Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Gene Regul Mech ; 1862(6): 643-656, 2019 06.
Article in English | MEDLINE | ID: mdl-30959128

ABSTRACT

Gluconeogenesis is essential for blood glucose homeostasis during fasting and is regulated by various enzymes, which are encoded by gluconeogenic genes. Those genes are controlled by various transcription factors. Zinc finger and BTB domain-containing 7c (Zbtb7c, also called Kr-pok) is a BTB-POZ family transcription factor with proto-oncogenic activity. Previous findings have indicated that Zbtb7c is involved in the regulation of fatty acid biosynthesis, suggesting an involvement also in primary metabolism. We found here that fasting induced Zbtb7c expression in the mouse liver and in primary liver hepatocytes. We also observed that Zbtb7c-knockout mice have decreased blood glucose levels, so we investigated whether Zbtb7c plays a role in gluconeogenesis. Indeed, differential gene expression analysis of Zbtb7c-knockout versus wild type mouse livers showed downregulated transcription of gluconeogenic genes encoding the glucose 6-phosphatase catalytic subunit (G6pc) and phosphoenolpyruvate carboxykinase 1 (Pck1), while Zbtb7c expression upregulated these two genes, under fasting conditions. Mechanistically, we found that when complexed with histone deacetylase 3 (Hdac3), Zbtb7c binds insulin response elements (IREs) within the G6pc and Pck1 promoters. Moreover, complexed Zbtb7c deacetylated forkhead box O1 (Foxo1), thereby increasing Foxo1 binding to the G6pc and Pck1 IREs, resulting in their transcriptional activation. These results demonstrate Zbtb7c to be a crucial metabolic regulator of blood glucose homeostasis, during mammalian fasting.


Subject(s)
Fasting , Gene Expression Regulation , Gluconeogenesis/physiology , Glucose-6-Phosphatase/genetics , Intracellular Signaling Peptides and Proteins/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Proteins/metabolism , Transcription Factors/metabolism , Zinc Fingers/physiology , Animals , Blood Glucose , DNA-Binding Proteins/metabolism , Fatty Acids/biosynthesis , Forkhead Box Protein O1/metabolism , Gluconeogenesis/genetics , Glucose/metabolism , Glucose-6-Phosphatase/metabolism , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Histone Deacetylases/metabolism , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Mutagenesis, Site-Directed , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Promoter Regions, Genetic , Proteins/genetics , Transcriptome , Zinc Fingers/genetics
2.
Diabetes Metab J ; 42(6): 465-471, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30398040

ABSTRACT

My professional journey to understand the glucose homeostasis began in the 1990s, starting from cloning of the promoter region of glucose transporter type 2 (GLUT2) gene that led us to establish research foundation of my group. When I was a graduate student, I simply thought that hyperglycemia, a typical clinical manifestation of type 2 diabetes mellitus (T2DM), could be caused by a defect in the glucose transport system in the body. Thus, if a molecular mechanism controlling glucose transport system could be understood, treatment of T2DM could be possible. In the early 70s, hyperglycemia was thought to develop primarily due to a defect in the muscle and adipose tissue; thus, muscle/adipose tissue type glucose transporter (GLUT4) became a major research interest in the diabetology. However, glucose utilization occurs not only in muscle/adipose tissue but also in liver and brain. Thus, I was interested in the hepatic glucose transport system, where glucose storage and release are the most actively occurring.

3.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2097-2107, 2018 06.
Article in English | MEDLINE | ID: mdl-29601978

ABSTRACT

Prolactin regulatory element-binding (PREB) protein is a transcription factor that regulates prolactin (PRL) gene expression. PRL, also known as luteotropic hormone or luteotropin, is well known for its role in producing milk. However, the role of PREB, in terms of hepatic glucose metabolism, is not well elucidated. Here, we observed expression of Preb in the mouse liver, in connection with glucose homeostasis. Morevoer, Preb was downregulated in db/db, ob/ob and high-fat diet-induced obese (DIO) mice, concurrent with upregulation of the liver genes glucose-6-phosphatase (G6pc) and phosphoenolpyruvate carboxykinase-1 (Pck). Administration of adenovirus-Preb (Ad-Preb) to db/db, ob/ob, and DIO mice diminished glucose, insulin, and pyruvate tolerance, which analogously, were impaired in normal (C57BL/6) mice knocked down for Preb, via infection with Ad-shPreb (anti-Preb RNA), indicating Preb to be a negative regulator of liver gluconeogenic genes. We further demonstrate that Preb negatively influences gluconeogenic gene expression, by directly binding to their promoters at a prolactin core-binding element (PCBE). A better understanding of Preb gene expression, during the pathogenesis of hepatic insulin resistance, could ultimately provide new avenues for therapies for metabolic syndrome, obesity, and type-2 diabetes mellitus, disorders whose worldwide incidences are increasing drastically.


Subject(s)
DNA-Binding Proteins/metabolism , Gluconeogenesis , Glucose/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Liver/metabolism , Transcription Factors/metabolism , Animals , Blood Glucose , DNA-Binding Proteins/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Down-Regulation , Fasting , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Hepatocytes/metabolism , Humans , Insulin/metabolism , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/blood , Obesity/etiology , Obesity/metabolism , Primary Cell Culture , Prolactin/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics , Up-Regulation
4.
Sci Rep ; 7: 45300, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28338058

ABSTRACT

The incidence of prostate cancer (PC) is growing rapidly throughout the world, in probable association with the adoption of western style diets. Thus, understanding the molecular pathways triggering the development of PC is crucial for both its prevention and treatment. Here, we investigated the role of the metabolism-associated protein, CREB3L4, in the proliferation of PC cells. CREB3L4 was upregulated by the synthetic androgen, R1881, in LNCaP PC cells (an androgen-dependent cell line). Knockdown of CREB3L4 resulted in decreased androgen-dependent PC cell growth. LNCaP cells transfected with siCREB3L4 underwent G2/M arrest, with upregulation of the proteins cyclin B1, phospho-CDK1, p21Waf1/Cip1, and INCA1, and downregulation of cyclin D1. Moreover, depletion of CREB3L4 resulted in significantly decreased expression of a subset of androgen-receptor (AR) target genes, including PSA, FKBP5, HPGD, KLK2, and KLK4. We also demonstrated that CREB3L4 directly interacts with the AR, and increases the binding of AR to androgen response elements (AREs). We also identified a role for the unfolded protein response (and its surrogate, IRE1α), in activating CREB3L4. Cumulatively, we postulate that CREB3L4 expression is mediated by an AR-IRE1α axis, but is also directly regulated by AR-to-ARE binding. Thus, our study demonstrates that CREB3L4 plays a key role in PC cell proliferation, which is promoted by both AR and IRE1α.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Proliferation , Nuclear Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein , Down-Regulation/drug effects , Endoribonucleases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Metribolone/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Unfolded Protein Response/drug effects , Up-Regulation/drug effects
5.
Biochem Biophys Res Commun ; 478(3): 1060-6, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27524233

ABSTRACT

Elongation of very long chain fatty acids protein 6 (ELOVL6), a rate-limiting enzyme for the elongation of saturated and monounsaturated fatty acids with 12, 14, and 16 carbons, plays a key role in energy metabolism and insulin sensitivity. Hepatic Elovl6 expression is upregulated in the fasting-refeeding response and in leptin-deficient ob/ob mice. Mouse Elovl6 has been shown to be a direct target of sterol regulatory element binding protein-1 (SREBP-1) in response to insulin. In the present study, we demonstrated that mouse and human Elovl6 expression is under the direct transcriptional control of carbohydrate response element binding protein (ChREBP), a mediator of glucose-induced gene expression. Serial deletion and site-directed mutagenesis studies revealed functional carbohydrate response elements (ChoREs) in the mouse and human Elovl6 promoters and gel shift assays and chromatin immunoprecipitation assays confirmed the binding of ChREBP to the Elovl6-ChoRE sites. In addition, the ectopic co-expression of ChREBP and SREBP-1c in HepG2 cells synergistically stimulated Elovl6 promoter activity and this synergistic activation was abolished by mutating the Elovl6 promoter ChoREs. Taken together, these results suggest that the synergistic action of ChREBP and SREBP-1c is necessary for the maximal induction of Elovl6 expression in the liver.


Subject(s)
Acetyltransferases/genetics , Gene Expression Regulation , Liver/metabolism , Nuclear Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription Factors/metabolism , Acetyltransferases/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Fatty Acid Elongases , Feeding Behavior , Hep G2 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics
7.
Sci Rep ; 5: 17395, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26620281

ABSTRACT

Glucokinase (GK), mainly expressed in the liver and pancreatic ß-cells, is critical for maintaining glucose homeostasis. GK expression and kinase activity, respectively, are both modulated at the transcriptional and post-translational levels. Post-translationally, GK is regulated by binding the glucokinase regulatory protein (GKRP), resulting in GK retention in the nucleus and its inability to participate in cytosolic glycolysis. Although hepatic GKRP is known to be regulated by allosteric mechanisms, the precise details of modulation of GKRP activity, by post-translational modification, are not well known. Here, we demonstrate that GKRP is acetylated at Lys5 by the acetyltransferase p300. Acetylated GKRP is resistant to degradation by the ubiquitin-dependent proteasome pathway, suggesting that acetylation increases GKRP stability and binding to GK, further inhibiting GK nuclear export. Deacetylation of GKRP is effected by the NAD(+)-dependent, class III histone deacetylase SIRT2, which is inhibited by nicotinamide. Moreover, the livers of db/db obese, diabetic mice also show elevated GKRP acetylation, suggesting a broader, critical role in regulating blood glucose. Given that acetylated GKRP may affiliate with type-2 diabetes mellitus (T2DM), understanding the mechanism of GKRP acetylation in the liver could reveal novel targets within the GK-GKRP pathway, for treating T2DM and other metabolic pathologies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucokinase/metabolism , Glucose/metabolism , Protein Processing, Post-Translational , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Animals , Carrier Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Glucokinase/genetics , Glucose/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Obese , Sirtuin 2/genetics , Sirtuin 2/metabolism
8.
Protein Cell ; 6(11): 804-13, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26334401

ABSTRACT

Post-translational modifications (PTMs) of transcription factors play a crucial role in regulating metabolic homeostasis. These modifications include phosphorylation, methylation, acetylation, ubiquitination, SUMOylation, and O-GlcNAcylation. Recent studies have shed light on the importance of lysine acetylation at nonhistone proteins including transcription factors. Acetylation of transcription factors affects subcellular distribution, DNA affinity, stability, transcriptional activity, and current investigations are aiming to further expand our understanding of the role of lysine acetylation of transcription factors. In this review, we summarize recent studies that provide new insights into the role of protein lysine-acetylation in the transcriptional regulation of metabolic homeostasis.


Subject(s)
Transcription Factors/metabolism , Acetylation , Animals , Diabetes Mellitus, Type 2/metabolism , Homeostasis/genetics , Homeostasis/physiology , Humans , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology
9.
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
10.
Diabetologia ; 56(12): 2723-32, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24037087

ABSTRACT

AIMS/HYPOTHESIS: Thioredoxin-interacting protein (TXNIP) is upregulated in the hyperglycaemic state and represses glucose uptake, resulting in imbalanced glucose homeostasis. In this study, we propose a mechanism of how TXNIP impairs hepatic glucose tolerance at the transcriptional level. METHODS: We administered adenoviral Txnip (Ad-Txnip) to normal mice and performed intraperitoneal glucose tolerance tests (IPGTT), insulin tolerance tests (ITT) and pyruvate tolerance tests (PTT). After Ad-Txnip administration, the expression of genes involved in glucose metabolism, including G6pc and Gck, was analysed using quantitative real-time PCR and western blot. To understand the increased G6pc expression in liver resulting from Txnip overexpression, we performed pull-down assays for TXNIP and small heterodimer partner (SHP). Luciferase reporter assays and chromatin immunoprecipitation using the Txnip promoter were performed to elucidate the interrelationship between carbohydrate response element-binding protein (ChREBP) and transcription factor E3 (TFE3) in the regulation of Txnip expression. RESULTS: Overabundance of TXNIP resulted in impaired glucose, insulin and pyruvate tolerance in normal mice. Ad-Txnip transduction upregulated G6pc expression and caused a decrease in Gck levels in the liver of normal mice and primary hepatocytes. TXNIP increased G6pc expression by forming a complex with SHP, which is known to be a negative modulator of gluconeogenesis. Txnip expression in mouse models of diabetes was decreased by Ad-Tfe3 administration, suggesting that TFE3 may play a negative role through competition with ChREBP at the E-box of the Txnip promoter. CONCLUSIONS/INTERPRETATION: We demonstrated that TXNIP impairs glucose and insulin tolerance in mice by upregulating G6pc through interaction with SHP.


Subject(s)
Carrier Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Gluconeogenesis , Glucose Intolerance/metabolism , Glucose-6-Phosphatase/metabolism , Liver/metabolism , Thioredoxins/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Blotting, Western , Carrier Proteins/genetics , Chromatin Immunoprecipitation , Glucose Tolerance Test , Glucose-6-Phosphatase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Thioredoxins/genetics , Transcription Factors/metabolism , Transcriptional Activation , Up-Regulation
11.
Yonsei Med J ; 54(3): 545-59, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23549795

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) belongs to a nuclear receptor superfamily; members of which play key roles in the control of body metabolism principally by acting on adipose tissue. Ligands of PPARγ, such as thiazolidinediones, are widely used in the treatment of metabolic syndromes and type 2 diabetes mellitus (T2DM). Although these drugs have potential benefits in the treatment of T2DM, they also cause unwanted side effects. Thus, understanding the molecular mechanisms governing the transcriptional activity of PPARγ is of prime importance in the development of new selective drugs or drugs with fewer side effects. Recent advancements in molecular biology have made it possible to obtain a deeper understanding of the role of PPARγ in body homeostasis. The transcriptional activity of PPARγ is subject to regulation either by interacting proteins or by modification of the protein itself. New interacting partners of PPARγ with new functions are being unveiled. In addition, post-translational modification by various cellular signals contributes to fine-tuning of the transcriptional activities of PPARγ. In this review, we will summarize recent advancements in our understanding of the post-translational modifications of, and proteins interacting with, PPARγ, both of which affect its transcriptional activities in relation to adipogenesis.


Subject(s)
Models, Genetic , PPAR gamma/physiology , Protein Processing, Post-Translational , Gene Expression Regulation , Homeostasis , PPAR gamma/genetics , PPAR gamma/metabolism , Sumoylation , Transcription Factors/metabolism , Transcription Factors/physiology , Ubiquitination
12.
Exp Diabetes Res ; 2012: 716425, 2012.
Article in English | MEDLINE | ID: mdl-22110478

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is characterized by fat accumulation in the liver not due to alcohol abuse. NAFLD is accompanied by variety of symptoms related to metabolic syndrome. Although the metabolic link between NAFLD and insulin resistance is not fully understood, it is clear that NAFLD is one of the main cause of insulin resistance. NAFLD is shown to affect the functions of other organs, including pancreas, adipose tissue, muscle and inflammatory systems. Currently efforts are being made to understand molecular mechanism of interrelationship between NAFLD and insulin resistance at the transcriptional level with specific focus on post-translational modification (PTM) of transcription factors. PTM of transcription factors plays a key role in controlling numerous biological events, including cellular energy metabolism, cell-cycle progression, and organ development. Cell type- and tissue-specific reversible modifications include lysine acetylation, methylation, ubiquitination, and SUMOylation. Moreover, phosphorylation and O-GlcNAcylation on serine and threonine residues have been shown to affect protein stability, subcellular distribution, DNA-binding affinity, and transcriptional activity. PTMs of transcription factors involved in insulin-sensitive tissues confer specific adaptive mechanisms in response to internal or external stimuli. Our understanding of the interplay between these modifications and their effects on transcriptional regulation is growing. Here, we summarize the diverse roles of PTMs in insulin-sensitive tissues and their involvement in the pathogenesis of insulin resistance.


Subject(s)
Fatty Liver/physiopathology , Insulin Resistance/physiology , Protein Processing, Post-Translational , Transcription Factors/metabolism , Adipokines/physiology , Animals , DNA/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Fatty Acids, Nonesterified/physiology , Fatty Liver/complications , Humans , Insulin-Secreting Cells/physiology , Liver/metabolism , Macrophages/physiology , Metabolic Syndrome/complications , Metabolic Syndrome/physiopathology , Non-alcoholic Fatty Liver Disease
13.
Behav Brain Res ; 229(1): 82-90, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22185695

ABSTRACT

The present study examined the effects of Tremella fuciformis (TF) on the learning and memory function and the neural activity in rats with trimethyltin (TMT)-induced memory deficits. The rats were administered saline or TF (TF 25, 50, 100 mg/kg, p.o.) daily for 21 days. The cognitive improving efficacy of TF on the amnesic rats, which was induced by TMT, was investigated by assessing the Morris water maze test and by performing Choline acetyltransferase (ChAT) and cAMP responsive element binding protein (CREB) immunohistochemistry. In order to confirm the underlying mechanisms of the memory enhancing effects of TF, we assessed the neurite outgrowth of PC12 cells. We also administered 18F-fluorodeoxyglucose and performed a PET scan of the frontal lobe. The rats with TMT injection showed impaired learning and memory of the tasks and treatment with TF produced a significant improvement of the escape latency to find the platform in the Morris water maze compared to that of the control group. In the retention test, the TF50 group showed increased time spent around the platform compared to that of the control group. Consistent with the behavioral data, TF50 mg/kg significantly alleviated the loss of ChAT-ir neurons in the hippocampus compared to that of the control group. Treatment with TF significantly increased the CREB positive neurons in the hippocampal CA1 area as compared to that of the control group. In addition, TF treatment (50 mg/kg) increased the glucose uptake approximately sevenfold in the frontal lobe and it significantly promoted neurite outgrowth of the PC12 cells, as compared to that of the controls. These results suggest that TF may be useful for improving the cognitive function via regulation of the CREB signaling pathway and cholinergic system in the hippocampus.


Subject(s)
CREB-Binding Protein/metabolism , Cholinergic Agents/metabolism , Memory Disorders/drug therapy , Neurites/drug effects , Neuroprotective Agents/therapeutic use , Polysaccharides/therapeutic use , Analysis of Variance , Animals , Choline O-Acetyltransferase/metabolism , Dose-Response Relationship, Drug , Fluorodeoxyglucose F18 , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/diagnostic imaging , Neuroprotective Agents/pharmacology , PC12 Cells/cytology , Polysaccharides/pharmacology , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Time Factors , Trimethyltin Compounds/toxicity
14.
Biomol Ther (Seoul) ; 20(2): 207-13, 2012 Mar.
Article in English | MEDLINE | ID: mdl-24116297

ABSTRACT

The present study examined the effects of krill-derived phosphatidylserine (Krill-PS) on the learning and memory function and the neural activity in rats with trimethyltin (TMT)-induced memory deficits. The rats were administered vehicle (medium-chain triglyceride: MCT) or Krill-PS (50, 100 mg/kg, p.o.) daily for 21 days. The cognitive improving efficacy of Krill-PS in TMT-induced amnesic rats was investigated by assessing the Morris water maze test and by performing choline acetyltransferase (ChAT), acetylcholinesterase (AChE) and cAMP responsive element binding protein (CREB) immunohistochemistry. The rats with TMT injection showed impaired learning and memory of the tasks and treatment with Krill-PS produced a significant improvement of the escape latency to find the platform in the Morris water maze at the 2(nd) and 4(th) day compared to that of the MCT group (p<0.05). In the retention test, the Krill-PS+MCT groups showed increased time spent around the platform compared to that of the MCT group. Consistent with the behavioral data, Krill-PS 50+MCT group significantly alleviated the loss of acetylcholinergic neurons in the hippocampus and medial septum compared to that of the MCT group. Treatment with Krill-PS significantly increased the CREB positive neurons in the hippocampal CA1 area as compared to that of the MCT group. These results suggest that Krill-PS may be useful for improving the cognitive function via regulation of cholinergic marker enzyme activity and neural activity.

15.
PLoS One ; 6(7): e22544, 2011.
Article in English | MEDLINE | ID: mdl-21811631

ABSTRACT

The carbohydrate response element binding protein (ChREBP), a basic helix-loop-helix/leucine zipper transcription factor, plays a critical role in the control of lipogenesis in the liver. To identify the direct targets of ChREBP on a genome-wide scale and provide more insight into the mechanism by which ChREBP regulates glucose-responsive gene expression, we performed chromatin immunoprecipitation-sequencing and gene expression analysis. We identified 1153 ChREBP binding sites and 783 target genes using the chromatin from HepG2, a human hepatocellular carcinoma cell line. A motif search revealed a refined consensus sequence (CABGTG-nnCnG-nGnSTG) to better represent critical elements of a functional ChREBP binding sequence. Gene ontology analysis shows that ChREBP target genes are particularly associated with lipid, fatty acid and steroid metabolism. In addition, other functional gene clusters related to transport, development and cell motility are significantly enriched. Gene set enrichment analysis reveals that ChREBP target genes are highly correlated with genes regulated by high glucose, providing a functional relevance to the genome-wide binding study. Furthermore, we have demonstrated that ChREBP may function as a transcriptional repressor as well as an activator.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genome, Human/genetics , Glucose/pharmacology , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Binding Sites , Chromatin Immunoprecipitation , DNA/metabolism , Databases, Genetic , Genetic Loci/genetics , HEK293 Cells , Hep G2 Cells , Humans , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/drug effects , Liver/metabolism , Molecular Sequence Data , Protein Binding/drug effects , Reproducibility of Results , Signal Transduction/drug effects , Signal Transduction/genetics
16.
J Biol Chem ; 286(2): 1157-64, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21081500

ABSTRACT

Glucose-6-phosphatase (G6Pase) is a key enzyme that is responsible for the production of glucose in the liver during fasting or in type 2 diabetes mellitus (T2DM). During fasting or in T2DM, peroxisome proliferator-activated receptor α (PPARα) is activated, which may contribute to increased hepatic glucose output. However, the mechanism by which PPARα up-regulates hepatic G6Pase gene expression in these states is not well understood. We evaluated the mechanism by which PPARα up-regulates hepatic G6Pase gene expression in fasting and T2DM states. In PPARα-null mice, both hepatic G6Pase and phosphoenolpyruvate carboxykinase levels were not increased in the fasting state. Moreover, treatment of primary cultured hepatocytes with Wy14,643 or fenofibrate increased the G6Pase mRNA level. In addition, we have localized and characterized a PPAR-responsive element in the promoter region of the G6Pase gene. Chromatin immunoprecipitation (ChIP) assay revealed that PPARα binding to the putative PPAR-responsive element of the G6Pase promoter was increased in fasted wild-type mice and db/db mice. These results indicate that PPARα is responsible for glucose production through the up-regulation of hepatic G6Pase gene expression during fasting or T2DM animal models.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation, Enzymologic , Glucose-6-Phosphatase/genetics , PPAR alpha/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Eating/physiology , Fasting/physiology , Gluconeogenesis/physiology , Hep G2 Cells , Humans , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , PPAR alpha/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Up-Regulation/physiology
17.
Biochem Biophys Res Commun ; 403(3-4): 329-34, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-21078299

ABSTRACT

During a state of fasting, the blood glucose level is maintained by hepatic gluconeogenesis. SIRT1 is an important metabolic regulator during nutrient deprivation and the liver-specific knockdown of SIRT1 resulted in decreased glucose production. We hypothesize that SIRT1 is responsible for the upregulation of insulin-suppressed gluconeogenic genes through the deacetylation of FOXO1. Treatment of primary cultured hepatocytes with resveratrol increased insulin-repressed PEPCK and G6Pase mRNA levels, which depend on SIRT1 activity. We found that the resveratrol treatment resulted in a decrease in the phosphorylation of Akt and FOXO1, which are independent of SIRT1 action. Fluorescence microscopy revealed that resveratrol caused the nuclear localization of FOXO1. In the nucleus, FOXO1 is deacetylated by SIRT1, which might make it more accessible to the IRE of the PEPCK and G6Pase promoter, causing an increase in their gene expression. Our results indicate that resveratrol upregulates the expression of gluconeogenic genes by attenuating insulin signaling and by deacetylating FOXO1, which are SIRT1-independent in the cytosol and SIRT1-dependent in the nucleus, respectively.


Subject(s)
Antioxidants/pharmacology , Forkhead Transcription Factors/metabolism , Gene Expression/drug effects , Gluconeogenesis/drug effects , Liver/drug effects , Stilbenes/pharmacology , Animals , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gluconeogenesis/genetics , Humans , Liver/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Resveratrol , Sirtuin 1/genetics , Sirtuin 1/metabolism , Up-Regulation
18.
Sensors (Basel) ; 10(5): 5031-53, 2010.
Article in English | MEDLINE | ID: mdl-22399922

ABSTRACT

Pancreatic ß-cells and the liver play a key role in glucose homeostasis. After a meal or in a state of hyperglycemia, glucose is transported into the ß-cells or hepatocytes where it is metabolized. In the ß-cells, glucose is metabolized to increase the ATP:ADP ratio, resulting in the secretion of insulin stored in the vesicle. In the hepatocytes, glucose is metabolized to CO(2), fatty acids or stored as glycogen. In these cells, solute carrier family 2 (SLC2A2) and glucokinase play a key role in sensing and uptaking glucose. Dysfunction of these proteins results in the hyperglycemia which is one of the characteristics of type 2 diabetes mellitus (T2DM). Thus, studies on the molecular mechanisms of their transcriptional regulations are important in understanding pathogenesis and combating T2DM. In this paper, we will review a recent update on the progress of gene regulation of glucose sensors in the liver and ß-cells.


Subject(s)
Gene Expression Regulation , Glucokinase/genetics , Glucose Transporter Type 2/genetics , Glucose/metabolism , Liver/metabolism , Transcription, Genetic , Animals , Glucokinase/metabolism , Glucose Transporter Type 2/metabolism , Humans , Insulin-Secreting Cells/metabolism , Liver/enzymology
19.
J Biol Chem ; 284(22): 15071-83, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19366697

ABSTRACT

Liver glucokinase (LGK) plays an essential role in controlling blood glucose levels and maintaining cellular metabolic functions. Expression of LGK is induced mainly regulated by insulin through sterol regulatory element-binding protein-1c (SREBP-1c) as a mediator. Since LGK expression is known to be decreased in the liver of liver X receptor (LXR) knockout mice, we have investigated whether LGK might be directly activated by LXRalpha. Furthermore, we have studied interrelationship between transcription factors that control gene expression of LGK. In the current studies, we demonstrated that LXRalpha increased LGK expression in primary hepatocytes and that there is a functional LXR response element in the LGK gene promoter as shown by electrophoretic mobility shift and chromatin precipitation assay. In addition, our studies demonstrate that LXRalpha and insulin activation of the LGK gene promoter occurs through a multifaceted indirect mechanism. LXRalpha increases SREBP-1c expression and then insulin stimulates the processing of the membrane-bound precursor SREBP-1c protein, and it activates LGK expression through SREBP sites in its promoter. LXRalpha also activates the LGK promoter by increasing the transcriptional activity and induction of peroxisome proliferator-activated receptor (PPAR)-gamma, which also stimulates LGK expression through a peroxisome proliferator-responsive element. This activation is tempered through a negative mechanism, where a small heterodimer partner (SHP) decreases LGK gene expression by inhibiting the transcriptional activity of LXRalpha and PPARgamma by directly interacting with their common heterodimer partner RXRalpha. From these data, we propose a mechanism for LXRalpha in controlling the gene expression of LGK that involves activation through SREBP-1c and PPARgamma and inhibition through SHP.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Glucokinase/genetics , Liver/enzymology , PPAR gamma/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Base Sequence , Cell Line , Glucokinase/metabolism , Hepatocytes/enzymology , Humans , Liver X Receptors , Mice , Models, Biological , Molecular Sequence Data , Orphan Nuclear Receptors , PPAR gamma/genetics , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Response Elements/genetics , Transcription, Genetic
20.
Biochem J ; 417(1): 313-22, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18774944

ABSTRACT

KLF5 (Krüppel-like factor 5) is a zinc-finger transcription factor that plays a critical role in the regulation of cellular signalling involved in cell proliferation, differentiation and oncogenesis. In the present study, we showed that KLF5 acts as a key regulator controlling the expression of FASN (fatty acid synthase) through an interaction with SREBP-1 (sterol-regulatory-element-binding protein-1) in the androgen-dependent LNCaP prostate cancer cell line. The mRNA level of KLF5 increased when cells were treated with a synthetic androgen, R1881. Furthermore, KLF5 bound to SREBP-1 and enhanced the SREBP-1-mediated increase in FASN promoter activity. The results also demonstrated that the expression of KLF5 in LNCaP prostate cancer cells enhanced FASN expression, whereas silencing of KLF5 by small interfering RNA down-regulated FASN expression. The proximal promoter region and the first intron of the FASN gene contain multiple CACCC elements that mediate the transcriptional regulation of the gene by KLF5. However, other lipogenic and cholesterogenic genes, such as those encoding acetyl-CoA carboxylase, ATP-citrate lyase, the LDL (low-density lipoprotein) receptor, HMG-CoA (3-hydroxy-3-methylglutaryl-CoA) synthase and HMG-CoA reductase are irresponsive to KLF5 expression, owing to the absence of CACCC elements in their promoter regions. Taken together, these results suggest that the FASN gene is activated by the synergistic action of KLF5 and SREBP-1, which was induced by androgen in androgen-dependent prostate cancer cells.


Subject(s)
Androgens/pharmacology , Fatty Acid Synthases/metabolism , Kruppel-Like Transcription Factors/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Blotting, Western , Cell Line, Tumor , Fatty Acid Synthases/genetics , Gene Expression/drug effects , Humans , Immunoprecipitation , Introns/genetics , Kruppel-Like Transcription Factors/genetics , Male , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sterol Regulatory Element Binding Protein 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...