Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Acta Physiol (Oxf) ; 222(3)2018 03.
Article in English | MEDLINE | ID: mdl-29032602

ABSTRACT

AIM: Muscle wasting is one of the factors most strongly predicting mortality and morbidity in critically ill intensive care unit (ICU). This muscle wasting affects both limb and respiratory muscles, but the understanding of underlying mechanisms and muscle-specific differences remains incomplete. This study aimed at investigating the temporal expression and phosphorylation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway in muscle wasting associated with the ICU condition to characterize the JAK/STAT proteins and the related changes leading or responding to their activation during exposure to the ICU condition. METHODS: A novel experimental ICU model allowing long-term exposure to the ICU condition, immobilization and mechanical ventilation, was used in this study. Rats were pharmacologically paralysed by post-synaptic neuromuscular blockade and mechanically ventilated for durations varying between 6 hours and 14 days to study muscle-specific differences in the temporal activation of the JAK/STAT pathway in plantaris, intercostal and diaphragm muscles. RESULTS: The JAK2/STAT3 pathway was significantly activated irrespective of muscle, but muscle-specific differences were observed in the temporal activation pattern between plantaris, intercostal and diaphragm muscles. CONCLUSION: The JAK2/STAT3 pathway was differentially activated in plantaris, intercostal and diaphragm muscles in response to the ICU condition. Thus, JAK2/STAT3 inhibitors may provide an attractive pharmacological intervention strategy in immobilized ICU patients, but further experimental studies are required in the study of muscle-specific effects on muscle mass and function in response to both short- and long-term exposure to the ICU condition prior to the translation into clinical research and practice.


Subject(s)
Janus Kinase 2/metabolism , Muscle, Skeletal/metabolism , Respiration, Artificial/adverse effects , Restraint, Physical/adverse effects , STAT3 Transcription Factor/metabolism , Animals , Female , Muscle, Skeletal/pathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley
2.
Diabetes Obes Metab ; 18(7): 648-53, 2016 07.
Article in English | MEDLINE | ID: mdl-27027662

ABSTRACT

Hyperglycaemia is commonly observed on admission and during hospitalization for medical illness, traumatic injury, burn and surgical intervention. This transient hyperglycaemia is referred to as stress-induced hyperglycaemia (SIH) and frequently occurs in individuals without a history of diabetes. SIH has many of the same underlying hormonal disturbances as diabetes mellitus, specifically absolute or relative insulin deficiency and glucagon excess. SIH has the added features of elevated blood levels of catecholamines and cortisol, which are not typically present in people with diabetes who are not acutely ill. The seriousness of SIH is highlighted by its greater morbidity and mortality rates compared with those of hospitalized patients with normal glucose levels, and this increased risk is particularly high in those without pre-existing diabetes. Insulin is the treatment standard for SIH, but new therapies that reduce glucose variability and hypoglycaemia are desired. In the present review, we focus on the key role of glucagon in SIH and discuss the potential use of glucagon receptor blockers and glucagon-like peptide-1 receptor agonists in SIH to achieve target glucose control.


Subject(s)
Glucagon/physiology , Hyperglycemia/etiology , Stress, Physiological/physiology , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Glucagon/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Humans , Hyperglycemia/drug therapy , Hyperglycemia/physiopathology
3.
Exp Clin Endocrinol Diabetes ; 116 Suppl 1: S46-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18777454

ABSTRACT

Mice, deficient for vascular endothelial growth factor VEGF-A in pancreatic islets, have reduced insulin gene expression levels and an impaired glucose tolerance. Here, we investigated whether VEGF-A was required for physiological glucose-stimulated insulin secretion and insulin content. We performed in situ pancreas perfusions and islet perifusions on mice lacking VEGF-A in the pancreatic epithelium in order to study their ability to secrete insulin in response to glucose. We identified insulin secretion defects in the pancreata of VEGF-A deficient mice, including a delayed and blunted response to glucose. Islet perifusion experiments revealed a missing first phase and weaker second phase of insulin secretion, in two of three VEGF-A deficient mice. On average, insulin content in VEGF-A deficient islets was significantly reduced when compared with control islets. We conclude that VEGF-A is required in pancreatic islets for normal glucose-stimulated insulin secretion and physiological insulin content. Thus, VEGF-A is a key factor for pancreatic islet function.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Arginine/pharmacology , Cells, Cultured , Down-Regulation/drug effects , Glucose/pharmacology , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Mice , Mice, Knockout , Time Factors , Vascular Endothelial Growth Factor A/metabolism
6.
Horm Metab Res ; 36(11-12): 822-9, 2004.
Article in English | MEDLINE | ID: mdl-15655714

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a potent incretin hormone currently under investigation for use as a novel therapeutic agent in the treatment of type 2 diabetes. One of several therapeutically important biological actions of GLP-1 in type 2 diabetic subjects is ability to induce strong suppression of glucagon secretion. The glucagonostatic action of GLP-1 results from its interaction with a specific G-protein coupled receptor resulting in the activation of adenylate cyclase and an increase in cAMP generation. In the pancreatic alpha-cell, cAMP, via activation of protein kinase A, interacts with a plethora of signal transduction processes including ion-channel activity and exocytosis of the glucagon-containing granules. In this short review, we will focus on recent advances in our understanding on the cellular mechanisms proposed to underlie the glucagonotropic action of GLP-1 and attempt to incorporate this knowledge into a working model for the control of glucagon secretion. Studies on the effects of GLP-1 on glucagon secretion are relevant to the pathogenesis of type 2 diabetes due to the likely contribution of hyperglucagonemia to impaired glucose tolerance in type 2 diabetes.


Subject(s)
Glucagon/metabolism , Glucagon/physiology , Islets of Langerhans/metabolism , Peptide Fragments/physiology , Protein Precursors/physiology , Animals , Diabetes Mellitus, Type 2/physiopathology , Glucagon-Like Peptide 1 , Humans , Receptors, Glucagon/metabolism
7.
Ann N Y Acad Sci ; 1009: 332-40, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15028607

ABSTRACT

The effect of the imidazoline compound LY374284 has been studied in pancreatic islets of db/db mice, a progressive model of diabetes. In perifusion experiments, pancreatic islets of db/db mice showed a progressive deterioration of glucose-induced insulin release with increasing age, whereby the first phase of insulin secretion was almost completely abolished and the second phase was substantially decreased by 15 weeks of age. LY374284 restored the first phase of glucose-induced insulin secretion in islets of 16-week-old db/db mice to 70% of that observed in islets isolated from age-matched nondiabetic db/1 mice. LY374284 did not affect insulin secretion at a low glucose concentration (3.3 mmol/L). A similar restoration of first phase insulin secretion was observed after application of glucagon-like peptide-1, whereas a sulfonylurea agent, tolbutamide, was inactive. LY374284 did not affect cytosolic Ca(2+) concentration or cellular ATP content. Furthermore, LY374284 strongly enhanced insulin secretion in islets of db/db and db/1 mice maximally depolarized by 30 mmol/L K(+) and 250 micromol/L diazoxide. The present data suggest that the imidazoline compound LY374284 restores biphasic insulin secretion in islets of diabetic db/db mice by amplifying glucose-induced insulin secretion at a site distal to Ca(2+)-influx.


Subject(s)
Diabetes Mellitus/metabolism , Imidazoles/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Culture Techniques , Disease Models, Animal , Glucose/metabolism , Humans , Imidazoles/chemistry , Insulin Secretion , Islets of Langerhans/drug effects , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL
8.
Diabetes Nutr Metab ; 15(6 Suppl): 15-8, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12702003

ABSTRACT

To investigate the hormonal and cellular selectivity of the prandial glucose regulators, we have undertaken a series of experiments, in which we characterised the effects of repaglinide and nateglinide on ATP-sensitive potassium ion (KATP) channel activity, membrane potential and exocytosis in rat pancreatic alpha-cells and somatotrophs. We found a pharmacological dissociation between the actions on KATP channels and exocytosis and suggest that compounds that, unlike repaglinide, have direct stimulatory effects on exocytosis in somatotrophs and alpha- and beta-cells, such as sulphonylureas and nateglinide, may have a clinically undesirable general stimulatory effect on cells within the endocrine system.


Subject(s)
Carbamates/pharmacology , Exocytosis/drug effects , Hypoglycemic Agents/pharmacology , Islets of Langerhans/metabolism , Piperidines/pharmacology , Potassium Channels/drug effects , Animals , Cyclohexanes/pharmacology , Electrophysiology , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Membrane Potentials/drug effects , Nateglinide , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Postprandial Period , Potassium Channels/metabolism , Rats
9.
Biochem Biophys Res Commun ; 288(2): 309-12, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11606044

ABSTRACT

Clonidine-displacing substance (CDS) is a potent stimulator of insulin release from pancreatic beta-cells and has been suggested to constitute the endogenous ligand for the islet imidazoline-binding site. Here we have explored the effects of CDS on glucagon release from mouse pancreatic alpha-cells. CDS (5 U/ml) produced a 35% inhibition (P < 0.05) of glucagon release from intact islets. This effect was dose-dependent and half-maximal inhibition by CDS was observed at 0.03 U/ml. Inhibition of glucagon release was not associated with a change in whole-cell ATP-sensitive K(+)-channel activity in single alpha-cells. However, during intracellular application through the recording pipette, CDS produced a 36% (P < 0.05) decrease in the rate of exocytosis, measured as changes in cell capacitance. The inhibitory effect of CDS on exocytosis resulted from activation of the protein phosphatase calcineurin and was abolished by cyclosporin A. These data provide further evidence for a role of CDS as an endogenous ligand controlling islet hormone secretion.


Subject(s)
Clonidine/analogs & derivatives , Clonidine/pharmacology , Exocytosis/drug effects , Glucagon/metabolism , Islets of Langerhans/drug effects , Animals , Calcium/metabolism , Electrophysiology , Exocytosis/physiology , Female , In Vitro Techniques , Islets of Langerhans/metabolism , Membrane Proteins/physiology , Mice , Patch-Clamp Techniques , Potassium Channels
10.
J Physiol ; 535(Pt 2): 519-32, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11533141

ABSTRACT

1. Measurements of cell capacitance were used to investigate the molecular mechanisms by which somatostatin inhibits Ca(2+)-induced exocytosis in single rat glucagon-secreting pancreatic alpha-cells. 2. Somatostatin decreased the exocytotic responses elicited by voltage-clamp depolarisations by 80 % in the presence of cyclic AMP-elevating agents such as isoprenaline and forskolin. Inhibition was time dependent and half-maximal within 22 s. 3. The inhibitory action of somatostatin was concentration dependent with an IC(50) of 68 nM and prevented by pretreatment of the cells with pertussis toxin. The latter effect was mimicked by intracellular dialysis with specific antibodies to G(i1/2) and by antisense oligonucleotides against G proteins of the subtype G(i2). 4. Somatostatin lacked inhibitory action when applied in the absence of forskolin or in the presence of the L-type Ca(2+) channel blocker nifedipine. The size of the omega-conotoxin-sensitive and forskolin-independent component of exocytosis was limited to 60 fF. By contrast, somatostatin abolished L-type Ca(2+) channel-dependent exocytosis in alpha-cells exposed to forskolin. The magnitude of the latter pool amounted to 230 fF. 5. The inhibitory effect of somatostatin on exocytosis was mediated by activation of the serine/threonine protein phosphatase calcineurin and was prevented by pretreatment with cyclosporin A and deltamethrin or intracellularly applied calcineurin autoinhibitory peptide. Experiments using the stable ATP analogue AMP-PCP indicate that somatostatin acts by depriming of granules. 6. We propose that somatostatin receptors associate with L-type Ca(2+) channels and couple to G(i2) proteins leading to a localised activation of calcineurin and depriming of secretory granules situated close to the L-type Ca(2+) channels.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Calcineurin/metabolism , Exocytosis/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Islets of Langerhans/metabolism , Proto-Oncogene Proteins/metabolism , Somatostatin/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Colforsin/pharmacology , Cytoplasm/metabolism , Exocytosis/drug effects , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nifedipine/pharmacology , Oligoribonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Rats , Rats, Inbred Lew , Secretory Vesicles/metabolism , omega-Conotoxin GVIA/pharmacology
11.
Biochem Biophys Res Commun ; 284(4): 918-22, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11409880

ABSTRACT

Effects of the imidazoline compound RX871024 on cytosolic free Ca(2+) concentration ([Ca(2+)]i) and insulin secretion in pancreatic beta-cells from SUR1 deficient mice have been studied. In beta-cells from wild-type mice RX871024 increased [Ca(2+)]i by blocking ATP-dependent K(+)-current (K(ATP)) and inducing membrane depolarization. In beta-cells lacking a component of the K(ATP)-channel, SUR1 subunit, RX871024 failed to increase [Ca(2+)]i. However, insulin secretion in these cells was strongly stimulated by the imidazoline. Thus, a major component of the insulinotropic activity of RX871024 is stimulation of insulin exocytosis independently from changes in K(ATP)-current and [Ca(2+)]i. This means that effects of RX871024 on insulin exocytosis are partly mediated by interaction with proteins distinct from those composing the K(ATP)-channel.


Subject(s)
Calcium/metabolism , Imidazoles/pharmacology , Indoles/pharmacology , Insulin/metabolism , Islets of Langerhans/physiology , Potassium Channels, Inwardly Rectifying , Potassium Channels/physiology , Animals , Cells, Cultured , Exocytosis/drug effects , Exocytosis/physiology , Female , In Vitro Techniques , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Oocytes/drug effects , Oocytes/physiology , Potassium Channel Blockers , Potassium Channels/deficiency , Potassium Channels/genetics , Promoter Regions, Genetic , Reference Values , Xenopus laevis
12.
Pflugers Arch ; 442(1): 19-26, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11374064

ABSTRACT

Somatostatin hyperpolarized rat pancreatic alpha-cells and inhibited spontaneous electrical activity by activating a low-conductance K+ channel (0.9 pS with physiological ionic gradients). This channel was insensitive to tolbutamide (a blocker of ATP-sensitive K+ channels) and apamin (an inhibitor of small-conductance Ca(2+)-activated K+ channels). Channel activation was prevented by pre-treating the cells with pertussis toxin, indicating the involvement of G-proteins. A direct interaction between an inhibitory G-protein and the somatostatin-activated K+ channel is suggested by the finding that intracellular application of guanosine 5'-O-(3-thiotriphosphate) (GTP gamma-S) and the G beta gamma subunit of G-proteins resulted in a transient stimulation of the current. Activation of the K+ current by somatostatin was inhibited by intracellular dialysis with specific antibodies to Gi1/2 and was not seen in cells treated with antisense oligonucleotides against G-proteins of the subtype Gi2. We conclude that somatostatin suppresses alpha-cell electrical activity by a Gi2-protein-dependent mechanism, which culminates in the activation of a sulphonylurea- and apamin-insensitive low-conductance K+ channel.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Islets of Langerhans/physiology , Potassium Channels/physiology , Receptors, Somatostatin/physiology , Animals , Antibodies/pharmacology , Apamin/pharmacology , Calcium/pharmacology , Dialysis , Electric Conductivity , GTP-Binding Protein alpha Subunits, Gi-Go/immunology , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Male , Pertussis Toxin , Potassium Channel Blockers , Potassium Channels/drug effects , Rats , Rats, Inbred Lew , Somatostatin/pharmacology , Tolbutamide/pharmacology , Virulence Factors, Bordetella/pharmacology
13.
Arch Oral Biol ; 46(4): 365-73, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11269870

ABSTRACT

Stimulation of muscarinic cholinergic receptors on rat parotid acinar cells causes a rapid production of inositol phosphates, with the key metabolic event being the breakdown of phosphatidylinositol 4,5-bisphosphate into inositol 1,4,5-trisphosphate (Ins(1,4,5)P3) and diacylglycerol. Here a high-performance liquid chromatographic technique was used to measure the effects of intracellular lithium ions on the amount of various inositol phosphates produced. When acini were stimulated maximally with acetylcholine (ACh), the sum of all inositol phosphates produced followed a monoexponential function with a production rate constant for Ins(1,4,5)P3 of 0.07 +/- 0.01 solidus/sec. The presence of 23 mM LiCl intracellularly reduced the production rate constant of Ins(1,4,5)P3 induced by ACh to 0.03 +/- 0.01 solidus/sec, resulting in a decrease in the Ins(1,4,5)P3 production as well as in the magnitude of the rise in the intracellular free Ca2+ concentration. The lithium ion (Li+) did not affect the rate of conversion of Ins(1,4,5)P3 to either inositol 1,4-bisphosphate or inositol 1,3,4,5-tetrakisphosphate. The rate of the inositol phosphate production after the addition of the Ca2+ ionophore ionomycin was unaffected by intracellular Li+ (23 mM), which implies that the action of Li+ was at the muscarinic cholinergic receptor, on G-protein or on the interactions between G-proteins and phospholipase C. Thus, in the early events after receptor stimulation with ACh, Li+ causes a reduction in the concentration of the cellular messengers Ins(1,4,5)P3 and Ca2+.


Subject(s)
Inositol 1,4,5-Trisphosphate/antagonists & inhibitors , Lithium/pharmacology , Parotid Gland/drug effects , Acetylcholine/pharmacology , Animals , Calcium Signaling/drug effects , Inositol 1,4,5-Trisphosphate/metabolism , Male , Parotid Gland/metabolism , Rats , Rats, Wistar , Receptors, Muscarinic/drug effects
14.
J Biol Chem ; 276(2): 924-30, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-10995774

ABSTRACT

Capacitance measurements were used to investigate the molecular mechanisms by which imidazoline compounds inhibit glucagon release in rat pancreatic alpha-cells. The imidazoline compound phentolamine reversibly decreased depolarization-evoked exocytosis >80% without affecting the whole-cell Ca(2+) current. During intracellular application through the recording pipette, phentolamine produced a concentration-dependent decrease in the rate of exocytosis (IC(50) = 9.7 microm). Another imidazoline compound, RX871024, exhibited similar effects on exocytosis (IC(50) = 13 microm). These actions were dependent on activation of pertussis toxin-sensitive G(i2) proteins but were not associated with stimulation of ATP-sensitive K(+) channels or adenylate cyclase activity. The inhibitory effect of phentolamine on exocytosis resulted from activation of the protein phosphatase calcineurin and was abolished by cyclosporin A and deltamethrin. Exocytosis was not affected by intracellular application of specific alpha(2), I(1), and I(2) ligands. Phentolamine reduced glucagon release (IC(50) = 1.2 microm) from intact islets by 40%, an effect abolished by pertussis toxin, cyclosporin A, and deltamethrin. These data suggest that imidazoline compounds inhibit glucagon secretion via G(i2)-dependent activation of calcineurin in the pancreatic alpha-cell. The imidazoline binding site is likely to be localized intracellularly and probably closely associated with the secretory granules.


Subject(s)
Calcineurin/metabolism , Cystamine/analogs & derivatives , Exocytosis/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Glucagon/metabolism , Islets of Langerhans/physiology , Oligodeoxyribonucleotides, Antisense/pharmacology , Phentolamine/pharmacology , Potassium Channels/physiology , Proto-Oncogene Proteins/metabolism , Adenylate Cyclase Toxin , Animals , Cells, Cultured , Clorgyline/pharmacology , Cyclosporine/pharmacology , Cystamine/pharmacology , Diazoxide/pharmacology , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Imidazoles/pharmacology , Indoles/pharmacology , Islets of Langerhans/drug effects , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Nitriles , Pertussis Toxin , Potassium Channels/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrethrins/pharmacology , Rats , Rats, Inbred Lew , Virulence Factors, Bordetella/pharmacology
15.
J Physiol ; 528(Pt 3): 509-20, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11060128

ABSTRACT

The perforated patch whole-cell configuration of the patch-clamp technique was applied to superficial glucagon-secreting alpha-cells in intact mouse pancreatic islets. alpha-cells were distinguished from the beta- and delta-cells by the presence of a large TTX-blockable Na+ current, a TEA-resistant transient K+ current sensitive to 4-AP (A-current) and the presence of two kinetically separable Ca2+ current components corresponding to low- (T-type) and high-threshold (L-type) Ca2+ channels. The T-type Ca2+, Na+ and A-currents were subject to steady-state voltage-dependent inactivation, which was half-maximal at -45, -47 and -68 mV, respectively. Pancreatic alpha-cells were equipped with tolbutamide-sensitive, ATP-regulated K+ (KATP) channels. Addition of tolbutamide (0.1 mM) evoked a brief period of electrical activity followed by a depolarisation to a plateau of -30 mV with no regenerative electrical activity. Glucagon secretion in the absence of glucose was strongly inhibited by TTX, nifedipine and tolbutamide. When diazoxide was added in the presence of 10 mM glucose, concentrations up to 2 microM stimulated glucagon secretion to the same extent as removal of glucose. We conclude that electrical activity and secretion in the alpha-cells is dependent on the generation of Na+-dependent action potentials. Glucagon secretion depends on low activity of KATP channels to keep the membrane potential sufficiently negative to prevent voltage-dependent inactivation of voltage-gated membrane currents. Glucose may inhibit glucagon release by depolarising the alpha-cell with resultant inactivation of the ion channels participating in action potential generation.


Subject(s)
Adenosine Triphosphate/physiology , Glucagon/metabolism , Islets of Langerhans/metabolism , Potassium Channels/physiology , Sodium Channel Blockers , Sodium Channels/drug effects , Tetrodotoxin/pharmacology , Animals , Calcium Channels/physiology , Drug Resistance , Electrophysiology , Homeostasis , Ion Channel Gating , Mice , Potassium Channels/drug effects , Sodium Channels/physiology , Tetraethylammonium/pharmacology
16.
Eur J Pharmacol ; 403(3): 221-4, 2000 Sep 08.
Article in English | MEDLINE | ID: mdl-10973622

ABSTRACT

Using capacitance measurements, we have explored the effects of three different scinderin actin-binding peptides (Sc(77-89); Sc(138-146); Sc(511-523)) on Ca(2+)- and GTPgammaS-induced exocytosis in single mouse pancreatic beta-cells. Sc(77-89) (10 microM) reduced exocytosis by 43% in whole-cell experiments in which secretion was triggered by intracellular dialysis with a Ca(2+)-EGTA buffer with a free Ca(2+) concentration of 2 microM. A more pronounced reduction of the rate of exocytosis was observed with Sc(138-146) (72%) but not with Sc(511-523) (39%). Sc(138-146) also reduced depolarisation-induced exocytosis by 61% without affecting the whole-cell Ca(2+) current. When exocytosis was triggered by infusion of GTPgammaS, all scinderin-binding peptides reduced exocytosis by 59-75%. These data suggest that scinderin might be important for controlling cortical actin network dynamics in mouse pancreatic beta-cells and that scinderin-induced cortical filamentous actin disassembly is required for insulin secretion.


Subject(s)
Calcium/antagonists & inhibitors , Exocytosis/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/antagonists & inhibitors , Islets of Langerhans/drug effects , Microfilament Proteins/pharmacology , Animals , Cells, Cultured , Gelsolin , Insulin/metabolism , Insulin Secretion , Mice , Microfilament Proteins/chemistry , Patch-Clamp Techniques
17.
J Physiol ; 527 Pt 1: 109-20, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10944174

ABSTRACT

1. Capacitance measurements were used to examine the effects of the sulphonylurea tolbutamide on Ca2+-dependent exocytosis in isolated glucagon-secreting rat pancreatic A-cells. 2. When applied extracellularly, tolbutamide stimulated depolarization-evoked exocytosis 4.2-fold without affecting the whole-cell Ca2+ current. The concentration dependence of the stimulatory action was determined by intracellular application through the recording pipette. Tolbutamide produced a concentration-dependent increase in cell capacitance. Half-maximal stimulation was observed at 33 microM and the maximum stimulation corresponded to a 3.4-fold enhancement of exocytosis. 3. The stimulatory action of tolbutamide was dependent on protein kinase C activity. The action of tolbutamide was mimicked by the general K+ channel blockers TEA (10 mM) and quinine (10 microM). A similar stimulation was elicited by 5-hydroxydecanoate (5-HD; 10 microM), an inhibitor of mitochondrial ATP-sensitive K+ (KATP) channels. 4. Tolbutamide-stimulated, but not TEA-induced, exocytosis was antagonized by the K+ channel openers diazoxide, pinacidil and cromakalim. 5. Dissipating the transgranular K+ gradient with nigericin and valinomycin inhibited tolbutamide- and Ca2+-evoked exocytosis. Furthermore, tolbutamide- and Ca2+-induced exocytosis were abolished by the H+ ionophore FCCP or by arresting the vacuolar (V-type) H+-ATPase with bafilomycin A1 or DCCD. Finally, ammonium chloride stimulated exocytosis to a similar extent to that obtained with tolbutamide. 6. We propose that during granular maturation, a granular V-type H+-ATPase pumps H+ into the secretory granule leading to the generation of a pH gradient across the granular membrane and the development of a positive voltage inside the granules. The pumping of H+ is facilitated by the concomitant exit of K+ through granular K+ channels with pharmacological properties similar to those of mitochondrial KATP channels. Release of granules that have been primed is then facilitated by the addition of K+ channel blockers. The resulting increase in membrane potential promotes exocytosis by unknown mechanisms, possibly involving granular alkalinization.


Subject(s)
Exocytosis/drug effects , Glucagon/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Membrane Proteins/metabolism , Tolbutamide/pharmacology , Vacuolar Proton-Translocating ATPases , Animals , Calcium/metabolism , Cell Culture Techniques , Electric Conductivity , Hydrogen-Ion Concentration/drug effects , Ionophores/pharmacology , Islets of Langerhans/drug effects , Male , Membrane Potentials , Membrane Proteins/antagonists & inhibitors , Models, Biological , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Potassium/metabolism , Potassium Channels , Protein Kinase C/antagonists & inhibitors , Proton-Translocating ATPases/antagonists & inhibitors , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , Sulfonylurea Compounds/pharmacology
18.
Pflugers Arch ; 439(5): 634-45, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10764224

ABSTRACT

Capacitance measurements of exocytosis were combined with carbon fibre amperometry for time-resolved measurements of the properties of secretion in single, insulin-secreting, mouse pancreatic beta-cells pre-loaded with the amine serotonin (5-HT). Glucose-induced electrical activity was associated with the appearance of brief and transient amperometric currents reflecting the serotonin co-released with insulin. The integrated amperometric responses resulting from voltage-clamp depolarisations were proportional to the corresponding increase in cell capacitance. Both parameters exhibited U-shaped relationships to the membrane potential with maximums around 0 mV. There was a variable latency (40-730 ms, average 230 ms) between the onset of the depolarisation and the amperometric current. During high-frequency repetitive stimulation, a progressive decrease in the exocytotic capacity ("depression") was observed. This was paralleled by a corresponding reduction of the amperometric responses. Using the carbon fibre to map the beta-cell for release sites indicated that exocytosis was confined to the part of the cell containing the highest density of secretory granules. Two types of amperometric responses were observed. In about 50% of the cells, a smooth increase was observed with no discernible discrete events. In the remaining cells, the amperometric records contained large spikes. These were ten or more times larger than that expected for the fusion of individual secretory granules. We propose that these large spikes reflect the exocytosis of multigranular complexes formed inside the beta-cell prior to exocytosis.


Subject(s)
Exocytosis/physiology , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium/pharmacology , Carbon , Electric Conductivity , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Microelectrodes , Patch-Clamp Techniques , Reaction Time/drug effects , Reaction Time/physiology , Serotonin/pharmacology , Temperature
19.
J Biol Chem ; 275(13): 9363-8, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10734079

ABSTRACT

Non-insulin-dependent diabetes mellitus is associated with, in addition to impaired insulin release, elevated levels of free fatty acids (FFA) in the blood. Insulin release is stimulated when beta-cells are acutely exposed to FFA, whereas chronic exposure may inhibit glucose-induced insulin secretion. In the present study we investigated the direct effects of long chain acyl-CoA (LC-CoA), the active intracellular form of FFA, on insulin exocytosis. Palmitoyl-CoA stimulated both insulin release from streptolysin-O-permeabilized HIT cells and fusion of secretory granules to the plasma membrane of mouse pancreatic beta-cells, as measured by cell capacitance. The LC-CoA effect was chain length-dependent, requiring chain lengths of at least 14 carbons. LC-CoA needed to be present to stimulate insulin release, and consequently there was no effect following its removal. The stimulatory effect was observed after inhibition of protein kinase activity and in the absence of ATP, even though both kinases and ATP, themselves, modulate exocytosis. The effect of LC-CoA was inhibited by cerulenin, which has been shown to block protein acylation. The data suggest that altered LC-CoA levels, resulting from FFA or glucose metabolism, may act directly on the exocytotic machinery to stimulate insulin release by a mechanism involving LC-CoA protein binding.


Subject(s)
Exocytosis/drug effects , Insulin/metabolism , Islets of Langerhans/drug effects , Palmitoyl Coenzyme A/pharmacology , Animals , Cell Line , Insulin Secretion , Islets of Langerhans/metabolism , Kinetics , Mice
20.
Eur J Pharmacol ; 386(1): 105-11, 1999 Dec 10.
Article in English | MEDLINE | ID: mdl-10611470

ABSTRACT

The effects of the two prandial glucose regulators, repaglinide and nateglinide, on ATP-sensitive K(+) (K(ATP)) channel activity, membrane potential and exocytosis in single rat pancreatic A-cells were investigated using the patch-clamp technique. K(ATP) channel activity was reversibly blocked by repaglinide (K(d)=22 nM) and nateglinide (K(d)=410 nM) and this was associated with membrane depolarisation and initiation of electrical activity. The effect of repaglinide and nateglinide on stimulation of glucagon secretion by direct interference with the exocytotic machinery was investigated by the use of capacitance measurements. Nateglinide, but not repaglinide, at concentrations similar to those required to block K(ATP) channels potentiated Ca(2+)-evoked exocytosis 3-fold. In alphaTC1-9 glucagonoma cells addition of nateglinide, but not repaglinide, was associated with stimulation of glucagon secretion. These results indicate that the fast-acting insulin secretagogue nateglinide is glucagonotropic primarily by stimulating Ca(2+)-dependent exocytosis.


Subject(s)
Carbamates/pharmacology , Cyclohexanes/pharmacology , Exocytosis/drug effects , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Islets of Langerhans/drug effects , Phenylalanine/analogs & derivatives , Piperidines/pharmacology , ATP-Binding Cassette Transporters , Animals , Calcium/antagonists & inhibitors , Calcium/pharmacology , Eating/physiology , Electrophysiology , Glucagon/metabolism , In Vitro Techniques , Islets of Langerhans/metabolism , KATP Channels , Kinetics , Male , Nateglinide , Patch-Clamp Techniques , Phenylalanine/pharmacology , Potassium Channel Blockers , Potassium Channels/metabolism , Potassium Channels, Inwardly Rectifying , Rats , Rats, Inbred Lew , Stimulation, Chemical
SELECTION OF CITATIONS
SEARCH DETAIL
...