Integrative genomics identifies new genes associated with severe COPD and emphysema

Phuwanat Sakornsakolpat, Jarrett D Morrow, Peter J Castaldi, Craig P Hersh, Yohan Bossé, Edwin K Silverman, Ani Manichaikul, Michael H Cho, Phuwanat Sakornsakolpat, Jarrett D Morrow, Peter J Castaldi, Craig P Hersh, Yohan Bossé, Edwin K Silverman, Ani Manichaikul, Michael H Cho

Abstract

Background: Genome-wide association studies have identified several genetic risk loci for severe chronic obstructive pulmonary disease (COPD) and emphysema. However, these studies do not fully explain disease heritability and in most cases, fail to implicate specific genes. Integrative methods that combine gene expression data with GWAS can provide more power in discovering disease-associated genes and give mechanistic insight into regulated genes.

Methods: We applied a recently described method that imputes gene expression using reference transcriptome data to genome-wide association studies for two phenotypes (severe COPD and quantitative emphysema) and blood and lung tissue gene expression datasets. We further tested the potential causality of individual genes using multi-variant colocalization.

Results: We identified seven genes significantly associated with severe COPD, and five genes significantly associated with quantitative emphysema in whole blood or lung. We validated results in independent transcriptome databases and confirmed colocalization signals for PSMA4, EGLN2, WNT3, DCBLD1, and LILRA3. Three of these genes were not located within previously reported GWAS loci for either phenotype. We also identified genetically driven pathways, including those related to immune regulation.

Conclusions: An integrative analysis of GWAS and gene expression identified novel associations with severe COPD and quantitative emphysema, and also suggested disease-associated genes in known COPD susceptibility loci.

Trial registration: NCT00608764 , Registry: ClinicalTrials.gov, Date of Enrollment of First Participant: November 2007, Date Registered: January 28, 2008 (retrospectively registered); NCT00292552 , Registry: ClinicalTrials.gov, Date of Enrollment of First Participant: December 2005, Date Registered: February 14, 2006 (retrospectively registered).

Keywords: Chronic obstructive pulmonary disease; Emphysema; Gene expression; Genome-wide association studies.

Conflict of interest statement

Ethics approval and consent to participate

Local institutional review boards provided ethical approval for the clinical centers. Written informed consent was obtained in all studies.

Consent for publication

Not applicable

Competing interests

The 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.

Figures

Fig. 1
Fig. 1
Summary of analyses. First, we discovered transcriptome-disease associations (predicted gene expression-disease) using reference data from DGN-Blood and GTEx-Lung. Then, we validated these associations using another set of reference data (GTEx-Blood and Lung-eQTL Consortium). Finally, we confirmed the transcriptome-disease associations using colocalization analysis. COPD = chronic obstructive pulmonary disease; DGN = Depression Gene Network; GTEx = Genotype-Tissue Expression project; Perc15 HU at 15th percentile of the density histogram; severe COPD is defined as FEV1 < 50% predicted and FEV1/FVC < 0.7
Fig. 2
Fig. 2
Manhattan plots of associations of imputed gene expression and phenotypes (severe COPD in the upper panel; %LAA-950 and Perc15 in the lower panel). Color indicates phenotypes and shape indicates tissue (see figure legend)
Fig. 3
Fig. 3
Regional association plots within 50 kb of WNT3. GWAS of severe COPD and lung eQTL are shown in the upper panel. Chromatin states and epigenomic marks of normal human lung fibroblasts are shown in the lower panel (see Additional file 1: Supplementary Methods)

References

    1. 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;188:941–947. doi: 10.1164/rccm.201302-0263OC.
    1. Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 report. GOLD executive summary. Am J Respir Crit Care Med. 2017;195:557–582. doi: 10.1164/rccm.201701-0218PP.
    1. Cho MH, McDonald M-LLN, Zhou X, Mattheisen M, Castaldi PJ, Hersh CP, et al. Risk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysis. Lancet Respir Med. 2014;2:214–225. doi: 10.1016/S2213-2600(14)70002-5.
    1. Hobbs BD, de Jong K, Lamontagne M, Bosse Y, Shrine N, Artigas MS, et al. Genetic loci associated with chronic obstructive pulmonary disease overlap with loci for lung function and pulmonary fibrosis. Nat Genet. 2017;49:426–432. doi: 10.1038/ng.3752.
    1. Cho MH, Castaldi PJ, Hersh CP, Hobbs BD, Barr RG, Tal-Singer R, et al. A genome-wide association study of emphysema and airway quantitative imaging phenotypes. Am J Respir Crit Care Med. 2015;192:559–569. doi: 10.1164/rccm.201501-0148OC.
    1. Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS, et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A. 2009;106:9362–9367. doi: 10.1073/pnas.0903103106.
    1. Nicolae DL, Gamazon E, Zhang W, Duan S, Dolan ME, Cox NJ. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 2010;6:e1000888. doi: 10.1371/journal.pgen.1000888.
    1. Gamazon ER, Wheeler HE, Shah KP, Mozaffari SV, Aquino-Michaels K, Carroll RJ, et al. A gene-based association method for mapping traits using reference transcriptome data. Nat Genet. 2015;47:1091–1098. doi: 10.1038/ng.3367.
    1. Gusev A, Ko A, Shi H, Bhatia G, Chung W, Penninx BWJH, et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat Genet. 2016;48:245–252. doi: 10.1038/ng.3506.
    1. Barbeira A, Dickinson SP, Torres JM, Torstenson ES, Zheng J, Wheeler HE, et al. Integrating tissue specific mechanisms into GWAS summary results. bioRxiv. 2017.
    1. Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, Brown MA, et al. 10 years of GWAS discovery: biology, function, and translation. Am J Hum Genet. 2017;101:5–22. doi: 10.1016/j.ajhg.2017.06.005.
    1. Zhou X, Baron RM, Hardin M, Cho MH, Zielinski J, Hawrylkiewicz I, et al. Identification of a chronic obstructive pulmonary disease genetic determinant that regulates HHIP. Hum Mol Genet. 2012;21:1325–1335. doi: 10.1093/hmg/ddr569.
    1. Flister MJ, Tsaih S-W, O’Meara CC, Endres B, Hoffman MJ, Geurts AM, et al. Identifying multiple causative genes at a single GWAS locus. Genome Res. 2013;23:1996–2002. doi: 10.1101/gr.160283.113.
    1. Claussnitzer M, Hui C-C, Kellis M. FTO obesity variant and adipocyte Browning in humans. N Engl J Med. 2016;374:192–193.
    1. Hormozdiari F, van de Bunt M, Segrè AV, Li X, Joo JWJ, Bilow M, et al. Colocalization of GWAS and eQTL signals detects target genes. Am J Hum Genet. 2016;99:1245–1260. doi: 10.1016/j.ajhg.2016.10.003.
    1. Cho MH, Castaldi PJ, Wan ES, Siedlinski M, Hersh CP, Demeo DL, et al. A genome-wide association study of COPD identifies a susceptibility locus on chromosome 19q13. Hum Mol Genet. 2012;21:947–957. doi: 10.1093/hmg/ddr524.
    1. Liu JZ, McRae AF, Nyholt DR, Medland SE, Wray NR, Brown KM, et al. A versatile gene-based test for genome-wide association studies. Am J Hum Genet. 2010;87:139–145. doi: 10.1016/j.ajhg.2010.06.009.
    1. Lutz SM, Cho MH, Young K, Hersh CP, Castaldi PJ, McDonald M-LL, et al. A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry. BMC Genet. 2015;16:138. doi: 10.1186/s12863-015-0299-4.
    1. Wain LV, Shrine N, Miller S, Jackson VE, Ntalla I, Soler Artigas M, et al. Novel insights into the genetics of smoking behaviour, lung function, and chronic obstructive pulmonary disease (UK BiLEVE): a genetic association study in UK biobank. Lancet Respir Med. 2015;3:769–781. doi: 10.1016/S2213-2600(15)00283-0.
    1. Fingerlin TE, Murphy E, Zhang W, Peljto AL, Brown KK, Steele MP, et al. Genome-wide association study identifies multiple susceptibility loci for pulmonary fibrosis. Nat Genet. 2013;45:613–620. doi: 10.1038/ng.2609.
    1. Noth I, Zhang Y, Ma S-F, Flores C, Barber M, Huang Y, et al. Genetic variants associated with idiopathic pulmonary fibrosis susceptibility and mortality: a genome-wide association study. Lancet Respir Med. 2013;1:309–317. doi: 10.1016/S2213-2600(13)70045-6.
    1. Clevers H, Nusse R. Wnt/beta-catenin signaling and disease. Cell. 2012;149:1192–1205. doi: 10.1016/j.cell.2012.05.012.
    1. Barrow JR, Thomas KR, Boussadia-Zahui O, Moore R, Kemler R, Capecchi MR, et al. Ectodermal Wnt3/beta-catenin signaling is required for the establishment and maintenance of the apical ectodermal ridge. Genes Dev. 2003;17:394–409. doi: 10.1101/gad.1044903.
    1. Hill-Burns EM, Wissemann WT, Hamza TH, Factor SA, Zabetian CP, Payami H. Identification of a novel Parkinson’s disease locus via stratified genome-wide association study. BMC Genomics. 2014;15:118. doi: 10.1186/1471-2164-15-118.
    1. Dubois PCA, Trynka G, Franke L, Hunt KA, Romanos J, Curtotti A, et al. Multiple common variants for celiac disease influencing immune gene expression. Nat Genet. 2010;42:295–302. doi: 10.1038/ng.543.
    1. Wang R, Ahmed J, Wang G, Hassan I, Strulovici-Barel Y, Hackett NR, et al. Down-regulation of the canonical Wnt beta-catenin pathway in the airway epithelium of healthy smokers and smokers with COPD. PLoS One. 2011;6:e14793. doi: 10.1371/journal.pone.0014793.
    1. Jiang Z, Lao T, Qiu W, Polverino F, Gupta K, Guo F, et al. A chronic obstructive pulmonary disease susceptibility gene, FAM13A, regulates protein stability of beta-catenin. Am J Respir Crit Care Med. 2016;194:185–197. doi: 10.1164/rccm.201505-0999OC.
    1. Baarsma HA, Königshoff M. “WNT-er is coming”: WNT signalling in chronic lung diseases. Thorax. 2017;72:746–759. doi: 10.1136/thoraxjnl-2016-209753.
    1. Spracklen CN, Chen P, Kim YJ, Wang X, Cai H, Li S, et al. Association analyses of east Asian individuals and trans-ancestry analyses with European individuals reveal new loci associated with cholesterol and triglyceride levels. Hum Mol Genet. 2017;26:1770–1784. doi: 10.1093/hmg/ddx062.
    1. Xu J, Mo Z, Ye D, Wang M, Liu F, Jin G, et al. Genome-wide association study in Chinese men identifies two new prostate cancer risk loci at 9q31.2 and 19q13.4. Nat Genet. 2012;44:1231–1235. doi: 10.1038/ng.2424.
    1. Melé M, Ferreira PG, Reverter F, DeLuca DS, Monlong J, Sammeth M, et al. Human genomics. The human transcriptome across tissues and individuals. Science. 2015;348:660–665. doi: 10.1126/science.aaa0355.
    1. Wright FA, Sullivan PF, Brooks AI, Zou F, Sun W, Xia K, et al. Heritability and genomics of gene expression in peripheral blood. Nat Genet. 2014;46:430–437. doi: 10.1038/ng.2951.
    1. Consortium GTe Human genomics. The genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348:648–660. doi: 10.1126/science.1262110.
    1. Reference Genome Group of the Gene Ontology Consortium The Gene Ontology’s Reference Genome Project: a unified framework for functional annotation across species. PLoS Comput Biol. 2009;5:e1000431. doi: 10.1371/journal.pcbi.1000431.
    1. Lan Q, Hsiung CA, Matsuo K, Hong Y-CC, Seow A, Wang Z, et al. Genome-wide association analysis identifies new lung cancer susceptibility loci in never-smoking women in Asia. Nat Genet. 2012;44:1330–1335. doi: 10.1038/ng.2456.
    1. Maurano MT, Humbert R, Rynes E, Thurman RE, Haugen E, Wang H, et al. Systematic localization of common disease-associated variation in regulatory DNA. Science. 2012;337:1190–1195. doi: 10.1126/science.1222794.
    1. Shooshtari P, Huang H, Cotsapas C. Integrative genetic and epigenetic analysis uncovers regulatory mechanisms of autoimmune disease. Am J Hum Genet. 2017;101:75–86. doi: 10.1016/j.ajhg.2017.06.001.
    1. Cloonan SM, Glass K, Laucho-Contreras ME, Bhashyam AR, Cervo M, Pabón MA, et al. Mitochondrial iron chelation ameliorates cigarette smoke-induced bronchitis and emphysema in mice. Nat Med. 2016;22:163–174. doi: 10.1038/nm.4021.
    1. Weathington NM, Sznajder JI, Mallampalli RK. The emerging role of the ubiquitin proteasome in pulmonary biology and disease. Am J Respir Crit Care Med. 2013;188:530–537. doi: 10.1164/rccm.201304-0754PP.
    1. Fu J, Wolfs MGM, Deelen P, Westra H-J, Fehrmann RSN, Te Meerman GJ, et al. Unraveling the regulatory mechanisms underlying tissue-dependent genetic variation of gene expression. PLoS Genet. 2012;8:e1002431. doi: 10.1371/journal.pgen.1002431.
    1. Hauberg ME, Zhang W, Giambartolomei C, Franzén O, Morris DL, Vyse TJ, et al. Large-scale identification of common trait and disease variants affecting gene expression. Am J Hum Genet. 2017;100:885–894. doi: 10.1016/j.ajhg.2017.04.016.
    1. Jiang Z, Knudsen NH, Wang G, Qiu W, Naing ZZC, Bai Y, et al. Genetic control of fatty acid β-oxidation in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2017;56:738–748. doi: 10.1165/rcmb.2016-0282OC.
    1. Ligthart S, Vaez A, Hsu Y-H. Inflammation working group of the CHARGE consortium, PMI-WG-XCP, LifeLines cohort study, et al. bivariate genome-wide association study identifies novel pleiotropic loci for lipids and inflammation. BMC Genomics. 2016;17:443. doi: 10.1186/s12864-016-2712-4.
    1. Obeidat M, Nie Y, Fishbane N, Li X, Bossé Y, Joubert P, et al. Integrative Genomics of Emphysema Associated Genes Reveals Potential Disease Biomarkers. Am J Respir Cell Mol Biol. 2017.
    1. Tobacco and Genetics Consortium Genome-wide meta-analyses identify multiple loci associated with smoking behavior. Nat Genet. 2010;42:441–447. doi: 10.1038/ng.571.
    1. Nedeljkovic I, Lahousse L, Carnero-Montoro E, Faiz A, Vonk JM, de Jong K, et al. COPD GWAS variant at 19q13.2 in relation with DNA methylation and gene expression. Hum Mol Genet. 2018;27:396–405. doi: 10.1093/hmg/ddx390.
    1. Epstein AC, Gleadle JM, McNeill LA, Hewitson KS, O’Rourke J, Mole DR, et al. C. Elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell. 2001;107:43–54. doi: 10.1016/S0092-8674(01)00507-4.
    1. Yasuo M, Mizuno S, Kraskauskas D, Bogaard HJ, Natarajan R, Cool CD, et al. Hypoxia inducible factor-1α in human emphysema lung tissue. Eur Respir J. 2011;37:775–783. doi: 10.1183/09031936.00022910.
    1. Kelsen SG. The unfolded protein response in chronic obstructive pulmonary disease. Ann am Thorac Soc. 2016;13(Suppl 2):S138–S145.
    1. Renda T, Baraldo S, Pelaia G, Bazzan E, Turato G, Papi A, et al. Increased activation of p38 MAPK in COPD. Eur Respir J. 2008;31:62–69. doi: 10.1183/09031936.00036707.
    1. Ravanti L, Heino J, López-Otín C, Kähäri VM. Induction of collagenase-3 (MMP-13) expression in human skin fibroblasts by three-dimensional collagen is mediated by p38 mitogen-activated protein kinase. J Biol Chem. 1999;274:2446–2455. doi: 10.1074/jbc.274.4.2446.
    1. Wain LV, Shrine N, Artigas MS, Erzurumluoglu AM, Noyvert B, Bossini-Castillo L, et al. Genome-wide association analyses for lung function and chronic obstructive pulmonary disease identify new loci and potential druggable targets. Nat Genet. 2017;49:416–425. doi: 10.1038/ng.3787.
    1. Gosselink JV, Hayashi S, Elliott WM, Xing L, Chan B, Yang L, et al. Differential expression of tissue repair genes in the pathogenesis of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;181:1329–1335. doi: 10.1164/rccm.200812-1902OC.
    1. Parker MM, Chase RP, Lamb A, Reyes A, Saferali A, Yun JH, et al. RNA sequencing identifies novel non-coding RNA and exon-specific effects associated with cigarette smoking. BMC Med Genet. 2017;10:58.
    1. Morrow JD, Zhou X, Lao T, Jiang Z, DeMeo DL, Cho MH, et al. Functional interactors of three genome-wide association study genes are differentially expressed in severe chronic obstructive pulmonary disease lung tissue. Sci Rep. 2017;7:44232. doi: 10.1038/srep44232.
    1. Mathai SK, Pedersen BS, Smith K, Russell P, Schwarz MI, Brown KK, et al. Desmoplakin variants are associated with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2016;193:1151–1160. doi: 10.1164/rccm.201509-1863OC.
    1. Cheng DT, Kim DK, Cockayne DA, Belousov A, Bitter H, Cho MH, et al. Systemic soluble receptor for advanced glycation endproducts is a biomarker of emphysema and associated with AGER genetic variants in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188:948–957. doi: 10.1164/rccm.201302-0247OC.
    1. Sullivan AK, Simonian PL, Falta MT, Mitchell JD, Cosgrove GP, Brown KK, et al. Oligoclonal CD4+ T cells in the lungs of patients with severe emphysema. Am J Respir Crit Care Med. 2005;172:590–596. doi: 10.1164/rccm.200410-1332OC.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102.
    1. Bhat TA, Panzica L, Kalathil SG, Thanavala Y. Immune dysfunction in patients with chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2015;12(Suppl 2):S169–S175.
    1. Grundy S, Plumb J, Lea S, Kaur M, Ray D, Singh D. Down regulation of T cell receptor expression in COPD pulmonary CD8 cells. PLoS One. 2013;8:e71629. doi: 10.1371/journal.pone.0071629.
    1. Biernacki W, Redpath AT, Best JJ, MacNee W. Measurement of CT lung density in patients with chronic asthma. Eur Respir J. 1997;10:2455–2459. doi: 10.1183/09031936.97.10112455.
    1. Sun W, Kechris K, Jacobson S, Drummond MB, Hawkins GA, Yang J, et al. Common genetic polymorphisms influence blood biomarker measurements in COPD. PLoS Genet. 2016;12:–e1006011.
    1. Chun S, Casparino A, Patsopoulos NA, Croteau-Chonka DC, Raby BA, De Jager PL, et al. Limited statistical evidence for shared genetic effects of eQTLs and autoimmune-disease-associated loci in three major immune-cell types. Nat Genet. 2017;49:600–605. doi: 10.1038/ng.3795.
    1. Petretto E. Single cell expression quantitative trait loci and complex traits. Genome med. 2013;5:72. Available from: 10.1186/gm476
    1. Joehanes R, Zhang X, Huan T, Yao C, Ying S-X, Nguyen QT, et al. Integrated genome-wide analysis of expression quantitative trait loci aids interpretation of genomic association studies. Genome Biol. 2017;18:16. doi: 10.1186/s13059-016-1142-6.

Source: PubMed

3
S'abonner