D. M. Schwartz, M. Bonelli, M. Gadina, O. Shea, and J. J. , Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases, Nat Rev Rheumatol, vol.12, pp.25-36, 2016.
DOI : 10.1038/nrrheum.2015.167

URL : http://europepmc.org/articles/pmc4688091?pdf=render

C. A. Dinarello, Introduction to the interleukin-1 family of cytokines and receptors: drivers of innate inflammation and acquired immunity, Immunol Rev, vol.281, pp.5-7, 2018.

K. Fujio, T. Komai, M. Inoue, K. Morita, T. Okamura et al., Revisiting the regulatory roles of the TGF-beta family of cytokines, Autoimmun Rev, vol.15, pp.917-939, 2016.

M. Croft and R. M. Siegel, Beyond TNF: TNF superfamily cytokines as targets for the treatment of rheumatic diseases, Nat Rev Rheumatol, vol.13, pp.217-250, 2017.

M. Choura and A. Rebai, Receptor tyrosine kinases: from biology to pathology, J Recept Signal Transduct Res, vol.31, pp.387-94, 2011.
DOI : 10.3109/10799893.2011.625425

J. W. Griffith, C. L. Sokol, and A. D. Luster, Chemokines and chemokine receptors: positioning cells for host defense and immunity, Annu Rev Immunol, vol.32, pp.659-702, 2014.

M. Akdis, A. Aab, C. Altunbulakli, K. Azkur, R. A. Costa et al., Interleukins (from IL-1 to IL-38), interferons, transforming growth factor beta, and TNF-alpha: receptors, functions, and roles in diseases, J Allergy Clin Immunol, vol.138, pp.984-1010, 2016.

C. Brocker, D. Thompson, A. Matsumoto, D. W. Nebert, and V. Vasiliou, Evolutionary divergence and functions of the human interleukin (IL) gene family, Hum Genomics, vol.5, pp.30-55, 2010.

W. Ouyang, S. Rutz, N. K. Crellin, P. A. Valdez, and S. G. Hymowitz, Regulation and functions of the IL-10 family of cytokines in inflammation and disease, Annu Rev Immunol, vol.29, pp.71-109, 2011.

S. Rutz, C. Eidenschenk, and W. Ouyang, IL-22, not simply a Th17 cytokine, Immunol Rev, vol.252, pp.116-148, 2013.
DOI : 10.1111/imr.12027

A. Knappe, S. Hor, S. Wittmann, and H. Fickenscher, Induction of a novel cellular homolog of interleukin-10, AK155, by transformation of T lymphocytes with herpesvirus saimiri, J Virol, vol.74, pp.3881-3888, 2000.

R. P. Donnelly, F. Sheikh, H. Dickensheets, R. Savan, H. A. Young et al., Interleukin-26: an IL-10-related cytokine produced by Th17 cells, Cytokine Growth Factor Rev, vol.21, pp.393-401, 2010.
DOI : 10.1016/j.cytogfr.2010.09.001

URL : http://europepmc.org/articles/pmc2997847?pdf=render

E. Stephen-victor, H. Fickenscher, and J. Bayry, IL-26: an emerging proinflammatory member of the IL-10 cytokine family with multifaceted actions in antiviral, antimicrobial, and autoimmune responses, PLoS Pathog, vol.12, p.1005624, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01340570

G. Lutfalla, R. Crollius, H. Stange-thomann, N. Jaillon, O. Mogensen et al., Comparative genomic analysis reveals independent expansion of a lineage-specific gene family in vertebrates: the class II cytokine receptors and their ligands in mammals and fish, BMC Genomics, vol.4, p.29, 2003.

K. Vandenbroeck, S. Cunningham, A. Goris, I. Alloza, S. Heggarty et al., Polymorphisms in the interferon-gamma/interleukin-26 gene region contribute to sex bias in susceptibility to rheumatoid arthritis, Arthritis Rheum, vol.48, pp.2773-2781, 2003.

A. Goris, M. G. Marrosu, and K. Vandenbroeck, Novel polymorphisms in the IL10 related AK155 gene (chromosome 12q15), Genes Immun, vol.2, pp.284-290, 2001.

P. L. Collins, M. A. Henderson, and T. M. Aune, Lineage-specific adjacent IFNG and IL26 genes share a common distal enhancer element, Genes Immun, vol.13, pp.481-489, 2012.
DOI : 10.1038/gene.2012.22

URL : https://www.nature.com/articles/gene201222.pdf

S. Meller, D. Domizio, J. Voo, K. S. Friedrich, H. C. Chamilos et al., T(H)17 cells promote microbial killing and innate immune sensing of DNA via interleukin 26, Nat Immunol, vol.16, pp.970-979, 2015.
DOI : 10.1038/ni.3211

URL : http://europepmc.org/articles/pmc4776746?pdf=render

C. Poli, J. F. Augusto, J. Dauve, C. Adam, L. Preisser et al., , p.26

, Confers Proinflammatory properties to extracellular DNA, J Immunol, vol.198, pp.3650-61, 2017.

D. M. Copolovici, K. Langel, E. Eriste, and U. Langel, Cell-penetrating peptides: design, synthesis, and applications, ACS Nano, vol.8, pp.1972-94, 2014.
DOI : 10.1021/nn4057269

Y. S. Choi and A. E. David, Cell penetrating peptides and the mechanisms for intracellular entry, Curr Pharm Biotechnol, vol.15, pp.192-201, 2014.
DOI : 10.2174/1389201015666140617093331

R. Sabat, IL-10 family of cytokines, Cytokine Growth Factor Rev, vol.21, pp.315-339, 2010.
DOI : 10.1016/j.cytogfr.2010.11.001

F. Sheikh, V. V. Baurin, A. Lewis-antes, N. K. Shah, S. V. Smirnov et al., Cutting edge: IL-26 signals through a novel receptor complex composed of IL-20 receptor 1 and IL-10 receptor 2, J Immunol, vol.172, pp.2006-2016, 2004.

S. Hor, H. Pirzer, L. Dumoutier, F. Bauer, S. Wittmann et al., The T-cell lymphokine interleukin-26 targets epithelial cells through the interleukin20 receptor 1 and interleukin-10 receptor 2 chains, J Biol Chem, vol.279, pp.33343-51, 2004.

K. F. Che, S. Tengvall, B. Levanen, E. Silverpil, M. E. Smith et al., Interleukin-26 in antibacterial host defense of human lungs. Effects on neutrophil mobilization, Am J Respir Crit Care Med, vol.190, pp.1022-1053, 2014.

P. Gough, S. Ganesan, and S. K. Datta, IL-20 signaling in activated human neutrophils inhibits neutrophil migration and function, J Immunol, vol.198, pp.4373-82, 2017.
DOI : 10.4049/jimmunol.1700253

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5476316

C. Miot, E. Beaumont, D. Duluc, L. Guillou-guillemette, H. Preisser et al., IL-26 is overexpressed in chronically HCV-infected patients and enhances TRAIL-mediated cytotoxicity and interferon production by human NK cells, Gut, vol.64, pp.1466-75, 2015.

L. Hummelshoj, L. P. Ryder, and L. K. Poulsen, The role of the interleukin-10 subfamily members in immunoglobulin production by human B cells, Scand J Immunol, vol.64, pp.40-47, 2006.

M. Corvaisier, Y. Delneste, H. Jeanvoine, L. Preisser, S. Blanchard et al., IL-26 is overexpressed in rheumatoid arthritis and induces proinflammatory cytokine production and Th17 cell generation, PLoS Biol, vol.10, 2012.
DOI : 10.1371/journal.pbio.1001395

URL : https://journals.plos.org/plosbiology/article/file?id=10.1371/journal.pbio.1001395&type=printable

K. F. Che, E. Tufvesson, S. Tengvall, E. Lappi-blanco, R. Kaarteenaho et al., The neutrophil-mobilizing cytokine interleukin-26 in the airways of long-term tobacco smokers, Clin Sci, vol.132, pp.959-83, 2018.

O. Braum, M. Klages, and H. Fickenscher, The cationic cytokine IL-26 differentially modulates virus infection in culture, PLoS ONE, vol.8, p.70281, 2013.

N. Sapay, Y. Guermeur, and G. Deleage, Prediction of amphipathic in-plane membrane anchors in monotopic proteins using a SVM classifier, BMC Bioinformatics, vol.7, p.255, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00315233

F. G. Avci, B. S. Akbulut, and E. Ozkirimli, Membrane active peptides and their biophysical characterization, Biomolecules, vol.8, p.77, 2018.
DOI : 10.20944/preprints201807.0008.v1

URL : https://www.preprints.org/manuscript/201807.0008/v1/download

S. Tengvall, K. F. Che, and A. Linden, Interleukin-26: an emerging player in host defense and inflammation, J Innate Immun, vol.8, pp.15-22, 2016.

J. Dambacher, F. Beigel, K. Zitzmann, D. Toni, E. N. Goke et al., The role of the novel Th17 cytokine IL-26 in intestinal inflammation, Gut, vol.58, pp.1207-1224, 2009.

J. Pene, S. Chevalier, L. Preisser, E. Venereau, M. H. Guilleux et al., Chronically inflamed human tissues are infiltrated by highly differentiated Th17 lymphocytes, J Immunol, vol.180, pp.7423-7453, 2008.

Y. Zhou, W. Hou, K. Xu, D. Han, C. Jiang et al., The elevated expression of Th17-related cytokines and receptors is associated with skin lesion severity in early systemic sclerosis, Hum Immunol, vol.76, pp.22-31, 2015.

J. R. Konradsen, B. Nordlund, B. Levanen, G. Hedlin, and A. Linden, The cytokine interleukin-26 as a biomarker in pediatric asthma, Respir Res, vol.17, p.32, 2016.

K. Ohnuma, R. Hatano, T. M. Aune, H. Otsuka, S. Iwata et al., Regulation of pulmonary graft-versus-host disease by IL26+CD26+CD4 T lymphocytes, J Immunol, vol.194, pp.3697-712, 2015.
DOI : 10.4049/jimmunol.1402785

URL : http://www.jimmunol.org/content/194/8/3697.full.pdf

P. K. Dagur, A. Biancotto, L. Wei, H. N. Sen, M. Yao et al., MCAMexpressing CD4(+) T cells in peripheral blood secrete IL-17A and are significantly elevated in inflammatory autoimmune diseases, J Autoimmun, vol.37, pp.319-346, 2011.

R. Anuradha, P. J. George, L. E. Hanna, P. Kumaran, V. Chandrasekaran et al., Expansion of parasite-specific CD4+ and CD8+ T cells expressing IL-10 superfamily cytokine members and their regulation in human lymphatic filariasis, PLoS Negl Trop Dis, vol.8, 2014.

K. Wolk, S. Kunz, K. Asadullah, and R. Sabat, Cutting edge: immune cells as sources and targets of the IL-10 family members?, J Immunol, vol.168, pp.5397-402, 2002.

Z. Chen, O. Shea, and J. J. , Regulation of IL-17 production in human lymphocytes, Cytokine, vol.41, pp.71-79, 2008.

E. Volpe, M. Touzot, N. Servant, M. A. Marloie-provost, P. Hupe et al., Multiparametric analysis of cytokine-driven human Th17 differentiation reveals a differential regulation of IL-17 and IL-22 production, Blood, vol.114, pp.3610-3614, 2009.

N. J. Wilson, K. Boniface, J. R. Chan, B. S. Mckenzie, W. M. Blumenschein et al., Development, cytokine profile and function of human interleukin 17producing helper T cells, Nat Immunol, vol.8, pp.950-957, 2007.
DOI : 10.1038/ni1497

W. Kaabachi, E. Bouali, A. Berraies, I. B. Dhifallh, B. Hamdi et al., Interleukin-26 is overexpressed in Behcet's disease and enhances Th17 related-cytokines, Immunol Lett, vol.190, pp.177-84, 2017.
DOI : 10.1016/j.imlet.2017.08.008

K. Ohnuma, R. Hatano, T. Itoh, N. Iwao, N. H. Dang et al., Role of IL-26+CD26+CD4 T cells in pulmonary chronic graft-versus-host disease and treatment with caveolin-1-Ig Fc conjugate, Crit Rev Immunol, vol.36, pp.239-67, 2016.

J. A. Dudakov, A. M. Hanash, and M. R. Van-den-brink, Interleukin-22: immunobiology and pathology, Annu Rev Immunol, vol.33, pp.747-85, 2015.
DOI : 10.1146/annurev-immunol-032414-112123

URL : http://europepmc.org/articles/pmc4407497?pdf=render

Q. Tang, Y. O. Ahn, P. Southern, B. R. Blazar, J. S. Miller et al., Development of IL-22-producing NK lineage cells from umbilical cord blood hematopoietic stem cells in the absence of secondary lymphoid tissue, Blood, vol.117, pp.4052-4057, 2011.

M. Cella, A. Fuchs, W. Vermi, F. Facchetti, K. Otero et al., A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity, Nature, vol.457, pp.722-727, 2009.

M. Cols, A. Rahman, P. J. Maglione, Y. Garcia-carmona, N. Simchoni et al., Expansion of inflammatory innate lymphoid cells in patients with common variable immune deficiency, J Allergy Clin Immunol, vol.137, pp.1206-1221, 2016.

T. Hughes, B. Becknell, S. Mcclory, E. Briercheck, A. G. Freud et al., Stage 3 immature human natural killer cells found in secondary lymphoid tissue constitutively and selectively express the TH 17 cytokine interleukin-22, Blood, vol.113, pp.4008-4018, 2009.

A. Woetmann, M. Alhede, S. Dabelsteen, T. Bjarnsholt, M. Rybtke et al., Interleukin-26 (IL-26) is a novel anti-microbial peptide produced by T cells in response to staphylococcal enterotoxin, Oncotarget, vol.9, pp.19481-19490, 2018.

K. Wolk, K. Witte, E. Witte, S. Proesch, G. Schulze-tanzil et al., Maturing dendritic cells are an important source of IL-29 and IL-20 that may cooperatively increase the innate immunity of keratinocytes, J Leukoc Biol, vol.83, pp.1181-93, 2008.

J. M. Guerra-laso, S. Raposo-garcia, S. Garcia-garcia, C. Diez-tascon, and O. M. Riverolezcano, Microarray analysis of Mycobacterium tuberculosis-infected monocytes reveals IL26 as a new candidate gene for tuberculosis susceptibility, Immunology, vol.144, pp.291-301, 2015.

M. L. Nagalakshmi, E. Murphy, T. Mcclanahan, and R. De-waal-malefyt, Expression patterns of IL-10 ligand and receptor gene families provide leads for biological characterization, Int Immunopharmacol, vol.4, pp.577-92, 2004.

K. F. Che, R. Kaarteenaho, E. Lappi-blanco, B. Levanen, J. Sun et al., Interleukin-26 production in human primary bronchial epithelial cells in response to viral stimulation: modulation by Th17 cytokines, Mol Med, vol.23, pp.247-57, 2017.

L. D. Heftdal, T. Andersen, D. Jaehger, A. Woetmann, R. Ostgard et al., Synovial cell production of IL-26 induces bone mineralization in spondyloarthritis, J Mol Med, vol.95, pp.779-87, 2017.

M. C. Li and S. H. He, IL-10 and its related cytokines for treatment of inflammatory bowel disease, World J Gastroenterol, vol.10, pp.620-625, 2004.

M. R. Yeaman, D. Cheng, B. Desai, L. I. Kupferwasser, Y. Q. Xiong et al., Susceptibility to thrombin-induced platelet microbicidal protein is associated with increased fluconazole efficacy against experimental endocarditis due to Candida albicans, Antimicrob Agents Chemother, vol.48, pp.3051-3057, 2004.

M. R. Yeaman and N. Y. Yount, Unifying themes in host defence effector polypeptides, Nat Rev Microbiol, vol.5, pp.727-767, 2007.

M. R. Yeaman, N. Y. Yount, A. J. Waring, K. D. Gank, D. Kupferwasser et al., Modular determinants of antimicrobial activity in platelet factor-4 family kinocidins, Biochim Biophys Acta, vol.1768, pp.609-628, 2007.

K. Fan, C. Ritter, P. Nghiem, A. Blom, M. E. Verhaegen et al., Circulating cell-free miR-375 as surrogate marker of tumor burden in Merkel cell carcinoma, Clin Cancer Res, vol.24, pp.5873-82, 2018.

P. A. Harris, S. B. Berger, J. U. Jeong, R. Nagilla, D. Bandyopadhyay et al., Discovery of a first-in-class receptor interacting protein 1 (RIP1) kinase specific clinical candidate (GSK2982772) for the treatment of inflammatory diseases, J Med Chem, vol.60, pp.1247-61, 2017.

C. E. Zielinski, F. Mele, D. Aschenbrenner, D. Jarrossay, F. Ronchi et al., Pathogen-induced human TH17 cells produce IFN-gamma or IL-10 and are regulated by IL-1beta, Nature, vol.484, pp.514-522, 2012.

Y. J. Peng, C. Y. Wang, Y. H. Lin, G. J. Lin, S. H. Huang et al., Interleukin 26 suppresses receptor activator of nuclear factor kappaB ligand induced osteoclastogenesis via down-regulation of nuclear factor of activated T-cells, cytoplasmic 1 and nuclear factor kappaB activity, Rheumatology, vol.55, pp.2074-83, 2016.

P. Fitzgerald-bocarsly, J. Dai, and S. Singh, Plasmacytoid dendritic cells and type I IFN: 50 years of convergent history, Cytokine Growth Factor Rev, vol.19, pp.3-19, 2008.

. Robinson, . We, B. Mcdougall, D. Tran, and M. E. Selsted, Anti-HIV-1 activity of indolicidin, an antimicrobial peptide from neutrophils, J Leukoc Biol, vol.63, pp.94-100, 1998.

A. Belaid, M. Aouni, R. Khelifa, A. Trabelsi, M. Jemmali et al., In vitro antiviral activity of dermaseptins against herpes simplex virus type 1, J Med Virol, vol.66, pp.229-263, 2002.

H. Tamamura, T. Ishihara, A. Otaka, T. Murakami, T. Ibuka et al., Analysis of the interaction of an anti-HIV peptide, T22 ([Tyr5, 12, Lys7]-polyphemusin II), with gp120 and CD4 by surface plasmon resonance, Biochim Biophys Acta, vol.1298, pp.37-44, 1996.

B. H. Song, G. C. Lee, M. S. Moon, Y. H. Cho, and C. H. Lee, Human cytomegalovirus binding to heparan sulfate proteoglycans on the cell surface and/or entry stimulates the expression of human leukocyte antigen class I, J Gen Virol, pp.2405-2418, 2001.

B. Yasin, W. Wang, M. Pang, N. Cheshenko, T. Hong et al., Theta defensins protect cells from infection by herpes simplex virus by inhibiting viral adhesion and entry, J Virol, vol.78, pp.5147-56, 2004.

W. L. Wang, W. Liu, H. Y. Gong, J. R. Hong, C. C. Lin et al., Activation of cytokine expression occurs through the TNFalpha/NF-kappaB-mediated pathway in birnavirus-infected cells, Fish Shellfish Immunol, vol.31, pp.10-21, 2011.

J. Cruz, C. Ortiz, F. Guzman, R. Fernandez-lafuente, and R. Torres, Antimicrobial peptides: promising compounds against pathogenic microorganisms, Curr Med Chem, vol.21, pp.2299-321, 2014.

E. M. Kosciuczuk, P. Lisowski, J. Jarczak, N. Strzalkowska, A. Jozwik et al., Cathelicidins: family of antimicrobial peptides. A review, Mol Biol Rep, vol.39, pp.10957-70, 2012.

G. W. Agak, S. Kao, K. Ouyang, M. Qin, D. Moon et al., Phenotype and antimicrobial activity of Th17 cells induced by propionibacterium acnes strains associated with healthy and acne skin, J Invest Dermatol, vol.138, pp.316-340, 2017.

B. Bechinger, The structure, dynamics and orientation of antimicrobial peptides in membranes by multidimensional solidstate NMR spectroscopy, Biochim Biophys Acta, vol.1462, pp.157-83, 1999.

A. A. Bahar and D. Ren, Antimicrobial peptides, Pharmaceuticals, vol.6, pp.1543-75, 2013.

J. T. Crowl, E. E. Gray, K. Pestal, H. E. Volkman, and D. B. Stetson, Intracellular nucleic acid detection in autoimmunity, Annu Rev Immunol, vol.35, pp.313-349, 2017.

M. S. Silverberg, J. H. Cho, J. D. Rioux, D. P. Mcgovern, J. Wu et al., Ulcerative colitis-risk loci on chromosomes 1p36 and 12q15 found by genome-wide association study, Nat Genet, vol.41, pp.216-236, 2009.

M. Fujii, A. Nishida, H. Imaeda, M. Ohno, K. Nishino et al., Expression of Interleukin-26 is upregulated in inflammatory bowel disease, World J Gastroenterol, vol.23, pp.5519-5548, 2017.

S. Kugathasan, R. N. Baldassano, J. P. Bradfield, P. M. Sleiman, M. Imielinski et al., Loci on 20q13 and 21q22 are associated with pediatric-onset inflammatory bowel disease, Nat Genet, vol.40, pp.1211-1216, 2008.

S. Bogaert, D. Laukens, H. Peeters, L. Melis, K. Olievier et al., Differential mucosal expression of Th17-related genes between the inflamed colon and ileum of patients with inflammatory bowel disease, BMC Immunol, vol.11, p.61, 2010.

G. Caiazzo, D. Caprio, R. Lembo, S. Raimondo, A. Scala et al., IL-26 in allergic contact dermatitis: resource in a state of readiness, Exp Dermatol, vol.27, pp.681-685, 2018.

P. Pinero, O. Juanola, E. Caparros, P. Zapater, P. Gimenez et al., Toll-like receptor polymorphisms compromise the inflammatory response against bacterial antigen translocation in cirrhosis, vol.7, p.46425, 2017.

A. Goris, S. Heggarty, M. G. Marrosu, C. Graham, A. Billiau et al., Linkage disequilibrium analysis of chromosome 12q14-15 in multiple sclerosis: delineation of a 118-kb interval around interferon-gamma (IFNG) that is involved in male versus female differential susceptibility, Genes Immun, vol.3, pp.470-476, 2002.

P. Pinero, O. Juanola, A. Gutierrez, P. Zapater, P. Gimenez et al., IL26 modulates cytokine response and anti-TNF consumption in Crohn's disease patients with bacterial DNA, J Mol Med, vol.95, pp.1227-1263, 2017.

C. Pagnoux, Updates in ANCA-associated vasculitis, Eur J Rheumatol, vol.3, pp.122-155, 2016.

P. A. Jarrot and G. Kaplanski, Pathogenesis of ANCA-associated vasculitis: an update, Autoimmun Rev, vol.15, pp.704-717, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01459842

J. C. Jennette and R. J. Falk, Pathogenesis of antineutrophil cytoplasmic autoantibody-mediated disease, Nat Rev Rheumatol, vol.10, pp.463-73, 2014.

G. Lopalco, O. M. Lucherini, A. Lopalco, V. Venerito, C. Fabiani et al., Cytokine signatures in mucocutaneous and ocular behcet's disease. Front Immunol, vol.8, p.200, 2017.

G. Esendagli, A. T. Kurne, G. Sayat, A. K. Kilic, D. Guc et al., Evaluation of Th17-related cytokines and receptors in multiple sclerosis patients under interferon beta-1 therapy, J Neuroimmunol, vol.255, pp.81-85, 2013.

N. Muls, Z. Nasr, H. A. Dang, C. Sindic, and V. Van-pesch, IL-22, GM-CSF and IL-17 in peripheral CD4+ T cell subpopulations during multiple sclerosis relapses and remission. Impact of corticosteroid therapy, PLoS ONE, vol.12, 2017.

S. Ueha, F. H. Shand, and K. Matsushima, Cellular and molecular mechanisms of chronic inflammation-associated organ fibrosis, Front Immunol, vol.3, p.71, 2012.

E. Sziksz, D. Pap, R. Lippai, N. J. Beres, A. Fekete et al., Fibrosis related inflammatory mediators: role of the IL-10 cytokine family, Mediat Inflamm, p.764641, 2015.

Y. Zhang, J. Li, C. Wang, and L. Zhang, Association between the interaction of key genes involved in effector T-cell pathways and susceptibility to develop allergic rhinitis: a population-based case-control association study, PLoS ONE, vol.10, 2015.

K. Wolk, H. Mitsui, K. Witte, S. Gellrich, N. Gulati et al., Deficient cutaneous antibacterial competence in cutaneous T-cell lymphomas: role of Th2-mediated biased Th17 function, Clin Cancer Res, vol.20, pp.5507-5523, 2014.

, Conflict of Interest Statement: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest

. Copyright-©-2019-larochette, . Miot, . Poli, . Beaumont, . Roingeard et al., This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited