Allergy Asthma Immunol Res.  2019 Sep;11(5):604-621. 10.4168/aair.2019.11.5.604.

Genes and Pathways Regulating Decline in Lung Function and Airway Remodeling in Asthma

Affiliations
  • 1Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea.
  • 2Department of Medicine, University of California San Diego, La Jolla, CA, USA. dbroide@ucsd.edu

Abstract

Asthma is a common disorder of the airways characterized by airway inflammation and by decline in lung function and airway remodeling in a subset of asthmatics. Airway remodeling is characterized by structural changes which include airway smooth muscle hypertrophy/hyperplasia, subepithelial fibrosis due to thickening of the reticular basement membrane, mucus metaplasia of the epithelium, and angiogenesis. Epidemiologic studies suggest that both genetic and environmental factors may contribute to decline in lung function and airway remodeling in a subset of asthmatics. Environmental factors include respiratory viral infection-triggered asthma exacerbations, and tobacco smoke. There is also evidence that several asthma candidate genes may contribute to decline in lung function, including ADAM33, PLAUR, VEGF, IL13, CHI3L1, TSLP, GSDMB, TGFB1, POSTN, ESR1 and ARG2. In addition, mediators or cytokines, including cysteinyl leukotrienes, matrix metallopeptidase-9, interleukin-33 and eosinophil expression of transforming growth factor-β, may contribute to airway remodeling in asthma. Although increased airway smooth muscle is associated with reduced lung function (i.e. forced expiratory volume in 1 second) in asthma, there have been few long-term studies to determine how individual pathologic features of airway remodeling contribute to decline in lung function in asthma. Clinical studies with inhibitors of individual gene products, cytokines or mediators are needed in asthmatic patients to identify their individual role in decline in lung function and/or airway remodeling.

Keyword

Airway remodeling; gene polymorphisms; lung function tests

MeSH Terms

Airway Remodeling*
Asthma*
Basement Membrane
Cytokines
Eosinophils
Epidemiologic Studies
Epithelium
Fibrosis
Forced Expiratory Volume
Humans
Inflammation
Interleukin-13
Interleukin-33
Leukotrienes
Lung*
Metaplasia
Mucus
Muscle, Smooth
Respiratory Function Tests
Smoke
Tobacco
Vascular Endothelial Growth Factor A
Cytokines
Interleukin-13
Interleukin-33
Leukotrienes
Smoke
Vascular Endothelial Growth Factor A

Reference

1. Clarke TC, Norris T, Schiller JS. Early release of selected estimates based on data from the 2016 national health interview survey. Hyattsville (MA): National Center for Health Statistics;2016.
2. Park SY, Kim JH, Kim HJ, Seo B, Kwon OY, Chang HS, et al. High prevalence of asthma in elderly women: findings from a Korean national health database and adult asthma cohort. Allergy Asthma Immunol Res. 2018; 10:387–396.
Article
3. Broide DH. Immunologic and inflammatory mechanisms that drive asthma progression to remodeling. J Allergy Clin Immunol. 2008; 121:560–570.
Article
4. Lange P, Parner J, Vestbo J, Schnohr P, Jensen G. A 15-year follow-up study of ventilatory function in adults with asthma. N Engl J Med. 1998; 339:1194–1200.
Article
5. Oldham PD. Decline of FEV1. Thorax. 1987; 42:161–164.
Article
6. McGeachie MJ, Yates KP, Zhou X, Guo F, Sternberg AL, Van Natta ML, et al. Patterns of growth and decline in lung function in persistent childhood asthma. N Engl J Med. 2016; 374:1842–1852.
Article
7. Hayden LP, Cho MH, Raby BA, Beaty TH, Silverman EK, Hersh CP. Childhood asthma is associated with COPD and known asthma variants in COPDGene: a genome-wide association study. Respir Res. 2018; 19:209.
Article
8. Ober C. Asthma genetics in the post-GWAS era. Ann Am Thorac Soc. 2016; 13:Suppl 1. S85–S90.
9. Stone AL, Kroeger M, Sang QX. Structure-function analysis of the ADAM family of disintegrin-like and metalloproteinase-containing proteins (review). J Protein Chem. 1999; 18:447–465.
10. Van Eerdewegh P, Little RD, Dupuis J, Del Mastro RG, Falls K, Simon J, et al. Association of the ADAM33 gene with asthma and bronchial hyperresponsiveness. Nature. 2002; 418:426–430.
Article
11. Holgate ST, Davies DE, Rorke S, Cakebread J, Murphy G, Powell RM, et al. ADAM 33 and its association with airway remodeling and hyperresponsiveness in asthma. Clin Rev Allergy Immunol. 2004; 27:23–34.
Article
12. Jongepier H, Boezen HM, Dijkstra A, Howard TD, Vonk JM, Koppelman GH, et al. Polymorphisms of the ADAM33 gene are associated with accelerated lung function decline in asthma. Clin Exp Allergy. 2004; 34:757–760.
Article
13. van Diemen CC, Postma DS, Vonk JM, Bruinenberg M, Schouten JP, Boezen HM. A disintegrin and metalloprotease 33 polymorphisms and lung function decline in the general population. Am J Respir Crit Care Med. 2005; 172:329–333.
Article
14. Poon AH, Houseman EA, Ryan L, Sparrow D, Vokonas PS, Litonjua AA. Variants of asthma and chronic obstructive pulmonary disease genes and lung function decline in aging. J Gerontol A Biol Sci Med Sci. 2014; 69:907–913.
Article
15. Mondino A, Resnati M, Blasi F. Structure and function of the urokinase receptor. Thromb Haemost. 1999; 82:Suppl 1. 19–22.
Article
16. Brooks AM, Bates ME, Vrtis RF, Jarjour NN, Bertics PJ, Sedgwick JB. Urokinase-type plasminogen activator modulates airway eosinophil adhesion in asthma. Am J Respir Cell Mol Biol. 2006; 35:503–511.
Article
17. Blasi F, Carmeliet P. uPAR: a versatile signalling orchestrator. Nat Rev Mol Cell Biol. 2002; 3:932–943.
Article
18. Barton SJ, Koppelman GH, Vonk JM, Browning CA, Nolte IM, Stewart CE, et al. PLAUR polymorphisms are associated with asthma, PLAUR levels, and lung function decline. J Allergy Clin Immunol. 2009; 123:1391–1400.e17.
Article
19. Ierodiakonou D, Portelli MA, Postma DS, Koppelman GH, Gerritsen J, Ten Hacken NH, et al. Urokinase plasminogen activator receptor polymorphisms and airway remodelling in asthma. Eur Respir J. 2016; 47:1568–1571.
Article
20. Lee CG, Link H, Baluk P, Homer RJ, Chapoval S, Bhandari V, et al. Vascular endothelial growth factor (VEGF) induces remodeling and enhances TH2-mediated sensitization and inflammation in the lung. Nat Med. 2004; 10:1095–1103.
Article
21. Hoshino M, Nakamura Y, Hamid QA. Gene expression of vascular endothelial growth factor and its receptors and angiogenesis in bronchial asthma. J Allergy Clin Immunol. 2001; 107:1034–1038.
Article
22. Simpson A, Custovic A, Tepper R, Graves P, Stern DA, Jones M, et al. Genetic variation in vascular endothelial growth factor-a and lung function. Am J Respir Crit Care Med. 2012; 185:1197–1204.
Article
23. Sharma S, Murphy AJ, Soto-Quiros ME, Avila L, Klanderman BJ, Sylvia JS, et al. Association of VEGF polymorphisms with childhood asthma, lung function and airway responsiveness. Eur Respir J. 2009; 33:1287–1294.
Article
24. Zhu Z, Lee CG, Zheng T, Chupp G, Wang J, Homer RJ, et al. Airway inflammation and remodeling in asthma. Lessons from interleukin 11 and interleukin 13 transgenic mice. Am J Respir Crit Care Med. 2001; 164:S67–S70.
25. Park HW, Lee JE, Kim SH, Kim YK, Min KU, Kim YY, et al. Genetic variation of IL13 as a risk factor of reduced lung function in children and adolescents: a cross-sectional population-based study in Korea. Respir Med. 2009; 103:284–288.
Article
26. Nagashima H, Nakamura Y, Kanno H, Sawai T, Inoue H, Yamauchi K. Effect of genetic variation of IL-13 on airway remodeling in bronchial asthma. Allergol Int. 2011; 60:291–298.
Article
27. Nakamura Y, Suzuki R, Mizuno T, Abe K, Chiba S, Horii Y, et al. Therapeutic implication of genetic variants of IL13 and STAT4 in airway remodelling with bronchial asthma. Clin Exp Allergy. 2016; 46:1152–1161.
28. Zhu Z, Zheng T, Homer RJ, Kim YK, Chen NY, Cohn L, et al. Acidic mammalian chitinase in asthmatic Th2 inflammation and IL-13 pathway activation. Science. 2004; 304:1678–1682.
Article
29. Ober C, Chupp GL. The chitinase and chitinase-like proteins: a review of genetic and functional studies in asthma and immune-mediated diseases. Curr Opin Allergy Clin Immunol. 2009; 9:401–408.
Article
30. Ober C, Tan Z, Sun Y, Possick JD, Pan L, Nicolae R, et al. Effect of variation in CHI3L1 on serum YKL-40 level, risk of asthma, and lung function. N Engl J Med. 2008; 358:1682–1691.
31. Rathcke CN, Holmkvist J, Husmoen LL, Hansen T, Pedersen O, Vestergaard H, et al. Association of polymorphisms of the CHI3L1 gene with asthma and atopy: a populations-based study of 6514 Danish adults. PLoS One. 2009; 4:e6106.
Article
32. Gomez JL, Crisafi GM, Holm CT, Meyers DA, Hawkins GA, Bleecker ER, et al. Genetic variation in chitinase 3-like 1 (CHI3L1) contributes to asthma severity and airway expression of YKL-40. J Allergy Clin Immunol. 2015; 136:51–58.e10.
Article
33. Abe K, Nakamura Y, Yamauchi K, Maemondo M. Role of genetic variations of chitinase 3-like 1 in bronchial asthmatic patients. Clin Mol Allergy. 2018; 16:9.
Article
34. Chupp GL, Lee CG, Jarjour N, Shim YM, Holm CT, He S, et al. A chitinase-like protein in the lung and circulation of patients with severe asthma. N Engl J Med. 2007; 357:2016–2027.
Article
35. Soumelis V, Reche PA, Kanzler H, Yuan W, Edward G, Homey B, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol. 2002; 3:673–680.
36. Ying S, O'Connor B, Ratoff J, Meng Q, Mallett K, Cousins D, et al. Thymic stromal lymphopoietin expression is increased in asthmatic airways and correlates with expression of Th2-attracting chemokines and disease severity. J Immunol. 2005; 174:8183–8190.
Article
37. Harada M, Hirota T, Jodo AI, Hitomi Y, Sakashita M, Tsunoda T, et al. Thymic stromal lymphopoietin gene promoter polymorphisms are associated with susceptibility to bronchial asthma. Am J Respir Cell Mol Biol. 2011; 44:787–793.
Article
38. Moffatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S, et al. Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature. 2007; 448:470–473.
Article
39. Das S, Miller M, Broide DH. Chromosome 17q21 genes ORMDL3 and GSDMB in asthma and immune diseases. Adv Immunol. 2017; 135:1–52.
Article
40. Moffatt MF, Gut IG, Demenais F, Strachan DP, Bouzigon E, Heath S, et al. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010; 363:1211–1221.
Article
41. Loss GJ, Depner M, Hose AJ, Genuneit J, Karvonen AM, Hyvärinen A, et al. The early development of wheeze. environmental determinants and genetic susceptibility at 17q21. Am J Respir Crit Care Med. 2016; 193:889–897.
Article
42. Toncheva AA, Potaczek DP, Schedel M, Gersting SW, Michel S, Krajnov N, et al. Childhood asthma is associated with mutations and gene expression differences of ORMDL genes that can interact. Allergy. 2015; 70:1288–1299.
43. Das S, Miller M, Beppu AK, Mueller J, McGeough MD, Vuong C, et al. GSDMB induces an asthma phenotype characterized by increased airway responsiveness and remodeling without lung inflammation. Proc Natl Acad Sci U S A. 2016; 113:13132–13137.
Article
44. Tang MF, Sy HY, Kong AP, Ko FW, Wang SS, Liu TC, et al. Genetic effects of multiple asthma loci identified by genomewide association studies on asthma and spirometric indices. Pediatr Allergy Immunol. 2016; 27:185–194.
Article
45. Miller M, Rosenthal P, Beppu A, Mueller JL, Hoffman HM, Tam AB, et al. ORMDL3 transgenic mice have increased airway remodeling and airway responsiveness characteristic of asthma. J Immunol. 2014; 192:3475–3487.
Article
46. Ojiaku CA, Yoo EJ, Panettieri RA Jr. Transforming growth factor β1 function in airway remodeling and hyperresponsiveness. the missing link? Am J Respir Cell Mol Biol. 2017; 56:432–442.
Article
47. Le AV, Cho JY, Miller M, McElwain S, Golgotiu K, Broide DH. Inhibition of allergen-induced airway remodeling in Smad 3-deficient mice. J Immunol. 2007; 178:7310–7316.
Article
48. Gregory LG, Mathie SA, Walker SA, Pegorier S, Jones CP, Lloyd CM. Overexpression of Smad2 drives house dust mite-mediated airway remodeling and airway hyperresponsiveness via activin and IL-25. Am J Respir Crit Care Med. 2010; 182:143–154.
Article
49. Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A, Ludwig MS, et al. Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest. 2003; 112:1029–1036.
Article
50. Ierodiakonou D, Postma DS, Koppelman GH, Gerritsen J, ten Hacken NH, Timens W, et al. TGF-β1 polymorphisms and asthma severity, airway inflammation, and remodeling. J Allergy Clin Immunol. 2013; 131:582–585.
Article
51. Sidhu SS, Yuan S, Innes AL, Kerr S, Woodruff PG, Hou L, et al. Roles of epithelial cell-derived periostin in TGF-beta activation, collagen production, and collagen gel elasticity in asthma. Proc Natl Acad Sci U S A. 2010; 107:14170–14175.
52. Izuhara K, Conway SJ, Moore BB, Matsumoto H, Holweg CT, Matthews JG, et al. Roles of periostin in respiratory disorders. Am J Respir Crit Care Med. 2016; 193:949–956.
Article
53. Kanemitsu Y, Matsumoto H, Izuhara K, Tohda Y, Kita H, Horiguchi T, et al. Increased periostin associates with greater airflow limitation in patients receiving inhaled corticosteroids. J Allergy Clin Immunol. 2013; 132:305–312.e3.
Article
54. Dijkstra A, Howard TD, Vonk JM, Ampleford EJ, Lange LA, Bleecker ER, et al. Estrogen receptor 1 polymorphisms are associated with airway hyperresponsiveness and lung function decline, particularly in female subjects with asthma. J Allergy Clin Immunol. 2006; 117:604–611.
Article
55. Vonk JM, Postma DS, Maarsingh H, Bruinenberg M, Koppelman GH, Meurs H. Arginase 1 and arginase 2 variations associate with asthma, asthma severity and beta2 agonist and steroid response. Pharmacogenet Genomics. 2010; 20:179–186.
56. Sampsonas F, Kaparianos A, Lykouras D, Karkoulias K, Spiropoulos K. DNA sequence variations of metalloproteinases: their role in asthma and COPD. Postgrad Med J. 2007; 83:244–250.
Article
57. Gagliardo R, La Grutta S, Chanez P, Profita M, Paternò A, Cibella F, et al. Non-invasive markers of airway inflammation and remodeling in childhood asthma. Pediatr Allergy Immunol. 2009; 20:780–790.
Article
58. Lim DH, Cho JY, Miller M, McElwain K, McElwain S, Broide DH. Reduced peribronchial fibrosis in allergen-challenged MMP-9-deficient mice. Am J Physiol Lung Cell Mol Physiol. 2006; 291:L265–71.
Article
59. Singh RK, Tandon R, Dastidar SG, Ray A. A review on leukotrienes and their receptors with reference to asthma. J Asthma. 2013; 50:922–931.
Article
60. Holgate ST, Peters-Golden M, Panettieri RA, Henderson WR Jr. Roles of cysteinyl leukotrienes in airway inflammation, smooth muscle function, and remodeling. J Allergy Clin Immunol. 2003; 111:S18–34.
Article
61. Henderson WR Jr, Tang LO, Chu SJ, Tsao SM, Chiang GK, Jones F, et al. A role for cysteinyl leukotrienes in airway remodeling in a mouse asthma model. Am J Respir Crit Care Med. 2002; 165:108–116.
Article
62. In KH, Asano K, Beier D, Grobholz J, Finn PW, Silverman EK, et al. Naturally occurring mutations in the human 5-lipoxygenase gene promoter that modify transcription factor binding and reporter gene transcription. J Clin Invest. 1997; 99:1130–1137.
Article
63. Mougey E, Lang JE, Allayee H, Teague WG, Dozor AJ, Wise RA, et al. ALOX5 polymorphism associates with increased leukotriene production and reduced lung function and asthma control in children with poorly controlled asthma. Clin Exp Allergy. 2013; 43:512–520.
64. Ro M, Kim S, Pyun JA, Shin C, Cho NH, Lee JY, et al. Association between arachidonate 5-lipoxygenase-activating protein (ALOX5AP) and lung function in a Korean population. Scand J Immunol. 2012; 76:151–157.
Article
65. Drake LY, Kita H. IL-33: biological properties, functions, and roles in airway disease. Immunol Rev. 2017; 278:173–184.
Article
66. Mizutani N, Nabe T, Yoshino S. Interleukin-33 and alveolar macrophages contribute to the mechanisms underlying the exacerbation of IgE-mediated airway inflammation and remodelling in mice. Immunology. 2013; 139:205–218.
Article
67. Bai TR, Vonk JM, Postma DS, Boezen HM. Severe exacerbations predict excess lung function decline in asthma. Eur Respir J. 2007; 30:452–456.
Article
68. Matsunaga K, Hirano T, Oka A, Tanaka A, Kanai K, Kikuchi T, et al. Progression of irreversible airflow limitation in asthma: correlation with severe exacerbations. J Allergy Clin Immunol Pract. 2015; 3:759–764.e1.
Article
69. Ortega H, Yancey SW, Keene ON, Gunsoy NB, Albers FC, Howarth PH. Asthma exacerbations associated with lung function decline in patients with severe eosinophilic asthma. J Allergy Clin Immunol Pract. 2018; 6:980–986.e1.
Article
70. O'Byrne PM, Pedersen S, Lamm CJ, Tan WC, Busse WW. START Investigators Group. Severe exacerbations and decline in lung function in asthma. Am J Respir Crit Care Med. 2009; 179:19–24.
71. Leigh R, Oyelusi W, Wiehler S, Koetzler R, Zaheer RS, Newton R, et al. Human rhinovirus infection enhances airway epithelial cell production of growth factors involved in airway remodeling. J Allergy Clin Immunol. 2008; 121:1238–1245.e4.
Article
72. Calışkan M, Bochkov YA, Kreiner-Møller E, Bønnelykke K, Stein MM, Du G, et al. Rhinovirus wheezing illness and genetic risk of childhood-onset asthma. N Engl J Med. 2013; 368:1398–1407.
Article
73. Lemiere C, Boulet LP. Cigarette smoking and asthma: a dangerous mix. Can Respir J. 2005; 12:79–80.
Article
74. Toledo-Pons N, van Boven JF, Román-Rodríguez M, Pérez N, Valera Felices JL, Soriano JB, et al. ACO: time to move from the description of different phenotypes to the treatable traits. PLoS One. 2019; 14:e0210915.
Article
75. Niwa M, Fujisawa T, Karayama M, Furuhashi K, Mori K, Hashimoto D, et al. Differences in airway structural changes assessed by 3-dimensional computed tomography in asthma and asthma-chronic obstructive pulmonary disease overlap. Ann Allergy Asthma Immunol. 2018; 121:704–710.e1.
Article
76. Min MG, Song DJ, Miller M, Cho JY, McElwain S, Ferguson P, et al. Coexposure to environmental tobacco smoke increases levels of allergen-induced airway remodeling in mice. J Immunol. 2007; 178:5321–5328.
Article
77. Boulet LP, Turcotte H, Laviolette M, Naud F, Bernier MC, Martel S, et al. Airway hyperresponsiveness, inflammation, and subepithelial collagen deposition in recently diagnosed versus long-standing mild asthma. Influence of inhaled corticosteroids. Am J Respir Crit Care Med. 2000; 162:1308–1313.
78. Chakir J, Shannon J, Molet S, Fukakusa M, Elias J, Laviolette M, et al. Airway remodeling-associated mediators in moderate to severe asthma: effect of steroids on TGF-beta, IL-11, IL-17, and type I and type III collagen expression. J Allergy Clin Immunol. 2003; 111:1293–1298.
79. Sont JK, Willems LN, Bel EH, van Krieken JH, Vandenbroucke JP, Sterk PJ. Clinical control and histopathologic outcome of asthma when using airway hyperresponsiveness as an additional guide to long-term treatment. The AMPUL study group. Am J Respir Crit Care Med. 1999; 159:1043–1051.
80. Ward C, Pais M, Bish R, Reid D, Feltis B, Johns D, et al. Airway inflammation, basement membrane thickening and bronchial hyperresponsiveness in asthma. Thorax. 2002; 57:309–316.
Article
Full Text Links
  • AAIR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr