Ensemble genomic analysis in human lung tissue identifies novel genes for chronic obstructive pulmonary disease

Jarrett D Morrow, Michael H Cho, John Platig, Xiaobo Zhou, Dawn L DeMeo, Weiliang Qiu, Bartholome Celli, Nathaniel Marchetti, Gerard J Criner, Raphael Bueno, George R Washko, Kimberly Glass, John Quackenbush, Edwin K Silverman, Craig P Hersh, Jarrett D Morrow, Michael H Cho, John Platig, Xiaobo Zhou, Dawn L DeMeo, Weiliang Qiu, Bartholome Celli, Nathaniel Marchetti, Gerard J Criner, Raphael Bueno, George R Washko, Kimberly Glass, John Quackenbush, Edwin K Silverman, Craig P Hersh

Abstract

Background: Genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) significantly associated with chronic obstructive pulmonary disease (COPD). However, many genetic variants show suggestive evidence for association but do not meet the strict threshold for genome-wide significance. Integrative analysis of multiple omics datasets has the potential to identify novel genes involved in disease pathogenesis by leveraging these variants in a functional, regulatory context.

Results: We performed expression quantitative trait locus (eQTL) analysis using genome-wide SNP genotyping and gene expression profiling of lung tissue samples from 86 COPD cases and 31 controls, testing for SNPs associated with gene expression levels. These results were integrated with a prior COPD GWAS using an ensemble statistical and network methods approach to identify relevant genes and observe them in the context of overall genetic control of gene expression to highlight co-regulated genes and disease pathways. We identified 250,312 unique SNPs and 4997 genes in the cis(local)-eQTL analysis (5% false discovery rate). The top gene from the integrative analysis was MAPT, a gene recently identified in an independent GWAS of lung function. The genes HNRNPAB and PCBP2 with RNA binding activity and the gene ACVR1B were identified in network communities with validated disease relevance.

Conclusions: The integration of lung tissue gene expression with genome-wide SNP genotyping and subsequent intersection with prior GWAS and omics studies highlighted candidate genes within COPD loci and in communities harboring known COPD genes. This integration also identified novel disease genes in sub-threshold regions that would otherwise have been missed through GWAS.

Trial registration: ClinicalTrials.gov NCT00608764 NCT00292552.

Keywords: Bayesian methods; Ensemble methods; Expression QTL; Integrative genomics; Network medicine; eQTL.

Conflict of interest statement

Ethics approval and consent to participate

Subjects provided written informed consent for the use of excess lung tissue for research. IRB approval was obtained at Partners Healthcare (parent company of Brigham and Women’s Hospital), Temple University, and St. Elizabeth’s Hospital. The methods for lung tissue research were carried out in accordance with the relevant guidelines.

Competing interests

Drs. Morrow, Platig, Zhou, Qiu, Marchetti, Criner, Celli, Glass, Quackenbush report no competing interests related to this manuscript.

Dr. Cho has received compensation from GSK.

Dr. DeMeo has received compensation from Novartis.

Dr. Bueno has received compensation from Myriad Genetics, Inc., Siemens, Verastem, Inc., Genentech, Inc., Gritstone Oncology, Inc., HTG Molecular Diagnostics, Inc., Neil Leifer, Esq, Morrison Mahoney, David Weiss, LLC, Balick & Balick, LLC, Hartford Hospital, Cleveland Clinic/Conference, Aspen Lung Conference, Case Western Reserve, North Shore University, Castle Bioscience, Novartis Institutes for Biomedical Research, AstraZebeca, imCORE/Roche, Arthur Tuverson, LLC, Ferraro Law Firm, Rice Dolan & Kershaw, Satterly & Kelly, LLC, Exosome, Inc.

Dr. Washko has received compensation from Boehringer Ingelheim, GlaxoSmithKline, Janssen Pharmaceuticals, BTG Interventional Medicine, Regeneron, Quantitative Imaging Solutions and ModoSpira, and his Spouse works for Biogen.

Dr. Silverman has received compensation from COPD Foundation, GlaxoSmithKline, Merck and Novartis,

Dr. Hersh has received consulting fees from AstraZeneca, Concert Pharmaceuticals, Mylan, and grant support from Boehrinher Ingelheim.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Graphical overview of the study methods and process. The cis- and trans-eQTLs identified in lung tissue were integrated with prior GWAS using Bayesian and network methods. The network communities identified were interrogated for evidence of differential gene expression and differential DNA methylation by COPD status
Fig. 2
Fig. 2
Plot of COPD GWAS p values vs. the cis-eQTL p values. Each point in the plot represents a cis-eQTL result with an rsID found in the prior GWAS. GWAS p values (y axis) are plotted against the expression QTL p values (x axis). A vertical dotted line indicates the threshold of significance (FDR < 5%) for the eQTL. Horizontal lines delineate genome-wide significant (red) and sub-threshold (blue) GWAS p values. The significant (red; FDR < 5%) and nominally significant (blue; p < 0.05) eGenes from gene expression profiling in COPD lung tissue are highlighted
Fig. 3
Fig. 3
Community 202 from CONDOR analysis that contains the Sherlock-derived genes CHRNA5, HNRNPAB, IREB2, and PCBP2. Community genes are listed in (Additional file 1: Table S6). (Red = SNP, yellow = SNP with GWAS p < 10−4, square = Sherlock gene, gray = gene, green = gene with differentially methylated site (p < 0.05 and effect > 5%), light blue = gene with differentially expressed probe (p < 0.05), and cyan = gene with differentially methylated site and differentially expressed probe)
Fig. 4
Fig. 4
Community 222 from CONDOR analysis that contains the Sherlock-derived gene ACVR1B. Community genes are listed in (Additional file 1: Table S6). (Red = SNP, yellow = SNP with GWAS p < 10−4, square = Sherlock gene, gray = gene, green = gene with differentially methylated site (p < 0.05 and effect > 5%), light blue = gene with differentially expressed probe (p < 0.05), and cyan = gene with differentially methylated site and differentially expressed probe)
Fig. 5
Fig. 5
Community 113 from CONDOR analysis that contains the Sherlock-derived gene CDH23. The central genes PSMC1 and CTDSPL2 partially overlap and are obstructed in the figure. Community genes are listed in (Additional file 1: Table S6). (Red = SNP, yellow = SNP with GWAS p < 10−4, square = Sherlock gene, gray = gene, green = gene with differentially methylated site (p < 0.05 and effect > 5%), light blue = gene with differentially expressed probe (p < 0.05), cyan = gene with differentially methylated site and differentially expressed probe)

References

    1. Vestbo J, Hurd SS, Agustí AG, Jones PW, Vogelmeier C, Anzueto A, 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;187:347–365. doi: 10.1164/rccm.201204-0596PP.
    1. Hersh CP, Hokanson JE, Lynch DA, Washko GR, Make BJ, Crapo JD, et al. Family history is a risk factor for copd. Chest. 2011;140:343–350. doi: 10.1378/chest.10-2761.
    1. McCloskey S, Patel B, Hinchliffe S, Reid E, Wareham N, Lomas D. Siblings of patients with severe chronic obstructive pulmonary disease have a significant risk of airflow obstruction. Am J Respir Crit Care Med. 2001;164:1419–1424. doi: 10.1164/ajrccm.164.8.2105002.
    1. Silverman EK. Genetics of chronic obstructive pulmonary disease. In: Organizer DC, Goode JA, editors. Chronic Obstr. Pulm. Dis. Pathog. Treat. John Wiley & Sons, Ltd; 2000. p. 45–64.
    1. Silverman EK, Chapman HA, Drazen JM, Weiss ST, Rosner B, Campbell EJ, et al. Genetic epidemiology of severe, early-onset chronic obstructive pulmonary disease. Risk to relatives for airflow obstruction and chronic bronchitis. Am J Respir Crit Care Med. 1998;157:1770–1778. doi: 10.1164/ajrccm.157.6.9706014.
    1. Cho MH, Boutaoui N, Klanderman BJ, Sylvia JS, Ziniti JP, Hersh CP, et al. Variants in FAM13A are associated with chronic obstructive pulmonary disease. Nat Genet. 2010;42:200–202. doi: 10.1038/ng.535.
    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. Cho MH, McDonald M-LN, 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, Bossé 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. Bjornsson H. An integrated epigenetic and genetic approach to common human disease. Trends Genet. 2004;20:350–358. doi: 10.1016/j.tig.2004.06.009.
    1. Feinberg AP. Epigenomics reveals a functional genome anatomy and a new approach to common disease. Nat Biotechnol. 2010;28:1049–1052. doi: 10.1038/nbt1010-1049.
    1. Wang X, Tucker NR, Rizki G, Mills R, Krijger PH, de Wit E, et al. Discovery and validation of sub-threshold genome-wide association study loci using epigenomic signatures. elife. 2016;5:e10557.
    1. Bush WS, Moore JH. Chapter 11: genome-wide association studies. Lewitter F, Kann M, editors. PLoS Comput Biol. 2012;8:e1002822.
    1. Civelek M, Lusis AJ. Systems genetics approaches to understand complex traits. Nat Rev Genet. 2013;15:34–48. doi: 10.1038/nrg3575.
    1. Li L, Kabesch M, Bouzigon E, Demenais F, Farrall M, Moffatt MF, et al. Using eQTL weights to improve power for genome-wide association studies: a genetic study of childhood asthma. Front Genet. 2013;4:103. doi:.
    1. Nica AC, Montgomery SB, Dimas AS, Stranger BE, Beazley C, Barroso I, et al. Candidate causal regulatory effects by integration of expression QTLs with complex trait genetic associations. Gibson G, editor. PLoS Genet. 2010;6:e1000895.
    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. 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. He X, Fuller CK, Song Y, Meng Q, Zhang B, Yang X, et al. Sherlock: detecting gene-disease associations by matching patterns of expression QTL and GWAS. Am J Hum Genet. 2013;92:667–680. doi: 10.1016/j.ajhg.2013.03.022.
    1. Platig J, Castaldi PJ, DeMeo D, Quackenbush J. Bipartite community structure of eQTLs. Markowetz F, editor. PLoS Comput Biol. 2016;12:e1005033.
    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. Kamburov A, Stelzl U, Lehrach H, Herwig R. The ConsensusPathDB interaction database: 2013 update. Nucleic Acids Res. 2013;41:D793–D800. doi: 10.1093/nar/gks1055.
    1. DeMeo DL, Mariani T, Bhattacharya S, Srisuma S, Lange C, Litonjua A, et al. Integration of genomic and genetic approaches implicates IREB2 as a COPD susceptibility gene. Am J Hum Genet. 2009;85:493–502. doi: 10.1016/j.ajhg.2009.09.004.
    1. Hardin M, Zielinski J, Wan ES, Hersh CP, Castaldi PJ, Schwinder E, et al. CHRNA3/5, IREB2, and ADCY2 are associated with severe chronic obstructive pulmonary disease in Poland. Am J Respir Cell Mol Biol. 2012;47:203–208. doi: 10.1165/rcmb.2012-0011OC.
    1. Pillai SG, Ge D, Zhu G, Kong X, Shianna KV, Need AC, et al. A genome-wide association study in chronic obstructive pulmonary disease (COPD): identification of two major susceptibility loci. PLoS Genet. 2009;5:e1000421. doi: 10.1371/journal.pgen.1000421.
    1. Navratilova Z, Krsjakova T, Novosadova E, Zatloukal J, Kolek V, Petrek M. Genes affecting mRNA stability and metalloproteinase inhibitor RECK are down-regulated in patients with COPD. Eur Respir J. 2013;42(Suppl 57):3512.
    1. Hou P, Li L, Chen F, Chen Y, Liu H, Li J, et al. PTBP3-mediated regulation of ZEB1 mRNA stability promotes epithelial-mesenchymal transition in breast cancer. Cancer Res. 2017;canres.0883.2017.
    1. Zhou Z-J, Dai Z, Zhou S-L, Hu Z-Q, Chen Q, Zhao Y-M, et al. HNRNPAB induces epithelial-mesenchymal transition and promotes metastasis of hepatocellular carcinoma by transcriptionally activating SNAIL. Cancer Res. 2014;74:2750–2762. doi: 10.1158/0008-5472.CAN-13-2509.
    1. Nowrin K, Sohal SS, Peterson G, Patel R, Walters EH. Epithelial-mesenchymal transition as a fundamental underlying pathogenic process in COPD airways: fibrosis, remodeling and cancer. Expert Rev Respir Med. 2014;8:547–559. doi: 10.1586/17476348.2014.948853.
    1. Sohal SS, Mahmood MQ, Walters EH. Clinical significance of epithelial mesenchymal transition (EMT) in chronic obstructive pulmonary disease (COPD): potential target for prevention of airway fibrosis and lung cancer. Clin Transl Med. 2014;3:33. doi: 10.1186/s40169-014-0033-2.
    1. Castaldi PJ, Cho MH, Zhou X, Qiu W, Mcgeachie M, Celli B, et al. Genetic control of gene expression at novel and established chronic obstructive pulmonary disease loci. Hum Mol Genet. 2015;24:1200–1210. doi: 10.1093/hmg/ddu525.
    1. Lutz SM, Cho MH, Young K, Hersh CP, Castaldi PJ, McDonald M-L, et al. A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry. BMC Genet. 2015;16(1):138. doi: 10.1186/s12863-015-0299-4.
    1. Ezzie ME, Crawford M, Cho J-H, Orellana R, Zhang S, Gelinas R, et al. Gene expression networks in COPD: microRNA and mRNA regulation. Thorax. 2012;67:122–131. doi: 10.1136/thoraxjnl-2011-200089.
    1. Gheldof A, Berx G. Cadherins and epithelial-to-mesenchymal transition. Prog Mol Biol Transl Sci. 2013;116:317–36. doi: 10.1016/B978-0-12-394311-8.00014-5.
    1. Imboden M, Bouzigon E, Curjuric I, Ramasamy A, Kumar A, Hancock DB, et al. Genome-wide association study of lung function decline in adults with and without asthma. J Allergy Clin Immunol. 2012;129:1218–1228. doi: 10.1016/j.jaci.2012.01.074.
    1. Yucesoy B, Kaufman KM, Lummus ZL, Weirauch MT, Zhang G, Cartier A, et al. Genome-wide association study identifies novel loci associated with diisocyanate-induced occupational asthma. Toxicol Sci. 2015;146:192–201. doi: 10.1093/toxsci/kfv084.
    1. Siedlinski M, Cho MH, Bakke P, Gulsvik A, Lomas DA, Anderson W, et al. Genome-wide association study of smoking behaviours in patients with COPD. Thorax. 2011;66:894–902. doi: 10.1136/thoraxjnl-2011-200154.
    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. Wain LV, Shrine N, Miller S, Jackson VE, Ntalla I, Artigas MS, 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. 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. Hobbs BD, Parker MM, Chen H, Lao T, Hardin M, Qiao D, et al. Exome array analysis identifies a common variant in IL27 associated with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2016;194:48–57. doi: 10.1164/rccm.201510-2053OC.
    1. Morrow JD, Cho MH, Hersh CP, Pinto-Plata V, Celli B, Marchetti N, et al. DNA methylation profiling in human lung tissue identifies genes associated with COPD. Epigenetics. 2016;11:730–739. doi: 10.1080/15592294.2016.1226451.
    1. Chen Y-C, Statt S, Wu R, Chang H-T, Liao J-W, Wang C-N, et al. High mobility group box 1-induced epithelial mesenchymal transition in human airway epithelial cells. Sci Rep. 2016;6:18815. doi:.
    1. Delaneau O, Marchini J, Zagury J-F. A linear complexity phasing method for thousands of genomes. Nat Methods. 2011;9:179–181. doi: 10.1038/nmeth.1785.
    1. Howie B, Marchini J, Stephens M. Genotype imputation with thousands of genomes. Chakravarti a, editor. G3amp58 GenesGenomesGenetics. 2011;1:457–470.
    1. Howie BN, Donnelly P, Marchini J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. Schork NJ, editor. PLoS Genet. 2009;5:e1000529.
    1. Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich D. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006;38:904–909. doi: 10.1038/ng1847.
    1. Shabalin AA. Matrix eQTL: ultra fast eQTL analysis via large matrix operations. Bioinformatics. 2012;28:1353–1358. doi: 10.1093/bioinformatics/bts163.
    1. Auton A, Abecasis GR, Altshuler DM, Durbin RM, Abecasis GR, Bentley DR, et al. A global reference for human genetic variation. Nature. 2015;526:68–74. doi: 10.1038/nature15393.
    1. Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ. Second-generation PLINK: rising to the challenge of larger and richer datasets. GigaScience. 2015;4:7. doi: 10.1186/s13742-015-0047-8.
    1. Aguet F, Ardlie KG, Cummings BB, Gelfand ET, Getz G, Hadley K, et al. Genetic effects on gene expression across human tissues. Nature. 2017;550:204–213. doi: 10.1038/nature24277.
    1. Csardi G, Nepusz T. The igraph software package for complex network research. InterJournal 2006;Complex Systems:1695.
    1. Phanstiel DH, Boyle AP, Araya CL, Snyder MP. Sushi.R: flexible, quantitative and integrative genomic visualizations for publication-quality multi-panel figures. Bioinformatics. 2014;30:2808–2810. doi: 10.1093/bioinformatics/btu379.
    1. Durinck S, Moreau Y, Kasprzyk A, Davis S, De Moor B, Brazma A, et al. BioMart and bioconductor: a powerful link between biological databases and microarray data analysis. Bioinformatics. 2005;21:3439–3440. doi: 10.1093/bioinformatics/bti525.
    1. Zerbino DR, Wilder SP, Johnson N, Juettemann T, Flicek PR. The Ensembl Regulatory Build. Genome Biol. 2015;16:56. doi: 10.1186/s13059-015-0621-5.
    1. Rosenbloom KR, Sloan CA, Malladi VS, Dreszer TR, Learned K, Kirkup VM, et al. ENCODE Data in the UCSC Genome Browser: year 5 update. Nucleic Acids Res. 2013;41:D56–D63. doi: 10.1093/nar/gks1172.

Source: PubMed

3
Suscribir