4.7 Article

Exome Sequencing Analysis in Severe, Early-Onset Chronic Obstructive Pulmonary Disease

Journal

Publisher

AMER THORACIC SOC
DOI: 10.1164/rccm.201506-1223OC

Keywords

genetic association studies; segregation analysis; chronic obstructive pulmonary disease

Funding

  1. NHLBI [R01 HL084323, P01 HL083069, P01 HL105339, R01 HL075478, R01 HL089856, K08 HL097029, R01 HL113264, K01 HL129039, R01 HL089897, RC2 HL-103010, RC2 HL-102923, RC2 HL-102924, RC2 HL-102925, RC2 HL-102926, 1 U54HG006493]
  2. Alpha-1 Foundation
  3. National Human Genome Research Institute
  4. NHLBI Exome Sequencing Program [RC2HL102923, RC2HL102926]
  5. COPD Foundation
  6. AstraZeneca
  7. Boehringer Ingelheim
  8. Novartis
  9. Pfizer
  10. GlaxoSmithKline
  11. Siemens
  12. Sunovion

Ask authors/readers for more resources

Rationale: Genomic regions identified by genome-wide association studies explain only a small fraction of heritability for chronic obstructive pulmonary disease (COPD). Alpha-1 antitrypsin deficiency shows that rare coding variants of large effect also influence COPD susceptibility. We hypothesized that exome sequencing in families identified through a proband with severe, early-onset COPD would identify additional rare genetic determinants of large effect. Objectives: To identify rare genetic determinants of severe COPD. Methods: We applied filtering approaches to identify potential causal variants for COPD in whole exomes from 347 subjects in 49 extended pedigrees from the Boston Early-Onset COPD Study. We assessed the power of this approach under different levels of genetic heterogeneity using simulations. We tested genes identified in these families using gene-based association tests in exomes of 204 cases with severe COPD and 195 resistant smokers from the COPDGene study. In addition, we examined previously described loci associated with COPD using these datasets. Measurements and Main Results: We identified 69 genes with predicted deleterious nonsynonymous, stop, or splice variants that segregated with severe COPD in at least two pedigrees. Four genes (DNAH8, ALCAM, PARS, and GBF1) also demonstrated an increase in rare nonsynonymous, stop, and/or splice mutations in cases compared with resistant smokers from the COPDGene study; however, these results were not statistically significant. We demonstrate the limitations of the power of this approach under genetic heterogeneity through simulation. Conclusions: Rare deleterious coding variants may increase risk for COPD, but multiple genes likely contribute to COPD susceptibility.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available