Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Pharmacol ; 78(6): 1046-58, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20837678

ABSTRACT

The liver X receptors (LXRα and LXRß) are members of the nuclear receptor superfamily that function as key transcriptional regulators of a number of biological processes, including cholesterol homeostasis, lipid metabolism, and keratinocyte differentiation. Natural ligands that activate LXRs include oxysterol derivatives such as 25-hydroxycholesterol, 27-hydroxycholesterol, 22(R)-hydroxycholesterol, 20(S)-hydroxycholesterol, and 24(S),25-epoxycholesterol. Related oxysterols, such as 5α,6α-epoxycholesterol (5,6-EC) are present in a number of foods and have been shown to induce atherosclerosis in animal models. Intriguingly, these oxysterols have also been detected in atherosclerotic plaques. Using a variety of biochemical and cellular assays, we demonstrate that 5,6-EC is the first dietary modulator and an endogenous LXR ligand with cell and gene context-dependent antagonist, agonist, and inverse agonist activities. In a multiplexed LXR-cofactor peptide interaction assay, 5,6-EC induced the recruitment of a number of cofactor peptides onto both LXRα and LXRß and showed an EC(50) of approximately 2 µM in peptide recruitment. Furthermore, 5,6-EC bound to LXRα in a radiolabeled ligand displacement assay (EC(50) = 76 nM), thus demonstrating it to be one of the most potent natural LXRα ligands known to date. Analysis of endogenous gene expression in various cell-based systems indicated the potential of 5,6-EC to antagonize LXR-mediated gene expression. Furthermore, it also induced the expression of some LXR-responsive genes in keratinocytes. These results clearly demonstrate that 5,6-EC is an LXR modulator that may play a role in the development of lipid disorders, such as atherosclerosis, by antagonizing the agonistic action of endogenous LXR ligands.


Subject(s)
Cholesterol/analogs & derivatives , Orphan Nuclear Receptors/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Cholesterol/chemistry , Cholesterol/metabolism , Cholesterol/physiology , Humans , Liver X Receptors , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/deficiency , Protein Binding/physiology , Protein Transport/physiology
2.
J Med Chem ; 53(8): 3296-304, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20350005

ABSTRACT

A series of phenyl sulfone substituted quinoxaline were prepared and the lead compound 13 (WYE-672) was shown to be a tissue selective LXR Agonist. Compound 13 demonstrated partial agonism for LXRbeta in kidney HEK-293 cells but did not activate Gal4 LXRbeta fusion proteins in huh-7 liver cells. Although 13 showed potent binding affinity to LXRbeta (IC(50) = 53 nM), it had little binding affinity for LXRalpha (IC(50) > 1.0 microM) and did not recruit any coactivator/corepressor peptides in the LXRalpha multiplex assay. However, compound 13 showed good agonism in THP-1 cells with respect to increasing ABCA1 gene expression and good potency on cholesterol efflux in THP-1 foam cells. In an eight-week lesion study in LDLR -/- mice, compound 13 showed reduction of aortic arch lesion progression and no plasma or hepatic triglyceride increase. These results suggest quinoxaline 13 may have an improved biological profile for potential use as a therapeutic agent.


Subject(s)
Orphan Nuclear Receptors/agonists , Quinoxalines/chemical synthesis , Sulfones/chemical synthesis , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/biosynthesis , Animals , Area Under Curve , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cell Line , Cholesterol/metabolism , Duodenum/metabolism , Half-Life , Humans , Kidney/metabolism , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Organ Specificity , Orphan Nuclear Receptors/genetics , Quinoxalines/chemistry , Quinoxalines/pharmacology , Radioligand Assay , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/pharmacology , Transcriptional Activation , Triglycerides/metabolism
3.
J Lipid Res ; 50(12): 2358-70, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19318684

ABSTRACT

Liver X receptors (LXRs) are ligand-activated transcription factors that coordinate regulation of gene expression involved in several cellular functions but most notably cholesterol homeostasis encompassing cholesterol transport, catabolism, and absorption. WAY-252623 (LXR-623) is a highly selective and orally bioavailable synthetic modulator of LXR, which demonstrated efficacy for reducing lesion progression in the murine LDLR(-/-) atherosclerosis model with no associated increase in hepatic lipogenesis either in this model or Syrian hamsters. In nonhuman primates with normal lipid levels, WAY-252623 significantly reduced total (50-55%) and LDL-cholesterol (LDLc) (70-77%) in a time- and dose-dependent manner as well as increased expression of the target genes ABCA1/G1 in peripheral blood cells. Statistically significant decreases in LDLc were noted as early as day 7, reached a maximum by day 28, and exceeded reductions observed for simvastatin alone (20 mg/kg). Transient increases in circulating triglycerides and liver enzymes reverted to baseline levels over the course of the study. Complementary microarray analysis of duodenum and liver gene expression revealed differential activation of LXR target genes and suggested no direct activation of hepatic lipogenesis. WAY-252623 displays a unique and favorable pharmacological profile suggesting synthetic LXR ligands with these characteristics may be suitable for evaluation in patients with atherosclerotic dyslipidemia.


Subject(s)
Atherosclerosis/drug therapy , Cholesterol, LDL/drug effects , Cholesterol, LDL/metabolism , Indazoles/pharmacology , Lipid Metabolism/drug effects , Macaca fascicularis/metabolism , Orphan Nuclear Receptors/agonists , Animals , Atherosclerosis/metabolism , Caco-2 Cells , Cricetinae , Disease Models, Animal , Humans , Indazoles/blood , Indazoles/chemistry , Ligands , Liver/enzymology , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors/metabolism
4.
J Transl Med ; 6: 59, 2008 Oct 16.
Article in English | MEDLINE | ID: mdl-18925943

ABSTRACT

BACKGROUND: LXRs (Liver X Receptor alpha and beta) are nuclear receptors that act as ligand-activated transcription factors. LXR activation causes upregulation of genes involved in reverse cholesterol transport (RCT), including ABCA1 and ABCG1 transporters, in macrophage and intestine. Anti-atherosclerotic effects of synthetic LXR agonists in murine models suggest clinical utility for such compounds. OBJECTIVE: Blood markers of LXR agonist exposure/activity were sought to support clinical development of novel synthetic LXR modulators. METHODS: Transcript levels of LXR target genes ABCA1 and ABCG1 were measured using quantitative reverse transcriptase/polymerase chain reaction assays (qRT-PCR) in peripheral blood from mice and rats (following a single oral dose) and monkeys (following 7 daily oral doses) of synthetic LXR agonists. LXRalpha, LXRbeta, ABCA1, and ABCG1 mRNA were measured by qRT-PCR in human peripheral blood mononuclear cells (PBMC), monocytes, T- and B-cells treated ex vivo with WAY-252623 (LXR-623), and protein levels in human PBMC were measured by Western blotting. ABCA1/G1 transcript levels in whole-blood RNA were measured using analytically validated assays in human subjects participating in a Phase 1 SAD (Single Ascending Dose) clinical study of LXR-623. RESULTS: A single oral dose of LXR agonists induced ABCA1 and ABCG1 transcription in rodent peripheral blood in a dose- and time-dependent manner. Induction of gene expression in rat peripheral blood correlated with spleen expression, suggesting LXR gene regulation in blood has the potential to function as a marker of tissue gene regulation. Transcriptional response to LXR agonist was confirmed in primates, where peripheral blood ABCA1 and ABCG1 levels increased in a dose-dependent manner following oral treatment with LXR-623. Human PBMC, monocytes, T- and B cells all expressed both LXRalpha and LXRbeta, and all cell types significantly increased ABCA1 and ABCG1 expression upon ex vivo LXR-623 treatment. Peripheral blood from a representative human subject receiving a single oral dose of LXR-623 showed significant time-dependent increases in ABCA1 and ABCG1 transcription. CONCLUSION: Peripheral blood cells express LXRalpha and LXRbeta, and respond to LXR agonist treatment by time- and dose-dependently inducing LXR target genes. Transcript levels of LXR target genes in peripheral blood are relevant and useful biological indicators for clinical development of synthetic LXR modulators.


Subject(s)
Blood Cells/metabolism , DNA-Binding Proteins/agonists , Receptors, Cytoplasmic and Nuclear/agonists , Transcription, Genetic , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Administration, Oral , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/pharmacology , Biomarkers , Blood Cells/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Liver X Receptors , Orphan Nuclear Receptors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
5.
Mol Pharmacol ; 70(4): 1340-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16825483

ABSTRACT

The nuclear receptors liver X receptor (LXR) LXRalpha and LXRbeta are differentially expressed ligand-activated transcription factors that induce genes controlling cholesterol homeostasis and lipogenesis. Synthetic ligands for both receptor subtypes activate ATP binding cassette transporter A1 (ABCA1)-mediated cholesterol metabolism, increase reverse cholesterol transport, and provide atheroprotection in mice. However, these ligands may also increase hepatic triglyceride (TG) synthesis via a sterol response element binding protein 1c (SREBP-1c)-dependent mechanism through a process reportedly regulated by LXRalpha. We studied pan-LXRalpha/beta agonists in LXRalpha knockout mice to assess the contribution of LXRbeta to the regulation of selected target genes. In vitro dose-response studies with macrophages from LXRalpha-/- and beta-/- mice confirm an equivalent role for LXRalpha and LXRbeta in the regulation of ABCA1 and SREBP-1c gene expression. Cholesterol-efflux studies verify that LXRbeta can drive apoA1-dependent cholesterol mobilization from macrophages. The in vivo role of LXRbeta in liver was further evaluated by treating LXRalpha-/- mice with a pan-LXRalpha/beta agonist. High-density lipoprotein (HDL) cholesterol increased without significant changes in plasma TG or very low density lipoprotein. Analysis of hepatic gene expression consistently revealed less activation of ABCA1 and SREBP-1c genes in the liver of LXRalpha null animals than in treated wild-type controls. In addition, hepatic CYP7A1 and several genes involved in fatty acid/TG biosynthesis were not induced. In peripheral tissues from these LXRalpha-null mice, LXRbeta activation increases ABCA1 and SREBP-1c gene expression in a parallel manner. However, putative elevation of SREBP-1c activity in these tissues did not cause hypertriglyceridemia. In summary, selective LXRbeta activation is expected to stimulate ABCA1 gene expression in macrophages, contribute to favorable HDL increases, but circumvent hepatic LXRalpha-dominated lipogenesis.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Liver/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Hepatocytes/metabolism , Lipid Metabolism , Lipoproteins, HDL/blood , Liver X Receptors , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Orphan Nuclear Receptors , Protein Isoforms , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
6.
Arterioscler Thromb Vasc Biol ; 26(10): 2209-15, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16825598

ABSTRACT

OBJECTIVE: The effect of a novel small molecule plasminogen activator inhibitor (PAI-1) inhibitor on adipose tissue physiology was investigated. METHODS AND RESULTS: In human preadipocyte cultures, PAI-039 inhibited both basal and glucose-stimulated increases in active PAI-1 antigen, yet had no effect on PAI-1 mRNA, suggesting a direct inactivation of PAI-1. Differentiation of human preadipocytes to adipocytes was associated with leptin synthesis, which was significantly reduced in the presence of PAI-039, together with an atypical adipocyte morphology characterized by a reduction in the size and number of lipid containing vesicles. In a model of diet-induced obesity, pair-fed C57 Bl/6 mice administered PAI-039 in a high-fat diet exhibited a dose-dependent reduction in body weight, epididymal adipose tissue weight, adipocyte volume, and circulating plasma active PAI-1. Plasma glucose, triglycerides, and leptin were also significantly reduced in drug-treated mice, and concentrations of PAI-039 associated with these physiological effects were near the in vitro IC50 for the inhibition of PAI-1. CONCLUSIONS: Our results indicate that a small molecule inactivator of PAI-1 can neutralize glucose-stimulated increases in PAI-1 in human preadipocyte cultures, reduce adipocyte differentiation, and prevent the development of diet-induced obesity. These data suggest the pharmacological inhibition of PAI-1 could be beneficial in diseases associated with expansion of adipose tissue mass.


Subject(s)
Acetates/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/growth & development , Indoles/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/anatomy & histology , Adult , Animals , Body Weight/drug effects , Cell Count , Cell Differentiation/drug effects , Cells, Cultured , Female , Glucose/pharmacology , Humans , Indoleacetic Acids , Mice , Mice, Inbred C57BL , Organ Size/drug effects , Plasminogen Activator Inhibitor 1/blood , Stem Cells/drug effects , Stem Cells/metabolism
7.
J Lipid Res ; 45(10): 1929-42, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15292374

ABSTRACT

Liver X receptors (LXRs) play key roles in the regulation of cholesterol homeostasis by limiting cholesterol accumulation in macrophages within arterial wall lesion sites by a mechanism that includes the upregulation of ATP binding cassette transporters. These atheroprotective properties distinguish LXRs as potential targets for pharmaceutical intervention in cardiovascular disease. Their associated activity for promoting lipogenesis and triglyceride accretion through the activation of sterol-response element binding protein 1c (SREBP-1c) expression, however, represents a potential proatherogenic liability. A newly characterized synthetic oxysterol, N,N-dimethyl-3beta-hydroxycholenamide (DMHCA), represents a gene-selective LXR modulator that mediates potent transcriptional activation of ABCA1 gene expression while exhibiting minimal effects on SREBP-1c both in vitro and in vivo in mice. DMHCA has the potential to stimulate cholesterol transport through the upregulation of LXR target genes, including ABCA1, in liver, small intestine, and peritoneal macrophages. Compared with known nonsteroidal LXR agonists, however, DMHCA exhibits only limited activity for increasing hepatic SREBP-1c mRNA and does not alter circulating plasma triglycerides. Cell-based studies also indicate that DMHCA enhances cholesterol efflux in macrophages and suggest a mechanism whereby this selective modulator can potentially inhibit cholesterol accumulation. DMHCA and related gene-selective ligands of LXR may have application to the study and treatment of atherosclerosis.


Subject(s)
Cholic Acids/pharmacology , Receptors, Cytoplasmic and Nuclear/genetics , Transcriptional Activation/drug effects , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Arteriosclerosis/drug therapy , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line, Tumor , Cholesterol/metabolism , DNA-Binding Proteins/genetics , Hepatocytes , Humans , Hydroxycholesterols/pharmacology , Ligands , Liver X Receptors , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors , RNA, Messenger/drug effects , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Steroid/drug effects , Receptors, Steroid/genetics , Sterol Regulatory Element Binding Protein 1 , Transcription Factors/genetics , Triglycerides/blood
8.
J Med Chem ; 47(3): 681-95, 2004 Jan 29.
Article in English | MEDLINE | ID: mdl-14736248

ABSTRACT

A novel series of substituted sulfanyldihydroimidazolones (1) that modulates high-density lipoprotein cholesterol (HDL-C) has been reported to have HDL-elevating properties in several animal models. Concerns about the chemical and metabolic stability of 1 directed us to explore the structure-activity relationship (SAR) of a related series of substituted thiohydantoins (2). Expansion of the scope of the thiohydantoin series led to exploration of compounds in related thio-containing ring systems 3-7 and the N-cyanoguanidine derivative 8. Compounds were tested sequentially in three animal models to assess their HDL-C elevating efficacy and safety profiles. Further evaluation of selected compounds in a dose-response paradigm culminated in the identification of compound 2.39 as a candidate compound for advanced preclinical studies.


Subject(s)
Cholesterol, HDL/blood , Imidazoles/chemical synthesis , Thiohydantoins/chemical synthesis , Thiones/chemical synthesis , Administration, Oral , Animals , Cricetinae , Drug Design , Guanidines/chemical synthesis , Guanidines/chemistry , Guanidines/pharmacology , Hypercholesterolemia/blood , Imidazoles/chemistry , Imidazoles/pharmacology , Male , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Stimulation, Chemical , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/chemistry , Thiazolidinediones/pharmacology , Thiohydantoins/chemistry , Thiohydantoins/pharmacology , Thiones/chemistry , Thiones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...