Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Appl Biochem Biotechnol ; 184(3): 909-918, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28918449

ABSTRACT

The compound 2-deoxy-2-fluoro-α-D-glucopyranosyl fluoride (F2Glc), which is a nonmetabolized superior glucose analogue, is a potent inhibitor of glycogen phosphorylase and pharmacological properties are reported. Glycogen phosphorylase (GP) and glycogen synthase (GS) are responsible of the degradation and synthesis, respectively, of glycogen which is a polymer of glucose units that provides a readily available source of energy in mammals. GP and GS are two key enzymes that modulate cellular glucose and glycogen levels; therefore, these proteins are suggested as potential targets for the treatment of diseases related to glycogen metabolism disorders. We studied by Western Blot technique that F2Glc decreased GP activity, and we also showed that F2Glc did not affect GS activity and its translocation from a uniform cytosolic distribution to the hepatocyte periphery, which is crucial for glycogen synthesis, using immunoblotting and immunofluorescence labeling techniques. F2Glc specifically inhibits glycogenolysis pathway and permits a greater deposition of glycogen. These observations open up the possibility of further develop drugs that act specifically on GP. The ability to selectively inhibit GP, which is a key enzyme for the release of glucose from the hepatic glycogen reserve, may represent a new approach for the treatment of hyperglycemia in type 2 diabetes.


Subject(s)
Deoxyglucose/analogs & derivatives , Glycogen Synthase/biosynthesis , Glycogen Synthase/metabolism , Hepatocytes/metabolism , Animals , Deoxyglucose/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Glycogen , Hepatocytes/cytology , Male , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley
2.
Mol Cell Biochem ; 424(1-2): 203-208, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27785702

ABSTRACT

Glycogen synthase kinase 3ß (GSK3ß) is a ubiquitous serine/threonine kinase and has important roles in glycogen metabolism biosynthesis. Studies have revealed that GSK3ß can directly regulate the glycogen synthase activity, yet little is known about the regulation of GSK3ß on GYS1 gene transcription. Here, we show that overexpression of GSK3ß decreased the mRNA expression level of GYS1. Then we cloned approximately 1.5 kb of pig GYS1 gene promoter region, generated sequential deletion constructs, and evaluated their activity. A gradual increase of the promoter activity was seen with increasing length of the promoter sequence, reaching its highest activity to the sequence corresponding to nt -350 to +224, and then decreased. However, the activities of constructed promoter fragments show different responses to GSK3ß co-transfection. By analyzing a series of GYS1 promoter reporter constructs, we have defined two crucial regions (-1488 to -539, -350 to -147) that are responsible for GSK3ß-induced transcriptional repression. Furthermore, the ChIP results revealed that only the first and second NF-κB sites of GYS1 promoter could bind to p65, and overexpression of GSK3ß induced a significant decrease in p65 binding to the second NF-κB binding site, suggesting that GSK3ß may regulate expression of GYS1 gene through binding to the second rather than the first NF-κB site. These data suggest that the NF-κB plays important roles in the transcriptional activity of pig GYS1 gene regulated by GSK3ß.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase/biosynthesis , Response Elements/physiology , Transcription Factor RelA/metabolism , Transcription, Genetic/physiology , Animals , Cell Line , Glycogen Synthase/genetics , Glycogen Synthase Kinase 3 beta/genetics , Swine , Transcription Factor RelA/genetics
3.
Diabetologia ; 58(7): 1569-78, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25870023

ABSTRACT

AIMS/HYPOTHESIS: Insulin and exercise stimulate skeletal muscle glycogen synthase (GS) activity by dephosphorylation and changes in kinetic properties. The aim of this study was to investigate the effects of insulin, exercise and post-exercise insulin stimulation on GS phosphorylation, activity and substrate affinity in obesity and type 2 diabetes. METHODS: Obese men with type 2 diabetes (n = 13) and weight-matched controls (n = 14) underwent euglycaemic-hyperinsulinaemic clamps in the rested state and 3 h after 60 min of cycling (70% maximal pulmonary oxygen uptake [VO2max]). Biopsies from vastus lateralis muscle were obtained before and after clamps, and before and immediately after exercise. RESULTS: Insulin-stimulated glucose uptake was lower in diabetic patients vs obese controls with or without prior exercise. Post exercise, glucose partitioning shifted away from oxidation and towards storage in both groups. Insulin and, more potently, exercise increased GS activity (fractional velocity [FV]) and substrate affinity in both groups. Both stimuli caused dephosphorylation of GS at sites 3a + 3b, with exercise additionally decreasing phosphorylation at sites 2 + 2a. In both groups, changes in GS activity, substrate affinity and dephosphorylation at sites 3a + 3b by exercise were sustained 3 h post exercise and further enhanced by insulin. Post exercise, reduced GS activity and substrate affinity as well as increased phosphorylation at sites 2 + 2a were found in diabetic patients vs obese controls. CONCLUSIONS/INTERPRETATION: Exercise-induced activation of muscle GS in obesity and type 2 diabetes involves dephosphorylation of GS at sites 3a + 3b and 2 + 2a and enhanced substrate affinity, which is likely to facilitate glucose partitioning towards storage. Lower GS activity and increased phosphorylation at sites 2 + 2a in type 2 diabetes in the recovery period imply an impaired response to exercise.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Exercise , Glycogen Synthase/biosynthesis , Muscle, Skeletal/enzymology , Bicycling , Biopsy , Cohort Studies , Diabetes Mellitus, Type 2/complications , Glucose Clamp Technique , Glycogen/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin/pharmacology , Kinetics , Male , Middle Aged , Obesity/complications , Obesity/metabolism , Phosphorylation , Uridine Diphosphate Glucose/metabolism
4.
Histochem Cell Biol ; 143(3): 313-24, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25371328

ABSTRACT

Diabetic nephropathy (DN) is a major complication of diabetic patients and the leading cause of end-stage renal disease. Glomerular dysfunction plays a critical role in DN, but deterioration of renal function also correlates with tubular alterations. Human DN is characterized by glycogen accumulation in tubules. Although this pathological feature has long been recognized, little information exists about the triggering mechanism. In this study, we detected over-expression of muscle glycogen synthase (MGS) in diabetic human kidney. This enhanced expression suggests the participation of MGS in renal metabolic changes associated with diabetes. HK2 human renal cell line exhibited an intrinsic ability to synthesize glycogen, which was enhanced after over-expression of protein targeting to glycogen. A correlation between increased glycogen amount and cell death was observed. Based on a previous transcriptome study on human diabetic kidney disease, significant differences in the expression of genes involved in glycogen metabolism were analyzed. We propose that glucose, but not insulin, is the main modulator of MGS activity in HK2 cells, suggesting that blood glucose control is the best approach to modulate renal glycogen-induced damage during long-term diabetes.


Subject(s)
Diabetes Mellitus, Type 2/enzymology , Diabetic Nephropathies/enzymology , Gene Expression Regulation, Enzymologic , Glycogen Synthase/biosynthesis , Muscles/enzymology , Aged , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/pathology , Female , Gene Expression Profiling , Glycogen Synthase/metabolism , Humans , Immunohistochemistry , Male , Real-Time Polymerase Chain Reaction
5.
Protein Expr Purif ; 108: 23-29, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25527037

ABSTRACT

We report the successful expression and purification of functional human muscle glycogen synthase (GYS1) in complex with human glycogenin-1 (GN1). Stoichiometric GYS1:GN1 complex was produced by co-expression of GYS1 and GN1 using a bicistronic pFastBac™-Dual expression vector, followed by affinity purification and subsequent size-exclusion chromatography. Mass spectrometry analysis identified that GYS1 is phosphorylated at several well-characterised and uncharacterised Ser/Thr residues. Biochemical analysis, including activity ratio (in the absence relative to that in the presence of glucose-6-phosphate) measurement, covalently attached phosphate estimation as well as phosphatase treatment, revealed that recombinant GYS1 is substantially more heavily phosphorylated than would be observed in intact human or rodent muscle tissues. A large quantity of highly-pure stoichiometric GYS1:GN1 complex will be useful to study its structural and biochemical properties in the future, which would reveal mechanistic insights into its functional role in glycogen biosynthesis.


Subject(s)
Gene Expression , Glucosyltransferases , Glycogen Synthase , Glycoproteins , Multienzyme Complexes , Animals , Glucosyltransferases/biosynthesis , Glucosyltransferases/genetics , Glucosyltransferases/isolation & purification , Glycogen Synthase/biosynthesis , Glycogen Synthase/genetics , Glycogen Synthase/isolation & purification , Glycoproteins/biosynthesis , Glycoproteins/genetics , Glycoproteins/isolation & purification , Humans , Multienzyme Complexes/biosynthesis , Multienzyme Complexes/genetics , Multienzyme Complexes/isolation & purification , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sf9 Cells , Spodoptera
6.
Nat Commun ; 4: 2316, 2013.
Article in English | MEDLINE | ID: mdl-23939267

ABSTRACT

During fasting, animals maintain their energy balance by shifting their energy source from carbohydrates to triglycerides. However, the trigger for this switch has not yet been entirely elucidated. Here we show that a selective hepatic vagotomy slows the speed of fat consumption by attenuating sympathetic nerve-mediated lipolysis in adipose tissue. Hepatic glycogen pre-loading by the adenoviral overexpression of glycogen synthase or the transcription factor TFE3 abolished this liver-brain-adipose axis activation. Moreover, the blockade of glycogenolysis [corrected] through the knockdown of the glycogen phosphorylase gene and the resulting elevation in the glycogen content abolished the lipolytic signal from the liver, indicating that glycogen is the key to triggering this neurocircuitry. These results demonstrate that liver glycogen shortage activates a liver-brain-adipose neural axis that has an important role in switching the fuel source from glycogen to triglycerides under prolonged fasting conditions.


Subject(s)
Adipose Tissue/innervation , Fasting/metabolism , Liver Glycogen/metabolism , Sympathetic Nervous System/metabolism , Triglycerides/metabolism , Adipose Tissue/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Brain/metabolism , Energy Metabolism , Glycogen Phosphorylase/genetics , Glycogen Phosphorylase/metabolism , Glycogen Synthase/biosynthesis , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Glycogenolysis/genetics , Guanethidine/pharmacology , Lipolysis/physiology , Liver/innervation , Liver/metabolism , Male , Mice , Mice, Inbred ICR , Nerve Block , Sympathetic Nervous System/drug effects , Sympatholytics/pharmacology , Vagus Nerve/surgery
7.
J Neurochem ; 118(4): 596-610, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21668450

ABSTRACT

Abnormal regulation of brain glycogen metabolism is believed to underlie insulin-induced hypoglycaemia, which may be serious or fatal in diabetic patients on insulin therapy. A key regulator of glycogen levels is glycogen targeted protein phosphatase 1 (PP1), which dephosphorylates and activates glycogen synthase (GS) leading to an increase in glycogen synthesis. In this study, we show that the gene PPP1R3F expresses a glycogen-binding protein (R3F) of 82.8 kDa, present at the high levels in rodent brain. R3F binds to PP1 through a classical 'RVxF' binding motif and substitution of Phe39 for Ala in this motif abrogates PP1 binding. A hydrophobic domain at the carboxy-terminus of R3F has similarities to the putative membrane binding domain near the carboxy-terminus of striated muscle glycogen targeting subunit G(M)/R(GL), and R3F is shown to bind not only to glycogen but also to membranes. GS interacts with PP1-R3F and is hyperphosphorylated at glycogen synthase kinase-3 sites (Ser640 and Ser644) when bound to R3F(Phe39Ala). Deprivation of glucose or stimulation with adenosine or noradrenaline leads to an increased phosphorylation of PP1-R3F bound GS at Ser640 and Ser644 curtailing glycogen synthesis and facilitating glycogen degradation to provide glucose in astrocytoma cells. Adenosine stimulation also modulates phosphorylation of R3F at Ser14/Ser18.


Subject(s)
Astrocytoma/enzymology , Brain Neoplasms/enzymology , Carrier Proteins/physiology , Extracellular Space/physiology , Glucose/pharmacology , Glycogen Synthase/biosynthesis , Phosphoprotein Phosphatases/physiology , Protein Phosphatase 1/physiology , Signal Transduction/drug effects , Adenosine/pharmacology , Adrenergic alpha-Agonists/pharmacology , Amino Acid Sequence , Animals , Astrocytoma/genetics , Brain/drug effects , Brain/enzymology , Brain Neoplasms/genetics , Carrier Proteins/genetics , Cell Line, Tumor , DNA/biosynthesis , DNA/genetics , Glycogen/metabolism , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutagenesis , Norepinephrine/pharmacology , Phosphoprotein Phosphatases/genetics , Phosphorylation , Protein Phosphatase 1/genetics , RNA/biosynthesis , RNA/genetics , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
8.
Infect Immun ; 79(3): 1044-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21199910

ABSTRACT

We previously demonstrated that plasmid-deficient Chlamydia muridarum retains the ability to infect the murine genital tract but does not elicit oviduct pathology because it fails to activate Toll-like receptor 2 (TLR2). We derived a plasmid-cured derivative of the human genital isolate Chlamydia trachomatis D/UW-3/Cx, strain CTD153, which also fails to activate TLR2, indicating this virulence phenotype is associated with plasmid loss in both C. trachomatis and C. muridarum. As observed with plasmid-deficient C. muridarum, CTD153 displayed impaired accumulation of glycogen within inclusions. Transcriptional profiling of the plasmid-deficient strains by using custom microarrays identified a conserved group of chromosomal loci, the expression of which was similarly controlled in plasmid-deficient C. muridarum strains CM972 and CM3.1 and plasmid-deficient C. trachomatis CTD153. However, although expression of glycogen synthase, encoded by glgA, was greatly reduced in CTD153, it was unaltered in plasmid-deficient C. muridarum strains. Thus, additional plasmid-associated factors are required for glycogen accumulation by this chlamydial species. Furthermore, in C. trachomatis, glgA and other plasmid-responsive chromosomal loci (PRCLs) were transcriptionally responsive to glucose limitation, indicating that additional regulatory elements may be involved in the coordinated expression of these candidate virulence effectors. Glucose-limited C. trachomatis displayed reduced TLR2 stimulation in an in vitro assay. During human chlamydial infection, glucose limitation may decrease chlamydial virulence through its effects on plasmid-responsive chromosomal genes.


Subject(s)
Chlamydia Infections/genetics , Chlamydia muridarum/genetics , Chlamydia trachomatis/genetics , Gene Expression Regulation, Bacterial/genetics , Plasmids/genetics , Toll-Like Receptor 2/metabolism , Animals , Cell Line , Chlamydia Infections/metabolism , Chlamydia muridarum/metabolism , Chlamydia muridarum/pathogenicity , Chlamydia trachomatis/metabolism , Chlamydia trachomatis/pathogenicity , Chromosomes, Bacterial/genetics , Gene Expression , Genetic Loci , Glucose/metabolism , Glycogen/metabolism , Glycogen Synthase/biosynthesis , Glycogen Synthase/genetics , Humans , Inclusion Bodies/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Virulence/genetics
9.
J Biol Chem ; 284(6): 3425-32, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19073609

ABSTRACT

O-Linked N-acetylglucosamine (O-GlcNAc) is a post-translational modification of proteins that functions as a nutrient sensing mechanism. We have previously shown a significant induction of O-GlcNAc modification under conditions of glucose deprivation. Increased O-GlcNAc modification was mediated by increased mRNA for nucleocytoplasmic O-linked N-acetylglucosaminyltransferase (ncOGT). We have investigated the mechanism mediating ncOGT induction with glucose deprivation. The signal does not appear to be general energy depletion because no differences in AMP-dependent kinase protein levels or phosphorylation were observed between glucose-deprived and normal glucose-treated cells. However, treatment of glucose-deprived cells with a small dose (1 mm) of glucosamine blocked the induction of ncOGT mRNA and subsequent increase in O-GlcNAc protein modification, suggesting that decreased hexosamine flux is the signal for ncOGT up-regulation. Consistent with this, treatment of glucose-deprived cells with an inhibitor of O-GlcNAcase (O-(2-acetamido-2-deoxy-D-glucopyranosylidene) amino N-phenyl carbamat) completely prevented the subsequent up-regulation of ncOGT. Glucosamine treatment also resulted in a 40% rescue of the down-regulation of glycogen synthase activity normally seen after glucose deprivation. We conclude that deglycosylation of proteins within the first few hours of glucose deprivation promotes ncOGT induction. These findings suggest a novel negative feedback regulatory loop for OGT and O-GlcNAc regulation.


Subject(s)
Acetylglucosamine/metabolism , Glucose/metabolism , N-Acetylglucosaminyltransferases/biosynthesis , Protein Processing, Post-Translational/physiology , AMP-Activated Protein Kinases/metabolism , Acetylglucosamine/pharmacology , Cell Line, Tumor , Cell Nucleus/enzymology , Cytoplasm/enzymology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Glycogen Synthase/biosynthesis , Humans , Phosphorylation/drug effects , Phosphorylation/physiology , Protein Processing, Post-Translational/drug effects , RNA, Messenger/biosynthesis
10.
Am J Physiol Endocrinol Metab ; 295(4): E798-809, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18577693

ABSTRACT

Cloned mouse embryos display a marked preference for glucose-containing culture medium, with enhanced development to the blastocyst stage in glucose-containing medium attributable mainly to an early beneficial effect during the first cell cycle. This early beneficial effect of glucose is not displayed by parthenogenetic, fertilized, or tetraploid nuclear transfer control embryos, indicating that it is specific to diploid clones. Precocious localization of the glucose transporter SLC2A1 to the cell surface, as well as increased expression of glucose transporters and increased uptake of glucose at the one- and two-cell stages, is also seen in cloned embryos. To examine the role of glucose in early cloned embryo development, we examined glucose metabolism and associated metabolites, as well as mitochondrial ultrastructure, distribution, and number. Clones prepared with cumulus cell nuclei displayed significantly enhanced glucose metabolism at the two-cell stage relative to parthenogenetic controls. Despite the increase in metabolism, ATP content was reduced in clones relative to parthenotes and fertilized controls. Clones at both stages displayed elevated concentrations of glycogen compared with parthenogenetic controls. There was no difference in the number of mitochondria, but clone mitochondria displayed ultrastructural alterations. Interestingly, glucose availability positively affected mitochondrial structure and localization. We conclude that cloned embryos may be severely compromised in terms of ATP-dependent processes during the first two cell cycles and that glucose may exert its early beneficial effects via positive effects on the mitochondria.


Subject(s)
Embryonic Development/physiology , Glucose/physiology , Adenosine Triphosphate/metabolism , Adenylate Kinase/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/physiology , Cloning, Organism , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Female , Fertilization in Vitro , Glycogen/metabolism , Glycogen Synthase/biosynthesis , Glycogen Synthase/genetics , Hybrid Cells , Mice , Microscopy, Electron, Transmission , Oocytes/drug effects , Parthenogenesis , Pregnancy
11.
Arch Biochem Biophys ; 466(2): 283-9, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17880910

ABSTRACT

To investigate the effect of elevation of liver glycogen synthase (GYS2) activity on glucose and glycogen metabolism, we performed adenoviral overexpression of the mutant GYS2 with six serine-to-alanine substitutions in rat primary hepatocytes. Cell-free assays demonstrated that the serine-to-alanine substitutions caused constitutive activity and electrophoretic mobility shift. In rat primary hepatocytes, overexpression of the mutant GYS2 significantly reduced glucose production by 40% and dramatically induced glycogen synthesis via the indirect pathway rather than the direct pathway. Thus, we conclude that elevation of glycogen synthase activity has an inhibitory effect on glucose production in hepatocytes by shunting gluconeogenic precursors into glycogen. In addition, although intracellular compartmentation of glucose-6-phosphate (G6P) remains unclear in hepatocytes, our results imply that there are at least two G6P pools via gluconeogenesis and due to glucose phosphorylation, and that G6P via gluconeogenesis is preferentially used for glycogen synthesis in hepatocytes.


Subject(s)
Alanine/genetics , Glycogen Synthase/biosynthesis , Hepatocytes/metabolism , Serine/genetics , Amino Acid Substitution , Animals , Cells, Cultured , Glucose/biosynthesis , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Glycogen Synthase/genetics , Male , Rats , Rats, Wistar
12.
Cancer Res ; 65(22): 10330-7, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16288022

ABSTRACT

The CCAAT/enhancer binding protein alpha (C/EBPalpha) is vital for establishing normal hepatic energy homeostasis and moderating hepatocellular growth. CEBPA loss-of-function mutations identified in acute myeloid leukemia patients support a tumor suppressor role for C/EBPalpha. Recent work showed reductions of C/EBPalpha levels in human hepatocellular carcinoma with the reductions correlating to tumor size and progression. We investigated the potential of reactivating c/ebpalpha expression during hepatic carcinogenesis to prevent tumor cell growth. We have developed a c/ebpalpha knock-in mouse in which a single-copy c/ebpalpha is regulated by one allele of the alpha-fetoprotein (AFP) gene promoter. The knock-in mice are physically indistinguishable from wild-type (WT) controls. However, knock-in animals were found to deposit fetal hepatic glycogen earlier than WT animals. Quantitative real-time PCR confirmed early c/ebpalpha expression and early glycogen synthase gene activation in knock-in fetuses. We then used diethylnitrosamine to induce hepatocellular carcinoma in our animals. Diethylnitrosamine produced half the number of hepatocellular nodules in knock-in mice as in WT mice. Immunohistochemistry showed reduced C/EBPalpha content in WT nodules whereas knock-in nodules stained strongly for C/EBPalpha. The p21 protein was examined because it mediates a C/EBPalpha growth arrest pathway. Nuclear p21 was absent in WT nodules whereas cytoplasmic p21 was abundant; knock-in nodules were positive for nuclear p21. Interestingly, only C/EBPalpha-positive nodules were positive for nuclear p21, suggesting that C/EBPalpha may be required to direct p21 to the cell nucleus to inhibit growth. Our data establish that controlled C/EBPalpha production can inhibit liver tumor growth in vivo.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/physiology , Liver Glycogen/metabolism , Liver Neoplasms, Experimental/metabolism , Alleles , Animals , CCAAT-Enhancer-Binding Protein-alpha/biosynthesis , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Growth Processes/genetics , Cell Growth Processes/physiology , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Genetic Predisposition to Disease , Glycogen Synthase/biosynthesis , Liver/metabolism , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/prevention & control , Mice , Mice, Transgenic , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , alpha-Fetoproteins/genetics
13.
J Biol Chem ; 279(43): 44740-8, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15292250

ABSTRACT

C/EBPalpha is highly expressed in liver and regulates many genes that are preferentially expressed in liver. Because C/EBPalpha-null mice die soon after birth, it is impossible to analyze the function of C/EBPalpha in the adult with this model. To address the function of C/EBPalpha in adult hepatocytes, liver-specific C/EBPalpha-null mice were produced using a floxed C/EBPalpha allele and the albumin-Cre transgene. Unlike whole body C/EBPalpha-null mice, mice lacking hepatic C/EBPalpha expression did not exhibit hypoglycemia, nor did they show reduced hepatic glycogen in adult. Expression of liver glycogen synthase, phosphoenolpyruvate carboxykinase, and glucose-6-phosphatase remained at normal levels. However, these mice exhibited impaired glucose tolerance due in part to reduced expression of hepatic glucokinase, and hyperammonemia from reduced expression of hepatic carbamoyl phosphate synthase-I. These mice also had reduced serum cholesterol and steatotic livers that was exacerbated with aging. This phenotype could be explained by increased expression of hepatic lipoprotein lipase and reduced expression of microsomal triglyceride transfer protein, apolipoproteins B100, and A-IV. These data demonstrate that hepatic C/EBPalpha is critical for ammonia detoxification and glucose and lipid homeostasis in adult mice.


Subject(s)
Ammonia/metabolism , CCAAT-Enhancer-Binding Protein-alpha/biosynthesis , Glucose/metabolism , Liver/metabolism , Aging , Ammonia/blood , Animals , Apolipoprotein B-100 , Apolipoproteins B/metabolism , Blotting, Northern , Blotting, Western , Cholesterol/metabolism , Chromatography, Liquid , Down-Regulation , Genotype , Glucokinase/biosynthesis , Glucose Tolerance Test , Glucose-6-Phosphatase/biosynthesis , Glycogen/metabolism , Glycogen Synthase/biosynthesis , Hypoglycemia/metabolism , Immunohistochemistry , Lipid Metabolism , Lipoprotein Lipase/biosynthesis , Mice , Mice, Knockout , Organ Size , Phenotype , Phosphoenolpyruvate Carboxykinase (ATP)/biosynthesis , Polymerase Chain Reaction , Time Factors , Tissue Distribution , Urea/metabolism
14.
Am J Physiol Endocrinol Metab ; 286(4): E523-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14656717

ABSTRACT

Insulin stimulates muscle glucose disposal via both glycolysis and glycogen synthesis. Insulin activates glycogen synthase (GS) in skeletal muscle by phosphorylating PKB (or Akt), which in turn phosphorylates and inactivates glycogen synthase kinase 3 (GSK-3), with subsequent activation of GS. A rapamycin-sensitive pathway, most likely acting via ribosomal 70-kDa protein S6 kinase (p70(S6K)), has also been implicated in the regulation of GSK-3 and GS by insulin. Amino acids potently stimulate p70(S6K), and recent studies on cultured muscle cells suggest that amino acids also inactivate GSK-3 and/or activate GS via activating p70(S6K). To assess the physiological relevance of these findings to normal human physiology, we compared the effects of amino acids and insulin on whole body glucose disposal, p70(S6K), and GSK-3 phosphorylation, and on the activity of GS in vivo in skeletal muscle of 24 healthy human volunteers. After an overnight fast, subjects received intravenously either a mixed amino acid solution (1.26 micromol.kg(-1).min(-1) x 6 h, n = 9), a physiological dose of insulin (1 mU.kg(-1).min(-1) euglycemic hyperinsulinemic clamp x 2 h, n = 6), or a pharmacological dose of insulin (20 mU.kg(-1).min(-1) euglycemic hyperinsulinemic clamp x 2 h, n = 9). Whole body glucose disposal rates were assessed by calculating the steady-state glucose infusion rates, and vastus lateralis muscle was biopsied before and at the end of the infusion. Both amino acid infusion and physiological hyperinsulinemia enhanced p70(S6K) phosphorylation without affecting GSK-3 phosphorylation, but only physiological hyperinsulinemia also increased whole body glucose disposal and GS activity. In contrast, a pharmacological dose of insulin significantly increased whole body glucose disposal, p70(S6K), GSK-3 phosphorylation, and GS activity. We conclude that amino acids at physiological concentrations mediate p70(S6K) but, unlike insulin, do not regulate GSK-3 and GS phosphorylation/activity in human skeletal muscle.


Subject(s)
Amino Acids/pharmacology , Glycogen Synthase Kinase 3/biosynthesis , Glycogen Synthase/biosynthesis , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Muscle, Skeletal/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis , Adult , Blood Glucose/metabolism , Blotting, Western , Dose-Response Relationship, Drug , Female , Humans , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Phosphorylation , Stimulation, Chemical
15.
Metabolism ; 52(5): 535-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12759880

ABSTRACT

To examine whether the effects of high-fat feeding on glycogen synthase (GS) activity and mRNA levels differ between diabetes-prone (C57BL/6J) and diabetes-resistant mice (NMRI), we measured GS activity and mRNA levels in muscle from C57BL/6J and NMRI mice fed a high-fat or normal chow diet for 3, 6, and 15 months. As compared with chow feeding, fat feeding increased plasma insulin levels in C57BL/6J mice at 15 months (464 +/- 29 v 267 +/- 47 pmol/L, P =.005), which was associated with elevated plasma glucose levels at 15 months (5.3 +/- 0.3 v 3.8 +/- 0.2 mmol/L, P =.001). Fat feeding increased plasma insulin levels also in NMRI mice at 15 months (705 +/- 145 v 275 +/- 64 pmol/L, P =.01) without, however, a rise of plasma glucose levels. In parallel with increased insulin levels, decreased muscle GS fractional velocity (FV) was observed at 6 (49.0% +/- 2.6% v 69.1% +/- 7.3%, P =.04) and 15 (45.8% +/- 1.8% v 53.4% +/- 1.6 %, P <.01) months but not at 3 months in the fat-fed C57BL/6J mice. Similarly, there was a significant decrease in GS fractional activity at 3 (57.9% +/- 4.3% v 70.4% +/- 2.6 %, P <.03) and 15 (47.3% +/- 2.4% v 56.4% +/- 2.1%, P =.02) but not at 6 months in the fat-fed NMRI mice. The decrease in GS activity was not associated with changes in mRNA levels at any time points. We conclude that (1) fat feeding results in similar elevation of plasma insulin levels and impairs GS activity in C57BL/6J and NMRI mice, and (2) the changes in GS activity do not involve effects on gene expression.


Subject(s)
Dietary Fats/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glycogen Synthase/metabolism , Muscle, Skeletal/enzymology , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus/genetics , Diet , Female , Glycogen Synthase/biosynthesis , Glycogen Synthase/genetics , Insulin/blood , Mice , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
16.
Metabolism ; 49(8): 962-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10954011

ABSTRACT

Glycogen synthase (GS) is the rate-limiting enzyme controlling nonoxidative glucose disposal in skeletal muscle. A reduction in GS activity and an impaired insulin responsiveness are characteristic features of skeletal muscle in type 2 diabetes. These properties also exist in human skeletal muscle cell cultures from type 2 diabetic subjects. To determine the effect of an isolated reduction in GS on skeletal muscle insulin action, cultures from nondiabetic subjects were treated with antisense oligonucleotides (ODNs) to GS to interfere with expression of the gene. Treatment with antisense ODNs reduced GS protein expression by 70% compared with control (scrambled) ODNs (P < .01). GS activity measured at 0.01 mmol/L glucose-6-phosphate (G-6-P) was reduced by antisense ODN treatment. The insulin responsiveness of GS was impaired. Insulin also failed to stimulate glucose incorporation into glycogen after antisense ODN treatment. The cellular glycogen content was lower in antisense ODN-treated cells compared with control ODN. The insulin responsiveness of glucose uptake was abolished by antisense ODN treatment. Thus, reductions in GS expression in human skeletal muscle cells lead to impairments in insulin responsiveness and may play an important role in insulin-resistant states.


Subject(s)
Gene Silencing , Glycogen Synthase/biosynthesis , Insulin Resistance/physiology , Muscle Proteins , Muscle, Skeletal/physiology , ATP-Binding Cassette Transporters/biosynthesis , Adult , Amino Acid Transport System X-AG , Culture Techniques , Down-Regulation/drug effects , Gene Expression Regulation, Enzymologic , Glucose/pharmacokinetics , Glucose Transporter Type 4 , Glycogen/biosynthesis , Glycogen/metabolism , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Humans , Insulin/blood , Insulin Resistance/genetics , Monosaccharide Transport Proteins/biosynthesis , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Oligoribonucleotides, Antisense/genetics , Oligoribonucleotides, Antisense/pharmacology , Phosphatidylethanolamines/pharmacology , RNA, Messenger/genetics
17.
Arch Biochem Biophys ; 368(2): 291-7, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10441380

ABSTRACT

Intensively treating type I diabetics with continuous subcutaneous insulin infusions or multiple daily insulin injections to normalize mean blood glucose concentrations significantly reduces the onset of secondary diabetic complications when compared to conventionally treated diabetics. Our studies focused on characterizing hepatic enzyme expression in intensively and conventionally treated diabetic rats. Alloxan-induced diabetic rats were conventionally treated with insulin injected twice daily or intensively treated with similar daily dosages of insulin administered via a surgically implanted osmotic pump. Our results demonstrate a significant difference in hepatic enzyme expression when these treatment regimes are compared. In conventionally treated diabetic rats, phosphoenolpyruvate carboxykinase (PEPCK) protein and mRNA levels remained slightly elevated when compared to normal animals, glycogen phosphorylase (GP) protein levels were still slightly decreased, and glycogen synthase (GS) protein and mRNA levels remained at the elevated levels observed in untreated diabetics. In contrast, the protein and mRNA levels of all three enzymes were normalized in the insulin pump-treated animals. These results suggest that intensive insulin therapy improves glycemia directly by normalizing hepatic gene expression while conventional insulin therapy normalizes plasma glucose concentrations indirectly.


Subject(s)
Carbohydrate Metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Insulin/administration & dosage , Liver/metabolism , Alloxan , Animals , Diabetes Mellitus, Experimental/chemically induced , Glycogen Synthase/biosynthesis , Infusion Pumps , Male , Phosphoenolpyruvate Carboxykinase (GTP)/biosynthesis , Phosphorylases/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley
18.
Dan Med Bull ; 46(1): 13-34, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10081651

ABSTRACT

When whole body insulin-stimulated glucose disposal rate is measured in man applying the euglycaemic, hyperinsulinaemic clamp technique it has been shown that approximately 75% of glucose is taken up by skeletal muscle. After the initial transport step, glucose is rapidly phosphorylated to glucose-6-phosphate and routed into the major pathways of either glucose storage as glycogen or the glycolytic/tricarboxylic acid pathway. Glucose uptake in skeletal muscle involves-the activity of specific glucose transporters and hexokinases, whereas, phosphofructokinase and glycogen synthase hold critical roles in glucose oxidation/glycolysis and glucose storage, respectively. Glucose transporters and glycogen synthase activities are directly and acutely stimulated by insulin whereas the activities of hexokinases and phosphofructokinase may primarily be allosterically regulated. The aim of the review is to discuss our present knowledge of the activities and gene expression of hexokinase II (HKII), phosphofructokinase (PFK) and glycogen synthase (GS) in human skeletal muscle in states of altered insulin-stimulated glucose metabolism. My own experimental studies have comprised patients with disorders characterized by insulin resistance like non-insulin-dependent diabetes mellitus (NIDDM) and insulin-dependent diabetes mellitus (IDDM) before and after therapeutic interventions, patients with microvascular angina and patients with severe insulin resistant diabetes mellitus and congenital muscle fiber type disproportion myopathy as well as athletes who are in a state of improved insulin sensitivity. By applying the glucose insulin clamp method in combination with nuclear magnetic resonance 31P spectroscopy to normoglycaemic or hyperglycaemic insulin resistant subjects impairment of insulin-stimulated glucose transport and/or phosphorylation in skeletal muscle has been shown. In states characterized by insulin resistance but normoglycaemia, the activity of HKII measured in needle revealed any genetic variability that contributes to explain the decreased muscle levels of GS mRNA or the decreased activity and activation of muscle GS in NIDDM patients and their glucose tolerant but insulin resistant relatives. Thus, the causes of impaired insulin-stimulated glycogen synthesis of skeletal muscle in normoglycaemic insulin resistant subjects are likely to be found in the insulin signalling network proximal to the GS protein. In insulin resistant diabetic patients the impact of these yet unknown abnormalities may be accentuated by the prevailing hyperglycaemia and hyperlipidaemia. Endurance training in young healthy subjects results in improved insulin-stimulated glucose disposal rates, predominantly due to an increased glycogen synthesis rate in muscle, which is paralleled by an increased total GS activity, increased GS mRNA levels and enhanced insulin-stimulated activation of GS. These changes are probably due to local contraction-dependent mechanisms. Likewise, one-legged exercise training has been reported to increase the basal concentration of muscle GS mRNA in NIDDM patients to a level similar to that seen in control subjects although insulin-stimulated glucose disposal rates remain reduced in NIDDM patients. In the insulin resistant states examined so far, basal and insulin-stimulated glucose oxidation rate at the whole body level and PFK activity in muscle are normal. In parallel, no changes have been found in skeletal muscle levels of PFK mRNA and immunoreactive protein in NIDDM or IDDM patients. In endurance trained subjects insulin-stimulated whole body glucose oxidation rate is often increased. However, depending on the intensity and frequency, physical exercise may induce an increased, a decreased or an unaltered level of muscle PFK activity. In athletes the muscle PFK mRNA is similar to what is found in sedentary subjects whereas the immunoreactive PFK protein concentration is decreased.


Subject(s)
Gene Expression Regulation , Glucose/metabolism , Glycogen Synthase/metabolism , Hexokinase/metabolism , Insulin/pharmacology , Muscle, Skeletal/enzymology , Phosphofructokinase-1/metabolism , Animals , Glycogen Synthase/biosynthesis , Glycogen Synthase/genetics , Hexokinase/biosynthesis , Hexokinase/genetics , Humans , Hypoglycemic Agents/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phosphofructokinase-1/biosynthesis , Phosphofructokinase-1/genetics
19.
FASEB J ; 12(15): 1701-12, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9837860

ABSTRACT

Complete spinal cord lesion leads to profound metabolic abnormalities and striking changes in muscle morphology. Here we assess the effects of electrically stimulated leg cycling (ESLC) on whole body insulin sensitivity, skeletal muscle glucose metabolism, and muscle fiber morphology in five tetraplegic subjects with complete C5-C7 lesions. Physical training (seven ESLC sessions/wk for 8 wk) increased whole body insulin-stimulated glucose uptake by 33+/-13%, concomitant with a 2.1-fold increase in insulin-stimulated (100 microU/ml) 3-O-methylglucose transport in isolated vastus lateralis muscle. Physical training led to a marked increase in protein expression of GLUT4 (378+/-85%), glycogen synthase (526+/-146%), and hexokinase II (204+/-47%) in vastus lateralis muscle, whereas phosphofructokinase expression (282+/-97%) was not significantly changed. Hexokinase II activity was significantly increased, whereas activity of phosphofructokinase, glycogen synthase, and citrate synthase was not changed after training. Muscle fiber type distribution and fiber area were markedly altered compared to able-bodied subjects before ESLC training, with no change noted in either parameter after ECSL training. In conclusion, muscle contraction improves insulin action on whole body and cellular glucose uptake in cervical cord-injured persons through a major increase in protein expression of key genes involved in the regulation of glucose metabolism. Furthermore, improvements in insulin action on glucose metabolism are independent of changes in muscle fiber type distribution.


Subject(s)
Exercise , Glucose/metabolism , Homeostasis , Muscle Proteins , Quadriplegia/metabolism , Spinal Cord Injuries/metabolism , 3-O-Methylglucose/metabolism , Biological Transport , Electric Stimulation Therapy , Glucose Clamp Technique , Glucose Transporter Type 4 , Glycogen/analysis , Glycogen Synthase/biosynthesis , Hexokinase/biosynthesis , Humans , Insulin/pharmacology , Leg , Male , Monosaccharide Transport Proteins/biosynthesis , Muscle Fibers, Skeletal/ultrastructure , Quadriplegia/therapy , Spinal Cord Injuries/therapy
20.
Diabetes ; 47(9): 1392-8, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9726226

ABSTRACT

Insulin resistance, as is found in skeletal muscle of individuals with obesity and NIDDM, appears to involve a reduced capacity of the hormone to stimulate glucose uptake and/or phosphorylation. The glucose phosphorylation step, as catalyzed by hexokinase II, has been described as rate limiting for glucose disposal in muscle, but overexpression of this enzyme under control of a muscle-specific promoter in transgenic mice has had limited metabolic impact. In the current study, we investigated in a cultured muscle model whether expression of glucokinase, which in contrast to hexokinase II is not inhibited by glucose-6-phosphate (G-6-P), would have a pronounced metabolic impact. We used a recombinant adenovirus containing the cDNA-encoding rat liver glucokinase (AdCMV-GKL) to increase the glucose phosphorylating activity in cultured human muscle cells by fourfold. G-6-P levels increased in AdCMV-GKL-treated cells in a glucose concentration-dependent manner over the range of 1-30 mmol/l, whereas the much smaller increases in G-6-P in control cells were maximal at glucose concentrations <5 mmol/l. Further, cells expressing glucokinase accumulated 17 times more 2-deoxyglucose-6-phosphate than control cells. In AdCMV-GKL-treated cells, the time-dependent rise in G-6-P correlated with an increase in the activity ratio of glycogen synthase. AdCMV-GKL-treated cells also exhibited a 2.5- to 3-fold increase in glycogen content and a four- to fivefold increase in glycolytic flux, proportional to the increase in glucose phosphorylating capacity. All of these observations were made in the absence of insulin. Thus we concluded that expression of glucokinase in cultured human muscle cells results in proportional increases in insulin-independent glucose disposal, and that muscle glucose storage and utilization becomes controlled in a glucose concentration-dependent manner in AdCMV-GKL-treated cells. These results encourage testing whether delivery of glucokinase to muscle in vivo has an impact on glycemic control, which could be a method for circumventing the failure of insulin to stimulate glucose uptake and/or phosphorylation in muscle normally in insulin-resistant subjects.


Subject(s)
Glucokinase/biosynthesis , Glucose/metabolism , Insulin/pharmacology , Muscle, Skeletal/metabolism , Adenoviridae , Animals , Biological Transport , Cells, Cultured , DNA, Complementary , Deoxyglucose/metabolism , Gene Expression , Genetic Vectors , Glucokinase/genetics , Glucosephosphates/metabolism , Glycogen/biosynthesis , Glycogen Synthase/biosynthesis , Humans , Kinetics , Liver/enzymology , Mice , Mice, Transgenic , Muscle, Skeletal/drug effects , Phosphorylases/biosynthesis , Rats , Recombinant Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...