melo et al., Amplification of cytokine production through synergistic activation of NFAT and AP-1 following stimulation of mast cells with antigen and IL-33, Eur J Immunol, vol.41, pp.760-72, 2011.

P. N. Pushparaj, H. K. Tay, H. 'ng, S. C. Pitman, N. Xu et al., The cytokine interleukin-33 mediates anaphylactic shock, Proc Natl Acad Sci, vol.106, pp.9773-9781, 2009.

T. Y. Jang and Y. H. Kim, Interleukin-33 and mast cells bridge innate and adaptive immunity: from the allergologist's perspective, Int Neurourol J, vol.19, p.142, 2015.

D. C. Fong, O. Malbec, M. Arock, J. C. Cambier, W. H. Fridman et al., Selective in vivo recruitment of the phosphatidylinositol phosphatase SHIP by phosphorylated Fc gammaRIIB during negative regulation of IgE-dependent mouse mast cell activation, Immunol Lett, vol.54, pp.83-91, 1996.

A. M. Gilfillan and M. A. Beaven, Phagocytosis-independent antimicrobial activity of mast cells by means of extracellular trap formation, Crit Rev Immunol, vol.31, 2011.

A. M. Gilfillan and M. A. Beaven, Regulation of mast cell responses in health and disease, Crit Rev Immunol, vol.31, 2011.

J. Kalesnikoff and S. J. Galli, Anaphylaxis: mechanisms of mast cell activation, Chem Immunol Allergy, vol.95, pp.45-66, 2010.
DOI : 10.1159/000315937

S. C. Bischoff, Role of mast cells in allergic and non-allergic immune responses: comparison of human and murine data, Nat Rev Immunol, vol.7, pp.93-104, 2007.

O. T. Burton and H. C. Oettgen, Beyond immediate hypersensitivity: evolving roles for IgE antibodies in immune homeostasis and allergic diseases, Immunol Rev, vol.242, pp.128-171, 2011.

F. Rivellese, A. Nerviani, F. W. Rossi, G. Marone, M. Matucci-cerinic et al., Mast cells in rheumatoid arthritis: friends or foes?, Autoimmun Rev, vol.16, pp.557-63, 2017.

D. M. Lee, D. S. Friend, M. F. Gurish, C. Benoist, D. Mathis et al., Mast cells: a cellular link between autoantibodies and inflammatory arthritis, Science, vol.297, pp.1689-92, 2002.

D. Van-der-velden, H. M. Lagraauw, A. Wezel, P. Launay, J. Kuiper et al., Mast cell depletion in the preclinical phase of collagen-induced arthritis reduces clinical outcome by lowering the inflammatory cytokine profile, Arthritis Res Ther, vol.18, 2016.

N. Schubert, J. Dudeck, P. Liu, A. Karutz, S. Speier et al., Mast cell promotion of T celldriven antigen-induced arthritis despite being dispensable for antibody-induced arthritis in which T cells are bypassed, Arthritis Rheumatol Hoboken NJ, vol.67, pp.903-916, 2015.

M. G. Buckley, C. Walters, W. M. Wong, M. I. Cawley, S. Ren et al., Mast cell activation in arthritis: detection of alpha-and beta-tryptase, histamine and eosinophil cationic protein in synovial fluid, Clin Sci Lond Engl, vol.93, pp.363-70, 1979.

C. Sandler, K. A. Lindstedt, S. Joutsiniemi, J. Lappalainen, T. Juutilainen et al., Selective activation of mast cells in rheumatoid synovial tissue results in production of TNF-alpha, IL1beta and IL-1Ra, Inflamm Res Off J Eur Histamine Res Soc Al, vol.56, pp.230-239, 2007.

F. Rivellese, J. Suurmond, K. Habets, A. L. Dorjée, N. Ramamoorthi et al., Ability of Interleukin-33-and Immune Complex-Triggered Activation of Human Mast Cells to DownRegulate Monocyte-Mediated Immune Responses: MAST CELL SUPPRESSION OF MONOCYTE ACTIVATION, Arthritis Rheumatol, vol.67, pp.2343-53, 2015.

D. B. Leveson-gower, E. I. Sega, J. Kalesnikoff, M. Florek, Y. Pan et al., Mast cells suppress murine GVHD in a mechanism independent of CD4+CD25+ regulatory T cells, Blood, vol.122, pp.3659-65, 2013.

C. Sunderkötter, M. Goebeler, K. Schulze-osthoff, R. Bhardwaj, and C. Sorg, Macrophage-derived angiogenesis factors, Pharmacol Ther, vol.51, pp.195-216, 1991.

S. M. Stamatovic, R. F. Keep, M. Mostarica-stojkovic, and A. V. Andjelkovic, CCL2 regulates angiogenesis via activation of Ets-1 transcription factor, J Immunol Baltim Md, vol.177, pp.2651-61, 1950.
DOI : 10.4049/jimmunol.177.4.2651

URL : http://www.jimmunol.org/content/177/4/2651.full.pdf

J. Ehling, M. Bartneck, X. Wei, F. Gremse, V. Fech et al., CCL2-dependent infiltrating macrophages promote angiogenesis in progressive liver fibrosis, Gut, vol.63, pp.1960-71, 2014.

A. Ridiandries, J. Tan, and C. Bursill, The Role of CC-Chemokines in the Regulation of Angiogenesis, Int J Mol Sci, vol.17, p.1856, 2016.

M. Heil, T. Ziegelhoeffer, F. Pipp, S. Kostin, S. Martin et al., Blood monocyte concentration is critical for enhancement of collateral artery growth, Am J Physiol Heart Circ Physiol, vol.283, pp.2411-2419, 2002.

H. Niiyama, H. Kai, T. Yamamoto, T. Shimada, K. Sasaki et al., Roles of endogenous monocyte chemoattractant protein-1 in ischemia-induced neovascularization, J Am Coll Cardiol, vol.44, pp.661-667, 2004.

V. Van-weel, R. Toes, L. Seghers, M. Deckers, M. R. De-vries et al., Natural killer cells and CD4+ T-cells modulate collateral artery development, Arterioscler Thromb Vasc Biol, vol.27, pp.2310-2318, 2007.

E. Stabile, M. S. Burnett, C. Watkins, T. Kinnaird, A. Bachis et al., Impaired arteriogenic response to acute hindlimb ischemia in CD4-knockout mice, Circulation, vol.108, pp.205-215, 2003.

E. Stabile, T. Kinnaird, L. Sala, A. Hanson, S. K. Watkins et al., CD8+ T lymphocytes regulate the arteriogenic response to ischemia by infiltrating the site of collateral vessel development and recruiting CD4+ mononuclear cells through the expression of interleukin-16, Circulation, vol.113, pp.118-142, 2006.

M. Kneilling, L. Hültner, B. J. Pichler, R. Mailhammer, L. Morawietz et al., Targeted mast cell silencing protects against joint destruction and angiogenesis in experimental arthritis in mice, Arthritis Rheum, vol.56, pp.1806-1822, 2007.

Y. Zouggari, H. Ait-oufella, L. Waeckel, J. Vilar, C. Loinard et al., Regulatory T Cells Modulate Postischemic Neovascularization, Circulation, vol.120, pp.1415-1440, 2009.

J. S. Silvestre, Z. Mallat, M. Duriez, R. Tamarat, M. F. Bureau et al., Antiangiogenic effect of interleukin-10 in ischemia-induced angiogenesis in mice hindlimb, Circ Res, vol.87, pp.448-52, 2000.

A. Facciabene, X. Peng, I. S. Hagemann, K. Balint, A. Barchetti et al., Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells, Nature, vol.475, pp.226-256, 2011.

D. Palma, M. Biziato, D. Petrova, and T. V. , Microenvironmental regulation of tumour angiogenesis, Nat Rev Cancer, vol.17, pp.457-74, 2017.

D. Palma, M. Biziato, D. Petrova, and T. V. , Microenvironmental regulation of tumour angiogenesis, Nat Rev Cancer, vol.17, pp.457-74, 2017.

T. A. Wynn and K. M. Vannella, Macrophages in Tissue Repair, Regeneration, and Fibrosis, Immunity, vol.44, pp.450-62, 2016.

S. K. Verma, V. Garikipati, P. Krishnamurthy, S. M. Schumacher-bass, L. A. Grisanti et al., Interleukin 10 Inhibits Bone Marrow Fibroblast Progenitor Cell-Mediated Cardiac Fibrosis in Pressure Overloaded Myocardium, Circulation, 2017.

K. M. Vannella and T. A. Wynn, Mechanisms of Organ Injury and Repair by Macrophages, Annu Rev Physiol, vol.79, pp.593-617, 2017.

A. L. Mescher, Macrophages and fibroblasts during inflammation and tissue repair in models of organ regeneration, Mescher. Regeneration, vol.4, pp.39-53, 2017.

A. E. Postlethwaite, M. A. Holness, H. Katai, and R. Raghow, Human fibroblasts synthesize elevated levels of extracellular matrix proteins in response to interleukin 4, J Clin Invest, vol.90, pp.1479-85, 1992.

T. A. Wynn, Fibrotic disease and the TH1/TH2 paradigm, Nat Rev Immunol, vol.4, pp.583-94, 2004.

T. Nakashima, M. Jinnin, K. Yamane, N. Honda, I. Kajihara et al., Impaired IL-17 signaling pathway contributes to the increased collagen expression in scleroderma fibroblasts, J Immunol Baltim Md, vol.188, pp.3573-83, 1950.

N. C. Brembilla, E. Montanari, M. Truchetet, E. Raschi, P. Meroni et al., Th17 cells favor inflammatory responses while inhibiting type I collagen deposition by dermal fibroblasts: differential effects in healthy and systemic sclerosis fibroblasts, Arthritis Res Ther, vol.15, p.151, 2013.
DOI : 10.1186/ar4334

URL : https://arthritis-research.biomedcentral.com/track/pdf/10.1186/ar4334

M. Truchetet, N. Brembilla, E. Montanari, P. Lonati, E. Raschi et al., Interleukin-17A+ cell counts are increased in systemic sclerosis skin and their number is inversely correlated with the extent of skin involvement, Arthritis Rheum, vol.65, pp.1347-56, 2013.

D. Burzyn, W. Kuswanto, D. Kolodin, J. L. Shadrach, M. Cerletti et al., A special population of regulatory T cells potentiates muscle repair, Cell, vol.155, pp.1282-95, 2013.

W. Kuswanto, D. Burzyn, M. Panduro, K. K. Wang, Y. C. Jang et al., Poor Repair of Skeletal Muscle in Aging Mice Reflects a Defect in Local, Interleukin-33-Dependent Accumulation of Regulatory T Cells, Immunity, vol.44, pp.355-67, 2016.

G. D. Rak, L. C. Osborne, M. C. Siracusa, B. S. Kim, K. Wang et al.,

, Innate Lymphoid Cells Promote Cutaneous Wound Healing, J Invest Dermatol, vol.136, pp.487-96, 2016.

L. A. Monticelli, L. C. Osborne, M. Noti, S. V. Tran, D. Zaiss et al., IL-33 promotes an innate immune pathway of intestinal tissue protection dependent on amphiregulin-EGFR interactions, Proc Natl Acad Sci U S A, vol.112, pp.10762-10769, 2015.

I. G. Luzina, P. Kopach, V. Lockatell, P. H. Kang, A. Nagarsekar et al., Interleukin-33 potentiates bleomycin-induced lung injury, Am J Respir Cell Mol Biol, vol.49, pp.999-1008, 2013.
DOI : 10.1165/rcmb.2013-0093oc

URL : http://europepmc.org/articles/pmc3931117?pdf=render

D. Li, R. Guabiraba, A. Besnard, M. Komai-koma, M. S. Jabir et al., IL-33 promotes ST2dependent lung fibrosis by the induction of alternatively activated macrophages and innate lymphoid cells in mice, J Allergy Clin Immunol, vol.134, pp.1422-1432, 2014.

S. Sanada, D. Hakuno, L. J. Higgins, E. R. Schreiter, A. Mckenzie et al., IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system, J Clin Invest, vol.117, pp.1538-1587, 2007.
DOI : 10.1172/jci30634

URL : http://www.jci.org/articles/view/30634/files/pdf

J. S. Lee, E. Seppanen, J. Patel, M. P. Rodero, and K. Khosrotehrani, ST2 receptor invalidation maintains wound inflammation, delays healing and increases fibrosis, Exp Dermatol, vol.25, pp.71-75, 2016.
DOI : 10.1111/exd.12833

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1111/exd.12833

E. Martínez-martínez, M. Miana, R. Jurado-lópez, E. Rousseau, P. Rossignol et al., A Role for Soluble ST2 in Vascular Remodeling Associated with Obesity in Rats, PLoS ONE, vol.8, p.79176, 2013.

N. K. Wilson, R. A. Gould, G. Macfarlane, E. Consortium, and M. L. , Pathophysiology of aortic aneurysm: insights from human genetics and mouse models, Pharmacogenomics, vol.17, pp.2071-80, 2016.
DOI : 10.2217/pgs-2016-0127

J. R. Stone, P. Bruneval, A. Angelini, G. Bartoloni, C. Basso et al., Consensus statement on surgical pathology of the aorta from the Society for Cardiovascular Pathology and the Association for European Cardiovascular Pathology: I. Inflammatory diseases, Cardiovasc Pathol Off J Soc Cardiovasc Pathol, vol.24, pp.267-78, 2015.

A. C. Lindsay, R. A. Foale, O. Warren, and J. A. Henry, Cannabis as a precipitant of cardiovascular emergencies, Int J Cardiol, vol.104, pp.230-232, 2005.

J. A. Jones, C. Beck, J. R. Barbour, J. A. Zavadzkas, R. Mukherjee et al., Alterations in aortic cellular constituents during thoracic aortic aneurysm development: myofibroblast-mediated vascular remodeling, Am J Pathol, vol.175, pp.1746-56, 2009.

R. He, D. Guo, A. L. Estrera, H. J. Safi, T. T. Huynh et al., Characterization of the inflammatory and apoptotic cells in the aortas of patients with ascending thoracic aortic aneurysms and dissections, J Thorac Cardiovasc Surg, vol.131, pp.671-679, 2006.

U. Schönbeck, G. K. Sukhova, N. Gerdes, and P. Libby, T H 2 predominant immune responses prevail in human abdominal aortic aneurysm, Am J Pathol, vol.161, pp.499-506, 2002.

C. Galle, L. Schandene, P. Stordeur, Y. Peignois, J. Ferreira et al., Predominance of type 1 CD4+T cells in human abdominal aortic aneurysm, Clin Exp Immunol, vol.0, p.051006055454003, 2005.

K. Shimizu, M. Shichiri, P. Libby, R. T. Lee, and R. N. Mitchell, Th2-predominant inflammation and blockade of IFN-? signaling induce aneurysms in allografted aortas, J Clin Invest, vol.114, pp.300-308, 2004.

H. Ait-oufella, Y. Wang, O. Herbin, S. Bourcier, S. Potteaux et al., Natural Regulatory T Cells Limit Angiotensin II-Induced Aneurysm Formation and Rupture in Mice, Arterioscler Thromb Vasc Biol, vol.33, pp.2374-2379, 2013.

Y. Wang, H. Ait-oufella, O. Herbin, P. Bonnin, B. Ramkhelawon et al., TGF-? activity protects against inflammatory aortic aneurysm progression and complications in angiotensin II-infused mice, J Clin Invest, vol.120, pp.422-454, 2010.

D. Allali and C. Chizzolini,

, Rev Med Suisse, vol.10, pp.854-862, 2014.

J. L. Lee, S. M. Naguwa, G. S. Cheema, and M. E. Gershwin, The geo-epidemiology of temporal (giant cell) arteritis, Clin Rev Allergy Immunol, vol.35, pp.88-95, 2008.

W. Rahman and F. Z. Rahman, Giant cell (temporal) arteritis: an overview and update, Surv Ophthalmol, vol.50, pp.415-443, 2005.

C. L. Hill, R. J. Black, J. C. Nossent, C. Ruediger, L. Nguyen et al., Risk of mortality in patients with giant cell arteritis: A systematic review and meta-analysis, Seminars in arthritis and rheumatism, pp.513-519, 2017.

S. S. Hayreh, P. A. Podhajsky, and B. Zimmerman, Ocular manifestations of giant cell arteritis, Am J Ophthalmol, vol.125, pp.509-529, 1998.

J. C. Robson, A. Kiran, J. Maskell, A. Hutchings, N. Arden et al., The relative risk of aortic aneurysm in patients with giant cell arteritis compared with the general population of the UK, Ann Rheum Dis, vol.74, pp.129-164, 2015.

J. M. Evans, W. M. O'fallon, and G. G. Hunder, Increased incidence of aortic aneurysm and dissection in giant cell (temporal) arteritis. A population-based study, Ann Intern Med, vol.122, pp.502-509, 1995.

F. Onen and N. Akkoc, Epidemiology of Takayasu arteritis, 1983.

K. Ishikawa, Diagnostic approach and proposed criteria for the clinical diagnosis of Takayasu's arteriopathy, J Am Coll Cardiol, vol.12, pp.964-72, 1988.

M. Park, S. Lee, Y. Park, N. S. Chung, and S. Lee, Clinical characteristics and outcomes of Takayasu's arteritis: analysis of 108 patients using standardized criteria for diagnosis, activity assessment, and angiographic classification, Scand J Rheumatol, vol.34, pp.284-92, 2005.

S. Sharma, M. Rajani, and K. K. Talwar, Angiographic morphology in nonspecific aortoarteritis (Takayasu's arteritis): a study of 126 patients from north India, Cardiovasc Intervent Radiol, vol.15, pp.160-165, 1992.

G. S. Kerr, C. W. Hallahan, J. Giordano, R. Y. Leavitt, A. S. Fauci et al., Takayasu arteritis, Ann Intern Med, vol.120, pp.919-948, 1994.

Y. Seko, S. Minota, A. Kawasaki, Y. Shinkai, K. Maeda et al., Perforin-secreting killer cell infiltration and expression of a 65-kD heat-shock protein in aortic tissue of patients with Takayasu's arteritis, J Clin Invest, vol.93, p.750, 1994.

D. Saadoun, M. Garrido, C. Comarmond, A. C. Desbois, F. Domont et al., Th1 and Th17 cytokines drive inflammation in Takayasu arteritis, Arthritis Rheumatol Hoboken NJ, vol.67, pp.1353-60, 2015.

L. Arnaud, J. Haroche, A. Mathian, G. Gorochov, and Z. Amoura, Pathogenesis of Takayasu's arteritis: a 2011 update, Autoimmun Rev, vol.11, pp.61-68, 2011.

A. Aggarwal, M. Chag, N. Sinha, and S. Naik, Takayasu's arteritis: role of Mycobacterium tuberculosis and its 65 kDa heat shock protein, Int J Cardiol, vol.55, pp.49-55, 1996.

E. S. Carvalho, A. W. Souza, S. C. Leão, M. Levy-neto, R. S. Oliveira et al., Absence of mycobacterial DNA in peripheral blood and artery specimens in patients with Takayasu arteritis, Clin Rheumatol, vol.36, pp.205-208, 2017.

A. Brack, A. Geisler, V. M. Martinez-taboada, B. R. Younge, J. J. Goronzy et al., Giant cell vasculitis is a T cell-dependent disease, Mol Med, vol.3, p.530, 1997.

C. M. Weyand and J. J. Goronzy, Giant-Cell Arteritis and Polymyalgia Rheumatica, N Engl J Med, vol.371, pp.50-57, 2014.

J. Deng, B. R. Younge, R. A. Olshen, J. J. Goronzy, and C. M. Weyand, Th17 and Th1 T-cell responses in giant cell arteritis, Circulation, vol.121, pp.906-921, 2010.

M. Samson, S. Audia, J. Fraszczak, M. Trad, P. Ornetti et al., Th1 and Th17 lymphocytes expressing CD161 are implicated in giant cell arteritis and polymyalgia rheumatica pathogenesis, Arthritis Rheum, vol.64, pp.3788-98, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00799573

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

G. Espígol-frigolé, M. Corbera-bellalta, E. Planas-rigol, E. Lozano, M. Segarra et al., Increased IL-17A expression in temporal artery lesions is a predictor of sustained response to glucocorticoid treatment in patients with giant-cell arteritis, Ann Rheum Dis, vol.72, pp.1481-1488, 2013.

K. Piggott, J. Deng, K. Warrington, B. Younge, J. T. Kubo et al., Blocking the NOTCH pathway inhibits vascular inflammation in large-vessel vasculitis, Circulation, vol.123, pp.309-327, 2011.

F. Ciccia, A. R. Rizzo, A. Principe, S. Raiata, F. Cavazza et al., Expression of interleukin32 in the inflamed arteries of patients with giant cell arteritis, Arthritis Rheum, vol.63, pp.2097-104, 2011.

F. Ciccia, A. Rizzo, G. Guggino, A. Cavazza, A. R. Maugeri et al., Difference in the expression of IL-9 and IL-17 correlates with different histological pattern of vascular wall injury in giant cell arteritis, Rheumatology, vol.54, pp.1596-604, 2015.

M. Samson, K. H. Ly, B. Tournier, N. Janikashvili, M. Trad et al., Involvement and prognosis value of CD8(+) T cells in giant cell arteritis, J Autoimmun, vol.72, pp.73-83, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01431809

K. Van-der-geest, W. H. Abdulahad, P. Chalan, A. Rutgers, G. Horst et al., Disturbed B Cell Homeostasis in Newly Diagnosed Giant Cell Arteritis and Polymyalgia Rheumatica: B Cells in GCA and PMR, Arthritis Rheumatol, vol.66, pp.1927-1965, 2014.

M. A. Nagel, T. White, N. Khmeleva, A. Rempel, P. J. Boyer et al., Analysis of VaricellaZoster Virus in Temporal Arteries Biopsy Positive and Negative for Giant Cell Arteritis, JAMA Neurol, vol.72, p.1281, 2015.

D. Gilden, T. M. White, L. Nagae, W. H. Gurdin, P. J. Boyer et al., Successful Antiviral Treatment of Giant Cell Arteritis and Takayasu Arteritis, JAMA Neurol, vol.72, pp.943-949, 2015.

F. Muratore, S. Croci, I. Tamagnini, A. Zerbini, S. Bellafiore et al., No detection of varicella-zoster virus in temporal arteries of patients with giant cell arteritis. Semin Arthritis Rheum, 2017.

R. Watanabe, H. Zhang, G. Berry, J. J. Goronzy, and C. M. Weyand, Immune checkpoint dysfunction in large and medium vessel vasculitis, Am J Physiol Heart Circ Physiol, vol.312, pp.1052-1061, 2017.

J. Deng, W. Ma-krupa, A. T. Gewirtz, B. R. Younge, J. J. Goronzy et al., Toll-like receptors 4 and 5 induce distinct types of vasculitis, Circ Res, vol.104, pp.488-495, 2009.

O. Pryshchep, W. Ma-krupa, B. R. Younge, J. J. Goronzy, and C. M. Weyand, Vessel-Specific Toll-Like Receptor Profiles in Human Medium and Large Arteries, Circulation, vol.118, pp.1276-84, 2008.

M. Soussan, P. Nicolas, C. Schramm, S. Katsahian, G. Pop et al., Management of largevessel vasculitis with FDG-PET: a systematic literature review and meta-analysis, Medicine (Baltimore), vol.94, p.622, 2015.

F. Ciccia, A. R. Rizzo, A. Raimondo, S. Giardina, A. Raiata et al., IL-33 is overexpressed in the inflamed arteries of patients with giant cell arteritis, Ann Rheum Dis, vol.72, pp.258-264, 2013.

F. Numano, M. Okawara, H. Inomata, and Y. Kobayashi, Takayasu's arteritis, Lancet Lond Engl, vol.356, pp.1023-1028, 2000.

D. Saadoun, M. Garrido, C. Comarmond, A. C. Desbois, F. Domont et al., Th1 and Th17 cytokines drive Takayasu Arteritis inflammation, Arthritis Rheumatol Hoboken NJ, 2015.
DOI : 10.1002/art.39037

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1002/art.39037

A. Márquez, R. Solans, J. Hernández-rodríguez, M. C. Cid, S. Castañeda et al., A candidate gene approach identifies an IL33 genetic variant as a novel genetic risk factor for GCA, PloS One, vol.9, p.113476, 2014.

G. G. Hunder, D. A. Bloch, B. A. Michel, M. B. Stevens, W. P. Arend et al., The American College of Rheumatology 1990 criteria for the classification of giant cell arteritis, Arthritis Rheum, vol.33, pp.1122-1130, 1990.

, The American College of Rheumatology 1990 criteria for the classification of Takayasu arteritis, 2017.

G. S. Kerr, C. W. Hallahan, J. Giordano, R. Y. Leavitt, A. S. Fauci et al., Takayasu arteritis, Ann Intern Med, vol.120, pp.919-948, 1994.

R. Saleh, G. Wedeh, H. Herrmann, S. Bibi, S. Cerny-reiterer et al., A new human mast cell line expressing a functional IgE receptor converts to tumorigenic growth by KIT D816V transfection, Blood, vol.124, pp.111-131, 2014.
DOI : 10.1182/blood-2013-10-534685

URL : https://hal.archives-ouvertes.fr/hal-01514454

B. Maneglier, C. Rogez-kreuz, P. Cordonnier, P. Therond, C. Advenier et al., Simultaneous measurement of kynurenine and tryptophan in human plasma and supernatants of cultured human cells by HPLC with coulometric detection, Clin Chem, vol.50, pp.2166-2174, 2004.

E. S. Baekkevold, M. Roussigné, T. Yamanaka, F. Johansen, F. L. Jahnsen et al., Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules, Am J Pathol, vol.163, pp.69-79, 2003.

G. Haraldsen, J. Balogh, J. Pollheimer, J. Sponheim, and A. M. Küchler, Interleukin-33-cytokine of dual function or novel alarmin?, Trends Immunol, vol.30, pp.227-260, 2009.
DOI : 10.1016/j.it.2009.03.003

A. M. Miller, Role of IL-33 in inflammation and disease, J Inflamm Lond, vol.8, p.22, 2011.

J. Bartunek, L. Delrue, F. Van-durme, O. Muller, F. Casselman et al., Nonmyocardial production of ST2 protein in human hypertrophy and failure is related to diastolic load, J Am Coll Cardiol, vol.52, pp.2166-74, 2008.

F. Y. Liew, N. I. Pitman, and I. B. Mcinnes, Disease-associated functions of IL-33: the new kid in the IL-1 family, Nat Rev Immunol, vol.10, pp.103-113, 2010.

C. Schiering, T. Krausgruber, A. Chomka, A. Fröhlich, K. Adelmann et al., The alarmin IL-33 promotes regulatory T-cell function in the intestine, Nature, vol.513, pp.564-572, 2014.

C. Orabona and U. Grohmann, Indoleamine 2,3-dioxygenase and regulatory function: tryptophan starvation and beyond, Methods Mol Biol Clifton NJ, vol.677, pp.269-80, 2011.
DOI : 10.1007/978-1-60761-869-0_19

L. Lu, E. F. Lind, D. C. Gondek, K. A. Bennett, M. W. Gleeson et al., Mast cells are essential intermediaries in regulatory T-cell tolerance, Nature, vol.442, pp.997-1002, 2006.

C. P. Rodrigues, A. Ferreira, M. P. Pinho, C. J. De-moraes, P. C. Bergami-santos et al., Tolerogenic IDO+ dendritic cells are induced by PD-1-expressing mast cells. Front Immunol, vol.7, 2016.

D. B. Leveson-gower, E. I. Sega, J. Kalesnikoff, M. Florek, Y. Pan et al., Mast cells suppress murine GVHD in a mechanism independent of CD4+CD25+ regulatory T cells, Blood, vol.122, pp.3659-65, 2013.

H. Morita, K. Arae, H. Unno, K. Miyauchi, S. Toyama et al., An interleukin-33-mast cellinterleukin-2 axis suppresses papain-induced allergic inflammation by promoting regulatory T cell numbers, Immunity, vol.43, pp.175-186, 2015.

C. M. Weyand and J. J. Goronzy, Medium-and large-vessel vasculitis, N Engl J Med, vol.349, pp.160-169, 2003.
DOI : 10.1056/nejmra022694

Y. Seko, S. Minota, A. Kawasaki, Y. Shinkai, K. Maeda et al., Perforin-secreting killer cell infiltration and expression of a 65-kD heat-shock protein in aortic tissue of patients with Takayasu's arteritis, J Clin Invest, vol.93, p.750, 1994.

Y. Seko, O. Sato, A. Takagi, Y. Tada, H. Matsuo et al., Restricted usage of T-cell receptor Valpha-Vbeta genes in infiltrating cells in aortic tissue of patients with Takayasu's arteritis, Circulation, vol.93, pp.1788-90, 1996.

M. Kaiser, B. Younge, J. Björnsson, J. J. Goronzy, and C. M. Weyand, Formation of new vasa vasorum in vasculitis. Production of angiogenic cytokines by multinucleated giant cells, Am J Pathol, vol.155, pp.765-74, 1999.

A. M. Miller, D. Xu, D. L. Asquith, L. Denby, Y. Li et al., IL-33 reduces the development of atherosclerosis, J Exp Med, vol.205, pp.339-385, 2008.

I. Ligi, S. Simoncini, E. Tellier, P. F. Vassallo, F. Sabatier et al., A switch toward angiostatic gene expression impairs the angiogenic properties of endothelial progenitor cells in low birth weight preterm infants, Blood, vol.118, pp.1699-709, 2011.

M. C. Cid, M. Cebrián, C. Font, B. Coll-vinent, J. Hernández-rodríguez et al., Cell adhesion molecules in the development of inflammatory infiltrates in giant cell arteritis: inflammation-induced angiogenesis as the preferential site of leukocyte-endothelial cell interactions, Arthritis Rheum, vol.43, pp.184-94, 2000.

Q. Espinassous, E. Garcia-de-paco, I. Garcia-verdugo, M. Synguelakis, S. Von-aulock et al., IL-33 enhances lipopolysaccharide-induced inflammatory cytokine production from mouse macrophages by regulating lipopolysaccharide receptor complex, J Immunol Baltim Md, vol.183, pp.1446-55, 1950.

R. D. Rudic, E. G. Shesely, N. Maeda, O. Smithies, S. S. Segal et al., Direct evidence for the importance of endothelium-derived nitric oxide in vascular remodeling, J Clin Invest, vol.101, pp.731-737, 1998.

K. Seki, S. Sanada, A. Y. Kudinova, M. L. Steinhauser, V. Handa et al., Interleukin-33 prevents apoptosis and improves survival after experimental myocardial infarction through ST2 signaling, Circ Heart Fail, p.109, 2009.

Y. Matsuyama, H. Okazaki, H. Tamemoto, H. Kimura, Y. Kamata et al., Increased levels of interleukin 33 in sera and synovial fluid from patients with active rheumatoid arthritis, J Rheumatol, vol.37, pp.18-25, 2010.

K. Yanaba, A. Yoshizaki, Y. Asano, T. Kadono, and S. Sato, Serum IL-33 levels are raised in patients with systemic sclerosis: association with extent of skin sclerosis and severity of pulmonary fibrosis, Clin Rheumatol, vol.30, pp.825-855, 2011.

T. Mchedlidze, M. Waldner, S. Zopf, J. Walker, A. L. Rankin et al., Interleukin-33dependent innate lymphoid cells mediate hepatic fibrosis, Immunity, vol.39, pp.357-371, 2013.

H. Zhang, R. Watanabe, G. J. Berry, A. Vaglio, Y. J. Liao et al., Immunoinhibitory checkpoint deficiency in medium and large vessel vasculitis, Proc Natl Acad Sci, vol.114, pp.970-979, 2017.

J. Deng, W. Ma-krupa, A. T. Gewirtz, B. R. Younge, J. J. Goronzy et al., Toll-Like Receptors 4 and 5 Induce Distinct Types of Vasculitis, Circ Res, vol.104, pp.488-95, 2009.

F. Ciccia, A. Rizzo, A. Ferrante, G. Guggino, S. Croci et al., New insights into the pathogenesis of giant cell arteritis, Autoimmun Rev, vol.16, pp.675-83, 2017.

M. Clement, A. Galy, P. Bruneval, M. Morvan, F. Hyafil et al., Tertiary lymphoid Organs in Takayasu arteritis. Front Immunol, vol.7, 2016.

F. Ciccia, A. Rizzo, R. Maugeri, A. R. Croci, S. Guggino et al., Ectopic expression of CXCL13, BAFF, APRIL and LT-? is associated with artery tertiary lymphoid organs in giant cell arteritis, Ann Rheum Dis, vol.76, pp.235-278, 2017.

S. Kumar-chauhan, K. Tripathy, N. Sinha, N. Singh, M. Nityanand et al., Cellular and humoral immune responses to mycobacterial heat shock protein-65 and its human homologue in Takayasu's arteritis, Clin Exp Immunol, vol.138, pp.547-553, 2004.

D. R. Schultz and P. I. Arnold, Heat shock (stress) proteins and autoimmunity in rheumatic diseases, Semin Arthritis Rheum, vol.22, pp.357-74, 1993.

L. Arnaud, E. Cambau, I. Brocheriou, F. Koskas, E. Kieffer et al., Absence of Mycobacterium tuberculosis in arterial lesions from patients with Takayasu's arteritis, J Rheumatol, vol.36, pp.1682-1687, 2009.

D. J. Rawlings, G. Metzler, M. Wray-dutra, and J. Sw, Altered B cell signalling in autoimmunity, Nat Rev Immunol, vol.17, pp.421-457, 2017.

M. Noris, D. E. Gamba, S. Bonazzola, S. Remuzzi, and G. , Interleukin-6 and RANTES in Takayasu arteritis: a guide for therapeutic decisions?, Circulation, vol.100, pp.55-60, 1999.

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

A. Peters, L. A. Pitcher, J. M. Sullivan, M. Mitsdoerffer, S. E. Acton et al., Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation, Immunity, vol.35, pp.986-96, 2011.

B. F. Hoyer, I. M. Mumtaz, K. Loddenkemper, A. Bruns, C. Sengler et al., Takayasu arteritis is characterised by disturbances of B cell homeostasis and responds to B cell depletion therapy with rituximab, Ann Rheum Dis, vol.71, pp.75-84, 2012.

J. M. Odegard, B. R. Marks, L. D. Diplacido, A. C. Poholek, D. H. Kono et al., ICOS-dependent extrafollicular helper T cells elicit IgG production via IL-21 in systemic autoimmunity, J Exp Med, vol.205, pp.2873-86, 2008.

P. Guilpain and L. Mouthon, Antiendothelial cells autoantibodies in vasculitis-associated systemic diseases, Clin Rev Allergy Immunol, vol.35, pp.59-65, 2008.

S. K. Chauhan, N. K. Tripathy, and S. Nityanand, Antigenic targets and pathogenicity of anti-aortic endothelial cell antibodies in Takayasu arteritis, Arthritis Rheum, vol.54, pp.2326-2359, 2006.

M. Park, Y. Park, S. Y. Jung, K. H. Lee, and S. Lee, Anti-endothelial cell antibodies and antiphospholipid antibodies in Takayasu's arteritis: correlations of their titers and isotype distributions with disease activity, Clin Exp Rheumatol, vol.24, p.10, 2006.

S. Kang, Y. Fedoriw, E. K. Brenneman, Y. K. Truong, K. Kikly et al., BAFF Induces Tertiary Lymphoid Structures and Positions T Cells within the Glomeruli during Lupus Nephritis, J Immunol Baltim Md, vol.198, pp.2602-2613, 1950.

C. Pitzalis, G. W. Jones, M. Bombardieri, and S. A. Jones, Ectopic lymphoid-like structures in infection, cancer and autoimmunity, Nat Rev Immunol, vol.14, pp.447-62, 2014.

L. Pattarini, C. Trichot, S. Bogiatzi, M. Grandclaudon, S. Meller et al., TSLP-activated dendritic cells induce human T follicular helper cell differentiation through OX40-ligand, J Exp Med, vol.214, pp.1529-1575, 2017.

O. Karadag, K. Aksu, A. Sahin, F. Y. Zihni, B. Sener et al., Assessment of latent tuberculosis infection in Takayasu arteritis with tuberculin skin test and Quantiferon-TB Gold test, Rheumatol Int, vol.30, pp.1483-1490, 2010.

Y. Seko, N. Takahashi, Y. Tada, H. Yagita, K. Okumura et al., Restricted usage of T-cell receptor Vgamma-Vdelta genes and expression of costimulatory molecules in Takayasu's arteritis, Int J Cardiol, vol.75, 2000.

S. K. Chauhan, N. K. Tripathy, and S. Nityanand, Antigenic targets and pathogenicity of anti-aortic endothelial cell antibodies in Takayasu arteritis, Arthritis Rheum, vol.54, pp.2326-2359, 2006.

M. Li, Y. Li, X. Liu, X. Gao, and Y. Wang, IL-33 blockade suppresses the development of experimental autoimmune encephalomyelitis in C57BL/6 mice, J Neuroimmunol, vol.247, pp.25-31, 2012.

H. Jiang, M. Milovanovi?, D. Allan, W. Niedbala, A. Besnard et al., IL-33 attenuates EAE by suppressing IL-17 and IFN-? production and inducing alternatively activated macrophages, Eur J Immunol, vol.42, pp.1804-1818, 2012.

U. Grohmann, F. Fallarino, and P. P. Tolerance, DCs and tryptophan: much ado about IDO, Trends Immunol, vol.24, pp.242-250, 2003.

B. M. Matta, J. M. Lott, L. R. Mathews, Q. Liu, B. R. Rosborough et al., IL-33 is an unconventional Alarmin that stimulates IL-2 secretion by dendritic cells to selectively expand IL-33R/ST2+ regulatory T cells, J Immunol Baltim Md, vol.193, pp.4010-4030, 1950.

C. Baumann, W. V. Bonilla, A. Fröhlich, C. Helmstetter, M. Peine et al., T-bet-and STAT4-dependent IL-33 receptor expression directly promotes antiviral Th1 cell responses, Proc Natl Acad Sci, vol.112, pp.4056-4061, 2015.

J. E. Rood, S. Rao, M. Paessler, P. A. Kreiger, N. Chu et al., ST2 contributes to T-cell hyperactivation and fatal hemophagocytic lymphohistiocytosis in mice, Blood, vol.127, pp.426-435, 2016.

D. K. Reichenbach, V. Schwarze, B. M. Matta, V. Tkachev, E. Lieberknecht et al., The IL-33/ST2 axis augments effector T-cell responses during acute GVHD, Blood, vol.125, pp.3183-92, 2015.

B. M. Matta, D. K. Reichenbach, X. Zhang, L. Mathews, B. H. Koehn et al., Peri-alloHCT IL33 administration expands recipient T-regulatory cells that protect mice against acute GVHD, Blood, vol.128, pp.427-466, 2016.

P. Gro?, K. Doser, W. Falk, F. Obermeier, and C. Hofmann, IL-33 attenuates development and perpetuation of chronic intestinal inflammation, Inflamm Bowel Dis, vol.18, pp.1900-1909, 2012.

M. A. Grimbaldeston, S. Nakae, J. Kalesnikoff, M. Tsai, and S. J. Galli, Mast cell-derived interleukin 10 limits skin pathology in contact dermatitis and chronic irradiation with ultraviolet B, Nat Immunol, vol.8, pp.1095-104, 2007.

S. J. Galli, S. Nakae, and M. Tsai, Mast cells in the development of adaptive immune responses, Nat Immunol, vol.6, pp.135-177, 2005.

Z. Wen, Y. Shen, G. Berry, F. Shahram, Y. Li et al., The microvascular niche instructs T cells in large vessel vasculitis via the VEGF-Jagged1-Notch pathway, Sci Transl Med, vol.9, 2017.

L. A. Monticelli, G. F. Sonnenberg, M. C. Abt, T. Alenghat, C. Ziegler et al., Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus, Nat Immunol, vol.12, pp.1045-54, 2011.
DOI : 10.1038/ni.2131

M. Samson and B. Bonnotte, Pathogénie de l'artérite à cellules géantes, Presse Médicale, vol.41, pp.937-984, 2012.

J. J. Maleszewski, B. R. Younge, J. T. Fritzlen, G. G. Hunder, J. J. Goronzy et al., Clinical and pathological evolution of giant cell arteritis: a prospective study of follow-up temporal artery biopsies in 40 treated patients, Mod Pathol, 2017.

M. Samson, T. Ghesquière, S. Berthier, and B. Bonnotte, Ustekinumab inhibits Th1 and Th17 polarisation in a patient with giant cell arteritis, Ann Rheum Dis, 2017.