Skip to main content
Top
Published in: Respiratory Research 1/2018

Open Access 01-12-2018 | Research

Modelling the asthma phenotype: impact of cigarette smoke exposure

Authors: Maria G. Belvisi, Katie Baker, Nicole Malloy, Kristof Raemdonck, Bilel Dekkak, Michael Pieper, Anthony T. Nials, Mark A. Birrell

Published in: Respiratory Research | Issue 1/2018

Login to get access

Abstract

Background

Asthmatics that are exposed to inhaled pollutants such as cigarette smoke (CS) have increased symptom severity. Approximately 25% of adult asthmatics are thought to be active smokers and many sufferers, especially in the third world, are exposed to high levels of inhaled pollutants. The mechanism by which CS or other airborne pollutants alter the disease phenotype and the effectiveness of treatment in asthma is not known. The aim of this study was to determine the impact of CS exposure on the phenotype and treatment sensitivity of rodent models of allergic asthma.

Methods

Models of allergic asthma were configured that mimicked aspects of the asthma phenotype and the effect of CS exposure investigated. In some experiments, treatment with gold standard asthma therapies was investigated and end-points such as airway cellular burden, late asthmatic response (LAR) and airway hyper-Reactivity (AHR) assessed.

Results

CS co-exposure caused an increase in the LAR but interestingly attenuated the AHR. The effectiveness of LABA, LAMA and glucocorticoid treatment on LAR appeared to be retained in the CS-exposed model system. The eosinophilia or lymphocyte burden was not altered by CS co-exposure, nor did CS appear to alter the effectiveness of glucocorticoid treatment. Steroids, however failed to reduce the neutrophilic inflammation in sensitized mice exposed to CS.

Conclusions

These model data have certain parallels with clinical findings in asthmatics, where CS exposure did not impact the anti-inflammatory efficacy of steroids but attenuated AHR and enhanced symptoms such as the bronchospasm associated with the LAR. These model systems may be utilised to investigate how CS and other airborne pollutants impact the asthma phenotype; providing the opportunity to identify novel targets.
Literature
1.
go back to reference Zmirou D, Gauvin S, Pin I, et al. Traffic related air pollution and incidence of childhood asthma: results of the Vesta case-control study. J Epidemiol Community Health. 2004;58:18–23.CrossRefPubMedPubMedCentral Zmirou D, Gauvin S, Pin I, et al. Traffic related air pollution and incidence of childhood asthma: results of the Vesta case-control study. J Epidemiol Community Health. 2004;58:18–23.CrossRefPubMedPubMedCentral
8.
go back to reference Apostol GG, Jacobs DRJ, Tsai AW, et al. Early life factors contribute to the decrease in lung function between ages 18 and 40: the coronary artery risk development in young adults study. Am J Respir Crit Care Med. 2002;166:166–72.CrossRefPubMed Apostol GG, Jacobs DRJ, Tsai AW, et al. Early life factors contribute to the decrease in lung function between ages 18 and 40: the coronary artery risk development in young adults study. Am J Respir Crit Care Med. 2002;166:166–72.CrossRefPubMed
12.
go back to reference Siroux V, Pin I, Oryszczyn M, et al. Relationships of active smoking to asthma and asthma severity in the EGEA study. Epidemiological study on the genetics and environment of asthma. Eur Respir J. 2000;15:470–7.CrossRefPubMed Siroux V, Pin I, Oryszczyn M, et al. Relationships of active smoking to asthma and asthma severity in the EGEA study. Epidemiological study on the genetics and environment of asthma. Eur Respir J. 2000;15:470–7.CrossRefPubMed
18.
go back to reference Stankus RP, Menon PK, Rando RJ, et al. Cigarette smoke-sensitive asthma: challenge studies. J Allergy Clin Immunol. 1988;82:331–8.CrossRefPubMed Stankus RP, Menon PK, Rando RJ, et al. Cigarette smoke-sensitive asthma: challenge studies. J Allergy Clin Immunol. 1988;82:331–8.CrossRefPubMed
19.
go back to reference Althuis MD, Sexton M, Prybylski D. Cigarette smoking and asthma symptom severity among adult asthmatics. J Asthma. 1999;36:257–64.CrossRefPubMed Althuis MD, Sexton M, Prybylski D. Cigarette smoking and asthma symptom severity among adult asthmatics. J Asthma. 1999;36:257–64.CrossRefPubMed
20.
go back to reference Gallefoss F, Bakke PS. Does smoking affect the outcome of patient education and self-management in asthmatics? Patient Educ Couns. 2003;49:91–7.CrossRefPubMed Gallefoss F, Bakke PS. Does smoking affect the outcome of patient education and self-management in asthmatics? Patient Educ Couns. 2003;49:91–7.CrossRefPubMed
21.
go back to reference Dahms TE, Bolin JF, Slavin RG. Passive smoking. Effects on bronchial asthma. Chest. 1981;80:530–4.CrossRefPubMed Dahms TE, Bolin JF, Slavin RG. Passive smoking. Effects on bronchial asthma. Chest. 1981;80:530–4.CrossRefPubMed
22.
go back to reference Menon PK, Stankus RP, Rando RJ, et al. Asthmatic responses to passive cigarette smoke: persistence of reactivity and effect of medications. J Allergy Clin Immunol. 1991;88:861–9.CrossRefPubMed Menon PK, Stankus RP, Rando RJ, et al. Asthmatic responses to passive cigarette smoke: persistence of reactivity and effect of medications. J Allergy Clin Immunol. 1991;88:861–9.CrossRefPubMed
23.
go back to reference Menon P, Rando RJ, Stankus RP, et al. Passive cigarette smoke-challenge studies: increase in bronchial hyperreactivity. J Allergy Clin Immunol. 1992;89:560–6.CrossRefPubMed Menon P, Rando RJ, Stankus RP, et al. Passive cigarette smoke-challenge studies: increase in bronchial hyperreactivity. J Allergy Clin Immunol. 1992;89:560–6.CrossRefPubMed
24.
go back to reference Chalmers GW, Macleod KJ, Little SA, et al. Influence of cigarette smoking on inhaled corticosteroid treatment in mild asthma. Thorax. 2002;57:226–30.CrossRefPubMedPubMedCentral Chalmers GW, Macleod KJ, Little SA, et al. Influence of cigarette smoking on inhaled corticosteroid treatment in mild asthma. Thorax. 2002;57:226–30.CrossRefPubMedPubMedCentral
32.
go back to reference Underwood S, Foster M, Raeburn D, et al. Time-course of antigen-induced airway inflammation in the Guinea-pig and its relationship to airway hyperresponsiveness. Eur Respir J. 1995;8:2104–13.CrossRefPubMed Underwood S, Foster M, Raeburn D, et al. Time-course of antigen-induced airway inflammation in the Guinea-pig and its relationship to airway hyperresponsiveness. Eur Respir J. 1995;8:2104–13.CrossRefPubMed
33.
go back to reference Belvisi MG, Birrell MA, Khalid S, Wortley MA, Dockry R, Coote J, Holt K, Dubuis E, Kelsall A, Maher SA, Bonvini S, Woodcock A, Smith JA. Neurophenotypes in Airway Diseases. Insights from Translational Cough Studies. Am J Respir Crit Care Med. 2016;193(12):1364–72. https://doi.org/10.1164/rccm.201508-1602OC Belvisi MG, Birrell MA, Khalid S, Wortley MA, Dockry R, Coote J, Holt K, Dubuis E, Kelsall A, Maher SA, Bonvini S, Woodcock A, Smith JA. Neurophenotypes in Airway Diseases. Insights from Translational Cough Studies. Am J Respir Crit Care Med. 2016;193(12):1364–72. https://​doi.​org/​10.​1164/​rccm.​201508-1602OC
34.
go back to reference Eltom S, Stevenson C, Birrell MA. Cigarette smoke exposure as a model of inflammation associated with COPD. Curr Protoc Pharmacol. 2013;5:5.64. Eltom S, Stevenson C, Birrell MA. Cigarette smoke exposure as a model of inflammation associated with COPD. Curr Protoc Pharmacol. 2013;5:5.64.
36.
go back to reference Birrell M a, Battram CH, Woodman P, et al. Dissociation by steroids of eosinophilic inflammation from airway hyperresponsiveness in murine airways. Respir Res. 2003;4:3.CrossRefPubMedPubMedCentral Birrell M a, Battram CH, Woodman P, et al. Dissociation by steroids of eosinophilic inflammation from airway hyperresponsiveness in murine airways. Respir Res. 2003;4:3.CrossRefPubMedPubMedCentral
39.
go back to reference Birrell MA, De Alba J, Catley MC, et al. Liver X receptor agonists increase airway reactivity in a model of asthma via increasing airway smooth muscle growth. J Immunol. 2008;181:4265–71.CrossRefPubMed Birrell MA, De Alba J, Catley MC, et al. Liver X receptor agonists increase airway reactivity in a model of asthma via increasing airway smooth muscle growth. J Immunol. 2008;181:4265–71.CrossRefPubMed
48.
go back to reference Min MG, Song DJ, Miller M, et al. Coexposure to environmental tobacco smoke increases levels of allergen-induced airway remodeling in mice. J Immunol. 2007;178:5321–8.CrossRefPubMed Min MG, Song DJ, Miller M, et al. Coexposure to environmental tobacco smoke increases levels of allergen-induced airway remodeling in mice. J Immunol. 2007;178:5321–8.CrossRefPubMed
49.
go back to reference Seymour BW, Pinkerton KE, Friebertshauser KE, et al. Second-hand smoke is an adjuvant for T helper-2 responses in a murine model of allergy. J Immunol (Baltimore, Md 1950). 1997;159:6169–75. Seymour BW, Pinkerton KE, Friebertshauser KE, et al. Second-hand smoke is an adjuvant for T helper-2 responses in a murine model of allergy. J Immunol (Baltimore, Md 1950). 1997;159:6169–75.
50.
go back to reference Rumold R, Jyrala M, Diaz-Sanchez D. Secondhand smoke induces allergic sensitization in mice. J Immunol (Baltimore, Md 1950). 2001;167:4765–70.CrossRef Rumold R, Jyrala M, Diaz-Sanchez D. Secondhand smoke induces allergic sensitization in mice. J Immunol (Baltimore, Md 1950). 2001;167:4765–70.CrossRef
55.
go back to reference Robbins CS, Pouladi MA, Fattouh R, et al. Mainstream cigarette smoke exposure attenuates airway immune inflammatory responses to surrogate and common environmental allergens in mice, despite evidence of increased systemic sensitization. J Immunol. 2005;175:2834–42.CrossRefPubMed Robbins CS, Pouladi MA, Fattouh R, et al. Mainstream cigarette smoke exposure attenuates airway immune inflammatory responses to surrogate and common environmental allergens in mice, despite evidence of increased systemic sensitization. J Immunol. 2005;175:2834–42.CrossRefPubMed
62.
go back to reference Pedersen B, Dahl R, Karlström R, et al. Eosinophil and neutrophil activity in asthma in a one-year trial with inhaled budesonide. The impact of smoking. Am J Respir Crit Care Med. 1996;153:1519–29.CrossRefPubMed Pedersen B, Dahl R, Karlström R, et al. Eosinophil and neutrophil activity in asthma in a one-year trial with inhaled budesonide. The impact of smoking. Am J Respir Crit Care Med. 1996;153:1519–29.CrossRefPubMed
64.
go back to reference Chang WC, Lee YC, Liu CL, et al. Increased expression of iNOS and c-fos via regulation of protein tyrosine phosphorylation and MEK1/ERK2 proteins in terminal bronchiole lesions in the lungs of rats exposed to cigarette smoke. Arch Toxicol. 2001;75:28–35.CrossRefPubMed Chang WC, Lee YC, Liu CL, et al. Increased expression of iNOS and c-fos via regulation of protein tyrosine phosphorylation and MEK1/ERK2 proteins in terminal bronchiole lesions in the lungs of rats exposed to cigarette smoke. Arch Toxicol. 2001;75:28–35.CrossRefPubMed
65.
go back to reference Foster PS, Hogan SP, Ramsay AJ, et al. Interleukin 5 deficiency abolishes eosinophilia, airways hyperreactivity, and lung damage in a mouse asthma model. J Exp Med. 1996;183:195–201.CrossRefPubMed Foster PS, Hogan SP, Ramsay AJ, et al. Interleukin 5 deficiency abolishes eosinophilia, airways hyperreactivity, and lung damage in a mouse asthma model. J Exp Med. 1996;183:195–201.CrossRefPubMed
66.
go back to reference Hamelmann E, Oshiba A, Loader J, et al. Antiinterleukin-5 antibody prevents airway hyperresponsiveness in a murine model of airway sensitization. Am J Respir Crit Care Med. 1997;155:819–25.CrossRefPubMed Hamelmann E, Oshiba A, Loader J, et al. Antiinterleukin-5 antibody prevents airway hyperresponsiveness in a murine model of airway sensitization. Am J Respir Crit Care Med. 1997;155:819–25.CrossRefPubMed
67.
go back to reference Hamelmann E, Takeda K, Schwarze J, et al. Development of eosinophilic airway inflammation and airway hyperresponsiveness requires interleukin-5 but not immunoglobulin E or B lymphocytes. Am J Respir Cell Mol Biol. 1999;21:480–9.CrossRefPubMed Hamelmann E, Takeda K, Schwarze J, et al. Development of eosinophilic airway inflammation and airway hyperresponsiveness requires interleukin-5 but not immunoglobulin E or B lymphocytes. Am J Respir Cell Mol Biol. 1999;21:480–9.CrossRefPubMed
69.
go back to reference Mattes J, Yang M, Siqueira A, et al. IL-13 induces airways hyperreactivity independently of the IL-4R alpha chain in the allergic lung. J Immunol (Baltimore, Md 1950). 2001;167:1683–92.CrossRef Mattes J, Yang M, Siqueira A, et al. IL-13 induces airways hyperreactivity independently of the IL-4R alpha chain in the allergic lung. J Immunol (Baltimore, Md 1950). 2001;167:1683–92.CrossRef
70.
go back to reference Barczyk A, Pierzchala W, Sozañska E. Interleukin-17 in sputum correlates with airway hyperresponsiveness to methacholine. Respir Med. 2003;97:726–33.CrossRefPubMed Barczyk A, Pierzchala W, Sozañska E. Interleukin-17 in sputum correlates with airway hyperresponsiveness to methacholine. Respir Med. 2003;97:726–33.CrossRefPubMed
73.
go back to reference Schramm C, Herz U, Podlech J, et al. TGF-beta regulates airway responses via T cells. J Immunol (Baltimore, Md 1950). 2003;170:1313–9.CrossRef Schramm C, Herz U, Podlech J, et al. TGF-beta regulates airway responses via T cells. J Immunol (Baltimore, Md 1950). 2003;170:1313–9.CrossRef
77.
go back to reference Ebina M, Takahashi T, Chiba T, et al. Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis. 1993;148:720–6.CrossRefPubMed Ebina M, Takahashi T, Chiba T, et al. Cellular hypertrophy and hyperplasia of airway smooth muscles underlying bronchial asthma. A 3-D morphometric study. Am Rev Respir Dis. 1993;148:720–6.CrossRefPubMed
78.
go back to reference Fang Q, Zhao M, Ren G. Effects of cigarette smoke extract on proliferation and ET-1 release of airway smooth muscle cells. Zhonghua Yi Xue Za Zhi. 1997;77:201–4.PubMed Fang Q, Zhao M, Ren G. Effects of cigarette smoke extract on proliferation and ET-1 release of airway smooth muscle cells. Zhonghua Yi Xue Za Zhi. 1997;77:201–4.PubMed
79.
go back to reference Lin J, Xu Y, Zhang Z, et al. Effect of cigarette smoke extract on the role of protein kinase C in the proliferation of passively sensitized human airway smooth muscle cells. J Huazhong Univ Sci Technolog Med Sci. 2005;25:269–73.CrossRefPubMed Lin J, Xu Y, Zhang Z, et al. Effect of cigarette smoke extract on the role of protein kinase C in the proliferation of passively sensitized human airway smooth muscle cells. J Huazhong Univ Sci Technolog Med Sci. 2005;25:269–73.CrossRefPubMed
81.
go back to reference Zhang X-Y, Xu Y-J, Liu X-S, et al. Cigarette smoke extract promotes proliferation of airway smooth muscle cells in asthmatic rats via regulating cyclin D1 expression. Chin Med J. 2010;123:1709–14.PubMed Zhang X-Y, Xu Y-J, Liu X-S, et al. Cigarette smoke extract promotes proliferation of airway smooth muscle cells in asthmatic rats via regulating cyclin D1 expression. Chin Med J. 2010;123:1709–14.PubMed
83.
go back to reference Stavenow L, Falke P, Berglund A. Effects of different injurious stimuli on cell death, proliferation, and collagen secretion by rabbit aortic smooth muscle cells and human umbilical vein endothelial cells in culture. Med Biol. 1983;61:214–8.PubMed Stavenow L, Falke P, Berglund A. Effects of different injurious stimuli on cell death, proliferation, and collagen secretion by rabbit aortic smooth muscle cells and human umbilical vein endothelial cells in culture. Med Biol. 1983;61:214–8.PubMed
85.
go back to reference Zhang W, Case S, Bowler RP, et al. Cigarette smoke modulates PGE2 and host defence against Moraxella catarrhalis infection in human airway epithelial cells. Respirology. 2011;16:508–16.CrossRefPubMed Zhang W, Case S, Bowler RP, et al. Cigarette smoke modulates PGE2 and host defence against Moraxella catarrhalis infection in human airway epithelial cells. Respirology. 2011;16:508–16.CrossRefPubMed
89.
go back to reference Simon SA, Liedtke W. How irritating: the role of TRPA1 in sensing cigarette smoke and aerogenic oxidants in the airways. J Clin Invest. 2008;118:2383–6.PubMedPubMedCentral Simon SA, Liedtke W. How irritating: the role of TRPA1 in sensing cigarette smoke and aerogenic oxidants in the airways. J Clin Invest. 2008;118:2383–6.PubMedPubMedCentral
91.
92.
go back to reference Takahashi N, Kuwaki T, Kiyonaka S, et al. TRPA1 underlies a sensing mechanism for O2. Nat Chem Biol. 2011;7:701–11.CrossRefPubMed Takahashi N, Kuwaki T, Kiyonaka S, et al. TRPA1 underlies a sensing mechanism for O2. Nat Chem Biol. 2011;7:701–11.CrossRefPubMed
93.
go back to reference Sawada S, Suehisa H, Yamashita M. Inhalation of corticosteroid and β-agonist for persistent cough following pulmonary resection. Gen Thorac Cardiovasc Surg. 2012;60:285–8.CrossRefPubMed Sawada S, Suehisa H, Yamashita M. Inhalation of corticosteroid and β-agonist for persistent cough following pulmonary resection. Gen Thorac Cardiovasc Surg. 2012;60:285–8.CrossRefPubMed
Metadata
Title
Modelling the asthma phenotype: impact of cigarette smoke exposure
Authors
Maria G. Belvisi
Katie Baker
Nicole Malloy
Kristof Raemdonck
Bilel Dekkak
Michael Pieper
Anthony T. Nials
Mark A. Birrell
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Respiratory Research / Issue 1/2018
Electronic ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-018-0799-7

Other articles of this Issue 1/2018

Respiratory Research 1/2018 Go to the issue