figshare
Browse
PIIS2213260017303879.pdf (2.72 MB)

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

Download (2.72 MB)
journal contribution
posted on 2017-11-21, 16:18 authored by Richard J Allen, Joanne Porte, Rebecca Braybrooke, Carlos Flores, Tasha E. Fingerlin, Justin M. Oldham, Beatriz Guillen-Guio, Shwu-Fan Ma, Tsukasa Okamoto, Alison E. John, Ma'en Obeidat, Ivana V. Yang, Amanda Henry, Richard B. Hubbard, Vidya Navaratnam, Gauri Saini, Norma Thompson, Helen L. Booth, Simon P. Hart, Mike R. Hill, Nik Hirani, Toby M. Maher, Robin J. McAnulty, Ann B. Millar, Philip L. Molyneaux, Helen Parfrey, Doris M. Rassl, Moira K. B. Whyte, William A. Fahy, Richard P. Marshall, Eunice Oballa, Yohan Bossé, David C. Nickle, Don D. Sin, Wim Timens, Nick Shrine, Ian Sayers, Ian P. Hall, Imre Noth, David A. Schwartz, Martin D. Tobin, Louise V. Wain, R Gisli Jenkins
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

This research has been done using the UK Biobank Resource under application 8389 (led by IS). This Article presents independent research funded partially by the UK National Institute for Health Research (NIHR). The views expressed are our own and not necessarily those of the NHS, the NIHR, or the UK Department of Health. This research used the ALICE High Performance Computing Facility at the University of Leicester. We would like to thank the staff at the Respiratory Health Network Tissue Bank of the Fonds de reserche Sante Quebec for their valuable assistance with the lung eQTL dataset at Laval University. Genotyping of the stage 1 samples was funded by an MRC Strategic Award to IPH, MDT, LVW, and David Strachan (MC_PC_12010). RGJ has an MRC grant (G0901226). IPH has an MRC grant (G1000861). DAS has grants from the National Heart, Lung, and Blood Institute (R01 HL097163 and P01 HL092870). BG-G was supported by a fellowship from ACIISI (TESIS2015010057). TMM is supported by an NIHR Clinician Scientist Fellowship (NIHR reference CS-2013–13–017). LVW holds a GlaxoSmithKline/British Lung Foundation Chair in Respiratory Research. MO is a fellow of the Parker B Francis Foundation.

History

Citation

The Lancet Respiratory Medicine, 2017, 5(11), pp. 869-880

Author affiliation

/Organisation/COLLEGE OF LIFE SCIENCES/School of Medicine/Department of Health Sciences

Version

  • VoR (Version of Record)

Published in

The Lancet Respiratory Medicine

Publisher

Elsevier

issn

2213-2600

eissn

2213-2619

Acceptance date

2017-09-27

Copyright date

2017

Available date

2017-11-21

Publisher version

http://www.sciencedirect.com/science/article/pii/S2213260017303879?via=ihub

Language

en

Usage metrics

    University of Leicester Publications

    Categories

    No categories selected

    Keywords

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC