136
Views
16
CrossRef citations to date
0
Altmetric
Original Research

Sputum IL-26 Is Overexpressed in Severe Asthma and Induces Proinflammatory Cytokine Production and Th17 Cell Generation: A Case–Control Study of Women

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon &
Pages 95-107 | Published online: 03 Feb 2020

References

  • Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol. 2015;16(1):45–56. doi:10.1038/ni.304925521684
  • Forno E, Wang T, Yan Q, et al. A multi-omics approach to identify genes associated with childhood asthma risk and morbidity. Am J Respir Cell Mol Biol. 2017;57(4):439–447. doi:10.1165/rcmb.2017-0002OC28574721
  • Silva MJ, de Santana MBR, Tosta BR, et al. Variants in the IL17 pathway genes are associated with atopic asthma and atopy makers in a South American population. Allergy Asthma Clin Immunol. 2019;15:28. doi:10.1186/s13223-019-0340-731168303
  • Whitehead GS, Kang HS, Thomas SY, et al. Therapeutic suppression of pulmonary neutrophilia and allergic airway hyperresponsiveness by a RORγt inverse agonist. JCI Insight. 2019. doi:10.1172/jci.insight.125528
  • Charrad R, Berraïes A, Hamdi B, et al. Anti inflammatory activity of IL-37 in asthmatic children: correlation with inflammatory cytokines TNF-α, IL-β, IL-6 and IL-17A. Immunobiology. 2016;221(2):182–187. doi:10.1016/j.imbio.2015.09.00926454413
  • Corren J, Parnes JR, Wang L, et al. Tezepelumab in adults with uncontrolled asthma. N Engl J Med. 2017;377:936–946. doi:10.1056/NEJMoa170406428877011
  • Donnelly RP, Sheikh F, Dickensheets H, et al. Interleukin-26: an il-10-related cytokine produced by th17 cells. Cytokine Growth Factor Rev. 2010;21:393–401. doi:10.1016/j.cytogfr.2010.09.00120947410
  • Konradsen JR, Nordlund B, Levänen B, et al. The cytokine interleukin-26 as a biomarker in pediatric asthma. Respir Res. 2016;17:32. doi:10.1186/s12931-016-0351-627029915
  • Ouyang W, Rutz S, Crellin NK, et al. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu Rev Immunol. 2011;29:71–109. doi:10.1146/annurev-immunol-031210-10131221166540
  • Cella M, Fuchs A, Vermi W, et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature. 2009;457(7230):722–725. doi:10.1038/nature0753718978771
  • Fujii M, Nishida A, Imaeda H, et al. Expression of Interleukin-26 is upregulated in inflammatory bowel disease. World J Gastroenterol. 2017;23(30):5519–5529. doi:10.3748/wjg.v23.i30.551928852311
  • Che KF, Tengvall S, Levänen B, et al. Interleukin-26 in antibacterial host defense of human lungs: effects on neutrophil mobilization. Am J Respir Crit Care Med. 2014;190(9):1022–1031. doi:10.1164/rccm.201404-0689OC25291379
  • Hor S, Pirzer H, Dumoutier L, et al. The T-cell lymphokine interleukin-26 targets epithelial cells through the interleukin-20 receptor 1 and interleukin-10 receptor 2 chains. J Biol Chem. 2004;279:33343–33351. doi:10.1074/jbc.M40500020015178681
  • Bals R, Hiemstra PS. Innate immunity in the lung: how epithelial cells fight against respiratory pathogens. Eur Respir J. 2004;23:327–333. doi:10.1183/09031936.03.0009880314979512
  • Larochette V, Miot C, Poli C, et al. IL-26, a cytokine with roles in extracellular DNA-induced inflammation and microbial defense. Front Immunol. 2019;10:204. doi:10.3389/fimmu.2019.0020430809226
  • Griffiths KL, Khader SA. Bringing in the cavalry: IL-26 mediates neutrophil recruitment to the lungs. Am J Respir Crit Care Med. 2014;190:1079–1080. doi:10.1164/rccm.201410-1870ED25398102
  • Meller S, Di Domizio J, Voo KS, et al. T(H)17 cells promote microbial killing and innate immune sensing of DNA via interleukin 26. Nat Immunol. 2015;16:970–979. doi:10.1038/ni.321126168081
  • Che KF, Kaarteenaho R, Lappi-Blanco E, et al. Interleukin-26 production in human primary bronchial epithelial cells in response to viral stimulation: modulation by Th17 cytokines. Mol Med. 2017;23:247–257. doi:10.2119/molmed.2016.0006428853490
  • Che KF, Tufvesson E, Tengvall S, et al. The neutrophil-mobilizing cytokine interleukin-26 in the airways of long-term tobacco smokers. Clin Sci (Lond). 2018;132:959–983. doi:10.1042/CS2018005729780024
  • Kortekaas Krohn I, Shikhagaie MM, Golebski K, et al. Emerging roles of innate lymphoid cells in inflammatory diseases: clinical implications. Allergy. 2017;73:837–850. doi:10.1111/all.1334029069535
  • Rajput C, Han M, Bentley JK, et al. Enterovirus D68 infection induces IL-17-dependent neutrophilic airway inflammation and hyperresponsiveness. JCI Insight. 2018;23:3.
  • Hekking PP, Loza MJ, Pavlidis S, et al. Study group. Pathway discovery using transcriptomic profiles in adult-onset severe asthma. J Allergy Clin Immunol. 2018;141(4):1280–1290. doi:10.1016/j.jaci.2017.06.03728756296
  • Chung KF, Wenzel SE, Brozek JL, et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J. 2014;43(2):343–373. doi:10.1183/09031936.0020201324337046
  • Brusasco V, Crapo R, Viegi G. Coming together: the ATS/ERS consensus on clinical pulmonary function testing. Eur Respir J. 2005;26(1):1–2. doi:10.1183/09031936.05.0003420515994380
  • Fahy JV, Liu J, Wong H, Boushey HA. Cellular and biochemical analysis of induced sputum from asthmatic and from healthy subjects. Am Rev Respir Dis. 1993;147(5):1126–1131. doi:10.1164/ajrccm/147.5.11268484620
  • Rosewich M, Zissler UM, Kheiri T, et al. Airway inflammation in children and adolescents with bronchiolitis obliterans. Cytokine. 2015;73(1):156–162. doi:10.1016/j.cyto.2014.10.02625748838
  • Gunawardhana LP, Gibson PG, Simpson JL, et al. Activity and expression of histone acetylases and deacetylases in inflammatory phenotypes of asthma. Clin Exp Allergy. 2014;44:47–57. doi:10.1111/cea.1216824355018
  • Sikkeland LI, Kongerud J, Stangeland AM, et al. Macrophage enrichment from induced sputum. Thorax. 2007;62(6):558–559. doi:10.1136/thx.2006.07354417536036
  • Kaabachi W, Bouali E, Berraïes A, et al. Interleukin-26 is overexpressed in Behçet’s disease and enhances Th17 related -cytokines. Immunol Lett. 2017;190:177–184. doi:10.1016/j.imlet.2017.08.00828811236
  • Corren J. New Targeted Therapies for uncontrolled asthma. J Allergy Clin Immunol Pract. 2019;7(5):1394–1403. doi:10.1016/j.jaip.2019.03.02231076057
  • Pavord ID. Oral corticosteroid-dependent asthma. Curr Opin Pulm Med. 2019;25:51–58. doi:10.1097/MCP.000000000000054130461530
  • Fahy JV. Type 2 inflammation in asthma–present in most, absent in many. Nat Rev Immunol. 2015;15:57–65. doi:10.1038/nri378625534623
  • Busse WW, Holgate S, Kerwin E, et al. Randomized, 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. 2013;188:1294–1302. doi:10.1164/rccm.201212-2318OC24200404
  • Corvaisier M, Delneste Y, Jeanvoine H, et al. IL-26 is overexpressed in rheumatoid arthritis and induces proinflammatory cytokine production and Th17 cell generation. PLoS Biol. 2012;10(9):e1001395. doi:10.1371/journal.pbio.100139523055831
  • Tufvesson E, Jogdand P, Che KF, et al. Enhanced local production of IL-26 in uncontrolled compared with controlled adult asthma. J Allergy Clin Immunol. 2019;144(4):1134–1136.e10. doi:10.1016/j.jaci.2019.06.03531288043
  • Che KF, Sun J, Linden A. Pharmacological modulation of endotoxin-induced release of IL-26 in human primary lung fibroblasts. Front Pharmacol. 2019;10:956. doi:10.3389/fphar.2019.0095631543817
  • Veldhoen M, Hocking RJ, Atkins CJ, et al. TGFβ in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24:179–189. doi:10.1016/j.immuni.2006.01.00116473830
  • Mangan PR, Harrington LE, O’Quinn DB, et al. Transforming growth factor-b induces development of the TH17 lineage. Nature. 2006;441:231–234. doi:10.1038/nature0475416648837
  • Cho ML, Kang JW, Moon YM, et al. STAT3 and NF-kB signal pathway is required for IL-23-mediated IL-17 production in spontaneous arthritis animal model IL-1 receptor antagonist-deficient mice. J Immunol. 2006;176:5652–5661. doi:10.4049/jimmunol.176.9.565216622035
  • Bao A, Che KF, Bozinovski S, et al. Recombinant human IL-26 facilitates the innate immune response to endotoxin in the bronchoalveolar space of mice in vivo. PLoS One. 2017;12:e0188909. doi:10.1371/journal.pone.018890929206862
  • Duenas Meza E, Jaramillo CA, Correa E, et al. Virus and mycoplasma pneumoniae prevalence in a selected pediatric population with acute asthma exacerbation. J Asthma. 2016;53(3):253–260. doi:10.3109/02770903.2015.107554826799194
  • Li N, Qiu R, Yang Z, et al. Sputum microbiota in severe asthma patients: relationship to eosinophilic inflammation. Respir Med. 2017;131:192–198. doi:10.1016/j.rmed.2017.08.01628947029
  • Webley WC, Hahn DL. Infection-mediated asthma: etiology, mechanisms and treatment options, with focus on Chlamydia pneumoniae and macrolides. Respir Res. 2017;18(1):98. doi:10.1186/s12931-017-0584-z28526018
  • Gon Y, Hashimoto S. Role of airway epithelial barrier dysfunction in pathogenesis of asthma. Allergol Int. 2018;67(1):12–17. doi:10.1016/j.alit.2017.08.01128941636
  • Wilson NJ, Boniface K, Chan JR, et al. Development cytokine profile and function of human interleukin 17–producing helper T cells. Nat Immunol. 2007;8(9):950–957. doi:10.1038/ni149717676044
  • Chang HS, Lee TH, Jun JA, et al. Neutrophilic inflammation in asthma: mechanisms and therapeutic considerations. Expert Rev Respir Med. 2017;11(1):29–40. doi:10.1080/17476348.2017.126891927918221
  • Berraïes A, Hamdi B, Ammar J, Hamzaoui K, Hamzaoui A. Increased expression of thymic stromal lymphopoietin in induced sputum from asthmatic children. Immunol Lett. 2016;178:85–91. doi:10.1016/j.imlet.2016.08.00427528425
  • Paplińska-Goryca M, Nejman-Gryz P, Proboszcz M, et al. Expression of TSLP and IL-33 receptors on sputum macrophages of asthma patients and healthy subjects. J Asthma. 2018;27:1–10.