scholarly journals Efficient and flexible Integration of variant characteristics in rare variant association studies using integrated nested Laplace approximation

2021 ◽  
Vol 17 (2) ◽  
pp. e1007784
Author(s):  
Hana Susak ◽  
Laura Serra-Saurina ◽  
German Demidov ◽  
Raquel Rabionet ◽  
Laura Domènech ◽  
...  

Rare variants are thought to play an important role in the etiology of complex diseases and may explain a significant fraction of the missing heritability in genetic disease studies. Next-generation sequencing facilitates the association of rare variants in coding or regulatory regions with complex diseases in large cohorts at genome-wide scale. However, rare variant association studies (RVAS) still lack power when cohorts are small to medium-sized and if genetic variation explains a small fraction of phenotypic variance. Here we present a novel Bayesian rare variant Association Test using Integrated Nested Laplace Approximation (BATI). Unlike existing RVAS tests, BATI allows integration of individual or variant-specific features as covariates, while efficiently performing inference based on full model estimation. We demonstrate that BATI outperforms established RVAS methods on realistic, semi-synthetic whole-exome sequencing cohorts, especially when using meaningful biological context, such as functional annotation. We show that BATI achieves power above 70% in scenarios in which competing tests fail to identify risk genes, e.g. when risk variants in sum explain less than 0.5% of phenotypic variance. We have integrated BATI, together with five existing RVAS tests in the ‘Rare Variant Genome Wide Association Study’ (rvGWAS) framework for data analyzed by whole-exome or whole genome sequencing. rvGWAS supports rare variant association for genes or any other biological unit such as promoters, while allowing the analysis of essential functionalities like quality control or filtering. Applying rvGWAS to a Chronic Lymphocytic Leukemia study we identified eight candidate predisposition genes, including EHMT2 and COPS7A.

2020 ◽  
Author(s):  
Hana Susak ◽  
Laura Serra-Saurina ◽  
Raquel Rabionet Janssen ◽  
Laura Domènech ◽  
Mattia Bosio ◽  
...  

AbstractRare variants are thought to play an important role in the etiology of complex diseases and may explain a significant fraction of the missing heritability in genetic disease studies. Next-generation sequencing facilitates the association of rare variants in coding or regulatory regions with complex diseases in large cohorts at genome-wide scale. However, rare variant association studies (RVAS) still lack power when cohorts are small to medium-sized and if genetic variation explains a small fraction of phenotypic variance. Here we present a novel Bayesian rare variant Association Test using Integrated Nested Laplace Approximation (BATI). Unlike existing RVAS tests, BATI allows integration of individual or variant-specific features as covariates, while efficiently performing inference based on full model estimation. We demonstrate that BATI outperforms established RVAS methods on realistic, semi-synthetic whole-exome sequencing cohorts, especially when using meaningful biological context, such as functional annotation. We show that BATI achieves power above 75% in scenarios in which competing tests fail to identify risk genes, e.g. when risk variants in sum explain less than 0.5% of phenotypic variance. We have integrated BATI, together with five existing RVAS tests in the ‘Rare Variant Genome Wide Association Study’ (rvGWAS) framework for data analyzed by whole-exome or whole genome sequencing. rvGWAS supports rare variant association for genes or any other biological unit such as promoters, while allowing the analysis of essential functionalities like quality control or filtering. Applying rvGWAS to a Chronic Lymphocytic Leukemia study we identified eight candidate predisposition genes, including EHMT2 and COPS7A.Data availability and implementationAll relevant data are within the manuscript and pipeline implementation on https://github.com/hanasusak/rvGWASAuthor summaryComplex diseases are characterized by being related to genetic factors and environmental factors such as air pollution, diet etc. that together define the susceptibility of each individual to develop a given disease. Much effort has been applied to advance the knowledge of the genetic bases of such diseases, specially in the discovery of frequent genetic variants in the population increasing disease risk. However, these variants usually explain a little part of the etiology of such diseases. Previous studies have shown that rare variants, i.e. variants present in less than 1% of the population, may explain the rest of the variability related to genetic aspects of the disease.Genome sequencing offers the opportunity to discover rare variants, but powerful statistical methods are needed to discriminate those variants that induce susceptibility to the disease. Here we have developed a powerful and flexible statistical approach for the detection of rare variants associated with a disease and we have integrated it into a computer tool that is easy and intuitive for the researchers and clinicians to use. We have shown that our approach outperformed other common statistical methods specially in a situation where these variants explain just a small part of the disease. The discovery of these rare variants will contribute to the knowledge of the molecular mechanism of complex diseases.


2019 ◽  
Vol 101 ◽  
Author(s):  
Lifeng Liu ◽  
Pengfei Wang ◽  
Jingbo Meng ◽  
Lili Chen ◽  
Wensheng Zhu ◽  
...  

Abstract In recent years, there has been an increasing interest in detecting disease-related rare variants in sequencing studies. Numerous studies have shown that common variants can only explain a small proportion of the phenotypic variance for complex diseases. More and more evidence suggests that some of this missing heritability can be explained by rare variants. Considering the importance of rare variants, researchers have proposed a considerable number of methods for identifying the rare variants associated with complex diseases. Extensive research has been carried out on testing the association between rare variants and dichotomous, continuous or ordinal traits. So far, however, there has been little discussion about the case in which both genotypes and phenotypes are ordinal variables. This paper introduces a method based on the γ-statistic, called OV-RV, for examining disease-related rare variants when both genotypes and phenotypes are ordinal. At present, little is known about the asymptotic distribution of the γ-statistic when conducting association analyses for rare variants. One advantage of OV-RV is that it provides a robust estimation of the distribution of the γ-statistic by employing the permutation approach proposed by Fisher. We also perform extensive simulations to investigate the numerical performance of OV-RV under various model settings. The simulation results reveal that OV-RV is valid and efficient; namely, it controls the type I error approximately at the pre-specified significance level and achieves greater power at the same significance level. We also apply OV-RV for rare variant association studies of diastolic blood pressure.


2011 ◽  
Vol 26 (S2) ◽  
pp. 1346-1346
Author(s):  
D. Benmessaoud ◽  
A.-M. Lepagnol-Bestel ◽  
M. Delepine ◽  
J. Hager ◽  
J.-M. Moalic ◽  
...  

Genome wide association studies (GWAS) of Schizophrenia (SZ) patients have identified common variants in ten genes including SMARCA2 (Koga et al., HMG, 2009). We found that the SZ-GWAS genes are part of an interacting network centered on SMARCA2 (Loe-Mie et al., HMG, 2010). Furthermore, SMARCA2 was found disrupted in SZ (Walsh et al., Science, 2008). SMARCA2 encodes the ATPase (BRM) of the SWI/SNF chromatin remodeling complex that is at the interface of genome and environmental adaptation.Taking advantage of an Algerian trio cohort of one hundred SZ patients (Benmessaoud et al., BMC Psychiatry, 2008), we replicated the association of SNP rs2296212 localized in exon 33, already shown associated in Koga study and resulting in D1546E amino acid change in the SMARCA2 protein. We studied SMARCA2 codons and found that exon 33 displays a signature of positive evolution in the primate lineage.Our working hypothesis is that the coding regions displaying positive selection are target of novel rare variants. To address this question, we sequenced two exons displaying positive evolution and one exon without evidence of positive evolution.We found (i) that rare variants are significantly in excess in SZ-patients compared to their parents (p = 0.038, Fisher test) and (ii) a higher proportion of rare variants in the primate-accelerated exons compared with the non-evolutionary exon in SZ-patients (p = 0.032, Fisher test).SMARCA2 exon sequencing and whole exome sequencing from patients harboring SNP rs2296212 common variant are under progress. Altogether, these results are expected to give new insights into the genetic architecture of SZ.


2019 ◽  
Author(s):  
George Kanoungi ◽  
Michael Nothnagel ◽  
Tim Becker ◽  
Dmitriy Drichel

AbstractRegion-based genome-wide scans are usually performed by use of a priori chosen analysis regions. Such an approach will likely miss the region comprising the strongest signal and, thus, may result in increased type II error rates and decreased power. Here, we propose a genomic exhaustive scan approach that analyzes all possible subsequences and does not rely on a prior definition of the analysis regions. As a prime instance, we present a computationally ultra-efficient implementation using the rare-variant collapsing test for phenotypic association, the genomic exhaustive collapsing scan (GECS). Our implementation allows for the identification of regions comprising the strongest signals in large, genome-wide rare-variant association studies while controlling the family-wise error rate via permutation. Application of GECS to two genomic data sets revealed several novel significantly associated regions for age-related macular degeneration and for schizophrenia. Our approach also offers a high potential for genome-wide scans for selection, methylation and other analyses.


2017 ◽  
Vol 103 (2) ◽  
pp. 649-659 ◽  
Author(s):  
Sasha R Howard ◽  
Leonardo Guasti ◽  
Ariel Poliandri ◽  
Alessia David ◽  
Claudia P Cabrera ◽  
...  

Abstract Context Self-limited delayed puberty (DP) is often associated with a delay in physical maturation, but although highly heritable the causal genetic factors remain elusive. Genome-wide association studies of the timing of puberty have identified multiple loci for age at menarche in females and voice break in males, particularly in pathways controlling energy balance. Objective/Main Outcome Measures We sought to assess the contribution of rare variants in such genes to the phenotype of familial DP. Design/Patients We performed whole-exome sequencing in 67 pedigrees (125 individuals with DP and 35 unaffected controls) from our unique cohort of familial self-limited DP. Using a whole-exome sequencing filtering pipeline one candidate gene [fat mass and obesity–associated gene (FTO)] was identified. In silico, in vitro, and mouse model studies were performed to investigate the pathogenicity of FTO variants and timing of puberty in FTO+/− mice. Results We identified potentially pathogenic, rare variants in genes in linkage disequilibrium with genome-wide association studies of age at menarche loci in 283 genes. Of these, five genes were implicated in the control of body mass. After filtering for segregation with trait, one candidate, FTO, was retained. Two FTO variants, found in 14 affected individuals from three families, were also associated with leanness in these patients with DP. One variant (p.Leu44Val) demonstrated altered demethylation activity of the mutant protein in vitro. Fto+/− mice displayed a significantly delayed timing of pubertal onset (P < 0.05). Conclusions Mutations in genes implicated in body mass and timing of puberty in the general population may contribute to the pathogenesis of self-limited DP.


2018 ◽  
Author(s):  
Christopher DeBoever ◽  
Matthew Aguirre ◽  
Yosuke Tanigawa ◽  
Chris C. A. Spencer ◽  
Timothy Poterba ◽  
...  

AbstractWhole genome sequencing studies applied to large populations or biobanks with extensive phenotyping raise new analytic challenges. The need to consider many variants at a locus or group of genes simultaneously and the potential to study many correlated phenotypes with shared genetic architecture provide opportunities for discovery and inference that are not addressed by the traditional one variant-one phenotype association study. Here we introduce a model comparison approach we refer to as MRP for rare variant association studies that considers correlation, scale, and location of genetic effects across a group of genetic variants, phenotypes, and studies. We consider the use of summary statistic data to apply univariate and multivariate gene-based meta-analysis models for identifying rare variant associations with an emphasis on protective protein-truncating variants that can expedite drug discovery. Through simulation studies, we demonstrate that the proposed model comparison approach can improve ability to detect rare variant association signals. We also apply the model to two groups of phenotypes from the UK Biobank: 1) asthma diagnosis, eosinophil counts, forced expiratory volume, and forced vital capacity; and 2) glaucoma diagnosis, intra-ocular pressure, and corneal resistance factor. We are able to recover known associations such as the protective association between rs146597587 in IL33 and asthma. We also find evidence for novel protective associations between rare variants in ANGPTL7 and glaucoma. Overall, we show that the MRP model comparison approach is able to retain and improve upon useful features from widely-used meta-analysis approaches for rare variant association analyses and prioritize protective modifiers of disease risk.Author summaryDue to the continually decreasing cost of acquiring genetic data, we are now beginning to see large collections of individuals for which we have both genetic information and trait data such as disease status, physical measurements, biomarker levels, and more. These datasets offer new opportunities to find relationships between inherited genetic variation and disease. While it is known that there are relationships between different traits, typical genetic analyses only focus on analyzing one genetic variant and one phenotype at a time. Additionally, it is difficult to identify rare genetic variants that are associated with disease due to their scarcity, even among large sample sizes. In this work, we present a method for identifying associations between genetic variation and disease that considers multiple rare variants and phenotypes at the same time. By sharing information across rare variant and phenotypes, we improve our ability to identify rare variants associated with disease compared to considering a single rare variant and a single phenotype. The method can be used to identify candidate disease genes as well as genes that might represent attractive drug targets.


2020 ◽  
Vol 66 (1) ◽  
pp. 11-23
Author(s):  
Yukihide Momozawa ◽  
Keijiro Mizukami

AbstractGenome-wide association studies have identified >10,000 genetic variants associated with various phenotypes and diseases. Although the majority are common variants, rare variants with >0.1% of minor allele frequency have been investigated by imputation and using disease-specific custom SNP arrays. Rare variants sequencing analysis mainly revealed have played unique roles in the genetics of complex diseases in humans due to their distinctive features, in contrast to common variants. Unique roles are hypothesis-free evidence for gene causality, a precise target of functional analysis for understanding disease mechanisms, a new favorable target for drug development, and a genetic marker with high disease risk for personalized medicine. As whole-genome sequencing continues to identify more rare variants, the roles associated with rare variants will also increase. However, a better estimation of the functional impact of rare variants across whole genome is needed to enhance their contribution to improvements in human health.


2019 ◽  
Author(s):  
Mart Kals ◽  
Tiit Nikopensius ◽  
Kristi Läll ◽  
Kalle Pärn ◽  
Timo Tõnis Sikka ◽  
...  

AbstractGenotype imputation has become a standard procedure prior genome-wide association studies (GWASs). For common and low-frequency variants, genotype imputation can be performed sufficiently accurately with publicly available and ethnically heterogeneous reference datasets like 1000 Genomes Project (1000G) and Haplotype Reference Consortium panels. However, the imputation of rare variants has been shown to be significantly more accurate when ethnically matched reference panel is used. Even more, greater genetic similarity between reference panel and target samples facilitates the detection of rare (or even population-specific) causal variants. Notwithstanding, the genome-wide downstream consequences and differences of using ethnically mixed and matched reference panels have not been yet comprehensively explored.We determined and quantified these differences by performing several comparative evaluations of the discovery-driven analysis scenarios. A variant-wise GWAS was performed on seven complex diseases and body mass index by using genome-wide genotype data of ∼37,000 Estonians imputed with ethnically mixed 1000G and ethnically matched imputation reference panels. Although several previously reported common (minor allele frequency; MAF > 5%) variant associations were replicated in both resulting imputed datasets, no major differences were observed among the genome-wide significant findings or in the fine-mapping effort. In the analysis of rare (MAF < 1%) coding variants, 46 significantly associated genes were identified in the ethnically matched imputed data as compared to four genes in the 1000G panel based imputed data. All resulting genes were consequently studied in the UK Biobank data.These associations provide a solid example of how rare variants can be efficiently analysed to discover novel, potentially functional genetic variants in relevant phenotypes. Furthermore, our work serves as proof of a cost-efficient study design, demonstrating that the usage of ethnically matched imputation reference panels can enable substantially improved imputation of rare variants, facilitating novel high-confidence findings in rare variant GWAS scans.Author summaryOver the last decade, genome-wide association studies (GWASs) have been widely used for detecting genetic biomarkers in a wide range of traits. Typically, GWASs are carried out using chip-based genotyping data, which are then combined with a more densely genotyped reference panel to infer untyped genetic variants in chip-typed individuals. The latter method is called genotype imputation and its accuracy depends on multiple factors. Publicly available and ethnically heterogeneous imputation reference panels (IRPs) such as 1000 Genomes Project (1000G) are sufficiently accurate for imputation of common and low-frequency variants, but custom ethnically matched IRPs outperform these in case of rare variants. In this work, we systematically compare downstream association analysis effects on eight complex traits in ∼37,000 Estonians imputed with ethnically mixed and ethnically matched IRPs. We do not observe major differences in the single variant analysis, where both imputed datasets replicate previously reported significant loci. But in the gene-based analysis of rare protein-coding variants we show that ethnically matched panel clearly outperforms 1000G panel based imputation, providing 10-fold increase in significant gene-trait associations. Our study demonstrates empirically that imputed data based on ethnically matched panel is very promising for rare variant analysis – it captures more population-specific variants and makes it possible to efficiently identify novel findings.


2015 ◽  
Author(s):  
Lawrence H. Uricchio ◽  
John S. Witte ◽  
Ryan D. Hernandez

Much recent debate has focused on the role of rare variants in complex phenotypes. However, it is well known that rare alleles can only contribute a substantial proportion of the phenotypic variance when they have much larger effect sizes than common variants, which is most easily explained by natural selection constraining trait-altering alleles to low frequency. It is also plausible that demographic events will influence the genetic architecture of complex traits. Unfortunately, most rare variant association tests do not explicitly model natural selection or non-equilibrium demography. Here, we develop a novel evolutionary model of complex traits. We perform numerical calculations and simulate phenotypes under this model using inferred human demographic and selection parameters. We show that rare variants only contribute substantially to complex traits under very strong assumptions about the relationship between effect size and selection strength. We then assess the performance of state-of-the-art rare variant tests using our simulations across a broad range of model parameters. Counterintuitively, we find that statistical power is lowest when rare variants make the greatest contribution to the additive variance, and that power is substantially lower under our model than previously studied models. While many empirical studies have attempted to identify causal loci using rare variant association methods, few have reported novel associations. Some authors have interpreted this to mean that rare variants contribute little to heritability, but our results show that an alternative explanation is that rare variant tests have less power than previously estimated.


Nature ◽  
2021 ◽  
Vol 590 (7845) ◽  
pp. 290-299 ◽  
Author(s):  
Daniel Taliun ◽  
◽  
Daniel N. Harris ◽  
Michael D. Kessler ◽  
Jedidiah Carlson ◽  
...  

AbstractThe Trans-Omics for Precision Medicine (TOPMed) programme seeks to elucidate the genetic architecture and biology of heart, lung, blood and sleep disorders, with the ultimate goal of improving diagnosis, treatment and prevention of these diseases. The initial phases of the programme focused on whole-genome sequencing of individuals with rich phenotypic data and diverse backgrounds. Here we describe the TOPMed goals and design as well as the available resources and early insights obtained from the sequence data. The resources include a variant browser, a genotype imputation server, and genomic and phenotypic data that are available through dbGaP (Database of Genotypes and Phenotypes)1. In the first 53,831 TOPMed samples, we detected more than 400 million single-nucleotide and insertion or deletion variants after alignment with the reference genome. Additional previously undescribed variants were detected through assembly of unmapped reads and customized analysis in highly variable loci. Among the more than 400 million detected variants, 97% have frequencies of less than 1% and 46% are singletons that are present in only one individual (53% among unrelated individuals). These rare variants provide insights into mutational processes and recent human evolutionary history. The extensive catalogue of genetic variation in TOPMed studies provides unique opportunities for exploring the contributions of rare and noncoding sequence variants to phenotypic variation. Furthermore, combining TOPMed haplotypes with modern imputation methods improves the power and reach of genome-wide association studies to include variants down to a frequency of approximately 0.01%.


Sign in / Sign up

Export Citation Format

Share Document