Genetic variants associated with susceptibility to idiopathic pulmonary fibrosis in people of European ancestry: a genome-wide association study

Summary Background Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with high mortality, uncertain cause, and few treatment options. Studies have identified a significant genetic risk associated with the development of IPF; however, mechanisms by which genetic risk factors promote IPF remain unclear. We aimed to identify genetic variants associated with IPF susceptibility and provide mechanistic insight using gene and protein expression analyses. Methods We used a two-stage approach: a genome-wide association study in patients with IPF of European ancestry recruited from nine different centres in the UK and controls selected from UK Biobank (stage 1) matched for age, sex, and smoking status; and a follow-up of associated genetic variants in independent datasets of patients with IPF and controls from two independent US samples from the Chicago consortium and the Colorado consortium (stage 2). We investigated the effect of novel signals on gene expression in large transcriptomic and genomic data resources, and examined expression using lung tissue samples from patients with IPF and controls. Findings 602 patients with IPF and 3366 controls were selected for stage 1. For stage 2, 2158 patients with IPF and 5195 controls were selected. We identified a novel genome-wide significant signal of association with IPF susceptibility near A-kinase anchoring protein 13 (AKAP13; rs62025270, odds ratio [OR] 1·27 [95% CI 1·18–1·37], p=1·32 × 10−9) and confirmed previously reported signals, including in mucin 5B (MUC5B; rs35705950, OR 2·89 [2·56–3·26], p=1·12 × 10−66) and desmoplakin (DSP; rs2076295, OR 1·44 [1·35–1·54], p=7·81 × 10−28). For rs62025270, the allele A associated with increased susceptibility to IPF was also associated with increased expression of AKAP13 mRNA in lung tissue from patients who had lung resection procedures (n=1111). We showed that AKAP13 is expressed in the alveolar epithelium and lymphoid follicles from patients with IPF, and AKAP13 mRNA expression was 1·42-times higher in lung tissue from patients with IPF (n=46) than that in lung tissue from controls (n=51). Interpretation AKAP13 is a Rho guanine nucleotide exchange factor regulating activation of RhoA, which is known to be involved in profibrotic signalling pathways. The identification of AKAP13 as a susceptibility gene for IPF increases the prospect of successfully targeting RhoA pathway inhibitors in patients with IPF. Funding UK Medical Research Council, National Heart, Lung, and Blood Institute of the US National Institutes of Health, Agencia Canaria de Investigación, Innovación y Sociedad de la Información, Spain, UK National Institute for Health Research, and the British Lung Foundation.

[1]  A. Azuma,et al.  A Phase 3 Trial of Pirfenidone in Patients with Idiopathic Pulmonary Fibrosis , 2015 .

[2]  John P. Overington,et al.  ChEMBL: a large-scale bioactivity database for drug discovery , 2011, Nucleic Acids Res..

[3]  Paul J. Friedman,et al.  American Thoracic Society/European Respiratory Society International Multidisciplinary Consensus Classification of the Idiopathic Interstitial Pneumonias. This joint statement of the American Thoracic Society (ATS), and the European Respiratory Society (ERS) was adopted by the ATS board of directors , 2002, American journal of respiratory and critical care medicine.

[4]  Naoto Hoshi,et al.  Bioinformatic design of A-kinase anchoring protein-in silico: A potent and selective peptide antagonist of type II protein kinase A anchoring , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[5]  D. Lamb,et al.  THE IMMUNOLOGICAL ARCHITECTURE OF B‐LYMPHOCYTE AGGREGATES IN CRYPTOGENIC FIBROSING ALVEOLITIS , 1996, The Journal of pathology.

[6]  Zhi-Xin Wang,et al.  Characterization of the interactions between the small GTPase RhoA and its guanine nucleotide exchange factors. , 2002, Analytical biochemistry.

[7]  Tom R. Gaunt,et al.  The UK10K project identifies rare variants in health and disease , 2015, Nature.

[8]  M. Burdick,et al.  Cultured lung fibroblasts isolated from patients with idiopathic pulmonary fibrosis have a diminished capacity to synthesize prostaglandin E2 and to express cyclooxygenase-2. , 1995, The Journal of clinical investigation.

[9]  Mark I. McCarthy,et al.  Evaluating the Performance of Fine-Mapping Strategies at Common Variant GWAS Loci , 2015, PLoS genetics.

[10]  M. Peters,et al.  Systematic identification of trans eQTLs as putative drivers of known disease associations , 2013, Nature Genetics.

[11]  Jun S. Liu,et al.  The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue gene regulation in humans , 2015, Science.

[12]  Jon Wakefield,et al.  A Bayesian measure of the probability of false discovery in genetic epidemiology studies. , 2007, American journal of human genetics.

[13]  P. Weinreb,et al.  Lysophosphatidic acid induces alphavbeta6 integrin-mediated TGF-beta activation via the LPA2 receptor and the small G protein G alpha(q). , 2009, The American journal of pathology.

[14]  L. Wain,et al.  GSTCD and INTS12 Regulation and Expression in the Human Lung , 2013, PloS one.

[15]  Michael Boehnke,et al.  Recommended Joint and Meta‐Analysis Strategies for Case‐Control Association Testing of Single Low‐Count Variants , 2013, Genetic epidemiology.

[16]  S. Traynelis,et al.  Pharmacology of dextromethorphan: Relevance to dextromethorphan/quinidine (Nuedexta®) clinical use. , 2016, Pharmacology & therapeutics.

[17]  Naftali Kaminski,et al.  Genetic variants associated with idiopathic pulmonary fibrosis susceptibility and mortality: a genome-wide association study. , 2013, The Lancet. Respiratory medicine.

[18]  Brent S. Pedersen,et al.  Genome-wide association study identifies multiple susceptibility loci for pulmonary fibrosis , 2013, Nature Genetics.

[19]  G. Laurent,et al.  Cyclooxygenase-2 deficiency results in a loss of the anti-proliferative response to transforming growth factor-beta in human fibrotic lung fibroblasts and promotes bleomycin-induced pulmonary fibrosis in mice. , 2001, The American journal of pathology.

[20]  Don D. Sin,et al.  Refining Susceptibility Loci of Chronic Obstructive Pulmonary Disease with Lung eqtls , 2013, PloS one.

[21]  J. Leader,et al.  C-X-C motif chemokine 13 (CXCL13) is a prognostic biomarker of idiopathic pulmonary fibrosis. , 2014, American journal of respiratory and critical care medicine.

[22]  P. Elliott,et al.  UK Biobank: An Open Access Resource for Identifying the Causes of a Wide Range of Complex Diseases of Middle and Old Age , 2015, PLoS medicine.

[23]  Ellen T. Gelfand,et al.  The Genotype-Tissue Expression (GTEx) project , 2013, Nature Genetics.

[24]  Tomoshige Kino,et al.  Rho Family Guanine Nucleotide Exchange Factor Brx Couples Extracellular Signals to the Glucocorticoid Signaling System* , 2006, Journal of Biological Chemistry.

[25]  O. Delaneau,et al.  A linear complexity phasing method for thousands of genomes , 2011, Nature Methods.

[26]  Brent S. Pedersen,et al.  Genome-wide imputation study identifies novel HLA locus for pulmonary fibrosis and potential role for auto-immunity in fibrotic idiopathic interstitial pneumonia , 2016, BMC Genetics.

[27]  Zora Modrusan,et al.  Heterogeneous gene expression signatures correspond to distinct lung pathologies and biomarkers of disease severity in idiopathic pulmonary fibrosis , 2014, Thorax.

[28]  David C. Nickle,et al.  Lung eQTLs to Help Reveal the Molecular Underpinnings of Asthma , 2012, PLoS genetics.

[29]  Gabor T. Marth,et al.  A global reference for human genetic variation , 2015, Nature.

[30]  Davide Heller,et al.  STRING v10: protein–protein interaction networks, integrated over the tree of life , 2014, Nucleic Acids Res..

[31]  L. Wain,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 , 2015, The Lancet. Respiratory medicine.

[32]  W. Coward,et al.  The pathogenesis of idiopathic pulmonary fibrosis , 2010, Therapeutic advances in respiratory disease.

[33]  A. Hall,et al.  Activated Gαq family members induce Rho GTPase activation and Rho‐dependent actin filament assembly , 2002, FEBS letters.

[34]  R. Hubbard,et al.  The rising incidence of idiopathic pulmonary fibrosis in the UK , 2011, Thorax.

[35]  Alan M. Kwong,et al.  A reference panel of 64,976 haplotypes for genotype imputation , 2015, Nature Genetics.

[36]  Jonathan Haines,et al.  Heterozygosity for a surfactant protein C gene mutation associated with usual interstitial pneumonitis and cellular nonspecific interstitial pneumonitis in one kindred. , 2002, American journal of respiratory and critical care medicine.

[37]  Cathy H. Wu,et al.  The Universal Protein Resource (UniProt) , 2004, Nucleic Acids Res..

[38]  F. Sciurba,et al.  Plasma B Lymphocyte Stimulator and B Cell Differentiation in Idiopathic Pulmonary Fibrosis Patients , 2013, The Journal of Immunology.

[39]  Rachel C Chambers,et al.  Exploration of a potent PI3 kinase/mTOR inhibitor as a novel anti-fibrotic agent in IPF , 2016, Thorax.

[40]  W. Coward,et al.  A central role for G9a and EZH2 in the epigenetic silencing of cyclooxygenase-2 in idiopathic pulmonary fibrosis , 2014, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[41]  G. Laurent,et al.  Epigenetic regulation of cyclooxygenase-2 by methylation of c8orf4 in pulmonary fibrosis , 2016, Clinical science.

[42]  M. Giel-Moloney,et al.  The Lbc Rho Guanine Nucleotide Exchange Factor/α-Catulin Axis Functions in Serotonin-induced Vascular Smooth Muscle Cell Mitogenesis and RhoA/ROCK Activation* , 2010, The Journal of Biological Chemistry.

[43]  I. Noth,et al.  Association between the MUC5B promoter polymorphism and survival in patients with idiopathic pulmonary fibrosis. , 2013, JAMA.

[44]  Joyce S Lee,et al.  Effect of telomere length on survival in patients with idiopathic pulmonary fibrosis: an observational cohort study with independent validation. , 2014, The Lancet. Respiratory medicine.

[45]  S. Kudoh,et al.  A genome-wide association study identifies an association of a common variant in TERT with susceptibility to idiopathic pulmonary fibrosis , 2008, Journal of Medical Genetics.

[46]  David S. Wishart,et al.  DrugBank: a comprehensive resource for in silico drug discovery and exploration , 2005, Nucleic Acids Res..

[47]  G. Abecasis,et al.  Optimal designs for two‐stage genome‐wide association studies , 2007, Genetic epidemiology.

[48]  S. Knapp,et al.  The crystal structure of the RhoA–AKAP-Lbc DH–PH domain complex , 2014, The Biochemical journal.

[49]  J. Scott,et al.  Type II regulatory subunit (RII) of the cAMP-dependent protein kinase interaction with A-kinase anchor proteins requires isoleucines 3 and 5. , 1994, The Journal of biological chemistry.

[50]  Mulin Jun Li,et al.  Nature Genetics Advance Online Publication a N a Ly S I S the Support of Human Genetic Evidence for Approved Drug Indications , 2022 .

[51]  P. D. de Jong,et al.  Surfactant protein C mutations are the basis of a significant portion of adult familial pulmonary fibrosis in a dutch cohort. , 2010, American journal of respiratory and critical care medicine.

[52]  L. Quarles,et al.  Calcium-sensing receptor activation of rho involves filamin and rho-guanine nucleotide exchange factor. , 2002, Endocrinology.

[53]  S. Mane,et al.  Exome Sequencing Links Mutations in PARN and RTEL1 with Familial Pulmonary Fibrosis and Telomere Shortening , 2015, Nature Genetics.

[54]  J. Scott,et al.  Akap-lbc Anchors Protein Kinase a and Nucleates G␣ 12 -selective Rho-mediated Stress Fiber Formation* , 2022 .

[55]  D. Diviani,et al.  A-Kinase-Anchoring Protein–Lbc Anchors IκB Kinase β To Support Interleukin-6-Mediated Cardiomyocyte Hypertrophy , 2012, Molecular and Cellular Biology.

[56]  D. Schwartz,et al.  Pulmonary fibrosis in the era of stratified medicine , 2016, Thorax.

[57]  B. Crestani,et al.  Dendritic cells accumulate in human fibrotic interstitial lung disease. , 2007, American journal of respiratory and critical care medicine.

[58]  C. Buckmaster,et al.  A Rho Exchange Factor Mediates Thrombin and Gα12-induced Cytoskeletal Responses* , 1999, The Journal of Biological Chemistry.

[59]  D. Diviani,et al.  A-kinase anchoring protein-Lbc promotes pro-fibrotic signaling in cardiac fibroblasts. , 2014, Biochimica et biophysica acta.

[60]  Andrew D. Johnson,et al.  SNAP: a web-based tool for identification and annotation of proxy SNPs using HapMap , 2008, Bioinform..

[61]  H. Collard,et al.  Clinical course and prediction of survival in idiopathic pulmonary fibrosis. , 2011, American journal of respiratory and critical care medicine.

[62]  Yan A. Su,et al.  Brx Mediates the Response of Lymphocytes to Osmotic Stress Through the Activation of NFAT5 , 2009, Science Signaling.

[63]  V. Soumelis,et al.  Cutting Edge: Nonproliferating Mature Immune Cells Form a Novel Type of Organized Lymphoid Structure in Idiopathic Pulmonary Fibrosis1 , 2006, The Journal of Immunology.

[64]  J. Marchini,et al.  Fast and accurate genotype imputation in genome-wide association studies through pre-phasing , 2012, Nature Genetics.

[65]  Takeshi Johkoh,et al.  American Thoracic Society Documents An Official ATS / ERS / JRS / ALAT Statement : Idiopathic Pulmonary Fibrosis : Evidence-based Guidelines for Diagnosis and Management , 2011 .

[66]  Efficacy and Safety of Nintedanib in Idiopathic Pulmonary Fibrosis. , 2015, The New England journal of medicine.

[67]  M. Venere,et al.  The Rho Guanine Nucleotide Exchange Factor AKAP13 (BRX) Is Essential for Cardiac Development in Mice* , 2010, The Journal of Biological Chemistry.

[68]  P. Donnelly,et al.  A new multipoint method for genome-wide association studies by imputation of genotypes , 2007, Nature Genetics.

[69]  R. Chambers,et al.  Ligation of protease-activated receptor 1 enhances alpha(v)beta6 integrin-dependent TGF-beta activation and promotes acute lung injury. , 2006, The Journal of clinical investigation.

[70]  P. Donnelly,et al.  A Flexible and Accurate Genotype Imputation Method for the Next Generation of Genome-Wide Association Studies , 2009, PLoS genetics.

[71]  R. D. du Bois,et al.  Immunohistological analysis of lung tissue from patients with cryptogenic fibrosing alveolitis suggesting local expression of immune hypersensitivity. , 1985, Thorax.

[72]  Shandra L. Protzko,et al.  An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline: Treatment of Idiopathic Pulmonary Fibrosis. An Update of the 2011 Clinical Practice Guideline. , 2015, American journal of respiratory and critical care medicine.

[73]  I. Fraser,et al.  Association of the type II cAMP-dependent protein kinase with a human thyroid RII-anchoring protein. Cloning and characterization of the RII-binding domain. , 1992, The Journal of biological chemistry.

[74]  J. Liao,et al.  The Rho Kinases: Critical Mediators of Multiple Profibrotic Processes and Rational Targets for New Therapies for Pulmonary Fibrosis , 2015, Pharmacological Reviews.

[75]  T. Maher PROFILEing idiopathic pulmonary fibrosis: rethinking biomarker discovery , 2013, European Respiratory Review.

[76]  Ivana V. Yang,et al.  Expression of cilium-associated genes defines novel molecular subtypes of idiopathic pulmonary fibrosis , 2013, Thorax.

[77]  Harry J de Koning,et al.  Genetic loci associated with chronic obstructive pulmonary disease overlap with loci for lung function and pulmonary fibrosis , 2017, Nature Genetics.

[78]  Wj Gauderman,et al.  QUANTO 1.1: A computer program for power and sample size calculations for genetic-epidemiology studies , 2006 .