학술논문

Canonical correlation analysis for multi-omics: Application to cross-cohort analysis.
Document Type
Article
Source
PLoS Genetics. 5/22/2023, Vol. 19 Issue 5, p1-22. 22p.
Subject
*MULTIOMICS
*BLOOD cell count
*STATISTICAL correlation
*SYSTEMS biology
*BIOLOGICAL systems
*BLOOD cells
Language
ISSN
1553-7390
Abstract
Integrative approaches that simultaneously model multi-omics data have gained increasing popularity because they provide holistic system biology views of multiple or all components in a biological system of interest. Canonical correlation analysis (CCA) is a correlation-based integrative method designed to extract latent features shared between multiple assays by finding the linear combinations of features–referred to as canonical variables (CVs)–within each assay that achieve maximal across-assay correlation. Although widely acknowledged as a powerful approach for multi-omics data, CCA has not been systematically applied to multi-omics data in large cohort studies, which has only recently become available. Here, we adapted sparse multiple CCA (SMCCA), a widely-used derivative of CCA, to proteomics and methylomics data from the Multi-Ethnic Study of Atherosclerosis (MESA) and Jackson Heart Study (JHS). To tackle challenges encountered when applying SMCCA to MESA and JHS, our adaptations include the incorporation of the Gram-Schmidt (GS) algorithm with SMCCA to improve orthogonality among CVs, and the development of Sparse Supervised Multiple CCA (SSMCCA) to allow supervised integration analysis for more than two assays. Effective application of SMCCA to the two real datasets reveals important findings. Applying our SMCCA-GS to MESA and JHS, we identified strong associations between blood cell counts and protein abundance, suggesting that adjustment of blood cell composition should be considered in protein-based association studies. Importantly, CVs obtained from two independent cohorts also demonstrate transferability across the cohorts. For example, proteomic CVs learned from JHS, when transferred to MESA, explain similar amounts of blood cell count phenotypic variance in MESA, explaining 39.0% ~ 50.0% variation in JHS and 38.9% ~ 49.1% in MESA. Similar transferability was observed for other omics-CV-trait pairs. This suggests that biologically meaningful and cohort-agnostic variation is captured by CVs. We anticipate that applying our SMCCA-GS and SSMCCA on various cohorts would help identify cohort-agnostic biologically meaningful relationships between multi-omics data and phenotypic traits. Author summary: Comprehensive understanding of human complex traits may benefit from incorporation of molecular features from multiple biological layers such as genome, epigenome, transcriptome, proteome, and metabolome. CCA is a correlation-based method for multi-omics data which reduces the dimension of each omic assay to several orthogonal components–commonly referred to as canonical variables (CVs). The widely-used SMCCA method allows effective dimension reduction and integration of multi-omics data, but suffers from potentially highly correlated CVs when applied to high-dimensional omics data. Here, we improve the statistical independence among the CVs by adopting a variation of the GS algorithm. We applied our SMCCA-GS method to proteomic and methylomic data from two cohort studies, MESA and JHS. Our results reveal a pronounced effect of blood cell counts on protein abundance, suggesting blood cell composition adjustment in protein-based association studies may be necessary. Finally, we present SSMCCA which allows supervised CCA analysis for the association between one phenotype of interest and more than two assays. We anticipate that SMCCA-GS would help reveal meaningful system-level factors from biological processes involving features from multiple assays; and SSMCCA would further empower interrogation of these factors for phenotypic traits related to health and diseases. [ABSTRACT FROM AUTHOR]