34 results on '"Kirchgessner TG"'
Search Results
2. A novel approach to measuring macrophage-specific reverse cholesterol transport in vivo in humans.
- Author
-
Cuchel M, Raper AC, Conlon DM, Pryma DA, Freifelder RH, Poria R, Cromley D, Li X, Dunbar RL, French B, Qu L, Farver W, Su CC, Lund-Katz S, Baer A, Ruotolo G, Akerblad P, Ryan CS, Xiao L, Kirchgessner TG, Millar JS, Billheimer JT, and Rader DJ
- Subjects
- 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, Atherosclerosis blood, Cholesterol blood, Cholesterol, HDL blood, Lipoproteins, HDL metabolism
- 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
3 H-cholesterol nanoparticles to selectively trace macrophage-specific RCT in vivo in humans. Use of3 H-cholesterol nanoparticles was initially tested in mice to assess the distribution of tracer and response to interventions known to increase RCT. Thirty healthy subjects received3 H-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 of3 H-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 of3 H-cholesterol nanoparticles as a feasible approach for the measurement of macrophage RCT in vivo in humans., (Copyright © 2017 by the American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
3. Beneficial and Adverse Effects of an LXR Agonist on Human Lipid and Lipoprotein Metabolism and Circulating Neutrophils.
- Author
-
Kirchgessner TG, Sleph P, Ostrowski J, Lupisella J, Ryan CS, Liu X, Fernando G, Grimm D, Shipkova P, Zhang R, Garcia R, Zhu J, He A, Malone H, Martin R, Behnia K, Wang Z, Barrett YC, Garmise RJ, Yuan L, Zhang J, Gandhi MD, Wastall P, Li T, Du S, Salvador L, Mohan R, Cantor GH, Kick E, Lee J, and Frost RJ
- Subjects
- 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, 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, Cell Movement drug effects, Imidazoles adverse effects, Imidazoles pharmacology, Lipid Metabolism, Lipoproteins metabolism, Liver X Receptors agonists, Neutrophils metabolism
- 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 LXRβ-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., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
4. Loss of Function of GALNT2 Lowers High-Density Lipoproteins in Humans, Nonhuman Primates, and Rodents.
- Author
-
Khetarpal SA, Schjoldager KT, Christoffersen C, Raghavan A, Edmondson AC, Reutter HM, Ahmed B, Ouazzani R, Peloso GM, Vitali C, Zhao W, Somasundara AV, Millar JS, Park Y, Fernando G, Livanov V, Choi S, Noé E, Patel P, Ho SP, Kirchgessner TG, Wandall HH, Hansen L, Bennett EP, Vakhrushev SY, Saleheen D, Kathiresan S, Brown CD, Abou Jamra R, LeGuern E, Clausen H, and Rader DJ
- Subjects
- 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, Lipoproteins, HDL metabolism, N-Acetylgalactosaminyltransferases deficiency
- 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., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
5. Genetic architecture of insulin resistance in the mouse.
- Author
-
Parks BW, Sallam T, Mehrabian M, Psychogios N, Hui ST, Norheim F, Castellani LW, Rau CD, Pan C, Phun J, Zhou Z, Yang WP, Neuhaus I, Gargalovic PS, Kirchgessner TG, Graham M, Lee R, Tontonoz P, Gerszten RE, Hevener AL, and Lusis AJ
- Subjects
- 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, Insulin Resistance genetics
- 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., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
6. Pharmacological characterization of a novel liver X receptor agonist with partial LXRα activity and a favorable window in nonhuman primates.
- Author
-
Kirchgessner TG, Martin R, Sleph P, Grimm D, Liu X, Lupisella J, Smalley J, Narayanan R, Xie Y, Ostrowski J, Cantor GH, Mohan R, and Kick E
- Subjects
- 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, Anticholesteremic Agents pharmacology, Imidazoles pharmacology, Liver drug effects, Orphan Nuclear Receptors agonists, Sulfones pharmacology
- Abstract
Liver X Receptors (LXRs) α and β 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 LXRβ 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 LXRβ 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., (Copyright © 2014 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
7. Genetic regulation of human adipose microRNA expression and its consequences for metabolic traits.
- Author
-
Civelek M, Hagopian R, Pan C, Che N, Yang WP, Kayne PS, Saleem NK, Cederberg H, Kuusisto J, Gargalovic PS, Kirchgessner TG, Laakso M, and Lusis AJ
- Subjects
- 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, Adipose Tissue metabolism, Gene Expression Regulation, MicroRNAs genetics, Quantitative Trait, Heritable
- 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 β, a key fatty acid oxidation enzyme that has been shown to play a role in regulating body fat and insulin resistance in adipose tissue.
- Published
- 2013
- Full Text
- View/download PDF
8. Genome-wide study of gene variants associated with differential cardiovascular event reduction by pravastatin therapy.
- Author
-
Shiffman D, Trompet S, Louie JZ, Rowland CM, Catanese JJ, Iakoubova OA, Kirchgessner TG, Westendorp RG, de Craen AJ, Slagboom PE, Buckley BM, Stott DJ, Sattar N, Devlin JJ, Packard CJ, Ford I, Sacks FM, and Jukema JW
- Subjects
- Aged, Coronary Disease drug therapy, Female, Genetic Predisposition to Disease genetics, Humans, Male, Middle Aged, Pravastatin therapeutic use, Coronary Disease genetics, Coronary Disease prevention & control, Genome-Wide Association Study, Polymorphism, Single Nucleotide, Pravastatin pharmacology
- 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.
- Published
- 2012
- Full Text
- View/download PDF
9. Genetic variants in the KIF6 region and coronary event reduction from statin therapy.
- Author
-
Li Y, Sabatine MS, Tong CH, Ford I, Kirchgessner TG, Packard CJ, Robertson M, Rowland CM, Bare LA, Shepherd J, Devlin JJ, and Iakoubova OA
- Subjects
- Chromosome Mapping, Coronary Disease prevention & control, Female, Humans, Linkage Disequilibrium, Male, Meta-Analysis as Topic, Middle Aged, Randomized Controlled Trials as Topic, Coronary Disease drug therapy, Coronary Disease genetics, Hydroxymethylglutaryl-CoA Reductase Inhibitors therapeutic use, Kinesins genetics, Polymorphism, Single Nucleotide
- 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.
- Published
- 2011
- Full Text
- View/download PDF
10. Effect of pravastatin therapy on coronary events in carriers of the KIF6 719Arg allele from the cholesterol and recurrent events trial.
- Author
-
Shiffman D, Sabatine MS, Louie JZ, Kirchgessner TG, Iakoubova OA, Campos H, Devlin JJ, and Sacks FM
- Subjects
- 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, Atherosclerosis genetics, Cholesterol blood, DNA genetics, Kinesins genetics, Myocardial Infarction drug therapy, Polymorphism, Genetic, Pravastatin therapeutic use
- 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., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
11. Systems genetics analysis of gene-by-environment interactions in human cells.
- Author
-
Romanoski CE, Lee S, Kim MJ, Ingram-Drake L, Plaisier CL, Yordanova R, Tilford C, Guan B, He A, Gargalovic PS, Kirchgessner TG, Berliner JA, and Lusis AJ
- Subjects
- Cell Line, Endothelial Cells drug effects, Endothelial Cells metabolism, Environment, Female, 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, Gene Expression Regulation drug effects
- 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., (Copyright 2010 The American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
12. Association between ADAMTS1 matrix metalloproteinase gene variation, coronary heart disease, and benefit of statin therapy.
- Author
-
Sabatine MS, Ploughman L, Simonsen KL, Iakoubova OA, Kirchgessner TG, Ranade K, Tsuchihashi Z, Zerba KE, Long DU, Tong CH, Packard CJ, Pfeffer MA, Devlin JJ, Shepherd J, Campos H, Sacks FM, and Braunwald E
- Subjects
- 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, Coronary Disease drug therapy, Coronary Disease genetics, Matrix Metalloproteinases genetics, Polymorphism, Genetic, Pravastatin therapeutic use
- 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.
- Published
- 2008
- Full Text
- View/download PDF
13. Association of the Trp719Arg polymorphism in kinesin-like protein 6 with myocardial infarction and coronary heart disease in 2 prospective trials: the CARE and WOSCOPS trials.
- Author
-
Iakoubova OA, Tong CH, Rowland CM, Kirchgessner TG, Young BA, Arellano AR, Shiffman D, Sabatine MS, Campos H, Packard CJ, Pfeffer MA, White TJ, Braunwald E, Shepherd J, Devlin JJ, and Sacks FM
- Subjects
- 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, Coronary Disease genetics, Kinesins genetics, Myocardial Infarction genetics, Polymorphism, Genetic
- 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.
- Published
- 2008
- Full Text
- View/download PDF
14. Polymorphism in KIF6 gene and benefit from statins after acute coronary syndromes: results from the PROVE IT-TIMI 22 study.
- Author
-
Iakoubova OA, Sabatine MS, Rowland CM, Tong CH, Catanese JJ, Ranade K, Simonsen KL, Kirchgessner TG, Cannon CP, Devlin JJ, and Braunwald E
- Subjects
- 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, Coronary Disease drug therapy, Coronary Disease genetics, Hydroxymethylglutaryl-CoA Reductase Inhibitors therapeutic use, Kinesins genetics, Polymorphism, Genetic
- 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.
- Published
- 2008
- Full Text
- View/download PDF
15. Asp92Asn polymorphism in the myeloid IgA Fc receptor is associated with myocardial infarction in two disparate populations: CARE and WOSCOPS.
- Author
-
Iakoubova OA, Tong CH, Chokkalingam AP, Rowland CM, Kirchgessner TG, Louie JZ, Ploughman LM, Sabatine MS, Campos H, Catanese JJ, Leong DU, Young BA, Lew D, Tsuchihashi Z, Luke MM, Packard CJ, Zerba KE, Shaw PM, Shepherd J, Devlin JJ, and Sacks FM
- Subjects
- 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, Antigens, CD genetics, Asparagine genetics, Aspartic Acid genetics, Myocardial Infarction genetics, Polymorphism, Single Nucleotide genetics, Receptors, Fc genetics
- 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.
- Published
- 2006
- Full Text
- View/download PDF
16. The unfolded protein response is an important regulator of inflammatory genes in endothelial cells.
- Author
-
Gargalovic PS, Gharavi NM, Clark MJ, Pagnon J, Yang WP, He A, Truong A, Baruch-Oren T, Berliner JA, Kirchgessner TG, and Lusis AJ
- Subjects
- 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, Endothelial Cells metabolism, Gene Expression Regulation, Inflammation genetics, Protein Folding
- 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.
- Published
- 2006
- Full Text
- View/download PDF
17. Identification of inflammatory gene modules based on variations of human endothelial cell responses to oxidized lipids.
- Author
-
Gargalovic PS, Imura M, Zhang B, Gharavi NM, Clark MJ, Pagnon J, Yang WP, He A, Truong A, Patel S, Nelson SF, Horvath S, Berliner JA, Kirchgessner TG, and Lusis AJ
- Subjects
- Activating Transcription Factor 4 genetics, Cells, Cultured, Gene Expression Profiling, Gene Expression Regulation, Humans, Interleukin-8 genetics, Oxidation-Reduction, Phospholipids chemistry, Protein Folding, Endothelial Cells drug effects, Endothelial Cells metabolism, Inflammation Mediators metabolism, Phospholipids pharmacology
- 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.
- Published
- 2006
- Full Text
- View/download PDF
18. Raising HDL cholesterol without inducing hepatic steatosis and hypertriglyceridemia by a selective LXR modulator.
- Author
-
Miao B, Zondlo S, Gibbs S, Cromley D, Hosagrahara VP, Kirchgessner TG, Billheimer J, and Mukherjee R
- Subjects
- Animals, DNA-Binding Proteins, Ligands, Liver X Receptors, Male, Mice, Mice, Inbred C57BL, Organ Specificity, Orphan Nuclear Receptors, Benzoates pharmacology, Benzylamines pharmacology, Cholesterol, HDL blood, Fatty Liver metabolism, Hypertriglyceridemia metabolism, Receptors, Cytoplasmic and Nuclear drug effects
- 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., (Copyright 2004 American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
19. A novel human hepatic organic anion transporting polypeptide (OATP2). Identification of a liver-specific human organic anion transporting polypeptide and identification of rat and human hydroxymethylglutaryl-CoA reductase inhibitor transporters.
- Author
-
Hsiang B, Zhu Y, Wang Z, Wu Y, Sasseville V, Yang WP, and Kirchgessner TG
- Subjects
- Amino Acid Sequence, Animals, Anion Transport Proteins, Base Sequence, Biological Transport, Carrier Proteins physiology, Dehydroepiandrosterone Sulfate pharmacokinetics, Humans, Molecular Sequence Data, Pravastatin pharmacokinetics, Rats, Taurocholic Acid pharmacokinetics, Carrier Proteins isolation & purification, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacokinetics, Liver metabolism
- Abstract
A novel human organic transporter, OATP2, has been identified that transports taurocholic acid, the adrenal androgen dehydroepiandrosterone sulfate, and thyroid hormone, as well as the hydroxymethylglutaryl-CoA reductase inhibitor, pravastatin. OATP2 is expressed exclusively in liver in contrast to all other known transporter subtypes that are found in both hepatic and nonhepatic tissues. OATP2 is considerably diverged from other family members, sharing only 42% sequence identity with the four other subtypes. Furthermore, unlike other subtypes, OATP2 did not transport digoxin or aldosterone. The rat isoform oatp1 was also shown to transport pravastatin, whereas other members of the OATP family, i.e. rat oatp2, human OATP, and the prostaglandin transporter, did not. Cis-inhibition studies indicate that both OATP2 and roatp1 also transport other statins including lovastatin, simvastatin, and atorvastatin. In summary, OATP2 is a novel organic anion transport protein that has overlapping but not identical substrate specificities with each of the other subtypes and, with its liver-specific expression, represents a functionally distinct OATP isoform. Furthermore, the identification of oatp1 and OATP2 as pravastatin transporters suggests that they are responsible for the hepatic uptake of this liver-specific hydroxymethylglutaryl-CoA reductase inhibitor in rat and man.
- Published
- 1999
- Full Text
- View/download PDF
20. Functional bioactive recombinant acylation stimulating protein is distinct from C3a anaphylatoxin.
- Author
-
Murray I, Parker RA, Kirchgessner TG, Tran J, Zhang ZJ, Westerlund J, and Cianflone K
- Subjects
- Animals, Arginine metabolism, Binding, Competitive, Biological Transport drug effects, Blood Proteins genetics, Blood Proteins pharmacology, Calcium pharmacokinetics, Cells, Cultured, Chromatography, High Pressure Liquid, Escherichia coli genetics, Glucose metabolism, Humans, Peptide Fragments pharmacology, Precipitin Tests, Receptors, Cell Surface metabolism, Recombinant Proteins genetics, Recombinant Proteins metabolism, Solubility, Triglycerides biosynthesis, Adipocytes metabolism, Blood Proteins metabolism, Complement C3a metabolism
- Abstract
Acylation stimulating protein (ASP) acts upon adipose tissue to stimulate triglyceride synthesis and glucose transport. The aim of the present study was to produce recombinant ASP and to measure its bioactivity. The cDNA region of the parent complement C3 sequence coding for ASP (C3adesArg) was cloned and expressed in E. coli. Bioactivity of the purified recombinant material was tested by determining its effect on triglyceride synthesis, glucose transport, and competition binding assays. In standard assays, concentrations of 5.5 microM recombinant ASP (rASP) stimulated triglyceride synthesis comparably to plasma ASP (pASP): 228% versus 237%, respectively, in 3T3 preadipocytes and 568% versus 440% in human differentiated adipocytes. rASP also increased glucose transport in L6 myocytes (163% at 10 microm rASP) and in human differentiated adipocytes (334% rASP vs. 329% pASP at 5 microM). rASP competitively displaced radiolabeled plasma ASP from high affinity association with the cell surface in both human differentiated adipocytes and 3T3 preadipocyte fibroblasts. Furthermore, immunoprecipitation of rASP and pASP with a specific monoclonal antibody abolished stimulation of cellular triglyceride synthesis. Lastly, we contrasted the structure:function activities of the arginated (C3a) and desarginated (ASP) proteins. The lipogenic activity and the anaphylatoxic activity result from distinct structural domains of the polypeptides. Thus rASP retains full biologic ASP activity and may provide a tool to study structure-function relationships in this physiologic system.
- Published
- 1997
21. Tumor necrosis factor-alpha contributes to obesity-related hyperleptinemia by regulating leptin release from adipocytes.
- Author
-
Kirchgessner TG, Uysal KT, Wiesbrock SM, Marino MW, and Hotamisligil GS
- Subjects
- Adipocytes drug effects, Animals, Cell Line, Cells, Cultured, Leptin, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mice, Obese, Protein Biosynthesis, Proteins genetics, RNA, Messenger metabolism, Recombinant Proteins pharmacology, Tumor Necrosis Factor-alpha deficiency, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha pharmacology, Adipocytes metabolism, Adipose Tissue metabolism, Obesity metabolism, Proteins metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
Cytokines, in particular tumor necrosis factor-alpha (TNF-alpha), have significant effects on energy metabolism and appetite although their mechanisms of action are largely unknown. Here, we examined whether TNF-alpha modulates the production of leptin, the recently identified fat-specific energy balance hormone, in cultured adipocytes and in mice. TNF-alpha treatment of 3T3-L1 adipocytes resulted in rapid stimulation of leptin accumulation in the media, with a maximum effect at 6 h. This stimulation was insensitive to cycloheximide, a protein synthesis inhibitor, but was completely inhibited by the secretion inhibitor brefeldin A, indicating a posttranslational effect. Treatment of mice with TNF-alpha also caused a similar increase in plasma leptin levels. Finally, in obese TNF-alpha-deficient mice, circulating leptin levels were significantly lower, whereas adipose tissue leptin was higher compared with obese wild-type animals. These data provide evidence that TNF-alpha can act directly on adipocytes to regulate the release of a preformed pool of leptin. Furthermore, they suggest that the elevated adipose tissue expression of TNF-alpha that occurs in obesity may contribute to obesity-related hyperleptinemia.
- Published
- 1997
- Full Text
- View/download PDF
22. Regulation of expression of the lipoprotein lipase gene in brown adipose tissue.
- Author
-
Mitchell JR, Jacobsson A, Kirchgessner TG, Schotz MC, Cannon B, and Nedergaard J
- Subjects
- Animals, Cold Temperature, Drug Stability, Half-Life, Insulin pharmacology, Male, Norepinephrine pharmacology, RNA, Messenger chemistry, Rats, Rats, Sprague-Dawley, Sympathetic Nervous System physiology, Adipose Tissue, Brown physiology, Gene Expression Regulation drug effects, Lipoprotein Lipase genetics
- Abstract
The regulation of lipoprotein lipase gene expression in brown adipose tissue was studied. Rats were preacclimated to 21 degrees C. Exposure to cold (4 degrees C) resulted in a rapid increase in the level of lipoprotein lipase mRNA in the tissue. The level peaked (expressed per microgram total RNA) after approximately 8 h and then slowly declined. The increased lipoprotein lipase mRNA level was not due to an increased stability of the mRNA, but, in a transition event from a high to a low expression of the lipoprotein lipase gene, a transcription-dependent process was recruited that accelerated the breakdown of lipoprotein lipase mRNA. Norepinephrine injections increased lipoprotein lipase mRNA levels in the tissue; this effect was mediated via a beta-adrenergic receptor. The effect of cold could be mimicked by norepinephrine injections, and these two effects were not additive, indicating that the cold effect was mediated by norepinephrine. The lipoprotein lipase mRNA level was also increased by insulin injections (into fasted animals); thus an increase in lipoprotein lipase gene expression in brown adipose tissue may be induced via two different stimuli, which, intracellularly, would be mediated via different signaling systems. In all investigated conditions, the changes in lipoprotein lipase mRNA levels observed here were parallelled by alterations in lipoprotein lipase activity reported earlier from this laboratory. It was therefore concluded that, under the conditions studied, lipoprotein lipase activity in brown adipose tissue was primarily regulated at the transcriptional level.
- Published
- 1992
- Full Text
- View/download PDF
23. Mechanisms of increased lipoprotein lipase in fat cells of obese Zucker rats.
- Author
-
Fried SK, Turkenkopf IJ, Goldberg IJ, Doolittle MH, Kirchgessner TG, Schotz MC, Johnson PR, and Greenwood MR
- Subjects
- Adipose Tissue pathology, Animals, Insulin pharmacology, Lipoprotein Lipase genetics, Obesity pathology, Precipitin Tests, RNA, Messenger metabolism, Rats, Rats, Zucker, Reference Values, Adipose Tissue enzymology, Lipoprotein Lipase metabolism, Obesity enzymology
- Abstract
The mechanisms underlying the increased activity of lipoprotein lipase (LPL) in adipocytes of genetically obese Zucker rats was studied. Relative rates of LPL synthesis (percent of total protein synthesis) determined by biosynthetic labeling and specific immunoprecipitation were similar in isolated fat cells from lean and obese rats, in the absence or presence of insulin. Insulin stimulated LPL synthesis as a result of a general increase in protein synthesis, and this effect was more marked in the obese fat cells. Levels of LPL mRNA, as a percent of total RNA, were also similar in fat cells from lean and obese rats. In contrast, when the data are calculated on a per fat cell basis, rates of LPL synthesis per fat cell are ninefold higher in obese compared with lean cells, accounting for the increase in LPL activity per fat cell. Fat cells from lean and obese rats showed similar rates of binding and degradation of purified bovine milk 125I-labeled LPL per unit fat cell surface area. Thus, on a per cell basis, rates of LPL turnover are increased in enlarged Zucker rat adipocytes, but there is no specific abnormality in the cellular regulation of LPL. Increases in LPL activity in obese rat adipocytes are related to an overall hyperresponsiveness to insulin effects on protein synthesis.
- Published
- 1991
- Full Text
- View/download PDF
24. The response of lipoprotein lipase to feeding and fasting. Evidence for posttranslational regulation.
- Author
-
Doolittle MH, Ben-Zeev O, Elovson J, Martin D, and Kirchgessner TG
- Subjects
- Adipose Tissue ultrastructure, Amino Acid Sequence, Animals, Endoplasmic Reticulum enzymology, Glycosylation, Golgi Apparatus enzymology, Hexosaminidases metabolism, Kinetics, Lipoprotein Lipase genetics, Male, Mannose analysis, Molecular Sequence Data, Myocardium enzymology, RNA, Messenger metabolism, Rats, Rats, Inbred Strains, Adipose Tissue enzymology, Fasting, Food, Lipoprotein Lipase metabolism, Protein Processing, Post-Translational
- Abstract
The regulation of adipose tissue lipoprotein lipase (LPL) was examined in rats fed or fasted overnight, and was found to be controlled posttranslationally. LPL catalytic activity decreased by 50% after fasting while LPL mRNA levels and rates of synthesis increased nearly 2-fold; enzyme mass remained unchanged. The distribution of LPL within the endoplasmic reticulum (ER) and Golgi/post-Golgi secretory pathway was assessed by differentiating between LPL high mannose and complex forms. After fasting, the majority of LPL is in the high mannose ER form (65%, 0.97 micrograms/g wet weight tissue), whereas the LPL complex form comprises only 35% (or 0.52 micrograms/g). After refeeding, however, the Golgi-derived LPL complex form predominates (65%, 1.03 micrograms/g) over the high mannose ER form (35%, 0.55 micrograms/g). Kinetic analysis suggests that high mannose LPL disappears with a half-life of t0.5 = 40 min in both fed and fasted rats, indicating that the redistribution of LPL mass during feeding/fasting does not arise by differential retention within ER. Instead, the fractional catabolic rate of complex LPL within the Golgi/post-Golgi secretory compartment can be calculated to be 3.5-fold greater in fasting. In heart, changes in LPL activity in response to feeding/fasting are also not due to differences in mRNA levels or rates of synthesis. Based on these findings, a model of LPL posttranslational regulation is proposed and discussed.
- Published
- 1990
25. Hormone-sensitive lipase: sequence, expression, and chromosomal localization to 19 cent-q13.3.
- Author
-
Holm C, Kirchgessner TG, Svenson KL, Fredrikson G, Nilsson S, Miller CG, Shively JE, Heinzmann C, Sparkes RS, and Mohandas T
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Chromosome Mapping, Cloning, Molecular, DNA genetics, Gene Expression Regulation, Humans, Molecular Sequence Data, Rats, Chromosomes, Human, Pair 19, Sterol Esterase genetics
- Abstract
Hormone-sensitive lipase, a key enzyme in fatty acid mobilization, overall energy homeostasis, and possibly steroidogenesis, is acutely controlled through reversible phosphorylation by catecholamines and insulin. The 757-amino acid sequence predicted from a cloned rat adipocyte complementary DNA showed no homology with any other known lipase or protein. The activity-controlling phosphorylation site was localized to Ser563 in a markedly hydrophilic domain, and a lipid-binding consensus site was tentatively identified. One or several messenger RNA species (3.3, 3.5, or 3.9 kilobases) were expressed in adipose and steroidogenic tissues and heart and skeletal muscle. The human hormone-sensitive lipase gene mapped to chromosome 19 cent-q13.3.
- Published
- 1988
- Full Text
- View/download PDF
26. Human lipoprotein lipase complementary DNA sequence.
- Author
-
Wion KL, Kirchgessner TG, Lusis AJ, Schotz MC, and Lawn RM
- Subjects
- Amino Acid Sequence, Base Sequence, Fatty Acids, Nonesterified metabolism, Humans, Lipase analysis, Lipase genetics, Lipoprotein Lipase analysis, Liver enzymology, Nucleic Acid Hybridization, DNA analysis, Lipoprotein Lipase genetics
- Abstract
Lipoprotein lipase is a key enzyme of lipid metabolism that acts to hydrolyze triglycerides, providing free fatty acids for cells and affecting the maturation of circulating lipoproteins. It has been proposed that the enzyme plays a role in the development of obesity and atherosclerosis. The human enzyme has been difficult to purify and its protein sequence was heretofore undetermined. A complementary DNA for human lipoprotein lipase that codes for a mature protein of 448 amino acids has now been cloned and sequenced. Analysis of the sequence indicates that human lipoprotein lipase, hepatic lipase, and pancreatic lipase are members of a gene family. Two distinct species of lipoprotein lipase messenger RNA that arise from alternative sites of 3'-terminal polyadenylation were detected in several different tissues.
- Published
- 1987
- Full Text
- View/download PDF
27. Genetic and developmental regulation of the lipoprotein lipase gene: loci both distal and proximal to the lipoprotein lipase structural gene control enzyme expression.
- Author
-
Kirchgessner TG, LeBoeuf RC, Langner CA, Zollman S, Chang CH, Taylor BA, Schotz MC, Gordon JI, and Lusis AJ
- Subjects
- Adipose Tissue metabolism, Animals, Gene Expression Regulation, Mice, Mice, Inbred Strains, Myocardium metabolism, Polymorphism, Restriction Fragment Length, RNA, Messenger metabolism, Genes, Lipoprotein Lipase genetics
- Abstract
We report here a study of the developmental and genetic control of tissue-specific expression of lipoprotein lipase, the enzyme responsible for hydrolysis of triglycerides in chylomicrons and very low density lipoproteins. Lipoprotein lipase (LPL) mRNA is present in a wide variety of adult rat and mouse tissues examined, albeit at very different levels. A remarkable increase in the levels of LPL mRNA occurs in heart over a period of several weeks following birth, closely paralleling developmental changes in lipase activity and myocardial beta-oxidation capacity. Large increases in LPL mRNA also occur during differentiation of 3T3L1 cells to adipocytes. As previously reported, at least two separate genetic loci control the tissue-specific expression of LPL activity in mice. One of the loci, controlling LPL activity in heart, is associated with an alteration in LPL mRNA size, while the other, controlling LPL activity in adipose tissue, appears to affect the translation or post-translational expression of LPL. To examine whether these genetic variations are due to mutations of the LPL structural locus, we mapped the LPL gene to a region of mouse chromosome 8 using restriction fragment-length polymorphisms and analysis of hamster-mouse somatic cell hybrids. This region is homologous to the region of human chromosome 8 which contains the human LPL gene as judged by the conservation of linked genetic markers. Genetic variations affecting LPL expression in heart cosegregated with the LPL gene, while variations affecting LPL expression in adipose tissue did not. Furthermore, Southern blotting analysis indicates that LPL is encoded by a single gene and, thus, the genetic differences are not a consequence of independent regulation of two separate genes in the two tissues. These results suggest the existence of cis-acting elements for LPL gene expression that operate in heart but not adipose tissue. Our results also indicate that two genetic mutations resulting in deficiencies of LPL in mice, the W mutation on chromosome 5 and the cld mutation on mouse chromosome 17, do not involve the LPL structural gene locus. Finally, we show that the gene for hepatic lipase, a member of a gene family with LPL, is unlinked to the gene for LPL. This indicates that combined deficiencies of LPL and hepatic lipase, observed in humans as well as in certain mutant strains of mice, do not result from focal disruptions of a cluster of lipase genes.
- Published
- 1989
28. The sequence of cDNA encoding lipoprotein lipase. A member of a lipase gene family.
- Author
-
Kirchgessner TG, Svenson KL, Lusis AJ, and Schotz MC
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Lipase genetics, Liver enzymology, Macrophages enzymology, Mice, Pancreas enzymology, Cloning, Molecular, DNA analysis, Genes, Lipoprotein Lipase genetics
- Abstract
cDNA clones corresponding to the entire coding region of mature lipoprotein lipase were identified by antibody screening of a mouse macrophage library and sequenced. The predicted amino acid sequence indicates that the mature protein contains 447 amino acids with a molecular weight of 50,314. Comparison of the nucleotide and amino acid sequence with those of rat hepatic lipase and porcine pancreatic lipase reveals extensive homology among the enzymes, indicating that they are members of a gene family of lipases. Most striking is a conservation of five disulfide bridges in all three enzymes, strongly suggesting that the enzymes have similar overall folding patterns. Lipoprotein lipase is also shown to be extraordinarily conserved among mouse, human, and bovine species. The mRNA for lipoprotein lipase is abundant in heart and adipose tissue but is also present in a wide variety of other tissues. There are two major species of mRNA in mouse and human tissues examined, 3.6 and 3.4 kilobases (kb) in size. Rat tissues, on the other hand, contain only the 3.6-kb species while bovine tissues contain an additional 1.7-kb species.
- Published
- 1987
29. Insulin increases the synthetic rate and messenger RNA level of lipoprotein lipase in isolated rat adipocytes.
- Author
-
Ong JM, Kirchgessner TG, Schotz MC, and Kern PA
- Subjects
- Animals, Cells, Cultured, Heparin pharmacology, Lipoprotein Lipase metabolism, Rats, Adipose Tissue enzymology, Insulin pharmacology, Lipoprotein Lipase genetics, RNA, Messenger metabolism
- Abstract
Lipoprotein lipase (LPL) is the enzyme responsible for hydrolysis of circulating triglyceride-rich lipoproteins and is important for storage of adipocyte lipid. To study the regulation of LPL synthetic rate in adipose tissue, primary cultures of isolated rat adipocytes were pulse-labeled with [35S]methionine, and LPL was immunoprecipitated with an LPL-specific antibody. A pulse-chase experiment identified the cellular and secreted forms of LPL as a 55-57-kDa protein. In the presence of heparin, there was a large increase in secretion of newly synthesized LPL from the cells, although heparin did not stimulate cellular LPL synthetic rate. When cells were exposed to insulin for 2 h, pulse-labeling revealed that insulin stimulated a maximal dose-related increase in LPL synthetic rate of 300% of control. This increase in LPL synthetic rate was observed after an exposure to insulin for as little as 60 min and was accompanied by only a 10-25% increase in total protein synthesis. In addition, insulin had no effect on the turnover of intracellular LPL. Using a cDNA probe for LPL, insulin induced a 2-fold increase in the LPL mRNA. Thus, insulin stimulated an increase in specific LPL mRNA in isolated rat adipocytes. This increase in LPL mRNA then leads to an increase in the synthetic rate of the LPL protein.
- Published
- 1988
30. Human genes involved in lipolysis of plasma lipoproteins: mapping of loci for lipoprotein lipase to 8p22 and hepatic lipase to 15q21.
- Author
-
Sparkes RS, Zollman S, Klisak I, Kirchgessner TG, Komaromy MC, Mohandas T, Schotz MC, and Lusis AJ
- Subjects
- Animals, Humans, Hybrid Cells, Lipoproteins blood, Lipoproteins genetics, Liver enzymology, Mice, Chromosome Mapping, Chromosomes, Human, Pair 15, Chromosomes, Human, Pair 8, Lipase genetics, Lipoprotein Lipase genetics
- Abstract
We have used cDNA probes for lipoprotein lipase and hepatic lipase to determine the chromosomal and subchromosomal locations of the human genes for these lipolytic enzymes. Southern blot analysis of genomic DNA from 17 independent mouse-human somatic cell hybrids demonstrated the presence of the gene for human lipoprotein lipase on chromosome 8, whereas the gene for hepatic lipase was on chromosome 15. Regional mapping of the genes by in situ hybridization to human chromosomes indicated that the lipoprotein lipase gene (LPL) resides in the p22 region of chromosome 8, while hepatic lipase gene (HL) resides in the q21 region of chromosome 15. We previously reported, on the basis of nucleotide and amino acid homologies, that these genes are members of a gene family of lipases, and, thus, the present findings indicate that the members of this family are dispersed. The results are also of significance with respect to disorders involving deficiencies of the enzymes. In particular, they suggest that certain rare combined deficiencies of both enzymes do not involve mutations of the structural loci.
- Published
- 1987
- Full Text
- View/download PDF
31. Regulation of chicken apolipoprotein B: cloning, tissue distribution, and estrogen induction of mRNA.
- Author
-
Kirchgessner TG, Heinzmann C, Svenson KL, Gordon DA, Nicosia M, Lebherz HG, Lusis AJ, and Williams DL
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Chickens, DNA genetics, DNA isolation & purification, Female, Liver metabolism, Male, Molecular Sequence Data, RNA, Messenger drug effects, Apolipoproteins B genetics, Cloning, Molecular, Estradiol pharmacology, Genes, Genes, Regulator, RNA, Messenger genetics, Transcription, Genetic drug effects
- Abstract
Apolipoprotein (apo) B is a major protein component of plasma very low-density and low-density lipoproteins (VLDL and LDL, respectively) and serves as a recognition signal for the cellular binding and internalization of LDL by the apoB/E receptor. In contrast to the situation in mammals, avian apoB is also a component of specialized VLDL particles that are produced by the liver in response to estrogen. These particles transport cholesterol and triglyceride from the liver to the ovary for deposition in egg yolk. We report here the identification and characterization of cDNA clones for chicken apoB and their use in examining the tissue distribution and hormonal regulation of chicken apoB mRNA. The cDNA clones were identified by immunological screening of a phage lambda gt11 library constructed with hen liver mRNA and their identity was supported by sequence comparisons with mammalian apoB. The chicken apoB mRNA is approximately the same size as mammalian apoB mRNA (14 kb), and, as occurs in mammals, is present at high levels in liver and small intestine. Unlike mammals, the chicken apoB mRNA is also found at high levels in the kidney, consistent with previous protein biosynthetic studies. A DNA-excess solution-hybridization assay was used to quantitate apoB mRNA in these tissues and to examine its hormonal regulation. In control roosters the liver and kidney contained 65% and 10%, respectively, as much apoB mRNA as the small intestine. Within 24 h after estradiol administration, apoB mRNA was increased five- to seven-fold in liver but was unchanged in intestine and kidney. The increase in apoB mRNA content and the kinetics of induction parallel hepatic apoB synthesis, indicating that estrogen regulates apoB production through changes in the cellular abundance of apoB mRNA. The apoB mRNA increased rapidly following hormone treatment while the mRNA for another VLDL protein (apoII) showed a lag or slow phase of several hours before significant mRNA accumulation occurred. These data indicate that the liver can respond immediately to estrogen to increase apoB mRNA accumulation, while apoII mRNA accumulation appears to involve additional events or signals which occur slowly and are specific to this gene.
- Published
- 1987
- Full Text
- View/download PDF
32. Nucleotide sequence of rat adipose hormone sensitive lipase cDNA.
- Author
-
Holm C, Kirchgessner TG, Svenson KL, Lusis AJ, Belfrage P, and Schotz MC
- Subjects
- Adipose Tissue metabolism, Animals, Base Sequence, Molecular Sequence Data, Rats, Sterol Esterase physiology, Adipose Tissue enzymology, DNA isolation & purification, Lipolysis, Sterol Esterase genetics
- Published
- 1988
- Full Text
- View/download PDF
33. Organization of the human lipoprotein lipase gene and evolution of the lipase gene family.
- Author
-
Kirchgessner TG, Chuat JC, Heinzmann C, Etienne J, Guilhot S, Svenson K, Ameis D, Pilon C, d'Auriol L, and Andalibi A
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Exons, Gene Library, Humans, Molecular Sequence Data, Restriction Mapping, Sequence Homology, Nucleic Acid, Transcription, Genetic, Biological Evolution, Genes, Lipase genetics, Lipoprotein Lipase genetics, Multigene Family
- Abstract
The human lipoprotein lipase gene was cloned and characterized. It is composed of 10 exons spanning approximately equal to 30 kilobases. The first exon encodes the 5'-untranslated region, the signal peptide plus the first two amino acids of the mature protein. The next eight exons encode the remaining 446 amino acids, and the tenth exon encodes the long 3'-untranslated region of 1948 nucleotides. The lipoprotein lipase transcription start site and the sequence of the 5'-flanking region were also determined. We compared the organization of genes for lipoprotein lipase, hepatic lipase, pancreatic lipase, and Drosophila yolk protein 1, which are members of a family of related genes. A model for the evolution of the lipase gene family is presented that involves multiple rounds of gene duplication plus exon-shuffling and intron-loss events.
- Published
- 1989
- Full Text
- View/download PDF
34. Localization of lipoprotein lipase mRNA in selected rat tissues.
- Author
-
Goldberg IJ, Soprano DR, Wyatt ML, Vanni TM, Kirchgessner TG, and Schotz MC
- Subjects
- Adrenal Medulla enzymology, Animals, Blotting, Northern, Cerebral Cortex enzymology, Female, Hippocampus enzymology, Kidney Medulla enzymology, Macaca fascicularis, Neurons enzymology, Nucleic Acid Hybridization, Organ Specificity, Pituitary Gland enzymology, RNA Probes, Rats, Rats, Inbred Strains, Lipoprotein Lipase metabolism, RNA, Messenger analysis
- Abstract
Measurements of enzymatic activity have demonstrated that lipoprotein lipase (LPL), the principal enzyme responsible for hydrolysis of circulating triglyceride, is present in a number of tissues including brain, kidney, and adrenal gland. To determine the sites of synthesis of LPL in these tissues, in situ hybridization studies were performed using a non-sense 35S-labeled RNA probe produced from a 624-bp mouse LPL cDNA fragment. Control studies were performed with a sense RNA strand. Using 5-10-micron sections of 5-day-old rat brain, strong hybridization was found in pyramidal neurons of the hippocampus. Positive hybridization, indicating the presence of LPL mRNA, was also found in brain cortex and in the intermediate lobe of adult rat pituitary gland. Specific areas of adrenal and kidney medulla showed hybridization with the probe. LPL mRNA is, therefore, present in a number of specific regions of the body. LPL in these areas may not be important in regulating circulating levels of lipoproteins, but may be essential for cellular uptake, binding, and transfer of free fatty acids or other lipophilic substances.
- Published
- 1989
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.