Research articles
 

By Dr. Yohan Bosse , Dr. Don Sin , Dr. Michel Laviolette , Dr. Andrew Sandford , Dr. James Hogg , Dr. Denise Daley , Dr. Lude Franke , Dr. David Nickle , Dr. Ke Hao , Dr. Wim Timens , Prof. Dirkje Postma , Dr. Peter D Pare
Corresponding Author Dr. Peter D Pare
University of British Columbia, St Paul's Hospital, iCAPTURE Centre, - Canada V6Z 1Y6
Submitting Author Dr. Peter D Pare
Other Authors Dr. Yohan Bosse
Institut universitaire de cardiologie et de pneumologie de Quebec and ) Department of Molecular Medi, - Canada

Dr. Don Sin
University of British Columbia James Hogg Research Centre, Division of Respiratory Medicine, Departm, - Canada

Dr. Michel Laviolette
Institut universitaire de cardiologie et de pneumologie de Quebec, - Canada

Dr. Andrew Sandford
University of British Columbia James Hogg Research Centre, Division of Respiratory Medicine, Departm, - Canada

Dr. James Hogg
University of British Columbia James Hogg Research Centre, Department of Pathology and Laboratory Me, - Canada

Dr. Denise Daley
University of British Columbia James Hogg Research Centre, Division of Respiratory Medicine, Departm, - Canada

Dr. Lude Franke
Departments of Pathology and Pulmonology, University Medical Center Groningen, - Netherlands

Dr. David Nickle
Molecular Profiling and Research Informatics, Merck Research Labs, Merck , - United States of America

Dr. Ke Hao
Molecular Profiling and Research Informatics, Merck Research Labs, Merck , - United States of America

Dr. Wim Timens
Departments of Pathology and Pulmonology, University Medical Center Groningen, - Netherlands

Prof. Dirkje Postma
Departments of Pathology and Pulmonology, University Medical Center Groningen, - Netherlands

LUNG

Genome Wide Association Study, Chronic Obstructive Lung Disease, Gene Expression, Expression Quantitative Trait loci, Emphysema

Bosse Y, Sin D, Laviolette M, Sandford A, Hogg J, Daley D, et al. Hypothesis-driven Research on Genomic Data Derived from a Large Scale Lung EQTL Mapping Study. WebmedCentral LUNG 2010;1(9):WMC00724
doi: 10.9754/journal.wmc.2010.00724
No
Submitted on: 24 Sep 2010 02:38:23 AM GMT
Published on: 24 Sep 2010 07:28:52 AM GMT

Abstract


The introduction of Genome Wide Association Studies (GWAS) has allowed the identification of reproducible disease-associated loci for more than 100 complex disorders and biological traits. Although these discoveries show the power of a hypothesis-free, unbiased approach to gene association studies, the downstream effects of most of these genetic variants remain to be clarified. The simultaneous genome-wide assay of gene expression in a specific tissue from many individuals can be coupled with the interrogation of the same individuals’ genetic variation. This allows the discovery of gene variants that contribute to inter-individual differences in gene expression (tissue-specific expression of quantitative trait loci (expression QTLs or eQTLs)). Discovery of eQTLs gives rise to a new level of data for the dissection of complex traits. The drawback of coupling GWAS data to genome-wide expression data is that it increases the number of possible comparisons by several orders of magnitude and requires large sample sizes. One approach to decrease multiple comparisons is to limit the analysis to relatively few candidate genes based on biologically plausible hypotheses. To increase creditability and to avoid data dredging, the hypothesis to be tested should to be stated and registered in the public domain before the data are examined. The purpose of this report is to register a priori hypotheses to be tested on a unique data set that will soon be available. The study is based on genome wide-genotypes and genome-wide lung tissue-specific gene expression for ~1200 well phenotyped individuals.  The aim of this study is to discover lung specific eQTLs and their relationship to COPD, asthma and lung cancer phenotypes.
The following is a list of hypotheses that will be tested once the data are available.
1.Confirm candidate genes previously associated with rate of decline in lung function.
2.Replicate the top previously reported and most likely susceptibility genes for COPD.
3.Replicate genome-wide association loci for COPD and related-phenotypes.
4.Replicate susceptibility loci for lung cancer.
5.Test for overlapping susceptibility loci of COPD and lung cancer.

Hypothesis and methods


The following is a list of hypotheses that we will test. They are grouped in two main categories: 1) Studies of genetic variants without reference to gene expression, and 2) Studies of gene expression without reference to genotype. However, it should be noted that some hypotheses take advantage of both genotyping and expression data.
Studies of genetic variants without reference to gene expression
1) Confirm candidate genes previously associated with rate of decline in lung function. The NHLBI funded Lung Health Study represents one of the largest longitudinal COPD cohorts worldwide (N=5,887 smokers). The UBC group has previously reported multiple candidate gene association studies using a nested case-control design in which the allele frequencies in candidate genes in the ~300 individuals who had the fastest rate of decline in lung function over the first 5 years of the study were compared with those of the ~300 who had the slowest decline over this time course [10-24]. The odds ratios associated with the alleles and the attributable risk for being in the fast decline group are shown in Table 2. Here we propose to test these genes for association with lung function related phenotypes in the present data set. These genes were chosen based on the strength of the associations and the prevalence of the polymorphisms. SNPs present on the Illumina Human1M BeadChip covering ten kilobases up- and downstream of these 17 genes will be tested in the present study.
2) Confirm additional candidate genes previously associated with level of lung function and rate of decline in the Vlagtwedde-Vlaardingen cohort study [25-29]. This cohort contains 1,390 subjects who had 8,159 measurements of forced expiratory volume in one second (FEV1) over eight surveys. This cohort was prospectively followed for 25 years with FEV1 measurements performed every three years. The genes which were significantly associated with lung function level or decline are presented with their respective chromosomal region, rs number and in vivo function in Table 3. SNPs present on the Illumina Human1M BeadChip covering ten kilobases up- and downstream of these genes will be tested in the present study.
3) Replicate the top previously reported and most likely susceptibility genes for COPD. In addition to the above mentioned genes and SNPs associated with rate of decline of lung function, a large number of genes have been associated with COPD and related phenotypes [30] and/or have been reported to associated with COPD in a literature based meta-analysis [31]. Varying levels of evidence support the contribution of these genes to the development of COPD. By reviewing the literature, we have identified the top genes that are most likely to be true susceptibility genes for COPD (see Table 4). SNPs present on the Illumina Human1M BeadChip covering ten kilobases up- and downstream of these ten genes will be tested in the present study.
4) Replicate genome-wide association loci for COPD and related-phenotypes. The results of GWAS for COPD and lung function were recently published [32-36]. Table 5 shows the susceptibility loci that were identified. We plan to test a set of uncorrelated SNPs within these loci for COPD and lung function measures using the samples genotyped in the present study.
5) Replicate susceptibility loci for lung cancer. Recent GWAS and follow-up investigations unequivocally identified three genetic loci associated with lung cancer, namely 15q25, 5p15, and 6p21. Based on previous publications, we have selected uncorrelated SNPs strongly associated with lung cancer in these regions (Table 6). We will attempt to replicate these findings by combining the present data set and a shared control group (Illumina’s iControlDB).
6) Test for overlapping susceptibility loci of COPD and lung cancer. Since COPD is an established risk factor for the development of lung cancer, we will assess which genes impart overlapping susceptibility for COPD and lung cancer.
Studies of gene expression without reference to genotype
In addition to testing for genetic associations, we have also formulated several hypotheses regarding the relationships of gene expression to lung function and disease (sub)phenotypes. We will employ both an unbiased approach, as with steroid treatment, and a hypothesis driven approach, as with heterogeneity of COPD.
1) Effects of steroids and combined steroids with long-acting β2 agonists (LABA). Lapperre and co-workers [37] recently described the effects of a treatment intervention on the long-term course of inflammation and airway function in patients with moderate to severe COPD. The COPD cohort was followed for over 2.5 years with clinical data, airway wall biopsies and sputum. Airway wall biopsies were taken at baseline, 6 months and 30 months following treatment with fluticasone (either 6 months followed by 24 months of placebo, or 30-month treatment), fluticasone/salmeterol combination for 30 months, or placebo for 30 months. Inhaled corticosteroid therapy decreased inflammation and attenuated the decline in lung function in previously steroid-naive COPD patients with moderate to severe airflow limitation. Adding LABAs did not modify these effects [37]. We are currently performing arrays on these airway wall biopsies in order to associate these with effects of treatment. It will be of great interest to compare the outcome of this study with a large group of lung tissues in individuals either treated or not treated with the same drug regimens. Furthermore, we will assess whether specific genes that are differentially expressed are also associated with SNPs in these respective genes, as derived from GWAS findings.
2) Heterogeneity of COPD: smoking, severity and gender. COPD is a heterogeneous disease and it has been shown that gender is one factor that may drive its heterogeneity, i.e. gender differences exist in susceptibility as well as in the type and progression of disease [38-42]. We will assess gene expression in the following pathways of interest with respect to their association with COPD, in conjunction with COPD severity, current smoking, treatments and gender effects. This will be followed by association with SNPs in genes in these pathways of interest. The pathways have been chosen based on previous findings in relation to COPD pathogenesis.
a. B-cell related pathways. Hogg et al. [43] were the first to show that infiltration of the airway wall by innate and adaptive inflammatory immune cells that form lymphoid follicles are coupled to a repair and remodeling process that thickens the walls of the small airways in COPD. Additionally we found that the large airways of patients with COPD contain higher numbers of B-cells compared with healthy controls, and that the number of B cells increases with the severity of COPD [44]. When studying the peripheral blood of patients who have COPD and controls, current smokers had higher percentages of (class-switched) memory B cells than ex-smokers and never smokers, irrespective of COPD [45]. This increase in (class-switched) memory B cells in current smokers is intriguing and suggests that smoke-induced neo-antigens may be constantly induced in the lung. The negative correlation between B cells and Tregs in blood is in line with previously published observations that Tregs can suppress B cells. Future studies focusing on the presence of these (class switched) memory B cells in the lung, their antigen specificity and their interaction with Tregs are necessary to further elucidate the specific B-cell response in COPD. Furthermore, B-cell follicles with an oligoclonal, antigen-specific reaction are present in both men and mice with emphysema. We hypothesize that these B cells contribute to the inflammatory process and/or the development and perpetuation of emphysema by producing antibodies against either tobacco smoke residues or ECM components [46]. Given these findings we will investigate whether differential B- and T-reg signatures exist in lung tissue.
b. Multidrug resistance (MDR) pathway, in particular MRP1 (ABCC1). Multidrug resistance-associated protein 1 (MRP1) is a member of the ATP-binding cassette (ABC) superfamily of transporters, which transport physiologic and toxic substrates across cell membranes. MRP1 is highly expressed in lung epithelium. Since it protects against toxic compounds and oxidative stress it might play a role in protection against smoke-induced disease progression [47]. We have found reduced expression of MRP1 in lung tissue of COPD patients [48] and polymorphisms in the MRP1 (ABCC1) gene are associated with a risk for the development of COPD in two independent populations [25]. Reduced inflammatory response to cigarette smoke is observed in MRP1 deficient mice [49]. Furthermore, SNPs in MRP1 are associated with airway wall inflammation in COPD patients as well [50]. Interestingly, cigarette smoke as well as drugs used in the management of COPD symptoms affect MRP function in an epithelial cell line [51, 52]. Thus, MRP1 as well as its downstream pathway is an interesting gene to study both for expression and genetics in the context of COPD severity, treatments, smoking and gender.
3) Lung injury and repair pathways.
a. Damage and repair. Gosselink et al. [53] used real time PCR to interrogate 54 genes which are involved in lung injury and repair in the lungs of patients with varying degrees of airflow obstruction. They hypothesized that an imbalance of expression of these genes in the small airway and lung parenchyma can lead to small airway remodelling and emphysema, respectively. The expression of the 54 genes was measured in 126 paired samples of small bronchiolar tissue and surrounding lung parenchymal tissue separated by laser capture microdissection in the lungs of 63 patients who required surgery for either lung cancer or lung transplantation for very severe COPD. Gene expression was measured by quantitative polymerase chain reaction and compared to the FEV1 by linear regression analysis. Following corrections for false discovery rates expression of only 2 out of 10 genes (SERPINE2 and MMP10) increased while 8 (MMP2, ITGA1, VEGF, ADAM33, SF/HGF, TIMP2, FN1 and COL3A1) decreased in small airways in association with FEV1. In contrast, 8/12 genes (EGR1, MMP1, MMP9, MMP10, PLAU, PLAUR, TNF and IL13) increased and 4/12 (MMP2, TIMP1, COL1A1 and TGFB3) declined in the surrounding lung tissue in association with progression of COPD. Table 7 shows the genes, the p and R2 values and the direction of the relationships.
We will test for relationships between the level of expression of these genes and the FEV1% predicted of the smokers in the eQTL data set at the time of their surgery. We do not have separate measures of emphysema and airway disease in these subjects but we do have a measure of lung diffusing capacity (DLCO) from 206 current-smokers and since diffusing capacity is a measure of emphysema we will determine if the level of expression of any of the genes which were correlated with lung parenchymal destruction in the study of Gosselink et al. [53] are related to the DLCO.
b. The TGFβ-SMAD pathways. Transforming growth factor (TGF)β1, is one of the important cytokines that induces fibroblast proliferation and stimulates production of extracellular matrix (ECM) constituents like decorin, biglycan, versican and collagens. Additionally, TGFβ1 is also a chemotactic factor for many inflammatory cells involved in COPD pathogenesis [54, 55]. We have demonstrated previously that small airways of patients with severe COPD contain less decorin, an important proteoglycan of the ECM [56, 57]. In addition, our studies have shown that pulmonary fibroblasts of COPD patients with GOLD state IV (very severe COPD) produce less decorin after TGFβ stimulation than those of controls [58].
The major intracellular signalling effector of TGFß1 is the Smad protein family. We previously showed that epithelial expression of the inhibitory Smad 7 gene is significantly lower in patients with GOLD stages II and IV than in controls. The expression of TGFß1 and TGFß receptor type I was significantly lower in stage II patients, whereas decorin staining of the adventitia and alveolar walls was significantly lower in COPD stage IV. Therefore we put forward that the TGFß1 - Smad pathway is aberrantly expressed in COPD, leading to abnormal tissue repair that ultimately results in reduced decorin production. We subsequently showed that the fibroblasts of COPD patients and controls differ in their regulation of the Smad pathway, the contrast being most pronounced during exposure to cigarette smoke. Smad3, 4 and 7 have to be considered as important factors in the defective repair process of COPD fibroblasts, since smoke exposure affects expression of these genes in COPD but not in control fibroblasts [59, 60]. Our preliminary data (unpublished) have shown that these effects may be changed by steroid treatment, whilst Chen et al. [61] have shown that VEGF production is affected as well, possibly via the SMAD pathway.
Similar as the pathways above the TGFβ and SMAD-pathway will be studied in conjunction with COPD severity, current smoking, treatments and gender effects.
4) Replicate previous molecular signatures of COPD and emphysema. A number of gene expression profiling experiments in human lung tissues have been conducted to identify molecular signatures for COPD and emphysema and/or for level of lung function. We will attempt to validate these molecular signatures in our data set using the Gene Set Enrichment Analysis (GSEA) program [62]. Briefly, genes tested in our microarray experiment will be ordered according to their differential expression between disease classes (cases and controls). Molecular signatures (or gene sets) derived from previous publications will be tested against our ranked list using the GSEA program. This analysis will determine whether the members of the gene sets are randomly distributed throughout our ranked list or primarily found at the top or bottom. Table 8 shows the gene sets that will be tested.
5) Genes differentially expressed in asthma. Laprise et al. [63] have conducted a microarray experiment comparing gene expression levels in bronchial biopsies of nonallergic healthy controls and patients with allergic asthma. They identified 74 genes that show significant differences in expression in asthmatics compared to controls. More recently, a second microarray experiment identified 40 genes differentially expressed in bronchial biopsies of patient with or without asthma [64]. Despite the use of different subjects, microarrays, and data analyses, 17 genes overlapped between the first and the second microarray experiments (Table 9). We hypothesize that these 17 genes will also be differentially expressed in the same direction in tumor-free lung tissues of asthmatics compared to non-asthmatics.
6) Replicate previous molecular signatures of lung cancer. Gene expression profiling with microarrays has the ability to clarify disease subtypes and could aid in prognosis and choice of therapies. By analyzing mRNA expression levels of human lung tumors, Bhattacharjee et al. [65] demonstrated that major lung cancer classes (small-cell lung carcinomas, adenocarcinomas, squamous cell carcinomas, and large-cell carcinomas) can be defined by a unique set of marker genes. They were also able to generate a subclassification of lung adenocarcinomas and detect metastases of extra-pulmonary origin from the microarray gene expression data. It has also been demonstrated that cancer-specific gene expression signatures can be detected in histologically normal large-airway epithelial cells from patients with lung cancer [66]. Accordingly, we propose that the molecular signatures defining lung cancer subtypes found in tumors will also be present in histologically normal lung tissues of patients with carcinomas.
7) Molecular signature of smoking. Molecular signatures of smoking have been identified in human airway epithelial cells. Spira et al. [67] obtained bronchial airway epithelial cells from brushings and identified 97 genes differentially expressed between current and never smokers. They also demonstrated that the vast majority of these genes return to normal levels in former smokers after 2 years of smoking cessation. Interestingly, a small list of smoking-induced genes (n = 13) were irreversibly altered by cigarette smoke. Boelens et al. [68] have also identified a molecular signature of smoking consisting of 246 genes differentially expressed in laser-microdissected bronchial epithelium between current and ex-smokers. Whether these reversible and irreversible molecular signatures of smoking are observed in the whole lung remained to be tested. The large number of former smokers in the lung eQTL data set, as well as the wide distribution in the number of years of smoking cessation among them, will provide a unique opportunity to differentiate genes that respond favourably from those that are resistant to a smoking cessation regimen.
8) Replication of >200 trans-eQTLs that have been detected in peripheral blood. Recently we have completed a meta-analysis on 1,469 unrelated individuals for whom we have genome-wide genotype and peripheral blood gene expression data. By applying a novel statistical method [69] that enables the removal of expression differences due to physiological effects we have been able to identify over 200 human trans-eQTLs. Some of these trans-eQTLs are clearly blood specific (as supported by very recent GWAS studies that assessed haematological traits), while others seem not to be tissue specific. We will compare lung eQTLs with these data to assess what the proportion of peripheral blood trans-eQTLs is present and to assess which additional trans-eQTLs can be identified when doing a combined meta-analysis (which will increase statistical power considerably).We expect that through meta-analysis we should be able to identify many genetic variants that affect gene-expression levels in trans, which is likely to result in the identification of previously unknown pathways.

Conclusion(s)


This manuscript proposes a series of replication and pathways analyses which will be conducted on a large and unique data set. The intent of this manuscript is to state, up front, the hypotheses that will be tested a priori. We will also analyze the data set in an unbiased manner to discover new associations and develop new hypotheses. However it seems parsimonious to utilize the data using a different statistical approach, to validate previously reported associations and correlations of genetic variation and gene expression with lung phenotypes.
The strategy will enable us to 1) replicate genetic associations with disease, 2) replicate reported gene expression profiles with disease, 3) dissect gene expression profiles for disease heterogeneity and treatments, and 4) to detect whether the gene expression- disease association is linked by underlying genetic mechanisms. The analyses will help to provide insight into the biological pathways affected by these diseases and unravel the regulatory networks underlying disease development and disease heterogeneity. It will help to identify new targets for disease detection and prevention. In this collaborative research effort, we will use a combination of hypothesis driven and unbiased approaches to investigate the rich tissue samples we have combined from three academic centers. This strategy will increase the power to ultimately find new targets for disease intervention.

References


1. Hindorff LA, Junkins HA, Mehta JP, Manolio TA: A catalog of published genome-wide association studies. National Human Genome Research Institute [online],  (accessed 1 Feb 2010).
2. Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS, Manolio TA: Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A 2009, 106(23):9362-9367.
3. Hirschhorn JN: Genomewide association studies--illuminating biologic pathways. N Engl J Med 2009, 360(17):1699-1701.
4. Moffatt MF, Kabesch M, Liang L, Dixon AL, Strachan D, Heath S, Depner M, von Berg A, Bufe A, Rietschel E et al: Genetic variants regulating ORMDL3 expression contribute to the risk of childhood asthma. Nature 2007, 448(7152):470-473.
5. Hunt KA, Zhernakova A, Turner G, Heap GA, Franke L, Bruinenberg M, Romanos J, Dinesen LC, Ryan AW, Panesar D et al: Newly identified genetic risk variants for celiac disease related to the immune response. Nat Genet 2008, 40(4):395-402.
6. Schadt EE, Molony C, Chudin E, Hao K, Yang X, Lum PY, Kasarskis A, Zhang B, Wang S, Suver C et al: Mapping the genetic architecture of gene expression in human liver. PLoS Biol 2008, 6(5):e107.
7. Huang RS, Duan S, Bleibel WK, Kistner EO, Zhang W, Clark TA, Chen TX, Schweitzer AC, Blume JE, Cox NJ et al: A genome-wide approach to identify genetic variants that contribute to etoposide-induced cytotoxicity. Proc Natl Acad Sci U S A 2007, 104(23):9758-9763.
8. Emilsson V, Thorleifsson G, Zhang B, Leonardson AS, Zink F, Zhu J, Carlson S, Helgason A, Walters GB, Gunnarsdottir S et al: Genetics of gene expression and its effect on disease. Nature 2008, 452(7186):423-428.
9. Cookson W, Liang L, Abecasis G, Moffatt M, Lathrop M: Mapping complex disease traits with global gene expression. Nat Rev Genet 2009, 10(3):184-194.
10. He JQ, Burkett K, Connett JE, Anthonisen NR, Pare PD, Sandford AJ: Interferon gamma polymorphisms and their interaction with smoking are associated with lung function. Hum Genet 2006, 119(4):365-375.
11. He JQ, Connett JE, Anthonisen NR, Pare PD, Sandford AJ: Glutathione S-transferase variants and their interaction with smoking on lung function. Am J Respir Crit Care Med 2004, 170(4):388-394.
12. He JQ, Connett JE, Anthonisen NR, Sandford AJ: Polymorphisms in the IL13, IL13RA1, and IL4RA genes and rate of decline in lung function in smokers. American journal of respiratory cell and molecular biology 2003, 28(3):379-385.
13. He JQ, Foreman MG, Shumansky K, Zhang X, Akhabir L, Sin DD, Man SF, DeMeo DL, Litonjua AA, Silverman EK et al: Associations of IL6 polymorphisms with lung function decline and COPD. Thorax 2009, 64(8):698-704.
14. He JQ, Ruan J, Connett JE, Anthonisen NR, Pare PD, Sandford AJ: Antioxidant gene polymorphisms and susceptibility to a rapid decline in lung function in smokers. Am J Respir Crit Care Med 2002, 166(3):323-328.
15. He JQ, Shumansky K, Connett JE, Anthonisen NR, Pare PD, Sandford AJ: Association of genetic variations in the CSF2 and CSF3 genes with lung function in smoking-induced COPD. Eur Respir J 2008, 32(1):25-34.
16. He JQ, Shumansky K, Zhang X, Connett JE, Anthonisen NR, Sandford AJ: Polymorphisms of interleukin-10 and its receptor and lung function in COPD. Eur Respir J 2007, 29(6):1120-1126.
17. Joos L, He JQ, Shepherdson MB, Connett JE, Anthonisen NR, Pare PD, Sandford AJ: The role of matrix metalloproteinase polymorphisms in the rate of decline in lung function. Human molecular genetics 2002, 11(5):569-576.
18. Joos L, McIntyre L, Ruan J, Connett JE, Anthonisen NR, Weir TD, Pare PD, Sandford AJ: Association of IL-1beta and IL-1 receptor antagonist haplotypes with rate of decline in lung function in smokers. Thorax 2001, 56(11):863-866.
19. Joos L, Weir TD, Connett JE, Anthonisen NR, Woods R, Pare PD, Sandford AJ: Polymorphisms in the beta2 adrenergic receptor and bronchodilator response, bronchial hyperresponsiveness, and rate of decline in lung function in smokers. Thorax 2003, 58(8):703-707.
20. Kasuga I, Pare PD, Ruan J, Connett JE, Anthonisen NR, Sandford AJ: Lack of association of group specific component haplotypes with lung function in smokers. Thorax 2003, 58(9):790-793.
21. Sandford AJ, Chagani T, Weir TD, Connett JE, Anthonisen NR, Pare PD: Susceptibility genes for rapid decline of lung function in the lung health study. Am J Respir Crit Care Med 2001, 163(2):469-473.
22. Tanaka G, Sandford AJ, Burkett K, Connett JE, Anthonisen NR, Pare PD, He JQ: Tumour necrosis factor and lymphotoxin A polymorphisms and lung function in smokers. Eur Respir J 2007, 29(1):34-41.
23. Wallace AM, He JQ, Burkett KM, Ruan J, Connett JE, Anthonisen NR, Pare PD, Sandford AJ: Contribution of alpha- and beta-defensins to lung function decline and infection in smokers: an association study. Respir Res 2006, 7:76.
24. Zhang X, Mahmudi-Azer S, Connett JE, Anthonisen NR, He JQ, Pare PD, Sandford AJ: Association of Hck genetic polymorphisms with gene expression and COPD. Hum Genet 2007, 120(5):681-690.
25. Siedlinski M, Boezen HM, Boer JM, Smit HA, Postma DS: ABCC1 polymorphisms contribute to level and decline of lung function in two population-based cohorts. Pharmacogenet Genomics 2009, 19(9):675-684.
26. Siedlinski M, Postma DS, Boer JM, van der Steege G, Schouten JP, Smit HA, Boezen HM: Level and course of FEV1 in relation to polymorphisms in NFE2L2 and KEAP1 in the general population. Respir Res 2009, 10:73.
27. Siedlinski M, Postma DS, van Diemen CC, Blokstra A, Smit HA, Boezen HM: Lung function loss, smoking, vitamin C intake, and polymorphisms of the glutamate-cysteine ligase genes. Am J Respir Crit Care Med 2008, 178(1):13-19.
28. Siedlinski M, van Diemen CC, Postma DS, Boezen HM: Heme oxygenase 1 variations and lung function decline in smokers: proof of replication. J Med Genet 2008, 45(6):400.
29. Siedlinski M, van Diemen CC, Postma DS, Vonk JM, Boezen HM: Superoxide dismutases, lung function and bronchial responsiveness in a general population. Eur Respir J 2009, 33(5):986-992.
30. Bosse Y: Genetics of chronic obstructive pulmonary disease: a succinct review, future avenues and prospective clinical applications. Pharmacogenomics 2009, 10(4):655-667.
31. Smolonska J, Wijmenga C, Postma DS, Boezen HM: Meta-analyses on suspected chronic obstructive pulmonary disease genes: a summary of 20 years' research. Am J Respir Crit Care Med 2009, 180(7):618-631.
32. Hancock DB, Eijgelsheim M, Wilk JB, Gharib SA, Loehr LR, Marciante KD, Franceschini N, van Durme YM, Chen TH, Barr RG et al: Meta-analyses of genome-wide association studies identify multiple loci associated with pulmonary function. Nat Genet, 42(1):45-52.
33. Pillai SG, Ge D, Zhu G, Kong X, Shianna KV, Need AC, Feng S, Hersh CP, Bakke P, Gulsvik A et al: A genome-wide association study in chronic obstructive pulmonary disease (COPD): identification of two major susceptibility loci. PLoS Genet 2009, 5(3):e1000421.
34. Repapi E, Sayers I, Wain LV, Burton PR, Johnson T, Obeidat M, Zhao JH, Ramasamy A, Zhai G, Vitart V et al: Genome-wide association study identifies five loci associated with lung function. Nat Genet, 42(1):36-44.
35. Wilk JB, Chen TH, Gottlieb DJ, Walter RE, Nagle MW, Brandler BJ, Myers RH, Borecki IB, Silverman EK, Weiss ST et al: A genome-wide association study of pulmonary function measures in the Framingham Heart Study. PLoS Genet 2009, 5(3):e1000429.
36. Cho MH, Boutaoui N, Klanderman BJ, Sylvia JS, Ziniti JP, Hersh CP, DeMeo DL, Hunninghake GM, Litonjua AA, Sparrow D et al: Variants in FAM13A are associated with chronic obstructive pulmonary disease. Nat Genet, 42(3):200-202.
37. Lapperre TS, Snoeck-Stroband JB, Gosman MM, Jansen DF, van Schadewijk A, Thiadens HA, Vonk JM, Boezen HM, Ten Hacken NH, Sont JK et al: Effect of fluticasone with and without salmeterol on pulmonary outcomes in chronic obstructive pulmonary disease: a randomized trial. Ann Intern Med 2009, 151(8):517-527.
38. Dransfield MT, Washko GR, Foreman MG, Estepar RS, Reilly J, Bailey WC: Gender differences in the severity of CT emphysema in COPD. Chest 2007, 132(2):464-470.
39. Han MK, Postma D, Mannino DM, Giardino ND, Buist S, Curtis JL, Martinez FJ: Gender and chronic obstructive pulmonary disease: why it matters. Am J Respir Crit Care Med 2007, 176(12):1179-1184.
40. Soriano JB, Sin DD, Zhang X, Camp PG, Anderson JA, Anthonisen NR, Buist AS, Burge PS, Calverley PM, Connett JE et al: A pooled analysis of FEV1 decline in COPD patients randomized to inhaled corticosteroids or placebo. Chest 2007, 131(3):682-689.
41. Gan WQ, Man SF, Postma DS, Camp P, Sin DD: Female smokers beyond the perimenopausal period are at increased risk of chronic obstructive pulmonary disease: a systematic review and meta-analysis. Respir Res 2006, 7:52.
42. Watson L, Vonk JM, Lofdahl CG, Pride NB, Pauwels RA, Laitinen LA, Schouten JP, Postma DS: Predictors of lung function and its decline in mild to moderate COPD in association with gender: results from the Euroscop study. Respir Med 2006, 100(4):746-753.
43. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO et al: The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med 2004, 350(26):2645-2653.
44. Gosman MM, Willemse BW, Jansen DF, Lapperre TS, van Schadewijk A, Hiemstra PS, Postma DS, Timens W, Kerstjens HA: Increased number of B-cells in bronchial biopsies in COPD. Eur Respir J 2006, 27(1):60-64.
45. Brandsma CA, Hylkema MN, Geerlings M, van Geffen WH, Postma DS, Timens W, Kerstjens HA: Increased levels of (class switched) memory B cells in peripheral blood of current smokers. Respir Res 2009, 10:108.
46. van der Strate BW, Postma DS, Brandsma CA, Melgert BN, Luinge MA, Geerlings M, Hylkema MN, van den Berg A, Timens W, Kerstjens HA: Cigarette smoke-induced emphysema: A role for the B cell? Am J Respir Crit Care Med 2006, 173(7):751-758.
47. van der Deen M, de Vries EG, Timens W, Scheper RJ, Timmer-Bosscha H, Postma DS: ATP-binding cassette (ABC) transporters in normal and pathological lung. Respir Res 2005, 6:59.
48. van der Deen M, Marks H, Willemse BW, Postma DS, Muller M, Smit EF, Scheffer GL, Scheper RJ, de Vries EG, Timens W: Diminished expression of multidrug resistance-associated protein 1 (MRP1) in bronchial epithelium of COPD patients. Virchows Arch 2006, 449(6):682-688.
49. van der Deen M, Timens W, Timmer-Bosscha H, van der Strate BW, Scheper RJ, Postma DS, de Vries EG, Kerstjens HA: Reduced inflammatory response in cigarette smoke exposed Mrp1/Mdr1a/1b deficient mice. Respir Res 2007, 8:49.
50. Budulac SE, Postma DS, Hiemstra PS, Kunz LI, Siedlinski M, Smit HA, Vonk JM, Rutgers B, Timens W, Boezen HM et al: Multidrug resistance-associated protein-1 (MRP1) genetic variants, MRP1 protein levels and severity of COPD. Respir Res, 11(1):60.
51. van der Deen M, Homan S, Timmer-Bosscha H, Scheper RJ, Timens W, Postma DS, de Vries EG: Effect of COPD treatments on MRP1-mediated transport in bronchial epithelial cells. Int J Chron Obstruct Pulmon Dis 2008, 3(3):469-475.
52. van der Deen M, de Vries EG, Visserman H, Zandbergen W, Postma DS, Timens W, Timmer-Bosscha H: Cigarette smoke extract affects functional activity of MRP1 in bronchial epithelial cells. J Biochem Mol Toxicol 2007, 21(5):243-251.
53. Gosselink JV, Hayashi S, Elliott WM, Xing L, Chan B, Yang L, Wright C, Sin D, Pare PD, Pierce JA et al: Differential Expression of Tissue Repair Genes in the Pathogenesis of COPD. Am J Respir Crit Care Med.
54. Wahl SM, Hunt DA, Wakefield LM, McCartney-Francis N, Wahl LM, Roberts AB, Sporn MB: Transforming growth factor type beta induces monocyte chemotaxis and growth factor production. Proc Natl Acad Sci U S A 1987, 84(16):5788-5792.
55. Gruber BL, Marchese MJ, Kew RR: Transforming growth factor-beta 1 mediates mast cell chemotaxis. J Immunol 1994, 152(12):5860-5867.
56. van Straaten JF, Coers W, Noordhoek JA, Huitema S, Flipsen JT, Kauffman HF, Timens W, Postma DS: Proteoglycan changes in the extracellular matrix of lung tissue from patients with pulmonary emphysema. Mod Pathol 1999, 12(7):697-705.
57. Van der Geld YM, Van Straaten JF, Postma DS, Timens W: Role of proteoglycans in development and pathogenesis of emphysema. In: Proteoglycans in Lung Disease. Edited by Garg HG, Roughley PJ, Hales PJ. New York: Marcel Dekker, Inc.; 2002: 241-267.
58. Noordhoek JA, Postma DS, Chong LL, Menkema L, Kauffman HF, Timens W, van Straaten JF, van der Geld YM: Different modulation of decorin production by lung fibroblasts from patients with mild and severe emphysema. COPD 2005, 2(1):17-25.
59. Zandvoort A, Postma DS, Jonker MR, Noordhoek JA, Vos JT, Timens W: Smad gene expression in pulmonary fibroblasts: indications for defective ECM repair in COPD. Respir Res 2008, 9:83.
60. Zandvoort A, Postma DS, Jonker MR, Noordhoek JA, Vos JT, van der Geld YM, Timens W: Altered expression of the Smad signalling pathway: implications for COPD pathogenesis. Eur Respir J 2006, 28(3):533-541.
61. Chen YJ, Wang T, Liu DS, Xu D, Chen L, An J, Wen FQ: Effect of Budesonide on TGF-beta1 enhanced VEGF Production by Lung Fibroblasts. Cell Biol Int.
62. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES et al: Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 2005, 102(43):15545-15550.
63. Laprise C, Sladek R, Ponton A, Bernier MC, Hudson TJ, Laviolette M: Functional classes of bronchial mucosa genes that are differentially expressed in asthma. BMC Genomics 2004, 5(1):21.
64. Chamberland A, Madore AM, Tremblay K, Laviolette M, Laprise C: A comparison of two sets of microarray experiments to define allergic asthma expression pattern. Exp Lung Res 2009, 35(5):399-410.
65. Bhattacharjee A, Richards WG, Staunton J, Li C, Monti S, Vasa P, Ladd C, Beheshti J, Bueno R, Gillette M et al: Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci U S A 2001, 98(24):13790-13795.
66. Spira A, Beane JE, Shah V, Steiling K, Liu G, Schembri F, Gilman S, Dumas YM, Calner P, Sebastiani P et al: Airway epithelial gene expression in the diagnostic evaluation of smokers with suspect lung cancer. Nat Med 2007, 13(3):361-366.
67. Spira A, Beane J, Shah V, Liu G, Schembri F, Yang X, Palma J, Brody JS: Effects of cigarette smoke on the human airway epithelial cell transcriptome. Proc Natl Acad Sci U S A 2004, 101(27):10143-10148.
68. Boelens MC, van den Berg A, Fehrmann RS, Geerlings M, de Jong WK, te Meerman GJ, Sietsma H, Timens W, Postma DS, Groen HJ: Current smoking-specific gene expression signature in normal bronchial epithelium is enhanced in squamous cell lung cancer. J Pathol 2009, 218(2):182-191.
69. Dubois PC, Trynka G, Franke L, Hunt KA, Romanos J, Curtotti A, Zhernakova A, Heap GA, Adany R, Aromaa A et al: Multiple common variants for celiac disease influencing immune gene expression. Nat Genet, 42(4):295-302.
70. Laurell CB, Eriksson S: The electrophoretic alpha1-globulin pattern of serum in alpha1-antitrypsin deficiency. Scand J Clin Lab Invest 1963, 15:132-140.
71. Demeo DL, Mariani TJ, Lange C, Srisuma S, Litonjua AA, Celedon JC, Lake SL, Reilly JJ, Chapman HA, Mecham BH et al: The SERPINE2 gene is associated with chronic obstructive pulmonary disease. Am J Hum Genet 2006, 78(2):253-264.
72. Zhu G, Warren L, Aponte J, Gulsvik A, Bakke P, Anderson WH, Lomas DA, Silverman EK, Pillai SG: The SERPINE2 gene is associated with chronic obstructive pulmonary disease in two large populations. Am J Respir Crit Care Med 2007, 176(2):167-173.
73. Wilk JB, Herbert A, Shoemaker CM, Gottlieb DJ, Karamohamed S: Secreted modular calcium-binding protein 2 haplotypes are associated with pulmonary function. Am J Respir Crit Care Med 2007, 175(6):554-560.
74. Celedon JC, Lange C, Raby BA, Litonjua AA, Palmer LJ, DeMeo DL, Reilly JJ, Kwiatkowski DJ, Chapman HA, Laird N et al: The transforming growth factor-beta1 (TGFB1) gene is associated with chronic obstructive pulmonary disease (COPD). Human molecular genetics 2004, 13(15):1649-1656.
75. Hersh CP, Demeo DL, Lazarus R, Celedon JC, Raby BA, Benditt JO, Criner G, Make B, Martinez FJ, Scanlon PD et al: Genetic association analysis of functional impairment in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2006, 173(9):977-984.
76. Hu G, Yao W, Zhou Y, Hu J, Shi Z, Li B, Ran P: Meta- and pooled analyses of the effect of glutathione S-transferase M1 and T1 deficiency on chronic obstructive pulmonary disease. Int J Tuberc Lung Dis 2008, 12(12):1474-1481.
77. Wilk JB, Walter RE, Laramie JM, Gottlieb DJ, O'Connor GT: Framingham Heart Study genome-wide association: results for pulmonary function measures. BMC medical genetics 2007, 8 Suppl 1:S8.
78. Gingo MR, Silveira LJ, Miller YE, Friedlander AL, Cosgrove GP, Chan ED, Maier LA, Bowler RP: Tumour necrosis factor gene polymorphisms are associated with COPD. Eur Respir J 2008, 31(5):1005-1012.
79. Hersh CP, Demeo DL, Lange C, Litonjua AA, Reilly JJ, Kwiatkowski D, Laird N, Sylvia JS, Sparrow D, Speizer FE et al: Attempted replication of reported chronic obstructive pulmonary disease candidate gene associations. American journal of respiratory cell and molecular biology 2005, 33(1):71-78.
80. Hu G, Shi Z, Hu J, Zou G, Peng G, Ran P: Association between polymorphisms of microsomal epoxide hydrolase and COPD: results from meta-analyses. Respirology 2008, 13(6):837-850.
81. Hunninghake GM, Cho MH, Tesfaigzi Y, Soto-Quiros ME, Avila L, Lasky-Su J, Stidley C, Melen E, Soderhall C, Hallberg J et al: MMP12, lung function, and COPD in high-risk populations. N Engl J Med 2009, 361(27):2599-2608.
82. Amos CI, Wu X, Broderick P, Gorlov IP, Gu J, Eisen T, Dong Q, Zhang Q, Gu X, Vijayakrishnan J et al: Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1. Nat Genet 2008, 40(5):616-622.
83. Broderick P, Wang Y, Vijayakrishnan J, Matakidou A, Spitz MR, Eisen T, Amos CI, Houlston RS: Deciphering the impact of common genetic variation on lung cancer risk: a genome-wide association study. Cancer Res 2009, 69(16):6633-6641.
84. Hung RJ, McKay JD, Gaborieau V, Boffetta P, Hashibe M, Zaridze D, Mukeria A, Szeszenia-Dabrowska N, Lissowska J, Rudnai P et al: A susceptibility locus for lung cancer maps to nicotinic acetylcholine receptor subunit genes on 15q25. Nature 2008, 452(7187):633-637.
85. Landi MT, Chatterjee N, Yu K, Goldin LR, Goldstein AM, Rotunno M, Mirabello L, Jacobs K, Wheeler W, Yeager M et al: A genome-wide association study of lung cancer identifies a region of chromosome 5p15 associated with risk for adenocarcinoma. Am J Hum Genet 2009, 85(5):679-691.
86. Liu P, Vikis HG, Wang D, Lu Y, Wang Y, Schwartz AG, Pinney SM, Yang P, de Andrade M, Petersen GM et al: Familial aggregation of common sequence variants on 15q24-25.1 in lung cancer. J Natl Cancer Inst 2008, 100(18):1326-1330.
87. McKay JD, Hung RJ, Gaborieau V, Boffetta P, Chabrier A, Byrnes G, Zaridze D, Mukeria A, Szeszenia-Dabrowska N, Lissowska J et al: Lung cancer susceptibility locus at 5p15.33. Nat Genet 2008, 40(12):1404-1406.
88. Thorgeirsson TE, Geller F, Sulem P, Rafnar T, Wiste A, Magnusson KP, Manolescu A, Thorleifsson G, Stefansson H, Ingason A et al: A variant associated with nicotine dependence, lung cancer and peripheral arterial disease. Nature 2008, 452(7187):638-642.
89. Wang Y, Broderick P, Webb E, Wu X, Vijayakrishnan J, Matakidou A, Qureshi M, Dong Q, Gu X, Chen WV et al: Common 5p15.33 and 6p21.33 variants influence lung cancer risk. Nat Genet 2008, 40(12):1407-1409.
90. Ning W, Li CJ, Kaminski N, Feghali-Bostwick CA, Alber SM, Di YP, Otterbein SL, Song R, Hayashi S, Zhou Z et al: Comprehensive gene expression profiles reveal pathways related to the pathogenesis of chronic obstructive pulmonary disease. Proc Natl Acad Sci U S A 2004, 101(41):14895-14900.
91. Spira A, Beane J, Pinto-Plata V, Kadar A, Liu G, Shah V, Celli B, Brody JS: Gene expression profiling of human lung tissue from smokers with severe emphysema. American journal of respiratory cell and molecular biology 2004, 31(6):601-610.
92. Golpon HA, Coldren CD, Zamora MR, Cosgrove GP, Moore MD, Tuder RM, Geraci MW, Voelkel NF: Emphysema lung tissue gene expression profiling. American journal of respiratory cell and molecular biology 2004, 31(6):595-600.
93. Francis SM, Larsen JE, Pavey SJ, Bowman RV, Hayward NK, Fong KM, Yang IA: Expression profiling identifies genes involved in emphysema severity. Respir Res 2009, 10:81.
94. Bhattacharya S, Srisuma S, Demeo DL, Reilly JJ, Bueno R, Silverman EK, Mariani TJ: Microarray data-based prioritization of chronic obstructive pulmonary disease susceptibility genes. Proc Am Thorac Soc 2006, 3(6):472.
95. Wang IM, Stepaniants S, Boie Y, Mortimer JR, Kennedy B, Elliott M, Hayashi S, Loy L, Coulter,    Cervino S et al: Gene expression profiling in patients with chronic obstructive pulmonary disease and lung cancer. Am J Respir Crit Care Med 2008, 177(4):402-411.

Source(s) of Funding


Funding for this study was provided by Merck

Competing Interests


none

Disclaimer


This article has been downloaded from WebmedCentral. With our unique author driven post publication peer review, contents posted on this web portal do not undergo any prepublication peer or editorial review. It is completely the responsibility of the authors to ensure not only scientific and ethical standards of the manuscript but also its grammatical accuracy. Authors must ensure that they obtain all the necessary permissions before submitting any information that requires obtaining a consent or approval from a third party. Authors should also ensure not to submit any information which they do not have the copyright of or of which they have transferred the copyrights to a third party.
Contents on WebmedCentral are purely for biomedical researchers and scientists. They are not meant to cater to the needs of an individual patient. The web portal or any content(s) therein is neither designed to support, nor replace, the relationship that exists between a patient/site visitor and his/her physician. Your use of the WebmedCentral site and its contents is entirely at your own risk. We do not take any responsibility for any harm that you may suffer or inflict on a third person by following the contents of this website.

Comments
0 comments posted so far

Please use this functionality to flag objectionable, inappropriate, inaccurate, and offensive content to WebmedCentral Team and the authors.

 

Author Comments
0 comments posted so far

 

What is article Popularity?

Article popularity is calculated by considering the scores: age of the article
Popularity = (P - 1) / (T + 2)^1.5
Where
P : points is the sum of individual scores, which includes article Views, Downloads, Reviews, Comments and their weightage

Scores   Weightage
Views Points X 1
Download Points X 2
Comment Points X 5
Review Points X 10
Points= sum(Views Points + Download Points + Comment Points + Review Points)
T : time since submission in hours.
P is subtracted by 1 to negate submitter's vote.
Age factor is (time since submission in hours plus two) to the power of 1.5.factor.

How Article Quality Works?

For each article Authors/Readers, Reviewers and WMC Editors can review/rate the articles. These ratings are used to determine Feedback Scores.

In most cases, article receive ratings in the range of 0 to 10. We calculate average of all the ratings and consider it as article quality.

Quality=Average(Authors/Readers Ratings + Reviewers Ratings + WMC Editor Ratings)