Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
Bioorg Med Chem ; 26(15): 4493-4501, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30077610

ABSTRACT

Pregnane X receptor (PXR) is a ligand-dependent transcription factor that is considered to be a potential therapeutic target for multiple diseases. Herein, we report the development and structure-activity relationship studies of a new series of hPXR agonists. Focusing on our recently developed silanol-sulfonamide scaffold, we developed the potent hPXR agonist 28, which shows good selectivity over hLXRα and ß, hFXR, and hRORα and γ. Examination of the structure-activity relationship suggested a possible strategy to manipulate the selectivity. Docking simulation indicated the presence of an additional binding cavity and polar contacts in the ligand-binding pocket of hPXR. This information should be helpful for the future development of more potent and selective hPXR ligands.


Subject(s)
Pregnane X Receptor/agonists , Silanes/chemistry , Binding Sites , Drug Design , Humans , Liver X Receptors/agonists , Liver X Receptors/metabolism , Molecular Docking Simulation , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/metabolism , Pregnane X Receptor/metabolism , Protein Structure, Tertiary , Silanes/chemical synthesis , Silanes/metabolism , Structure-Activity Relationship
2.
Crit Rev Biochem Mol Biol ; 50(5): 380-92, 2015.
Article in English | MEDLINE | ID: mdl-26383043

ABSTRACT

Health during aging can be improved by genetic, dietary and pharmacological interventions. Many of these increase resistance to various stressors, including xenobiotics. Up-regulation of xenobiotic detoxification genes is a transcriptomic signature shared by long-lived nematodes, flies and mice, suggesting that protection of cells from toxicity of xenobiotics may contribute to longevity. Expression of genes involved in xenobiotic detoxification is controlled by evolutionarily conserved transcriptional regulators. Three closely related subgroups of nuclear hormone receptors (NHRs) have a major role, and these include DAF-12 and NHR-8 in C. elegans, DHR96 in Drosophila and FXR, LXRs, PXR, CAR and VDR in mammals. In the invertebrates, these NHRs have been experimentally demonstrated to play a role in extension of lifespan by genetic and environmental interventions. NHRs represent critical hubs in that they regulate detoxification enzymes with broad substrate specificities, metabolizing both endo- and xeno-biotics. They also modulate homeostasis of steroid hormones and other endogenous cholesterol derivatives and lipid metabolism, and these roles, as well as xenobiotic detoxification, may contribute to the effects of NHRs on lifespan and health during aging, an issue that is being increasingly addressed in C. elegans and Drosophila. Disentangling the contribution of these processes to longevity will require more precise understanding of the molecular mechanisms by which each is effected, including identification of ligands and co-regulators of NHRs, patterns of tissue-specificity and mechanisms of interaction between tissues. The roles of vertebrate NHRs in determination of health during aging and lifespan have yet to be investigated.


Subject(s)
Aging/drug effects , Cell Nucleus/drug effects , Gene Expression Regulation, Developmental/drug effects , Models, Biological , Receptors, Cytoplasmic and Nuclear/metabolism , Xenobiotics/toxicity , Animals , Biotransformation , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Humans , Ligands , Nuclear Envelope/drug effects , Nuclear Envelope/metabolism , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/chemistry , Orphan Nuclear Receptors/metabolism , Protein Conformation , Protein Isoforms/agonists , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/chemistry , Sterols/metabolism , Toxicokinetics , Xenobiotics/metabolism , Xenobiotics/pharmacokinetics
3.
J Biol Chem ; 291(3): 1115-22, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26511317

ABSTRACT

Diabetes is characterized by increased lipogenesis as well as increased endoplasmic reticulum (ER) stress and inflammation. The nuclear hormone receptor liver X receptor (LXR) is induced by insulin and is a key regulator of lipid metabolism. It promotes lipogenesis and cholesterol efflux, but suppresses endoplasmic reticulum stress and inflammation. The goal of these studies was to dissect the effects of insulin on LXR action. We used antisense oligonucleotides to knock down Lxrα in mice with hepatocyte-specific deletion of the insulin receptor and their controls. We found, surprisingly, that knock-out of the insulin receptor and knockdown of Lxrα produced equivalent, non-additive effects on the lipogenic genes. Thus, insulin was unable to induce the lipogenic genes in the absence of Lxrα, and LXRα was unable to induce the lipogenic genes in the absence of insulin. However, insulin was not required for LXRα to modulate the phospholipid profile, or to suppress genes in the ER stress or inflammation pathways. These data show that insulin is required specifically for the lipogenic effects of LXRα and that manipulation of the insulin signaling pathway could dissociate the beneficial effects of LXR on cholesterol efflux, inflammation, and ER stress from the negative effects on lipogenesis.


Subject(s)
Endoplasmic Reticulum Stress , Gene Expression Regulation , Hepatitis/metabolism , Insulin/metabolism , Lipogenesis , Liver/metabolism , Orphan Nuclear Receptors/agonists , Animals , Crosses, Genetic , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Hepatitis/complications , Hepatitis/enzymology , Hepatitis/immunology , Insulin Resistance , Liver/enzymology , Liver/immunology , Liver X Receptors , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Phospholipids/metabolism , Receptor, Insulin/agonists , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction
4.
Bioorg Med Chem Lett ; 27(7): 1608-1610, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28242276

ABSTRACT

Targeting the transcriptional activity of nuclear hormone receptors has proven an effective strategy to treat certain human diseases, and they have become a major focus point to develop novel therapies for the treatment of cancer, inflammation, autoimmune diseases, metabolic disorders, and others. One family of nuclear receptors that has attracted most interest in recent years is the retinoic acid receptor-related orphan receptors (RORs), in particular RORγ. RORγ is a critical regulator of the immune system and RORγ antagonists have shown activity in animal models of inflammatory autoimmune diseases. Here we present the synthesis and biological evaluation of dihydroimidazole tethered imidazolinethiones. We have identified several dual RORγ/α and pan-ROR antagonists with significant activity in cellular assays that could serve as starting points for future optimization efforts to generate potent and selective RORγ modulators.


Subject(s)
Imidazolines/pharmacology , Orphan Nuclear Receptors/antagonists & inhibitors , Thiones/pharmacology , Animals , CHO Cells , Cricetulus , Imidazolines/chemical synthesis , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 2/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Thiones/chemical synthesis
5.
Biochim Biophys Acta ; 1851(12): 1577-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26434697

ABSTRACT

BACKGROUND: Liver X receptors (LXRs) are transcription factors activated by cholesterol metabolites containing an oxidized side chain. Due to their ability to regulate lipid metabolism and cholesterol transport, they have become attractive pharmacological targets. LXRs are closely related to DAF-12, a nuclear receptor involved in nematode lifespan and regulated by the binding of C-27 steroidal acids. Based on our recent finding that the lack of the C-25 methyl group does not abolish their DAF-12 activity, we evaluated the effect of removing it from the (25R)-cholestenoic acid, a LXR agonist. METHODS: The binding mode and the molecular basis of action of 27-nor-5-cholestenoic acid were evaluated using molecular dynamics simulations. The biological activity was investigated using reporter gene expression assays and determining the expression levels of endogenous target genes. The in vitro MARCoNI assay was used to analyze the interaction with cofactors. RESULTS: 27-Nor-5-cholestenoic acid behaves as an inverse agonist. This correlates with the capacity of the complex to better bind corepressors rather than coactivators. The C-25 methyl moiety would be necessary for the maintenance of a torsioned conformation of the steroid side chain that stabilizes an active LXRß state. CONCLUSION: We found that a 27-nor analog is able to act as a LXR ligand. Interestingly, this minimal structural change on the steroid triggered a drastic change in the LXR response. GENERAL SIGNIFICANCE: Results contribute to improve our understanding on the molecular basis of LXRß mechanisms of action and provide a new scaffold in the quest for selective LXR modulators.


Subject(s)
Cholestenes/pharmacology , Gene Expression Regulation/drug effects , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/metabolism , Binding Sites , HEK293 Cells , Hep G2 Cells , Humans , Ligands , Liver X Receptors , Orphan Nuclear Receptors/genetics
6.
J Pathol ; 235(5): 710-20, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25557254

ABSTRACT

Chronic ethanol consumption causes hepatic steatosis and inflammation, which are associated with liver hypoxia. Monocyte chemoattractant protein-1 (MCP-1) is a hypoxia response factor that determines recruitment and activation of monocytes to the site of tissue injury. The level of MCP-1 is elevated in the serum and liver of patients with alcoholic liver disease (ALD); however, the molecular details regarding the regulation of MCP-1 expression are not yet understood completely. Here, we show the role of liver X receptor α (LXRα) in the regulation of MCP-1 expression during the development of ethanol-induced fatty liver injury, using an antagonist, 22-S-hydroxycholesterol (22-S-HC). First, administration of 22-S-HC attenuated the signs of liver injury with decreased levels of MCP-1 and its receptor CCR2 in ethanol-fed mice. Second, hypoxic conditions or treatment with the LXRα agonist GW3965 significantly induced the expression of MCP-1, which was completely blocked by treatment with 22-S-HC or infection by shLXRα lentivirus in the primary hepatocytes. Third, over-expression of LXRα or GW3965 treatment increased MCP-1 promoter activity by increasing the binding of hypoxia-inducible factor-1α to the hypoxia response elements, together with LXRα. Finally, treatment with recombinant MCP-1 increased the level of expression of LXRα and LXRα-dependent lipid droplet accumulation in both hepatocytes and Kupffer cells. These data show that LXRα and its ligand-induced up-regulation of MCP-1 and MCP-1-induced LXRα-dependent lipogenesis play a key role in the autocrine and paracrine activation of MCP-1 in the pathogenesis of alcoholic fatty liver disease, and that this activation may provide a promising new target for ALD therapy.


Subject(s)
Autocrine Communication/drug effects , Chemokine CCL2/metabolism , Fatty Liver, Alcoholic/prevention & control , Hydroxycholesterols/pharmacology , Liver/drug effects , Orphan Nuclear Receptors/antagonists & inhibitors , Paracrine Communication/drug effects , Animals , Binding Sites , Cell Hypoxia , Cells, Cultured , Chemokine CCL2/genetics , Cytoprotection , Disease Models, Animal , Ethanol , Fatty Liver, Alcoholic/metabolism , Fatty Liver, Alcoholic/pathology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Kupffer Cells/pathology , Lipogenesis/drug effects , Liver/metabolism , Liver/pathology , Liver X Receptors , Male , Mice, Inbred C57BL , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Promoter Regions, Genetic , Signal Transduction/drug effects , Transfection , Up-Regulation
7.
Biochim Biophys Acta ; 1841(6): 847-58, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24576892

ABSTRACT

ATP-binding cassette transporter G1 (ABCG1) plays an important role in macrophage reverse cholesterol transport in vivo by promoting cholesterol efflux onto lipidated apoA-I. However, the underlying mechanism is unclear. Here, we found that ABCG1 co-immunoprecipitated with caveolin-1 (CAV1) but not with flotillin-1 and -2. Knockdown of CAV1 expression using siRNAs significantly reduced ABCG1-mediated cholesterol efflux without detectable effect on ABCA1-mediated cholesterol efflux. Disruption of the putative CAV1 binding site in ABCG1, through replacement of tyrosine residues at positions 487 and 489 or at positions 494 and 495 with alanine (Y487AY489A and Y494AY495A), impaired the interaction of ABCG1 with CAV1 and significantly decreased ABCG1-mediated cholesterol efflux. The substitution of Tyr494 and Tyr495 with Phe or Trp that resulted in an intact CAV1 binding site had no effect. Furthermore, Y494AY495A affected trafficking of ABCG1 to the cell surface. The mutant protein is mainly located intracellularly. Finally, we found that CAV1 co-immunoprecipitated with ABCG1 and regulated cholesterol efflux to reconstituted HDL in THP-1-derived macrophages upon the liver X receptor agonist treatment. These findings indicate that CAV1 interacts with ABCG1 and regulates ABCG1-mediated cholesterol efflux.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Caveolin 1/metabolism , Cholesterol/metabolism , Protein Binding/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Binding Sites , Biological Transport/genetics , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Cholesterol/genetics , Humans , Liver X Receptors , Membrane Proteins/metabolism , Orphan Nuclear Receptors/antagonists & inhibitors , RNA, Small Interfering
8.
Biochim Biophys Acta ; 1841(2): 259-66, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24239766

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is a rare neurodegenerative disorder characterized by the accumulation of very-long-chain fatty acids resulting from a beta-oxidation defect. Oxidative stress and inflammation are also key components of the pathogenesis. X-ALD is caused by mutations in the ABCDI gene, which encodes for a peroxisomal half ABC transporter predicted to participate in the entry of VLCFA-CoA into the peroxisome, the unique site of their beta-oxidation. Two homologous peroxisomal ABC transporters, ABCD2 and ABCD3 have been proven to compensate for ABCD1 deficiency when overexpressed. Pharmacological induction of these target genes could therefore represent an alternative therapy for X-ALD patients. Since LXR activation was shown to repress ABCD2 expression, we investigated the effects of LXR antagonists in different cell lines. Cells were treated with GSK(17) (a LXR antagonist recently discovered from the GlaxoSmithKline compound collection), 22(S)-hydroxycholesterol (22S-HC, another LXR antagonist) and 22R-HC (an endogenous LXR agonist). We observed up-regulation of ABCD2,ABCD3 and CTNNB1 (the gene encoding for beta-catenin, which was recently demonstrated to induce ABCD2 expression) in human HepG2 hepatoma cells and in X-ALD skin fibroblasts treated with LXR antagonists. Interestingly, induction in X-ALD fibroblasts was concomitant with a decrease in oxidative stress. Rats treated with 22S-HC showed hepatic induction of the 3 genes of interest. In human, we show by multiple tissue expression array that expression of ABCD2 appears to be inversely correlated with NR1H3 (LXRalpha) expression. Altogether, antagonists of LXR that are currently developed in the context of dyslipidemia may find another indication with X-ALD.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Orphan Nuclear Receptors/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily D , Adrenoleukodystrophy/metabolism , Fatty Acids/analysis , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Hydroxycholesterols/pharmacology , Liver X Receptors , Oxidative Stress
9.
Biochem Biophys Res Commun ; 462(3): 233-8, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-25956064

ABSTRACT

OBJECTIVES: Cholesterol efflux has been thought to be the main and basic mechanism by which free cholesterol is transferred from extra hepatic cells to the liver or intestine for excretion. Salvianolic acid B (Sal B) has been widely used for the prevention and treatment of atherosclerotic diseases. Here, we sought to investigate the effects of Sal B on the cholesterol efflux in THP-1 macrophages. METHODS: After PMA-stimulated THP-1 cells were exposed to 50 mg/L of oxLDL and [(3)H] cholesterol (1.0 µCi/mL) for another 24 h, the effect of Sal B on cholesterol efflux was evaluated in the presence of apoA-1, HDL2 or HDL3. The expression of ATP binding cassette transporter A1 (ABCA1), peroxisome proliferator-activated receptor-gamma (PPAR-γ), and liver X receptor-alpha (LXRα) was detected both at protein and mRNA levels in THP-1 cells after the stimulation of Sal B. Meanwhile, specific inhibition of PPAR-γ and LXRα were performed to investigate the mechanism. RESULTS: The results showed that Sal B significantly accelerated apoA-I- and HDL-mediated cholesterol efflux in both dose- and time-dependent manners. Meanwhile, Sal B treatment also enhanced the expression of ABCA1 at both mRNA and protein levels. Then the data demonstrated that Sal B increased the expression of PPAR-γ and LXRα. And the application of specific agonists and inhibitors of further confirmed that Sal exert the function through PPAR-γ and LXRα. CONCLUSION: These results demonstrate that Sal B promotes cholesterol efflux in THP-1 macrophages through ABCA1/PPAR-γ/LXRα pathway.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Benzofurans/pharmacology , Cholesterol/metabolism , Orphan Nuclear Receptors/metabolism , PPAR gamma/metabolism , ATP Binding Cassette Transporter 1/genetics , Apolipoprotein A-I/metabolism , Biological Transport, Active/drug effects , Cell Line , Drugs, Chinese Herbal/pharmacology , Humans , Lipoproteins, HDL2/metabolism , Lipoproteins, HDL3/metabolism , Liver X Receptors , Macrophages/drug effects , Macrophages/metabolism , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects
10.
J Neuroinflammation ; 12: 27, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25889344

ABSTRACT

BACKGROUND: The nuclear receptor liver X receptor (LXR) exerts transcriptional control over lipid metabolism and inflammatory response in cells of the myeloid lineage, suggesting that LXR may be a potential target in a number of chronic neuroinflammatory and neurodegenerative diseases where persistent microglial activation has been implicated in the pathogenesis. METHODS: The effect of LXR activation on microglia and central nervous system (CNS) inflammation was studied using a synthetic LXR agonist in cultured microglia, a microglial cell line and experimental allergic encephalomyelitis (EAE), an animal model of CNS inflammation. RESULTS: LXR activation inhibited nitric oxide synthase 2, inducible (Nos2) expression and nitric oxide production in lipopolysaccharide (LPS)-stimulated microglia. Inhibition of microglial activation in response to interferon-γ was less reliable. In LPS-stimulated cells, LXR activation did not inhibit nuclear translocation of NF-kappaB1 p50. Instead, LXR-dependent Nos2 repression was associated with inhibition of histone 4 acetylation and inhibition of NF-kappaB1 p50 binding at the Nos2 promoter. Histone acetylation and NF-kappaB1 p50 binding were mechanistically linked, and histone deacetylase (HDAC) activity appeared to be important for LXR-dependent transcriptional repression of Nos2. Analysis of CNS gene expression in animals undergoing EAE showed that the expressions of Lxr and LXR-dependent genes were downregulated during CNS inflammation. Nevertheless, administration of LXR agonist GW3965 during the effector phase of EAE delayed the onset of clinical disease and reversed the diminished expression of LXR-dependent reverse cholesterol transport genes. However, the CNS expressions of Nos2 and other inflammatory genes were not significantly inhibited by LXR activation in EAE, and clinical disease severity was comparable to vehicle controls at later time points in LXR agonist treated animals. CONCLUSIONS: LXR can be targeted to modulate microglial activation. LXR-dependent repression of inflammatory genes may be stimulus-dependent and impaired by HDAC inhibition. Endogenous LXR activity does not appear to modulate CNS inflammation, but LXR activity can be partially restored in the CNS by administration of exogenous LXR agonist with an impact on clinical disease severity at early, but not late, time points in EAE.


Subject(s)
Cytokines/metabolism , Microglia/metabolism , Nitric Oxide Synthase Type II/metabolism , Orphan Nuclear Receptors/metabolism , Animals , Animals, Newborn , Benzoates/therapeutic use , Benzylamines/therapeutic use , Cells, Cultured , Chromatin Immunoprecipitation , Deoxyribonucleases/metabolism , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Gene Expression Profiling , Histones/metabolism , Lipopolysaccharides/pharmacology , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Microglia/drug effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Orphan Nuclear Receptors/antagonists & inhibitors , Peptide Fragments/immunology , Peptide Fragments/toxicity , Protein Binding/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
11.
J Pharmacol Exp Ther ; 355(3): 362-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26384859

ABSTRACT

Liver X receptor (LXR) is a member of the nuclear receptor superfamily, and it regulates various biologic processes, including de novo lipogenesis, cholesterol metabolism, and inflammation. Selective inhibition of LXR may aid the treatment of nonalcoholic fatty liver diseases. In the present study, we evaluated the effects of three cinnamamide derivatives on ligand-induced LXRα activation and explored whether these derivatives could attenuate steatosis in mice. N-(4-trifluoromethylphenyl) 3,4-dimethoxycinnamamide (TFCA) decreased the luciferase activity in LXRE-tk-Luc-transfected cells and also suppressed ligand-induced lipid accumulation and expression of the lipogenic genes in murine hepatocytes. Furthermore, it significantly attenuated hepatic neutral lipid accumulation in a ligand-induced fatty liver mouse system. Modeling study indicated that TFCA inhibited activation of the LXRα ligand-binding domain by hydrogen bonding to Arg305 in the H5 region of that domain. It regulated the transcriptional control exerted by LXRα by influencing coregulator exchange; this process involves dissociation of the thyroid hormone receptor-associated proteins (TRAP)/DRIP coactivator and recruitment of the nuclear receptor corepressor. These results show that TFCA has the potential to attenuate ligand-induced lipogenesis and fatty liver by selectively inhibiting LXRα in the liver.


Subject(s)
Cinnamates/pharmacology , Fatty Liver/prevention & control , Lipogenesis/drug effects , Orphan Nuclear Receptors/antagonists & inhibitors , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Adipogenesis , Animals , Cell Line , Hepatocytes/drug effects , Hepatocytes/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Ligands , Lipid Metabolism/drug effects , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Tartrate-Resistant Acid Phosphatase , Transfection , Triglycerides/metabolism
12.
Hepatology ; 59(5): 1791-802, 2014 May.
Article in English | MEDLINE | ID: mdl-24277692

ABSTRACT

UNLABELLED: Liver X receptor (LXR) activation stimulates triglyceride (TG) accumulation in the liver. Several lines of evidence indicate that estradiol-17ß (E2) reduces TG levels in the liver; however, the molecular mechanism underlying the E2 effect remains unclear. Here, we show that administration of E2 attenuated sterol regulatory element-binding protein (SREBP)-1 expression and TG accumulation induced by LXR activation in mouse liver. In estrogen receptor alpha (ERα) knockout (KO) and liver-specific ERα KO mice, E2 did not affect SREBP-1 expression or TG levels. Molecular analysis revealed that ERα is recruited to the SREBP-1c promoter through direct binding to LXR and inhibits coactivator recruitment to LXR in an E2-dependent manner. Our findings demonstrate the existence of a novel liver-dependent mechanism controlling TG accumulation through the nonclassical ER/LXR pathway. To confirm that a nonclassical ER/LXR pathway regulates ERα-dependent inhibition of LXR activation, we screened ERα ligands that were able to repress LXR activation without enhancing ERα transcriptional activity, and, as a result, we identified the phytoestrogen, phloretin. In mice, phloretin showed no estrogenic activity; however, it did reduce SREBP-1 expression and TG levels in liver of mice fed a high-fat diet to an extent similar to that of E2. CONCLUSION: We propose that ER ligands reduce TG levels in the liver by inhibiting LXR activation through a nonclassical pathway. Our results also indicate that the effects of ER on TG accumulation can be distinguished from its estrogenic effects by a specific ER ligand.


Subject(s)
Fatty Liver/prevention & control , Orphan Nuclear Receptors/physiology , Receptors, Estrogen/physiology , Animals , Diet, High-Fat , Estradiol/pharmacology , Female , Ligands , Liver X Receptors , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/antagonists & inhibitors , Phloretin/pharmacology , Promoter Regions, Genetic , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics , Transcriptional Activation , Triglycerides/metabolism
13.
J Pharmacol Sci ; 128(2): 71-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26109499

ABSTRACT

20(S)-protopanaxatriol (PPT) is an aglycone of ginsenosides isolated from Panax ginseng and has several interesting activities, including anti-inflammatory and anti-oxidative stress effects. Herein, PPT was identified as an inhibitor against the ligand-dependent transactivation of liver X receptor α (LXRα) using a Gal4-TK-luciferase reporter system. LXRα is a transcription factor of nuclear hormone receptor family and stimulates the transcription of many metabolic genes, such as lipogenesis- or reverse cholesterol transport (RCT)-related genes. Quantitative RT-PCR analysis showed that PPT inhibited the LXRα-dependent transcription of lipogenic genes, such as sterol regulatory element binding protein-1c (SREBP-1c), fatty acid synthase, and stearoyl CoA desaturase 1. These inhibitory effects of PPT are, at least in part, a consequence of the reduced recruitment of RNA polymerase II to the LXR response element (LXRE) of the SREBP-1c promoter. Furthermore, LXRα-dependent triglyceride accumulation in primary mouse hepatocytes was significantly reduced by PPT. Interestingly, PPT did not inhibit the LXRα-dependent transcription of ABCA1, a crucial LXRα target gene involved in RCT. Chromatin immunoprecipitation assays revealed that PPT repressed recruitment of the lipogenic coactivator TRAP80 to the SREBP-1c LXRE, but not the ABCA1 LXRE. Overall, these data suggest that PPT has selective inhibitory activity against LXRα-mediated lipogenesis, but not LXRα-stimulated RCT.


Subject(s)
Ginsenosides/pharmacology , Hepatocytes/metabolism , Lipogenesis/drug effects , Lipogenesis/genetics , Orphan Nuclear Receptors/antagonists & inhibitors , Sapogenins/pharmacology , Sterol Regulatory Element Binding Protein 1/genetics , Transcription, Genetic/drug effects , Animals , Biological Transport/drug effects , Biological Transport/genetics , Cells, Cultured , Cholesterol/metabolism , Liver X Receptors , Male , Mediator Complex/metabolism , Mice, Inbred C57BL , Orphan Nuclear Receptors/physiology , Triglycerides/metabolism
14.
Proc Natl Acad Sci U S A ; 109(32): 13112-7, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22826221

ABSTRACT

Parkinson disease (PD) is a progressive neurodegenerative disease whose progression may be slowed, but at present there is no pharmacological intervention that would stop or reverse the disease. Liver X receptor ß (LXRß) is a member of the nuclear receptor super gene family expressed in the central nervous system, where it is important for cortical layering during development and survival of dopaminergic neurons throughout life. In the present study we have used the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD to investigate the possible use of LXRß as a target for prevention or treatment of PD. The dopaminergic neurons of the substantia nigra of LXRß(-/-) mice were much more severely affected by MPTP than were those of their WT littermates. In addition, the number of activated microglia and GFAP-positive astrocytes was higher in the substantia nigra of LXRß(-/-) mice than in WT littermates. Administration of the LXR agonist GW3965 to MPTP-treated WT mice protected against loss of dopaminergic neurons and of dopaminergic fibers projecting to the striatum, and resulted in fewer activated microglia and astroglia. Surprisingly, LXRß was not expressed in the neurons of the substantia nigra but in the microglia and astroglia. We conclude that LXR agonists may have beneficial effects in treatment of PD by modulating the cytotoxic functions of microglia.


Subject(s)
Dopaminergic Neurons/metabolism , Orphan Nuclear Receptors/metabolism , Parkinsonian Disorders/metabolism , Substantia Nigra/cytology , Analysis of Variance , Animals , Astrocytes/metabolism , Benzoates/pharmacology , Benzylamines/pharmacology , Glial Fibrillary Acidic Protein , Immunohistochemistry , Liver X Receptors , Male , Mice , Mice, Knockout , Microglia/metabolism , Nerve Tissue Proteins/metabolism , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , Parkinsonian Disorders/prevention & control , Parkinsonian Disorders/therapy
15.
Chem Pharm Bull (Tokyo) ; 63(8): 628-35, 2015.
Article in English | MEDLINE | ID: mdl-26062802

ABSTRACT

Fourteen novel compounds were prepared and their antagonistic activities against liver X receptors (LXR) α/ß were tested in vitro. Compound 26 had an IC50 value of 6.4 µM against LXRα and an IC50 value of 5.6 µM against LXRß. Docking studies and the results of structure-activity relationships support the further development of this chemical series as LXRα/ß antagonists.


Subject(s)
Fenofibrate/analogs & derivatives , Hypolipidemic Agents/chemistry , Orphan Nuclear Receptors/antagonists & inhibitors , Drug Discovery , Fenofibrate/pharmacology , Humans , Hypolipidemic Agents/pharmacology , Ligands , Liver X Receptors , Molecular Docking Simulation , Orphan Nuclear Receptors/metabolism , Structure-Activity Relationship
16.
J Sci Food Agric ; 95(9): 1903-10, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25186103

ABSTRACT

BACKGROUND: Chlorogenic acids (CGAs) are widely distributed in plant material, including foods and beverages. 5-Caffeoylquinic acid (5-CQA) is the most studied CGA, but the mechanism of its hypolipidaemic effect remains unclear. This study aimed to determine the effect of 5-CQA on lipid metabolism in the liver of Sprague-Dawley rats fed a high-fat diet (HFD). RESULTS: 5-CQA suppressed HFD-induced increases in body weight and visceral fat-pad weight, serum lipid levels, and serum and hepatic free fatty acids in a dose-dependent manner. Real-time polymerase chain reaction revealed that 5-CQA altered the mRNA expression of the transcription factors peroxisome proliferator-activated receptor α (PPARα) and liver X receptor α (LXRα) and target genes involved in hepatic fatty acid uptake, ß-oxidation, fatty acid synthesis, and cholesterol synthesis. Moreover, hepatic tissue sections from HFD-fed rats showed many empty vacuoles, suggesting that liver cells were filled with more fat droplets. However, 5-CQA significantly ameliorated this effect. CONCLUSION: 5-CQA may improve lipid metabolism disorders by altering the expression of PPARα and LXRα, which are involved in multiple intracellular signalling pathways.


Subject(s)
Anti-Obesity Agents/therapeutic use , Chlorogenic Acid/analogs & derivatives , Dietary Supplements , Liver/metabolism , Obesity/prevention & control , Orphan Nuclear Receptors/antagonists & inhibitors , PPAR alpha/agonists , Quinic Acid/analogs & derivatives , Adiposity , Animals , Anti-Obesity Agents/administration & dosage , Antioxidants/administration & dosage , Antioxidants/therapeutic use , Chlorogenic Acid/administration & dosage , Chlorogenic Acid/therapeutic use , Diet, High-Fat/adverse effects , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Gene Expression Regulation , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Hyperlipidemias/prevention & control , Hypolipidemic Agents/administration & dosage , Hypolipidemic Agents/therapeutic use , Lipid Metabolism , Lipids/blood , Liver/pathology , Liver X Receptors , Male , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , PPAR alpha/genetics , PPAR alpha/metabolism , Quinic Acid/administration & dosage , Quinic Acid/therapeutic use , Random Allocation , Rats, Sprague-Dawley
17.
Biochim Biophys Acta ; 1835(2): 219-29, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23357067

ABSTRACT

Prostate cancer (PCa) is the most common cancer in men in developed countries. Epidemiological studies have associated high blood-cholesterol levels with an increased risk of PCa, whilst cholesterol-lowering drugs (statins) reduce the risk of advanced PCa. Furthermore, normal prostate epithelial cells have an abnormally high cholesterol content, with cholesterol levels increasing further during progression to PCa. In this review, we explore why and how this occurs. Concurrent to this observation, intense efforts have been expended in cardiovascular research to better understand the regulators of cholesterol homeostasis. Here, we apply this knowledge to elucidate the molecular mechanisms driving the accumulation of cholesterol in PCa. For instance, recent evidence from our group and others shows that major signalling players in prostate growth and differentiation, such as androgens and Akt, modulate the key transcriptional regulators of cholesterol homeostasis to enhance cholesterol levels. This includes adjusting central carbon metabolism to sustain greater lipid synthesis. Perturbations in cholesterol homeostasis appear to be maintained even when PCa approaches the advanced, 'castration-resistant' state. Overall, this provides a link between cholesterol accumulation and PCa cell growth. Given there is currently no cure for castration-resistant PCa, could cholesterol metabolism be a novel target for PCa therapy? Overall, this review presents a picture that cholesterol metabolism is important for PCa development: growth-promoting factors stimulate cholesterol accumulation, which in turn presents a possible target for chemotherapy. Consequently, we recommend future investigations, both to better elucidate the mechanisms driving this accumulation and applying it in novel chemotherapeutic strategies.


Subject(s)
Cholesterol/metabolism , Prostatic Neoplasms/metabolism , Cell Proliferation , Homeostasis , Humans , Liver X Receptors , Male , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/physiology , Prostatic Neoplasms/drug therapy , Receptors, Androgen/physiology , Sterol Regulatory Element Binding Protein 2/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 2/physiology
18.
Biochim Biophys Acta ; 1831(6): 1134-45, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23466610

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) facilitates cholesterol efflux and thereby inhibits lipid-laden macrophage/foam cell formation and atherosclerosis. ABCA1 expression is transcriptionally regulated by activation of liver X receptor (LXR). Both etoposide and teniposide are DNA topoisomerase II (Topo II) inhibitors and are chemotherapeutic medications used in the treatment of various cancers. Interestingly, etoposide inhibits atherosclerosis in rabbits by unclear mechanisms. Herein, we report the effects of etoposide and teniposide on macrophage ABCA1 expression and cholesterol efflux. Both etoposide and teniposide increased macrophage free cholesterol efflux. This increase was associated with increased ABCA1 mRNA and protein expression. Etoposide and teniposide also increased ABCA1 promoter activity in an LXR-dependent manner and formation of the LXRE-LXR/RXR complex indicating that transcriptional induction had occurred. Expression of ABCG1 and fatty acid synthase (FAS), another two LXR-targeted genes, was also induced by etoposide and teniposide. In vivo, administration of mice with either etoposide or teniposide induced macrophage ABCA1 expression and enhanced reverse cholesterol transport from macrophages to feces. Taken together, our study indicates that etoposide and teniposide increase macrophage ABCA1 expression and cholesterol efflux that may be attributed to the anti-atherogenic properties of etoposide. Our study also describes a new function for Topo II inhibitors in addition to their role in anti-tumorigenesis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cholesterol/metabolism , Foam Cells/drug effects , Gene Expression Regulation/drug effects , Macrophages/drug effects , Orphan Nuclear Receptors/metabolism , Topoisomerase II Inhibitors/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cells, Cultured , Electrophoretic Mobility Shift Assay , Etoposide/pharmacology , Fluorescent Antibody Technique , Foam Cells/cytology , Foam Cells/metabolism , Liver X Receptors , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , Promoter Regions, Genetic/genetics , RNA, Small Interfering/genetics , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Teniposide/pharmacology
19.
Biochem Biophys Res Commun ; 446(3): 651-5, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24480443

ABSTRACT

The regulation of the ABCD2 gene is recognized as a possible therapeutic target for X-linked adrenoleukodystrophy, a rare neurodegenerative disease caused by mutations in the ABCD1 gene. Up-regulation of ABCD2 expression has indeed been demonstrated to compensate for ABCD1 deficiency, restoring peroxisomal ß-oxidation of very-long-chain fatty acids. Besides the known inducers of the ABCD2 gene (phenylbutyrate and histone deacetylase inhibitors, fibrates, dehydroepiandrosterone, thyroid hormone and thyromimetics), this review will focus on LXR antagonists and 22S-hydroxycholesterol, recently described as inducers of ABCD2 expression. Several LXR antagonists have been identified and their possible indication for neurodegenerative disorders will be discussed.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Adrenoleukodystrophy/genetics , Gene Expression Regulation/drug effects , Orphan Nuclear Receptors/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily D , ATP-Binding Cassette Transporters/metabolism , Adrenoleukodystrophy/drug therapy , Humans , Hydroxycholesterols/metabolism , Hydroxycholesterols/pharmacology , Liver X Receptors , Molecular Targeted Therapy/methods , Orphan Nuclear Receptors/genetics
20.
Biochem Biophys Res Commun ; 448(3): 329-34, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24751522

ABSTRACT

Cholesterol efflux from macrophages is a critical mechanism to prevent the development of atherosclerosis. Although PPARγ is known to be a potent sterol sensor that play a fundamental role in cholesterol metabolism, the potential effects of PPARγ responsive miRNA still need to be revealed. In this study, we found that miR-613 is inversely correlated with LXRα and ABCA1 in PPARγ activated THP-1 cells. PPARγ negatively regulates the expression of miR-613 at transcriptional level, and miR-613 suppressed LXRα and ABCA1 by targeting the 3'-UTR of their mRNAs. Furthermore, downregulation of LXRα and ABCA1 by miR-613 inhibited cholesterol efflux from PPARγ activated THP-1 macrophages. These results revealed an alternative mechanism for PPARγ regulation and provided a potential target for the treatment of cholesterol metabolic diseases.


Subject(s)
ATP Binding Cassette Transporter 1/antagonists & inhibitors , ATP Binding Cassette Transporter 1/genetics , Cholesterol/metabolism , MicroRNAs/genetics , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/genetics , PPAR gamma/metabolism , 3' Untranslated Regions , Atherosclerosis/genetics , Atherosclerosis/metabolism , Base Sequence , Binding Sites/genetics , Biological Transport, Active , Cell Line , Down-Regulation , Humans , Liver X Receptors , Macrophage Activation , Macrophages/metabolism , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL