182
Views
9
CrossRef citations to date
0
Altmetric
Review

Candidate genes for COPD: current evidence and research

&
Pages 2249-2255 | Published online: 19 Oct 2015

Abstract

COPD is a common complex disease characterized by progressive airflow limitation. Several genome-wide association studies (GWASs) have discovered genes that are associated with COPD. Recently, candidate genes for COPD identified by GWASs include CHRNA3/5 (cholinergic nicotine receptor alpha 3/5), IREB2 (iron regulatory binding protein 2), HHIP (hedgehog-interacting protein), FAM13A (family with sequence similarity 13, member A), and AGER (advanced glycosylation end product–specific receptor). Their association with COPD susceptibility has been replicated in multiple populations. Since these candidate genes have not been considered in COPD, their pathological roles are still largely unknown. Herein, we review some evidences that they can be effective drug targets or serve as biomarkers for diagnosis or subtyping. However, more study is required to understand the functional roles of these candidate genes. Future research is needed to characterize the effect of genetic variants, validate gene function in humans and model systems, and elucidate the genes’ transcriptional and posttranscriptional regulatory mechanisms.

Introduction

COPD is a common complex disease characterized by progressive airflow limitation; the condition is not fully reversible or curable.Citation1 The global burden of this disease is high, especially in countries with older populations, and this burden is expected to increase as the populace continues aging.Citation2 Although environmental factors, including cigarette smoking and biofuel smoke exposure, are major risk factors, genetic risk factors are also important.Citation3 In addition, an interaction between genetics and environment is believed to drive the development of COPD.

Pathways that play a role in COPD pathogenesis include the response to oxidative stress, the protease–antiprotease imbalance, cell death, and inflammation.Citation4Citation7 Genetic studies have been performed to identify genetic risk factors and to understand the pathogenesis of COPD. Family-based studies and candidate gene association studies have found associations to many genes and loci. However, α-1 antitrypsin deficiency caused by mutations in SERPINA1 is the only established genetically driven cause of COPD that has a potential intervention so far.Citation8

Although whole-genome and exome sequencing may be the next tools used for the genetic study of COPD, genome-wide association study (GWAS) is currently the most widely used method for the discovery of candidate genes. Several GWASs have discovered novel genes and pathways that are associated with COPD susceptibility. Even more genes have been found to be significantly associated with lung function in the general population. Some of these lung function genes are also associated with COPD susceptibility. The genetic basis of different COPD-related phenotypes, including emphysema and chronic bronchitis, also overlaps with that of COPD susceptibility. After being implicated in disease pathogenesis, these genes can be used as potential drug targets or as biomarkers that can influence diagnosis and personalized treatment.

Currently, the most well-known candidate genes for COPD are CHRNA3/5 (cholinergic nicotine receptor alpha 3/5), IREB2 (iron regulatory binding protein 2), HHIP (hedgehog-interacting protein), FAM13A (family with sequence similarity 13, member A), and AGER (advanced glycosylation end product–specific receptor). They have been replicated in multiple populations. None of them are targeted by treatments for COPD yet, and the mechanisms by which they alter COPD risk are still largely unknown. There is some emerging evidence that they may be good targets for treatments or useful as biomarkers. However, more study is required to understand the functional roles of these candidate genes.

Future research is needed to characterize the effect of genetic variants, validate gene function in humans and model systems, and elucidate the genes’ transcriptional and post-transcriptional regulatory mechanisms.Citation9 In this review, we will cover the genetic and genomic association studies of COPD that identified the candidate genes and review what is currently known about those genes’ functions.

CHRNA3, CHRNA5, and IREB2

There are several genes at chromosome 15q25 that have been identified by GWAS for affecting COPD risk, including CHRNA3, CHRNA5, and IREB2.Citation10Citation12 The COPD cohorts investigated were the Norway case-control cohort (GenKOLS), the family-based ICGN cohort, the NETT (National Emphysema Treatment Trial)/NAS (normative aging study) cohorts, the Boston Early-Onset COPD cohort, and the COPDGene study cohort. The association between CHRNA3/5 and COPD has been replicated in multiple ethnic populations by direct genotyping.Citation13Citation15 The CHRNA3/5 locus is associated with emphysema and smoking intensity in COPD.Citation16,Citation17 The CHRNA3/5 region is also associated with lung cancer and nicotine addiction. It has been debated whether this common susceptibility region is the result of a common pathogenic pathway for lung cancer and COPD, or if it is simply associated with nicotine addiction, a risk factor for both diseases. In addition, the causal variant within the CHRNA3/5 locus may be different in lung cancer than in COPD. There is some evidence that this locus has independent roles in the pathogenesis of COPD and smoking behavior.Citation18

CHRNA3/5 and CHRNB4 are subunits of the nicotine cholinergic receptor, and the cholinergic system is active not only in cholinergic neuronal cells, but also in bronchial epithelial cells and airway inflammatory cells. The proteins are responsive to nicotine and are upregulated during chronic tobacco exposure. A recent study integrating GWAS results with expression quantitative trait loci (eQTL) study results found that single nucleotide polymorphisms (SNPs) in the 15q25 region were associated with the expression of IREB2 and CHRNA3 in blood and sputum samples.Citation19 CHRNA3/5 and IREB2 may play different roles in the pathogenesis of COPD.

IREB2 was first identified by characterizing the differential gene expression in lung tissue between COPD patients and controls, and genotyping the SNPs within the candidate regions.Citation20 IREB2 is a protein that binds iron-responsive elements (IREs), maintains cellular iron metabolism, and is regulated in response to oxygen and iron supply. IREB2 expression is higher in the lung tissue of COPD cases. The Ireb2-knockout mouse has abnormal iron metabolism in the brain, which causes cellular dysfunction.Citation21 However, the role of IREB2 in COPD pathogenesis is still not known. A GWAS of the pulmonary artery measurement obtained by computed tomography (CT) in cohorts from the COPD-Gene study and the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) study found a genome-wide significant association to IREB2.Citation22 This suggests a role for IREB2 in the pathogenesis of pulmonary hypertension in COPD, particularly in the vascular subtype.

HHIP

Many recently identified COPD-associated variants are located at chromosome 4q31, upstream of HHIP. This intergenic region has been associated with COPD susceptibility in several GWASsCitation10,Citation11,Citation23 and has consistently been replicated in multiple ethnicities.Citation24Citation27 This region is also associated with lung function in the general population.Citation28Citation30 The HHIP locus is associated with fat-free mass and exacerbations in COPD subjects.Citation16 HHIP is associated with various CT phenotypes in COPD including distinct patterns of emphysemaCitation17 and the severity of emphysema.Citation31 HHIP is more associated with emphysema measurements than with airway phenotypes and has a more significant association in emphysema subgroups.Citation32 This difference may reflect a different pathogenic process driven by HHIP, or may be driven by correlations between COPD status and imaging measurements. HHIP is also associated with adult height in the general population.Citation33 Considering that the FEV1 (forced expiratory volume in 1 second) prediction is determined by height, there may be genetic factors that control both phenotypes. Using the candidate gene strategy, it was found that the HHIP gene interacted with environmental tobacco smoke in utero, suggesting that this gene is involved in the lung response to smoke exposure in early life.Citation34

HHIP encodes a membrane glycoprotein that is an endogenous antagonist for the Hedgehog pathway. Hedgehog signaling is important for the morphogenesis of the lung and other organs.Citation35 Although the role of HHIP in COPD is not fully understood, several studies have validated the function of this gene in COPD pathogenesis. The associated SNPs are located upstream of HHIP, suggesting they may affect promoter activity. A lung eQTL study revealed that SNPs associated with COPD affect the expression of HHIP, and the risk allele of rs1828591 decreases its expression.Citation36 Zhou et alCitation37 reported that HHIP expression is reduced in COPD lung tissues and the genomic region upstream of HHIP interacts with the HHIP promoter. The risk allele of a variant in the HHIP enhancer region reduces promoter activity via a differential binding affinity to transcription factors.

These studies suggest that the genetic variation of the HHIP region affects the risk of COPD by affecting HHIP expression in lung tissues. HHIP silencing in an airway epithelial cell line leads to a change in gene expression, and these differentially expressed genes are enriched in pathways related to the extracellular matrix and cell growth, which are processes relevant to COPD pathogenesis.Citation38 Recently, Lao et alCitation39 found that Hhip-haploinsufficient mice have increased airspace size after cigarette smoke exposure, increased lung compliance, and increased numbers of lymphoid aggregates. The functions of the genes with altered expression in Hhip+/− mice exposed to cigarette smoke were enriched in the pathway of lymphocyte activation. They used haploinsufficient mice because Hhip−/− mice die shortly after birth due to lung branching morphogenesis failure.

HHIP was also found to be associated with lung cancer by a candidate gene study.Citation40 The Hedgehog pathway is a critical mediator of cigarette-smoke-induced lung cancer, and it may act as a common pathway for the development of COPD and lung cancer.Citation41

FAM13A

A GWAS using three COPD cohorts, GenKOLS, NETT/NAS, and the ECLIPSE study, identified variants at chromosome 4q22 in the gene FAM13A.Citation42 These are some of the most highly associated SNPs in COPD and are located in an intron. These associations have been replicated in a subset of the patients in the COPDGene study and the cohort of the International COPD Genetics Network. They have also been replicated in Asian populations, assayed using the candidate gene strategy.Citation43,Citation44 FAM13A was first found to associate with lung function in a GWAS using the general population,Citation28 and it is associated with lung function in asthmatic subjects.Citation45 GWASs using COPD cases with chronic bronchitis in the COPDGene study, GenKOLS, and ECLIPSE cohorts identified a significant association with FAM13A,Citation46 whereas several GWASs for emphysema did not identify a genome-wide association. The odds ratios of FAM13A SNPs for COPD with chronic bronchitis were significantly higher than those for nonchronic bronchitis COPD, suggesting that FAM13A is more related to the pathogenesis of the chronic bronchitis subtype. Of note, FAM13A is also associated with idiopathic pulmonary disease,Citation47 but the expression of FAM13A in lung tissues does not differ by case/control status or by genotype.

FAM13A was originally identified in cattle near a quantitative trait locus affecting milk production, and is expressed in the kidney, pancreas, lung, and thymus.Citation48 Although the function of FAM13A has not been extensively studied, its RhoGAP domain may be related to COPD. Rho GTPases are key regulators of cytoskeletal dynamics, are involved in the pulmonary endothelial barrier, and are dysregulated in several lung diseases.Citation49 A lung eQTL study suggested that the expression of FAM13A may be associated with particular SNPs.Citation36 In the case of COPD, the FAM13A risk allele is associated with increased FAM13A expression in the lung, although expression does not differ in lung tissues between COPD cases and controls.Citation43 A recent study by Jin et alCitation50 found that FAM13A activates Wnt signaling by increasing the stability of β-catenin. Although depletion of FAM13A in a lung cancer cell line reduces Wnt signaling activity, FAM13A-knockout mice are viable and FAM13A-mutant lungs are morphologically indistinguishable from wild-type lungs, and Wnt signaling remains normal in Fam13a-knockout lungs. They also found that Akt regulates the phosphorylation of FAM13A, which can lead to cytoplasmic sequestration of FAM13A. Considering that Akt has a role in the pathogenesis of COPD,Citation51 FAM13A may contribute to lung disease through aberrant Akt signaling. Further work is needed to validate the functional role of FAM13A in the pathogenesis of COPD.

AGER

GWASs of lung function in the general population have found that chromosome 6p21 is associated to FEV1/FVC (forced vital capacity) and FEV1, which are important physiologic parameters of COPD.Citation30,Citation28,Citation52 This association was investigated in COPD patients identified from the population cohort using spirometry criteria, and the study found a suggestive association between COPD risk and AGER, although it was not statistically significant.Citation53 A candidate gene study in NETT/NAS, GenKOLS, ECLIPSE, and a subset of the COPDGene study cohort found that it was associated with COPD susceptibility, although a subsequent GWAS did not find a significant association.Citation54 On the other hand, an association has been found in multiple ethnic populations.Citation55

The 6p21 region that showed significant association with COPD includes many genes, including TNXB, PPT2, AGER, and NOTCH4. However, AGER has a potential functional variant, rs2070600, and it has been studied the most in the pathogenesis of COPD. A GWAS of percent emphysema determined by CT using the Multi-Ethnic Study of Atherosclerosis cohort identified a significant association with the AGER/PPT region.Citation56 This region did associate with emphysema severity and gas trapping in a GWAS using cohorts from the COPDGene, ECLIPSE, NETT, and GenKOLS studies.Citation31

The protein product of AGER, the receptor for advanced glycan end products (RAGE), is a multiligand receptor of the immunoglobulin superfamily and interacts with molecules implicated in homeostatic function, inflammation, and development.Citation57 RAGE levels are increased in the airway and alveolar walls of COPD lungs.Citation58 RAGE expression in mice increases after cigarette smoke exposure, and cigarette-smoking-induced inflammatory responses by alveolar macrophages are diminished in RAGE-knockout mice.Citation59 Transgenic mice with upregulated RAGE have impaired alveolar morphogenesis during lung development, distal airspace enlargement, and increased alveolar cell apoptosis.Citation60 Another study using RAGE transgenic mice found incremental dilation of alveolar spaces, as well as pronounced inflammation in the peripheral lung and alveolar destabilization.Citation61 A promoter variant of AGER in cystic fibrosis patients is associated with poor lung function, and it increases expression in airway epithelial cell lines, suggesting it is a modifier of lung disease severity.Citation62

The soluble isoform of RAGE (sRAGE) contains the RAGE extracellular domain and can bind to circulating proinflammatory ligands, preventing RAGE activation. Mice that are exposed to chronic hypoxia have downregulated pulmonary RAGE protein and increased levels of sRAGE, which might be adaptive to and protective against chronic hypoxia.Citation63 Circulatory levels of sRAGE are reduced in COPD patients.Citation64 Reduced sRAGE levels are associated with increased emphysema in two COPD cohorts.Citation65 Decreased plasma sRAGE levels are also associated with the progression of airflow limitation over time.Citation66 In patients of the Treatment of Emphysema with a Selective Retinoid Agonist (TESRA) and ECLIPSE studies, sRAGE is associated with diffusing capacity, emphysema, and COPD disease status, and the variant rs2070600 is associated with circulating sRAGE levels.Citation67 The significant association between rs2070600 and plasma sRAGE levels was also found in Dutch diabetes mellitus and control subjects.Citation68 RAGE has been studied in metabolic diseases, and decreased levels of sRAGE are linked to vascular complications. RAGE contributes to the pathogenesis of COPD in the lung probably via the regulation of inflammation and apoptosis, and further study of the functions of this gene may lead to it being identified as a potential therapeutic target.

Other candidate genes

There have been several more regions identified in GWASs of COPD. A GWAS using subjects from the ECLIPSE, NETT/NAS, GenKOLS, and COPDGene studies identified chromosome 19q13 as being associated with COPD, along with the previously identified HHIP, FAM13A, and 15q25 regions.Citation11 Chromosome 19q13 contains CYP2A6, RAB4B, MIA, and EGLN, which could potentially be involved in COPD pathogenesis, and EGLN2 was found to be dysregulated in the airway epithelium of smokers.Citation69 A GWAS using the full COPDGene cohort identified additional associations with TGFB2, MMP12, and RIN3.Citation23 TGFB2 and MMP12 have been previously studied in COPD or related phenotypes,Citation70,Citation71 whereas RIN3 has not been studied in COPD and needs to be investigated further. SERPINE2 was identified using a linkage analysis of gene expression changes in lung tissue.Citation72 A recent GWAS of airway thickness identified rs734556 on chromosome 2q, which is associated with SERPINE2 expression.Citation73

An association study of SNP in GSTDS on chromosome 4q24, which was originally shown to be associated with lung function in general population, was performed in COPD case and control subjects. Variant in GSTDS was significantly associated with COPD susceptibility.Citation53 SNPs in this region were also tested in GenKOLS, NETT, ECLIPSE, and part of COPDGene, and association with COPD susceptibility was significant.Citation54 GSTCD and INTS12 genes in 4q24 are expressed in human lung tissues and airway cells and expression of these genes was altered by TGFβ1 in human airway smooth muscle cells.Citation74 Top SNPs associated with airflow obstruction defined by FEV1 and FEV1/FVC less than lower limits of normal were tested in the COPD meta-analysis. In this analysis, ADAM19 on chromosome 5q33 was nominally associated with COPD.Citation12 This gene was also associated with FEV1/FVC in the CHARGE consortium.Citation28 This region was nominally associated with COPD susceptibility.Citation54 A GWAS of emphysema in the three COPD cohorts including the Norway cohort, ECLIPSE, and NETT revealed that BICD1 was the most associated with the presence or absence of emphysema.Citation75 The emphysema-predominant mild COPD of COPDGene study was nominally associated with BICD1.

These associations require more replications and further fine-mapping studies are needed to find the causal variants of COPD, as well as studies to functionally validate the identified genes.

Future directions

Subtyping or classification, drug development, and risk prediction are potential clinical applications of the associated genes. Phenotype heterogeneity can be investigated using association studies on various COPD-related phenotypes. Although the associations merely reflect the correlations between COPD status and phenotypes, there may be subtypes with different genetic alterations. Considering that treatment with PDE4 inhibitor is effective only for the chronic bronchitis subtype, there may be a mechanism that is unique to this subtype. Candidate genes can be used to determine personalized treatment because they may help identify a subtype-unique pathogenesis, variation in a drug-action site, or variable drug metabolism.Citation76

More regions have been identified in GWASs on FEV1 and FEV1/FVC in the general population, probably because of the larger sample sizes. GWASs using a greater sample size of COPD subjects may find more candidate genes.Citation77 Another approach for finding more candidate genes is to identify rare variation using exome sequencing or arrays.

Functional studies for COPD candidate genes are summarized in . Although there are a few potential drug targets that are implicated in the novel COPD pathogenesis, it will take many years to apply them in the treatment.

Table 1 Current candidate genes for COPD and their potential functional mechanisms in the COPD pathogenesis

Conclusion

Recently, several candidate genes associated with COPD risk have been identified using GWAS. Replication and functional validation studies may lead to clinical applications for these genes such as novel therapeutics, subtyping, and risk prediction for COPD.

Disclosure

The authors report no conflicts of interest in this work.

References

  • VestboJHurdSSAgustíAGGlobal strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary diseaseAm J Respir Crit Care Med201318734736522878278
  • PrinceMJWuFGuoYThe burden of disease in older people and implications for health policy and practiceLancet201538554956225468153
  • PostmaDSBushAvan den BergeMRisk factors and early origins of chronic obstructive pulmonary diseaseLancet201538589990925123778
  • CosioMGSaettaMAgustiAImmunologic aspects of chronic obstructive pulmonary diseaseN Engl J Med20093602445245419494220
  • TuderRPetracheIPathogenesis of chronic obstructive pulmonary diseaseJ Clin Invest20121222749275522850885
  • KirkhamPABarnesPJOxidative stress in COPDChest201314426627323880677
  • BagdonasERaudoniuteJBruzauskaiteINovel aspects of pathogenesis and regeneration mechanisms in COPDInt J Chron Obstruct Pulmon Dis201510995101326082624
  • SilvermanEKSandhausRAAlpha1-antitrypsin deficiencyN Engl J Med20093602749275719553648
  • OberCButteAJEliasJAGetting from genes to function in lung diseaseAm J Respir Crit Care Med201018273273720558629
  • PillaiSGGeDZhuGA genome-wide association study in chronic obstructive pulmonary disease (COPD): identification of two major susceptibility lociPLoS Genet20095e100042119300482
  • ChoMHCastaldiPJWanESA genome-wide association study of COPD identifies a susceptibility locus on chromosome 19q13Hum Mol Genet20122194795722080838
  • WilkJBShrineNRLoehrLRGenome-wide association studies identify CHRNA5/3 and HTR4 in the development of airflow obstructionAm J Respir Crit Care Med201218662263222837378
  • HardinMZielinskiJWanESCHRNA3/5, IREB2, and ADCY2 are associated with severe chronic obstructive pulmonary disease in PolandAm J Respir Cell Mol Biol20124720320822461431
  • KimWJOhYMKimTHCHRNA3 variant for lung cancer is associated with chronic obstructive pulmonary disease in KoreaRespiration20138611712223207642
  • YangLQiuFLuXFunctional polymorphisms of CHRNA3 predict risks of chronic obstructive pulmonary disease and lung cancer in ChinesePLoS One20127e4607123056235
  • PillaiSGKongXEdwardsLDLoci identified by genome-wide association studies influence different disease-related phenotypes in chronic obstructive pulmonary diseaseAm J Respir Crit Care Med20101821498150520656943
  • CastaldiPJChoMHSan Jose EsteparRGenome-wide association identifies regulatory Loci associated with distinct local histogram emphysema patternsAm J Respir Crit Care Med201419039940925006744
  • SiedlinskiMTingleyDLipmanPJDissecting direct and indirect genetic effects on chronic obstructive pulmonary disease (COPD) susceptibilityHum Genet201313243144123299987
  • CastaldiPJChoMHZhouXGenetic control of gene expression at novel and established chronic obstructive pulmonary disease lociHum Mol Genet2015241200121025315895
  • DeMeoDLMarianiTBhattacharyaSIntegration of genomic and genetic approaches implicates IREB2 as a COPD susceptibility geneAm J Hum Genet20098549350219800047
  • Zumbrennen-BulloughKBBeckerLGarrettLAbnormal brain iron metabolism in Irp2 deficient mice is associated with mild neurological and behavioral impairmentsPLoS One20149e9807224896637
  • LeeJHChoMHHershCPIREB2 and GALC are associated with pulmonary artery enlargement in chronic obstructive pulmonary diseaseAm J Respir Cell Mol Biol20155236537625101718
  • ChoMHMcDonaldMLZhouXRisk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysisLancet Respir Med2014221422524621683
  • Van DurmeYMEijgelsheimMJoosGFHedgehog-interacting protein is a COPD susceptibility gene: the Rotterdam StudyEur Respir J201036899519996190
  • KimWJOhYMLeeJHGenetic variants in HHIP are associated with FEV1 in subjects with chronic obstructive pulmonary diseaseRespirology2013181202120923731023
  • XieJWuHXuYGene susceptibility identification in a longitudinal study confirms new loci in the development of chronic obstructive pulmonary disease and influences lung function declineRespir Res2015164925928290
  • WangBZhouHYangJAssociation of HHIP polymorphisms with COPD and COPD-related phenotypes in a Chinese Han populationGene201353110110523994291
  • HancockDBEijgelsheimMWilkJBMeta-analyses of genome-wide association studies identify multiple loci associated with pulmonary functionNat Genet201042455220010835
  • WilkJBChenTHGottliebDJA genome-wide association study of pulmonary function measures in the Framingham Heart StudyPLoS Genet20095e100042919300500
  • RepapiESayersIWainLVGenome-wide association study identifies five loci associated with lung functionNat Genet201042364420010834
  • ChoMHCastaldiPJHershCPA genome-wide association study of emphysema and airway quantitative imaging phenotypesAm J Respir Crit Care Med201519255956926030696
  • CastaldiPJDyJRossJCluster analysis in the COPDGene study identifies subtypes of smokers with distinct patterns of airway disease and emphysemaThorax201469416423
  • van der ValkRJPKreiner-MøllerEKooijmanMNA novel common variant in DCST2 is associated with length in early life and height in adulthoodHum Mol Genet2015241155116825281659
  • KerkhofMBoezenHMGranellRTransient early wheeze and lung function in early childhood associated with chronic obstructive pulmonary disease genesJ Allergy Clin Immunol20141336876.e6423886569
  • ChuangPTKawcakTMcMahonAPFeedback control of mammalian Hedgehog signaling by the Hedgehog-binding protein, Hip1, modulates Fgf signaling during branching morphogenesis of the lungGenes Dev20031734234712569124
  • LamontagneMCoutureCPostmaDSRefining susceptibility loci of chronic obstructive pulmonary disease with lung EQTLSPLoS One20138e7022023936167
  • ZhouXBaronRMHardinMIdentification of a chronic obstructive pulmonary disease genetic determinant that regulates HHIPHum Mol Genet2012211325133522140090
  • ZhouXQiuWSathirapongsasutiJFGene expression analysis uncovers novel hedgehog interacting protein (HHIP) effects in human bronchial epithelial cellsGenomics201310126327223459001
  • LaoTGlassKQiuWHaploinsufficiency of Hedgehog interacting protein causes increased emphysema induced by cigarette smoke through network rewiringGenome Med201571225763110
  • YoungRPWhittingtonCFHopkinsRJChromosome 4q31 locus in COPD is also associated with lung cancerEur Respir J2010361375138221119205
  • Lemjabbar-AlaouiHDasariVSidhuSSWnt and Hedgehog are critical mediators of cigarette smoke-induced lung cancerPLoS One20061e9317183725
  • ChoMHBoutaouiNKlandermanBJVariants in FAM13A are associated with chronic obstructive pulmonary diseaseNat Genet20104220020220173748
  • KimWJLimMNHongYAssociation of lung function genes with chronic obstructive pulmonary diseaseLung201419247348024737086
  • GuoYLinHGaoKGenetic analysis of IREB2, FAM13A and XRCC5 variants in Chinese Han patients with chronic obstructive pulmonary diseaseBiochem Biophys Res Commun201141528428722027142
  • LiXHawkinsGAAmplefordEJGenome-wide association study identifies TH1 pathway genes associated with lung function in asthmatic patientsJ Allergy Clin Immunol2013132313320.e31523541324
  • LeeJHChoMHHershCPGenetic susceptibility for chronic bronchitis in chronic obstructive pulmonary diseaseRespir Res20141511325241909
  • FingerlinTEMurphyEZhangWGenome-wide association study identifies multiple susceptibility loci for pulmonary fibrosisNat Genet20134561362023583980
  • CohenMReichensteinMEverts-van der WindACloning and characterization of FAM13A1 – a gene near a milk protein QTL on BTA6: evidence for population-wide linkage disequilibrium in Israeli HolsteinsGenomics20048437438315234000
  • CorvolHHodgesCADrummMLMoving beyond genetics: is FAM13A a major biological contributor in lung physiology and chronic lung diseases?J Med Genet20145164664925163686
  • JinZChungJWMeiWRegulation of nuclear-cytoplasmic shuttling and function of Family with sequence similarity 13, member A (Fam13a), by B56-containing PP2As and AktMol Biol Cell2015261160117325609086
  • BozinovskiSVlahosRHansenMAkt in the pathogenesis of COPDInt J Chron Obstruct Pulmon Dis20061313818046900
  • KimWJLeeMKShinCGenome-wide association studies identify locus on 6p21 influencing lung function in the Korean populationRespirology20141936036824387323
  • Soler ArtigasMWainLVRepapiEEffect of five genetic variants associated with lung function on the risk of chronic obstructive lung disease, and their joint effects on lung functionAm J Respir Crit Care Med201118478679521965014
  • CastaldiPJChoMHLitonjuaAAThe association of genome-wide significant spirometric loci with chronic obstructive pulmonary disease susceptibilityAm J Respir Cell Mol Biol2011451147115321659657
  • LiYYangCMaGAssociation of polymorphisms of the receptor for advanced glycation end products gene with COPD in the Chinese populationDNA Cell Biol20143325125824520905
  • ManichaikulAHoffmanEASmolonskaJGenome-wide study of percent emphysema on computed tomography in the general population. The multi-ethnic study of atherosclerosis lung/SNP health association resource studyAm J Respir Crit Care Med201418940841824383474
  • SorciGRiuzziFGiambancoIRAGE in tissue homeostasis, repair and regenerationBiochim Biophys Acta2013183310110923103427
  • WuLMaLNicholsonLFBAdvanced glycation end products and its receptor (RAGE) are increased in patients with COPDRespir Med201110532933621112201
  • RobinsonABJohnsonKDBennionBGRAGE signaling by alveolar macrophages influences tobacco smoke-induced inflammationAm J Physiol Lung Cell Mol Physiol2012302L1192L119922505673
  • FineschiSDe CuntoGFacchinettiFReceptor for advanced glycation end products contributes to postnatal pulmonary development and adult lung maintenance program in miceAm J Respir Cell Mol Biol20134816417123144333
  • StogsdillMPStogsdillJABodineBGConditional overexpression of receptors for advanced glycation end-products in the adult murine lung causes airspace enlargement and induces inflammationAm J Respir Cell Mol Biol20134912813423526218
  • BeucherJBoëlleP-YBussonP-FAGER-429T/C is associated with an increased lung disease severity in cystic fibrosisPLoS One20127e4191322860029
  • GopalPGoskerHRTheijeCCEffect of chronic hypoxia on RAGE and its soluble forms in lungs and plasma of miceBiochim Biophys Acta20151852992100025703138
  • MiniatiMMontiSBastaGSoluble receptor for advanced glycation end products in COPD: relationship with emphysema and chronic cor pulmonale: a case-control studyRespir Res2011123721450080
  • CarolanBHughesGMorrowJThe association of plasma biomarkers with computed tomography-assessed emphysema phenotypesRespir Res20141512725306249
  • IwamotoHGaoJPulkkinenVSoluble receptor for advanced glycation end-products and progression of airway diseaseBMC Pulm Med2014146824758342
  • ChengDTKimDKCockayneDASystemic soluble receptor for advanced glycation endproducts is a biomarker of emphysema and associated with AGER genetic variants in patients with chronic obstructive pulmonary diseaseAm J Respir Crit Care Med201318894895723947473
  • GaensKHJFerreiraIvan der KallenCJHAssociation of polymorphism in the receptor for advanced glycation end products (RAGE) gene with circulating RAGE levelsJ Clin Endocrinol Metabol20099451745180
  • RyanDMVincentTLSalitJSmoking dysregulates the human airway basal cell transcriptome at COPD risk locus 19q13.2PLoS One20149e8805124498427
  • HunninghakeGMChoMHTesfaigziYMMP12, lung function, and COPD in high-risk populationsN Engl J Med20093612599260820018959
  • ArtigasMSLothDWWainLVGenome-wide association and large-scale follow up identifies 16 new loci influencing lung functionNat Genet2011431082109021946350
  • DeMeoDLMarianiTJLangeCThe SERPINE2 gene is associated with chronic obstructive pulmonary diseaseAm J Hum Genet20067825326416358219
  • DijkstraAEPostmaDSvan GinnekenBNovel genes for airway wall thickness identified with combined genome-wide association and expression analysesAm J Respir Crit Care Med201419154755625517131
  • ObeidatMMillerSProbertKGSTCD and INTS12 regulation and expression in the human lungPLoS One20138e7463024058608
  • KongXChoMHAndersonWGenome-wide association study identifies BICD1 as a susceptibility gene for emphysemaAm J Respir Crit Care Med2011183434920709820
  • HershCPPharmacogenetics of chronic obstructive pulmonary disease: challenges and opportunitiesPharmacogenomics20101123724720136362
  • SilvermanEKVestboJAgustiAOpportunities and challenges in the genetics of COPD 2010: an international COPD genetics conference reportCOPD2011812113521495840