Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
2.
PLoS One ; 17(12): e0279560, 2022.
Article in English | MEDLINE | ID: mdl-36580474

ABSTRACT

The risk of type-2 diabetes and cardiovascular disease is higher in subjects with metabolic syndrome, a cluster of clinical conditions characterized by obesity, impaired glucose metabolism, hyperinsulinemia, hyperlipidemia and hypertension. Diuretics are frequently used to treat hypertension in these patients, however, their use has long been associated with poor metabolic outcomes which cannot be fully explained by their diuretic effects. Here, we show that mice lacking the diuretic-sensitive Na+K+2Cl-cotransporter-1 Nkcc1 (Slc12a2) in insulin-secreting ß-cells of the pancreatic islet (Nkcc1ßKO) have reduced in vitro insulin responses to glucose. This is associated with islet hypoplasia at the expense of fewer and smaller ß-cells. Remarkably, Nkcc1ßKO mice excessively gain weight and progressive metabolic syndrome when fed a standard chow diet ad libitum. This is characterized by impaired hepatic insulin receptor activation and altered lipid metabolism. Indeed, overweight Nkcc1ßKO but not lean mice had fasting and fed hyperglycemia, hypertriglyceridemia and non-alcoholic steatohepatitis. Notably, fasting hyperinsulinemia was detected earlier than hyperglycemia, insulin resistance, glucose intolerance and increased hepatic de novo gluconeogenesis. Therefore, our data provide evidence supporting the novel hypothesis that primary ß-cell defects related to Nkcc1-regulated intracellular Cl-homeostasis and ß-cell growth can result in the development of metabolic syndrome shedding light into additional potential mechanisms whereby chronic diuretic use may have adverse effects on metabolic homeostasis in susceptible individuals.


Subject(s)
Hyperglycemia , Hyperinsulinism , Hypertension , Insulin Resistance , Insulin-Secreting Cells , Metabolic Syndrome , Mice , Animals , Metabolic Syndrome/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Hyperinsulinism/metabolism , Hyperglycemia/metabolism , Diuretics , Hypertension/metabolism
3.
PLoS One ; 15(12): e0242749, 2020.
Article in English | MEDLINE | ID: mdl-33264332

ABSTRACT

Cystic fibrosis (CF) is due to mutations in the CF-transmembrane conductance regulator (CFTR) and CF-related diabetes (CFRD) is its most common co-morbidity, affecting ~50% of all CF patients, significantly influencing pulmonary function and longevity. Yet, the complex pathogenesis of CFRD remains unclear. Two non-mutually exclusive underlying mechanisms have been proposed in CFRD: i) damage of the endocrine cells secondary to the severe exocrine pancreatic pathology and ii) intrinsic ß-cell impairment of the secretory response in combination with other factors. The later has proven difficult to determine due to low expression of CFTR in ß-cells, which results in the general perception that this Cl-channel does not participate in the modulation of insulin secretion or the development of CFRD. The objective of the present work is to demonstrate CFTR expression at the molecular and functional levels in insulin-secreting ß-cells in normal human islets, where it seems to play a role. Towards this end, we have used immunofluorescence confocal and immunofluorescence microscopy, immunohistochemistry, RT-qPCR, Western blotting, pharmacology, electrophysiology and insulin secretory studies in normal human, rat and mouse islets. Our results demonstrate heterogeneous CFTR expression in human, mouse and rat ß-cells and provide evidence that pharmacological inhibition of CFTR influences basal and stimulated insulin secretion in normal mouse islets but not in islets lacking this channel, despite being detected by electrophysiological means in ~30% of ß-cells. Therefore, our results demonstrate a potential role for CFTR in the pancreatic ß-cell secretory response suggesting that intrinsic ß-cell dysfunction may also participate in the pathogenesis of CFRD.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Insulin-Secreting Cells/metabolism , Adult , Aged , Animals , Antibodies/metabolism , Antigens/metabolism , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Female , Humans , Infant , Insulin Secretion , Male , Mice , Middle Aged , Rats , Reproducibility of Results , Young Adult
4.
Hum Mutat ; 41(5): 884-905, 2020 05.
Article in English | MEDLINE | ID: mdl-32027066

ABSTRACT

The most common genetic cause of neonatal diabetes and hyperinsulinism is pathogenic variants in ABCC8 and KCNJ11. These genes encode the subunits of the ß-cell ATP-sensitive potassium channel, a key component of the glucose-stimulated insulin secretion pathway. Mutations in the two genes cause dysregulated insulin secretion; inactivating mutations cause an oversecretion of insulin, leading to congenital hyperinsulinism, whereas activating mutations cause the opposing phenotype, diabetes. This review focuses on variants identified in ABCC8 and KCNJ11, the phenotypic spectrum and the treatment implications for individuals with pathogenic variants.


Subject(s)
Congenital Hyperinsulinism/genetics , Diabetes Mellitus/genetics , Insulin-Secreting Cells/metabolism , Mutation , Potassium Channels, Inwardly Rectifying/genetics , Sulfonylurea Receptors/genetics , Congenital Hyperinsulinism/diagnosis , Diabetes Mellitus/diagnosis , Gain of Function Mutation , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Infant, Newborn , Loss of Function Mutation
5.
Biochem Soc Trans ; 47(6): 1843-1855, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31697318

ABSTRACT

It is accepted that insulin-secreting ß-cells release insulin in response to glucose even in the absence of functional ATP-sensitive K+ (KATP)-channels, which play a central role in a 'consensus model' of secretion broadly accepted and widely reproduced in textbooks. A major shortcoming of this consensus model is that it ignores any and all anionic mechanisms, known for more than 40 years, to modulate ß-cell electrical activity and therefore insulin secretion. It is now clear that, in addition to metabolically regulated KATP-channels, ß-cells are equipped with volume-regulated anion (Cl-) channels (VRAC) responsive to glucose concentrations in the range known to promote electrical activity and insulin secretion. In this context, the electrogenic efflux of Cl- through VRAC and other Cl- channels known to be expressed in ß-cells results in depolarization because of an outwardly directed Cl- gradient established, maintained and regulated by the balance between Cl- transporters and channels. This review will provide a succinct historical perspective on the development of a complex hypothesis: Cl- transporters and channels modulate insulin secretion in response to nutrients.


Subject(s)
Chlorides/metabolism , Insulin-Secreting Cells/physiology , Ion Channels/metabolism , Models, Biological , Animals , Humans , Insulin Secretion , Insulin-Secreting Cells/metabolism , Ion Transport
6.
Sci Rep ; 7(1): 17231, 2017 12 08.
Article in English | MEDLINE | ID: mdl-29222447

ABSTRACT

Cystic fibrosis (CF)-related diabetes (CFRD) is thought to result from beta-cell injury due in part to pancreas exocrine damage and lipofibrosis. CFRD pancreata exhibit reduced islet density and altered cellular composition. To investigate a possible etiology, we tested the hypothesis that such changes are present in CF pancreata before the development of lipofibrosis. We evaluated pancreas and islet morphology in tissues from very young CF children (<4 years of age), and adult patients with CF and CFRD. The relative number of beta-cells in young CF tissues was reduced by 50% or more when compared to age-matched controls. Furthermore, young CF tissues displayed significantly smaller insulin-positive areas, lower proportion of beta-cells positive for the proliferation marker Ki67 or the ductal marker CK19 vs. control subjects, and islet inflammatory cell infiltrates, independently of the severity of the exocrine lesion and in the absence of amyloid deposits. CFRD pancreata exhibited greater islet injury with further reduction in islet density, decreased relative beta-cell number, and presence of amyloid deposits. Together, these results strongly suggest that an early deficiency in beta-cell number in infants with CF may contribute to the development of glucose intolerance in the CF pediatric population, and to CFRD, later in life.


Subject(s)
Cystic Fibrosis/pathology , Diabetes Complications/pathology , Islets of Langerhans/pathology , Cell Proliferation , Child, Preschool , Cystic Fibrosis/metabolism , Diabetes Complications/metabolism , Female , Glucose Tolerance Test , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Male , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology
7.
Sci Rep ; 7(1): 1732, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28496181

ABSTRACT

Intracellular chloride concentration ([Cl-]i) in pancreatic ß-cells is kept above electrochemical equilibrium due to the predominant functional presence of Cl- loaders such as the Na+K+2Cl- co-transporter 1 (Slc12a2) over Cl-extruders of unidentified nature. Using molecular cloning, RT-PCR, Western blotting, immunolocalization and in vitro functional assays, we establish that the "neuron-specific" K+Cl- co-transporter 2 (KCC2, Slc12a5) is expressed in several endocrine cells of the pancreatic islet, including glucagon secreting α-cells, but particularly in insulin-secreting ß-cells, where we provide evidence for its role in the insulin secretory response. Three KCC2 splice variants were identified: the formerly described KCC2a and KCC2b along with a novel one lacking exon 25 (KCC2a-S25). This new variant is undetectable in brain or spinal cord, the only and most abundant known sources of KCC2. Inhibition of KCC2 activity in clonal MIN6 ß-cells increases basal and glucose-stimulated insulin secretion and Ca2+ uptake in the presence of glibenclamide, an inhibitor of the ATP-dependent potassium (KATP)-channels, thus suggesting a possible mechanism underlying KCC2-dependent insulin release. We propose that the long-time considered "neuron-specific" KCC2 co-transporter is expressed in pancreatic islet ß-cells where it modulates Ca2+-dependent insulin secretion.


Subject(s)
Insulin Secretion , Neurons/metabolism , Symporters/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Base Sequence , Calcium/metabolism , Cell Line , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , KATP Channels/metabolism , Mice , Pyridazines , Symporters/chemistry , Symporters/genetics , Thiazoles , K Cl- Cotransporters
8.
J Endocrinol ; 227(3): 153-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26400961

ABSTRACT

The products of the Slc12a1 and Slc12a2 genes, commonly known as Na(+)-dependent K(+)2Cl(-) co-transporters NKCC2 and NKCC1, respectively, are the targets for the diuretic bumetanide. NKCCs are implicated in the regulation of intracellular chloride concentration ([Cl(-)]i) in pancreatic ß-cells, and as such, they may play a role in glucose-stimulated plasma membrane depolarization and insulin secretion. Unexpectedly, permanent elimination of NKCC1 does not preclude insulin secretion, an event potentially linked to the homeostatic regulation of additional Cl(-) transporters expressed in ß-cells. In this report we provide evidence for such a mechanism. Mice lacking a single allele of Slc12a2 exhibit lower fasting glycemia, increased acute insulin response (AIR) and lower blood glucose levels 15-30 min after a glucose load when compared to mice harboring both alleles of the gene. Furthermore, heterozygous expression or complete absence of Slc12a2 associates with increased NKCC2 protein expression in rodent pancreatic ß-cells. This has been confirmed by using chronic pharmacological down-regulation of NKCC1 with bumetanide in the mouse MIN6 ß-cell line or permanent molecular silencing of NKCC1 in COS7 cells, which results in increased NKCC2 expression. Furthermore, MIN6 cells chronically pretreated with bumetanide exhibit increased initial rates of Cl(-) uptake while preserving glucose-stimulated insulin secretion. Together, our results suggest that NKCCs are involved in insulin secretion and that a single Slc12a2 allele may protect ß-cells from failure due to increased homeostatic expression of Slc12a1.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Solute Carrier Family 12, Member 1/metabolism , Solute Carrier Family 12, Member 2/metabolism , Animals , Bumetanide/pharmacology , Down-Regulation/drug effects , Gene Expression/drug effects , Glucose Tolerance Test , Heterozygote , Insulin-Secreting Cells/drug effects , Mice , Mice, Transgenic , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 1/genetics , Solute Carrier Family 12, Member 2/genetics
9.
Diabetes ; 61(1): 85-93, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22187376

ABSTRACT

Amino acids profoundly affect insulin action and glucose metabolism in mammals. Here, we investigated the role of the mediobasal hypothalamus (MBH), a key center involved in nutrient-dependent metabolic regulation. Specifically, we tested the novel hypothesis that the metabolism of leucine within the MBH couples the central sensing of leucine with the control of glucose production by the liver. We performed either central (MBH) or systemic infusions of leucine in Sprague-Dawley male rats during basal pancreatic insulin clamps in combination with various pharmacological and molecular interventions designed to modulate leucine metabolism in the MBH. We also examined the role of hypothalamic ATP-sensitive K(+) channels (K(ATP) channels) in the effects of leucine. Enhancing the metabolism of leucine acutely in the MBH lowered blood glucose through a biochemical network that was insensitive to rapamycin but strictly dependent on the hypothalamic metabolism of leucine to α-ketoisocaproic acid and, further, insensitive to acetyl- and malonyl-CoA. Functional K(ATP) channels were also required. Importantly, molecular attenuation of this central sensing mechanism in rats conferred susceptibility to developing hyperglycemia. We postulate that the metabolic sensing of leucine in the MBH is a previously unrecognized mechanism for the regulation of hepatic glucose production required to maintain glucose homeostasis.


Subject(s)
Glucose/metabolism , Hypothalamus/metabolism , Leucine/metabolism , Liver/metabolism , Animals , Cells, Cultured , Gluconeogenesis/drug effects , Gluconeogenesis/physiology , Humans , Leucine/pharmacology , Leucine/physiology , Liver/drug effects , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Mice , Mice, Knockout , Models, Biological , Rats , Rats, Sprague-Dawley
10.
J Biol Chem ; 286(10): 8481-8492, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21199866

ABSTRACT

The ATP-binding cassette (ABC) transporter ABCB6 is a mitochondrial porphyrin transporter that activates porphyrin biosynthesis. ABCB6 lacks a canonical mitochondrial targeting sequence but reportedly traffics to other cellular compartments such as the plasma membrane. How ABCB6 reaches these destinations is unknown. In this study, we show that endogenous ABCB6 is glycosylated in multiple cell types, indicating trafficking through the endoplasmic reticulum (ER), and has only one atypical site for glycosylation (NXC) in its amino terminus. ABCB6 remained glycosylated when the highly conserved cysteine (Cys-8) was substituted with serine to make a consensus site, NXS. However, this substitution blocked ER exit and produced ABCB6 degradation, which was mostly reversed by the proteasomal inhibitor MG132. The amino terminus of ABCB6 has an additional highly conserved ER luminal cysteine (Cys-26). When Cys-26 was mutated alone or in combination with Cys-8, it also resulted in instability and ER retention. Further analysis revealed that these two cysteines form a disulfide bond. We discovered that other ABC transporters with an amino terminus in the ER had similarly configured conserved cysteines. This analysis led to the discovery of a disease-causing mutation in the sulfonylurea receptor 1 (SUR1)/ABCC8 from a patient with hyperinsulinemic hypoglycemia. The mutant allele only contains a mutation in a conserved amino-terminal cysteine, producing SUR1 that fails to reach the cell surface. These results suggest that for ABC transporters the propensity to form a disulfide bond in the ER defines a unique checkpoint that determines whether a protein is ER-retained.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Disulfides/metabolism , Endoplasmic Reticulum/metabolism , Mitochondrial Proteins/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/metabolism , ATP-Binding Cassette Transporters/genetics , Alleles , Amino Acid Substitution , Animals , Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Endoplasmic Reticulum/genetics , Glycosylation , HEK293 Cells , Humans , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Hypoglycemia/genetics , Hypoglycemia/metabolism , K562 Cells , Leupeptins/pharmacology , Mice , Mitochondrial Proteins/genetics , Mutation, Missense , NIH 3T3 Cells , Potassium Channels, Inwardly Rectifying/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Structure, Tertiary , Protein Transport/genetics , Receptors, Drug/genetics , Sulfonylurea Receptors
11.
J Clin Invest ; 119(11): 3246-56, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19805912

ABSTRACT

The enhanced oxidative stress associated with type 2 diabetes mellitus contributes to disease pathogenesis. We previously identified plasma membrane-associated ATP-sensitive K+ (KATP) channels of pancreatic beta cells as targets for oxidants. Here, we examined the effects of genetic and pharmacologic ablation of KATP channels on loss of mouse beta cell function and viability following oxidative stress. Using mice lacking the sulfonylurea receptor type 1 (Sur1) subunit of KATP channels, we found that, compared with insulin secretion by WT islets, insulin secretion by Sur1-/- islets was less susceptible to oxidative stress induced by the oxidant H2O2. This was likely, at least in part, a result of the reduced ability of H2O2 to hyperpolarize plasma membrane potential and reduce cytosolic free Ca2+ concentration ([Ca2+]c) in the Sur1-/- beta cells. Remarkably, Sur1-/- beta cells were less prone to apoptosis induced by H2O2 or an NO donor than WT beta cells, despite an enhanced basal rate of apoptosis. This protective effect was attributed to upregulation of the antioxidant enzymes SOD, glutathione peroxidase, and catalase. Upregulation of antioxidant enzymes and reduced sensitivity of Sur1-/- cells to H2O2-induced apoptosis were mimicked by treatment with the sulfonylureas tolbutamide and gliclazide. Enzyme upregulation and protection against oxidant-induced apoptosis were abrogated by agents lowering [Ca2+]c. Sur1-/- mice were less susceptible than WT mice to streptozotocin-induced beta cell destruction and subsequent hyperglycemia and death, which suggests that loss of KATP channel activity may protect against streptozotocin-induced diabetes in vivo.


Subject(s)
Insulin-Secreting Cells/physiology , KATP Channels/antagonists & inhibitors , KATP Channels/physiology , Oxidative Stress/physiology , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blood Glucose/analysis , Calcium/metabolism , Diabetes Mellitus, Experimental , Enzymes/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gliclazide/pharmacology , Hydrogen Peroxide/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , KATP Channels/genetics , Mice , Mice, Knockout , Oxidants/pharmacology , Oxidative Stress/drug effects , Streptozocin/pharmacology , Tolbutamide/pharmacology
12.
Endocrinology ; 150(3): 1132-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18988673

ABSTRACT

Glucose-stimulated insulin and glucagon release regulates glucose homeostasis by an excitation-secretion coupling pathway beginning with ATP-sensitive K(+) channel closure, membrane depolarization, and entry of calcium ions to stimulate exocytosis. The contribution of voltage-gated sodium channels to this release pathway is still being elucidated. We demonstrate that loss of Scn1b, a major regulatory subunit expressed with Na(v)1.7 protein in mouse pancreatic islets, reduces glucose-stimulated insulin and glucagon secretion in vitro and in vivo, resulting in severe fed and fasting hypoglycemia. This genetic mouse model is the first to demonstrate that sodium channelopathy impairs the physiological excitation-release coupling pathway for pancreatic insulin and glucagon release.


Subject(s)
Glucagon/metabolism , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Sodium Channels/genetics , Animals , Cells, Cultured , Down-Regulation/drug effects , Glucose Tolerance Test , Hypoglycemia/genetics , Hypoglycemia/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Sodium Channels/metabolism , Voltage-Gated Sodium Channel beta-1 Subunit
13.
Am J Physiol Endocrinol Metab ; 295(6): E1298-306, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18940941

ABSTRACT

Mammalian beta-cells are acutely and chronically regulated by sensing surrounding glucose levels that determine the rate at which insulin is secreted, to maintain euglycemia. Experimental research in vitro and in vivo has shown that, when these cells are exposed to adverse conditions like long periods of hypoglycemia or hyperglycemia, their capability to sense glucose is decreased. Understanding the normal physiology and identifying the main players along this route becomes paramount. In this review, we have taken on the task of looking at the role that ion channels play in the regulation of this process, delineating the different families, and describing the signaling that parallels the glucose sensing process that results in insulin release.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , Potassium Channels/physiology , Animals , Calcium Signaling/physiology , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Membrane Potentials/physiology , Models, Biological , Transient Receptor Potential Channels/physiology
15.
Endocr Rev ; 29(3): 265-91, 2008 May.
Article in English | MEDLINE | ID: mdl-18436707

ABSTRACT

An explosion of work over the last decade has produced insight into the multiple hereditary causes of a nonimmunological form of diabetes diagnosed most frequently within the first 6 months of life. These studies are providing increased understanding of genes involved in the entire chain of steps that control glucose homeostasis. Neonatal diabetes is now understood to arise from mutations in genes that play critical roles in the development of the pancreas, of beta-cell apoptosis and insulin processing, as well as the regulation of insulin release. For the basic researcher, this work is providing novel tools to explore fundamental molecular and cellular processes. For the clinician, these studies underscore the need to identify the genetic cause underlying each case. It is increasingly clear that the prognosis, therapeutic approach, and genetic counseling a physician provides must be tailored to a specific gene in order to provide the best medical care.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/physiopathology , Infant, Newborn, Diseases/genetics , Infant, Newborn, Diseases/physiopathology , Animals , Diabetes Mellitus, Type 1/therapy , Humans , Hyperglycemia/genetics , Hyperglycemia/physiopathology , Hyperglycemia/therapy , Infant, Newborn , Infant, Newborn, Diseases/therapy
16.
Cell Physiol Biochem ; 20(1-4): 65-74, 2007.
Article in English | MEDLINE | ID: mdl-17595516

ABSTRACT

K(ATP) channel activity influences beta cell Ca(2+) homeostasis by regulating Ca(2+) influx through L-type Ca(2+) channels. The present paper demonstrates that loss of K(ATP) channel activity due to pharmacologic or genetic ablation affects Ca(2+) storage in intracellular organelles. ATP depletion, by the mitochondrial inhibitor FCCP, led to Ca(2+) release from the endoplasmic reticulum (ER) of wildtype beta cells. Blockade of ER Ca(2+) ATPases by cyclopiazonic acid abolished the FCCP-induced Ca(2+) transient. In beta cells treated with K(ATP) channel inhibitors FCCP elicited a significantly larger Ca(2+) transient. Cyclopiazonic acid did not abolish this Ca(2+) transient suggesting that non-ER compartments are recruited as additional Ca(2+) stores in beta cells lacking K(ATP) channel activity. Genetic ablation of K(ATP) channels in SUR1KO mice produced identical results. In INS-1 cells transfected with a mitochondrial-targeted Ca(2+)-sensitive fluorescence dye (ratiometric pericam) the increase in mitochondrial Ca(2+) evoked by tolbutamide was 5-fold larger compared to 15 mM glucose. These data show that genetic or pharmacologic ablation of K(ATP) channel activity conveys Ca(2+) release from a non-ER store. Based on the sensitivity to FCCP and the property of tolbutamide to increase mitochondrial Ca(2+) it is suggested that mitochondria are the recruited store. The change in Ca(2+) sequestration in beta cells treated with insulinotropic antidiabetics may have implications for beta cell survival and the therapeutic use of these drugs.


Subject(s)
Calcium/metabolism , Insulin-Secreting Cells/metabolism , Potassium Channels/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cell Compartmentation , Cell Line , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Glucose/pharmacology , In Vitro Techniques , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Potassium Channels/deficiency , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/deficiency , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/deficiency , Receptors, Drug/genetics , Receptors, Drug/metabolism , Sulfonylurea Receptors , Tolbutamide/pharmacology
17.
J Biol Chem ; 282(3): 1747-56, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17138557

ABSTRACT

In adult beta-cells glucose-induced insulin secretion involves two mechanisms (a) a K(ATP) channel-dependent Ca(2+) influx and rise of cytosolic [Ca(2+)](c) and (b) a K(ATP) channel-independent amplification of secretion without further increase of [Ca(2+)](c). Mice lacking the high affinity sulfonylurea receptor (Sur1KO), and thus K(ATP) channels, have been developed as a model of congenital hyperinsulinism. Here, we compared [Ca(2+)](c) and insulin secretion in overnight cultured islets from 2-week-old normal and Sur1KO mice. Control islets proved functionally mature: the magnitude and biphasic kinetics of [Ca(2+)](c) and insulin secretion changes induced by glucose, and operation of the amplifying pathway, were similar to adult islets. Sur1KO islets perifused with 1 mm glucose showed elevation of both basal [Ca(2+)](c) and insulin secretion. Stimulation with 15 mm glucose produced a transient drop of [Ca(2+)](c) followed by an overshoot and a sustained elevation, accompanied by a monophasic, 6-fold increase in insulin secretion. Glucose also increased insulin secretion when [Ca(2+)](c) was clamped by KCl. When Sur1KO islets were cultured in 5 instead of 10 mm glucose, [Ca(2+)](c) and insulin secretion were unexpectedly low in 1 mm glucose and increased following a biphasic time course upon stimulation by 15 mm glucose. This K(ATP) channel-independent first phase [Ca(2+)](c) rise was attributed to a Na(+)-, Cl(-)-, and Na(+)-pump-independent depolarization of beta-cells, leading to Ca(2+) influx through voltage-dependent calcium channels. Glucose indeed depolarized Sur1KO islets under these conditions. It is suggested that unidentified potassium channels are sensitive to glucose and subserve the acute and long-term metabolic control of [Ca(2+)](c) in beta-cells without functional K(ATP) channels.


Subject(s)
Adenosine Triphosphate/chemistry , Calcium/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Potassium Channels/chemistry , ATP-Binding Cassette Transporters/genetics , Animals , Cyclic AMP/metabolism , Humans , Insulin Secretion , Islets of Langerhans/metabolism , Kinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Sulfonylurea Receptors
18.
Pflugers Arch ; 453(5): 703-18, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16897043

ABSTRACT

The sulfonylurea receptors (SURs) ABCC8/SUR1 and ABCC9/SUR2 are members of the C-branch of the transport adenosine triphosphatase superfamily. Unlike their brethren, the SURs have no identified transport function; instead, evolution has matched these molecules with K(+) selective pores, either K(IR)6.1/KCNJ8 or K(IR)6.2/KCNJ11, to assemble adenosine triphosphate (ATP)-sensitive K(+) channels found in endocrine cells, neurons, and both smooth and striated muscle. Adenine nucleotides, the major regulators of ATP-sensitive K(+) (K(ATP)) channel activity, exert a dual action. Nucleotide binding to the pore reduces the activity or channel open probability, whereas Mg-nucleotide binding and/or hydrolysis in the nucleotide-binding domains of SUR antagonize this inhibitory action to stimulate channel openings. Mutations in either subunit can alter this balance and, in the case of the SUR1/KIR6.2 channels found in neurons and insulin-secreting pancreatic beta cells, are the cause of monogenic forms of hyperinsulinemic hypoglycemia and neonatal diabetes. Additionally, the subtle dysregulation of K(ATP) channel activity by a K(IR)6.2 polymorphism has been suggested as a predisposing factor in type 2 diabetes mellitus. Studies on K(ATP) channel null mice are clarifying the roles of these metabolically sensitive channels in a variety of tissues.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Potassium Channels, Inwardly Rectifying/physiology , Potassium Channels/physiology , Receptors, Drug/physiology , Amino Acids/physiology , Animals , Calcium/physiology , Catecholamines/metabolism , Congenital Hyperinsulinism/physiopathology , Diabetes Mellitus/congenital , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/physiology , Glucose/metabolism , Humans , Infant , Infant, Newborn , Insulin/metabolism , Insulin Secretion , Liver/metabolism , Mice , Mice, Transgenic , Models, Molecular , Protein Structure, Tertiary , Sulfonylurea Receptors
19.
N Engl J Med ; 355(5): 456-66, 2006 Aug 03.
Article in English | MEDLINE | ID: mdl-16885549

ABSTRACT

BACKGROUND: The ATP-sensitive potassium (K(ATP)) channel, composed of the beta-cell proteins sulfonylurea receptor (SUR1) and inward-rectifying potassium channel subunit Kir6.2, is a key regulator of insulin release. It is inhibited by the binding of adenine nucleotides to subunit Kir6.2, which closes the channel, and activated by nucleotide binding or hydrolysis on SUR1, which opens the channel. The balance of these opposing actions determines the low open-channel probability, P(O), which controls the excitability of pancreatic beta cells. We hypothesized that activating mutations in ABCC8, which encodes SUR1, cause neonatal diabetes. METHODS: We screened the 39 exons of ABCC8 in 34 patients with permanent or transient neonatal diabetes of unknown origin. We assayed the electrophysiologic activity of mutant and wild-type K(ATP) channels. RESULTS: We identified seven missense mutations in nine patients. Four mutations were familial and showed vertical transmission with neonatal and adult-onset diabetes; the remaining mutations were not transmitted and not found in more than 300 patients without diabetes or with early-onset diabetes of similar genetic background. Mutant channels in intact cells and in physiologic concentrations of magnesium ATP had a markedly higher P(O) than did wild-type channels. These overactive channels remained sensitive to sulfonylurea, and treatment with sulfonylureas resulted in euglycemia. CONCLUSIONS: Dominant mutations in ABCC8 accounted for 12 percent of cases of neonatal diabetes in the study group. Diabetes results from a newly discovered mechanism whereby the basal magnesium-nucleotide-dependent stimulatory action of SUR1 on the Kir pore is elevated and blockade by sulfonylureas is preserved.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Diabetes Mellitus/genetics , Mutation, Missense , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels/genetics , Receptors, Drug/genetics , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Birth Weight , Developmental Disabilities/complications , Diabetes Complications , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Female , Heterozygote , Humans , Hypoglycemic Agents/therapeutic use , Infant, Newborn , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Pedigree , Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/antagonists & inhibitors , Receptors, Drug/metabolism , Sulfonylurea Compounds/therapeutic use , Sulfonylurea Receptors , Tolbutamide/pharmacology
20.
Diabetes ; 54(10): 2946-51, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16186397

ABSTRACT

The mechanisms involved in the release of glucagon in response to hypoglycemia are unclear. Proposed mechanisms include the activation of the autonomic nervous system via glucose-sensing neurons in the central nervous system, via the regulation of glucagon secretion by intra-islet insulin and zinc concentrations, or via direct ionic control, all mechanisms that involve high-affinity sulfonylurea receptor/inwardly rectifying potassium channel-type ATP-sensitive K(+) channels. Patients with congenital hyperinsulinism provide a unique physiological model to understand glucagon regulation. In this study, we compare serum glucagon responses to hyperinsulinemic hypoglycemia versus nonhyperinsulinemic hypoglycemia. In the patient group (n = 20), the mean serum glucagon value during hyperinsulinemic hypoglycemia was 17.6 +/- 5.7 ng/l compared with 59.4 +/- 7.8 ng/l in the control group (n = 15) with nonhyperinsulinemic hypoglycemia (P < 0.01). There was no difference between the serum glucagon responses in children with diffuse, focal, and diazoxide-responsive forms of hyperinsulinism. The mean serum epinephrine and norepinephrine concentrations in the hyperinsulinemic group were 2,779 +/- 431 pmol/l and 2.9 +/- 0.7 nmol/l and appropriately rose despite the blunted glucagon response. In conclusion, the loss of ATP-sensitive K(+) channels and or elevated intraislet insulin cannot explain the blunted glucagon release in all patients with congenital hyperinsulinism. Other possible mechanisms such as the suppressive effect of prolonged hyperinsulinemia on alpha-cell secretion should be considered.


Subject(s)
Glucagon/blood , Homeostasis , Hormones/blood , Hyperglycemia/blood , Hyperinsulinism/congenital , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate/pharmacology , Child, Preschool , Diazoxide/therapeutic use , Epinephrine/blood , Fatty Acids, Nonesterified/blood , Female , Humans , Hyperinsulinism/blood , Hyperinsulinism/genetics , Infant , Insulin/analysis , Insulin/blood , Islets of Langerhans/chemistry , Ketone Bodies/blood , Male , Mutation , Norepinephrine/blood , Potassium Channels/genetics , Potassium Channels/physiology , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Sulfonylurea Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...