Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 183
Filter
1.
Biochim Biophys Acta ; 1824(5): 769-84, 2012 May.
Article in English | MEDLINE | ID: mdl-22446163

ABSTRACT

We investigated how glycerol, urea, glucose and a GKA influence kinetics and stability of wild-type and mutant GK. Glycerol and glucose stabilized GK additively. Glycerol barely affected the TF spectra of all GKs but decreased k(cat), glucose S(0.5) and K(D) values and ATP K(M) while leaving cooperativity unchanged. Glycerol sensitized all GKs to GKA as shown by TF. Glucose increased TF of GKs without influence of glycerol on the effect. Glycerol and GKA affected kinetics and binding additively. The activation energies for thermal denaturation of GK were a function of glucose with K(D)s of 3 and 1mM without and with glycerol, respectively. High urea denatured wild type GK reversibly at 20 and 60°C and urea treatment of irreversibly heat denatured GK allowed refolding as demonstrated by TF including glucose response. We concluded: Glycerol stabilizes GK indirectly without changing the folding structure of the apoenzyme, by restructuring the surface water of the protein, whereas glucose stabilizes GK directly by binding to its substrate site and inducing a compact conformation. Glucose or glycerol (alone or combined) is unable to prevent irreversible heat denaturation above 40°C. However, urea denatures GK reversibly even at 60°C by binding to the protein backbone and directly interacting with hydrophobic side chains. It prevents irreversible aggregation allowing complete refolding when urea is removed. This study establishes the foundation for exploring numerous instability mutants among the more than 600 variant GKs causing diabetes in animals and humans.


Subject(s)
Apoenzymes/chemistry , Enzyme Activators/chemistry , Glucokinase/chemistry , Glucose/chemistry , Glycerol/chemistry , Urea/chemistry , Adenosine Triphosphate/chemistry , Allosteric Regulation , Apoenzymes/genetics , Enzyme Stability , Escherichia coli/genetics , Glucokinase/genetics , Humans , Kinetics , Models, Molecular , Mutation , Osmotic Pressure , Protein Conformation , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Temperature , Thermodynamics , Water/chemistry
2.
Biochem Soc Trans ; 33(Pt 1): 306-10, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15667334

ABSTRACT

The enzyme GK (glucokinase), which phosphorylates glucose to form glucose 6-phosphate, serves as the glucose sensor of insulin-producing beta-cells. GK has thermodynamic, kinetic, regulatory and molecular genetic characteristics that are ideal for its glucose sensor function and allow it to control glycolytic flux of the beta-cells as indicated by control-, elasticity- and response-coefficients close to or larger than 1.0. GK operates in tandem with the K(+) and Ca(2+) channels of the beta-cell membrane, resulting in a threshold for glucose-stimulated insulin release of approx. 5 mM, which is the set point of glucose homoeostasis for most laboratory animals and humans. Point mutations of GK cause 'glucokinase disease' in humans, which includes hypo- and hyper-glycaemia syndromes resulting from activating or inactivating mutations respectively. GK is allosterically activated by pharmacological agents (called GK activators), which lower blood glucose in normal animals and animal models of T2DM. On the basis of crystallographic studies that identified a ligand-free 'super-open' and a liganded closed structure of GK, on thermostability studies using glucose or mannoheptulose as ligands and studies showing that mannoheptulose alone or combined with GK activators induces expression of GK in pancreatic islets and partially preserves insulin secretory competency, a new hypothesis was developed that GK may function as a metabolic switch per se without involvement of enhanced glucose metabolism. Current research has the goal to find molecular targets of this putative 'GK-switch'. The case of GK research illustrates how basic science may culminate in therapeutic advances of human medicine.


Subject(s)
Glucokinase/metabolism , Glucose/metabolism , Homeostasis , Crystallography, X-Ray , Glucokinase/antagonists & inhibitors , Glucokinase/chemistry , Glucokinase/genetics , Humans , Hypoglycemic Agents/pharmacology , Kinetics , Point Mutation , Protein Conformation
3.
Biotechnol Bioeng ; 87(7): 835-48, 2004 Sep 30.
Article in English | MEDLINE | ID: mdl-15334410

ABSTRACT

A method was developed for obtaining high signal-to-noise 13C NMR spectra of intracellular compounds in metabolically active cultured cells. The method allows TCA cycle labeling kinetics to be determined in real time without significant oxygen transport limitations. Cells were immobilized on the surface of nonporous microcarriers that were either uncoated or coated with polypeptides and used in a 12-cm3 packed bed. The methods were tested with two EMT6 mouse mammary tumor cell lines, one strongly adherent and the other moderately adherent, and a weakly adherent mouse insulinoma line (betaHC9). For both EMT6 lines, NTP and oxygen consumption measurements indicated that the number of cells in the spectrometer ranged from 6 x 10(8) to 1 x 10(9). During infusion of [1-13C]glucose, labeling in C-4 glutamate (indicative of flux into the first half of the TCA cycle) could be detected with 15-min resolution. However, labeling for C-3 and C-2 glutamate (indicative of complete TCA cycle activity) was fivefold lower and difficult to quantify. To increase TCA cycle labeling, cells were infused with medium containing [1,6-13C2]glucose. A 2.5-fold increase was observed in C-4 glutamate labeling and C-3 and C-2 glutamate labeling could be monitored with 30-min resolution. Citrate synthase activity was indirectly detected in real time, as [3,4-13C2]glutamate was formed from [2-13C]oxaloacetate and [2-13C]acetate (of acetyl-CoA). Cell mass levels observed with betaHC9 cells were somewhat lower. However, the 13C S/N was sufficient to allow real-time monitoring of the response of intracellular metabolite labeling to a step change in glucose and a combined glutamine/serum pulse.


Subject(s)
Cell Culture Techniques/methods , Glucose/metabolism , Glutamic Acid/metabolism , Insulinoma/metabolism , Magnetic Resonance Spectroscopy/methods , Mammary Neoplasms, Experimental/metabolism , Online Systems , Animals , Carbon Isotopes , Cell Culture Techniques/instrumentation , Cell Line, Tumor , Cells, Immobilized/metabolism , Isotope Labeling/methods , Kinetics , Magnetic Resonance Spectroscopy/instrumentation , Metabolic Clearance Rate , Mice , Reproducibility of Results , Sensitivity and Specificity
4.
Diabetologia ; 46(3): 386-93, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12687337

ABSTRACT

AIMS/HYPOTHESIS: Recent success in islet transplantation renews the hope for the complete cure of patients afflicted with Type 1 diabetes. However, in the Edmonton series, two to four pancreas donors were required to obtain a sufficient islet mass to reverse the diabetes of each patient. In view of the donor shortage, this represents a major obstacle preventing greater application of islet transplantation to diabetic patients. We hypothesised that increasing the expression of the insulin gene in transplanted islets would augment their capacity for insulin production, thereby allowing reversal of diabetes with a reduced islet mass. METHODS: We used a replication defective adenovirus to deliver the human proinsulin gene (Ad-Ins) to isolated human islets. The function of Ad-Ins-transduced human islets was compared to islets transduced with a control vector (Ad-lacz). RESULTS: Ad-Ins-transduced islets produced two to three times more insulin than normal islets or those infected with Ad-lacz, as assessed by in vitro perifusion tests of glucose stimulated insulin release. When transplanted, Ad-Ins-transduced islets normalised the blood glucose of diabetic immunodeficient NOD-Scid mice, and less than half as many Ad-Ins islets were required for reversal of diabetes than when normal islets were transplanted. CONCLUSION/INTERPRETATION: Our results suggest a simple and effective approach that could enhance the efficiency of islet transplantation for treatment of diabetes in humans.


Subject(s)
Insulin/genetics , Islets of Langerhans Transplantation/physiology , Islets of Langerhans/physiology , Adenoviridae/genetics , Animals , Blood Glucose/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Gene Transfer Techniques , Glucagon/metabolism , Humans , Insulin/biosynthesis , Islets of Langerhans/ultrastructure , Lac Operon/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Electron , Proinsulin/genetics , Promoter Regions, Genetic/genetics , Subcellular Fractions/metabolism , Transgenes/genetics , Transplantation, Heterologous/physiology
5.
Diabetologia ; 44(7): 898-905, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11508276

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to assess the prevalence of glucokinase gene mutations in Italian children with MODY and to investigate genotype/phenotype correlations of the mutants. METHODS: Screening for sequence variants in the glucokinase gene was performed by denaturing gradient gel electrophoresis and direct sequencing in 132 children with maturity onset diabetes of the young (MODY) and in 9 children with chronic fasting hyperglycaemia but without laboratory evidence for Type I (insulin-dependent) diabetes mellitus and with normoglycaemic parents ("non-classical" MODY). RESULTS: Altogether 54 mutations were identified in the MODY group (54/132 or 41%) and 3 among the "non-classical" MODY individuals (3/9 or 33%). Paternity testing indicated that the latter mutations have arisen de novo. Mean fasting plasma glucose concentrations of the children with the mutant glucokinase was in the expected impaired fasting glucose range. In contrast, results of the oral glucose tolerance test showed a wide range from normal glucose tolerance (Group 1: 2-h OGTT = 6.7 +/- 1.1 mmol/l; 11 patients) to diabetes (Group 2: 2-h OGTT = 11.5 +/- 0.5 mmol/l; 9 patients), with the remaining in the impaired glucose tolerance range. Disruptive mutations (i.e. nonsense, frameshifts, splice-site) were equally represented in Groups 1 and 2 and were not clearly associated with an impaired first-phase insulin response. Surprisingly, 5 out of 11 children (or 45%) in Group 1 were found to be overweight but no children in Group 2 were overweight. Sensitivity index (SI), calculated by a recently described method, was found to be significantly lower in Group 2 than in Group 1 (SI Group 2 = 0.0013 +/- 0.0009 ml Kg(-1) min(-1)/muU/ml; SI Group 1 = 0.0068 +/- 0.0048, p < 0.0035). CONCLUSION/INTERPRETATION: Mutations in glucokinase are the first cause of MODY among Italian children selected through a low threshold limit of fasting plasma glucose (i. e. > 5.5 mmol). The lack of correlation between the molecular severity of glucokinase mutations, insulin secretion at intravenous glucose tolerance test and differences in glucose tolerance suggests that factors outside the beta cell are also involved in determining post-load glucose concentrations in these subjects. Our results seem to indicate that the differences observed in the 2-h responses at the OGTT among children with MODY 2 could be related to individual differences in insulin sensitivity.


Subject(s)
Blood Glucose/metabolism , Body Mass Index , Diabetes Mellitus, Type 2/genetics , Glucokinase/genetics , Insulin/metabolism , Mutation , Amino Acid Substitution , Child , Conserved Sequence , Diabetes Mellitus/enzymology , Diabetes Mellitus/physiopathology , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/physiopathology , Fasting , Female , Glucose Tolerance Test , Humans , Hyperglycemia/blood , Insulin/blood , Insulin Resistance/genetics , Insulin Secretion , Italy , Male , Mutation, Missense , Obesity , Pedigree , Sensitivity and Specificity
6.
Genes Dev ; 15(13): 1706-15, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11445544

ABSTRACT

We have used conditional gene ablation to uncover a dramatic and unpredicted role for the winged-helix transcription factor Foxa2 (formerly HNF-3 beta) in pancreatic beta-cell differentiation and metabolism. Mice that lack Foxa2 specifically in beta cells (Foxa2(loxP/loxP); Ins.Cre mice) are severely hypoglycemic and show dysregulated insulin secretion in response to both glucose and amino acids. This inappropriate hypersecretion of insulin in the face of profound hypoglycemia mimics pathophysiological and molecular aspects of familial hyperinsulinism. We have identified the two subunits of the beta-cell ATP-sensitive K(+) channel (K(ATP)), the most frequently mutated genes linked to familial hyperinsulinism, as novel Foxa2 targets in islets. The Foxa2(loxP/loxP); Ins.Cre mice will serve as a unique model to investigate the regulation of insulin secretion by the beta cell and suggest the human FOXA2 as a candidate gene for familial hyperinsulinism.


Subject(s)
DNA-Binding Proteins/physiology , Hyperinsulinism/etiology , Hypoglycemia/etiology , Islets of Langerhans/metabolism , Nuclear Proteins/physiology , Transcription Factors , Adenosine Triphosphate/metabolism , Animals , Cell Lineage , DNA-Binding Proteins/genetics , Hepatocyte Nuclear Factor 3-beta , Humans , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Mice , Mice, Knockout , Nuclear Proteins/genetics , Potassium Channels/metabolism
7.
Life Sci ; 68(25): 2813-24, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11432447

ABSTRACT

It has been shown that all-trans retinoic acid induces prematurely hepatic glucokinase mRNA in ten days-old neonatal rat hepatocytes, however, this effect could be related to the capacity of the retinoid to promote a more differentiated state of the hepatocyte. In this report we demonstrate that physiological concentrations of all-trans retinoic acid stimulate glucokinase activity in both mature fully differentiated hepatocytes and at the onset of the induction of the enzyme in 15 to 17 days-old neonatal hepatocytes. The effects produced by the retinoid were similar both in magnitude and in time, to those elicited by insulin, a well-known stimulator of hepatic glucokinase expression. No additive effect was observed when insulin and retinoic acid were tested together. Using the branched DNA assay, a sensitive signal amplification technique, we detected relative increases in glucokinase mRNA levels of about 70% at 3 and 24 h after the treatment with 10(-6) M all-trans retinoic acid, in both neonatal and adult hepatocytes. These data show that retinoic acid exerts a stimulatory effect on hepatic glucokinase independent of the hepatocyte stage of maturity and suggest a physiological role of retinoic acid on glucose metabolism. The action of retinoic acid on hepatic glucokinase might explain previous observations on the relationship between vitamin A status and liver glycogen synthesis. These findings may serve as basis for further investigations on the biological functions of retinoic acid derivatives on hepatic glucose metabolism.


Subject(s)
Gene Expression/drug effects , Glucokinase/biosynthesis , Hepatocytes/drug effects , Hepatocytes/enzymology , Tretinoin/pharmacology , Animals , Animals, Newborn , Animals, Suckling , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Induction/drug effects , Glucokinase/genetics , Insulin/pharmacology , Liver/drug effects , Liver/enzymology , RNA, Messenger/metabolism , Rats , Rats, Wistar
9.
Endocrinology ; 142(4): 1448-52, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11250924

ABSTRACT

Comparison of the pancreatic and hepatic glucokinase gene transcripts reveals tissue-specific control of expression and the existence of two distinct promoters in a single glucokinase gene. The existence of alternate promoters suggests that separate factors regulate glucokinase transcription in the two tissues. Hepatic glucokinase expression has been shown to be repressed by cAMP; however, in the pancreatic beta-cell it is unlikely that cAMP represses glucokinase activity, as cAMP is known to positively affect glucose-induced insulin secretion, a process that in mature islets requires pancreatic glucokinase activity. In this work we demonstrate that cAMP indeed has a stimulatory effect on pancreatic glucokinase. The cyclic nucleotide stimulates pancreatic glucokinase activity after 3-h incubation, and maximal effects are observed after 6 and 12 h of treatment. Using the bDNA assay, a sensitive signal amplification technique, we detected relative increases in glucokinase messenger RNA levels of 40.5 +/- 7.5% after 3-h incubation with cAMP. This stimulatory effect was increased to 106.3 +/- 22% after 6-h incubation and sustained up to 12 h of incubation. Inhibition of gene transcription by actinomycin D abolishes cAMP-induced glucokinase activity. In transfected fetal islets, cAMP increased the activity of the -1000 bp rat glucokinase promoter by 60 +/- 6%. These data demonstrate that cAMP has a stimulatory effect on pancreatic glucokinase gene expression and that the nucleotide has opposite effects on pancreatic and hepatic glucokinase, supporting the concept that glucokinase transcription in the liver and that in the beta-cell differ.


Subject(s)
Cyclic AMP/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glucokinase/genetics , Glucokinase/metabolism , Pancreas/enzymology , Animals , Dactinomycin/pharmacology , Female , Fetus/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Plasmids/genetics , Pregnancy , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Transfection
10.
11.
J Biol Chem ; 275(11): 7826-31, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10713097

ABSTRACT

The glucokinase regulatory protein (GKRP) inhibits glucokinase competitively with respect to glucose by forming a protein-protein complex with this enzyme. The physiological role of GKRP in controlling hepatic glucokinase activity was addressed using gene targeting to disrupt GKRP gene expression. Heterozygote and homozygote knockout mice have a substantial decrease in hepatic glucokinase expression and enzymatic activity as measured at saturating glucose concentrations when compared with wild-type mice, with no change in basal blood glucose levels. Interestingly, when assayed under conditions to promote the association between glucokinase and GKRP, liver glucokinase activity in wild-type and null mice displayed comparable glucose phosphorylation capacities at physiological glucose concentrations (5 mM). Thus, despite reduced hepatic glucokinase expression levels in the null mice, glucokinase activity in the liver homogenates was maintained at nearly normal levels due to the absence of the inhibitory effects of GKRP. However, following a glucose tolerance test, the homozygote knockout mice show impaired glucose clearance, indicating that they cannot recruit sufficient glucokinase due to the absence of a nuclear reserve. These data suggest both a regulatory and a stabilizing role for GKRP in maintaining adequate glucokinase in the liver. Furthermore, this study provides evidence for the important role GKRP plays in acutely regulating of hepatic glucose metabolism.


Subject(s)
Carrier Proteins , Glucokinase/antagonists & inhibitors , Glucose/metabolism , Liver/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Age Factors , Animals , Enzyme Inhibitors/metabolism , Gene Targeting , Glucose Tolerance Test , Heterozygote , Homeostasis , Homozygote , Insulin/blood , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Mutagenesis , Proteins/genetics
12.
Diabetologia ; 42(10): 1175-86, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10525657

ABSTRACT

AIMS/HYPOTHESIS: Mutations of the glucokinase gene cause hyperglycaemia or hypoglycaemia. A quantitative understanding of these defects of glucose homeostasis linked to the glucokinase gene was lacking. Therefore a database of kinetic variables of wild-type and 20 missense mutants of glucokinase was developed and used in mathematical modelling to predict the thresholds for glucose-stimulated insulin release. METHODS: Recombinant human glucokinase was generated in E. coli. The k(cat), glucose S(0.5), ATP K(m), and Hill number of glucokinase were determined. Inhibition by Stearoyl CoA and glucokinase regulatory protein and thermal stability were assayed for all mutants kinetically similar to wild-type glucokinase. A mathematical model predicting the threshold for glucose-stimulated insulin release was constructed. This model is based on the two substrate kinetics of glucokinase and the kinetic variables of the database. It is assumed that both glucokinase gene alleles are equally expressed in beta-cells and that induction of glucokinase occurs as a function of basal blood glucose. RESULTS: Large changes, varying greatly between mutants were found in nearly all variables. Glucokinase flux at threshold for glucose-stimulated insulin release was about 25 % of total phosphorylating potential in the normal beta-cell and this was used to predict thresholds for the mutant heterozygotes. Clinical data for maturity onset diabetes of the young type linked to the glucokinase gene and familial hyperinsulinaemic hypoglycaemia linked to the glucokinase gene and the glucokinase kinetic data of this study were used to test the model. The model predicts fasting blood glucose between 3 and 7 mmol/l in these cases. CONCLUSION/INTERPRETATION: A kinetics database of wild-type and 20 mutants of glucokinase was developed. Many kinetic differences were found for the mutants. The mathematical model to calculate the threshold for glucose-stimulated insulin release predicts fasting blood glucose between 3 and 7 mmol/l in subjects with glucokinase gene mutations. [Diabetologia 42: 1175-1186]


Subject(s)
Carrier Proteins , Glucokinase/genetics , Glucokinase/metabolism , Glucose/physiology , Hyperglycemia/genetics , Hypoglycemia/genetics , Adaptor Proteins, Signal Transducing , Adenosine Triphosphate/metabolism , Blood Glucose/metabolism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Stability , Glucokinase/antagonists & inhibitors , Glucose/pharmacology , Glutathione Transferase/genetics , Homeostasis , Humans , Hyperglycemia/blood , Hypoglycemia/blood , Insulin/blood , Models, Biological , Mutation , Proteins/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Syndrome
13.
Endocrinology ; 140(10): 4595-600, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10499515

ABSTRACT

Biotin has been reported to affect glucose homeostasis; however, its role on pancreatic islets of Langerhans has not been assessed. In this report, we demonstrate that physiologic concentrations of biotin stimulate glucokinase activity in rat islets in culture. Using the branched DNA (bDNA) assay, a sensitive signal amplification technique, we detected relative increases in glucokinase mRNA levels of 41.5 +/- 13% and 81.3 +/- 19% at 12 and 24 h respectively in islets treated with [10(-6) M] biotin. Because glucokinase activity controls insulin secretion, we also investigated the effect of biotin on insulin release. Treatment with [10(-6) M] biotin for 24 h increased insulin secretion. We extended our studies by analyzing the effect of biotin deficiency on pancreatic islet glucokinase expression and activity, as well as insulin secretion. Our results show that islet glucokinase activity and mRNA are reduced by 50% in the biotin deficient rat. Insulin secretion in response to glucose was also impaired in islets isolated from the deficient rat. These data show that biotin affects pancreatic islet glucokinase activity and expression and insulin secretion in cultured islets.


Subject(s)
Biotin/deficiency , Biotin/physiology , Glucokinase/metabolism , Insulin/metabolism , Pancreas/metabolism , Animals , Biotin/pharmacology , Cells, Cultured , Glucokinase/genetics , Insulin Secretion , Male , Pancreas/drug effects , Pancreas/enzymology , RNA, Messenger/metabolism , Rats , Rats, Wistar
14.
Diabetes ; 48(9): 1698-705, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10480597

ABSTRACT

Mutations in human glucokinase are implicated in the development of diabetes and hypoglycemia. Human glucokinase shares 54% identical amino acid residues with human brain hexokinase I. This similarity was used to model the structure of glucokinase by analogy to the crystal structure of brain hexokinase. Glucokinase was modeled with both its substrates, glucose and MgATP, to understand the effect of mutations. The glucose is predicted to form hydrogen bond interactions with the side chains of glucokinase residues Thr 168, Lys 169, Asn 204, Asp 205, Asn 231, and Glu 290, similar to those observed for brain hexokinase I. The magnesium ion is coordinated by the carboxylates of Asp 78 and Asp 205 and the gamma-phosphate of ATP. ATP is predicted to form hydrogen bond interactions with residues Gly 81, Thr 82, Asn 83, Arg 85, Lys 169, Thr 228, Lys 296, Thr 332, and Ser 336. Mutations of residues close to the predicted ATP binding site produced dramatic changes in the Km for ATP, the catalytic rate, and a loss of cooperativity, which confirmed our model. Mutations of residues in the glucose binding site dramatically reduced the catalytic activity, as did a mutation that was predicted to disrupt an alpha-helix. Other mutations located far from the active site gave smaller changes in kinetic parameters. In the absence of a crystal structure for glucokinase, our models help rationalize the potential effects of mutations in diabetes and hypoglycemia, and the models may also facilitate the discovery of pharmacological glucokinase activators and inhibitors.


Subject(s)
Adenosine Triphosphate/chemistry , Glucokinase/chemistry , Glucose/chemistry , Hyperglycemia/genetics , Hypoglycemia/genetics , Models, Molecular , Amino Acid Sequence , Binding Sites , Humans , Molecular Sequence Data , Mutation , Protein Conformation
15.
Biochem J ; 342 ( Pt 2): 345-52, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10455021

ABSTRACT

Mutations in the glucokinase (GK) gene cause type-2 maturity-onset diabetes of the young type 2 (MODY-2) and GK-linked hyperinsulinaemia (GK-HI). Recombinant adenoviruses expressing the human wild-type islet GK or one of four mutant forms of GK, (the MODY-2 mutants E70K, E300K and V203A and the GK-HI mutant V455M) were transduced into glucose-responsive insulin-secreting beta-HC9 cells and tested functionally in order to initiate the first analysis in vivo of recombinant wild-type and mutant human islet GK. Kinetic analysis of wild-type human GK showed that the glucose S(0. 5) and Hill coefficient were similar to previously published data in vitro (S(0.5) is the glucose level at the half-maximal rate). E70K had half the glucose affinity of wild-type, but similar enzyme activity. V203A demonstrated decreased catalytic activity and an 8-fold increase in glucose S(0.5) when compared with wild-type human islet GK. E300K had a glucose S(0.5) similar to wild-type but a 10-fold reduction in enzyme activity. E300K mRNA levels were comparable with wild-type GK mRNA levels, but Western-blot analyses demonstrated markedly reduced levels of immunologically detectable protein, consistent with an instability mutation. V455M was just as active as wild-type GK, but with a markedly reduced S(0.5). The effects of the different GK mutants on glucose-stimulated insulin release support the kinetic and expression data. These experiments show the utility of a combined genetic, biochemical and cell-biological approach to the quantification of functional and structural changes of human GK that result from MODY-2 and GK-HI mutations.


Subject(s)
Diabetes Mellitus/enzymology , Diabetes Mellitus/genetics , Glucokinase/genetics , Insulin/blood , Point Mutation , Adenoviridae/genetics , Animals , Cell Line , Gene Expression Regulation, Enzymologic , Glucokinase/metabolism , Glucose/pharmacology , Humans , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Kinetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
16.
Diabetes ; 48(8): 1535-42, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10426370

ABSTRACT

Leucine or the nonmetabolized leucine analog +/- 2-amino-2-norbornane-carboxylic acid (BCH) (both at 10 mmol/l) induced biphasic insulin secretion in the presence of 2 mmol/l glutamine (Q2) in cultured mouse islets pretreated for 40 min without glucose but with Q2 present. The beta-cell response consisted of an initial peak of 20- to 25-fold above basal and a less marked secondary phase. However, BCH produced only a delayed response, while leucine was totally ineffective when islets were pretreated with 25 mmol/l glucose plus Q2. With Q2, 10 mmol/l BCH or leucine caused a nearly threefold increase, a twofold increase, or had no effect on cytosolic Ca2+ levels in islets pretreated for 40 min with 0, 5, or 15 mmol/l glucose, respectively. Thus, pretreatment of islets with high glucose inhibited BCH- and leucine-induced cytosolic Ca2+ changes and insulin release. Glucose decreased glutamine oxidation in cultured rat islets when BCH was present at 10 mmol/l, but not in its absence, with a lowest effective level of approximately 0.1 mmol/l, a maximum of 18-30 mmol/l, and an inhibitory concentration, 50%, of approximately 3 mmol/l. The data are consistent with the hypothesis that glucose inhibits glutaminolysis in pancreatic beta-cells in a concentration-dependent manner and hence blocks leucine-stimulated insulin secretion. We postulate that in the basal interprandial state, glutaminolysis of beta-cells is partly turned on because glutamate dehydrogenase (GDH) is activated by a decreased P-potential due to partial fuel depletion and sensitization to endogenous activators such as leucine. Additionally, it may contribute significantly to basal insulin release, which is known to be responsible for about half of the insulin released daily. The data explain "leucine-hypersensitivity" of beta-cells during hypoglycemia and contribute to the elucidation of the GDH-linked syndrome of hyperinsulinism associated with elevated serum ammonia levels. Thus, understanding the precise regulation and role of beta-cell glutaminolysis is probably central to our concept of normal blood glucose control.


Subject(s)
Amino Acids, Cyclic , Glucose/pharmacology , Glutamine/metabolism , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Amino Acids/pharmacology , Animals , Calcium/metabolism , Culture Techniques , Cytosol/metabolism , Dose-Response Relationship, Drug , Glycolysis/physiology , Insulin Secretion , Leucine/pharmacology , Mice , Oxidation-Reduction , Rats
17.
Endocrinology ; 140(7): 3091-6, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385401

ABSTRACT

Retinoic acid has manifold effects on pancreatic beta-cells. Previously we reported that retinoic acid increases glucokinase activity and messenger RNA (mRNA) levels in the insulinoma cell line RIN-m5F; however, we could not rule out the possibility that the effect of retinoic acid on RIN-m5F glucokinase was inherent to the cell line or related to its differentiating capacity. In this report, we demonstrate that physiologic concentrations of retinoic acid stimulate glucokinase activity in both fetal islets and differentiated adult islets in culture. In the adult tissue, the response to the retinoid was less pronounced, achieving about half of the maximal effect produced on the fetal tissue. Using the branched DNA (bDNA) assay, a sensitive signal amplification technique, we detected relative increases in glucokinase mRNA levels of 51.8+/-13.3% and 62.8+/-16.1% at 12 and 24 h, respectively, in adult islets treated with] 10(-6) M retinoic acid. In fetal islets, increases of 55+/-14.9% and 107+/-30.5% at 12 and 24 h, respectively, were observed. In transfected fetal islets, retinoic acid increased the activity of the -1000 kb rat glucokinase promoter by 51.3%. Because glucokinase activity controls insulin secretion, we also investigated the effect of retinoic acid on insulin secretion. Treatment with 10(-6) M retinoic acid for 24 h increased insulin secretion in both fetal and adult islets; however, the increases on insulin secretion were more pronounced in the mature islets; in contrast, retinoic acid produced higher levels of insulin mRNA in the fetal islets. These data show that retinoic acid increases pancreatic glucokinase in cultured islets and that the mechanism may involve a stimulatory effect on the glucokinase promoter.


Subject(s)
Glucokinase/genetics , Glucokinase/metabolism , Insulin/metabolism , Islets of Langerhans/physiology , Tretinoin/pharmacology , Animals , Culture Techniques , Fetus/metabolism , Gene Expression/physiology , Insulin/genetics , Insulin Secretion , Islets of Langerhans/embryology , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Male , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , RNA, Messenger/metabolism , Rats/embryology , Rats, Wistar
18.
Eur J Neurosci ; 10(7): 2416-25, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9749769

ABSTRACT

We have studied Ca2+ homeostasis in a unique model of human neurons, the NT2N cell, which differentiates from a human teratocarcinoma cell line, NTera2/C1.D1 by retinoic acid treatment. When perifused with Krebs-HEPES buffer containing 2.5 mM CaCl2, fura-2 loaded NT2N cells produced spontaneous cytosolic Ca2+ oscillations, or Ca2+ transients. These cytosolic Ca2+ transients were not blocked by antagonists of glutamate (6-cyano-7-nitroquinoxaline-2,3-dione and D(-)-2-amino-5-phosphonopentanoic acid) or muscarinic (atropine) receptors. Omission of extracellular Ca2+ completely abolished Ca2+ oscillations and decreased the average Ca2+ level from 106 +/- 14 nM to 59 +/- 8 nM. Addition of the L-type Ca2+ channel blocker nifedipine (1 or 10 microM) or of the N-type inhibitor omega-conotoxin GVIA (5 microM) significantly, although incompletely, suppressed Ca2+ oscillations, while omega-conotoxin MVIIC (5 microM), a selective antagonist of P- and Q-channels, had no effect. Ni2+, at 100 microM, a concentration selective for T-type channels, did not inhibit Ca2+ transients. Non-specific blockage of Ca2+ channels by higher concentrations of Ni2+ (2-5 mM) or Co2+ (1 mM) abolished Ca2+ oscillations completely. The endoplasmic reticulum Ca2+-ATPase inhibitor, thapsigargin (1 microM), slightly decreased Ca2+ oscillation frequency, and induced a small transitory increase in the average cytosolic Ca2+ concentration. The mRNAs of L- (alpha1D subunit) and N-type (alpha1B subunit) Ca2+ channel were present in NT2N cells, while that of a T-type Ca2+ channel (alpha1-subunit) was not present in the NT2N cells as shown by reverse transcription-polymerase chain reaction. In conclusion, NT2N neuronal cells generate cytosolic Ca2+ oscillations mainly by influx of extracellular Ca2+ through multiple channels, which include L- and N-type channels, and do not require activation of glutamate or muscarinic receptors.


Subject(s)
Calcium/metabolism , Cytosol/metabolism , Neurons/cytology , Neurons/metabolism , Calcium Channels/genetics , Cell Differentiation/physiology , Cells, Cultured , Electrophysiology , Humans , Oscillometry , RNA, Messenger/metabolism
19.
Biochem Biophys Res Commun ; 247(2): 407-13, 1998 Jun 18.
Article in English | MEDLINE | ID: mdl-9642141

ABSTRACT

Ca2+ channel expression and regulation of intracellular Ca2+ homeostasis were studied during retinoic acid (RA)-induced differentiation of the human teratocarcinoma cell line Ntera 2/C1.D1 (NT2- cells) into NT2N neurons, a unique model of human neurons in culture. The cytosolic Ca2+ level of undifferentiated NT2- cells was low (75 +/- 5 nM) and stable under basal conditions, and it was only marginally decreased (by 9%) upon removal of extracellular Ca2+. After 10 microM RA treatment, NT2- cells were irreversibly differentiated into a phenotype of neuron-like NT2N cells. Cytosolic Ca2+ level of NT2N neurons was higher (106 +/- 14 nM) than that of NT2- cells and spontaneously fluctuated (0.208 +/- 0.038 transients/min) under basal conditions. Although K+ increased 86Rb fluxes in both NT2- cells and NT2N neurons, it only increased cytosolic Ca2+ level in NT2N neurons. The K+-induced increase in cytosolic Ca2+ in NT2N neurons was antagonized by 0.1-10 microM nifedipine or verapamil, 5 microM omega-CgTx GVIA, but not by 1 microM omega-agatoxin IVA, 1 microM omega-agatoxin TK, 1 microM FTX-3.3, or 100 microM Ni+ implicating L- and N-type voltage-dependent Ca2+ channels. In L- and N-type channels, but not in P- and Q-types, mRNAs were expressed in NT2N neurons as well as NT2- cells. Quantitative analysis of L- and N-type Ca2+ protein levels showed major differences between NT2- cells and NT2N neurons. In NT2- cells, N-type Ca2+ channels were undetectable while L-type channels levels were fivefold lower compared to NT2N neurons. Our findings show that L- and N-type channels are expressed during differentiation of NT2- cells into neurons, and that these voltage-dependent Ca2+ channels have a major role in regulating intracellular Ca2+ homeostasis and neuronal excitability.


Subject(s)
Calcium Channels/genetics , Tretinoin/pharmacology , Base Sequence , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/classification , Calcium Channels/metabolism , Cell Differentiation/drug effects , DNA Primers/genetics , Gene Expression/drug effects , Homeostasis , Humans , Intracellular Fluid/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Nifedipine/pharmacology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Verapamil/pharmacology
20.
J Neurochem ; 70(4): 1394-400, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9523555

ABSTRACT

The effects of synthetic beta-amyloid (A beta1-42) on cell viability and cellular Ca2+ homeostasis have been studied in the human neuron-like NT2N cell, which differentiates from a teratocarcinoma cell line, NTera2/C1.D1, by retinoic acid treatment. NT2N viability was measured using morphological criteria and fluorescent live/dead staining and quantified using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide metabolism. A beta1-42 dose-dependently caused NT2N cell death when it was present in the cell culture for 14 days but had no effect on viability when it was present for 4 days. The lowest effective concentration was 4 microM, and the strongest effect was produced by 40 microM. Control NT2N cells produced spontaneous cytosolic Ca2+ oscillations under basal conditions. These oscillations were inhibited dose-dependently (0.4-40 microM) by A beta1-42 that was present in the cell culture for 1 or 4 days. Ca2+ wave frequency was decreased from 0.21 +/- 0.02 to 0.05 +/- 0.02/min, amplitude from 88 +/- 8 to 13 +/- 4 nM, and average Ca2+ level from 130 +/- 8 to 58 +/- 3 nM. The Ca2+ responses to 30 mM K+ and 100 microM glutamate were not different between control and A beta-treated cells. Thus, the results do not support the hypothesis that cytosolic early Ca2+ accumulation mediates A beta-induced NT2N cell death.


Subject(s)
Amyloid beta-Peptides/pharmacology , Calcium/metabolism , Cytoplasm/metabolism , Neurons/drug effects , Peptide Fragments/pharmacology , Cell Differentiation/physiology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glutamic Acid/pharmacology , Homeostasis/drug effects , Humans , Neurons/cytology , Potassium/pharmacology , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...