SEVERE ASTHMA SERIES
Genes and severe asthma
 
More details
Hide details
1
Department of Pneumonology, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
 
2
Airway Disease Unit, National Heart and Lung Institute, Imperial College, London, UK
 
 
Corresponding author
Paschalis Steiropoulos   

Medical School, Democritus University of Thrace Department of Pneumonology, University Hospital of Alexandroupolis 68100 Alexandroupolis, Greece
 
 
Pneumon 2011;24(3):321-329
 
KEYWORDS
ABSTRACT
Asthma is a chronic inflammatory disease of the airways, affecting approximately 155 million people worldwide. The causes are multifactorial and are attributed to several interactions between various different genes and environmental exposures. These interactions can affect either the susceptibility of a person to develop of asthma or the severity of the disease. This review article presents recent approaches to the understanding of the genetic influences on the severity of asthma expression. Specifically, genetic studies conducted in adults with severe asthma, and the spectrum of the genetics of lung function are reviewed, with special focus on pharmacogenetics and the genetic factors that affect the response to asthma treatment. Finally, environmental interactions and epigenetic mechanisms are discussed.
 
REFERENCES (90)
1.
Asher MI, Keil U, Anderson HR, et al. International Study of Asthma and Allergies in Childhood (ISAAC): rationale and methods. Eur Respir J 1995;8:483-491.
 
2.
Beasley R. The burden of asthma with specific reference to the United States. J Allergy Clin Immunol 2002;109:482-489.
 
3.
Duffy DL, Martin NG, Battistuta D, et al. Genetics of asthma and hay feverin Australian twins. Am Rev Respir Dis 1990;152:1351- 1358.
 
4.
Nieminen NM, Kaprio J, Koskenvuo M. A population-based study of bronchial asthmain adult twin pairs. Chest 1991;100:70-77.
 
5.
Hopp RJ, Bewtra AK, Watt GD, et al. Genetic analysis of allergic disease in twins. J Allergy Clin Immunol 1984;73:265-270.
 
6.
Los H, Postmus PE, Boomsma DI. Asthma genetics and intermediate phenotypes: a review from twin studies. Twin Res 2001;4:81–93.
 
7.
Burke W, Fesinmeyer M, Reed K, et al. Family history as a predictor of asthma risk. Am J Prev Med 2003;24:160–169.
 
8.
Bleecker ER. Pharmacogenetics of asthma. US Resp Disease 2008;4:39–44.
 
9.
Jongepier H, Boezen HM, Dijkstra A, et al. Polymorphisms of the ADAM33 gene are associated with accelerated lung function decline in asthma. Clin Exp Allergy 2004; 34: 757–760.
 
10.
Slager RE, Li X, Meyers DA, Bleecker ER. Recent developments in the genetics of asthma susceptibility and severity. Eur Respir Mon 2011;51:82-96.
 
11.
Meyers DA. Genetics of asthma and allergy: What have we learned? J Allergy Clin Immunol 2010; 126:439-446.
 
12.
Zhang J, Pare PD, Sandford AJ. Recent advances in asthma genetics. Respir Res 2008;9;4.
 
13.
Moraitaki P, Papamichail D, Georgatou N. Severe Asthma: Definitions, risk factors and phenotype characterization. Pneumon 2010;23:276-292.
 
14.
Hoffjan S, Ober C. Present status on the genetic studies of asthma. Curr Opin Immunol 2002;14:709-717.
 
15.
Chen Q, Book M, Fang X, et al. Screening of copynumber polymorphisms in human beta-defensin genes using modified realtime quantitative PCR. J Immunol Methods 2006;308:231-240.
 
16.
Li X, Howard TD, Zheng SL, et al. Genome-wide association study of asthma identifies RAD 50-IL13 and HLA DR/DQ regions. J Allergy Clin Immunology 2010,125:328-335.
 
17.
Dolan CM, Fraher KE, Bleecker ER, et al. Design and baseline characteristics of the epidemiology and natural history of asthma: Outcomes and Treatment Regimens (TENOR) study: a large cohort of patients with severe or difficult-to-treat asthma. Ann Allergy Asthma Immunol 2004;92:32–39.
 
18.
Haselkorn T, Fish JE, Zeiger RS, et al. Consistently very poorly controlled asthma, as defined by the impairment domain of the Expert Panel Report 3 guidelines, increases risk for future severe asthma exacerbations in The Epidemiology and Natural History of Asthma: Outcomes and Treatment Regimens (TENOR) study. J Allergy Clin Immunol 2009; 124: 895–902.
 
19.
Grunig G, Warnock M, Wakil AE, et al. Requirement for IL-13 independently of IL-4 in experimental asthma. Science 1998; 282: 2261–2263.
 
20.
Li X, Howard TD, Ampleford EJ, et al. Genome-wide association study of asthma severity using proportional odds model identifies TMEM154. Am J Respir Crit Care Med 2010;181:A3728.
 
21.
Wills-Karp M , Luyimbazi J, Xu X, et al. Interleukin-13: central mediator of allergic asthma. Science 1998; 282: 2258–2261.
 
22.
Booth M, Shaw MA, Carpenter D, et al. Carriage of DRB1*13 is associated with increased posttreatment IgE levels against Schistosoma mansoni antigens and lower long-term reinfection levels. J Immunol 2006;176:7112–7118.
 
23.
Munthe-Kaas MC, Carlsen KL, Carlsen KH, et al. HLA Dr-Dq haplotypes and the TNFA-308 polymorphism: associations with asthma and allergy. Allergy 2007;62:991–998.
 
24.
Moffatt MF, Gut IG, Demenais F, et al. A large-scale, consortiumbased genomewide association study of asthma. N Engl J Med 2010;363:1211–1221.
 
25.
Weidinger S, Gieger C, Rodriguez E, et al. Genome-wide scan on total serum IgE levels identifies FCER1A as novel susceptibility locus. PLoS Genetics 2008;4:e1000166.
 
26.
Moore WC, Bleecker ER, Curran-Everett D et al. Characterization of the severe asthma phenotype by the National Heart, Lung, and Blood Institute’s Severe Asthma Research Program J Allergy Clin Immunol 2007;119(2):405-413.
 
27.
Moore WC, Meyers DA, Wenzel SE, et al. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med 2010;181:315–323.
 
28.
Van Eerdewegh P, Little RD, Dupuis J, et al. Association of the ADAM33 gene with asthma and bronchial hyperresponsiveness. Nature 2002;418:426–430.
 
29.
Howard TD, Postma DS, Jongepier H, et al. Association of a disintegrin and metalloprotease 33 (ADAM33) gene with asthma in ethnically diverse populations. J Allergy Clin Immunol 2003;112:717–722.
 
30.
Van Diemen CC, Postma DS, Vonk JM, et al. A disintegrin and metalloprotease 33 polymorphisms and lung function decline in the general population. Am J Respir Crit Care Med 2005;172:329–333.
 
31.
Sadeghnejad A, Ohar JA, Zheng SL, et al. Adam33 polymorphisms are associated with COPD and lung function in long-term tobacco smokers. Resp Res 2009;10:21.
 
32.
Hancock DB, Eijgelsheim M, Wilk JB, et al. Meta-analyses of genome-wide association studies identify multiple loci associated with pulmonary function. Nat Gen 2010;42:45–52.
 
33.
Repapi E, Sayers I, Wain LV, et al. Genome-wide association study identifies five loci associated with lung function. Nat Gen 2010; 42: 36–44.
 
34.
Li X, Howard TD, Moore WC, et al. Importance of HHIP and other lung function genes in asthma. J Allergy Clin Immunol: 2011 [DOI: 10.1016/J.JACI.2011.01.056].
 
35.
Gudbjartsson DF, Bjornsdottir US, Halapi E, et al. Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Gen 2009;41:342–347.
 
36.
Wenzel SE, Balzar S, Ampleford E, et al. IL4R a mutations are associated with asthma exacerbations and mast cell/IgE expression. Am J Respir Crit Care Med 2007;175:570–576.
 
37.
Israel E, Chinchilli VM, Ford JG, et al. Use of regularly scheduled albuterol treatment in asthma: genotypestratified, randomised, placebo-controlled cross-over trial. Lancet 2004;364:1505–1512.
 
38.
Wechsler ME, Kunselman SJ, Chinchilli VM, et al. Effect of β2- adrenergic receptor polymorphism on response to longacting b2 agonist in asthma (LARGE trial): a genotype-stratified, randomised, placebo-controlled, crossover trial. Lancet 2009; 374:1754–1764.
 
39.
Bleecker ER, Nelson HS, Kraft M, et al. Β2-receptor polymorphisms in patients receiving salmeterol with or without fluticasone propionate. Am J Respir Crit Care Med 2010;181:676–687.
 
40.
Bleecker ER, Postma DS, Lawrance RM, et al. Effect of ADRB2 polymorphisms on response to longacting β2- agonist therapy: a pharmacogenetic analysis of two randomised studies. Lancet 2007;370:2118–2125.
 
41.
Vercelli D. Discovering susceptibility genes for asthma and allergy. Nat Rev Immunol 2008;8:169–182.
 
42.
Pascual RM, Bleecker ER. Pharmacogenetics of asthma. Curr Opin Pharmacol 2010;10:226–235.
 
43.
Lima JJ, Blake KV, Tantisira KG, et al. Pharmacogenetics of asthma. Curr Opin Pulm Med 2009;15:57–62.
 
44.
Green SA, Turki J, Innis M, et al. Amino-terminal polymorphisms of the human β2-adrenergic receptor impart distinct agonistpromoted regulatory properties. Biochemistry 1994;33:9414- 9419.
 
45.
Green SA, Turki J, Bejarano P, et al. Influence of β2-adrenergic receptor genotypes on signal transduction in human airway smooth muscle cells. Am J Resp Cell Mol Biol 1995;13:25–33.
 
46.
Martinez FD, Graves PE, Baldini M, et al. Association between genetic polymorphisms of the β2-adrenoceptor and response to albuterol in children with and without a history of wheezing. J Clin Invest 1997;100:3184–3188.
 
47.
Cho SH, Oh SY, Bahn JW, et al. Association between bronchodilating response to short-acting β-agonist and non-synonymous single-nucleotide polymorphisms of b-adrenoceptor gene. Clin Exp Allergy 2005;35:1162–1167.
 
48.
Kotani Y, Nishimura Y, Maeda H, et al. Β2-adrenergic receptor polymorphisms affect airway responsiveness to salbutamol in asthmatics. J Asthma 1999;36:583–590.
 
49.
Tan S, Hall IP, Dewar J, et al. Association between β2-adrenoceptor polymorphism and susceptibility to bronchodilator desensitisation in moderately severe stable asthmatics. Lancet 1997;350:995–999.
 
50.
Hancox RJ, Sears MR, Taylor DR. Polymorphism of the b2- adrenoceptor and the response to long-term β2- agonist therapy in asthma. Eur Respir J 1998;11:589–593.
 
51.
Taylor DR, Drazen JM, Herbison GP, et al. Asthma exacerbations during long term b agonist use: influence of β2 adrenoceptor polymorphism. Thorax 2000;55:762–767.
 
52.
Castle W, Fuller R, Hall J, et al. Serevent nationwide surveillance study: comparison of salmeterol with salbutamol in asthmatic patients who require regular bronchodilator treatment. BMJ 1993;306:1034–1037.
 
53.
Nelson HS, Weiss ST, Bleecker ER, et al. The Salmeterol Multicenter Asthma Research Trial: a comparison of usual pharmacotherapy for asthma or usual pharmacotherapy plus salmeterol. Chest 2006;129:15–26.
 
54.
Tantisira KG, Lake S, Silverman ES, et al. Corticosteroid pharmacogenetics: association of sequence variants in CRHR1 with improved lung function in asthmatics treated with inhaled corticosteroids. Hum Mol Gen 2004;13:1353–1359.
 
55.
Hawkins GA, Lazarus R, Smith RS, et al. The glucocorticoid receptor heterocomplex gene STIP1 is associated with improved lung function in asthmatic subjects treated with inhaled corticosteroids. J Allergy Clin Immunol 2009;123:1376–1383.
 
56.
Adcock IM, Barnes PJ. Molecular mechanisms of corticosteroid resistance. Chest 2008;134:394–401.
 
57.
Barnes PJ, Adcock IM. Glucocorticoid resistance in inflammatory diseases. Lancet 2009;373:1905–1917.
 
58.
Huizenga NA, Koper JW, De Lange P, et al. A polymorphism in the glucocorticoid receptor gene may be associated with an increased sensitivity to glucocorticoids in vivo. J Clin Endocrinol Metab 1998;83:144–151.
 
59.
Drazen JM, Yandava CN, Dube L, et al. Pharmacogenetic association between ALOX5 promoter genotype and the response to anti-asthma treatment. Nat Gen 1999;22:168–170.
 
60.
Coffey M, Peters-Golden M. Extending the understanding of leukotrienes in asthma. Curr Opin Allergy Clin Immunol 2003;3:57–63.
 
61.
Hui Y, Funk CD. Cysteinyl leukotriene receptors. Biochem Pharmacol 2002;64:1549–1557.
 
62.
Sampson AP, Siddiqui S, Buchanan D, et al. Variant LTC(4) synthase allele modifies cysteinyl leukotriene synthesis in eosinophils and predicts clinical response to zafirlukast. Thorax 2000; 55(Suppl. 2):S28–S31.
 
63.
Corrigan C, Mallett K, Ying S, et al. Expression of the cysteinyl leukotriene receptors cysLT1; and cysLT2; in aspirin-sensitive and aspirin-tolerant chronic rhinosinusitis. J Allergy Clin Immunol 2005;115:316–322.
 
64.
Fukai H, Ogasawara Y, Migita O, et al. Association between a polymorphism in cysteinyl leukotriene receptor 2 on chromosome 13q14 and atopic asthma. Pharmacogenetics 2004;14:683–690.
 
65.
Hao L, Sayers I, Cakebread JA, et al. The cysteinyl-leukotriene type 1 receptor polymorphism 927T/C is associated with atopy severity but not with asthma. Clin Exp Allergy Immunol 2006;36:735–741.
 
66.
Pillai SG, Cousens DJ, Barnes AA, et al. A coding polymorphism in the CYSLT2 receptor with reduced affinity to LTD4 is associated with asthma. Pharmacogenetics 2004;14:627–633.
 
67.
Lima JJ, Zhang S, Grant A, et al. Influence of leukotriene pathway polymorphisms on response to montelukast in asthma. Am J Respir Crit Care Med 2006;173:379–385.
 
68.
Meyers DA, Hawkins GA, Wenzel SE, et al. Pharmacogenetic identification of increased responsiveness in severe asthma with anti-TNF (Golimumab) therapy. J Allergy Clin Immunol 2008;121:S798.
 
69.
Slager RE, Hawkins GA, Ampleford EJ, et al. IL-4 receptor a polymorphisms are predictors of a pharmacogenetic response to a novel IL-4/IL-13 antagonist. J Allergy Clin Immunol 2010;126:875–878.
 
70.
Wenzel S, Wilbraham D, Fuller R, et al. Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. Lancet 2007;370:1422–1431.
 
71.
Slager RE, Hawkins GA, Otulana BA, et al. Interleukin 4 receptor polymorphisms predict therapeutic pitrakinra treatment response in moderate to severe asthma. Am J Respir Crit Care Med 2011;183: (abstract).
 
72.
Zambelli-Weiner A, Ehrlich E, Stockton ML, et al. Evaluation of the CD14/-260 polymorphism and house dust endotoxin exposure in the Barbados Asthma Genetics Study. J Allergy Clin Immunol 2005;115:1203–1209.
 
73.
Bottema RWB, Reijmerink NE, Kerkhof M, et al. Interleukin 13, CD14, pet and tobacco smoke influence atopy in three Dutch cohorts: the allergenic study. Eur Respir J 2008;32:593–602.
 
74.
Pacheco KA, Rose CS, Silveira LJ, et al. Gene-environment interactions influence airways function in laboratory animal workers. J Allergy Clin Immunol 2010;126:232–240.
 
75.
Simpson A, John SL, Jury F, et al. Endotoxin exposure, CD14, and allergic disease: an interaction between genes and the environment. Am J Respir Crit Care Med 2006;174:386–392.
 
76.
Ober C, Vercelli D. Gene-environment interactions in human disease: nuisance or opportunity? Trends Genet 2011; [Epub ahead of print, DOI: 10.1016/j.tig.2010.12.004].
 
77.
Koppelman GH. Gene by environment interaction in asthma. Curr Allergy Asthma Rep 2006;6:103–111.
 
78.
Meyers DA, Postma DS, Stine OC, et al. Genome screen for asthma and bronchial hyperresponsiveness: interactions with passive smoke exposure. J Allergy Clin Immunol 2005;115:1169–1175.
 
79.
Colilla S, Nicolae D, Pluzhnikov A, et al. Evidence for geneenvironment interactions in a linkage study of asthma and smoking exposure. J Allergy Clin Immunol 2003;111:840–846.
 
80.
Adcock IM, Ford P, Ito K, et al. Epigenetics and airways disease. Respir Res 2006;7:21.
 
81.
Ito K, Barnes PJ, Adcock IM. Glucocorticoid receptor recruitment of histone deacetylase 2 inhibits interleukin-1betainduced histone H4 acetylation on lysines 8 and 12. Mol Cell Biol 2000;20:6891-6903.
 
82.
Ito K, Yamamura S, Essilfie-Quaye S, et al. Histone deacetylase 2-mediated deacetylation of the glucocorticoid receptor enables NF-kappaB suppression. J Exp Med 2006;203:7-13.
 
83.
Hew M, Bhavsar P, Torreqo P, et al. Relative corticosteroid insensitivity of peripheral blood mononuclear cells in severe asthma. Am J Respir Crit Care Med 2006;174:134-141.
 
84.
Ito K, Ito M, Elliot WM, et al. Decreased histone deacetylase activity in chronic obstructive pulmonary disease. N Engl J Med 2005;352:1967-1976.
 
85.
Ito K, Caramori G, Lim S, et al. Expression and activity of histone deacetylases in human asthmatic airways. Am J Respir Crit Care Med 2002;166:392-396.
 
86.
Cosío BG, Mann B, Ito K, et al. Histone acetylase and deacetylase activity in alveolar macrophages and blood mononocytes in asthma. Am J Respir Crit Care Med 2004;170:141-147.
 
87.
Kumar-R, Hitchins MP, Foster PS. Epigenetic changes in childhood asthma. Dis Mod Mechan 2009; 2:549-553.
 
88.
Breton CV, Byun HM, Wenten M, et al. Prenatal tobacco smoke exposure affects global and gene-specific DNA methylation. Am J Respir Crit Care Med 2009; 180: 462–467.
 
89.
Jiang X. The emerging role of microRNAs in asthma. Mol Cell Biochem 2011 Mar 6 [E-pub ahead of print].
 
90.
Chiba Y, Misawa M. MicroRNAs and their therapeutic potential for human diseases: MiR-133a and bronchial smooth muscle hyperresponsiveness in asthma. J Pharmacol Sci 2010;114:264-26.
 
eISSN:1791-4914
ISSN:1105-848X
Journals System - logo
Scroll to top