Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biol Pharm Bull ; 45(10): 1458-1465, 2022.
Article in English | MEDLINE | ID: mdl-36184503

ABSTRACT

Obesity is currently the most common cause of metabolic diseases including type 2 diabetes and hyperlipidemia. Obesity results from excess lipid accumulation in adipose tissue. Several studies have investigated the inhibitory effects of natural plant-derived products on adipocyte differentiation and lipid accumulation. In this study, we examined the effect of hydrolysable tannins composed of gallic acid and glucose on adipocyte differentiation in 3T3-L1 cells. 1,2,3,4,6-Penta-O-galloyl-ß-D-glucose (PGG) (1), a representative gallotannin, inhibited lipid accumulation in 3T3-L1 cells, whereas ellagitannins (tellimagrandin I, eugeniin and casuarictin) did not. The expression of adipocyte differentiation-related genes, including peroxisome proliferator activator γ2 (Pparγ2), CCAAT/enhancer binding protein α (C/EBPα) and adipocyte fatty acid binding protein (aP2), was significantly suppressed in PGG (1)-treated 3T3-L1 cells beginning at day 2 after induction of differentiation. While PGG (1) did not directly reduce Pparγ2 expression, it reduced the expression of its target genes in mature adipocytes. In addition, PGG (1) treatment inhibited mitotic clonal expansion, one of earliest events of adipocyte differentiation. These findings indicate that PGG (1) has an inhibitory effect on adipocyte differentiation through the suppression of mitotic clonal expansion.


Subject(s)
Diabetes Mellitus, Type 2 , Hydrolyzable Tannins , 3T3-L1 Cells , Adipocytes , Adipogenesis , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Diabetes Mellitus, Type 2/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/pharmacology , Gallic Acid/pharmacology , Glucose/metabolism , Hydrolyzable Tannins/metabolism , Hydrolyzable Tannins/pharmacology , Lipids , Mice , Obesity/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferators/metabolism , Peroxisome Proliferators/pharmacology
2.
J Endocrinol ; 254(2): 121-133, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35662074

ABSTRACT

Adiponectin is a cytokine secreted from adipocytes and regulates metabolism. Although serum adiponectin levels show diurnal variations, it is not clear if the effects of adiponectin are time-dependent. Therefore, this study conducted locomotor activity analyses and various metabolic studies using the adiponectin knockout (APN (-/-)) and the APN (+/+) mice to understand whether adiponectin regulates the circadian rhythm of glucose and lipid metabolism. We observed that the adiponectin gene deficiency does not affect the rhythmicity of core circadian clock genes expression in several peripheral tissues. In contrast, the adiponectin gene deficiency alters the circadian rhythms of liver and serum lipid levels and results in the loss of the time dependency of very-low-density lipoprotein-triglyceride secretion from the liver. In addition, the whole-body glucose tolerance of the APN (-/-) mice was normal at CT10 but reduced at CT22, compared to the APN (+/+) mice. The decreased glucose tolerance at CT22 was associated with insulin hyposecretion in vivo. In contrast, the gluconeogenesis activity was higher in the APN (-/-) mice than in the APN (+/+) mice throughout the day. These results indicate that adiponectin regulates part of the circadian rhythm of metabolism in the liver.


Subject(s)
Adiponectin , Circadian Clocks , Adiponectin/deficiency , Adiponectin/genetics , Adiponectin/metabolism , Animals , Circadian Clocks/genetics , Circadian Rhythm/genetics , Glucose/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Metabolism, Inborn Errors , Mice
3.
Int J Mol Sci ; 19(9)2018 Sep 18.
Article in English | MEDLINE | ID: mdl-30231537

ABSTRACT

Brain and muscle arnt-like protein 1 (BMAL1), is a transcription factor known to regulate circadian rhythm. BMAL1 was originally characterized by its high expression in the skeletal muscle. Since the skeletal muscle is the dominant organ system in energy metabolism, the possible functions of BMAL1 in the skeletal muscle include the control of metabolism. Here, we established that its involvement in the regulation of oxidative capacity in the skeletal muscle. Muscle-specific Bmal1 KO mice (MKO mice) displayed several physiological hallmarks for the increase of oxidative capacity. This included increased energy expenditure and oxygen consumption, high running endurance and resistance to obesity with improved metabolic profiles. Also, the phosphorylation status of AMP-activated protein kinase and its downstream signaling substrate acetyl-CoA carboxylase in the MKO mice were substantially higher than those in the Bmal1flox/flox mice. In addition, biochemical and histological studies confirmed the substantial activation of oxidative fibers in the skeletal muscle of the MKO mice. The mechanism includes the regulation of Cacna1s expression, followed by the activation of calcium-nuclear factor of activated T cells (NFAT) axis. We thus conclude that BMAL1 is a critical regulator of the muscular fatty acid level under nutrition overloading and that the mechanism involves the control of oxidative capacity.


Subject(s)
ARNTL Transcription Factors/genetics , Fats/metabolism , Gene Deletion , Muscle, Skeletal/metabolism , Obesity/genetics , Oxidative Stress , ARNTL Transcription Factors/metabolism , Animals , Diet, High-Fat/adverse effects , Insulin Resistance , Locomotion , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology
4.
Neurosci Lett ; 677: 26-31, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29680250

ABSTRACT

It has been shown that the incidence of cognitive impairment increases with the severity of chronic kidney disease (CKD). A previous study has demonstrated that hippocampal oxidative stress contributes to cognitive dysfunction in CKD model mice. Endoplasmic reticulum (ER) stress is thought to contribute significantly to neuronal dysfunction, but its role in the hippocampal dysfunction seen in CKD still remains unclear. The present study examined whether the ER stress response as well as oxidative stress was activated in the hippocampus of CKD model mice. Western blotting revealed that the expression level of 4-hydroxy-2-nonenal (HNE)-protein adducts, a marker of oxidative stress, was increased in the hippocampus 8 weeks after 5/6 nephrectomy. In these mice, the expression level of glucose-regulated protein 78 (GRP78), a typical ER stress marker, also showed a pronounced increase in the hippocampus. Correlation analyses showed that the levels of these two marker proteins in the hippocampus are positively correlated with the serum concentrations of BUN and creatinine. These results suggest that ER stress as well as oxidative stress are induced in the hippocampus of CKD mice and that the levels of these stress markers in the hippocampus are correlated with the renal impairment caused by CKD.


Subject(s)
Endoplasmic Reticulum Stress , Hippocampus/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Male , Mice, Inbred C57BL , Oxidative Stress
5.
Biochem Biophys Res Commun ; 496(1): 7-11, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29274785

ABSTRACT

Hepatocellular carcinoma (HCC) is a disease with poor prognosis. Nuclear accumulation of YB-1 is closely related to the malignancy of HCC. Treatment with anticancer agents often induces translocation of YB-1 from cytoplasm to nucleus and activates the expression of multidrug resistance gene 1 (MDR1). Therefore, any effective inhibitor of this phenomenon would be useful for cancer treatment. Here we examined various indirubin derivatives and found that indirubin 3'-oxime inhibits actinomycin D-induced nuclear transport of YB-1 and suppresses the activation of MDR1 gene expression in the human hepatocellular carcinoma cell line HepG2. Furthermore, use of both indirubin 3'-oxime and actinomycin D in combination increased the anticancer effect on HepG2 cells. Indirubin 3'-oxime is a novel and efficient inhibitor of anticancer agent-induced YB-1 nuclear translocation.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Indoles/administration & dosage , Oximes/administration & dosage , Y-Box-Binding Protein 1/metabolism , Active Transport, Cell Nucleus/drug effects , Angiogenesis Inhibitors/administration & dosage , Carcinoma, Hepatocellular/pathology , Cell Nucleus/pathology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Hep G2 Cells , Humans
6.
J Pharmacol Sci ; 130(3): 185-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27032909

ABSTRACT

S-allyl-l-cysteine (SAC) is known to have neuroprotective properties. We synthesized various SAC derivatives and tested their effects on endoplasmic reticulum stress-induced neurotoxicity in cultured hippocampal neurons (HPNs). Among the compounds tested, S-propyl-l-cysteine (SPC) exhibited the strongest neuroprotective activity in HPNs, followed by S-ethyl-l-cysteine (SEC) and S-methyl-l-cysteine (SMC). Unlike SAC and SMC, SPC and SEC did not have inhibitory activity on µ-calpain, suggesting that the mechanism underlying the protective activity of SPC and SEC differs from that of SAC.


Subject(s)
Calpain/antagonists & inhibitors , Cysteine/analogs & derivatives , Endoplasmic Reticulum Stress/drug effects , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents , Animals , Cells, Cultured , Cysteine/pharmacology , Endoplasmic Reticulum Stress/physiology , Hippocampus/cytology , Rats, Wistar
7.
J Nat Med ; 70(3): 502-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26880616

ABSTRACT

As obesity is a global health concern the demand for anti-obesity drugs is high. In this study, we investigated the anti-obesity effect of the dried branches and leaves of murta (Myrceugenia euosma Legrand, Myrtaceae). A methanol extract of the dried branches and leaves of murta inhibited adipogenesis in 3T3-L1 cells. Three known flavanones-cryptostrobin (1), pinocembrin (4), and 5,7-dihydroxy-6,8-dimethylflavanone (6), and three chalcones-2',6'-dihydroxy-3'-methyl-4'-methoxychalcone (2), pinostrobin chalcone (3), and 2',6'-dihydroxy-4'-methoxy-3',5'-dimethylchalcone (5) were isolated from the active fraction. Structures of these compounds were identified using various spectral data. Each of these compounds also inhibited adipogenesis in 3T3-L1 cells. In particular, compound 3 was a more potent inhibitor of triglyceride accumulation than the positive control berberine. Gene expression studies revealed that treatment of 3T3-L1 cells with 3 lowers the expression levels of CCAAT/enhancer-binding protein α and peroxisome proliferator activator γ2 during adipogenesis without affecting cell viability. Treatment of 3T3-L1 cells with 3 reduced the expression levels of mRNAs encoding sterol regulatory element-binding protein 1c and several lipogenic enzymes, including fatty acid synthase and stearoyl CoA desaturase-1. These results indicate that the methanol extract and compounds isolated from the dried branches and leaves of murta exert their anti-obesity effects through the inhibition of adipogenesis.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Anti-Obesity Agents/pharmacology , Eugenia/chemistry , Lipid Metabolism/drug effects , Plant Extracts/pharmacology , 3T3-L1 Cells , Animals , Cell Differentiation/drug effects , Mice , Plant Leaves/chemistry
8.
J Biol Chem ; 291(13): 7004-16, 2016 Mar 25.
Article in English | MEDLINE | ID: mdl-26865635

ABSTRACT

Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor regulating the expression of genes involved in xenobiotic response. Recent studies have suggested that AhR plays essential roles not only in xenobiotic detoxification but also energy metabolism. Thus, in this study, we studied the roles of AhR in lipid metabolism. Under high fat diet (HFD) challenge, liver-specific AhR knock-out (AhR LKO) mice exhibited severe steatosis, inflammation, and injury in the liver. Gene expression analysis and biochemical study revealed thatde novolipogenesis activity was significantly increased in AhR LKO mice. In contrast, induction of suppressor of cytokine signal 3 (Socs3) expression by HFD was attenuated in the livers of AhR LKO mice. Rescue of theSocs3gene in the liver of AhR LKO mice cancelled the HFD-induced hepatic lipotoxicities. Promoter analysis established Socs3 as novel transcriptional target of AhR. These results indicated that AhR plays a protective role against HFD-induced hepatic steatosis and the subsequent lipotoxicity effects, such as inflammation, and that the mechanism of protection involves the direct transcriptional regulation ofSocs3expression by AhR.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Diet, High-Fat , Dietary Fats/adverse effects , Fatty Liver/genetics , Receptors, Aryl Hydrocarbon/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Binding Sites , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation , Lipid Metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Receptors, Aryl Hydrocarbon/deficiency , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription, Genetic
9.
Mol Cell Biol ; 34(9): 1682-94, 2014 May.
Article in English | MEDLINE | ID: mdl-24567372

ABSTRACT

Estrogen sulfotransferase (EST/SULT1E1) is known to catalyze the sulfoconjugation and deactivation of estrogens. The goal of this study is to determine whether and how EST plays a role in human adipogenesis. By using human primary adipose-derived stem cells (ASCs) and whole-fat tissues from the abdominal subcutaneous fat of obese and nonobese subjects, we showed that the expression of EST was low in preadipocytes but increased upon differentiation. Overexpression and knockdown of EST in ASCs promoted and inhibited differentiation, respectively. The proadipogenic activity of EST in humans was opposite to the antiadipogenic effect of the same enzyme in rodents. Mechanistically, EST promoted adipogenesis by deactivating estrogens. The proadipogenic effect of EST can be recapitulated by using an estrogen receptor (ER) antagonist or ERα knockdown. In contrast, activation of ER in ASCs inhibited adipogenesis by decreasing the recruitment of the adipogenic peroxisome proliferator-activated receptor γ (PPARγ) onto its target gene promoters, whereas ER antagonism increased the recruitment of PPARγ to its target gene promoters. Linear regression analysis revealed a positive correlation between the expression of EST and body mass index (BMI), as well as a negative correlation between ERα expression and BMI. We conclude that EST is a proadipogenic factor which may serve as a druggable target to inhibit the turnover and accumulation of adipocytes in obese patients.


Subject(s)
Adipogenesis , Sulfotransferases/genetics , Sulfotransferases/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adiposity , Adult , Cells, Cultured , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Sulfotransferases/antagonists & inhibitors , Up-Regulation
10.
Mol Pharmacol ; 83(5): 1133-40, 2013 May.
Article in English | MEDLINE | ID: mdl-23478803

ABSTRACT

A member of the NADPH oxidase subunits, p40(phox) plays an important role in the regulation of NADPH oxidase activity and the subsequent production of reactive oxygen species (ROS). In this study, we show that mouse p40(phox) is a novel transcriptional target of the aryl hydrocarbon receptor (AhR), known as a dioxin receptor or xenobiotic receptor, in the liver. Treatment of mice with 3-methylcholanthrene (3MC) increased p40(phox) gene expression in the liver, but this induction of p40(phox) gene expression was diminished by the deletion of the AhR gene in the liver. Consistent with the in vivo results, the expression of the p40(phox) gene was increased in 3MC-treated Hepa1c1c7 cells in an AhR-dependent manner. In addition, promoter analysis established p40(phox) as a transcriptional target of AhR. Studies using the RNA-interference technique revealed that p40(phox) is involved in the increase of NADPH oxidase activity and the subsequent ROS production in AhR-activated Hepa1c1c7 cells. Consequently, the results obtained here may provide a novel molecular mechanism for ROS production after exposure to dioxins.


Subject(s)
NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Line, Tumor , Gene Expression Regulation , Hep G2 Cells , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/metabolism , Promoter Regions, Genetic , Reactive Oxygen Species/metabolism , Transcription, Genetic
11.
Drug Metab Dispos ; 41(1): 1-11, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23043185

ABSTRACT

This article is a report on a symposium sponsored by the American Society for Pharmacology and Experimental Therapeutics and held at the Experimental Biology 12 meeting in San Diego, CA. The presentations discussed the roles of a number of nuclear receptors in regulating glucose and lipid homeostasis, the pathophysiology of obesity-related disease states, and the promise associated with targeting their activities to treat these diseases. While many of these receptors (in particular, constitutive androstane receptor and pregnane X receptor) and their target enzymes have been thought of as regulators of drug and xenobiotic metabolism, this symposium highlighted the advances made in our understanding of the endogenous functions of these receptors. Similarly, as we gain a better understanding of the mechanisms underlying bile acid signaling pathways in the regulation of body weight and glucose homeostasis, we see the importance of using complementary approaches to elucidate this fascinating network of pathways. The observation that some receptors, like the farnesoid X receptor, can function in a tissue-specific manner via well defined mechanisms has important clinical implications, particularly in the treatment of liver diseases. Finally, the novel findings that agents that selectively activate estrogen receptor ß can effectively inhibit weight gain in a high-fat diet model of obesity identifies a new role for this member of the steroid superfamily. Taken together, the significant findings reported during this symposium illustrate the promise associated with targeting a number of nuclear receptors for the development of new therapies to treat obesity and other metabolic disorders.


Subject(s)
Lipids/physiology , Obesity/physiopathology , Receptors, Cytoplasmic and Nuclear/physiology , Bile Acids and Salts/metabolism , Energy Metabolism , Glucose/metabolism , Humans , Receptors, Cytoplasmic and Nuclear/metabolism , Xenobiotics/metabolism
12.
Mol Endocrinol ; 25(9): 1612-23, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21816900

ABSTRACT

The estrogen sulfotransferase (EST) is a phase II drug-metabolizing enzyme known to catalyze the sulfoconjugation of estrogens. EST is highly expressed in the white adipose tissue of male mice, but the role of EST in the development and function of adipocytes remains largely unknown. In this report, we showed that EST played an important role in adipocyte differentiation. EST was highly expressed in 3T3-L1 preadipocytes and primary mouse preadipocytes. The expression of EST was dramatically reduced in differentiated 3T3-L1 cells and mature primary adipocytes. Overexpression of EST in 3T3-L1 cells prevented adipocyte differentiation. In contrast, preadipocytes isolated from EST knockout (EST-/-) mice exhibited enhanced differentiation. The inhibitory effect of EST on adipogenesis likely resulted from the sustained activation of ERK1/2 MAPK and inhibition of insulin signaling, leading to a failure of switch from clonal expansion to differentiation. The enzymatic activity of EST was required for the inhibitory effect of EST on adipogenesis, because an enzyme-dead EST mutant failed to inhibit adipocyte differentiation. In vivo, overexpression of EST in the adipose tissue of female transgenic mice resulted in smaller adipocyte size. Taken together, our results suggest that EST functions as a negative regulator of adipogenesis.


Subject(s)
Adipocytes/cytology , Adipocytes/enzymology , Cell Differentiation , Sulfotransferases/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Adipose Tissue/drug effects , Adipose Tissue/enzymology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Size/drug effects , Enzyme Activation/drug effects , Estradiol/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Deletion , Gene Expression Regulation, Enzymologic/drug effects , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , PPAR gamma/agonists , PPAR gamma/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Sulfotransferases/genetics
13.
Physiol Genomics ; 43(13): 818-28, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21540300

ABSTRACT

Retinoid-related orphan receptor (ROR)α4 is the major RORα isoform expressed in adipose tissues and liver. In this study we demonstrate that RORα-deficient staggerer mice (RORα(sg/sg)) fed with a high-fat diet (HFD) exhibited reduced adiposity and hepatic triglyceride levels compared with wild-type (WT) littermates and were resistant to the development of hepatic steatosis, adipose-associated inflammation, and insulin resistance. Gene expression profiling showed that many genes involved in triglyceride synthesis and storage, including Cidec, Cidea, and Mogat1, were expressed at much lower levels in liver of RORα(sg/sg) mice. In contrast, overexpression of RORα in mouse hepatoma Hepa1-6 cells significantly increased the expression of genes that were repressed in RORα(sg/sg) liver, including Sult1b1, Adfp, Cidea, and ApoA4. ChIP and promoter analysis suggested that several of these genes were regulated directly by RORα. In addition to reduced lipid accumulation, inflammation was greatly diminished in white adipose tissue (WAT) of RORα(sg/sg) mice fed with an HFD. The infiltration of macrophages and the expression of many immune response and proinflammatory genes, including those encoding various chemo/cytokines, Toll-like receptors, and TNF signaling proteins, were significantly reduced in RORα(sg/sg) WAT. Moreover, RORα(sg/sg) mice fed with an HFD were protected from the development of insulin resistance. RORα(sg/sg) mice consumed more oxygen and produced more carbon dioxide, suggesting increased energy expenditure in this genotype. Our study indicates that RORα plays a critical role in the regulation of several aspects of metabolic syndrome. Therefore, RORα may provide a novel therapeutic target in the management of obesity and associated metabolic diseases.


Subject(s)
Fatty Liver/genetics , Gene Expression Profiling , Gene Expression Regulation , Inflammation/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/deficiency , Obesity/genetics , Transcription, Genetic , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Aging/genetics , Aging/pathology , Animals , Dietary Fats , Energy Metabolism/genetics , Fatty Liver/complications , Fatty Liver/pathology , Glucose Intolerance/complications , Glucose Intolerance/genetics , Inflammation/complications , Inflammation/pathology , Insulin Resistance/genetics , Lipogenesis/genetics , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Obesity/complications , Obesity/pathology
14.
Mol Endocrinol ; 25(4): 584-96, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21310851

ABSTRACT

The nuclear receptor liver X receptor (LXR) is a ligand-dependent transcription factor that plays an important role in the metabolism and homeostasis of cholesterol, lipids, bile acids, and steroid hormones. MicroRNAs (miRNAs) are recently recognized important negative regulators of gene expression. In this report, we showed that miRNA hsa-miR-613 played an important role in the autoregulation of the human LXRα gene. hsa-miR-613 targeted the endogenous LXRα through its specific miRNA response element (613MRE) within the LXRα 3'-untranslated region. Interestingly and paradoxically, the expression of hsa-miR-613 itself was induced upon the activation of LXR. However, hsa-miR-613 did not appear to be a direct LXR target gene. Instead, the positive regulation of hsa-miR-613 by LXR was mediated by the sterol regulatory element binding protein (SREBP)-1c, a known LXR target gene. Promoter analysis revealed an SREBP response element in the hsa-miR-613 gene promoter. Treatment with insulin also induced the expression of hsa-miR-613 in an SREBP-1c-dependent manner, further supporting the role of SREBP-1c in the positive regulation of this miRNA species. Finally, the autoinduction of LXRα by a LXR agonist was enhanced when hsa-miR-613 was inhibited or SREBP-1c was down-regulated. hsa-miR-613 appeared to specifically target the human LXRα. We propose that the negative regulation mediated by hsa-miR-613 and SREBP-1c and the previously reported positive regulation mediated by an LXR response element in the LXRα gene promoter constitute a ying-yang mechanism to ensure a tight regulation of this nuclear receptor of many metabolic functions.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , 3' Untranslated Regions/genetics , Blotting, Western , Electrophoretic Mobility Shift Assay , Feedback, Physiological , Gene Silencing , Hep G2 Cells , Humans , Insulin/metabolism , Liver X Receptors , Polymerase Chain Reaction , Promoter Regions, Genetic , RNA, Small Interfering , Response Elements/genetics , Signal Transduction , Sterol Regulatory Element Binding Protein 1/genetics
15.
Diabetes ; 60(1): 177-88, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20864514

ABSTRACT

OBJECTIVE: The nuclear receptor TAK1/TR4/NR2C2 is expressed in several tissues that are important in the control of energy homeostasis. In this study, we investigate whether TAK1 functions as a regulator of lipid and energy homeostasis and has a role in metabolic syndrome. RESEARCH DESIGN AND METHODS: We generated TAK1-deficient (TAK1⁻(/)⁻) mice to study the function of TAK1 in the development of metabolic syndrome in aged mice and mice fed a high-fat diet (HFD). (Immuno)histochemical, biochemical, and gene expression profile analyses were performed to determine the effect of the loss of TAK1 expression on lipid homeostasis in liver and adipose tissues. In addition, insulin sensitivity, energy expenditure, and adipose-associated inflammation were compared in wild-type (WT) and TAK1⁻(/)⁻ mice fed a HFD. RESULTS: TAK1-deficient (TAK1⁻(/)⁻) mice are resistant to the development of age- and HFD-induced metabolic syndrome. Histo- and biochemical analyses showed significantly lower hepatic triglyceride levels and reduced lipid accumulation in adipose tissue in TAK1⁻(/)⁻ mice compared with WT mice. Gene expression profiling analysis revealed that the expression of several genes encoding proteins involved in lipid uptake and triglyceride synthesis and storage, including Cidea, Cidec, Mogat1, and CD36, was greatly decreased in the liver and primary hepatocytes of TAK1⁻(/)⁻ mice. Restoration of TAK1 expression in TAK1⁻(/)⁻ hepatocytes induced expression of several lipogenic genes. Moreover, TAK1⁻(/)⁻ mice exhibited reduced infiltration of inflammatory cells and expression of inflammatory genes in white adipose tissue, and were resistant to the development of glucose intolerance and insulin resistance. TAK1⁻(/)⁻ mice consume more oxygen and produce more carbon dioxide than WT mice, suggesting increased energy expenditure. CONCLUSIONS: Our data reveal that TAK1 plays a critical role in the regulation of energy and lipid homeostasis, and promotes the development of metabolic syndrome. TAK1 may provide a new therapeutic target in the management of obesity, diabetes, and liver steatosis.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Fatty Liver/prevention & control , Inflammation/prevention & control , Obesity/complications , Receptors, Steroid/deficiency , Receptors, Thyroid Hormone/deficiency , Adipose Tissue/anatomy & histology , Adipose Tissue/pathology , Animals , Dietary Fats , Epididymis , Fatty Liver/pathology , Flow Cytometry , Inflammation/pathology , Insulin Resistance , Male , Metabolic Syndrome/genetics , Metabolic Syndrome/pathology , Metabolic Syndrome/prevention & control , Mice , Mice, Knockout , Organ Size , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
16.
Gastroenterology ; 139(2): 653-63, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20303349

ABSTRACT

BACKGROUND & AIMS: The aryl hydrocarbon receptor (AhR) also known as the dioxin receptor or xenobiotic receptor is a member of the basic helix-loop-helix/period AhR nuclear translocator single minded family. The goal of this study was to determine the endobiotic role of AhR in hepatic steatosis. METHODS: Wild-type, constitutively activated AhR transgenic, AhR null and CD36/fatty acid translocase null mice were used to investigate the role of AhR in steatosis and the involvement of CD36 in the steatotic effect of AhR. The promoters of the mouse and human CD36 genes were cloned and their regulation by AhR was analyzed. RESULTS: Activation of AhR induced spontaneous hepatic steatosis characterized by the accumulation of triglycerides. The steatotic effect of AhR likely is owing to the combined up-regulation of CD36 and fatty acid transport proteins, suppression of fatty acid oxidation, inhibition of hepatic export of triglycerides, increase in peripheral fat mobilization, and increased hepatic oxidative stress. Promoter analysis established CD36 as a novel transcriptional target of AhR. Activation of AhR in liver cells induced CD36 gene expression and enhanced fatty acid uptake. The steatotic effect of an AhR agonist was inhibited in CD36-/- mice. CONCLUSIONS: Our study reveals a novel link between AhR-induced steatosis and the expression of CD36. Industrial or military exposures to dioxin and related compounds have been linked to increased prevalence of fatty liver in human beings. Results from this study may help to establish AhR and its target CD36 as novel therapeutic and preventive targets for fatty liver disease.


Subject(s)
CD36 Antigens/metabolism , Fatty Acids/metabolism , Fatty Liver/metabolism , Liver/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Adiposity , Animals , Basic Helix-Loop-Helix Transcription Factors , Body Weight , CD36 Antigens/genetics , Cell Line , Fatty Acid Transport Proteins/metabolism , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/physiopathology , Female , Humans , Liver/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oxidation-Reduction , Oxidative Stress , Polychlorinated Dibenzodioxins/toxicity , Promoter Regions, Genetic , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Transfection , Triglycerides/metabolism
17.
Mol Endocrinol ; 24(5): 923-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20203100

ABSTRACT

The retinoic acid-related orphan receptor gamma (RORgamma) has important roles in development and metabolic homeostasis. Although the biological functions of RORgamma have been studied extensively, no ligands for RORgamma have been identified, and no structure of RORgamma has been reported. In this study, we showed that hydroxycholesterols promote the recruitment of coactivators by RORgamma using biochemical assays. We also report the crystal structures of the RORgamma ligand-binding domain bound with hydroxycholesterols. The structures reveal the binding modes of various hydroxycholesterols in the RORgamma pocket, with the receptors all adopting the canonical active conformation. Mutations that disrupt the binding of hydroxycholesterols abolish the constitutive activity of RORgamma. Our observations suggest an important role for the endogenous hydroxycholesterols in modulating RORgamma-dependent biological processes.


Subject(s)
Hydroxycholesterols/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Hydroxycholesterols/chemistry , Hydroxycholesterols/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Protein Binding/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology
18.
J Biol Chem ; 284(38): 25984-92, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19617349

ABSTRACT

Obesity and type 2 diabetes are related metabolic disorders of high prevalence. The constitutive androstane receptor (CAR) was initially characterized as a xenobiotic receptor regulating the responses of mammals to xenotoxicants. In this study, we have uncovered an unexpected role of CAR in preventing obesity and alleviating type 2 diabetes. Using a high fat diet (HFD)-induced obesity model, we showed that treatment of wild type mice with the CAR agonist 1,4-bis[2-(3,5 dichloropyridyloxy)] benzene (TCPOBOP) efficiently prevented obesity from happening or reversed preinduced obesity. Treatment with TCPOBOP improved insulin sensitivity in both the HFD-induced type 2 diabetic model and the ob/ob mice. In contrast, CAR null mice maintained on a chow diet showed spontaneous insulin insensitivity, which cannot be relieved by TOPOBOP treatment. The hepatic steatosis in HFD-treated mice and ob/ob mice was markedly reduced by the TCPOBOP treatment. The metabolic benefits of CAR activation may have resulted from the combined effect of inhibition of lipogenesis, very low density lipoprotein secretion and export of triglycerides, and gluconeogenesis as well as increases in brown adipose tissue energy expenditure and peripheral fat mobilization. Moreover, the skeletal muscle of CAR-activated mice showed a decreased incomplete oxidation, despite having a lower expression level of peroxisome proliferator-activated receptor alpha and its target genes involved in fatty acid oxidation. In summary, our results have revealed an important metabolic function of CAR and may establish this "xenobiotic receptor" as a novel therapeutic target for the prevention and treatment of obesity and type 2 diabetes.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance , Muscle, Skeletal/metabolism , Obesity/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Constitutive Androstane Receptor , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/prevention & control , Diet , Dietary Fats/adverse effects , Disease Models, Animal , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Acids/genetics , Fatty Acids/metabolism , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/prevention & control , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Humans , Lipoproteins, VLDL/metabolism , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/genetics , Obesity/prevention & control , Oxidation-Reduction/drug effects , PPAR alpha/genetics , PPAR alpha/metabolism , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Triglycerides/metabolism
19.
Trends Endocrinol Metab ; 20(6): 273-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19595610

ABSTRACT

The nuclear receptors pregnane X receptor (PXR, or NR1I2) and constitutive androstane receptor (CAR, or NR1I3) were originally identified as xenosensors that regulate the expression of Phase I and Phase II drug-metabolizing enzymes and transporters. Recent results suggest that PXR and CAR also have important endobiotic roles in energy metabolism by affecting the metabolism of fatty acids, lipids and glucose. PXR and CAR exert their effects on energy metabolism through direct gene regulation or through crosstalk with other transcriptional regulators. This review focuses on the roles of CAR and PXR in energy metabolism and offers a perspective on whether PXR and CAR represent novel therapeutic targets for the management of metabolic syndrome.


Subject(s)
Energy Metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Animals , Constitutive Androstane Receptor , Glucose/metabolism , Homeostasis , Humans , Lipid Metabolism , Pregnane X Receptor , Transcription Factors/metabolism , Xenobiotics/metabolism
20.
Mol Pharmacol ; 76(3): 604-11, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19542321

ABSTRACT

Aldo-keto reductase (AKR) family 1, member 7 (AKR1B7), a member of the AKR superfamily, has been suggested to play an important role in the detoxification of lipid peroxidation by-products. The nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) are xenosensors postulated to alleviate xeno- and endobiotic chemical insults. In this study, we show that the mouse Akr1b7 is a shared transcriptional target of PXR and CAR in the liver and intestine. Treatment of wild-type mice with the PXR agonist pregnenolone-16alpha-carbonitrile (PCN) activated Akr1b7 gene expression, whereas the effect was abrogated in PXR(-/-) mice. Similarly, the activation of Akr1b7 gene expression by the CAR agonist 1,4-bis[2-(3,5-dichlorpyridyloxyl)]-benzene, seen in wild-type mice, was abolished in CAR(-/-) mice. The promoter of Akr1b7 gene was activated by PXR and CAR, and this activation was achieved through the binding of PXR-retinoid X receptor (RXR) or CAR-RXR heterodimers to direct repeat-4 type nuclear receptor-binding sites found in the Akr1b7 gene promoter. At the functional level, treatment with PCN in wild-type mice, but not PXR(-/-) mice, led to a decreased intestinal accumulation of malondialdehyde, a biomarker of lipid peroxidation. The regulation of Akr1b7 by PXR was independent of the liver X receptor (LXR), another nuclear receptor known to regulate this AKR isoform. Because a major function of Akr1b7 is to detoxify lipid peroxidation, the PXR-, CAR-, and LXR-controlled regulatory network of Akr1b7 may have contributed to alleviate toxicity associated with lipid peroxidation.


Subject(s)
Aldehyde Reductase/genetics , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Animals , Constitutive Androstane Receptor , Lipid Peroxidation , Mice , Mice, Mutant Strains , Pregnane X Receptor , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/agonists , Receptors, Steroid/genetics , Response Elements , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...