Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
Add more filters

Publication year range
2.
PLoS Genet ; 16(4): e1008629, 2020 04.
Article in English | MEDLINE | ID: mdl-32282858

ABSTRACT

Analyzing 12,361 all-cause cirrhosis cases and 790,095 controls from eight cohorts, we identify a common missense variant in the Mitochondrial Amidoxime Reducing Component 1 gene (MARC1 p.A165T) that associates with protection from all-cause cirrhosis (OR 0.91, p = 2.3*10-11). This same variant also associates with lower levels of hepatic fat on computed tomographic imaging and lower odds of physician-diagnosed fatty liver as well as lower blood levels of alanine transaminase (-0.025 SD, 3.7*10-43), alkaline phosphatase (-0.025 SD, 1.2*10-37), total cholesterol (-0.030 SD, p = 1.9*10-36) and LDL cholesterol (-0.027 SD, p = 5.1*10-30) levels. We identified a series of additional MARC1 alleles (low-frequency missense p.M187K and rare protein-truncating p.R200Ter) that also associated with lower cholesterol levels, liver enzyme levels and reduced risk of cirrhosis (0 cirrhosis cases for 238 R200Ter carriers versus 17,046 cases of cirrhosis among 759,027 non-carriers, p = 0.04) suggesting that deficiency of the MARC1 enzyme may lower blood cholesterol levels and protect against cirrhosis.


Subject(s)
Fatty Liver/genetics , Fatty Liver/prevention & control , Genetic Predisposition to Disease , Liver Cirrhosis/genetics , Liver Cirrhosis/prevention & control , Mitochondrial Proteins/genetics , Mutation, Missense/genetics , Oxidoreductases/genetics , Alleles , Cholesterol, LDL/blood , Coronary Artery Disease/genetics , Datasets as Topic , Fatty Liver/blood , Fatty Liver/enzymology , Female , Homozygote , Humans , Liver/enzymology , Liver Cirrhosis/blood , Liver Cirrhosis/enzymology , Liver Cirrhosis, Alcoholic/blood , Liver Cirrhosis, Alcoholic/enzymology , Liver Cirrhosis, Alcoholic/genetics , Liver Cirrhosis, Alcoholic/prevention & control , Loss of Function Mutation/genetics , Male , Middle Aged
3.
Clin Sci (Lond) ; 136(14): 1081-1110, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35892309

ABSTRACT

Metabolic demands of skeletal muscle are substantial and are characterized normally as highly flexible and with a large dynamic range. Skeletal muscle composition (e.g., fiber type and mitochondrial content) and metabolism (e.g., capacity to switch between fatty acid and glucose substrates) are altered in obesity, with some changes proceeding and some following the development of the disease. Nonetheless, there are marked interindividual differences in skeletal muscle composition and metabolism in obesity, some of which have been associated with obesity risk and weight loss capacity. In this review, we discuss related molecular mechanisms and how current and novel treatment strategies may enhance weight loss capacity, particularly in diet-resistant obesity.


Subject(s)
Muscle, Skeletal , Obesity , Fatty Acids/metabolism , Humans , Mitochondria/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Weight Loss/physiology
4.
Arterioscler Thromb Vasc Biol ; 41(8): 2252-2262, 2021 08.
Article in English | MEDLINE | ID: mdl-34134519

ABSTRACT

OBJECTIVE: Leveraging microRNA-Seq data and the 1000 Genomes imputed genotypes, we identified rs174561 as a strong microRNA quantitative trait loci for circulating microRNA-1908-5p with higher miR-1908-5p and reduced LDL (lowdensity lipoprotein)-cholesterol, fasting glucose and A1c concentrations in carriers of the rs-174561-C allele. Here, we have investigated the molecular mechanism(s) linking miR-1908-5p to LDL-C concentrations. APPROACH AND RESULTS: Transfection experiments demonstrate that the presence of the C allele significantly increases miR- 1908-5p abundance relative to the T allele. LDLR mRNA and low-density lipoprotein receptor (LDLR) total protein were unchanged in response to differential miR-1908-5p expression. However, the ratio of the cleaved to full-length form of LDLR decreased with miR-1908-5p mimic and increased with miR-1908-5p inhibitor treatment. BMP1 (bone morphogenetic protein 1) is a protease responsible for LDLR cleavage, and we show that miR-1908-5p mimic reduces BMP1 mRNA. Using a reporter array, we identified the TGF-ß (transforming growth factor-beta) signaling pathway activity to be reduced by miR- 1908-5p mimic treatment, and this was associated with reduced TGFB1 expression. TGF-ß signaling increases BMP1, and we further demonstrate that the effect of miR-1908-5p on LDLR cleavage is abolished by exogenous TGF-ß treatment. CONCLUSIONS: These findings uncover a mechanism whereby miR-1908-5p reduces TGFB1 abundance resulting in lower expression of BMP1, ultimately leading to reduced LDLR cleavage. Cleavage of the mature LDLR is known to reduce cell surface affinity for LDL, thereby linking miR-1908-5p to lower circulating LDL-cholesterol levels.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , Cholesterol, LDL/metabolism , Fatty Acid Desaturases/genetics , Hepatocytes/enzymology , MicroRNAs/metabolism , Polymorphism, Genetic , Bone Morphogenetic Protein 1/genetics , Cell Line , Delta-5 Fatty Acid Desaturase , Fatty Acid Desaturases/metabolism , Gene Expression Regulation , Humans , MicroRNAs/genetics , Protein Stability , Proteolysis , RNA Stability , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
5.
Arterioscler Thromb Vasc Biol ; 39(10): 1925-1937, 2019 10.
Article in English | MEDLINE | ID: mdl-31462092

ABSTRACT

Recent studies have led to a broader understanding of the genetic architecture of coronary artery disease and demonstrate that it largely derives from the cumulative effect of multiple common risk alleles individually of small effect size rather than rare variants with large effects on coronary artery disease risk. The tools applied include genome-wide association studies encompassing over 200 000 individuals complemented by bioinformatic approaches including imputation from whole-genome data sets, expression quantitative trait loci analyses, and interrogation of ENCODE (Encyclopedia of DNA Elements), Roadmap Epigenetic Project, and other data sets. Over 160 genome-wide significant loci associated with coronary artery disease risk have been identified using the genome-wide association studies approach, 90% of which are situated in intergenic regions. Here, I will describe, in part, our research over the last decade performed in collaboration with a series of bright trainees and an extensive number of groups and individuals around the world as it applies to our understanding of the genetic basis of this complex disease. These studies include computational approaches to better understand missing heritability and identify causal pathways, experimental approaches, and progress in understanding at the molecular level the function of the multiple risk loci identified and potential applications of these genomic data in clinical medicine and drug discovery.


Subject(s)
Coronary Artery Disease/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Quality Improvement , Computational Biology , Forecasting , Genetics/trends , Genomics/trends , Genotype , Humans , Phenotype , Polymorphism, Single Nucleotide/genetics
6.
Circulation ; 137(3): 222-232, 2018 01 16.
Article in English | MEDLINE | ID: mdl-28982690

ABSTRACT

BACKGROUND: Nitric oxide signaling plays a key role in the regulation of vascular tone and platelet activation. Here, we seek to understand the impact of a genetic predisposition to enhanced nitric oxide signaling on risk for cardiovascular diseases, thus informing the potential utility of pharmacological stimulation of the nitric oxide pathway as a therapeutic strategy. METHODS: We analyzed the association of common and rare genetic variants in 2 genes that mediate nitric oxide signaling (Nitric Oxide Synthase 3 [NOS3] and Guanylate Cyclase 1, Soluble, Alpha 3 [GUCY1A3]) with a range of human phenotypes. We selected 2 common variants (rs3918226 in NOS3 and rs7692387 in GUCY1A3) known to associate with increased NOS3 and GUCY1A3 expression and reduced mean arterial pressure, combined them into a genetic score, and standardized this exposure to a 5 mm Hg reduction in mean arterial pressure. Using individual-level data from 335 464 participants in the UK Biobank and summary association results from 7 large-scale genome-wide association studies, we examined the effect of this nitric oxide signaling score on cardiometabolic and other diseases. We also examined whether rare loss-of-function mutations in NOS3 and GUCY1A3 were associated with coronary heart disease using gene sequencing data from the Myocardial Infarction Genetics Consortium (n=27 815). RESULTS: A genetic predisposition to enhanced nitric oxide signaling was associated with reduced risks of coronary heart disease (odds ratio, 0.37; 95% confidence interval [CI], 0.31-0.45; P=5.5*10-26], peripheral arterial disease (odds ratio 0.42; 95% CI, 0.26-0.68; P=0.0005), and stroke (odds ratio, 0.53; 95% CI, 0.37-0.76; P=0.0006). In a mediation analysis, the effect of the genetic score on decreased coronary heart disease risk extended beyond its effect on blood pressure. Conversely, rare variants that inactivate the NOS3 or GUCY1A3 genes were associated with a 23 mm Hg higher systolic blood pressure (95% CI, 12-34; P=5.6*10-5) and a 3-fold higher risk of coronary heart disease (odds ratio, 3.03; 95% CI, 1.29-7.12; P=0.01). CONCLUSIONS: A genetic predisposition to enhanced nitric oxide signaling is associated with reduced risks of coronary heart disease, peripheral arterial disease, and stroke. Pharmacological stimulation of nitric oxide signaling may prove useful in the prevention or treatment of cardiovascular disease.


Subject(s)
Blood Pressure/genetics , Coronary Disease/genetics , Mutation , Nitric Oxide Synthase Type III/genetics , Nitric Oxide/metabolism , Peripheral Arterial Disease/genetics , Polymorphism, Single Nucleotide , Signal Transduction/genetics , Soluble Guanylyl Cyclase/genetics , Stroke/genetics , Coronary Disease/enzymology , Coronary Disease/epidemiology , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Nitric Oxide Synthase Type III/metabolism , Peripheral Arterial Disease/enzymology , Peripheral Arterial Disease/epidemiology , Phenotype , Protective Factors , Risk Factors , Soluble Guanylyl Cyclase/metabolism , Stroke/enzymology , Stroke/epidemiology
7.
Cardiovasc Diabetol ; 18(1): 71, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31164165

ABSTRACT

In the era of precision medicine, treatments that target specific modifiable characteristics of high-risk patients have the potential to lower further the residual risk of atherosclerotic cardiovascular events. Correction of atherogenic dyslipidemia, however, remains a major unmet clinical need. Elevated plasma triglycerides, with or without low levels of high-density lipoprotein cholesterol (HDL-C), offer a key modifiable component of this common dyslipidemia, especially in insulin resistant conditions such as type 2 diabetes mellitus. The development of selective peroxisome proliferator-activated receptor alpha modulators (SPPARMα) offers an approach to address this treatment gap. This Joint Consensus Panel appraised evidence for the first SPPARMα agonist and concluded that this agent represents a novel therapeutic class, distinct from fibrates, based on pharmacological activity, and, importantly, a safe hepatic and renal profile. The ongoing PROMINENT cardiovascular outcomes trial is testing in 10,000 patients with type 2 diabetes mellitus, elevated triglycerides, and low levels of HDL-C whether treatment with this SPPARMα agonist safely reduces residual cardiovascular risk.


Subject(s)
Benzoxazoles/therapeutic use , Butyrates/therapeutic use , Cardiovascular Diseases/prevention & control , Dyslipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Lipids/blood , PPAR alpha/agonists , Animals , Benzoxazoles/adverse effects , Biomarkers/blood , Butyrates/adverse effects , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Consensus , Dyslipidemias/blood , Dyslipidemias/diagnosis , Humans , Hypolipidemic Agents/adverse effects , Molecular Targeted Therapy , PPAR alpha/metabolism , Patient Safety , Risk Assessment , Risk Factors , Signal Transduction , Treatment Outcome
8.
Circ Res ; 121(1): 81-88, 2017 Jun 23.
Article in English | MEDLINE | ID: mdl-28506971

ABSTRACT

RATIONALE: Therapies that inhibit CETP (cholesteryl ester transfer protein) have failed to demonstrate a reduction in risk for coronary heart disease (CHD). Human DNA sequence variants that truncate the CETP gene may provide insight into the efficacy of CETP inhibition. OBJECTIVE: To test whether protein-truncating variants (PTVs) at the CETP gene were associated with plasma lipid levels and CHD. METHODS AND RESULTS: We sequenced the exons of the CETP gene in 58 469 participants from 12 case-control studies (18 817 CHD cases, 39 652 CHD-free controls). We defined PTV as those that lead to a premature stop, disrupt canonical splice sites, or lead to insertions/deletions that shift frame. We also genotyped 1 Japanese-specific PTV in 27561 participants from 3 case-control studies (14 286 CHD cases, 13 275 CHD-free controls). We tested association of CETP PTV carrier status with both plasma lipids and CHD. Among 58 469 participants with CETP gene-sequencing data available, average age was 51.5 years and 43% were women; 1 in 975 participants carried a PTV at the CETP gene. Compared with noncarriers, carriers of PTV at CETP had higher high-density lipoprotein cholesterol (effect size, 22.6 mg/dL; 95% confidence interval, 18-27; P<1.0×10-4), lower low-density lipoprotein cholesterol (-12.2 mg/dL; 95% confidence interval, -23 to -0.98; P=0.033), and lower triglycerides (-6.3%; 95% confidence interval, -12 to -0.22; P=0.043). CETP PTV carrier status was associated with reduced risk for CHD (summary odds ratio, 0.70; 95% confidence interval, 0.54-0.90; P=5.1×10-3). CONCLUSIONS: Compared with noncarriers, carriers of PTV at CETP displayed higher high-density lipoprotein cholesterol, lower low-density lipoprotein cholesterol, lower triglycerides, and lower risk for CHD.


Subject(s)
Cholesterol Ester Transfer Proteins/genetics , Coronary Disease/diagnosis , Coronary Disease/genetics , Genetic Variation/genetics , Adult , Aged , Case-Control Studies , Cholesterol Ester Transfer Proteins/blood , Coronary Disease/blood , Female , Humans , Male , Middle Aged , Risk Factors
9.
Nature ; 504(7480): 432-6, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24213632

ABSTRACT

Myocardial infarction, a leading cause of death in the Western world, usually occurs when the fibrous cap overlying an atherosclerotic plaque in a coronary artery ruptures. The resulting exposure of blood to the atherosclerotic material then triggers thrombus formation, which occludes the artery. The importance of genetic predisposition to coronary artery disease and myocardial infarction is best documented by the predictive value of a positive family history. Next-generation sequencing in families with several affected individuals has revolutionized mutation identification. Here we report the segregation of two private, heterozygous mutations in two functionally related genes, GUCY1A3 (p.Leu163Phefs*24) and CCT7 (p.Ser525Leu), in an extended myocardial infarction family. GUCY1A3 encodes the α1 subunit of soluble guanylyl cyclase (α1-sGC), and CCT7 encodes CCTη, a member of the tailless complex polypeptide 1 ring complex, which, among other functions, stabilizes soluble guanylyl cyclase. After stimulation with nitric oxide, soluble guanylyl cyclase generates cGMP, which induces vasodilation and inhibits platelet activation. We demonstrate in vitro that mutations in both GUCY1A3 and CCT7 severely reduce α1-sGC as well as ß1-sGC protein content, and impair soluble guanylyl cyclase activity. Moreover, platelets from digenic mutation carriers contained less soluble guanylyl cyclase protein and consequently displayed reduced nitric-oxide-induced cGMP formation. Mice deficient in α1-sGC protein displayed accelerated thrombus formation in the microcirculation after local trauma. Starting with a severely affected family, we have identified a link between impaired soluble-guanylyl-cyclase-dependent nitric oxide signalling and myocardial infarction risk, possibly through accelerated thrombus formation. Reversing this defect may provide a new therapeutic target for reducing the risk of myocardial infarction.


Subject(s)
Disease Susceptibility/metabolism , Myocardial Infarction/metabolism , Nitric Oxide/metabolism , Signal Transduction , Animals , Chaperonin Containing TCP-1/genetics , Chaperonin Containing TCP-1/metabolism , Cyclic GMP/metabolism , Exome/genetics , Female , Genetic Predisposition to Disease , Guanylate Cyclase/deficiency , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , HEK293 Cells , Humans , Male , Mice , Mutation/genetics , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Pedigree , Platelet Activation , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Reproducibility of Results , Solubility , Soluble Guanylyl Cyclase , Thrombosis/metabolism , Vasodilation
10.
Am J Hum Genet ; 97(2): 228-37, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26166477

ABSTRACT

Runs of homozygosity (ROHs) are recognized signature of recessive inheritance. Contributions of ROHs to the genetic architecture of coronary artery disease and regulation of gene expression in cells relevant to atherosclerosis are not known. Our combined analysis of 24,320 individuals from 11 populations of white European ethnicity showed an association between coronary artery disease and both the count and the size of ROHs. Individuals with coronary artery disease had approximately 0.63 (95% CI: 0.4-0.8) excess of ROHs when compared to coronary-artery-disease-free control subjects (p = 1.49 × 10(-9)). The average total length of ROHs was approximately 1,046.92 (95% CI: 634.4-1,459.5) kb greater in individuals with coronary artery disease than control subjects (p = 6.61 × 10(-7)). None of the identified individual ROHs was associated with coronary artery disease after correction for multiple testing. However, in aggregate burden analysis, ROHs favoring increased risk of coronary artery disease were much more common than those showing the opposite direction of association with coronary artery disease (p = 2.69 × 10(-33)). Individual ROHs showed significant associations with monocyte and macrophage expression of genes in their close proximity-subjects with several individual ROHs showed significant differences in the expression of 44 mRNAs in monocytes and 17 mRNAs in macrophages when compared to subjects without those ROHs. This study provides evidence for an excess of homozygosity in coronary artery disease in outbred populations and suggest the potential biological relevance of ROHs in cells of importance to the pathogenesis of atherosclerosis.


Subject(s)
Coronary Artery Disease/genetics , Gene Expression Regulation/genetics , Genes, Recessive/genetics , Homozygote , Macrophages/metabolism , Monocytes/metabolism , Age Factors , Humans , RNA, Messenger/metabolism , White People/genetics
11.
N Engl J Med ; 372(17): 1608-18, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25853659

ABSTRACT

BACKGROUND: The nature and underlying mechanisms of an inverse association between adult height and the risk of coronary artery disease (CAD) are unclear. METHODS: We used a genetic approach to investigate the association between height and CAD, using 180 height-associated genetic variants. We tested the association between a change in genetically determined height of 1 SD (6.5 cm) with the risk of CAD in 65,066 cases and 128,383 controls. Using individual-level genotype data from 18,249 persons, we also examined the risk of CAD associated with the presence of various numbers of height-associated alleles. To identify putative mechanisms, we analyzed whether genetically determined height was associated with known cardiovascular risk factors and performed a pathway analysis of the height-associated genes. RESULTS: We observed a relative increase of 13.5% (95% confidence interval [CI], 5.4 to 22.1; P<0.001) in the risk of CAD per 1-SD decrease in genetically determined height. There was a graded relationship between the presence of an increased number of height-raising variants and a reduced risk of CAD (odds ratio for height quartile 4 versus quartile 1, 0.74; 95% CI, 0.68 to 0.84; P<0.001). Of the 12 risk factors that we studied, we observed significant associations only with levels of low-density lipoprotein cholesterol and triglycerides (accounting for approximately 30% of the association). We identified several overlapping pathways involving genes associated with both development and atherosclerosis. CONCLUSIONS: There is a primary association between a genetically determined shorter height and an increased risk of CAD, a link that is partly explained by the association between shorter height and an adverse lipid profile. Shared biologic processes that determine achieved height and the development of atherosclerosis may explain some of the association. (Funded by the British Heart Foundation and others.).


Subject(s)
Body Height/genetics , Coronary Artery Disease/genetics , Genetic Variation , Adult , Cholesterol, LDL/blood , Coronary Artery Disease/etiology , Humans , Hyperlipidemias/complications , Odds Ratio , Risk Factors , Triglycerides/blood
12.
Circ Res ; 118(4): 564-78, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26892958

ABSTRACT

Genetic factors contribute importantly to the risk of coronary artery disease (CAD), and in the past decade, there has been major progress in this area. The tools applied include genome-wide association studies encompassing >200,000 individuals complemented by bioinformatic approaches, including 1000 Genomes imputation, expression quantitative trait locus analyses, and interrogation of Encyclopedia of DNA Elements, Roadmap, and other data sets. close to 60 common SNPs (minor allele frequency>0.05) associated with CAD risk and reaching genome-wide significance (P<5 × 10(-8)) have been identified. Furthermore, a total of 202 independent signals in 109 loci have achieved a false discovery rate (q<0.05) and together explain 28% of the estimated heritability of CAD. These data have been used successfully to create genetic risk scores that can improve risk prediction beyond conventional risk factors and identify those individuals who will benefit most from statin therapy. Such information also has important applications in clinical medicine and drug discovery by using a Mendelian randomization approach to interrogate the causal nature of many factors found to associate with CAD risk in epidemiological studies. In contrast to genome-wide association studies, whole-exome sequencing has provided valuable information directly relevant to genes with known roles in plasma lipoprotein metabolism but has, thus far, failed to identify other rare coding variants linked to CAD. Overall, recent studies have led to a broader understanding of the genetic architecture of CAD and demonstrate that it largely derives from the cumulative effect of multiple common risk alleles individually of small effect size rather than rare variants with large effects on CAD risk. Despite this success, there has been limited progress in understanding the function of the novel loci; the majority of which are in noncoding regions of the genome.


Subject(s)
Coronary Artery Disease/genetics , Polymorphism, Single Nucleotide , Animals , Coronary Artery Disease/diagnosis , Coronary Artery Disease/epidemiology , Coronary Artery Disease/therapy , Gene Frequency , Genetic Association Studies , Genetic Markers , Genetic Predisposition to Disease , Genetic Testing , Humans , Phenotype , Predictive Value of Tests , Prognosis , Risk Assessment , Risk Factors
13.
Biochim Biophys Acta ; 1862(2): 223-32, 2016 02.
Article in English | MEDLINE | ID: mdl-26657055

ABSTRACT

Genome-wide association studies for plasma triglycerides and hepatic steatosis identified a risk locus on chromosome 8q24 close to the TRIB1 gene, encoding Tribbles Pseudokinase 1 (TRIB1). In previous studies conducted in murine models, hepatic over-expression of Trib1 was shown to increase fatty acid oxidation and decrease triglyceride synthesis whereas Trib1 knockdown mice exhibited hypertriglyceridemia. Here we have examined the impact of TRIB1 suppression in human and mouse hepatocytes. Examination of a panel of lipid regulator transcripts revealed species-specific effects, prompting us to focus on human models for the remainder of the study. Acute knockdown of TRIB1 in human primary hepatocytes resulted in decreased expression of MTTP and APOB, required for very low density lipoprotein (VLDL) assembly although particle secretion was not significantly affected. A parallel analysis performed in HepG2 revealed reduced MTTP, but not APOB, protein as a result of TRIB1 suppression. Global gene expression changes of human primary hepatocytes upon TRIB1 suppression were analyzed by clustering algorithms and found to be consistent with dysregulation of several pathways fundamental to liver function, including altered CEBPA and B transcript levels and impaired glucose handling. Indeed, TRIB1 expression in HepG2 cells was found to be inversely proportional to glucose concentration. Lastly TRIB1 downregulation in primary hepatocytes was associated with suppression of the HNF4A axis. In HepG2 cells, TRIB1 suppression resulted in reduced HNF4A protein levels while HNF4A suppression increased TRIB1 expression. Taken together these studies reveal an important role for TRIB1 in human hepatocyte biology.


Subject(s)
Hepatocytes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Fatty Liver/genetics , Fatty Liver/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
14.
Hum Mol Genet ; 24(2): 559-71, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25187575

ABSTRACT

C-reactive protein (CRP) concentration is a heritable systemic marker of inflammation that is associated with cardiovascular disease risk. Genome-wide association studies have identified CRP-associated common variants associated in ∼25 genes. Our aims were to apply exome sequencing to (1) assess whether the candidate loci contain rare coding variants associated with CRP levels and (2) perform an exome-wide search for rare variants in novel genes associated with CRP levels. We exome-sequenced 6050 European-Americans (EAs) and 3109 African-Americans (AAs) from the NHLBI-ESP and the CHARGE consortia, and performed association tests of sequence data with measured CRP levels. In single-variant tests across candidate loci, a novel rare (minor allele frequency = 0.16%) CRP-coding variant (rs77832441-A; p.Thr59Met) was associated with 53% lower mean CRP levels (P = 2.9 × 10(-6)). We replicated the association of rs77832441 in an exome array analysis of 11 414 EAs (P = 3.0 × 10(-15)). Despite a strong effect on CRP levels, rs77832441 was not associated with inflammation-related phenotypes including coronary heart disease. We also found evidence for an AA-specific association of APOE-ε2 rs7214 with higher CRP levels. At the exome-wide significance level (P < 5.0 × 10(-8)), we confirmed associations for reported common variants of HNF1A, CRP, IL6R and TOMM40-APOE. In gene-based tests, a burden of rare/lower frequency variation in CRP in EAs (P ≤ 6.8 × 10(-4)) and in retinoic acid receptor-related orphan receptor α (RORA) in AAs (P = 1.7 × 10(-3)) were associated with CRP levels at the candidate gene level (P < 2.0 × 10(-3)). This inquiry did not elucidate novel genes, but instead demonstrated that variants distributed across the allele frequency spectrum within candidate genes contribute to CRP levels.


Subject(s)
C-Reactive Protein/genetics , C-Reactive Protein/metabolism , Cardiovascular Diseases/blood , Exome , Adult , Black or African American/genetics , Cardiovascular Diseases/genetics , Cohort Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Genome-Wide Association Study , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Male , Plasma/chemistry , Polymorphism, Single Nucleotide , Receptors, Interleukin-6/genetics , Risk Factors , White People/genetics
15.
Circ Res ; 117(8): 671-83, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26195219

ABSTRACT

RATIONALE: Inflammation impairs macrophage cholesterol clearance from vascular tissues and promotes atherosclerosis. Inflammatory macrophages suppress expression of the transcription cofactor interferon regulatory factor 2-binding protein 2 (IRF2BP2), and genetic variants near IRF2BP2 associate with ischemic heart disease progression in humans. OBJECTIVES: To test whether IRF2BP2 in macrophages affects atherosclerosis in mice and humans. METHODS AND RESULTS: We generated mice that delete IRF2BP2 in macrophages. IRF2BP2-deficient macrophages worsened atherosclerosis in irradiated low-density lipoprotein receptor null-recipient mice and in apolipoprotein E null mice. IRF2BP2-deficient macrophages were inflammatory and had impaired cholesterol efflux because of their inability to activate the cholesterol transporter ABCA1 in response to cholesterol loading. Their expression of the anti-inflammatory transcription factor Krüppel-like factor 2 was markedly reduced. Promoter studies revealed that IRF2BP2 is required for MEF2-dependent activation of Krüppel-like factor 2. Importantly, restoring Krüppel-like factor 2 in IRF2BP2-deficient macrophages attenuated M1 inflammatory and rescued M2 anti-inflammatory gene activation and improved the cholesterol efflux deficit by restoring ABCA1 activation in response to cholesterol loading. In a cohort of 1066 angiographic cases and 1011 controls, homozygous carriers of a deletion polymorphism (rs3045215) in the 3' untranslated region sequence of human IRF2BP2 mRNA had a higher risk of coronary artery disease (recessive model, odds ratio [95% confidence interval]=1.560 [1.179-2.065], P=1.73E-03) and had lower IRF2BP2 (and Krüppel-like factor 2) protein levels in peripheral blood mononuclear cells. The effect of this deletion polymorphism to suppress protein expression was confirmed in luciferase reporter studies. CONCLUSION: Ablation of IRF2BP2 in macrophages worsens atherosclerosis in mice, and a deletion variant that lowers IRF2BP2 expression predisposes to coronary artery disease in humans.


Subject(s)
Atherosclerosis/prevention & control , Carrier Proteins/metabolism , Cholesterol/metabolism , Coronary Artery Disease/prevention & control , Inflammation/prevention & control , Macrophage Activation , Macrophages/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , ATP Binding Cassette Transporter 1/metabolism , Aged , Aged, 80 and over , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Carrier Proteins/genetics , Case-Control Studies , Cells, Cultured , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , DNA-Binding Proteins , Disease Models, Animal , Female , Genetic Predisposition to Disease , Homozygote , Humans , Inflammation/genetics , Inflammation/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , MEF2 Transcription Factors/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nuclear Proteins/genetics , Odds Ratio , Phenotype , Polymorphism, Genetic , Promoter Regions, Genetic , Protective Factors , Radiography , Receptors, LDL/deficiency , Receptors, LDL/genetics , Risk Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Transfection
16.
Arterioscler Thromb Vasc Biol ; 36(5): 972-83, 2016 05.
Article in English | MEDLINE | ID: mdl-26966274

ABSTRACT

OBJECTIVE: A recent genome-wide association study meta-analysis identified an intronic single nucleotide polymorphism in SMAD3, rs56062135C>T, the minor allele (T) which associates with protection from coronary artery disease. Relevant to atherosclerosis, SMAD3 is a key contributor to transforming growth factor-ß pathway signaling. Here, we seek to identify ≥1 causal coronary artery disease-associated single nucleotide polymorphisms at the SMAD3 locus and characterize mechanisms whereby the risk allele(s) contribute to coronary artery disease risk. APPROACH AND RESULTS: By genetic and epigenetic fine mapping, we identified a candidate causal single nucleotide polymorphism rs17293632C>T (D', 0.97; r(2), 0.94 with rs56062135) in intron 1 of SMAD3 with predicted functional effects. We show that the sequence encompassing rs17293632 acts as a strong enhancer in human arterial smooth muscle cells. The common allele (C) preserves an activator protein (AP)-1 site and enhancer function, whereas the protective (T) allele disrupts the AP-1 site and significantly reduces enhancer activity (P<0.001). Pharmacological inhibition of AP-1 activity upstream demonstrates that this allele-specific enhancer effect is AP-1 dependent (P<0.001). Chromatin immunoprecipitation experiments reveal binding of several AP-1 component proteins with preferential binding to the (C) allele. We show that rs17293632 is an expression quantitative trait locus for SMAD3 in blood and atherosclerotic plaque with reduced expression of SMAD3 in carriers of the protective allele. Finally, siRNA knockdown of SMAD3 in human arterial smooth muscle cells increases cell viability, consistent with an antiproliferative role. CONCLUSIONS: The coronary artery disease-associated rs17293632C>T single nucleotide polymorphism represents a novel functional cis-acting element at the SMAD3 locus. The protective (T) allele of rs17293632 disrupts a consensus AP-1 binding site in a SMAD3 intron 1 enhancer, reduces enhancer activity and SMAD3 expression, altering human arterial smooth muscle cell proliferation.


Subject(s)
Coronary Artery Disease/genetics , Coronary Artery Disease/prevention & control , Polymorphism, Single Nucleotide , Smad3 Protein/genetics , Binding Sites , Cell Proliferation , Cell Survival , Cells, Cultured , Coronary Artery Disease/diagnosis , Coronary Artery Disease/metabolism , Coronary Vessels/metabolism , Coronary Vessels/pathology , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Genome-Wide Association Study , Haplotypes , Humans , Introns , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Phenotype , Plaque, Atherosclerotic , Promoter Regions, Genetic , Protective Factors , Quantitative Trait Loci , RNA Interference , Risk Factors , Smad3 Protein/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transfection
17.
PLoS Genet ; 10(7): e1004502, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25033284

ABSTRACT

The majority of the heritability of coronary artery disease (CAD) remains unexplained, despite recent successes of genome-wide association studies (GWAS) in identifying novel susceptibility loci. Integrating functional genomic data from a variety of sources with a large-scale meta-analysis of CAD GWAS may facilitate the identification of novel biological processes and genes involved in CAD, as well as clarify the causal relationships of established processes. Towards this end, we integrated 14 GWAS from the CARDIoGRAM Consortium and two additional GWAS from the Ottawa Heart Institute (25,491 cases and 66,819 controls) with 1) genetics of gene expression studies of CAD-relevant tissues in humans, 2) metabolic and signaling pathways from public databases, and 3) data-driven, tissue-specific gene networks from a multitude of human and mouse experiments. We not only detected CAD-associated gene networks of lipid metabolism, coagulation, immunity, and additional networks with no clear functional annotation, but also revealed key driver genes for each CAD network based on the topology of the gene regulatory networks. In particular, we found a gene network involved in antigen processing to be strongly associated with CAD. The key driver genes of this network included glyoxalase I (GLO1) and peptidylprolyl isomerase I (PPIL1), which we verified as regulatory by siRNA experiments in human aortic endothelial cells. Our results suggest genetic influences on a diverse set of both known and novel biological processes that contribute to CAD risk. The key driver genes for these networks highlight potential novel targets for further mechanistic studies and therapeutic interventions.


Subject(s)
Coronary Artery Disease/genetics , Gene Regulatory Networks , Genetic Predisposition to Disease , Signal Transduction/genetics , Animals , Coronary Artery Disease/pathology , Gene Expression Regulation , Genome-Wide Association Study , Genomics , Humans , Mice
18.
PLoS Genet ; 10(7): e1004494, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25078778

ABSTRACT

Exome sequencing studies in complex diseases are challenged by the allelic heterogeneity, large number and modest effect sizes of associated variants on disease risk and the presence of large numbers of neutral variants, even in phenotypically relevant genes. Isolated populations with recent bottlenecks offer advantages for studying rare variants in complex diseases as they have deleterious variants that are present at higher frequencies as well as a substantial reduction in rare neutral variation. To explore the potential of the Finnish founder population for studying low-frequency (0.5-5%) variants in complex diseases, we compared exome sequence data on 3,000 Finns to the same number of non-Finnish Europeans and discovered that, despite having fewer variable sites overall, the average Finn has more low-frequency loss-of-function variants and complete gene knockouts. We then used several well-characterized Finnish population cohorts to study the phenotypic effects of 83 enriched loss-of-function variants across 60 phenotypes in 36,262 Finns. Using a deep set of quantitative traits collected on these cohorts, we show 5 associations (p<5×10⁻8) including splice variants in LPA that lowered plasma lipoprotein(a) levels (P = 1.5×10⁻¹¹7). Through accessing the national medical records of these participants, we evaluate the LPA finding via Mendelian randomization and confirm that these splice variants confer protection from cardiovascular disease (OR = 0.84, P = 3×10⁻4), demonstrating for the first time the correlation between very low levels of LPA in humans with potential therapeutic implications for cardiovascular diseases. More generally, this study articulates substantial advantages for studying the role of rare variation in complex phenotypes in founder populations like the Finns and by combining a unique population genetic history with data from large population cohorts and centralized research access to National Health Registers.


Subject(s)
Founder Effect , Genetic Diseases, Inborn , Genetic Drift , Genetics, Population , Exome/genetics , Female , Finland , Gene Frequency , Genetic Variation , Genome-Wide Association Study , Humans , Male , Phenotype , White People
19.
JAMA ; 317(9): 937-946, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28267856

ABSTRACT

Importance: The activity of lipoprotein lipase (LPL) is the rate-determining step in clearing triglyceride-rich lipoproteins from the circulation. Mutations that damage the LPL gene (LPL) lead to lifelong deficiency in enzymatic activity and can provide insight into the relationship of LPL to human disease. Objective: To determine whether rare and/or common variants in LPL are associated with early-onset coronary artery disease (CAD). Design, Setting, and Participants: In a cross-sectional study, LPL was sequenced in 10 CAD case-control cohorts of the multinational Myocardial Infarction Genetics Consortium and a nested CAD case-control cohort of the Geisinger Health System DiscovEHR cohort between 2010 and 2015. Common variants were genotyped in up to 305 699 individuals of the Global Lipids Genetics Consortium and up to 120 600 individuals of the CARDIoGRAM Exome Consortium between 2012 and 2014. Study-specific estimates were pooled via meta-analysis. Exposures: Rare damaging mutations in LPL included loss-of-function variants and missense variants annotated as pathogenic in a human genetics database or predicted to be damaging by computer prediction algorithms trained to identify mutations that impair protein function. Common variants in the LPL gene region included those independently associated with circulating triglyceride levels. Main Outcomes and Measures: Circulating lipid levels and CAD. Results: Among 46 891 individuals with LPL gene sequencing data available, the mean (SD) age was 50 (12.6) years and 51% were female. A total of 188 participants (0.40%; 95% CI, 0.35%-0.46%) carried a damaging mutation in LPL, including 105 of 32 646 control participants (0.32%) and 83 of 14 245 participants with early-onset CAD (0.58%). Compared with 46 703 noncarriers, the 188 heterozygous carriers of an LPL damaging mutation displayed higher plasma triglyceride levels (19.6 mg/dL; 95% CI, 4.6-34.6 mg/dL) and higher odds of CAD (odds ratio = 1.84; 95% CI, 1.35-2.51; P < .001). An analysis of 6 common LPL variants resulted in an odds ratio for CAD of 1.51 (95% CI, 1.39-1.64; P = 1.1 × 10-22) per 1-SD increase in triglycerides. Conclusions and Relevance: The presence of rare damaging mutations in LPL was significantly associated with higher triglyceride levels and presence of coronary artery disease. However, further research is needed to assess whether there are causal mechanisms by which heterozygous lipoprotein lipase deficiency could lead to coronary artery disease.


Subject(s)
Coronary Artery Disease/genetics , Lipoprotein Lipase/genetics , Mutation , Adult , Age of Onset , Case-Control Studies , Cross-Sectional Studies , Female , Genotype , Heterozygote , Humans , Lipoproteins/blood , Male , Middle Aged , Odds Ratio , Triglycerides/blood
20.
FASEB J ; 29(1): 336-45, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25342132

ABSTRACT

Insulin resistance may be linked to incomplete fatty acid ß-oxidation and the subsequent increase in acylcarnitine species in different tissues including skeletal muscle. It is not known if acylcarnitines participate in muscle insulin resistance or simply reflect dysregulated metabolism. The aims of this study were to determine whether acylcarnitines can elicit muscle insulin resistance and to better understand the link between incomplete muscle fatty acid ß-oxidation, oxidative stress, inflammation, and insulin-resistance development. Differentiated C2C12, primary mouse, and human myotubes were treated with acylcarnitines (C4:0, C14:0, C16:0) or with palmitate with or without carnitine acyltransferase inhibition by mildronate. Treatment with C4:0, C14:0, and C16:0 acylcarnitines resulted in 20-30% decrease in insulin response at the level of Akt phosphorylation and/or glucose uptake. Mildronate reversed palmitate-induced insulin resistance concomitant with an ∼25% decrease in short-chain acylcarnitine and acetylcarnitine secretion. Although proinflammatory cytokines were not affected under these conditions, oxidative stress was increased by 2-3 times by short- or long-chain acylcarnitines. Acylcarnitine-induced oxidative stress and insulin resistance were reversed by treatment with antioxidants. Results are consistent with the conclusion that incomplete muscle fatty acid ß-oxidation causes acylcarnitine accumulation and associated oxidative stress, raising the possibility that these metabolites play a role in muscle insulin resistance.


Subject(s)
Carnitine/analogs & derivatives , Insulin Resistance/physiology , Muscle, Skeletal/metabolism , Adult , Animals , Antioxidants/pharmacology , Carnitine/metabolism , Case-Control Studies , Cell Line , Cells, Cultured , Cytokines/metabolism , Fatty Acids/metabolism , Female , Humans , Inflammation/metabolism , Mice , Middle Aged , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Obesity/metabolism , Oxidation-Reduction , Oxidative Stress
SELECTION OF CITATIONS
SEARCH DETAIL