Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters










Publication year range
1.
Curr Opin Investig Drugs ; 2(3): 375-81, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11575708

ABSTRACT

The apparent protective effect of high density lipoprotein cholesterol (HDL) with respect to coronary heart disease (CHD) is generally thought to reside in its ability to transport cholesterol from peripheral cells to the liver for excretion from the body. Knozon as reverse cholesterol transport (RCT), this process involves many key steps and lipoprotein interconversions, and there is no consensus as to which step is most suitable for possible drug intervention. The membrane proteins, scavenger receptor class B, type 1 (SR-B1) and the ATP-binding cassette 1 (ABC1), have been strongly implicated as being important in cholesterol efflux; the former as a bona fide receptor for HDL and the latter as a lipid transporter. Lecithin:cholesterol acyltransferase (LCAT) then esterifies the effluxed cholesterol to form cholesteryl esters (Step 2), which are then transferred to apoB-containing lipoproteins by cholesteryl ester transfer protein (CETP, Step 3). Despite the complexities and uncertainties, drugs should be developed which impact all of the above steps, and short-term endpoints need to be defined for a cautious, systematic approach to clinical evaluation.


Subject(s)
Apolipoprotein A-I/biosynthesis , Cholesterol, HDL/biosynthesis , Cholesterol/metabolism , Coronary Artery Disease/prevention & control , Glycoproteins , Membrane Proteins , Receptors, Immunologic , Receptors, Lipoprotein , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Apolipoprotein A-I/genetics , Biological Transport, Active , CD36 Antigens/genetics , CD36 Antigens/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cholesterol/chemistry , Cholesterol Ester Transfer Proteins , Cholesterol Esters/metabolism , Cholesterol, HDL/blood , Coronary Artery Disease/metabolism , Disease Models, Animal , Gene Targeting , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Mice , Mice, Knockout , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Receptors, Scavenger , Scavenger Receptors, Class B , Triglycerides/metabolism , Up-Regulation
2.
Atherosclerosis ; 157(1): 97-105, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11427208

ABSTRACT

We hypothesized that coadministration of avasimibe and simvastatin would limit size, composition and extent of atherosclerotic lesions and potentially promote lesion regression, since bioavailable ACAT inhibitors decrease monocyte-macrophage enrichment and HMG-CoA reductase inhibitors limit smooth muscle cell migration and proliferation. Male New Zealand white rabbits were sequentially fed a 0.5% cholesterol, 3% peanut oil, 3% coconut oil diet for 9 weeks and a chow-fat diet for 6 weeks prior to drug administration. A time zero control group was necropsied prior to drug administration and the progression control was fed various diets but untreated. Avasimibe (10 mg/kg), simvastatin (2.5 mg/kg) or combination of avasimibe (10 mg/kg) with simvastatin (2.5 mg/kg) were administered in the chow-fat diet for 8 weeks. Plasma total cholesterol exposure was unchanged by avasimibe but was reduced 21% by both simvastatin alone and in combination with avasimibe. Combination of avasimibe and simvastatin decreased VLDL-cholesterol exposure by 56%. VLDL+IDL lipid composition was similar in the progression control and simvastatin-treated animals. Administration of avasimibe alone or in combination with simvastatin reduced the cholesteryl ester fraction and increased the triglyceride fraction to comparable extents. Relative to the progression control, avasimibe plus simvastatin markedly decreased thoracic aortic cholesteryl ester content and lesion coverage by 50% and aortic arch lesion size and macrophage area by 75 and 73%, respectively. With respect to lesion regression, avasimibe+simvastatin decreased aortic arch lesion size by 64% and monocyte-macrophage area by 73% when compared to time zero. Based on these data, we conclude that despite changes in plasma total and lipoprotein cholesterol exposure and lipoprotein composition comparable to monotherapy, inhibition of both ACAT and HMG-CoA reductase may not only directly blunt lesion progression but also promote regression of pre-established atherosclerotic lesions.


Subject(s)
Acetates/therapeutic use , Arteriosclerosis/drug therapy , Hypolipidemic Agents/therapeutic use , Simvastatin/therapeutic use , Sulfonic Acids/therapeutic use , Acetamides , Acetates/pharmacology , Animals , Arteriosclerosis/enzymology , Arteriosclerosis/pathology , Coenzyme A-Transferases/antagonists & inhibitors , Drug Therapy, Combination , Hydroxymethylglutaryl CoA Reductases/metabolism , Hypolipidemic Agents/pharmacology , Rabbits , Simvastatin/pharmacology , Sulfonamides , Sulfonic Acids/pharmacology
3.
Life Sci ; 66(18): 1683-94, 2000 Mar 24.
Article in English | MEDLINE | ID: mdl-10809165

ABSTRACT

Past studies have shown that a high saturated fatty acid diet containing coconut oil elevates plasma HDL cholesterol and apolipoprotein A-I (apoA-1) in rabbits through a mechanism involving increased synthesis. We have extended those studies by investigating expression of the hepatic apolipoprotein A-I gene and other lipid related genes in that model. Rabbits fed a diet containing 14% coconut oil for 4 weeks showed HDL-C elevations of 170% to 250% over chow-fed controls with peak differences occurring at 1 week. Plasma apoA-I levels were also increased over this time frame (160% to 180%) reflecting the HDL-C changes. After 4 weeks, there were no differences in plasma VLDL-C or LDL-C levels in chow versus coconut oil-fed rabbits. Hepatic levels of apoA-I mRNA in coconut oil-fed animals were elevated 150% after 4 weeks compared to chow-fed controls; hepatic mRNA levels for ten other genes either decreased slightly (apoB, LCAT, hepatic lipase, albumin, ACAT, and HMG CoA reductase) or were unchanged (CETP, apoE, LDL-receptor, and acyl CoA oxidase). Nuclear run-on transcription assays revealed that coconut oil feeding for 4 weeks caused a 220% increase in hepatic apoA-I transcription rate compared to controls; no change was observed for CETP and apoE. Treatment of cultured rabbit liver cells with various saturated fatty acids and sera from chow-fed and coconut oil-fed rabbits did not alter apoA-I mRNA levels as observed in vivo. These data demonstrate that coconut oil elevates plasma HDL-C and apoA-I by increasing hepatic apoA-I transcription while expression of other genes involved in lipid metabolism are reduced or unchanged in response to coconut oil feeding.


Subject(s)
Apolipoprotein A-I/biosynthesis , Hyperlipoproteinemias/metabolism , Lipoproteins, HDL/blood , Liver/metabolism , Transcription, Genetic/physiology , Animals , Apolipoprotein A-I/genetics , Cell Nucleus/metabolism , Cells, Cultured , Coconut Oil , DNA/biosynthesis , DNA/genetics , Diet , Dietary Fats/pharmacology , Fatty Acids/blood , Fatty Acids/metabolism , Fatty Acids/pharmacology , Humans , Hyperlipoproteinemias/genetics , Leptin/biosynthesis , Liver/cytology , Liver/drug effects , Male , Plant Oils/pharmacology , RNA, Messenger/biosynthesis , Rabbits , Reverse Transcriptase Polymerase Chain Reaction
4.
Arterioscler Thromb Vasc Biol ; 20(1): 70-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634802

ABSTRACT

Given the significance of cholesteryl ester (CE) accumulation in macrophage foam cell formation, we hypothesized that inhibitors of acyl-CoA:cholesterol O-acyltransferase (ACAT) would produce a histologically stable lesion by limiting macrophage enrichment and thereby a source of matrix metalloproteinases (MMPs). Male New Zealand White rabbits were sequentially fed a cholesterol/fat diet for 9 weeks, a fat-only diet for 6 weeks, and 25 mg/kg avasimibe for 7 to 8 weeks. Avasimibe had no effect on plasma total cholesterol exposure. Plasma avasimibe maximal concentration and 24-hour area-under-the-curve levels were 178 ng/mL and 2525 ng. h/mL, respectively, after 7 weeks of treatment with 25 mg/kg avasimibe. The median inhibitory concentration against human monocyte-macrophage ACAT was 12 ng/mL when determined in the absence of albumin, and aortic arch avasimibe levels were 25 ng/g of tissue wet weight. Avasimibe reduced thoracic aortic and iliac-femoral CE content by 39%, the extent of thoracic aortic lesions by 41%, aortic arch cross-sectional lesions area by 35%, and monocyte-macrophage area by 27%. The reduction in monocyte-macrophage area reflected a change in cell number and not cell size. In the iliac-femoral artery, avasimibe decreased monocyte-macrophage content by 77% and reduced the macrophage-to-lesion ratio from 0.16 to 0.05. Within the aortic arch, the catalytic activity of latent and active MMP-9 was reduced by 65% and 33%, respectively; latent and active MMP-1 and MMP-3 activity measured collectively was decreased by 52% and 60%, respectively, and MMP-2 was unchanged. Aortic arch MMP-9, tissue inhibitor of matrix metalloproteinase (TIMP)-1, and TIMP-2 mRNA levels were reduced 29% to 39%, and MMP-2 mRNA levels increased. We conclude that the bioavailable ACAT inhibitor avasimibe can directly limit macrophage accumulation, resulting in the histological appearance of mainly fibromuscular lesions, and can potentially stabilize preestablished atherosclerotic lesions by reducing MMP expression within the lesion.


Subject(s)
Acetates , Anticholesteremic Agents/pharmacology , Arteriosclerosis/drug therapy , Enzyme Inhibitors/pharmacology , Hypercholesterolemia/drug therapy , Macrophages/drug effects , Matrix Metalloproteinases/metabolism , Sterol O-Acyltransferase/antagonists & inhibitors , Sulfonic Acids/pharmacology , Acetamides , Animals , Aorta, Thoracic/enzymology , Arteriosclerosis/enzymology , Arteriosclerosis/pathology , Base Sequence , Cholesterol/blood , DNA Primers/genetics , Gene Expression/drug effects , Humans , Hypercholesterolemia/enzymology , Hypercholesterolemia/pathology , Macrophages/pathology , Male , Matrix Metalloproteinases/genetics , Rabbits , Sulfonamides , Tissue Inhibitor of Metalloproteinases/genetics , Tissue Inhibitor of Metalloproteinases/metabolism
5.
J Biol Chem ; 273(41): 26755-64, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9756919

ABSTRACT

The synthesis of cholesterol esters by acyl-CoA:cholesterol acyltransferase (ACAT, EC 2.3.1.26) is an important component of cellular cholesterol homeostasis. Cholesterol ester formation also is hypothesized to be important in several physiologic processes, including intestinal cholesterol absorption, hepatic lipoprotein production, and macrophage foam cell formation in atherosclerotic lesions. Mouse tissue expression studies and the disruption of the mouse ACAT gene (Acact) have indicated that more than one ACAT exists in mammals and specifically that another enzyme is important in mouse liver and intestine. We now describe a second mammalian ACAT enzyme, designated ACAT-2, that is 44% identical to the first cloned mouse ACAT (henceforth designated ACAT-1). Infection of H5 insect cells with an ACAT-2 recombinant baculovirus resulted in expression of a approximately 46-kDa protein in cell membranes that was associated with high levels of cholesterol esterification activity. Both ACAT-1 and ACAT-2 also catalyzed the esterification of the 3beta-hydroxyl group of a variety of oxysterols. Cholesterol esterification activities for ACAT-1 and ACAT-2 exhibited different IC50 values when assayed in the presence of several ACAT-specific inhibitors, demonstrating that ACAT inhibitors can selectively target specific forms of ACAT. ACAT-2 was expressed primarily in mouse liver and small intestine, supporting the hypothesis that ACAT-2 contributes to cholesterol esterification in these tissues. The mouse ACAT-2 gene (Acact2) maps to chromosome 15 in a region containing a quantitative trait locus influencing plasma cholesterol levels. The identification and cloning of ACAT-2 will facilitate molecular approaches to understanding the role of ACAT enzymes in mammalian biology.


Subject(s)
Isoenzymes/genetics , Sterol O-Acyltransferase/genetics , Amino Acid Sequence , Animals , Baculoviridae/genetics , Base Sequence , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , Genetic Linkage , Humans , Isoenzymes/metabolism , Mice , Molecular Sequence Data , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Spodoptera , Sterol O-Acyltransferase/metabolism
6.
Atherosclerosis ; 140(1): 15-24, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9733211

ABSTRACT

Hypolipidemic drugs that are efficacious in man are not always active in classical animal models of dyslipidemia. Inhibitors of HMG-CoA reductase (statins) do not lower plasma cholesterol in rats, but yet this species was alone in providing activity for fibrate-type drugs. Nicotinic acid possesses many desirable features with regard to clinical use, but most of these actions are lacking in rats and monkeys. The metabolism of low density lipoproteins in hamsters is widely thought to be similar to that in humans, yet neither statins or fibrates lower plasma lipids in these species. With the advent of mouse models expressing specific human genes (or disruption of genes) it is now possible to re-examine the effect of established drugs and to characterize new hypolipidemic compounds with respect to site and mechanism of action. Drug responses observed in humans are now being seen in such mouse models (e.g. HDL elevation with fenofibrate in mice with the human apo A-I gene). Moreover, mice are now being screened for compounds that lower plasma (human) Lp(a), or lower plasma cholesterol in the absence of LDL receptors. It is proposed that these new genetic mouse models may afford a more focused examination of drug action and provide, for new compounds, better prediction of the human response.


Subject(s)
Disease Models, Animal , Hyperlipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Lipids/blood , Lipoproteins/blood , Animals , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mice , Niacin/analogs & derivatives
7.
Lipids ; 33(5): 489-98, 1998 May.
Article in English | MEDLINE | ID: mdl-9625596

ABSTRACT

WAY-121,898 is an inhibitor of pancreatic cholesteryl ester hydrolase (pCEH). After confirming its in vitro potency and relative lack of a major effect on acyl-CoA:cholesterol acyltransferase (ACAT), it was found that this compound lowers plasma cholesterol in cholesterol-fed, but not chow-fed, rats. Measures of liver cholesteryl ester content and the direct determination of cholesterol absorption (lymph-fistula model) show that inhibition of cholesterol absorption is at least one mechanism for the observed cholesterol lowering. However, WAY-121,898 was also active when administered parenterally to cholesterol-fed rats, and in cholesterol-fed hamsters cholesterol-lowering occurred with oral dosing despite no change in cholesterol absorption, suggesting other modes of action possibly relating to inhibition of liver CEH. Combination treatment in cholesterol-fed rats with the ACAT inhibitor CI-976 resulted in a greater-than-additive reduction in plasma cholesterol, implying that both pCEH and ACAT may play a role in cholesterol absorption in this species. In rabbits, WAY-121,898 prevented the rise in plasma cholesterol due to the feeding of cholesteryl ester but not in rabbits fed (free) cholesterol. In guinea pigs, the compound induced an increase in adrenal cholesteryl ester mass. Taken together, the overall profile in these animal models suggests that WAY-121,898 inhibits more than just the intestinal (lumenal) pCEH, and that the role of this enzyme in cholesterol metabolism may be different within and across species, the former depending upon the dietary cholesterol load.


Subject(s)
Carbamates/pharmacology , Cholesterol, Dietary , Hypolipidemic Agents/pharmacology , Lipids/blood , Liver/metabolism , Pancreas/enzymology , Sterol Esterase/antagonists & inhibitors , Administration, Oral , Animals , Carbamates/administration & dosage , Cholesterol/blood , Cholesterol Esters/metabolism , Cricetinae , Guinea Pigs , Injections, Subcutaneous , Intestinal Absorption/drug effects , Liver/drug effects , Male , Mesocricetus , Rabbits , Rats , Rats, Sprague-Dawley , Species Specificity , Swine , Triglycerides/blood
8.
Atherosclerosis ; 137(1): 77-85, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9568739

ABSTRACT

The hypocholesterolemic and anti-atherogenic properties of sulfamic acid ((2,4,6-tris (1-methylethyl) phenyl) acetyl) 2,6-bis(1-methylethyl) phenyl ester, the ACAT inhibitor, CI-1011, was tested in 120 male F1B hamsters fed a hypercholesterolemic chow-based diet containing 10%, coconut oil and 0.05% cholesterol plus: (i) no drug treatment (HCD); (ii) 3 mg/kg per day (HCD+3): (iii)10 mg/kg per day (HCD+10); (iv) 30 mg/kg per day (HCD+30) of CI-1011; or (v) 500 mg/kg per day of cholestyramine (CSTY). Plasma samples were collected at 8 and 10 weeks for measurement of total cholesterol (TC), very low-density lipoprotein cholesterol (VLDL-C), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C) and triglycerides (TG). For the progression studies, animals were euthanized after 10 weeks for aortic fatty streak area and hepatic cholesterol analysis. For the regression study, a cohort of the HCD was treated with 30 mg/kg per day of CI-1011 (regression) for an additional 8 weeks. The HCD+3, HCD+10, HCD+30 and CSTY lowered plasma TC (25, 32, 34 and 32%, respectively), VLDL-C (62, 74, 71 and 75%, respectively), LDL-C (25, 38, 47 and 46%, respectively) and TG (48, 47, 42 and 45%, respectively). All treatments resulted in a significant lowering of aortic fatty streak area (68, 86, 93 and 94%, respectively) and reduction in hepatic cholesteryl esters (57, 65, 67 and 70%, respectively). Regression of aortic fatty streak area was 90% after 8 weeks of HCD+30 treatment. Also during the regression phase, plasma TC, LDL-C and TG were lowered 23, 33 and 47%, respectively, as well as, hepatic cholesteryl esters (76%). Significant correlations between plasma LDL-C concentration and aortic fatty streak area (r=0.62, P < 0.004) in the HCD+10 group, suggest that CI-1101 altered aortic lipid infiltration primarily by its effect on plasma lipids. However the 30 mg/kg per day dose of CI-1011 which additionally reduced aortic fatty streak area by 51% relative to the 10 mg/kg per day dose was only associated with a 14% further decrease in plasma LDL-C. Finally the 10-fold regression of aortic fatty streak area was associated with only a 35% reduction in plasma LDL-C. These exceptions to the lipid-lesion relationship raise the possibility of additional effects of CI-1011, which may occur independent of or in concert with lipoprotein cholesterol lowering. It is concluded that in hypercholesterolemic hamsters, CI-1011 is approximately 50 times more potent than cholestyramine in cholesterol-lowering, reduction and regression of aortic fatty streak area.


Subject(s)
Acetates , Arteriosclerosis/prevention & control , Enzyme Inhibitors/pharmacology , Sulfonic Acids/pharmacology , Acetamides , Animals , Aorta/drug effects , Aorta/pathology , Arteriosclerosis/drug therapy , Cholesterol/blood , Cholesterol/metabolism , Cholesterol Esters/metabolism , Cholesterol, HDL/blood , Cholesterol, HDL/drug effects , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Cholesterol, VLDL/blood , Cholesterol, VLDL/drug effects , Cricetinae , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Liver/chemistry , Liver/drug effects , Liver/metabolism , Male , Reference Values , Sterol O-Acyltransferase/antagonists & inhibitors , Sulfonamides , Sulfonic Acids/administration & dosage , Sulfonic Acids/therapeutic use , Time Factors , Triglycerides/blood
9.
J Med Chem ; 41(5): 682-90, 1998 Feb 26.
Article in English | MEDLINE | ID: mdl-9513596

ABSTRACT

We prepared a series of alpha-substituted malonester amides that were evaluated for their ability to inhibit acyl-CoA:cholesterol O-acyl transferase activity in vitro and to lower plasma total cholesterol levels in a variety of cholesterol-fed animal models. Compounds of this series were also useful in examining the relationship between adrenal toxicity and ACAT inhibition. One compound from this series, 9f, was a potent inhibitor of ACAT in both the microsomal and cellular assays. It was also bioavailable as determined by both a bioassay and a HPLC-UV assay. This compound was evaluated in both guinea pig and dog models of adrenal toxicity and compared to tetrazole amide 15. In the most sensitive species, the dog, both of these compounds achieved good plasma levels; however, compound 9f caused adrenal necrosis, whereas compound 15 had no effect on the adrenal gland. This adds to the growing body of evidence that the adrenal toxicity observed with ACAT inhibitors may not be mechanism related.


Subject(s)
Adrenal Glands/drug effects , Adrenal Glands/pathology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Malonates/pharmacology , Malonates/toxicity , Phenylacetates/pharmacology , Phenylacetates/toxicity , Sterol O-Acyltransferase/antagonists & inhibitors , Amides/pharmacology , Amides/toxicity , Animals , Anticholesteremic Agents/chemical synthesis , Biological Availability , Cholesterol/blood , Cholesterol, Dietary/administration & dosage , Chromatography, High Pressure Liquid , Dogs , Enzyme Inhibitors/chemical synthesis , Female , Guinea Pigs , Male , Malonates/chemical synthesis , Mice , Microsomes, Liver/enzymology , Necrosis , Phenylacetates/chemical synthesis , Rats , Rats, Sprague-Dawley , Tetrazoles/pharmacology , Tetrazoles/toxicity
11.
Bioorg Med Chem ; 5(4): 739-47, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9158873

ABSTRACT

Our continued interest in developing novel, potent acyl-CoA:cholesterol acyltransferase (ACAT) inhibitors, and our discovery of several active series of disubstituted urea ACAT inhibitors, have led us to investigate a series of trisubstituted ureas that are structural hybrids of our disubstituted series and of a trisubstituted urea ACAT inhibitor series disclosed by scientists at Lederle. This investigation has led to the discovery of novel trisubstituted ureas, several of which inhibit ACAT in the nanomolar range and effectively lower total plasma cholesterol when administered as a diet admixture in an acute model of hypercholesterolemia in rats. One analogue (35) also lowered total cholesterol as efficaciously as CL 277,082 in our chronic hypercholesterolemic rat model. The most notable finding of this study is that the SAR of the trisubstituted ureas diverges from that seen in our previously disclosed disubstituted urea series. This series showed optimal activity with 2,4-difluoro and 2,4,6-trifluoro substitution on the urea N-phenyl, whereas the disubstituted series showed optimal activity with bulky 2,6-disubstitution on the phenyl ring.


Subject(s)
Anticholesteremic Agents/chemical synthesis , Sterol O-Acyltransferase/antagonists & inhibitors , Urea/analogs & derivatives , Administration, Oral , Animals , Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Benzene Derivatives/chemistry , Cholesterol/blood , Diet , Disease Models, Animal , Fluorides/chemistry , Hypercholesterolemia/drug therapy , Magnetic Resonance Spectroscopy , Rabbits , Rats , Structure-Activity Relationship , Urea/chemistry , Urea/pharmacology
12.
Clin Chem ; 42(12): 1996-2001, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8969639

ABSTRACT

Low-density lipoprotein-cholesterol (LDL-C) is currently estimated clinically by using the Friedewald formula, when plasma triglycerides are < 4000 mg/L, or as the difference between infranatant and high-density lipoprotein-cholesterol (HDL-C) values after ultracentrifugation of plasma at native density, when plasma triglycerides are > or = 4000 mg/L (beta quantification). HDL-C is measured by selective precipitation of apolipoprotein B-containing lipoproteins from whole plasma or from the density > 1.006 kg/L infranatant. We compared these conventional methods for LDL-C and HDL-C with "high-performance" gel chromatography (HPGC), a method that directly and simultaneously measures both LDL-C and HDL-C in a single, microliter volume of plasma. Not surprisingly, we found that the results by all these methods were highly correlated. However, LDL-C values were significantly higher and HDL-C values significantly lower by the direct HPGC method than by the conventional methods (paired t-test). In addition, both Bland-Altman plots and concordance correlation analyses indicated lack of agreement between the methods' results in the majority of patients' subgroups.


Subject(s)
Cholesterol, HDL/blood , Cholesterol, LDL/blood , Chromatography, High Pressure Liquid , Apolipoproteins B/blood , Humans , Mathematics , Triglycerides/blood
14.
J Lipid Res ; 37(11): 2372-82, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8978489

ABSTRACT

Guinea pigs were fed 15% (w/W) fat, high in lauric and myristic acids, a diet known to produce hypercholesterolemia in these animals. The diet was given alone or in combination with four doses of atorvastatin equivalent to 1, 3, 10, and 20 mg/kg per day. Atorvastatin reduced plasma LDL cholesterol concentrations by 46, 50, 53, and 70%, respectively (P < 0.001). Plasma apoB concentrations were reduced by atorvastatin (P < 0.001) and compositional changes occurred in VLDL and LDL with reductions of the relative proportion of cholesteryl ester and increases in triacylglycerol. A reduction in hepatic cholesteryl ester (66%) was observed only with the highest atorvastatin dose (20 mg/kg per day) while microsomal cholesterol was reduced by 30% with 3-20 mg/kg per day. Hepatic ACAT activity was down-regulated and apoB/E receptor number was increased by atorvastatin. In contrast, HMG-CoA reductase activity and cholesterol 7 alpha-hydroxylase were not affected by the drug. VLDL apoB secretion rates were decreased by atorvastatin treatment 59 and 76% with 3 and 20 mg/kg per day, respectively. Nascent VLDL particles were larger after drug treatment, showing an increased number in triacylglycerol molecules. These results support the hypothesis that the plasma LDL lowering induced by atorvastatin is due to a decreased secretion of apoB in combination with an increase of hepatic apoB/E receptors.


Subject(s)
Anticholesteremic Agents/pharmacology , Cholesterol/blood , Heptanoic Acids/pharmacology , Lipoproteins/blood , Liver/metabolism , Pyrroles/pharmacology , Animals , Atorvastatin , Cholesterol, LDL/blood , Guinea Pigs , Lipids/blood , Lipoproteins/chemistry , Lipoproteins, LDL/chemistry , Lipoproteins, VLDL/chemistry , Liver/drug effects , Male , Microsomes, Liver/metabolism , Receptors, Lipoprotein/metabolism , Triglycerides/blood
15.
Atherosclerosis ; 127(1): 91-101, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-9006809

ABSTRACT

Chow and sucrose-fed rats were used as animal models to study the dose-responses of bezafibrate and gemfibrozil in normolipidemic and hypertriglyceridemic states, respectively. Although both drugs lowered plasma triglycerides (TG) to about the same extent in chow-fed rats, gemfibrozil lowered liver TG as well as plasma total and LDL-cholesterol (LDL-C), but elevated HDL-cholesterol (HDL-C) and plasma apo E concentrations. Bezafibrate produced opposite effects, namely, decreased HDL-C, apo E and liver TG, and tended to increase LDL-C. TG lowering for both drugs in chow-fed rats was not due to changes in TG secretion (production) in normal rats but was associated with enhanced LPL activity. In hypertriglyceridemic rats both drugs modestly reduced TG secretion rates about 40% at a dose producing maximal TG lowering, but again, gemfibrozil elevated and bezafibrate lowered HDL-C and apo E. Unlike gemfibrozil, bezafibrate induced the appearance of LDL-C in hypertriglyceridemic rats which was not detected in control animals, and also tended to increase rather than decrease plasma apo B levels. Finally, changes in liver TG concentration (mg/g) in hypertriglyceridemic rats were opposite for these drugs, resulting in significant drug-related differences in liver TG content (mg/organ). From these data we postulate that, although similar with regard to TG lowering activity and mechanisms thereof, gemfibrozil and bezafibrate produce fundamentally different effects on LDL, HDL and apolipoprotein metabolism (apo B and apo E) in rats which may relate to potential differential effects on reverse cholesterol transport and atherogenesis.


Subject(s)
Bezafibrate/pharmacology , Gemfibrozil/pharmacology , Hypertriglyceridemia/drug therapy , Hypolipidemic Agents/pharmacology , Animals , Apolipoproteins B/blood , Apolipoproteins E/blood , Bezafibrate/administration & dosage , Body Weight , Cholesterol/metabolism , Cholesterol, LDL/blood , Dose-Response Relationship, Drug , Gemfibrozil/administration & dosage , Hypertriglyceridemia/metabolism , Hypolipidemic Agents/administration & dosage , Immunoelectrophoresis , Lipoprotein Lipase/drug effects , Lipoprotein Lipase/metabolism , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley , Spectrophotometry , Triglycerides/metabolism
16.
J Med Chem ; 39(22): 4382-95, 1996 Oct 25.
Article in English | MEDLINE | ID: mdl-8893833

ABSTRACT

A series of diaryl-substituted heterocyclic ureas was prepared, and their ability to inhibit acyl-CoA: cholesterol O-acyltransferase (ACAT) in vitro and to lower plasma total cholesterol in cholesterol-fed animal models in vivo was examined. N-(2,6-Diisopropylphenyl)-N'-tetrazole or isoxazole-substituted heterocyclic ureas proved optimal. A carbon chain of 11-14 carbons substituted 1,3 with respect to the amine provided the optimal side chain. Substitution of the alkyl chain generally lowered activity. Tetrazole urea 2i dosed at 3 mg/kg lowered plasma total cholesterol (TC) 67% in an acute, cholesterol-fed (C-fed) rat model of hypercholesterolemia and 47% in C-fed dogs. Tetrazole 2i, dosed at 10 mg/kg, also lowered TC 52% and raised HDL cholesterol 113% in rats with pre-established hypercholesterolemia.


Subject(s)
Anticholesteremic Agents/chemistry , Sterol O-Acyltransferase/antagonists & inhibitors , Urea/analogs & derivatives , Animals , Cholesterol, HDL/blood , Chromatography, High Pressure Liquid , Disease Models, Animal , Dogs , Female , Hypercholesterolemia/drug therapy , Male , Rats , Tetrazoles/chemistry
17.
J Med Chem ; 39(20): 3908-19, 1996 Sep 27.
Article in English | MEDLINE | ID: mdl-8831757

ABSTRACT

A series of heterocyclic amides were synthesized and evaluated as inhibitors of acyl-CoA: cholesterol O-acyltransferase (ACAT) in vitro and for cholesterol lowering in cholesterol-fed rats. Compounds were evaluated for cell-based macrophage ACAT inhibition, bioactivity, and adrenal toxicity. Candidates were selected for evaluation in cholesterol-fed dogs and, ultimately, the injured cholesterol-fed rabbit model of atherosclerosis. The heterocyclic amides potently inhibited rabbit liver ACAT (IC50's = 0.014-0.11 microM), and the majority of compounds significantly lowered plasma cholesterol (42-68%) in an acute cholesterol-fed rat model at 3 mg/kg. The most efficacious compounds in the rat were evaluated for bioactivity in vivo and arterial ACAT inhibition in a cell-based macrophage ACAT assay. Two highly bioactive analogs, (+/-)-2-(3-dodecylisoxazol-5-yl)-2-phenyl-N-(2,4,6-trimethoxypheny l) acetamide (13a) and (+/-)-2-(5-dodecylisoxazol-3-yl)-2-phenyl-N-(2,4,6-trimethoxypheny l) acetamide (16a), were selected for further study and were found to be nontoxic in a guinea pig model of adrenal toxicity. Compounds 13a and 16a lowered total cholesterol in the cholesterol-fed rat, rabbit, and dog models of pre-established hypercholesterolemia. Compound 13a in the injured cholesterol-fed rabbit model of atherosclerosis was effective in slowing the development of cholesteryl ester-rich thoracic aortic lesions, reducing lesion coverage by 53% at a dose of 1 mg/kg.


Subject(s)
Acetamides/chemical synthesis , Anticholesteremic Agents/chemical synthesis , Arteriosclerosis/drug therapy , Enzyme Inhibitors/chemical synthesis , Isoxazoles/chemical synthesis , Sterol O-Acyltransferase/antagonists & inhibitors , Acetamides/therapeutic use , Acetamides/toxicity , Adrenal Gland Diseases/chemically induced , Animals , Anticholesteremic Agents/therapeutic use , Cholesterol/blood , Dogs , Enzyme Inhibitors/therapeutic use , Guinea Pigs , Isoxazoles/therapeutic use , Isoxazoles/toxicity , Liver/enzymology , Male , Molecular Structure , Rabbits , Rats
18.
J Med Chem ; 39(12): 2354-66, 1996 Jun 07.
Article in English | MEDLINE | ID: mdl-8691430

ABSTRACT

A series of tetrazole amide derivatives of (+/-)-2-dodecyl-alpha-phenyl-N-(2,4,6-trimethoxyphenyl)-2H-tetrazole-5- acetamide (1) was prepared and evaluated for their ability to inhibit acyl-CoA: cholesterol O-acyltransferase (ACAT) in vitro and to lower plasma total cholesterol in vivo. For this series of compounds, our objective was to systematically replace substituents appended to the amide and tetrazole moieties of 1 with structurally diverse functionalities and assess the effect that these changes have on biological activity. The ensuing structure-activity relationship (SAR) studies identified aryl (7b) and heteroaryl (7f,g) replacements for 2,4,6-trimethoxyphenyl that potently inhibit liver microsomal and macrophage ACAT in vitro and exhibit good cholesterol lowering activity (56-66% decreases in plasma total cholesterol at 30 mg/kg), relative to 1, when compared in the acute rat model of hypercholesterolemia. Replacement of the alpha-phenyl moiety with electron-withdrawing substituents (13e-h), however, significantly reduced liver microsomal ACAT inhibitory activity (IC50 > 1 microM). This is in contrast to electron-donating substituents (13ij,m-q), which produce IC50 values ranging from 5 to 75 nM in the hepatic microsomal assay. For selected tetrazole amides (1, 7b, 13n,o), reversing the order of substituents appended to the 2- and 5-positions in the tetrazole ring (36a-d), in general, improved macrophage ACAT inhibitory activity and provided excellent cholesterol-lowering activity (ranging from 65% to 77% decreases in plasma total cholesterol at 30 mg/kg) in the acute rat screen. The most potent isomeric pair in this set of unsubstituted methylene derivatives (13n and 36a) caused adrenocortical cell degeneration in guinea pigs treated with these inhibitors. In contrast, adrenal glands taken from guinea pigs treated with the corresponding alpha-phenyl-substituted analogs (7b and 36c) were essentially unchanged compared to untreated controls. Subsequent evaluation of 7b and 36c in a rabbit bioassay showed that both compounds and/or their metabolities were present in plasma after oral dosing. Unlike 7b and 36c, compound 1 and related 2,4,6-trimethoxyanilides (13j, 30c,d) showed poor oral activity in the rabbit bioassay. Nevertheless, in cholesterol-fed rabbits, both systemically available (7b, 36c) and poorly absorbed inhibitors (1, 36d) were more effective in lowering plasma total cholesterol than the fatty acid amide CI-976.


Subject(s)
Anticholesteremic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Sterol O-Acyltransferase/antagonists & inhibitors , Tetrazoles/pharmacology , Animals , Anticholesteremic Agents/chemical synthesis , Anticholesteremic Agents/chemistry , Arteriosclerosis/prevention & control , Cholesterol/blood , Cholesterol, Dietary/pharmacokinetics , Drug Design , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Guinea Pigs , Hypercholesterolemia/chemically induced , Hypercholesterolemia/drug therapy , Macrophages/enzymology , Male , Microsomes, Liver/enzymology , Molecular Structure , Rabbits , Rats , Structure-Activity Relationship , Tetrazoles/chemical synthesis , Tetrazoles/chemistry
19.
J Med Chem ; 39(6): 1243-52, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8632431

ABSTRACT

Several series of acyl-CoA:cholesterol O-acyltransferase inhibitors were prepared by the stepwise addition of nitrogen, oxygen, and sulfur nucleophiles to N-chlorosulfonyl isocyanate. The (aminosulfonyl)ureas 3-44 were the most potent inhibitors in vitro, with several compounds having IC50 values < 1 microM. Although the other series of compounds were not as potent in vitro, many compounds did display good in vivo activity in cholesterol-fed rats. Several of the oxysulfonyl carbamates (including CI-999, 115) showed excellent lipid-lowering activity in the chronic in vivo screen, demonstrating significant cholesterol lowering in a pre-established hypercholesterolemic state.


Subject(s)
Enzyme Inhibitors/pharmacology , Isocyanates/pharmacology , Sterol O-Acyltransferase/antagonists & inhibitors , Animals , Rats , Structure-Activity Relationship
20.
J Lipid Res ; 36(12): 2541-51, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8847480

ABSTRACT

For the last 30 years fibrates have been widely prescribed to treat human dyslipidemia. However, the primary mechanism by which they lower plasma lipid levels is still unknown. Studies with transgenic mice have suggested that changes in apoC-III expression levels have a dramatic influence on plasma triglyceride levels. These results suggested that fibrates could reduce lipid levels by lowering apoC-III gene expression. In the current studies, we sought to determine whether the selected fibrates, bezafibrate, clofibrate, fenofibrate, and gemfibrozil, could reduce hepatic apoC-III mRNA and plasma apoC-III levels. Chow-fed rats were orally gavaged daily with a dosing vehicle alone or with 100 mg/kg of each of the fibrates for 1 week and in addition with gemfibrozil for 2 weeks. Bezafibrate and fenofibrate lowered plasma triglyceride by approximately half and dramatically reduced hepatic apoC-III mRNA and plasma apoC-III levels. In contrast, clofibrate did not reduce plasma triglyceride levels and only partially reduced apoC-III mRNA and plasma protein levels. Gemfibrozil strongly reduced plasma triglyceride levels and had an intermediate but significant effect on apoC-III mRNA and plasma apoC-III levels. Some of the fibrates, especially gemfibrozil also reduced plasma apoC-II levels, an effect that could contribute to the observed triglyceride-lowering effect. In addition, the ratio of plasma apoE to plasma apoC-II plus apoC-III was strongly and inversely correlated with plasma triglyceride levels. As plasma apoE levels were not reduced in gemfibrozil-treated animals, this could also have contributed to the triglyceride-lowering effect of this fibrate. Fibrate-mediated triglyceride lowering was not the result of a decreased apoB or VLDL production and, therefore, suggested an enhanced VLDL remnant catabolism. Our results suggest that the mechanism by which fibrates lower plasma triglycerides is by reducing the level of hepatic apoC-III expression.


Subject(s)
Apolipoproteins C/metabolism , Hypolipidemic Agents/pharmacology , Liver/drug effects , Animals , Apolipoprotein C-III , Apolipoproteins C/genetics , Bezafibrate/pharmacology , Cholesterol/blood , Clofibrate/pharmacology , Fenofibrate/pharmacology , Gemfibrozil/pharmacology , Liver/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...