Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2018

Open Access 01.12.2018 | Research article

RNAseq analysis of bronchial epithelial cells to identify COPD-associated genes and SNPs

verfasst von: Jiyoun Yeo, Diego A. Morales, Tian Chen, Erin L. Crawford, Xiaolu Zhang, Thomas M. Blomquist, Albert M. Levin, Pierre P. Massion, Douglas A. Arenberg, David E. Midthun, Peter J. Mazzone, Steven D. Nathan, Ronald J. Wainz, Patrick Nana-Sinkam, Paige F. S. Willey, Taylor J. Arend, Karanbir Padda, Shuhao Qiu, Alexei Federov, Dawn-Alita R. Hernandez, Jeffrey R. Hammersley, Youngsook Yoon, Fadi Safi, Sadik A. Khuder, James C. Willey

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2018

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Background

There is a need for more powerful methods to identify low-effect SNPs that contribute to hereditary COPD pathogenesis. We hypothesized that SNPs contributing to COPD risk through cis-regulatory effects are enriched in genes comprised by bronchial epithelial cell (BEC) expression patterns associated with COPD.

Methods

To test this hypothesis, normal BEC specimens were obtained by bronchoscopy from 60 subjects: 30 subjects with COPD defined by spirometry (FEV1/FVC < 0.7, FEV1% < 80%), and 30 non-COPD controls. Targeted next generation sequencing was used to measure total and allele-specific expression of 35 genes in genome maintenance (GM) genes pathways linked to COPD pathogenesis, including seven TP53 and CEBP transcription factor family members. Shrinkage linear discriminant analysis (SLDA) was used to identify COPD-classification models. COPD GWAS were queried for putative cis-regulatory SNPs in the targeted genes.

Results

On a network basis, TP53 and CEBP transcription factor pathway gene pair network connections, including key DNA repair gene ERCC5, were significantly different in COPD subjects (e.g., Wilcoxon rank sum test for closeness, p-value = 5.0E-11). ERCC5 SNP rs4150275 association with chronic bronchitis was identified in a set of Lung Health Study (LHS) COPD GWAS SNPs restricted to those in putative regulatory regions within the targeted genes, and this association was validated in the COPDgene non-hispanic white (NHW) GWAS. ERCC5 SNP rs4150275 is linked (D’ = 1) to ERCC5 SNP rs17655 which displayed differential allelic expression (DAE) in BEC and is an expression quantitative trait locus (eQTL) in lung tissue (p = 3.2E-7). SNPs in linkage (D’ = 1) with rs17655 were predicted to alter miRNA binding (rs873601). A classifier model that comprised gene features CAT, CEBPG, GPX1, KEAP1, TP73, and XPA had pooled 10-fold cross-validation receiver operator characteristic area under the curve of 75.4% (95% CI: 66.3%–89.3%). The prevalence of DAE was higher than expected (p = 0.0023) in the classifier genes.

Conclusions

GM genes comprised by COPD-associated BEC expression patterns were enriched for SNPs with cis-regulatory function, including a putative cis-rSNP in ERCC5 that was associated with COPD risk. These findings support additional total and allele-specific expression analysis of gene pathways with high prior likelihood for involvement in COPD pathogenesis.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12890-018-0603-y) contains supplementary material, which is available to authorized users.
Abkürzungen
ANCOVA
Analysis of covariance
ASE
Allele specific expression
AUC
Area under the curve
BEC
Bronchial epithelial cell
CAT
Correlation adjusted t
CB
Chronic bronchitis
cis-rSNP
cis-regulatory single nucleotide polymorphism
COPD
Chronic obstructive pulmonary disease
DAE
Differential allelic expression
EM
Emphysema
eQTL
Expression quantitative trait loci
GM
Genome maintenance
GTEX
Genotype-Tissue Expression
GWAS
Genome-wide association studies
IRB
Institutional Review Board
IS
Internal standard
LCRT
Lung cancer risk test
LHS
Lung health study
NGS
Next generation sequencing
NHW
Non-Hispanic white
ROC
Receiver Operator Characteristic
SLDA
Shrinkage linear discriminant analysis
SNP
Single nucleotide polymorphism

Background

Tobacco smoking is the predominant exogenous risk factor for chronic obstructive pulmonary disease (COPD) [1]. However, not all smokers develop COPD, implying that genetic factors contribute to COPD predisposition and pathogenesis. COPD genome-wide association studies (GWAS) have significantly increased our understanding of COPD pathogenesis, yet genomic variants identified through GWAS still explain only a small fraction of hereditary risk [2]. Each variant identified by COPD GWAS required large sample size for detection due to small effect on heritability [3, 4]. Those that remain to be discovered will likely have an even lower effect. To address this challenge, one recent study was designed to discover rare coding variants with large effect on COPD risk, similar to that of alpha-1-antitrypsin deficiency [5]. In another approach, GWAS meta-analyses were used to identify common SNPs with very low effect through very large sample size [3, 6, 7].
The purpose of this study was to identify candidate COPD-risk variants through their role in generation of COPD-associated transcript abundance patterns in bronchial epithelial cells (BEC). BEC coat the surfaces of lung airways and protect lung tissue from the environment. Sub-optimal BEC function is implicated in COPD pathogenesis [810]. We hypothesized that hereditary risk for COPD is due to the combined effect of multiple regulatory SNPs that each contribute to generation of BEC transcript abundance patterns associated with hereditary predisposition to COPD. If the proposed hypothesis is correct, a) a study to identify COPD-associated transcript abundance patterns in BEC will have high power and require a relatively small sample size, and b) genes comprised by such patterns will be enriched for cis-regulatory (r) SNPs and possibly SNPs significant in COPD GWAS using reduced stringency correction for multiple testing. Recent knowledge regarding predisposition for diseases caused by complex genetics supports this hypothesis in that SNPs associated with COPD and other complex phenotypes are enriched for cis- and/or trans-regulatory function [1119]. For example, in a report on the recently updated GTEx database nearly 50% of common GWAS variants of interest were significantly associated with the expression of one or more genes (P < 0.05, after correcting for multiple tissue testing) [17].
A common way to identify cis-rSNPs is to measure dose effect of alleles at candidate cis-rSNPs on total expression [17]. SNPs significant by this analysis are referred to as cis-expression quantitative trait loci (eQTL). Another way to identify cis-rSNPs is by measurement of allele-specific expression (ASE) to identify differential allelic expression (DAE) [15, 20, 21]. Importantly, with appropriate methodological conditions, the power to identify cis-regulatory SNPs by measurement of ASE is higher than that for measurement of total expression [22], possibly because ASE controls for variation in trans-effects, including those resulting from variation in the environment. Importantly, there is a high correlation between cis-rSNPs identified by ASE measurement and those identified by as e-QTL by total expression [17]. The recent increase in throughput and reduction in cost of next generation sequencing (NGS) now facilitates large-scale measurement of ASE Tools and best practices for data processing in allelic expression analysis [19, 23].
We used targeted NGS technology to measure total and allele-specific expression (ASE) of 35 selected genome maintenance (GM) genes through RNA sequencing (RNAseq) of BEC RNA from 30 COPD subjects and 30 non-COPD controls. This targeted NGS method employs multiplex competitive PCR-amplicon libraries that provide excellent sequencing depth for all target analytes [24]. This approach is based on RT-PCR technology proven to ensure optimal quality-control characteristics, including high linear dynamic range, signal-to-analyte response, precision, and accuracy, and high correlation with qPCR [2433].
The targeted NGS RNAseq method used in this study simplified allele-specific transcript abundance measurement and assessment of genes for cis-regulatory (r) SNPs manifesting as DAE. Gene expression is well-documented to be a heritable trait [3436]. Heritable differences in gene expression between individuals are caused by DNA variants that affect the expression level of one allele (cis-acting) or both alleles (trans-acting) of a gene [37]. Measurement of DAE is recognized as a powerful approach for identifying cis-acting regulatory variation [27, 35, 38]. We applied this technology to assess cis-rSNP activity and disease association at the exon level. As previously reported, multivariate exon-level analysis provides a more powerful approach than univariate gene-level analysis for identification of cis-rSNPs as well as disease association [39].
The 35 genes selected for analysis have a high prior likelihood for a role in COPD pathogenesis based on studies from this laboratory as well as those from other investigators. They represent antioxidant (AO), DNA repair (DNAR), and cell cycle control (CCC) pathways that play a key role in protecting BEC from inhaled cigarette smoke and toxins from the environment or occupational exposure [8, 25, 27, 29, 30, 4043]. Importantly, there is significant inter-individual variation in BEC regulation of these key GM pathway genes [30, 40, 44, 45] and many function differently in BEC of COPD subjects compared to matched controls [8, 4143, 4654]. For example, genetic variants in CAT, GSTM1, GSTT1, GSTP1, SOD3, NFE2L2, KEAP1, OGG1, XRCC1, XRCC3, XRCC4, XRCC5, CDKN1A, and p53 are reported to be associated with COPD [5469]. Further, regulation of both CEBP and TP53 family genes as well as many of their targets is different in subjects with COPD [52, 53, 67, 70].
A focus on members of the CEBP transcription factor family (i.e., CEBPA, CEBPD and CEBPG) and TP53 transcription factor family (i.e. TP53, TP63, and TP73) was based on evidence that they play an important role in regulating lung development and differentiation, and regulation of AO, DNAR, and CCC genes in BEC [25, 29, 30, 52, 7177]. CEBPG is a truncated transcription factor that does not trans-activate but plays a regulatory role through heterodimer formation [78]. CEBPG regulates key GM genes in BEC [25, 29, 30]. CEBPA and CEBPE are anti-proliferative transcription factors that lead to cell differentiation [71, 72, 78]. CEBPB and CEBPD contribute to regulation of cell-cycle progression [7375]. CEBPG or CEBPA knockout mice die at birth or in adulthood respectively from emphysematous lungs [52, 76], providing experimental confirmation of the important role that sub-optimal function of these regulatory pathways plays in risk for COPD. The role of the TP53 gene transcription factor family in COPD is supported by association of TP53 and CDKN1A alleles with COPD risk and the role of CDKN1A in response to cigarette smoke [66, 67, 79]. TP63 plays a key role in airway epithelial cell proliferation and differentiation [8088], and is important in maintaining airway epithelial integrity and repair [89]. TP53 and TP73 work together to differentiate BEC into ciliated cells, and TP73 knockout in mice is associated with epithelial cell loss and inflammation of epithelium [90].

Methods

The goal of this study was to identify BEC gene expression patterns and hereditary DNA variants associated with COPD pathogenesis. Toward this goal we conducted a nested case-control study to a) identify genes associated with COPD based on BEC transcript abundance values and b) assess COPD-associated genes for cis-rSNP enrichment, measured as DAE. In parallel, we queried COPD GWAS for significance of putative cis-rSNPs in COPD-associated genes. The study design is presented schematically in Fig. 1.

Study subjects and biospecimens

Homogeneous BEC biospecimens were obtained by bronchoscopic brush biopsy of normal appearing airway (main bronchus) epithelium from 30 COPD and 30 non-COPD control subjects who were enrolled in the Lung Cancer Risk Test (LCRT) study (NCT 01130285 at Clinicaltrials.​gov) [91]. The purpose of the LCRT study is to assess clinical validity of the previously reported LCRT to predict risk for lung cancer [25]. LCRT enrollment criteria included high demographic risk for lung cancer (age 50 or more and 20 pack-years smoking or more) and absence of lung cancer at time of enrollment based on chest CT. Summary statistics for the demographic and clinical characteristics of the subjects used in studies presented here are provided in Table 1 and information for each subject are provided in Additional file 1: Table S1 rows 3–12. Additional relevant details of the LCRT study are provided in Additional file 2. In the study presented here, COPD was defined by spirometry as FEV1/FVC < 0.7 and FEV1% expected < 80%. This corresponds to GOLD Stage II-IV COPD [92]. The LCRT enrolled 385 subjects at 11 clinical centers between 2011 and 2013. At each site, BEC were collected into ice-cold normal saline, then pelleted at 300 g and suspended in RNAlater. Biospecimens collected from each LCRT subject were shipped on dry ice overnight to ResearchDx, Irvine, CA, USA) for RNA extraction and storage. All subjects provided written informed consent. Use of tissue samples and corresponding medical/demographic data for this study is approved under UT IRB protocols #108538 and #107844. For this study, COPD and control subjects were selected with a goal to match for age, smoking history, and gender.
Table 1
Clinical characteristics of study population
 
Non-COPD (n = 30)
COPD (n = 30)
p-value
Age, yr
64.3
63.6
0.713
Sex
  
0.009
 Male
11
22
 
 Female
19
8
 
Smoking status
  
1.0
 Current
10
9
 
 Former
20
21
 
 Never
0
0
 
Pack-years
49
60
0.088
FEV1/FVC
0.81
0.53
5.81E-13
Ethnicity
   
 White
28
26
 
 AA
2
4
 
p-values were calculated using Student’s t-test for age and Pack-years, and Fisher exact test for sex and smoking history

BEC and peripheral blood cell (PBC) samples for differential allelic expression (DAE) measurement

Reliable measurement of DAE required a sample size large enough to include a sufficient number of heterozygotes. Thus, in addition to the BEC samples and matched PBC samples from 60 LCRT subjects used in COPD classifier development, we evaluated archival BEC (120) and PBC (117) samples from additional subjects who were not characterized for COPD status for the purpose of DAE analysis. Of the total 180 BEC and 177 PBC samples, matched samples were available from 98 subjects.
Summary statistics for the cohort used in allele-specific expression analysis are provided in Additional file 1: Table S2.

RNA and DNA extraction

For the 60 LCRT subject samples, RNA was extracted from BEC samples at ResearchDx using the RNeasy Kit (Qiagen, Valencia, CA). RNA was treated with DNAse to remove gDNA contamination and assessed for RNA integrity (see below). The RNA was split into two aliquots and frozen at − 80 degrees C. One of the BEC RNA aliquots from each subject was shipped to the University of Toledo where it was first re-tested for gDNA contamination through PCR. Any samples with signal for gDNA were re-treated with DNAse. Samples then were reverse transcribed into cDNA with M-MLV reverse transcriptase (Invitrogen, Carlsbad, CA) using oligo dT primer according to the manufacturer’s protocol. Genomic DNA was extracted from PBC at each clinical site [91], frozen and shipped to the NCI-EDRN bio-specimen bank at NCI-Frederick. One vial of DNA from each subject was provided to this lab for this study. For non-LCRT subject archival samples (120 BEC samples and 117 PBC samples), both BEC RNA and PBC DNA were extracted in this laboratory according to previously described methods [93].

RNA integrity analysis

RNA extracted from each BEC sample used in this study was assessed for RNA integrity and quantity as previously described [91]. In addition, after careful review of available RNA integrity measurement methods, we chose the 5′/3′ ratio mRNA integrity assay to assess NBEC sample RNA integrity, based on a recent comparison with other methods, including quantitative microfluidic electrophoresis [94]. This method was particularly informative because, since RNA samples were reverse transcribed with poly dT, low quality, fragmented, short length RNA would be associated with lower transcript abundance measured in assays located further away from the 3′ end.

Competitive multiplex PCR amplicon library preparation

Targeted competitive multiplex PCR amplicon libraries were prepared to quantify total and allele-specific expression at 68 target assays on 35 genes by next generation sequencing (NGS) (in Additional file 1: Table S1) according to previously described methods [24, 26]. Genes with high prior likelihood for association with COPD-risk were selected for analysis based on careful literature review, as described in the background section. Included among these was a set of genes previously reported to be relatively unaffected by environmental variation based on cigarette smoking history [30, 40]. Primers. We designed a pool of forward and reverse primer sets targeting 68 assays in the 35 genes, using methods described in detail previously [24, 26]. When possible, we measured transcript abundance at multiple sites for each gene because it is not uncommon that probes assessing different alternative transcripts yield different expression patterns due to inter-individual biological variation [28, 95]. For classifier gene DAE analysis, assays were selected from all exonic SNPs with minor allele frequency of > 0.05 identified based on data from the 1000 Genomes Project [96]. Additional primer design methods are provided in Additional file 2.

Internal standards (IS) and internal standard mixture

Methods for preparation of each internal standard and the internal standard mixture were described previously [24, 26]. Details for this study are provided in Additional file 2. Preparation of a PCR amplicon library for each sample involved four sequential PCR reactions, described in Additional file 2. All primer and IS sequences are provided in Additional file 1: Table S1.

Sequencing

PCR library products were analyzed on an Illumina HiSeq 2500 with TruSeq SBS Kit v4 reagent at Macrogen (Macrogen, Inc., Seoul, South Korea). Macrogen then returned raw sequencing data in FASTQ format. Data Processing, calculation of total or allele-specific target transcript abundance, and filtering to avoid stochastic sampling error were described in detail in [24, 26]. Details for this study are provided in Additional file 2.

Interaction network differences between COPD cases and control

Bivariate interactions among genes in control or COPD subjects were assessed by Pearson’s correlation. Gene pair network connections were assessed for difference between COPD and control cohorts by Wilcoxon rank sum test for in- and out-degree, betweenness, and closeness, using igraph analysis as described in Additional file 2.

Test for cis-rSNP enrichment among classifier genes

Inter-individual variation in DAE of a gene is a manifestation of one or more cis-rSNPs [15, 17, 19, 22, 23]. To test the question of whether cis-rSNPs are enriched in genes with COPD-associated gene expression pattern, we assessed significant difference in prevalence of DAE among COPD-associated genes compared with prevalence of DAE among all genes in multiple different tissues in prior studies [15, 17, 97].

Lung health study (LHS) and COPDgene non-Hispanic white (NHW) GWAS analysis

Detailed methods for analysis of the LHS dataset (phs000335) and COPDgene dataset phs000765 (NHW) [98, 99] are provided in the Additional file 2.

Sub-phenotyping of LHS and COPDgene NHW subjects

As presented in Additional file 2: Figure S1 for LHS, prior to analysis the LHS and COPDgene NHW subjects were stratified into Chronic Bronchitis (CB) or Emphysema (EM) sub-phenotypes based on clinical annotation parameters pertaining to chronic productive cough. For LHS, following quality-control analysis a total of 3230 subjects were included. Of these, 527 were chronic bronchitis, 1198 were emphysema, and 1505 were controls. For COPDgene NHW there were a total of 5269 subjects, of which 556 were chronic bronchitis, 2223 were emphysema, and 2490 were controls.

Statistical analysis

All statistical analyses were performed using R (v 3.2.5) (http://​www.​R-project.​org).

Shrinkage linear discriminant analysis (SLDA) to develop COPD classifier

BEC transcript abundance values for each assay in each subject were used in development of the COPD classifier model. After filtering, 32 target assays in 23 genes yielded BEC transcript abundance data for reliable total transcript abundance quantification in at least 70% of subjects and were included in statistical analyses. Typically, the cause of low subject representation for an assay was insufficient target molecules loaded into library preparation due to low BEC expression. Missing values were imputed using the corresponding mean value for each assay. Shrinkage Linear Discriminant Analysis (SLDA) was performed to select assays. The overall ranking of each assay was determined by correlation-adjusted t-score (CAT scores) [100]. The 10-fold cross-validation (CV) receiver operating characteristic (ROC) area under the curve (AUC) was applied to identify the best sets of assays, and the pooled 10-fold cross-validated ROC AUC was reported for the selected classifier. For each patient, the classifier assigned a probability score for COPD phenotype. An optimal cut-point to predict the class label based on Youden Index (J) was determined by repeated cross-validation step. Each patient was classified according to optimal cut-point. We compared the model predicted class label to the “true” state of COPD or control subject then calculated the diagnostic odds ratio and confidence interval.

Inter-individual variation in allele-specific transcript abundance

For each gene, and at each measured transcribed polymorphic locus, we used the F-test to compare inter-individual variation in allelic imbalance in cDNA samples with that in peripheral blood genomic DNA samples. Specifically, gDNA was used as a control because it is expected that every cell will have two copies of gDNA. Therefore, under ordinary circumstances in non-malignant cells, it is expected that the measured ratio between alleles will be close to one, subject to analytical variation. In contrast, inter-individual variation in cis-regulation due to polymorphisms may cause inter-individual variation in transcription of one allele to the other. Each allele ratio of read counts was log base 2 transformed prior to further analysis. F-test was performed using R (v 3.2.5) (http://​www.​R-project.​org). GraphPad Prism was used to plot figures.

Test for cis-rSNP enrichment in COPD classifier genes

We measured DAE as an indicator that a gene contained one or more cis-rSNPs. To assess for cis-rSNP enrichment among classifier genes, we compared the fraction of genes with DAE among the genes comprised by the classifier to the fraction of genes with DAE in a large prior study [15, 101] using the N-1 Chi-Squared test [102] at MEDCALC (https://​www.​medcalc.​org/​calc/​comparison_​of_​proportions.​php).

Bivariate analysis

We assessed difference in inter-gene correlation of log-transformed transcript abundance by Pearson correlation coefficient (r-value). We used the Fisher r-to-z transformation (Z-score) to assess the significance of difference between two correlation coefficients in two groups.

Covariate analysis

Analysis of covariance (ANCOVA) was used to assess COPD vs control group difference in transcript abundance after controlling for single covariates.

Correction for multiple testing

We used Bonferroni adjustment to correct for multiple testing in GWAS analysis. GTEx data regarding eQTL in lung tissue are reported with the p-values corrected for multiple testing and calculated by GTEx. We report analyses of individual gene-pair correlation changes between control and COPD and analyses of differences in means after correction for covariates (ANCOVA) without correction for multiple testing. This is justified because each of the features tested was selected for analysis based on prior association with COPD, thereby reducing likelihood of false discovery.

Results

RNA and DNA samples

RNA extracted from each BEC sample met previously described thresholds for RNA integrity and quantity as described in methods [91].

Targeted RNAseq expression data

Total transcript abundance values meeting the QC threshold were obtained in BEC samples from at least 70% of subjects for 32 target assays in 23 genes and this set was used in univariate and multivariate analyses.

Interaction network differences between COPD cases and control

On a network basis, TP53 and CEBP transcription factor pathway gene pair network connections were different between COPD and control cohorts as measured by Wilcoxon rank sum test for in- and out-degree (p-value = 7.0E-05), betweenness (p-value = 0.00437), and closeness (p-value = 5.0E-11) (Fig. 2). Consistent with this, the total number of inter-gene correlation connections (lines) among the tested assays was higher among COPD individuals. Inter-gene correlation data in relationship to transcription factors are presented in Additional file 1: Table S3. Notably, ERCC5 was more highly correlated with both CEBPD and TP73 in COPD compared with controls (Fig. 3, Additional file 1: Table S3). In contrast, CEPBD correlation with TP53 was decreased in COPD.

Analysis of covariance (ANCOVA)

After adjustment for covariate effects, there was a difference in mean expression between COPD and controls for several classifier genes. Specifically CEBPG, GPX1, and TP73 were expressed at a higher level in COPD compared to controls while KEAP1 was expressed at a lower level (Additional file 1: Table S4).

Identification of genes with COPD-associated expression pattern

SLDA was used to rank each feature for classification ability according to correlation-adjusted t (CAT)-score (See Additional file 1: Table S1). After 10-fold cross-validation, the classifier with best ROC AUC comprised nine features, including the three demographic variables sex, age, and smoking history in pack-years, and six genes: CAT, CEBPG, GPX1, KEAP1, TP73, and XPA (Fig. 4, Table 2, and Additional file 1: Table S1 rows 14–19). The 10-fold cross-validation AUC for the model was 75.4% (95% CI: 66.3%–89.3%). As reported above, ERCC5 has altered correlation with transcription factor TP73 comprised by the classifier (Fig. 3) and other genes as measured by iGraph (Fig. 2).
Table 2
COPD classifier gene features selected by SLDA1 and 10-fold cross-validation
Feature
Gene Function
CAT2 score
Ranking
Missing value (%)
KEAP1
AO3
3.35
1
8%
GPX1
AO
3.32
2
5%
CEBPG
TF4
2.98
3
18%
XPA
DNAR5
2.64
4
28%
CAT-2
AO
2.64
5
22%
TP73–2
CCC6/DNAR
2.30
6
28%
1SLDA shrinkage linear discriminant analysis, 2CAT score correlation-adjusted t-scores, 3AO antioxidant, 4TF transcription factor, 5DNAR DNA repair, 6CCC cell cycle control

Query of LHS GWAS for targeted genes

Details of LHS GWAS analysis are provided in Additional file 2: Figure S1). Briefly, the LHS COPD GWAS subjects were stratified into chronic bronchitis (CB) or emphysema (EM) sub-phenotypes. All LHS SNPs that passed quality control were restricted to putative cis-regulatory regions. Then, SNPs in this restricted set with p < 0.05 for association with LHS CB or LHS EM subjects were identified. The resulting SNP sets were designated {LHS CB Restricted} and {LHS EM Restricted}.

Integration of restricted LHS SNP sets with genes targeted in this study

The intersection of the 35 genes targeted in this study with the {LHS CB Restricted} SNP set comprised six linked SNPs in ERCC5, each with P < 0.05 (Additional file 1: Table S5). In contrast, no SNPs were identified in the intersection of the 35 targeted genes and the {LHS EM Restricted} SNP set.

Validation of ERCC5 SNP association with chronic bronchitis in the COPDgene cohort

The independent COPDgene NHW phs000765 cohort [3, 98] was stratified according to CB or EM sub-phenotype using the same criteria used to sub-phenotype LHS. COPDgene NHW CB and COPDgene NHW EM were each queried for rs4150275, which was chosen to represent the 6 linked SNPs (Additional file 2: Figure S2). In this validation test, because one SNP was queried in two sub-phenotypes, the Bonferonni adjusted threshold for significance was a = 0.05/2 = 0.025. As with LHS analysis, rs4150275 was significant in COPDgene NHW CB (p = 0.0046) but not COPDgene NHW EM. Importantly, the haplotype represented by rs4150275 allele A was associated with CB in both LHS and COPDgene NHW.

Assessment of genes with COPD-associated RNAseq patterns for DAE in BEC

ERCC5 displays inter-individual variation in DAE in BEC, measured at multiple SNPs, including rs17655, which is linked (D’) to rs4150275) [93]. Further, rs873601 which is linked to rs17655 is predicted to alter miRNA binding sites and likely plays a key functional cis-regulatory role [103].
We identified at least one expressed SNP with MAF > 0.05 in four of the six COPD classifier genes selected by SLDA; CAT: rs1049982, CEBPG: rs3745968, KEAP1:rs1048287, and TP73:rs1801174. For each of these SNPs, the number of heterozygotes among the cDNA samples was close to Hardy-Weinberg Equilibrium expectations (Table 3) and comparable to that observed among gDNA samples. Inter-individual variation in allelic-imbalance in cDNA was significantly higher (p < 0.05) than that in gDNA at each of these four sites after Bonferroni adjustment for multiple testing (Table 3, Fig. 5). The rate of DAE among ERCC5 and the four measurable classifier genes was 100% (4/4) which was significantly higher (p = 0.0023) than the 30% of genes that demonstrated DAE in lung tissue (5884/19,725) [17] or lymphoblastoid cell lines (2935/9751) [101].
Table 3
SLDA COPD classifier gene differential allelic expression (DAE) in bronchial epithelial cell (BEC) or in GTEx lung tissue database
 
DAE in BEC
GTEx Lung Tissue (n = 278)
1SNP
MAF
Subjects Assessed (n)
2Heterozygote Subjects with DAE data (n)
3p-value
eQTL
4p-value
KEAP1-rs1048287
0.1
159
30
9.02E-10
KEAP1
5N.R.
CEBPG-rs3745968
0.11
128
17
6.35E-04
CEBPG
N.R.
CAT-rs1049982
0.34
156
52
1.51E-24
CAT
1.95E-08
TP73-rs1801174
0.09
158
27
1.34E-10
TP73
N.R.
Significant p-values indicated in italicized font
1SNP that served as marker for DAE. SNPs with highest minor allele frequency chosen
2n = number of subjects for whom each SNP allele was measurable in BEC after filtering to prevent stochastic sampling error. The fraction of gDNA samples with heterozygotes was comparable to that for cDNA samples and both approximated Hardy Weinberg Equilibrium expectations
3p-value for F-test comparing inter-individual variation in cDNA to inter-individual variation in gDNA samples
4p-value reported in GTEx database
5N.R not reported

Assessment of BEC putative cis-rSNPs for lung tissue quantitative trait loci (eQTL) status in GTEx database

We queried the Genotype-Tissue Expression (GTEx) database [17, 104] for lung tissue eQTL at SNPs in that were associated with DAE in BEC, including classifier genes CAT, CEBPG, GPX1, and TP73 (Table 3), and ERCC5. GTEx measured eQTL as dose-effect of genotype on total transcript abundance. SNPs associated with lung tissue eQTL include rs17655 (p = 3.2E-7), located in the 3′ untranslated region of ERCC5, and rs1049982 (p = 1.95E-08), located in the promoter of CAT. Notably, for each of these SNPs, the same allele associated with higher expression in GTEx lung tissue was significantly more likely to be expressed at higher level than the opposite allele in BEC. SNPs associated with BEC DAE in the other genes were not associated with significant eQTL in lung tissue in GTEx (Table 3).

Discussion

Data reported here support the hypothesis that low-effect COPD risk/pathogenesis SNPs may be discovered through enrichment as cis-regulatory SNPs in genes that display COPD-associated BEC expression patterns. Moreover, by integrating the BEC RNAseq data with COPD GWAS [98, 99], ENCODE [105], and GTEx [17, 104] databases, we identified ERCC5 SNP rs873601 as a plausible functional connection between ERCC5 DAE (measured at rs17655), and association of rs4150275 with chronic bronchitis in COPD GWAS (Additional file 1: Table S6). [103, 106]. Thus, these data support the role of rs873601 in ERCC5 cis-regulation associated with COPD pathogenesis and risk. That said, other SNPs linked to rs4150275 also are predicted to affect binding of transacting proteins and also could play a role (Additional file 1: Table S5).

Putative cis-regulatory SNPs in SLDA classifier genes

CAT SNP rs1049982 is predicted to have cis-regulatory function because it is in the 5′ untranslated region near the promoter and affects binding of POLR2A. In addition, this SNP was identified as a lung tissue eQTL in lung tissue [17, 104]. Based on these characteristics, this SNP would be a suitable target for experimental confirmation of function in BEC [107]. With respect to CEBPG, KEAP1, and TP73 it is likely that SNPs other than those used to measure DAE in this study are responsible for cis-regulation of these genes in BEC and additional experimental studies will be necessary to answer this question.
COPD-associated SNPs have been reported for several of the classifier genes (see background section) but, to our knowledge, not validated in individual GWAS. It will be worthwhile to assess recently completed COPD meta-analysis GWAS for SNPs in these genes using reduced stringency for false reporting.

Effect of study design characteristics selected to optimize power

Several methodological approaches were implemented in this study to maximize the power to identify BEC transcription patterns associated with COPD. For example, genes with high prior likelihood for hereditary COPD-risk association were targeted. In addition, a transcript abundance measurement platform with excellent analytical performance characteristics was used. Specifically, there was no measurable signal-to-analyte compression with the method used in this study [25, 28] and the targeted PCR method used resulted in abundant signal for each analyte. This resulted in robust collection of data from each specimen, comparing favorably with other methods, such as microarrays or whole exome sequencing analysis [8, 17, 41]. Moreover, analysis of cell populations homogeneous for a particular cell type, such as the homogeneous BEC population samples in this study, increases the power to identify disease-associated transcript abundance patterns and eQTL [95, 108, 109]. According to ASE measurement by targeted RNAseq, DAE was clearly detected for marker SNPs in classifier genes CEBPG, KEAP1, and TP73 in BEC (Table 3). However, lung tissue eQTL was not reported for any SNPs in these genes measured by whole transcriptome RNAseq in GTEx study [17]. This observation is likely due to difference in specimen type (i.e. homogeneous BEC vs heterogeneous lung tissue), and deeper coverage obtained by targeted RNAseq for ASE in this study.

Limits of study and opportunities

It is evident that COPD risk SNPs discovered in the future will have low effect because they are very rare and/or they are common but their individual contribution to risk is low. Results presented here exemplify both the challenge to identify low-effect complex disease risk variants and the opportunity of the approach used. The low effect of rs4150275 is likely due to multiple factors. For example, the rs4150275 A allele prevalence in European populations is 5%. Moreover, rs873601, the putative functional SNP linked to DAE SNP rs17655 and COPD risk SNP rs4150275, may be one of multiple SNPs that contribute to ERCC5 transcription regulation [93]. As such, there is a need to directly measure function of putative cis-regulatory SNPs through recently developed high throughput NGS methods [110112]. Another factor contributing to low effect of rs4150275 is that, based on data presented here, ERCC5 is likely one of many genes that contribute to COPD risk when sub-optimally regulated. In this study we increased the power to identify COPD risk SNPs with cis-regulatory function in BEC by studying homogeneous BEC biospecimens. It is likely that hereditary variants affecting gene expression in lung fibroblasts and immune cells also contribute to COPD predisposition. Thus, homogeneous populations of these cell types should be included in future studies.

Conclusion

We report that low-effect COPD risk SNPs may be identified through enrichment as cis-regulatory SNPs in genes that display COPD-associated BEC expression patterns. These findings support broader application of the approach presented here, including further targeted RNAseq analysis of BEC and homogeneous populations of other lung cell types to identify COPD associated expression patterns and cis-rSNPs in genes comprised by the expression patterns, followed by test of associated cis-rSNPs in large GWAS meta-analyses. This approach promises to facilitate progress toward the important goal of identifying a set of COPD risk variants with sufficient effect on COPD pathogenesis and variation in hereditary risk to have clinical utility. This knowledge is expected to lead to better COPD prevention and treatment strategies.

Acknowledgements

Inchul Yang (Korean Research Institute for Standards Science: KRISS) for many insightful comments; Fluidigm Corporation (South San Francisco, CA) for generous loan and support of Access Array™ instrument.

Funding

NIH grants CA148572 and HL108016; The George Isaac Cancer Research Fund; Fluidigm Corporation (South San Francisco, CA), loan and support of Access Array™ instrument.

Availability of data and materials

The majority of the data generated for this study are available in the additional files. The complete datasets analyzed during this study are available from the corresponding author upon reasonable request.
Collection and use of samples and corresponding medical/demographic data was approved under University of Toledo IRB protocols #108538 and #107844. Each subject included in this study provided written informed consent.
Not applicable.

Competing interests

JCW is a consultant for and has equity interest in Accugenomics, Inc. which has a financial interest in the data presented here. JCW, TB and ELC have issued and pending patents for the technology and biomarkers presented here. Other authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat Rennard SI. COPD: overview of definitions, epidemiology, and factors influencing its development. Chest. 1998;113(4 Suppl):235S–41S.PubMedCrossRef Rennard SI. COPD: overview of definitions, epidemiology, and factors influencing its development. Chest. 1998;113(4 Suppl):235S–41S.PubMedCrossRef
2.
Zurück zum Zitat Zhou JJ, Cho MH, Castaldi PJ, Hersh CP, Silverman EK, Laird NM. Heritability of chronic obstructive pulmonary disease and related phenotypes in smokers. Am J Respir Crit Care Med. 2013;188(8):941–7.PubMedPubMedCentralCrossRef Zhou JJ, Cho MH, Castaldi PJ, Hersh CP, Silverman EK, Laird NM. Heritability of chronic obstructive pulmonary disease and related phenotypes in smokers. Am J Respir Crit Care Med. 2013;188(8):941–7.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Cho MH, McDonald ML, Zhou X, Mattheisen M, Castaldi PJ, Hersh CP, Demeo DL, Sylvia JS, Ziniti J, Laird NM, et al. Risk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysis. Lancet Respir Med. 2014;2(3):214–25.PubMedPubMedCentralCrossRef Cho MH, McDonald ML, Zhou X, Mattheisen M, Castaldi PJ, Hersh CP, Demeo DL, Sylvia JS, Ziniti J, Laird NM, et al. Risk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysis. Lancet Respir Med. 2014;2(3):214–25.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Todd JL, Goldstein DB, Ge D, Christie J, Palmer SM. The state of genome-wide association studies in pulmonary disease: a new perspective. Am J Respir Crit Care Med. 2011;184(8):873–80.PubMedPubMedCentralCrossRef Todd JL, Goldstein DB, Ge D, Christie J, Palmer SM. The state of genome-wide association studies in pulmonary disease: a new perspective. Am J Respir Crit Care Med. 2011;184(8):873–80.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Qiao D, Lange C, Beaty TH, Crapo JD, Barnes KC, Bamshad M, Hersh CP, Morrow J, Pinto-Plata VM, Marchetti N, et al. Exome sequencing analysis in severe, early-onset chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2016;193(12):1353–63.PubMedPubMedCentralCrossRef Qiao D, Lange C, Beaty TH, Crapo JD, Barnes KC, Bamshad M, Hersh CP, Morrow J, Pinto-Plata VM, Marchetti N, et al. Exome sequencing analysis in severe, early-onset chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2016;193(12):1353–63.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Busch R, Hobbs BD, Zhou J, Castaldi PJ, McGeachie MJ, Hardin ME, Hawrylkiewicz I, Sliwinski P, Yim JJ, Kim WJ, et al. Genetic association and risk scores in a COPD meta-analysis of 16,707 subjects. Am J Respir Cell Mol Biol. 2017;57(1):35–45.PubMedCrossRef Busch R, Hobbs BD, Zhou J, Castaldi PJ, McGeachie MJ, Hardin ME, Hawrylkiewicz I, Sliwinski P, Yim JJ, Kim WJ, et al. Genetic association and risk scores in a COPD meta-analysis of 16,707 subjects. Am J Respir Cell Mol Biol. 2017;57(1):35–45.PubMedCrossRef
7.
Zurück zum Zitat Wain LV, Shrine N, Artigas MS, Erzurumluoglu AM, Noyvert B, Bossini-Castillo L, Me O, Henry AP, Portelli MA, Hall RJ, et al. Genome-wide association analyses for lung function and chronic obstructive pulmonary disease identify new loci and potential druggable targets. Nat Genet. 2017;49(3):416–25.PubMedPubMedCentralCrossRef Wain LV, Shrine N, Artigas MS, Erzurumluoglu AM, Noyvert B, Bossini-Castillo L, Me O, Henry AP, Portelli MA, Hall RJ, et al. Genome-wide association analyses for lung function and chronic obstructive pulmonary disease identify new loci and potential druggable targets. Nat Genet. 2017;49(3):416–25.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Steiling K, van den Berge M, Hijazi K, Florido R, Campbell J, Liu G, Xiao J, Zhang XH, Duclos G, Drizik E, et al. A dynamic bronchial airway gene expression signature of chronic obstructive pulmonary disease and lung function impairment. Am J Respir Crit Care Med. 2013;187(9):933–42.PubMedPubMedCentralCrossRef Steiling K, van den Berge M, Hijazi K, Florido R, Campbell J, Liu G, Xiao J, Zhang XH, Duclos G, Drizik E, et al. A dynamic bronchial airway gene expression signature of chronic obstructive pulmonary disease and lung function impairment. Am J Respir Crit Care Med. 2013;187(9):933–42.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Shaykhiev R, Crystal RG. Early events in the pathogenesis of chronic obstructive pulmonary disease. Smoking-induced reprogramming of airway epithelial basal progenitor cells. Ann Am Thorac Soc. 2014;11(Suppl 5):S252–8.PubMedPubMedCentralCrossRef Shaykhiev R, Crystal RG. Early events in the pathogenesis of chronic obstructive pulmonary disease. Smoking-induced reprogramming of airway epithelial basal progenitor cells. Ann Am Thorac Soc. 2014;11(Suppl 5):S252–8.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Pierrou S, Broberg P, O'Donnell RA, Pawlowski K, Virtala R, Lindqvist E, Richter A, Wilson SJ, Angco G, Moller S, et al. Expression of genes involved in oxidative stress responses in airway epithelial cells of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;175(6):577–86.PubMedCrossRef Pierrou S, Broberg P, O'Donnell RA, Pawlowski K, Virtala R, Lindqvist E, Richter A, Wilson SJ, Angco G, Moller S, et al. Expression of genes involved in oxidative stress responses in airway epithelial cells of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;175(6):577–86.PubMedCrossRef
11.
Zurück zum Zitat 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):423–8.PubMedCrossRef 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):423–8.PubMedCrossRef
12.
Zurück zum Zitat Albert FW, Kruglyak L. The role of regulatory variation in complex traits and disease. Nat Rev Genet. 2015;16(4):197–212.PubMedCrossRef Albert FW, Kruglyak L. The role of regulatory variation in complex traits and disease. Nat Rev Genet. 2015;16(4):197–212.PubMedCrossRef
13.
Zurück zum Zitat Fehrmann RS, Jansen RC, Veldink JH, Westra HJ, Arends D, Bonder MJ, Fu J, Deelen P, Groen HJ, Smolonska A, et al. Trans-eQTLs reveal that independent genetic variants associated with a complex phenotype converge on intermediate genes, with a major role for the HLA. PLoS Genet. 2011;7(8):e1002197.PubMedPubMedCentralCrossRef Fehrmann RS, Jansen RC, Veldink JH, Westra HJ, Arends D, Bonder MJ, Fu J, Deelen P, Groen HJ, Smolonska A, et al. Trans-eQTLs reveal that independent genetic variants associated with a complex phenotype converge on intermediate genes, with a major role for the HLA. PLoS Genet. 2011;7(8):e1002197.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Grundberg E, Small KS, Hedman AK, Nica AC, Buil A, Keildson S, Bell JT, Yang TP, Meduri E, Barrett A, et al. Mapping cis- and trans-regulatory effects across multiple tissues in twins. Nat Genet. 2012;44(10):1084–9.PubMedPubMedCentralCrossRef Grundberg E, Small KS, Hedman AK, Nica AC, Buil A, Keildson S, Bell JT, Yang TP, Meduri E, Barrett A, et al. Mapping cis- and trans-regulatory effects across multiple tissues in twins. Nat Genet. 2012;44(10):1084–9.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Hamdi Y, Soucy P, Adoue V, Michailidou K, Canisius S, Lemacon A, Droit A, Andrulis IL, Anton-Culver H, Arndt V, et al. Association of breast cancer risk with genetic variants showing differential allelic expression: identification of a novel breast cancer susceptibility locus at 4q21. Oncotarget. 2016;7(49):80140–63.PubMedPubMedCentralCrossRef Hamdi Y, Soucy P, Adoue V, Michailidou K, Canisius S, Lemacon A, Droit A, Andrulis IL, Anton-Culver H, Arndt V, et al. Association of breast cancer risk with genetic variants showing differential allelic expression: identification of a novel breast cancer susceptibility locus at 4q21. Oncotarget. 2016;7(49):80140–63.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E, Wang H, Reynolds AP, Sandstrom R, Qu H, Brody J, et al. Systematic localization of common disease-associated variation in regulatory DNA. Science. 2012;337(6099):1190–5.PubMedPubMedCentralCrossRef Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E, Wang H, Reynolds AP, Sandstrom R, Qu H, Brody J, et al. Systematic localization of common disease-associated variation in regulatory DNA. Science. 2012;337(6099):1190–5.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Battle A, Brown CD, Engelhardt BE, Montgomery SB. Genetic effects on gene expression across human tissues. Nature. 2017;550(7675):204–13.PubMedCrossRef Battle A, Brown CD, Engelhardt BE, Montgomery SB. Genetic effects on gene expression across human tissues. Nature. 2017;550(7675):204–13.PubMedCrossRef
18.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Castaldi PJ, Cho MH, Zhou X, Qiu W, McGeachie M, Celli B, Bakke P, Gulsvik A, Lomas DA, Crapo JD, et al. Genetic control of gene expression at novel and established chronic obstructive pulmonary disease loci. Hum Mol Genet. 2015;24(4):1200–10.PubMedCrossRef Castaldi PJ, Cho MH, Zhou X, Qiu W, McGeachie M, Celli B, Bakke P, Gulsvik A, Lomas DA, Crapo JD, et al. Genetic control of gene expression at novel and established chronic obstructive pulmonary disease loci. Hum Mol Genet. 2015;24(4):1200–10.PubMedCrossRef
20.
Zurück zum Zitat Yan H, Yuan W, Velculescu VE, Vogelstein B, Kinzler KW. Allelic variation in human gene expression. Science. 2002;297(5584):1143.PubMedCrossRef Yan H, Yuan W, Velculescu VE, Vogelstein B, Kinzler KW. Allelic variation in human gene expression. Science. 2002;297(5584):1143.PubMedCrossRef
21.
Zurück zum Zitat Kang EY, Martin LJ, Mangul S, Isvilanonda W, Zou J, Ben-David E, Han B, Lusis AJ, Shifman S, Eskin E. Discovering single nucleotide polymorphisms regulating human gene expression using allele specific expression from RNA-seq data. Genetics. 2016;204(3):1057–64.PubMedPubMedCentralCrossRef Kang EY, Martin LJ, Mangul S, Isvilanonda W, Zou J, Ben-David E, Han B, Lusis AJ, Shifman S, Eskin E. Discovering single nucleotide polymorphisms regulating human gene expression using allele specific expression from RNA-seq data. Genetics. 2016;204(3):1057–64.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Almlof JC, Lundmark P, Lundmark A, Ge B, Maouche S, Goring HH, Liljedahl U, Enstrom C, Brocheton J, Proust C, et al. Powerful identification of cis-regulatory SNPs in human primary monocytes using allele-specific gene expression. PLoS One. 2012;7(12):e52260.PubMedPubMedCentralCrossRef Almlof JC, Lundmark P, Lundmark A, Ge B, Maouche S, Goring HH, Liljedahl U, Enstrom C, Brocheton J, Proust C, et al. Powerful identification of cis-regulatory SNPs in human primary monocytes using allele-specific gene expression. PLoS One. 2012;7(12):e52260.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Castel SE, Levy-Moonshine A, Mohammadi P, Banks E, Lappalainen T. Tools and best practices for data processing in allelic expression analysis. Genome Biol. 2015;16:195.PubMedPubMedCentralCrossRef Castel SE, Levy-Moonshine A, Mohammadi P, Banks E, Lappalainen T. Tools and best practices for data processing in allelic expression analysis. Genome Biol. 2015;16:195.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Blomquist TM, Crawford EL, Lovett JL, Yeo J, Stanoszek LM, Levin A, Li J, Lu M, Shi L, Muldrew K, et al. Targeted RNA-sequencing with competitive multiplex-PCR amplicon libraries. PLoS One. 2013;8(11):e79120.PubMedPubMedCentralCrossRef Blomquist TM, Crawford EL, Lovett JL, Yeo J, Stanoszek LM, Levin A, Li J, Lu M, Shi L, Muldrew K, et al. Targeted RNA-sequencing with competitive multiplex-PCR amplicon libraries. PLoS One. 2013;8(11):e79120.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Blomquist T, Crawford EL, Mullins D, Yoon Y, Hernandez DA, Khuder S, Ruppel PL, Peters E, Oldfield DJ, Austermiller B, et al. Pattern of antioxidant and DNA repair gene expression in normal airway epithelium associated with lung cancer diagnosis. Cancer Res. 2009;69(22):8629–35.PubMedPubMedCentralCrossRef Blomquist T, Crawford EL, Mullins D, Yoon Y, Hernandez DA, Khuder S, Ruppel PL, Peters E, Oldfield DJ, Austermiller B, et al. Pattern of antioxidant and DNA repair gene expression in normal airway epithelium associated with lung cancer diagnosis. Cancer Res. 2009;69(22):8629–35.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Blomquist T, Crawford EL, Yeo J, Zhang X, Willey JC. Control for stochastic sampling variation and qualitative sequencing error in next generation sequencing. Biomol Detect Quantif. 2015;5:30–7.PubMedPubMedCentralCrossRef Blomquist T, Crawford EL, Yeo J, Zhang X, Willey JC. Control for stochastic sampling variation and qualitative sequencing error in next generation sequencing. Biomol Detect Quantif. 2015;5:30–7.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Blomquist TM, Crawford EL, Willey JC. Cis-acting genetic variation at an E2F1/YY1 response site and putative p53 site is associated with altered allele-specific expression of ERCC5 (XPG) transcript in normal human bronchial epithelium. Carcinogenesis. 2010;31(7):1242–50.PubMedPubMedCentralCrossRef Blomquist TM, Crawford EL, Willey JC. Cis-acting genetic variation at an E2F1/YY1 response site and putative p53 site is associated with altered allele-specific expression of ERCC5 (XPG) transcript in normal human bronchial epithelium. Carcinogenesis. 2010;31(7):1242–50.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Canales RD, Luo Y, Willey JC, Austermiller B, Barbacioru CC, Boysen C, Hunkapiller K, Jensen RV, Knight CR, Lee KY, et al. Evaluation of DNA microarray results with quantitative gene expression platforms. Nat Biotechnol. 2006;24(9):1115–22.PubMedCrossRef Canales RD, Luo Y, Willey JC, Austermiller B, Barbacioru CC, Boysen C, Hunkapiller K, Jensen RV, Knight CR, Lee KY, et al. Evaluation of DNA microarray results with quantitative gene expression platforms. Nat Biotechnol. 2006;24(9):1115–22.PubMedCrossRef
29.
Zurück zum Zitat Crawford EL, Blomquist T, Mullins DN, Yoon Y, Hernandez DR, Al-Bagdhadi M, Ruiz J, Hammersley J, Willey JC. CEBPG regulates ERCC5/XPG expression in human bronchial epithelial cells and this regulation is modified by E2F1/YY1 interactions. Carcinogenesis. 2007;28(12):2552–9.PubMedCrossRef Crawford EL, Blomquist T, Mullins DN, Yoon Y, Hernandez DR, Al-Bagdhadi M, Ruiz J, Hammersley J, Willey JC. CEBPG regulates ERCC5/XPG expression in human bronchial epithelial cells and this regulation is modified by E2F1/YY1 interactions. Carcinogenesis. 2007;28(12):2552–9.PubMedCrossRef
30.
Zurück zum Zitat Mullins DN, Crawford EL, Khuder SA, Hernandez DA, Yoon Y, Willey JC. CEBPG transcription factor correlates with antioxidant and DNA repair genes in normal bronchial epithelial cells but not in individuals with bronchogenic carcinoma. BMC Cancer. 2005;5:141.PubMedPubMedCentralCrossRef Mullins DN, Crawford EL, Khuder SA, Hernandez DA, Yoon Y, Willey JC. CEBPG transcription factor correlates with antioxidant and DNA repair genes in normal bronchial epithelial cells but not in individuals with bronchogenic carcinoma. BMC Cancer. 2005;5:141.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Yeo J, Crawford EL, Blomquist TM, Stanoszek LM, Dannemiller RE, Zyrek J, De Las Casas LE, Khuder SA, Willey JC. A multiplex two-color real-time PCR method for quality-controlled molecular diagnostic testing of FFPE samples. PLoS One. 2014;9(2):e89395.PubMedPubMedCentralCrossRef Yeo J, Crawford EL, Blomquist TM, Stanoszek LM, Dannemiller RE, Zyrek J, De Las Casas LE, Khuder SA, Willey JC. A multiplex two-color real-time PCR method for quality-controlled molecular diagnostic testing of FFPE samples. PLoS One. 2014;9(2):e89395.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Crawford EL, Warner KA, Khuder SA, Zahorchak RJ, Willey JC. Multiplex standardized RT-PCR for expression analysis of many genes in small samples. Biochem Biophys Res Commun. 2002;293(1):509–16.PubMedCrossRef Crawford EL, Warner KA, Khuder SA, Zahorchak RJ, Willey JC. Multiplex standardized RT-PCR for expression analysis of many genes in small samples. Biochem Biophys Res Commun. 2002;293(1):509–16.PubMedCrossRef
33.
Zurück zum Zitat Crawford EL, Peters GJ, Noordhuis P, Rots MG, Vondracek M, Grafstrom RC, Lieuallen K, Lennon G, Zahorchak RJ, Georgeson MJ, et al. Reproducible gene expression measurement among multiple laboratories obtained in a blinded study using standardized RT (StaRT)-PCR. Mol Diagn. 2001;6(4):217–25.PubMedCrossRef Crawford EL, Peters GJ, Noordhuis P, Rots MG, Vondracek M, Grafstrom RC, Lieuallen K, Lennon G, Zahorchak RJ, Georgeson MJ, et al. Reproducible gene expression measurement among multiple laboratories obtained in a blinded study using standardized RT (StaRT)-PCR. Mol Diagn. 2001;6(4):217–25.PubMedCrossRef
34.
Zurück zum Zitat Cheung VG, Spielman RS, Ewens KG, Weber TM, Morley M, Burdick JT. Mapping determinants of human gene expression by regional and genome-wide association. Nature. 2005;437(7063):1365–9.PubMedPubMedCentralCrossRef Cheung VG, Spielman RS, Ewens KG, Weber TM, Morley M, Burdick JT. Mapping determinants of human gene expression by regional and genome-wide association. Nature. 2005;437(7063):1365–9.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Schadt EE, Monks SA, Drake TA, Lusis AJ, Che N, Colinayo V, Ruff TG, Milligan SB, Lamb JR, Cavet G, et al. Genetics of gene expression surveyed in maize, mouse and man. Nature. 2003;422(6929):297–302.PubMedCrossRef Schadt EE, Monks SA, Drake TA, Lusis AJ, Che N, Colinayo V, Ruff TG, Milligan SB, Lamb JR, Cavet G, et al. Genetics of gene expression surveyed in maize, mouse and man. Nature. 2003;422(6929):297–302.PubMedCrossRef
36.
Zurück zum Zitat Stranger BE, Forrest MS, Clark AG, Minichiello MJ, Deutsch S, Lyle R, Hunt S, Kahl B, Antonarakis SE, Tavare S, et al. Genome-wide associations of gene expression variation in humans. PLoS Genet. 2005;1(6):e78.PubMedPubMedCentralCrossRef Stranger BE, Forrest MS, Clark AG, Minichiello MJ, Deutsch S, Lyle R, Hunt S, Kahl B, Antonarakis SE, Tavare S, et al. Genome-wide associations of gene expression variation in humans. PLoS Genet. 2005;1(6):e78.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Skelly DA, Johansson M, Madeoy J, Wakefield J, Akey JM. A powerful and flexible statistical framework for testing hypotheses of allele-specific gene expression from RNA-seq data. Genome Res. 2011;21(10):1728–37.PubMedPubMedCentralCrossRef Skelly DA, Johansson M, Madeoy J, Wakefield J, Akey JM. A powerful and flexible statistical framework for testing hypotheses of allele-specific gene expression from RNA-seq data. Genome Res. 2011;21(10):1728–37.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Blomquist TM, Brown RD, Crawford EL, de la Serna I, Williams K, Yoon Y, Hernandez DA, Willey JC. CEBPG exhibits allele-specific expression in human bronchial epithelial cells. Gene regul syst biol. 2013;7:125–38. Blomquist TM, Brown RD, Crawford EL, de la Serna I, Williams K, Yoon Y, Hernandez DA, Willey JC. CEBPG exhibits allele-specific expression in human bronchial epithelial cells. Gene regul syst biol. 2013;7:125–38.
39.
Zurück zum Zitat Brumpton BM, Ferreira MA. Multivariate eQTL mapping uncovers functional variation on the X-chromosome associated with complex disease traits. Hum Genet. 2016;135(7):827–39.PubMedCrossRef Brumpton BM, Ferreira MA. Multivariate eQTL mapping uncovers functional variation on the X-chromosome associated with complex disease traits. Hum Genet. 2016;135(7):827–39.PubMedCrossRef
40.
Zurück zum Zitat Crawford EL, Khuder SA, Durham SJ, Frampton M, Utell M, Thilly WG, Weaver DA, Ferencak WJ, Jennings CA, Hammersley JR, et al. Normal bronchial epithelial cell expression of glutathione transferase P1, glutathione transferase M3, and glutathione peroxidase is low in subjects with bronchogenic carcinoma. Cancer Res. 2000;60(6):1609–18.PubMed Crawford EL, Khuder SA, Durham SJ, Frampton M, Utell M, Thilly WG, Weaver DA, Ferencak WJ, Jennings CA, Hammersley JR, et al. Normal bronchial epithelial cell expression of glutathione transferase P1, glutathione transferase M3, and glutathione peroxidase is low in subjects with bronchogenic carcinoma. Cancer Res. 2000;60(6):1609–18.PubMed
41.
Zurück zum Zitat Tilley AE, O'Connor TP, Hackett NR, Strulovici-Barel Y, Salit J, Amoroso N, Zhou XK, Raman T, Omberg L, Clark A, et al. Biologic phenotyping of the human small airway epithelial response to cigarette smoking. PLoS One. 2011;6(7):e22798.PubMedPubMedCentralCrossRef Tilley AE, O'Connor TP, Hackett NR, Strulovici-Barel Y, Salit J, Amoroso N, Zhou XK, Raman T, Omberg L, Clark A, et al. Biologic phenotyping of the human small airway epithelial response to cigarette smoking. PLoS One. 2011;6(7):e22798.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Barnes PJ. Cellular and molecular mechanisms of chronic obstructive pulmonary disease. Clin Chest Med. 2014;35(1):71–86.PubMedCrossRef Barnes PJ. Cellular and molecular mechanisms of chronic obstructive pulmonary disease. Clin Chest Med. 2014;35(1):71–86.PubMedCrossRef
43.
Zurück zum Zitat Tang W, Bentley AR, Kritchevsky SB, Harris TB, Newman AB, Bauer DC, Meibohm B, Cassano PA, Health ABCs. Genetic variation in antioxidant enzymes, cigarette smoking, and longitudinal change in lung function. Free Radic Biol Med. 2013;63:304–12.PubMedPubMedCentralCrossRef Tang W, Bentley AR, Kritchevsky SB, Harris TB, Newman AB, Bauer DC, Meibohm B, Cassano PA, Health ABCs. Genetic variation in antioxidant enzymes, cigarette smoking, and longitudinal change in lung function. Free Radic Biol Med. 2013;63:304–12.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Willey JC, Frampton MW, Utell MJ, Apostolakos MJ, Coy EL, Olson DE, Hammersley JR, Matteson D, Thilly WG. Patterns of gene expression in human airway epithelial cells. Chest. 1997;111(6 Suppl):83S.PubMedCrossRef Willey JC, Frampton MW, Utell MJ, Apostolakos MJ, Coy EL, Olson DE, Hammersley JR, Matteson D, Thilly WG. Patterns of gene expression in human airway epithelial cells. Chest. 1997;111(6 Suppl):83S.PubMedCrossRef
45.
Zurück zum Zitat DeMuth JP, Jackson CM, Weaver DA, Crawford EL, Durzinsky DS, Durham SJ, Zaher A, Phillips ER, Khuder SA, Willey JC. The gene expression index c-myc x E2F-1/p21 is highly predictive of malignant phenotype in human bronchial epithelial cells. Am J Respir Cell Mol Biol. 1998;19(1):18–24.PubMedCrossRef DeMuth JP, Jackson CM, Weaver DA, Crawford EL, Durzinsky DS, Durham SJ, Zaher A, Phillips ER, Khuder SA, Willey JC. The gene expression index c-myc x E2F-1/p21 is highly predictive of malignant phenotype in human bronchial epithelial cells. Am J Respir Cell Mol Biol. 1998;19(1):18–24.PubMedCrossRef
46.
Zurück zum Zitat Bentley AR, Kritchevsky SB, Harris TB, Newman AB, Bauer DC, Meibohm B, Clark AG, Cassano PA, Health ABCS. Genetic variation in antioxidant enzymes and lung function. Free Radic Biol Med. 2012;52(9):1577–83.PubMedPubMedCentralCrossRef Bentley AR, Kritchevsky SB, Harris TB, Newman AB, Bauer DC, Meibohm B, Clark AG, Cassano PA, Health ABCS. Genetic variation in antioxidant enzymes and lung function. Free Radic Biol Med. 2012;52(9):1577–83.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Tsuji T, Aoshiba K, Nagai A. Alveolar cell senescence in patients with pulmonary emphysema. Am J Respir Crit Care Med. 2006;174(8):886–93.PubMedCrossRef Tsuji T, Aoshiba K, Nagai A. Alveolar cell senescence in patients with pulmonary emphysema. Am J Respir Crit Care Med. 2006;174(8):886–93.PubMedCrossRef
48.
Zurück zum Zitat Shaykhiev R, Sackrowitz R, Fukui T, Zuo WL, Chao IW, Strulovici-Barel Y, Downey RJ, Crystal RG. Smoking-induced CXCL14 expression in the human airway epithelium links chronic obstructive pulmonary disease to lung cancer. Am J Respir Cell Mol Biol. 2013;49(3):418–25.PubMedPubMedCentralCrossRef Shaykhiev R, Sackrowitz R, Fukui T, Zuo WL, Chao IW, Strulovici-Barel Y, Downey RJ, Crystal RG. Smoking-induced CXCL14 expression in the human airway epithelium links chronic obstructive pulmonary disease to lung cancer. Am J Respir Cell Mol Biol. 2013;49(3):418–25.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Heijink IH, Noordhoek JA, Timens W, van Oosterhout AJ, Postma DS. Abnormalities in airway epithelial junction formation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2014;189(11):1439–42.PubMedCrossRef Heijink IH, Noordhoek JA, Timens W, van Oosterhout AJ, Postma DS. Abnormalities in airway epithelial junction formation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2014;189(11):1439–42.PubMedCrossRef
50.
Zurück zum Zitat Hersh CP, Pillai SG, Zhu G, Lomas DA, Bakke P, Gulsvik A, DeMeo DL, Klanderman BJ, Lazarus R, Litonjua AA, et al. Multistudy fine mapping of chromosome 2q identifies XRCC5 as a chronic obstructive pulmonary disease susceptibility gene. Am J Respir Crit Care Med. 2010;182(5):605–13.PubMedPubMedCentralCrossRef Hersh CP, Pillai SG, Zhu G, Lomas DA, Bakke P, Gulsvik A, DeMeo DL, Klanderman BJ, Lazarus R, Litonjua AA, et al. Multistudy fine mapping of chromosome 2q identifies XRCC5 as a chronic obstructive pulmonary disease susceptibility gene. Am J Respir Crit Care Med. 2010;182(5):605–13.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Didon L, Roos AB, Elmberger GP, Gonzalez FJ, Nord M. Lung-specific inactivation of CCAAT/enhancer binding protein alpha causes a pathological pattern characteristic of COPD. Eur Respir J. 2010;35(1):186–97.PubMedCrossRef Didon L, Roos AB, Elmberger GP, Gonzalez FJ, Nord M. Lung-specific inactivation of CCAAT/enhancer binding protein alpha causes a pathological pattern characteristic of COPD. Eur Respir J. 2010;35(1):186–97.PubMedCrossRef
53.
Zurück zum Zitat Didon L, Qvarfordt I, Andersson O, Nord M, Riise GC. Decreased CCAAT/enhancer binding protein transcription factor activity in chronic bronchitis and COPD. Chest. 2005;127(4):1341–6.PubMed Didon L, Qvarfordt I, Andersson O, Nord M, Riise GC. Decreased CCAAT/enhancer binding protein transcription factor activity in chronic bronchitis and COPD. Chest. 2005;127(4):1341–6.PubMed
54.
Zurück zum Zitat da Silva AL, da Rosa HT, Karnopp TE, Charlier CF, Ellwanger JH, Moura DJ, Possuelo LG, Valim AR, Guecheva TN, Henriques JA. Evaluation of DNA damage in COPD patients and its correlation with polymorphisms in repair genes. BMC Med Genet. 2013;14:93.PubMedPubMedCentralCrossRef da Silva AL, da Rosa HT, Karnopp TE, Charlier CF, Ellwanger JH, Moura DJ, Possuelo LG, Valim AR, Guecheva TN, Henriques JA. Evaluation of DNA damage in COPD patients and its correlation with polymorphisms in repair genes. BMC Med Genet. 2013;14:93.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Lakhdar R, Denden S, Knani J, Leban N, Daimi H, Hassine M, Lefranc G, Chibani JB, Khelil AH. Combined analysis of EPHX1, GSTP1, GSTM1 and GSTT1 gene polymorphisms in relation to chronic obstructive pulmonary disease risk and lung function impairment. Dis Markers. 2011;30(5):253–63.PubMedPubMedCentralCrossRef Lakhdar R, Denden S, Knani J, Leban N, Daimi H, Hassine M, Lefranc G, Chibani JB, Khelil AH. Combined analysis of EPHX1, GSTP1, GSTM1 and GSTT1 gene polymorphisms in relation to chronic obstructive pulmonary disease risk and lung function impairment. Dis Markers. 2011;30(5):253–63.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat 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):323–8.PubMedCrossRef 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):323–8.PubMedCrossRef
57.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Yang SF, Xu YJ, Xie JG, Zhang ZX. hOGG1 Ser326Cys and XRCC1 Arg399Gln polymorphisms associated with chronic obstructive pulmonary disease. Chin Med J. 2009;122(8):960–6.PubMed Yang SF, Xu YJ, Xie JG, Zhang ZX. hOGG1 Ser326Cys and XRCC1 Arg399Gln polymorphisms associated with chronic obstructive pulmonary disease. Chin Med J. 2009;122(8):960–6.PubMed
59.
Zurück zum Zitat Young RP, Hopkins R, Black PN, Eddy C, Wu L, Gamble GD, Mills GD, Garrett JE, Eaton TE, Rees MI. Functional variants of antioxidant genes in smokers with COPD and in those with normal lung function. Thorax. 2006;61(5):394–9.PubMedPubMedCentralCrossRef Young RP, Hopkins R, Black PN, Eddy C, Wu L, Gamble GD, Mills GD, Garrett JE, Eaton TE, Rees MI. Functional variants of antioxidant genes in smokers with COPD and in those with normal lung function. Thorax. 2006;61(5):394–9.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Sorheim IC, DeMeo DL, Washko G, Litonjua A, Sparrow D, Bowler R, Bakke P, Pillai SG, Coxson HO, Lomas DA, et al. Polymorphisms in the superoxide dismutase-3 gene are associated with emphysema in COPD. Copd. 2010;7(4):262–8.PubMedPubMedCentralCrossRef Sorheim IC, DeMeo DL, Washko G, Litonjua A, Sparrow D, Bowler R, Bakke P, Pillai SG, Coxson HO, Lomas DA, et al. Polymorphisms in the superoxide dismutase-3 gene are associated with emphysema in COPD. Copd. 2010;7(4):262–8.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Korytina GF, Akhmadishina LZ, Tselousova OS, Zagidullin Sh Z, Viktorova TV. Polymorphism of the genes for antioxidant defense enzymes and their association with the development of chronic obstructive pulmonary disease in the population of Bashkortostan. Genetika. 2009;45(7):967–76.PubMed Korytina GF, Akhmadishina LZ, Tselousova OS, Zagidullin Sh Z, Viktorova TV. Polymorphism of the genes for antioxidant defense enzymes and their association with the development of chronic obstructive pulmonary disease in the population of Bashkortostan. Genetika. 2009;45(7):967–76.PubMed
62.
Zurück zum Zitat Juul K, Tybjaerg-Hansen A, Marklund S, Lange P, Nordestgaard BG. Genetically increased antioxidative protection and decreased chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006;173(8):858–64.PubMedCrossRef Juul K, Tybjaerg-Hansen A, Marklund S, Lange P, Nordestgaard BG. Genetically increased antioxidative protection and decreased chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006;173(8):858–64.PubMedCrossRef
63.
Zurück zum Zitat Dahl M, Bowler RP, Juul K, Crapo JD, Levy S, Nordestgaard BG. Superoxide dismutase 3 polymorphism associated with reduced lung function in two large populations. Am J Respir Crit Care Med. 2008;178(9):906–12.PubMedPubMedCentralCrossRef Dahl M, Bowler RP, Juul K, Crapo JD, Levy S, Nordestgaard BG. Superoxide dismutase 3 polymorphism associated with reduced lung function in two large populations. Am J Respir Crit Care Med. 2008;178(9):906–12.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Ganguly K, Depner M, Fattman C, Bein K, Oury TD, Wesselkamper SC, Borchers MT, Schreiber M, Gao F, von Mutius E, et al. Superoxide dismutase 3, extracellular (SOD3) variants and lung function. Physiol Genomics. 2009;37(3):260–7.PubMedPubMedCentralCrossRef Ganguly K, Depner M, Fattman C, Bein K, Oury TD, Wesselkamper SC, Borchers MT, Schreiber M, Gao F, von Mutius E, et al. Superoxide dismutase 3, extracellular (SOD3) variants and lung function. Physiol Genomics. 2009;37(3):260–7.PubMedPubMedCentralCrossRef
65.
Zurück zum Zitat Bosse Y. Updates on the COPD gene list. Int J Chron Obstruct Pulmon Dis. 2012;7:607–31. Bosse Y. Updates on the COPD gene list. Int J Chron Obstruct Pulmon Dis. 2012;7:607–31.
66.
Zurück zum Zitat Arif E, Vibhuti A, Deepak D, Singh B, Siddiqui MS, Pasha MAQ. COX2 and p53 risk-alleles coexist in COPD. Clin Chim Acta. 2008;397(1–2):46–50. Arif E, Vibhuti A, Deepak D, Singh B, Siddiqui MS, Pasha MAQ. COX2 and p53 risk-alleles coexist in COPD. Clin Chim Acta. 2008;397(1–2):46–50.
67.
Zurück zum Zitat Lee YL, Chen W, Tsai WK, Lee JC, Chiou HL, Shih CM, Wang YC. Polymorphisms of p53 and p21 genes in chronic obstructive pulmonary disease. J Lab Clin Med. 2006;147(5):228–33.PubMedCrossRef Lee YL, Chen W, Tsai WK, Lee JC, Chiou HL, Shih CM, Wang YC. Polymorphisms of p53 and p21 genes in chronic obstructive pulmonary disease. J Lab Clin Med. 2006;147(5):228–33.PubMedCrossRef
68.
Zurück zum Zitat Hua CC, Chang LC, Tseng JC, Chu CM, Liu YC, Shieh WB. Functional haplotypes in the promoter region of transcription factor Nrf2 in chronic obstructive pulmonary disease. Dis Markers. 2010;28(3):185–93.PubMedPubMedCentralCrossRef Hua CC, Chang LC, Tseng JC, Chu CM, Liu YC, Shieh WB. Functional haplotypes in the promoter region of transcription factor Nrf2 in chronic obstructive pulmonary disease. Dis Markers. 2010;28(3):185–93.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Yan F, Chen C, Jing J, Li W, Shen H, Wang X. Association between polymorphism of glutathione S-transferase P1 and chronic obstructive pulmonary disease: a meta-analysis. Respir Med. 2010;104(4):473–80.PubMedCrossRef Yan F, Chen C, Jing J, Li W, Shen H, Wang X. Association between polymorphism of glutathione S-transferase P1 and chronic obstructive pulmonary disease: a meta-analysis. Respir Med. 2010;104(4):473–80.PubMedCrossRef
70.
Zurück zum Zitat Mori M, Bjermer L, Erjefalt JS, Stampfli MR, Roos AB. Small airway epithelial-C/EBPbeta is increased in patients with advanced COPD. Respir Res. 2015;16:133.PubMedPubMedCentralCrossRef Mori M, Bjermer L, Erjefalt JS, Stampfli MR, Roos AB. Small airway epithelial-C/EBPbeta is increased in patients with advanced COPD. Respir Res. 2015;16:133.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Timchenko NA, Harris TE, Wilde M, Bilyeu TA, Burgess-Beusse BL, Finegold MJ, Darlington GJ. CCAAT/enhancer binding protein alpha regulates p21 protein and hepatocyte proliferation in newborn mice. Mol Cell Biol. 1997;17(12):7353–61.PubMedPubMedCentralCrossRef Timchenko NA, Harris TE, Wilde M, Bilyeu TA, Burgess-Beusse BL, Finegold MJ, Darlington GJ. CCAAT/enhancer binding protein alpha regulates p21 protein and hepatocyte proliferation in newborn mice. Mol Cell Biol. 1997;17(12):7353–61.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Timchenko NA, Wilde M, Nakanishi M, Smith JR, Darlington GJ. CCAAT/enhancer-binding protein alpha (C/EBP alpha) inhibits cell proliferation through the p21 (WAF-1/CIP-1/SDI-1) protein. Genes Dev. 1996;10(7):804–15.PubMedCrossRef Timchenko NA, Wilde M, Nakanishi M, Smith JR, Darlington GJ. CCAAT/enhancer-binding protein alpha (C/EBP alpha) inhibits cell proliferation through the p21 (WAF-1/CIP-1/SDI-1) protein. Genes Dev. 1996;10(7):804–15.PubMedCrossRef
74.
Zurück zum Zitat Eaton EM, Hanlon M, Bundy L, Sealy L. Characterization of C/EBPbeta isoforms in normal versus neoplastic mammary epithelial cells. J Cell Physiol. 2001;189(1):91–105.PubMedCrossRef Eaton EM, Hanlon M, Bundy L, Sealy L. Characterization of C/EBPbeta isoforms in normal versus neoplastic mammary epithelial cells. J Cell Physiol. 2001;189(1):91–105.PubMedCrossRef
75.
Zurück zum Zitat O'Rourke JP, Hutt JA, DeWille J. Transcriptional regulation of C/EBPdelta in G(0) growth-arrested mouse mammary epithelial cells. Biochem Biophys Res Commun. 1999;262(3):696–701.PubMedCrossRef O'Rourke JP, Hutt JA, DeWille J. Transcriptional regulation of C/EBPdelta in G(0) growth-arrested mouse mammary epithelial cells. Biochem Biophys Res Commun. 1999;262(3):696–701.PubMedCrossRef
76.
Zurück zum Zitat Kaisho T, Tsutsui H, Tanaka T, Tsujimura T, Takeda K, Kawai T, Yoshida N, Nakanishi K, Akira S. Impairment of natural killer cytotoxic activity and interferon gamma production in CCAAT/enhancer binding protein gamma-deficient mice. J Exp Med. 1999;190(11):1573–82.PubMedPubMedCentralCrossRef Kaisho T, Tsutsui H, Tanaka T, Tsujimura T, Takeda K, Kawai T, Yoshida N, Nakanishi K, Akira S. Impairment of natural killer cytotoxic activity and interferon gamma production in CCAAT/enhancer binding protein gamma-deficient mice. J Exp Med. 1999;190(11):1573–82.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Korytina GF, Akhmadishina LZ, Kochetova OV, Burdiuk Iu V, Aznabaeva Iu G, Zagidullin Sh Z, Victorova TV. Association of the nicotine and cigarette smoke toxicants metabolic (CHRNA3/5, CYP2A6, NQO1) and DNA repair genes (XRCC1, XRCC3, XPC, XPA) with chronic obstructive pulmonary disease. Mol Biol. 2014;48(6):939–51.CrossRef Korytina GF, Akhmadishina LZ, Kochetova OV, Burdiuk Iu V, Aznabaeva Iu G, Zagidullin Sh Z, Victorova TV. Association of the nicotine and cigarette smoke toxicants metabolic (CHRNA3/5, CYP2A6, NQO1) and DNA repair genes (XRCC1, XRCC3, XPC, XPA) with chronic obstructive pulmonary disease. Mol Biol. 2014;48(6):939–51.CrossRef
78.
Zurück zum Zitat Parkin SE, Baer M, Copeland TD, Schwartz RC, Johnson PF. Regulation of CCAAT/enhancer-binding protein (C/EBP) activator proteins by heterodimerization with C/EBPgamma (Ig/EBP). J Biol Chem. 2002;277(26):23563–72.PubMedCrossRef Parkin SE, Baer M, Copeland TD, Schwartz RC, Johnson PF. Regulation of CCAAT/enhancer-binding protein (C/EBP) activator proteins by heterodimerization with C/EBPgamma (Ig/EBP). J Biol Chem. 2002;277(26):23563–72.PubMedCrossRef
79.
Zurück zum Zitat Tuder RM, Yun JH, Graham BB. Cigarette smoke triggers code red: p21CIP1/WAF1/SDI1 switches on danger responses in the lung. Am J Respir Cell Mol Biol. 2008;39(1):1–6.PubMedPubMedCentralCrossRef Tuder RM, Yun JH, Graham BB. Cigarette smoke triggers code red: p21CIP1/WAF1/SDI1 switches on danger responses in the lung. Am J Respir Cell Mol Biol. 2008;39(1):1–6.PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Warner SM, Hackett TL, Shaheen F, Hallstrand TS, Kicic A, Stick SM, Knight DA. Transcription factor p63 regulates key genes and wound repair in human airway epithelial basal cells. Am J Respir Cell Mol Biol. 2013;49(6):978–88.PubMedPubMedCentralCrossRef Warner SM, Hackett TL, Shaheen F, Hallstrand TS, Kicic A, Stick SM, Knight DA. Transcription factor p63 regulates key genes and wound repair in human airway epithelial basal cells. Am J Respir Cell Mol Biol. 2013;49(6):978–88.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Daniely Y, Liao G, Dixon D, Linnoila RI, Lori A, Randell SH, Oren M, Jetten AM. Critical role of p63 in the development of a normal esophageal and tracheobronchial epithelium. Am J Physiol Cell Physiol. 2004;287(1):C171–81.PubMedCrossRef Daniely Y, Liao G, Dixon D, Linnoila RI, Lori A, Randell SH, Oren M, Jetten AM. Critical role of p63 in the development of a normal esophageal and tracheobronchial epithelium. Am J Physiol Cell Physiol. 2004;287(1):C171–81.PubMedCrossRef
82.
Zurück zum Zitat Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. In vivo differentiation potential of tracheal basal cells: evidence for multipotent and unipotent subpopulations. Am j physiol Lung cell mol physiol. 2004;286(4):L643–9.PubMedCrossRef Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. In vivo differentiation potential of tracheal basal cells: evidence for multipotent and unipotent subpopulations. Am j physiol Lung cell mol physiol. 2004;286(4):L643–9.PubMedCrossRef
83.
Zurück zum Zitat Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. Basal cells are a multipotent progenitor capable of renewing the bronchial epithelium. Am J Pathol. 2004;164(2):577–88.PubMedPubMedCentralCrossRef Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. Basal cells are a multipotent progenitor capable of renewing the bronchial epithelium. Am J Pathol. 2004;164(2):577–88.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Koster MI, Kim S, Mills AA, DeMayo FJ, Roop DR. p63 is the molecular switch for initiation of an epithelial stratification program. Genes Dev. 2004;18(2):126–31.PubMedPubMedCentralCrossRef Koster MI, Kim S, Mills AA, DeMayo FJ, Roop DR. p63 is the molecular switch for initiation of an epithelial stratification program. Genes Dev. 2004;18(2):126–31.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Romano RA, Smalley K, Magraw C, Serna VA, Kurita T, Raghavan S, Sinha S. DeltaNp63 knockout mice reveal its indispensable role as a master regulator of epithelial development and differentiation. Development. 2012;139(4):772–82.PubMedPubMedCentralCrossRef Romano RA, Smalley K, Magraw C, Serna VA, Kurita T, Raghavan S, Sinha S. DeltaNp63 knockout mice reveal its indispensable role as a master regulator of epithelial development and differentiation. Development. 2012;139(4):772–82.PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Candi E, Cipollone R, Rivetti di Val Cervo P, Gonfloni S, Melino G, Knight R. p63 in epithelial development. Cell Mol Life Sci. 2008;65(20):3126–33.PubMedCrossRef Candi E, Cipollone R, Rivetti di Val Cervo P, Gonfloni S, Melino G, Knight R. p63 in epithelial development. Cell Mol Life Sci. 2008;65(20):3126–33.PubMedCrossRef
87.
Zurück zum Zitat Mills AA, Zheng B, Wang XJ, Vogel H, Roop DR, Bradley A. p63 is a p53 homologue required for limb and epidermal morphogenesis. Nature. 1999;398(6729):708–13.PubMedCrossRef Mills AA, Zheng B, Wang XJ, Vogel H, Roop DR, Bradley A. p63 is a p53 homologue required for limb and epidermal morphogenesis. Nature. 1999;398(6729):708–13.PubMedCrossRef
88.
Zurück zum Zitat Yang A, Schweitzer R, Sun D, Kaghad M, Walker N, Bronson RT, Tabin C, Sharpe A, Caput D, Crum C, et al. p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature. 1999;398(6729):714–8.PubMedCrossRef Yang A, Schweitzer R, Sun D, Kaghad M, Walker N, Bronson RT, Tabin C, Sharpe A, Caput D, Crum C, et al. p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development. Nature. 1999;398(6729):714–8.PubMedCrossRef
89.
Zurück zum Zitat Arason AJ, Jonsdottir HR, Halldorsson S, Benediktsdottir BE, Bergthorsson JT, Ingthorsson S, Baldursson O, Sinha S, Gudjonsson T, Magnusson MK. deltaNp63 has a role in maintaining epithelial integrity in airway epithelium. PLoS One. 2014;9(2):e88683.PubMedPubMedCentralCrossRef Arason AJ, Jonsdottir HR, Halldorsson S, Benediktsdottir BE, Bergthorsson JT, Ingthorsson S, Baldursson O, Sinha S, Gudjonsson T, Magnusson MK. deltaNp63 has a role in maintaining epithelial integrity in airway epithelium. PLoS One. 2014;9(2):e88683.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Marshall CB, Mays DJ, Beeler JS, Rosenbluth JM, Boyd KL, Santos Guasch GL, Shaver TM, Tang LJ, Liu Q, Shyr Y, et al. p73 is required for Multiciliogenesis and regulates the Foxj1-associated gene network. Cell Rep. 2016;14(10):2289–300.PubMedPubMedCentralCrossRef Marshall CB, Mays DJ, Beeler JS, Rosenbluth JM, Boyd KL, Santos Guasch GL, Shaver TM, Tang LJ, Liu Q, Shyr Y, et al. p73 is required for Multiciliogenesis and regulates the Foxj1-associated gene network. Cell Rep. 2016;14(10):2289–300.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Crawford EL, Levin A, Safi F, Lu M, Baugh A, Zhang X, Yeo J, Khuder SA, Boulos AM, Nana-Sinkam P, et al. Lung cancer risk test trial: study design, participant baseline characteristics, bronchoscopy safety, and establishment of a biospecimen repository. BMC pulm med. 2016;16:16.PubMedPubMedCentralCrossRef Crawford EL, Levin A, Safi F, Lu M, Baugh A, Zhang X, Yeo J, Khuder SA, Boulos AM, Nana-Sinkam P, et al. Lung cancer risk test trial: study design, participant baseline characteristics, bronchoscopy safety, and establishment of a biospecimen repository. BMC pulm med. 2016;16:16.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Vestbo J, Hurd SS, Agusti AG, Jones PW, Vogelmeier C, Anzueto A, Barnes PJ, Fabbri LM, Martinez FJ, Nishimura M, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187(4):347–65.PubMedCrossRef Vestbo J, Hurd SS, Agusti AG, Jones PW, Vogelmeier C, Anzueto A, Barnes PJ, Fabbri LM, Martinez FJ, Nishimura M, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187(4):347–65.PubMedCrossRef
93.
Zurück zum Zitat Zhang X, Crawford EL, Blomquist TM, Khuder SA, Yeo J, Levin AM, Willey JC. Haplotype and diplotype analyses of variation in ERCC5 transcription cis-regulation in normal bronchial epithelial cells. Physiol Genomics. 2016;48(7):537–43.PubMedPubMedCentralCrossRef Zhang X, Crawford EL, Blomquist TM, Khuder SA, Yeo J, Levin AM, Willey JC. Haplotype and diplotype analyses of variation in ERCC5 transcription cis-regulation in normal bronchial epithelial cells. Physiol Genomics. 2016;48(7):537–43.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Vermeulen J, De Preter K, Lefever S, Nuytens J, De Vloed F, Derveaux S, Hellemans J, Speleman F, Vandesompele J. Measurable impact of RNA quality on gene expression results from quantitative PCR. Nucleic Acids Res. 2011;39(9):e63.PubMedPubMedCentralCrossRef Vermeulen J, De Preter K, Lefever S, Nuytens J, De Vloed F, Derveaux S, Hellemans J, Speleman F, Vandesompele J. Measurable impact of RNA quality on gene expression results from quantitative PCR. Nucleic Acids Res. 2011;39(9):e63.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Kusko RL, Brothers JF 2nd, Tedrow J, Pandit K, Huleihel L, Perdomo C, Liu G, Juan-Guardela B, Kass D, Zhang S, et al. Integrated genomics reveals convergent transcriptomic networks underlying chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2016;194(8):948–60.PubMedPubMedCentralCrossRef Kusko RL, Brothers JF 2nd, Tedrow J, Pandit K, Huleihel L, Perdomo C, Liu G, Juan-Guardela B, Kass D, Zhang S, et al. Integrated genomics reveals convergent transcriptomic networks underlying chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2016;194(8):948–60.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Sudmant PH, Rausch T, Gardner EJ, Handsaker RE, Abyzov A, Huddleston J, Zhang Y, Ye K, Jun G, Hsi-Yang Fritz M, et al. An integrated map of structural variation in 2,504 human genomes. Nature. 2015;526(7571):75–81.PubMedPubMedCentralCrossRef Sudmant PH, Rausch T, Gardner EJ, Handsaker RE, Abyzov A, Huddleston J, Zhang Y, Ye K, Jun G, Hsi-Yang Fritz M, et al. An integrated map of structural variation in 2,504 human genomes. Nature. 2015;526(7571):75–81.PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat Pastinen T, Sladek R, Gurd S, Sammak A, Ge B, Lepage P, Lavergne K, Villeneuve A, Gaudin T, Brandstrom H, et al. A survey of genetic and epigenetic variation affecting human gene expression. Physiol Genomics. 2004;16(2):184–93.PubMedCrossRef Pastinen T, Sladek R, Gurd S, Sammak A, Ge B, Lepage P, Lavergne K, Villeneuve A, Gaudin T, Brandstrom H, et al. A survey of genetic and epigenetic variation affecting human gene expression. Physiol Genomics. 2004;16(2):184–93.PubMedCrossRef
98.
Zurück zum Zitat Regan EA, Hokanson JE, Murphy JR, Make B, Lynch DA, Beaty TH, Curran-Everett D, Silverman EK, Crapo JD. Genetic epidemiology of COPD (COPDGene) study design. Copd. 2010;7(1):32–43.PubMedPubMedCentralCrossRef Regan EA, Hokanson JE, Murphy JR, Make B, Lynch DA, Beaty TH, Curran-Everett D, Silverman EK, Crapo JD. Genetic epidemiology of COPD (COPDGene) study design. Copd. 2010;7(1):32–43.PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Connett JE, Kusek JW, Bailey WC, O'Hara P, Wu M. Design of the Lung Health Study: a randomized clinical trial of early intervention for chronic obstructive pulmonary disease. Control Clin Trials. 1993;14(2 Suppl):3s–19s.PubMedCrossRef Connett JE, Kusek JW, Bailey WC, O'Hara P, Wu M. Design of the Lung Health Study: a randomized clinical trial of early intervention for chronic obstructive pulmonary disease. Control Clin Trials. 1993;14(2 Suppl):3s–19s.PubMedCrossRef
100.
Zurück zum Zitat Ahdesmaki M, Strimmer K. Feature selection in omics prediction problems using cat scores and false nondiscovery rate control. Ann Appl Stat. 2010;4(1):503–19.CrossRef Ahdesmaki M, Strimmer K. Feature selection in omics prediction problems using cat scores and false nondiscovery rate control. Ann Appl Stat. 2010;4(1):503–19.CrossRef
101.
Zurück zum Zitat Ge B, Pokholok DK, Kwan T, Grundberg E, Morcos L, Verlaan DJ, Le J, Koka V, Lam KC, Gagne V, et al. Global patterns of cis variation in human cells revealed by high-density allelic expression analysis. Nat Genet. 2009;41(11):1216–22.PubMedCrossRef Ge B, Pokholok DK, Kwan T, Grundberg E, Morcos L, Verlaan DJ, Le J, Koka V, Lam KC, Gagne V, et al. Global patterns of cis variation in human cells revealed by high-density allelic expression analysis. Nat Genet. 2009;41(11):1216–22.PubMedCrossRef
102.
Zurück zum Zitat Campbell I. Chi-squared and Fisher-Irwin tests of two-by-two tables with small sample recommendations. Stat Med. 2007;26(19):3661–75.PubMedCrossRef Campbell I. Chi-squared and Fisher-Irwin tests of two-by-two tables with small sample recommendations. Stat Med. 2007;26(19):3661–75.PubMedCrossRef
103.
Zurück zum Zitat Liu C, Zhang F, Li T, Lu M, Wang L, Yue W, Zhang D. MirSNP, a database of polymorphisms altering miRNA target sites, identifies miRNA-related SNPs in GWAS SNPs and eQTLs. BMC Genomics. 2012;13:661.PubMedPubMedCentralCrossRef Liu C, Zhang F, Li T, Lu M, Wang L, Yue W, Zhang D. MirSNP, a database of polymorphisms altering miRNA target sites, identifies miRNA-related SNPs in GWAS SNPs and eQTLs. BMC Genomics. 2012;13:661.PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Consortium GT. The genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348(6235):648–60.CrossRef Consortium GT. The genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348(6235):648–60.CrossRef
105.
Zurück zum Zitat Rosenbloom KR, Armstrong J, Barber GP, Casper J, Clawson H, Diekhans M, Dreszer TR, Fujita PA, Guruvadoo L, Haeussler M, et al. The UCSC genome browser database: 2015 update. Nucleic Acids Res. 2015;43(Database issue):D670–81.PubMedCrossRef Rosenbloom KR, Armstrong J, Barber GP, Casper J, Clawson H, Diekhans M, Dreszer TR, Fujita PA, Guruvadoo L, Haeussler M, et al. The UCSC genome browser database: 2015 update. Nucleic Acids Res. 2015;43(Database issue):D670–81.PubMedCrossRef
106.
Zurück zum Zitat Zhu ML, Shi TY, Hu HC, He J, Wang M, Jin L, Yang YJ, Wang JC, Sun MH, Chen H, et al. Polymorphisms in the ERCC5 gene and risk of esophageal squamous cell carcinoma (ESCC) in eastern Chinese populations. PLoS One. 2012;7(7):e41500.PubMedPubMedCentralCrossRef Zhu ML, Shi TY, Hu HC, He J, Wang M, Jin L, Yang YJ, Wang JC, Sun MH, Chen H, et al. Polymorphisms in the ERCC5 gene and risk of esophageal squamous cell carcinoma (ESCC) in eastern Chinese populations. PLoS One. 2012;7(7):e41500.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat Boyle AP, Hong EL, Hariharan M, Cheng Y, Schaub MA, Kasowski M, Karczewski KJ, Park J, Hitz BC, Weng S, et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 2012;22(9):1790–7.PubMedPubMedCentralCrossRef Boyle AP, Hong EL, Hariharan M, Cheng Y, Schaub MA, Kasowski M, Karczewski KJ, Park J, Hitz BC, Weng S, et al. Annotation of functional variation in personal genomes using RegulomeDB. Genome Res. 2012;22(9):1790–7.PubMedPubMedCentralCrossRef
108.
Zurück zum Zitat Dimas AS, Deutsch S, Stranger BE, Montgomery SB, Borel C, Attar-Cohen H, Ingle C, Beazley C, Gutierrez Arcelus M, Sekowska M, et al. Common regulatory variation impacts gene expression in a cell type-dependent manner. Science. 2009;325(5945):1246–50.PubMedPubMedCentralCrossRef Dimas AS, Deutsch S, Stranger BE, Montgomery SB, Borel C, Attar-Cohen H, Ingle C, Beazley C, Gutierrez Arcelus M, Sekowska M, et al. Common regulatory variation impacts gene expression in a cell type-dependent manner. Science. 2009;325(5945):1246–50.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Fairfax BP, Makino S, Radhakrishnan J, Plant K, Leslie S, Dilthey A, Ellis P, Langford C, Vannberg FO, Knight JC. Genetics of gene expression in primary immune cells identifies cell type-specific master regulators and roles of HLA alleles. Nat Genet. 2012;44(5):502–10.PubMedPubMedCentralCrossRef Fairfax BP, Makino S, Radhakrishnan J, Plant K, Leslie S, Dilthey A, Ellis P, Langford C, Vannberg FO, Knight JC. Genetics of gene expression in primary immune cells identifies cell type-specific master regulators and roles of HLA alleles. Nat Genet. 2012;44(5):502–10.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Melnikov A, Murugan A, Zhang X, Tesileanu T, Wang L, Rogov P, Feizi S, Gnirke A, Callan CG Jr, Kinney JB, et al. Systematic dissection and optimization of inducible enhancers in human cells using a massively parallel reporter assay. Nat Biotechnol. 2012;30(3):271–7.PubMedPubMedCentralCrossRef Melnikov A, Murugan A, Zhang X, Tesileanu T, Wang L, Rogov P, Feizi S, Gnirke A, Callan CG Jr, Kinney JB, et al. Systematic dissection and optimization of inducible enhancers in human cells using a massively parallel reporter assay. Nat Biotechnol. 2012;30(3):271–7.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Kwasnieski JC, Mogno I, Myers CA, Corbo JC, Cohen BA. Complex effects of nucleotide variants in a mammalian cis-regulatory element. Proc Natl Acad Sci U S A. 2012;109(47):19498–503.PubMedPubMedCentralCrossRef Kwasnieski JC, Mogno I, Myers CA, Corbo JC, Cohen BA. Complex effects of nucleotide variants in a mammalian cis-regulatory element. Proc Natl Acad Sci U S A. 2012;109(47):19498–503.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat White MA, Myers CA, Corbo JC, Cohen BA. Massively parallel in vivo enhancer assay reveals that highly local features determine the cis-regulatory function of ChIP-seq peaks. Proc Natl Acad Sci U S A. 2013;110(29):11952–7.PubMedPubMedCentralCrossRef White MA, Myers CA, Corbo JC, Cohen BA. Massively parallel in vivo enhancer assay reveals that highly local features determine the cis-regulatory function of ChIP-seq peaks. Proc Natl Acad Sci U S A. 2013;110(29):11952–7.PubMedPubMedCentralCrossRef
Metadaten
Titel
RNAseq analysis of bronchial epithelial cells to identify COPD-associated genes and SNPs
verfasst von
Jiyoun Yeo
Diego A. Morales
Tian Chen
Erin L. Crawford
Xiaolu Zhang
Thomas M. Blomquist
Albert M. Levin
Pierre P. Massion
Douglas A. Arenberg
David E. Midthun
Peter J. Mazzone
Steven D. Nathan
Ronald J. Wainz
Patrick Nana-Sinkam
Paige F. S. Willey
Taylor J. Arend
Karanbir Padda
Shuhao Qiu
Alexei Federov
Dawn-Alita R. Hernandez
Jeffrey R. Hammersley
Youngsook Yoon
Fadi Safi
Sadik A. Khuder
James C. Willey
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2018
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-018-0603-y

Weitere Artikel der Ausgabe 1/2018

BMC Pulmonary Medicine 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.