Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Commun Biol ; 7(1): 122, 2024 01 24.
Article in English | MEDLINE | ID: mdl-38267566

ABSTRACT

Type 2 diabetes (T2D) is known as one of the important risk factors for the severity and mortality of COVID-19. Here, we evaluate the impact of T2D and its genetic susceptibility on the severity and mortality of COVID-19, using 459,119 individuals in UK Biobank. Utilizing the polygenic risk scores (PRS) for T2D, we identified a significant association between T2D or T2D PRS, and COVID-19 severity. We further discovered the efficacy of vaccination and the pivotal role of T2D-related genetics in the pathogenesis of severe COVID-19. Moreover, we found that individuals with T2D or those in the high T2D PRS group had a significantly increased mortality rate. We also observed that the mortality rate for SARS-CoV-2-infected patients was approximately 2 to 7 times higher than for those not infected, depending on the time of infection. These findings emphasize the potential of T2D PRS in estimating the severity and mortality of COVID-19.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , COVID-19/epidemiology , COVID-19/genetics , UK Biobank , Biological Specimen Banks , SARS-CoV-2 , Genetic Predisposition to Disease , Genetic Risk Score
2.
J Am Heart Assoc ; : e030211, 2023 Nov 10.
Article in English | MEDLINE | ID: mdl-37947095

ABSTRACT

Background Patients with rheumatoid arthritis (RA) have a 2- to 10-fold increased risk of cardiovascular disease (CVD), but the biological mechanisms and existence of causality underlying such associations remain to be investigated. We aimed to investigate the genetic associations and underlying mechanisms between RA and CVD by leveraging large-scale genomic data and genetic cross-trait analytic approaches. Methods and Results Within UK Biobank data, we examined the genetic correlation, shared genetics, and potential causality between RA (Ncases=6754, Ncontrols=452 384) and cardiovascular diseases (CVD, Ncases=44 238, Ncontrols=414 900) using linkage disequilibrium score regression, cross-trait meta-analysis, and Mendelian randomization. We observed significant genetic correlations of RA with myocardial infarction (rg:0.40 [95% CI, 0.24-0.56), angina (rg:0.42 [95% CI, 0.28-0.56]), coronary heart diseases (rg:0.41 [95% CI, 0.27-0.55]), and CVD (rg:0.43 [95% CI, 0.29-0.57]) after correcting for multiple testing (P<0.05/5). When stratified by frequent use of analgesics, we found increased genetic correlation between RA and CVD among participants without aspirin usage (rg:0.54 [95% CI, 0.30-0.78] for angina; Pvalue=6.69×10-6) and among participants with paracetamol usage (rg:0.75 [95% CI, 0.20-1.29] for myocardial infarction; Pvalue=8.90×10-3), whereas others remained similar. Cross-trait meta-analysis identified 9 independent shared loci between RA and CVD, including PTPN22 at chr1p13.2, BCL2L11 at chr2q13, and CCR3 at chr3p21.31 (Psingle trait<1×10-3 and Pmeta<5×10-8), highlighting potential shared pathogenesis including accelerating atherosclerosis, upregulating oxidative stress, and vascular permeability. Finally, Mendelian randomization estimates showed limited evidence of causality between RA and CVD. Conclusions Our results supported shared genetic pathogenesis rather than causality in explaining the observed association between RA and CVD. The identified shared genetic factors provided insights into potential novel therapeutic target for RA-CVD comorbidities.

3.
Front Endocrinol (Lausanne) ; 14: 1165744, 2023.
Article in English | MEDLINE | ID: mdl-37680885

ABSTRACT

Introduction: The influence of dietary patterns measured using Recommended Food Score (RFS) with foods with high amounts of antioxidant nutrients for Type 2 diabetes (T2D) was analyzed. Our analysis aims to find associations between dietary patterns and T2D and conduct a gene-diet interaction analysis related to T2D. Methods: Data analyzed in the current study were obtained from the Korean Genome and Epidemiology Study Cohort. The dietary patterns of 46 food items were assessed using a validated food frequency questionnaire. To maximize the predictive power of the RFS, we propose two weighted food scores, namely HisCoM-RFS calculated using the novel Hierarchical Structural Component model (HisCoM) and PLSDA-RFS calculated using Partial Least Squares-Discriminant Analysis (PLS-DA) method. Results: Both RFS (OR: 1.11; 95% CI: 1.03- 1.20; P = 0.009) and PLSDA-RFS (OR: 1.10; 95% CI: 1.02-1.19, P = 0.011) were positively associated with T2D. Mapping of SNPs (P < 0.05) from the interaction analysis between SNPs and the food scores to genes and pathways yielded some 12 genes (CACNA2D3, RELN, DOCK2, SLIT3, CTNNA2, etc.) and pathways associated with T2D. The strongest association was observed with the adipocytokine signalling pathway, highlighting 32 genes (STAT3, MAPK10, MAPK8, IRS1, AKT1-3, ADIPOR2, etc.) most likely associated with T2D. Finally, the group of the subjects in low, intermediate and high using both the food scores and a polygenic risk score found an association between diet quality groups with issues at high genetic risk of T2D. Conclusion: A dietary pattern of poor amounts of antioxidant nutrients is associated with the risk of T2D, and diet affects pathway mechanisms involved in developing T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Antioxidants , Diet , Signal Transduction/genetics , Adipokines
4.
Genomics Inform ; 20(2): e16, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35794696

ABSTRACT

Various methodologies for the genetic analysis of longitudinal data have been proposed and applied to data from large-scale genome-wide association studies (GWAS) to identify single nucleotide polymorphisms (SNPs) associated with traits of interest and to detect SNP-time interactions. We recently proposed a grid-based Bayesian mixed model for longitudinal genetic data and showed that our Bayesian method increased the statistical power compared to the corresponding univariate method and well detected SNP-time interactions. In this paper, we further analyze longitudinal obesity-related traits such as body mass index, hip circumference, waist circumference, and waist-hip ratio from Korea Association Resource data to evaluate the proposed Bayesian method. We first conducted GWAS analyses of cross-sectional traits and combined the results of GWAS analyses through a meta-analysis based on a trajectory model and a random-effects model. We then applied our Bayesian method to a subset of SNPs selected by meta-analysis to further discover SNPs associated with traits of interest and SNP-time interactions. The proposed Bayesian method identified several novel SNPs associated with longitudinal obesity-related traits, and almost 25% of the identified SNPs had significant p-values for SNP-time interactions.

5.
Genetics ; 221(4)2022 07 30.
Article in English | MEDLINE | ID: mdl-35689615

ABSTRACT

We develop a computationally efficient alternative, TwinEQTL, to a linear mixed-effects model for twin genome-wide association study data. Instead of analyzing all twin samples together with linear mixed-effects model, TwinEQTL first splits twin samples into 2 independent groups on which multiple linear regression analysis can be validly performed separately, followed by an appropriate meta-analysis-like approach to combine the 2 nonindependent test results. Through mathematical derivations, we prove the validity of TwinEQTL algorithm and show that the correlation between 2 dependent test statistics at each single-nucleotide polymorphism is independent of its minor allele frequency. Thus, the correlation is constant across all single-nucleotide polymorphisms. Through simulations, we show empirically that TwinEQTL has well controlled type I error with negligible power loss compared with the gold-standard linear mixed-effects models. To accommodate expression quantitative loci analysis with twin subjects, we further implement TwinEQTL into an R package with much improved computational efficiency. Our approaches provide a significant leap in terms of computing speed for genome-wide association study and expression quantitative loci analysis with twin samples.


Subject(s)
Genome-Wide Association Study , Polymorphism, Single Nucleotide , Humans , Gene Frequency , Genome-Wide Association Study/methods , Linear Models , Quantitative Trait Loci
6.
Genomics Inform ; 20(1): e8, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35399007

ABSTRACT

Despite the success of recent genome-wide association studies investigating longitudinal traits, a large fraction of overall heritability remains unexplained. This suggests that some of the missing heritability may be accounted for by gene-gene and gene-time/environment interactions. In this paper, we develop a Bayesian variable selection method for longitudinal genetic data based on mixed models. The method jointly models the main effects and interactions of all candidate genetic variants and non-genetic factors and has higher statistical power than previous approaches. To account for the within-subject dependence structure, we propose a grid-based approach that models only one fixed-dimensional covariance matrix, which is thus applicable to data where subjects have different numbers of time points. We provide the theoretical basis of our Bayesian method and then illustrate its performance using data from the 1000 Genome Project with various simulation settings. Several simulation studies show that our multivariate method increases the statistical power compared to the corresponding univariate method and can detect gene-time/ environment interactions well. We further evaluate our method with different numbers of individuals, variants, and causal variants, as well as different trait-heritability, and conclude that our method performs reasonably well with various simulation settings.

7.
PLoS Med ; 19(4): e1003972, 2022 04.
Article in English | MEDLINE | ID: mdl-35472203

ABSTRACT

BACKGROUND: Both genetic and lifestyle factors contribute to the risk of type 2 diabetes, but the extent to which there is a synergistic effect of the 2 factors is unclear. The aim of this study was to examine the joint associations of genetic risk and diet quality with incident type 2 diabetes. METHODS AND FINDINGS: We analyzed data from 35,759 men and women in the United States participating in the Nurses' Health Study (NHS) I (1986 to 2016) and II (1991 to 2017) and the Health Professionals Follow-up Study (HPFS; 1986 to 2016) with available genetic data and who did not have diabetes, cardiovascular disease, or cancer at baseline. Genetic risk was characterized using both a global polygenic score capturing overall genetic risk and pathway-specific polygenic scores denoting distinct pathophysiological mechanisms. Diet quality was assessed using the Alternate Healthy Eating Index (AHEI). Cox models were used to calculate hazard ratios (HRs) for type 2 diabetes after adjusting for potential confounders. With over 902,386 person-years of follow-up, 4,433 participants were diagnosed with type 2 diabetes. The relative risk of type 2 diabetes was 1.29 (95% confidence interval [CI] 1.25, 1.32; P < 0.001) per standard deviation (SD) increase in global polygenic score and 1.13 (1.09, 1.17; P < 0.001) per 10-unit decrease in AHEI. Irrespective of genetic risk, low diet quality, as compared to high diet quality, was associated with approximately 30% increased risk of type 2 diabetes (Pinteraction = 0.69). The joint association of low diet quality and increased genetic risk was similar to the sum of the risk associated with each factor alone (Pinteraction = 0.30). Limitations of this study include the self-report of diet information and possible bias resulting from inclusion of highly educated participants with available genetic data. CONCLUSIONS: These data provide evidence for the independent associations of genetic risk and diet quality with incident type 2 diabetes and suggest that a healthy diet is associated with lower diabetes risk across all levels of genetic risk.


Subject(s)
Diabetes Mellitus, Type 2 , Adult , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/genetics , Diet/adverse effects , Female , Follow-Up Studies , Humans , Male , Prospective Studies , Risk Factors , United States/epidemiology
8.
Nanotechnology ; 33(12)2021 Dec 24.
Article in English | MEDLINE | ID: mdl-34852337

ABSTRACT

Nano-membrane tri-gateß-gallium oxide (ß-Ga2O3) field-effect transistors (FETs) on SiO2/Si substrate fabricated via exfoliation have been demonstrated for the first time. By employing electron beam lithography, the minimum-sized features can be defined with the footprint channel width of 50 nm. For high-quality interface betweenß-Ga2O3and gate dielectric, atomic layer-deposited 15 nm thick aluminum oxide (Al2O3) was utilized with tri-methyl-aluminum (TMA) self-cleaning surface treatment. The fabricated devices demonstrate extremely low subthreshold slope (SS) of 61 mV dec-1, high drain current (IDS) ON/OFF ratio of 1.5 × 109, and negligible transfer characteristic hysteresis. We also experimentally demonstrated robustness of these devices with current-voltage (I-V) characteristics measured at temperatures up to 400 °C.

9.
Ophthalmol Sci ; 1(1)2021 Mar.
Article in English | MEDLINE | ID: mdl-34382031

ABSTRACT

PURPOSE: Large-scale genome-wide association studies (GWAS) have reported important single nucleotide polymorphisms (SNPs) with significant associations with age-related macular degeneration (AMD). However, their role in disease development remains elusive. This study aimed to assess SNP-metabolite associations (i.e., metabolite quantitative trait loci [met-QTL]) and to provide insights into the biological mechanisms of AMD risk SNPs. DESIGN: Cross-sectional multicenter study (Boston, Massachusetts, and Coimbra, Portugal). PARTICIPANTS: Patients with AMD (n = 388) and control participants (n = 98) without any vitreoretinal disease (> 50 years). METHODS: Age-related macular degeneration grading was performed using color fundus photographs according to the Age-Related Eye Disease Study classification scheme. Fasting blood samples were collected and evaluated with mass spectrometry for metabolomic profiling and Illumina OmniExpress for SNPs profiling. Analyses of met-QTL of endogenous metabolites were conducted using linear regression models adjusted for age, gender, smoking, 10 metabolite principal components (PCs), and 10 SNP PCs. Additionally, we analyzed the cumulative effect of AMD risk SNPs on plasma metabolites by generating genetic risk scores and assessing their associations with metabolites using linear regression models, accounting for the same covariates. Modeling was performed first for each cohort, and then combined by meta-analysis. Multiple comparisons were accounted for using the false discovery rate (FDR). MAIN OUTCOME MEASURES: Plasma metabolite levels associated with AMD risk SNPs. RESULTS: After quality control, data for 544 plasma metabolites were included. Meta-analysis of data from all individuals (AMD patients and control participants) identified 28 significant met-QTL (ß = 0.016-0.083; FDR q-value < 1.14 × 10-2), which corresponded to 5 metabolites and 2 genes: ASPM and LIPC. Polymorphisms in the LIPC gene were associated with phosphatidylethanolamine metabolites, which are glycerophospholipids, and polymorphisms in the ASPM gene with branched-chain amino acids. Similar results were observed when considering only patients with AMD. Genetic risk score-metabolite associations further supported a global impact of AMD risk SNPs on the plasma metabolome. CONCLUSIONS: This study demonstrated that genomic-metabolomic associations can provide insights into the biological relevance of AMD risk SNPs. In particular, our results support that the LIPC gene and the glycerophospholipid metabolic pathway may play an important role in AMD, thus offering new potential therapeutic targets for this disease.

10.
Epigenomics ; 13(21): 1761-1770, 2021 11.
Article in English | MEDLINE | ID: mdl-33719520

ABSTRACT

Health disparities correspond to differences in disease burden and mortality among socially defined population groups. Such disparities may emerge according to race/ethnicity, socioeconomic status and a variety of other social contexts, and are documented for a wide range of diseases. Here, we provide a transdisciplinary perspective on the contribution of epigenetics to the understanding of health disparities, with a special emphasis on disparities across socially defined racial/ethnic groups. Scientists in the fields of biological anthropology, bioinformatics and molecular epidemiology provide a summary of theoretical, statistical and practical considerations for conducting epigenetic health disparities research, and provide examples of successful applications from cancer research using this approach.


Subject(s)
Ethnicity , Racial Groups , Epigenesis, Genetic , Epigenomics , Ethnicity/genetics , Humans , Racial Groups/genetics , Social Class
11.
Genomics Inform ; 19(4): e36, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35012283

ABSTRACT

Predicting individual traits and diseases from genetic variants is critical to fulfilling the promise of personalized medicine. The genetic variants from genome-wide association studies (GWAS), including variants well below GWAS significance, can be aggregated into highly significant predictions across a wide range of complex traits and diseases. The recent arrival of large-sample public biobanks enables highly accurate polygenic predictions based on genetic variants across the whole genome. Various statistical methodologies and diverse computational tools have been introduced and developed to computed the polygenic risk score (PRS) more accurately. However, many researchers utilize PRS tools without a thorough understanding of the underlying model and how to specify the parameters for the best performance. It is advantageous to study the statistical models implemented in computational tools for PRS estimation and the formulas of parameters to be specified. Here, we review a variety of recent statistical methodologies and computational tools for PRS computation.

12.
Eur Heart J ; 41(28): 2645-2656, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32406924

ABSTRACT

AIMS: To investigate whether metabolic signature composed of multiple plasma metabolites can be used to characterize adherence and metabolic response to the Mediterranean diet and whether such a metabolic signature is associated with cardiovascular disease (CVD) risk. METHODS AND RESULTS: Our primary study cohort included 1859 participants from the Spanish PREDIMED trial, and validation cohorts included 6868 participants from the US Nurses' Health Studies I and II, and Health Professionals Follow-up Study (NHS/HPFS). Adherence to the Mediterranean diet was assessed using a validated Mediterranean Diet Adherence Screener (MEDAS), and plasma metabolome was profiled by liquid chromatography-tandem mass spectrometry. We observed substantial metabolomic variation with respect to Mediterranean diet adherence, with nearly one-third of the assayed metabolites significantly associated with MEDAS (false discovery rate < 0.05). Using elastic net regularized regressions, we identified a metabolic signature, comprised of 67 metabolites, robustly correlated with Mediterranean diet adherence in both PREDIMED and NHS/HPFS (r = 0.28-0.37 between the signature and MEDAS; P = 3 × 10-35 to 4 × 10-118). In multivariable Cox regressions, the metabolic signature showed a significant inverse association with CVD incidence after adjusting for known risk factors (PREDIMED: hazard ratio [HR] per standard deviation increment in the signature = 0.71, P < 0.001; NHS/HPFS: HR = 0.85, P = 0.001), and the association persisted after further adjustment for MEDAS scores (PREDIMED: HR = 0.73, P = 0.004; NHS/HPFS: HR = 0.85, P = 0.004). Further genome-wide association analysis revealed that the metabolic signature was significantly associated with genetic loci involved in fatty acids and amino acids metabolism. Mendelian randomization analyses showed that the genetically inferred metabolic signature was significantly associated with risk of coronary heart disease (CHD) and stroke (odds ratios per SD increment in the genetically inferred metabolic signature = 0.92 for CHD and 0.91 for stroke; P < 0.001). CONCLUSIONS: We identified a metabolic signature that robustly reflects adherence and metabolic response to a Mediterranean diet, and predicts future CVD risk independent of traditional risk factors, in Spanish and US cohorts.


Subject(s)
Cardiovascular Diseases , Diet, Mediterranean , Cardiovascular Diseases/epidemiology , Follow-Up Studies , Genome-Wide Association Study , Humans , Metabolome , Risk Factors
13.
J Allergy Clin Immunol ; 145(2): 537-549, 2020 02.
Article in English | MEDLINE | ID: mdl-31669095

ABSTRACT

BACKGROUND: Clinical and epidemiologic studies have shown that obesity is associated with asthma and that these associations differ by asthma subtype. Little is known about the shared genetic components between obesity and asthma. OBJECTIVE: We sought to identify shared genetic associations between obesity-related traits and asthma subtypes in adults. METHODS: A cross-trait genome-wide association study (GWAS) was performed using 457,822 subjects of European ancestry from the UK Biobank. Experimental evidence to support the role of genes significantly associated with both obesity-related traits and asthma through a GWAS was sought by using results from obese versus lean mouse RNA sequencing and RT-PCR experiments. RESULTS: We found a substantial positive genetic correlation between body mass index and later-onset asthma defined by asthma age of onset at 16 years or greater (Rg = 0.25, P = 9.56 × 10-22). Mendelian randomization analysis provided strong evidence in support of body mass index causally increasing asthma risk. Cross-trait meta-analysis identified 34 shared loci among 3 obesity-related traits and 2 asthma subtypes. GWAS functional analyses identified potential causal relationships between the shared loci and Genotype-Tissue Expression (GTEx) quantitative trait loci and shared immune- and cell differentiation-related pathways between obesity and asthma. Finally, RNA sequencing data from lungs of obese versus control mice found that 2 genes (acyl-coenzyme A oxidase-like [ACOXL] and myosin light chain 6 [MYL6]) from the cross-trait meta-analysis were differentially expressed, and these findings were validated by using RT-PCR in an independent set of mice. CONCLUSIONS: Our work identified shared genetic components between obesity-related traits and specific asthma subtypes, reinforcing the hypothesis that obesity causally increases the risk of asthma and identifying molecular pathways that might underlie both obesity and asthma.


Subject(s)
Asthma/genetics , Genetic Predisposition to Disease/genetics , Obesity/genetics , Adult , Animals , Biological Specimen Banks , Body Mass Index , Female , Genome-Wide Association Study , Humans , Male , Mice , United Kingdom
14.
ACS Omega ; 4(24): 20756-20761, 2019 Dec 10.
Article in English | MEDLINE | ID: mdl-31858062

ABSTRACT

Herein, we present a solar-blind ultraviolet photodetector realized using atomic layer-deposited p-type cuprous oxide (Cu2O) underneath a mechanically exfoliated n-type ß-gallium oxide (ß-Ga2O3) nanomembrane. The atomic layer deposition process of the Cu2O film applies bis(N,N'-di-secbutylacetamidinato)dicopper(I) [Cu(5Bu-Me-amd)]2 as a novel Cu precursor and water vapor as an oxidant. The exfoliated ß-Ga2O3 nanomembrane was transferred to the top of the Cu2O layer surface to realize a unique oxide pn heterojunction, which is not easy to realize by conventional oxide epitaxy techniques. The current-voltage (I-V) characteristics of the fabricated pn heterojunction diode show the typical rectifying behavior. The fabricated Cu2O/ß-Ga2O3 photodetector achieves sensitive detection of current at the picoampere scale in the reverse mode. This work provides a new approach to integrate all oxide heterojunctions using membrane transfer and bonding techniques, which goes beyond the limitation of conventional heteroepitaxy.

15.
J Am Coll Cardiol ; 74(17): 2162-2174, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31648709

ABSTRACT

BACKGROUND: High resting heart rate (RHR) occurs in parallel with type 2 diabetes (T2D) and metabolic disorders, implying shared etiology between them. However, it is unknown if they are causally related, and no study has been conducted to investigate the shared mechanisms underlying these associations. OBJECTIVES: The objective of this study was to understand the genetic basis of the association between resting heart rate and cardiometabolic disorders/T2D. METHODS: This study examined the genetic correlation, causality, and shared genetics between RHR and T2D using LD Score regression, generalized summary data-based Mendelian randomization, and transcriptome wide association scan (TWAS) in UK Biobank data (n = 428,250) and summary-level data for T2D (74,124 cases and 824,006 control subjects) and 8 cardiometabolic traits (sample size ranges from 51,750 to 236,231). RESULTS: Significant genetic correlation between RHR and T2D (rg = 0.22; 95% confidence interval: 0.18 to 0.26; p = 1.99 × 10-22), and 6 cardiometabolic traits (fasting insulin, fasting glucose, waist-hip ratio, triglycerides, high-density lipoprotein, and body mass index; rg range -0.12 to 0.24; all p < 0.05) were observed. RHR has significant estimated causal effect on T2D (odds ratio: 1.12 per 10-beats/min increment; p = 7.79 × 10-11) and weaker causal estimates from T2D to RHR (0.32 beats/min per doubling increment in T2D prevalence; p = 6.14 × 10-54). Sensitivity analysis by controlling for the included cardiometabolic traits did not modify the relationship between RHR and T2D. TWAS found locus chr2q23.3 (rs1260326) was highly pleiotropic among RHR, cardiometabolic traits, and T2D, and identified 7 genes (SMARCAD1, RP11-53O19.3, CTC-498M16.4, PDE8B, AKTIP, KDM4B, and TSHZ3) that were statistically independent and shared between RHR and T2D in tissues from the nervous and cardiovascular systems. These shared genes suggested the involvement of epigenetic regulation of energy and glucose metabolism, and AKT activation-related telomere dysfunction and vascular endothelial aging in the shared etiologies between RHR and T2D. Finally, FADS1 was found to be shared among RHR, fasting glucose, high-density lipoprotein, and triglycerides. CONCLUSIONS: These findings provide evidence of significant genetic correlations and causation between RHR and T2D/cardiometabolic traits, advance our understanding of RHR, and provide insight into shared etiology for high RHR and T2D.


Subject(s)
Cardiovascular Diseases/genetics , Diabetes Mellitus, Type 2/genetics , Genome-Wide Association Study , Heart Rate , Transcriptome , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Biological Specimen Banks , Blood Glucose/analysis , Cardiovascular System , Comorbidity , DNA Helicases/genetics , Delta-5 Fatty Acid Desaturase , Endothelium, Vascular/pathology , Epigenesis, Genetic , Homeodomain Proteins/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Linkage Disequilibrium , Lipoproteins, HDL/metabolism , Mendelian Randomization Analysis , Phosphorylation , Prevalence , Telomere/ultrastructure , Triglycerides/metabolism , United Kingdom
16.
Nat Commun ; 10(1): 3095, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31300640

ABSTRACT

The nasal cellular epigenome may serve as biomarker of airway disease and environmental response. Here we collect nasal swabs from the anterior nares of 547 children (mean-age 12.9 y), and measure DNA methylation (DNAm) with the Infinium MethylationEPIC BeadChip. We perform nasal Epigenome-Wide Association analyses (EWAS) of current asthma, allergen sensitization, allergic rhinitis, fractional exhaled nitric oxide (FeNO) and lung function. We find multiple differentially methylated CpGs (FDR < 0.05) and Regions (DMRs; ≥ 5-CpGs and FDR < 0.05) for asthma (285-CpGs), FeNO (8,372-CpGs; 191-DMRs), total IgE (3-CpGs; 3-DMRs), environment IgE (17-CpGs; 4-DMRs), allergic asthma (1,235-CpGs; 7-DMRs) and bronchodilator response (130-CpGs). Discovered DMRs annotated to genes implicated in allergic asthma, Th2 activation and eosinophilia (EPX, IL4, IL13) and genes previously associated with asthma and IgE in EWAS of blood (ACOT7, SLC25A25). Asthma, IgE and FeNO were associated with nasal epigenetic age acceleration. The nasal epigenome is a sensitive biomarker of asthma, allergy and airway inflammation.


Subject(s)
Asthma/diagnosis , DNA Methylation/immunology , Inflammation/diagnosis , Nasal Mucosa/immunology , Adolescent , Asthma/genetics , Asthma/immunology , Asthma/pathology , Biomarkers/analysis , Child , CpG Islands/genetics , CpG Islands/immunology , Epigenesis, Genetic/immunology , Epigenomics/methods , Female , Humans , Inflammation/immunology , Inflammation/pathology , Male , Nasal Mucosa/pathology
17.
Metabolites ; 9(7)2019 Jul 02.
Article in English | MEDLINE | ID: mdl-31269701

ABSTRACT

The pathogenesis of age-related macular degeneration (AMD), a leading cause of blindness worldwide, remains only partially understood. This has led to the current lack of accessible and reliable biofluid biomarkers for diagnosis and prognosis, and absence of treatments for dry AMD. This study aimed to assess the plasma metabolomic profiles of AMD and its severity stages with the ultimate goal of contributing to addressing these needs. We recruited two cohorts: Boston, United States (n = 196) and Coimbra, Portugal (n = 295). Fasting blood samples were analyzed using ultra-high performance liquid chromatography mass spectrometry. For each cohort, we compared plasma metabolites of AMD patients versus controls (logistic regression), and across disease stages (permutation-based cumulative logistic regression considering both eyes). Meta-analyses were then used to combine results from the two cohorts. Our results revealed that 28 metabolites differed significantly between AMD patients versus controls (false discovery rate (FDR) q-value: 4.1 × 10-2-1.8 × 10-5), and 67 across disease stages (FDR q-value: 4.5 × 10-2-1.7 × 10-4). Pathway analysis showed significant enrichment of glycerophospholipid, purine, taurine and hypotaurine, and nitrogen metabolism (p-value ≤ 0.04). In conclusion, our findings support that AMD patients present distinct plasma metabolomic profiles, which vary with disease severity. This work contributes to the understanding of AMD pathophysiology, and can be the basis of future biomarkers and precision medicine for this blinding condition.

18.
Nat Commun ; 10(1): 569, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718517

ABSTRACT

We introduce cross-trait penalized regression (CTPR), a powerful and practical approach for multi-trait polygenic risk prediction in large cohorts. Specifically, we propose a novel cross-trait penalty function with the Lasso and the minimax concave penalty (MCP) to incorporate the shared genetic effects across multiple traits for large-sample GWAS data. Our approach extracts information from the secondary traits that is beneficial for predicting the primary trait based on individual-level genotypes and/or summary statistics. Our novel implementation of a parallel computing algorithm makes it feasible to apply our method to biobank-scale GWAS data. We illustrate our method using large-scale GWAS data (~1M SNPs) from the UK Biobank (N = 456,837). We show that our multi-trait method outperforms the recently proposed multi-trait analysis of GWAS (MTAG) for predictive performance. The prediction accuracy for height by the aid of BMI improves from R2 = 35.8% (MTAG) to 42.5% (MCP + CTPR) or 42.8% (Lasso + CTPR) with UK Biobank data.


Subject(s)
Genome-Wide Association Study/methods , Models, Genetic , Algorithms , Genotype , Humans , Phenotype , Quantitative Trait Loci/genetics
19.
Nat Genet ; 50(12): 1753, 2018 12.
Article in English | MEDLINE | ID: mdl-30390058

ABSTRACT

In the version of this article originally published, there were two errors in the text of the second paragraph of the Methods section. In the sentence "To identify genetic variants that contribute to doctor-diagnosed asthma and allergic diseases (detailed phenotype information described in the Supplementary Note) and link them with other conditions, we performed GWASs using phenotype measures in UK Biobank participants (N = 487,409)" the number of participants should have been 150,509. In the sentence "Thus, a total of 110,361 European descendants with high-quality genotyping and complete phenotype/covariate data were used for these analyses, including 25,685 allergic diseases subjects (hay fever/allergic rhinitis or eczema, without doctor-diagnosed asthma), 14,085 asthma subjects and 76,768 controls for the analysis" the phrase "without doctor-diagnosed asthma" should have read "some with doctor-diagnosed asthma." The errors have been corrected in the HTML and PDF versions of the article.

20.
Nat Genet ; 50(6): 857-864, 2018 06.
Article in English | MEDLINE | ID: mdl-29785011

ABSTRACT

Clinical and epidemiological data suggest that asthma and allergic diseases are associated and may share a common genetic etiology. We analyzed genome-wide SNP data for asthma and allergic diseases in 33,593 cases and 76,768 controls of European ancestry from UK Biobank. Two publicly available independent genome-wide association studies were used for replication. We have found a strong genome-wide genetic correlation between asthma and allergic diseases (rg = 0.75, P = 6.84 × 10-62). Cross-trait analysis identified 38 genome-wide significant loci, including 7 novel shared loci. Computational analysis showed that shared genetic loci are enriched in immune/inflammatory systems and tissues with epithelium cells. Our work identifies common genetic architectures shared between asthma and allergy and will help to advance understanding of the molecular mechanisms underlying co-morbid asthma and allergic diseases.


Subject(s)
Asthma/genetics , Hypersensitivity/genetics , Adult , Aged , Biological Specimen Banks , Case-Control Studies , Female , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , United Kingdom
SELECTION OF CITATIONS
SEARCH DETAIL
...