Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 23(3): 475-82, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12615696

ABSTRACT

OBJECTIVE: Peroxisome proliferator-activated receptor gamma (PPARgamma), a ligand-activated transcription factor, has pleiotropic effects, including regulation of macrophage differentiation and lipid homeostasis. The PPARgamma ligands, thiazolidinediones (TZDs), attenuate atherosclerosis in mice by uncertain mechanisms. The objective of this study was to determine whether activation of PPARgamma or its obligate heterodimer, retinoid X receptor (RXR), modulates macrophage foam cell formation induced by oxidized (ox) lipoproteins. METHODS AND RESULTS: Incubation of THP-1 macrophages with oxHTG-VLDL, oxREM, or oxLDL increased cellular cholesteryl ester over 6-fold. Preincubation with the TZD, ciglitazone, the RXR-specific ligand, 9-cis retinoic acid (9cRA) or the combination reduced CE mass accumulation by up to 65%. Ciglitazone and 9cRA increased CD36 mRNA (up to 4-fold); however, uptake of [125I]oxLDL was only modestly enhanced (up to 1.8-fold) becaues of a concomitant PPARgamma:RXR-induced decrease in SRAI/II activity (up to 40%). This suggested that PPARgamma:RXR activation inhibited cholesteryl ester accumulation by enhancing cholesterol efflux. Ciglitazone and 9cRA were found to increase the expression of ATP-binding cassette proteins A1 and G1, resulting in enhanced cholesterol efflux to lipoprotein-deficient serum, apoAI and HDL3. CONCLUSIONS: PPARgamma and/or RXR activation inhibit foam cell formation through enhanced cholesterol efflux despite increased oxLDL uptake. These observations explain the reduced atherosclerosis in TZD-treated mice and may extend the therapeutic implications of these ligands.


Subject(s)
Arteriosclerosis/metabolism , Cholesterol/metabolism , Lipoproteins/metabolism , Macrophages/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Humans , Membrane Transport Proteins/metabolism , Mice , Oxidation-Reduction , Retinoid X Receptors
2.
Arterioscler Thromb Vasc Biol ; 21(12): 2011-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11742878

ABSTRACT

Transforming growth factor beta1 (TGF-beta1) is secreted by various cells, including macrophages, smooth muscle cells, and endothelial cells. TGF-beta1 is present in atherosclerotic lesions, but its role in regulating macrophage foam cell formation is not understood. Hypertriglyceridemic very low density lipoprotein (VLDL) remnants (VLDL-REMs) in their native or oxidized form will induce cholesteryl ester (CE) and triglyceride (TG) accumulation in macrophages. Therefore, we examined whether TGF-beta1 can modulate the macrophage uptake of native or oxidized VLDL-REMs (oxVLDL-REMs). Incubation of J774A.1 macrophages with VLDL-REMs and oxVLDL-REMs compared with control cells increased cellular CE (13- and 21-fold, respectively) and TG mass (21-and 18-fold, respectively). Preincubation with TGF-beta1 before incubation with VLDL-REMs or oxVLDL-REMs significantly decreased CE (73% and 54%, respectively) and TG mass (42% and 41%, respectively). TGF-beta1 inhibited the activity and expression of 2 key components needed for VLDL-REM uptake: lipoprotein lipase and low density lipoprotein receptor. TGF-beta1 inhibited CE mass induced by oxVLDL-REMs in part by decreasing the expression of scavenger receptor type AI/II and CD36. Furthermore, TGF-beta1 enhanced cholesterol efflux through upregulation of the ATP-binding cassette (ABC) transporters ABCA1 and ABCG1. Thus, TGF-beta1 inhibits macrophage foam cell formation induced by VLDL-REMs or oxVLDL-REMs, which suggests an antiatherogenic role for this cytokine.


Subject(s)
Arteriosclerosis/metabolism , Cholesterol Esters/metabolism , Lipoproteins, VLDL/metabolism , Macrophages/metabolism , Transforming Growth Factor beta/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Cells, Cultured , Down-Regulation , Humans , Lipid Metabolism , Lipoprotein Lipase/metabolism , Lipoproteins, LDL/metabolism , Mice , Oxidation-Reduction , RNA, Messenger/analysis , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Transforming Growth Factor beta1 , Up-Regulation
3.
J Clin Endocrinol Metab ; 86(6): 2747-51, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11397881

ABSTRACT

We discovered that rare mutations in LMNA, which encodes lamins A and C, underlie autosomal dominant Dunnigan-type familial partial lipodystrophy. Because familial partial lipodystrophy is an extreme example of genetically disturbed adipocyte differentiation, it is possible that common variation in LMNA is associated with obesity-related phenotypes. We subsequently discovered a common single nucleotide polymorphism (SNP) in LMNA, namely 1908C/T, which was associated with obesity-related traits in Canadian Oji-Cree. We now report association of this LMNA SNP with anthropometric indexes in 186 nondiabetic Canadian Inuit. We found that physical indexes of obesity, such as body mass index, waist circumference, waist to hip circumference ratio, subscapular skinfold thickness, and subscapular to triceps skinfold thickness ratio were each significantly higher among Inuit subjects with the LMNA 1908T allele than in subjects with the 1908C/1908C genotype. For each significantly associated obesity-related trait, the LMNA 1908C/T SNP genotype accounted for between approximately 10--100% of the attributable variation. The results indicate that common genetic variation in LMNA is an important determinant of obesity-related quantitative traits.


Subject(s)
Genetic Variation , Genome , Inuit/genetics , Laminin/genetics , Nuclear Proteins/genetics , Obesity/genetics , Obesity/pathology , Adult , Alleles , Body Mass Index , Female , Gene Frequency , Genotype , Humans , Lamins , Leptin/blood , Male , Middle Aged , Obesity/blood , Phenotype , Sex Characteristics , Skinfold Thickness
4.
J Lipid Res ; 42(5): 725-34, 2001 May.
Article in English | MEDLINE | ID: mdl-11352979

ABSTRACT

The citrus flavonoids, naringenin and hesperetin, lower plasma cholesterol in vivo. However, the underlying mechanisms are not fully understood. The ability of these flavonoids to modulate apolipoprotein B (apoB) secretion and cellular cholesterol homeostasis was determined in the human hepatoma cell line, HepG2. apoB accumulation in the media decreased in a dose-dependent manner following 24-h incubations with naringenin (up to 82%, P < 0.00001) or hesperetin (up to 74%, P < 0.002). Decreased apoB secretion was associated with reduced cellular cholesteryl ester mass. Cholesterol esterification was decreased, dose-dependently, up to 84% (P < 0.0001) at flavonoid concentrations of 200 microM. Neither flavonoid demonstrated selective inhibition of either form of acyl CoA:cholesterol acyltransferase (ACAT) as determined using CHO cells stably transfected with either ACAT1 or ACAT2. However, in HepG2 cells, ACAT2 mRNA was selectively decreased (- 50%, P < 0.001) by both flavonoids, whereas ACAT1 mRNA was unaffected. In addition, naringenin and hesperetin decreased both the activity (- 20% to - 40%, P < 0.00004) and expression (- 30% to - 40%, P < 0.02) of microsomal triglyceride transfer protein (MTP). Both flavonoids caused a 5- to 7-fold increase (P < 0.02) in low density lipoprotein (LDL) receptor mRNA, which resulted in a 1.5- to 2-fold increase in uptake and degradation of (125)I-LDL. We conclude that both naringenin and hesperetin decrease the availability of lipids for assembly of apoB-containing lipoproteins, an effect mediated by 1) reduced activities of ACAT1 and ACAT2, 2) a selective decrease in ACAT2 expression, and 3) reduced MTP activity. Together with an enhanced expression of the LDL receptor, these mechanisms may explain the hypocholesterolemic properties of the citrus flavonoids.


Subject(s)
Apolipoproteins B/metabolism , Carrier Proteins/metabolism , Citrus/chemistry , Flavanones , Flavonoids/pharmacology , Hepatocytes/metabolism , Hesperidin , Sterol O-Acyltransferase/metabolism , Anticholesteremic Agents/pharmacology , Carcinoma, Hepatocellular , Carrier Proteins/genetics , Cholesterol/metabolism , Cholesterol Esters/metabolism , Enzyme-Linked Immunosorbent Assay , Hepatocytes/drug effects , Humans , Receptors, LDL/metabolism , Sterol O-Acyltransferase/chemistry , Triglycerides/metabolism , Tumor Cells, Cultured , Sterol O-Acyltransferase 2
5.
J Clin Endocrinol Metab ; 85(9): 3089-93, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10999791

ABSTRACT

Mutations in LMNA, which encodes lamins A and C, have been found in patients with autosomal dominant Dunnigan-type familial partial lipodystrophy (FPLD). We analyzed the relationship between plasma leptin and the rare LMNA R482Q mutation in 23 adult FPLD subjects compared with 25 adult family controls with normal LMNA in an extended Canadian FPLD kindred. We found that the LMNA Q482/R482 genotype was a significant determinant of plasma leptin, the ratio of plasma leptin to body mass index (BMI), plasma insulin, and plasma C peptide (P= 0.015, P = 0.0007, P = 0.0004, and P < 0.0001, respectively), but not BMI (P = 0.67). Family members who were heterozygous for LMNA Q482/R482 had significantly lower plasma leptin and leptin:BMI ratio than unaffected R482/R482 homozygotes. Fasting plasma concentrations of insulin and C peptide were both significantly higher in LMNA Q482/R482 heterozygotes than in R482/R482 homozygotes. Multivariate regression analysis revealed that the LMNA R482Q genotype accounted for 40.9%, 48.2%, 86.9%, and 81.0%, respectively, of the attributable variation in log leptin, leptin:BMI ratio, log insulin, and log C peptide (P = 0.013, P = 0.0007, P = 0.0002 and P < 0.0001, respectively). The results indicate that a rare FPLD mutation in LMNA determines the plasma leptin concentration. It remains to be established whether the reduction in leptin results from the reduced adipose tissue mass in FPLD or from another subcellular effect of mutant LMNA. It also remains to be established whether the insulin resistance in FPLD is a consequence of the reduced plasma leptin or of another functional change resulting from mutant LMNA.


Subject(s)
Leptin/blood , Lipodystrophy/genetics , Mutation/genetics , Nuclear Proteins/genetics , Adult , Alleles , Body Mass Index , C-Peptide/blood , Female , Genotype , Humans , Insulin/blood , Lamins , Leptin/deficiency , Lipodystrophy/blood , Male , Multivariate Analysis
6.
Am J Hypertens ; 13(5 Pt 1): 518-22, 2000 May.
Article in English | MEDLINE | ID: mdl-10826403

ABSTRACT

Little is known about the effects of antihypertensive drugs on hemodynamic responses to mental stress. We studied 24 patients with mild-to-moderate hypertension in a double-blind random-sequence crossover study comparing placebo with amlodipine titrated up from 5 to 10 mg daily. After 1 month of treatment, the subjects performed 20 min of a frustrating cognitive task. At baseline before task, amlodipine significantly reduced systolic pressure (128.9+/-8.2 mm Hg v 140.3+/-10.7 mm Hg, P < .001), diastolic pressure (81.7+/-7.7 mm Hg v 90+/-7.5 mm Hg, P < .001), and total peripheral resistance (37.5+/-15 v 45.6+/-23.7 mm Hg/L/min, P < .05), while elevating baseline norepinephrine levels (2286+/-731 pmol/L v 1788+/-546 pmol/L, P < .001). Blood pressure during the stress task was significantly less with amlodipine than with placebo (systolic 142.3+/-12.3 mm Hg v 150.9+/-14.6 mm Hg, P < .001; diastolic 87.9+/-8.4 mm Hg v 97.7+/-9.3 mm Hg, P < .001), whereas norepinephrine was significantly higher (2754+/-1007 pmol/L v 1970+/-740 pmol/L, P < .001). There were no significant differences in cardiac output, plasma lipids or lipoproteins, or markers of platelet activation. Heart rate increased significantly during stress, but there was no significant difference between amlodipine and placebo either at baseline or during stress. Our conclusion is that amlodipine reduces blood pressure at baseline and during mental stress, but raises basal and stress-related plasma catecholamines. This finding may have implications for the recent controversy over the safety of calcium channel antagonists, and suggests the potential relevance of combining amlodipine with adrenergic blockers.


Subject(s)
Amlodipine/therapeutic use , Calcium Channel Blockers/therapeutic use , Endocrine System/metabolism , Hemodynamics/drug effects , Norepinephrine/blood , Stress, Psychological/physiopathology , Adolescent , Adult , Aged , Biomarkers/blood , Chromatography, High Pressure Liquid , Cross-Over Studies , Double-Blind Method , Endocrine System/drug effects , Female , Humans , Hypertension/blood , Hypertension/drug therapy , Hypertension/physiopathology , Lipids/blood , Male , Middle Aged , Stress, Psychological/blood , Stress, Psychological/drug therapy
7.
J Lipid Res ; 41(3): 368-75, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10706584

ABSTRACT

The effects of continuously administering both conjugated equine estrogens (CEE) and micronized progesterone (MP) on the concentration, composition, production and catabolism of very low density (VLDL) and low density lipoproteins (LDL) have not previously been reported. The mechanism of the hormonally induced reductions of plasma LDL cholesterol of S(f) 0;-20 (mean 16%, P < 0.005) and LDL apoB (mean 6%, P < 0.025) were investigated by studying the kinetics of VLDL and LDL apolipoprotein (apo) B turnover after injecting autologous (131)I-labeled VLDL and (125)I-labeled LDL into each of the 6 moderately hypercholesterolemic postmenopausal subjects under control conditions and again in the fourth week of a 7-week course of therapy (0.625 mg/d of CEE + 200 mg/d of MP). The combined hormones significantly lowered plasma LDL apoB by increasing the mean fractional catabolic rate of LDL apoB by 20% (0. 32 vs. 0.27 pools/d, P < 0.03). Treatment also induced a significant increase in IDL production (6.3 vs. 3.7 mg/kg/d, P = 0.028). However, this did not result in an increase in LDL production because of an increase in IDL apoB direct catabolism (mean 102%, P = 0.033). VLDL kinetic parameters were unchanged and the concentrations of plasma total triglycerides (TG), VLDL-TG, VLDL-apoB did not rise as often seen with estrogen alone. Plasma HDL-cholesterol rose significantly (P < 0.02). Our major conclusion is that increased fractional catabolism of LDL underlies the LDL-lowering effect of the combined hormones.


Subject(s)
Estrogens/therapeutic use , Hormone Replacement Therapy , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/metabolism , Postmenopause , Progesterone/administration & dosage , Aged , Female , Humans , Kinetics , Middle Aged
8.
Clin Chim Acta ; 286(1-2): 231-42, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10511295

ABSTRACT

Acyl coenzyme A: cholesterol acyltransferase (ACAT) is postulated to play a role in hepatic and intestinal lipoprotein secretion. There is accumulating evidence, both in vitro and in vivo, that cholesterol and/or cholesteryl ester availability can regulate hepatic VLDL secretion. How ACAT inhibition regulates the assembly and secretion of apolipoprotein (apo) B containing lipoproteins within the hepatocyte has not been clearly established. ApoB kinetic studies performed in animals indicate that reduction in VLDL apoB secretion is an important mechanism whereby ACAT inhibitors decrease the plasma concentrations of these lipoproteins. However, in cultured hepatocytes, the effect of ACAT inhibition on apoB secretion has been inconsistent. Recent evidence has suggested the existence of more than one ACAT enzyme in mammals, which has culminated in the recent cloning of ACAT2. ACAT1 and ACAT2 respond differently to ACAT inhibitors of differing structures and classes. ACAT2 is present in the liver and intestine, the sites of apoB containing lipoprotein secretion and may represent the enzyme responsible for generating cholesteryl esters destined for lipoprotein assembly and secretion.


Subject(s)
Apolipoproteins B/metabolism , Liver/metabolism , Sterol O-Acyltransferase/antagonists & inhibitors , Animals , Cholesterol Esters/metabolism , Enzyme Inhibitors/pharmacology , Sterol O-Acyltransferase/genetics
9.
Endocrinology ; 140(11): 5293-302, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537160

ABSTRACT

It has been postulated that the rate of hepatic very low density lipoprotein (VLDL) apolipoprotein (apo) B secretion is dependent upon the activity of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase. To test this hypothesis in vivo, apoB kinetic studies were carried out in miniature pigs before and after 21 days treatment with high-dose (10 mg/kg/day), atorvastatin (A) or simvastatin (S) (n = 5). Pigs were fed a diet containing fat (34% of calories) and cholesterol (400 mg/day; 0.1%). Statin treatment decreased plasma total cholesterol [31 (A) vs. 20% (S)] and low density lipoprotein (LDL) cholesterol concentrations [42 (A) vs. 24% (S)]. Significant reductions in plasma total triglyceride (46%) and VLDL triglyceride (50%) concentrations were only observed with (A). Autologous [131I]VLDL, [125I]LDL, and [3H]leucine were injected simultaneously, and apoB kinetic parameters were determined by triple-isotope multicompartmental analysis using SAAM II. Statin treatment decreased the VLDL apoB pool size [49 (A) vs. 24% (S)] and the hepatic VLDL apoB secretion rate [50 (A) vs. 33% (S)], with no change in the fractional catabolic rate (FCR). LDL apoB pool size decreased [39 (A) vs. 26% (S)], due to reductions in both the total LDL apoB production rate [30 (A) vs. 21% (S)] and LDL direct synthesis [32 (A) vs. 23% (S)]. A significant increase in the LDL apoB FCR (15%) was only seen with (A). Neither plasma VLDL nor LDL lipoprotein compositions were significantly altered. Hepatic HMG-CoA reductase was inhibited to a greater extent with (A), when compared with (S), as evidenced by 1) a greater induction in hepatic mRNA abundances for HMG-CoA reductase (105%) and the LDL receptor (40%) (both P < 0.05); and 2) a greater decrease in hepatic free (9%) and esterified cholesterol (25%) (both P < 0.05). We conclude that both (A) and (S) decrease hepatic VLDL apoB secretion, in vivo, but that the magnitude is determined by the extent of HMG-CoA reductase inhibition.


Subject(s)
Apolipoproteins B/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins, VLDL/metabolism , Liver/metabolism , Animals , Atorvastatin , Cholesterol/blood , Cholesterol, LDL/blood , Heptanoic Acids/pharmacology , Kinetics , Lipoproteins/blood , Lipoproteins, IDL , Lipoproteins, LDL/administration & dosage , Lipoproteins, LDL/blood , Lipoproteins, VLDL/administration & dosage , Lipoproteins, VLDL/blood , Liver/enzymology , Microsomes, Liver/enzymology , Pyrroles/pharmacology , Simvastatin/pharmacology , Swine , Swine, Miniature , Triglycerides/blood
10.
J Lipid Res ; 40(7): 1317-27, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10393217

ABSTRACT

An orally bioavailable acyl coenzyme A:cholesterol acyltransferase (ACAT) inhibitor, avasimibe (CI-1011), was used to test the hypothesis that inhibition of cholesterol esterification, in vivo, would reduce hepatic very low density (VLDL) apolipoprotein (apo) B secretion into plasma. ApoB kinetic studies were carried out in 10 control miniature pigs, and in 10 animals treated with avasimibe (10 mg/kg/d, n = 6; 25 mg/kg/d, n = 4). Pigs were fed a diet containing fat (34% of calories) and cholesterol (400 mg/d; 0.1%). Avasimibe decreased the plasma concentrations of total triglyceride, VLDL triglyceride, and VLDL cholesterol by 31;-40% 39-48%, and 31;-35%, respectively. Significant reductions in plasma total cholesterol (35%) and low density lipoprotein (LDL) cholesterol (51%) concentrations were observed only with high dose avasimibe. Autologous 131I-labeled VLDL, 125I-labeled LDL, and [3H]leucine were injected simultaneously into each pig and apoB kinetic data were analyzed using multicompartmental analysis (SAAM II). Avasimibe decreased the VLDL apoB pool size by 40;-43% and the hepatic secretion rate of VLDL apoB by 38;-41%, but did not alter its fractional catabolism. Avasimibe decreased the LDL apoB pool size by 13;-57%, largely due to a dose-dependent 25;-63% in the LDL apoB production rate. Hepatic LDL receptor mRNA abundances were unchanged, consistent with a marginal decrease in LDL apoB FCRs. Hepatic ACAT activity was decreased by 51% (P = 0.050) and 68% (P = 0.087) by low and high dose avasimibe, respectively. The decrease in total apoB secretion correlated with the decrease in hepatic ACAT activity (r = 0.495; P = 0.026). We conclude that inhibition of hepatic ACAT by avasimibe reduces both plasma VLDL and LDL apoB concentrations, primarily by decreasing apoB secretion.


Subject(s)
Acetates , Anticholesteremic Agents/pharmacology , Apolipoproteins B/biosynthesis , Enzyme Inhibitors/pharmacology , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/metabolism , Sterol O-Acyltransferase/antagonists & inhibitors , Sulfonic Acids/pharmacology , Swine, Miniature/metabolism , Acetamides , Animals , Female , Liver/drug effects , Liver/metabolism , Male , Sulfonamides , Swine
11.
J Lipid Res ; 40(6): 1017-28, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10357833

ABSTRACT

Hypertriglyceridemic (HTG) very low density lipoproteins (VLDL) from subjects with type IV hyperlipoproteinemia induce both cholesteryl ester (CE) and triglyceride (TG) accumulation in cultured J774 macrophages. We examined whether the cytokine interferon-gamma (IFN-gamma), which is expressed by lymphocytes in atherosclerotic lesions, would modulate macrophage uptake of HTG -VLDL. Incubation of cells with HTG -VLDL alone significantly increased cellular CE and TG mass 17- and 4.3-fold, respectively, while cellular free cholesterol (FC) was unaffected. Pre-incubation of cells with IFN-gamma (50 U/ml) prior to incubation with HTG -VLDL caused a marked enhancement in cellular CE and TG 27- and 6-fold over no additions (controls), respectively, and a 1.5-fold increase in FC. IFN-gamma increased low density lipoprotein (LDL)-induced cellular CE 2-fold compared to LDL alone. IFN-gamma did not enhance the uptake of type III (apoE2/E2) HTG -VLDL or VLDL from apoE knock-out mice. Incubations in the presence of a lipoprotein lipase (LPL) inhibitor or an acylCoA:cholesterol acyltransferase (ACAT) inhibitor demonstrated that the IFN-gamma-enhanced HTG -VLDL uptake was dependent on LPL and ACAT activities. IFN-gamma significantly increased the binding and degradation of 125I-labeled LDL. Binding studies with 125I-labeled alpha2-macroglobulin, a known LDL receptor-related protein (LRP) ligand, and experiments with copper-oxidized LDL indicated that the IFN-gamma-enhanced uptake was not due to increased expression of the LRP or scavenger receptors. Thus, IFN-gamma may promote foam cell formation by accelerating macrophage uptake of native lipoproteins. IFN-gamma-stimulated CE accumulation in the presence of HTG -VLDL occurs via a process that requires receptor binding-competent apoE and active LPL. IFN-gamma-enhanced uptake of both HTG -VLDL and LDL is mediated by the LDL-receptor and requires ACAT-mediated cholesterol esterification.


Subject(s)
Hypertriglyceridemia/blood , Interferon-gamma/pharmacology , Lipoproteins, VLDL/metabolism , Macrophages/metabolism , Animals , Apolipoproteins E/genetics , Cell Line , Cholesterol Esters/metabolism , Esterification , Humans , Lipoprotein Lipase/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Triglycerides/metabolism , alpha-Macroglobulins/metabolism
12.
J Lipid Res ; 40(6): 1078-89, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10357840

ABSTRACT

The concept that hepatic cholesterol synthesis regulates hepatocyte assembly and secretion of apoB-containing lipoproteins remains controversial. The present study was carried out in HepG2 cells to examine the regulation of apoB secretion by the HMG-CoA reductase inhibitor atorvastatin. ApoB accumulation in the media was decreased by 24% and 36% at 10 microm (P < 0.02) and 20 microm (P < 0.01) of atorvastatin, respectively. Atorvastatin inhibited HepG2 cell cholesterol synthesis by up to 96% (P < 0.001) and cellular cholesteryl ester (CE) mass by 54% (P < 0.001). Another HMG-CoA reductase inhibitor, simvastatin, decreased cellular cholesterol synthesis and CE mass by up to 96% (P < 0.001) and 52% (P < 0.001), respectively. However, in contrast to atorvastatin, simvastatin had no effect on apoB secretion. To characterize the reduction in apoB secretion by atorvastatin (10 microm), pulse-chase experiments were performed and the kinetic data were analyzed by multicompartmental modeling using SAAM II. Atorvastatin had no affect on the synthesis of apoB, however, apoB secretion into the media was decreased by 44% (P = 0.048). Intracellular apoB degradation increased proportionately (P = 0.048). Simvastatin (10 microm) treatment did not significantly alter either the secretion or intracellular degradation of apoB, relative to control. The kinetics of apoB degradation were best described by a rapidly and a slowly turning over degradation compartment. The effect of atorvastatin on apoB degradation was largely confined to the rapid compartment. Neither inhibitor affected apoB mRNA concentrations, however, both significantly increased LDL receptor and HMG-CoA reductase mRNA levels. Atorvastatin treatment also decreased the mRNA for the microsomal triglyceride transfer protein (MTP) by 22% (P < 0.02). These results show that atorvastatin decreases apoB secretion, by a mechanism that results in an enhanced intracellular degradation in apoB.


Subject(s)
Apolipoproteins B/metabolism , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver/metabolism , Pyrroles/pharmacology , Simvastatin/pharmacology , Apolipoproteins B/analysis , Apolipoproteins B/genetics , Atorvastatin , Carrier Proteins/genetics , Cell Line , Cholesterol Esters/metabolism , Hydroxymethylglutaryl CoA Reductases/genetics , Lipid Metabolism , Lipids/biosynthesis , Lipoproteins/analysis , Lipoproteins/metabolism , Liver/drug effects , RNA, Messenger/analysis , Receptors, LDL/genetics
13.
Arterioscler Thromb Vasc Biol ; 19(5): 1238-49, 1999 May.
Article in English | MEDLINE | ID: mdl-10323775

ABSTRACT

Very low density lipoproteins (VLDLs) from apolipoprotein (apo) E2/E2 subjects with type III hyperlipoproteinemia, VLDL remnants, and VLDL from apoE-knockout (EKO) mice are taken up poorly by macrophages. The present study examined whether VLDL modification by the reactive aldehyde p-hydroxyphenylacetaldehyde (pHA) enhances cholesteryl ester (CE) accumulation by J774A.1 macrophages. pHA is the major product derived from the oxidation of L-tyrosine by myeloperoxidase and is a component of human atherosclerotic lesions. Incubation of J774A.1 cells with native type III VLDL, their remnants, and EKO-VLDL increased cellular CE by only 3-, 5-, and 5-fold, respectively, compared with controls. In striking contrast, cells exposed to VLDL modified by purified pHA (pHA-VLDL) exhibited marked increases in cellular CE of 38-, 47-, and 35-fold, respectively (P95%, CE accumulation induced by copper-oxidized VLDL. These results demonstrate a novel mechanism for the conversion of type III VLDLs, their remnants, and EKO-VLDL into atherogenic particles and suggest that macrophage uptake of pHA-VLDL (1) requires catalytically active lipoprotein lipase, (2) involves acyl coenzyme A:cholesterol acyltransferase-mediated cholesterol esterification, and (3) involves pathways distinct from the SR-A.


Subject(s)
Acetaldehyde/analogs & derivatives , Apolipoproteins E/deficiency , Cholesterol Esters/metabolism , Hyperlipoproteinemia Type III/blood , Lipoproteins, VLDL/blood , Macrophages/metabolism , Membrane Proteins , Peroxidase/metabolism , Receptors, Lipoprotein , Acetaldehyde/pharmacology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Cell Line , Esterification , Humans , Hypochlorous Acid/metabolism , Interferon-gamma/pharmacology , Lipoprotein Lipase/antagonists & inhibitors , Lipoprotein Lipase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Phenol , Poly I/pharmacology , Receptors, Immunologic/metabolism , Receptors, Scavenger , Scavenger Receptors, Class A , Scavenger Receptors, Class B , Sterol O-Acyltransferase/metabolism , Triglycerides/metabolism , Tyrosine/metabolism
14.
Arterioscler Thromb Vasc Biol ; 19(4): 939-49, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10195921

ABSTRACT

The concept that hepatic cholesteryl ester (CE) mass and the rate of cholesterol esterification regulate hepatocyte assembly and secretion of apoB-containing lipoproteins remains controversial. The present study was carried out in HepG2 cells to correlate the rate of cholesterol esterification and CE mass with apoB secretion by CI-1011, an acyl CoA:cholesterol acyltransferase (ACAT) inhibitor that is known to decrease apoB secretion, in vivo, in miniature pigs. HepG2 cells were incubated with CI-1011 (10 nmol/L, 1 micromol/L, and 10 micromol/L) for 24 hours. ApoB secretion into media was decreased by 25%, 27%, and 43%, respectively (P<0.0012). CI-1011 (10 micromol/L) inhibited HepG2 cell ACAT activity by 79% (P<0.002) and cellular CE mass by 32% (P<0.05). In contrast, another ACAT inhibitor, DuP 128 (10 micromol/L), decreased cellular ACAT activity and CE mass by 85% (P<0.002) and 42% (P=0.01), respectively, but had no effect on apoB secretion into media. To characterize the reduction in apoB secretion by CI-1011, pulse-chase experiments were performed and analyzed by multicompartmental modelling using SAAM II. CI-1011 did not affect the synthesis of apoB or albumin. However, apoB secretion into the media was decreased by 42% (P=0.019). Intracellular apoB degradation increased proportionately (P=0.019). The secretion of albumin and cellular reuptake of labeled lipoproteins were unchanged. CI-1011 and DuP 128 did not affect apoB mRNA concentrations. These results show that CI-1011 decreases apoB secretion by a mechanism that involves an enhanced intracellular degradation of apoB. This study demonstrates that ACAT inhibitors can exert differential effects on apoB secretion from HepG2 cells that do not reflect their efficacy in inhibiting cholesterol esterification.


Subject(s)
Acetates , Apolipoproteins B/antagonists & inhibitors , Apolipoproteins B/metabolism , Coenzyme A/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Sterol O-Acyltransferase/antagonists & inhibitors , Sulfonic Acids/pharmacology , Acetamides , Apolipoprotein B-100 , Carbon Radioisotopes , Hepatoblastoma/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Intracellular Fluid/metabolism , Lipid Metabolism , Lipids/biosynthesis , RNA, Messenger/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sulfonamides , Tumor Cells, Cultured
15.
Int J Clin Pract ; 53(8): 584-92, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10692751

ABSTRACT

The study compared valsartan 80 mg or 160 mg o.d. with captopril 25 mg t.i.d. or placebo on plasma lipids in normotensive and treated hypertensive patients with type II diabetes and microalbuminuria. One hundred and twenty-two adult outpatients were randomised to receive either valsartan 80 mg or 160 mg, captopril 25 mg or placebo for 360 days. Changes from baseline to endpoint in plasma lipid parameters were measured. The primary criterion for tolerability was the incidence of adverse events. All treatment groups showed minor changes in lipid parameters. Triglyceride increased by 2.7% (valsartan 160 mg) to 9.1% (placebo). Total cholesterol decreased under valsartan 80 mg, while other groups showed increases of up to 0.031 mmol/l. Decreases in total cholesterol (p = 0.018), apolipoprotein B (p = 0.042) and apolipoprotein A1 (p = 0.025), were significant for the comparison of 80 mg valsartan and captopril. Valsartan 80 mg or 160 mg o.d. does not cause deleterious changes in the diabetic lipid profile and, unlike captopril, is not associated with dry cough.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Antihypertensive Agents/administration & dosage , Captopril/administration & dosage , Lipids/blood , Tetrazoles/administration & dosage , Valine/analogs & derivatives , Adult , Aged , Albuminuria/blood , Albuminuria/drug therapy , Diabetes Mellitus, Type 2/blood , Double-Blind Method , Female , Humans , Hypertension/blood , Male , Middle Aged , Valine/administration & dosage , Valsartan
16.
Arterioscler Thromb Vasc Biol ; 18(12): 1906-14, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9848883

ABSTRACT

We have previously shown in vivo that the 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor atorvastatin decreases hepatic apolipoprotein B (apoB) secretion into plasma. To test the hypothesis that atorvastatin modulates exogenous triglyceride-rich lipoprotein (TRL) metabolism in vivo, an oral fat load (2 g fat/kg body wt) containing retinol (50 000 IU) was given to 6 control miniature pigs and to 6 animals after 28 days of treatment with atorvastatin 3 mg. kg-1. d-1. A multicompartmental model was developed by use of SAAM II and kinetic analysis performed on the plasma retinyl palmitate (RP) data. Peak TRL (d<1.006 g/mL; Sf>20) triglyceride concentrations were decreased 29% by atorvastatin, and the time to achieve this peak was delayed (5.2 versus 2.3 hours; P<0.01). The TRL triglyceride 0- to 12-hour area under the curve was decreased by 24%. In contrast, atorvastatin treatment had no effect on peak TRL RP concentrations, time to peak, or its rate of appearance into plasma; however, the TRL RP 0- to 12-hour area under the curve was decreased by 20%. Analysis of the RP kinetic parameters revealed that the TRL fractional clearance rate was increased significantly, 1.4-fold (3.093 versus 2.276 pools/h; P=0.012), with atorvastatin treatment. The percent conversion of TRL RP from the rapid-turnover to the slow-turnover compartment was decreased by 47% with atorvastatin treatment. The TRL RP fractional clearance rate was negatively correlated with very low density lipoprotein apoB production rate measured in the fasting state (r=-0.49). Thus, although atorvastatin had no effect on intestinal TRL assembly and secretion, plasma TRL clearance was significantly increased, an effect that may relate to a decreased competition for removal processes by hepatic very low density lipoprotein.


Subject(s)
Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins/metabolism , Postprandial Period , Pyrroles/pharmacology , Triglycerides/metabolism , Animals , Atorvastatin , Chylomicrons/metabolism , Lipoproteins, VLDL/metabolism , Metabolic Clearance Rate , Receptors, LDL/physiology , Swine , Swine, Miniature
17.
Biochim Biophys Acta ; 1393(1): 63-79, 1998 Jul 31.
Article in English | MEDLINE | ID: mdl-9714740

ABSTRACT

To further test the hypothesis that newly synthesized cholesteryl esters regulate hepatic apolipoprotein B (apoB) secretion into plasma, apoB kinetic studies were carried out in seven control miniature pigs and in seven animals after 21 days intravenous administration of the acyl coenzyme A:cholesterol acyltransferase (ACAT) inhibitor DuP 128 (2.2 mg/kg/day). Pigs were fed a fat (34% of calories; polyunsaturated/monounsaturated/saturated ratio, 1:1:1) and cholesterol (400 mg/day; 0.1%; 0.2 mg/kcal) containing pig chow based diet. DuP 128 significantly reduced total plasma triglyceride and very low density lipoprotein (VLDL) triglyceride concentrations by 36 and 31%, respectively (P<0.05). Autologous 131I-VLDL and 125I-LDL were injected simultaneously into each pig and apoB kinetic data was analyzed using multicompartmental analysis (SAAM II). The VLDL apoB pool size decreased by 26% (0.443 vs. 0.599 mg/kg; P<0. 001) which was due entirely to a 28% reduction in VLDL apoB production or secretion rate (1.831 vs. 2.548 mg/kg/h; P=0.006). The fractional catabolic rate (FCR) for VLDL apoB was unchanged. The LDL apoB pool size and production rate were unaffected by DuP 128 treatment. Hepatic microsomal ACAT activity decreased by 51% (0.44 vs. 0.90 nmol/min/mg; P<0.001). Although an increase in hepatic free cholesterol and subsequent decrease in both LDL receptor expression and LDL apoB FCR might be expected, this did not occur. The concentration of hepatic free cholesterol decreased 12% (P=0.008) and the LDL apoB FCR were unaffected by DuP 128 treatment. In addition, DuP 128 treatment did not alter the concentration of hepatic triglyceride or the activity of diacylglycerol acyltransferase, indicating a lack of effect of DuP 128 on hepatic triglyceride metabolism. In our previous studies, DuP 128 treatment of miniature pigs fed a low fat, cholesterol free diet, decreased VLDL apoB secretion by 65% resulting in a reduction in plasma apoB of 60%. We conclude that in miniature pigs fed a high fat, cholesterol containing diet, the inhibition of hepatic cholesteryl ester synthesis by DuP 128 decreases apoB secretion into plasma, but the effect is attenuated relative to a low fat, cholesterol free diet.


Subject(s)
Anticholesteremic Agents/pharmacology , Apolipoproteins B/blood , Cholesterol, Dietary/pharmacology , Dietary Fats/pharmacology , Imidazoles/pharmacology , Lipoproteins, VLDL/blood , Sterol O-Acyltransferase/antagonists & inhibitors , Urea/analogs & derivatives , Animals , Down-Regulation , Enzyme Inhibitors/pharmacology , Intestines/enzymology , Liver/enzymology , Swine , Swine, Miniature , Urea/pharmacology
18.
J Lipid Res ; 39(5): 1008-20, 1998 May.
Article in English | MEDLINE | ID: mdl-9610767

ABSTRACT

We have previously shown that very low density lipoproteins (VLDL, Sf 60-400) from subjects with type IV hyperlipoproteinemia (HTG-VLDL) will induce appreciable cholesteryl ester accumulation in cultured macrophages (J774A.1). The present study examined whether copper-mediated oxidative modification of HTG-VLDL and their remnants would further enhance cholesteryl ester accumulation in J774A.1 cells. Incubation with oxidized VLDL-remnants caused the greatest increase in cellular cholesteryl ester concentrations (54-fold) relative to control cells (P = 0.001). HTG-VLDL and VLDL-remnants each induced similar increases in cholesteryl ester levels (32.3- and 35.8-fold, respectively; both P = 0.001), whereas incubation with oxidized HTG-VLDL brought about only a 20.6-fold increase in cholesteryl ester concentrations (P = 0.014). The increase in cellular cholesteryl ester concentrations induced by oxidized VLDL-remnants was significantly higher (P < or = 0.04) than that induced by all other lipoproteins tested including low density lipoprotein (LDL) and oxidized LDL which caused a 6.7- and a 35.1-fold increase (P < or = 0.0002 for both), respectively. Unlike HTG-VLDL and to a lesser extent VLDL-remnants, uptake of oxidized VLDL and oxidized VLDL-remnants did not require catalytically active, cell secreted lipoprotein lipase. Co-incubation with polyinosine, which blocks binding to the type I scavenger receptor, completely inhibited the cholesteryl ester accumulation induced by oxidized HTG-VLDL, oxidized VLDL-remnants and oxidized LDL (P < or = 0.02). We conclude that oxidation of VLDL-remnants significantly enhances macrophage cholesteryl ester accumulation compared to either HTG-VLDL, VLDL-remnants, or oxidized LDL. Uptake of oxidized VLDL and oxidized VLDL-remnants does not require catalytically active lipoprotein lipase, and involves a receptor that can be competed for by polyinosine.


Subject(s)
Cholesterol Esters/metabolism , Hyperlipoproteinemia Type IV/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, VLDL/metabolism , Macrophages/metabolism , Animals , Cattle , Cell Line , Humans , Lipoprotein Lipase/metabolism , Mice , Milk/enzymology
19.
Arterioscler Thromb Vasc Biol ; 17(11): 2589-600, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9409231

ABSTRACT

In the present studies, the 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor atorvastatin was used to test the hypothesis that inhibition of cholesterol biosynthesis in vivo with a consequent reduction in the availability of hepatic cholesterol for lipoprotein synthesis, would (1) reduce very low density lipoprotein (VLDL) apolipoprotein B (apoB) secretion into the plasma, (2) reduce the conversion of VLDL apoB to LDL apoB, and (3) reduce LDL apoB direct synthesis. ApoB kinetic studies were carried out in six control miniature pigs and in six animals after 21 days of administration of atorvastatin (3 mg/kg per day). Pigs were fed a fat- (34% of calories; polyunsaturated to monounsaturated to saturated ratio, 1:1:1) and cholesterol- (400 mg/d cholesterol; 0.1%; 0.2 mg/kcal) containing pig chow-based diet. Atorvastatin treatment significantly reduced plasma total cholesterol, LDL cholesterol, total triglyceride, and VLDL triglyceride concentrations by 16%, 31%, 19%, and 28%, respectively (P < .01). Autologous 131I-VLDL, 125I-LDL, and [3H]leucine were injected simultaneously into each pig, and apoB kinetic data were analyzed using multicompartmental analysis (SAAM II). The VLDL apoB pool size decreased by 29% (0.46 versus 0.65 mg/kg; P = .002), which was entirely due to a 34% reduction in the VLDL apoB production rate (PR) (1.43 versus 2.19 mg/kg per hour; P = .027). The fractional catabolic rate (FCR) was unchanged. The LDL apoB pool size decreased by 30% (4.74 versus 6.75 mg/kg; P = .0004), which was due to a 22% reduction in the LDL apoB PR (0.236 versus 0.301 mg/kg per hour; P = .004), since the FCR was unchanged. The reduction in LDL apoB PR was primarily due to a 34% decrease in conversion of VLDL apoB to LDL apoB; however, this reduction was not statistically significant (P = .114). Hepatic apoB mRNA abundance quantitated by RNase protection assay was decreased by 13% in the atorvastatin-treated animals (P = .003). Hepatic and intestinal LDL receptor mRNA abundances were not affected. We conclude that inhibition of hepatic HMG-CoA reductase by atorvastatin reduces both VLDL and LDL apoB concentrations, primarily by decreasing apoB secretion into the plasma and not by an increase in hepatic LDL receptor expression. This decrease in apoB secretion may, in part, be due to a reduction in apoB mRNA abundance.


Subject(s)
Anticholesteremic Agents/pharmacology , Apolipoproteins B/biosynthesis , Gene Expression Regulation/drug effects , Heptanoic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins, LDL/biosynthesis , Lipoproteins, VLDL/biosynthesis , Liver/drug effects , Pyrroles/pharmacology , Animals , Apolipoproteins B/blood , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Atorvastatin , Cholesterol/biosynthesis , Cholesterol/blood , Depression, Chemical , Dietary Fats/administration & dosage , Female , Intestine, Small/drug effects , Intestine, Small/metabolism , Kinetics , Lipoproteins, LDL/blood , Lipoproteins, VLDL/blood , Liver/metabolism , Male , Models, Biological , RNA, Messenger/biosynthesis , Receptors, LDL/biosynthesis , Receptors, LDL/genetics , Swine , Swine, Miniature , Triglycerides/blood
20.
Arterioscler Thromb Vasc Biol ; 17(9): 1707-15, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9327767

ABSTRACT

We previously showed that hypertriglyceridemic VLDL (HTG-VLDL, Sf 60 to 400) from subjects with type III (E2/E2) hyperlipoproteinemia do not induce appreciable cholesteryl ester (CE) accumulation in cultured macrophages (J774A.1). In the present study, we examined whether oxidation of type III HTG-VLDL would enhance their uptake by J774A.1 cells. Type III HTG-VLDL were oxidized as measured by both conjugated-diene formation and increased electrophoretic mobility on agarose gels. Both LDL and type III HTG-VLDL undergo oxidation, albeit under different kinetic parameters. From the conjugated-diene curve, type III HTG-VLDL, compared with LDL, were found to have a 6-fold longer lag time, to take 6-fold longer to reach maximal diene production, and to produce a 2-fold greater amount of dienes but at half the rate (all P < .005). Incubation of macrophages with either native type III HTG-VLDL or LDL (50 micrograms lipoprotein cholesterol/mL media for 16 hours) caused small increases (4-fold and 2.7-fold, respectively) in cellular CE levels relative to control cells (both P = .0001). After 24 hours of CuSO4 exposure, we found that oxidized type III HTG-VLDL and LDL caused a 9.4-fold and 10.5-fold increase, respectively, in cellular CE levels (P = .0001). We next examined whether extending the exposure period for type III HTG-VLDL to CuSO4 beyond 24 hours would further enhance its ability to induce macrophage CE accumulation. After 48 hours of CuSO4 exposure, type III HTG-VLDL and LDL caused 21.3-fold and 11.6-fold increases, respectively, in cellular CE levels (P = .0001). The cellular CE loading achieved with 48 hour-oxidized type III HTG-VLDL was significantly higher than either 24 hour-oxidized type III HTG-VLDL (2.3-fold, P = .003) or 48 hour-oxidized LDL (1.8-fold, P = .012). There was no significant difference between the CE loading achieved by incubation of cells with either 24 hour-oxidized type III HTG-VLDL, 24 hour-oxidized LDL, or 48 hour-oxidized LDL (P > or = .518). In this study, we also examined whether partial lipolysis (19% to 50% triglyceride hydrolysis) of type III HTG-VLDL to produce remnants would increase the susceptibility of the lipoprotein to oxidative modification and subsequent cellular CE loading. Forty-eight hour-oxidized type III VLDL-remnants stimulated CE accumulation 30.4-fold over baseline (P = .0001). In contrast, nonoxidized type III VLDL-remnants caused the same very low level of CE loading as did native type III HTG-VLDL (P = .680). The increase in cellular CE levels achieved with 48 hour-oxidized type III VLDL-remnants was significantly higher than that achieved with 48 hour-oxidized type III HTG-VLDL (P = .047). In conclusion, we have shown that oxidized type III HTG-VLDL will induce macrophage CE accumulation well above levels achieved with oxidized LDL. In addition, we also showed that by forming a VLDL-remnant before oxidative modification, we can further enhance macrophage CE accumulation. These results provide a potential mechanism for the atherogenicity of type III HTG-VLDL and their remnants.


Subject(s)
Hypertriglyceridemia/blood , Lipoproteins, VLDL/metabolism , Macrophages/metabolism , Animals , Cattle , Cell Line , Cells, Cultured , Cholesterol Esters/metabolism , Copper Sulfate/pharmacology , Electrophoresis, Agar Gel , Foam Cells/physiology , Humans , Hypertriglyceridemia/classification , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Mice , Oxidation-Reduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...