Enhanced oxidative stress in smoking and ex-smoking severe asthma in the U-BIOPRED cohort

Rosalia Emma, Aruna T Bansal, Johan Kolmert, Craig E Wheelock, Swen-Erik Dahlen, Matthew J Loza, Bertrand De Meulder, Diane Lefaudeux, Charles Auffray, Barbro Dahlen, Per S Bakke, Pascal Chanez, Stephen J Fowler, Ildiko Horvath, Paolo Montuschi, Norbert Krug, Marek Sanak, Thomas Sandstrom, Dominick E Shaw, Louise J Fleming, Ratko Djukanovic, Peter H Howarth, Florian Singer, Ana R Sousa, Peter J Sterk, Julie Corfield, Ioannis Pandis, Kian F Chung, Ian M Adcock, René Lutter, Lorena Fabbella, Massimo Caruso, U-BIOPRED Study Group, Rosalia Emma, Aruna T Bansal, Johan Kolmert, Craig E Wheelock, Swen-Erik Dahlen, Matthew J Loza, Bertrand De Meulder, Diane Lefaudeux, Charles Auffray, Barbro Dahlen, Per S Bakke, Pascal Chanez, Stephen J Fowler, Ildiko Horvath, Paolo Montuschi, Norbert Krug, Marek Sanak, Thomas Sandstrom, Dominick E Shaw, Louise J Fleming, Ratko Djukanovic, Peter H Howarth, Florian Singer, Ana R Sousa, Peter J Sterk, Julie Corfield, Ioannis Pandis, Kian F Chung, Ian M Adcock, René Lutter, Lorena Fabbella, Massimo Caruso, U-BIOPRED Study Group

Abstract

Oxidative stress is believed to be a major driver of inflammation in smoking asthmatics. The U-BIOPRED project recruited a cohort of Severe Asthma smokers/ex-smokers (SAs/ex) and non-smokers (SAn) with extensive clinical and biomarker information enabling characterization of these subjects. We investigated oxidative stress in severe asthma subjects by analysing urinary 8-iso-PGF2α and the mRNA-expression of the main pro-oxidant (NOX2; NOSs) and anti-oxidant (SODs; CAT; GPX1) enzymes in the airways of SAs/ex and SAn. All the severe asthma U-BIOPRED subjects were further divided into current smokers with severe asthma (CSA), ex-smokers with severe asthma (ESA) and non-smokers with severe asthma (NSA) to deepen the effect of active smoking. Clinical data, urine and sputum were obtained from severe asthma subjects. A bronchoscopy to obtain bronchial biopsy and brushing was performed in a subset of subjects. The main clinical data were analysed for each subset of subjects (urine-8-iso-PGF2α; IS-transcriptomics; BB-transcriptomics; BBr-transcriptomics). Urinary 8-iso-PGF2α was quantified using mass spectrometry. Sputum, bronchial biopsy and bronchial brushing were processed for mRNA expression microarray analysis. Urinary 8-iso-PGF2α was increased in SAs/ex, median (IQR) = 31.7 (24.5-44.7) ng/mmol creatinine, compared to SAn, median (IQR) = 26.6 (19.6-36.6) ng/mmol creatinine (p< 0.001), and in CSA, median (IQR) = 34.25 (24.4-47.7), vs. ESA, median (IQR) = 29.4 (22.3-40.5), and NSA, median (IQR) = 26.5 (19.6-16.6) ng/mmol creatinine (p = 0.004). Sputum mRNA expression of NOX2 was increased in SAs/ex compared to SAn (probe sets 203922_PM_s_at fold-change = 1.05 p = 0.006; 203923_PM_s_at fold-change = 1.06, p = 0.003; 233538_PM_s_at fold-change = 1.06, p = 0.014). The mRNA expression of antioxidant enzymes were similar between the two severe asthma cohorts in all airway samples. NOS2 mRNA expression was decreased in bronchial brushing of SAs/ex compared to SAn (fold-change = -1.10; p = 0.029). NOS2 mRNA expression in bronchial brushing correlated with FeNO (Kendal's Tau = 0.535; p< 0.001). From clinical and inflammatory analysis, FeNO was lower in CSA than in ESA in all the analysed subject subsets (p< 0.01) indicating an effect of active smoking. Results about FeNO suggest its clinical limitation, as inflammation biomarker, in severe asthma active smokers. These data provide evidence of greater systemic oxidative stress in severe asthma smokers as reflected by a significant changes of NOX2 mRNA expression in the airways, together with elevated urinary 8-iso-PGF2α in the smokers/ex-smokers group. Trial registration ClinicalTrials.gov-Identifier: NCT01976767.

Conflict of interest statement

M. Caruso, R. Emma, A.T. Bansal, C.E. Wheelock, M.J. Loza, L. Fabbella, T. Sandstrom, F. Singer, A.R. Sousa, D.E. Shaw, R. Lutter, S.J. Fowler, I. Horvath, I. Pandis, P. Montuschi declare non conflict of interest. J. Kolmert, B. De Meulder, D. Lefaudeux, C. Auffray, P.J. Sterk, N. Krug, M. Sanak, J. Corfield declare grants from IMI (Innovative Medicine Initiatives) during the conduct of the study. S.E. Dahlen declares financial supporting by Swedish Research Academic Foundations, RSPS Incentive, AZ, Teva and GSK consultancies. B. Dahlen declares consultancies for TEVA. P. Chanez reports grants and personal fees from Almirall, BI, Centocor, GSK, MSD, AstraZeneca, Novartis, Teva, Chiesi, Schering Plough, outside the submitted work. P.S. Bakke reports personal fees from GlaxoSmithKline, Boehringer-Ingelheim, AstraZeneca, Orionpharma, Mundipharma, outside the submitted work. R. Djukanovic reports receiving fees for lectures at symposia organised by Novartis and TEVA and consultation for these two companies as member of advisory boards. He is a co-founder and current consultant, and has shares in Synairgen, a University of Southampton spin out company. P.H. Howarth reports fees from GSK, outside the submitted work. L. Fleming reports personal fees and other from Vectura, Novartis, Boehringer ingelheim, outside the submitted work. I. Adcock reports grants from EU-IMI, during the conduct of the study; grants from MRC, BHF, Dunhill Medical Trust, personal fees from Chiesi, GSK, Boehringer Ingelheim, outside the submitted work. K.F. Chung reports personal fees from Advisory Board membership, grants for research, and personal fees from payments for lectures, outside the submitted work. ATB is employed by Acclarogen Ltd., MJL by Janssen Research & Development, LLC., ARS by GSK and JC by Astra Zeneca R&D and Areteva R&D. There are no patents, products in development or marketed products to declare. This does not alter our adherence to all the PLOS ONE policies on sharing data and materials, as detailed online in the guide for authors.

Figures

Fig 1
Fig 1
(A) Comparison of 8-iso-PGF2α in urine between SAn and SAs/ex. (B) Comparison of urinary 8-iso-PGF2α between severe asthma smoking subgroups. CSA: current smokers with severe asthma; ESA: ex-smokers with severe asthma; NSA: non smokers with severe asthma; SAn: Severe Asthma non smokers; SAs/ex: Severe Asthma smokers/ex-smokers.
Fig 2. Scatter plot of the relationship…
Fig 2. Scatter plot of the relationship between NOS2 expression in bronchial brushing and FeNO.
NOS2 expression in bronchial brushing were strongly correlated to FeNO. Kendall’s Tau = 0.535, p< 0.001, (n = 62). FeNO (ppb) were log2-transformed. SAn: Severe Asthma non smokers; SAs/ex: Severe Asthma smokers/ex-smokers.

References

    1. Holgate ST, Polosa R. Treatment strategies for allergy and asthma. Nat Rev Immunol. 2008;8: 218–30. 10.1038/nri2262
    1. Wenzel SE. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med. 2012;18: 716–25. 10.1038/nm.2678
    1. Chung KF. Defining phenotypes in asthma: a step towards personalized medicine. Drugs. 2014;74: 719–28. 10.1007/s40265-014-0213-9
    1. Chung KF, Wenzel SE, Brozek JL, Bush A, Castro M, Sterk PJ, et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur Respir J. 2014;43: 343–373. 10.1183/09031936.00202013
    1. Polosa R, Thomson NC. Smoking and asthma: dangerous liaisons. Eur Respir J. 2013;41: 716–26. 10.1183/09031936.00073312
    1. Kelly FJ, Mudway I, Blomberg A, Frew A, Sandström T. Altered lung antioxidant status in patients with mild asthma. Lancet (London, England). 1999;354: 482–3.
    1. Zuo L, Otenbaker NP, Rose BA, Salisbury KS. Molecular mechanisms of reactive oxygen species-related pulmonary inflammation and asthma. Mol Immunol. 2013;56: 57–63. 10.1016/j.molimm.2013.04.002
    1. Holguin F. Oxidative stress in airway diseases. Ann Am Thorac Soc. 2013;10 Suppl: S150–7.
    1. Bogdan C. Nitric oxide synthase in innate and adaptive immunity: an update. Trends Immunol. 2015;36: 161–178. 10.1016/j.it.2015.01.003
    1. Rahman I, Biswas SK, Kode A. Oxidant and antioxidant balance in the airways and airway diseases. Eur J Pharmacol. 2006;533: 222–39. 10.1016/j.ejphar.2005.12.087
    1. Wood LG, Gibson PG, Garg ML. Biomarkers of lipid peroxidation, airway inflammation and asthma. Eur Respir J. 2003;21: 177–86.
    1. Valavanidis A, Vlachogianni T, Fiotakis K. Tobacco smoke: involvement of reactive oxygen species and stable free radicals in mechanisms of oxidative damage, carcinogenesis and synergistic effects with other respirable particles. Int J Environ Res Public Health. 2009;6: 445–62. 10.3390/ijerph6020445
    1. Shaw DE, Sousa AR, Fowler SJ, Fleming LJ, Roberts G, Corfield J, et al. Clinical and inflammatory characteristics of the European U-BIOPRED adult severe asthma cohort. Eur Respir J. 2015;
    1. Bousquet J. Global initiative for asthma (GINA) and its objectives. Clin Exp Allergy. 2000;30 Suppl 1: 2–5.
    1. Athey BD, Braxenthaler M, Haas M, Guo Y. tranSMART: An Open Source and Community-Driven Informatics and Data Sharing Platform for Clinical and Translational Research. AMIA Jt Summits Transl Sci proceedings AMIA Jt Summits Transl Sci. 2013;2013: 6–8.
    1. Balgoma D, Larsson J, Rokach J, Lawson JA, Daham K, Dahlén B, et al. Quantification of lipid mediator metabolites in human urine from asthma patients by electrospray ionization mass spectrometry: controlling matrix effects. Anal Chem. American Chemical Society; 2013;85: 7866–74.
    1. Liu H, Bebu I, Li X. Microarray probes and probe sets. Front Biosci (Elite Ed). 2010;2: 325–38.
    1. Mak JCW, Ho SP, Ho ASS, Law BKW, Cheung AHK, Ho JCM, et al. Sustained elevation of systemic oxidative stress and inflammation in exacerbation and remission of asthma. ISRN Allergy. 2013;2013: 561831 10.1155/2013/561831
    1. Papaioannou AI, Koutsokera A, Tanou K, Kiropoulos TS, Tsilioni I, Oikonomidi S, et al. The acute effect of smoking in healthy and asthmatic smokers. Eur J Clin Invest. 2010;40: 103–109. 10.1111/j.1365-2362.2009.02221.x
    1. MONTUSCHI P, CORRADI M, CIABATTONI G, NIGHTINGALE J, KHARITONOV SA, BARNES PJ. Increased 8-Isoprostane, a Marker of Oxidative Stress, in Exhaled Condensate of Asthma Patients. Am J Respir Crit Care Med. 1999;160: 216–220. 10.1164/ajrccm.160.1.9809140
    1. Rahman I, Adcock IM. Oxidative stress and redox regulation of lung inflammation in COPD. Eur Respir J. 2006;28: 219–42. 10.1183/09031936.06.00053805
    1. Lee I-T, Yang C-M. Role of NADPH oxidase/ROS in pro-inflammatory mediators-induced airway and pulmonary diseases. Biochem Pharmacol. 2012;84: 581–90. 10.1016/j.bcp.2012.05.005
    1. Bedard K, Krause K-H. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87: 245–313. 10.1152/physrev.00044.2005
    1. Cheng S-E, Lee I-T, Lin C-C, Kou YR, Yang C-M. Cigarette smoke particle-phase extract induces HO-1 expression in human tracheal smooth muscle cells: role of the c-Src/NADPH oxidase/MAPK/Nrf2 signaling pathway. Free Radic Biol Med. 2010;48: 1410–22. 10.1016/j.freeradbiomed.2010.02.026
    1. Yao H, Yang S-R, Kode A, Rajendrasozhan S, Caito S, Adenuga D, et al. Redox regulation of lung inflammation: role of NADPH oxidase and NF-kappaB signalling. Biochem Soc Trans. 2007;35: 1151–5. 10.1042/BST0351151
    1. Bruijnzeel PLB, Uddin M, Koenderman L. Targeting neutrophilic inflammation in severe neutrophilic asthma: can we target the disease-relevant neutrophil phenotype? J Leukoc Biol. 2015;98: 549–56. 10.1189/jlb.3VMR1214-600RR
    1. Xue J, Schmidt S V, Sander J, Draffehn A, Krebs W, Quester I, et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 2014;40: 274–88. 10.1016/j.immuni.2014.01.006
    1. Batra J, Chatterjee R, Ghosh B. Inducible nitric oxide synthase (iNOS): role in asthma pathogenesis. Indian J Biochem Biophys. 2007;44: 303–9.
    1. Mattila JT, Thomas AC. Nitric oxide synthase: non-canonical expression patterns. Front Immunol. 2014;5: 478 10.3389/fimmu.2014.00478
    1. Voraphani N, Gladwin MT, Contreras AU, Kaminski N, Tedrow JR, Milosevic J, et al. An airway epithelial iNOS-DUOX2-thyroid peroxidase metabolome drives Th1/Th2 nitrative stress in human severe asthma. Mucosal Immunol. 2014;7: 1175–85. 10.1038/mi.2014.6
    1. Yamamoto M, Tochino Y, Chibana K, Trudeau JB, Holguin F, Wenzel SE. Nitric oxide and related enzymes in asthma: relation to severity, enzyme function and inflammation. Clin Exp Allergy. 2012;42: 760–8. 10.1111/j.1365-2222.2011.03860.x
    1. Roos AB, Mori M, Grönneberg R, Österlund C, Claesson H-E, Wahlström J, et al. Elevated exhaled nitric oxide in allergen-provoked asthma is associated with airway epithelial iNOS. PLoS One. 2014;9: e90018 10.1371/journal.pone.0090018
    1. Lane C, Knight D, Burgess S, Franklin P, Horak F, Legg J, et al. Epithelial inducible nitric oxide synthase activity is the major determinant of nitric oxide concentration in exhaled breath. Thorax. 2004;59: 757–60. 10.1136/thx.2003.014894
    1. Kelleher ZT, Matsumoto A, Stamler JS, Marshall HE. NOS2 regulation of NF-kappaB by S-nitrosylation of p65. J Biol Chem. 2007;282: 30667–72. 10.1074/jbc.M705929200
    1. Horváth I, Donnelly LE, Kiss A, Balint B, Kharitonov SA, Barnes PJ. Exhaled nitric oxide and hydrogen peroxide concentrations in asthmatic smokers. Respiration. 2004;71: 463–468. 10.1159/000080630
    1. Thomson NC, Chaudhuri R, Heaney LG, Bucknall C, Niven RM, Brightling CE, et al. Clinical outcomes and inflammatory biomarkers in current smokers and exsmokers with severe asthma. J Allergy Clin Immunol. 2013;131: 1008–16. 10.1016/j.jaci.2012.12.1574
    1. Kinnula VL, Crapo JD. Superoxide dismutases in the lung and human lung diseases. Am J Respir Crit Care Med. 2003;167: 1600–19. 10.1164/rccm.200212-1479SO
    1. Comhair SAA, Ricci KS, Arroliga M, Lara AR, Dweik RA, Song W, et al. Correlation of systemic superoxide dismutase deficiency to airflow obstruction in asthma. Am J Respir Crit Care Med. 2005;172: 306–13. 10.1164/rccm.200502-180OC
    1. Comhair SAA, Xu W, Ghosh S, Thunnissen FBJM, Almasan A, Calhoun WJ, et al. Superoxide dismutase inactivation in pathophysiology of asthmatic airway remodeling and reactivity. Am J Pathol. 2005;166: 663–74. 10.1016/S0002-9440(10)62288-2
    1. Marklund SL. Regulation by cytokines of extracellular superoxide dismutase and other superoxide dismutase isoenzymes in fibroblasts. J Biol Chem. 1992;267: 6696–701.
    1. Gilks CB, Price K, Wright JL, Churg A. Antioxidant gene expression in rat lung after exposure to cigarette smoke. Am J Pathol. 1998;152: 269–78.
    1. York GK, Peirce TH, Schwartz LW, Cross CE. Stimulation by cigarette smoke of glutathione peroxidase system enzyme activities in rat lung. Arch Environ Health. 31: 286–90.
    1. Jenifer HD, Bhola S, Kalburgi V, Warad S, Kokatnur VM. The influence of cigarette smoking on blood and salivary super oxide dismutase enzyme levels among smokers and nonsmokers—A cross sectional study. J Tradit Complement Med. 2015;5: 100–105. 10.1016/j.jtcme.2014.11.003
    1. Hoffmann RF, Zarrintan S, Brandenburg SM, Kol A, de Bruin HG, Jafari S, et al. Prolonged cigarette smoke exposure alters mitochondrial structure and function in airway epithelial cells. Respir Res. 2013;14: 97 10.1186/1465-9921-14-97
    1. Betsuyaku T, Fuke S, Inomata T, Kaga K, Morikawa T, Odajima N, et al. Bronchiolar epithelial catalase is diminished in smokers with mild COPD. Eur Respir J. 2013;42: 42–53. 10.1183/09031936.00058912

Source: PubMed

3
購読する