Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
medRxiv ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39072045

ABSTRACT

Discerning the mechanisms driving type 2 diabetes (T2D) pathophysiology from genome-wide association studies (GWAS) remains a challenge. To this end, we integrated omics information from 16 multi-tissue and multi-ancestry expression, protein, and metabolite quantitative trait loci (QTL) studies and 46 multi-ancestry GWAS for T2D-related traits with the largest, most ancestrally diverse T2D GWAS to date. Of the 1,289 T2D GWAS index variants, 716 (56%) demonstrated strong evidence of colocalization with a molecular or T2D-related trait, implicating 657 cis-effector genes, 1,691 distal-effector genes, 731 metabolites, and 43 T2D-related traits. We identified 773 of these cis- and distal-effector genes using either expression QTL data from understudied ancestry groups or inclusion of T2D index variants enriched in underrepresented populations, emphasizing the value of increasing population diversity in functional mapping. Linking these variants, genes, metabolites, and traits into a network, we elucidated mechanisms through which T2D-associated variation may impact disease risk. Finally, we showed that drugs targeting effector proteins were enriched in those approved to treat T2D, highlighting the potential of these results to prioritize drug targets for T2D. These results represent a leap in the molecular characterization of T2D-associated genetic variation and will aid in translating genetic findings into novel therapeutic strategies.

2.
Nature ; 628(8006): 130-138, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448586

ABSTRACT

Genome-wide association analyses using high-throughput metabolomics platforms have led to novel insights into the biology of human metabolism1-7. This detailed knowledge of the genetic determinants of systemic metabolism has been pivotal for uncovering how genetic pathways influence biological mechanisms and complex diseases8-11. Here we present a genome-wide association study for 233 circulating metabolic traits quantified by nuclear magnetic resonance spectroscopy in up to 136,016 participants from 33 cohorts. We identify more than 400 independent loci and assign probable causal genes at two-thirds of these using manual curation of plausible biological candidates. We highlight the importance of sample and participant characteristics that can have significant effects on genetic associations. We use detailed metabolic profiling of lipoprotein- and lipid-associated variants to better characterize how known lipid loci and novel loci affect lipoprotein metabolism at a granular level. We demonstrate the translational utility of comprehensively phenotyped molecular data, characterizing the metabolic associations of intrahepatic cholestasis of pregnancy. Finally, we observe substantial genetic pleiotropy for multiple metabolic pathways and illustrate the importance of careful instrument selection in Mendelian randomization analysis, revealing a putative causal relationship between acetone and hypertension. Our publicly available results provide a foundational resource for the community to examine the role of metabolism across diverse diseases.


Subject(s)
Biomarkers , Genome-Wide Association Study , Metabolomics , Female , Humans , Pregnancy , Acetone/blood , Acetone/metabolism , Biomarkers/blood , Biomarkers/metabolism , Cholestasis, Intrahepatic/blood , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Cohort Studies , Genome-Wide Association Study/methods , Hypertension/blood , Hypertension/genetics , Hypertension/metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Magnetic Resonance Spectroscopy , Mendelian Randomization Analysis , Metabolic Networks and Pathways/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Pregnancy Complications/blood , Pregnancy Complications/genetics , Pregnancy Complications/metabolism
3.
Nature ; 627(8003): 347-357, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38374256

ABSTRACT

Type 2 diabetes (T2D) is a heterogeneous disease that develops through diverse pathophysiological processes1,2 and molecular mechanisms that are often specific to cell type3,4. Here, to characterize the genetic contribution to these processes across ancestry groups, we aggregate genome-wide association study data from 2,535,601 individuals (39.7% not of European ancestry), including 428,452 cases of T2D. We identify 1,289 independent association signals at genome-wide significance (P < 5 × 10-8) that map to 611 loci, of which 145 loci are, to our knowledge, previously unreported. We define eight non-overlapping clusters of T2D signals that are characterized by distinct profiles of cardiometabolic trait associations. These clusters are differentially enriched for cell-type-specific regions of open chromatin, including pancreatic islets, adipocytes, endothelial cells and enteroendocrine cells. We build cluster-specific partitioned polygenic scores5 in a further 279,552 individuals of diverse ancestry, including 30,288 cases of T2D, and test their association with T2D-related vascular outcomes. Cluster-specific partitioned polygenic scores are associated with coronary artery disease, peripheral artery disease and end-stage diabetic nephropathy across ancestry groups, highlighting the importance of obesity-related processes in the development of vascular outcomes. Our findings show the value of integrating multi-ancestry genome-wide association study data with single-cell epigenomics to disentangle the aetiological heterogeneity that drives the development and progression of T2D. This might offer a route to optimize global access to genetically informed diabetes care.


Subject(s)
Diabetes Mellitus, Type 2 , Disease Progression , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Adipocytes/metabolism , Chromatin/genetics , Chromatin/metabolism , Coronary Artery Disease/complications , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 2/classification , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/complications , Diabetic Nephropathies/genetics , Endothelial Cells/metabolism , Enteroendocrine Cells , Epigenomics , Genetic Predisposition to Disease/genetics , Islets of Langerhans/metabolism , Multifactorial Inheritance/genetics , Peripheral Arterial Disease/complications , Peripheral Arterial Disease/genetics , Single-Cell Analysis
4.
medRxiv ; 2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37425837

ABSTRACT

Metabolites are small molecules that are useful for estimating disease risk and elucidating disease biology. Nevertheless, their causal effects on human diseases have not been evaluated comprehensively. We performed two-sample Mendelian randomization to systematically infer the causal effects of 1,099 plasma metabolites measured in 6,136 Finnish men from the METSIM study on risk of 2,099 binary disease endpoints measured in 309,154 Finnish individuals from FinnGen. We identified evidence for 282 causal effects of 70 metabolites on 183 disease endpoints (FDR<1%). We found 25 metabolites with potential causal effects across multiple disease domains, including ascorbic acid 2-sulfate affecting 26 disease endpoints in 12 disease domains. Our study suggests that N-acetyl-2-aminooctanoate and glycocholenate sulfate affect risk of atrial fibrillation through two distinct metabolic pathways and that N-methylpipecolate may mediate the causal effect of N6, N6-dimethyllysine on anxious personality disorder. This study highlights the broad causal impact of plasma metabolites and widespread metabolic connections across diseases.

5.
Asian J Pharm Sci ; 18(3): 100800, 2023 May.
Article in English | MEDLINE | ID: mdl-37274924

ABSTRACT

Glioblastoma is acknowledged as the most aggressive cerebral tumor in adults. However, the efficacy of current standard therapy is seriously undermined by drug resistance and suppressive immune microenvironment. Ferroptosis is a recently discovered form of iron-dependent cell death that may have excellent prospect as chemosensitizer. The utilization of ferropotosis inducer Erastin could significantly mediate chemotherapy sensitization of Temozolomide and exert anti-tumor effects in glioblastoma. In this study, a combination of hydrogel-liposome nanoplatform encapsulated with Temozolomide and ferroptosis inducer Erastin was constructed. The αvß3 integrin-binding peptide cyclic RGD was utilized to modify codelivery system to achieve glioblastoma targeting strategy. As biocompatible drug reservoirs, cross-linked GelMA (gelatin methacrylamide) hydrogel and cRGD-coated liposome realized the sustained release of internal contents. In the modified intracranial tumor resection model, GelMA-liposome system achieved slow release of Temozolomide and Erastin in situ for more than 14 d. The results indicated that nanoplatform (T+E@LPs-cRGD+GelMA) improved glioblastoma sensitivity to chemotherapeutic temozolomide and exerted satisfactory anti-tumor effects. It was demonstrated that the induction of ferroptosis could be utilized as a therapeutic strategy to overcome drug resistance. Furthermore, transcriptome sequencing was conducted to reveal the underlying mechanism that the nanoplatform (T+E@LPs-cRGD+GelMA) implicated in. It is suggested that GelMA-liposome system participated in the immune response and immunomodulation of glioblastoma via interferon/PD-L1 pathway. Collectively, this study proposed a potential combinatory therapeutic strategy for glioblastoma treatment.

6.
Diabetologia ; 66(8): 1472-1480, 2023 08.
Article in English | MEDLINE | ID: mdl-37280435

ABSTRACT

AIMS/HYPOTHESIS: Determining how high BMI at different time points influences the risk of developing type 2 diabetes and affects insulin secretion and insulin sensitivity is critical. METHODS: By estimating childhood BMI in 441,761 individuals in the UK Biobank, we identified which genetic variants had larger effects on adulthood BMI than on childhood BMI, and vice versa. All genome-wide significant genetic variants were then used to separate the independent genetic effects of high childhood BMI from those of high adulthood BMI on the risk of type 2 diabetes and insulin-related phenotypes using Mendelian randomisation. We performed two-sample MR using external studies of type 2 diabetes, and oral and intravenous measures of insulin secretion and sensitivity. RESULTS: We found that a childhood BMI that was one standard deviation (1.97 kg/m2) higher than the mean, corrected for the independent genetic liability to adulthood BMI, was associated with a protective effect for seven measures of insulin sensitivity and secretion, including increased insulin sensitivity index (ß=0.15; 95% CI 0.067, 0.225; p=2.79×10-4) and reduced fasting glucose levels (ß=-0.053; 95% CI -0.089, -0.017; p=4.31×10-3). However, there was little to no evidence of a direct protective effect on type 2 diabetes (OR 0.94; 95% CI 0.85, 1.04; p=0.228) independently of genetic liability to adulthood BMI. CONCLUSIONS/INTERPRETATION: Our results provide evidence of the protective effect of higher childhood BMI on insulin secretion and sensitivity, which are crucial intermediate diabetes traits. However, we stress that our results should not currently lead to any change in public health or clinical practice, given the uncertainty regarding the biological pathway of these effects and the limitations of this type of study.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/genetics , Body Mass Index , Phenotype , Insulin/genetics , Mendelian Randomization Analysis , Genome-Wide Association Study , Polymorphism, Single Nucleotide
7.
Nat Genet ; 55(6): 973-983, 2023 06.
Article in English | MEDLINE | ID: mdl-37291194

ABSTRACT

Distinct tissue-specific mechanisms mediate insulin action in fasting and postprandial states. Previous genetic studies have largely focused on insulin resistance in the fasting state, where hepatic insulin action dominates. Here we studied genetic variants influencing insulin levels measured 2 h after a glucose challenge in >55,000 participants from three ancestry groups. We identified ten new loci (P < 5 × 10-8) not previously associated with postchallenge insulin resistance, eight of which were shown to share their genetic architecture with type 2 diabetes in colocalization analyses. We investigated candidate genes at a subset of associated loci in cultured cells and identified nine candidate genes newly implicated in the expression or trafficking of GLUT4, the key glucose transporter in postprandial glucose uptake in muscle and fat. By focusing on postprandial insulin resistance, we highlighted the mechanisms of action at type 2 diabetes loci that are not adequately captured by studies of fasting glycemic traits.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Insulin/genetics , Genome-Wide Association Study , Insulin Resistance/genetics , Diabetes Mellitus, Type 2/genetics , Glucose/metabolism , Blood Glucose/genetics
8.
medRxiv ; 2023 Mar 31.
Article in English | MEDLINE | ID: mdl-37034649

ABSTRACT

Type 2 diabetes (T2D) is a heterogeneous disease that develops through diverse pathophysiological processes. To characterise the genetic contribution to these processes across ancestry groups, we aggregate genome-wide association study (GWAS) data from 2,535,601 individuals (39.7% non-European ancestry), including 428,452 T2D cases. We identify 1,289 independent association signals at genome-wide significance (P<5×10-8) that map to 611 loci, of which 145 loci are previously unreported. We define eight non-overlapping clusters of T2D signals characterised by distinct profiles of cardiometabolic trait associations. These clusters are differentially enriched for cell-type specific regions of open chromatin, including pancreatic islets, adipocytes, endothelial, and enteroendocrine cells. We build cluster-specific partitioned genetic risk scores (GRS) in an additional 137,559 individuals of diverse ancestry, including 10,159 T2D cases, and test their association with T2D-related vascular outcomes. Cluster-specific partitioned GRS are more strongly associated with coronary artery disease and end-stage diabetic nephropathy than an overall T2D GRS across ancestry groups, highlighting the importance of obesity-related processes in the development of vascular outcomes. Our findings demonstrate the value of integrating multi-ancestry GWAS with single-cell epigenomics to disentangle the aetiological heterogeneity driving the development and progression of T2D, which may offer a route to optimise global access to genetically-informed diabetes care.

9.
Discov Nano ; 18(1): 40, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36969494

ABSTRACT

Abstract: Ischemic stroke is one of the most severe neurological disorders with limited therapeutic strategies. The utilization of nanoparticle drug delivery systems is a burgeoning field and has been widely investigated. Among these, biomimetic drug delivery systems composed of biogenic membrane components and synthetic nanoparticles have been extensively highlighted in recent years. Biomimetic membrane camouflage presents an effective strategy to prolong circulation, reduce immunogenicity and enhance targeting. For one thing, biomimetic nanoparticles reserve the physical and chemical properties of intrinsic nanoparticle. For another, the biological functions of original source cells are completely inherited. Compared to conventional surface modification methods, this approach is more convenient and biocompatible. In this review, membrane-based nanoparticles derived from different donor cells were exemplified. The prospect of future biomimetic nanoparticles in ischemic stroke therapy was discussed.

10.
J Neurol Neurosurg Psychiatry ; 94(7): 526-531, 2023 07.
Article in English | MEDLINE | ID: mdl-36732044

ABSTRACT

BACKGROUND: There are currently no specific biomarkers for multiple sclerosis (MS). Identifying robust biomarkers for MS is crucial to improve disease diagnosis and management. METHODS: This study first used six Mendelian randomisation methods to assess causal relationship of 174 metabolites with MS, incorporating data from European-ancestry metabolomics (n=8569-86 507) and MS (n=14 802 MS cases, 26 703 controls) genomewide association studies. Genetic scores for identified causal metabolite(s) were then computed to predict MS disability progression in an independent longitudinal cohort (AusLong study) of 203 MS cases with up to 15-year follow-up. RESULTS: We found a novel genetic causal effect of serine on MS onset (OR=1.67, 95% CI 1.51 to 1.84, p=1.73×10-20), such that individuals whose serine level is 1 SD above the population mean will have 1.67 times the risk of developing MS. This is robust across all sensitivity methods (OR ranges from 1.49 to 1.67). In an independent longitudinal MS cohort, we then constructed time-dynamic and time-fixed genetic scores based on serine genetic instrument single-nucleotide polymorphisms, where higher scores for raised serum serine level were associated with increased risk of disability worsening, especially in the time-dynamic model (RR=1.25, 95% CI 1.10 to 1.42, p=7.52×10-4). CONCLUSIONS: These findings support investigating serine as an important candidate biomarker for MS onset and disability progression.


Subject(s)
Disabled Persons , Multiple Sclerosis , Humans , Multiple Sclerosis/diagnosis , Multiple Sclerosis/genetics , Causality , Metabolomics , Biomarkers , Disease Progression
11.
medRxiv ; 2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36798216

ABSTRACT

Determining how high body-mass index (BMI) at different time points influences the risk of developing type two diabetes (T2D), and affects insulin secretion and insulin sensitivity, is critical. By estimating childhood BMI in 441,761 individuals in the UK Biobank, we identified which genetic variants had larger effects on adulthood BMI than on childhood BMI, and vice-versa. All genome-wide significant genetic variants were then used to separate the independent genetic effects of high childhood BMI from high adulthood BMI on the risk of T2D and insulin related phenotypes using Mendelian randomisation and studies of T2D, and oral and intravenous measures of insulin secretion and sensitivity. We found that a 1.s.d. (= 1.97kg/m 2 ) higher childhood BMI, corrected for the independent genetic liability to adulthood BMI, was associated with a protective effect for seven measures of insulin sensitivity and secretion, including an increased insulin sensitivity index (ß = 0.15 [0.067, 0.225], p = 2.79×10 -4 ), and reduced fasting glucose (ß = -0.053 [-0.089, -0.017], p = 4.31×10 -3 ). There was however little to no evidence of a direct protective effect on T2D (OR = 0.94 [0.85 - 1.04], p = 0.228), independently of genetic liability to adulthood BMI. Our results thus cumulatively provide evidence of the protective effect of higher childhood BMI on insulin secretion and sensitivity, which are crucial intermediate diabetes traits. However, we stress that our results should not currently lead to any change in public health or clinical practice, given the uncertainty in biological pathway of these effects, and the limitations of this type of study. Research in Context: High BMI in adulthood is associated with higher risk of type two diabetes, coupled with lower insulin sensitivity and secretion.Richardson et al [2020] used genetics to show that high BMI in childhood does not appear to increase the risk of type diabetes independently from its effect on adult BMI.We asked: does high childhood BMI affect insulin related traits such as fasting glucose and insulin sensitivity, independently of adulthood BMI?We used genetics to show that high childhood BMI has a protective effect on seven insulin sensitivity and secretion traits, including fasting glucose and measures of insulin sensitivity and secretion, independently of adulthood BMI.Our work has the potential to turn conventional understanding on its head - high BMI in childhood improves insulin sensitivity (when adjusting for knock on effects to high adult BMI) and opens up important questions about plasticity in childhood and compensatory mechanisms.

13.
Am J Hum Genet ; 110(2): 284-299, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36693378

ABSTRACT

Insulin secretion is critical for glucose homeostasis, and increased levels of the precursor proinsulin relative to insulin indicate pancreatic islet beta-cell stress and insufficient insulin secretory capacity in the setting of insulin resistance. We conducted meta-analyses of genome-wide association results for fasting proinsulin from 16 European-ancestry studies in 45,861 individuals. We found 36 independent signals at 30 loci (p value < 5 × 10-8), which validated 12 previously reported loci for proinsulin and ten additional loci previously identified for another glycemic trait. Half of the alleles associated with higher proinsulin showed higher rather than lower effects on glucose levels, corresponding to different mechanisms. Proinsulin loci included genes that affect prohormone convertases, beta-cell dysfunction, vesicle trafficking, beta-cell transcriptional regulation, and lysosomes/autophagy processes. We colocalized 11 proinsulin signals with islet expression quantitative trait locus (eQTL) data, suggesting candidate genes, including ARSG, WIPI1, SLC7A14, and SIX3. The NKX6-3/ANK1 proinsulin signal colocalized with a T2D signal and an adipose ANK1 eQTL signal but not the islet NKX6-3 eQTL. Signals were enriched for islet enhancers, and we showed a plausible islet regulatory mechanism for the lead signal in the MADD locus. These results show how detailed genetic studies of an intermediate phenotype can elucidate mechanisms that may predispose one to disease.


Subject(s)
Diabetes Mellitus, Type 2 , Proinsulin , Humans , Proinsulin/genetics , Proinsulin/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Genome-Wide Association Study/methods , Insulin/genetics , Insulin/metabolism , Glucose , Transcription Factors/genetics , Homeodomain Proteins/genetics
14.
Am J Hum Genet ; 110(1): 44-57, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36608684

ABSTRACT

Integrative genetic association methods have shown great promise in post-GWAS (genome-wide association study) analyses, in which one of the most challenging tasks is identifying putative causal genes and uncovering molecular mechanisms of complex traits. Recent studies suggest that prevailing computational approaches, including transcriptome-wide association studies (TWASs) and colocalization analysis, are individually imperfect, but their joint usage can yield robust and powerful inference results. This paper presents INTACT, a computational framework to integrate probabilistic evidence from these distinct types of analyses and implicate putative causal genes. This procedure is flexible and can work with a wide range of existing integrative analysis approaches. It has the unique ability to quantify the uncertainty of implicated genes, enabling rigorous control of false-positive discoveries. Taking advantage of this highly desirable feature, we further propose an efficient algorithm, INTACT-GSE, for gene set enrichment analysis based on the integrated probabilistic evidence. We examine the proposed computational methods and illustrate their improved performance over the existing approaches through simulation studies. We apply the proposed methods to analyze the multi-tissue eQTL data from the GTEx project and eight large-scale complex- and molecular-trait GWAS datasets from multiple consortia and the UK Biobank. Overall, we find that the proposed methods markedly improve the existing putative gene implication methods and are particularly advantageous in evaluating and identifying key gene sets and biological pathways underlying complex traits.


Subject(s)
Genome-Wide Association Study , Transcriptome , Humans , Transcriptome/genetics , Genome-Wide Association Study/methods , Multifactorial Inheritance/genetics , Quantitative Trait Loci/genetics , Computer Simulation , Polymorphism, Single Nucleotide/genetics , Genetic Predisposition to Disease
16.
Cell Death Differ ; 30(1): 54-68, 2023 01.
Article in English | MEDLINE | ID: mdl-35871232

ABSTRACT

Glioblastoma multiforme (GBM) is acknowledged as the most aggressive primary brain tumor in adults. It is typically characterized by the high heterogeneity which corresponds to extensive genetic mutations and complex alternative splicing (AS) profiles. Known as a major repressive splicing factor in AS, polypyrimidine tract-binding protein 1 (PTBP1) is involved in the exon skipping events of multiple precursor mRNAs (pre-mRNAs) in GBM. However, precise mechanisms that modulate the expression and activity of PTBP1 remain to be elucidated. In present study, we provided evidences for the role of a long intergenic noncoding RNA (LINREP) implicated in the regulation of PTBP1-induced AS. LINREP interacted with PTBP1 and human antigen R (HuR, ELAVL1) protein complex and protected PTBP1 from the ubiquitin-proteasome degradation. Consequently, a broad spectrum of PTBP1-induced spliced variants was generated by exon skipping, especially for the skipping of reticulon 4 (RTN4) exon 3. Interestingly, LINREP also promoted the dissociation of nuclear UPF1 from PTBP1, which increased the binding of PTBP1 to RTN4 transcripts, thus enhancing the skipping of RTN4 exon 3 to some extent. Besides, HuR recruitment was essential for the stabilization of LINREP via a manner dependent on N6-methyladenosine (m6A) formation and identification. Taken together, our results demonstrated the functional significance of LINREP in human GBM for its dual regulation of PTBP1-induced AS and its m6A modification modality, implicating that HuR/LINREP/PTBP1 axis might serve as a potential therapeutic target for GBM.


Subject(s)
Glioblastoma , RNA, Long Noncoding , Adult , Humans , Glioblastoma/genetics , Glioblastoma/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Alternative Splicing/genetics , Trans-Activators/metabolism , RNA Helicases/metabolism
17.
18.
Nutrients ; 14(24)2022 Dec 11.
Article in English | MEDLINE | ID: mdl-36558443

ABSTRACT

BACKGROUND: Vitamin D level has been reported to be associated with psoriasis, atopic dermatitis, and vitiligo. However, its causal relationship with the risk of these three diseases remains unclear. METHODS: We obtained genome-wide association statistics for three measures of circulating vitamin D levels (25(OH)D in 120,618 individuals, and 25(OH)D3 and epimeric form C3-epi-25(OH)D3 in 40,562 individuals) and for the diseases psoriasis (3871 cases and 333,288 controls), atopic dermatitis (21,399 cases and 95,464 controls), and vitiligo (4680 cases and 39,586 controls). We performed Mendelian randomization using inverse-variance weighted, weighted median, MR-Egger, and MR-pleiotropy residual sum and outlier methods. We carried out sensitivity analyses to evaluate the robustness of the results. RESULTS: We showed that elevated vitamin D levels protected individuals from developing psoriasis (OR = 0.995, p = 8.84 × 10-4 for 25(OH)D; OR = 0.997, p = 1.81 × 10-3 for 25(OH)D3; and OR = 0.998, p = 0.044 for C3-epi-25(OH)D3). Genetically predicted risk of atopic dermatitis increased the levels of 25(OH)D (OR = 1.040, p = 7.14 × 10-4) and 25(OH)D3 (OR = 1.208, p = 0.048). A sensitivity analysis suggested the robustness of these causal associations. CONCLUSIONS: This study reported causal relationships between circulating vitamin D levels and the risk of psoriasis, atopic dermatitis, and vitiligo. These findings provide potential disease intervention and monitoring targets.


Subject(s)
Dermatitis, Atopic , Psoriasis , Vitiligo , Humans , Vitamin D , Mendelian Randomization Analysis/methods , Dermatitis, Atopic/epidemiology , Dermatitis, Atopic/genetics , Vitiligo/epidemiology , Vitiligo/genetics , Genome-Wide Association Study , Vitamins , Psoriasis/epidemiology , Psoriasis/genetics , Polymorphism, Single Nucleotide
19.
Nature ; 610(7933): 704-712, 2022 10.
Article in English | MEDLINE | ID: mdl-36224396

ABSTRACT

Common single-nucleotide polymorphisms (SNPs) are predicted to collectively explain 40-50% of phenotypic variation in human height, but identifying the specific variants and associated regions requires huge sample sizes1. Here, using data from a genome-wide association study of 5.4 million individuals of diverse ancestries, we show that 12,111 independent SNPs that are significantly associated with height account for nearly all of the common SNP-based heritability. These SNPs are clustered within 7,209 non-overlapping genomic segments with a mean size of around 90 kb, covering about 21% of the genome. The density of independent associations varies across the genome and the regions of increased density are enriched for biologically relevant genes. In out-of-sample estimation and prediction, the 12,111 SNPs (or all SNPs in the HapMap 3 panel2) account for 40% (45%) of phenotypic variance in populations of European ancestry but only around 10-20% (14-24%) in populations of other ancestries. Effect sizes, associated regions and gene prioritization are similar across ancestries, indicating that reduced prediction accuracy is likely to be explained by linkage disequilibrium and differences in allele frequency within associated regions. Finally, we show that the relevant biological pathways are detectable with smaller sample sizes than are needed to implicate causal genes and variants. Overall, this study provides a comprehensive map of specific genomic regions that contain the vast majority of common height-associated variants. Although this map is saturated for populations of European ancestry, further research is needed to achieve equivalent saturation in other ancestries.


Subject(s)
Body Height , Chromosome Mapping , Polymorphism, Single Nucleotide , Humans , Body Height/genetics , Gene Frequency/genetics , Genome, Human/genetics , Genome-Wide Association Study , Haplotypes/genetics , Linkage Disequilibrium/genetics , Polymorphism, Single Nucleotide/genetics , Europe/ethnology , Sample Size , Phenotype
20.
Am J Hum Genet ; 109(10): 1727-1741, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36055244

ABSTRACT

Transcriptomics data have been integrated with genome-wide association studies (GWASs) to help understand disease/trait molecular mechanisms. The utility of metabolomics, integrated with transcriptomics and disease GWASs, to understand molecular mechanisms for metabolite levels or diseases has not been thoroughly evaluated. We performed probabilistic transcriptome-wide association and locus-level colocalization analyses to integrate transcriptomics results for 49 tissues in 706 individuals from the GTEx project, metabolomics results for 1,391 plasma metabolites in 6,136 Finnish men from the METSIM study, and GWAS results for 2,861 disease traits in 260,405 Finnish individuals from the FinnGen study. We found that genetic variants that regulate metabolite levels were more likely to influence gene expression and disease risk compared to the ones that do not. Integrating transcriptomics with metabolomics results prioritized 397 genes for 521 metabolites, including 496 previously identified gene-metabolite pairs with strong functional connections and suggested 33.3% of such gene-metabolite pairs shared the same causal variants with genetic associations of gene expression. Integrating transcriptomics and metabolomics individually with FinnGen GWAS results identified 1,597 genes for 790 disease traits. Integrating transcriptomics and metabolomics jointly with FinnGen GWAS results helped pinpoint metabolic pathways from genes to diseases. We identified putative causal effects of UGT1A1/UGT1A4 expression on gallbladder disorders through regulating plasma (E,E)-bilirubin levels, of SLC22A5 expression on nasal polyps and plasma carnitine levels through distinct pathways, and of LIPC expression on age-related macular degeneration through glycerophospholipid metabolic pathways. Our study highlights the power of integrating multiple sets of molecular traits and GWAS results to deepen understanding of disease pathophysiology.


Subject(s)
Genome-Wide Association Study , Transcriptome , Bilirubin , Carnitine , Glycerophospholipids , Humans , Male , Metabolomics , Quantitative Trait Loci/genetics , Solute Carrier Family 22 Member 5/genetics , Transcriptome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL