Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 167(3): 643-656.e17, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27768888

ABSTRACT

Humans differ in the outcome that follows exposure to life-threatening pathogens, yet the extent of population differences in immune responses and their genetic and evolutionary determinants remain undefined. Here, we characterized, using RNA sequencing, the transcriptional response of primary monocytes from Africans and Europeans to bacterial and viral stimuli-ligands activating Toll-like receptor pathways (TLR1/2, TLR4, and TLR7/8) and influenza virus-and mapped expression quantitative trait loci (eQTLs). We identify numerous cis-eQTLs that contribute to the marked differences in immune responses detected within and between populations and a strong trans-eQTL hotspot at TLR1 that decreases expression of pro-inflammatory genes in Europeans only. We find that immune-responsive regulatory variants are enriched in population-specific signals of natural selection and show that admixture with Neandertals introduced regulatory variants into European genomes, affecting preferentially responses to viral challenges. Together, our study uncovers evolutionarily important determinants of differences in host immune responsiveness between human populations.


Subject(s)
Adaptation, Physiological/genetics , Adaptation, Physiological/immunology , Adaptive Immunity , Neanderthals/genetics , Neanderthals/immunology , Adaptive Immunity/genetics , Alleles , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Base Sequence , Biological Evolution , Black People/genetics , Gene Expression Regulation , Genetic Variation , Humans , Immune System , Quantitative Trait Loci , RNA/genetics , Selection, Genetic , Sequence Analysis, RNA , Toll-Like Receptors/genetics , Transcription, Genetic , Virus Diseases/genetics , Virus Diseases/immunology , White People/genetics
2.
Nature ; 626(8000): 827-835, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355791

ABSTRACT

Individuals differ widely in their immune responses, with age, sex and genetic factors having major roles in this inherent variability1-6. However, the variables that drive such differences in cytokine secretion-a crucial component of the host response to immune challenges-remain poorly defined. Here we investigated 136 variables and identified smoking, cytomegalovirus latent infection and body mass index as major contributors to variability in cytokine response, with effects of comparable magnitudes with age, sex and genetics. We find that smoking influences both innate and adaptive immune responses. Notably, its effect on innate responses is quickly lost after smoking cessation and is specifically associated with plasma levels of CEACAM6, whereas its effect on adaptive responses persists long after individuals quit smoking and is associated with epigenetic memory. This is supported by the association of the past smoking effect on cytokine responses with DNA methylation at specific signal trans-activators and regulators of metabolism. Our findings identify three novel variables associated with cytokine secretion variability and reveal roles for smoking in the short- and long-term regulation of immune responses. These results have potential clinical implications for the risk of developing infections, cancers or autoimmune diseases.


Subject(s)
Adaptive Immunity , Smoking , Female , Humans , Male , Adaptive Immunity/drug effects , Adaptive Immunity/genetics , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Body Mass Index , Cytokines/blood , Cytokines/immunology , Cytomegalovirus/immunology , Cytomegalovirus/pathogenicity , Cytomegalovirus/physiology , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Immunity, Innate/drug effects , Immunity, Innate/genetics , Infections/etiology , Infections/immunology , Neoplasms/etiology , Neoplasms/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Smoking/adverse effects , Smoking/blood , Smoking/genetics , Smoking/immunology
3.
Nature ; 621(7977): 120-128, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37558883

ABSTRACT

Humans display substantial interindividual clinical variability after SARS-CoV-2 infection1-3, the genetic and immunological basis of which has begun to be deciphered4. However, the extent and drivers of population differences in immune responses to SARS-CoV-2 remain unclear. Here we report single-cell RNA-sequencing data for peripheral blood mononuclear cells-from 222 healthy donors of diverse ancestries-that were stimulated with SARS-CoV-2 or influenza A virus. We show that SARS-CoV-2 induces weaker, but more heterogeneous, interferon-stimulated gene activity compared with influenza A virus, and a unique pro-inflammatory signature in myeloid cells. Transcriptional responses to viruses display marked population differences, primarily driven by changes in cell abundance including increased lymphoid differentiation associated with latent cytomegalovirus infection. Expression quantitative trait loci and mediation analyses reveal a broad effect of cell composition on population disparities in immune responses, with genetic variants exerting a strong effect on specific loci. Furthermore, we show that natural selection has increased population differences in immune responses, particularly for variants associated with SARS-CoV-2 response in East Asians, and document the cellular and molecular mechanisms by which Neanderthal introgression has altered immune functions, such as the response of myeloid cells to viruses. Finally, colocalization and transcriptome-wide association analyses reveal an overlap between the genetic basis of immune responses to SARS-CoV-2 and COVID-19 severity, providing insights into the factors contributing to current disparities in COVID-19 risk.


Subject(s)
COVID-19 , Genetics, Population , SARS-CoV-2 , Single-Cell Gene Expression Analysis , Animals , Humans , Cell Differentiation , COVID-19/genetics , COVID-19/immunology , COVID-19/virology , Cytomegalovirus/physiology , East Asian People/genetics , Genetic Introgression , Influenza A virus/pathogenicity , Influenza A virus/physiology , Interferons/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Myeloid Cells/immunology , Neanderthals/genetics , Neanderthals/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Selection, Genetic , Virus Latency
4.
Cell ; 154(3): 691-703, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23890820

ABSTRACT

Large numbers of inbred laboratory rat strains have been developed for a range of complex disease phenotypes. To gain insights into the evolutionary pressures underlying selection for these phenotypes, we sequenced the genomes of 27 rat strains, including 11 models of hypertension, diabetes, and insulin resistance, along with their respective control strains. Altogether, we identified more than 13 million single-nucleotide variants, indels, and structural variants across these rat strains. Analysis of strain-specific selective sweeps and gene clusters implicated genes and pathways involved in cation transport, angiotensin production, and regulators of oxidative stress in the development of cardiovascular disease phenotypes in rats. Many of the rat loci that we identified overlap with previously mapped loci for related traits in humans, indicating the presence of shared pathways underlying these phenotypes in rats and humans. These data represent a step change in resources available for evolutionary analysis of complex traits in disease models.


Subject(s)
Rats/classification , Rats/genetics , Animals , Disease Models, Animal , Genome , Phenotype , Phylogeny , Polymorphism, Single Nucleotide , Rats, Inbred Strains
5.
PLoS Genet ; 16(11): e1009090, 2020 11.
Article in English | MEDLINE | ID: mdl-33147208

ABSTRACT

Interferon ß (IFN-ß) is a cytokine that induces a global antiviral proteome, and regulates the adaptive immune response to infections and tumors. Its effects strongly depend on its level and timing of expression. Therefore, the transcription of its coding gene IFNB1 is strictly controlled. We have previously shown that in mice, the TRIM33 protein restrains Ifnb1 transcription in activated myeloid cells through an upstream inhibitory sequence called ICE. Here, we show that the deregulation of Ifnb1 expression observed in murine Trim33-/- macrophages correlates with abnormal looping of both ICE and the Ifnb1 gene to a 100 kb downstream region overlapping the Ptplad2/Hacd4 gene. This region is a predicted myeloid super-enhancer in which we could characterize 3 myeloid-specific active enhancers, one of which (E5) increases the response of the Ifnb1 promoter to activation. In humans, the orthologous region contains several single nucleotide polymorphisms (SNPs) known to be associated with decreased expression of IFNB1 in activated monocytes, and loops to the IFNB1 gene. The strongest association is found for the rs12553564 SNP, located in the E5 orthologous region. The minor allele of rs12553564 disrupts a conserved C/EBP-ß binding motif, prevents binding of C/EBP-ß, and abolishes the activation-induced enhancer activity of E5. Altogether, these results establish a link between a genetic variant preventing binding of a transcription factor and a higher order phenotype, and suggest that the frequent minor allele (around 30% worldwide) might be associated with phenotypes regulated by IFN-ß expression in myeloid cells.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation/immunology , Interferon-beta/genetics , Myeloid Cells/metabolism , Alleles , Animals , Blood Buffy Coat/cytology , Cells, Cultured , Humans , Interferon-beta/immunology , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Point Mutation , Polymorphism, Single Nucleotide , Primary Cell Culture , Promoter Regions, Genetic , Quantitative Trait Loci , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Am J Hum Genet ; 104(6): 1241-1250, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31155285

ABSTRACT

Archaic admixture is increasingly recognized as an important source of diversity in modern humans, and Neanderthal haplotypes cover 1%-3% of the genome of present-day Eurasians. Recent work has shown that archaic introgression has contributed to human phenotypic diversity, mostly through the regulation of gene expression. Yet the mechanisms through which archaic variants alter gene expression and the forces driving the introgression landscape at regulatory regions remain elusive. Here, we explored the impact of archaic introgression on transcriptional and post-transcriptional regulation. We focused on promoters and enhancers across 127 different tissues as well as on microRNA (miRNA)-mediated regulation. Although miRNAs themselves harbor few archaic variants, we found that some of these variants may have a strong impact on miRNA-mediated gene regulation. Enhancers were by far the regulatory elements most affected by archaic introgression: up to one-third of the tissues we tested presented significant enrichments. Specifically, we found strong enrichments of archaic variants in adipose-related tissues and primary T cells, even after accounting for various genomic and evolutionary confounders such as recombination rate and background selection. Interestingly, we identified signatures of adaptive introgression at enhancers of some key regulators of adipogenesis, raising the interesting hypothesis of a possible adaptation of early Eurasians to colder climates. Collectively, this study sheds new light on the mechanisms through which archaic admixture has impacted gene regulation in Eurasians and, more generally, increases our understanding of the contribution of Neanderthals to the regulation of acquired immunity and adipose homeostasis in modern humans.


Subject(s)
Evolution, Molecular , Gene Expression Regulation, Developmental , Neanderthals/genetics , Polymorphism, Single Nucleotide , RNA Processing, Post-Transcriptional , Selection, Genetic , Transcription, Genetic , Adaptation, Physiological , Adipogenesis , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Genome, Human , Genomics , Humans , MicroRNAs/genetics , Recombination, Genetic , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
7.
Genome Res ; 27(3): 440-450, 2017 03.
Article in English | MEDLINE | ID: mdl-28250018

ABSTRACT

The recoding of genetic information through RNA editing contributes to proteomic diversity, but the extent and significance of RNA editing in disease is poorly understood. In particular, few studies have investigated the relationship between RNA editing and disease at a genome-wide level. Here, we developed a framework for the genome-wide detection of RNA sites that are differentially edited in disease. Using RNA-sequencing data from 100 hippocampi from mice with epilepsy (pilocarpine-temporal lobe epilepsy model) and 100 healthy control hippocampi, we identified 256 RNA sites (overlapping with 87 genes) that were significantly differentially edited between epileptic cases and controls. The degree of differential RNA editing in epileptic mice correlated with frequency of seizures, and the set of genes differentially RNA-edited between case and control mice were enriched for functional terms highly relevant to epilepsy, including "neuron projection" and "seizures." Genes with differential RNA editing were preferentially enriched for genes with a genetic association to epilepsy. Indeed, we found that they are significantly enriched for genes that harbor nonsynonymous de novo mutations in patients with epileptic encephalopathy and for common susceptibility variants associated with generalized epilepsy. These analyses reveal a functional convergence between genes that are differentially RNA-edited in acquired symptomatic epilepsy and those that contribute risk for genetic epilepsy. Taken together, our results suggest a potential role for RNA editing in the epileptic hippocampus in the occurrence and severity of epileptic seizures.


Subject(s)
Epilepsy/genetics , RNA Editing , Animals , Genome-Wide Association Study , Hippocampus/metabolism , Male , Mice , Transcriptome
9.
Mol Cell Proteomics ; 14(3): 484-98, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25532521

ABSTRACT

Macrophage multinucleation (MM) is essential for various biological processes such as osteoclast-mediated bone resorption and multinucleated giant cell-associated inflammatory reactions. Here we study the molecular pathways underlying multinucleation in the rat through an integrative approach combining MS-based quantitative phosphoproteomics (LC-MS/MS) and transcriptome (high-throughput RNA-sequencing) to identify new regulators of MM. We show that a strong metabolic shift toward HIF1-mediated glycolysis occurs at transcriptomic level during MM, together with modifications in phosphorylation of over 50 proteins including several ARF GTPase activators and polyphosphate inositol phosphatases. We use shortest-path analysis to link differential phosphorylation with the transcriptomic reprogramming of macrophages and identify LRRFIP1, SMARCA4, and DNMT1 as novel regulators of MM. We experimentally validate these predictions by showing that knock-down of these latter reduce macrophage multinucleation. These results provide a new framework for the combined analysis of transcriptional and post-translational changes during macrophage multinucleation, prioritizing essential genes, and revealing the sequential events leading to the multinucleation of macrophages.


Subject(s)
Cell Nucleus/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Helicases/metabolism , Gene Expression Profiling/methods , Macrophages/metabolism , Nuclear Proteins/metabolism , Proteome/analysis , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Helicases/genetics , High-Throughput Nucleotide Sequencing/methods , Nuclear Proteins/genetics , Phosphorylation , RNA-Binding Proteins/genetics , Rats , Rats, Inbred Lew , Rats, Inbred WKY , Sequence Analysis, RNA/methods , Transcription Factors/genetics
10.
Nucleic Acids Res ; 43(3): 1418-32, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25605797

ABSTRACT

The large number of chemical modifications that are found on the histone proteins of eukaryotic cells form multiple complex combinations, which can act as recognition signals for reader proteins. We have used peptide capture in conjunction with super-SILAC quantification to carry out an unbiased high-throughput analysis of the composition of protein complexes that bind to histone H3K9/S10 and H3K27/S28 methyl-phospho modifications. The accurate quantification allowed us to perform Weighted correlation network analysis (WGCNA) to obtain a systems-level view of the histone H3 histone tail interactome. The analysis reveals the underlying modularity of the histone reader network with members of nuclear complexes exhibiting very similar binding signatures, which suggests that many proteins bind to histones as part of pre-organized complexes. Our results identify a novel complex that binds to the double H3K9me3/S10ph modification, which includes Atrx, Daxx and members of the FACT complex. The super-SILAC approach allows comparison of binding to multiple peptides with different combinations of modifications and the resolution of the WGCNA analysis is enhanced by maximizing the number of combinations that are compared. This makes it a useful approach for assessing the effects of changes in histone modification combinations on the composition and function of bound complexes.


Subject(s)
Histones/metabolism , Proteins/metabolism , Animals , Cell Line, Tumor , Combinatorial Chemistry Techniques , Mass Spectrometry , Mice
11.
PLoS Genet ; 10(1): e1004006, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24391511

ABSTRACT

Recent high-throughput efforts such as ENCODE have generated a large body of genome-scale transcriptional data in multiple conditions (e.g., cell-types and disease states). Leveraging these data is especially important for network-based approaches to human disease, for instance to identify coherent transcriptional modules (subnetworks) that can inform functional disease mechanisms and pathological pathways. Yet, genome-scale network analysis across conditions is significantly hampered by the paucity of robust and computationally-efficient methods. Building on the Higher-Order Generalized Singular Value Decomposition, we introduce a new algorithmic approach for efficient, parameter-free and reproducible identification of network-modules simultaneously across multiple conditions. Our method can accommodate weighted (and unweighted) networks of any size and can similarly use co-expression or raw gene expression input data, without hinging upon the definition and stability of the correlation used to assess gene co-expression. In simulation studies, we demonstrated distinctive advantages of our method over existing methods, which was able to recover accurately both common and condition-specific network-modules without entailing ad-hoc input parameters as required by other approaches. We applied our method to genome-scale and multi-tissue transcriptomic datasets from rats (microarray-based) and humans (mRNA-sequencing-based) and identified several common and tissue-specific subnetworks with functional significance, which were not detected by other methods. In humans we recapitulated the crosstalk between cell-cycle progression and cell-extracellular matrix interactions processes in ventricular zones during neocortex expansion and further, we uncovered pathways related to development of later cognitive functions in the cortical plate of the developing brain which were previously unappreciated. Analyses of seven rat tissues identified a multi-tissue subnetwork of co-expressed heat shock protein (Hsp) and cardiomyopathy genes (Bag3, Cryab, Kras, Emd, Plec), which was significantly replicated using separate failing heart and liver gene expression datasets in humans, thus revealing a conserved functional role for Hsp genes in cardiovascular disease.


Subject(s)
Cardiomyopathies/genetics , Gene Regulatory Networks , Genome, Human , Transcription, Genetic , Algorithms , Animals , Cardiomyopathies/pathology , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Gene Expression , Gene Expression Profiling , Humans , Organ Specificity , Rats , Signal Transduction/genetics
12.
PLoS Genet ; 10(12): e1004813, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25474312

ABSTRACT

Epigenetic marks such as cytosine methylation are important determinants of cellular and whole-body phenotypes. However, the extent of, and reasons for inter-individual differences in cytosine methylation, and their association with phenotypic variation are poorly characterised. Here we present the first genome-wide study of cytosine methylation at single-nucleotide resolution in an animal model of human disease. We used whole-genome bisulfite sequencing in the spontaneously hypertensive rat (SHR), a model of cardiovascular disease, and the Brown Norway (BN) control strain, to define the genetic architecture of cytosine methylation in the mammalian heart and to test for association between methylation and pathophysiological phenotypes. Analysis of 10.6 million CpG dinucleotides identified 77,088 CpGs that were differentially methylated between the strains. In F1 hybrids we found 38,152 CpGs showing allele-specific methylation and 145 regions with parent-of-origin effects on methylation. Cis-linkage explained almost 60% of inter-strain variation in methylation at a subset of loci tested for linkage in a panel of recombinant inbred (RI) strains. Methylation analysis in isolated cardiomyocytes showed that in the majority of cases methylation differences in cardiomyocytes and non-cardiomyocytes were strain-dependent, confirming a strong genetic component for cytosine methylation. We observed preferential nucleotide usage associated with increased and decreased methylation that is remarkably conserved across species, suggesting a common mechanism for germline control of inter-individual variation in CpG methylation. In the RI strain panel, we found significant correlation of CpG methylation and levels of serum chromogranin B (CgB), a proposed biomarker of heart failure, which is evidence for a link between germline DNA sequence variation, CpG methylation differences and pathophysiological phenotypes in the SHR strain. Together, these results will stimulate further investigation of the molecular basis of locally regulated variation in CpG methylation and provide a starting point for understanding the relationship between the genetic control of CpG methylation and disease phenotypes.


Subject(s)
Cardiovascular Diseases/genetics , DNA Methylation , Genome , Myocardium/metabolism , Animals , Base Sequence , Cardiovascular Diseases/pathology , Cells, Cultured , Disease Models, Animal , Humans , Male , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Polymorphism, Single Nucleotide , Rats , Rats, Inbred BN , Rats, Inbred SHR , Sequence Analysis, DNA/methods
13.
Nature ; 467(7314): 460-4, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20827270

ABSTRACT

Combined analyses of gene networks and DNA sequence variation can provide new insights into the aetiology of common diseases that may not be apparent from genome-wide association studies alone. Recent advances in rat genomics are facilitating systems-genetics approaches. Here we report the use of integrated genome-wide approaches across seven rat tissues to identify gene networks and the loci underlying their regulation. We defined an interferon regulatory factor 7 (IRF7)-driven inflammatory network (IDIN) enriched for viral response genes, which represents a molecular biomarker for macrophages and which was regulated in multiple tissues by a locus on rat chromosome 15q25. We show that Epstein-Barr virus induced gene 2 (Ebi2, also known as Gpr183), which lies at this locus and controls B lymphocyte migration, is expressed in macrophages and regulates the IDIN. The human orthologous locus on chromosome 13q32 controlled the human equivalent of the IDIN, which was conserved in monocytes. IDIN genes were more likely to associate with susceptibility to type 1 diabetes (T1D)-a macrophage-associated autoimmune disease-than randomly selected immune response genes (P = 8.85 × 10(-6)). The human locus controlling the IDIN was associated with the risk of T1D at single nucleotide polymorphism rs9585056 (P = 7.0 × 10(-10); odds ratio, 1.15), which was one of five single nucleotide polymorphisms in this region associated with EBI2 (GPR183) expression. These data implicate IRF7 network genes and their regulatory locus in the pathogenesis of T1D.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Immunity, Innate/genetics , Viruses/immunology , Animals , Base Sequence , Chromosomes, Human, Pair 13/genetics , Chromosomes, Mammalian/genetics , Diabetes Mellitus, Type 1/immunology , Gene Regulatory Networks/genetics , Genome-Wide Association Study , Humans , Inflammation/genetics , Inflammation/immunology , Interferon Regulatory Factor-7/immunology , Macrophages/immunology , Macrophages/metabolism , Organ Specificity , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Rats , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
14.
Hum Mol Genet ; 21(12): 2815-24, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22403184

ABSTRACT

One mechanism by which disease-associated DNA variation can alter disease risk is altering gene expression. However, linkage disequilibrium (LD) between variants, mostly single-nucleotide polymorphisms (SNPs), means it is not sufficient to show that a particular variant associates with both disease and expression, as there could be two distinct causal variants in LD. Here, we describe a formal statistical test of colocalization and apply it to type 1 diabetes (T1D)-associated regions identified mostly through genome-wide association studies and expression quantitative trait loci (eQTLs) discovered in a recently determined large monocyte expression data set from the Gutenberg Health Study (1370 individuals), with confirmation sought in an additional data set from the Cardiogenics Transcriptome Study (558 individuals). We excluded 39 out of 60 overlapping eQTLs in 49 T1D regions from possible colocalization and identified 21 coincident eQTLs, representing 21 genes in 14 distinct T1D regions. Our results reflect the importance of monocyte (and their derivatives, macrophage and dendritic cell) gene expression in human T1D and support the candidacy of several genes as causal factors in autoimmune pancreatic beta-cell destruction, including AFF3, CD226, CLECL1, DEXI, FKRP, PRKD2, RNLS, SMARCE1 and SUOX, in addition to the recently described GPR183 (EBI2) gene.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease/genetics , Monocytes/metabolism , Polymorphism, Single Nucleotide , Transcriptome , Adult , Aged , Algorithms , Female , Genome-Wide Association Study , Genotype , Humans , Linkage Disequilibrium , Male , Middle Aged , Models, Genetic , Quantitative Trait Loci/genetics , Risk Factors
15.
PLoS Genet ; 7(12): e1002367, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22144904

ABSTRACT

One major expectation from the transcriptome in humans is to characterize the biological basis of associations identified by genome-wide association studies. So far, few cis expression quantitative trait loci (eQTLs) have been reliably related to disease susceptibility. Trans-regulating mechanisms may play a more prominent role in disease susceptibility. We analyzed 12,808 genes detected in at least 5% of circulating monocyte samples from a population-based sample of 1,490 European unrelated subjects. We applied a method of extraction of expression patterns-independent component analysis-to identify sets of co-regulated genes. These patterns were then related to 675,350 SNPs to identify major trans-acting regulators. We detected three genomic regions significantly associated with co-regulated gene modules. Association of these loci with multiple expression traits was replicated in Cardiogenics, an independent study in which expression profiles of monocytes were available in 758 subjects. The locus 12q13 (lead SNP rs11171739), previously identified as a type 1 diabetes locus, was associated with a pattern including two cis eQTLs, RPS26 and SUOX, and 5 trans eQTLs, one of which (MADCAM1) is a potential candidate for mediating T1D susceptibility. The locus 12q24 (lead SNP rs653178), which has demonstrated extensive disease pleiotropy, including type 1 diabetes, hypertension, and celiac disease, was associated to a pattern strongly correlating to blood pressure level. The strongest trans eQTL in this pattern was CRIP1, a known marker of cellular proliferation in cancer. The locus 12q15 (lead SNP rs11177644) was associated with a pattern driven by two cis eQTLs, LYZ and YEATS4, and including 34 trans eQTLs, several of them tumor-related genes. This study shows that a method exploiting the structure of co-expressions among genes can help identify genomic regions involved in trans regulation of sets of genes and can provide clues for understanding the mechanisms linking genome-wide association loci to disease.


Subject(s)
Celiac Disease/genetics , Diabetes Mellitus, Type 1/genetics , Gene Expression Regulation/genetics , Genetic Variation/genetics , Hypertension/genetics , Monocytes/metabolism , Quantitative Trait Loci/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Female , Gene Expression Profiling , Genetic Predisposition to Disease , Genome, Human , Genome-Wide Association Study , Humans , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Muramidase/genetics , Polymorphism, Single Nucleotide , Proteins/genetics , Ribosomal Proteins/genetics , Transcription Factors/genetics
16.
Nat Commun ; 14(1): 8347, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102153

ABSTRACT

Genome-wide association studies (GWASs) have identified thousands of non-coding variants that are associated with human complex traits and diseases. The analysis of such GWAS variants in different contexts and physiological states is essential for deciphering the regulatory mechanisms underlying human disease. Alternative polyadenylation (APA) is a key post-transcriptional modification for most human genes that substantially impacts upon cell behavior. Here, we mapped 9,493 3'-untranslated region APA quantitative trait loci in 18 human immune baseline cell types and 8 stimulation conditions (immune 3'aQTLs). Through the comparison between baseline and stimulation data, we observed the high responsiveness of 3'aQTLs to immune stimulation (response 3'aQTLs). Co-localization and mendelian randomization analyses of immune 3'aQTLs identified 678 genes where 3'aQTL are associated with variation in complex traits, 27.3% of which were derived from response 3'aQTLs. Overall, these analyses reveal the role of immune 3'aQTLs in the determination of complex traits, providing new insights into the regulatory mechanisms underlying disease etiologies.


Subject(s)
Polyadenylation , Quantitative Trait Loci , Humans , Quantitative Trait Loci/genetics , Polyadenylation/genetics , 3' Untranslated Regions/genetics , Genome-Wide Association Study , Multifactorial Inheritance
17.
Blood ; 115(23): 4644-50, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20212171

ABSTRACT

Through its binding with protein S (PS), a key element of the coagulation/fibrinolysis cascade, the C4b-binding protein (C4BP) has been hypothesized to be involved in the susceptibility to venous thrombosis (VT). To identify genetic factors that may influence the plasma levels of the 3 C4BP existing isoforms, alpha(7)beta(1), alpha(6)beta(1), and alpha(7)beta(0), we conducted a genome-wide association study by analyzing 283 437 single nucleotide polymorphisms (SNPs) in the Genetic Analysis of Idiopathic Thrombophilia (GAIT) study composed of 352 persons. Three SNPs at the C4BPB/C4BPA locus were found genome-wide significantly associated with alpha(7)beta(0) levels. One of these SNPs was further found to explain approximately 11% of the variability of mRNA C4BPA expression in the Gutenberg Heart Study composed of 1490 persons, with no effect on C4BPB mRNA expression. The allele associated with increased alpha(7)beta(0) plasma levels and increased C4BPA expression was further found associated with increased risk of VT (odds ratio [OR] = 1.24 [1.03-1.53]) in 2 independent case-control studies (MARseille THrombosis Association study [MARTHA] and FActeurs de RIsque et de récidives de la maladie thromboembolique VEineuse [FARIVE]) gathering 1706 cases and 1379 controls. This SNP was not associated with free PS or total PS. In conclusion, we observed strong evidence that the C4BPB/C4BPA locus is a new susceptibility locus for VT through a PS-independent mechanism that remains to be elucidated.


Subject(s)
Case-Control Studies , Genetic Loci , Histocompatibility Antigens/genetics , Polymorphism, Single Nucleotide , Venous Thrombosis/genetics , Clinical Trials as Topic , Complement C4b-Binding Protein , Female , Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Histocompatibility Antigens/blood , Humans , Male , Protein S , Risk Factors , Venous Thrombosis/blood
18.
Genomics ; 98(5): 320-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21763416

ABSTRACT

In humans, the fraction of X-linked genes with higher expression in females has been estimated to be 5% from microarray studies, a proportion lower than the 25% of genes thought to escape X inactivation. We analyzed 715 X-linked transcripts in circulating monocytes from 1,467 subjects and found an excess of female-biased transcripts on the X compared to autosomes (9.4% vs 5.5%, p<2×10(-5)). Among the genes not previously known to escape inactivation, the most significant one was EFHC2 whose 20% of variability was explained by sex. We also investigated cis expression quantitative trait loci (eQTLs) by analyzing 15,703 X-linked SNPs. The frequency and magnitude of X-linked cis eQTLs were quite similar in males and females. Few genes exhibited a stronger genetic effect in females than in males (ARSD, DCX, POLA1 and ITM2A). These genes would deserve further investigation since they may contribute to sex pathophysiological differences.


Subject(s)
Genes, X-Linked , Genetic Variation , Monocytes/cytology , Quantitative Trait Loci , Adult , Aged , Calcium-Binding Proteins/genetics , Chromosomes, Human, X/genetics , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Sex Factors , Transcription, Genetic , X Chromosome Inactivation
19.
Genome Med ; 14(1): 28, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35264221

ABSTRACT

BACKGROUND: Blood plasma proteins play an important role in immune defense against pathogens, including cytokine signaling, the complement system, and the acute-phase response. Recent large-scale studies have reported genetic (i.e., protein quantitative trait loci, pQTLs) and non-genetic factors, such as age and sex, as major determinants to inter-individual variability in immune response variation. However, the contribution of blood-cell composition to plasma protein heterogeneity has not been fully characterized and may act as a mediating factor in association studies. METHODS: Here, we evaluated plasma protein levels from 400 unrelated healthy individuals of western European ancestry, who were stratified by sex and two decades of life (20-29 and 60-69 years), from the Milieu Intérieur cohort. We quantified 229 proteins by Luminex in a clinically certified laboratory and their levels of variation were analyzed together with 5.2 million single-nucleotide polymorphisms. With respect to non-genetic variables, we included 254 lifestyle and biochemical factors, as well as counts of seven circulating immune cell populations measured by hemogram and standardized flow cytometry. RESULTS: Collectively, we found 152 significant associations involving 49 proteins and 20 non-genetic variables. Consistent with previous studies, age and sex showed a global, pervasive impact on plasma protein heterogeneity, while body mass index and other health status variables were among the non-genetic factors with the highest number of associations. After controlling for these covariates, we identified 100 and 12 pQTLs acting in cis and trans, respectively, collectively associated with 87 plasma proteins and including 19 novel genetic associations. Genetic factors explained the largest fraction of the variability of plasma protein levels, as compared to non-genetic factors. In addition, blood-cell fractions, including leukocytes, lymphocytes, monocytes, neutrophils, eosinophils, basophils, and platelets, had a larger contribution to inter-individual variability than age and sex and appeared as confounders of specific genetic associations. Finally, we identified new genetic associations with plasma protein levels of five monogenic Mendelian disease genes including two primary immunodeficiency genes (Ficolin-3 and FAS). CONCLUSIONS: Our study identified novel genetic and non-genetic factors associated to plasma protein levels which may inform health status and disease management.


Subject(s)
Blood Proteins , Primary Immunodeficiency Diseases , Blood Proteins/genetics , Genome-Wide Association Study , Health Status , Humans , Polymorphism, Single Nucleotide , Quantitative Trait Loci
20.
Nat Commun ; 13(1): 5895, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36202838

ABSTRACT

Epigenetic changes are required for normal development, yet the nature and respective contribution of factors that drive epigenetic variation in humans remain to be fully characterized. Here, we assessed how the blood DNA methylome of 884 adults is affected by DNA sequence variation, age, sex and 139 factors relating to life habits and immunity. Furthermore, we investigated whether these effects are mediated or not by changes in cellular composition, measured by deep immunophenotyping. We show that DNA methylation differs substantially between naïve and memory T cells, supporting the need for adjustment on these cell-types. By doing so, we find that latent cytomegalovirus infection drives DNA methylation variation and provide further support that the increased dispersion of DNA methylation with aging is due to epigenetic drift. Finally, our results indicate that cellular composition and DNA sequence variation are the strongest predictors of DNA methylation, highlighting critical factors for medical epigenomics studies.


Subject(s)
DNA Methylation , Epigenomics , Adult , Aging/genetics , Epigenesis, Genetic , Epigenomics/methods , Humans , Immunologic Factors
SELECTION OF CITATIONS
SEARCH DETAIL