Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Sci Rep ; 8(1): 13733, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30214074

ABSTRACT

Vitamin K plays a crucial role in the regulation of vascular calcifications by allowing activation of matrix Gla protein. The dietary requirement for vitamin K is low because of an efficient recycling of vitamin K by vitamin K epoxide reductase (VKORC1). However, decreased VKORC1 activity may result in vascular calcification. More than 30 coding mutations of VKORC1 have been described. While these mutations have been suspected of causing anticoagulant resistance, their association with an increase in the risk of vascular calcification has never been considered. We thus investigated functional cardiovascular characteristics in a rat model mutated in VKORC1. This study revealed that limited intake in vitamin K in mutated rat induced massive calcified areas in the media of arteries of lung, aortic arch, kidneys and testis. Development of calcifications could be inhibited by vitamin K supplementation. In calcified areas, inactive Matrix Gla protein expression increased, while corresponding mRNA expression was not modified. Mutation in VKORC1 associated with a limited vitamin K intake is thus a major risk for cardiovascular disease. Our model is the first non-invasive rat model that shows spontaneous medial calcifications and would be useful for studying physiological function of vitamin K.


Subject(s)
Arteriosclerosis/genetics , Monckeberg Medial Calcific Sclerosis/genetics , Vascular Calcification/genetics , Vitamin K Epoxide Reductases/genetics , Vitamin K/metabolism , Animals , Anticoagulants/administration & dosage , Aorta, Thoracic/pathology , Arteries/pathology , Arteriosclerosis/pathology , Disease Models, Animal , Humans , Kidney/blood supply , Kidney/pathology , Lung/blood supply , Lung/pathology , Male , Monckeberg Medial Calcific Sclerosis/pathology , Mutation , Mutation, Missense/genetics , Polymorphism, Single Nucleotide/genetics , Rats , Testis/blood supply , Testis/pathology , Vascular Calcification/pathology , Vitamin K/genetics , Vitamin K Epoxide Reductases/metabolism
2.
J Med Food ; 17(3): 310-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24433070

ABSTRACT

This study was performed to investigate the hypolipidemic, antiobese, and antiatherogenic effects of resveratrol in apoE-deficient mice fed an atherogenic diet (20% fat and 1% cholesterol). These animals were fed an atherogenic diet containing 0.02% lovastatin (w/w) or 0.02% resveratrol (w/w) for 12 weeks. Resveratrol and lovastatin supplementation significantly reduced either the body weight or epididymal fat weight without altering the food intake and food efficiency ratio. Resveratrol significantly decreased the plasma total cholesterol (total-C), low-density lipoprotein cholesterol (LDL-C), non-high-density lipoprotein cholesterol (non-HDL-C) concentrations, apoB/apoA-I ratio, hepatic cholesterol, and triglyceride (TG) contents, whereas significantly it increased the plasma HDL-C concentration compared with the control and lovastatin groups. Plasma and hepatic TG and plasma apoB levels were significantly lower in both the lovastatin and resveratrol groups than in the control group without altering the plasma apoA-I concentration. Both resveratrol and lovastatin significantly decreased hepatic fatty acid and TG synthesis, whereas they increased fatty acid oxidation (ß-oxidation) except for the carnitine palmitoyltransferase activity compared with the control group. However, there was no difference in hepatic 3-hydroxyl-3-methylglutaryl-CoA reductase activity among the groups, although hepatic acyl-CoA: cholesterol acyltransferase activity was significantly lower in the lovastatin groups than in the control group. In epididymal adipose tissue, resveratrol supplementation led to an increase in ß-oxidation and decrease in TG synthesis, compared with the control group. Tissue morphology revealed that there were dramatic decreases in hepatic lipid droplets and aortic fatty streaks by resveratrol and lovastatin supplementation. This study demonstrates that resveratrol exerts not only antiobesity and hypolipidemic effects, but also protective effects for the liver and aorta through the modulation of lipid metabolism in both the liver and white adipose tissues.


Subject(s)
Anti-Obesity Agents/administration & dosage , Apolipoproteins E/genetics , Arteriosclerosis/prevention & control , Obesity/drug therapy , Protective Agents/administration & dosage , Stilbenes/administration & dosage , Animals , Anticholesteremic Agents/administration & dosage , Aorta/drug effects , Apolipoproteins E/deficiency , Arteriosclerosis/drug therapy , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Diet, Atherogenic/adverse effects , Humans , Lovastatin/administration & dosage , Male , Mice , Mice, Knockout , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Resveratrol
3.
J Oleo Sci ; 62(12): 1049-58, 2013.
Article in English | MEDLINE | ID: mdl-24292357

ABSTRACT

To investigate the effects of dietary Grifola frondosa on cholesterol, normal mice were fed a diet containing 1% cholesterol (HC group) or 1% cholesterol and 10% freeze-dried G. frondosa powder (HC+G group) for 4 weeks and hepatic and plasma lipid levels were compared with those of a cholesterol-free diet-fed mice (N group). Hepatic total cholesterol (TC), triacylglycerol contents were considerably increased and plasma TC / phospholipid (PL) was also increased significantly in the HC group compared with the N group. However, plasma TC content decreased in the HC+G group compared with the HC group. To characterize the mechanisms responsible for lowered plasma cholesterol in G. frondosa-supplemented mice, hepatic gene expression was profiled using DNA microarray and gene ontology. Genome analyses revealed that de novo cholesterol synthesis genes were suppressed following cholesterol intake. However, expression of bile acid biosynthesis and low-density lipoprotein receptor genes showed little change. Scarb1, Abcg5, and Abcg8, involved in cholesterol transport and excretion, were slightly upregulated in the HC+G group compared with the HC group. These data indicate the plasma cholesterol-lowering effect of G. frondosa. Moreover, fatty acid (FA) ß-oxidation was promoted via adipocytokine signaling pathways, and Saa, encodes serum amyloid A related to arteriosclerosis, was suppressed in the HC+G group.


Subject(s)
Cholesterol, Dietary/administration & dosage , Cholesterol/metabolism , Dietary Supplements , Gene Expression Regulation , Grifola , Liver/metabolism , Triglycerides/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/metabolism , Adipokines/physiology , Animals , Arteriosclerosis/genetics , Arteriosclerosis/prevention & control , Cholesterol/blood , Fatty Acids/metabolism , Lipid Peroxidation , Lipoproteins/metabolism , Male , Mice, Inbred ICR , Oligonucleotide Array Sequence Analysis , Phospholipids , Scavenger Receptors, Class B/metabolism , Signal Transduction/physiology , Up-Regulation
4.
Biomed Res ; 33(1): 9-13, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22361881

ABSTRACT

Many biological activities of green tea have been attributed to a major constituent, (minus;)-epigallocatechin gallate (EGCG). We previously reported that EGCG and an EGCG-free fraction derived from green tea modulated the gene expression of gluconeogenic enzymes, glucose-6-phosphatase and phosphoenolpyruvate carboxykinase, in the mouse liver. EGCG is also known to affect the gene expression of enzymes related to lipid metabolism. However, it remains to be examined whether or not a constituent other than EGCG contributes to the change in gene expression of these enzymes. In this study, we prepared an EGCG-free water-soluble fraction (GT-W), and examined its effects on the hepatic gene expression of lipogenic enzymes in mice. The results of quantitative real-time PCR assays indicated that the dietary administration of GT-W for 4 weeks reduced the hepatic gene expression of lipogenic enzymes: fatty acid synthase, hydroxymethylglutaryl coenzyme A reductase, and acetyl-coenzyme A carboxylase alpha. Also, the gene expression of sterol regulatory element-binding transcription factor (Srebf)1 and/or Srebf2 was reduced, suggesting that the reduction of Srebfs contributed to the down-regulation of the lipogenic enzymes, since these transcription factors bind the promoter region to enhance their expression. The plasma levels of triglycerides and cholesterol were reduced with statistical significance in the group given a diet containing GT-W. These results suggest that in addition to EGCG, green tea contains some component(s) which may help to prevent arteriosclerosis and obesity.


Subject(s)
Catechin/analogs & derivatives , Lipid Metabolism/genetics , Liver/drug effects , Plant Extracts/pharmacology , Tea/chemistry , Animals , Arteriosclerosis/genetics , Arteriosclerosis/prevention & control , Blood Glucose/analysis , Catechin/metabolism , Catechin/pharmacology , Cholesterol/blood , Down-Regulation/drug effects , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Liver/enzymology , Male , Mice , Mice, Inbred BALB C , Obesity/genetics , Obesity/prevention & control , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Plant Leaves/chemistry , Real-Time Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Triglycerides/blood
5.
J Hum Genet ; 50(5): 241-248, 2005.
Article in English | MEDLINE | ID: mdl-15895286

ABSTRACT

This study aimed at assessing the effect of folic acid supplementation quantitatively in each MTHFR C677T genotype and considered the efficiency of tailor-made prevention of atherosclerosis. Study design was genotype-stratified, randomized, double-blind, placebo-controlled trials. The setting was a Japanese company in the chemical industry. Subjects were 203 healthy men after exclusion of those who took folic acid or drugs known to effect folic acid metabolism. Intervention was folic acid 1 mg/day p.o. for 3 months. The primary endpoint was plasma total homocysteine level (tHcy). In all three genotypes, there were significant tHcy decreases. The greatest decrease was in the TT homozygote [6.61 (3.47-9.76) micromol/l] compared with other genotypes [CC: 2.59 (1.81-3.36), CT: 2.64 (2.16-3.13)], and there was a significant trend between the mutated allele number and the decrease. The tHcy were significantly lowered in all the genotypes, but the amount of the decrease differed significantly in each genotype, which was observed at both 1 and 3 months. Using these time-series data, the largest benefit obtained by the TT homozygote was appraised as 2.4 times compared with the CC homozygote. Taking into account the high allele frequency of this SNP, this quantitative assessment should be useful when considering tailor-made prevention of atherosclerosis with folic acid.


Subject(s)
Arteriosclerosis/genetics , Arteriosclerosis/prevention & control , Dietary Supplements , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mutation/genetics , Alleles , Arteriosclerosis/enzymology , Double-Blind Method , Folic Acid/blood , Folic Acid Deficiency , Genotype , Homocysteine/blood , Homozygote , Humans , Male , Middle Aged
6.
J Nutr ; 135(4): 722-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15795424

ABSTRACT

The beneficial health effects of red wine have been attributed to the antioxidant activity of its polyphenols. The present study investigated the effects of a standardized freeze-dried powder made from fresh grapes, rich in grape-specific polyphenols and free of alcohol, on oxidative stress, atherogenicity of macrophages, and the development of atherosclerotic lesions in apolipoprotein E deficient (E(0)) mice. Thirty E(0) mice were assigned to 3 groups. Mice consumed water alone (control), 150 mug total polyphenols/d in the form of grape powder (grape powder), or the equivalent amount of glucose and fructose (placebo) in drinking water for 10 wk. Consumption of grape powder reduced the atherosclerotic lesion area by 41% (P < 0.0002) compared to the control or placebo mice. The antiatherosclerotic effect was at least partly due to a significant 8% reduction in serum oxidative stress, an up to 22% increase in serum antioxidant capacity, a significant 33% reduction in macrophage uptake of oxidized LDL, and a 25% decrease in macrophage-mediated oxidation of LDL relative to controls. Grape powder directly protected both plasma LDL and macrophages from oxidative stress in vitro. We conclude that polyphenols from fresh grape powder directly affect macrophage atherogenicity by reducing macrophage-mediated oxidation of LDL and cellular uptake of oxidized LDL. Both of these processes can eventually reduce macrophage cholesterol accumulation and foam cell formation and hence attenuate atherosclerosis development.


Subject(s)
Apolipoproteins E/physiology , Arteriosclerosis/prevention & control , Flavonoids/pharmacology , Macrophages, Peritoneal/physiology , Phenols/pharmacology , Phytotherapy , Plant Extracts/pharmacology , Vitis , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/genetics , CD36 Antigens/genetics , Cell Line , Lipid Peroxides/metabolism , Lipoproteins, LDL/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/pathology , Mice , Mice, Knockout , Polyphenols , RNA, Messenger/genetics , Receptors, LDL/genetics , Thiobarbituric Acid Reactive Substances/metabolism
7.
Curr Atheroscler Rep ; 6(6): 485-91, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15485595

ABSTRACT

The dietary intake of omega-3 (n-3) polyunsaturated fatty acids has emerged, over the past 20 years, as an important way to modify cardiovascular risk. This likely occurs through beneficial effects at all stages in the natural history of vascular disease, from the inception of atherosclerotic lesions, to their growth and acute complications (plaque rupture in most instances), up to protection of myocardium from the consequences of ensuing acute myocardial ischemia. This review specifically focuses on the modulating effects of n-3 fatty acids on biologic events involved in early atherogenesis, including important properties of these natural substances on endothelial expression of adhesion molecules and cytokines, processes collectively denoted as "endothelial activation." By decreasing the endothelial responsiveness to proinflammatory and proatherogenic stimuli, n-3 fatty acids act on molecular events not targeted by any other drugs or interventions, and thereby complementary to those of already implemented pharmacologic treatments.


Subject(s)
Arteriosclerosis/prevention & control , Cell Adhesion Molecules/metabolism , Cytokines/metabolism , Endothelium, Vascular/drug effects , Fatty Acids, Omega-3/pharmacology , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Endothelium, Vascular/metabolism , Gene Expression Regulation/physiology , Humans
9.
Circulation ; 109(25): 3215-20, 2004 Jun 29.
Article in English | MEDLINE | ID: mdl-15197147

ABSTRACT

BACKGROUND: These studies were designed to determine the mechanism of action of an oral apolipoprotein (apo) A-I mimetic peptide, D-4F, which previously was shown to dramatically reduce atherosclerosis in mice. METHODS AND RESULTS: Twenty minutes after 500 microg of D-4F was given orally to apoE-null mice, small cholesterol-containing particles (CCPs) of 7 to 8 nm with pre-beta mobility and enriched in apoA-I and paraoxonase activity were found in plasma. Before D-4F, both mature HDL and the fast protein liquid chromatography fractions containing the CCPs were proinflammatory. Twenty minutes after oral D-4F, HDL and CCPs became antiinflammatory, and there was an increase in HDL-mediated cholesterol efflux from macrophages in vitro. Oral D-4F also promoted reverse cholesterol transport from intraperitoneally injected cholesterol-loaded macrophages in vivo. In addition, oral D-4F significantly reduced lipoprotein lipid hydroperoxides (LOOH), except for pre-beta HDL fractions, in which LOOH increased. CONCLUSIONS: The mechanism of action of oral D-4F in apoE-null mice involves rapid formation of CCPs, with pre-beta mobility enriched in apoA-I and paraoxonase activity. As a result, lipoprotein LOOH are reduced, HDL becomes antiinflammatory, and HDL-mediated cholesterol efflux and reverse cholesterol transport from macrophages are stimulated.


Subject(s)
Apolipoprotein A-I/pharmacology , Apolipoproteins E/deficiency , Arteriosclerosis/genetics , Cholesterol/metabolism , Hyperlipoproteinemia Type II/genetics , Lipoproteins, HDL/biosynthesis , Macrophages, Peritoneal/drug effects , Administration, Oral , Amino Acid Sequence , Animals , Apolipoprotein A-I/therapeutic use , Apolipoproteins E/genetics , Arteriosclerosis/blood , Aryldialkylphosphatase/blood , Biological Transport/drug effects , Cells, Cultured , Chemotaxis/drug effects , Coculture Techniques , Drug Evaluation, Preclinical , Female , High-Density Lipoproteins, Pre-beta , Humans , Hyperlipoproteinemia Type II/blood , Inflammation , Lipid Peroxidation/drug effects , Lipoproteins, HDL/blood , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism
10.
Food Chem Toxicol ; 42(8): 1259-67, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15207376

ABSTRACT

Eggplant (Solanum melongena) has been used as hypocholesterolemic agent in many countries. However, few controlled studies were addressed to this subject and atherogenesis. We have evaluated the effect of eggplant on cholesterol metabolism and atherogenesis in LDLR(-/-) mice. Animals were fed on chow (n=17) or atherogenic (n=21) diet during 12 weeks receiving water (control) or eggplant extract. Liver, serum and fecal lipids, together with serum lipoproteins were measured. Oxidative stress was evaluated through conjugate diene formation and ox-LDL antibodies by enzyme immunoassay. Atherosclerotic lesions were measured in different sites of aorta. Total cholesterol and atherogenic lipoproteins did not decrease after eggplant intake. Animals receiving eggplant and chow diet showed increased anti-ox-LDL antibodies and a decreased lag phase of conjugated diene formation, indicating a higher oxidative stress than controls. No differences were seen in lesion area of aortic valve. Eggplant extract had high histamine and other amine levels that could enhance LDL oxidation and its endocytosis. Eggplant did not decrease plasma cholesterol nor prevent the development of atherosclerosis in LDLR(-/-) mice. Surprisingly, eggplant increased oxidative stress, representing a risk factor for atherosclerosis. These results did not support the use of eggplant extract as hypocholesterolemic agent.


Subject(s)
Arteriosclerosis/genetics , Arteriosclerosis/pathology , Oxidative Stress/physiology , Receptors, LDL/genetics , Solanum melongena/chemistry , Animals , Antibodies/analysis , Aorta/pathology , Body Weight/drug effects , Cholesterol/blood , Diet , Diet, Atherogenic , Feces/chemistry , Lipids/blood , Lipoproteins/blood , Lipoproteins/immunology , Lipoproteins/metabolism , Liver/metabolism , Mice , Mice, Knockout , Myocardium/pathology , Plant Extracts/pharmacology
11.
Mil Med ; 169(4): 325-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15132238

ABSTRACT

Within the past four decades, the efforts of investigators worldwide have established the amino acid homocysteine as an important factor in arteriosclerosis and diseases of aging. After its discovery in 1932, homocysteine was demonstrated to be an important intermediate in the metabolism of amino acids. However, little was known about the broader biomedical significance of homocysteine until 1962, when children with mental retardation, accelerated growth, dislocated ocular lenses, and frequent vascular thrombosis were found to excrete homocysteine in the urine. My study of two patients with homocystinuria caused by different inherited enzymatic disorders in 1968 disclosed advanced widespread arteriosclerotic plaques in both cases. This discovery led to the conclusion that homocysteine causes vascular disease by a direct effect on the cells and tissues of the arteries. This interpretation suggests that homocysteine is important in the pathogenesis of arteriosclerosis in persons with hereditary, dietary, environmental, hormonal, metabolic, and other factors predisposing them to hyperhomocysteinemia. Within the past decade, many major clinical and epidemiological studies have proven that hyperhomocysteinemia is a potent independent risk factor for vascular disease. According to the homocysteine theory of arteriosclerosis, insufficient dietary intake of the B vitamins, folic acid and pyridoxine, caused by losses of these nutrients during processing of foods, leads to elevation of blood homocysteine and vascular disease in the general population. The dramatic decline in cardiovascular mortality since the 1960s in the United States is attributed to fortification of the food supply by synthetic pyridoxine and folic acid. The recent Swiss Heart Study showed that B vitamins slowed restenosis in patients with coronary arteriosclerosis treated with angioplasty. Currently, more than 20 prospective, worldwide, interventional trials involving at least 100,000 participants are examining whether lowering plasma homocysteine levels with supplemental B vitamins will prevent mortality and morbidity from arteriosclerotic vascular disease.


Subject(s)
Arteriosclerosis/prevention & control , Homocysteine/blood , Vitamin B Complex/therapeutic use , Animals , Arteriosclerosis/etiology , Arteriosclerosis/genetics , Arteriosclerosis/physiopathology , Dietary Proteins/administration & dosage , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/genetics , Models, Theoretical , Risk Factors , Vitamin B Complex/administration & dosage
12.
Circ Res ; 94(9): 1168-78, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15142970

ABSTRACT

A large body of data gathered over the past couple of years has identified the peroxisome proliferator-activated receptors (PPAR) alpha, gamma, and beta/delta as transcription factors exerting modulatory actions in vascular cells. PPARs, which belong to the nuclear receptor family of ligand-activated transcription factors, were originally described as gene regulators of various metabolic pathways. Although the PPARalpha, gamma, and beta/delta subtypes are approximately 60% to 80% homologous in their ligand- and DNA-binding domains, significant differences in ligand and target gene specificities are observed. PPARalpha is activated by polyunsaturated fatty acids and oxidized derivatives and by lipid-modifying drugs of the fibrate family, including fenofibrate or gemfibrozil. PPARalpha controls expression of genes implicated in lipid metabolism. PPARgamma, in contrast, is a key regulator of glucose homeostasis and adipogenesis. Ligands of PPARgamma include naturally occurring FA derivatives, such as hydroxyoctadecadienoic acids (HODEs), prostaglandin derivatives such as 15-deoxyDelta12,14-prostaglandin J2, and glitazones, insulin-sensitizing drugs presently used to treat patients with type 2 diabetes. Ligands for PPARbeta/delta are polyunsaturated fatty acids, prostaglandins, and synthetic compounds, some of which are presently in clinical development. PPARbeta/delta stimulates fatty acid oxidation predominantly acting in muscle. All PPARs are expressed in vascular cells, where they exhibit antiinflammatory and antiatherogenic properties. In addition, studies in various animal models as well as clinical data suggest that PPARalpha and PPARgamma activators can modulate atherogenesis in vivo. At present, no data are available relating to possible effects of PPARbeta/delta agonists on atherogenesis. Given the widespread use of PPARalpha and PPARgamma agonists in patients at high risk for cardiovascular disease, the understanding of their function in the vasculature is not only of basic interest but also has important clinical implications. This review will focus on the role of PPARs in the vasculature and summarize the present understanding of their effects on atherogenesis and its cardiovascular complications.


Subject(s)
Arteriosclerosis/prevention & control , Gene Expression Regulation/drug effects , Peroxisome Proliferators/pharmacology , Animals , Arteriosclerosis/chemically induced , Arteriosclerosis/genetics , Clinical Trials as Topic , Consensus Sequence , Dimerization , Drug Design , Drug Evaluation, Preclinical , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Peroxisome Proliferators/therapeutic use , Peroxisome Proliferators/toxicity , Rats , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Transcription Factors/drug effects , Transcription Factors/metabolism , Transcription Factors/physiology , Transcription, Genetic/drug effects
14.
Biochem Cell Biol ; 82(1): 212-24, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15052339

ABSTRACT

Atherosclerosis is the leading cause of death in North America and within the next two decades will be the leading cause worldwide. Atherosclerosis is characterized by vascular obstruction from the deposits of plaque, resulting in reduced blood flow. Plaque rupture and the consequent thrombosis may lead to sudden blockage of the arteries and cause heart attack. High serum lipid levels, especially the elevated level of low-density lipoprotein (LDL), have been shown to be strongly related to the development of atherosclerosis. It is generally accepted that atherosclerotic lesions are initiated via an enhancement of LDL uptake by monocytes and macrophages. In the liver, uptake of plasma LDL is mediated via specific LDL receptors, but a scavenger receptor system is employed by macrophages. Plasma LDL must be modified prior to uptake by macrophages. Analysis of the lipid content in the oxidatively modified LDL from hyper lipidemic patients revealed that the level of lysophosphatidylcholine was greatly elevated, and the high level of the lysolipid was shown to impair the endothelium-dependent relaxation of the blood vessels. In a separate study, we showed that a high level of homocysteine caused the increase in cholesterol production and apolipoprotein B-100 secretion in hepatic cells. Statins have been used effectively to control the production of cholesterol in the liver, and recently, ezetimibe has been shown to supplement the efficacy of statins by inhibiting cholesterol absorption. The factor of elevated levels of triglyceride-rich lipoproteins in association with depressed high-density lipoproteins, usually in the context of insulin resistance, is an important contributor to atherosclerosis and can be effectively treated with fibric acid derivatives. In hyperhomocysteinemia, folic acid supplements may have a role in the control of cholesterol by reducing the plasma homocysteine level.


Subject(s)
Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Lipids/blood , Arteriosclerosis/etiology , Chemokines/genetics , Chemokines/metabolism , Cholesterol/metabolism , Endothelium, Vascular/metabolism , Homocysteine/blood , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/etiology , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Hyperlipidemias/therapy , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/physiology , Risk Factors
15.
Phytother Res ; 18(2): 177-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15022174

ABSTRACT

The genomics of atherosclerosis can arise as a result of cross-talk between the genes coding for the LDL-receptor (LDL-R), LXR-alpha, PPARs (alpha, gamma), CD36 and C-myc because these genes control lipid metabolism, cytokine production and cellular activity within the arterial wall. The effect of green tea polyphenols (GTPs) upon such genomics revealed their ability to down-regulate genes coding for PPAR-gamma, CD36, LXR-alpha, C-myc coupled with up-regulation of genes coding for LDL-R and PPAR-alpha at the transcriptional level. Based upon these results, it is proposed that GTPs have the inherent capacity to inhibit the development of atherosclerotic lesions.


Subject(s)
Arteriosclerosis/genetics , Arteriosclerosis/prevention & control , Flavonoids/pharmacology , Phenols/pharmacology , Phytotherapy , Tea , DNA-Binding Proteins , Dose-Response Relationship, Drug , Flavonoids/administration & dosage , Flavonoids/therapeutic use , Gene Expression Regulation , Genes, myc/genetics , Humans , Leukocytes, Mononuclear/drug effects , Liver X Receptors , Orphan Nuclear Receptors , Phenols/administration & dosage , Phenols/therapeutic use , Polyphenols , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, LDL/genetics , Transcription Factors/genetics
16.
Proc Natl Acad Sci U S A ; 100(24): 14235-40, 2003 Nov 25.
Article in English | MEDLINE | ID: mdl-14614151

ABSTRACT

An intercross between atherosclerosis susceptible (C57BL/6J ApoE0) and resistant (FVB/N ApoE0) mice revealed a susceptibility locus on chromosome 10 (11 cM, logarithm of odds 7.8). Surprisingly, the genotypic means for this locus revealed that heterozygosity or homozygosity for the C57BL/6J allele was associated with decreased atherosclerosis. A candidate gene in this region is A20, which is involved in the feedback suppression of NFkappaB activation induced by tumor necrosis factor alpha (TNFalpha). We sequenced the A20 gene coding region from the parental strains and found a single-nucleotide polymorphism resulting in a single amino acid exchange, Glu627Ala (C57BL/6J vs. FVB/N). This mutation introduces a putative casein kinase 2 phosphorylation site in C57BL/6J-A20 not present in FVB/N-A20. NFkappaB reporter gene assays showed that this amino acid change results in less effective termination of TNFalpha-stimulated NFkappaB activation by C57BL/6J-A20. In accordance, the TNFalpha-induced expression of NFkappaB target genes (A20, IkappaBalpha) in vascular smooth muscle cells was prolonged in cells isolated from C57BL/6J compared with FVB/N mice. In light of the genotypic means for atherosclerosis at the chromosome 10 locus in F2 mice from this intercross, the observations now reported suggest that prolonged expression of genes induced by NFkappaB might be antirather than proatherogenic.


Subject(s)
Arteriosclerosis/genetics , Arteriosclerosis/metabolism , NF-kappa B/metabolism , Proteins/genetics , Proteins/metabolism , Amino Acid Sequence , Animals , Apolipoproteins E/genetics , Base Sequence , Chromosome Mapping , Cysteine Endopeptidases , DNA, Complementary/genetics , Gene Expression/drug effects , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , NF-KappaB Inhibitor alpha , Nuclear Proteins , Polymorphism, Single Nucleotide , Recombinant Proteins/pharmacology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Species Specificity , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/pharmacology
17.
Biochem Soc Trans ; 31(Pt 5): 1075-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14505483

ABSTRACT

Conjugated linoleic acid (CLA) refers to a group of positional and geometric isomers of linoleic acid that has been shown to suppress the development of atherosclerosis in a rabbit model. We investigated whether CLA acts as a cyclo-oxygenase (COX) inhibitor or as an agonist of the peroxisome-proliferator-activator receptor (PPAR) gamma in the ApoE(-/-) mouse model. In vitro, a 9-cis, 11-trans isomer of CLA inhibited prostaglandin formation and oxygen consumption by both isoforms of COX, with no evidence by MS of alternative products being generated. In vivo, supplementation with CLA was found to induce resolution of atherosclerosis. The effect of CLA in vivo could not be explained by COX inhibition alone, as urinary prostaglandin levels were unchanged in animals receiving CLA supplementation, and administration of selective COX inhibitors did not induce lesion regression. There was however induction of PPAR gamma, a known response to agonists of this nuclear orphan receptor.


Subject(s)
Apolipoproteins E/genetics , Arteriosclerosis/genetics , Arteriosclerosis/pathology , Linoleic Acid/genetics , Animals , Cell Nucleus/metabolism , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Eicosanoids/metabolism , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/metabolism , Linoleic Acid/metabolism , Membrane Proteins , Mice , Models, Biological , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , Protein Isoforms , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism
18.
Acta Physiol Scand ; 179(2): 123-35, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14510775

ABSTRACT

Nitric oxide synthases (NOSs) generate nitric oxide (NO) and the by-product l-citrulline, via the catalytic combination of l-arginine and molecular oxygen. In mammals, there are three NOS genes: nNOS (NOS1), iNOS (NOS2) and eNOS (NOS3). The molecular structure, enzymology and pharmacology of these enzymes have been well defined, and reveal critical roles for the NOS system in a variety of important processes. The studies of NOS enzymes using knockout and transgenic mouse models have provided an invaluable contribution, highlighting critical roles in neuronal, renal, pulmonary, gastro-intestinal, skeletal muscle, reproductive and cardiovascular biology. This review will outline the data gleaned from complementary knockout and transgenic over-expression models in mice, and focus on the interactions between NOS enzymes and pathophysiology of the vascular system. These studies are a paradigm for the near future, which will involve the translation of an enormous amount of genomic data into physiological insights that penetrate the realms of both health care and biology.


Subject(s)
Nitric Oxide Synthase/metabolism , Animals , Arteriosclerosis/genetics , Coronary Vessels/physiology , Disease Models, Animal , Endotoxins/adverse effects , Liver Circulation/genetics , Male , Mice , Mice, Knockout , Muscle, Skeletal/blood supply , Neurons/physiology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Penis/blood supply , Pulmonary Circulation/genetics , Renal Circulation/genetics
19.
Arterioscler Thromb Vasc Biol ; 23(1): 97-103, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12524231

ABSTRACT

OBJECTIVE: L-arginine serves as a substrate for the formation of NO by the NO synthase (NOS) enzymes. In some studies, dietary supplementation of L-arginine reduces atherosclerosis through the restoration of NO release and improvement in endothelial function. In the present study, we investigate the effect of L-arginine supplementation on the development of atherosclerosis in a mouse model. METHODS AND RESULTS: Apolipoprotein E (apoE) knockout (ko) and apoE/inducible NOS (iNOS) double-ko mice were fed a western-type diet with or without L-arginine supplementation in the drinking water (25 g/L). L-Arginine did not affect the lesion area after 16 weeks or 24 weeks in apoE ko mice. However, L-arginine negates the protective effect of iNOS gene deficiency. In contrast to apoE/iNOS dko mice without arginine supplementation, lesion areas were increased in apoE/iNOS double-ko mice with arginine supplementation at 24 weeks. This was associated with an increase in thiobarbituric acid-reactive malondialdehyde adducts, nitrotyrosine staining within lesions, and a decrease in the ratio of reduced tetrahydrobiopterin to total biopterins. CONCLUSIONS: Although L-arginine supplementation does not affect lesion formation in the western-type diet-fed apoE ko mice, it negates the protective effect of iNOS gene deficiency in this model. This raises the possibility that L-arginine supplementation may paradoxically contribute to, rather than reduce, lesion formation by mechanisms that involve lipid oxidation, peroxynitrite formation, and NOS uncoupling.


Subject(s)
Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arginine/administration & dosage , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Disease Models, Animal , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/genetics , Tyrosine/analogs & derivatives , Animals , Aorta/chemistry , Aorta/enzymology , Aorta/physiology , Apolipoproteins E/physiology , Arginine/metabolism , Arteriosclerosis/enzymology , Arteriosclerosis/physiopathology , Biopterins/blood , Blotting, Western , Cholesterol/blood , Immunohistochemistry , Isoenzymes/immunology , Malondialdehyde/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase/physiology , Nitric Oxide Synthase Type II , Staining and Labeling , Thiobarbituric Acid Reactive Substances/metabolism , Tyrosine/immunology
20.
Arterioscler Thromb Vasc Biol ; 23(1): 104-10, 2003 Jan 01.
Article in English | MEDLINE | ID: mdl-12524232

ABSTRACT

OBJECTIVE: The role of insulin in atherosclerosis progression in diabetes is uncertain. We examined the effects of oral insulin supplementation on atherogenesis in apolipoprotein E-deficient (E(0)) mice. METHODS AND RESULTS: One-month-old male E(0) mice were orally supplemented with human insulin (0.1, 0.5, and 1 U/mL) or placebo for 3 months. At the end of the study, serum and macrophage oxidative stress and atherosclerosis progression were studied. Insulin reduced lesion size by 22% to 37% (P<0.05) in all study groups. Lipid peroxides serum levels were 18% lower (P<0.01), and serum paraoxonase activity was 30% higher (P<0.01) in mice supplemented with 1.0 U/mL insulin compared with controls. Insulin reduced mouse peritoneal macrophage (MPM) lipid peroxides content and superoxide anion release by up to 44% and 62%, respectively (P<0.01). In addition, oral insulin reduced MPM cholesterol content and cholesterol biosynthesis by up to 36% and 53%, respectively (P<0.01). In vitro incubation of E(0) mice MPM with increasing insulin concentrations (0 to 100 micro U/mL) resulted in a dose-dependent reduction of cholesterol synthesis by up to 66% (P<0.05). CONCLUSIONS: In E(0) mice, oral insulin supplementation attenuates the atherosclerotic process. This may be attributable to insulin-mediated reduction of oxidative stress in serum and macrophages as well as reduction in macrophage cholesterol content.


Subject(s)
Apolipoproteins E/deficiency , Arteriosclerosis/drug therapy , Arteriosclerosis/genetics , Insulin/therapeutic use , Administration, Oral , Animals , Aorta/drug effects , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/drug therapy , Apolipoproteins E/genetics , Arteriosclerosis/blood , Aryldialkylphosphatase , Blood Glucose/metabolism , Blood Pressure/drug effects , Body Weight/drug effects , Cholesterol/biosynthesis , Dietary Supplements , Dose-Response Relationship, Drug , Esterases/blood , Humans , Insulin/administration & dosage , Insulin/blood , Lipid Peroxidation/drug effects , Lipids/blood , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Oxidative Stress/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL