Motif-Raptor: A Cell Type-Specific and Transcription Factor Centric Approach for Post-GWAS Prioritization of Causal Regulators

Author(s):  
Qiuming Yao ◽  
Paolo Ferragina ◽  
Yakir Reshef ◽  
Guillaume Lettre ◽  
Daniel E Bauer ◽  
...  

Abstract Motivation Genome-wide association studies (GWAS) have identified thousands of common trait-associated genetic variants but interpretation of their function remains challenging. These genetic variants can overlap the binding sites of transcription factors (TFs) and therefore could alter gene expression. However, we currently lack a systematic understanding on how this mechanism contributes to phenotype. Results We present Motif-Raptor, a TF-centric computational tool that integrates sequence-based predictive models, chromatin accessibility, gene expression datasets and GWAS summary statistics to systematically investigate how TF function is affected by genetic variants. Given trait associated non-coding variants, Motif-Raptor can recover relevant cell types and critical TFs to drive hypotheses regarding their mechanism of action. We tested Motif-Raptor on complex traits such as rheumatoid arthritis and red blood cell count and demonstrated its ability to prioritize relevant cell types, potential regulatory TFs and non-coding SNPs which have been previously characterized and validated. Availability Motif-Raptor is freely available as a Python package at: https://github.com/pinellolab/MotifRaptor. Supplementary information Supplementary data are available at Bioinformatics online.

2019 ◽  
Author(s):  
Tom G Richardson ◽  
Gibran Hemani ◽  
Tom R Gaunt ◽  
Caroline L Relton ◽  
George Davey Smith

AbstractBackgroundDeveloping insight into tissue-specific transcriptional mechanisms can help improve our understanding of how genetic variants exert their effects on complex traits and disease. By applying the principles of Mendelian randomization, we have undertaken a systematic analysis to evaluate transcriptome-wide associations between gene expression across 48 different tissue types and 395 complex traits.ResultsOverall, we identified 100,025 gene-trait associations based on conventional genome-wide corrections (P < 5 × 10−08) that also provided evidence of genetic colocalization. These results indicated that genetic variants which influence gene expression levels in multiple tissues are more likely to influence multiple complex traits. We identified many examples of tissue-specific effects, such as genetically-predicted TPO, NR3C2 and SPATA13 expression only associating with thyroid disease in thyroid tissue. Additionally, FBN2 expression was associated with both cardiovascular and lung function traits, but only when analysed in heart and lung tissue respectively.We also demonstrate that conducting phenome-wide evaluations of our results can help flag adverse on-target side effects for therapeutic intervention, as well as propose drug repositioning opportunities. Moreover, we find that exploring the tissue-dependency of associations identified by genome-wide association studies (GWAS) can help elucidate the causal genes and tissues responsible for effects, as well as uncover putative novel associations.ConclusionsThe atlas of tissue-dependent associations we have constructed should prove extremely valuable to future studies investigating the genetic determinants of complex disease. The follow-up analyses we have performed in this study are merely a guide for future research. Conducting similar evaluations can be undertaken systematically at http://mrcieu.mrsoftware.org/Tissue_MR_atlas/.


2019 ◽  
Author(s):  
Yi Yang ◽  
Xingjie Shi ◽  
Yuling Jiao ◽  
Jian Huang ◽  
Min Chen ◽  
...  

AbstractMotivationAlthough genome-wide association studies (GWAS) have deepened our understanding of the genetic architecture of complex traits, the mechanistic links that underlie how genetic variants cause complex traits remains elusive. To advance our understanding of the underlying mechanistic links, various consortia have collected a vast volume of genomic data that enable us to investigate the role that genetic variants play in gene expression regulation. Recently, a collaborative mixed model (CoMM) [42] was proposed to jointly interrogate genome on complex traits by integrating both the GWAS dataset and the expression quantitative trait loci (eQTL) dataset. Although CoMM is a powerful approach that leverages regulatory information while accounting for the uncertainty in using an eQTL dataset, it requires individual-level GWAS data and cannot fully make use of widely available GWAS summary statistics. Therefore, statistically efficient methods that leverages transcriptome information using only summary statistics information from GWAS data are required.ResultsIn this study, we propose a novel probabilistic model, CoMM-S2, to examine the mechanistic role that genetic variants play, by using only GWAS summary statistics instead of individual-level GWAS data. Similar to CoMM which uses individual-level GWAS data, CoMM-S2 combines two models: the first model examines the relationship between gene expression and genotype, while the second model examines the relationship between the phenotype and the predicted gene expression from the first model. Distinct from CoMM, CoMM-S2 requires only GWAS summary statistics. Using both simulation studies and real data analysis, we demonstrate that even though CoMM-S2 utilizes GWAS summary statistics, it has comparable performance as CoMM, which uses individual-level GWAS [email protected] and implementationThe implement of CoMM-S2 is included in the CoMM package that can be downloaded from https://github.com/gordonliu810822/CoMM.Supplementary informationSupplementary data are available at Bioinformatics online.


2021 ◽  
Author(s):  
Karthik A. Jagadeesh ◽  
Kushal K Dey ◽  
Daniel T. Montoro ◽  
Steven Gazal ◽  
Jesse M Engreitz ◽  
...  

Cellular dysfunction is a hallmark of disease. Genome-wide association studies (GWAS) have provided a powerful means to identify loci and genes contributing to disease risk, but in many cases the related cell types/states through which genes confer disease risk remain unknown. Deciphering such relationships is important both for our understanding of disease, and for developing therapeutic interventions. Here, we introduce a framework for integrating single-cell RNA-seq (scRNA-seq), epigenomic maps and GWAS summary statistics to infer the underlying cell types and processes by which genetic variants influence disease. We analyzed 1.6 million scRNA-seq profiles from 209 individuals spanning 11 tissue types and 6 disease conditions, and constructed gene programs capturing cell types, disease progression in cell types, and cellular processes both within and across cell types. We evaluated these gene programs for disease enrichment by transforming them to SNP annotations with tissue-specific epigenomic maps and computing enrichment scores across 60 diseases and complex traits (average N=297K). The inferred disease enrichments recapitulated known biology and highlighted novel relationships for different conditions, including GABAergic neurons in major depressive disorder (MDD), disease progression programs in M cells in ulcerative colitis, and a disease-specific complement cascade process in multiple sclerosis. Our framework provides a powerful approach for identifying the cell types and cellular processes by which genetic variants influence disease.


2021 ◽  
Vol 11 ◽  
Author(s):  
Huanhuan Zhu ◽  
Lulu Shang ◽  
Xiang Zhou

Genome-wide association studies (GWASs) have identified and replicated many genetic variants that are associated with diseases and disease-related complex traits. However, the biological mechanisms underlying these identified associations remain largely elusive. Exploring the biological mechanisms underlying these associations requires identifying trait-relevant tissues and cell types, as genetic variants likely influence complex traits in a tissue- and cell type-specific manner. Recently, several statistical methods have been developed to integrate genomic data with GWASs for identifying trait-relevant tissues and cell types. These methods often rely on different genomic information and use different statistical models for trait-tissue relevance inference. Here, we present a comprehensive technical review to summarize ten existing methods for trait-tissue relevance inference. These methods make use of different genomic information that include functional annotation information, expression quantitative trait loci information, genetically regulated gene expression information, as well as gene co-expression network information. These methods also use different statistical models that range from linear mixed models to covariance network models. We hope that this review can serve as a useful reference both for methodologists who develop methods and for applied analysts who apply these methods for identifying trait relevant tissues and cell types.


2020 ◽  
Vol 36 (12) ◽  
pp. 3811-3817 ◽  
Author(s):  
Songshan Yang ◽  
Jiawei Wen ◽  
Scott T Eckert ◽  
Yaqun Wang ◽  
Dajiang J Liu ◽  
...  

Abstract Motivation Large scale genome-wide association studies (GWAS) have resulted in the identification of a wide range of genetic variants related to a host of complex traits and disorders. Despite their success, the individual single-nucleotide polymorphism (SNP) analysis approach adopted in most current GWAS can be limited in that it is usually biologically simple to elucidate a comprehensive genetic architecture of phenotypes and statistically underpowered due to heavy multiple-testing correction burden. On the other hand, multiple-SNP analyses (e.g. gene-based or region-based SNP-set analysis) are usually more powerful to examine the joint effects of a set of SNPs on the phenotype of interest. However, current multiple-SNP approaches can only draw an overall conclusion at the SNP-set level and does not directly inform which SNPs in the SNP-set are driving the overall genotype–phenotype association. Results In this article, we propose a new permutation-assisted tuning procedure in lasso (plasso) to identify phenotype-associated SNPs in a joint multiple-SNP regression model in GWAS. The tuning parameter of lasso determines the amount of shrinkage and is essential to the performance of variable selection. In the proposed plasso procedure, we first generate permutations as pseudo-SNPs that are not associated with the phenotype. Then, the lasso tuning parameter is delicately chosen to separate true signal SNPs and non-informative pseudo-SNPs. We illustrate plasso using simulations to demonstrate its superior performance over existing methods, and application of plasso to a real GWAS dataset gains new additional insights into the genetic control of complex traits. Availability and implementation R codes to implement the proposed methodology is available at https://github.com/xyz5074/plasso. Supplementary information Supplementary data are available at Bioinformatics online.


2019 ◽  
Vol 36 (7) ◽  
pp. 2009-2016 ◽  
Author(s):  
Yi Yang ◽  
Xingjie Shi ◽  
Yuling Jiao ◽  
Jian Huang ◽  
Min Chen ◽  
...  

Abstract Motivation Although genome-wide association studies (GWAS) have deepened our understanding of the genetic architecture of complex traits, the mechanistic links that underlie how genetic variants cause complex traits remains elusive. To advance our understanding of the underlying mechanistic links, various consortia have collected a vast volume of genomic data that enable us to investigate the role that genetic variants play in gene expression regulation. Recently, a collaborative mixed model (CoMM) was proposed to jointly interrogate genome on complex traits by integrating both the GWAS dataset and the expression quantitative trait loci (eQTL) dataset. Although CoMM is a powerful approach that leverages regulatory information while accounting for the uncertainty in using an eQTL dataset, it requires individual-level GWAS data and cannot fully make use of widely available GWAS summary statistics. Therefore, statistically efficient methods that leverages transcriptome information using only summary statistics information from GWAS data are required. Results In this study, we propose a novel probabilistic model, CoMM-S2, to examine the mechanistic role that genetic variants play, by using only GWAS summary statistics instead of individual-level GWAS data. Similar to CoMM which uses individual-level GWAS data, CoMM-S2 combines two models: the first model examines the relationship between gene expression and genotype, while the second model examines the relationship between the phenotype and the predicted gene expression from the first model. Distinct from CoMM, CoMM-S2 requires only GWAS summary statistics. Using both simulation studies and real data analysis, we demonstrate that even though CoMM-S2 utilizes GWAS summary statistics, it has comparable performance as CoMM, which uses individual-level GWAS data. Availability and implementation The implement of CoMM-S2 is included in the CoMM package that can be downloaded from https://github.com/gordonliu810822/CoMM. Supplementary information Supplementary data are available at Bioinformatics online.


2020 ◽  
Vol 48 (W1) ◽  
pp. W193-W199 ◽  
Author(s):  
Nina Baumgarten ◽  
Dennis Hecker ◽  
Sivarajan Karunanithi ◽  
Florian Schmidt ◽  
Markus List ◽  
...  

Abstract A current challenge in genomics is to interpret non-coding regions and their role in transcriptional regulation of possibly distant target genes. Genome-wide association studies show that a large part of genomic variants are found in those non-coding regions, but their mechanisms of gene regulation are often unknown. An additional challenge is to reliably identify the target genes of the regulatory regions, which is an essential step in understanding their impact on gene expression. Here we present the EpiRegio web server, a resource of regulatory elements (REMs). REMs are genomic regions that exhibit variations in their chromatin accessibility profile associated with changes in expression of their target genes. EpiRegio incorporates both epigenomic and gene expression data for various human primary cell types and tissues, providing an integrated view of REMs in the genome. Our web server allows the analysis of genes and their associated REMs, including the REM’s activity and its estimated cell type-specific contribution to its target gene’s expression. Further, it is possible to explore genomic regions for their regulatory potential, investigate overlapping REMs and by that the dissection of regions of large epigenomic complexity. EpiRegio allows programmatic access through a REST API and is freely available at https://epiregio.de/.


2019 ◽  
Author(s):  
Ana Viñuela ◽  
Arushi Varshney ◽  
Martijn van de Bunt ◽  
Rashmi B. Prasad ◽  
Olof Asplund ◽  
...  

AbstractMost signals detected by genome-wide association studies map to non-coding sequence and their tissue-specific effects influence transcriptional regulation. However, many key tissues and cell-types required for appropriate functional inference are absent from large-scale resources such as ENCODE and GTEx. We explored the relationship between genetic variants influencing predisposition to type 2 diabetes (T2D) and related glycemic traits, and human pancreatic islet transcription using RNA-Seq and genotyping data from 420 islet donors. We find: (a) eQTLs have a variable replication rate across the 44 GTEx tissues (<73%), indicating that our study captured islet-specific cis-eQTL signals; (b) islet eQTL signals show marked overlap with islet epigenome annotation, though eQTL effect size is reduced in the stretch enhancers most strongly implicated in GWAS signal location; (c) selective enrichment of islet eQTL overlap with the subset of T2D variants implicated in islet dysfunction; and (d) colocalization between islet eQTLs and variants influencing T2D or related glycemic traits, delivering candidate effector transcripts at 23 loci, including DGKB and TCF7L2. Our findings illustrate the advantages of performing functional and regulatory studies in tissues of greatest disease-relevance while expanding our mechanistic insights into complex traits association loci activity with an expanded list of putative transcripts implicated in T2D development.


2019 ◽  
Vol 20 (10) ◽  
pp. 765-780 ◽  
Author(s):  
Diana Cruz ◽  
Ricardo Pinto ◽  
Margarida Freitas-Silva ◽  
José Pedro Nunes ◽  
Rui Medeiros

Atrial fibrillation (AF) and stroke are included in a group of complex traits that have been approached regarding of their study by susceptibility genetic determinants. Since 2007, several genome-wide association studies (GWAS) aiming to identify genetic variants modulating AF risk have been conducted. Thus, 11 GWAS have identified 26 SNPs (p < 5 × 10-2), of which 19 reached genome-wide significance (p < 5 × 10-8). From those variants, seven were also associated with cardioembolic stroke and three reached genome-wide significance in stroke GWAS. These associations may shed a light on putative shared etiologic mechanisms between AF and cardioembolic stroke. Additionally, some of these identified variants have been incorporated in genetic risk scores in order to elucidate new approaches of stroke prediction, prevention and treatment.


2020 ◽  
Vol 36 (Supplement_1) ◽  
pp. i194-i202
Author(s):  
Berk A Alpay ◽  
Pinar Demetci ◽  
Sorin Istrail ◽  
Derek Aguiar

Abstract Motivation Genome-wide association studies (GWAS) have discovered thousands of significant genetic effects on disease phenotypes. By considering gene expression as the intermediary between genotype and disease phenotype, expression quantitative trait loci studies have interpreted many of these variants by their regulatory effects on gene expression. However, there remains a considerable gap between genotype-to-gene expression association and genotype-to-gene expression prediction. Accurate prediction of gene expression enables gene-based association studies to be performed post hoc for existing GWAS, reduces multiple testing burden, and can prioritize genes for subsequent experimental investigation. Results In this work, we develop gene expression prediction methods that relax the independence and additivity assumptions between genetic markers. First, we consider gene expression prediction from a regression perspective and develop the HAPLEXR algorithm which combines haplotype clusterings with allelic dosages. Second, we introduce the new gene expression classification problem, which focuses on identifying expression groups rather than continuous measurements; we formalize the selection of an appropriate number of expression groups using the principle of maximum entropy. Third, we develop the HAPLEXD algorithm that models haplotype sharing with a modified suffix tree data structure and computes expression groups by spectral clustering. In both models, we penalize model complexity by prioritizing genetic clusters that indicate significant effects on expression. We compare HAPLEXR and HAPLEXD with three state-of-the-art expression prediction methods and two novel logistic regression approaches across five GTEx v8 tissues. HAPLEXD exhibits significantly higher classification accuracy overall; HAPLEXR shows higher prediction accuracy on approximately half of the genes tested and the largest number of best predicted genes (r2&gt;0.1) among all methods. We show that variant and haplotype features selected by HAPLEXR are smaller in size than competing methods (and thus more interpretable) and are significantly enriched in functional annotations related to gene regulation. These results demonstrate the importance of explicitly modeling non-dosage dependent and intragenic epistatic effects when predicting expression. Availability and implementation Source code and binaries are freely available at https://github.com/rapturous/HAPLEX. Supplementary information Supplementary data are available at Bioinformatics online.


Sign in / Sign up

Export Citation Format

Share Document