scispace - formally typeset
Search or ask a question
Author

Martijn Vochteloo

Bio: Martijn Vochteloo is an academic researcher from University Medical Center Groningen. The author has contributed to research in topics: Expression quantitative trait loci & Context (archaeology). The author has an hindex of 1, co-authored 1 publications receiving 9 citations. Previous affiliations of Martijn Vochteloo include Hanze University of Applied Sciences.

Papers
More filters
Posted ContentDOI
02 Mar 2021-bioRxiv
TL;DR: In this article, the authors harmonized and integrated 8,727 RNA-seq samples with accompanying genotype data from multiple brain-regions from 14 datasets and performed both cis-and trans-expression quantitative locus (eQTL) mapping.
Abstract: Gaining insight into the downstream consequences of non-coding variants is an essential step towards the identification of therapeutic targets from genome-wide association study (GWAS) findings. Here we have harmonized and integrated 8,727 RNA-seq samples with accompanying genotype data from multiple brain-regions from 14 datasets. This sample size enabled us to perform both cis- and trans-expression quantitative locus (eQTL) mapping. Upon comparing the brain cortex cis-eQTLs (for 12,307 unique genes at FDR We inferred the brain cell type for 1,515 cis-eQTLs by using cell type proportion information. We conducted Mendelian Randomization on 31 brain-related traits using cis-eQTLs as instruments and found 159 significant findings that also passed colocalization. Furthermore, two multiple sclerosis (MS) findings had cell type specific signals, a neuron-specific cis-eQTL for CYP24A1 and a macrophage specific cis-eQTL for CLECL1. To further interpret GWAS hits, we performed trans-eQTL analysis. We identified 2,589 trans-eQTLs (at FDR We also generated a brain-specific gene-coregulation network that we used to predict which genes have brain-specific functions, and to perform a novel network analysis of Alzheimer’s disease (AD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS) and Parkinson’s disease (PD) GWAS data. This resulted in the identification of distinct sets of genes that show significantly enriched co-regulation with genes inside the associated GWAS loci, and which might reflect drivers of these diseases.

32 citations

Journal ArticleDOI
TL;DR: In this paper , a detailed dissection of this using single-cell RNA-sequencing of 1.3M peripheral blood mononuclear cells from 120 individuals, longitudinally exposed to three different pathogens was provided.
Abstract: Abstract The host’s gene expression and gene regulatory response to pathogen exposure can be influenced by a combination of the host’s genetic background, the type of and exposure time to pathogens. Here we provide a detailed dissection of this using single-cell RNA-sequencing of 1.3M peripheral blood mononuclear cells from 120 individuals, longitudinally exposed to three different pathogens. These analyses indicate that cell-type-specificity is a more prominent factor than pathogen-specificity regarding contexts that affect how genetics influences gene expression (i.e., eQTL) and co-expression (i.e., co-expression QTL). In monocytes, the strongest responder to pathogen stimulations, 71.4% of the genetic variants whose effect on gene expression is influenced by pathogen exposure (i.e., response QTL) also affect the co-expression between genes. This indicates widespread, context-specific changes in gene expression level and its regulation that are driven by genetics. Pathway analysis on the CLEC12A gene that exemplifies cell-type-, exposure-time- and genetic-background-dependent co-expression interactions, shows enrichment of the interferon (IFN) pathway specifically at 3-h post-exposure in monocytes. Similar genetic background-dependent association between IFN activity and CLEC12A co-expression patterns is confirmed in systemic lupus erythematosus by in silico analysis, which implies that CLEC12A might be an IFN-regulated gene. Altogether, this study highlights the importance of context for gaining a better understanding of the mechanisms of gene regulation in health and disease.

18 citations

Posted ContentDOI
30 Jul 2022-bioRxiv
TL;DR: PICALO is robust, works well with heterogeneous datasets, yields reproducible interaction components, and identifies eQTL interactions and contexts that would have been missed when using cell counts or expression based principal components.
Abstract: Expression quantitative trait loci (eQTL) can reveal the regulatory mechanisms of trait associated variants. eQTLs are highly cell-type and context-specific, but often these contexts are unknown or not measured. Here, we introduce PICALO (Principal Interaction Component Analysis through Likelihood Optimization), an unbiased method to identify known and hidden contexts that influence eQTLs. PICALO uses expectation maximization to identify latent components, referred to as Principal Interaction Components (PIC), that interact with genotypes to maximize explained eQTL effect-sizes. We applied PICALO to bulk RNA-seq eQTL datasets in blood (n=2,932) and brain (n=2,440). We identify 31 PICs in blood, interacting with 4,169 (32%) unique cis-eQTLs (BH-FDR≤0.05). In brain, we identified 21 PICs, interacting with 4,058 (39%) unique cis-eQTLs (BH-FDR≤0.05). These PICs are associated with RNA quality, cell type composition or environmental influences. Furthermore, PICs clearly disentangle distinct eQTL contexts, for example technical from non-technical factors. Combined, 3,065 unique genes showed a cis-eQTL effect that is dependent on a cell type or other non-technical context, emphasizing the value of methods like PICALO. PICALO is robust, works well with heterogeneous datasets, yields reproducible interaction components, and identifies eQTL interactions and contexts that would have been missed when using cell counts or expression based principal components. Since PICALO allows for the identification of many context-dependent eQTLs without any prior knowledge of such contexts, this method can help to reveal and quantify the influence of previously unknown environmental factors that play a role in common diseases.

1 citations


Cited by
More filters
Posted ContentDOI
18 Mar 2021-medRxiv
TL;DR: All ALS associated signals combined reveal a role for perturbations in vesicle mediated transport and autophagy, and provide evidence for cell-autonomous disease initiation in glutamatergic neurons.
Abstract: Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with a life-time risk of 1 in 350 people and an unmet need for disease-modifying therapies. We conducted a cross-ancestry GWAS in ALS including 29,612 ALS patients and 122,656 controls which identified 15 risk loci in ALS. When combined with 8,953 whole-genome sequenced individuals (6,538 ALS patients, 2,415 controls) and the largest cortex-derived eQTL dataset (MetaBrain), analyses revealed locus-specific genetic architectures in which we prioritized genes either through rare variants, repeat expansions or regulatory effects. ALS associated risk loci were shared with multiple traits within the neurodegenerative spectrum, but with distinct enrichment patterns across brain regions and cell-types. Across environmental and life-style risk factors obtained from literature, Mendelian randomization analyses indicated a causal role for high cholesterol levels. All ALS associated signals combined reveal a role for perturbations in vesicle mediated transport and autophagy, and provide evidence for cell-autonomous disease initiation in glutamatergic neurons.

110 citations

Journal ArticleDOI
TL;DR: This article performed an eQTL analysis using single-nuclei RNA sequencing from 192 individuals in eight brain cell types derived from the prefrontal cortex, temporal cortex and white matter, and identified 7,607 eGenes, a substantial fraction (46%, 3,537/7,607) of which show cell-type-specific effects with strongest effects in microglia.
Abstract: To date, most expression quantitative trait loci (eQTL) studies, which investigate how genetic variants contribute to gene expression, have been performed in heterogeneous brain tissues rather than specific cell types. In this study, we performed an eQTL analysis using single-nuclei RNA sequencing from 192 individuals in eight brain cell types derived from the prefrontal cortex, temporal cortex and white matter. We identified 7,607 eGenes, a substantial fraction (46%, 3,537/7,607) of which show cell-type-specific effects, with strongest effects in microglia. Cell-type-level eQTLs affected more constrained genes and had larger effect sizes than tissue-level eQTLs. Integration of brain cell type eQTLs with genome-wide association studies (GWAS) revealed novel relationships between expression and disease risk for neuropsychiatric and neurodegenerative diseases. For most GWAS loci, a single gene co-localized in a single cell type, providing new clues into disease etiology. Our findings demonstrate substantial contrast in genetic regulation of gene expression among brain cell types and reveal potential mechanisms by which disease risk genes influence brain disorders. Bryois et al. mapped genetic variants regulating gene expression in eight major brain cell types. They found a large number of cell-type-specific genetic effects and leveraged their results to identify novel putative risk genes for brain disorders.

48 citations

Journal ArticleDOI
TL;DR: This article performed an eQTL analysis using single-nuclei RNA sequencing from 192 individuals in eight brain cell types derived from the prefrontal cortex, temporal cortex and white matter, and identified 7,607 eGenes, a substantial fraction (46%, 3,537/7,607) of which show cell-type-specific effects with strongest effects in microglia.
Abstract: To date, most expression quantitative trait loci (eQTL) studies, which investigate how genetic variants contribute to gene expression, have been performed in heterogeneous brain tissues rather than specific cell types. In this study, we performed an eQTL analysis using single-nuclei RNA sequencing from 192 individuals in eight brain cell types derived from the prefrontal cortex, temporal cortex and white matter. We identified 7,607 eGenes, a substantial fraction (46%, 3,537/7,607) of which show cell-type-specific effects, with strongest effects in microglia. Cell-type-level eQTLs affected more constrained genes and had larger effect sizes than tissue-level eQTLs. Integration of brain cell type eQTLs with genome-wide association studies (GWAS) revealed novel relationships between expression and disease risk for neuropsychiatric and neurodegenerative diseases. For most GWAS loci, a single gene co-localized in a single cell type, providing new clues into disease etiology. Our findings demonstrate substantial contrast in genetic regulation of gene expression among brain cell types and reveal potential mechanisms by which disease risk genes influence brain disorders. Bryois et al. mapped genetic variants regulating gene expression in eight major brain cell types. They found a large number of cell-type-specific genetic effects and leveraged their results to identify novel putative risk genes for brain disorders.

46 citations

Journal ArticleDOI
TL;DR: In this paper, the authors map e/sQTLs and allele-specific expression in cultured cells representing two major developmental stages, primary human neural progenitors and their sorted neuronal progeny, identifying numerous loci not detected in either bulk developing cortical wall or adult cortex.
Abstract: Interpretation of the function of non-coding risk loci for neuropsychiatric disorders and brain-relevant traits via gene expression and alternative splicing quantitative trait locus (e/sQTL) analyses is generally performed in bulk post-mortem adult tissue. However, genetic risk loci are enriched in regulatory elements active during neocortical differentiation, and regulatory effects of risk variants may be masked by heterogeneity in bulk tissue. Here, we map e/sQTLs, and allele-specific expression in cultured cells representing two major developmental stages, primary human neural progenitors (n = 85) and their sorted neuronal progeny (n = 74), identifying numerous loci not detected in either bulk developing cortical wall or adult cortex. Using colocalization and genetic imputation via transcriptome-wide association, we uncover cell-type-specific regulatory mechanisms underlying risk for brain-relevant traits that are active during neocortical differentiation. Specifically, we identified a progenitor-specific eQTL for CENPW co-localized with common variant associations for cortical surface area and educational attainment.

27 citations

Posted ContentDOI
11 Jun 2021-medRxiv
TL;DR: In this article, the authors identified 139 genes in which protein-coding variants cause severe or familial forms of nine human traits, and then computed the association between common complex forms of the same traits and noncoding variation.
Abstract: The genetic basis of most complex traits is highly polygenic and dominated by non-coding alleles, and it is widely assumed that such alleles exert small regulatory effects on the expression of cis-linked genes. However, despite availability of expansive gene expression and epigenomic data sets, few variant-to-gene links have emerged. We identified 139 genes in which protein-coding variants cause severe or familial forms of nine human traits. We then computed the association between common complex forms of the same traits and non-coding variation, revealing that most such traits are also associated with non-coding variation in the vicinity of the same genes. However, we found colocalization evidence--the same variant influencing both the physiological trait and gene expression--for only 7% of genes, and transcriptome-wide association evidence with correct direction of effect for only 4% of genes, despite an abundance of eQTLs in most loci. Fine mapping variants to regulatory elements and assigning these to genes by linear distance similarly failed to implicate most genes in complex traits. These results contradict the hypothesis that most complex trait-associated variants coincide with currently ascertained expression quantitative trait loci. The field must confront this deficit, and pursue the "missing regulation."

24 citations