Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Curr Atheroscler Rep ; 21(12): 48, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31741187

ABSTRACT

PURPOSE OF REVIEW: The goal of this review is to evaluate the role of inhibiting the synthesis of lipoproteins when there is no or little residual LDL-receptor function as in patients with homozygous familial hypercholesterolaemia. Lomitapide is administered orally once a day while mipomersen is given by subcutaneous injection once a week. Lomitapide inhibits microsomal triglyceride transfer protein while mipomersen is an antisense oligonucleotide directed against apoB100. RECENT FINDINGS: The pivotal registration trials for lomitapide and mipomersen were published in 2013 and 2010, respectively. More recently published data from extension trials and cohort studies provides additional information on long-term safety and efficacy. The mean LDL cholesterol reduction was 50% with lomitapide in its single-arm open-label registration trial. Mipomersen reduced LDL cholesterol by approximately 25% in its double-blind, placebo-controlled registration study. Both lomitapide and mipomersen therapy are associated with variable increases in hepatic fat content. The long-term safety of increased hepatic fat content in patients receiving these therapies is uncertain and requires further study. Both drugs may cause elevated transaminase in some patients, but no cases of severe liver injury have been reported. Lomitapide may also cause gastrointestinal discomfort and diarrhoea, especially if patients consume high-fat meals and patients are advised to follow a low-fat diet supplemented with essential fatty acids and fat-soluble vitamins. Mipomersen may cause injection-site and influenza-like reactions. The effect of lomitapide and mipomersen on cardiovascular outcomes has not been studied, but circumstantial evidence suggests that the LDL cholesterol lowering achieved with these two agents may reduce cardiovascular event rates.


Subject(s)
Apolipoprotein B-100/biosynthesis , Benzimidazoles/pharmacology , Hyperlipoproteinemia Type II , Oligonucleotides/pharmacology , Anticholesteremic Agents/pharmacology , Cardiovascular Diseases/prevention & control , Carrier Proteins/antagonists & inhibitors , Humans , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/metabolism , Microsomes
2.
Biochim Biophys Acta Mol Basis Dis ; 1864(12): 3697-3713, 2018 12.
Article in English | MEDLINE | ID: mdl-30292637

ABSTRACT

Familial Hypercholesterolemia (FH) is an autosomal co-dominant genetic disorder characterized by elevated low-density lipoprotein (LDL) cholesterol levels and increased risk for premature cardiovascular disease. Here, we examined FH pathophysiology in skin fibroblasts derived from FH patients harboring heterozygous mutations in the LDL-receptor. Fibroblasts from FH patients showed a reduced LDL-uptake associated with increased intracellular cholesterol levels and coenzyme Q10 (CoQ10) deficiency, suggesting dysregulation of the mevalonate pathway. Secondary CoQ10 deficiency was associated with mitochondrial depolarization and mitophagy activation in FH fibroblasts. Persistent mitophagy altered autophagy flux and induced inflammasome activation accompanied by increased production of cytokines by mutant cells. All the pathological alterations in FH fibroblasts were also reproduced in a human endothelial cell line by LDL-receptor gene silencing. Both increased intracellular cholesterol and mitochondrial dysfunction in FH fibroblasts were partially restored by CoQ10 supplementation. Dysregulated mevalonate pathway in FH, including increased expression of cholesterogenic enzymes and decreased expression of CoQ10 biosynthetic enzymes, was also corrected by CoQ10 treatment. Reduced CoQ10 content and mitochondrial dysfunction may play an important role in the pathophysiology of early atherosclerosis in FH. The diagnosis of CoQ10 deficiency and mitochondrial impairment in FH patients may also be important to establish early treatment with CoQ10.


Subject(s)
Ataxia/complications , Cholesterol/metabolism , Fibroblasts/pathology , Hyperlipoproteinemia Type II/complications , Mitochondrial Diseases/complications , Muscle Weakness/complications , Ubiquinone/deficiency , Ataxia/metabolism , Ataxia/pathology , Cells, Cultured , Fibroblasts/metabolism , Humans , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type II/pathology , Lipoproteins, LDL/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitophagy , Muscle Weakness/metabolism , Muscle Weakness/pathology , Reactive Oxygen Species/metabolism , Receptors, LDL/metabolism , Ubiquinone/metabolism
3.
J Clin Invest ; 125(10): 3819-30, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26368306

ABSTRACT

Recent genome-wide association studies have revealed that variations near the gene locus encoding the transcription factor Krüppel-like factor 14 (KLF14) are strongly associated with HDL cholesterol (HDL-C) levels, metabolic syndrome, and coronary heart disease. However, the precise mechanisms by which KLF14 regulates lipid metabolism and affects atherosclerosis remain largely unexplored. Here, we report that KLF14 is dysregulated in the liver of 2 dyslipidemia mouse models. We evaluated the effects of both KLF14 overexpression and genetic inactivation and determined that KLF14 regulates plasma HDL-C levels and cholesterol efflux capacity by modulating hepatic ApoA-I production. Hepatic-specific Klf14 deletion in mice resulted in decreased circulating HDL-C levels. In an attempt to pharmacologically target KLF14 as an experimental therapeutic approach, we identified perhexiline, an approved therapeutic small molecule presently in clinical use to treat angina and heart failure, as a KLF14 activator. Indeed, in WT mice, treatment with perhexiline increased HDL-C levels and cholesterol efflux capacity via KLF14-mediated upregulation of ApoA-I expression. Moreover, perhexiline administration reduced atherosclerotic lesion development in apolipoprotein E-deficient mice. Together, these results provide comprehensive insight into the KLF14-dependent regulation of HDL-C and subsequent atherosclerosis and indicate that interventions that target the KLF14 pathway should be further explored for the treatment of atherosclerosis.


Subject(s)
Apolipoprotein A-I/biosynthesis , Atherosclerosis/prevention & control , Cholesterol, HDL/blood , Cholesterol/metabolism , Hyperlipoproteinemia Type II/drug therapy , Kruppel-Like Transcription Factors/physiology , Liver/metabolism , Perhexiline/pharmacology , Animals , Apolipoprotein A-I/genetics , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/therapy , Diet, Atherogenic , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Genetic Therapy , Genetic Vectors/therapeutic use , Genome-Wide Association Study , Hep G2 Cells , Humans , Hyperlipoproteinemia Type II/metabolism , Kruppel-Like Transcription Factors/agonists , Leptin/deficiency , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Obese , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Sp Transcription Factors/genetics , Sp Transcription Factors/metabolism , Sterol Regulatory Element Binding Proteins/biosynthesis , Sterol Regulatory Element Binding Proteins/genetics
4.
Rheum Dis Clin North Am ; 39(2): 481-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23597975

ABSTRACT

Rheumatologic manifestations of hyperlipidemia and lipid-associated arthritis are rarely seen in the rheumatologist's office. On the other hand, a rheumatologist may be the clinician who identifies and initiates proper therapy for disorders related to hyperlipidemia when the musculoskeletal manifestations of these syndromes are recognized. In this article both the joint and tendon manifestations are reviewed, including the lesser known lipid liquid crystal form of arthritis. The relationship between gout and hyperuricemia is briefly discussed, as are the autoimmune manifestations of lipid-lowering therapy.


Subject(s)
Arthritis/complications , Hyperlipoproteinemia Type III/complications , Hyperlipoproteinemia Type II/complications , Xanthomatosis/complications , Arthritis/immunology , Arthritis/metabolism , Autoimmune Diseases/complications , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Gout/complications , Gout/immunology , Gout/metabolism , Humans , Hyperlipoproteinemia Type II/immunology , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type III/immunology , Hyperlipoproteinemia Type III/metabolism , Hyperuricemia/complications , Hyperuricemia/immunology , Hyperuricemia/metabolism , Lipid Metabolism , Tendons , Xanthomatosis/immunology , Xanthomatosis/metabolism
5.
Drugs ; 72(11): 1445-55, 2012 Jul 30.
Article in English | MEDLINE | ID: mdl-22799743

ABSTRACT

High levels of low-density lipoprotein cholesterol (LDL-C) and lipoprotein(a) [Lp(a)] are associated with early morbidity and mortality caused by cardiovascular disease (CVD). There are hints that a reduction of LDL-C levels beyond currently advocated targets, and the use of drugs that also have Lp(a)-lowering potential, could provide further clinical benefit. Today, LDL apheresis is the only available treatment option to achieve further lowering of apolipoprotein-B (apo-B)-containing lipoproteins, especially Lp(a). Mipomersen is currently being studied in patients with mild to severe hypercholesterolaemia as add-on therapy to other lipid-lowering therapy, as monotherapy in patients who are intolerant of HMG-CoA reductase inhibitors (statins) and who are at high risk for CVD. Patients affected by homozygous or heterozygous familial hypercholesterolaemia (FH), which are inherited autosomal co-dominant disorders characterized by a marked elevation of serum LDL-C concentration, remain a clinical challenge, especially when their CVD risk is aggravated by additionally elevated Lp(a) levels. Mipomersen is a 20-mer oligonucleotide [2'-O-(2-methoxy) ethyl-modified oligonucleotide], a second-generation antisense oligonucleotide (AOS), complementary to the coding region for human-specific apo-B-100 messenger RNA (mRNA). Mipomersen inhibits apo-B-100 synthesis and is consequently a new treatment strategy to lower apo-B-containing lipoproteins like LDL-C and Lp(a) in patients at high risk for CVD not on target or intolerant to statins. This article focuses on mipomersen and gives an overview of the current status of mipomersen as a promising treatment option. Recent studies have shown a decrease in LDL-C levels of 22-42.2% and in Lp(a) of 19.6-31.1% from baseline, depending on study design. Dose-dependent reductions of very low-density lipoprotein cholesterol (VLDL-C) and triglyceride levels have also been observed. Although the short-term efficacy and safety of mipomersen have been proven, side effects like injection-site reactions (up to 90-100%), increased liver enzymes, cephalgias, nasopharyngitis, myalgia, nausea and fatigue must be mentioned and critically discussed. Furthermore, we need more data on the long-term side effects, especially regarding the long-term potential for hepatic steatosis. Data on cardiovascular outcomes with mipomersen are also not yet available.


Subject(s)
Anticholesteremic Agents/therapeutic use , Hyperlipoproteinemia Type II/drug therapy , Oligonucleotides/therapeutic use , Animals , Anticholesteremic Agents/adverse effects , Apolipoprotein B-100/antagonists & inhibitors , Apolipoprotein B-100/blood , Apolipoprotein B-100/metabolism , Cholesterol, LDL/blood , Cholesterol, LDL/metabolism , Cholesterol, VLDL/blood , Cholesterol, VLDL/metabolism , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Double-Blind Method , Drug Evaluation, Preclinical , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/metabolism , Lipoprotein(a)/blood , Lipoprotein(a)/metabolism , Oligonucleotides/adverse effects , Randomized Controlled Trials as Topic , Triglycerides/blood , Triglycerides/metabolism
6.
Free Radic Res ; 44(7): 821-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20528577

ABSTRACT

Oxidative stress contributes to lipid peroxidation and decreases nitric oxide (NO) bioavailability in atherosclerosis. While long-chain (n-3) polyunsaturated fatty acids (PUFA) are easily oxidized in vitro, they improve endothelial function. Hence, this study postulates that long-chain (n-3) PUFA decrease atherogenic oxidative stress in vivo. To test this, apoE(-/-) mice were fed a corn oil- or a fish oil (FO)-rich diet for 8, 14 or 20 weeks and parameters related to NO and superoxide (O(2)(.-)) plus markers of lipid peroxidation and protein oxidative damage in the aortic root were evaluated. The FO-rich diet increased NO production and endothelial NO synthase (NOS) expression and lowered inducible NOS, p22(phox) expression and O(2)(.-)production after 14 and 20 weeks of diet. Protein lipoxidative damage (including 4-hydroxynonenal) was decreased after a long-term FO-diet. This supports the hypothesis that a FO-rich diet could counteract atherogenic oxidative stress, showing beneficial effects of long-chain (n-3) PUFA.


Subject(s)
Apolipoproteins E/deficiency , Endothelium, Vascular/drug effects , Fish Oils/therapeutic use , Hyperlipoproteinemia Type II/diet therapy , Oxidative Stress/drug effects , Aldehydes/analysis , Animals , Aorta/chemistry , Aorta/drug effects , Aorta/enzymology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Corn Oil/administration & dosage , Corn Oil/pharmacology , Cytochrome b Group/biosynthesis , Cytochrome b Group/genetics , Fish Oils/administration & dosage , Fish Oils/pharmacology , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/metabolism , Lipid Peroxidation/drug effects , Male , Mice , Mice, Knockout , NADPH Oxidases/biosynthesis , NADPH Oxidases/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics
7.
Physiol Res ; 58(2): 179-184, 2009.
Article in English | MEDLINE | ID: mdl-18380539

ABSTRACT

A recently discussed cardiovascular risk factor, asymmetric dimethylarginine (ADMA), is known to act as an endogenous inhibitor of endothelial nitric oxide synthase. The aim of this study was to establish 1) the relationship between ADMA and ultrasonographically or biochemically determined endothelial dysfunction in children, and 2) the effect of folate supplementation on these parameters. The study cohort included 32 children with familial hypercholesterolemia (FH), 30 with diabetes mellitus type 1 (DM1) and 30 age-matched healthy children as the control group. Furthermore, twenty-eight randomly selected FH and DM1 children were re-examined after 3-months supplementation with folic acid. Baseline levels of ADMA and oxidized low density lipoproteins (oxLDL) were significantly higher in FH group than in DM1 and healthy children. Children in DM1 group had significantly lower concentration of homocysteine, but ADMA levels were normal. Folic acid supplementation significantly lowered homocysteine and hsCRP levels in both FH and DM1 group; however, ADMA and oxLDL concentrations remained unaltered. In conclusion, ADMA and oxLDL appear to be associated with endothelial dysfunction in children with FH. Administration of folic acid did not influence these markers in both FH and DM1 children.


Subject(s)
Arginine/analogs & derivatives , Diabetes Mellitus, Type 1/metabolism , Folic Acid/administration & dosage , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/metabolism , Vitamin B Complex/administration & dosage , Adolescent , Anticholesteremic Agents/administration & dosage , Arginine/blood , Azetidines/administration & dosage , Biomarkers/blood , Child , Drug Therapy, Combination , Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Ezetimibe , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hyperlipoproteinemia Type II/epidemiology , Lipoproteins, LDL/blood , Male , Risk Factors , Ultrasonography
8.
Br J Nutr ; 98(5): 890-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17537294

ABSTRACT

Foods containing plant sterol or stanol esters can be beneficial in lowering LDL-cholesterol concentration, a major risk factor for CVD. The present study examined whether high dietary intake of rapeseed oil (RSO) derived plant sterol and stanol esters is associated with increased levels of these components in brain tissue of homozygous and heterozygous Watanabe rabbits, an animal model for familial hypercholesterolemia. Homozygous animals received either a standard diet, RSO stanol or RSO sterol ester while heterozygous animals were additionally fed with 2 g cholesterol/kg to the respective diet form for 120 d (n 9 for each group). Concentrations of cholesterol, its precursor lathosterol, plant sterols and stanols in brain and additionally in liver and plasma were determined by highly sensitive GC-MS. High-dose intake of RSO derived plant sterols and stanols resulted in increased levels of these components in plasma and liver. In brain a limited uptake of plant sterols and stanols was proven, indicating that these compounds passed the blood-brain barrier and may be retained in the brain tissue of Watanabe rabbits. Plant stanol ester feeding lowered plant sterol levels in brain, liver, and plasma. Cholesterol synthesis in brain, indicated by lathosterol, a local surrogate cholesterol synthesis marker, does not seem to be affected by plant sterol or stanol ester feeding. We conclude that high dose intake of plant sterol and stanol esters in Watanabe rabbits results in elevated concentrations of these components not only in the periphery but also in the central nervous system.


Subject(s)
Brain/metabolism , Phytosterols/pharmacokinetics , Plant Oils/chemistry , Sitosterols/pharmacokinetics , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Blood-Brain Barrier , Cholesterol/metabolism , Disease Models, Animal , Fatty Acids, Monounsaturated , Female , Heterozygote , Homozygote , Hyperlipoproteinemia Type II/metabolism , Liver/metabolism , Male , Phytosterols/blood , Rabbits , Rapeseed Oil , Sitosterols/blood
9.
Proc Natl Acad Sci U S A ; 101(23): 8797-802, 2004 Jun 08.
Article in English | MEDLINE | ID: mdl-15169957

ABSTRACT

The pathogenic mechanisms by which physical exercise influences atherosclerotic lesion formation remain poorly understood. Because vigorous physical training increases oxidative stress, this study tested the hypothesis that graduated and moderate physical exercise together with metabolic intervention (l-arginine and antioxidants) may contribute to increased vascular protection. Exercise training in mice was induced by graduated swimming. In hypercholesterolemic male mice on an atherogenic high-cholesterol diet, graduated and moderate exercise lowered plasma cholesterol and decreased atherosclerotic lesions compared with sedentary control mice. Antioxidants (1.0% vitamin E added to the chow and 0.05% vitamin C added to the drinking water) and l-arginine (6% in drinking water) supplementation to exercising hypercholesterolemic mice further and synergistically reduced atherosclerosis compared with untreated exercised mice. Arterial oxidation-specific epitopes and systemic oxidative stress were reduced by metabolic intervention. Graduated chronic exercise elicited an increase in production of nitric oxide through increased endothelial nitric oxide synthase expression and ameliorated scavenger activities. Thus, metabolic intervention with l-arginine and antioxidants together with graduated and moderate exercise training reduce atherosclerotic lesion formation.


Subject(s)
Antioxidants/therapeutic use , Arteriosclerosis/prevention & control , Hyperlipoproteinemia Type II/complications , Hyperlipoproteinemia Type II/therapy , Physical Conditioning, Animal , Animals , Arginine/therapeutic use , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Ascorbic Acid/therapeutic use , Diet, Atherogenic , Hyperlipoproteinemia Type II/metabolism , Male , Mice , Mice, Knockout , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Oxidative Stress , Receptors, LDL/deficiency , Receptors, LDL/genetics , Vitamin E/therapeutic use
10.
Nucl Med Rev Cent East Eur ; 6(1): 35-9, 2003.
Article in English | MEDLINE | ID: mdl-14600931

ABSTRACT

BACKGROUND: The hypoglycemic effect of prickly pear is well known by native local Indian population since a long time. Beside the beneficial effects on lipid metabolism, oxidation injury and platelet function has been claimed in experimental animals. We recently found an upregulation of apo-B/E receptor. MATERIAL AND METHODS: We therefore examined 10 patients with isolated heterozygous familial hypercholesterolemia (FH) being enrolled in a dietary run-in phase of 6 weeks after dietary counselling and a further 6 weeks of prickly pear addition. Uptake of autologous (123)I-radiolabeled LDL was determined at entry as well as after 6 weeks of daily prickly pear ingestion. RESULTS: We found a significant (p < 0.0001) increase in LDL-uptake by the liver (24.5 +/- 4.9 vs. 31.1 +/- 5.2%) and an enhanced decay in circulating blood. Total (298.0 --> 268.0 mg/dl; p < 0.0001) and LDL-cholesterol (210.5 --> 176.4 mg/dl; p = 0.0001) were significantly affected, while HDL (p = 0.0629) and triglycerides were not. CONCLUSIONS: These findings demonstrate a significant upregulation of (123)I-LDL binding by prickly pear in FH-patients invivo and indicate that prickly pear exerts a significant hypolipidemic action via receptor upregulation.


Subject(s)
Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/metabolism , Lipoproteins, LDL/metabolism , Liver/drug effects , Liver/metabolism , Opuntia/chemistry , Phytotherapy/methods , Plant Extracts/therapeutic use , Adult , Female , Humans , Hyperlipoproteinemia Type II/diagnostic imaging , Hyperlipoproteinemia Type II/diet therapy , Iodine Radioisotopes/pharmacokinetics , Liver/diagnostic imaging , Male , Metabolic Clearance Rate , Middle Aged , Protein Binding , Radionuclide Imaging , Radiopharmaceuticals , Treatment Outcome , Up-Regulation
11.
Arterioscler Thromb Vasc Biol ; 23(11): 2078-82, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14512370

ABSTRACT

OBJECTIVE: The combination of LDL apheresis with high doses of a potent hepatic hydroxymethylglutaryl coenzyme A reductase inhibitor, such as atorvastatin, has been the best therapy available for the prevention of cardiovascular disease in patients with homozygous familial hypercholesterolemia (HFH). However, some concerns have been made about the effect of atorvastatin on HDL cholesterol levels in these patients. METHODS AND RESULTS: HDL cholesterol levels were determined bimonthly over the course of 2 years of treatment with high-dose atorvastatin in genotypically defined HFH patients either receptor-defective (n=6) or receptor-negative (n=6) under long-term treatment with LDL apheresis. We additionally stratified the atorvastatin effect on HDL cholesterol according to the genotype as an indicator of residual in vivo LDL receptor activity. Our findings indicate that (1) an early and transitory reduction of plasma HDL cholesterol levels occurs during the first 4 weeks of atorvastatin treatment; (2) the degree of the transient HDL reduction is higher in receptor-negative than in receptor-defective patients (-21+/-11 versus -10+/-4%; P=0.01); and (3) after long-term treatment, HDL cholesterol concentration remains higher in receptor-defective than receptor-negative patients (P=0.026). CONCLUSIONS: The present study reveals that HDL cholesterol reduction after high-dose atorvastatin is an early and transient event in HFH patients which magnitude depends on the presence of a residual LDL-R activity.


Subject(s)
Cholesterol, HDL/genetics , Cholesterol, HDL/metabolism , Heptanoic Acids/administration & dosage , Hyperlipoproteinemia Type II/drug therapy , Hyperlipoproteinemia Type II/genetics , Pyrroles/administration & dosage , Receptors, LDL/metabolism , Adolescent , Adult , Atorvastatin , Blood Component Removal , Child , Cholesterol, HDL/drug effects , Female , Heterozygote , Homozygote , Humans , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type II/therapy , Male , Mutation , Phenotype , Receptors, LDL/genetics
12.
Bull Exp Biol Med ; 134(3): 230-2, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12511988

ABSTRACT

The development of type IIa alimentary hyperlipoproteinemia in rats was accompanied by changes in activity of glutathione-dependent enzymes, exhaustion of the reserves of antioxidant vitamins in the liver and blood, and intensification of lipid peroxidation. The hepatoprotective preparation Maksar obtained from far-eastern plants Maachia amurensis and containing natural biological antioxidants normalized blood lipid composition, corrected liver lipidosis, and improved the antioxidant defense system.


Subject(s)
Antioxidants/pharmacology , Flavonoids , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type II/therapy , Phenols/pharmacology , Plant Extracts/pharmacology , Polymers/pharmacology , Animals , Disease Models, Animal , Glutathione/metabolism , Lipid Peroxidation , Lipids/blood , Liver/metabolism , Male , Plant Extracts/chemistry , Polyphenols , Rats , Rats, Wistar
13.
Arterioscler Thromb Vasc Biol ; 20(11): 2422-7, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073847

ABSTRACT

Familial hypercholesterolemia (FH) and disturbances in postprandial lipoprotein metabolism are both associated with premature atherosclerosis. The effect of beta-hydroxy-beta-methylglutaryl coenzyme A reductase inhibitors on plasma cholesterol levels in patients with FH is well established; however, it is not known whether postprandial lipoproteins are also influenced. In this case-controlled intervention study, we investigated the effects of high-dose simvastatin on postprandial lipoproteins. We used a new method to analyze remnant lipoproteins based on the immunoseparation principle (remnant-like particle cholesterol [RLP-C] assay) and the well-established measurement of retinyl ester (RE) analysis in plasma and in the Svedberg flotation unit (Sf)<1000 fraction. Seven heterozygous FH patients and 7 control subjects matched for sex, age, body mass index, triglycerides, and apolipoprotein E genotype were enrolled in the study. An oral vitamin A (RE) fat-loading test was performed at baseline in both groups and after 3 months of high-dose simvastatin (80 mg/d) treatment in the FH patients. Before treatment, FH patients had significantly higher fasting and postprandial concentrations of lipoprotein remnants (plasma RLP-C 42+/-19 mg/dL and area under the RLP-C curve 415+/-82 mg. L(-1). h(-1), respectively) than did control subjects (7+/-3 mg/dL and 101+/-35 mg. L( -1). h(-1), respectively; P<0.05), suggesting a delayed clearance of chylomicron remnant particles in the FH patients. Treatment with simvastatin significantly reduced fasting and postprandial remnant lipoprotein cholesterol concentrations (13+/-3 mg/dL and 136+/-53 mg. L(-1). h(-1), respectively; P<0.05 for both). Postprandial RE in the Sf<1000 fraction, not total RE in plasma, was also significantly higher in FH patients than in control subjects (24+/-10 versus 6.3+/-5.9 mg. L( -1). h(-1), P<0.05), but treatment with simvastatin did not result in improvement of the postprandial RE response, either in the Sf<1000 fraction or in plasma. It is concluded that heterozygous FH patients have increased fasting and postprandial remnant lipoprotein concentrations. Treatment with simvastatin significantly reduced the fasting and postprandial RLP-C concentrations but did not result in improved postprandial RE response.


Subject(s)
Apolipoproteins/metabolism , Cholesterol , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/metabolism , Lipoproteins/metabolism , Postprandial Period/drug effects , Simvastatin/administration & dosage , Triglycerides/metabolism , Apolipoproteins B/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Genetic Carrier Screening , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Hyperlipoproteinemia Type II/genetics , Lipoproteins/blood , Male , Middle Aged , Retinol-Binding Proteins , Retinol-Binding Proteins, Plasma , Retinyl Esters , Simvastatin/blood
14.
Cardiovasc Res ; 42(3): 752-60, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10533616

ABSTRACT

OBJECTIVE: Dihydropyridine calcium antagonists have been shown to retard atherogenesis in animal models and to prevent the development of early angiographic lesions in human coronary arteries. Endothelial dysfunction is an early event in the pathogenesis of cardiovascular disease. We investigated whether nifedipine could improve endothelial function in hypercholesterolemia, independently of changes in blood pressure or plasma lipids. METHODS: First, we compared in vivo forearm vascular responses to the endothelium-dependent and independent vasodilators serotonin (5-HT) and sodium nitroprusside (SNP) in 11 patients with familial hypercholesterolemia before and after 6-weeks treatment with nifedipine GITS (60 mg, OD) and in 12 matched controls. In a subgroup of six control subjects forearm vascular function was also assessed before and after 6-weeks nifedipine GITS treatment. In vitro, we subsequently explored possible mechanisms underlying the effect of nifedipine on endothelial function. We investigated the effects of nifedipine on both NO production by recombinant endothelial NO synthase (eNOS) and endothelial cells, using 3H-arginine conversion, as well as on superoxide generation by endothelial cell lysates, using lucigenin enhanced chemiluminescence. RESULTS: In hypercholesterolemia 5-HT-induced vasodilation was impaired (47 +/- 9% increase in forearm bloodflow vs. 99 +/- 8% in controls). Treatment with nifedipine completely restored 5-HT-induced vasodilation (113 +/- 13%), whereas it did not influence basal forearm vasomotion or SNP-induced vasodilation. Nifedipine did not alter forearm vascular responses in control subjects and did not alter blood pressure or plasma lipids. In vitro, we found no direct effect of nifedipine on NO production by recombinant eNOS or endothelial cells. However, we did observe a reduction in endothelial superoxide generation. CONCLUSIONS: Our data show that nifedipine improves endothelial function in hypercholesterolemia. It is suggested from our in vitro experiments that this effect is due to reduced NO degradation.


Subject(s)
Calcium Channel Blockers/therapeutic use , Endothelium, Vascular/drug effects , Hyperlipoproteinemia Type II/drug therapy , Nifedipine/therapeutic use , Adult , Case-Control Studies , Cells, Cultured , Endothelium, Vascular/metabolism , Female , Humans , Hyperlipoproteinemia Type II/metabolism , Male , Middle Aged , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Nitroprusside/pharmacology , Recombinant Proteins/metabolism , Regional Blood Flow/drug effects , Serotonin/pharmacology , Time Factors , Vasodilator Agents/pharmacology
15.
J Lipid Res ; 40(2): 213-20, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9925649

ABSTRACT

Screening for structural alterations of the low density lipoprotein (LDL) receptor gene by Southern blot analysis revealed an abnormal band pattern in one subject with a clinical diagnosis of homozygous familial hypercholesterolemia (FH). The molecular defect was further characterized by polymerase chain reaction and cDNA sequencing. These analyses identified a 4.8 kb in-frame deletion of exons 2 and 3, where exon 1 was spliced to exon 4. This deletion is expected to produce a receptor that has lost the two first cysteine-rich repeats of the ligand-binding domain. Previously published data of in vitro site-directed mutagenesis has shown that binding of LDL to such a receptor is reduced to 70% of normal. A mild phenotype in our FH homozygote is consistent with that observation. In contrast, heterozygotes carrying this deletion have a relatively more severe phenotype that is comparable to that of heterozygotes carrying a null-allele. A severe phenotype was also found in a compound heterozygote carrying this deletion. Possible mechanisms for this phenotypic variability are discussed.-Rødningen, O. K., S. Tonstad, J. D. Medh, D. A. Chappell, L. Ose, and T. P. Leren. Phenotypic consequences of a deletion of exons 2 and 3 of the LDL receptor gene.


Subject(s)
Exons/genetics , Gene Deletion , Hyperlipoproteinemia Type II/genetics , Receptors, LDL/genetics , Base Sequence , Blotting, Northern , Blotting, Southern , Cells, Cultured , DNA, Complementary/genetics , Fibroblasts/metabolism , Genotype , Humans , Hyperlipoproteinemia Type II/metabolism , Iodine Radioisotopes , Lipoproteins, LDL/blood , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/blood , Lipoproteins, VLDL/metabolism , Pedigree , Phenotype , Point Mutation , Polymerase Chain Reaction/methods , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
16.
J Biochem ; 116(4): 747-51, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7883748

ABSTRACT

In an effort to characterize mRNAs that are highly expressed during atherosclerosis, we employed differential hybridization screening of a cDNA library constructed from total RNA derived from the aorta of Watanabe heritable hyperlipidemic (WHHL) rabbits. Characterizing the cDNAs for mRNAs that are present in large amounts in WHHL rabbit aortae, we identified a positive clone encoding matrix gamma-carboxyglutamic acid protein (MGP). The primary structure of rabbit MGP was deduced from nucleotide sequence analysis of the cDNA. Northern blot analysis of total RNA prepared from aortae of WHHL and normal rabbits of various ages indicated that the expression of MGP mRNA increased in proportion to the progression of atherosclerosis in WHHL rabbits. Analysis of MGP mRNA by in situ hybridization revealed that a significant amount of MGP mRNA is accumulated in atherosclerotic lesions of WHHL rabbits, suggesting that the expression of MGP mRNA is correlated with the progression of atherosclerosis.


Subject(s)
Aorta/metabolism , Arteriosclerosis/metabolism , Calcium-Binding Proteins/biosynthesis , Extracellular Matrix Proteins , Hyperlipoproteinemia Type II/metabolism , Amino Acid Sequence , Animals , Arteriosclerosis/genetics , Arteriosclerosis/physiopathology , Base Sequence , Blotting, Northern , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/physiology , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Disease Progression , Gene Expression , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/physiopathology , In Situ Hybridization , Male , Molecular Sequence Data , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Sequence Homology, Amino Acid , Matrix Gla Protein
17.
Vopr Pitan ; (5): 33-6, 1994.
Article in Russian | MEDLINE | ID: mdl-7871774

ABSTRACT

Effect of antiatherosclerotic diet of 112 patients with cardioishemic disease IIa, IIb, and IV types was observed. Positive dynamics of clinical picture of disease, values of lipid metabolism and T-cell cents immunity were shown, particularly for patients with hyperlipidemy, which developed as result of effect of medium cents and genetic cents factors. Including to antiatherosclerosis diet polyunsaturated fatty acids of family (w-3) intensified hypolipidemic effect.


Subject(s)
Diet, Atherogenic , Fatty Acids, Omega-3/administration & dosage , Hyperlipoproteinemia Type II/diet therapy , Hypertension/diet therapy , Myocardial Ischemia/diet therapy , Adult , Aged , Female , Humans , Hyperlipoproteinemia Type II/immunology , Hyperlipoproteinemia Type II/metabolism , Hypertension/immunology , Hypertension/metabolism , Male , Middle Aged , Myocardial Ischemia/immunology , Myocardial Ischemia/metabolism , T-Lymphocytes/immunology
18.
Biochim Biophys Acta ; 958(3): 352-60, 1988 Feb 19.
Article in English | MEDLINE | ID: mdl-2963664

ABSTRACT

Fibroblasts from patients with homozygous familial hypercholesterolemia (FH), a disease characterized by accelerated atherogenesis, are known to lack functional low-density-lipoprotein receptors, which ultimately results in increased cholesterol biosynthesis in the cultured cells. [14C]Acetate incorporation in these cells was compared to that of normal fibroblasts and to fibroblasts from patients with Down's syndrome, a disease in which atherosclerosis is rare. Total [14C]acetate incorporation did not differ significantly between normal and Down's fibroblasts, nor did its partitioning into the hexane-extractable and aqueous fractions of the cell hydrolysates. [14C]Acetate incorporation was much greater in FH cells in both the aqueous and hexane-extractable fractions. Preincubation in fetal bovine serum increased acetate incorporation only by FH cells, while 50 micrograms low-density lipoprotein/ml medium depressed acetate incorporation in all three groups. A C27 sterol, identified by gas chromatography-mass spectrometry as a probable isomer of cholesterol, was present in small amounts in FH fibroblasts, but was not detectable in the normal or Down's cells. The absolute amounts of [14C]acetate incorporated into the non-sterol lipids were greater in the FH fibroblasts, indicating that these cells may have to synthesize, in addition to cholesterol, other required cellular lipids which are delivered to the normal cells by low-density lipoproteins.


Subject(s)
Acetates/metabolism , Down Syndrome/metabolism , Fibroblasts/metabolism , Hyperlipoproteinemia Type II/metabolism , Acetic Acid , Carbon Radioisotopes , Cells, Cultured , Cholesterol/biosynthesis , Fibroblasts/drug effects , Gas Chromatography-Mass Spectrometry , Humans , Insulin/pharmacology , Leucine/metabolism , Protein Biosynthesis , Receptors, LDL/metabolism , Selenium/pharmacology , Transferrin/pharmacology
19.
Vopr Pitan ; (2): 39-43, 1986.
Article in Russian | MEDLINE | ID: mdl-3705542

ABSTRACT

The effect of diets containing soybean proteins substituting a significant portion of meat proteins was studied in patients with hyperlipidemia, type IIa, and in experiments on rats. It was found that soybean proteins included into the diet induced blood plasma amino acid imbalance, changed the excretion of some amino acids with urine, and significantly diminished the blood plasma cholesterol level. In the experiments on rats a reduced rate was noted in the low-density lipoprotein apoprotein formation and high-density lipoprotein apoprotein regeneration in the blood plasma. It is suggested that the hypocholesterolemic effect of soybean proteins is caused by decreased apoprotein E synthesis and metabolism due to low levels of blood plasma lysine and arginine.


Subject(s)
Anticholesteremic Agents , Dietary Proteins/therapeutic use , Glycine max , Plant Proteins/therapeutic use , Adult , Amino Acids/analysis , Animals , Apolipoproteins/blood , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Coronary Disease/diet therapy , Coronary Disease/metabolism , Drug Evaluation , Drug Evaluation, Preclinical , Half-Life , Humans , Hyperlipoproteinemia Type II/diet therapy , Hyperlipoproteinemia Type II/metabolism , Male , Middle Aged , Rats , Rats, Inbred Strains
20.
J Nutr ; 113(2): 328-36, 1983 Feb.
Article in English | MEDLINE | ID: mdl-6822906

ABSTRACT

Two experiments were conducted to determine the effect of dietary manganese on cholesterol and lipid metabolism in the Wistar rat and the genetically hypercholesterolemic RICO rat. Weanling animals were placed on a manganese-deficient (0.12 microgram/g) and a supplemented diet (100.12 micrograms/g). Mean body weights, hepatic fatty acid synthesis and liver manganese concentration significantly decreased in the deficient group of Wistar rats. Plasma cholesterol, VLDL (very low density lipoprotein) and HDL (high-density lipoprotein) cholesterol, hepatic cholesterol synthesis, liver cholesterol and lipid concentrations were not significantly affected by manganese deficiency. Mean body weights and hepatic manganese content were lower in the manganese-deficient group in both normal and hypercholesterolemic RICO rats. Manganese deficiency significantly decreased LDL cholesterol concentration in the hypercholesterolemic RICO rats. Manganese deficiency had no significant effect on hepatic cholesterol and fatty acid synthesis, plasma cholesterol, VLDL and HDL cholesterol concentrations, liver lipid and liver cholesterol concentration in either group of RICO rats. These results indicate that dietary manganese deficiency does not result in significant alterations in cholesterol and lipid metabolism in the rat.


Subject(s)
Cholesterol/metabolism , Diet , Hyperlipoproteinemia Type II/metabolism , Lipid Metabolism , Manganese/deficiency , Animals , Female , Lipoproteins/blood , Liver/metabolism , Rats , Rats, Inbred Strains/growth & development , Rats, Inbred Strains/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL