BMC Genomics (Sep 2024)

Proteomic networks and related genetic variants associated with smoking and chronic obstructive pulmonary disease

  • Iain R Konigsberg,
  • Thao Vu,
  • Weixuan Liu,
  • Elizabeth M Litkowski,
  • Katherine A Pratte,
  • Luciana B Vargas,
  • Niles Gilmore,
  • Mohamed Abdel-Hafiz,
  • Ani Manichaikul,
  • Michael H Cho,
  • Craig P Hersh,
  • Dawn L DeMeo,
  • Farnoush Banaei-Kashani,
  • Russell P Bowler,
  • Leslie A Lange,
  • Katerina J Kechris

DOI
https://doi.org/10.1186/s12864-024-10619-1
Journal volume & issue
Vol. 25, no. 1
pp. 1 – 20

Abstract

Read online

Abstract Background Studies have identified individual blood biomarkers associated with chronic obstructive pulmonary disease (COPD) and related phenotypes. However, complex diseases such as COPD typically involve changes in multiple molecules with interconnections that may not be captured when considering single molecular features. Methods Leveraging proteomic data from 3,173 COPDGene Non-Hispanic White (NHW) and African American (AA) participants, we applied sparse multiple canonical correlation network analysis (SmCCNet) to 4,776 proteins assayed on the SomaScan v4.0 platform to derive sparse networks of proteins associated with current vs. former smoking status, airflow obstruction, and emphysema quantitated from high-resolution computed tomography scans. We then used NetSHy, a dimension reduction technique leveraging network topology, to produce summary scores of each proteomic network, referred to as NetSHy scores. We next performed a genome-wide association study (GWAS) to identify variants associated with the NetSHy scores, or network quantitative trait loci (nQTLs). Finally, we evaluated the replicability of the networks in an independent cohort, SPIROMICS. Results We identified networks of 13 to 104 proteins for each phenotype and exposure in NHW and AA, and the derived NetSHy scores significantly associated with the variable of interests. Networks included known (sRAGE, ALPP, MIP1) and novel molecules (CA10, CPB1, HIS3, PXDN) and interactions involved in COPD pathogenesis. We observed 7 nQTL loci associated with NetSHy scores, 4 of which remained after conditional analysis. Networks for smoking status and emphysema, but not airflow obstruction, demonstrated a high degree of replicability across race groups and cohorts. Conclusions In this work, we apply state-of-the-art molecular network generation and summarization approaches to proteomic data from COPDGene participants to uncover protein networks associated with COPD phenotypes. We further identify genetic associations with networks. This work discovers protein networks containing known and novel proteins and protein interactions associated with clinically relevant COPD phenotypes across race groups and cohorts.

Keywords