Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Immunity ; 47(5): 875-889.e10, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29166588

ABSTRACT

Migration of activated regulatory T (Treg) cells to inflamed tissue is crucial for their immune-modulatory function. While metabolic reprogramming during Treg cell differentiation has been extensively studied, the bioenergetics of Treg cell trafficking remains undefined. We have investigated the metabolic demands of migrating Treg cells in vitro and in vivo. We show that glycolysis was instrumental for their migration and was initiated by pro-migratory stimuli via a PI3K-mTORC2-mediated pathway culminating in induction of the enzyme glucokinase (GCK). Subsequently, GCK promoted cytoskeletal rearrangements by associating with actin. Treg cells lacking this pathway were functionally suppressive but failed to migrate to skin allografts and inhibit rejection. Similarly, human carriers of a loss-of-function GCK regulatory protein gene-leading to increased GCK activity-had reduced numbers of circulating Treg cells. These cells displayed enhanced migratory activity but similar suppressive function, while conventional T cells were unaffected. Thus, GCK-dependent glycolysis regulates Treg cell migration.


Subject(s)
Glucokinase/physiology , Glycolysis , T-Lymphocytes, Regulatory/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , CD28 Antigens/physiology , CTLA-4 Antigen/physiology , Cells, Cultured , Humans , Mechanistic Target of Rapamycin Complex 1/physiology , Mechanistic Target of Rapamycin Complex 2/physiology , Mice , Mice, Inbred Strains , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology
2.
Nihon Rinsho ; 71(11): 2030-7, 2013 Nov.
Article in Japanese | MEDLINE | ID: mdl-24397179

ABSTRACT

Clinical development of new antidiabetic drugs such as SGLT2 inhibitor, GPR40 agonist, GPR119 agonist, and GKA has been progressing world wide. Action mechanism of each drug is unique and clearly distinguished from the existing drugs. The effect of SGLT2 inhibitors is independent of insulin action and characterized by inhibition of glucose reabsorption in the kidney accompanying with significant body weight reduction. It is known that GPR40 agonist stimulates insulin secretion in glucose independent manner. GKA potentiates glucose-stimulated insulin secretion from pancreatic beta cells and stimulates glucose uptake into the liver. Selective SGLT2 inhibitors and GPR40 agonist are expected to enter into the market in near future.


Subject(s)
Diabetes Mellitus/drug therapy , Drug Discovery/trends , Hypoglycemic Agents , Sodium-Glucose Transporter 2/physiology , Diabetes Mellitus/physiopathology , Enzyme Activators , Glucokinase/metabolism , Glucokinase/physiology , Humans , Insulin-Secreting Cells/physiology , Molecular Targeted Therapy , Pyridines , Receptors, G-Protein-Coupled/agonists , Sodium-Glucose Transporter 2 Inhibitors
3.
Lancet ; 378(9786): 182-97, 2011 Jul 09.
Article in English | MEDLINE | ID: mdl-21705062

ABSTRACT

The increasing prevalence, variable pathogenesis, progressive natural history, and complications of type 2 diabetes emphasise the urgent need for new treatment strategies. Longacting (eg, once weekly) agonists of the glucagon-like-peptide-1 receptor are advanced in development, and they improve prandial insulin secretion, reduce excess glucagon production, and promote satiety. Trials of inhibitors of dipeptidyl peptidase 4, which enhance the effect of endogenous incretin hormones, are also nearing completion. Novel approaches to glycaemic regulation include use of inhibitors of the sodium-glucose cotransporter 2, which increase renal glucose elimination, and inhibitors of 11ß-hydroxysteroid dehydrogenase 1, which reduce the glucocorticoid effects in liver and fat. Insulin-releasing glucokinase activators and pancreatic-G-protein-coupled fatty-acid-receptor agonists, glucagon-receptor antagonists, and metabolic inhibitors of hepatic glucose output are being assessed. Early proof of principle has been shown for compounds that enhance and partly mimic insulin action and replicate some effects of bariatric surgery.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , 11-beta-Hydroxysteroid Dehydrogenase Type 1/antagonists & inhibitors , Allylamine/analogs & derivatives , Allylamine/pharmacology , Allylamine/therapeutic use , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Bariatric Surgery , Bile Acids and Salts , Cardiovascular System/drug effects , Colesevelam Hydrochloride , Comorbidity , Diabetes Mellitus, Type 2/physiopathology , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Exenatide , Glucagon-Like Peptide 1/analysis , Glucokinase/physiology , Humans , Hyperglycemia/physiopathology , Hypoglycemic Agents/administration & dosage , Indoles/pharmacology , Indoles/therapeutic use , Insulin/pharmacology , Insulin Resistance/physiology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Liver/drug effects , Liver/physiopathology , Obesity/epidemiology , Obesity/surgery , Peptides/administration & dosage , Randomized Controlled Trials as Topic , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium-Glucose Transporter 2 Inhibitors , Treatment Outcome , Venoms/administration & dosage
4.
Biochem Biophys Res Commun ; 417(1): 495-500, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22177951

ABSTRACT

Uncoupling Protein 2 (UCP2) is expressed in the pancreatic ß-cell, where it partially uncouples the mitochondrial proton gradient, decreasing both ATP-production and glucose-stimulated insulin secretion (GSIS). Increased glucose levels up-regulate UCP2 mRNA and protein levels, but the mechanism for UCP2 up-regulation in response to increased glucose is unknown. The aim was to examine the effects of glucokinase (GK) deficiency on UCP2 mRNA levels and to characterize the interaction between UCP2 and GK with regard to glucose-stimulated insulin secretion in pancreatic islets. UCP2 mRNA expression was reduced in GK+/- islets and GK heterozygosity prevented glucose-induced up-regulation of islet UCP2 mRNA. In contrast to UCP2 protein function UCP2 mRNA regulation was not dependent on superoxide generation, but rather on products of glucose metabolism, because MnTBAP, a superoxide dismutase mimetic, did not prevent the glucose-induced up-regulation of UCP2. Glucose-stimulated insulin secretion was increased in UCP2-/- and GK+/- islets compared with GK+/- islets and UCP2 deficiency improved glucose tolerance of GK+/- mice. Accordingly, UCP2 deficiency increased ATP-levels of GK+/- mice. Thus, the compensatory down-regulation of UCP2 is involved in preserving the insulin secretory capacity of GK mutant mice and might also be implicated in limiting disease progression in MODY2 patients.


Subject(s)
Glucokinase/physiology , Glucose/metabolism , Ion Channels/biosynthesis , Islets of Langerhans/metabolism , Mitochondrial Proteins/biosynthesis , RNA, Messenger/biosynthesis , Adenosine Triphosphate/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Glucokinase/genetics , Glucose/pharmacology , Glucose Tolerance Test , Insulin/metabolism , Insulin Secretion , Ion Channels/genetics , Male , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , RNA, Messenger/genetics , Superoxides/metabolism , Uncoupling Protein 2
5.
IUBMB Life ; 64(6): 545-50, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22535686

ABSTRACT

Biochemistry in Spain owes much to the figure of Alberto Sols. In words of Nobel Prize winner Severo Ochoa: "He has been the first scientist to establish successfully biochemistry in Spain." His intellectual rigour, care in experimental design, emphasis on quality, and attention to the presentation of results permeated far beyond his inner circle to the then fledging Spanish biochemical community. It would be difficult to find some Spanish biochemist of the generation that now starts to retire who has not been influenced in a way or another by the work of Sols. However, it is also likely that the new generations of biochemists and molecular biologists in the country ignore who was Sols and what their field owns to him. The following lines try to highlight some key points of his scientific biography, the circumstances in which they took place and the state of the corresponding research area at that moment.


Subject(s)
Biochemistry/history , Biochemistry/education , Biological Transport , Glucokinase/physiology , History, 20th Century , Intestines/enzymology , Lactase/physiology , Liver/enzymology , Societies, Scientific/history , Spain
6.
J Pediatr Endocrinol Metab ; 25(7-8): 801-3, 2012.
Article in English | MEDLINE | ID: mdl-23155715

ABSTRACT

Maturity-onset diabetes of the young type 2 (MODY2) is an autosomal dominant inherited disease caused by heterozygous inactivating mutations in the glucokinase (GCK) gene and is characterized by mild noninsulin-dependent fasting hyperglycemia. It is treated with diet only, and complications are extremely rare. We present a report of a family with MODY2 caused by a novel NM_000162.3:c.878T>C mutation in exon 8 of the GCK gene. Testing for MODY2 and reporting all novel mutations are important to avoid difficulties in the interpretation of genetic test results and to provide fast and definitive diagnosis for all patients with this disease.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucokinase/genetics , Mutation , Adolescent , Blood Glucose/analysis , Circadian Rhythm , DNA Mutational Analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Genetic Predisposition to Disease , Glucokinase/physiology , Humans , Male , Mutation/physiology
7.
An R Acad Nac Med (Madr) ; 129(2): 541-63; discussion 563, 2012.
Article in Spanish | MEDLINE | ID: mdl-24298863

ABSTRACT

Metabolic sensors play an important role in the control of food intake, utilization of nutrients and demonstration of feeding behaviour. In this work we describe the study done in our laboratory on glucokinase (GK) as brain glucose sensor, the AMP kinase (AMPK) as detector of the fall of intracellular energy charge and as the S6K in the signaling pathway of mTOR with opposite effects to AMPK. Glucose sensors are molecular designs that detect with accuracy glucose concentrations, facilitating therefore the homeostasis of this hexose. We consider GK as a component of a glucose sensor system that might modulates the feeding behaviour and indirectly the control of body weight. Our findings indicate that GK and GLUT-2 mRNAs and proteins are coexpressed mainly in areas of the hypothalamus implied in the control of food intake. We have also found a high glucose phosphorylating activity with kinetic properties similar to that reported in the liver, with a high apparent Km for glucose that displays no product inhibition by glucose-6-phosphate. GK may be also regulated by the presence of glucokinase regulatory protein (GKRP), which has been identified in the same brain areas than GK. The coexpression of these molecules might play a role as glucose sensors in which GLUT-2 has a permissive role and the interactions of GK with GKRP made possible a real sensor activity. Furthermore, the effects of anorexigenic peptides in this system should facilitate the transduction of signals required to produce a state of satiety. Thus, GLP-1 reduced significantly the glucose metabolism in areas of the hypothalamus and brainstem related with food intake, which open new ways to the study of pathophysiologicals aspects of feeding behaviour. Besides we have studied the functions of AMPK and mTOR pathway in the hypothalamic areas ventromedial (VMH) and lateral (LH) under situations with alterations of the nutritional status and energy balance. Our results revealed that the activation of AMPK and S6K in VMH y LH occur in response to the changes of glucose concentrations or in the changes in the nutritional state, as well as GLP-1/exendin-4 act by counteracting the activation/inactivation of these kinases, which support a modulating role of these peptides on the kinases. On the other hand, GLP-1/exendin-4 might contribute to the normalization of the altered values of these kinases in pathophysiological states such as obesity.


Subject(s)
Body Weight/physiology , Brain/metabolism , Energy Metabolism , Feeding Behavior/physiology , Adenylate Kinase/physiology , Exenatide , Glucagon-Like Peptide 1/physiology , Glucokinase/physiology , Homeostasis , Humans , Hypothalamus/physiology , Peptides/physiology , Ribosomal Protein S6 Kinases/physiology , Signal Transduction , TOR Serine-Threonine Kinases/physiology , Venoms
8.
J Diabetes Investig ; 12(9): 1545-1554, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33638884

ABSTRACT

AIMS/INTRODUCTION: We aimed to determine whether glucokinase is required for ß-cell mass expansion induced by high-starch diet (HSTD)-feeding, as has been shown in its high-fat diet-induced expansion. MATERIALS AND METHODS: Eight-week-old male wild-type (Gck+/+ ) or glucokinase haploinsufficient (Gck+/- ) mice were fed either a normal chow (NC) or an HSTD for 15 weeks. The bodyweight, glucose tolerance, insulin sensitivity, insulin secretion and ß-cell mass were assessed. RESULTS: Both HSTD-fed Gck+/+ and Gck+/- mice had significantly higher bodyweight than NC-fed mice. Insulin and oral glucose tolerance tests revealed that HSTD feeding did not affect insulin sensitivity nor glucose tolerance in either the Gck+/+ or Gck+/- mice. However, during the oral glucose tolerance test, the 15-min plasma insulin concentration after glucose loading was significantly higher in the HSTD group than that in the NC group for Gck+/+ , but not for Gck+/- mice. ß-Cell mass was significantly larger in HSTD-fed Gck+/+ mice than that in NC-fed Gck+/+ mice. In contrast, the ß-cell mass of the HSTD-fed Gck+/- mice was not different from that of the NC-fed Gck+/- mice. CONCLUSIONS: The results showed that HSTD feeding would increase pancreatic ß-cell mass and insulin secretion in Gck+/+ , but not Gck+/- mice. This observation implies that glucokinase in ß-cells would be required for the increase in ß-cell mass induced by HSTD feeding.


Subject(s)
Diet, High-Fat , Glucokinase/physiology , Glucose/metabolism , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/cytology , Starch/administration & dosage , Animals , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Knockout
9.
J Bacteriol ; 192(11): 2892-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20363943

ABSTRACT

Glucose metabolism in Legionella pneumophila was studied by focusing on the Entner-Doudoroff (ED) pathway with a combined genetic and biochemical approach. The bacterium utilized exogenous glucose for synthesis of acid-insoluble cell components but manifested no discernible increase in the growth rate. Assays with permeabilized cell preparations revealed the activities of three enzymes involved in the pathway, i.e., glucokinase, phosphogluconate dehydratase, and 2-dehydro-3-deoxy-phosphogluconate aldolase, presumed to be encoded by the glk, edd, and eda genes, respectively. Gene-disrupted mutants for the three genes and the ywtG gene encoding a putative sugar transporter were devoid of the ability to metabolize exogenous glucose, indicating that the pathway is almost exclusively responsible for glucose metabolism and that the ywtG gene product is the glucose transporter. It was also established that these four genes formed part of an operon in which the gene order was edd-glk-eda-ywtG, as predicted by genomic information. Intriguingly, while the mutants exhibited no appreciable change in growth characteristics in vitro, they were defective in multiplication within eukaryotic cells, strongly indicating that the ED pathway must be functional for the intracellular growth of the bacterium to occur. Curiously, while the deficient glucose metabolism of the ywtG mutant was successfully complemented by the ywtG(+) gene supplied in trans via plasmid, its defect in intracellular growth was not. However, the latter defect was also manifested in wild-type cells when a plasmid carrying the mutant ywtG gene was introduced. This phenomenon, resembling so-called dominant negativity, awaits further investigation.


Subject(s)
Gene Expression Regulation, Bacterial/physiology , Glucose/metabolism , Legionella pneumophila/growth & development , Legionella pneumophila/metabolism , Signal Transduction/physiology , Aldehyde-Lyases/genetics , Aldehyde-Lyases/physiology , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Blotting, Northern , Gene Expression Regulation, Bacterial/genetics , Glucokinase/genetics , Glucokinase/physiology , Hydro-Lyases/genetics , Hydro-Lyases/physiology , Legionella pneumophila/genetics , Plasmids , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
10.
J Clin Invest ; 117(1): 246-57, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200721

ABSTRACT

Glucokinase (Gck) functions as a glucose sensor for insulin secretion, and in mice fed standard chow, haploinsufficiency of beta cell-specific Gck (Gck(+/-)) causes impaired insulin secretion to glucose, although the animals have a normal beta cell mass. When fed a high-fat (HF) diet, wild-type mice showed marked beta cell hyperplasia, whereas Gck(+/-) mice demonstrated decreased beta cell replication and insufficient beta cell hyperplasia despite showing a similar degree of insulin resistance. DNA chip analysis revealed decreased insulin receptor substrate 2 (Irs2) expression in HF diet-fed Gck(+/-) mouse islets compared with wild-type islets. Western blot analyses confirmed upregulated Irs2 expression in the islets of HF diet-fed wild-type mice compared with those fed standard chow and reduced expression in HF diet-fed Gck(+/-) mice compared with those of HF diet-fed wild-type mice. HF diet-fed Irs2(+/-) mice failed to show a sufficient increase in beta cell mass, and overexpression of Irs2 in beta cells of HF diet-fed Gck(+/-) mice partially prevented diabetes by increasing beta cell mass. These results suggest that Gck and Irs2 are critical requirements for beta cell hyperplasia to occur in response to HF diet-induced insulin resistance.


Subject(s)
Dietary Fats/pharmacology , Glucokinase/physiology , Insulin Resistance/physiology , Insulin-Secreting Cells/pathology , Insulin/physiology , Intracellular Signaling Peptides and Proteins/physiology , Phosphoproteins/physiology , Animals , Glucokinase/deficiency , Glucokinase/genetics , Humans , Hyperplasia , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphoproteins/deficiency , Phosphoproteins/genetics , Signal Transduction
11.
Rev Endocr Metab Disord ; 11(3): 179-83, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20878480

ABSTRACT

Glucokinase is a key enzyme involved in regulating insulin secretion from the pancreatic ß-cell. The unique role of glucokinase in human glucose physiology is illustrated by the fact that genetic mutations in glucokinase can either cause hyperglycaemia or hypoglycaemia. Heterozygous inactivating mutations in glucokinase cause maturity-onset diabetes of the young (MODY), homozygous inactivating in glucokinase mutations result in permanent neonatal diabetes whereas heterozygous activating glucokinase mutations cause hyperinsulinaemic hypoglycaemia.


Subject(s)
Congenital Hyperinsulinism/genetics , Diabetes Mellitus/congenital , Diabetes Mellitus/genetics , Glucokinase/genetics , Insulin-Secreting Cells/enzymology , Diabetes Mellitus, Type 2/congenital , Diabetes Mellitus, Type 2/genetics , Glucokinase/metabolism , Glucokinase/physiology , Humans , Infant, Newborn , Insulin-Secreting Cells/metabolism , Models, Biological , Mutation/physiology
12.
Biochem J ; 414(1): 1-18, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18651836

ABSTRACT

Conversion of glucose into glycogen is a major pathway that contributes to the removal of glucose from the portal vein by the liver in the postprandial state. It is regulated in part by the increase in blood-glucose concentration in the portal vein, which activates glucokinase, the first enzyme in the pathway, causing an increase in the concentration of glucose 6-P (glucose 6-phosphate), which modulates the phosphorylation state of downstream enzymes by acting synergistically with other allosteric effectors. Glucokinase is regulated by a hierarchy of transcriptional and post-transcriptional mechanisms that are only partially understood. In the fasted state, glucokinase is in part sequestered in the nucleus in an inactive state, complexed to a specific regulatory protein, GKRP (glucokinase regulatory protein). This reserve pool is rapidly mobilized to the cytoplasm in the postprandial state in response to an elevated concentration of glucose. The translocation of glucokinase between the nucleus and cytoplasm is modulated by various metabolic and hormonal conditions. The elevated glucose 6-P concentration, consequent to glucokinase activation, has a synergistic effect with glucose in promoting dephosphorylation (inactivation) of glycogen phosphorylase and inducing dephosphorylation (activation) of glycogen synthase. The latter involves both a direct ligand-induced conformational change and depletion of the phosphorylated form of glycogen phosphorylase, which is a potent allosteric inhibitor of glycogen synthase phosphatase activity associated with the glycogen-targeting protein, GL [hepatic glycogen-targeting subunit of PP-1 (protein phosphatase-1) encoded by PPP1R3B]. Defects in both the activation of glucokinase and in the dephosphorylation of glycogen phosphorylase are potential contributing factors to the dysregulation of hepatic glucose metabolism in Type 2 diabetes.


Subject(s)
Glucokinase/metabolism , Liver Glycogen/metabolism , Liver/enzymology , Animals , Glucokinase/chemistry , Glucokinase/physiology , Glucose-6-Phosphate/chemistry , Glucose-6-Phosphate/metabolism , Glucose-6-Phosphate/physiology , Humans , Liver/blood supply , Liver/physiology , Liver Glycogen/chemistry , Liver Glycogen/physiology , Phosphorylation , Signal Transduction/physiology
13.
Mol Metab ; 17: 17-27, 2018 11.
Article in English | MEDLINE | ID: mdl-30146176

ABSTRACT

OBJECTIVE: Appropriate glucose levels are essential for survival; thus, the detection and correction of low blood glucose is of paramount importance. Hypoglycemia prompts an integrated response involving reduction in insulin release and secretion of key counter-regulatory hormones glucagon and epinephrine that together promote endogenous glucose production to restore normoglycemia. However, specifically how this response is orchestrated remains to be fully clarified. The low affinity hexokinase glucokinase is found in glucose-sensing cells involved in glucose homeostasis including pancreatic ß-cells and in certain brain areas. Here, we aimed to examine the role of glucokinase in triggering counter-regulatory hormonal responses to hypoglycemia, hypothesizing that reduced glucokinase activity would lead to increased and/or earlier triggering of responses. METHODS: Hyperinsulinemic glucose clamps were performed to examine counter-regulatory responses to controlled hypoglycemic challenges created in humans with monogenic diabetes resulting from heterozygous glucokinase mutations (GCK-MODY). To examine the relative importance of glucokinase in different sensing areas, we then examined responses to clamped hypoglycemia in mice with molecularly defined disruption of whole body and/or brain glucokinase. RESULTS: GCK-MODY patients displayed increased and earlier glucagon responses during hypoglycemia compared with a group of glycemia-matched patients with type 2 diabetes. Consistent with this, glucagon responses to hypoglycemia were also increased in I366F mice with mutated glucokinase and in streptozotocin-treated ß-cell ablated diabetic I366F mice. Glucagon responses were normal in conditional brain glucokinase-knockout mice, suggesting that glucagon release during hypoglycemia is controlled by glucokinase-mediated glucose sensing outside the brain but not in ß-cells. For epinephrine, we found increased responses in GCK-MODY patients, in ß-cell ablated diabetic I366F mice and in conditional (nestin lineage) brain glucokinase-knockout mice, supporting a role for brain glucokinase in triggering epinephrine release. CONCLUSIONS: Our data suggest that glucokinase in brain and other non ß-cell peripheral hypoglycemia sensors is important in glucose homeostasis, allowing the body to detect and respond to a falling blood glucose.


Subject(s)
Diabetes Mellitus/metabolism , Glucokinase/physiology , Hypoglycemia/metabolism , Adult , Animals , Blood Glucose/analysis , Diabetes Mellitus/genetics , Disease Models, Animal , Epinephrine , Female , Glucagon/blood , Glucokinase/metabolism , Glucose/metabolism , Glucose Clamp Technique , Humans , Hyperinsulinism , Hypoglycemia/physiopathology , Hypoglycemic Agents , Insulin/blood , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred BALB C , Middle Aged
14.
Ann N Y Acad Sci ; 1411(1): 65-82, 2018 01.
Article in English | MEDLINE | ID: mdl-29044608

ABSTRACT

Pancreatic beta cells sense changes in nutrients during the cycles of fasting and feeding and release insulin accordingly to maintain glucose homeostasis. Abnormal beta cell nutrient sensing resulting from gene mutations leads to hypoglycemia or diabetes. Glucokinase (GCK) plays a key role in beta cell glucose sensing. As one form of congenital hyperinsulinism (CHI), activating mutations of GCK result in a decreased threshold for glucose-stimulated insulin secretion and hypoglycemia. In contrast, inactivating mutations of GCK result in diabetes, including a mild form (MODY2) and a severe form (permanent neonatal diabetes mellitus (PNDM)). Mutations of beta cell ion channels involved in insulin secretion regulation also alter glucose sensing. Activating or inactivating mutations of ATP-dependent potassium (KATP ) channel genes result in severe but completely opposite clinical phenotypes, including PNDM and CHI. Mutations of the other ion channels, including voltage-gated potassium channels (Kv 7.1) and voltage-gated calcium channels, also lead to abnormal glucose sensing and CHI. Furthermore, amino acids can stimulate insulin secretion in a glucose-independent manner in some forms of CHI, including activating mutations of the glutamate dehydrogenase gene, HDAH deficiency, and inactivating mutations of KATP channel genes. These genetic defects have provided insight into a better understanding of the complicated nature of beta cell fuel-sensing mechanisms.


Subject(s)
Congenital Hyperinsulinism/physiopathology , Diabetes Mellitus/physiopathology , Glucokinase/physiology , Ion Channels/physiology , Islets of Langerhans/physiology , Nutrients/pharmacokinetics , 3-Hydroxyacyl CoA Dehydrogenases/deficiency , Amino Acids/pharmacokinetics , Animals , Blood Glucose/metabolism , Congenital Hyperinsulinism/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Fatty Acids/metabolism , Glucokinase/deficiency , Glucokinase/genetics , Glucose/pharmacokinetics , Humans , Insulin/physiology , Ion Channels/genetics , Metabolism, Inborn Errors/physiopathology , Mice , Mice, Knockout , Mutation , Oxidation-Reduction
15.
Endocrinology ; 148(4): 1928-32, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17218412

ABSTRACT

Because appetite, hypothalamic gene expression, reproductive function, and adrenal function are highly sensitive to acute changes in plasma glucose levels, it has been hypothesized hypothalamic neurons sensitive to glucose play a role in regulating these functions. To assess this hypothesis, we examined these neuronendocrine functions in mice in which the glucokinase gene, which plays an essential role in neuroendocrine glucose sensing, has been ablated. Haploinsufficiency in heterozygous glucokinase knockout mice produced effects similar to those produced by hypoglycemia: impaired reproductive function, elevated plasma corticosterone, increased food intake, and hypothalamic gene expression similar to that observed in fasted or leptin-deficient obese mice (increased hypothalamic neuropeptide Y mRNA and reduced hypothalamic proopiomelanocortin mRNA). Plasma glucose was elevated 2-fold in glucokinase knockout mice, consistent with a maturity-onset diabetes of the young phenotype, but plasma insulin and leptin levels were normal. These data support the hypothesis that glucokinase plays a key role in the neuroendocrine regulation of metabolic economy.


Subject(s)
Eating/genetics , Glucocorticoids/metabolism , Glucokinase/physiology , Hypothalamus/metabolism , Reproduction/genetics , Animals , Female , Fertility/genetics , Gene Expression Regulation , Glucokinase/genetics , Hyperglycemia/genetics , Insulin/blood , Insulin/genetics , Leptin/blood , Male , Mice , Mice, Knockout , Promoter Regions, Genetic
16.
Endocrinology ; 147(11): 5196-204, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16916947

ABSTRACT

Insulin synthesis and release activities of beta-cells can be acutely regulated by glucose through its glycolytic and mitochondrial breakdown involving a glucokinase-dependent rate-limiting step. Isolated beta-cell populations are composed of cells with intercellular differences in acute glucose responsiveness that have been attributed to differences in glucokinase (GK) expression and activity. This study first shows that glyceraldehyde can be used as GK-bypassing oxidative substrate and then examines whether the triose can metabolically activate beta-cells with low glucose responsiveness. Glyceraldehyde 1 mm induced a similar cellular (14)CO(2) output and metabolic redox state as glucose 4 mM. Using flow cytometric analysis, glyceraldehyde (0.25-2 mM) was shown to concentration-dependently increase the percent metabolically activated cells at all tested glucose concentrations (2.5-20 mM). Its ability to activate beta-cells that are unresponsive to the prevailing glucose level was further illustrated in glucose low-responsive cells that were isolated by flow sorting. Metabolic activation by glyceraldehyde was associated with an activation of nutrient-driven translational control proteins and an increased protein synthetic response to glucose, however not beyond the maximal rates that are inducible by glucose alone. It is concluded that glucose low-responsive beta-cells can be metabolically activated by the GK-bypassing glyceraldehyde, increasing their acute biosynthetic response to glucose but not their maximal glucose-inducible biosynthetic capacity, which is considered subject to chronic regulation.


Subject(s)
Glucose/pharmacology , Glyceraldehyde/pharmacology , Islets of Langerhans/metabolism , Protein Biosynthesis/drug effects , Animals , Glucokinase/physiology , Glucose/metabolism , Islets of Langerhans/drug effects , Male , Oxidation-Reduction , Rats , Rats, Wistar
17.
Endocrinology ; 147(12): 5768-76, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16980436

ABSTRACT

The glucose sensor enzyme glucokinase plays a pivotal role in the regulation of glucose-induced insulin secretion in pancreatic beta-cells. Activation of glucokinase represents a promising concept for the treatment of type 2 diabetes. Therefore, we analyzed the glucokinase activation through its physiological interaction partner, the bifunctional enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFK-2/FBPase-2) and the resulting effect on glucose metabolism in insulin-producing cells. In RINm5F-GK-PFK-2/FBPase-2 cells stably overexpressing glucokinase plus islet PFK-2/FBPase-2, colocalization between both enzymes as well as elevation of glucokinase activity were significantly increased at a stimulatory glucose concentration of 10 mmol/liter. RINm5F-GK-PFK-2/FBPase-2 cells showed under this culture condition a significant increase in glucose utilization and in the ATP/ADP ratio compared with RINm5F-GK cells, which only overexpress glucokinase. Also glucose-induced insulin secretion was elevated in RINm5F-GK-PFK-2/FBPase-2 cells in comparison to RINm5F-GK cells. Furthermore, pyruvate accumulation and lactate production in RINm5F-GK-PFK-2/FBPase-2 cells were significantly lower at both 10 and 30 mmol/liter glucose than in RINm5F-GK and RINm5F cells. The significant improvement of glucose metabolism after PFK-2/FBPase-2 overexpression is apparently not exclusively the result of high glucokinase enzyme activity. Stabilization of the closed glucokinase conformation by PFK-2/FBPase-2 may not only activate the enzyme but also improve metabolic channeling in beta-cells.


Subject(s)
Glucokinase/physiology , Glucose/metabolism , Insulin/metabolism , Phosphofructokinase-2/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cell Survival , Glucokinase/metabolism , Glucose/pharmacology , Humans , Insulin Secretion , Islets of Langerhans/metabolism , Lactic Acid/biosynthesis , Metabolic Networks and Pathways , Protein Binding , Pyruvic Acid/metabolism , Rats , Transfection
18.
Diabetes ; 54(10): 2925-31, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16186394

ABSTRACT

As the rate-limiting controller of glucose metabolism, glucokinase represents the primary beta-cell "glucose sensor." Inactivation of both glucokinase (GK) alleles results in permanent neonatal diabetes; inactivation of a single allele causes maturity-onset diabetes of the young type 2 (MODY-2). Similarly, mice lacking both alleles (GK(-/-)) exhibit severe neonatal diabetes and die within a week, whereas heterozygous GK(+/-) mice exhibit markedly impaired glucose tolerance and diabetes, resembling MODY-2. Glucose metabolism increases the cytosolic [ATP]-to-[ADP] ratio, which closes ATP-sensitive K(+) channels (K(ATP) channels), leading to membrane depolarization, Ca(2+) entry, and insulin exocytosis. Glucokinase insufficiency causes defective K(ATP) channel regulation, which may underlie the impaired secretion. To test this prediction, we crossed mice lacking neuroendocrine glucokinase (nGK(+/-)) with mice lacking K(ATP) channels (Kir6.2(-/-)). Kir6.2 knockout rescues perinatal lethality of nGK(-/-), although nGK(-/-)Kir6.2(-/-) animals are postnatally diabetic and still die prematurely. nGK(+/-) animals are diabetic on the Kir6.2(+/+) background but only mildly glucose intolerant on the Kir6.2(-/-) background. In the presence of glutamine, isolated nGK(+/-)Kir6.2(-/-) islets show improved insulin secretion compared with nGK(+/-)Kir6.2(+/+). The significant abrogation of nGK(-/-) and nGK(+/-) phenotypes in the absence of K(ATP) demonstrate that a major factor in glucokinase deficiency is indeed altered K(ATP) signaling. The results have implications for understanding and therapy of glucokinase-related diabetes.


Subject(s)
Diabetes Mellitus/enzymology , Glucokinase/deficiency , Potassium Channels, Inwardly Rectifying/physiology , Signal Transduction , Animals , Animals, Newborn , Blood Glucose/analysis , Crosses, Genetic , Diabetes Mellitus/genetics , Diabetes Mellitus/mortality , Genotype , Glucokinase/physiology , Glutamine/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/enzymology , Islets of Langerhans/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium Channels, Inwardly Rectifying/deficiency
19.
Diabetes ; 49(2): 195-201, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10868935

ABSTRACT

To identify the amino acids involved in the specific regulatory properties of glucokinase, and particularly its low affinity for glucose, mutants of the human islet enzyme have been prepared, in which glucokinase-specific residues have been replaced. Two mutations increased the affinity for glucose by twofold (K296M) and sixfold (Y214A), the latter also decreasing the Hill coefficient from 1.75 to 1.2 with minimal change in the affinity for ATP. Combining these two mutations with N166R resulted in a 50-fold decrease in the half-saturating substrate concentration (S0.5) value, which became then comparable to the Km of hexokinase II. The location of N166, Y214, and K296 in the three-dimensional structure of glucokinase suggests that these mutations act by favoring closure of the catalytic cleft. As a rule, mutations changed the affinity for glucose and for the competitive inhibitor mannoheptulose (MH) in parallel, whereas they barely affected the affinity for N-acetylglucosamine (NAG). These and other results suggest that NAG and MH bind to the same site but to different conformations of glucokinase. A small reduction in the affinity for the regulatory protein was observed with mutations of residues on the smaller domain and in the hinge region, confirming the bipartite nature of the binding site for the regulatory protein. The K296M mutant was found to have a threefold decreased affinity for palmitoyl CoA; this effect was additive to that previously observed for the E279Q mutant, indicating that the binding site for long-chain acyl CoAs is located on the upper face of the larger domain.


Subject(s)
Glucokinase/physiology , Adenosine Triphosphate/metabolism , Binding, Competitive , Enzyme Inhibitors/pharmacology , Glucokinase/chemistry , Glucokinase/genetics , Glucokinase/metabolism , Glucose/metabolism , Humans , Molecular Structure , Mutagenesis, Site-Directed , Mutation/physiology
20.
Diabetes ; 35(1): 61-7, 1986 Jan.
Article in English | MEDLINE | ID: mdl-3510141

ABSTRACT

The enzyme glucokinase controls glucose metabolism in islets and is proposed to be the glucose sensor in pancreatic beta-cells. This concept was developed from studies with rodents and it remained to be explored whether it also applies to man. Studies in man were hampered, however, by the difficulty in obtaining well-preserved pancreatic islet tissue and also because the high activity of hexokinase made it difficult to measure glucokinase. To overcome these obstacles, quantitative histochemical sampling techniques were developed allowing precise dissection of pure human islet tissue and a newly designed radiometric microassay was used, avoiding hexokinase interference, and providing the sensitivity necessary to measure the relatively low glucokinase activity in small samples of tissue obtained from brain-dead tissue donors. The present data indicate that glucokinase is present in human pancreatic islet tissue and is not found in the exocrine pancreas. The enzyme's Vmax with D-glucose as substrate was similar to the Vmax for glucose utilization reported previously for intact, isolated human islets and the enzyme's Km for D-glucose was about 5 mM. Since glucokinase was also present in islet tissue of hamster, mouse, and rat, it is suggested that the glucokinase-glucose sensor concept has general applicability and that it could explain many aspects of the physiology and pathology of glucose homeostasis. This well-defined pancreatic islet glucokinase-glucose sensor should, therefore, be incorporated in any comprehensive model of glucose homeostasis.


Subject(s)
Glucokinase/physiology , Glucose/metabolism , Islets of Langerhans/physiology , Adolescent , Adult , Aged , Animals , Child , Cricetinae , Cricetulus , Female , Glucagon/analysis , Hexokinase/metabolism , Humans , Insulin/analysis , Islets of Langerhans/analysis , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred CBA , Phosphorylation , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL