Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 9(1): e1003116, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23326232

ABSTRACT

Mycobacterium tuberculosis (Mtb) is thought to preferentially rely on fatty acid metabolism to both establish and maintain chronic infections. Its metabolic network, however, allows efficient co-catabolism of multiple carbon substrates. To gain insight into the importance of carbohydrate substrates for Mtb pathogenesis we evaluated the role of glucose phosphorylation, the first reaction in glycolysis. We discovered that Mtb expresses two functional glucokinases. Mtb required the polyphosphate glucokinase PPGK for normal growth on glucose, while its second glucokinase GLKA was dispensable. (13)C-based metabolomic profiling revealed that both enzymes are capable of incorporating glucose into Mtb's central carbon metabolism, with PPGK serving as dominant glucokinase in wild type (wt) Mtb. When both glucokinase genes, ppgK and glkA, were deleted from its genome, Mtb was unable to use external glucose as substrate for growth or metabolism. Characterization of the glucokinase mutants in mouse infections demonstrated that glucose phosphorylation is dispensable for establishing infection in mice. Surprisingly, however, the glucokinase double mutant failed to persist normally in lungs, which suggests that Mtb has access to glucose in vivo and relies on glucose phosphorylation to survive during chronic mouse infections.


Subject(s)
Bacterial Proteins/metabolism , Glucokinase/metabolism , Glucose/metabolism , Host-Pathogen Interactions , Mycobacterium tuberculosis/pathogenicity , Phosphotransferases/metabolism , Tuberculosis/metabolism , Animals , Bacterial Proteins/genetics , Carbon Radioisotopes/metabolism , Disease Models, Animal , Female , Gene Knockout Techniques , Glucokinase/deficiency , Glucokinase/genetics , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Mycobacterium tuberculosis/enzymology , Phosphorylation , Phosphotransferases/deficiency , Phosphotransferases/genetics , Substrate Specificity , Tuberculosis/immunology , Tuberculosis/microbiology
2.
Mol Metab ; 53: 101311, 2021 11.
Article in English | MEDLINE | ID: mdl-34325016

ABSTRACT

OBJECTIVES: Glucokinase (GCK) is critical for glucosensing. In rats, GCK is expressed in hypothalamic tanycytes and appears to play an essential role in feeding behavior. In this study, we investigated the distribution of GCK-expressing tanycytes in mice and their role in the regulation of energy balance. METHODS: In situ hybridization, reporter gene assay, and immunohistochemistry were used to assess GCK expression along the third ventricle in mice. To evaluate the impact of GCK-expressing tanycytes on arcuate neuron function and mouse physiology, Gck deletion along the ventricle was achieved using loxP/Cre recombinase technology in adult mice. RESULTS: GCK expression was low along the third ventricle, but detectable in tanycytes facing the ventromedial arcuate nucleus from bregma -1.5 to -2.2. Gck deletion induced the death of this tanycyte subgroup through the activation of the BAD signaling pathway. The ablation of GCK-expressing tanycytes affected different aspects of energy balance, leading to an increase in adiposity in mice. This phenotype was systematically associated with a defect in NPY neuron function. In contrast, the regulation of glucose homeostasis was mostly preserved, except for glucoprivic responses. CONCLUSIONS: This study describes the role of GCK in tanycyte biology and highlights the impact of tanycyte loss on the regulation of energy balance.


Subject(s)
Ependymoglial Cells/metabolism , Glucokinase/genetics , Adiposity , Animals , Energy Metabolism , Glucokinase/deficiency , Glucokinase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
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
4.
Article in English | MEDLINE | ID: mdl-32508747

ABSTRACT

Introduction: The prevalence among pregnant women with diabetes of monogenic diabetes due to glucokinase deficit (GCK-MODY) varies from 0 to 80% in different studies, based on the chosen selection criteria for genetic test. New pregnancy-specific Screening Criteria (NSC), validated on an Anglo-Celtic pregnant cohort, have been proposed and include pre-pregnancy BMI <25 kg/m2 and fasting glycemia >99 mg/dl. Our aim was to estimate the prevalence of GCK-MODY and to evaluate the diagnostic performance of NSC in our population of women with diabetes in pregnancy. Patients and Methods: We retrospectively selected from our database of 468 diabetic pregnant patients in Sant'Andrea Hospital, in Rome, from 2010 to 2018, all the women who received a genetic test for GCK deficit because of specific clinical features. We estimated the prevalence of GCK-MODY among tested women and the minimum prevalence in our entire population with non-autoimmune diabetes. We evaluated diagnostic performance of NSC on the tested cohort and estimated the eligibility to genetic test based on NSC in the entire population. Results: A total of 409 patients had diabetes in pregnancy, excluding those with autoimmune diabetes; 21 patients have been tested for GCK-MODY, 8 have been positive and 13 have been negative (2 of them had HNF1-alfa mutations and 1 had HNF4-alfa mutation). We found no significant differences in clinical features between positive and negative groups except for fasting glycemia, which was higher in the positive group. The minimum prevalence of monogenic diabetes in our population was 2.4%. The minimum prevalence of GCK-MODY was 1.95%. In the tested cohort, the prevalence of GCK-MODY was 38%. In this group, NSC sensitivity is 87% and specificity is 30%, positive predictive value is 43%, and negative predictive value is 80%. Applying NSC on the entire population of women with non-autoimmune diabetes in pregnancy, 41 patients (10%) would be eligible for genetic test; considering a fasting glycemia >92 mg/dl, 85 patients (20.7%) would be eligible. Discussion: In our population, NSC have good sensitivity but low specificity, probably because there are many GDM with GCK-MODY like features. It is mandatory to define selective criteria with a good diagnostic performance on Italian population, to avoid unnecessary genetic tests.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Diabetes, Gestational/epidemiology , Glucokinase/deficiency , Mutation , Pregnancy in Diabetics/epidemiology , Adult , Biomarkers/analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes, Gestational/blood , Diabetes, Gestational/enzymology , Diabetes, Gestational/genetics , Female , Follow-Up Studies , Genetic Testing , Glucokinase/genetics , Humans , Italy/epidemiology , Pregnancy , Pregnancy in Diabetics/blood , Pregnancy in Diabetics/enzymology , Pregnancy in Diabetics/genetics , Prevalence , Prognosis , Retrospective Studies
5.
Pediatr Diabetes ; 9(3 Pt 1): 245-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18298419

ABSTRACT

Glucokinase deficiency is an unfrequent cause of permanent neonatal diabetes (PND), as only seven patients have been reported, either homozygous for a missense or frameshift mutation or compound heterozygous for both of them. We report here the first known case caused by a homozygous nonsense mutation (Y61X) in the glucokinase gene (GCK) that introduces a premature stop codon, generating a truncated protein that is predicted to be completely inactive as it lacks both the glucose- and the adenosine triphosphate-binding sites. The proband, born to consanguineous parents, was a full-term, intra-uterine growth-retarded male newborn who presented with a glycaemia of 129 mg/dL (7.16 mmol/L) on his second day of life, increasing thereafter up to 288 mg/dL (15.98 mmol/L) and 530 mg/dL (29.41 mmol/L) over the next 24 h, in the face of low serum insulin (<3 muIU/mL; <20.83 pmol/L). He was put on insulin on the third day of life. Insulin has never been discontinued since then. The patient was tested negative for anti-insulin and islet cell antibodies at age 5 months. His father had non-progressive, impaired fasting glucose for several years. The mother was found to be mildly hyperglycaemic only when her glucose was checked after the child was diagnosed. In conclusion, biallelic GCK loss should be considered as a potential cause of PND in children born to consanguineous parents, even if they are not known to be diabetic at the time of PND presentation.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Glucokinase/deficiency , Glucokinase/genetics , Insulin/therapeutic use , Anti-Bacterial Agents/therapeutic use , Codon, Nonsense , Consanguinity , Diabetes Mellitus, Type 1/drug therapy , Escherichia coli Infections/complications , Escherichia coli Infections/diagnosis , Female , Homozygote , Humans , Hyperglycemia/enzymology , Hyperglycemia/genetics , Infant, Newborn , Injections, Subcutaneous , Insulin/administration & dosage , Male , Pedigree
6.
Nat Commun ; 9(1): 546, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416045

ABSTRACT

Glucagon secretion by pancreatic α-cells is triggered by hypoglycemia and suppressed by high glucose levels; impaired suppression of glucagon secretion is a hallmark of both type 1 and type 2 diabetes. Here, we show that α-cell glucokinase (Gck) plays a role in the control of glucagon secretion. Using mice with α-cell-specific inactivation of Gck (αGckKO mice), we find that glucokinase is required for the glucose-dependent increase in intracellular ATP/ADP ratio and the closure of KATP channels in α-cells and the suppression of glucagon secretion at euglycemic and hyperglycemic levels. αGckKO mice display hyperglucagonemia in the fed state, which is associated with increased hepatic gluconeogenic gene expression and hepatic glucose output capacity. In adult mice, fed hyperglucagonemia is further increased and glucose intolerance develops. Thus, glucokinase governs an α-cell metabolic pathway that suppresses secretion at or above normoglycemic levels; abnormal suppression of glucagon secretion deregulates hepatic glucose metabolism and, over time, induces a pre-diabetic phenotype.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Glucokinase/genetics , Glucose Intolerance/metabolism , Glucose/metabolism , Hypoglycemia/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Transport , Female , Gene Expression , Glucagon-Secreting Cells/pathology , Glucokinase/deficiency , Glucose Intolerance/genetics , Glucose Intolerance/pathology , Hypoglycemia/genetics , Hypoglycemia/pathology , Insulin/metabolism , KATP Channels/genetics , KATP Channels/metabolism , Liver/metabolism , Male , Mice , Mice, Knockout
7.
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
8.
J Clin Invest ; 98(8): 1755-61, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8878425

ABSTRACT

All glucokinase gene mutations identified to date have been localized to exons that are common to the pancreatic and hepatic isoforms of the enzyme. While impaired insulin secretion has been observed in glucokinase-deficient subjects the consequences of this mutation on hepatic glucose metabolism remain unknown. To examine this question hepatic glycogen concentration was measured in seven glucokinase-deficient subjects with normal glycosylated hemoglobin and 12 control subjects using 13C nuclear magnetic spectroscopy during a day in which three isocaloric mixed meals were ingested. The relative fluxes of the direct and indirect pathways of hepatic glycogen synthesis were also assessed using [1-13C]glucose in combination with acetaminophen to noninvasively sample the hepatic UDP-glucose pool. Average fasting hepatic glycogen content was similar in glucokinase-deficient and control subjects (279+/-20 vs 284+/-14 mM; mean+/-SEM), and increased in both groups after the meals with a continuous pattern throughout the day. However, the net increment in hepatic glycogen content after each meal was 30-60% lower in glucokinase-deficient than in the control subjects (breakfast, 46% lower, P < 0.02; lunch, 62% lower, P = 0.002; dinner; 30% lower, P = 0.04). The net increment over basal values 4 h after dinner was 105 +/-18 mM in glucokinase-deficient and 148+/-11 mM in control subjects (P = 0.04). In the 4 h after breakfast, flux through the gluconeogenic pathway relative to the direct pathway of hepatic glycogen synthesis was higher in glucokinase-deficient than in control subjects (50+/-2% vs 34+/-5%; P = 0.038). In conclusion glucokinase-deficient subjects have decreased net accumulation of hepatic glycogen and relatively augmented hepatic gluconeogenesis after meals. These results suggest that in addition to the altered beta cell function, abnormalities in liver glycogen metabolism play an important role in the pathogenesis of hyperglycemia in patients with glucokinase-deficient maturity onset diabetes of young.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucokinase/deficiency , Liver Glycogen/biosynthesis , Adult , Glucokinase/genetics , Gluconeogenesis , Humans , Insulin/blood , Male , Middle Aged
9.
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
10.
Eur J Emerg Med ; 13(6): 349-51, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17091057

ABSTRACT

Diabetes mellitus is a heterogeneous disorder that may occur at any age. Neonatal diabetes mellitus, defined as hyperglycaemia presenting within the first 6 weeks of life in term infants, is a rare disorder that may result in permanent or transient diabetes mellitus. Although reported in paediatric diabetes literature, there are no reports of this condition in emergency medicine journals and these children may present to emergency departments with a picture mimicking sepsis. We report the case of a 5-week-old infant with diabetes mellitus who presented with diabetic ketoacidosis and review the literature surrounding this rare condition.


Subject(s)
Diabetes Mellitus , Emergency Treatment/methods , Intensive Care, Neonatal/methods , ATP Phosphoribosyltransferase/genetics , Critical Illness , Diabetes Mellitus/congenital , Diabetes Mellitus/diagnosis , Diabetes Mellitus/therapy , Diabetic Ketoacidosis/etiology , Diagnosis, Differential , Fluid Therapy/methods , Genetic Diseases, Inborn/complications , Glucokinase/deficiency , Glucokinase/genetics , Homeodomain Proteins/genetics , Humans , Hypoglycemic Agents/therapeutic use , Infant , Insulin/therapeutic use , Male , Mutation/genetics , Rare Diseases , Sepsis/diagnosis , Trans-Activators/genetics , X-Linked Combined Immunodeficiency Diseases/complications
11.
Nat Commun ; 7: 11933, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27324299

ABSTRACT

Recent advances in genome engineering have further widened the gap between our ability to implement essentially any genetic change and understanding the impact of these changes on cellular function. We lack efficient methods to diagnose limiting steps in engineered pathways. Here, we develop a generally applicable approach to reveal limiting steps within a synthetic pathway. It is based on monitoring metabolite dynamics and simplified kinetic modelling to differentiate between putative causes of limiting product synthesis during the start-up phase of the pathway with near-maximal rates. We examine the synthetic N-acetylglucosamine (GlcNAc) pathway in Bacillus subtilis and find none of the acetyl-, amine- or glucose-moiety precursors to limit synthesis. Our dynamic metabolomics approach predicts an energy-dissipating futile cycle between GlcNAc6P and GlcNAc as the primary problem in the pathway. Deletion of the responsible glucokinase more than doubles GlcNAc productivity by restoring healthy growth of the overproducing strain.


Subject(s)
Acetylglucosamine/analogs & derivatives , Acetylglucosamine/metabolism , Bacillus subtilis/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Glucokinase/genetics , Bacillus subtilis/genetics , Bacterial Proteins/metabolism , Citric Acid Cycle/genetics , Computer Simulation , Culture Media/chemistry , Gene Deletion , Glucokinase/deficiency , Glucose/metabolism , Glycolysis/genetics , Kinetics , Metabolic Engineering/methods , Metabolomics/methods , Models, Chemical , Substrate Cycling
12.
Diabetes ; 49(1): 82-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10615953

ABSTRACT

Low birth weight has been reported to be associated with impaired insulin secretion and insulin resistance. It has been proposed that this association results from fetal programming in response to the intrauterine environment (the thrifty phenotype hypothesis). To elucidate the relationship between birth weight and genetically determined defects in insulin secretion, we measured the birth weights of neonates derived from crosses of male pancreatic beta-cell type glucokinase knockout (Gck+/-) mice and female wild-type (WT) or Gck+/- mice. In 135 offspring, birth weights were lower in the presence of a fetal heterozygous mutation and higher in the presence of a maternal heterozygous mutation. Moreover, Gck-/- neonates had significantly smaller birth weights than WT or Gck+/- neonates (means +/- SE 1.49+/-0.03 [n = 30] vs. 1.63+/-0.03 [n = 30] or 1.63+/-0.02 [n = 50] g, respectively; P<0.01). Thus, Gck mutations in beta-cells may impair insulin response to glucose and alter intrauterine growth as well as glucose metabolism after birth. This study has confirmed the results of a previous report that human subjects carrying mutations in Gck had reduced birth weights and has provided direct evidence for a link between insulin and fetal growth. Moreover, birth weights were reduced in insulin receptor substrate-1 knockout mice despite normal insulin levels. Taken together, these results suggest that a genetically programmed insulin effect during embryogenesis determines fetal growth and provides a possible molecular link between birth weight and susceptibility to type 2 diabetes.


Subject(s)
Fetus/physiology , Insulin/physiology , Animals , Animals, Newborn/physiology , Birth Weight/genetics , Diabetes Mellitus, Type 2/genetics , Embryonic and Fetal Development/physiology , Female , Genetic Predisposition to Disease , Glucokinase/deficiency , Glucokinase/genetics , Heterozygote , Insulin Receptor Substrate Proteins , Male , Mice , Mice, Knockout/genetics , Mutation/physiology , Phosphoproteins/deficiency , Phosphoproteins/genetics
13.
Diabetes ; 43(12): 1402-6, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7958490

ABSTRACT

Insulin and glucagon release and insulin sensitivity were investigated in patients with glucokinase deficiency. Five subjects with a missense mutation (Glu256Lys) were studied. They were compared with six healthy subjects with low insulin response but normal glucose tolerance. Insulin and glucagon levels were measured at blood glucose 7.1 +/- 0.1 mmol/l and at 10.9 +/- 0.2 mmol/l with or without arginine (5 g i.v.). Insulin sensitivity was assessed as the ratio between infused glucose and the insulin level (M:I) during hyperglycemic clamps. Glu256Lys subjects were nonobese and had fasting blood glucose 6.7 +/- 0.1 mmol/l (P < 0.001 vs. control group). Insulin release was reduced in response to 11 mmol/l glucose (61% of control group, P < 0.05) as well as to arginine in the presence of 11 mmol/l glucose (54% of control group, P < 0.01). Also, the slope of potentiation, i.e., the enhancement of arginine-induced release as a function of prevailing glucose concentration, was reduced (delta insulin/delta glucose, 47% of control group, P < 0.05). As for glucagon release, the response to arginine was not inhibited normally by glucose, resulting in threefold higher levels at 11 mmol/l glucose versus control subjects. Insulin sensitivity, assessed as M:I, was significantly (P < 0.05) reduced (55% of control group). Glucokinase deficiency thus affects not only insulin responses to glucose per se but also glucose potentiation of responses to non-nutrient secretagogues. Abnormalities in glucagon release and insulin sensitivity coexist with attenuated insulin responses in glucokinase-deficient subjects.


Subject(s)
Arginine/pharmacology , Glucokinase/deficiency , Glucose/pharmacology , Insulin/metabolism , Lysine/genetics , Mutation , Adult , Blood Glucose/metabolism , Drug Synergism , Female , Glucagon/metabolism , Glucokinase/genetics , Glucose Tolerance Test , Glutamic Acid/genetics , Humans , Insulin Secretion , Male , Middle Aged
14.
Diabetes ; 52(11): 2854-60, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14578306

ABSTRACT

Neonatal diabetes can be either permanent or transient. We have recently shown that permanent neonatal diabetes can result from complete deficiency of glucokinase activity. Here we report three new cases of glucokinase-related permanent neonatal diabetes. The probands had intrauterine growth retardation (birth weight <1,900 g) and insulin-treated diabetes from birth (diagnosis within the first week of life). One of the subjects was homozygous for the missense mutation Ala378Val (A378V), which is an inactivating mutation with an activity index of only 0.2% of wild-type glucokinase activity. The second subject was homozygous for a mutation in the splice donor site of exon 8 (intervening sequence 8 [IVS8] + 2T-->G), which is predicted to lead to the synthesis of an inactive protein. The third subject (second cousin of subject 2) was a compound heterozygote with one allele having the splice-site mutation IVS8 + 2T-->G and the other the missense mutation Gly264Ser (G264S), a mutation with an activity index of 86% of normal activity. The five subjects with permanent neonatal diabetes due to glucokinase deficiency identified to date are characterized by intrauterine growth retardation, permanent insulin-requiring diabetes from the first day of life, and hyperglycemia in both parents. Autosomal recessive inheritance and enzyme deficiency are features typical for an inborn error of metabolism, which occurred in the glucose-insulin signaling pathway in these subjects.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/enzymology , Diabetes Mellitus/congenital , Diabetes Mellitus/genetics , Glucokinase/deficiency , Glucokinase/genetics , Insulin/physiology , Base Sequence , Carbohydrate Metabolism, Inborn Errors/genetics , DNA Primers , Exons , Female , Genetic Markers , Glucokinase/chemistry , Glucokinase/metabolism , Glucose/physiology , Humans , Infant, Newborn , Introns , Kinetics , Male , Models, Molecular , Mutation , Pedigree , Polymerase Chain Reaction , Promoter Regions, Genetic , Protein Conformation , Signal Transduction/genetics , White People
15.
Diabetes ; 49(12): 2056-62, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11118007

ABSTRACT

Type 2 diabetes is characterized by impaired beta-cell function, hyperglycemia, and islet amyloid deposition. The primary constituent of islet amyloid is the 37-amino acid beta-cell product called islet amyloid polypeptide (IAPP) or amylin. To study mechanisms of islet amyloid formation, we developed a transgenic mouse model that produces and secretes the amyloidogenic human IAPP (hIAPP) molecule and have shown that 81% of male transgenic mice develop islet amyloid after 14 months on a high-fat diet. To test whether impaired beta-cell function and hyperglycemia could enhance islet amyloid formation, we cross-bred our hIAPP transgenic mice with beta-cell glucokinase-knockout mice (GKKO) that have impaired glucose-mediated insulin secretion and fasting hyperglycemia. The resulting new (hIAPPxGKKO) line of mice had higher basal plasma glucose concentrations than the hIAPP transgenic mice at 3, 6, and 12 months of age (P < 0.05), as did GKKO mice compared with hIAPP transgenic mice at 6 and 12 months of age (P < 0.05). Basal plasma immunoreactive insulin (IRI) levels were lower in hIAPP x GKKO mice than in hIAPP transgenic mice at 6 months of age (P < 0.05). The area under the glucose curve in response to an intraperitoneal glucose challenge (1 g/kg body weight) was larger in hIAPPxGKKO mice than in hIAPP transgenic mice at 3, 6, and 12 months of age (P < 0.005) and in GKKO mice compared with hIAPP transgenic mice at 6 and 12 months of age (P < 0.005). The area under the IRI curve was lower in hIAPPxGKKO mice at 6 and 12 months of age (P < 0.05) than in hIAPP transgenic mice and in GKKO mice compared with hIAPP transgenic mice at 12 months of age (P < 0.05). Despite the presence of hyperglycemia, hIAPPxGKKO mice had a lower incidence (4 of 17 vs. 12 of 19, P < 0.05) and amount (0.40 +/- 0.24 vs. 1.2 +/- 0.3 arbitrary units, P < 0.05) of islet amyloid than hIAPP transgenic mice had. As expected, no islet amyloid was observed in GKKO mice lacking the hIAPP transgene (0 of 13). There was no difference in pancreatic content of IRI and hIAPP among the three groups of mice. Thus, despite the presence of impaired islet function and hyperglycemia, hIAPPxGKKO mice had a decreased incidence and quantity of islet amyloid. Therefore, our data suggest that impaired beta-cell glucose metabolism or hyperglycemia are not likely to contribute to islet amyloid formation in diabetes. Furthermore, this finding may explain the lack of progression of glycemia in patients with maturity-onset diabetes of the young.


Subject(s)
Amyloid/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Glucokinase/deficiency , Hyperglycemia/etiology , Islets of Langerhans/metabolism , Aging/metabolism , Amyloid/blood , Amyloid/genetics , Animals , Body Weight , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Glucokinase/genetics , Glucose Tolerance Test , Humans , Injections, Intraperitoneal , Islet Amyloid Polypeptide , Mice , Mice, Transgenic/genetics , Osmolar Concentration
16.
Chem Biol ; 4(4): 249-57, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9235288

ABSTRACT

The beta-cells of the pancreas control the blood levels of glucose and other nutrients by secreting insulin. They sense blood nutrient levels not by using a classical receptor-signaling system, but by detecting the products of nutrient metabolism. Mutations in this pathway can cause diabetes or hypoglycemia.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Amino Acids/metabolism , Calcium/metabolism , Diabetes Mellitus/metabolism , Energy Metabolism , Glucokinase/deficiency , Glucokinase/metabolism , Glucose/metabolism , Homeostasis , Humans , Insulin Secretion , Lipid Metabolism , Potassium Channels/metabolism , Signal Transduction
17.
Diabetes Care ; 17(9): 1015-21, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7988299

ABSTRACT

OBJECTIVE: In eight glucokinase (GCK)-deficient subjects, we have investigated insulin secretion rates (ISRs) in response to intravenous arginine. Impairment in the enzymatic activity of mutant GCK leads to a reduced glycolytic flux in beta-cells. This defect translates in vivo as a right shift in the glucose/SR dose-response curve. Insulin secretion in response to other secretagogues has not been reported. RESEARCH DESIGN AND METHODS: The arginine test was performed immediately after a 2-h hyperglycemic (10 mM) clamp. ISR was computed by deconvolution of peripheral C-peptide levels. Linear regression analyses were performed to assess correlations between the beta-cell secretory responses to the arginine test, an intravenous glucose tolerance test (IVGTT), and a hyperglycemic clamp (areas under the C-peptide curves), and between these parameters and the glucose tolerance status (area under the glucose curve during an oral glucose tolerance test). RESULTS: Two minutes after the injection of arginine, the increment in ISR was 30.17 +/- 10.01 pmol insulin.kg-1.min-1 in patients and 36.25 +/- 15.46 pmol insulin.kg-1.min-1 in control subjects (P = 0.38). Throughout the experiment, increments in ISR were comparable in both groups. The amount of insulin secreted in response to arginine (0-5 min) was similar in patients and control subjects: 81 +/- 28 vs. 119 +/- 55 pmol/kg (P = 0.16), respectively. The arginine test C-peptide response was not correlated with the IVGTT or hyperglycemic clamp responses. The arginine test and hyperglycemic clamp responses were not correlated to the glucose tolerance status. The best predictor of the glucose tolerance was the C-peptide response to the IVGTT (r2 = 0.78; P = 0.002). CONCLUSIONS: beta-cell secretory increment in response to arginine was found to be in the normal range in GCK-deficient subjects. The arginine test does not seem to reflect either the beta-cell secretory defect or the glucose tolerance status of these subjects. IVGTT seems to be the best predictor of the latter parameter in this population.


Subject(s)
Arginine/pharmacology , Glucokinase/deficiency , Insulin/metabolism , Adolescent , Adult , Arginine/administration & dosage , C-Peptide/blood , Dose-Response Relationship, Drug , Female , Glucose/administration & dosage , Glucose/pharmacology , Glucose Clamp Technique , Glucose Tolerance Test , Humans , Hyperglycemia/blood , Hyperglycemia/enzymology , Infusions, Intravenous , Insulin/blood , Islets of Langerhans/metabolism , Male , Middle Aged , Radioimmunoassay , Regression Analysis
18.
Diabetes Care ; 19(9): 915-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8875082

ABSTRACT

OBJECTIVE: To assess the prevalence of diabetes complications and the severity of diabetes in kindreds with NIDDM linked to the MODY3 locus (chromosome 12q) and to compare these parameters with data obtained in glucokinase (GCK)-deficient and other-MODY (unlinked to any of the three known loci) families, as well as with data from families with a late age of onset of NIDDM. RESEARCH DESIGN AND METHODS: Clinical and biological data were obtained from 667 affected members of 7 MODY3, 25 GCK-deficient, 6 other-MODY, and 81 NIDDM families. Severity of diabetes (glucose tolerance status and insulin secretion) was assessed by an oral glucose tolerance test. Neurological examination and eye fundus examination were performed in 349 and 251 subjects, respectively, and proteinuria was tested with strips in 282 family members. RESULTS: A higher prevalence of proliferative retinopathy was observed in MODY3 (21%) and NIDDM subjects (23%) than in GCK-deficient (3%) and other-MODY subjects (8%; P = 0.004). Proteinuria was detected in 19, 7, 5, and 0% (P = 0.07) of subjects, respectively. Prevalence of neuropathy was higher in NIDDM (17%; P = 0.005) than in MODY3 (4%), GCK-deficient (5%) and other-MODY (0%) subjects. MODY3 and NIDDM subjects had significantly higher fasting glucose levels than subjects in the other groups. Glucose levels after 2 h were significantly higher, and the ratios of insulin to glucose levels were significantly lower in MODY3 subjects than in the other three groups. CONCLUSIONS: The MODY3 subtype of NIDDM is characterized by a severe insulin secretory defect and by major hyperglycemia that progresses rapidly to overt diabetes. Microvascular complications of diabetes were frequently observed in the MODY3 subjects and the subjects with a late age of onset of NIDDM in this cohort. Both the duration and the severity of diabetes were independently associated with these complications.


Subject(s)
Chromosomes, Human, Pair 12 , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Glucokinase/deficiency , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Analysis of Variance , Blood Glucose/metabolism , Blood Pressure , Child , Chromosome Mapping , Diabetic Neuropathies/epidemiology , Diabetic Neuropathies/genetics , Diabetic Retinopathy/epidemiology , Diabetic Retinopathy/genetics , Female , Genetic Linkage , Glucokinase/genetics , Humans , Male , Middle Aged , Prevalence , Proteinuria/epidemiology , Proteinuria/genetics
19.
Eur J Endocrinol ; 141(6): 609-18, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10601964

ABSTRACT

OBJECTIVE: To evaluate insulin secretion and sensitivity in affected (diabetes mellitus or impaired glucose tolerance; n=7) and in unaffected (normal glucose tolerance; n=3) carriers of hepatocyte nuclear factor-1alpha (maturity-onset diabetes of the young-3 (MODY3)) gene mutations. METHODS: Insulin secretion was assessed by an i.v. glucose tolerance test (IVGTT), hyperglycemic clamp and arginine test, and insulin sensitivity by an euglycemic hyperinsulinemic clamp. Results were compared with those of diabetic MODY2 (glucokinase-deficient) and control subjects. RESULTS: The amount of insulin secreted during an IVGTT was decreased in affected MODY3 subjects (46+/-24 (s.d.) pmol/kg body weight (BW)) as compared with values in MODY2 (120+/-49pmol/kg BW) and control (173+/-37pmol/kg BW; P=0.0004) subjects. The amount of insulin secreted during a 10mmol/l glucose clamp was decreased in affected MODY3 subjects (171+/-78pmol/kg BW) and MODY2 subjects (302+/-104pmol/kg BW) as compared with control subjects (770+/-199pmol/kg BW; P=0.0001). Insulin secretion in response to arginine was decreased in affected MODY3 subjects. Milder and heterogeneous defects were observed in the unaffected MODY3 subjects; the amount of insulin secreted during the hyperglycemic clamp was 40-79% of that of controls. The response to arginine was abnormally delayed. Insulin sensitivity was decreased in diabetic but not in non-diabetic MODY3 subjects. CONCLUSIONS: Beta-cell dysfunction in response to glucose and arginine is observed in affected and unaffected MODY3 subjects. The MODY3 and MODY2 subtypes present different insulin secretion profiles. Secondary insulin resistance might contribute to the chronic hyperglycemia of MODY3 patients and modulate their glucose tolerance.


Subject(s)
DNA-Binding Proteins , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Insulin/metabolism , Insulin/pharmacology , Mutation , Nuclear Proteins , Transcription Factors/genetics , Adolescent , Adult , Arginine , Female , Glucokinase/deficiency , Glucose Clamp Technique , Glucose Tolerance Test , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Hepatocyte Nuclear Factor 1-beta , Humans , Insulin Secretion , Islets of Langerhans/physiopathology , Kinetics , Male , Middle Aged
20.
FEMS Microbiol Lett ; 188(2): 209-15, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10913707

ABSTRACT

The glk gene from Corynebacterium glutamicum was isolated by complementation using Escherichia coli ZSC113 (ptsG ptsM glk). We sequenced a total of 3072 bp containing the 969-bp open reading frame encoding glucose kinase (Glk). The glk gene has a deduced molecular mass of 34.2 kDa and contains a typical ATP binding site. Comparison with protein sequences revealed homologies to Glk from Streptomyces coelicolor (43%) and Bacillus megaterium (35%). The glk gene in C. glutamicum was inactivated on the chromosome via single crossover homologous recombination and the resulting glk mutant was characterized. Interestingly, the C. glutamicum glk mutant showed poor growth on rich medium such as LB medium or brain heart infusion medium in the presence or absence of glucose, fructose, maltose or sucrose as the sole carbon source. Growth yield was reduced significantly when maltose was used as the sole carbon source using minimal medium. The growth defect of glk mutant on rich medium was complemented by a plasmid-encoded glk gene. A chromosomal glk-lacZ fusion was constructed and used to monitor glk expression, and it was found that glk was expressed constitutively under all tested conditions with different carbon sources.


Subject(s)
Corynebacterium/genetics , Genes, Bacterial , Glucokinase/genetics , Amino Acid Sequence , Binding Sites , Carbohydrates , Cloning, Molecular , Corynebacterium/enzymology , Corynebacterium/growth & development , Culture Media , Electroporation , Escherichia coli , Genetic Vectors , Glucokinase/deficiency , Glucokinase/metabolism , Lac Operon , Molecular Sequence Data , Mutation , Plasmids/genetics , Sequence Alignment , Transformation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL