University of Leicester
Browse
1-s2.0-S221326002200251X-main.pdf (1.86 MB)

Longitudinal lung function and gas transfer in individuals with idiopathic pulmonary fibrosis: a genome-wide association study

Download (1.86 MB)
journal contribution
posted on 2022-08-31, 09:26 authored by Richard Allen, Justin Oldham, David Jenkins, Olivia Leavy, Beatriz Guillen-Guio, Carl Melbourne, Shwu-Fan Ma, Jonathan Jou, John Kim, William Fahy, Eunice Oballa, Richard Hubbard, Vidya Navaratnam, Rebecca Braybrooke, Gauri Saini, Katy Roach, Martin Tobin, Nik Hirani, Moira Whyte, Naftali Kaminski, Yingzee Zhang, Fernando Martinez, Angela Linderholm, Ayodeji Adegunsoye, Mary Strek, Toby Maher, Philip Molyneaux, Carlos Flores, Imre Noth, Gisli Jenkins, Louise Wain

Background

Idiopathic pulmonary fibrosis (IPF) is an incurable lung disease characterised by progressive scarring leading to alveolar stiffness, reduced lung capacity, and impeded gas transfer. We aimed to identify genetic variants associated with declining lung capacity or declining gas transfer after diagnosis of IPF.


Methods

We did a genome-wide meta-analysis of longitudinal measures of forced vital capacity (FVC) and diffusing capacity of the lung for carbon monoxide (DLCO) in individuals diagnosed with IPF. Individuals were recruited to three studies between June, 1996, and August, 2017, from across centres in the US, UK, and Spain. Suggestively significant variants were investigated further in an additional independent study (CleanUP-IPF). All four studies diagnosed cases following American Thoracic Society/European Respiratory Society guidelines. Variants were defined as significantly associated if they had a meta-analysis p<5 × 10−8 when meta-analysing across all discovery and follow-up studies, had consistent direction of effects across all four studies, and were nominally significant (p<0·05) in each study.


Findings

1329 individuals with a total of 5216 measures were included in the FVC analysis. 975 individuals with a total of 3361 measures were included in the DLCO analysis. For the discovery genome-wide analyses, 7 611 174 genetic variants were included in the FVC analysis and 7 536 843 in the DLCO analysis. One variant (rs115982800) located in an antisense RNA gene for protein kinase N2 (PKN2) showed a genome-wide significant association with FVC decline (−140 mL/year per risk allele [95% CI –180 to –100]; p=9·14 × 10−12).


Interpretation

Our analysis identifies a genetic variant associated with disease progression, which might highlight a new biological mechanism for IPF. We found that PKN2, a Rho and Rac effector protein, is the most likely gene of interest from this analysis. PKN2 inhibitors are currently in development and signify a potential novel therapeutic approach for IPF.

Funding

Action for Pulmonary Fibrosis, Medical Research Council, Wellcome Trust, and National Institutes of Health National Heart, Lung, and Blood Institute.

History

Author affiliation

Department of Health Sciences, University of Leicester

Version

  • VoR (Version of Record)

Published in

The Lancet Respiratory Medicine

Publisher

Elsevier

issn

2213-2600

Copyright date

2022

Available date

2022-08-31

Language

en

Usage metrics

    University of Leicester Publications

    Categories

    No categories selected

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC