Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 286(45): 39560-72, 2011 Nov 11.
Article in English | MEDLINE | ID: mdl-21921030

ABSTRACT

We performed genome-wide mutagenesis in C57BL/6J mice using N-ethyl-N-nitrosourea to identify mutations causing high blood glucose early in life and to produce new animal models of diabetes. Of a total of 13 new lines confirmed by heritability testing, we identified two semi-dominant pedigrees with novel missense mutations (Gck(K140E) and Gck(P417R)) in the gene encoding glucokinase (Gck), the mammalian glucose sensor that is mutated in human maturity onset diabetes of the young type 2 and the target of emerging anti-hyperglycemic agents that function as glucokinase activators (GKAs). Diabetes phenotype corresponded with genotype (mild-to-severe: Gck(+/+) < Gck(P417R/+), Gck(K140E)(/+) < Gck(P417R/P417R), Gck(P417R/K140E), and Gck(K140E/K140E)) and with the level of expression of GCK in liver. Each mutant was produced as the recombinant enzyme in Escherichia coli, and analysis of k(cat) and tryptophan fluorescence (I(320/360)) during thermal shift unfolding revealed a correlation between thermostability and the severity of hyperglycemia in the whole animal. Disruption of the glucokinase regulatory protein-binding site (GCK(K140E)), but not the ATP binding cassette (GCK(P417R)), prevented inhibition of enzyme activity by glucokinase regulatory protein and corresponded with reduced responsiveness to the GKA drug. Surprisingly, extracts from liver of diabetic GCK mutants inhibited activity of the recombinant enzyme, a property that was also observed in liver extracts from mice with streptozotocin-induced diabetes. These results indicate a relationship between genotype, phenotype, and GKA efficacy. The integration of forward genetic screening and biochemical profiling opens a pathway for preclinical development of mechanism-based diabetes therapies.


Subject(s)
Alkylating Agents/adverse effects , Diabetes Mellitus, Experimental , Enzyme Activators/metabolism , Ethylnitrosourea/adverse effects , Glucokinase , Liver/enzymology , Mutation, Missense , Alkylating Agents/pharmacology , Amino Acid Substitution , Animals , Binding Sites/genetics , Blood Glucose/genetics , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Ethylnitrosourea/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Glucokinase/antagonists & inhibitors , Glucokinase/biosynthesis , Glucokinase/genetics , Humans , Hyperglycemia/chemically induced , Hyperglycemia/enzymology , Hyperglycemia/genetics , Liver/pathology , Male , Mice , Mice, Mutant Strains , Organ Specificity , Protein Folding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
2.
Biochem J ; 440(2): 203-15, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21831042

ABSTRACT

GK (glucokinase) is activated by glucose binding to its substrate site, is inhibited by GKRP (GK regulatory protein) and stimulated by GKAs (GK activator drugs). To explore further the mechanisms of these processes we studied pure recombinant human GK (normal enzyme and a selection of 31 mutants) using steady-state kinetics of the enzyme and TF (tryptophan fluorescence). TF studies of the normal binary GK-glucose complex corroborate recent crystallography studies showing that it exists in a closed conformation greatly different from the open conformation of the ligand-free structure, but indistinguishable from the ternary GK-glucose-GKA complex. GKAs did activate and GKRP did inhibit normal GK, whereas its TF was doubled by glucose saturation. However, the enzyme kinetics, GKRP inhibition, TF enhancement by glucose and responsiveness to GKA of the selected mutants varied greatly. Two predominant response patterns were identified accounting for nearly all mutants: (i) GK mutants with a normal or close to normal response to GKA, normally low basal TF (indicating an open conformation), some variability of kinetic parameters (k(cat), glucose S(0.5), h and ATP K(m)), but usually strong GKRP inhibition (13/31); and (ii) GK mutants that are refractory to GKAs, exhibit relatively high basal TF (indicating structural compaction and partial closure), usually show strongly enhanced catalytic activity primarily due to lowering of the glucose S(0.5), but with reduced or no GKRP inhibition in most cases (14/31). These results and those of previous studies are best explained by envisioning a common allosteric regulator region with spatially non-overlapping GKRP- and GKA-binding sites.


Subject(s)
Allosteric Regulation , Glucokinase/metabolism , Carrier Proteins , Fluorescence , Glucokinase/antagonists & inhibitors , Glucokinase/genetics , Glucose/pharmacology , Humans , Kinetics , Point Mutation , Protein Conformation , Tryptophan/chemistry
3.
Diabetes Care ; 33(2): 290-2, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19903754

ABSTRACT

OBJECTIVE: To evaluate the heterogeneity in the clinical expression in a family with glucokinase mature-onset diabetes of the young (GCK-MODY). RESEARCH DESIGN AND METHODS: Members (three generations) of the same family presented either with overt neonatal hyperglycemia, marked postprandial hyperglycemia, or glucosuria. Homeostasis model assessment of insulin resistance (HOMA(IR)) and insulinogenic and disposition indexes were calculated. Oral glucose tolerance test (OGTT) results in the GCK mutation carriers from this family were compared with those from other subjects with GCK mutations in the same codon (GCK(261)), with other missense and other types of GCK mutations in different codons from the European MODY Consortium database (GCK(m)). RESULTS: Mutation G261R was found in the GCK gene. During the OGTT, glucose (P = 0.02) and insulin (P = 0.009) response at 2 h as well as at the 2-h glucose increment (GCK(261) versus other missense GCK mutations, P = 0.003) were significantly higher in GCK(261) than in GCK(m) carriers. CONCLUSIONS: Differing from other GCK(m) carriers, the glucose and insulin response to oral glucose was significantly higher in GCK(261) carriers, indicating clinical heterogeneity in GCK-MODY.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucokinase/genetics , Mutation , Adult , Blood Glucose/metabolism , Codon/genetics , Family , Female , Glucose Tolerance Test , Humans , Male , Young Adult
4.
Diabetes ; 58(6): 1419-27, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19336674

ABSTRACT

OBJECTIVE: Heterozygous activating mutations of glucokinase have been reported to cause hypoglycemia attributable to hyperinsulinism in a limited number of families. We report three children with de novo glucokinase hyperinsulinism mutations who displayed a spectrum of clinical phenotypes corresponding to marked differences in enzyme kinetics. RESEARCH DESIGN AND METHODS: Mutations were directly sequenced, and mutants were expressed as glutathionyl S-transferase-glucokinase fusion proteins. Kinetic analysis of the enzymes included determinations of stability, activity index, the response to glucokinase activator drug, and the effect of glucokinase regulatory protein. RESULTS: Child 1 had an ins454A mutation, child 2 a W99L mutation, and child 3 an M197I mutation. Diazoxide treatment was effective in child 3 but ineffective in child 1 and only partially effective in child 2. Expression of the mutant glucokinase ins454A, W99L, and M197I enzymes revealed a continuum of high relative activity indexes in the three children (26, 8.9, and 3.1, respectively; wild type = 1.0). Allosteric responses to inhibition by glucokinase regulatory protein and activation by the drug RO0281675 were impaired by the ins454A but unaffected by the M197I mutation. Estimated thresholds for glucose-stimulated insulin release were more severely reduced by the ins454A than the M197I mutation and intermediate in the W99L mutation (1.1, 3.5, and 2.2 mmol/l, respectively; wild type = 5.0 mmol/l). CONCLUSIONS: These results confirm the potency of glucokinase as the pancreatic beta-cell glucose sensor, and they demonstrate that responsiveness to diazoxide varies with genotype in glucokinase hyperinsulinism resulting in hypoglycemia, which can be more difficult to control than previously believed.


Subject(s)
Diazoxide/therapeutic use , Glucokinase/genetics , Hyperinsulinism/enzymology , Hyperinsulinism/genetics , Mutation , Adolescent , Amino Acid Substitution , Birth Weight , Blood Glucose/metabolism , Child , Circadian Rhythm , DNA Transposable Elements , Glucokinase/metabolism , Humans , Hyperinsulinism/drug therapy , Insulin/metabolism , Insulin Secretion , Kinetics , Male , Phenotype , Recombinant Proteins/metabolism
5.
Biochem J ; 413(2): 269-80, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18370929

ABSTRACT

Tryptophan fluorescence was used to study GK (glucokinase), an enzyme that plays a prominent role in glucose homoeostasis which, when inactivated or activated by mutations, causes diabetes mellitus or hypoglycaemia in humans. GK has three tryptophan residues, and binding of D-glucose increases their fluorescence. To assess the contribution of individual tryptophan residues to this effect, we generated GST-GK [GK conjugated to GST (glutathione transferase)] and also pure GK with one, two or three of the tryptophan residues of GK replaced with other amino acids (i.e. W99C, W99R, W167A, W167F, W257F, W99R/W167F, W99R/W257F, W167F/W257F and W99R/W167F/W257F). Enzyme kinetics, binding constants for glucose and several other sugars and fluorescence quantum yields (varphi) were determined and compared with those of wild-type GK retaining its three tryptophan residues. Replacement of all three tryptophan residues resulted in an enzyme that retained all characteristic features of GK, thereby demonstrating the unique usefulness of tryptophan fluorescence as an indicator of GK conformation. Curves of glucose binding to wild-type and mutant GK or GST-GK were hyperbolic, whereas catalysis of wild-type and most mutants exhibited co-operativity with D-glucose. Binding studies showed the following order of affinities for the enzyme variants: N-acetyl-D-glucosamine>D-glucose>D-mannose>D-mannoheptulose>2-deoxy-D-glucose>>L-glucose. GK activators increased sugar binding of most enzymes, but not of the mutants Y214A/V452A and C252Y. Contributions to the fluorescence increase from Trp(99) and Trp(167) were large compared with that from Trp(257) and are probably based on distinct mechanisms. The average quantum efficiency of tryptophan fluorescence in the basal and glucose-bound state was modified by activating (Y214A/V452A) or inactivating (C213R and C252Y) mutations and was interpreted as a manifestation of distinct conformational states.


Subject(s)
Glucokinase/genetics , Mutation , Recombinant Proteins/chemistry , Tryptophan/chemistry , Binding Sites , Carbohydrates/chemistry , Escherichia coli/metabolism , Glutathione Transferase/metabolism , Humans , Kinetics , Molecular Conformation , Protein Binding , Protein Conformation , Recombinant Fusion Proteins/chemistry , Spectrometry, Fluorescence/methods
6.
J Biol Chem ; 282(18): 13906-16, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17353190

ABSTRACT

To better understand how glucokinase (GK) missense mutations associated with human glycemic diseases perturb glucose homeostasis, we generated and characterized mice with either an activating (A456V) or inactivating (K414E) mutation in the gk gene. Animals with these mutations exhibited alterations in their blood glucose concentration that were inversely related to the relative activity index of GK. Moreover, the threshold for glucose-stimulated insulin secretion from islets with either the activating or inactivating mutation were left- or right-shifted, respectively. However, we were surprised to find that mice with the activating mutation had markedly reduced amounts of hepatic GK activity. Further studies of bacterially expressed mutant enzymes revealed that GK(A456V) is as stable as the wild type enzyme, whereas GK(K414E) is thermolabile. However, the ability of GK regulatory protein to inhibit GK(A456V) was found to be less than that of the wild type enzyme, a finding consistent with impaired hepatic nuclear localization. Taken together, this study indicates that it is necessary to have knowledge of both thermolability and the interactions of mutant GK enzymes with GK regulatory protein when attempting to predict in vivo glycemic phenotypes based on the measurement of enzyme kinetics.


Subject(s)
Blood Glucose/metabolism , Carrier Proteins/metabolism , Glucokinase/metabolism , Glucose Metabolism Disorders/enzymology , Liver/enzymology , Mutation, Missense , Adaptor Proteins, Signal Transducing , Amino Acid Substitution , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Enzyme Activation/genetics , Enzyme Stability/genetics , Glucokinase/chemistry , Glucokinase/genetics , Glucose Metabolism Disorders/genetics , Glucose Metabolism Disorders/pathology , Hot Temperature , Insulin/metabolism , Insulin Secretion , Intracellular Signaling Peptides and Proteins , Liver/pathology , Mice , Mice, Mutant Strains , Mice, Transgenic , Phenotype , Protein Binding/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Diabetes ; 55(6): 1713-22, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16731834

ABSTRACT

Glucokinase functions as a glucose sensor in pancreatic beta-cells and regulates hepatic glucose metabolism. A total of 83 probands were referred for a diagnostic screening of mutations in the glucokinase (GCK) gene. We found 11 different mutations (V62A, G72R, L146R, A208T, M210K, Y215X, S263P, E339G, R377C, S453L, and IVS5 + 1G>C) in 14 probands. Functional characterization of recombinant glutathionyl S-transferase-G72R glucokinase showed slightly increased activity, whereas S263P and G264S had near-normal activity. The other point mutations were inactivating. S263P showed marked thermal instability, whereas the stability of G72R and G264S differed only slightly from that of wild type. G72R and M210K did not respond to an allosteric glucokinase activator (GKA) or the hepatic glucokinase regulatory protein (GKRP). Mutation analysis of the role of glycine at position 72 by substituting E, F, K, M, S, or Q showed that G is unique since all these mutants had very low or no activity and were refractory to GKRP and GKA. Structural analysis provided plausible explanations for the drug resistance of G72R and M210K. Our study provides further evidence that protein instability in combination with loss of control by a putative endogenous activator and GKRP could be involved in the development of hyperglycemia in maturity-onset diabetes of the young, type 2. Furthermore, based on data obtained on G264S, we propose that other and still unknown mechanisms participate in the regulation of glucokinase.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Glucokinase/metabolism , Mutant Proteins/metabolism , Mutation , Binding Sites , Blood Glucose/metabolism , Carrier Proteins/metabolism , Crystallography, X-Ray , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/prevention & control , Enzyme Stability/drug effects , Genetic Testing , Glucokinase/chemistry , Glucokinase/genetics , Glucose/pharmacology , Humans , Hyperglycemia/enzymology , Hyperglycemia/genetics , Hyperglycemia/metabolism , Kinetics , Mutant Proteins/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
8.
J Biol Chem ; 280(14): 14105-13, 2005 Apr 08.
Article in English | MEDLINE | ID: mdl-15677479

ABSTRACT

Glucokinase (GCK) serves as the pancreatic glucose sensor. Heterozygous inactivating GCK mutations cause hyperglycemia, whereas activating mutations cause hypoglycemia. We studied the GCK V62M mutation identified in two families and co-segregating with hyperglycemia to understand how this mutation resulted in reduced function. Structural modeling locates the mutation close to five naturally occurring activating mutations in the allosteric activator site of the enzyme. Recombinant glutathionyl S-transferase-V62M GCK is paradoxically activated rather than inactivated due to a decreased S0.5 for glucose compared with wild type (4.88 versus 7.55 mM). The recently described pharmacological activator (RO0281675) interacts with GCK at this site. V62M GCK does not respond to RO0281675, nor does it respond to the hepatic glucokinase regulatory protein (GKRP). The enzyme is also thermally unstable, but this lability is apparently less pronounced than in the proven instability mutant E300K. Functional and structural analysis of seven amino acid substitutions at residue Val62 has identified a non-linear relationship between activation by the pharmacological activator and the van der Waals interactions energies. Smaller energies allow a hydrophobic interaction between the activator and glucokinase, whereas larger energies prohibit the ligand from fitting into the binding pocket. We conclude that V62M may cause hyperglycemia by a complex defect of GCK regulation involving instability in combination with loss of control by a putative endogenous activator and/or GKRP. This study illustrates that mutations that cause hyperglycemia are not necessarily kinetically inactivating but may exert their effects by other complex mechanisms. Elucidating such mechanisms leads to a deeper understanding of the GCK glucose sensor and the biochemistry of beta-cells and hepatocytes.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Glucokinase/genetics , Glucokinase/metabolism , Point Mutation , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , Carrier Proteins/metabolism , Child , DNA Mutational Analysis , Enzyme Activation , Enzyme Stability , Female , Glucose/metabolism , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Infant, Newborn , Male , Models, Molecular , Pedigree , Pregnancy , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
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
10.
Diabetes ; 51(4): 1240-6, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11916951

ABSTRACT

In this study, a second case of hyperinsulinemic hypoglycemia due to activation of glucokinase is reported. The 14-year-old proband had a history of neonatal hypoglycemia, treated with diazoxide. He was admitted with coma and convulsions due to nonketotic hypoglycemia. His BMI was 34 kg/m(2), and his fasting blood glucose ranged from 2.1 to 2.7 mmol/l, associated with inappropriately high serum levels of insulin, C-peptide, and proinsulin. An oral glucose tolerance test (OGTT) showed exaggerated responses of these peptides followed by profound hypoglycemia. Treatment with diazoxide and chlorothiazide was effective. His mother never had clinical hypoglycemic symptoms, even though her fasting blood glucose ranged from 2.9 to 3.5 mmol/l. Increases in serum insulin, C-peptide, and proinsulin in response to an OGTT suggested a lower threshold for glucose-stimulated insulin release (GSIR). Screening for mutations in candidate genes revealed a heterozygous glucokinase mutation in exon 10, substituting valine for alanine at codon 456 (A456V) in the proband and his mother. The purified recombinant glutathionyl S-transferase fusion protein of the A456V glucokinase revealed a decreased glucose S(0.5) (the concentration of glucose needed to achieve the half-maximal rate of phosphorylation) from 8.04 (wild-type) to 2.53 mmol/l. The mutant's Hill coefficient was decreased, and its maximal specific activity k(cat) was increased. Mathematical modeling predicted a markedly lowered GSIR threshold of 1.5 mmol/l. The theoretical and practical implications are manifold and significant.


Subject(s)
Diabetes Mellitus/genetics , Glucokinase/genetics , Glucose/metabolism , Mutation, Missense , Adolescent , Allosteric Regulation , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Diazoxide/therapeutic use , Enzyme Activation , Glutathione Transferase/genetics , Homeostasis , Humans , Infant, Newborn , Infant, Small for Gestational Age , Insulin/blood , Insulin/metabolism , Insulin Secretion , Kinetics , Male , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...