Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
2.
Genet Epidemiol ; 47(5): 394-406, 2023 07.
Article in English | MEDLINE | ID: mdl-37021827

ABSTRACT

Genome-wide association studies (GWAS) have significantly advanced our understanding of the genetic underpinnings of diseases, but case and control cohort definitions for a given disease can vary between different published studies. For example, two GWAS for the same disease using the UK Biobank data set might use different data sources (i.e., self-reported questionnaires, hospital records, etc.) or different levels of granularity (i.e., specificity of inclusion criteria) to define cases and controls. The extent to which this variability in cohort definitions impacts the end-results of a GWAS study is unclear. In this study, we systematically evaluated the effect of the data sources used for case and control definitions on GWAS findings. Using the UK Biobank, we selected three diseases-glaucoma, migraine, and iron-deficiency anemia. For each disease, we designed 13 GWAS, each using different combinations of data sources to define cases and controls, and then calculated the pairwise genetic correlations between all GWAS for each disease. We found that the data sources used to define cases for a given disease can have a significant impact on GWAS end-results, but the extent of this depends heavily on the disease in question. This suggests the need for greater scrutiny on how case cohorts are defined for GWAS.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide , Self Report
3.
medRxiv ; 2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36865145

ABSTRACT

Chronic Obstructive Pulmonary Disease (COPD) has a simple physiological diagnostic criterion but a wide range of clinical characteristics. The mechanisms underlying this variability in COPD phenotypes are unclear. To investigate the potential contribution of genetic variants to phenotypic heterogeneity, we examined the association of genome-wide associated lung function, COPD, and asthma variants with other phenotypes using phenome-wide association results derived in the UK Biobank. Our clustering analysis of the variants-phenotypes association matrix identified three clusters of genetic variants with different effects on white blood cell counts, height, and body mass index (BMI). To assess the potential clinical and molecular effects of these groups of variants, we investigated the association between cluster-specific genetic risk scores and phenotypes in the COPDGene cohort. We observed differences in steroid use, BMI, lymphocyte counts, chronic bronchitis, and differential gene and protein expression across the three genetic risk scores. Our results suggest that multi-phenotype analysis of obstructive lung disease-related risk variants may identify genetically driven phenotypic patterns in COPD.

4.
ERJ Open Res ; 7(2)2021 Apr.
Article in English | MEDLINE | ID: mdl-33981765

ABSTRACT

Homozygosity for the SERPINA1 Z allele causes α1-antitrypsin deficiency, a rare condition that can cause lung and liver disease. However, the effects of Z allele heterozygosity on nonrespiratory phenotypes, and on lung function in the general population, remain unclear. We conducted a large, population-based study to determine Z allele effects on >2400 phenotypes in the UK Biobank (N=303 353). Z allele heterozygosity was strongly associated with increased height (ß=1.02 cm, p=3.91×10-68), and with other nonrespiratory phenotypes including increased risk of gall bladder disease, reduced risk of heart disease and lower blood pressure, reduced risk of osteoarthritis and reduced bone mineral density, increased risk of headache and enlarged prostate, as well as with blood biomarkers of liver function. Heterozygosity was associated with higher height-adjusted forced expiratory volume in 1 s (FEV1) (ß=19.36 mL, p=9.21×10-4) and FEV1/forced vital capacity (ß=0.0031, p=1.22×10-5) in nonsmokers, whereas in smokers, this protective effect was abolished. Furthermore, we show for the first time that sex modifies the association of the Z allele on lung function. We conclude that Z allele heterozygosity and homozygosity exhibit opposing effects on lung function in the UK population, and that these associations are modified by smoking and sex. In exploratory analyses, heterozygosity for the Z allele also showed pleiotropic associations with nonrespiratory health-related traits and disease risk.

6.
Nat Genet ; 51(3): 481-493, 2019 03.
Article in English | MEDLINE | ID: mdl-30804560

ABSTRACT

Reduced lung function predicts mortality and is key to the diagnosis of chronic obstructive pulmonary disease (COPD). In a genome-wide association study in 400,102 individuals of European ancestry, we define 279 lung function signals, 139 of which are new. In combination, these variants strongly predict COPD in independent populations. Furthermore, the combined effect of these variants showed generalizability across smokers and never smokers, and across ancestral groups. We highlight biological pathways, known and potential drug targets for COPD and, in phenome-wide association studies, autoimmune-related and other pleiotropic effects of lung function-associated variants. This new genetic evidence has potential to improve future preventive and therapeutic strategies for COPD.


Subject(s)
Genetic Predisposition to Disease/genetics , Lung/physiopathology , Pulmonary Disease, Chronic Obstructive/genetics , Aged , Aged, 80 and over , Case-Control Studies , Female , Genome-Wide Association Study/methods , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , Smoking/genetics
7.
Bioinformatics ; 33(17): 2784-2786, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28472345

ABSTRACT

SUMMARY: We developed the STOPGAP (Systematic Target OPportunity assessment by Genetic Association Predictions) database, an extensive catalog of human genetic associations mapped to effector gene candidates. STOPGAP draws on a variety of publicly available GWAS associations, linkage disequilibrium (LD) measures, functional genomic and variant annotation sources. Algorithms were developed to merge the association data, partition associations into non-overlapping LD clusters, map variants to genes and produce a variant-to-gene score used to rank the relative confidence among potential effector genes. This database can be used for a multitude of investigations into the genes and genetic mechanisms underlying inter-individual variation in human traits, as well as supporting drug discovery applications. AVAILABILITY AND IMPLEMENTATION: Shell, R, Perl and Python scripts and STOPGAP R data files (version 2.5.1 at publication) are available at https://github.com/StatGenPRD/STOPGAP . Some of the most useful STOPGAP fields can be queried through an R Shiny web application at http://stopgapwebapp.com . CONTACT: matthew.r.nelson@gsk.com. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Databases, Factual , Genetic Association Studies/methods , Genetic Variation , Linkage Disequilibrium , Algorithms , Humans , Sequence Analysis, DNA/methods
8.
J Allergy Clin Immunol ; 139(3): 797-803.e7, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27523435

ABSTRACT

BACKGROUND: Inhaled corticosteroids (ICSs) are considered the most effective anti-inflammatory therapy for asthma control and management; however, there is substantial treatment response variability. OBJECTIVE: We sought to identify genetic markers of ICS response by conducting the largest pharmacogenetic investigation to date in 2672 ICS-treated patients with asthma. METHODS: Genotyping and imputation was performed in fluticasone furoate (FF) or fluticasone propionate-treated patients with asthma from 3 phase IIB and 4 phase IIIA randomized, double-blind, placebo-controlled, parallel group, multicenter studies. The primary end point analyzed was change in trough FEV1 (ΔFEV1) from baseline to 8 to 12 weeks of treatment. RESULTS: More than 9.8 million common genetic variants (minor allele frequency ≥ 1%) were analyzed to test for association with ΔFEV1. No genetic variant met the prespecified threshold for statistical significance. CONCLUSIONS: This study provides no evidence to confirm previously reported associations between candidate genetic variants and ICS response (ΔFEV1) in patients with asthma. In addition, no variant satisfied the criterion for genome-wide significance in our study. Common genetic variants are therefore unlikely to prove useful as predictive biomarkers of ICS response in patients with asthma.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Androstadienes/therapeutic use , Anti-Asthmatic Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Asthma/genetics , Adult , Asthma/physiopathology , Double-Blind Method , Female , Forced Expiratory Volume , Genetic Variation , Genotype , Humans , Male , Middle Aged , Treatment Outcome
9.
Pharmacogenomics ; 17(5): 459-62, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27023328

ABSTRACT

AIM: Pyrexia is a common adverse event (AE) on dabrafenib treatment (monotherapy or combination with trametinib). Since germline SNPs and HLA alleles are implicated in drug-induced AEs, this study investigated their association with pyrexia. PATIENTS & METHODS: 1006 melanoma subjects from five dabrafenib-trametinib clinical studies underwent genotyping for genome-wide SNPs, which enabled imputation of 150 HLA alleles. SNP/HLA allele frequencies were compared between pyrexia cases (n = 218) and controls (n = 361) out of the 1006 subjects by meta-analysis. RESULTS: This analysis had adequate power to detect association of common SNPs or HLA alleles with moderate to large effects on pyrexia (odds ratio >6), but no significant association was found. CONCLUSION: The study suggests that common genetic variation or HLA polymorphisms do not contribute substantially to dabrafenib-induced pyrexia.


Subject(s)
Antineoplastic Agents/adverse effects , Fever/chemically induced , HLA Antigens/genetics , Imidazoles/adverse effects , Melanoma/drug therapy , Oximes/adverse effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Case-Control Studies , Fever/genetics , Genetic Association Studies , Humans , Melanoma/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins B-raf/genetics
10.
J Clin Pharmacol ; 53(10): 1078-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23934621

ABSTRACT

The mu-opioid system has a key role in hedonic and motivational processes critical to substance addiction. However, existing mu-opioid antagonists have had limited success as anti-addiction treatments. GSK1521498 is a selective and potent mu-opioid antagonist being developed for the treatment of overeating and substance addictions. In this study, 28 healthy participants were administered single doses of GSK1521498 20 mg, ethanol 0.5 g/kg body weight, or both in combination, in a double blind placebo controlled four-way crossover design. The primary objective was to determine the risk of significant adverse pharmacodynamic and pharmacokinetic (PK) interactions. The effects of GSK1521498 on hedonic and consummatory responses to alcohol and the attentional processing of alcohol-related stimuli, and their modulation by the OPRM1 A118G polymorphism were also explored. GSK1521498 20 mg was well tolerated alone and in combination with ethanol. There were mild transient effects of GSK1521498 on alertness and mood that were greater when it was combined with ethanol. These effects were not of clinical significance. There were no effects of GSK1521498 on reaction time, hedonic or consummatory responses. These findings provide encouraging safety and PK data to support continued development of GSK1521498 for the treatment of alcohol addiction.


Subject(s)
Ethanol/administration & dosage , Indans/administration & dosage , Triazoles/administration & dosage , Adult , Affect/drug effects , Cross-Over Studies , Double-Blind Method , Drug Interactions , Ethanol/adverse effects , Ethanol/pharmacokinetics , Humans , Indans/adverse effects , Indans/pharmacokinetics , Middle Aged , Polymorphism, Genetic , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/genetics , Triazoles/adverse effects , Triazoles/pharmacokinetics , Young Adult
12.
Am J Hum Genet ; 92(4): 547-57, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23541341

ABSTRACT

Clinical trials for preventative therapies are complex and costly endeavors focused on individuals likely to develop disease in a short time frame, randomizing them to treatment groups, and following them over time. In such trials, statistical power is governed by the rate of disease events in each group and cost is determined by randomization, treatment, and follow-up. Strategies that increase the rate of disease events by enrolling individuals with high risk of disease can significantly reduce study size, duration, and cost. Comprehensive study of common, complex diseases has resulted in a growing list of robustly associated genetic markers. Here, we evaluate the utility--in terms of trial size, duration, and cost--of enriching prevention trial samples by combining clinical information with genetic risk scores to identify individuals at greater risk of disease. We also describe a framework for utilizing genetic risk scores in these trials and evaluating the associated cost and time savings. With type 1 diabetes (T1D), type 2 diabetes (T2D), myocardial infarction (MI), and advanced age-related macular degeneration (AMD) as examples, we illustrate the potential and limitations of using genetic data for prevention trial design. We illustrate settings where incorporating genetic information could reduce trial cost or duration considerably, as well as settings where potential savings are negligible. Results are strongly dependent on the genetic architecture of the disease, but we also show that these benefits should increase as the list of robustly associated markers for each disease grows and as large samples of genotyped individuals become available.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 2/prevention & control , Genetic Testing/statistics & numerical data , Genetic Variation/genetics , Genotype , Macular Degeneration/prevention & control , Myocardial Infarction/prevention & control , Research Design , Clinical Trials as Topic , Cost-Benefit Analysis , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 2/genetics , Humans , Macular Degeneration/genetics , Models, Statistical , Myocardial Infarction/genetics , Phenotype , Risk Factors
13.
J Invest Dermatol ; 133(6): 1489-96, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23358095

ABSTRACT

The pathogenesis of androgenetic alopecia (AGA, male-pattern baldness) is driven by androgens, and genetic predisposition is the major prerequisite. Candidate gene and genome-wide association studies have reported that single-nucleotide polymorphisms (SNPs) at eight different genomic loci are associated with AGA development. However, a significant fraction of the overall heritable risk still awaits identification. Furthermore, the understanding of the pathophysiology of AGA is incomplete, and each newly associated locus may provide novel insights into contributing biological pathways. The aim of this study was to identify unknown AGA risk loci by replicating SNPs at the 12 genomic loci that showed suggestive association (5 × 10(-8)

Subject(s)
Alopecia/epidemiology , Alopecia/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway/physiology , Adult , Alopecia/etiology , Alopecia/metabolism , Cholestanetriol 26-Monooxygenase/genetics , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 2 , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 5 , Frizzled Receptors/genetics , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , White People/genetics , White People/statistics & numerical data , Wnt3 Protein/genetics
14.
PLoS One ; 8(12): e83137, 2013.
Article in English | MEDLINE | ID: mdl-24391740

ABSTRACT

Urotensin-II controls ion/water homeostasis in fish and vascular tone in rodents. We hypothesised that common genetic variants in urotensin-II pathway genes are associated with human blood pressure or renal function. We performed family-based analysis of association between blood pressure, glomerular filtration and genes of the urotensin-II pathway (urotensin-II, urotensin-II related peptide, urotensin-II receptor) saturated with 28 tagging single nucleotide polymorphisms in 2024 individuals from 520 families; followed by an independent replication in 420 families and 7545 unrelated subjects. The expression studies of the urotensin-II pathway were carried out in 97 human kidneys. Phylogenetic evolutionary analysis was conducted in 17 vertebrate species. One single nucleotide polymorphism (rs531485 in urotensin-II gene) was associated with adjusted estimated glomerular filtration rate in the discovery cohort (p = 0.0005). It showed no association with estimated glomerular filtration rate in the combined replication resource of 8724 subjects from 6 populations. Expression of urotensin-II and its receptor showed strong linear correlation (r = 0.86, p<0.0001). There was no difference in renal expression of urotensin-II system between hypertensive and normotensive subjects. Evolutionary analysis revealed accumulation of mutations in urotensin-II since the divergence of primates and weaker conservation of urotensin-II receptor in primates than in lower vertebrates. Our data suggest that urotensin-II system genes are unlikely to play a major role in genetic control of human blood pressure or renal function. The signatures of evolutionary forces acting on urotensin-II system indicate that it may have evolved towards loss of function since the divergence of primates.


Subject(s)
Blood Pressure/genetics , Blood Pressure/physiology , Kidney/physiology , Peptide Hormones/genetics , Peptide Hormones/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Urotensins/genetics , Urotensins/physiology , Adolescent , Adult , Aged , Animals , Cohort Studies , Evolution, Molecular , Female , Gene Expression , Genetic Association Studies , Glomerular Filtration Rate/genetics , Glomerular Filtration Rate/physiology , Humans , Hypertension/genetics , Hypertension/physiopathology , Intracellular Signaling Peptides and Proteins , Male , Middle Aged , Polymorphism, Single Nucleotide , Primates/genetics , Primates/physiology , Young Adult
15.
PLoS Genet ; 8(5): e1002746, 2012 May.
Article in English | MEDLINE | ID: mdl-22693459

ABSTRACT

Androgenetic alopecia (AGA) is a highly heritable condition and the most common form of hair loss in humans. Susceptibility loci have been described on the X chromosome and chromosome 20, but these loci explain a minority of its heritable variance. We conducted a large-scale meta-analysis of seven genome-wide association studies for early-onset AGA in 12,806 individuals of European ancestry. While replicating the two AGA loci on the X chromosome and chromosome 20, six novel susceptibility loci reached genome-wide significance (p = 2.62×10⁻9-1.01×10⁻¹²). Unexpectedly, we identified a risk allele at 17q21.31 that was recently associated with Parkinson's disease (PD) at a genome-wide significant level. We then tested the association between early-onset AGA and the risk of PD in a cross-sectional analysis of 568 PD cases and 7,664 controls. Early-onset AGA cases had significantly increased odds of subsequent PD (OR = 1.28, 95% confidence interval: 1.06-1.55, p = 8.9×10⁻³). Further, the AGA susceptibility alleles at the 17q21.31 locus are on the H1 haplotype, which is under negative selection in Europeans and has been linked to decreased fertility. Combining the risk alleles of six novel and two established susceptibility loci, we created a genotype risk score and tested its association with AGA in an additional sample. Individuals in the highest risk quartile of a genotype score had an approximately six-fold increased risk of early-onset AGA [odds ratio (OR) = 5.78, p = 1.4×10⁻88]. Our results highlight unexpected associations between early-onset AGA, Parkinson's disease, and decreased fertility, providing important insights into the pathophysiology of these conditions.


Subject(s)
Alopecia/genetics , Genome-Wide Association Study , Parkinson Disease/genetics , Adult , Aged , Alleles , Fertility/genetics , Genetic Predisposition to Disease , Genotype , Haplotypes , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Risk Factors
16.
J Cereb Blood Flow Metab ; 32(1): 1-5, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22008728

ABSTRACT

[(11)C]PBR28 binds the 18-kDa Translocator Protein (TSPO) and is used in positron emission tomography (PET) to detect microglial activation. However, quantitative interpretations of signal are confounded by large interindividual variability in binding affinity, which displays a trimodal distribution compatible with a codominant genetic trait. Here, we tested directly for an underlying genetic mechanism to explain this. Binding affinity of PBR28 was measured in platelets isolated from 41 human subjects and tested for association with polymorphisms in TSPO and genes encoding other proteins in the TSPO complex. Complete agreement was observed between the TSPO Ala147Thr genotype and PBR28 binding affinity phenotype (P value=3.1 × 10(-13)). The TSPO Ala147Thr polymorphism predicts PBR28 binding affinity in human platelets. As all second-generation TSPO PET radioligands tested hitherto display a trimodal distribution in binding affinity analogous to PBR28, testing for this polymorphism may allow quantitative interpretation of TSPO PET studies with these radioligands.


Subject(s)
Acetamides/metabolism , Polymorphism, Single Nucleotide , Pyridines/metabolism , Radiopharmaceuticals/metabolism , Receptors, GABA/genetics , Receptors, GABA/metabolism , Adult , Amino Acid Substitution/genetics , Binding, Competitive/genetics , Blood Platelets/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Female , Genetic Association Studies , Humans , Isoquinolines/metabolism , Male , Positron-Emission Tomography , Protein Binding , Radioligand Assay , Tritium
17.
Int J Mol Epidemiol Genet ; 2(3): 261-85, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21915365

ABSTRACT

Although experimental studies have suggested that insulin-like growth factor I (IGF-I) and its binding protein IGFBP-3 might have a role in the aetiology of coronary artery disease (CAD), the relevance of circulating IGFs and their binding proteins in the development of CAD in human populations is unclear. We conducted a nested case-control study, with a mean follow-up of six years, within the EPIC-Norfolk cohort to assess the association between circulating levels of IGF-I and IGFBP-3 and risk of CAD in up to 1,013 cases and 2,055 controls matched for age, sex and study enrolment date. After adjustment for cardiovascular risk factors, we found no association between circulating levels of IGF-I or IGFBP-3 and risk of CAD (odds ratio: 0.98 (95% Cl 0.90-1.06) per 1 SD increase in circulating IGF-I; odds ratio: 1.02 (95% Cl 0.94-1.12) for IGFBP-3). We examined associations between tagging single nucleotide polymorphisms (tSNPs) at the IGF1 and IGFBP3 loci and circulating IGF-I and IGFBP-3 levels in up to 1,133 cases and 2,223 controls and identified three tSNPs (rs1520220, rs3730204, rs2132571) that showed independent association with either circulating IGF-I or IGFBP-3 levels. In an assessment of 31 SNPs spanning the IGF1 or IGFBP3 loci, none were associated with risk of CAD in a meta-analysis that included EPIC-Norfolk and eight additional studies comprising up to 9,319 cases and 19,964 controls. Our results indicate that IGF-I and IGFBP-3 are unlikely to be importantly involved in the aetiology of CAD in human populations.

18.
PLoS One ; 6(4): e18208, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21490708

ABSTRACT

BACKGROUND: The Asia-specific PLA2G7 994G-T transversion leads to V279F substitution within the lipoprotein-associated phospholipase-A2 (Lp-PLA2) and to absence of enzyme activity in plasma. This variant offers a unique natural experiment to assess the role of Lp-PLA2 in the pathogenesis of coronary artery disease (CAD) in humans. Given conflicting results from mostly small studies, a large two-stage case-control study was warranted. METHODOLOGY/PRINCIPAL FINDINGS: PLA2G7 V279F genotypes were initially compared in 2890 male cases diagnosed with CAD before age 60 with 3128 male controls without CAD at age 50 and above and subsequently in a second independent male dataset of 877 CAD cases and 1230 controls. In the first dataset, the prevalence of the 279F null allele was 11.5% in cases and 12.8% in controls. After adjustment for age, body mass index, diabetes, smoking, glucose and lipid levels, the OR (95% CI) for CAD for this allele was 0.80 (0.66-0.97, p = 0.02). The results were very similar in the second dataset, despite lower power, with an allele frequency of 11.2% in cases and 12.5% in controls, leading to a combined OR of 0.80 (0.69-0.92), p = 0.002. The magnitude and direction of this genetic effect were fully consistent with large epidemiological studies on plasma Lp-PLA2 activity and CAD risk. CONCLUSIONS: Natural deficiency in Lp-PLA2 activity due to carriage of PLA2G7 279F allele protects from CAD in Korean men. These results provide evidence for a causal relationship between Lp-PLA2 and CAD, and support pharmacological inhibition of this enzyme as an innovative way to prevent CAD.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Coronary Artery Disease/genetics , Alleles , Asian People , Female , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Republic of Korea
19.
Hypertension ; 56(6): 1069-76, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21060006

ABSTRACT

Genetic determinants of blood pressure are poorly defined. We undertook a large-scale, gene-centric analysis to identify loci and pathways associated with ambulatory systolic and diastolic blood pressure. We measured 24-hour ambulatory blood pressure in 2020 individuals from 520 white European nuclear families (the Genetic Regulation of Arterial Pressure of Humans in the Community Study) and genotyped their DNA using the Illumina HumanCVD BeadChip array, which contains ≈ 50 000 single nucleotide polymorphisms in >2000 cardiovascular candidate loci. We found a strong association between rs13306560 polymorphism in the promoter region of MTHFR and CLCN6 and mean 24-hour diastolic blood pressure; each minor allele copy of rs13306560 was associated with 2.6 mm Hg lower mean 24-hour diastolic blood pressure (P = 1.2 × 10⁻8). rs13306560 was also associated with clinic diastolic blood pressure in a combined analysis of 8129 subjects from the Genetic Regulation of Arterial Pressure of Humans in the Community Study, the CoLaus Study, and the Silesian Cardiovascular Study (P=5.4 × 10⁻6). Additional analysis of associations between variants in gene ontology-defined pathways and mean 24-hour blood pressure in the Genetic Regulation of Arterial Pressure of Humans in the Community Study showed that cell survival control signaling cascades could play a role in blood pressure regulation. There was also a significant overrepresentation of rare variants (minor allele frequency: < 0.05) among polymorphisms showing at least nominal association with mean 24-hour blood pressure indicating that a considerable proportion of its heritability may be explained by uncommon alleles. Through a large-scale gene-centric analysis of ambulatory blood pressure, we identified an association of a novel variant at the MTHFR/CLNC6 locus with diastolic blood pressure and provided new insights into the genetic architecture of blood pressure.


Subject(s)
Blood Pressure Monitoring, Ambulatory , Blood Pressure/genetics , Chloride Channels/genetics , Hypertension/epidemiology , Hypertension/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Cohort Studies , Female , Gene Frequency , Genetic Association Studies , Genetic Loci , Genotype , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prevalence , Promoter Regions, Genetic , United Kingdom/epidemiology , White People/genetics , Young Adult
20.
Arterioscler Thromb Vasc Biol ; 30(11): 2264-76, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20864672

ABSTRACT

OBJECTIVE: Genetic studies might provide new insights into the biological mechanisms underlying lipid metabolism and risk of CAD. We therefore conducted a genome-wide association study to identify novel genetic determinants of low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and triglycerides. METHODS AND RESULTS: We combined genome-wide association data from 8 studies, comprising up to 17 723 participants with information on circulating lipid concentrations. We did independent replication studies in up to 37 774 participants from 8 populations and also in a population of Indian Asian descent. We also assessed the association between single-nucleotide polymorphisms (SNPs) at lipid loci and risk of CAD in up to 9 633 cases and 38 684 controls. We identified 4 novel genetic loci that showed reproducible associations with lipids (probability values, 1.6×10(-8) to 3.1×10(-10)). These include a potentially functional SNP in the SLC39A8 gene for HDL-C, an SNP near the MYLIP/GMPR and PPP1R3B genes for LDL-C, and at the AFF1 gene for triglycerides. SNPs showing strong statistical association with 1 or more lipid traits at the CELSR2, APOB, APOE-C1-C4-C2 cluster, LPL, ZNF259-APOA5-A4-C3-A1 cluster and TRIB1 loci were also associated with CAD risk (probability values, 1.1×10(-3) to 1.2×10(-9)). CONCLUSIONS: We have identified 4 novel loci associated with circulating lipids. We also show that in addition to those that are largely associated with LDL-C, genetic loci mainly associated with circulating triglycerides and HDL-C are also associated with risk of CAD. These findings potentially provide new insights into the biological mechanisms underlying lipid metabolism and CAD risk.


Subject(s)
Cholesterol, HDL/genetics , Cholesterol, LDL/genetics , Coronary Artery Disease/genetics , Lipid Metabolism/genetics , Triglycerides/genetics , Asian People , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Genetic Variation , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide , Risk Factors , Triglycerides/blood , White People
SELECTION OF CITATIONS
SEARCH DETAIL
...