Andrew A. Brown’s research while affiliated with University of Dundee and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (101)


Fig. 2 | Abundant pleiotropy identified across molecular phenotypes. A, B Distribution of the P values for SNP in significant (FDR < 0.05) cis-eQTL (A) as pQTLs and for SNPs in significant (FDR < 0.05) cis-pQTLs (B) as eQTLs. Most pairs showed consistent direction of effect. Data shown are the -log10 P values of the linear regressions between gene expression or protein abundances and SNPs. C Local network of QTLs for rs34097845, a SNP significantly associated with both the expression of MPO (P value = 1.7e-10, blue) and its protein (MPO, P value = 2.08e-14, orange) with a consistent direction of effect (ß expression = −0.87, ß protein = −0.40). D We identified 101 trios of expression-SNP-proteins, of which 48 involved a protein and its coding gene, while 53 involved the expression of a nearby gene different that the coding gene for the protein.
Fig. 3 | Tissue specific genetic regulation partially explains the lack of shared associations between gene expression and proteins. A Using n = 3027 biologically independent samples, we detected a cis-pQTL for CCL16 in whole blood (P value = 9.5e-243, n = 3029). The GTEx consortium reported a cis-eQTL, with the same SNP (rs10445391) affecting the expression of the gene in liver (n = 208). Violin plots show the median and first and last quartiles as defined by ggplot geom_violin function. Partially created with BioRender.com B Between 91.2% (pancreatic islets) and 71.6% (esophagus mucosa) of cis-eQTLs discovered by GTEx v8 were also active in whole blood DIRECT datasets (n = 3029) as shown by the π 1 values (y-axis). The number of P values per tissue used to calculate the π 1 estimates ranged from 334 in kidney to 14,920 in thyroid. C Comparison of the effect size of cis-eQTLs from pancreatic islets (InsPIRE) and whole blood (DIRECT). A total of 486 eQTLs were not significant in blood (P value > 0.035, orange color) but significant in pancreatic islets (n = 420) and 294 had opposite direction of effect (N = 2691). Data shown are the ß values (effect) resulting from the linear regressions between gene expression and SNPs identifying eQTLs in both studies. D Comparison of the π 1 enrichment analysis between an earlier version of GTEx (v6p) and a larger later version (v8). eQTLs from DIRECT blood detected in GTEx v8 decreased compared to v6p independently of the change in sample size across versions (Supplementary Fig. 5H). E Degree of sharing of pQTLs detected as eQTLs in GTEx v8 tissues. Up to 66.6% of plasma cis-pQTLs were also active as DIRECT whole blood cis-eQTLs. The number of overlapping QTLs across tissues oscillates between 13 (kidney) and 311 (Thyroid). F Degree of sharing of metabo-QTLs acting as cis-eQTLs in GTEx v8. Up to 16.88% (testis) of the metabo-QTLs detected in blood were active eQTLs in other tissues, with many tissues sharing no associations with metabolites-QTLs. The number of P values used to calculate π 1 values per tissues ranged from 4298 in whole blood to 6575 in testis.
Fig. 5 | QTL integration identifies regulatory networks associated to GWAS variants. A Of the GWAS signal overlapping SNPs in the full network (Supplementary Fig. 9), the largest number were cis-eSNPs followed by trans-eSNPs (Number). However, when considering the number of significant QTLs evaluated (Percentage), we observed that more metabo-SNPs were also reported GWAS followed by trans-eSNPs. The barplots show numbers and percentages of SNPs involved in QTLs that were also reported as lead GWAS by the GWAS catalogue (Supplementary Data 13). B Network of associations for the resistin gene (RETN). The RETN gene and its protein (orange node) have been associated with low density lipoproteins (LDL) levels. The regulatory network associated with the gene included GWAS variants (purple nodes) associated to RETN abundance (rs1477341); cardiovascular diseases and cholesterol levels (rs13284665); platelet counts
Genetic analysis of blood molecular phenotypes reveals common properties in the regulatory networks affecting complex traits
  • Article
  • Full-text available

August 2023

·

287 Reads

·

10 Citations

Andrew A. Brown

·

Juan J. Fernandez-Tajes

·

·

[...]

·

Ana Viñuela

We evaluate the shared genetic regulation of mRNA molecules, proteins and metabolites derived from whole blood from 3029 human donors. We find abundant allelic heterogeneity, where multiple variants regulate a particular molecular phenotype, and pleiotropy, where a single variant associates with multiple molecular phenotypes over multiple genomic regions. The highest proportion of share genetic regulation is detected between gene expression and proteins (66.6%), with a further median shared genetic associations across 49 different tissues of 78.3% and 62.4% between plasma proteins and gene expression. We represent the genetic and molecular associations in networks including 2828 known GWAS variants, showing that GWAS variants are more often connected to gene expression in trans than other molecular phenotypes in the network. Our work provides a roadmap to understanding molecular networks and deriving the underlying mechanism of action of GWAS variants using different molecular phenotypes in an accessible tissue.

Download

Figure 4. Forest plots showing random effects of XBP1 eQTL variant on HbA1c (mmol/mol).
Allelefrequencies across ancestries and effects of lead eQTL variant on XBP1 expression in pancreatic beta-cells, T2DM risk.
XBP1 expression in pancreatic islet cells is associated with poor glycaemic control across ancestries especially in young non-obese onset diabetes

May 2023

·

67 Reads

Objective Certain ethnicities such as South Asians and East Asians have higher rates of type 2 diabetes mellitus, in part, driven by insulin deficiency. Insulin deficiency can be due to beta-cell insufficiency, low beta-cell mass, or early cell death. Transcription factor XBP1 maintains beta-cell function and prevents early cell death by mitigating cellular endoplasmic reticulum stress. We examine the role of XBP1 expression in maintaining glucose homeostasis, glycaemic control, and response to diabetes therapeutics. Research Design and Methods Colocalisation analyses were used to determine if expression of XBP1 in pancreatic islets and type 2 diabetes shared common causal genetic variants. We identify a lead eQTL variant associated exclusively with XBP1 expression and examine its association HOMA-B and stimulated glucose in cohorts of newly diagnosed Asian Indians from Dr. Mohans Diabetes Specialities Centre, India (DMDSC) and the Telemedicine Project for Screening diabetes and complications in rural Tamil Nadu (TREND). We then examine longer term glycaemic control using HbA1c in Asian Indian cohorts, the Tayside Diabetes Study (TDS) of white European ancestry in Scoltand, and the Genes & Health (G&H) study of British South Asian Bangladeshi and Pakistani ancestry. Finally, we assess the effect of eQTL variant on drugs designed to improve insulin secretion (sulphonylureas and GLP1-RA). Results Variants affecting XBP1 expression in the pancreatic islets colocalised with variants associated with T2DM risk in East Asians but not in white Europeans. Lower expression of XBP1 was associated with higher risk of T2DM. rs7287124 was the lead eQTL variant and had a higher risk allele frequency in East (65%) and South Asians (50%) compared to white Europeans (25%). In 470 South Asian Indians, the variant was associated with lower beta-cell function and higher stimulated glucose (Beta log HOMAB =-0.14, P=5x10-3). Trans-ancestry meta-analysed effect of the variant in 179,668 individuals was 4.32 mmol/mol (95%CI:2.60,6.04, P=8x10-7) per allele. In 477 individuals with young onset diabetes with non-obese BMI, the per allele effect was 6.41 mmol/mol (95%CI:3.04, 9.79, P =2x10-4). Variant carriers showed impaired response to sulphonylureas. Conclusion XBP1 expression is a novel target for T2DM with particular value for individuals of under-researched ancestries who have greater risk of young, non-obese onset diabetes. The effect of XBP1 eQTL variant was found to be comparable with or greater that the effect of novel glucose-lowering therapies.


Multiallelic Copy Number Variation in ORM1 is Associated with Plasma Cell-Free DNA Levels as an Intermediate Phenotype for Venous Thromboembolism

January 2023

·

25 Reads

Thrombosis and Haemostasis

Venous thromboembolism (VTE) is a common disease with high heritability. However, only a small portion of the genetic variance of VTE can be explained by known genetic risk factors. Neutrophil extracellular traps (NETs) have been associated with prothrombotic activity. Therefore, the genetic basis of NETs could reveal novel risk factors for VTE. A recent genome-wide association study of plasma cell-free DNA (cfDNA) levels in the Genetic Analysis of Idiopathic Thrombophilia 2 (GAIT-2) Project showed a significant associated locus near ORM1. We aimed to further explore this candidate region by next-generation sequencing, copy number variation (CNV) quantification, and expression analysis using an extreme phenotype sampling design involving 80 individuals from the GAIT-2 Project. The RETROVE study with 400 VTE cases and 400 controls was used to replicate the results. A total of 105 genetic variants and a multiallelic CNV (mCNV) spanning ORM1 were identified in GAIT-2. Of these, 17 independent common variants, a region of 22 rare variants, and the mCNV were significantly associated with cfDNA levels. In addition, eight of these common variants and the mCNV influenced ORM1 expression. The association of the mCNV and cfDNA levels was replicated in RETROVE (p-value = 1.19 × 10−6). Additional associations between the mCNV and thrombin generation parameters were identified. Our results reveal that increased mCNV dosages in ORM1 decreased gene expression and upregulated cfDNA levels. Therefore, the mCNV in ORM1 appears to be a novel marker for cfDNA levels, which could contribute to VTE risk.


Fig. 3 LocusCompare plots of cis-eQTLs for ORM1 and ORM2 genes. Visualization plots displaying colocalization of GWAS and cis-eQTLs for ORM1 (A) and ORM2 (B) in the GAIT-2 study. Only cis-eQTL variants reaching the Bonferroni threshold of significance (0.05/28,133) are represented in LocusZoom plots showing eQTLs. The labeled SNP (purple dot) is the lead SNP in the GWAS that is a significant eQTL variant for both ORM1 and ORM2 genes. Other SNPs are colored according to their LD r 2 with the lead SNP, as presented in the color key. eQTL, expression quantitative trait loci; GWAS, genome-wide association study; LD, linkage disequilibrium; SNP, single nucleotide polymorphism.
Integrated GWAS and Gene Expression Suggest ORM1 as a Potential Regulator of Plasma Levels of Cell-Free DNA and Thrombosis Risk

March 2022

·

79 Reads

·

10 Citations

Thrombosis and Haemostasis

Plasma cell-free DNA (cfDNA) is a surrogate marker of neutrophil extracellular traps (NETs) that contribute to immunothrombosis. There is growing interest about the mechanisms underlying NET formation and elevated cfDNA, but little is known about the factors involved. We aimed to identify genes involved in the regulation of cfDNA levels using data from the Genetic Analysis of Idiopathic Thrombophilia (GAIT-2) Project. Imputed genotypes, whole blood RNA-Seq data, and plasma cfDNA quantification were available for 935 of the GAIT-2 participants from 35 families with idiopathic thrombophilia. We performed heritability and GWAS analysis for cfDNA. The heritability of cfDNA was 0.26 (p = 3.7 × 10−6), while the GWAS identified a significant association (rs1687391, p = 3.55 × 10−10) near the ORM1 gene, on chromosome 9. An eQTL (expression quantitative trait loci) analysis revealed a significant association between the lead GWAS variant and the expression of ORM1 in whole blood (p = 6.14 × 10−9). Additionally, ORM1 expression correlated with levels of cfDNA (p = 4.38 × 10−4). Finally, genetic correlation analysis between cfDNA and thrombosis identified a suggestive association (ρ g = 0.43, p = 0.089). All in all, we show evidence of the role of ORM1 in regulating cfDNA levels in plasma, which might contribute to the susceptibility to thrombosis through mechanisms of immunothrombosis.


Genetic analysis of blood molecular phenotypes reveals regulatory networks affecting complex traits: a DIRECT study

March 2021

·

266 Reads

·

2 Citations

Genetic variants identified by genome-wide association studies can contribute to disease risk by altering the production and abundance of mRNA, proteins and other molecules. However, the interplay between molecular intermediaries that define the pathway from genetic variation to disease is not well understood. Here, we evaluated the shared genetic regulation of mRNA molecules, proteins and metabolites derived from whole blood from 3,029 human donors. We find abundant allelic heterogeneity, where multiple variants regulate a particular molecular phenotype, and pleiotropy, where a single variant was associated with multiple molecular phenotypes over multiple genomic regions. We find varying proportions of shared genetic regulation across phenotypes, highest between expression and proteins (66.6%). We were able to recapitulate a substantial proportion of gene expression genetic regulation in a diverse set of 44 tissues, with a median of 88% shared associations for blood expression and 22.3% for plasma proteins. Finally, the genetic and molecular associations were represented in networks including 2,828 known GWAS variants. One sub-network shows the trans relationship between rs149007767 and RTEN, and identifies GRB10 and IKZF1 as candidates mediating genes. Our work provides a roadmap to understanding molecular networks and deriving the underlying mechanism of action of GWAS variants across different molecular phenotypes.


Islet eQTLs and their activity in other tissues
a Proportion of islet eQTLs active in GTEx tissues using p-value enrichment analysis (π1 estimate for replication). b Comparison between eQTLs discovered in islets and their p-values in β-cells (top figure, N = 26) and whole pancreas tissue from GTEX (bottom figure, n = 149). The axes show the −log10p-value of the eQTL associations adjusted by the eQTL direction (positive or negative) of effect with respect to the reference allele. Source data are provided as a Source data file.
Integration of islet eQTL with epigenomic information
a Distribution of absolute effect sizes for islet eQTLs in each islet chromatin state. b Distribution of absolute effect sizes for islet eQTL in ATAC-seq peaks in three islet chromatin states. eQTL SNPs in ATAC-seq peaks in stretch enhancers have significantly lower effect sizes than SNPs in ATAC-seq peaks in active TSS and typical enhancer states. P-values were obtained from a Wilcoxon rank-sum test. c Fold enrichment for transcription factor footprint motifs to overlap low vs high effect size islet eQTL SNPs. d TF footprint motif directionality fraction vs fold enrichment for the TF footprint motif to overlap islet eQTLs. TF footprint motif directionality fraction is calculated as the fraction of eQTL SNPs overlapping a TF footprint motif, where the base preferred in the motif is associated with increased expression of the eQTL eGene. Significance of skew of this fraction from a null expectation of 0.5 was calculated using the binomial test. Source data are provided as a Source data file.
GWAS SNPs in islet eQTLs
a Enrichment of eQTL effect sizes in different GTEx tissues at T2D (all) and glycemic GWAS-associated variants. Numbers within square brackets denote the number of variants implicated for each the trait. Also shown a subset of T2D GWAS associated with reduced insulin secretion or islet β-cell dysfunction (T2D (BC)) and type 1 diabetes (T1D)-associated signals. b LocusCompare plot for the T2D GWAS p-values in the TCF7L2 locus. Plots on the right −log10 p-values for the GWAS (top) and for the the eQTL for TCF7L2, highlighting in both the GWAS lead SNP in the cis window tested for eQTLs. On the left it shows a comparison of the p-values in both analyses. Source data are provided as a Source data file.
Functional assessment of DGKB eQTL locus
a We show the two of the three independent islet eQTL signals that colocalize with identified independent GWAS variants near the DGKB gene locus (lead SNP rs17168486 referred at as the 5′ signal and lead SNP rs10231021 referred to as the 3′ signal). These signals colocalize with two independent T2D GWAS signals shown in b, where rs17168486 is referred to as the 5′ signal and lead SNP rs2191349 referred to as the 3′ signal. LD information was not available for SNPs denoted by (×). The third GWAS variant and the third eSNP are not shown as both are located outside this region and in opposite location with respect to DGKB, showing no evidence of colocalization. c Normalized DGKB gene expression levels relative to the T2D-risk-allele dosage at the 3′ islet eQTL for DGKB lead SNP rs10231021. eQTL p-value adjusted to the beta distribution is shown. d Genome browser view of the region highlighted in purple in a and b that contains the 3′ DGKB eQTL and T2D GWAS signals. Two regulatory elements (element 1 highlighted in green, element 2 highlighted in blue) overlapping ATAC-seq peaks in islet β-cells (islet single nuclei ATAC-seq⁴⁹) and bulk islets (islet track represents one islet sample from Varshney et al.¹⁵) were cloned into a luciferase reporter assay construct for functional validation. All ATAC-seq tracks are normalized to 10 M reads and scaled from 0–15. e Log 2 luciferase assay activities (normalized to empty vector) are shown for in rat (832/13), mouse (MIN6), and human (EndoC-βH1) β-cell lines for the element 2 (cloned in the forward orientation), highlighted in blue in d. The risk haplotype shows significantly higher (p < 0.05) activity than the non-risk haplotype in 832/13 and MIN6, consistent with the eQTL direction shown in c. P-values were determined using unpaired two-sided t-tests. f EMSA for probes with risk and non-risk alleles at the four SNPs overlapping the regulatory element validated in e, using nuclear extract from MIN6 cells. Filled arrows, allele-specific binding; open arrows, non-allele-specific binding of proteins to probes. Source data are provided as a Source data file.
Genetic variant effects on gene expression in human pancreatic islets and their implications for T2D

September 2020

·

211 Reads

·

108 Citations

Most signals detected by genome-wide association studies map to non-coding sequence and their tissue-specific effects influence transcriptional regulation. However, key tissues and cell-types required for functional inference are absent from large-scale resources. Here we explore the relationship between genetic variants influencing predisposition to type 2 diabetes (T2D) and related glycemic traits, and human pancreatic islet transcription using data from 420 donors. We find: (a) 7741 cis-eQTLs in islets with a replication rate across 44 GTEx tissues between 40% and 73%; (b) marked overlap between islet cis-eQTL signals and active regulatory sequences in islets, with reduced eQTL effect size observed in the stretch enhancers most strongly implicated in GWAS signal location; (c) enrichment of islet cis-eQTL signals with T2D risk variants identified in genome-wide association studies; and (d) colocalization between 47 islet cis-eQTLs and variants influencing T2D or glycemic traits, including DGKB and TCF7L2. Our findings illustrate the advantages of performing functional and regulatory studies in disease relevant tissues. Mechanistic inference following GWAS is hampered by the lack of tissue-specific transcriptomic resources. Here the authors combine genetic variants predisposing to type 2 diabetes with human pancreatic islet RNA-seq data. They identify 7741 islet expression quantitative trait loci (eQTLs), providing a resource for functional interpretation of association signals mapping to non-coding sequence.


Figure 1 | Islet eQTL discovery. A) Proportion of islet eQTLs active in GTEx tissues using P-value enrichment analysis (π 1 estimate for replication). B) Comparison between eQTLs discovered in islets and their pvalues in beta-cells (top figure, N=26) and whole pancreas tissue from GTEX (bottom figure, n=149). The axes show thelog10 Pvalue of the eQTL associations adjusted by the eQTL direction of effect with respect to the reference allele.
Influence of genetic variants on gene expression in human pancreatic islets – implications for type 2 diabetes

May 2019

·

169 Reads

·

13 Citations

Most 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.



S1 Fig

September 2018

·

25 Reads

Enrichment plots for CommonMind, GTEx, GTEx/CommonMind consensus brain and non-CNS eQTLs. The top panels contain Q-Q and fold enrichment plots for the CommonMind, GTEx brain and TwinsUK non-CNS eQTLs. The bottom panels contain relative fold enrichment plots for CommonMind and GTEx and for GTEx/CommonMind consensus brain eQTLs compared to non-CNS eQTL variants. (PDF)


Schizophrenia association enrichment in eQTLs
Q-Q and fold enrichment plots for adipose, epidermal, LCL and whole blood eQTLs. The baseline is determined by respectively matched control SNP sets. The fold enrichment is displayed in logarithmic scale.
Schizophrenia association enrichment of eQTLs with different Roadmap functional annotations
Chi-squared general linear model coefficients for eQTLs of different tissues (adipose, epidermal, lymphoblastoid cell lines (LCL), whole blood) and location (proximal, distal) affiliated to different Roadmap functional elements. “All” stands for all eQTLs (* p < 0.05, ** p < 0.001).
Relationship between polygenicity and eQTL association enrichment across different GWASes
Differences (Mann-Whitney test p-values) in association p-values between eQTLs and control variants of various types as functions of the estimated proportions of non-null associations. The GWAS names or acronyms are color-coded to represent different categories (azure = anthropometric [height]; red = cardiovascular, systolic blood pressure [SBP]; green = immune, rheumatoid arthritis [RA]; gold = metabolic, body mass index [BMI], type-II diabetes [T2D]; black = schizophrenia) and their sizes are proportional to the respective chi-squared linear model coefficients (* p < 0.05, ** p < 0.001).
Enrichment statistics and general linear model coefficients for squared schizophrenia association z-scores differences between adipose tissue, epidermal tissue, lymphoblastoid cell lines (LCL) and whole blood eQTLs, and matching control variants
Cross-tissue eQTLs in the loci with genome-wide significant association with schizophrenia
Cross-tissue eQTL enrichment of associations in schizophrenia

September 2018

·

80 Reads

·

5 Citations

The genome-wide association study of the Psychiatric Genomics Consortium identified over one hundred schizophrenia susceptibility loci. The number of non-coding variants discovered suggests that gene regulation could mediate the effect of these variants on disease. Expression quantitative trait loci (eQTLs) contribute to variation in levels of mRNA. Given the co-occurrence of schizophrenia and several traits not involving the central nervous system (CNS), we investigated the enrichment of schizophrenia associations among eQTLs for four non-CNS tissues: adipose tissue, epidermal tissue, lymphoblastoid cells and blood. Significant enrichment was seen in eQTLs of all tissues: adipose (β = 0.18, p = 8.8 × 10⁻⁰⁶), epidermal (β = 0.12, p = 3.1 × 10⁻⁰⁴), lymphoblastoid (β = 0.19, p = 6.2 × 10⁻⁰⁸) and blood (β = 0.19, p = 6.4 × 10⁻⁰⁶). For comparison, we looked for enrichment of association with traits of known relevance to one or more of these tissues (body mass index, height, rheumatoid arthritis, systolic blood pressure and type-II diabetes) and found that schizophrenia enrichment was of similar scale to that observed when studying diseases in the context of a more likely causal tissue. To further investigate tissue specificity, we looked for differential enrichment of eQTLs with relevant Roadmap affiliation (enhancers and promoters) and varying distance from the transcription start site. Neither factor significantly contributed to the enrichment, suggesting that this is equally distributed in tissue-specific and cross-tissue regulatory elements. Our analyses suggest that functional correlates of schizophrenia risk are prevalent in non-CNS tissues. This could be because of pleiotropy or the effectiveness of variants affecting expression in different contexts. This suggests the utility of large, single-tissue eQTL experiments to increase eQTL discovery power in the study of schizophrenia, in addition to smaller, multiple-tissue approaches. Our results conform to the notion that schizophrenia is a systemic disorder involving many tissues.


Citations (42)


... After stringent quality control (see ESM Methods), we identified 132 (ESM Table 1) and 779 (ESM Table 2) metabolites from targeted and untargeted metabolomics measurements, respectively, that were profiled for 3000 samples (ESM Table 3) [28]. Baseline characteristics (Table 1) revealed that there were significant differences in BMI, fasting variables and health status observed between NGR, IGR and type 2 diabetes groups. ...

Reference:

Role of human plasma metabolites in prediabetes and type 2 diabetes from the IMI-DIRECT study
Genetic analysis of blood molecular phenotypes reveals common properties in the regulatory networks affecting complex traits

... S21 Table eQTLs and matched control variants census in the data set used. The eQTLs and control variants from [1] were projected onto templates of ∼2.5 million variants (∼9 million variants for analyses involving brain eQTLs) with known pairwise LD. ...

Reference:

S21 Table
Quantifying the degree of sharing of genetic and non-genetic causes of gene expression variability across four tissues

... The rs150611042 in the promoter of ORM1 has been reported to influence the interindividual capacity to generate thrombin [57]. Additionally, Lopez et al. pointed out that immunothrombosis as a mechanism by which ORM1 could contribute to the susceptibility of thrombosis [58]. It has been reported that the 5HT2a receptor is increased in platelets of patients with chronic migraine, thereby mediating the activation of phospholipase enzymes to promote platelet activation and thrombosis [59]. ...

Integrated GWAS and Gene Expression Suggest ORM1 as a Potential Regulator of Plasma Levels of Cell-Free DNA and Thrombosis Risk

Thrombosis and Haemostasis

... Many of these relevant tissues are difficult to collect pre-mortem, with consequences for our ability to discover the genetic and molecular processes involved in the development of T2D. On the other hand, gene expression studies with thousands of samples have shown that the expression of most genes is affected by multiple eQTLs and many of these effects are shared across multiple tissues 12,13 . Therefore, it is possible that larger molecular studies using samples from tissues that are easy to collect, such as blood, could identify disease relevant loci through these shared signals. ...

Genetic analysis of blood molecular phenotypes reveals regulatory networks affecting complex traits: a DIRECT study

... 5) The entire set of GWAS, with data from 7,051 post-mortem samples representing 44 tissues and 449 individuals was queried at the GTEx portal [15,36] in order to identify cis-eQTL. Particular to the GTEx data, we first queried the 716 SNPs not mapping in or near genes to retrieve eQTL data in any tissue. ...

Distant regulatory effects of genetic variation in multiple human tissues

... To understand the immediate consequences and function of non-coding variation, molecular studies have assayed gene expression in multiple human tissues and linked genetic variation to changes in gene expression. These studies provide a path for discovering disease causing genes and the tissues in which they are active by connecting GWAS loci to genes in a particular tissue [4][5][6] . Transcriptome wide association methods are a type of method using reference expression datasets to build predictive models of genetically regulated gene expression. ...

Genetic variant effects on gene expression in human pancreatic islets and their implications for T2D

... The associations were run with the FastQTL package. 20 The observed nominal p values were calculated by correlating the genotype and link quantifications, which were Gaussian transformed. Subsequently, we ran permutations for each link separately to assign empirical p values to each link. ...

Fast and efficient QTL mapper for thousands of molecular phenotypes

... For example, given that regulatory variations play critical roles in many human diseases (47), understanding how genetic variation contributes to increasing gene expression variability will facilitate the identification of disease-related variants. This is especially true when gene expression heterogeneity characterizes traits or diseases such as aging (48)(49)(50) and cancer (51). For many diseases that display a high degree of phenotypic heterogeneity among patients, we may consider that the increased phenotypic variability is due to variability-controlling mutations (such as evSNPs). ...

Age-dependent changes in mean and variance of gene expression across tissues in a twin cohort

... BANs were defined as genes connected in our Bayesian networks with more genes in the "known bone gene" list than would be expected by chance [29][30][31][32] However, a gene being a BAN is likely not strong evidence, by itself, that a particular gene is causal for a BMD GWAS association. Therefore, to provide additional evidence connecting BMD-associated variants to the regulation of BANs, we identified local eQTL for each BAN homolog in 48 human non-bone samples using the Genotype-Tissue Expression (GTEx) project 10,33,34 . Our rationale for using GTEx was that while these data do not include information on bone tissues or bone cells, a 4 5 6 7 8 9 10 1112 13 1415 16 17 18 Fig. 2 Characterization of the experimental Diversity Outbred cohort. ...

Local genetic effects on gene expression across 44 human tissues

... Some patients with diabetes can be misdiagnosed. Not all patients with diabetes are type 1 or type 2. There are also patients with diabetes caused by genetic mutation, all over the world [9][10][11]. The vast majority of these people are treated unnecessarily using insulin rather than low-dose medication. ...

Influence of genetic variants on gene expression in human pancreatic islets – implications for type 2 diabetes