Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
FEBS J ; 273(9): 1989-99, 2006 May.
Article in English | MEDLINE | ID: mdl-16640562

ABSTRACT

Hepatic insulin resistance in the leptin-receptor defective Zucker fa/fa rat is associated with impaired glycogen synthesis and increased activity of phosphorylase-a. We investigated the coupling between phosphorylase-a and glycogen synthesis in hepatocytes from fa/fa rats by modulating the concentration of phosphorylase-a. Treatment of hepatocytes from fa/fa rats and Fa/? controls with a selective phosphorylase inhibitor caused depletion of phosphorylase-a, activation of glycogen synthase and stimulation of glycogen synthesis. The flux-control coefficient of phosphorylase on glycogen synthesis was glucose dependent and at 10 mm glucose was higher in fa/fa than Fa/? hepatocytes. There was an inverse correlation between the activities of glycogen synthase and phosphorylase-a in both fa/fa and Fa/? hepatocytes. However, fa/fa hepatocytes had a higher activity of phosphorylase-a, for a corresponding activity of glycogen synthase. This defect was, in part, normalized by expression of the glycogen-targeting protein, PTG. Hepatocytes from fa/fa rats had normal expression of the glycogen-targeting proteins G(L) and PTG but markedly reduced expression of R6. Expression of R6 protein was increased in hepatocytes from Wistar rats after incubation with leptin and insulin. Diminished hepatic R6 expression in the leptin-receptor defective fa/fa rat may be a contributing factor to the elevated phosphorylase activity and/or its high control strength on glycogen synthesis.


Subject(s)
Glycogen/biosynthesis , Hepatocytes/enzymology , Insulin Resistance/genetics , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphorylase a/chemistry , Protein Subunits/antagonists & inhibitors , Protein Subunits/biosynthesis , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Cells, Cultured , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Female , Glycogen/metabolism , Glycogen/physiology , Insulin/chemistry , Intracellular Signaling Peptides and Proteins , Leptin/chemistry , Male , Obesity/enzymology , Obesity/genetics , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/metabolism , Phosphorylase a/physiology , Protein Subunits/metabolism , Rats , Rats, Wistar , Rats, Zucker , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Receptors, Leptin
2.
J Biol Chem ; 279(45): 46474-82, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15322104

ABSTRACT

Expression of the glycogen-targeting protein PTG promotes glycogen synthase activation and glycogen storage in various cell types. In this study, we tested the contribution of phosphorylase inactivation to the glycogenic action of PTG in hepatocytes by using a selective inhibitor of phosphorylase (CP-91149) that causes dephosphorylation of phosphorylase a and sequential activation of glycogen synthase. Similar to CP-91194, graded expression of PTG caused a concentration-dependent inactivation of phosphorylase and activation of glycogen synthase. The latter was partially counter-acted by the expression of muscle phosphorylase and was not additive with the activation by CP-91149, indicating that it is in part secondary to the inactivation of phosphorylase. PTG expression caused greater stimulation of glycogen synthesis and translocation of glycogen synthase than CP-91149, and the translocation of synthase could not be explained by accumulation of glycogen, supporting an additional role for glycogen synthase translocation in the glycogenic action of PTG. The effects of PTG expression on glycogen synthase and glycogen synthesis were additive with the effects of glucokinase expression, confirming the complementary roles of depletion of phosphorylase a (a negative modulator) and elevated glucose 6-phosphate (a positive modulator) in potentiating the activation of glycogen synthase. PTG expression mimicked the inactivation of phosphorylase caused by high glucose and counteracted the activation caused by glucagon. The latter suggests a possible additional role for PTG on phosphorylase kinase inactivation.


Subject(s)
Glycogen Synthase/metabolism , Glycogen/physiology , Hepatocytes/metabolism , Phosphorylases/metabolism , Adenoviridae/genetics , Amides/pharmacology , Animals , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Glucagon/chemistry , Glucokinase/metabolism , Glucose/metabolism , Glucose-6-Phosphate/metabolism , Glycogen/chemistry , Glycogen/metabolism , Immunoblotting , Indoles/pharmacology , Male , Models, Biological , Muscles/enzymology , Phosphorylase Kinase/metabolism , Phosphorylases/antagonists & inhibitors , Phosphorylation , Protein Binding , Protein Transport , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Time Factors
3.
Diabetes ; 53(3): 535-41, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14988235

ABSTRACT

Glucokinase (GK) has a major role in the control of blood glucose homeostasis and is a strong potential target for the pharmacological treatment of type 2 diabetes. We report here the mechanism of action of two novel and potent direct activators of GK: 6-[(3-isobutoxy-5-isopropoxybenzoyl)amino]nicotinic acid(GKA1) and 5-([3-isopropoxy-5-[2-(3-thienyl)ethoxy]benzoyl]amino)-1,3,4-thiadiazole-2-carboxylic acid(GKA2), which increase the affinity of GK for glucose by 4- and 11-fold, respectively. GKA1 increased the affinity of GK for the competitive inhibitor mannoheptulose but did not affect the affinity for the inhibitors palmitoyl-CoA and the endogenous 68-kDa regulator (GK regulatory protein [GKRP]), which bind to allosteric sites or to N-acetylglucosamine, which binds to the catalytic site. In hepatocytes, GKA1 and GKA2 stimulated glucose phosphorylation, glycolysis, and glycogen synthesis to a similar extent as sorbitol, a precursor of fructose 1-phosphate, which indirectly activates GK through promoting its dissociation from GKRP. Consistent with their effects on isolated GK, these compounds also increased the affinity of hepatocyte metabolism for glucose. GKA1 and GKA2 caused translocation of GK from the nucleus to the cytoplasm. This effect was additive with the effect of sorbitol and is best explained by a "glucose-like" effect of the GK activators in translocating GK to the cytoplasm. In conclusion, GK activators are potential antihyperglycemic agents for the treatment of type 2 diabetes through the stimulation of hepatic glucose metabolism by a mechanism independent of GKRP.


Subject(s)
Glucokinase/metabolism , Glucose/metabolism , Hepatocytes/enzymology , Liver/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/metabolism , Cloning, Molecular , DNA, Complementary/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/metabolism , Escherichia coli/enzymology , Escherichia coli/metabolism , Glucokinase/antagonists & inhibitors , Glucose/pharmacology , Humans , Intracellular Signaling Peptides and Proteins , Kinetics , Liver/enzymology , Male , Polymerase Chain Reaction , Rats , Rats, Wistar , Sorbitol/pharmacology
4.
Biochem J ; 377(Pt 1): 195-204, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-13678417

ABSTRACT

The role of glucose 6-P (glucose 6-phosphate) in regulating the activation state of glycogen synthase and its translocation is well documented. In the present study, we investigated the effects of glucose 6-P on the activation state and compartmentation of phosphorylase in hepatocytes. Glucose 6-P levels were modulated in hepatocytes by glucokinase overexpression or inhibition with 5-thioglucose and the effects of AMP were tested using AICAR (5-aminoimidazole-4-carboxamide 1-beta-D-ribofuranoside), which is metabolized to an AMP analogue. Inhibition of glucokinase partially counteracted the effect of glucose both on the inactivation of phosphorylase and on the translocation of phosphorylase a from a soluble to a particulate fraction. The increase in glucose 6-P caused by glucokinase overexpression caused translocation of phosphorylase a to the pellet and had additive effects with glucose on inactivation of phosphorylase. It decreased the glucose concentration that caused half-maximal inactivation from 20 to 11 mM, indicating that it acts synergistically with glucose. AICAR activated phosphorylase and counteracted the effect of glucose 6-P on phosphorylase inactivation. However, it did not counteract translocation of phosphorylase by glucose 6-P. Glucose 6-P and AICAR had opposite effects on the activation state of glycogen synthase, but they had additive effects on translocation of the enzyme to the pellet. There was a direct correlation between the translocation of phosphorylase a and of glycogen synthase to the pellet, suggesting that these enzymes translocate in tandem. In conclusion, glucose 6-P causes both translocation of phosphorylase and inactivation, indicating a more complex role in the regulation of glycogen metabolism than can be explained from regulation of glycogen synthase alone.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Glucose-6-Phosphate/physiology , Glucose/pharmacology , Hepatocytes/enzymology , Phosphorylases/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Compartmentation , Cells, Cultured , Glucokinase/metabolism , Glycogen Synthase/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Protein Transport , Rats , Rats, Wistar , Ribonucleotides/pharmacology
5.
Eur J Biochem ; 270(13): 2773-81, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12823547

ABSTRACT

Multiple signalling pathways are involved in the mechanism by which insulin stimulates hepatic glycogen synthesis. In this study we used selective inhibitors of glycogen synthase kinase-3 (GSK-3) and an allosteric inhibitor of phosphorylase (CP-91149) that causes dephosphorylation of phosphorylase a, to determine the relative contributions of inactivation of GSK-3 and dephosphorylation of phosphorylase a as alternative pathways in the stimulation of glycogen synthesis by insulin in hepatocytes. GSK-3 inhibitors (SB-216763 and Li+) caused a greater activation of glycogen synthase than insulin (90% vs. 40%) but a smaller stimulation of glycogen synthesis (30% vs. 150%). The contribution of GSK-3 inactivation to insulin stimulation of glycogen synthesis was estimated to be less than 20%. Dephosphorylation of phosphorylase a with CP-91149 caused activation of glycogen synthase and translocation of the protein from a soluble to a particulate fraction and mimicked the stimulation of glycogen synthesis by insulin. The stimulation of glycogen synthesis by phosphorylase inactivation cannot be explained by either inhibition of glycogen degradation or activation of glycogen synthase alone and suggests an additional role for translocation of synthase. Titrations with the phosphorylase inactivator showed that stimulation of glycogen synthesis by insulin can be largely accounted for by inactivation of phosphorylase over a wide range of activities of phosphorylase a. We conclude that a signalling pathway involving dephosphorylation of phosphorylase a leading to both activation and translocation of glycogen synthase is a critical component of the mechanism by which insulin stimulates hepatic glycogen synthesis. Selective inactivation of phosphorylase can mimic insulin stimulation of hepatic glycogen synthesis.


Subject(s)
Glycogen/biosynthesis , Hepatocytes/metabolism , Insulin/metabolism , Phosphorylase a/metabolism , Signal Transduction/physiology , Amides/metabolism , Animals , Biological Transport/physiology , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/metabolism , Glucose/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Hepatocytes/cytology , Indoles/metabolism , Male , Phosphorylase a/antagonists & inhibitors , Rats , Rats, Wistar
6.
Diabetes ; 52(6): 1333-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12765941

ABSTRACT

High glucose concentration suppresses hepatic glycogenolysis by allosteric inhibition and dephosphorylation (inactivation) of phosphorylase-a. The latter effect is attributed to a direct effect of glucose on the conformation of phosphorylase-a. Although glucose-6-phosphate (G6P), like glucose, stimulates dephosphorylation of phosphorylase-a by phosphorylase phosphatase, its physiological role in regulating glycogenolysis in intact hepatocytes has not been tested. We show in this study that metabolic conditions associated with an increase in G6P, including glucokinase overexpression and incubation with octanoate or dihydroxyacetone, cause inactivation of phosphorylase. The latter conditions also inhibit glycogenolysis. The activity of phosphorylase-a correlated inversely with the G6P concentration within the physiological range. The inhibition of glycogenolysis and inactivation of phosphorylase-a caused by 10 mmol/l glucose can be at least in part counteracted by inhibition of glucokinase with 5-thioglucose, which lowers G6P. In conclusion, metabolic conditions that alter the hepatic G6P content affect glycogen metabolism not only through regulation of glycogen synthase but also through regulation of the activation state of phosphorylase. Dysregulation of G6P in diabetes by changes in activity of glucokinase or glucose 6-phosphatase may be a contributing factor to impaired suppression of glycogenolysis by hyperglycemia.


Subject(s)
Dihydroxyacetone/pharmacology , Glucose-6-Phosphate/metabolism , Liver Glycogen/biosynthesis , Liver/metabolism , Phosphorylase a/metabolism , Animals , Caprylates/pharmacology , Cells, Cultured , Glucokinase/metabolism , Homeostasis , Kinetics , Male , Models, Biological , Phosphorylase a/antagonists & inhibitors , Rats , Rats, Wistar , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...