IL-17 mRNA in sputum of asthmatic patients: linking T cell driven inflammation and granulocytic influx?

Dominique M A Bullens, Els Truyen, Liesbeth Coteur, Ellen Dilissen, Peter W Hellings, Lieven J Dupont, Jan L Ceuppens, Dominique M A Bullens, Els Truyen, Liesbeth Coteur, Ellen Dilissen, Peter W Hellings, Lieven J Dupont, Jan L Ceuppens

Abstract

Background: The role of Th2 cells (producing interleukin (IL-)4, IL-5 and IL-13) in allergic asthma is well-defined. A distinct proinflammatory T cell lineage has recently been identified, called Th17 cells, producing IL-17A, a cytokine that induces CXCL8 (IL-8) and recruits neutrophils. Neutrophilic infiltration in the airways is prominent in severe asthma exacerbations and may contribute to airway gland hypersecretion, bronchial hyper-reactivity and airway wall remodelling in asthma.

Aim: to study the production of IL-17 in asthmatic airways at the mRNA level, and to correlate this with IL-8 mRNA, neutrophilic inflammation and asthma severity.

Methods: We obtained airway cells by sputum induction from healthy individuals (n = 15) and from asthmatic patients (n = 39). Neutrophils were counted on cytospins and IL-17A and IL-8 mRNA expression was quantified by real-time RT-PCR (n = 11 controls and 33 asthmatics).

Results: Sputum IL-17A and IL-8 mRNA levels are significantly elevated in asthma patients compared to healthy controls. IL-17 mRNA levels are significantly correlated with CD3gamma mRNA levels in asthmatic patients and mRNA levels of IL-17A and IL-8 correlated with each other and with sputum neutrophil counts. High sputum IL-8 and IL-17A mRNA levels were also found in moderate-to-severe (persistent) asthmatics on inhaled steroid treatment.

Conclusion: The data suggest that Th17 cell infiltration in asthmatic airways links T cell activity with neutrophilic inflammation in asthma.

Figures

Figure 1
Figure 1
IL-17A and IL-8 mRNA levels in healthy controls and asthmatics. RNA was isolated from induced sputum of healthy controls (n = 11) and asthmatic patients (n = 33) and real time RT-PCR was performed with β-actin, IL-17A and IL-8 specific primers and with VIC-(β-actin) or FAM-(IL-17A, IL-8) labelled specific probes. Results were quantified by the use of a cDNA plasmid-standard and expressed as the ratio of cDNA copy numbers for IL-17A (A-C) or IL-8 (B-D) divided by the cDNA copy numbers for β-actin multiplied by 104 (A-C) or 101 (B-D). Patients were divided in subgroups according to asthma severity following the reviewed GINA criteria [33] (A, B) and allergic state (allergic state was not known in one patient and two patients with only specific IgE for grass pollen were excluded for the comparison of allergic to non-allergic asthma) (C, D). Open symbols represent patients treated with corticosteroids, closed symbols represent patients without corticosteroid-treatment. Comparisons were performed using the Kruskall-Wallis and Mann-Whitney-U test. The median is indicated with a horizontal line. * = p < 0.05, ** = p < 0.01, *** = p < 0.001 and ns = not significant.
Figure 2
Figure 2
CD3γ mRNA levels in healthy controls and asthmatics, correlations between IL-17A and CD3γ and between IL-17A other cytokine/chemokine mRNA levels. CD3γ, IL-8, IL-17A and IL-5 mRNA levels in induced sputum of asthmatic patients were quantified as explained in figure 1 using IL-8, IL-17A and IL-5 specific primers and VIC-(β-actin) or FAM-(IL-8, IL-17A, IL-5) labeled specific probes. Asthma severity was determined as in figure 1A. Open symbols represent patients treated with corticosteroids, closed symbols represent patients without corticosteroid-treatment. Comparisons were performed using the Kruskall-Wallis test and Dunn's post test (C). Correlations were studied by Spearman non-parametric test (A-B, D). The median is indicated with a horizontal line. * = p < 0.05, ** = p < 0.01, *** = p < 0.001, and ns = not significant.
Figure 3
Figure 3
Sputum neutrophil counts in healthy controls and asthma patients and correlation of IL-17A and IL-8 mRNA levels with the neutrophil count in asthmatics. Cytospins of induced sputum of healthy controls (n = 11) and asthmatic patients (n = 32) were stained with May Grünwald Giemsa. Neutrophils are counted as percentage of the leukocytes. IL-17A and IL-8 mRNA levels in induced sputum of asthmatic patients (n = 27) were determined as explained in figure 1 and 2. Open symbols represent patients treated with corticosteroids, closed symbols represent patients without corticosteroid-treatment. The mean is indicated with a horizontal line. Differences are studied by student t-test (A) and correlation was studied by Spearman non-parametric test (B-C). * = p < 0.05, ** = p < 0.01 and *** = p < 0.001.

References

    1. Busse WW, Lemanske RF. Asthma. N Engl J Med. 2001;344:350–362. doi: 10.1056/NEJM200102013440507.
    1. Kay AB. The role of T lymphocytes in asthma. Chem Immunol Allergy. 2006;91:59–75.
    1. Robinson DS, Hamid Q, Ying S, Tsicopoulos A, Barkans J, Bentley AM, et al. Predominant TH2-like bronchoalveolar T-lymphocyte population in atopic asthma. N Engl J Med. 1992;326:298–304.
    1. Wills-Karp M. Immunologic basis of antigen-induced airway hyperresponsiveness. Annu Rev Immunol. 1999;17:255–281. doi: 10.1146/annurev.immunol.17.1.255.
    1. Heijink IH, van Oosterhout AJ. Targeting T cells for asthma. Curr Opin Pharmacol. 2005;5:227–231. doi: 10.1016/j.coph.2005.04.002.
    1. Kuipers H, Lambrecht BN. The interplay of dendritic cells, Th2 cells and regulatory T cells in asthma. Curr Opin Immunol. 2004;16:702–708. doi: 10.1016/j.coi.2004.09.010.
    1. Hansen G, Berry G, DeKruyff RH, Umetsu DT. Allergen-specific Th1 cells fail to counterbalance Th2 cell-induced airway hyperreactivity but cause severe airway inflammation. J Clin Invest. 1999;103:175–183.
    1. Cui J, Pazdziorko S, Miyashiro JS, Thakker P, Pelker JW, Declercq C, et al. TH1-mediated airway hyperresponsiveness independent of neutrophilic inflammation. J Allergy Clin Immunol. 2005;115:309–315. doi: 10.1016/j.jaci.2004.10.046.
    1. Truyen E, Coteur L, Dilissen E, Overbergh L, Dupont LJ, Ceuppens JL, et al. Evaluation of airway inflammation by quantitative Th1/Th2 cytokine mRNA measurement in sputum of asthma patients. Thorax. 2006;61:202–208. doi: 10.1136/thx.2005.052399.
    1. Park H, Li Z, O Yang X, Chang SH, Nurieva R, Wang Y-H, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. 2005;6:1133–1141. doi: 10.1038/ni1261.
    1. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 2005;6:1123–1132. doi: 10.1038/ni1254.
    1. Kolls JK, Lindén A. Interleukin-17 family members and inflammation. Immunity. 2004;21:467–476. doi: 10.1016/j.immuni.2004.08.018.
    1. Molet S, Hamid Q, Davoine F, Nutku E, Taha R, Pagé N, et al. IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines. J Allergy Clin Immunol. 2001;108:430–438. doi: 10.1067/mai.2001.117929.
    1. Sun Y-c, Zhou Q-t, Yao W-z. Sputum interleukin-17 is increased and associated with airway neutrophilia in patients with severe asthma. Chin Med J. 2005;118:953–956.
    1. Barczyk A, Pierzchala W, Sozanska E. Interleukin-17 in sputum correlates with airway hyperresponsiveness to methacholine. Respir Med. 2003;97:726–733. doi: 10.1053/rmed.2003.1507.
    1. Nakae S, Komiyama Y, Nambu A, Sudo K, Iwase M, Homma I, et al. Antigen-specific T cell sensitization is impaired in IL-17-deficient mice, causing suppression of allergic cellular and humoral responses. Immunity. 2002;17:375–387. doi: 10.1016/S1074-7613(02)00391-6.
    1. Hellings PW, Kasran A, Liu Z, Vandekerckhove P, Wuyts A, Overbergh L, et al. Interleukin-17 orchestrates the granulocyte influx into airways after allergen challenge inhalation in a mouse model of allergic asthma. Am J Respir Cell Mol Biol. 2003;28:42–50. doi: 10.1165/rcmb.4832.
    1. Jatakanon A, Uasuf C, Maziak W, Lim S, Chung KF, Barnes PJ. Neutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med. 1999;160:1532–1539.
    1. Tillie-Leblond I, Gosset P, Tonnel A-B. Review article: Inflammatory events in severe acute asthma. Allergy. 2005;60:23–29. doi: 10.1111/j.1398-9995.2005.00632.x.
    1. Wenzel S, Schwartz LB, Langmack EL, Halliday JL, Trudeau JB, Gibbs RL, 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. 1999;160:1001–1008.
    1. Green RH, Brightling CE, Woltmann G, Parker D, Wardlaw AJ, Pavord ID. Analysis of induced sputum in adults with asthma: identification of subgroup with isolated sputum neutrophilia and poor response to inhaled corticosteroids. Thorax. 2002;57:875–879. doi: 10.1136/thorax.57.10.875.
    1. Woodruff PG, Khashayar R, Lazarus SC, Janson S, Avila P, Boushey HA, et al. Relationship between airway inflammation, hyperresponsiveness, and obstruction in asthma. J Allergy Clin Immunol. 2001;108:753–758. doi: 10.1067/mai.2001.119411.
    1. Barnes PJ. Pharmacology of airway smooth muscle. Am J Respir Crit Care Med. 1998;158:S123–S132.
    1. Cundall M, Sun Y, C M, Trudeau JB, Barnes S, Wenzel SE. Neutrophil-derived matrix metalloproteinase-9 is increased in severe asthma and poorly inhibited by glucocorticoids. J Allergy Clin Immunol. 2003;112:1064–1071. doi: 10.1016/j.jaci.2003.08.013.
    1. Lindén A. Role of interleukin-17 and the neutrophil in asthma. Int Arch Allergy Immunol. 2001;126:179–184. doi: 10.1159/000049511.
    1. Gibson PG, Simpson JL, Saltos N. Heterogeneity of airway inflammation in persistent asthma: evidence of neutrophilic inflammation and increased sputum interleukin-8. Chest. 2001;119:1329–1336. doi: 10.1378/chest.119.5.1329.
    1. Folkard SG, Westwick J, Millar AB. Production of interleukin-8, RANTES and MCP-1 in intrinsic and extrinsic asthma. Eur Respir J. 1997;10:2097–2104. doi: 10.1183/09031936.97.10092097.
    1. Xiao W, Hsu Y-P, Ishizaka A, Kirikae T, Moss RB. Sputum cathelicidin, urokinase plasminogen activation system components and cytokines discriminate cystic fibrosis, COPD and asthma inflammation. Chest. 2005;128:2316–2326. doi: 10.1378/chest.128.4.2316.
    1. Ordonez CL, Shaughnessy TE, Matthay MA, Fahy JV. Increased neutrophil numbers and IL-8 levels in airway secretions in acute severe asthma: clinical and biologic significance. Am J Respir Crit Care Med. 2000;161:1185–1190.
    1. Paggiaro PL, Chanez P, Holz O, Ind PW, Djukanovic R, Maestrelli P, et al. Sputum induction. Eur Respir J. 2002;20(suppl 37):3s–8s. doi: 10.1183/09031936.02.00000302.
    1. Gelder CM, Thomas PS, Yates DH, Adcock IM, Morrison JF, Barnes PJ. Cytokine expression in normal, atopic, and asthmatic subjects using the combination of sputum induction and the polymerase chain reaction. Thorax. 1995;50:1033–1037.
    1. Giulietti A, Overbergh L, Valckx D, Decallonne B, Bouillon R, Mathieu C. An overview of real-time quantitative PCR: applications to quantify cytokine gene expression. Methods. 2001;25:386–401. doi: 10.1006/meth.2001.1261.
    1. Global Initiative for Asthma . Global strategy for Asthma management and Prevention. Bethesda (MD): NIH Publication; 2002.
    1. Dupont LJ, Rochette F, Demedts MG, Verleden GM. Exhaled nitric oxide correlates with airway hyperresponsiveness in steroid-naive patients with mild asthma. Am J Respir Crit Care Med. 1998;157:894–989.
    1. Juniper EF, O'Byrne PM, Guyatt GH, Ferrie PJ, King DR. Development and validation of a questionnaire to measure asthma control. Eur Respir J. 1999;14:902–907. doi: 10.1034/j.1399-3003.1999.14d29.x.
    1. Pizzichini E, Pizzichini MMM, Efthimiadis A, Hargreave FE, Dolovich J. Measurements of inflammatory indices in induced sputum: effects of selection of sputum to minimize salivary contamination. Eur Respir J. 1996;9:1174–1180. doi: 10.1183/09031936.96.09061174.
    1. Vanaudenaerde BM, Dupont LJ, Wuyts WA, Verbeken EK, Meyts I, Bullens DM, et al. The role of interleukin-17 during acute rejection after lung transplantation. Eur Respir J. 2006;27:779–787. doi: 10.1183/09031936.06.00019405.
    1. Kharitonov SA, Yates D, Robbins RA, Logan-Sinclair R, Shinebourne EA, Barnes PJ. Increased nitric oxide in exhaled air of asthmatic patients. The Lancet. 1994;343:133–135. doi: 10.1016/S0140-6736(94)90931-8.
    1. Prause O, Laan M, Lötvall J, Lindén A. Pharmacological modulation of interleukin-17-induced GCP-2-, GRO-α- and interleukin-8 release in human bronchial epithelial cells. Eur J Pharmacol. 2003;462:193–198. doi: 10.1016/S0014-2999(03)01341-4.
    1. Rahman MS, Yang J, Shan LY, Unruh H, Yang X, Halayko AJ, et al. IL-17R activation of human airway smooth muscle cells induces CXCL-8 production via a transcriptional-dependent mechanism. Clin Immunol. 2005;115:268–276. doi: 10.1016/j.clim.2005.01.014.
    1. Wenzel S. Severe asthma in adults. Am J Respir Crit Care Med. 2005;172:149–160. doi: 10.1164/rccm.200409-1181PP.
    1. Fukakusa M, Bergeron C, Tulic MK, Fiset P-O, Dewachi OA, Laviolette M, et al. Oral corticosteroids decrease eosinophil and CC chemokine expression but increase neutrophil, IL-8, and IFN-γ-inducible protein-10 expression in asthmatic airway mucosa. J Allergy Clin Immunol. 2005;115:280–286. doi: 10.1016/j.jaci.2004.10.036.
    1. Cox G. Glucocorticoid treatment inhibits apoptosis in human neutrophils. Separation of survival and activation outcomes. J Immunol. 1995;154:4719–4725.

Source: PubMed

3
S'abonner