Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Diabetes Technol Ther ; 24(10): 737-748, 2022 10.
Article in English | MEDLINE | ID: mdl-35758724

ABSTRACT

Repeated hypoglycemia exposure leads to impaired awareness of hypoglycemia (IAH) and the development of defective counterregulatory responses. To date, only pancreas or islet transplantation has demonstrated normalization of hypoglycemia awareness and the endogenous glucose production (EGP) response to defend against insulin-induced hypoglycemia in long-standing type 1 diabetes (T1D). This study aims to validate clinical metrics of IAH (Clarke score), hypoglycemia severity (HYPO score), glycemic lability (lability index), and continuous glucose monitoring (CGM) as predictors of absent autonomic symptom (AS) recognition and defective glucose counterregulation during insulin-induced hypoglycemia, thus enabling early identification of individuals with compromised physiologic defense against clinically significant hypoglycemia. Forty-three subjects with mean ± standard deviation age 43 ± 13 years and T1D duration 28 ± 13 years, including 32 with IAH and 11 with hypoglycemia awareness (Aware), and 12 nondiabetic control subjects, underwent single-blinded randomized-paired hyperinsulinemic-euglycemic and hypoglycemic clamp experiments. Receiver operating characteristic (ROC) curves and sensitivity analyses were performed to assess metric prediction of absent AS recognition and defective EGP responses to hypoglycemia. Clarke score and CGM measures of hypoglycemia exposure demonstrated good ability to predict absent AS recognition (area under the curve ≥0.80). A composite threshold of IAH-Clarke ≥4 with ROC curve-derived thresholds for CGM measures of hypoglycemia exposure showed high specificity and predictive value in identifying an absent AS response during the hypoglycemic clamp. Metrics demonstrated poor ability to predict defective glucose counterregulation by the EGP response, which was impaired even in the Aware group. Screening for IAH alongside assessment of CGM data can increase the specificity for identifying individuals with absent hypoglycemia symptom recognition who may benefit from further intervention.


Subject(s)
Diabetes Mellitus, Type 1 , Hypoglycemia , Insulins , Adult , Benchmarking , Blood Glucose , Blood Glucose Self-Monitoring , Diabetes Mellitus, Type 1/complications , Glucose , Humans , Hypoglycemia/diagnosis , Hypoglycemia/etiology , Hypoglycemic Agents/adverse effects , Insulin , Middle Aged
2.
Am J Physiol Endocrinol Metab ; 302(1): E87-E102, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21952036

ABSTRACT

It was reported previously that isolated human islets from individuals with type 2 diabetes mellitus (T2DM) show reduced glucose-stimulated insulin release. To assess the possibility that impaired bioenergetics may contribute to this defect, glucose-stimulated respiration (Vo(2)), glucose usage and oxidation, intracellular Ca(2+), and insulin secretion (IS) were measured in pancreatic islets isolated from three healthy and three type 2 diabetic organ donors. Isolated mouse and rat islets were studied for comparison. Islets were exposed to a "staircase" glucose stimulus, whereas IR and Vo(2) were measured. Vo(2) of human islets from normals and diabetics increased sigmoidally from equal baselines of 0.25 nmol/100 islets/min as a function of glucose concentration. Maximal Vo(2) of normal islets at 24 mM glucose was 0.40 ± 0.02 nmol·min(-1)·100 islets(-1), and the glucose S(0.5) was 4.39 ± 0.10 mM. The glucose stimulation of respiration of islets from diabetics was lower, V(max) of 0.32 ± 0.01 nmol·min(-1)·100 islets(-1), and the S(0.5) shifted to 5.43 ± 0.13 mM. Glucose-stimulated IS and the rise of intracellular Ca(2+) were also reduced in diabetic islets. A clinically effective glucokinase activator normalized the defective Vo(2), IR, and free calcium responses during glucose stimulation in islets from type 2 diabetics. The body of data shows that there is a clear relationship between the pancreatic islet energy (ATP) production rate and IS. This relationship was similar for normal human, mouse, and rat islets and the data for all species fitted a single sigmoidal curve. The shared threshold rate for IS was ∼13 pmol·min(-1)·islet(-1). Exendin-4, a GLP-1 analog, shifted the ATP production-IS curve to the left and greatly potentiated IS with an ATP production rate threshold of ∼10 pmol·min(-1)·islet(-1). Our data suggest that impaired ß-cell bioenergetics resulting in greatly reduced ATP production is critical in the molecular pathogenesis of type 2 diabetes mellitus.


Subject(s)
Benzeneacetamides/pharmacology , Diabetes Mellitus, Type 2/metabolism , Enzyme Activators/pharmacology , Glucokinase/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/drug effects , Adult , Animals , Calcium Signaling/drug effects , Cell Respiration/drug effects , Diabetes Mellitus, Type 2/drug therapy , Exenatide , Female , Glucagon-Like Peptide 1/analogs & derivatives , Glucokinase/chemistry , Glycolysis/drug effects , Humans , Hypoglycemic Agents/pharmacology , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mice , Middle Aged , Oxidative Phosphorylation/drug effects , Peptides/pharmacology , Rats , Species Specificity , Tissue Culture Techniques , Venoms/pharmacology
3.
J Biol Chem ; 285(41): 31806-18, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20670938

ABSTRACT

The mechanism of insulin dysregulation in children with hyperinsulinism associated with inactivating mutations of short-chain 3-hydroxyacyl-CoA dehydrogenase (SCHAD) was examined in mice with a knock-out of the hadh gene (hadh(-/-)). The hadh(-/-) mice had reduced levels of plasma glucose and elevated plasma insulin levels, similar to children with SCHAD deficiency. hadh(-/-) mice were hypersensitive to oral amino acid with decrease of glucose level and elevation of insulin. Hypersensitivity to oral amino acid in hadh(-/-) mice can be explained by abnormal insulin responses to a physiological mixture of amino acids and increased sensitivity to leucine stimulation in isolated perifused islets. Measurement of cytosolic calcium showed normal basal levels and abnormal responses to amino acids in hadh(-/-) islets. Leucine, glutamine, and alanine are responsible for amino acid hypersensitivity in islets. hadh(-/-) islets have lower intracellular glutamate and aspartate levels, and this decrease can be prevented by high glucose. hadh(-/-) islets also have increased [U-(14)C]glutamine oxidation. In contrast, hadh(-/-) mice have similar glucose tolerance and insulin sensitivity compared with controls. Perifused hadh(-/-) islets showed no differences from controls in response to glucose-stimulated insulin secretion, even with addition of either a medium-chain fatty acid (octanoate) or a long-chain fatty acid (palmitate). Pull-down experiments with SCHAD, anti-SCHAD, or anti-GDH antibodies showed protein-protein interactions between SCHAD and GDH. GDH enzyme kinetics of hadh(-/-) islets showed an increase in GDH affinity for its substrate, α-ketoglutarate. These studies indicate that SCHAD deficiency causes hyperinsulinism by activation of GDH via loss of inhibitory regulation of GDH by SCHAD.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/deficiency , Carbohydrate Metabolism, Inborn Errors/enzymology , Glutamate Dehydrogenase/metabolism , Hyperinsulinism/enzymology , Insulin-Secreting Cells/enzymology , Amino Acids/metabolism , Amino Acids/pharmacology , Animals , Blood Glucose/genetics , Blood Glucose/metabolism , Carbohydrate Metabolism, Inborn Errors/genetics , Enzyme Activation/drug effects , Enzyme Activation/genetics , Glutamate Dehydrogenase/genetics , Hyperinsulinism/genetics , Insulin/blood , Ketoglutaric Acids/metabolism , Mice , Mice, Knockout
5.
J Biol Chem ; 283(25): 17238-49, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18445600

ABSTRACT

Pancreatic beta cells are hyper-responsive to amino acids but have decreased glucose sensitivity after deletion of the sulfonylurea receptor 1 (SUR1) both in man and mouse. It was hypothesized that these defects are the consequence of impaired integration of amino acid, glucose, and energy metabolism in beta cells. We used gas chromatography-mass spectrometry methodology to study intermediary metabolism of SUR1 knock-out (SUR1(-/-)) and control mouse islets with d-[U-(13)C]glucose as substrate and related the results to insulin secretion. The levels and isotope labeling of alanine, aspartate, glutamate, glutamine, and gamma-aminobutyric acid (GABA) served as indicators of intermediary metabolism. We found that the GABA shunt of SUR1(-/-) islets is blocked by about 75% and showed that this defect is due to decreased glutamate decarboxylase synthesis, probably caused by elevated free intracellular calcium. Glutaminolysis stimulated by the leucine analogue d,l-beta-2-amino-2-norbornane-carboxylic acid was, however, enhanced in SUR1(-/-) and glyburide-treated SUR1(+/+) islets. Glucose oxidation and pyruvate cycling was increased in SUR1(-/-) islets at low glucose but was the same as in controls at high glucose. Malic enzyme isoforms 1, 2, and 3, involved in pyruvate cycling, were all expressed in islets. High glucose lowered aspartate and stimulated glutamine synthesis similarly in controls and SUR1(-/-) islets. The data suggest that the interruption of the GABA shunt and the lack of glucose regulation of pyruvate cycling may cause the glucose insensitivity of the SUR1(-/-) islets but that enhanced basal pyruvate cycling, lowered GABA shunt flux, and enhanced glutaminolytic capacity may sensitize the beta cells to amino acid stimulation.


Subject(s)
Adenosine Triphosphate/chemistry , Glucose/metabolism , Glutamine/chemistry , Potassium/chemistry , Pyruvates/chemistry , gamma-Aminobutyric Acid/metabolism , Amino Acids/chemistry , Animals , Gas Chromatography-Mass Spectrometry/methods , Genotype , Glutamate Decarboxylase/metabolism , Mice , Mice, Transgenic , Models, Biological , Oxygen/metabolism
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.
Am J Physiol Endocrinol Metab ; 292(6): E1507-19, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17264232

ABSTRACT

Fuel stimulation of insulin secretion from pancreatic beta-cells is thought to be mediated by metabolic coupling factors that are generated by energized mitochondria, including protons, adenine nucleotides, and perhaps certain amino acids (AA), as for instance aspartate, glutamate, or glutamine (Q). The goal of the present study was to evaluate the role of such factors when insulin release (IR) is stimulated by glucose or AA, alone or combined, using (31)P, (23)Na and (1)H NMR technology, respirometry, and biochemical analysis to study the metabolic events that occur in continuously superfused mouse beta-HC9 cells contained in agarose beads and enhanced by the phosphodiesterase inhibitor IBMX. Exposing beta-HC9 cells to high glucose or 3.5 mM of a physiological mixture of 18 AA (AAM) plus 2 mM glutamine caused a marked stimulation of insulin secretion associated with increased oxygen consumption, cAMP release, and phosphorylation potential as evidenced by higher phosphocreatine and lower P(i) peak areas of (31)P NMR spectra. Diazoxide blocked stimulation of IR completely, suggesting involvement of ATP-dependent potassium (K(ATP)) channels in this process. However, levels of MgATP and MgADP concentrations, which regulate channel activity, changed only slowly and little, whereas the rate of insulin release increased fast and very markedly. The involvement of other candidate coupling factors was therefore considered. High glucose or AAM + Q increased pH(i). The availability of temporal pH profiles allowed the precise computation of the phosphate potential (ATP/P(i) x ADP) in fuel-stimulated IR. Intracellular Na+ levels were greatly elevated by AAM + Q. However, glutamine alone or together with 2-amino-2-norbornanecarboxylic acid (which activates glutamate dehydrogenase) decreased beta-cell Na levels. Stimulation of beta-cells by glucose in the presence of AAM + Q (0.5 mM) was associated with rising cellular concentrations of glutamate and glutamine and strikingly lower aspartate levels. Methionine sulfoximine, an inhibitor of glutamine synthetase, blocked the glucose enhancement of AMM + Q-induced IR and associated changes in glutamine and aspartate but did not prevent the accumulation of glutamate. The results of this study demonstrate again that an increased phosphate potential and a functional K(ATP) channel are essential for metabolic coupling during fuel-stimulated insulin release but illustrate that determining the identity and relative importance of all participating coupling factors and second messengers remains a challenge largely unmet.


Subject(s)
Amino Acids/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amino Acids/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , Drug Combinations , Glucose/administration & dosage , Glucose/pharmacology , Glutamine/pharmacology , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Mice , Oxygen Consumption/drug effects , Phosphates/metabolism , Phosphorylation , Potassium Channels/metabolism , Sodium/metabolism , Time Factors
8.
Am J Physiol Endocrinol Metab ; 291(3): E525-35, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16638820

ABSTRACT

Neural and endocrine factors (i.e., Ach and GLP-1) restore defective glucose-stimulated insulin release in pancreatic islets lacking sulfonylurea type 1 receptors (SUR1(-/-)) (Doliba NM, Qin W, Vatamaniuk MZ, Li C, Zelent D, Najafi H, Buettger CW, Collins HW, Carr RD, Magnuson MA, and Matschinsky FM. Am J Physiol Endocrinol Metab 286: E834-E843, 2004). The goal of the present study was to assess fuel-induced respiration in SUR1(-/-) islets and to correlate it with changes in intracellular Ca(2+), insulin, and glucagon secretion. By use of a method based on O(2) quenching of phosphorescence, the O(2) consumption rate (OCR) of isolated islets was measured online in a perifusion system. Basal insulin release (IR) was 7-10 times higher in SUR1(-/-) compared with control (CON) islets, but the OCR was comparable. The effect of high glucose (16.7 mM) on IR and OCR was markedly reduced in SUR1(-/-) islets compared with CON. Ach (0.5 microM) in the presence of 16.7 mM glucose caused a large burst of IR in CON and SUR1(-/-) islets with minor changes in OCR in both groups of islets. In SUR1(-/-) islets, high glucose failed to inhibit glucagon secretion during stimulation with amino acids or Ach. We conclude that 1) reduced glucose responsiveness of SUR1(-/-) islets may be in part due to impaired energetics, as evidenced by significant decrease in glucose-stimulated OCR; 2) elevated intracellular Ca(2+) levels may contribute to altered insulin and glucagon secretion in SUR1(-/-) islets; and 3) The amplitudes of the changes in OCR during glucose and Ach stimulation do not correlate with IR in normal and SUR1(-/-) islets suggesting that the energy requirements for exocytosis are minor compared with other ATP-consuming reactions.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cell Respiration/physiology , Cholinergic Fibers/physiology , Glucose/pharmacology , Hormones/metabolism , Islets of Langerhans/metabolism , Multidrug Resistance-Associated Proteins/genetics , ATP-Binding Cassette Transporters/antagonists & inhibitors , Acetylcholine/pharmacology , Acetylcholinesterase/genetics , Amino Acids/pharmacology , Animals , Calcium/metabolism , Cell Respiration/drug effects , Gene Expression/genetics , Glucagon/metabolism , Glucose Transporter Type 2/genetics , Glyburide/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Kv1.3 Potassium Channel , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Oxygen Consumption/drug effects , Potassium Channels, Inwardly Rectifying/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Drug , Sulfonylurea Receptors
9.
J Biol Chem ; 281(22): 15064-72, 2006 Jun 02.
Article in English | MEDLINE | ID: mdl-16574664

ABSTRACT

Glutamate dehydrogenase (GDH) plays an important role in insulin secretion as evidenced in children by gain of function mutations of this enzyme that cause a hyperinsulinism-hyperammonemia syndrome (GDH-HI) and sensitize beta-cells to leucine stimulation. GDH transgenic mice were generated to express the human GDH-HI H454Y mutation and human wild-type GDH in islets driven by the rat insulin promoter. H454Y transgene expression was confirmed by increased GDH enzyme activity in islets and decreased sensitivity to GTP inhibition. The H454Y GDH transgenic mice had hypoglycemia with normal growth rates. H454Y GDH transgenic islets were more sensitive to leucine- and glutamine-stimulated insulin secretion but had decreased response to glucose stimulation. The fluxes via GDH and glutaminase were measured by tracing 15N flux from [2-15N]glutamine. The H454Y transgene in islets had higher insulin secretion in response to glutamine alone and had 2-fold greater GDH flux. High glucose inhibited both glutaminase and GDH flux, and leucine could not override this inhibition. 15NH4Cl tracing studies showed 15N was not incorporated into glutamate in either H454Y transgenic or normal islets. In conclusion, we generated a GDH-HI disease mouse model that has a hypoglycemia phenotype and confirmed that the mutation of H454Y is disease causing. Stimulation of insulin release by the H454Y GDH mutation or by leucine activation is associated with increased oxidative deamination of glutamate via GDH. This study suggests that GDH functions predominantly in the direction of glutamate oxidation rather than glutamate synthesis in mouse islets and that this flux is tightly controlled by glucose.


Subject(s)
Glutamate Dehydrogenase/genetics , Insulin/metabolism , Mutation , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium Signaling/drug effects , Glucose/pharmacology , Glutamate Dehydrogenase/antagonists & inhibitors , Glutamate Dehydrogenase/metabolism , Glutamine/pharmacology , Guanosine Triphosphate/pharmacology , Humans , Hyperinsulinism/enzymology , Hyperinsulinism/genetics , Hyperinsulinism/physiopathology , In Vitro Techniques , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Kinetics , Leucine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
10.
J Biol Chem ; 281(15): 10214-21, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16476731

ABSTRACT

Insulin secretion by pancreatic beta-cells is stimulated by glucose, amino acids, and other metabolic fuels. Glutamate dehydrogenase (GDH) has been shown to play a regulatory role in this process. The importance of GDH was underscored by features of hyperinsulinemia/hyperammonemia syndrome, where a dominant mutation causes the loss of inhibition by GTP and ATP. Here we report the effects of green tea polyphenols on GDH and insulin secretion. Of the four compounds tested, epigallocatechin gallate (EGCG) and epicatechin gallate were found to inhibit GDH with nanomolar ED(50) values and were therefore found to be as potent as the physiologically important inhibitor GTP. Furthermore, we have demonstrated that EGCG inhibits BCH-stimulated insulin secretion, a process that is mediated by GDH, under conditions where GDH is no longer inhibited by high energy metabolites. EGCG does not affect glucose-stimulated insulin secretion under high energy conditions where GDH is probably fully inhibited. We have further shown that these compounds act in an allosteric manner independent of their antioxidant activity and that the beta-cell stimulatory effects are directly correlated with glutamine oxidation. These results demonstrate that EGCG, much like the activator of GDH (BCH), can facilitate dissecting the complex regulation of insulin secretion by pharmacologically modulating the effects of GDH.


Subject(s)
Enzyme Inhibitors/pharmacology , Flavonoids/chemistry , Glutamate Dehydrogenase/antagonists & inhibitors , Insulin/metabolism , Phenols/chemistry , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Animals , Cattle , Dose-Response Relationship, Drug , Glutamate Dehydrogenase/metabolism , Guanosine Triphosphate/chemistry , Hyperammonemia/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Kinetics , Leucine/chemistry , Male , Models, Biological , Models, Chemical , Models, Molecular , Oxygen Consumption , Perfusion , Polyphenols , Protein Conformation , Rats , Rats, Wistar , Tea , Time Factors
11.
Am J Physiol Endocrinol Metab ; 286(5): E834-43, 2004 May.
Article in English | MEDLINE | ID: mdl-14736703

ABSTRACT

Inhibition of ATP-sensitive K+ (K(ATP)) channels by an increase in the ATP/ADP ratio and the resultant membrane depolarization are considered essential in the process leading to insulin release (IR) from pancreatic beta-cells stimulated by glucose. It is therefore surprising that mice lacking the sulfonylurea type 1 receptor (SUR1-/-) in beta-cells remain euglycemic even though the knockout is expected to cause hypoglycemia. To complicate matters, isolated islets of SUR1-/- mice secrete little insulin in response to high glucose, which extrapolates to hyperglycemia in the intact animal. It remains thus unexplained how euglycemia is maintained. In recognition of the essential role of neural and endocrine regulation of IR, we evaluated the effects of acetylcholine (ACh) and glucagon-like peptide-1 (GLP-1) on IR and free intracellular Ca2+ concentration ([Ca2+]i) of freshly isolated or cultured islets of SUR1-/- mice and B6D2F1 controls (SUR1+/+). IBMX, a phosphodiesterase inhibitor, was also used to explore cAMP-dependent signaling in IR. Most striking, and in contrast to controls, SUR1-/-) islets are hypersensitive to ACh and IBMX, as demonstrated by a marked increase of IR even in the absence of glucose. The hypersensitivity to ACh was reproduced in control islets by depolarization with the SUR1 inhibitor glyburide. Pretreatment of perifused SUR1-/- islets with ACh or IBMX restored glucose stimulation of IR, an effect expectedly insensitive to diazoxide. The calcium channel blocker verapamil reduced but did not abolish ACh-stimulated IR, supporting a role for intracellular Ca2+ stores in stimulus-secretion coupling. The effect of ACh on IR was greatly potentiated by GLP-1 (10 nM). ACh caused a dose-dependent increase in [Ca2+]i at 0.1-1 microM or biphasic changes (an initial sharp increase in [Ca2+]i followed by a sustained phase of low [Ca2+]i) at 1-100 microM. The latter effects were observed in substrate-free medium or in the presence of 16.7 mM glucose. We conclude that SUR1 deletion depolarizes the beta-cells and markedly elevates basal [Ca2+]i. Elevated [Ca2+]i in turn sensitizes the beta-cells to the secretory effects of ACh and IBMX. Priming by the combination of high [Ca2+]i, ACh, and GLP-1 restores the defective glucose responsiveness, precluding the development of diabetes but not effectively enough to cause hyperinsulinemic hypoglycemia.


Subject(s)
ATP-Binding Cassette Transporters , Acetylcholine/physiology , Blood Glucose/metabolism , Insulin/physiology , Islets of Langerhans/metabolism , Multidrug Resistance-Associated Proteins/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Calcium/metabolism , Glucagon/physiology , Glucagon-Like Peptide 1 , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Intracellular Fluid/metabolism , Islets of Langerhans/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred Strains , Mice, Knockout , Multidrug Resistance-Associated Proteins/deficiency , Peptide Fragments/physiology , Phosphodiesterase Inhibitors/pharmacology , Potassium Channels, Inwardly Rectifying , Protein Precursors/physiology , Receptors, Drug , Second Messenger Systems/physiology , Signal Transduction/physiology , Sulfonylurea Receptors
12.
J Biol Chem ; 279(14): 13393-401, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-14736887

ABSTRACT

Children with hypoglycemia due to recessive loss of function mutations of the beta-cell ATP-sensitive potassium (K(ATP)) channel can develop hypoglycemia in response to protein feeding. We hypothesized that amino acids might stimulate insulin secretion by unknown mechanisms, because the K(ATP) channel-dependent pathway of insulin secretion is defective. We therefore investigated the effects of amino acids on insulin secretion and intracellular calcium in islets from normal and sulfonylurea receptor 1 knockout (SUR1-/-) mice. Even though SUR1-/- mice are euglycemic, their islets are considered a suitable model for studies of the human genetic defect. SUR1-/- islets, but not normal islets, released insulin in response to an amino acid mixture ramp. This response to amino acids was decreased by 60% when glutamine was omitted. Insulin release by SUR1-/- islets was also stimulated by a ramp of glutamine alone. Glutamine was more potent than leucine or dimethyl glutamate. Basal intracellular calcium was elevated in SUR1-/- islets and was increased further by glutamine. In normal islets, methionine sulfoximine, a glutamine synthetase inhibitor, suppressed insulin release in response to a glucose ramp. This inhibition was reversed by glutamine or by 6-diazo-5-oxo-l-norleucine, a non-metabolizable glutamine analogue. High glucose doubled glutamine levels of islets. Methionine sulfoximine inhibition of glucose stimulated insulin secretion was associated with accumulation of glutamate and aspartate. We hypothesize that glutamine plays a critical role as a signaling molecule in amino acid- and glucose-stimulated insulin secretion, and that beta-cell depolarization and subsequent intracellular calcium elevation are required for this glutamine effect to occur.


Subject(s)
ATP-Binding Cassette Transporters , Glutamine/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Signal Transduction/physiology , Ammonium Chloride/pharmacokinetics , Animals , Calcium/metabolism , Glutamine/metabolism , Glyburide/pharmacology , Hypoglycemic Agents/pharmacology , Insulin Secretion , Islets of Langerhans/drug effects , Mice , Mice, Knockout , Multidrug Resistance-Associated Proteins/genetics , Nitrogen Isotopes , Potassium Channels, Inwardly Rectifying , Receptors, Drug , Signal Transduction/drug effects , Sulfonylurea Receptors
13.
Diabetes ; 52(2): 394-402, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12540613

ABSTRACT

In the present study, noninvasive (31)P and (23)Na(+)-nuclear magnetic resonance (NMR) technology and respirometry were used to compare the effect of high glucose (30 mmol/l) with the effect of the antidiabetic sulfonylurea (SU) compound glyburide (GLY) on energy metabolism, Na(+) flux, insulin, and cAMP release of continuously superfused beta-HC9 cells encapsulated in microscopic agarose beads. Both high glucose and GLY increased oxygen consumption in beta-HC9 cells (15-30%) with a maximal effect at 8 mmol/l for glucose and at 250 nmol/l for GLY. At the same time, insulin release from beta-cells increased by 15- and 25-fold with high glucose or GLY, respectively. The P-creatine (PCr) level was greatly increased and inorganic phosphate (P(i)) was decreased with 30 mmol/l glucose in contrast to the decreased level of PCr and increased P(i) with GLY. ATP levels remained unchanged during both interventions. Studies on isolated mitochondria of beta-HC9 cells showed that GLY added to mitochondria oxidizing glutamine or glutamate abolished the stimulation of respiration by ADP (state 3) meanwhile leaving state 3 respiration unchanged during oxidation of other substrates. Exposure of beta-HC9 cells to 5 mmol/l glucose decreased intracellular Na(+) levels monitored by (23)Na(+)-NMR spectroscopy and 30 mmol/l glucose resulted in a further decrease in cytosolic Na(+). In contrast, Na(+) increased when 1 micro mol/l GLY was added to the perfusate containing 5 mmol/l glucose. These data support the hypothesis that glucose activates the beta-cell through a "push mechanism" due to substrate pressure enhancing fuel flux, energy production, and extrusion of Na(+) from the cells in contrast to SU receptor (SUR)-1 inhibitors, which may modify intermediary and energy metabolism secondarily through a "pull mechanism" due to higher energy demand resulting from increased ion fluxes and the exocytotic work load.


Subject(s)
Energy Metabolism/drug effects , Glucose/pharmacology , Glyburide/pharmacology , Oxygen Consumption/drug effects , Sodium/metabolism , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Line , Glutamic Acid/metabolism , Glutamine/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Magnetic Resonance Spectroscopy/methods , Mitochondria/drug effects , Mitochondria/metabolism , Perfusion/methods , Phosphocreatine/metabolism , Rats , Succinic Acid/metabolism
14.
J Biol Chem ; 278(5): 2853-8, 2003 Jan 31.
Article in English | MEDLINE | ID: mdl-12444083

ABSTRACT

Glutamate dehydrogenase (GDH) is regulated by both positive (leucine and ADP) and negative (GTP and ATP) allosteric factors. We hypothesized that the phosphate potential of beta-cells regulates the sensitivity of leucine stimulation. These predictions were tested by measuring leucine-stimulated insulin secretion in perifused rat islets following glucose depletion and by tracing the nitrogen flux of [2-(15)N]glutamine using stable isotope techniques. The sensitivity of leucine stimulation was enhanced by long time (120-min) energy depletion and inhibited by glucose pretreatment. After limited 50-min glucose depletion, leucine, not alpha-ketoisocaproate, failed to stimulate insulin release. beta-Cells sensitivity to leucine is therefore proposed to be a function of GDH activation. Leucine increased the flux through GDH 3-fold compared with controls while causing insulin release. High glucose inhibited flux through both glutaminase and GDH, and leucine was unable to override this inhibition. These results clearly show that leucine induced the secretion of insulin by augmenting glutaminolysis through activating glutaminase and GDH. Glucose regulates beta-cell sensitivity to leucine by elevating the ratio of ATP and GTP to ADP and P(i) and thereby decreasing the flux through GDH and glutaminase. These mechanisms provide an explanation for hypoglycemia caused by mutations of GDH in children.


Subject(s)
Glutamine/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Leucine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Glutamate Dehydrogenase/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Kinetics , Male , Models, Biological , Perfusion , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...