Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Lipid Res ; 58(4): 752-762, 2017 04.
Article in English | MEDLINE | ID: mdl-28167703

ABSTRACT

Reverse cholesterol transport (RCT) is thought to be an atheroprotective function of HDL, and macrophage-specific RCT in mice is inversely associated with atherosclerosis. We developed a novel method using 3H-cholesterol nanoparticles to selectively trace macrophage-specific RCT in vivo in humans. Use of 3H-cholesterol nanoparticles was initially tested in mice to assess the distribution of tracer and response to interventions known to increase RCT. Thirty healthy subjects received 3H-cholesterol nanoparticles intravenously, followed by blood and stool sample collection. Tracer counts were assessed in plasma, nonHDL, HDL, and fecal fractions. Data were analyzed by using multicompartmental modeling. Administration of 3H-cholesterol nanoparticles preferentially labeled macrophages of the reticuloendothelial system in mice, and counts were increased in mice treated with a liver X receptor agonist or reconstituted HDL, as compared with controls. In humans, tracer disappeared from plasma rapidly after injection of nanoparticles, followed by reappearance in HDL and nonHDL fractions. Counts present as free cholesterol increased rapidly and linearly in the first 240 min after nadir; counts in cholesteryl ester increased steadily over time. Estimates of fractional transfer rates of key RCT steps were obtained. These results support the use of 3H-cholesterol nanoparticles as a feasible approach for the measurement of macrophage RCT in vivo in humans.


Subject(s)
Atherosclerosis/blood , Cholesterol, HDL/blood , Cholesterol/blood , Lipoproteins, HDL/metabolism , Adolescent , Adult , Aged , Animals , Atherosclerosis/pathology , Biological Transport/genetics , Cholesterol/chemistry , Cholesterol/genetics , Cholesterol, HDL/chemistry , Cholesterol, HDL/isolation & purification , Feces/chemistry , Female , Humans , Lipoproteins, HDL/isolation & purification , Liver/metabolism , Liver/pathology , Liver X Receptors/agonists , Liver X Receptors/blood , Macrophages/metabolism , Male , Mice , Middle Aged , Nanoparticles/administration & dosage , Nanoparticles/chemistry
2.
Cell Metab ; 24(2): 223-33, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27508871

ABSTRACT

The development of LXR agonists for the treatment of coronary artery disease has been challenged by undesirable properties in animal models. Here we show the effects of an LXR agonist on lipid and lipoprotein metabolism and neutrophils in human subjects. BMS-852927, a novel LXRß-selective compound, had favorable profiles in animal models with a wide therapeutic index in cynomolgus monkeys and mice. In healthy subjects and hypercholesterolemic patients, reverse cholesterol transport pathways were induced similarly to that in animal models. However, increased plasma and hepatic TG, plasma LDL-C, apoB, apoE, and CETP and decreased circulating neutrophils were also evident. Furthermore, similar increases in LDL-C were observed in normocholesterolemic subjects and statin-treated patients. The primate model markedly underestimated human lipogenic responses and did not predict human neutrophil effects. These studies demonstrate both beneficial and adverse LXR agonist clinical responses and emphasize the importance of further translational research in this area.


Subject(s)
Cell Movement , Imidazoles/adverse effects , Imidazoles/pharmacology , Lipid Metabolism , Lipoproteins/metabolism , Liver X Receptors/agonists , Neutrophils/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Adipose Tissue/metabolism , Adolescent , Adult , Animals , Cell Movement/drug effects , Cholesterol/blood , Cholesterol/metabolism , Healthy Volunteers , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Imidazoles/therapeutic use , Leukocyte Count , Lipoproteins/blood , Macaca fascicularis , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mononuclear Phagocyte System/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Triglycerides/blood , Young Adult
3.
Cell Metab ; 24(2): 234-45, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27508872

ABSTRACT

Human genetics studies have implicated GALNT2, encoding GalNAc-T2, as a regulator of high-density lipoprotein cholesterol (HDL-C) metabolism, but the mechanisms relating GALNT2 to HDL-C remain unclear. We investigated the impact of homozygous GALNT2 deficiency on HDL-C in humans and mammalian models. We identified two humans homozygous for loss-of-function mutations in GALNT2 who demonstrated low HDL-C. We also found that GALNT2 loss of function in mice, rats, and nonhuman primates decreased HDL-C. O-glycoproteomics studies of a human GALNT2-deficient subject validated ANGPTL3 and ApoC-III as GalNAc-T2 targets. Additional glycoproteomics in rodents identified targets influencing HDL-C, including phospholipid transfer protein (PLTP). GALNT2 deficiency reduced plasma PLTP activity in humans and rodents, and in mice this was rescued by reconstitution of hepatic Galnt2. We also found that GALNT2 GWAS SNPs associated with reduced HDL-C also correlate with lower hepatic GALNT2 expression. These results posit GALNT2 as a direct modulator of HDL metabolism across mammals.


Subject(s)
Lipoproteins, HDL/metabolism , N-Acetylgalactosaminyltransferases/deficiency , Amino Acid Sequence , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Animals , Base Sequence , Cholesterol, HDL/blood , Gene Knockdown Techniques , Glycoproteins/metabolism , Homozygote , Humans , Liver/enzymology , Mice , Mice, Knockout , Models, Animal , Mutation/genetics , N-Acetylgalactosaminyltransferases/chemistry , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Phenotype , Phospholipid Transfer Proteins/metabolism , Polymorphism, Single Nucleotide/genetics , Primates , Proteomics , Rats , Triglycerides/metabolism , Polypeptide N-acetylgalactosaminyltransferase
4.
Cell Metab ; 21(2): 334-347, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25651185

ABSTRACT

Insulin resistance (IR) is a complex trait with multiple genetic and environmental components. Confounded by large differences between the sexes, environment, and disease pathology, the genetic basis of IR has been difficult to dissect. Here we examine IR and related traits in a diverse population of more than 100 unique male and female inbred mouse strains after feeding a diet rich in fat and refined carbohydrates. Our results show dramatic variation in IR among strains of mice and widespread differences between sexes that are dependent on genotype. We uncover more than 15 genome-wide significant loci and validate a gene, Agpat5, associated with IR. We also integrate plasma metabolite levels and global gene expression from liver and adipose tissue to identify metabolite quantitative trait loci (mQTL) and expression QTL (eQTL), respectively. Our results provide a resource for analysis of interactions between diet, sex, and genetic background in IR.


Subject(s)
Insulin Resistance/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/genetics , 1-Acylglycerol-3-Phosphate O-Acyltransferase/metabolism , Animals , Diet, High-Fat , Dietary Carbohydrates , Female , Genetic Variation/genetics , Genotype , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA
5.
J Pharmacol Exp Ther ; 352(2): 305-14, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25467132

ABSTRACT

Liver X Receptors (LXRs) α and ß are nuclear hormone receptors that regulate multiple genes involved in reverse cholesterol transport (RCT) and are potential drug targets for atherosclerosis. However, full pan agonists also activate lipogenic genes, resulting in elevated plasma and hepatic lipids. We report the pharmacology of BMS-779788 [2-(2-(1-(2-chlorophenyl)-1-methylethyl)-1-(3'-(methylsulfonyl)-4-biphenylyl)-1H-imidazol-4-yl)-2-propanol], a potent partial LXR agonist with LXRß selectivity, which has an improved therapeutic window in the cynomolgus monkey compared with a full pan agonist. BMS-779788 induced LXR target genes in blood in vivo with an EC50 = 610 nM, a value similar to its in vitro blood gene induction potency. BMS-779788 was 29- and 12-fold less potent than the full agonist T0901317 in elevating plasma triglyceride and LDL cholesterol, respectively, with similar results for plasma cholesteryl ester transfer protein and apolipoprotein B. However, ABCA1 and ABCG1 mRNA inductions in blood, which are critical for RCT, were comparable. Increased liver triglyceride was observed after 7-day treatment with BMS-779788 at the highest dose tested and was nearly identical to the dose response for plasma triglyceride, consistent with the central role of liver LXR in these lipogenic effects. Dose-dependent increases in biliary cholesterol and decreases in phospholipid and bile acid occurred in BMS-779788-treated animals, similar to LXR agonist effects reported in mouse. In summary, BMS-779788, a partial LXRß selective agonist, has decreased lipogenic potential compared with a full pan agonist in cynomolgus monkeys, with similar potency in the induction of genes known to stimulate RCT. This provides support in nonhuman primates for improving LXR agonist therapeutic windows by limiting LXRα activity.


Subject(s)
Anticholesteremic Agents/pharmacology , Imidazoles/pharmacology , Liver/drug effects , Orphan Nuclear Receptors/agonists , Sulfones/pharmacology , ATP-Binding Cassette Transporters/blood , ATP-Binding Cassette Transporters/genetics , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/blood , Dose-Response Relationship, Drug , Drug Partial Agonism , Imidazoles/administration & dosage , Imidazoles/blood , Lipids/blood , Lipogenesis/drug effects , Liver/metabolism , Liver X Receptors , Macaca fascicularis , Male , Sulfones/administration & dosage , Sulfones/blood , Triglycerides/metabolism
6.
Hum Mol Genet ; 22(15): 3023-37, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23562819

ABSTRACT

The genetics of messenger RNA (mRNA) expression has been extensively studied in humans and other organisms, but little is known about genetic factors contributing to microRNA (miRNA) expression. We examined natural variation of miRNA expression in adipose tissue in a population of 200 men who have been carefully characterized for metabolic syndrome (MetSyn) phenotypes as part of the Metabolic Syndrome in Men (METSIM) study. We genotyped the subjects using high-density single-nucleotide polymorphism microarrays and quantified the mRNA abundance using genome-wide expression arrays and miRNA abundance using next-generation sequencing. We reliably quantified 356 miRNA species that were expressed in human adipose tissue, a limited number of which made up most of the expressed miRNAs. We mapped the miRNA abundance as an expression quantitative trait and determined cis regulation of expression for nine of the miRNAs and of the processing of one miRNA (miR-28). The degree of genetic variation of miRNA expression was substantially less than that of mRNAs. For the majority of the miRNAs, genetic regulation of expression was independent of the expression of mRNA from which the miRNA is transcribed. We also showed that for 108 miRNAs, mapped reads displayed widespread variation from the canonical sequence. We found a total of 24 miRNAs to be significantly associated with MetSyn traits. We suggest a regulatory role for miR-204-5p which was predicted to inhibit acetyl coenzyme A carboxylase ß, a key fatty acid oxidation enzyme that has been shown to play a role in regulating body fat and insulin resistance in adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Quantitative Trait, Heritable , Genetic Association Studies , Humans , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Phenotype , Polymorphism, Single Nucleotide , Quantitative Trait Loci , RNA Interference , RNA Processing, Post-Transcriptional , Transcription, Genetic , Transcriptome
7.
PLoS One ; 7(5): e38240, 2012.
Article in English | MEDLINE | ID: mdl-22666496

ABSTRACT

Statin therapy reduces the risk of coronary heart disease (CHD), however, the person-to-person variability in response to statin therapy is not well understood. We have investigated the effect of genetic variation on the reduction of CHD events by pravastatin. First, we conducted a genome-wide association study of 682 CHD cases from the Cholesterol and Recurrent Events (CARE) trial and 383 CHD cases from the West of Scotland Coronary Prevention Study (WOSCOPS), two randomized, placebo-controlled studies of pravastatin. In a combined case-only analysis, 79 single nucleotide polymorphisms (SNPs) were associated with differential CHD event reduction by pravastatin according to genotype (P<0.0001), and these SNPs were analyzed in a second stage that included cases as well as non-cases from CARE and WOSCOPS and patients from the PROspective Study of Pravastatin in the Elderly at Risk/PHArmacogenomic study of Statins in the Elderly at risk for cardiovascular disease (PROSPER/PHASE), a randomized placebo controlled study of pravastatin in the elderly. We found that one of these SNPs (rs13279522) was associated with differential CHD event reduction by pravastatin therapy in all 3 studies: P = 0.002 in CARE, P = 0.01 in WOSCOPS, P = 0.002 in PROSPER/PHASE. In a combined analysis of CARE, WOSCOPS, and PROSPER/PHASE, the hazard ratio for CHD when comparing pravastatin with placebo decreased by a factor of 0.63 (95% CI: 0.52 to 0.75) for each extra copy of the minor allele (P = 4.8 × 10(-7)). This SNP is located in DnaJ homolog subfamily C member 5B (DNAJC5B) and merits investigation in additional randomized studies of pravastatin and other statins.


Subject(s)
Coronary Disease/genetics , Coronary Disease/prevention & control , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Pravastatin/pharmacology , Aged , Coronary Disease/drug therapy , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Pravastatin/therapeutic use
8.
Hum Genet ; 129(1): 17-23, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20886236

ABSTRACT

A single nucleotide polymorphism (SNP) in KIF6, a member of the KIF9 family of kinesins, is associated with differential coronary event reduction from statin therapy in four randomized controlled trials; this SNP (rs20455) is also associated with the risk for coronary heart disease (CHD) in multiple prospective studies. We investigated whether other common SNPs in the KIF6 region were associated with event reduction from statin therapy. Of the 170 SNPs in the KIF6 region investigated in the Cholesterol and Recurrent Events trial (CARE), 28 were associated with differential event reduction from statin therapy (P (interaction) < 01 in Caucasians, adjusted for age and sex) and were further investigated in the Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis In Myocardial Infarction 22 (PROVE IT-TIMI22) and West of Scotland Coronary Prevention Study (WOSCOPS). These analyses revealed that two SNPs (rs9462535 and rs9471077), in addition to rs20455, were associated with event reduction from statin therapy (P (interaction) < 0.1 in each of the three studies). The relative risk reduction ranged from 37 to 50% (P < 0.01) in carriers of the minor alleles of these SNPs and from -4 to 13% (P > 0.4) in non-carriers. These three SNPs are in high linkage disequilibrium with one another (r (2) > 0.84). Functional studies of these variants may help to understand the role of KIF6 in the pathogenesis of CHD and differential response to statin therapy.


Subject(s)
Coronary Disease/drug therapy , Coronary Disease/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Kinesins/genetics , Polymorphism, Single Nucleotide , Chromosome Mapping , Coronary Disease/prevention & control , Female , Humans , Linkage Disequilibrium , Male , Meta-Analysis as Topic , Middle Aged , Randomized Controlled Trials as Topic
9.
Am J Cardiol ; 105(9): 1300-5, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20403483

ABSTRACT

A previous genetic analysis of the Cholesterol and Recurrent Events (CARE) trial found that carriers of the 719Arg allele of the kinesin family member 6 gene (KIF6) (rs20455), but not noncarriers, received significant event reduction from pravastatin therapy. However, that previous analysis of CARE included only Caucasian patients and was limited to the myocardial infarction components of the primary end point. Therefore, the aim of this study was to investigate whether pravastatin therapy reduced primary end point events in KIF6 719Arg carriers and noncarriers, separately, in the combined ethnic groups of CARE. The effect of pravastatin therapy on primary end point events (fatal coronary event or nonfatal myocardial infarction) was investigated in Cox regression models that adjusted for population structure using either self-reported ethnicity or the principal components of genetic heterogeneity. After adjustment for age, gender, and self-reported ethnicity, pravastatin therapy reduced events in carriers of KIF6 719Arg (hazard ratio [HR] 0.63, 95% confidence interval [CI] 0.49 to 0.83) but not in noncarriers (HR 1.01, 95% CI 0.69 to 1.45) (p for interaction = 0.049). After adjustment for age, gender, traditional risk factors, and principal components, pravastatin therapy reduced events in carriers of 719Arg (HR 0.64, 95% CI 0.49 to 0.85) but not in noncarriers (HR 0.90, 95% CI 0.62 to 1.32) (p for interaction = 0.14). In conclusion, in an analysis that included CARE patients of all ethnic groups, pravastatin therapy significantly and substantially reduced primary end point events in carriers of the KIF6 719Arg allele but not in noncarriers.


Subject(s)
Atherosclerosis/genetics , Cholesterol/blood , DNA/genetics , Kinesins/genetics , Myocardial Infarction/drug therapy , Polymorphism, Genetic , Pravastatin/therapeutic use , Alleles , Atherosclerosis/blood , Atherosclerosis/complications , Dose-Response Relationship, Drug , Double-Blind Method , Female , Genetic Predisposition to Disease , Genotype , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Middle Aged , Myocardial Infarction/epidemiology , Myocardial Infarction/genetics , Polymerase Chain Reaction , Pravastatin/administration & dosage , Prevalence , Prospective Studies , Risk Factors , Secondary Prevention , Survival Rate/trends , Treatment Outcome , United States/epidemiology
10.
Am J Hum Genet ; 86(3): 399-410, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20170901

ABSTRACT

Gene by environment (GxE) interactions are clearly important in many human diseases, but they have proven to be difficult to study on a molecular level. We report genetic analysis of thousands of transcript abundance traits in human primary endothelial cell (EC) lines in response to proinflammatory oxidized phospholipids implicated in cardiovascular disease. Of the 59 most regulated transcripts, approximately one-third showed evidence of GxE interactions. The interactions resulted primarily from effects of distal-, trans-acting loci, but a striking example of a local-GxE interaction was also observed for FGD6. Some of the distal interactions were validated by siRNA knockdown experiments, including a locus involved in the regulation of multiple transcripts involved in the ER stress pathway. Our findings add to the understanding of the overall architecture of complex human traits and are consistent with the possibility that GxE interactions are responsible, in part, for the failure of association studies to more fully explain common disease variation.


Subject(s)
Gene Expression Regulation , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Environment , Female , Gene Expression Regulation/drug effects , Genetic Variation , Genome-Wide Association Study , Humans , Male , Models, Genetic , Oligonucleotide Array Sequence Analysis , Phosphatidylcholines/pharmacology , Polymorphism, Single Nucleotide , Quantitative Trait Loci , RNA, Small Interfering/genetics , Systems Biology , Transcription, Genetic
11.
Arterioscler Thromb Vasc Biol ; 28(3): 562-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18174457

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the association between the Ala227Pro polymorphism in the ADAMTS1 metalloproteinase gene and coronary heart disease and benefit from statin therapy in 2 independent cohorts. METHODS AND RESULTS: The frequency of the ADAMTS1 227Pro minor allele was 0.24 in 2421 male subjects from CARE, a randomized trial of pravastatin versus placebo. In the placebo arm, homozygotes (6.3% of study population) had a significantly increased risk of fatal coronary disease or nonfatal myocardial infarction (D/MI) compared with noncarriers (OR 2.12, 95% CI 1.07 to 4.19, P=0.03), and in the entire study the benefit of pravastatin in reducing the risk of D/MI was greater in these subjects (OR 0.21, 95% CI 0.06 to 0.69) than in heterozygotes (OR 0.74, 95% CI 0.48 to 1.14) or noncarriers (OR 0.99, 95% CI 0.68 to 1.42; P(interaction)=0.044). Results were tested in 1565 male subjects from WOSCOPS, also a randomized trial of pravastatin versus placebo. Similar to the results in CARE, in the placebo arm subjects homozygous for the minor allele were at increased risk of D/MI (OR 1.72, P=0.052) and in the entire study the benefit of pravastatin in reducing D/MI was greater in these subjects (OR 0.24, 95% CI 0.09 to 0.68) than in heterozygotes (OR 0.73, 95% CI 0.48 to 1.11) or noncarriers (OR 0.65, 95% CI 0.20 to 2.09) (P(interaction)=0.029). CONCLUSIONS: In men not on pravastatin, those homozygous for the 227Pro allele of ADAMTS1 have a nearly 2-fold increased risk of coronary heart disease events compared with noncarriers. In this high-risk group, treatment with pravastatin is highly efficacious, reducing the odds of fatal coronary disease or nonfatal MI by approximately 75%, as compared with 25% in noncarriers or heterozygotes.


Subject(s)
Coronary Disease/drug therapy , Coronary Disease/genetics , Matrix Metalloproteinases/genetics , Polymorphism, Genetic , Pravastatin/therapeutic use , Adult , Age Factors , Analysis of Variance , Coronary Disease/mortality , Dose-Response Relationship, Drug , Drug Administration Schedule , Follow-Up Studies , Genetic Variation , Genotype , Humans , Male , Matrix Metalloproteinases/metabolism , Middle Aged , Predictive Value of Tests , Probability , Reference Values , Risk Assessment , Severity of Illness Index , Survival Analysis , Treatment Outcome
12.
J Am Coll Cardiol ; 51(4): 435-43, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18222353

ABSTRACT

OBJECTIVES: We asked whether 35 genetic polymorphisms, previously found to be associated with cardiovascular disease, were associated with myocardial infarction (MI) in the CARE (Cholesterol and Recurrent Events) trial and with coronary heart disease (CHD) in the WOSCOPS (West of Scotland Coronary Prevention Study) trial and whether the risk associated with these polymorphisms could be reduced by pravastatin treatment. BACKGROUND: Identification of genetic polymorphisms associated with CHD may improve assessment of CHD risk and understanding of disease pathophysiology. METHODS: We tested the association between genotype and recurrent MI in the CARE study and between genotype and primary CHD in the WOSCOPS trial using regression models that adjusted for conventional risk factors: Cox proportional hazards models for the CARE study and conditional logistic regression models for a nested case-control study of the WOSCOPS trial. RESULTS: We found that Trp719Arg (rs20455) in KIF6 was associated with coronary events. KIF6 encodes kinesin-like protein 6, a member of the molecular motor superfamily. In placebo-treated patients, carriers of the KIF6 719Arg allele (59.4% of the CARE trial cohort) had a hazard ratio of 1.50 (95% confidence interval [CI] 1.05 to 2.15) in the CARE trial and an odds ratio of 1.55 (95% CI 1.14 to 2.09) in the WOSCOPS trial. Among carriers, the absolute risk reduction by pravastatin was 4.89% (95% CI 1.81% to 7.97%) in the CARE trial and 5.49% (95% CI 3.52% to 7.46%) in the WOSCOPS trial. CONCLUSIONS: In both the CARE and the WOSCOPS trials, carriers of the KIF6 719Arg allele had an increased risk of coronary events, and pravastatin treatment substantially reduced that risk.


Subject(s)
Coronary Disease/genetics , Kinesins/genetics , Myocardial Infarction/genetics , Polymorphism, Genetic , Anticholesteremic Agents/therapeutic use , Case-Control Studies , Coronary Disease/drug therapy , Female , Gene Frequency , Genotype , Humans , Logistic Models , Male , Middle Aged , Myocardial Infarction/drug therapy , Pravastatin/therapeutic use , Proportional Hazards Models , Prospective Studies , Risk Factors , Treatment Outcome
13.
J Am Coll Cardiol ; 51(4): 449-55, 2008 Jan 29.
Article in English | MEDLINE | ID: mdl-18222355

ABSTRACT

OBJECTIVES: We explored whether the benefit of intensive versus moderate statin therapy would be greater in carriers of KIF6 719Arg than in noncarriers. BACKGROUND: The 719Arg variant of Trp719Arg (rs20455), a polymorphism in kinesin-like protein 6, is associated with greater risk of coronary events and greater benefit from pravastatin versus placebo. METHODS: We genotyped 1,778 acute coronary syndrome patients within the PROVE IT-TIMI 22 (Pravastatin or Atorvastatin Evaluation and Infection Therapy: Thrombolysis in Myocardial Infarction 22) trial and investigated different intensities of statin therapy in carriers of 719Arg and in noncarriers using Cox proportional hazards models that adjusted for traditional risk factors. RESULTS: Benefit from intensive, compared with moderate, statin therapy was significantly greater in the 59% of the cohort who were carriers (hazard ratio [HR] 0.59, 95% confidence interval [CI] 0.45 to 0.77) than in those who were noncarriers (HR 0.94, 95% CI 0.70 to 1.27; p = 0.018 for interaction between 719Arg carrier status and treatment). Absolute risk reduction was 10.0% in carriers versus 0.8% in noncarriers. The benefit of intensive therapy in carriers was significant as early as day 30 of therapy. Carriers and noncarriers did not differ in on-treatment low-density lipoprotein cholesterol, triglyceride, or C-reactive protein (CRP) levels. CONCLUSIONS: Carriers of 719Arg receive significantly greater benefit from intensive statin therapy than do noncarriers, a superior benefit that appears to be due to a mechanism distinct from lipid or CRP lowering. Functional studies of the KIF6 kinesin are warranted, given the consistent association of Trp719Arg with risk of coronary events and statin benefit.


Subject(s)
Coronary Disease/drug therapy , Coronary Disease/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Kinesins/genetics , Polymorphism, Genetic , Atorvastatin , Cohort Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Heptanoic Acids/therapeutic use , Heterozygote , Humans , Male , Middle Aged , Pravastatin/therapeutic use , Pyrroles/therapeutic use , Risk Factors , Treatment Outcome
14.
Arterioscler Thromb Vasc Biol ; 26(12): 2763-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17008591

ABSTRACT

OBJECTIVE: Statins reduce inflammation and risk of myocardial infarction (MI). Because the myeloid IgA Fc receptor encoded by FCAR mediates inflammation, we hypothesized that the FCAR Asp92Asn polymorphism is associated with risk of MI and that this risk would be modified by pravastatin. METHODS AND RESULTS: In the placebo arm of the Cholesterol and Recurrent Events (CARE) study, male carriers of the 92Asn allele had an adjusted hazard ratio for incident MI of 1.68 (95% CI 1.10 to 2.57); relative risk reduction by pravastatin was 69% in carriers and 12% in noncarriers (P(interaction)=0.007). In the placebo arm of the all-male West of Scotland Coronary Prevention Study (WOSCOPS), carriers had an adjusted odds ratio for incident coronary heart disease (CHD) of 1.46 (90% CI 1.05 to 2.03); for pravastatin compared with placebo treatment, the adjusted odds ratios were 0.55 (95% CI 0.32 to 0.93) in carriers and 0.65 (95% CI 0.51 to 0.83) in noncarriers (P(interaction)=0.55). CONCLUSIONS: Carriers of 92Asn had increased risk of MI in CARE and increased odds of CHD in WOSCOPS. Pravastatin significantly reduced risk in carriers in both CARE and WOSCOPS. A genotype by treatment interaction was observed in CARE but not in WOSCOPS.


Subject(s)
Antigens, CD/genetics , Asparagine/genetics , Aspartic Acid/genetics , Myocardial Infarction/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Fc/genetics , Alleles , Anticholesteremic Agents/therapeutic use , Coronary Disease/etiology , Coronary Disease/genetics , Coronary Disease/prevention & control , Genetic Predisposition to Disease/genetics , Genotype , Humans , Male , Myocardial Infarction/etiology , Myocardial Infarction/prevention & control , Odds Ratio , Pravastatin/therapeutic use , Risk Factors , Scotland
15.
Proc Natl Acad Sci U S A ; 103(34): 12741-6, 2006 Aug 22.
Article in English | MEDLINE | ID: mdl-16912112

ABSTRACT

Oxidized phospholipids are thought to promote atherogenesis by stimulating endothelial cells (ECs) to produce inflammatory cytokines, such as IL-8. In studies with mouse models, we previously demonstrated that genetic variation in inflammatory responses of endothelial cells to oxidized lipids contributes importantly to atherosclerosis susceptibility. We now show that similar variations occur in cultured aortic ECs derived from multiple heart transplant donors. These variations were stably maintained between passages and, thus, reflect either genetic or epigenetic regulatory differences. Expression array analysis of aortic EC cultures derived from 12 individuals revealed that >1,000 genes were regulated by oxidized phospholipids. We have used the observed variations in the sampled population to construct a gene coexpression network comprised of 15 modules of highly connected genes. We show that several identified modules are significantly enriched in genes for known pathways and confirm a module enriched for unfolded protein response (UPR) genes using siRNA and the UPR inducer tunicamycin. On the basis of the constructed network, we predicted that a gene of unknown function (MGC4504) present in the UPR module is a target for UPR transcriptional activator ATF4. Our data also indicate that IL-8 is present in the UPR module and is regulated, in part, by the UPR. We validate these by using siRNA. In conclusion, we show that interindividual variability can be used to group genes into pathways and predict gene-gene regulatory relationships, thus identifying targets potentially involved in susceptibility to common diseases such as atherosclerosis.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Phospholipids/pharmacology , Activating Transcription Factor 4/genetics , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation , Humans , Interleukin-8/genetics , Oxidation-Reduction , Phospholipids/chemistry , Protein Folding
16.
Arterioscler Thromb Vasc Biol ; 26(11): 2490-6, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16931790

ABSTRACT

OBJECTIVE: Oxidized 1-palmitoyl-2-arachidonyl-sn-3-glycero-phosphorylcholine (oxPAPC) accumulates in atherosclerotic lesions and in vitro studies suggest that it mediates chronic inflammatory response in endothelial cells (ECs). The goal of our studies was to identify pathways mediating the induction of inflammatory genes by oxPAPC. METHODS AND RESULTS: Using expression arrays, quantitative polymerase chain reaction (PCR), and immunoblotting we demonstrate that oxPAPC leads to endoplasmic reticulum stress and activation of the unfolded protein response (UPR) in human aortic ECs. Immunohistochemistry analysis of human atherosclerotic lesions indicated that UPR is induced in areas containing oxidized phospholipids. Using the UPR inducing agent tunicamycin and selective siRNA targeting of the ATF4 and XBP1 branches of the UPR, we demonstrate that these transcription factors are essential mediators of IL8, IL6, and MCP1 expression in human aortic ECs required for maximal inflammatory gene expression in the basal state and after oxPAPC treatment. We also identify a novel oxPAPC-induced chemokine, the CXC motif ligand 3 (CXCL3), and show that its expression requires XBP1. CONCLUSIONS: These data suggest that the UPR pathway is a general mediator of vascular inflammation and EC dysfunction in atherosclerosis, and, likely, other inflammatory disorders.


Subject(s)
Endothelial Cells/metabolism , Gene Expression Regulation , Inflammation/genetics , Protein Folding , Activating Transcription Factor 4/metabolism , Aorta/cytology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Cells, Cultured , Chromosome Mapping , DNA-Binding Proteins/metabolism , Gene Expression/drug effects , Humans , Immunologic Techniques , Nuclear Proteins/metabolism , Oxidation-Reduction , Phosphatidylcholines/pharmacology , Phospholipids/metabolism , Regulatory Factor X Transcription Factors , Tissue Distribution , Transcription Factors , X-Box Binding Protein 1
17.
Stroke ; 36(11): 2346-50, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16239632

ABSTRACT

BACKGROUND AND PURPOSE: The paraoxonases are involved in protecting low-density lipoprotein (LDL) from lipid oxidation. Paraoxonase 1 (PON1) was implicated in susceptibility to coronary artery disease and stroke in previous studies. We evaluated, in a comprehensive way, all 3 paraoxonase genes for association with stroke observed in the Cholesterol and Recurrent Events (CARE) trial. METHODS: Over 2500 subjects enrolled in the CARE trial were genotyped for 14 single nucleotide polymorphisms, including 7 newly identified in this study, in the 3 paraoxonase genes. RESULTS: A glutamine (Gln)/arginine (Arg) polymorphism at amino acid residue 192 in PON1 was significantly associated with stroke (P=0.003 in multivariate analysis, including age, sex, LDL, hypertension, diabetes, smoking, and pravastatin treatment as covariates). The odds ratios were 2.28 (95% CI, 1.38 to 3.79) for Gln/Arg heterozygotes and 2.47 (95% CI, 1.18 to 5.19) for Arg/Arg homozygotes compared with Gln/Gln homozygotes. These results are consistent with 2 of 3 other published studies. In combined analysis of all 4 studies, the association between Gln192Arg SNP and stroke was highly significant (chi2(8df)=45.58, P<0.000001). Sequence analysis of the PON1 gene from seventy stroke cases revealed a novel nonsense mutation at codon 32 in one stroke case, which was not detected in over 2500 unaffected individuals. Polymorphisms in the PON2 and PON3 genes were not associated with stroke. CONCLUSIONS: These results suggest that Gln192Arg genotype is an important risk factor for stroke.


Subject(s)
Aryldialkylphosphatase/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Polymorphism, Genetic , Stroke/genetics , Arginine/chemistry , Female , Genotype , Glutamine/chemistry , Heterozygote , Homozygote , Humans , Lipoproteins, LDL/metabolism , Male , Multivariate Analysis , Odds Ratio , Polymorphism, Single Nucleotide , Pravastatin/therapeutic use , Risk , Risk Factors
18.
J Lipid Res ; 45(8): 1410-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15145986

ABSTRACT

Liver X receptors (LXRs) are ligand-activated transcription factors that belong to the nuclear receptor superfamily. LXRs activate transcription of a spectrum of genes that regulate reverse cholesterol transport, including the ATP binding cassette transporter A1 (ABCA1), and raise HDL cholesterol (HDL-C) levels. However, LXR agonists also induce genes that stimulate lipogenesis, including the sterol response element binding protein (SREBP1-c) and fatty acid synthetase (FAS). The induction of these genes in the liver cause increased hepatic triglyceride synthesis, hypertriglyceridemia, and hepatic steatosis. As LXR response elements have been identified in these promoters, it is not clear if these two processes can be separated. Herein, we demonstrate that plasma HDL-C elevation and intestinal ABCA1 induction can occur with relatively little induction of FAS and SREBP1-c in mouse liver via a selective LXR modulator GW3965. This is in contrast to the strong induction of hepatic lipogenic genes by the well-characterized LXR agonist T0901317 (T317). Consistent with the in vivo results, GW3965 is a very weak LXR activator compared with T317 in human hepatoma cells. GW3965-liganded LXR recruits selected coactivators less effectively than T317 and may explain in part the tissue selective gene induction. This demonstration that tissue and gene selective modulation is possible with selective LXR modulators has positive implications for the development of this class of antiatherosclerotic agents.


Subject(s)
Benzoates/pharmacology , Benzylamines/pharmacology , Cholesterol, HDL/blood , Fatty Liver/metabolism , Hypertriglyceridemia/metabolism , Receptors, Cytoplasmic and Nuclear/drug effects , Animals , DNA-Binding Proteins , Ligands , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Organ Specificity , Orphan Nuclear Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...