Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 6: 28367, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27380719

ABSTRACT

Identification of low cost, highly active, durable completely noble metal-free electro-catalyst for oxygen reduction reaction (ORR) in proton exchange membrane (PEM) fuel cells, oxygen evolution reaction (OER) in PEM based water electrolysis and metal air batteries remains one of the major unfulfilled scientific and technological challenges of PEM based acid mediated electro-catalysts. In contrast, several non-noble metals based electro-catalysts have been identified for alkaline and neutral medium water electrolysis and fuel cells. Herein we report for the very first time, F doped Cu1.5Mn1.5O4, identified by exploiting theoretical first principles calculations for ORR and OER in PEM based systems. The identified novel noble metal-free electro-catalyst showed similar onset potential (1.43 V for OER and 1 V for ORR vs RHE) to that of IrO2 and Pt/C, respectively. The system also displayed excellent electrochemical activity comparable to IrO2 for OER and Pt/C for ORR, respectively, along with remarkable long term stability for 6000 cycles in acidic media validating theory, while also displaying superior methanol tolerance and yielding recommended power densities in full cell configurations.

2.
Mol Endocrinol ; 29(4): 571-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25719402

ABSTRACT

The nuclear receptor farnesoid X receptor (FXR) (nuclear receptor subfamily 1, group H, member 4, or NR1H4) is highly expressed in the liver and intestine. Previous reports have suggested beneficial functions of FXR in the homeostasis of bile acids, lipids, and glucose, as well as in promoting liver regeneration and inhibiting carcinogenesis. To investigate the effect of chronic FXR activation in vivo, we generated transgenic mice that conditionally and tissue specifically express the activated form of FXR in the liver and intestine. Unexpectedly, the transgenic mice showed several intriguing phenotypes, including partial neonatal lethality, growth retardation, and spontaneous liver toxicity. The transgenic mice also displayed heightened sensitivity to a high-cholesterol diet-induced hepatotoxicity but resistance to the gallstone formation. The phenotypes were transgene specific, because they were abolished upon treatment with doxycycline to silence the transgene expression. The perinatal toxicity, which can be rescued by a maternal vitamin supplement, may have resulted from vitamin deficiency due to low biliary bile acid output as a consequence of inhibition of bile acid formation. Our results also suggested that the fibroblast growth factor-inducible immediate-early response protein 14 (Fn14), a member of the proinflammatory TNF family, is a FXR-responsive gene. However, the contribution of Fn14 induction in the perinatal toxic phenotype of the transgenic mice remains to be defined. Because FXR is being explored as a therapeutic target, our results suggested that a chronic activation of this nuclear receptor may have an unintended side effect especially during the perinatal stage.


Subject(s)
Cholesterol/toxicity , Intestinal Mucosa/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts/metabolism , Diet, High-Fat , Intestines/drug effects , Liver/drug effects , Liver Diseases/metabolism , Mice , Mice, Transgenic , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , TWEAK Receptor , Vitamin A/blood , Vitamin E/blood
3.
Mol Pharmacol ; 80(1): 191-200, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21511916

ABSTRACT

Liver fibrosis is a chronic disorder that is characterized by an alteration of the balance between fibrogenesis and fibrinolysis, which results in accumulation of excessive amounts of extracellular matrix (ECM) and distortion of the normal liver architecture. The activation and transformation of quiescent hepatic stellate cells (HSCs) into myofibroblast-like cells constitute a major mechanism for the increased production of ECM in the liver. The nuclear receptor farnesoid X receptor (FXR) shows potent antifibrotic activity in HSCs and protects animals in rodent models of liver fibrosis. However, the detailed mechanism remains incompletely understood. In this study, we report that treatment with 3-[2-[2-chloro-4-[[3-(2,6-dichlorophenyl)-5-(1-methylethyl)-4-isoxazolyl]methoxy]phenyl]ethenyl]benzoic acid (GW4064), a synthetic FXR ligand, led to up-regulation of microRNA-29a (miR-29a) in HSCs isolated from wild-type mice, rats, and humans but not from FXR(-/-) mice. miR-29a seems to play an inhibitory role in the regulation of ECM production because of the following: 1) transfection of HSCs with miR-29a mimic resulted in drastic down-regulation of the mRNA expression of several genes that encode ECM proteins; and 2) miR-29a significantly inhibited the expression of a reporter expression plasmid that contains the 3'-untranslated region of the corresponding ECM genes. Our results suggest that miR-29a is a FXR target gene because miR-29a promoter activity was significantly increased by pharmacologic or genetic activation of FXR. Functional analysis of human miR-29a promoter identified an imperfect inverted repeat spaced by one nucleotide DNA motif, inverted repeat-1 (5'-AGGTCAcAGACCT-3'), as a likely FXR-responsive element that is involved in miR-29a regulation. Our study uncovers a new mechanism by which FXR negatively regulates the expression of ECM in HSCs.


Subject(s)
Liver/metabolism , MicroRNAs/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Base Sequence , Blotting, Western , Chromatin Immunoprecipitation , Humans , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley
4.
PLoS One ; 5(11): e13955, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21085652

ABSTRACT

Activation of hepatic stellate cells (HSCs) plays an important role in the development of cirrhosis through the increased production of collagen and the enhanced contractile response to vasoactive mediators such as endothelin-1 (ET-1). The farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily that is highly expressed in liver, kidneys, adrenals, and intestine. FXR is also expressed in HSCs and activation of FXR in HSCs is associated with significant decreases in collagen production. However, little is known about the roles of FXR in the regulation of contraction of HSCs. We report in this study that treatment of quiescent HSCs with GW4064, a synthetic FXR agonist, significantly inhibited the HSC transdifferentiation, which was associated with an inhibition of the upregulation of ET-1 expression. These GW4064-treated cells also showed reduced contractile response to ET-1 in comparison to HSCs without GW4064 treatment. We have further shown that GW4064 treatment inhibited the ET-1-mediated contraction in fully activated HSCs. To elucidate the potential mechanism we showed that GW4064 inhibited ET-1-mediated activation of Rho/ROCK pathway in activated HSCs. Our studies unveiled a new mechanism that might contribute to the anti-cirrhotic effects of FXR ligands.


Subject(s)
Endothelin-1/metabolism , Hepatic Stellate Cells/drug effects , Isoxazoles/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Blotting, Western , Cell Shape/drug effects , Cell Transdifferentiation/drug effects , Cells, Cultured , Endothelin-1/genetics , Endothelin-1/pharmacology , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Male , Microfilament Proteins/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , rho-Associated Kinases/metabolism
5.
Atherosclerosis ; 213(2): 443-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20956002

ABSTRACT

OBJECTIVE: The farnesoid X receptor (FXR), a member of the nuclear receptor superfamily, has been proposed to play an important role in the pathogenesis of cardiovascular diseases by regulating the metabolism and transport of cholesterol and triglyceride. Scavenger receptor class B type I (SR-BI), a high-density lipoprotein receptor, plays an important role in decreasing lipid metabolism-associated cardiovascular diseases by regulating reverse cholesterol transport. Recent studies have shown that SR-BI expression is upregulated by several nuclear receptors. However, the role of FXR in the regulation of SR-BI expression is not well known. In the present study, we investigate the regulation of SR-BI by FXR in hepatocyte and the corresponding mechanism. METHODS AND RESULTS: Treatment of human hepatoma cell line HepG2 with FXR ligands resulted in upregulation of SR-BI at the levels of both mRNA and protein. Reporter assays showed that activation of FXR significantly enhanced the SR-BI promoter activity. Electrophoretic mobility shift and chromatin immunoprecipitation assays indicated that FXR induced SR-BI expression by binding to a novel FXR element (FXRE), a directed repeat DNA motif, DR8 (-703 AGGCCAcgttctagAGCTCA -684). The in vivo experiment demonstrated that gavaging mice with a natural ligand of FXR increased SR-BI expression in liver tissues. CONCLUSIONS: FXR can directly upregulate SR-BI expression in hepatocyte, and DR8 is a likely novel FXRE that is involved in SR-BI regulation. FXR may serve as a novel molecular target for manipulating SR-BI expression in hepatocyte.


Subject(s)
Receptors, Cytoplasmic and Nuclear/physiology , Scavenger Receptors, Class B/metabolism , Animals , DNA Probes , Female , Hep G2 Cells , Hepatocytes/metabolism , Humans , Male , Mice , Promoter Regions, Genetic/physiology , Scavenger Receptors, Class B/genetics , Up-Regulation
6.
Mol Pharmacol ; 78(4): 666-74, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20592274

ABSTRACT

The liver X receptor (LXR) and constitutive androstane receptor (CAR) are two nuclear receptors postulated to have distinct functions. LXR is a sterol sensor that promotes lipogenesis, whereas CAR is a xenosensor that controls xenobiotic responses. Here, we show that LXRα and CAR are functionally related in vivo. Loss of CAR increased the expression of lipogenic LXR target genes, leading to increased hepatic triglyceride accumulation, whereas activation of CAR inhibited the expression of LXR target genes and LXR ligand-induced lipogenesis. On the other hand, a combined loss of LXR α and ß increased the basal expression of xenobiotic CAR target genes, whereas activation of LXR inhibited the expression of CAR target genes and sensitized mice to xenobiotic toxicants. The mutual suppression between LXRα and CAR was also observed in cell culture and reporter gene assays. LXRα, like CAR, exhibited constitutive activity in the absence of an exogenously added ligand by recruiting nuclear receptor coactivators. Interestingly, although CAR competed with LXRα for coactivators, the constitutive activity and recruitment of coactivators was not required for CAR to suppress the activity of LXRα. In vivo chromatin immunoprecipitation assay showed that cotreatment of a CAR agonist compromised the LXR agonist responsive recruitment of LXRα to Srebp-1c, whereas an LXR agonist inhibited the CAR agonist-responsive recruitment of CAR to Cyp2b10. In conclusion, our results have revealed dual functions of LXRα and CAR in lipogenesis and xenobiotic responses, establishing a unique role of these two receptors in integrating xenobiotic and endobiotic homeostasis.


Subject(s)
Lipogenesis/physiology , Orphan Nuclear Receptors/metabolism , Receptor Cross-Talk/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cells, Cultured , Constitutive Androstane Receptor , Female , Lipogenesis/drug effects , Lipogenesis/genetics , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Orphan Nuclear Receptors/deficiency , Orphan Nuclear Receptors/physiology , Pyridines/pharmacology , Receptor Cross-Talk/drug effects , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/physiology , Xenobiotics/metabolism , Xenobiotics/pharmacology
7.
J Pharmacol Exp Ther ; 331(1): 234-43, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19605523

ABSTRACT

Asymmetric dimethylarginine (ADMA) is a potent endogenous inhibitor of endothelial nitric-oxide synthase (eNOS), and increased plasma concentrations of ADMA have been regarded as a risk factor for a number of cardiovascular diseases. Circulating ADMA is largely taken up by liver and kidney via system y(+) carriers of the cationic amino acid (CAT) family and subsequently metabolized by dimethylarginine dimethylaminohydrolases (DDAHs). As such, agents targeted at enhancing ADMA metabolism may prove to be useful in the prevention and/or treatment of various types of cardiovascular disease. Farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily and plays an important role in the maintenance of cholesterol and bile acid homeostasis. We report here that treatment of mice with an FXR agonist 3-(2,6-dichlorophenyl)-4-(3'-carboxy-2-chlorostilben-4-yl)oxymethyl-5-isopropylisoxazole; GW4064) led to increased expression of DDAH-1 and CAT-1 in both liver and kidney. In cultured human hepatocytes and kidney proximal tubular epithelial cells, GW4064 increased CAT-1 expression, and this was associated with a significant increase in the cellular uptake of ADMA. Promoter analyses suggest that CAT-1 is a likely target of FXR, and a functional FXR response element was found in the promoter region of CAT-1 gene. These data suggest that FXR may play an important role in regulating blood levels of ADMA via coordinated regulation of DDAH-1 and CAT-1 in liver and kidney.


Subject(s)
Amidohydrolases/metabolism , Arginine/analogs & derivatives , Cationic Amino Acid Transporter 1/metabolism , Kidney/enzymology , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Arginine/blood , Arginine/chemistry , Cell Line , Cells, Cultured , Haplorhini , Humans , Isoxazoles/pharmacology , Kidney/drug effects , Kidney/physiology , Liver/enzymology , Liver/physiology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/physiology , Receptors, Cytoplasmic and Nuclear/agonists
8.
J Control Release ; 137(1): 38-45, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19332088

ABSTRACT

A new series of cationic polymers, poly[N,N-bis-(2-hydroxylpropyl) alkylalcoholamine-co-ethylenediamine] were synthesized by directly cross-linking several dichloro alkylating agents with ethylenediamine and its derivatives. Co-polymerization with cystamine introduces biodegradable disulfide bonds to the polymer backbone. When tested on COS-1 cells, PHAs showed reduced cytotoxicity, broad polymer to DNA ratios, and enhanced transfection activity that was 2-9-fold better than that of polyethylenimine. Comparison studies also revealed several chemical and physical parameters related to the biological activities of these polymers. The length of the side chain groups affects both transfection activity and toxicity of the polymers; a side chain group of moderate size appeared to be optimal for both high transfection activity and low toxicity. Introduction of biodegradable disulfide bonds to the polymer backbone further enhanced transfection activity and reduced toxicity of the polymer. Fractionated PHAs with molecular weight of > or =5000 Da were equally effective but smaller polymers were ineffective in transfection. This flexible synthesis route enables the determination of key structural and physical parameters related to polymer activity and could aid further improvement of polymer-based transfection agents.


Subject(s)
Cations , Ethylenediamines , Gene Transfer Techniques , Polyamines , Polymers , Acid Phosphatase/analysis , Alkylation , Animals , COS Cells , Cations/chemistry , Cations/pharmacology , Cell Line , Cell Survival/drug effects , Chlorocebus aethiops , Cricetinae , Cross-Linking Reagents/pharmacology , Disulfides/chemistry , Ethylenediamines/chemistry , Ethylenediamines/pharmacology , Ethylenediamines/toxicity , Genes, Reporter , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , Hydrogen-Ion Concentration , Lethal Dose 50 , Luciferases/genetics , Molecular Structure , Molecular Weight , Polyamines/chemistry , Polyamines/pharmacology , Polyamines/toxicity , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacology , Polymers/chemistry , Polymers/pharmacology , Polymers/toxicity , Transfection , alpha-Galactosidase/genetics
9.
AAPS J ; 11(1): 23-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19132538

ABSTRACT

Targeted delivery of functional nucleic acids (genes and oligonucleotides) to pulmonary endothelium may become a novel therapy for the treatment of various types of lung diseases. It may also provide a new research tool to study the functions and regulation of novel genes in pulmonary endothelium. Its success is largely dependent on the development of a vehicle that is capable of efficient pulmonary delivery with minimal toxicity. This review summarizes the recent progress that has been made in our laboratory along these research directions. Factors that affect pulmonary nucleic acids delivery are also discussed.


Subject(s)
Drug Delivery Systems/methods , Lung/blood supply , Nucleic Acids/pharmacokinetics , Pulmonary Circulation , Administration, Inhalation , Animals , Antigens, Surface/drug effects , Cations/pharmacokinetics , Dependovirus/genetics , Drug Carriers/administration & dosage , Drug Carriers/pharmacokinetics , Endothelial Cells/drug effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacokinetics , Humans , Injections, Intravenous , Lipids/administration & dosage , Lipids/pharmacokinetics , Liposomes/administration & dosage , Liposomes/pharmacokinetics , Lung/cytology , Mice , Nucleic Acids/administration & dosage , Nucleic Acids/therapeutic use , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacokinetics , Oligonucleotides/therapeutic use , Peptides/administration & dosage , Peptides/pharmacokinetics , Polymers/pharmacokinetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/therapeutic use
10.
Biochem Biophys Res Commun ; 372(4): 746-51, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18514055

ABSTRACT

Decorin is a member of the family of small leucine-rich proteoglycans that are present in blood vessels and synthesized by vascular smooth muscle cells (VSMCs). Decorin plays complex roles in both normal vascular physiology and the pathogenesis of various types of vascular disorders. However, the mechanisms of regulation of decorin expression in vasculature are not clearly understood. Particularly little information is available about a role of nuclear receptors in the regulation of decorin expression. In the present study, we report that activation of vascular FXR by a specific ligand resulted in upregulation of decorin at the levels of both mRNA and protein. FXR appears to induce decorin expression at a transcriptional level because (1) upregulation of decorin mRNA expression was abolished by the treatment of a transcription inhibitor, actinomycin D; and (2) decorin promoter activity was significantly increased by activation of FXR. Functional analysis of human decorin promoter identified an imperfect inverted repeat DNA motif, IR8 (-2313TGGTCAtagtgtcaTGACCT-2294), as a likely FXR-responsive element that is involved in decorin regulation.


Subject(s)
DNA-Binding Proteins/metabolism , Extracellular Matrix Proteins/genetics , Gene Expression Regulation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Proteoglycans/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Cells, Cultured , DNA-Binding Proteins/agonists , Decorin , Humans , Isoxazoles/pharmacology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Repetitive Sequences, Nucleic Acid , Response Elements/drug effects , Sequence Analysis, DNA , Transcription Factors/agonists , Up-Regulation
11.
Gastroenterology ; 134(2): 556-67, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18242221

ABSTRACT

BACKGROUND & AIMS: Liver X receptor (LXR) is known to promote hepatic lipogenesis by activating the lipogenic transcriptional factor sterol regulatory element-binding protein (Srebp). Pregnane X receptor (PXR), a previously known "xenobiotic receptor," could mediate a Srebp-independent lipogenic pathway by activating the free fatty acid uptake transporter Cd36. The goal of this study is to investigate further the role of Cd36 in hepatic steatosis. METHODS: Wild-type, LXR transgenic, PXR transgenic, and Cd36 null mice were used to study the regulation of Cd36 and other hepatic lipogenic genes and the implication of this regulation in hepatic steatosis. Promoter sequences of Cd36 and peroxisome proliferator-activated receptor (PPAR) gamma were cloned, and their respective regulation by LXR and PXR was investigated by combinations of receptor-DNA binding and reporter gene assays. RESULTS: We showed that genetic (transgene) or pharmacologic (ligands) activation of LXR induced Cd36. Promoter analysis established Cd36 as a novel transcription target of LXRalpha. Moreover, the hepatic steatosis induced by LXR agonists was largely abolished in Cd36 null mice. We also showed that PPARgamma, a positive regulator of Cd36, is a transcriptional target of PXR, suggesting that PXR can regulate Cd36 directly or through its activation of PPARgamma. Interestingly, both LXR-mediated Cd36 regulation and PXR-mediated PPARgamma regulation are liver specific. CONCLUSIONS: We conclude that Cd36 is a shared target of LXR, PXR, and PPARgamma. The network of CD36 regulation by LXR, PXR, and PPARgamma establishes this free fatty acid transporter as a common target of orphan nuclear receptors in their mediation of lipid homeostasis.


Subject(s)
CD36 Antigens/metabolism , DNA-Binding Proteins/metabolism , Fatty Liver/metabolism , PPAR gamma/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Animals , Base Sequence , CD36 Antigens/genetics , DNA-Binding Proteins/genetics , Fatty Liver/pathology , Fatty Liver/physiopathology , Female , Gene Expression Regulation/physiology , Homeostasis/physiology , Lipid Metabolism/physiology , Liver/metabolism , Liver/pathology , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Orphan Nuclear Receptors , PPAR gamma/genetics , Pregnane X Receptor , Promoter Regions, Genetic/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/genetics
12.
Cardiovasc Res ; 77(3): 560-9, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18006431

ABSTRACT

AIMS: The farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily and plays an important role in the pathogenesis of cardiovascular diseases via regulating the metabolism and transport of cholesterol. We and others have recently shown that FXR is also expressed in the vasculature, including endothelial cells and smooth muscle cells (SMC). However, the biological significance of FXR activation in SMC is still poorly understood. In this study, we examine the effect of FXR ligands on the angiotensin system in rat aortic SMC (RASMC), as angiotensin II (Ang II) signalling contributes to various types of vascular lesions by promoting cell growth of vascular SMC. METHODS AND RESULTS: Treatment of RASMC with a FXR ligand showed no obvious effect on the expression of angiotensinogen, Ang II type 1 receptor (AT1R) or type 4 receptor (AT4R) but led to a significant increase in the expression of type 2 receptor (AT2R). FXR ligand treatment also resulted in an inhibition of Ang II-mediated extracellular signal-regulated kinase (ERK) activation and growth proliferation. Promoter reporter gene and electrophoretic mobility-shift assays suggest that FXR upregulates AT2R expression at a transcriptional level. Upregulation of AT2R appears to play a role in the FXR-mediated inhibition of ERK activation via upregulation of Rous sarcoma oncogene (Src) homology domain-containing tyrosine phosphatase 1 (SHP-1) because FXR-mediated upregulation of SHP-1 can be blocked by an AT2R antagonist and FXR-mediated ERK inactivation was significantly attenuated via treatment with either an AT2R antagonist or a SHP-1 inhibitor. CONCLUSION: FXR in SMC may serve as a novel molecular target for modulating Ang II signalling in the vasculature.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptor, Angiotensin, Type 2/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Angiotensin II/physiology , Animals , Cells, Cultured , Chenodeoxycholic Acid/pharmacology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Isoxazoles/pharmacology , Muscle, Smooth, Vascular/cytology , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Rats , Rats, Sprague-Dawley
13.
Cardiovasc Res ; 77(1): 169-77, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18006476

ABSTRACT

AIMS: The farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily that is highly expressed in liver, kidney, adrenals, and intestine. FXR was previously proposed to play an important role in the pathogenesis of cardiovascular diseases via regulating the metabolism and transport of cholesterol. We have recently shown that FXR is also expressed in rat pulmonary vascular endothelial cells (EC) and that activation of FXR leads to inhibition of endothelin-1 expression. In the present study, we examine whether activation of FXR also affects the expression of endothelial nitric oxide synthase (eNOS) in rat, bovine, and sheep vascular EC. METHODS AND RESULTS: Treatment of vascular EC with a FXR ligand resulted in upregulation of expression of eNOS mRNA and protein and an increased production of nitrite/nitrate. FXR appears to induce eNOS expression at a transcriptional level because (1) upregulation of eNOS mRNA expression was abolished by the treatment of a transcription inhibitor, actinomycin D; and (2) eNOS promoter activity was significantly increased by pharmacological or genetic activation of FXR. Functional analysis of rat eNOS promoter identified an imperfect inverted repeat DNA motif, IR2 (-628AGCTCAgtGGACCT-641), as a likely FXR-responsive element that is involved in eNOS regulation. CONCLUSION: These results support the notion that vascular FXR may serve as a novel molecular target for manipulating the expression of eNOS for the treatment of vascular diseases.


Subject(s)
DNA-Binding Proteins/physiology , Endothelial Cells/enzymology , Gene Expression Regulation, Enzymologic , Nitric Oxide Synthase Type III/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Cattle , Cells, Cultured , DNA/metabolism , DNA-Binding Proteins/agonists , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Promoter Regions, Genetic , RNA, Messenger/analysis , Rats , Receptors, Cytoplasmic and Nuclear/agonists , Response Elements/physiology , Sheep , Transcription Factors/agonists
14.
Circ Res ; 98(2): 192-9, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16357303

ABSTRACT

The farnesoid X receptor (FXR) is a member of the nuclear receptor superfamily that is highly expressed in liver, kidney, adrenals, and intestine. FXR may play an important role in the pathogenesis of cardiovascular diseases via regulating the metabolism and transport of cholesterol. In this study, we report that FXR is also expressed in rat pulmonary artery endothelial cells (EC), a "nonclassical" bile acid target tissue. FXR is functional in EC, as demonstrated by induction of its target genes such as small heterodimer partner (SHP) after treatment with chenodeoxycholic acid, a FXR agonist. Interestingly, activation of FXR in EC led to downregulation of endothelin (ET)-1 expression. Reporter assays showed that activation of FXR inhibited transcriptional activation of the human ET-1 gene promoter and also repressed the activity of a heterologous promoter driven by activator protein (AP)-1 response elements. Electrophoretic mobility-shift and chromatin immunoprecipitation assays indicated that FXR reduced the binding activity of AP-1 transcriptional factors, suggesting that FXR may suppress ET-1 expression via negatively interfering with AP-1 signaling. These studies suggest that FXR may play a role in endothelial homeostasis and may serve as a novel molecular target for manipulating ET-1 expression in vascular EC.


Subject(s)
DNA-Binding Proteins/physiology , Endothelial Cells/metabolism , Endothelin-1/genetics , Gene Expression Regulation , Transcription Factors/physiology , Animals , Cells, Cultured , Chenodeoxycholic Acid/pharmacology , Down-Regulation , Endothelial Cells/chemistry , Humans , Promoter Regions, Genetic , Pulmonary Artery/chemistry , Rabbits , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Response Elements , Transcription Factor AP-1/physiology , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...