Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nat Commun ; 14(1): 7096, 2023 11 04.
Article in English | MEDLINE | ID: mdl-37925459

ABSTRACT

Disrupted circadian rhythms have been linked to an increased risk of hypertension and cardiovascular disease. However, many studies show inconsistent findings and are not sufficiently powered for targeted subgroup analyses. Using the UK Biobank cohort, we evaluate the association between circadian rhythm-disrupting behaviours, blood pressure (SBP, DBP) and inflammatory markers in >350,000 adults with European white British ancestry. The independent U-shaped relationship between sleep length and SBP/DBP is most prominent with a low inflammatory status. Poor sleep quality and permanent night shift work are also positively associated with SBP/DBP. Although fully adjusting for BMI in the linear regression model attenuated effect sizes, these associations remain significant. Two-sample Mendelian Randomisation (MR) analyses support a potential causal effect of long sleep, short sleep, chronotype, daytime napping and sleep duration on SBP/DBP. Thus, in the current study, we present a positive association between circadian rhythm-disrupting behaviours and SBP/DBP regulation in males and females that is largely independent of age.


Subject(s)
Shift Work Schedule , Sleep Initiation and Maintenance Disorders , Adult , Male , Female , Humans , Blood Pressure/physiology , Biological Specimen Banks , Sleep/physiology , Circadian Rhythm/physiology , Inflammation , United Kingdom
2.
Nature ; 616(7955): 123-131, 2023 04.
Article in English | MEDLINE | ID: mdl-36991119

ABSTRACT

The use of omic modalities to dissect the molecular underpinnings of common diseases and traits is becoming increasingly common. But multi-omic traits can be genetically predicted, which enables highly cost-effective and powerful analyses for studies that do not have multi-omics1. Here we examine a large cohort (the INTERVAL study2; n = 50,000 participants) with extensive multi-omic data for plasma proteomics (SomaScan, n = 3,175; Olink, n = 4,822), plasma metabolomics (Metabolon HD4, n = 8,153), serum metabolomics (Nightingale, n = 37,359) and whole-blood Illumina RNA sequencing (n = 4,136), and use machine learning to train genetic scores for 17,227 molecular traits, including 10,521 that reach Bonferroni-adjusted significance. We evaluate the performance of genetic scores through external validation across cohorts of individuals of European, Asian and African American ancestries. In addition, we show the utility of these multi-omic genetic scores by quantifying the genetic control of biological pathways and by generating a synthetic multi-omic dataset of the UK Biobank3 to identify disease associations using a phenome-wide scan. We highlight a series of biological insights with regard to genetic mechanisms in metabolism and canonical pathway associations with disease; for example, JAK-STAT signalling and coronary atherosclerosis. Finally, we develop a portal ( https://www.omicspred.org/ ) to facilitate public access to all genetic scores and validation results, as well as to serve as a platform for future extensions and enhancements of multi-omic genetic scores.


Subject(s)
Coronary Artery Disease , Multiomics , Humans , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Metabolomics/methods , Phenotype , Proteomics/methods , Machine Learning , Black or African American/genetics , Asian/genetics , European People/genetics , United Kingdom , Datasets as Topic , Internet , Reproducibility of Results , Cohort Studies , Proteome/analysis , Proteome/metabolism , Metabolome , Plasma/metabolism , Databases, Factual
3.
Nat Commun ; 13(1): 7356, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36446790

ABSTRACT

Understanding how genetic variants influence disease risk and complex traits (variant-to-function) is one of the major challenges in human genetics. Here we present a model-driven framework to leverage human genome-scale metabolic networks to define how genetic variants affect biochemical reaction fluxes across major human tissues, including skeletal muscle, adipose, liver, brain and heart. As proof of concept, we build personalised organ-specific metabolic flux models for 524,615 individuals of the INTERVAL and UK Biobank cohorts and perform a fluxome-wide association study (FWAS) to identify 4312 associations between personalised flux values and the concentration of metabolites in blood. Furthermore, we apply FWAS to identify 92 metabolic fluxes associated with the risk of developing coronary artery disease, many of which are linked to processes previously described to play in role in the disease. Our work demonstrates that genetically personalised metabolic models can elucidate the downstream effects of genetic variants on biochemical reactions involved in common human diseases.


Subject(s)
Adipose Tissue , Coronary Artery Disease , Humans , Brain , Genome, Human , Heart
4.
Nat Commun ; 12(1): 74, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397965

ABSTRACT

The effective storage of lipids in white adipose tissue (WAT) critically impacts whole body energy homeostasis. Many genes have been implicated in WAT lipid metabolism, including tripartite motif containing 28 (Trim28), a gene proposed to primarily influence adiposity via epigenetic mechanisms in embryonic development. However, in the current study we demonstrate that mice with deletion of Trim28 specifically in committed adipocytes, also develop obesity similar to global Trim28 deletion models, highlighting a post-developmental role for Trim28. These effects were exacerbated in female mice, contributing to the growing notion that Trim28 is a sex-specific regulator of obesity. Mechanistically, this phenotype involves alterations in lipolysis and triglyceride metabolism, explained in part by loss of Klf14 expression, a gene previously demonstrated to modulate adipocyte size and body composition in a sex-specific manner. Thus, these findings provide evidence that Trim28 is a bona fide, sex specific regulator of post-developmental adiposity and WAT function.


Subject(s)
Adipocytes/metabolism , Gene Deletion , Glucose/metabolism , Obesity/pathology , Tripartite Motif-Containing Protein 28/genetics , 3T3-L1 Cells , Adipose Tissue, White/metabolism , Adiposity , Animals , Body Weight , Diet , Diet, High-Fat , Energy Metabolism , Female , Gene Regulatory Networks , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Phenotype , Triglycerides/metabolism , Tripartite Motif-Containing Protein 28/deficiency
5.
Nat Commun ; 11(1): 3761, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32724101

ABSTRACT

Chronic immune-mediated diseases of adulthood often originate in early childhood. To investigate genetic associations between neonatal immunity and disease, we map expression quantitative trait loci (eQTLs) in resting myeloid cells and CD4+ T cells from cord blood samples, as well as in response to lipopolysaccharide (LPS) or phytohemagglutinin (PHA) stimulation, respectively. Cis-eQTLs are largely specific to cell type or stimulation, and 31% and 52% of genes with cis-eQTLs have response eQTLs (reQTLs) in myeloid cells and T cells, respectively. We identified cis regulatory factors acting as mediators of trans effects. There is extensive colocalisation between condition-specific neonatal cis-eQTLs and variants associated with immune-mediated diseases, in particular CTSH had widespread colocalisation across diseases. Mendelian randomisation shows causal neonatal gene expression effects on disease risk for BTN3A2, HLA-C and others. Our study elucidates the genetics of gene expression in neonatal immune cells, and aetiological origins of autoimmune and allergic diseases.


Subject(s)
Autoimmune Diseases/genetics , Child Development/physiology , Gene Expression Regulation, Developmental/immunology , Hypersensitivity/genetics , Quantitative Trait Loci/immunology , Autoimmune Diseases/immunology , Butyrophilins/genetics , Butyrophilins/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cathepsin H/genetics , Cathepsin H/metabolism , Child , Child, Preschool , Datasets as Topic , Fetal Blood/cytology , Gene Expression Profiling , Gene Regulatory Networks/immunology , Genetic Predisposition to Disease , Genome-Wide Association Study , HLA-C Antigens/genetics , HLA-C Antigens/metabolism , Humans , Hypersensitivity/immunology , Infant , Infant, Newborn , Mendelian Randomization Analysis , Myeloid Cells/immunology , Myeloid Cells/metabolism , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prospective Studies
6.
Am J Hum Genet ; 105(6): 1076-1090, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31679650

ABSTRACT

Cytokines are essential regulatory components of the immune system, and their aberrant levels have been linked to many disease states. Despite increasing evidence that cytokines operate in concert, many of the physiological interactions between cytokines, and the shared genetic architecture that underlies them, remain unknown. Here, we aimed to identify and characterize genetic variants with pleiotropic effects on cytokines. Using three population-based cohorts (n = 9,263), we performed multivariate genome-wide association studies (GWAS) for a correlation network of 11 circulating cytokines, then combined our results in meta-analysis. We identified a total of eight loci significantly associated with the cytokine network, of which two (PDGFRB and ABO) had not been detected previously. In addition, conditional analyses revealed a further four secondary signals at three known cytokine loci. Integration, through the use of Bayesian colocalization analysis, of publicly available GWAS summary statistics with the cytokine network associations revealed shared causal variants between the eight cytokine loci and other traits; in particular, cytokine network variants at the ABO, SERPINE2, and ZFPM2 loci showed pleiotropic effects on the production of immune-related proteins, on metabolic traits such as lipoprotein and lipid levels, on blood-cell-related traits such as platelet count, and on disease traits such as coronary artery disease and type 2 diabetes.


Subject(s)
Biomarkers/analysis , Cardiovascular Diseases/genetics , Cytokines/genetics , Genetic Pleiotropy , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Adolescent , Adult , Aged , Blood Proteins/genetics , Blood Proteins/immunology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/pathology , Child , Cytokines/immunology , Female , Follow-Up Studies , Gene Regulatory Networks , Genetic Predisposition to Disease , Genome, Human , Humans , Longitudinal Studies , Male , Middle Aged , Prognosis , Prospective Studies , Young Adult
7.
PLoS One ; 14(10): e0223692, 2019.
Article in English | MEDLINE | ID: mdl-31644575

ABSTRACT

BACKGROUND: GlycA is a nuclear magnetic resonance (NMR) spectroscopy biomarker that predicts risk of disease from myriad causes. It is heterogeneous; arising from five circulating glycoproteins with dynamic concentrations: alpha-1 antitrypsin (AAT), alpha-1-acid glycoprotein (AGP), haptoglobin (HP), transferrin (TF), and alpha-1-antichymotrypsin (AACT). The contributions of each glycoprotein to the disease and mortality risks predicted by GlycA remain unknown. METHODS: We trained imputation models for AAT, AGP, HP, and TF from NMR metabolite measurements in 626 adults from a population cohort with matched NMR and immunoassay data. Levels of AAT, AGP, and HP were estimated in 11,861 adults from two population cohorts with eight years of follow-up, then each biomarker was tested for association with all common endpoints. Whole blood gene expression data was used to identify cellular processes associated with elevated AAT. RESULTS: Accurate imputation models were obtained for AAT, AGP, and HP but not for TF. While AGP had the strongest correlation with GlycA, our analysis revealed variation in imputed AAT levels was the most predictive of morbidity and mortality for the widest range of diseases over the eight year follow-up period, including heart failure (meta-analysis hazard ratio = 1.60 per standard deviation increase of AAT, P-value = 1×10-10), influenza and pneumonia (HR = 1.37, P = 6×10-10), and liver diseases (HR = 1.81, P = 1×10-6). Transcriptional analyses revealed association of elevated AAT with diverse inflammatory immune pathways. CONCLUSIONS: This study clarifies the molecular underpinnings of the GlycA biomarker's associated disease risk, and indicates a previously unrecognised association between elevated AAT and severe disease onset and mortality.


Subject(s)
Biomarkers , alpha 1-Antitrypsin/blood , Disease Susceptibility , Female , Glycoproteins , Humans , Magnetic Resonance Spectroscopy , Male , Morbidity , Mortality , Orosomucoid/adverse effects , Proportional Hazards Models
8.
PLoS One ; 14(2): e0210495, 2019.
Article in English | MEDLINE | ID: mdl-30742629

ABSTRACT

Tissue-resident CD8+ memory T (TRM) cells are immune cells that permanently reside at tissue sites where they play an important role in providing rapid protection against reinfection. They are not only phenotypically and functionally distinct from their circulating memory counterparts, but also exhibit a unique transcriptional profile. To date, the local tissue signals required for their development and long-term residency are not well understood. So far, the best-characterised tissue-derived signal is transforming growth factor-ß (TGF-ß), which has been shown to promote the development of these cells within tissues. In this study, we aimed to determine to what extent the transcriptional signatures of TRM cells from multiple tissues reflects TGF-ß imprinting. We activated murine CD8+ T cells, stimulated them in vitro by TGF-ß, and profiled their transcriptomes using RNA-seq. Upon comparison, we identified a TGF-ß-induced signature of differentially expressed genes between TGF-ß-stimulated and -unstimulated cells. Next, we linked this in vitro TGF-ß-induced signature to a previously identified in vivo TRM-specific gene set and found considerable (>50%) overlap between the two gene sets, thus showing that a substantial part of the TRM signature can be attributed to TGF-ß signalling. Finally, gene set enrichment analysis further revealed that the altered gene signature following TGF-ß exposure reflected transcriptional signatures found in TRM cells from both epithelial and non-epithelial tissues. In summary, these findings show that TGF-ß has a broad footprint in establishing the residency-specific transcriptional profile of TRM cells, which is detectable in TRM cells from diverse tissues. They further suggest that constitutive TGF-ß signaling might be involved for their long-term persistence at tissue sites.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Transcriptome , Transforming Growth Factor beta/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Female , Gene Expression Regulation , Immunologic Memory , Mice, Inbred C57BL
9.
PLoS Genet ; 14(8): e1007607, 2018 08.
Article in English | MEDLINE | ID: mdl-30142156

ABSTRACT

Schizophrenia and the affective disorders, here comprising bipolar disorder and major depressive disorder, are psychiatric illnesses that lead to significant morbidity and mortality worldwide. Whilst understanding of their pathobiology remains limited, large case-control studies have recently identified single nucleotide polymorphisms (SNPs) associated with these disorders. However, discerning the functional effects of these SNPs has been difficult as the associated causal genes are unknown. Here we evaluated whether schizophrenia and affective disorder associated-SNPs are correlated with gene expression within human brain tissue. Specifically, to identify expression quantitative trait loci (eQTLs), we leveraged disorder-associated SNPs identified from 11 genome-wide association studies with gene expression levels in post-mortem, neurologically-normal tissue from two independent human brain tissue expression datasets (UK Brain Expression Consortium (UKBEC) and Genotype-Tissue Expression (GTEx)). Utilizing stringent multi-region meta-analyses, we identified 2,224 cis-eQTLs associated with expression of 40 genes, including 11 non-coding RNAs. One cis-eQTL, rs16969968, results in a functionally disruptive missense mutation in CHRNA5, a schizophrenia-implicated gene. Importantly, comparing across tissues, we find that blood eQTLs capture < 10% of brain cis-eQTLs. Contrastingly, > 30% of brain-associated eQTLs are significant in tibial nerve. This study identifies putatively causal genes whose expression in region-specific tissue may contribute to the risk of schizophrenia and affective disorders.


Subject(s)
Mood Disorders/genetics , Quantitative Trait Loci , Schizophrenia/genetics , Brain/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Genotype , Humans , Mood Disorders/diagnosis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Polymorphism, Single Nucleotide , Principal Component Analysis , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Schizophrenia/diagnosis
10.
Nat Genet ; 50(6): 849-856, 2018 06.
Article in English | MEDLINE | ID: mdl-29785015

ABSTRACT

To examine the transmission dynamics of Mycobacterium tuberculosis (Mtb) isolated from tuberculosis patients in Ho Chi Minh City, Vietnam, we sequenced the whole genomes of 1,635 isolates and compared these with 3,144 isolates from elsewhere. The data identify an underlying burden of disease caused by the endemic Mtb lineage 1 associated with the activation of long-term latent infection, and a threefold higher burden associated with the more recently introduced Beijing lineage and lineage 4 Mtb strains. We find that Beijing lineage Mtb is frequently transferred between Vietnam and other countries, and detect higher levels of transmission of Beijing lineage strains within this host population than the endemic lineage 1 Mtb. Screening for parallel evolution of Beijing lineage-associated SNPs in other Mtb lineages as a signal of positive selection, we identify an alteration in the ESX-5 type VII-secreted protein EsxW, which could potentially contribute to the enhanced transmission of Beijing lineage Mtb in Vietnamese and other host populations.


Subject(s)
Mycobacterium tuberculosis/genetics , Tuberculosis/microbiology , Beijing , DNA, Bacterial/genetics , Genotype , Humans , Polymorphism, Single Nucleotide , Tuberculosis/transmission , Vietnam
11.
Genome Biol ; 18(1): 146, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28764798

ABSTRACT

BACKGROUND: Immunometabolism plays a central role in many cardiometabolic diseases. However, a robust map of immune-related gene networks in circulating human cells, their interactions with metabolites, and their genetic control is still lacking. Here, we integrate blood transcriptomic, metabolomic, and genomic profiles from two population-based cohorts (total N = 2168), including a subset of individuals with matched multi-omic data at 7-year follow-up. RESULTS: We identify topologically replicable gene networks enriched for diverse immune functions including cytotoxicity, viral response, B cell, platelet, neutrophil, and mast cell/basophil activity. These immune gene modules show complex patterns of association with 158 circulating metabolites, including lipoprotein subclasses, lipids, fatty acids, amino acids, small molecules, and CRP. Genome-wide scans for module expression quantitative trait loci (mQTLs) reveal five modules with mQTLs that have both cis and trans effects. The strongest mQTL is in ARHGEF3 (rs1354034) and affects a module enriched for platelet function, independent of platelet counts. Modules of mast cell/basophil and neutrophil function show temporally stable metabolite associations over 7-year follow-up, providing evidence that these modules and their constituent gene products may play central roles in metabolic inflammation. Furthermore, the strongest mQTL in ARHGEF3 also displays clear temporal stability, supporting widespread trans effects at this locus. CONCLUSIONS: This study provides a detailed map of natural variation at the blood immunometabolic interface and its genetic basis, and may facilitate subsequent studies to explain inter-individual variation in cardiometabolic disease.


Subject(s)
Cardiovascular Diseases/genetics , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Metabolic Syndrome/genetics , Metabolome/genetics , Quantitative Trait Loci/immunology , Amino Acids/immunology , Amino Acids/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Basophils/immunology , Basophils/pathology , Blood Platelets/immunology , Blood Platelets/pathology , C-Reactive Protein/genetics , C-Reactive Protein/immunology , Cardiovascular Diseases/immunology , Cardiovascular Diseases/metabolism , Fatty Acids/immunology , Fatty Acids/metabolism , Follow-Up Studies , Gene Ontology , Genome, Human , Humans , Immunity, Innate , Lipoproteins/genetics , Lipoproteins/immunology , Metabolic Syndrome/immunology , Metabolic Syndrome/metabolism , Metabolome/immunology , Neutrophils/immunology , Neutrophils/pathology , Polymorphism, Single Nucleotide , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/immunology
12.
Cell Syst ; 1(4): 293-301, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-27136058

ABSTRACT

The biomarker glycoprotein acetylation (GlycA) has been shown to predict risk of cardiovascular disease and all-cause mortality. Here, we characterize biological processes associated with GlycA by leveraging population-based omics data and health records from >10,000 individuals. Our analyses show that GlycA levels are chronic within individuals for up to a decade. In apparently healthy individuals, elevated GlycA corresponded to elevation of myriad inflammatory cytokines, as well as a gene coexpression network indicative of increased neutrophil activity, suggesting that individuals with high GlycA may be in a state of chronic inflammatory response. Accordingly, analysis of infection-related hospitalization and death records showed that increased GlycA increased long-term risk of severe non-localized and respiratory infections, particularly septicaemia and pneumonia. In total, our work demonstrates that GlycA is a biomarker for chronic inflammation, neutrophil activity, and risk of future severe infection. It also illustrates the utility of leveraging multi-layered omics data and health records to elucidate the molecular and cellular processes associated with biomarkers.

13.
PLoS Genet ; 9(3): e1003362, 2013.
Article in English | MEDLINE | ID: mdl-23516379

ABSTRACT

We describe a novel approach to capturing the covariance structure of peripheral blood gene expression that relies on the identification of highly conserved Axes of variation. Starting with a comparison of microarray transcriptome profiles for a new dataset of 189 healthy adult participants in the Emory-Georgia Tech Center for Health Discovery and Well-Being (CHDWB) cohort, with a previously published study of 208 adult Moroccans, we identify nine Axes each with between 99 and 1,028 strongly co-regulated transcripts in common. Each axis is enriched for gene ontology categories related to sub-classes of blood and immune function, including T-cell and B-cell physiology and innate, adaptive, and anti-viral responses. Conservation of the Axes is demonstrated in each of five additional population-based gene expression profiling studies, one of which is robustly associated with Body Mass Index in the CHDWB as well as Finnish and Australian cohorts. Furthermore, ten tightly co-regulated genes can be used to define each Axis as "Blood Informative Transcripts" (BITs), generating scores that define an individual with respect to the represented immune activity and blood physiology. We show that environmental factors, including lifestyle differences in Morocco and infection leading to active or latent tuberculosis, significantly impact specific axes, but that there is also significant heritability for the Axis scores. In the context of personalized medicine, reanalysis of the longitudinal profile of one individual during and after infection with two respiratory viruses demonstrates that specific axes also characterize clinical incidents. This mode of analysis suggests the view that, rather than unique subsets of genes marking each class of disease, differential expression reflects movement along the major normal Axes in response to environmental and genetic stimuli.


Subject(s)
Blood Proteins , Gene Expression Profiling , Gene Expression , Adult , Australia , B-Lymphocytes/metabolism , Blood Proteins/classification , Blood Proteins/genetics , Blood Proteins/metabolism , Body Mass Index , Female , Georgia , Humans , Microarray Analysis , Morocco , Precision Medicine
SELECTION OF CITATIONS
SEARCH DETAIL
...