S. E. Wenzel, Asthma phenotypes: the evolution from clinical to molecular approaches, Nat Med, vol.18, pp.716-725, 2012.

W. C. Moore, D. A. Meyers, S. E. Wenzel, W. G. Teague, H. Li et al., National Heart, Lung, and Blood Institute's Severe Asthma Research Program. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program, Am J Respir Crit Care Med, vol.181, pp.315-323, 2010.

T. Nagasaki, H. Matsumoto, Y. Kanemitsu, K. Izuhara, Y. Tohda et al., Integrating longitudinal information on pulmonary function and inflammation using asthma phenotypes, J Allergy Clin Immunol, vol.133, pp.1474-1477, 2014.

K. J. Baines, J. L. Simpson, L. G. Wood, R. J. Scott, N. L. Fibbens et al., Sputum gene expression signature of 6 biomarkers discriminates asthma inflammatory phenotypes, J Allergy Clin Immunol, vol.133, pp.997-1007, 2014.

W. Wu, E. Bleecker, W. Moore, W. W. Busse, M. Castro et al., Unsupervised phenotyping of Severe Asthma Research Program participants using expanded lung data, J Allergy Clin Immunol, vol.133, pp.1280-1288, 2014.

J. R. Arron, H. Scheerens, and J. G. Matthews, Redefining approaches to asthma: developing targeted biologic therapies, Adv Pharmacol, vol.66, pp.1-49, 2013.

P. G. Woodruff, B. Modrek, D. F. Choy, G. Jia, A. R. Abbas et al., T-helper type 2-driven inflammation defines major subphenotypes of asthma, Am J Respir Crit Care Med, vol.180, pp.388-395, 2009.

J. Corren, R. F. Lemanske, N. A. Hanania, P. E. Korenblat, M. V. Parsey et al., Lebrikizumab treatment in adults with asthma, N Engl J Med, vol.365, pp.1088-1098, 2011.

P. Haldar, C. E. Brightling, B. Hargadon, S. Gupta, W. Monteiro et al., Mepolizumab and exacerbations of refractory eosinophilic asthma, N Engl J Med, vol.360, pp.973-984, 2009.

P. Haldar, C. E. Brightling, A. Singapuri, B. Hargadon, S. Gupta et al., Outcomes after cessation of mepolizumab therapy in severe eosinophilic asthma: a 12-month follow-up analysis, J Allergy Clin Immunol, vol.133, pp.921-923, 2014.

H. Ortega, G. Chupp, P. Bardin, A. Bourdin, G. Garcia et al., The role of mepolizumab in atopic and nonatopic severe asthma with persistent eosinophilia, Eur Respir J, vol.44, pp.239-241, 2014.

P. Nair, M. Pizzichini, M. Kjarsgaard, M. D. Inman, A. Efthimiadis et al., Mepolizumab for prednisone-dependent asthma with sputum eosinophilia, N Engl J Med, vol.360, pp.985-993, 2009.

D. Boever, E. H. Ashman, C. Cahn, A. P. Locantore, N. W. Overend et al., Efficacy and safety of an anti-IL-13 mAb in patients with severe asthma: a randomized trial, J Allergy Clin Immunol, vol.133, pp.989-996, 2014.

W. Al-ramli, D. Préfontaine, F. Chouiali, J. G. Martin, R. Olivenstein et al., T(H)17-associated cytokines (IL-17A and IL-17F) in severe asthma, J Allergy Clin Immunol, vol.123, pp.1185-1187, 2009.

D. M. Bullens, E. Truyen, L. Coteur, E. Dilissen, P. W. Hellings et al., IL-17 mRNA in sputum of asthmatic patients: linking T cell driven inflammation and granulocytic influx?, Respir Res, vol.7, p.135, 2006.

M. Pelletier, L. Maggi, A. Micheletti, E. Lazzeri, N. Tamassia et al., Evidence for a cross-talk between human neutrophils and Th17 cells, Blood, vol.115, pp.335-343, 2010.

C. Nembrini, B. J. Marsland, and M. Kopf, IL-17-producing T cells in lung immunity and inflammation, J Allergy Clin Immunol, vol.123, pp.995-996, 2009.

S. Yu, H. Y. Kim, Y. Chang, R. H. Dekruyff, and D. T. Umetsu, Innate lymphoid cells and asthma, J Allergy Clin Immunol, vol.133, pp.943-950, 2014.

S. T. Scanlon and A. Mckenzie, Type 2 innate lymphoid cells: new players in asthma and allergy, Curr Opin Immunol, vol.24, pp.707-712, 2012.

J. A. Walker, J. L. Barlow, and A. Mckenzie, Innate lymphoid cells-how did we miss them?, Nat Rev Immunol, vol.13, pp.75-87, 2013.

K. R. Bartemes, K. Iijima, T. Kobayashi, G. M. Kephart, A. N. Mckenzie et al., IL-33-responsive lineage-CD251 CD44(hi) lymphoid cells mediate innate type 2 immunity and allergic inflammation in the lungs, J Immunol, vol.188, pp.1503-1513, 2012.

T. Halim, C. A. Steer, L. Mathä, M. J. Gold, I. Martinez-gonzalez et al., Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation, Immunity, vol.40, pp.425-435, 2014.

H. Y. Kim, Y. J. Chang, S. Subramanian, H. H. Lee, L. A. Albacker et al., Innate lymphoid cells responding to IL-33 mediate airway hyperreactivity independently of adaptive immunity, J Allergy Clin Immunol, vol.129, pp.216-227, 2012.

H. Y. Kim, H. J. Lee, Y. Chang, M. Pichavant, S. A. Shore et al., Interleukin-17-producing innate lymphoid cells and the NLRP3 inflammasome facilitate obesity-associated airway hyperreactivity, Nat Med, vol.20, pp.54-61, 2014.

A. Vazquez-tello, R. Halwani, R. Li, J. Nadigel, A. Bar-or et al., IL-17A and IL-17F expression in B lymphocytes, Int Arch Allergy Immunol, vol.157, pp.406-416, 2012.

P. R. Taylor, S. Roy, S. M. Leal, Y. Sun, S. J. Howell et al., Activation of neutrophils by autocrine IL-17A-IL-17RC interactions during fungal infection is regulated by IL-6, IL-23, RORgt and dectin-2, Nat Immunol, vol.15, pp.143-151, 2014.

J. R. Murdoch and C. M. Lloyd, Resolution of allergic airway inflammation and airway hyperreactivity is mediated by IL-17-producing gdT cells, Am J Respir Crit Care Med, vol.182, pp.464-476, 2010.

M. Pichavant, S. Goya, E. H. Meyer, R. A. Johnston, H. Y. Kim et al., Ozone exposure in a mouse model induces airway hyperreactivity that requires the presence of natural killer T cells and IL-17, J Exp Med, vol.205, pp.385-393, 2008.

S. E. Wenzel, L. B. Schwartz, E. L. Langmack, J. L. Halliday, J. B. Trudeau et al., Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics, Am J Respir Crit Care Med, vol.160, pp.1001-1008, 1999.

W. C. Moore, A. T. Hastie, X. Li, H. Li, W. W. Busse et al., National Heart, Lung, and Blood Institute's Severe Asthma Research Program. Sputum neutrophil counts are associated with more severe asthma phenotypes using cluster analysis, J Allergy Clin Immunol, vol.133, pp.1557-1563, 2014.

A. T. Hastie, W. C. Moore, D. A. Meyers, P. L. Vestal, H. Li et al., Bleecker ER; National Heart, Lung, and Blood Institute Severe Asthma Research Program. Analyses of asthma severity phenotypes and inflammatory proteins in subjects stratified by sputum granulocytes, J Allergy Clin Immunol, vol.125, pp.1028-1036, 2010.

S. Molet, Q. Hamid, F. Davoine, E. Nutku, R. Taha et al., IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines, J Allergy Clin Immunol, vol.108, pp.430-438, 2001.

A. Barczyk, W. Pierzchala, and E. Sozañska, Interleukin-17 in sputum correlates with airway hyperresponsiveness to methacholine, Respir Med, vol.97, pp.726-733, 2003.

Y. Sun, Q. Zhou, and W. Yao, Sputum interleukin-17 is increased and associated with airway neutrophilia in patients with severe asthma, Chin Med J (Engl), vol.118, pp.953-956, 2005.

C. Doe, M. Bafadhel, S. Siddiqui, D. Desai, V. Mistry et al., Expression of the T helper 17-associated cytokines IL-17A and IL-17F in asthma and COPD, Chest, vol.138, pp.1140-1147, 2010.

I. Agache, C. Ciobanu, C. Agache, and M. Anghel, Increased serum IL-17 is an independent risk factor for severe asthma, Respir Med, vol.104, pp.1131-1137, 2010.

Y. Zhao, J. Yang, Y. Gao, and W. Guo, Th17 immunity in patients with allergic asthma, Int Arch Allergy Immunol, vol.151, pp.297-307, 2010.

Y. H. Wang, K. S. Voo, B. Liu, C. Y. Chen, B. Uygungil et al., A novel subset of CD4(1) T(H)2 memory/effector cells that produce inflammatory IL-17 cytokine and promote the exacerbation of chronic allergic asthma, J Exp Med, vol.207, pp.2479-2491, 2010.

L. Cosmi, L. Maggi, V. Santarlasci, M. Capone, E. Cardilicchia et al., Identification of a novel subset of human circulating memory CD4(1) T cells that produce both IL-17A and IL-4, J Allergy Clin Immunol, vol.125, pp.222-230, 2010.

J. W. Chien, C. Y. Lin, K. D. Yang, C. H. Lin, J. K. Kao et al., Increased IL-17A secreting CD41 T cells, serum IL-17 levels and exhaled nitric oxide are correlated with childhood asthma severity, Clin Exp Allergy, vol.43, pp.1018-1026, 2013.

J. Pène, S. Chevalier, L. Preisser, E. Vénéreau, M. Guilleux et al., Chronically inflamed human tissues are infiltrated by highly differentiated Th17 lymphocytes, J Immunol, vol.180, pp.7423-7430, 2008.

C. Irvin, I. Zafar, J. Good, D. Rollins, C. Christianson et al., Increased frequency of dual-positive TH2/TH17 cells in bronchoalveolar lavage fluid characterizes a population of patients with severe asthma, J Allergy Clin Immunol, 2014.

E. B. Brandt, M. B. Kovacic, G. B. Lee, A. M. Gibson, T. H. Acciani et al., Diesel exhaust particle induction of IL-17A contributes to severe asthma, J Allergy Clin Immunol, vol.132, pp.1194-1204, 2013.

K. A. Papp, C. Leonardi, A. Menter, J. Ortonne, J. G. Krueger et al., Brodalumab, an antiinterleukin-17-receptor antibody for psoriasis, N Engl J Med, vol.366, pp.1181-1189, 2012.

J. G. Krueger, S. Fretzin, M. Suárez-fariñas, P. A. Haslett, K. M. Phipps et al., IL-17A is essential for cell activation and inflammatory gene circuits in subjects with psoriasis, J Allergy Clin Immunol, vol.130, pp.145-154, 2012.

R. H. Wilson, G. S. Whitehead, H. Nakano, M. E. Free, J. K. Kolls et al., Allergic sensitization through the airway primes Th17-dependent neutrophilia and airway hyperresponsiveness, Am J Respir Crit Care Med, vol.180, pp.720-730, 2009.

R. He, M. K. Oyoshi, H. Jin, and R. S. Geha, Epicutaneous antigen exposure induces a Th17 response that drives airway inflammation after inhalation challenge, Proc Natl Acad Sci, vol.104, pp.15817-15822, 2007.

J. Zhao, C. M. Lloyd, and A. Noble, Th17 responses in chronic allergic airway inflammation abrogate regulatory T-cell-mediated tolerance and contribute to airway remodeling, Mucosal Immunol, vol.6, pp.335-346, 2013.

S. Lajoie, I. P. Lewkowich, Y. Suzuki, J. R. Clark, A. A. Sproles et al., Complement-mediated regulation of the IL-17A axis is a central genetic determinant of the severity of experimental allergic asthma, Nat Immunol, vol.11, pp.928-935, 2010.

L. G. Wood, K. J. Baines, J. Fu, H. A. Scott, and P. G. Gibson, The neutrophilic inflammatory phenotype is associated with systemic inflammation in asthma, Chest, vol.142, pp.86-93, 2012.

Y. Chung, S. H. Chang, G. J. Martinez, X. O. Yang, R. Nurieva et al., Critical regulation of early Th17 cell differentiation by interleukin-1 signaling, Immunity, vol.30, pp.576-587, 2009.

K. J. Baines, J. L. Simpson, N. A. Bowden, R. J. Scott, and P. G. Gibson, Differential gene expression and cytokine production from neutrophils in asthma phenotypes, Eur Respir J, vol.35, pp.522-531, 2010.

M. L. Manni, J. B. Trudeau, E. V. Scheller, S. Mandalapu, M. M. Elloso et al., The complex relationship between inflammation and lung function in severe asthma, Mucosal Immunol, vol.7, pp.1186-1198, 2014.

D. Baeten, X. Baraliakos, J. Braun, J. Sieper, P. Emery et al., Antiinterleukin-17A monoclonal antibody secukinumab in treatment of ankylosing spondylitis: a randomised, double-blind, placebocontrolled trial, Lancet, vol.382, pp.1705-1713, 2013.

C. E. Jones and K. Chan, Interleukin-17 stimulates the expression of interleukin-8, growth-related oncogene-alpha, and granulocytecolony-stimulating factor by human airway epithelial cells, Am J Respir Cell Mol Biol, vol.26, pp.748-753, 2002.

F. Mcallister, A. Henry, J. L. Kreindler, P. J. Dubin, L. Ulrich et al., Role of IL-17A, IL-17F, and the IL-17 receptor in regulating growth-related oncogenealpha and granulocyte colony-stimulating factor in bronchial epithelium: implications for airway inflammation in cystic fibrosis, J Immunol, vol.175, pp.404-412, 2005.

B. Boisson, C. Wang, V. Pedergnana, L. Wu, S. Cypowyj et al., An ACT1 mutation selectively abolishes interleukin-17 responses in humans with chronic mucocutaneous candidiasis, Immunity, vol.39, pp.676-686, 2013.

A. Drewniak, R. P. Gazendam, A. Tool, M. Van-houdt, M. H. Jansen et al., Invasive fungal infection and impaired neutrophil killing in human CARD9 deficiency, Blood, vol.121, pp.2385-2392, 2013.

A. Bush and S. Saglani, Management of severe asthma in children, Lancet, vol.376, pp.814-825, 2010.

L. Mckinley, J. F. Alcorn, A. Peterson, R. B. Dupont, S. Kapadia et al., Th17 cells mediate steroid-resistant airway inflammation and airway hyperresponsiveness in mice, J Immunol, vol.181, pp.4089-4097, 2008.

S. Ano, Y. Morishima, Y. Ishii, K. Yoh, Y. Yageta et al., Transcription factors GATA-3 and RORgt are important for determining the phenotype of allergic airway inflammation in a murine model of asthma, J Immunol, vol.190, pp.1056-1065, 2013.

A. Vazquez-tello, R. Halwani, Q. Hamid, and S. Al-muhsen, Glucocorticoid receptor-beta up-regulation and steroid resistance induction by IL-17 and IL-23 cytokine stimulation in peripheral mononuclear cells, J Clin Immunol, vol.33, pp.466-478, 2013.

A. M. Nanzer, E. S. Chambers, K. Ryanna, D. F. Richards, C. Black et al., Enhanced production of IL-17A in patients with severe asthma is inhibited by 1a,25-dihydroxyvitamin D3 in a glucocorticoidindependent fashion, J Allergy Clin Immunol, vol.132, pp.297-304, 2013.

Q. Liang, L. Guo, S. Gogate, Z. Karim, A. Hanifi et al., IL-2 and IL-4 stimulate MEK1 expression and contribute to T cell resistance against suppression by TGF-beta and IL-10 in asthma, J Immunol, vol.185, pp.5704-5713, 2010.

D. C. Tsitoura and P. B. Rothman, Enhancement of MEK/ERK signaling promotes glucocorticoid resistance in CD41 T cells, J Clin Invest, vol.113, pp.619-627, 2004.

A. Gupta, S. Dimeloe, D. F. Richards, E. S. Chambers, C. Black et al., Defective IL-10 expression and in vitro steroid-induced IL-17A in paediatric severe therapy-resistant asthma, Thorax, vol.69, pp.508-515, 2014.

R. Ramesh, L. Kozhaya, K. Mckevitt, I. M. Djuretic, T. J. Carlson et al., Proinflammatory human Th17 cells selectively express P-glycoprotein and are refractory to glucocorticoids, J Exp Med, vol.211, pp.89-104, 2014.

J. Cao, G. Ren, Y. Gong, S. Dong, Y. Yin et al., Bronchial epithelial cells release IL-6, CXCL1 and CXCL8 upon mast cell interaction, Cytokine, vol.56, pp.823-831, 2011.

F. Huang, C. Y. Kao, S. Wachi, P. Thai, J. Ryu et al., Requirement for both JAK-mediated PI3K signaling and ACT1/TRAF6/TAK1-dependent NF-kappaB activation by IL-17A in enhancing cytokine expression in human airway epithelial cells, J Immunol, vol.179, pp.6504-6513, 2007.

G. J. Zijlstra, T. Hacken, N. H. Hoffmann, R. F. Van-oosterhout, A. J. Heijink et al., Interleukin-17A induces glucocorticoid insensitivity in human bronchial epithelial cells, Eur Respir J, vol.39, pp.439-445, 2012.

T. S. Hallstrand, Y. Lai, W. A. Altemeier, C. L. Appel, B. Johnson et al., Regulation and function of epithelial secreted phospholipase A2 group X in asthma, Am J Respir Crit Care Med, vol.188, pp.42-50, 2013.

A. Bellini, M. A. Marini, L. Bianchetti, M. Barczyk, M. Schmidt et al., Interleukin (IL)-4, IL-13, and IL-17A differentially affect the profibrotic and proinflammatory functions of fibrocytes from asthmatic patients, Mucosal Immunol, vol.5, pp.140-149, 2012.

S. Al-muhsen, S. Letuve, A. Vazquez-tello, M. A. Pureza, A. et al., Th17 cytokines induce profibrotic cytokines release from human eosinophils, Respir Res, vol.14, p.34, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00802220

J. Chakir, J. Shannon, S. Molet, M. Fukakusa, J. Elias et al., Airway remodeling-associated mediators in moderate to severe asthma: effect of steroids on TGF-b, IL-11, IL-17, and type I and type III collagen expression, J Allergy Clin Immunol, vol.111, pp.1293-1298, 2003.

H. Hayashi, A. Kawakita, S. Okazaki, M. Yasutomi, H. Murai et al., IL-17A/F modulates fibrocyte functions in cooperation with CD40-mediated signaling, Inflammation, vol.36, pp.830-838, 2013.

L. Loubaki, I. Hadj-salem, R. Fakhfakh, J. E. Plante, S. Boisvert et al., Co-culture of human bronchial fibroblasts and CD41 T cells increases Th17 cytokine signature, PLoS ONE, vol.8, p.81983, 2013.

T. Fujisawa, S. Velichko, P. Thai, L. Y. Hung, F. Huang et al., Regulation of airway MUC5AC expression by IL-1beta and IL-17A; the NF-kappaB paradigm, J Immunol, vol.183, pp.6236-6243, 2009.

Y. Chen, P. Thai, Y. Zhao, Y. Ho, M. M. Desouza et al., Stimulation of airway mucin gene expression by interleukin (IL)-17 through IL-6 paracrine/autocrine loop, J Biol Chem, vol.278, pp.17036-17043, 2003.

M. Pain, O. Bermudez, P. Lacoste, P. Royer, K. Botturi et al., Tissue remodelling in chronic bronchial diseases: from the epithelial to mesenchymal phenotype, Eur Respir Rev, vol.23, pp.118-130, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-02163334

R. Vittal, L. Fan, D. S. Greenspan, E. A. Mickler, B. Gopalakrishnan et al., IL-17 induces type V collagen overexpression and EMT via TGFb-dependent pathways in obliterative bronchiolitis, Am J Physiol Lung Cell Mol Physiol, vol.304, pp.401-414, 2013.

X. Ji, J. Li, L. Xu, W. Wang, M. Luo et al., IL4 and IL-17A provide a Th2/Th17-polarized inflammatory milieu in favor of TGF-b1 to induce bronchial epithelial-mesenchymal transition (EMT), Int J Clin Exp Pathol, vol.6, pp.1481-1492, 2013.

S. S. Sohal, C. Ward, and E. H. Walters, Importance of epithelial mesenchymal transition (EMT) in COPD and asthma, Thorax, vol.69, p.768, 2014.

Y. Chang, L. Al-alwan, P. Risse, A. J. Halayko, J. G. Martin et al., Th17-associated cytokines promote human airway smooth muscle cell proliferation, FASEB J, vol.26, pp.5152-5160, 2012.

P. O. Girodet, A. Ozier, I. Bara, J. M. Tunon-de-lara, R. Marthan et al., Airway remodeling in asthma: new mechanisms and potential for pharmacological intervention, Pharmacol Ther, vol.130, pp.325-337, 2011.

S. Dragon, M. S. Rahman, J. Yang, H. Unruh, A. J. Halayko et al., IL-17 enhances IL-1beta-mediated CXCL-8 release from human airway smooth muscle cells, Am J Physiol Lung Cell Mol Physiol, vol.292, pp.1023-1029, 2007.

L. A. Al-alwan, Y. Chang, C. J. Baglole, P. Risse, A. J. Halayko et al., Autocrine-regulated airway smooth muscle cell migration is dependent on IL-17-induced growth-related oncogenes, J Allergy Clin Immunol, vol.130, pp.977-985, 2012.

Y. Chang, L. Al-alwan, P. Risse, L. Roussel, S. Rousseau et al., TH17 cytokines induce human airway smooth muscle cell migration, J Allergy Clin Immunol, vol.127, pp.1046-1053, 2011.

M. Kudo, A. C. Melton, C. Chen, M. B. Engler, K. E. Huang et al., IL-17A produced by ab T cells drives airway hyper-responsiveness in mice and enhances mouse and human airway smooth muscle contraction, Nat Med, vol.18, pp.547-554, 2012.

G. Loirand, V. Sauzeau, P. Pacaud, and G. Small, Small G proteins in the cardiovascular system: physiological and pathological aspects, Physiol Rev, vol.93, pp.1659-1720, 2013.

W. W. Busse, S. Holgate, E. Kerwin, Y. Chon, J. Feng et al., double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma, Am J Respir Crit Care Med, vol.188, pp.1294-1302, 2013.

T. Kato, Y. Takeda, T. Nakada, and F. Sendo, Inhibition by dexamethasone of human neutrophil apoptosis in vitro, Nat Immun, vol.14, pp.198-208, 1995.

D. C. Cowan, J. O. Cowan, R. Palmay, A. Williamson, and D. R. Taylor, Effects of steroid therapy on inflammatory cell subtypes in asthma, Thorax, vol.65, pp.384-390, 2010.

W. C. Moore, E. R. Bleecker, D. Curran-everett, S. C. Erzurum, B. T. Ameredes et al., National Heart, Lung, Blood Institute's Severe Asthma Research Program. Characterization of the severe asthma phenotype by the National Heart, Lung, and Blood Institute's Severe Asthma Research Program, J Allergy Clin Immunol, vol.119, pp.405-413, 2007.