Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters










Publication year range
1.
FEBS Lett ; 587(10): 1579-86, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23587483

ABSTRACT

Both radiotherapy and most effective chemotherapeutic agents induce different types of DNA damage. Here we show that tungstate modulates cell response to DNA damaging agents. Cells treated with tungstate were more sensitive to etoposide, phleomycin and ionizing radiation (IR), all of which induce DNA double-strand breaks (DSBs). Tungstate also modulated the activation of the central DSB signalling kinase, ATM, in response to these agents. These effects required the functionality of the Mre11-Nbs1-Rad50 (MRN) complex and were mimicked by the inhibition of PP2A phosphatase. Therefore, tungstate may have adjuvant activity when combined with DNA-damaging agents in the treatment of several malignancies.


Subject(s)
Cell Cycle Proteins/physiology , DNA Damage/drug effects , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Proteins/physiology , Tungsten Compounds/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cells, Cultured , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , HEK293 Cells , HeLa Cells , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , Radiation Dosage , Radiation, Ionizing , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Tumor Suppressor Proteins/metabolism
2.
FEBS Lett ; 586(3): 270-6, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22245679

ABSTRACT

Tungstate counteracts diabetes and obesity in animal models, but its molecular mechanisms remain elusive. Our Saccharomyces cerevisiae-based approach has found that tungstate alleviated the growth defect induced by nutrient stress and enhanced the activation of the GCN pathway. Tungstate relieved the sensitivity to starvation of a gcn2-507 yeast hypomorphic mutant, indicating that tungstate modulated the GCN pathway downstream of Gcn2p. Interestingly, tungstate inhibited Glc7p and PP1 phosphatase activity, both negative regulators of the GCN pathway in yeast and humans, respectively. Accordingly, overexpression of a dominant-negative Glc7p mutant in yeast mimicked tungstate effects. Therefore tungstate alleviates nutrient stress in yeast by in vivo inhibition of Glc7p. These data uncover a potential role for tungstate in the treatment of PP1 and GCN related diseases.


Subject(s)
Anti-Obesity Agents/pharmacology , Antineoplastic Agents/pharmacology , Hypoglycemic Agents/pharmacology , Protein Phosphatase 1/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Tungsten Compounds/pharmacology , Amino Acids/deficiency , Drug Synergism , Food , Humans , Mutation , Protein Serine-Threonine Kinases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Tacrolimus/pharmacology , Triazoles/chemistry , Triazoles/pharmacology
3.
Diabetologia ; 54(10): 2639-48, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21811873

ABSTRACT

AIMS/HYPOTHESIS: Glycogen deposition is impaired in diabetes, thus contributing to the development of hyperglycaemia. Several glucose-lowering strategies have attempted to increase liver glycogen deposition by modulating targets, which eventually trigger the activation of liver glycogen synthase (LGS). However, these targets also alter several other biological processes, and therefore their therapeutic use may be limited. Here we tested the approach of directly activating LGS and evaluated the potential of this strategy as a possible treatment for diabetes. METHODS: In this study, we examined the efficacy of directly overproducing a constitutively active form of LGS in the liver to ameliorate streptozotocin-induced diabetes in rats. RESULTS: Activated mutant LGS overproduction in the liver of streptozotocin-induced diabetic rats normalised liver glycogen content, despite low levels of glucokinase and circulating insulin. Moreover, this overproduction led to a decrease in food intake and in the production of the main gluconeogenic enzymes, glucose-6-phosphatase, fructose-1,6-bisphosphatase and phosphoenolpyruvate carboxykinase. The resulting combined effect was a reduction in hyperglycaemia. CONCLUSIONS/INTERPRETATION: The restoration of liver glycogen ameliorated diabetes and therefore is considered a potential strategy for the treatment of this disease.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Hyperglycemia/metabolism , Hyperglycemia/therapy , Liver Glycogen/metabolism , Animals , Blood Glucose/metabolism , Eating/genetics , Eating/physiology , Fructose-Bisphosphatase/metabolism , Gluconeogenesis , Glucose-6-Phosphatase/metabolism , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Hepatocytes/metabolism , Hyperglycemia/genetics , Immunoblotting , Insulin/metabolism , Liver/metabolism , Male , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Rats , Rats, Wistar
4.
Reprod Fertil Dev ; 23(3): 468-80, 2011.
Article in English | MEDLINE | ID: mdl-21426864

ABSTRACT

The main aim of the present work was to test the effects of glucose and fructose on the phosphorylation levels of proteins linked to the control of overall sperm function in two species with very different metabolic characteristics, dog and boar. Incubation of dog spermatozoa with 10mM glucose increased serine phosphorylation of proteins related to cell cycle and signal transduction including cyclins B and E, Cdk2, Cdk6, Cdc6, PYK2, c-kit, Raf-1, TRK and several protein phosphatases. Incubation of dog spermatozoa with 10mM fructose decreased serine phosphorylation levels of cyclins B and D3, Cdk1/Cdc2, Cdk2, Cdk6, Akt, PI3 kinase, ERK-1 and protein kinase C. Incubation of boar spermatozoa with glucose or fructose did not modify any of the phosphorylation patterns studied. Given that one important difference between dog and boar spermatozoa is the presence of glucokinase (GK) in dog but not in boar, GK-transfected COS7 cells were incubated with either 10mM glucose or 10mM fructose. Incubation of GK-transfected cells with fructose decreased serine phosphorylation of cyclin A, ERK-2 and Hsp-70. In contrast, incubation of control COS7 cells with fructose increased serine phosphorylation of Cdk6, Cdk1/Cdc2, protein kinase C and Hsp-70. Incubation with glucose did not induce any significant effect. Our results indicate that monosaccharides act as signalling compounds in dog spermatozoa after ejaculation through changes in the phosphorylation levels of specific proteins. One of the factors that may be related to the action of sugars is the equilibrium of the total sperm hexokinase activity, in which the presence or absence of GK appears to be relevant.


Subject(s)
Dogs/physiology , Fructose/pharmacology , Glucose/pharmacology , Spermatozoa/physiology , Swine/physiology , Acrosome Reaction/drug effects , Acrosome Reaction/physiology , Animals , Blotting, Western , COS Cells , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chlorocebus aethiops , Male , Oligonucleotide Array Sequence Analysis , Phosphorylation , Signal Transduction , Sperm Capacitation/drug effects , Sperm Capacitation/physiology , Spermatozoa/drug effects , Transfection/veterinary
5.
Diabetologia ; 51(7): 1285-95, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18483800

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to investigate the action of the glucose-lowering compound sodium tungstate on glucose transport in muscle myotubes and to unravel the molecular events underlying the effects observed. METHODS: We studied the effects of tungstate on 2-deoxy-D: -glucose uptake, levels and translocation of the glucose transporters GLUT4 and GLUT1, and Glut4 (also known as Slc2a4) promoter activity. We also measured the modifications of individual components of the signalling pathways involved in the effects observed. RESULTS: Tungstate increased 2-deoxy-D: -glucose uptake in differentiated L6 myotubes through an increase in the total amount and translocation of GLUT4 transporter. The effects on glucose uptake were additive to those of insulin. Tungstate activated transcription of the Glut4 promoter, as shown by an increase in Glut4 mRNA, and by a promoter reporter assay. The assay of deletions of the Glut4 promoter indicated that the effect of tungstate is mediated by the myocyte enhancer factor 2 (MEF2)-binding domain. Accordingly, MEF2 levels and DNA binding activities were increased in response to the treatment. Tungstate-induced glucose uptake and GLUT4 transcriptional activation were dependent on the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2), while no changes were observed in the phosphorylation state of the beta subunit of the insulin receptor, in the phosphatidylinositol 3-kinase pathway or in the activation of 5'AMP-activated protein kinase. CONCLUSIONS/INTERPRETATION: Tungstate activates glucose uptake in myotubes through a novel ERK1/2-dependent mechanism. This effect is exerted by an increase in the content and translocation of the GLUT4 transporter. This is the first report of a glucose-lowering compound activating Glut4 transcription through an ERK1/2-dependent increase in MEF2 levels.


Subject(s)
Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , MAP Kinase Signaling System/drug effects , Myoblasts/drug effects , Myogenic Regulatory Factors/metabolism , Tungsten Compounds/pharmacology , Animals , Cells, Cultured , Deoxyglucose/metabolism , Deoxyglucose/pharmacology , Glucose/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/physiopathology , MAP Kinase Signaling System/physiology , MEF2 Transcription Factors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Skeletal/cytology , Myoblasts/cytology , Myoblasts/physiology , Promoter Regions, Genetic/physiology , Rats , Transcription, Genetic/drug effects , Transfection , Tritium
6.
Av. diabetol ; 23(6): 441-446, nov.-dic. 2007. tab, graf, ilus
Article in English | IBECS | ID: ibc-135704

ABSTRACT

Introduction: Sodium tungstate is an effective anti-diabetic agent in several animal models of diabetes mellitus in both short- and long-term treatments. Aims: To further characterize its therapeutic application, we studied whether this compound could act in autoimmune diabetes in the NOD (non-obese diabetic) mouse. Material and methods: Four-week-old female mice were given sodium tungstate for 24 weeks. Blood glucose was measured every 2 days throughout the entire experimental period. At the end of treatment, morphometric analysis of the pancreas was performed. Alternatively, diabetic mice were treated with tungstate and liver enzyme activity was determined. Results: We found that tungstate treatment delayed diabetes onset by 6 weeks. In addition, treated mice exhibited lower hyperglycemia at the onset of the disease and this parameter remained low until the end of treatment. Tungstate treatment had no effect on either the severity of insulitis or on β-cell mass. However, tungstate treatment induced a recovery of liver glucokinase and pyruvate kinase activities in diabetic animals. Conclusions: Administration of sodium tungstate to NOD mice corroborates its anti-diabetic properties, delaying diabetes onset and diminishing its incidence. The results indicate that, in the NOD mouse, as in other animal models, the liver is one of the main targets of tungstate actions (AU)


No disponible


Subject(s)
Animals , Tungsten Compounds/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Diabetes Mellitus/drug therapy , Disease Models, Animal , Protective Agents/pharmacokinetics , Mice, Inbred NOD , Liver , Pancreas , Immunohistochemistry/methods , B-Lymphocytes
7.
Hum Reprod ; 22(8): 2128-35, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17588954

ABSTRACT

BACKGROUND: Diabetes induces great alterations in female reproductive function. We analyzed the effects of tungstate, an anti-diabetic agent, on the reproductive function of healthy and diabetic female rats. METHODS: Healthy and streptozotocin-induced diabetic rats were treated with sodium tungstate (2 mg/ml in their drinking water) for 12 weeks. Markers of reproductive function and diabetes were measured in serum, and in uterus and ovaries by Western blot or RT-PCR. Reproductive function was also assessed by mating. RESULTS: Diabetic rats showed great impairment of libido, which was accompanied by a total loss of fertility (P < 0.05) and a decrease in the serum levels of FSH (P < 0.05) and LH (P < 0.05) compared with healthy rats. Tungstate treatment of diabetic rats partially recovered libido while fertility rate increased to 66.6%. This improvement was accompanied by a recovery of serum FSH (to a level higher than healthy rats) and LH. Moreover, tungstate treatment normalized ovarian expression of GLUT 3 hexose transporter, and estrogen, progesterone and FSH receptors, whereas only GLUT 3 and FSH receptors were normalized in the uterus. CONCLUSIONS: Our results indicate that the alterations in female reproduction in diabetes were partially reversed after tungstate treatment by a mechanism(s) involving the normalization of serum FSH and LH levels, and ovarian and uterine expression of FSH receptors and GLUT3.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Reproduction/drug effects , Sexual Behavior, Animal/drug effects , Tungsten Compounds/pharmacology , Animals , Diabetes Mellitus, Experimental/drug therapy , Female , Glucose Transporter Type 3/biosynthesis , Litter Size/drug effects , Ovary/drug effects , Ovary/metabolism , Pregnancy , Rats , Rats, Wistar , Receptor, Insulin/biosynthesis , Receptors, Estrogen/biosynthesis , Receptors, FSH/biosynthesis , Receptors, LH/biosynthesis , Receptors, Progesterone/biosynthesis , Receptors, Prolactin/biosynthesis , Streptozocin , Tungsten Compounds/therapeutic use , Uterus/drug effects , Uterus/metabolism
8.
Biol Reprod ; 71(5): 1437-45, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15215203

ABSTRACT

In vitro capacitation of dog spermatozoa in a medium without sugars and with lactate as the metabolic substrate (l-CCM) was accompanied by a progressive increase of intracellular glycogen during the first 2 h of incubation, which was followed by a subsequent decrease of glycogen levels after up to 4 h of incubation. Lactate from the medium is the source for the observed glycogen synthesis, as the presence of [(14)C]glycogen after the addition to l-CCM with [(14)C]lactate was demonstrated. The existence of functional gluconeogenesis in dog sperm was also sustained by the presence of key enzymes of this metabolic pathway, such as fructose 1,6-bisphophatase and aldolase B. On the other hand, glycogen metabolism from gluconeogenic sources was important in the maintenance of a correct in vitro fertilization after incubation in the l-CCM. This was demonstrated after the addition of phenylacetic acid (PAA) to l-CCM. In the presence of PAA, in vitro capacitation of dog spermatozoa suffered alterations, which translated into changes in capacitation functional markers, like the increase in the percentage of altered acrosomes, a distinct motion pattern, decrease or even disappearance of capacitation-induced tyrosine phosphorylation, and increased heterogeneity of the chlorotetracycline pattern in capacitated cells. Thus, this is the first report indicating the existence of a functional glyconeogenesis in mammalian spermatozoa. Moreover, gluconeogenesis-linked glycogen metabolism seems to be of importance in the maintenance of a correct in vitro capacitation in dog sperm in the absence of hexoses in the medium.


Subject(s)
Culture Media/chemistry , Dogs/physiology , Gluconeogenesis/physiology , Glycogen/metabolism , Sperm Capacitation/physiology , Spermatozoa/physiology , Animals , Cell Culture Techniques , Dogs/metabolism , Glucose , Lactic Acid/administration & dosage , Male , Phenylacetates/pharmacology , Sperm Capacitation/drug effects , Spermatozoa/enzymology , Spermatozoa/metabolism
9.
Diabetologia ; 47(3): 470-477, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14968293

ABSTRACT

AIMS/HYPOTHESIS: Sodium tungstate has recently emerged as an effective oral treatment for diabetes. We examined the effects of tungstate administration in the beta-cell mass of the pancreas as well as its therapeutic potential. METHODS: Sodium tungstate was administered via drinking water to healthy and neonatal streptozotocin (nSTZ)-diabetic rats for one month. The pancreas from each rat was removed and morphometric and immunocytochemical studies were carried out. The molecular mechanism of tungstate's action was also studied. RESULTS: In nSTZ rats administration of this compound normalised glycaemia, and increased insulinaemia and islet insulin content. Blood glucose concentrations were normalised as early as on day 4 of treatment, and tungstate treatment produced a partial recovery of beta-cell mass. The rats remained normoglycaemic after tungstate withdrawal. Morphometric studies showed that the increase in beta-cell mass was not due to beta-cell hypertrophy but to hyperplasia, with an increase in islet density in treated diabetic rats. Tungstate treatment increased extra-islet beta-cell replication without modifying intra-islet beta-cell replication rates. Moreover, the treatment induced increases in insulin-positive cells located close to ducts; and in PDX-1 positive cells scattered in the exocrine tissue, suggesting active neogenesis. In islets from treated diabetic rats, tungstate is able to increase the phosphorylation state of PDX-1 through the activation of p38. CONCLUSION/INTERPRETATION: These observations indicate that tungstate treatment is able to regenerate a stable, functional pancreatic beta-cell population which leads to and maintains normoglycaemia.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Islets of Langerhans/physiology , Regeneration/physiology , Tungsten Compounds/pharmacology , Animals , Cell Division/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/pathology , Organ Size/drug effects , Pancreas/anatomy & histology , Pancreas/drug effects , Rats , Rats, Wistar , Regeneration/drug effects
10.
Biochem Mol Biol Educ ; 32(6): 378-80, 2004 Nov.
Article in English | MEDLINE | ID: mdl-21706760

ABSTRACT

This article describes a course for reinforcing the knowledge of biochemistry in secondary school science teachers. The Department of Biochemistry and Molecular Biology of the University of Barcelona designed a course to bring these teachers up to date with this discipline. In addition to updating their knowledge of biochemistry and molecular biology, this course aims to provide teachers with a set of relevant laboratory practices that can be applied in their practical lessons.

11.
FEBS Lett ; 542(1-3): 84-8, 2003 May 08.
Article in English | MEDLINE | ID: mdl-12729903

ABSTRACT

Oral administration of sodium tungstate is an effective treatment for diabetes in animal models. We examined the effects of 6 weeks of oral administration of tungstate on glucose transporters (GLUT) in streptozotocin-induced diabetic rat diaphragm. Diabetes decreased GLUT4 expression while tungstate treatment normalized not only GLUT4 protein but also GLUT4 mRNA in the diabetic rats. Furthermore, treatment increased GLUT4 protein in plasma and internal membranes, suggesting a stimulation of its translocation to the plasma membrane. Tungstate had no effect on healthy animals. There were no differences in the total amount of GLUT1 transporter in any group. We conclude that the normoglycemic effect of tungstate may be partly due to a normalization of the levels and subcellular localization of GLUT4, which should result in an increase in muscle glucose uptake.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diaphragm/drug effects , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Tungsten Compounds/pharmacology , Animals , Blood Glucose/analysis , Cell Membrane/chemistry , Diabetes Mellitus, Experimental/diagnosis , Diaphragm/metabolism , Female , Glucose Transporter Type 1 , Glucose Transporter Type 4 , Monosaccharide Transport Proteins/analysis , Monosaccharide Transport Proteins/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Wistar
12.
Reproduction ; 123(4): 579-91, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11914120

ABSTRACT

Incubation of dog spermatozoa with 10 mmol l(-1) glucose or fructose rapidly increased the intracellular content of glucose 6-phosphate and fructose 6-phosphate, although the effect of fructose was greater. These effects were correlated with increases in ATP, ribose 5-phosphate and glycogen contents, and in the rates of formation of L-lactate and CO2. In all cases, except for ATP and glycogen, the effect of fructose was greater than that of glucose. The total hexokinase activity of the crude extracts of dog spermatozoa was more sensitive to fructose than to glucose at lower concentrations (0.1-3.0 mmol l(-1)). Both monosaccharides induced a fast and intense increase in the overall tyrosine phosphorylation of dog spermatozoa, although their specific induced-phosphorylation patterns differed slightly. Glut 3 and Glut 5 hexose transporters were the main hexose transporters in dog spermatozoa; however, other possible SGLT family-related hexose transporters were also localized. These data indicate that, at concentrations from 1 mmol l(-1) to 10 mmol l(-1), fructose has a stronger effect than glucose on hexose metabolism of dog spermatozoa. These differences appear to be related to variations in the sensitivity of hexokinase activity. Moreover, the differential hexose metabolism induced by the two sugars had distinct effects on the function of dog spermatozoa, as revealed by the diverse patterns of tyrosine phosphorylation.


Subject(s)
Fructose/pharmacology , Glucose/pharmacology , Hexoses/metabolism , Nerve Tissue Proteins , Spermatozoa/metabolism , Adenosine Triphosphate/metabolism , Animals , Carbon Dioxide/metabolism , Cells, Cultured , Dogs , Fructosephosphates/metabolism , Glucose Transporter Type 3 , Glucose Transporter Type 5 , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Lactic Acid/metabolism , Male , Monosaccharide Transport Proteins/metabolism , Phosphorylation , Ribosemonophosphates/metabolism , Spermatozoa/drug effects , Tyrosine/metabolism
13.
Biochem J ; 357(Pt 1): 17-24, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11415431

ABSTRACT

Changes in the intracellular distribution of liver glycogen synthase (GS) might constitute a new regulatory mechanism for the activity of this enzyme at cellular level. Our previous studies indicated that incubation of isolated hepatocytes with glucose activated GS and resulted in its translocation from a homogeneous cytosolic distribution to the cell periphery. These studies also suggested a relationship with insoluble elements of the cytoskeleton, in particular actin. Here we show the translocation of GS in a different experimental model that allows the analysis of this phenomenon in long-term studies. We describe the reversibility of translocation of GS and its effect on glycogen distribution. Incubation of cultured rat hepatocytes with glucose activated GS and triggered its translocation to the hepatocyte periphery. The relative amount of the enzyme concentrated near the plasma membrane increased with time up to 8 h of incubation with glucose, when the glycogen stores reached their maximal value. The lithium-induced covalent activation of GS was not sufficient to cause its translocation to the cell periphery. The intracellular distribution of GS closely resembled that of glycogen. Our results showed an interaction between GS and an insoluble element of the hepatocyte matrix. Although no co-localization between actin filaments and GS was observed in any condition, disruption of actin cytoskeleton resulted in a significantly lower percentage of cells in which the enzyme translocated to the cell periphery in response to glucose. This observation suggests that the microfilament network has a role in the translocation of GS.


Subject(s)
Glycogen Synthase/metabolism , Hepatocytes/metabolism , Liver Glycogen/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Cytosol/metabolism , Glucose/metabolism , Glucose/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , Kinetics , Male , Protein Transport , Rats , Rats, Wistar , Subcellular Fractions/metabolism
14.
Diabetologia ; 44(4): 507-13, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11357483

ABSTRACT

AIMS/HYPOTHESIS: Recent studies have shown the anti diabetic effects of oral sodium tungstate treatment in several animal models of diabetes based on short-term experiments. In this study, we examined the effectiveness of long-term tungstate treatment of streptozotocin-induced-diabetic rats. METHODS: Tungstate was administered to the drinking water of rats for eight months. RESULTS: The treatment resulted in a reduction in serum glucose concentrations in diabetic rats, but no change in glycaemia was detected in healthy rats. Alterations in the hepatic glucose metabolism due to diabetes were almost completely counteracted by tungstate treatment. The partial recovery of glucokinase activity, not found in diabetic animals, normalised glycogen and glucose 6-phosphate concentrations. Tungstate treatment also restored pyruvate kinase activity and fructose 2,6-bisphosphate concentrations. In healthy rats, tungstate treatment did not modify the majority of the hepatic parameters studied. Moreover, tungstate treatment prevented diabetes-induced morphological changes in the kidney and ocular lens and also reduced mortality. Furthermore, no hypoglycaemic episodes or undesirable side effects were observed in treated diabetic or healthy rats. In addition, there is no evidence of intolerance developing after prolonged use. CONCLUSION/INTERPRETATION: Tungstate could play a helpful part in the long-term treatment of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Tungsten Compounds/therapeutic use , Aging , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Blood Glucose/metabolism , Cornea/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Fructosediphosphates/analysis , Glucokinase/analysis , Glucose/metabolism , Glucose-6-Phosphate/analysis , Kidney/pathology , Liver/chemistry , Liver/metabolism , Liver/pathology , Male , Pyruvate Kinase/analysis , Rats , Rats, Wistar , Tungsten Compounds/administration & dosage , Tungsten Compounds/adverse effects
15.
J Biol Chem ; 276(26): 23858-66, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11309391

ABSTRACT

We used metabolic control analysis to determine the flux control coefficient of phosphorylase on glycogen synthesis in hepatocytes by titration with a specific phosphorylase inhibitor (CP-91149) or by expression of muscle phosphorylase using recombinant adenovirus. The muscle isoform was used because it is catalytically active in the b-state. CP-91149 inactivated phosphorylase with sequential activation of glycogen synthase. It increased glycogen synthesis by 7-fold at 5 mm glucose and by 2-fold at 20 mm glucose with a decrease in the concentration of glucose causing half-maximal rate (S(0.5)) from 26 to 19 mm. Muscle phosphorylase was expressed in hepatocytes mainly in the b-state. Low levels of phosphorylase expression inhibited glycogen synthesis by 50%, with little further inhibition at higher enzyme expression, and caused inactivation of glycogen synthase that was reversed by CP-91149. At endogenous activity, phosphorylase has a very high (greater than unity) negative control coefficient on glycogen synthesis, regardless of whether it is determined by enzyme inactivation or overexpression. This high control is attenuated by glucokinase overexpression, indicating dependence on other enzymes with high control. The high control coefficient of phosphorylase on glycogen synthesis affirms that phosphorylase is a strong candidate target for controlling hyperglycemia in type 2 diabetes in both the absorptive and postabsorptive states.


Subject(s)
Hepatocytes/metabolism , Liver Glycogen/biosynthesis , Phosphorylases/metabolism , Adenosine Monophosphate/pharmacology , Adenoviridae/genetics , Amides/pharmacology , Animals , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glucagon/pharmacology , Glucokinase/genetics , Glucokinase/metabolism , Glucose/pharmacology , Glycogen Synthase/genetics , Glycogen Synthase/metabolism , Hepatocytes/drug effects , Hepatocytes/enzymology , Indoles/pharmacology , Male , Phosphorylase b/antagonists & inhibitors , Phosphorylase b/metabolism , Phosphorylases/antagonists & inhibitors , Rats , Rats, Wistar , Transfection
16.
Biochem J ; 355(Pt 3): 787-93, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11311143

ABSTRACT

It is widely assumed that the control coefficient of an enzyme on pathway flux decreases as the concentration of enzyme increases. However, it has been shown [Kholodenko and Brown (1996) Biochem. J. 314, 753-760] that enzymes with sigmoidal kinetics can maintain or even gain control with an increase in enzyme activity or concentration. This has been described as 'paradoxical control'. Here we formulate the general requirements for allosteric enzyme kinetics to display this behaviour. We show that a necessary condition is that the Hill coefficient of the enzyme should increase with an increase in substrate concentration or decrease with an increase in product concentration. We also describe the necessary and sufficient requirements for the occurrence of paradoxical control in terms of the flux control coefficients and the derivatives of the elasticities. The derived expression shows that the higher the control coefficient of an allosteric enzyme, the more likely it is that the pathway will display this behaviour. Control of pathway flux is generally shared between a large number of enzymes and therefore the likelihood of observing sustained or increased control is low, even if the kinetic parameters are in the most favourable range to generate the phenomenon. We show that hepatic glucokinase, which has a very high flux control coefficient and displays sigmoidal behaviour within the hepatocyte in situ as a result of interaction with a regulatory protein, displays sustained or increased control over an extended range of enzyme concentrations when the regulatory protein is overexpressed.


Subject(s)
Glucokinase/chemistry , Allosteric Regulation , Animals , Hepatocytes/enzymology , Kinetics , Models, Chemical , Rats
17.
Diabetes ; 50(1): 131-8, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11147778

ABSTRACT

Tungstate was orally administered to 7.5-week-old male Zucker diabetic fatty (ZDF) rats that already showed moderate hyperglycemia (180 +/- 16 mg/dl). The animals became normoglycemic for approximately 10 days. Then, glycemia started to rise again, although it did not reach the initial values until day 24, when levels stabilized at approximately 200 mg/dl for the duration of the experiment. Untreated ZDF rats showed steadily increased blood glucose levels between 7.5 and 10 weeks of age, when they reached a maximum value of 450 +/- 19 mg/dl, which was maintained throughout the experiment. In addition, tolerance to intraperitoneal glucose load improved in treated diabetic rats. Serum levels of triglycerides were elevated in untreated diabetic rats compared with their lean counterparts (ZLC). In the liver of diabetic animals, glucokinase (GK), glycogen phosphorylase a (GPa), liver-pyruvate kinase (L-PK), and fatty acid synthase (FAS) activities decreased by 81, 30, 54, and 35%, respectively, whereas phosphoenolpyruvate carboxykinase (PEPCK) levels increased by 240%. Intracellular glucose-6-phosphate (G6P) decreased by 40%, whereas glycogen levels remained unaffected. Tungstate treatment of these rats induced a 42% decrease in serum levels of triglycerides and normalized hepatic G6P concentrations, GPa activity, and PEPCK levels. GK activity in treated diabetic rats increased to 50% of the values of untreated ZLC rats. L-PK and FAS activity increased to higher values than those in untreated lean rats (1.7-fold L-PK and 2.4-fold FAS). Hepatic glycogen levels were 55% higher than those in untreated diabetic and healthy rats. Tungstate treatment did not significantly change the phosphotyrosine protein profile of primary cultured hepatocytes from diabetic animals. These data suggest that tungstate administration to ZDF rats causes a considerable reduction of glycemia, mainly through a partial restoration of hepatic glucose metabolism and a decrease in lipotoxicity.


Subject(s)
Diabetes Mellitus/physiopathology , Hypoglycemic Agents/pharmacology , Obesity , Tungsten Compounds/pharmacology , Administration, Oral , Animals , Diabetes Mellitus/blood , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Hyperglycemia/chemically induced , Islets of Langerhans/physiopathology , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Phosphorylation/drug effects , Rats , Rats, Zucker , Tyrosine/metabolism
18.
Biochem J ; 351 Pt 3: 811-6, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11042138

ABSTRACT

We have used recombinant adenoviruses (AdCMV-RLGS and AdCMV-GK) to overexpress the liver isoforms of glycogen synthase (GS) and glucokinase (GK) in primary cultured rat hepatocytes. Glucose activated overexpressed GS in a dose-dependent manner and caused the accumulation of larger amounts of glycogen in the AdCMV-RLGS-treated hepatocytes. The concentration of intermediate metabolites of the glycogenic pathway, such as glucose 6-phosphate (Glc-6-P) and UDP-glucose, were not significantly altered. GK overexpression also conferred on the hepatocyte an enhanced capacity to synthesize glycogen in response to glucose, as described previously [Seoane, Gómez-Foix, O'Doherty, Gómez-Ara, Newgard and Guinovart (1996) J. Biol. Chem. 271, 23756-23760], although, in this case, they accumulated Glc-6-P. When GS and GK were simultaneously overexpressed, the accumulation of glycogen was enhanced in comparison with cells overexpressing either GS or GK. Our results are consistent with the hypothesis that liver GS catalyses the rate-limiting step of hepatic glycogen synthesis. However, hepatic glycogen deposition from glucose is submitted to a system of shared control in which the 'controller', GS, is, in turn, controlled by GK. This control is indirectly exerted through Glc-6-P, which 'switches on' GS dephosphorylation and activation.


Subject(s)
Glucokinase/metabolism , Glycogen Synthase/metabolism , Liver Glycogen/biosynthesis , Adenoviridae/genetics , Adenoviridae/physiology , Animals , Gene Expression Regulation, Viral/physiology , Hepatocytes/enzymology , Hepatocytes/metabolism , Lactic Acid/biosynthesis , Male , Rats , Rats, Wistar
19.
J Biol Chem ; 275(43): 33614-21, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-10924520

ABSTRACT

The detailed catalytic mechanism by which glycosyltransferases catalyze the transfer of a glycosyl residue from a donor sugar to an acceptor is not known. Through the multiple alignment of all known eukaryotic glycogen synthases we have found an invariant 17-amino acid stretch enclosed within the most conserved region of the members of this family. This peptide includes an E-X(7)-E motif, which is highly conserved in four families of retaining glycosyltransferases. Site-directed mutagenesis was performed in human muscle glycogen synthase to analyze the roles of the two conserved Glu residues (Glu-510 and Glu-518) of the motif. Proteins were transiently expressed in COS-1 cells as fusions to green fluorescence protein. The E510A and E518A mutant proteins retained the ability to translocate from the nucleus to the cytosol in response to glucose and to bind to intracellular glycogen. Although the E518A variant had approximately 6% of the catalytic activity shown by the green fluorescence protein-human muscle glycogen synthase fusion protein, the E510A mutation inactivated the enzyme. These results led us to conclude that the E-X(7)-E motif is part of the active site of eukaryotic glycogen synthases and that both conserved Glu residues are involved in catalysis. We propose that Glu-510 may function as the nucleophile and Glu-518 as the general acid/base catalyst.


Subject(s)
Glutamic Acid/physiology , Glycogen Synthase/chemistry , Amino Acid Motifs , Amino Acid Sequence , Catalytic Domain , Glycogen/metabolism , Glycogen Synthase/metabolism , Humans , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry
20.
Mol Reprod Dev ; 56(2): 207-19, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10813853

ABSTRACT

The glycogen content in fresh raw dog spermatozoa was 0.22+/-0.03 micromol/mg protein. This matched with the presence of a glycogen-like staining in the head and midpiece. Glycogen levels lowered to 0.05 micromol/mg protein after incubation for 60 min without sugars. Addition of either 10 mM fructose or 10 mM glucose increased glycogen content to 0.70 micromol/mg protein. On the other hand, glycogen synthase activity ratio of fresh dog sperm (0.35+/-0.07, measured in the absence and the presence of glucose 6-P) increased to 0.55 with 10 mM fructose for 20 min, whereas glucose had a smaller effect. Spermatozoa extracts had also a protein of about 100 Kd, which reacted against a rat liver glycogen synthase antibody. This was located in sperm head and midpiece. Furthermore, glycogen phosphorylase activity ratio measured in presence and absence of AMP (0.25+/-0.03 in fresh samples) decreased to 0.15 by 10 mM glucose for 20 min, whereas fructose was less potent in this regard. The maximal effect of glucose and fructose were observed from 10-20 mM onwards. This work is the first indication for a functional glycogen metabolism in mammal spermatozoa, which could play an important role in regulating sperm survival in vivo.


Subject(s)
Glycogen/metabolism , Spermatozoa/metabolism , Animals , Dogs , Fructose/metabolism , Glucose/metabolism , Glucose-6-Phosphate/metabolism , Glycogen Synthase/metabolism , Horses , Male , Phosphorylases/metabolism , Rats , Sheep , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...