scispace - formally typeset
Search or ask a question
Author

Nurlan Kerimov

Bio: Nurlan Kerimov is an academic researcher from University of Tartu. The author has contributed to research in topics: Expression quantitative trait loci & Quantitative trait locus. The author has an hindex of 4, co-authored 5 publications receiving 73 citations.

Papers
More filters
Journal ArticleDOI
TL;DR: The eQTL Catalogue as discussed by the authors is a set of gene expression quantitative trait locus (eQTL) studies published their summary statistics, which can be used to gain insight into complex human traits by downstream analyses, such as fine mapping and co-localization.
Abstract: Many gene expression quantitative trait locus (eQTL) studies have published their summary statistics, which can be used to gain insight into complex human traits by downstream analyses, such as fine mapping and co-localization. However, technical differences between these datasets are a barrier to their widespread use. Consequently, target genes for most genome-wide association study (GWAS) signals have still not been identified. In the present study, we present the eQTL Catalogue ( https://www.ebi.ac.uk/eqtl ), a resource of quality-controlled, uniformly re-computed gene expression and splicing QTLs from 21 studies. We find that, for matching cell types and tissues, the eQTL effect sizes are highly reproducible between studies. Although most QTLs were shared between most bulk tissues, we identified a greater diversity of cell-type-specific QTLs from purified cell types, a subset of which also manifested as new disease co-localizations. Our summary statistics are freely available to enable the systematic interpretation of human GWAS associations across many cell types and tissues.

122 citations

Posted ContentDOI
29 Jan 2020-bioRxiv
TL;DR: The eQTL Catalogue is presented, a resource which contains quality controlled, uniformly recomputed QTLs from 21 eQtl studies, and it is found that for matching cell types and tissues, the eZTL effect sizes are highly reproducible between studies, enabling the integrative analysis of these data.
Abstract: An increasing number of gene expression quantitative trait locus (QTL) studies have made summary statistics publicly available, which can be used to gain insight into human complex traits by downstream analyses such as fine-mapping and colocalisation. However, differences between these datasets in their variants tested, allele codings, and in the transcriptional features quantified are a barrier to their widespread use. Here, we present the eQTL Catalogue, a resource which contains quality controlled, uniformly re-computed QTLs from 19 eQTL publications. In addition to gene expression QTLs, we have also identified QTLs at the level of exon expression, transcript usage, and promoter, splice junction and 3ʹ end usage. Our summary statistics can be downloaded by FTP or accessed via a REST API and are also accessible via the Open Targets Genetics Portal. We demonstrate how the eQTL Catalogue and GWAS Catalog APIs can be used to perform colocalisation analysis between GWAS and QTL results without downloading and reformatting summary statistics. New datasets will continuously be added to the eQTL Catalogue, enabling systematic interpretation of human GWAS associations across a large number of cell types and tissues. The eQTL Catalogue is available at https://www.ebi.ac.uk/eqtl/.

62 citations

Posted ContentDOI
05 Sep 2021-medRxiv
TL;DR: In this paper, the authors performed and integrated fine-mapping across 148 complex traits in three large-scale biobanks (BioBank Japan4,5, FinnGen6, and UK Biobank7,8; total n = 811,261), resulting in 4,518 variant-trait pairs with high posterior probability (> 0.9) of causality.
Abstract: Despite the great success of genome-wide association studies (GWAS) in identifying genetic loci significantly associated with diseases, the vast majority of causal variants underlying disease-associated loci have not been identified1–3. To create an atlas of causal variants, we performed and integrated fine-mapping across 148 complex traits in three large-scale biobanks (BioBank Japan4,5, FinnGen6, and UK Biobank7,8; total n = 811,261), resulting in 4,518 variant-trait pairs with high posterior probability (> 0.9) of causality. Of these, we found 285 high-confidence variant-trait pairs replicated across multiple populations, and we characterized multiple contributors to the surprising lack of overlap among fine-mapping results from different biobanks. By studying the bottlenecked Finnish and Japanese populations, we identified 21 and 26 putative causal coding variants with extreme allele frequency enrichment (> 10-fold) in these two populations, respectively. Aggregating data across populations enabled identification of 1,492 unique fine-mapped coding variants and 176 genes in which multiple independent coding variants influence the same trait (i.e., with an allelic series of coding variants). Our results demonstrate that fine-mapping in diverse populations enables novel insights into the biology of complex traits by pinpointing high-confidence causal variants for further characterization.

35 citations

Journal ArticleDOI
03 Sep 2020-eLife
TL;DR: Co-expression modules inferred from gene expression data with five methods as traits as traits in trans-eQTL analysis are used to limit multiple testing and improve interpretability and highlight how co-expression combined with functional enrichment analysis improves the identification and prioritisation of trans- eQTLs when applied to emerging cell-type-specific datasets.
Abstract: Understanding the causal processes that contribute to disease onset and progression is essential for developing novel therapies. Although trans-acting expression quantitative trait loci (trans-eQTLs) can directly reveal cellular processes modulated by disease variants, detecting trans-eQTLs remains challenging due to their small effect sizes. Here, we analysed gene expression and genotype data from six blood cell types from 226 to 710 individuals. We used co-expression modules inferred from gene expression data with five methods as traits in trans-eQTL analysis to limit multiple testing and improve interpretability. In addition to replicating three established associations, we discovered a novel trans-eQTL near SLC39A8 regulating a module of metallothionein genes in LPS-stimulated monocytes. Interestingly, this effect was mediated by a transient cis-eQTL present only in early LPS response and lost before the trans effect appeared. Our analyses highlight how co-expression combined with functional enrichment analysis improves the identification and prioritisation of trans-eQTLs when applied to emerging cell-type-specific datasets.

20 citations

Posted ContentDOI
24 Apr 2020-bioRxiv
TL;DR: This analysis provides a rare detailed characterisation of a trans-eQTL effect cascade from a proximal cis effect to the affected signalling pathway, transcription factor, and target genes.
Abstract: Background Developing novel therapies for complex disease requires better understanding of the causal processes that contribute to disease onset and progression. Although trans-acting gene expression quantitative trait loci (trans-eQTLs) can be a powerful approach to directly reveal cellular processes modulated by disease variants, detecting trans-eQTLs remains challenging due to their small effect sizes and large number of genes tested. However, if a single trans-eQTL controls a group of co-regulated genes, then multiple testing burden can be greatly reduced by summarising gene expression at the level of co-expression modules prior to trans-eQTL analysis. Results We analysed gene expression and genotype data from six blood cell types from 226 to 710 individuals. We inferred gene co-expression modules with five methods on the full dataset, as well as in each cell type separately. We detected a number of established co-expression module trans-eQTLs, such as the monocyte-specific associations at the IFNB1 and LYZ loci, as well as a platelet-specific ARHGEF3 locus associated with mean platelet volume. We also discovered a novel trans association near the SLC39A8 gene in LPS-stimulated monocytes. Here, we linked an early-response cis-eQTL of the SLC39A8 gene to a module of co-expressed metallothionein genes upregulated more than 20 hours later and used motif analysis to identify zinc-induced activation of the MTF1 transcription factor as a likely mediator of this effect. Conclusions Our analysis provides a rare detailed characterisation of a trans-eQTL effect cascade from a proximal cis effect to the affected signalling pathway, transcription factor, and target genes. This highlights how co-expression analysis combined with functional enrichment analysis can greatly improve the identification and prioritisation of trans-eQTLs when applied to emerging cell-type specific datasets.

15 citations


Cited by
More filters
Journal ArticleDOI
08 Jul 2021-Nature
TL;DR: In this article, the role of human genetics in SARS-CoV-2 infection and COVID-19 severity was investigated and the results of three genome-wide association meta-analyses were presented.
Abstract: The genetic make-up of an individual contributes to the susceptibility and response to viral infection. Although environmental, clinical and social factors have a role in the chance of exposure to SARS-CoV-2 and the severity of COVID-191,2, host genetics may also be important. Identifying host-specific genetic factors may reveal biological mechanisms of therapeutic relevance and clarify causal relationships of modifiable environmental risk factors for SARS-CoV-2 infection and outcomes. We formed a global network of researchers to investigate the role of human genetics in SARS-CoV-2 infection and COVID-19 severity. Here we describe the results of three genome-wide association meta-analyses that consist of up to 49,562 patients with COVID-19 from 46 studies across 19 countries. We report 13 genome-wide significant loci that are associated with SARS-CoV-2 infection or severe manifestations of COVID-19. Several of these loci correspond to previously documented associations to lung or autoimmune and inflammatory diseases3-7. They also represent potentially actionable mechanisms in response to infection. Mendelian randomization analyses support a causal role for smoking and body-mass index for severe COVID-19 although not for type II diabetes. The identification of novel host genetic factors associated with COVID-19 was made possible by the community of human genetics researchers coming together to prioritize the sharing of data, results, resources and analytical frameworks. This working model of international collaboration underscores what is possible for future genetic discoveries in emerging pandemics, or indeed for any complex human disease.

485 citations

Journal ArticleDOI
TL;DR: This paper performed a two-stage genome-wide association study with 111,326 clinically diagnosed/proxy AD cases and 677,663 controls and found 75 risk loci, of which 42 were new at the time of analysis.
Abstract: Abstract Characterization of the genetic landscape of Alzheimer’s disease (AD) and related dementias (ADD) provides a unique opportunity for a better understanding of the associated pathophysiological processes. We performed a two-stage genome-wide association study totaling 111,326 clinically diagnosed/‘proxy’ AD cases and 677,663 controls. We found 75 risk loci, of which 42 were new at the time of analysis. Pathway enrichment analyses confirmed the involvement of amyloid/tau pathways and highlighted microglia implication. Gene prioritization in the new loci identified 31 genes that were suggestive of new genetically associated processes, including the tumor necrosis factor alpha pathway through the linear ubiquitin chain assembly complex. We also built a new genetic risk score associated with the risk of future AD/dementia or progression from mild cognitive impairment to AD/dementia. The improvement in prediction led to a 1.6- to 1.9-fold increase in AD risk from the lowest to the highest decile, in addition to effects of age and the APOE ε4 allele.

403 citations

Journal ArticleDOI
Douglas P Wightman1, Iris E. Jansen1, Jeanne E. Savage1, Alexey A. Shadrin2, Shahram Bahrami3, Shahram Bahrami2, Dominic Holland4, Arvid Rongve5, Sigrid Børte6, Sigrid Børte2, Sigrid Børte3, Bendik S. Winsvold6, Bendik S. Winsvold3, Ole Kristian Drange6, Amy E Martinsen2, Amy E Martinsen3, Amy E Martinsen6, Anne Heidi Skogholt6, Cristen J. Willer7, Geir Bråthen6, Ingunn Bosnes6, Ingunn Bosnes8, Jonas B. Nielsen7, Jonas B. Nielsen6, Jonas B. Nielsen9, Lars G. Fritsche7, Laurent F. Thomas6, Linda M. Pedersen3, Maiken Elvestad Gabrielsen6, Marianne Bakke Johnsen3, Marianne Bakke Johnsen2, Marianne Bakke Johnsen6, Tore Wergeland Meisingset6, Wei Zhou10, Wei Zhou7, Petroula Proitsi11, Angela Hodges11, Richard Dobson, Latha Velayudhan11, Karl Heilbron, Adam Auton, Julia M. Sealock12, Lea K. Davis12, Nancy L. Pedersen13, Chandra A. Reynolds14, Ida K. Karlsson13, Ida K. Karlsson15, Sigurdur H. Magnusson16, Hreinn Stefansson16, Steinunn Thordardottir, Palmi V. Jonsson17, Jon Snaedal, Anna Zettergren18, Ingmar Skoog18, Ingmar Skoog19, Silke Kern18, Silke Kern19, Margda Waern19, Margda Waern18, Henrik Zetterberg, Kaj Blennow18, Kaj Blennow19, Eystein Stordal8, Eystein Stordal6, Kristian Hveem6, John-Anker Zwart3, John-Anker Zwart2, John-Anker Zwart6, Lavinia Athanasiu3, Lavinia Athanasiu2, Per Selnes20, Ingvild Saltvedt6, Sigrid Botne Sando6, Ingun Ulstein3, Srdjan Djurovic5, Srdjan Djurovic3, Tormod Fladby20, Tormod Fladby2, Dag Aarsland21, Dag Aarsland11, Geir Selbæk3, Geir Selbæk2, Stephan Ripke22, Stephan Ripke23, Stephan Ripke10, Kari Stefansson16, Ole A. Andreassen3, Ole A. Andreassen2, Danielle Posthuma24, Danielle Posthuma1 
TL;DR: This paper identified microglia, immune cells and protein catabolism as relevant genes for late-onset Alzheimer's disease, while identifying and prioritizing previously unidentified genes of potential interest.
Abstract: Late-onset Alzheimer's disease is a prevalent age-related polygenic disease that accounts for 50-70% of dementia cases. Currently, only a fraction of the genetic variants underlying Alzheimer's disease have been identified. Here we show that increased sample sizes allowed identification of seven previously unidentified genetic loci contributing to Alzheimer's disease. This study highlights microglia, immune cells and protein catabolism as relevant to late-onset Alzheimer's disease, while identifying and prioritizing previously unidentified genes of potential interest. We anticipate that these results can be included in larger meta-analyses of Alzheimer's disease to identify further genetic variants that contribute to Alzheimer's pathology.

269 citations

Journal ArticleDOI
07 Apr 2021-Nature
TL;DR: In this article, the activity-by-contact (ABC) model was applied to create enhancer-gene maps in 131 human cell types and tissues, and use these maps to interpret the functions of GWAS variants.
Abstract: Genome-wide association studies (GWAS) have identified thousands of noncoding loci that are associated with human diseases and complex traits, each of which could reveal insights into the mechanisms of disease1. Many of the underlying causal variants may affect enhancers2,3, but we lack accurate maps of enhancers and their target genes to interpret such variants. We recently developed the activity-by-contact (ABC) model to predict which enhancers regulate which genes and validated the model using CRISPR perturbations in several cell types4. Here we apply this ABC model to create enhancer–gene maps in 131 human cell types and tissues, and use these maps to interpret the functions of GWAS variants. Across 72 diseases and complex traits, ABC links 5,036 GWAS signals to 2,249 unique genes, including a class of 577 genes that appear to influence multiple phenotypes through variants in enhancers that act in different cell types. In inflammatory bowel disease (IBD), causal variants are enriched in predicted enhancers by more than 20-fold in particular cell types such as dendritic cells, and ABC achieves higher precision than other regulatory methods at connecting noncoding variants to target genes. These variant-to-function maps reveal an enhancer that contains an IBD risk variant and that regulates the expression of PPIF to alter the membrane potential of mitochondria in macrophages. Our study reveals principles of genome regulation, identifies genes that affect IBD and provides a resource and generalizable strategy to connect risk variants of common diseases to their molecular and cellular functions. Mapping enhancer regulation across human cell types and tissues illuminates genome function and provides a resource to connect risk variants for common diseases to their molecular and cellular functions.

233 citations

Journal ArticleDOI
TL;DR: Open Targets Genetics offers tools that enable users to prioritise causal variants and genes at disease-associated loci and access systematic cross-disease and disease-molecular trait colocalization analysis across 92 cell types and tissues including the eQTL Catalogue.
Abstract: Open Targets Genetics (https://genetics.opentargets.org) is an open-access integrative resource that aggregates human GWAS and functional genomics data including gene expression, protein abundance, chromatin interaction and conformation data from a wide range of cell types and tissues to make robust connections between GWAS-associated loci, variants and likely causal genes. This enables systematic identification and prioritisation of likely causal variants and genes across all published trait-associated loci. In this paper, we describe the public resources we aggregate, the technology and analyses we use, and the functionality that the portal offers. Open Targets Genetics can be searched by variant, gene or study/phenotype. It offers tools that enable users to prioritise causal variants and genes at disease-associated loci and access systematic cross-disease and disease-molecular trait colocalization analysis across 92 cell types and tissues including the eQTL Catalogue. Data visualizations such as Manhattan-like plots, regional plots, credible sets overlap between studies and PheWAS plots enable users to explore GWAS signals in depth. The integrated data is made available through the web portal, for bulk download and via a GraphQL API, and the software is open source. Applications of this integrated data include identification of novel targets for drug discovery and drug repurposing.

218 citations