Cargando…

Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study

An enormous challenge in the post-genome era is to annotate and resolve the consequences of genetic variation on diverse phenotypes. The genome-wide association study (GWAS) is a well-known method to identify potential genetic loci for complex traits from huge genetic variations, following which it...

Descripción completa

Detalles Bibliográficos
Autores principales: Yuan, Kai, Zeng, Tao, Chen, Luonan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8826544/
https://www.ncbi.nlm.nih.gov/pubmed/35155440
http://dx.doi.org/10.3389/fcell.2021.720321
_version_ 1784647448988221440
author Yuan, Kai
Zeng, Tao
Chen, Luonan
author_facet Yuan, Kai
Zeng, Tao
Chen, Luonan
author_sort Yuan, Kai
collection PubMed
description An enormous challenge in the post-genome era is to annotate and resolve the consequences of genetic variation on diverse phenotypes. The genome-wide association study (GWAS) is a well-known method to identify potential genetic loci for complex traits from huge genetic variations, following which it is crucial to identify expression quantitative trait loci (eQTL). However, the conventional eQTL methods usually disregard the systematical role of single-nucleotide polymorphisms (SNPs) or genes, thereby overlooking many network-associated phenotypic determinates. Such a problem motivates us to recognize the network-based quantitative trait loci (QTL), i.e., network QTL (nQTL), which is to detect the cascade association as genotype → network → phenotype rather than conventional genotype → expression → phenotype in eQTL. Specifically, we develop the nQTL framework on the theory and approach of single-sample networks, which can identify not only network traits (e.g., the gene subnetwork associated with genotype) for analyzing complex biological processes but also network signatures (e.g., the interactive gene biomarker candidates screened from network traits) for characterizing targeted phenotype and corresponding subtypes. Our results show that the nQTL framework can efficiently capture associations between SNPs and network traits (i.e., edge traits) in various simulated data scenarios, compared with traditional eQTL methods. Furthermore, we have carried out nQTL analysis on diverse biological and biomedical datasets. Our analysis is effective in detecting network traits for various biological problems and can discover many network signatures for discriminating phenotypes, which can help interpret the influence of nQTL on disease subtyping, disease prognosis, drug response, and pathogen factor association. Particularly, in contrast to the conventional approaches, the nQTL framework could also identify many network traits from human bulk expression data, validated by matched single-cell RNA-seq data in an independent or unsupervised manner. All these results strongly support that nQTL and its detection framework can simultaneously explore the global genotype–network–phenotype associations and the underlying network traits or network signatures with functional impact and importance.
format Online
Article
Text
id pubmed-8826544
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-88265442022-02-10 Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study Yuan, Kai Zeng, Tao Chen, Luonan Front Cell Dev Biol Cell and Developmental Biology An enormous challenge in the post-genome era is to annotate and resolve the consequences of genetic variation on diverse phenotypes. The genome-wide association study (GWAS) is a well-known method to identify potential genetic loci for complex traits from huge genetic variations, following which it is crucial to identify expression quantitative trait loci (eQTL). However, the conventional eQTL methods usually disregard the systematical role of single-nucleotide polymorphisms (SNPs) or genes, thereby overlooking many network-associated phenotypic determinates. Such a problem motivates us to recognize the network-based quantitative trait loci (QTL), i.e., network QTL (nQTL), which is to detect the cascade association as genotype → network → phenotype rather than conventional genotype → expression → phenotype in eQTL. Specifically, we develop the nQTL framework on the theory and approach of single-sample networks, which can identify not only network traits (e.g., the gene subnetwork associated with genotype) for analyzing complex biological processes but also network signatures (e.g., the interactive gene biomarker candidates screened from network traits) for characterizing targeted phenotype and corresponding subtypes. Our results show that the nQTL framework can efficiently capture associations between SNPs and network traits (i.e., edge traits) in various simulated data scenarios, compared with traditional eQTL methods. Furthermore, we have carried out nQTL analysis on diverse biological and biomedical datasets. Our analysis is effective in detecting network traits for various biological problems and can discover many network signatures for discriminating phenotypes, which can help interpret the influence of nQTL on disease subtyping, disease prognosis, drug response, and pathogen factor association. Particularly, in contrast to the conventional approaches, the nQTL framework could also identify many network traits from human bulk expression data, validated by matched single-cell RNA-seq data in an independent or unsupervised manner. All these results strongly support that nQTL and its detection framework can simultaneously explore the global genotype–network–phenotype associations and the underlying network traits or network signatures with functional impact and importance. Frontiers Media S.A. 2022-01-26 /pmc/articles/PMC8826544/ /pubmed/35155440 http://dx.doi.org/10.3389/fcell.2021.720321 Text en Copyright © 2022 Yuan, Zeng and Chen. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Cell and Developmental Biology
Yuan, Kai
Zeng, Tao
Chen, Luonan
Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study
title Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study
title_full Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study
title_fullStr Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study
title_full_unstemmed Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study
title_short Interpreting Functional Impact of Genetic Variations by Network QTL for Genotype–Phenotype Association Study
title_sort interpreting functional impact of genetic variations by network qtl for genotype–phenotype association study
topic Cell and Developmental Biology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8826544/
https://www.ncbi.nlm.nih.gov/pubmed/35155440
http://dx.doi.org/10.3389/fcell.2021.720321
work_keys_str_mv AT yuankai interpretingfunctionalimpactofgeneticvariationsbynetworkqtlforgenotypephenotypeassociationstudy
AT zengtao interpretingfunctionalimpactofgeneticvariationsbynetworkqtlforgenotypephenotypeassociationstudy
AT chenluonan interpretingfunctionalimpactofgeneticvariationsbynetworkqtlforgenotypephenotypeassociationstudy