20 research outputs found

    Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease.

    Get PDF
    Scavenger receptor BI (SR-BI) is the major receptor for high-density lipoprotein (HDL) cholesterol (HDL-C). In humans, high amounts of HDL-C in plasma are associated with a lower risk of coronary heart disease (CHD). Mice that have depleted Scarb1 (SR-BI knockout mice) have markedly elevated HDL-C levels but, paradoxically, increased atherosclerosis. The impact of SR-BI on HDL metabolism and CHD risk in humans remains unclear. Through targeted sequencing of coding regions of lipid-modifying genes in 328 individuals with extremely high plasma HDL-C levels, we identified a homozygote for a loss-of-function variant, in which leucine replaces proline 376 (P376L), in SCARB1, the gene encoding SR-BI. The P376L variant impairs posttranslational processing of SR-BI and abrogates selective HDL cholesterol uptake in transfected cells, in hepatocyte-like cells derived from induced pluripotent stem cells from the homozygous subject, and in mice. Large population-based studies revealed that subjects who are heterozygous carriers of the P376L variant have significantly increased levels of plasma HDL-C. P376L carriers have a profound HDL-related phenotype and an increased risk of CHD (odds ratio = 1.79, which is statistically significant)

    Concept, Design and Implementation of a Cardiovascular Gene-Centric 50 K SNP Array for Large-Scale Genomic Association Studies

    Get PDF
    A wealth of genetic associations for cardiovascular and metabolic phenotypes in humans has been accumulating over the last decade, in particular a large number of loci derived from recent genome wide association studies (GWAS). True complex disease-associated loci often exert modest effects, so their delineation currently requires integration of diverse phenotypic data from large studies to ensure robust meta-analyses. We have designed a gene-centric 50 K single nucleotide polymorphism (SNP) array to assess potentially relevant loci across a range of cardiovascular, metabolic and inflammatory syndromes. The array utilizes a “cosmopolitan” tagging approach to capture the genetic diversity across ∼2,000 loci in populations represented in the HapMap and SeattleSNPs projects. The array content is informed by GWAS of vascular and inflammatory disease, expression quantitative trait loci implicated in atherosclerosis, pathway based approaches and comprehensive literature searching. The custom flexibility of the array platform facilitated interrogation of loci at differing stringencies, according to a gene prioritization strategy that allows saturation of high priority loci with a greater density of markers than the existing GWAS tools, particularly in African HapMap samples. We also demonstrate that the IBC array can be used to complement GWAS, increasing coverage in high priority CVD-related loci across all major HapMap populations. DNA from over 200,000 extensively phenotyped individuals will be genotyped with this array with a significant portion of the generated data being released into the academic domain facilitating in silico replication attempts, analyses of rare variants and cross-cohort meta-analyses in diverse populations. These datasets will also facilitate more robust secondary analyses, such as explorations with alternative genetic models, epistasis and gene-environment interactions

    Mining the LIPG Allelic Spectrum Reveals the Contribution of Rare and Common Regulatory Variants to HDL Cholesterol

    Get PDF
    Genome-wide association studies (GWAS) have successfully identified loci associated with quantitative traits, such as blood lipids. Deep resequencing studies are being utilized to catalogue the allelic spectrum at GWAS loci. The goal of these studies is to identify causative variants and missing heritability, including heritability due to low frequency and rare alleles with large phenotypic impact. Whereas rare variant efforts have primarily focused on nonsynonymous coding variants, we hypothesized that noncoding variants in these loci are also functionally important. Using the HDL-C gene LIPG as an example, we explored the effect of regulatory variants identified through resequencing of subjects at HDL-C extremes on gene expression, protein levels, and phenotype. Resequencing a portion of the LIPG promoter and 5′ UTR in human subjects with extreme HDL-C, we identified several rare variants in individuals from both extremes. Luciferase reporter assays were used to measure the effect of these rare variants on LIPG expression. Variants conferring opposing effects on gene expression were enriched in opposite extremes of the phenotypic distribution. Minor alleles of a common regulatory haplotype and noncoding GWAS SNPs were associated with reduced plasma levels of the LIPG gene product endothelial lipase (EL), consistent with its role in HDL-C catabolism. Additionally, we found that a common nonfunctional coding variant associated with HDL-C (rs2000813) is in linkage disequilibrium with a 5′ UTR variant (rs34474737) that decreases LIPG promoter activity. We attribute the gene regulatory role of rs34474737 to the observed association of the coding variant with plasma EL levels and HDL-C. Taken together, the findings show that both rare and common noncoding regulatory variants are important contributors to the allelic spectrum in complex trait loci

    Dense genotyping of candidate gene loci identifies variants associated with high-density lipoprotein cholesterol.

    No full text
    BACKGROUND: Plasma levels of high density lipoprotein cholesterol (HDL-C) are known to be heritable, but only a fraction of the heritability is explained. We used a high density genotyping array containing SNPs from HDL-C candidate genes selected on known biology of HDL-C metabolism, mouse genetic studies, and human genetic association studies. SNP selection was based on tagging-SNPs but also included low-frequency nonsynonymous SNPs. METHODS AND RESULTS: Association analysis in a cohort containing extremes of HDL-C (case-control, n=1733) provided a discovery phase, with replication in three additional populations for a total meta-analysis in 7,857 individuals. We replicated the majority of loci identified through genome wide association studies and present on the array (including ABCA1, APOA1/C3/A4/A5, APOB, APOE/C1/C2, CETP, CTCF-PRMT8, FADS1/2/3, GALNT2, LCAT, LILRA3, LIPC, LIPG, LPL, LRP4, SCARB1, TRIB1, ZNF664), and provide evidence suggestive of association in several previously unreported candidate gene loci (including ABCG1, GPR109A/B/81, NFKB1, PON1/2/3/4). There was evidence for multiple, independent association signals in five loci, including association with low frequency nonsynonymous variants. CONCLUSIONS: Genetic loci associated with HDL-C are likely to harbor multiple, independent causative variants, frequently with opposite effects on the HDL-C phenotype. Cohorts composed of extreme individuals may be efficiently used in a case-control discovery of quantitative traits

    High-density single-nucleotide polymorphism maps of the human genome

    No full text
    Here we report a large, extensively characterized set of single-nucleotide polymorphisms (SNPs) covering the human genome. We determined the allele frequencies of 55,018 SNPs in African Americans, Asians (Japanese-Chinese), and European Americans as part of The SNP Consortium's Allele Frequency Project. A subset of 8333 SNPs was also characterized in Koreans. Because these SNPs were ascertained in the same way, the data set is particularly useful for modeling. Our results document that much genetic variation is shared among populations. For autosomes, some 44% of these SNPs have a minor allele frequency >= 10% in each population, and the average allele frequency differences between populations with different continental origins are less than 19%. However, the several percentage point allele frequency differences among the closely related Korean, Japanese, and Chinese populations suggest caution in using mixtures of well-established populations for case-control genetic studies of complex traits. We estimate that similar to 7% of these SNPs are private SNPs with minor allele frequencies 60%) can be used for admixture studies. High-density maps of high-quality, characterized SNPs produced by this project are freely available. (c) 2005 Elsevier Inc. All rights reserved

    Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease Item Type Article Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease

    No full text
    Abstract Scavenger receptor BI (SR-BI) is the major receptor for high-density lipoprotein (HDL) cholesterol (HDL-C). In humans, high amounts of HDL-C in plasma are associated with a lower risk of coronary heart disease (CHD). Mice that have depleted Scarb1 (SR-BI knockout mice) have markedly elevated HDL-C levels but, paradoxically, increased atherosclerosis. The impact of SR-BI on HDL metabolism and CHD risk in humans remains unclear. Through targeted sequencing of coding regions of lipid-modifying genes in 328 individuals with extremely high plasma HDL-C levels, we identified a homozygote for a loss-of-function variant, in which leucine replaces proline 376 (P376L), in SCARB1, the gene encoding SR-BI. The P376L variant impairs posttranslational processing of SR-BI and abrogates selective HDL cholesterol uptake in transfected cells, in hepatocyte-like cells derived from induced pluripotent stem cells from the homozygous subject, and in mice. Large population-based studies revealed that subjects who are heterozygous carriers of the P376L variant have significantly increased levels of plasma HDL-C. Zanoni et al. Page 2 P376L carriers have a profound HDL-related phenotype and an increased risk of CHD (odds ratio = 1.79, which is statistically significant). The strong inverse association between amounts of high-density lipoprotein (HDL) cholesterol (HDL-C) and coronary heart disease (CHD) risk has generated interest in a potential causal relationship between HDL metabolism and CHD. However, clinical trials with drugs that raise HDL-C levels, niacin and cholesteryl ester transfer protein (CETP) inhibitors, have produced disappointing results ( 1 ). Furthermore, recent studies of human genetic variants that are associated with HDL-C levels have generally failed to show association with CHD ( 2 , 3 ). Most notably, a loss-of-function variant in LIPG, a gene encoding an endothelial lipase that, in the heterozygous state, raises HDL-C by ~5 mg/dl, was found to have no association with CHD ( 4 ). Although these previous studies suggest that higher HDL-C levels may not be causally protective against CHD, we reasoned that additional human genetic analyses might provide mechanistic insight into the complex relationship between HDL and CHD. The scavenger receptor class BI (SR-BI), encoded by the gene SCARB1, was discovered to be an HDL receptor two decades ago ( 5 ). SR-BI promotes the selective uptake of HDL cholesteryl esters (HDL-CEs) into cells, particularly hepatocytes and steroidogenic cells ( 5 , 6 ). In mice, overexpression of SR-BI in the liver reduces levels of HDL-C ( 7 -10 ), and genetic deletion of SR-BI results in higher HDL-C levels ( 11 -13 ). Remarkably, these genetic manipulations in mice have effects on atherosclerosis opposite to those predicted by human epidemiological data: Overexpression reduces atherosclerosis despite the lower HDL-C levels ( 14 -16 ), and gene deletion increases atherosclerosis despite the higher HDL-C levels ( 17 -20 ). One potential explanation relates to the flux of cholesterol from macrophages through the reverse cholesterol transport (RCT) pathway; SR-BI overexpression increases macrophage RCT, and SR-BI knockout reduces macrophage RCT ( 21 ). The human relevance of these observations has been unclear. Identification of SCARB1 P376L homozygote and association with extremely high HDL-C We hypothesized that humans with extremely high levels of HDL-C may harbor loss-offunction variants in SCARB1 and undertook a targeted resequencing discovery experiment in 328 participants with very high HDL-C (>95th percentile, mean HDL-C of 106.8 mg/dl) and a control group of 398 subjects with low HDL-C (<25th percentile, mean HDL-C of 30.4 mg/dl). In this cohort, we sequenced the exons of ~990 genes located within 300 kb of each of the 95 loci with significant associations (P < 5 × 10 −8 ) with plasma lipid levels identified by the Global Lipids Genetics Consortium as of 2010 C>T, p.P376L, rs74830677), a 67-year-old female with an HDL-C of 152 mg/dl, and confirmed this finding by Sanger sequencing. This subject harbored no mutations in other high HDL-C genes such as CETP and LIPG. In addition to this homozygote, four P376L heterozygotes were identified by targeted sequencing in the high HDL-C group; no heterozygotes were found in the low HDL-C group (P =0.008, Fisher's exact test). Zanoni et al. Page 3 To identify additional P376L carriers, we genotyped an expanded cohort of very high versus low HDL-C subjects. Among 524 additional subjects with very high HDL-C (mean HDL-C 95.0 mg/dl), we identified 11 heterozygotes for P376L; whereas among 758 subjects with low HDL-C (mean HDL-C 33.5 mg/dl), we identified 3 heterozygotes. In total, our combined sequencing and genotyping for discovery of the P376L variant showed that this variant is significantly overrepresented in subjects with high HDL-C [minor allele frequency (MAF) = 0.010 in high HDL-C versus 0.0013 in low HDL-C controls, P = 0.000127, Fisher's exact test, Because this variant is present on the exome array, we expanded our analysis to the Global Lipid Genetics Consortium exome array data in >300,000 individuals. The P376L variant was very rare in this population (MAF of ~0.0003). It was significantly associated with higher HDL-C levels with a relatively large effect size (beta = 8.4 mg/dl; P =1.4 × 10 −15 ). Notably, this variant was not associated with plasma levels of low-density lipoprotein cholesterol (LDL-C) or triglycerides (TGs) (table S1). Thus, we conclude that SCARB1 P376L is associated specifically with elevated HDL-C levels. HDL-related phenotypes of SCARB1 P376L homozygote and heterozygotes We next recruited the P376L homozygote, eight heterozygous carriers, and both high HDL-C and normal HDL-C noncarrier controls for deep phenotyping of HDL metabolism and related traits. All of the P376L study participants were of European ancestry, almost exclusively of Ashkenazi Jewish descent. Clinical characteristics and lipid profiles of the subjects are reported in SCARB1 P376L results in complete loss of function of SR-BI Given the profound HDL phenotype of the P376L carriers, we sought to understand the impact of the variant on SR-BI function. We generated induced pluripotent stem cells (iPSCs) using peripheral blood mononuclear cells from the P376L homozygote and a noncarrier control. We next differentiated these cells into hepatocyte-like cells (HLCs) to study HDL metabolism in the setting of endogenous cellular SCARB1 expression. HLCs differentiated through this protocol recapitulate phenotypes of cultured primary hepatocytes Zanoni et al. Page 4 such as albumin and VLDL (very low density lipoprotein) secretion ( 23 -26 ). The cell lines from the control donor and the P376L homozygous subject demonstrated expression of hepatocyte-specific genes ALB (albumin) and AFP (alpha-fetoprotein) and exhibited comparable SCARB1 gene expression ( To evaluate the physiological impact of the P376L variant on HDL-C levels and catabolism in vivo, we used adeno-associated virus (AAV) vectors to direct hepatic overexpression of WT SR-BI or the P376L variant in mice with depleted Scarb1 [Scarb1 knockout (KO) mice]. The two groups of mice demonstrated similar hepatic expression levels of Scarb1 mRNA ( We hypothesized that the markedly reduced HDL-CE uptake could be because of aberrant processing of the P376L SR-BI protein, which leads to impaired cell surface localization. To test this, we isolated cell surface proteins from COS7 cells transfected with WT and P376L SR-BI using biotinylation and found markedly reduced cell surface SR-BI in the P376L transfected cell lysates after streptavidin cell surface protein pull-down assays ( SCARB1 P376L is associated with increased risk of CHD in humans Despite a profound increase in HDL-C, SR-BI deficiency in mice causes accelerated atherosclerosis ( 17 -20 ). The relationship of reduced SR-BI function to atherosclerotic cardiovascular disease in humans has not been established. The P376L homozygous subject did not have clinical CHD, but her carotid intimal-medial thickness (cIMT) was 0.789 mm (left-right average), which is in the >75th percentile for females of her age; in addition, she had detectable plaque throughout the left internal carotid artery and at the bifurcation of her right internal carotid artery. cIMT measurements were not significantly different in the P376L heterozygotes compared with both groups of controls ( To achieve greater statistical power to address this question, we performed a meta-analysis of large exome array genotyping studies of CHD cases and healthy controls to determine the relationship of the P376L variant with risk of CHD ( Discussion Studies of mice have provided important insights into the effects of SR-BI on HDL metabolism, RCT, and atherosclerosis. These studies revealed that overexpression of SR-BI reduces HDL-C ( 7 -10 ) and reduces atherosclerosis ( 14 -16 ), whereas gene deletion of SR-BI increases HDL-C ( 11 -13 ) and accelerates atherosclerosis ( 17 -20 ). The clinical relevance of these findings has remained uncertain, however. Studies of injected labeled HDL-CE in humans suggested that the majority of the HDL-CE was transported to the liver via CETPmediated exchange to apoB-containing lipoproteins rather than by direct uptake from HDL by the liver ( 30 ), which brings into question the importance of hepatic SR-BI in human physiology. Common genetic variants near the SCARB1 locus were found to be significantly associated with plasma HDL-C levels, which suggests that SR-BI may play a role in HDL metabolism in humans ( 22 , 31 ). A family with a rare SCARB1 variant in which serine replaces proline 297 (P297S) was reported in which the heterozygous carriers of the variant had modestly elevated HDL-C levels ( 31 ). However, the variant retains substantial SR-BI activity, no homozygotes were identified, the apparent effect on HDL-C was modest, and there was insufficient power to address its effects on atherosclerosis. Through sequencing of subjects with extremely high plasma levels of HDL-C, we identified a homozygote for a P376L variant in SR-BI. Our complementary approaches consistently Zanoni et al. Page 6 demonstrated that this variant confers virtually complete loss of function of SR-BI. Our results demonstrate many similarities in the consequences of SR-BI deficiency on HDL composition between mice and humans, including a shift toward large, buoyant HDL particles and a significant increase in apoA-I, but not apoA-II, in plasma and HDL ( 12 , 32 , 33 ). The homozygote is a woman who had two healthy children without fertility issues or delivery complications, which suggests that, in humans, SR-BI deficiency may not impair reproductive function in the same manner as it does in mice ( 18 , 34 ). In mice, SR-BImediated CE uptake from HDL is a critical process underlying steroid hormone synthesis in adrenal and gonadal tissues, and SR-BI deficiency alters adrenal cholesterol content, impairs adrenal glucocorticoid response under stress, and can lead to fasting-induced hypoglycemia ( 6 , 35 , 36 ). We did not observe any differences in fasting glucose, serum cortisol, adrenocorticotropic hormone, or female gonadal hormones in P376L heterozygous subjects versus controls, and we saw only a modest increase in testosterone in male P376L heterozygotes relative to noncarriers. We postulate that differences in expression or capacity for up-regulation of apoB-containing lipoprotein receptors relative to SR-BI between mouse models and humans in steroidogenic tissues may account, at least partially, for the lack of recapitulation of some of the phenotypes of SR-BI deficiency in mice. We also observed no differences in platelet levels, cholesterol content, and activation from the P376L carriers, despite reports of thrombocytopenia and altered platelet activity in Scarb1 KO mice ( 31 ). These results suggest a relatively different contribution of SR-BI to platelet function between mice and humans. Note that the phenotypes of human SCARB1 P376L homozygote (elevated HDL-C and large HDL particles but relatively normal steroidogenesis, reproductive viability, and platelet function) are comparable to those observed in mice lacking PDZ domain containing 1 (PDZK1), an adaptor protein for SR-BI ( 37 ). Perhaps the most important finding of our study is that, despite the elevation in HDL-C, P376L carriers exhibit increased risk of CHD, as do Scarb1 KO mice. Our results are consistent with a growing theme in HDL biology that steady-state more important than absolute levels. Using an in vivo assay of macrophage RCT, we previously showed that Scarb1 KO mice have impaired macrophage RCT even though they have elevated HDL-C levels ( 21 ). Our results suggest that reduced hepatic SR-BI function in humans causes impaired RCT, which leads to increased risk of CHD despite elevation in HDL-C levels. However, SR-BI is also expressed in vascular cell types, including endothelial cells, vascular smooth muscle cells, and macrophages, where it could have protective effects against atherosclerosis as well ( 38 , 39 ). Our results are also consistent with the previously suggested concept ( 39 ) that up-regulation or enhancement of SR-BI could be a novel therapeutic approach to reducing CHD risk in the general population

    Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease

    No full text
    Scavenger receptor BI (SR-BI) is the major receptor for high-density lipoprotein (HDL) cholesterol (HDL-C). In humans, high amounts of HDL-C in plasma are associated with a lower risk of coronary heart disease (CHD). Mice that have depleted Scarb1 (SR-BI knockout mice) have markedly elevated HDL-C levels but, paradoxically, increased atherosclerosis. The impact of SR-BI on HDL metabolism and CHD risk in humans remains unclear. Through targeted sequencing of coding regions of lipid-modifying genes in 328 individuals with extremely high plasma HDL-C levels, we identified a homozygote for a lossof-function variant, in which leucine replaces proline 376 (P376L), in SCARB1, the gene encoding SR-BI. The P376L variant impairs posttranslational processing of SR-BI and abrogates selective HDL cholesterol uptake in transfected cells, in hepatocyte-like cells derived from induced pluripotent stem cells from the homozygous subject, and in mice. Large population-based studies revealed that subjects who are heterozygous carriers of the P376L variant have significantly increased levels of plasma HDL-C. P376L carriers have a profound HDL-related phenotype and an increased risk of CHD (odds ratio = 1.79, which is statistically significant)

    Identification of ADAMTS7 as a novel locus for coronary atherosclerosis and association of ABO with myocardial infarction in the presence of coronary atherosclerosis: two genome-wide association studies

    No full text
    BACKGROUND: We tested whether genetic factors distinctly contribute to either development of coronary atherosclerosis or, specifically, to myocardial infarction in existing coronary atherosclerosis. METHODS: We did two genome-wide association studies (GWAS) with coronary angiographic phenotyping in participants of European ancestry. To identify loci that predispose to angiographic coronary artery disease (CAD), we compared individuals who had this disorder (n=12,393) with those who did not (controls, n=7383). To identify loci that predispose to myocardial infarction, we compared patients who had angiographic CAD and myocardial infarction (n=5783) with those who had angiographic CAD but no myocardial infarction (n=3644). FINDINGS: In the comparison of patients with angiographic CAD versus controls, we identified a novel locus, ADAMTS7 (p=4·98×10(-13)). In the comparison of patients with angiographic CAD who had myocardial infarction versus those with angiographic CAD but no myocardial infarction, we identified a novel association at the ABO locus (p=7·62×10(-9)). The ABO association was attributable to the glycotransferase-deficient enzyme that encodes the ABO blood group O phenotype previously proposed to protect against myocardial infarction. INTERPRETATION: Our findings indicate that specific genetic predispositions promote the development of coronary atherosclerosis whereas others lead to myocardial infarction in the presence of coronary atherosclerosis. The relation to specific CAD phenotypes might modify how novel loci are applied in personalised risk assessment and used in the development of novel therapies for CAD
    corecore