Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 19(19): 5617-21, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19717304

ABSTRACT

The liver X receptors (LXR) play a key role in cholesterol homeostasis and lipid metabolism. SAR studies around tertiary-amine lead molecule 2, an LXR full agonist, revealed that steric and conformational changes to the acetic acid and propanolamine groups produce dramatic effects on agonist efficacy and potency. The new analogs possess good functional activity, demonstrating the ability to upregulate LXR target genes, as well as promote cholesterol efflux in macrophages.


Subject(s)
Amines/chemistry , Cholesterol/metabolism , Macrophages/drug effects , Orphan Nuclear Receptors/agonists , Amines/chemical synthesis , Amines/pharmacokinetics , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Humans , Liver X Receptors , Macrophages/immunology , Mice , Mice, Knockout , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
3.
Eur J Pharmacol ; 606(1-3): 109-14, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19168058

ABSTRACT

Heme oxygenase-1 (HO) metabolizes heme to form the vasodilator carbon monoxide and antioxidant biliverdin. Upregulation of HO-1 by hemin, which is also a substrate attenuates thrombosis in rodent models, however, whether protection is due to HO-1 upregulation or to increased substrate availability is unknown. This study tested the hypothesis that treatment of mice with cobalt protoporphyrin (CoPP), a non-substrate HO-1 inducer, would protect the endothelium from laser injury. C57Bl/J6 mice were treated with vehicle, CoPP (20 mg/kg), CoPP plus the HO-1 inhibitor tin protoporphyrin (SnPP; 20 mg/kg) or SnPP alone for 18 h. Intravital microscopy was used to quantitate thrombus formation in cremaster arterioles in response to laser ablation of the endothelium. CoPP treatment inhibited thrombosis by 43% compared to vehicle (P<0.05). SnPP co-treatment negated the inhibitory effect of CoPP while SnPP alone potentiated thrombosis compared to vehicle. In CoPP-treated animals, cremaster HO-1 mRNA expression was increased 59+/-17-fold over vehicle (P<0.001). Co-treatment with CoPP+SnPP attenuated this effect by 36%, however the increase in HO-1 protein induced by CoPP was unaffected by SnPP. Induction of HO-1 by the non-substrate inducer CoPP protects against laser induced endothelial injury without the need for increased substrate. Small molecule, substrate-independent upregulation of HO-1 expression represents a feasible approach to ameliorate endothelial dysfunction in cardiovascular disease.


Subject(s)
Arterioles/drug effects , Arterioles/pathology , Heme Oxygenase-1/biosynthesis , Protoporphyrins/pharmacology , Thrombosis/enzymology , Animals , Arterioles/metabolism , Enzyme Induction/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hemodynamics/drug effects , Lasers/adverse effects , Male , Mice , Mice, Inbred C57BL , Thrombosis/etiology , Thrombosis/genetics , Thrombosis/physiopathology , Up-Regulation/drug effects
4.
Bioorg Med Chem Lett ; 19(1): 27-30, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19058966

ABSTRACT

Endothelial lipase (EL) activity has been implicated in HDL catabolism, vascular inflammation, and atherogenesis, and inhibitors are therefore expected to be useful for the treatment of cardiovascular disease. Sulfonylfuran urea 1 was identified in a high-throughput screening campaign as a potent and non-selective EL inhibitor. A lead optimization effort was undertaken to improve potency and selectivity, and modifications leading to improved LPL selectivity were identified. Radiolabeling studies were undertaken to establish the mechanism of action for these inhibitors, which were ultimately demonstrated to be irreversible inhibitors.


Subject(s)
Furans , Lipase/antagonists & inhibitors , Sulfonylurea Compounds/chemical synthesis , Animals , Cardiovascular Diseases/drug therapy , Drug Discovery , Drug Evaluation, Preclinical , Endothelium/enzymology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Sulfonylurea Compounds/pharmacology
5.
Mol Endocrinol ; 22(10): 2241-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18669643

ABSTRACT

Classically, activated transcription by nuclear receptors (NRs) is due to a ligand-induced switch from corepressor- to coactivator-bound states. However, coactivators and corepressors recognize overlapping surfaces of liganded and unliganded NRs, respectively. Here we show that, at sufficiently high concentration, the NR corepressor (NCoR) influences the activity of the liver X receptor (LXR) even in the presence of a potent full agonist that destabilizes NCoR binding. Partial agonist ligands that less effectively dissociate NCoR from LXR are even more sensitive to NCoR levels, in a target gene-selective manner. Thus, differential recruitment of NCoR is a major determinant of partial agonism and selective LXR modulation of target genes.


Subject(s)
DNA-Binding Proteins/agonists , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Transcriptional Activation , Benzoates/chemistry , Benzoates/metabolism , Benzylamines/chemistry , Benzylamines/metabolism , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Humans , Hydrocarbons, Fluorinated , Ligands , Liver X Receptors , Molecular Structure , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Orphan Nuclear Receptors , RNA Interference , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Retinoid X Receptors/chemistry , Retinoid X Receptors/metabolism , Sulfonamides/chemistry , Sulfonamides/metabolism
6.
J Biomol Screen ; 13(6): 468-75, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18566479

ABSTRACT

Endothelial lipase (EL) is a 482-amino-acid protein from the triglyceride lipase gene family that uses a Ser-His-Asp triad for catalysis. Its expression in endothelial cells and preference for phospholipids rather than triglycerides are unique. Animal models in which it is overexpressed or knocked out indicate EL levels are inversely correlated with high-density lipoprotein cholesterol (HDL-C). HDL-C is commonly referred to as the good form of cholesterol because it is involved in the reverse cholesterol transport pathway, in which excess cholesterol is effluxed from peripheral tissues for excretion or reabsorption. Thus, EL inhibition in humans is expected to lead to increases in HDL levels and possibly a decrease in cardiovascular disease. To discover inhibitors of EL, a coupled assay for EL has been developed, using its native substrate, HDL. Hydrolysis of HDL by EL yields free fatty acids, which are coupled through acyl-CoA synthetase, acyl-CoA oxidase, and horseradish peroxidase to produce the fluorescent species resorufin. This assay was developed into a 5-microL, 1536-well assay format, and a high-throughput screen was executed against the GSK collection. In addition to describing the screening results, novel post-HTS mechanism-of-action studies were developed for EL and applied to 1 of the screening hits as an example.


Subject(s)
Lipase/metabolism , Lipoproteins, HDL/metabolism , Acyl-CoA Oxidase/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Assay , CHO Cells , Coenzyme A Ligases/metabolism , Cricetinae , Cricetulus , Fatty Acids, Nonesterified/metabolism , Horseradish Peroxidase/metabolism , Humans , Hydrolysis , Kinetics , Lipase/chemistry , Lipase/genetics , Lipase/isolation & purification , Models, Biological , Oxazines/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
7.
Arterioscler Thromb Vasc Biol ; 27(5): 1115-22, 2007 May.
Article in English | MEDLINE | ID: mdl-17322100

ABSTRACT

OBJECTIVE: Reduced plasma concentrations of high-density lipoprotein-cholesterol (HDL-C) are a significant risk factor for cardiovascular disease. Mechanisms that regulate HDL-C concentrations represent an important area of investigation. METHODS AND RESULTS: Comparative transcriptome analyses of monocyte-derived macrophages (MDM) from a large population of low HDL-C subjects and age- and sex-matched controls revealed a cluster of inflammatory genes highly expressed in low HDL-C subjects. The expression levels of peroxisome proliferator activated receptor (PPAR) gamma and several antioxidant metallothionein genes were decreased in MDM from all low HDL-C groups compared with controls, as was the expression of other genes regulated by PPARgamma, including CD36, adipocyte fatty acid binding protein (FABP4), and adipophilin (ADFP). In contrast, PPARdelta expression was increased in MDM from low HDL-C groups. Quantitative RT-PCR corroborated all major findings from the microarray analysis in two separate patient cohorts. Expression of several inflammatory cytokine genes including interleukin 1beta, interleukin 8, and tumor necrosis factor alpha were highly increased in low HDL-C subjects. CONCLUSIONS: The activated proinflammatory state of monocytes and MDM in low HDL-C subjects constitutes a novel parameter of risk associated with HDL deficiency, related to altered expression of metallothionein genes and the reciprocal regulation of PPARgamma and PPARdelta.


Subject(s)
Cholesterol, HDL/deficiency , Gene Expression , Hypolipoproteinemias/blood , Macrophages/metabolism , PPAR delta/genetics , PPAR gamma/genetics , RNA/genetics , Atherosclerosis/blood , Atherosclerosis/etiology , Biomarkers/blood , Cholesterol, HDL/blood , Fatty Acid-Binding Proteins/biosynthesis , Fatty Acid-Binding Proteins/genetics , Genotype , Humans , Hypolipoproteinemias/complications , Hypolipoproteinemias/genetics , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-8/biosynthesis , Interleukin-8/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Microarray Analysis , Mutation , PPAR delta/biosynthesis , PPAR gamma/biosynthesis , Perilipin-2 , Phenotype , Polymerase Chain Reaction , Risk Factors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
8.
FEBS J ; 273(15): 3498-510, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16884492

ABSTRACT

Lipid accumulation alters macrophage biology and contributes to lipid retention within the vessel wall. In this study, we investigated the role of adipophilin on triglyceride accumulation and lipid-droplet formation in THP-1-derived macrophages (THP-1 macrophages). In the presence of acetylated low-density lipoprotein, macrophages infected with an adenovirus expressing human adipophilin showed a 31% increase in triglyceride content and a greater number of lipid droplets compared with control cells. Incubation of macrophages with very low-density lipoprotein (VLDL) dramatically increased cellular triglyceride content similarly in control and adipophilin-overexpressing cells. By itself, VLDL increased adipophilin expression, which explains the lack of effect of adipophilin overexpression on cellular triglyceride content in macrophages loaded with VLDL. The lipid-droplet content of macrophages was increased by overexpression of adipophilin and/or loading with VLDL. In contrast, inhibition of adipophilin expression using siRNA prevented lipid-droplet formation and significantly reduced intracellular triglyceride content. Using inhibitors of beta-oxidation and acyl-coenzyme A synthetase, results were obtained which suggest that adipophilin elevates cellular lipids by inhibition of beta-oxidation and stimulation of long-chain fatty acid incorporation into triglycerides. Adipophilin expression in THP-1 macrophages altered the cellular content of different lipids and enhanced the size of lipid droplets, consistent with a role for adipophilin in human foam cell formation.


Subject(s)
Macrophages/metabolism , Peptides/physiology , Triglycerides/metabolism , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Immunohistochemistry , Membrane Proteins , Oxidation-Reduction , Perilipin-2
9.
Eur J Pharmacol ; 536(3): 232-40, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16603153

ABSTRACT

The purpose of this study was to determine whether poloxamer 407, a chemical known to increase plasma lipid levels in rodents following parenteral administration, decreased the gene expression of ATP-binding-cassette transporter A1. Using human macrophages cultured with poloxamer 407, there was a significant reduction in the gene expression of ATP-binding-cassette transporter A1; however, there was no effect on the gene expression of either fatty acid synthase or sterol regulatory element binding protein-1. Reduction of ATP-binding-cassette transporter A1 mRNA levels was also observed in both liver and intestine of poloxamer 407-treated rats. When macrophages were cultured with poloxamer 407, the percent of cholesterol effluxed decreased in a concentration-dependent fashion, both in the absence and presence of a synthetic liver X receptor agonist. Lastly, total and unesterified (free) cholesterol concentrations were determined in the liver and 9 peripheral tissues of poloxamer 407- and saline-injected (control) rats. In every tissue, the concentration of total cholesterol for poloxamer 407-treated rats was significantly greater than the corresponding value for controls. Our findings would seem to suggest that the poloxamer 407-mediated reduction in both ATP-binding-cassette transporter A1 gene expression and cellular cholesterol efflux may potentially be one factor that contributes to the accumulation of cholesterol and cholesteryl esters in the liver and 9 peripheral tissues of poloxamer 407-treated rats. Furthermore, the surprising specificity by poloxamer 407 for inhibition of ATP-binding-cassette transporter A1 gene expression over fatty acid synthase and sterol regulatory element binding protein-1 may potentially be due to either disruption of a transcriptional cofactor required for ATP-binding-cassette transporter A1 gene expression, or enhanced turnover of ATP-binding-cassette transporter A1 mRNA.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cholesterol/metabolism , Gene Expression/drug effects , Poloxamer/pharmacology , ATP Binding Cassette Transporter 1 , Animals , Cells, Cultured , Cholesterol/blood , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/genetics , Fatty Acid Synthases/genetics , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Lipids/analysis , Lipids/blood , Liver/drug effects , Liver/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sterol Regulatory Element Binding Protein 1/genetics , Surface-Active Agents/pharmacology
10.
J Lipid Res ; 47(6): 1250-60, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16567856

ABSTRACT

The liver X receptors (LXRs) alpha and beta are responsible for the transcriptional regulation of a number of genes involved in cholesterol efflux from cells and therefore may be molecular targets for the treatment of cardiovascular disease. However, the effects of LXR ligands on cholesterol turnover in cells has not been examined comprehensively. In this study, cellular cholesterol handling (e.g., synthesis, catabolism, influx, and efflux) was examined using a stable isotope labeling study and a two-compartment modeling scheme. In HepG2 cells, the incorporation of 13C into cholesterol from [1-13C]acetate was analyzed by mass isotopomer distribution analysis in conjunction with nonsteady state, multicompartment kinetic analysis to calculate the cholesterol fluxes. Incubation with synthetic, nonsteroidal LXR agonists (GW3965, T0901317, and SB742881) increased cholesterol synthesis (approximately 10-fold), decreased cellular cholesterol influx (71-82%), and increased cellular cholesterol efflux (1.7- to 1.9-fold) by 96 h. As a consequence of these altered cholesterol fluxes, cellular cholesterol decreased (36-39%) by 96 h. The increased cellular cholesterol turnover was associated with increased expression of the LXR-activated genes ABCA1, ABCG1, FAS, and sterol-regulatory element binding protein 1c. In summary, the mathematical model presented allows time-dependent calculations of cellular cholesterol fluxes. These data demonstrate that all of the cellular cholesterol fluxes were altered by LXR activation and that the increase in cholesterol synthesis did not compensate for the increased cellular cholesterol efflux, resulting in a net cellular cholesterol loss.


Subject(s)
Cholesterol/metabolism , DNA-Binding Proteins/physiology , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Benzoates/pharmacology , Benzylamines/pharmacology , Carbon Isotopes/analysis , Cell Line, Tumor , Cholesterol/analysis , Cholesterol Esters/metabolism , Culture Media, Serum-Free/pharmacology , DNA-Binding Proteins/agonists , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Humans , Hydrocarbons, Fluorinated , Kinetics , Liver/cytology , Liver/drug effects , Liver X Receptors , Models, Biological , Orphan Nuclear Receptors , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/physiology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Sulfonamides/pharmacology
11.
Atherosclerosis ; 189(1): 142-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16442115

ABSTRACT

Abnormal lipid deposition in human arteries leads to the formation of fatty streaks due to the accumulation of a large number of macrophage derived-foam cells. The formation and catabolism of intracellular lipid droplets is regulated by droplet-associated proteins. Among such proteins, the role of perilipin in human macrophages was unknown. In this study, we first showed that perilipin expression was increased during differentiation of human monocytes to macrophages. Interestingly, cellular perilipin content was unaffected by treatment of cells with OxLDL, AcLDL, VLDL or sterol esters. Moreover, its expression was not dependent on the presence of adipophilin, another lipid droplet-associated protein, since it was not affected by transfection of macrophages with siRNA-adipophilin. Perilipin overexpression in macrophages with an expression vector resulted in significant lipid droplet formation and TG accumulation and this was unaffected by decreasing adipophilin levels using siRNA. Consequently, perilipin, like adipophilin, might play an important role in the conversion of macrophages into foam cells and contribute to lesion formation. Therefore, inhibition of adipophilin might not be sufficient to prevent lesion formation as previously suggested, and perilipin inhibition might be additionally required.


Subject(s)
Atherosclerosis/metabolism , Macrophages/metabolism , Peptides/metabolism , Phosphoproteins/metabolism , Triglycerides/metabolism , Atherosclerosis/pathology , Biomarkers/metabolism , Blotting, Western , Carrier Proteins , Cell Differentiation , Cells, Cultured , Gene Expression , Humans , Intracellular Fluid/metabolism , Macrophages/pathology , Membrane Proteins , Monocytes/metabolism , Monocytes/pathology , Peptides/genetics , Perilipin-1 , Perilipin-2 , Phosphoproteins/genetics , RNA, Small Interfering/genetics
12.
Circulation ; 113(1): 90-7, 2006 Jan 03.
Article in English | MEDLINE | ID: mdl-16365197

ABSTRACT

BACKGROUND: Liver X receptors (LXRs) are ligand-activated transcription factors involved in the control of lipid metabolism and inflammation. Synthetic LXR agonists have been shown to inhibit the progression of atherosclerosis in mice, but the mechanism is uncertain. LXR agonism upregulates the genes encoding ATP binding cassette transporters A1 (ABCA1) and G1 (ABCG1) in macrophages, thus promoting efflux of cholesterol; it also upregulates liver and intestinal ABCG5 and ABCG8, helping to promote biliary and fecal excretion of cholesterol. Thus, LXR agonism may inhibit atherosclerosis through promotion of reverse cholesterol transport (RCT) in vivo, but this has not been proven. We previously described an in vivo method to trace the movement of cholesterol from 3H-cholesterol-labeled J774 macrophages into plasma, into liver, and ultimately into the bile and feces as free cholesterol or bile acids. In the present study we used this approach to test the hypothesis that administration of the synthetic LXR agonist GW3965 would increase the rate of macrophage RCT in vivo. METHODS AND RESULTS: Three different mouse models-wild-type C57BL/6 mice, LDLR/apobec-1 double knockout mice, and human apolipoprotein (apo)B/cholesteryl ester transfer protein (CETP) double transgenic mice-were treated with either vehicle or GW3965. Mice were injected intraperitoneally with 3H-cholesterol-labeled and cholesterol-loaded macrophages and monitored for the appearance of 3H-tracer in plasma, liver, and feces. Administration of GW3965 significantly increased the levels of 3H-tracer in plasma and feces in all 3 mouse models. CONCLUSIONS: These results demonstrate that administration of the LXR agonist GW3965 increases the rate of RCT from macrophages to feces in vivo.


Subject(s)
Cholesterol/metabolism , DNA-Binding Proteins/agonists , DNA-Binding Proteins/metabolism , Macrophages/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , APOBEC-1 Deaminase , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Biological Transport , Cholesterol/blood , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Feces/chemistry , Humans , Kinetics , Liver X Receptors , Macrophages/cytology , Mice , Mice, Knockout , Orphan Nuclear Receptors , Radioactive Tracers , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tritium
13.
J Med Chem ; 48(17): 5419-22, 2005 Aug 25.
Article in English | MEDLINE | ID: mdl-16107141

ABSTRACT

Substituted 3-(phenylamino)-1H-pyrrole-2,5-diones were identified from a high throughput screen as inducers of human ATP binding cassette transporter A1 expression. Mechanism of action studies led to the identification of GSK3987 as an LXR ligand. GSK3987 recruits the steroid receptor coactivator-1 to human LXRalpha and LXRbeta with EC(50)s of 40 nM, profiles as an LXR agonist in functional assays, and activates LXR though a mechanism that is similar to first generation LXR agonists.


Subject(s)
Aniline Compounds/chemical synthesis , DNA-Binding Proteins/agonists , Maleimides/chemical synthesis , Receptors, Cytoplasmic and Nuclear/agonists , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Binding Sites , Cell Line , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , Genes, Reporter , Histone Acetyltransferases , Humans , Ligands , Liver X Receptors , Luciferases/genetics , Maleimides/chemistry , Maleimides/pharmacology , Models, Molecular , Molecular Structure , Monocytes/drug effects , Monocytes/metabolism , Nuclear Receptor Coactivator 1 , Orphan Nuclear Receptors , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/chemistry , Structure-Activity Relationship , Transcription Factors/metabolism , Up-Regulation
14.
J Lipid Res ; 46(10): 2182-91, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16024916

ABSTRACT

Liver X receptor (LXR) nuclear receptors regulate the expression of genes involved in whole body cholesterol trafficking, including absorption, excretion, catabolism, and cellular efflux, and possess both anti-inflammatory and antidiabetic actions. Accordingly, LXR is considered an appealing drug target for multiple indications. Synthetic LXR agonists demonstrated inhibition of atherosclerosis progression in murine genetic models; however, these and other studies indicated that their major undesired side effect is an increase of plasma and hepatic triglycerides. A significant impediment to extrapolating results with LXR agonists from mouse to humans is the absence in mice of cholesteryl ester transfer protein, a known LXR target gene, and the upregulation in mice but not humans of cholesterol 7alpha-hydroxylase. To better predict the human response to LXR agonism, two synthetic LXR agonists were examined in hamsters and cynomolgus monkeys. In contrast to previously published results in mice, neither LXR agonist increased HDL-cholesterol in hamsters, and similar results were obtained in cynomolgus monkeys. Importantly, in both species, LXR agonists increased LDL-cholesterol, an unfavorable effect not apparent from earlier murine studies. These results reveal additional problems associated with current synthetic LXR agonists and emphasize the importance of profiling compounds in preclinical species with a more human-like LXR response and lipoprotein metabolism.


Subject(s)
Benzhydryl Compounds/pharmacology , Benzoates/pharmacology , Benzylamines/pharmacology , Carrier Proteins/biosynthesis , DNA-Binding Proteins/agonists , Glycoproteins/biosynthesis , Phenylacetates/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Cholesterol Ester Transfer Proteins , Cholesterol, HDL/drug effects , Cholesterol, LDL/drug effects , Cricetinae , Lipids/blood , Lipoproteins/blood , Liver X Receptors , Macaca fascicularis , Male , Mesocricetus , Orphan Nuclear Receptors
15.
Curr Opin Lipidol ; 15(2): 183-9, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15017361

ABSTRACT

PURPOSE OF REVIEW: In the past year, several laboratories taking independent approaches have provided compelling evidence that endothelial lipase, a relatively recent addition to the triglyceride lipase gene family, is a major determinant of HDL metabolism. This review summarizes recent findings from experiments in mice with altered levels of endothelial lipase, from an examination of endothelial lipase catalytic and non-catalytic functions in vitro, and from human genetic studies. RECENT FINDINGS: An analysis of lipids and lipoproteins in endothelial lipase knockout and transgenic mice and in mice with adenovirus-driven hepatic overexpression of endothelial lipase shows, without exception, that total cholesterol, phospholipid and HDL-cholesterol all vary inversely with the endothelial lipase gene dosage, and primarily depend on endothelial lipase catalytic activity. Endothelial lipase participates in HDL metabolism by promoting the turnover of HDL components and increasing the catabolism of apolipoprotein A-I. The measurement of lipase activity on lipoprotein substrates in vitro demonstrates that endothelial lipase is distinct from other triglyceride lipases in showing the highest activity on HDL. Endothelial lipase gene polymorphisms in humans appear to be associated with HDL-cholesterol or HDL3-cholesterol concentrations. SUMMARY: A low HDL-cholesterol level in humans is a risk factor for coronary heart disease. Although not yet demonstrated, it is possible that the action of endothelial lipase on HDL may promote atherogenesis, in which case endothelial lipase may represent an attractive target for pharmaceutical intervention.


Subject(s)
Lipase/metabolism , Lipoproteins, HDL/metabolism , Animals , Arteriosclerosis/genetics , Arteriosclerosis/metabolism , Humans , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Lipase/genetics , Lipase/physiology , Mice , Mice, Knockout , Polymorphism, Genetic
16.
Arterioscler Thromb Vasc Biol ; 24(3): 504-10, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14707038

ABSTRACT

OBJECTIVE: Uptake of modified low-density lipoprotein (LDL) by macrophages through scavenger receptors results in lipid droplets accumulation and foam cell formation. Excess lipid deposition in macrophages has been reported to modulate expression of several genes including adipophilin. In this study, we investigated the function of adipophilin in lipid accumulation and cholesterol efflux in THP-1 macrophages. METHODS AND RESULTS: Adipophilin mRNA expression was 3.5-fold higher in human atherosclerotic plaques compared with healthy areas of the same arteries. Moreover, in the presence of acetylated LDL (AcLDL), triglycerides and cholesteryl esters were increased in macrophages overexpressing adipophilin by 40% and 67%, respectively, whereas their accumulation was reduced when endogenous cellular adipophilin was depleted using siRNA approach. In addition, neither overexpression nor downregulation of adipophilin altered expression of genes involved in lipid efflux. However, the affinity and the number of AcLDL receptors were not affected. After 24-hour incubation of lipid-loaded macrophages with apolipoprotein A-I, cholesterol efflux was reduced by 47% in adipophilin transfected cells versus control cells. CONCLUSIONS: Our results showed that stimulation of adipophilin expression in macrophages by modified LDL promotes triglycerides and cholesterol storage and reduces cholesterol efflux. Therefore, adipophilin might contribute, in vivo, to lipid accumulation in the intima of the arterial wall.


Subject(s)
Carotid Artery Diseases/metabolism , Lipid Metabolism , Macrophages/drug effects , Peptides/physiology , Apolipoprotein A-I/pharmacology , Carotid Artery Diseases/etiology , Carotid Artery Diseases/surgery , Cell Differentiation/drug effects , Cell Line/drug effects , Cell Line/metabolism , Cholesterol/metabolism , Cholesterol Esters/metabolism , DNA, Complementary/genetics , Endarterectomy, Carotid , Foam Cells/metabolism , Gene Silencing , Humans , Lipoproteins, LDL/pharmacology , Macrophages/metabolism , Membrane Proteins , Monocytes/drug effects , Peptides/genetics , Perilipin-2 , RNA, Messenger/biosynthesis , RNA, Small Interfering/pharmacology , Receptors, Immunologic/metabolism , Receptors, Scavenger , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Triglycerides/metabolism
17.
Curr Opin Investig Drugs ; 4(9): 1053-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14582448

ABSTRACT

Liver X receptor (LXR) alpha/beta nuclear receptors are intracellular sterol sensors that regulate expression of genes controlling cholesterol absorption, excretion, catabolism and cellular efflux in target organs, including small intestine, liver and macrophages. Through co-ordination of the expression of target genes in multiple tissues, LXR agonists increase the flux of cholesterol from the periphery to the liver, where it is metabolized and excreted into the bile. Synthetic dual LXR alpha/beta agonists decrease atherosclerosis in mice, however, upregulation of lipogenic target genes and triglyceride elevation in rodents reveals a narrow therapeutic window. LXR subtype-selective agonists or LXR modulators may dissociate the anti-atherosclerotic and lipogenic effects of current dual LXR agonists.


Subject(s)
Anticholesteremic Agents/therapeutic use , Coronary Artery Disease/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Anticholesteremic Agents/chemistry , Cholesterol/metabolism , Coronary Artery Disease/metabolism , DNA-Binding Proteins , Gene Expression Regulation/drug effects , Humans , Liver X Receptors , Macrophages/metabolism , Molecular Structure , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/genetics
18.
J Pharmacol Exp Ther ; 307(2): 720-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12975486

ABSTRACT

Histone deacetylases (HDACs) represent an expanding family of protein modifying-enzymes that play important roles in cell proliferation, chromosome remodeling, and gene transcription. We have previously shown that recombinant human HDAC8 can be expressed in bacteria and retain its catalytic activity. To further explore the catalytic activity of HDACs, we expressed two additional human class I HDACs, HDAC1 and HDAC3, in baculovirus. Recombinant HDAC1 and HDAC3 fusion proteins remained soluble and catalytically active and were purified to near homogeneity. Interestingly, trichostatin (TSA) was found to be a potent inhibitor for all three HDACs (IC50 value of approximately 0.1-0.3 microM), whereas another HDAC inhibitor MS-27-275 (N-(2-aminophenyl)-4-[N-(pyridin-3-methyloxycarbonyl)-aminomethyl]benzamide) preferentially inhibited HDAC1 (IC50 value of approximately 0.3 microM) versus HDAC3 (IC50 value of approximately 8 microM) and had no inhibitory activity toward HDAC8 (IC50 value >100 microM). MS-27-275 as well as TSA increased histone H4 acetylation, induced apoptosis in the human colon cancer cell line SW620, and activated the simian virus 40 early promoter. HDAC1 protein was more abundantly expressed in SW620 cells compared with that of HDAC3 and HDAC8. Using purified recombinant HDAC proteins, we identified several novel HDAC inhibitors that preferentially inhibit HDAC1 or HDAC8. These inhibitors displayed distinct properties in inducing histone acetylation and reporter gene expression. These results suggest selective HDAC inhibitors could be identified using recombinantly expressed HDACs and that HDAC1 may be a promising therapeutic target for designing HDAC inhibitors for proliferative diseases such as cancer.


Subject(s)
Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Pyridines/pharmacology , Cloning, Molecular , Colonic Neoplasms/pathology , Drug Interactions , Gene Expression , Histone Deacetylase 1 , Histone Deacetylases , Humans , Protein Isoforms/antagonists & inhibitors , Recombinant Proteins/antagonists & inhibitors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...