Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Pineal Res ; 68(3): e12638, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32053237

ABSTRACT

Alcoholic liver disease is the most prevalent chronic liver disease. Melatonin is known to control many vital processes. Here, we explored a novel molecular mechanism by which melatonin-induced SIRT1 signaling protects against alcohol-mediated oxidative stress and liver injury. Gene expression profiles and metabolic changes were measured in liver specimens of mice and human subjects. Expression levels of Cb1r, Crbn, Btg2, Yy1, pro-inflammatory cytokines, and Cyp2e1 were significantly enhanced in chronic alcohol-challenged mice and human subjects. Levels of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), hepatic CYP2E1 protein, and reactive oxygen species (ROS) were elevated in alcohol-fed WT mice but not in Cb1r antagonist-treated, Crbn null, or Yy1-silenced mice. Importantly, alcohol-induced Yy1 and Cyp2e1 expression, ROS amount, and liver injury were markedly diminished by melatonin treatment and the transduction of Sirt1 in mice, whereas this phenomenon was prominently ablated by silencing of Sirt1. Notably, SIRT1 physically interacted with YY1 and attenuated YY1 occupancy on the Cyp2e1 gene promoter. Melatonin-SIRT1 signaling ameliorates alcohol-induced oxidative liver injury by disrupting the CRBN-YY1-CYP2E1 signaling pathway. The manipulation of CRBN-YY1-CYP2E1 signaling network by the melatonin-SIRT1 pathway highlights a novel entry point for treating alcoholic liver disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytochrome P-450 CYP2E1/metabolism , Liver Diseases, Alcoholic/metabolism , Melatonin/metabolism , Sirtuin 1/metabolism , Ubiquitin-Protein Ligases/metabolism , YY1 Transcription Factor/metabolism , Animals , Humans , Mice , Oxidative Stress/physiology , Signal Transduction/physiology
2.
J Pineal Res ; 62(2)2017 Mar.
Article in English | MEDLINE | ID: mdl-28095641

ABSTRACT

Alcoholic liver disease is a major cause of chronic liver disease worldwide, and cannabinoid receptor type 1 (CB1R) is involved in a diverse metabolic diseases. B-cell translocation gene 2 (BTG2) and yin yang 1 (YY1) are a potent regulator of biological conditions. Melatonin plays a crucial role in regulating diverse physiological functions and metabolic homeostasis. MicroRNAs are key regulators of various biological processes. Herein, we demonstrate that melatonin improves bile acid synthesis in the liver of alcohol-fed mice by controlling miR-497 expression. The level of bile acid and the expression of Cb1r, Btg2, Yy1, and bile acid synthetic enzymes were significantly elevated in the livers of Lieber-DeCarli alcohol-fed mice. The overexpression of Btg2 enhanced Yy1 gene expression and bile acid production, whereas disrupting the CB1R-BTG2-YY1 cascade protected against the bile acid synthesis caused by alcohol challenge. We identified an alcohol-mediated YY1 binding site on the cholesterol 7α-hydroxylase (Cyp7a1) gene promoter using promoter deletion analysis and chromatin immunoprecipitation assays. Notably, melatonin attenuated the alcohol-stimulated induction of Btg2, Yy1 mRNA levels and bile acid production by promoting miR-497. Overexpression of a miR-497 mimic dramatically diminished the increase of Btg2 and Yy1 gene expression as well as bile acid production by alcohol, whereas this phenomenon was reversed by miR-497 inhibitor. These results demonstrate that the upregulation of miR-497 by melatonin represses alcohol-induced bile acid synthesis by attenuating the BTG2-YY1 signaling pathway. The melatonin-miR497 signaling network may provide novel therapeutic targets for the treatment of hepatic metabolic dysfunction caused by the alcohol-dependent pathway.


Subject(s)
Antioxidants/pharmacology , Bile Acids and Salts/biosynthesis , Liver Diseases, Alcoholic/metabolism , Melatonin/pharmacology , MicroRNAs/biosynthesis , Animals , Blotting, Western , Chromatin Immunoprecipitation , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factor TFIIH/metabolism , YY1 Transcription Factor/metabolism
3.
Dis Aquat Organ ; 127(1): 41-47, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29256426

ABSTRACT

A 5 yr old, 184 kg, and 262 cm total length female bottlenose dolphin Tursiops truncatus was found dead in a display after bloody discharge from the blowhole was observed 3 h prior to death. Pathological examination revealed fibrinous bronchopneumonia with prominent areas of necrosis (sequestra) and numerous Gram-negative bacilli within alveoli and in blood vessels of the lungs and liver and between muscle fibers. The cause of death was attributed to septicemia. Often, cases of fibrinous bronchopneumonia are characterized by bacteremia in the latter stages of infection, resulting in the death of the animal. Septicemia likely accounts for the ecchymoses and petechiae noted on the spleen, pancreas, forestomach, lungs, visceral peritoneum, and small intestine. Additional lesions included hemothorax, stable red frothy fluid in the trachea, and lymphoid depletion in the spleen and lymph nodes. Pure growth of Morganella morganii was isolated from the lungs, blood, liver, and blowhole mucosa. Sequencing of 16s rRNA of the isolated bacteria showed more than 99.6% identity with M. morganii strain FDAARGOS_172. To our knowledge, this is the first report of fatal fibrinonecrotizing bronchopneumonia associated with M. morganii infection in a cetacean.


Subject(s)
Bottle-Nosed Dolphin , Bronchopneumonia/veterinary , Enterobacteriaceae Infections/veterinary , Morganella morganii/isolation & purification , Animals , Bronchopneumonia/microbiology , Bronchopneumonia/pathology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Fatal Outcome , Female
4.
Biochim Biophys Acta ; 1852(12): 2662-70, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26410577

ABSTRACT

Alcohol consumption exacerbates alcoholic liver disease by attenuating the activity of AMP-activated protein kinase (AMPK). AMPK is activated by fenofibrate, a peroxisome proliferator-activated receptor α (PPARα) agonist, and inhibited by direct interaction with cereblon (CRBN), a component of an E3 ubiquitin ligase complex. Based on these preliminary findings, we investigated that CRBN would be up-regulated in the liver by alcohol consumption and that CRBN deficiency would ameliorate hepatic steatosis and pro-inflammatory responses in alcohol-fed mice by increasing AMPK activity. Wild-type, CRBN and PPARα null mice were fed an alcohol-containing liquid diet and administered with fenofibrate. Gene expression profiles and metabolic changes were measured in the liver and blood of these mice. Expression of CRBN, cytochrome P450 2E1 (CYP2E1), lipogenic genes, pro-inflammatory cytokines, serum alanine aminotransferase (ALT), and aspartate aminotransferase (AST) were increased in the Lieber-DeCarli alcohol-challenged mice. Fenofibrate attenuated the induction of CRBN and reduced hepatic steatosis and pro-inflammatory markers in these mice. Ablation of the gene encoding CRBN produced the same effect as fenofibrate. The increase in CRBN gene expression by alcohol and the reduction of CRBN expression by fenofibrate were negated in PPARα null mice. Fenofibrate increased the recruitment of PPARα on CRBN gene promoter in WT mice but not in PPARα null mice. Silencing of AMPK prevented the beneficial effects of fenofibrate. These results demonstrate that activation of PPARα by fenofibrate alleviates alcohol-induced hepatic steatosis and inflammation by reducing the inhibition of AMPK by CRBN. CRBN is a potential therapeutic target for the alcoholic liver disease.

5.
Biochem J ; 467(3): 453-60, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25695641

ABSTRACT

Insulin-like growth factor (IGF)-binding protein-2 (IGFBP-2), one of the most abundant circulating IGFBPs, is known to attenuate the biological action of IGF-1. Although the effect of IGFBP-2 in preventing metabolic disorders is well known, its regulatory mechanism remains unclear. In the present study, we demonstrated the transcriptional regulation of the Igfbp-2 gene by peroxisome-proliferator-activated receptor (PPAR) α in the liver. During fasting, both Igfbp-2 and PPARα expression levels were increased. Wy14643, a selective PPARα agonist, significantly induced Igfbp-2 gene expression in primary cultured hepatocytes. However, Igfbp-2 gene expression in Pparα null mice was not affected by fasting or Wy14643. In addition, through transient transfection and chromatin immunoprecipitation assay in fasted livers, we determined that PPARα bound to the putative PPAR-responsive element between -511 bp and -499 bp on the Igfbp-2 gene promoter, indicating that the Igfbp-2 gene transcription is activated directly by PPARα. To explore the role of PPARα in IGF-1 signalling, we treated primary cultured hepatocytes with Wy14643 and observed a decrease in the number of IGF-1 receptors (IGF-1Rs) and in Akt phosphorylation. No inhibition was observed in the hepatocytes isolated from Pparα null mice. These results suggest that PPARα controls IGF-1 signalling through the up-regulation of hepatic Igfbp-2 transcription during fasting and Wy14643 treatment.


Subject(s)
Fasting/metabolism , Insulin-Like Growth Factor Binding Protein 2/genetics , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Insulin-Like Growth Factor I/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR alpha/deficiency , PPAR alpha/genetics , PPAR gamma/agonists , Peroxisome Proliferators/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rosiglitazone , Signal Transduction , Thiazolidinediones/pharmacology , Up-Regulation/drug effects
6.
J Biol Chem ; 289(2): 1079-91, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24265317

ABSTRACT

Small heterodimer partner interacting leucine zipper protein (SMILE) has been identified as a nuclear corepressor of the nuclear receptor (NRs) family. Here, we examined the role of SMILE in the regulation of nuclear receptor liver X receptor (LXR)-mediated sterol regulatory element binding protein-1c (SREBP-1c) gene expression. We found that SMILE inhibited T0901317 (T7)-induced transcriptional activity of LXR, which functions as a major regulator of lipid metabolism by inducing SREBP-1c, fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC) gene expression. Moreover, we demonstrated that SMILE physically interacts with LXR and represses T7-induced LXR transcriptional activity by competing with coactivator SRC-1. Adenoviral overexpression of SMILE (Ad-SMILE) attenuated fat accumulation and lipogenic gene induction in the liver of T7 administered or of high fat diet (HFD)-fed mice. Furthermore, we investigated the mechanism by which ursodeoxycholic acid (UDCA) inhibits LXR-induced lipogenic gene expression. Interestingly, UDCA treatment significantly increased SMILE promoter activity and gene expression in an adenosine monophosphate-activated kinase-dependent manner. Furthermore, UDCA treatment repressed T7-induced SREBP-1c, FAS, and ACC protein levels, whereas knockdown of endogenous SMILE gene expression by adenovirus SMILE shRNA (Ad-shSMILE) significantly reversed UDCA-mediated repression of SREBP-1c, FAS, and ACC protein levels. Collectively, these results demonstrate that UDCA activates SMILE gene expression through adenosine monophosphate-activated kinase phosphorylation, which leads to repression of LXR-mediated hepatic lipogenic enzyme gene expression.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Lipogenesis/drug effects , Liver/drug effects , Orphan Nuclear Receptors/metabolism , Ursodeoxycholic Acid/pharmacology , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Blotting, Western , Cells, Cultured , Diet, High-Fat , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Hydrocarbons, Fluorinated/pharmacology , Liver/cytology , Liver/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Protein Binding/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sulfonamides/pharmacology , Transcriptional Activation/drug effects
7.
Biochem Biophys Res Commun ; 460(4): 996-1001, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25839654

ABSTRACT

Hepcidin is a peptide hormone secreted in the liver and plays a key role in maintaining iron homeostasis. Here, we demonstrate that B-cell translocation gene 2 (BTG2) is a key player in hepatic hepcidin regulation via induction of Yin Yang 1 (YY1). Hepatic hepcidin gene expression significantly enhanced by fasting states and glucagon exposure led to induction of gluconeogenic gene expression, and elevated serum hepcidin production in mice. Notably, overexpression of BTG2 using adenoviral system (Ad-BTG2) significantly elevated serum hepcidin levels via a significant induction of YY1 gene transcription. Immunoprecipitation studies demonstrated that BTG2 physically interacted with YY1 and recruited on the hepcidin gene promoter. Finally, ablation of hepatic BTG2 gene by gene silencing markedly attenuated the elevation of serum hepcidin production along with YY1 and hepcidin mRNA expression in fasting state. Likewise, forskolin (FSK)-stimulated hepcidin promoter activity was dramatically disrupted by endogenous BTG2 knockdown. Overall, our current study provides a novel molecular mechanism of BTG2-mediated induction of hepcidin gene expression, thereby contributing to a better understanding of the hepatic hepcidin production involved in iron homeostasis.


Subject(s)
Hepcidins/biosynthesis , Immediate-Early Proteins/physiology , Tumor Suppressor Proteins/physiology , YY1 Transcription Factor/biosynthesis , Animals , Base Sequence , Cell Line, Transformed , DNA Primers , Gluconeogenesis , Hepcidins/genetics , Immediate-Early Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics
8.
J Allergy Clin Immunol ; 134(3): 714-721.e7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24948367

ABSTRACT

BACKGROUND: Extracellular signal-regulated kinases 1/2 (ERK1/2) make important contributions to allergic responses via their regulation of degranulation, eicosanoid production, and cytokine expression by mast cells, yet the mechanisms underlying their positive effects on FcεRI-dependent signaling are not fully understood. Recently, we reported that mast cell activation and anaphylaxis are negatively regulated by AMP-activated protein kinase (AMPK). However, little is known about the relationship between ERK1/2-mediated positive and the AMPK-mediated negative regulation of FcεRI signaling in mast cells. OBJECTIVE: We investigated possible interactions between ERK1/2 and AMPK in the modulation of mast cell signaling and anaphylaxis. METHODS: Wild-type or AMPKα2(-/-) mice, or bone marrow-derived mast cells obtained from these mice, were treated with either chemical agents or small interfering RNAs that modulated the activity or expression of ERK1/2 or AMPK to evaluate the functional interplay between ERK1/2 and AMPK in FcεRI-dependent signaling. RESULTS: The ERK1/2 pathway inhibitor U0126 and the AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-4-ribofuranoside similarly inhibited FcεRI-mediated mast cell signals in vitro and anaphylaxis in vivo. ERK1/2-specific small interfering RNA also mimicked this effect on FcεRI signals. Moreover, AMPKα2 knockdown or deficiency led to increased FcεRI-mediated mast cell activation and anaphylaxis that were insensitive to U0126 or activator 5-aminoimidazole-4-carboxamide-1-ß-4-ribofuranoside, suggesting that the suppression of FcεRI signals by the inhibition of the ERK1/2 pathway relies largely on AMPK activation. ERK1/2 controlled AMPK activity by regulating its subcellular translocation. CONCLUSIONS: ERK1/2 ablated the AMPK-dependent negative regulatory axis, thereby activating FcεRI signals in mast cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anaphylaxis/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hypersensitivity/immunology , Mast Cells/immunology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Anaphylaxis/etiology , Animals , Butadienes/pharmacology , Cell Degranulation/drug effects , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hypersensitivity/complications , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nitriles/pharmacology , Receptors, IgG/metabolism , Ribonucleosides/pharmacology , Signal Transduction/drug effects
9.
J Biol Chem ; 288(22): 15937-46, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23589295

ABSTRACT

Testosterone level is low in insulin-resistant type 2 diabetes. Whether this is due to negative effects of high level of insulin on the testes caused by insulin resistance has not been studied in detail. In this study, we found that insulin directly binds to insulin receptors in Leydig cell membranes and activates phospho-insulin receptor-ß (phospho-IR-ß), phospho-IRS1, and phospho-AKT, leading to up-regulation of DAX-1 (dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1) gene expression in the MA-10 mouse Leydig cell line. Insulin also inhibits cAMP-induced and liver receptor homolog-1 (LRH-1)-induced steroidogenic enzyme gene expression and steroidogenesis. In contrast, knockdown of DAX-1 reversed insulin-mediated inhibition of steroidogenesis. Whether insulin directly represses steroidogenesis through regulation of steroidogenic enzyme gene expression was assessed in insulin-injected mouse models and high fat diet-induced obesity. In insulin-injected mouse models, insulin receptor signal pathway was activated and subsequently inhibited steroidogenesis via induction of DAX-1 without significant change of luteinizing hormone or FSH levels. Likewise, the levels of steroidogenic enzyme gene expression and steroidogenesis were low, but interestingly, the level of DAX-1 was high in the testes of high fat diet-fed mice. These results represent a novel regulatory mechanism of steroidogenesis in Leydig cells. Insulin-mediated induction of DAX-1 in Leydig cells of testis may be a key regulatory step of serum sex hormone level in insulin-resistant states.


Subject(s)
DAX-1 Orphan Nuclear Receptor/metabolism , Gonadal Steroid Hormones/biosynthesis , Insulin Resistance , Insulin/metabolism , Leydig Cells/metabolism , Obesity/metabolism , Animals , Cell Line , DAX-1 Orphan Nuclear Receptor/genetics , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Disease Models, Animal , Gene Expression Regulation, Enzymologic/genetics , Gonadal Steroid Hormones/genetics , Insulin/genetics , Leydig Cells/pathology , Male , Mice , Obesity/chemically induced , Obesity/pathology , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction/genetics
10.
J Allergy Clin Immunol ; 132(3): 729-736.e12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23587332

ABSTRACT

BACKGROUND: Aggregation of FcεRI activates a cascade of signaling events leading to mast cell activation, followed by inhibitory signals that turn off the activating signals. However, the overall view of negative signals in mast cells is still incomplete. Although AMP-activated protein kinase (AMPK), which is generally known as a regulator of energy metabolism, is also associated with anti-inflammation, little is known about the role of AMPK in mast cells. OBJECTIVES: We investigated the role of AMPK and its regulatory mechanism in mast cells. METHOD: The roles of AMPK in FcεRI-dependent activation of bone marrow-derived mast cells (BMMCs) were evaluated by using chemical agents, small interfering RNAs (siRNAs), or adenovirus that modulated the activity or expression of AMPK signaling components. In addition, AMPKα2(-/-) mice were used to verify the role of AMPK in anaphylactic models. RESULTS: FcεRI signaling and associated effector functions in BMMCs were suppressed by the AMPK activator 5-aminoimidazole-4-carboxamide-1-ß-4-ribofuranoside (AICAR) and were conversely augmented by siRNA knockdown of AMPKα2 or liver kinase B1 (LKB1), an upstream kinase of AMPK. Furthermore, AMPKα2 deficiency led to increased FcεRI-mediated BMMC activation and anaphylaxis that were insensitive to AICAR, whereas enforced expression of AMPKα2 in AMPKα2(-/-) BMMCs reversed the hypersensitive FcεRI signaling to normal levels. Pharmacologic inhibition or siRNA knockdown of Fyn mimicked AMPK activation, suggesting that Fyn counterregulates the LKB1-AMPK axis. Mechanistically, Fyn controlled AMPK activity by regulating LKB1 localization. CONCLUSIONS: The Fyn-regulated LKB1-AMPK axis acts as a novel inhibitory module for mast cell activation, which points to AMPK activators as therapeutic drugs for allergic diseases.


Subject(s)
AMP-Activated Protein Kinases/immunology , Anaphylaxis/immunology , Mast Cells/immunology , Receptors, IgE/immunology , AMP-Activated Protein Kinases/genetics , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/immunology
11.
Mol Pharmacol ; 84(1): 147-57, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23658008

ABSTRACT

Low molecular weight fucoidan (LMWF) is widely used to treat metabolic disorders, but its physiologic effects have not been well determined. In the present study, we investigated the metabolic effects of LMWF in obese diabetic mice (leptin receptor-deficient db/db mice) and the underlying molecular mechanisms involved in endoplasmic reticulum (ER) stress-responsive L6 myotubes. The effect of LMWF-mediated AMP-activated protein kinase (AMPK) activation on insulin resistance via regulation of the ER stress-dependent pathway was examined in vitro and in vivo. In db/db mice, LMWF markedly reduced serum glucose, triglyceride, cholesterol, and low-density lipoprotein levels, and gradually reduced body weights by reducing lipid parameters. Furthermore, it effectively ameliorated glucose homeostasis by elevating glucose tolerance. In addition, the phosphorylation levels of AMPK and Akt were markedly reduced by ER stressor, and subsequently, glucose uptake and fatty acid oxidation were also reduced. However, these adverse effects of ER stress were significantly ameliorated by LMWF. Finally, in L6 myotubes, LMWF markedly reduced the ER stress-induced upregulation of the mammalian target of rapamycin-p70S61 kinase network and subsequently improved the action of insulin via AMPK stimulation. Our findings suggest that AMPK activation by LMWF could prevent metabolic diseases by controlling the ER stress-dependent pathway and that this beneficial effect of LMWF provides a potential therapeutic strategy for ameliorating ER stress-mediated metabolic dysfunctions.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , Homeostasis/drug effects , Insulin Resistance/physiology , Lipid Metabolism/drug effects , Muscle Fibers, Skeletal/metabolism , Polysaccharides/pharmacology , Animals , Body Weight , Cholesterol/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Lipids , Lipoproteins, LDL/blood , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Weight , Muscle Fibers, Skeletal/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Triglycerides/blood
12.
J Biol Chem ; 287(44): 37098-108, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22977252

ABSTRACT

Growth hormone (GH) is a key metabolic regulator mediating glucose and lipid metabolism. Ataxia telangiectasia mutated (ATM) is a member of the phosphatidylinositol 3-kinase superfamily and regulates cell cycle progression. The orphan nuclear receptor small heterodimer partner (SHP: NR0B2) plays a pivotal role in regulating metabolic processes. Here, we studied the role of ATM on GH-dependent regulation of hepatic gluconeogenesis in the liver. GH induced phosphoenolpyruvate carboxykinase (PEPCK) and glucose 6-phosphatase gene expression in primary hepatocytes. GH treatment and adenovirus-mediated STAT5 overexpression in hepatocytes increased glucose production, which was blocked by a JAK2 inhibitor, AG490, dominant negative STAT5, and STAT5 knockdown. We identified a STAT5 binding site on the PEPCK gene promoter using reporter assays and point mutation analysis. Up-regulation of SHP by metformin-mediated activation of the ATM-AMP-activated protein kinase pathway led to inhibition of GH-mediated induction of hepatic gluconeogenesis, which was abolished by an ATM inhibitor, KU-55933. Immunoprecipitation studies showed that SHP physically interacted with STAT5 and inhibited STAT5 recruitment on the PEPCK gene promoter. GH-induced hepatic gluconeogenesis was decreased by either metformin or Ad-SHP, whereas the inhibition by metformin was abolished by SHP knockdown. Finally, the increase of hepatic gluconeogenesis following GH treatment was significantly higher in the liver of SHP null mice compared with that of wild-type mice. Overall, our results suggest that the ATM-AMP-activated protein kinase-SHP network, as a novel mechanism for regulating hepatic glucose homeostasis via a GH-dependent pathway, may be a potential therapeutic target for insulin resistance.


Subject(s)
Gluconeogenesis/genetics , Hepatocytes/metabolism , Human Growth Hormone/physiology , Receptors, Cytoplasmic and Nuclear/physiology , STAT5 Transcription Factor/metabolism , Transcriptional Activation , Adenylate Kinase/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Enzyme Activators/pharmacology , Genes, Reporter , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Hep G2 Cells , Humans , Liver/cytology , Liver/metabolism , Metformin/pharmacology , Mice , Phosphoenolpyruvate Carboxykinase (GTP)/biosynthesis , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Rats , STAT5 Transcription Factor/genetics , Signal Transduction , Tumor Suppressor Proteins/metabolism
13.
J Biol Chem ; 287(50): 41875-87, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23074219

ABSTRACT

Cyclic AMP (cAMP) induces steroidogenic enzyme gene expression and stimulates testosterone production in Leydig cells. Phosphoenolpyruvate carboxykinase (PEPCK) is expressed in Leydig cells, but its role has not been defined. In this study, we found that PEPCK and glucose-6-phosphatase (Glc-6-Pase) are increased significantly following cAMP treatment of mouse Leydig cells. Moreover, cAMP treatment increased recruitment of the cAMP-response element-binding transcription factor and decreased recruitment of the corepressor DAX-1 on the pepck promoter. Furthermore, cAMP induced an increase in ATP that correlated with a decrease in phospho-AMP-activated protein kinase (AMPK). In contrast, knockdown or inhibition of PEPCK decreased ATP and increased phospho-AMPK. Treatment with an AMPK activator or overexpression of the constitutively active form of AMPK inhibited cAMP-induced steroidogenic enzyme promoter activities and gene expression. Liver receptor homolog-1 (LRH-1) was involved in cAMP-induced steroidogenic enzyme gene expression but was inhibited by AMPK activation in Leydig cells. Additionally, inhibition or knockdown of PEPCK and Glc-6-Pase decreased cAMP-mediated induction of steroidogenic enzyme gene expression and steroidogenesis. Finally, pubertal mouse (8-week-old) testes and human chorionic gonadotropin-induced prepubertal mouse testes showed increased PEPCK and Glc-6-Pase gene expression. Taken together, these results suggest that induction of PEPCK and Glc-6-Pase by cAMP plays an important role in Leydig cell steroidogenesis.


Subject(s)
Glucose-6-Phosphatase/biosynthesis , Leydig Cells/enzymology , Phosphoenolpyruvate Carboxykinase (ATP)/biosynthesis , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cyclic AMP/genetics , Cyclic AMP/metabolism , DAX-1 Orphan Nuclear Receptor/genetics , DAX-1 Orphan Nuclear Receptor/metabolism , Gene Expression Regulation, Enzymologic/physiology , Glucose-6-Phosphatase/genetics , HeLa Cells , Humans , Leydig Cells/cytology , Male , Mice , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Promoter Regions, Genetic/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
14.
Biochem Biophys Res Commun ; 430(4): 1246-52, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23266607

ABSTRACT

The aim of the present study was to determine the effect of Tanshinone IIA (Tan IIA) on endoplasmic reticulum (ER) stress-induced insulin resistance in L6 myotubes and db/db mice. ER stress markers, RNA-activated protein kinase-like ER resident kinase (PERK), JNK, and AMPK activity were determined in tunicamycin-treated L6 myotubes. Insulin resistance was monitored using glucose uptake assays in vitro and blood glucose levels in vivo. Tan IIA clearly suppressed the phosphorylations of PERK and JNK and potentiated insulin-mediated Akt phosphorylation as well as glucose uptake via AMPK activation under ER stress. Furthermore, these effects are completely abrogated by siRNA-mediated knockdown of AMPK or LKB1. In addition, Tan IIA reduced blood glucose levels and body weights in db/db mice without altering food intake. These findings suggest that Tan IIA enhances insulin sensitivity and improves glucose metabolic disorders by increasing AMPK activity and attenuating ER stress-induced insulin resistance.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Abietanes/pharmacology , Drugs, Chinese Herbal/pharmacology , Endoplasmic Reticulum Stress/drug effects , Hypoglycemic Agents/pharmacology , Insulin Resistance , Animals , Cell Line , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/enzymology , Tunicamycin/pharmacology
15.
Exp Mol Med ; 55(7): 1556-1569, 2023 07.
Article in English | MEDLINE | ID: mdl-37488285

ABSTRACT

Melatonin is involved in the regulation of various biological functions. Here, we explored a novel molecular mechanism by which the melatonin-induced sestrin2 (SESN2)-small heterodimer partner (SHP) signaling pathway protects against fasting- and diabetes-mediated hepatic glucose metabolism. Various key gene expression analyses were performed and multiple metabolic changes were assessed in liver specimens and primary hepatocytes of mice and human participants. The expression of the hepatic cereblon (CRBN) and b-cell translocation gene 2 (BTG2) genes was significantly increased in fasting mice, diabetic mice, and patients with diabetes. Overexpression of Crbn and Btg2 increased hepatic gluconeogenesis by enhancing cyclic adenosine monophosphate (cAMP)-responsive element-binding protein H (CREBH), whereas this phenomenon was prominently ablated in Crbn null mice and Btg2-silenced mice. Interestingly, melatonin-induced SESN2 and SHP markedly reduced hepatic glucose metabolism in diabetic mice and primary hepatocytes, and this protective effect of melatonin was strikingly reversed by silencing Sesn2 and Shp. Finally, the melatonin-induced SESN2-SHP signaling pathway inhibited CRBN- and BTG2-mediated hepatic gluconeogenic gene transcription via the competition of BTG2 and the interaction of CREBH. Mitigation of the CRBN-BTG2-CREBH axis by the melatonin-SESN2-SHP signaling network may provide a novel therapeutic strategy to treat metabolic dysfunction due to diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Immediate-Early Proteins , Melatonin , Animals , Humans , Mice , Gluconeogenesis/physiology , Melatonin/pharmacology , Melatonin/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Liver/metabolism , Signal Transduction , Glucose/metabolism , Mice, Inbred C57BL , Sestrins/metabolism , Immediate-Early Proteins/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/metabolism
16.
J Biol Chem ; 286(44): 38035-38042, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21911493

ABSTRACT

LIPINs have been reported to perform important roles in the regulation of intracellular lipid levels. Their mutations induce lipodystrophy, myoglobinuria, and inflammatory disorders. Recently, the phosphatidic acid phosphatase function of LIPINs has been associated with the perturbation of hepatic insulin receptor signaling via the diacylglycerol-mediated stimulation of PKCε activity. Here, we report that nuclear estrogen-related receptor (ERR) γ is a novel transcriptional regulator of LIPIN1. Overexpression of ERRγ significantly increased LIPIN1 expression in primary hepatocytes, whereas the abolition of ERRγ gene expression attenuated the expression of LIPIN1. Deletion and mutation analyses of the LIPIN1 promoter showed that ERRγ exerts its effect on the transcriptional regulation of LIPIN1 via ERRE1 of the LIPIN1 promoter, as confirmed by ChIP assay. We also determined that the gene transcription of LIPIN1 by ERRγ is controlled by the competition between PGC-1α and small heterodimer partner. Additionally, ERRγ leads to the induction of hepatic LIPIN1 expression and diacylglycerol production in vivo. Finally, an inverse agonist of ERRγ, GSK5182, restores the impaired insulin signaling induced by LIPIN1-mediated PKCε activation. Our findings indicate that the selective control of ERRγ transcriptional activity by its specific inverse agonist could provide a novel therapeutic approach to the amelioration of impaired hepatic insulin signaling induced by LIPIN1-mediated PKCε activation.


Subject(s)
Gene Expression Regulation, Enzymologic , Insulin/metabolism , Liver/metabolism , Nuclear Proteins/biosynthesis , Phosphatidate Phosphatase/genetics , Receptors, Estrogen/metabolism , Transcription, Genetic , Animals , Cell Line , Cell Nucleus/metabolism , Hepatocytes/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Phosphatidate Phosphatase/biosynthesis , Protein Kinase C/metabolism , Rats , Signal Transduction , Transfection
17.
Biochem Biophys Res Commun ; 427(4): 801-5, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23058912

ABSTRACT

Hepatic gluconeogenesis is mediated by the network of transcriptional factors and cofactors. Here, we show that B-cell translocation gene-2 (BTG2) plays a crucial cofactor in hepatic gluconeogenesis via upregulation of the cyclic AMP (cAMP) response element binding (CREB) in hepatocytes. cAMP/dexamethasone (Dex) significantly increased BTG2 and other gluconeogenic genes such as Nur77, phosphoenolpyruvate carboxykinase (PEPCK), and glucose-6-phosphatase (G6Pase) in hepatocytes. In contrast, insulin treatment completely blocks their expressions. Interestingly, overexpression of BTG2 using adenoviral system (Ad-BTG2) significantly elevated hepatic glucose production via the increase of transcriptional activity and gene expression of CREB, PEPCK, and G6Pase in hepatocytes, suggesting that BTG2 is the key player on hepatic glucose production. Physiological interaction studies demonstrated that BTG2 correlated CREB recruitment on the PEPCK gene promoter via a direct interaction. Finally, knockdown of endogenous BTG2 expression markedly inhibits the cAMP/Dex-induced transcriptional activity of gluconeogenic genes and glucose production in hepatocytes. Overall, the present study provides us with a novel molecular mechanism of BTG2-mediated induction of hepatic gluconeogenesis and suggests that BTG2 plays an important role in hepatic glucose metabolism.


Subject(s)
Gene Expression Regulation , Gluconeogenesis/genetics , Glucose/metabolism , Immediate-Early Proteins/metabolism , Liver/metabolism , Response Elements/genetics , Tumor Suppressor Proteins/metabolism , Animals , CREB-Binding Protein/metabolism , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Dexamethasone/pharmacology , Glucagon/metabolism , Glucagon/pharmacology , Hep G2 Cells , Humans , Immediate-Early Proteins/genetics , Mice , Protein Serine-Threonine Kinases/genetics , Rats , Signal Transduction , Tumor Suppressor Proteins/genetics
18.
J Biol Chem ; 285(42): 32182-91, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20688914

ABSTRACT

Orphan nuclear receptor small heterodimer partner (SHP) plays a key role in transcriptional repression of gluconeogenic enzyme gene expression. Here, we show that SHP inhibited protein kinase A-mediated transcriptional activity of cAMP-response element-binding protein (CREB), a major regulator of glucose metabolism, to modulate hepatic gluconeogenic gene expression. Deletion analysis of phosphoenolpyruvate carboxykinase (PEPCK) promoter demonstrated that SHP inhibited forskolin-mediated induction of PEPCK gene transcription via inhibition of CREB transcriptional activity. In vivo imaging demonstrated that SHP inhibited CREB-regulated transcription coactivator 2 (CRTC2)-mediated cAMP-response element-driven promoter activity. Furthermore, overexpression of SHP using adenovirus SHP decreased CRTC2-dependent elevations in blood glucose levels and PEPCK or glucose-6-phosphatase (G6Pase) expression in mice. SHP and CREB physically interacted and were co-localized in vivo. Importantly, SHP inhibited both wild type CRTC2 and S171A (constitutively active form of CRTC2) coactivator activity and disrupted CRTC2 recruitment on the PEPCK gene promoter. In addition, metformin or overexpression of a constitutively active form of AMPK (Ad-CA-AMPK) inhibited S171A-mediated PEPCK and G6Pase gene expression, and hepatic glucose production and knockdown of SHP partially relieved the metformin- and Ad-CA-AMPK-mediated repression of hepatic gluconeogenic enzyme gene expression in primary rat hepatocytes. In conclusion, our results suggest that a delayed effect of metformin-mediated induction of SHP gene expression inhibits CREB-dependent hepatic gluconeogenesis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Gluconeogenesis/physiology , Hepatocytes/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation , Gluconeogenesis/drug effects , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Hepatocytes/cytology , Humans , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Mice , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Promoter Regions, Genetic , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators/genetics
19.
Sci Rep ; 11(1): 18999, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34556771

ABSTRACT

Growth hormone (GH) is one of the critical factors in maintaining glucose metabolism. B-cell translocation gene 2 (BTG2) and yin yang 1 (YY1) are key regulators of diverse metabolic processes. In this study, we investigated the link between GH and BTG2-YY1 signaling pathway in glucose metabolism. GH treatment elevated the expression of hepatic Btg2 and Yy1 in primary mouse hepatocytes and mouse livers. Glucose production in primary mouse hepatocytes and serum blood glucose levels were increased during GH exposure. Overexpression of hepatic Btg2 and Yy1 induced key gluconeogenic enzymes phosphoenolpyruvate carboxykinase 1 (PCK1) and glucose-6 phosphatase (G6PC) as well as glucose production in primary mouse hepatocytes, whereas this phenomenon was markedly diminished by knockdown of Btg2 and Yy1. Here, we identified the YY1-binding site on the Pck1 and G6pc gene promoters using reporter assays and point mutation analysis. The regulation of hepatic gluconeogenic genes induced by GH treatment was clearly linked with YY1 recruitment on gluconeogenic gene promoters. Overall, this study demonstrates that BTG2 and YY1 are novel regulators of GH-dependent regulation of hepatic gluconeogenic genes and glucose production. BTG2 and YY1 may be crucial therapeutic targets to intervene in metabolic dysfunction in response to the GH-dependent signaling pathway.


Subject(s)
Gluconeogenesis/genetics , Growth Hormone/metabolism , Immediate-Early Proteins/metabolism , Tumor Suppressor Proteins/metabolism , YY1 Transcription Factor/metabolism , Animals , Cell Line , Glucose/biosynthesis , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Growth Hormone/administration & dosage , Hepatocytes , Humans , Injections, Intraperitoneal , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Male , Mice , Models, Animal , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Point Mutation , Primary Cell Culture , Promoter Regions, Genetic , Signal Transduction/genetics
20.
BMB Rep ; 54(4): 221-226, 2021 04.
Article in English | MEDLINE | ID: mdl-33795032

ABSTRACT

Hepcidin (HAMP) is synthesized in the liver. It is a key ironregulatory hormone that controls systemic iron homeostasis. Cereblon (CRBN) and Kruppel-like factor 15 (KLF15) are known to regulate diverse physiological functions. In this study, we investigated the role of CRBN on hepatic hepcidin gene expression and production under gluconeogenic stimuli. Fasted mice as well as forskolin (FSK)- and glucagon (GLU)-treated mice had reduced serum iron levels but increased expression levels of hepatic Crbn and Klf15 and hepcidin secretion. MicroRNA (miRNA) expression analysis of fasted and Ad-Crbninfected mice revealed significant reduction of microRNA-639 (miR-639). Hepatic overexpression of Crbn elevated hepcidin expression and production along with Klf15 gene expression, whereas knockdown of Crbn and Klf15 markedly decreased FSK- and fasting-mediated induction of hepcidin gene expression and its biosynthesis in mouse livers and primary hepatocytes. Moreover, expression of KLF15 significantly increased the activity of hepcidin reporter gene. It was exclusively dependent on the KLF15-binding site identified within the hepcidin gene promoter. Overall, this study demonstrates that CRBN and KLF15 are novel mediators of gluconeogenic signal-induced hepcidin gene expression and production. Thus, CRBN and KLF15 might be novel potential therapeutic targets to intervene metabolic dysfunction. [BMB Reports 2021; 54(4): 221-226].


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hepcidins/genetics , Kruppel-Like Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Hepcidins/metabolism , Kruppel-Like Transcription Factors/genetics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL