Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Diabetologia ; 55(9): 2432-44, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22760787

ABSTRACT

AIMS/HYPOTHESIS: Hypothalamic glucose-excited (GE) neurons contribute to whole-body glucose homeostasis and participate in the detection of hypoglycaemia. This system appears defective in type 1 diabetes, in which hypoglycaemia commonly occurs. Unfortunately, it is at present unclear which molecular components required for glucose sensing are produced in individual neurons and how these are functionally linked. We used the GT1-7 mouse hypothalamic cell line to address these issues. METHODS: Electrophysiological recordings, coupled with measurements of gene expression and protein levels and activity, were made from unmodified GT1-7 cells and cells in which AMP-activated protein kinase (AMPK) catalytic subunit gene expression and activity were reduced. RESULTS: Hypothalamic GT1-7 neurons express the genes encoding glucokinase and ATP-sensitive K(+) channel (K(ATP)) subunits K ( ir ) 6.2 and Sur1 and exhibit GE-type glucose-sensing behaviour. Lowered extracellular glucose concentration hyperpolarised the cells in a concentration-dependent manner, an outcome that was reversed by tolbutamide. Inhibition of glucose uptake or metabolism hyperpolarised cells, showing that energy metabolism is required to maintain their resting membrane potential. Short hairpin (sh)RNA directed to Ampkα2 (also known as Prkaa2) reduced GT1-7 cell AMPKα2, but not AMPKα1, activity and lowered the threshold for hypoglycaemia-induced hyperpolarisation. shAmpkα1 (also known as Prkaa1) had no effect on glucose-sensing or AMPKα2 activity. Decreased uncoupling protein 2 (Ucp2) mRNA was detected in AMPKα2-reduced cells, suggesting that AMPKα2 regulates UCP2 levels. CONCLUSIONS/INTERPRETATION: We have demonstrated that GT1-7 cells closely mimic GE neuron glucose-sensing behaviour, and reducing AMPKα2 blunts their responsiveness to hypoglycaemic challenge, possibly by altering UCP2 activity. These results show that suppression of AMPKα2 activity inhibits normal glucose-sensing behaviour and may contribute to defective detection of hypoglycaemia.


Subject(s)
AMP-Activated Protein Kinases/genetics , Cell Line/metabolism , Hypoglycemia/genetics , Hypothalamus/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Hypoglycemia/physiopathology , Insulin Secretion , Ion Channels/metabolism , Mice , Mitochondrial Proteins/metabolism , RNA, Small Interfering/metabolism , Signal Transduction , Uncoupling Protein 2
2.
Neuroscience ; 159(1): 83-93, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19135134

ABSTRACT

A number of anti-obesity agents have been developed that enhance hypothalamic 5-HT transmission. Various studies have demonstrated that arcuate neurons, which express proopiomelanocortin peptides (POMC neurons), and neuropeptide Y with agouti-related protein (NPY/AgRP) neurons, are components of the hypothalamic circuits responsible for energy homeostasis. An additional arcuate neuron population, rat insulin 2 promoter Cre recombinase transgene (RIPCre) neurons, has recently been implicated in hypothalamic melanocortin circuits involved in energy balance. It is currently unclear how 5-HT modifies neuron excitability in these local arcuate neuronal circuits. We show that 5-HT alters the excitability of the majority of mouse arcuate RIPCre neurons, by either hyperpolarization and inhibition or depolarization and excitation. RIPCre neurons sensitive to 5-HT, predominantly exhibit hyperpolarization and pharmacological studies indicate that inhibition of neuronal firing is likely to be through 5-HT(1F) receptors increasing current through a voltage-dependent potassium conductance. Indeed, 5-HT(1F) receptor immunoreactivity co-localizes with RIPCre green fluorescent protein expression. A minority population of POMC neurons also respond to 5-HT by hyperpolarization, and this appears to be mediated by the same receptor-channel mechanism. As neither POMC nor RIPCre neuronal populations display a common electrical response to 5-HT, this may indicate that sub-divisions of POMC and RIPCre neurons exist, perhaps serving different outputs.


Subject(s)
Arcuate Nucleus of Hypothalamus/cytology , Neural Inhibition/drug effects , Neurons/drug effects , Neurons/physiology , Pro-Opiomelanocortin/metabolism , Serotonin/pharmacology , Action Potentials/drug effects , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Biophysical Phenomena/drug effects , Electric Stimulation/methods , Green Fluorescent Proteins/genetics , In Vitro Techniques , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Integrases/genetics , Integrases/metabolism , Mice , Mice, Transgenic , Neuropeptide Y/genetics , Patch-Clamp Techniques/methods , Potassium Channel Blockers/pharmacology , Potassium Channels/physiology , Pro-Opiomelanocortin/genetics , Rats , Serotonin Antagonists/pharmacology , Time Factors
3.
Nat Neurosci ; 3(8): 757-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10903566

ABSTRACT

Insulin and leptin receptors are present in hypothalamic regions that control energy homeostasis, and these hormones reduce food intake and body weight in lean, but not obese, Zucker rats. Here we demonstrate that insulin, like leptin, hyperpolarizes lean rat hypothalamic glucose-responsive (GR) neurons by opening KATP channels. These findings suggest hypothalamic K ATP channel function is crucial to physiological regulation of food intake and body weight.


Subject(s)
Adenosine Triphosphate/pharmacology , Hypothalamus/physiology , Insulin/pharmacology , Neurons/physiology , Obesity/physiopathology , Potassium Channels/physiology , Animals , Arcuate Nucleus of Hypothalamus/physiology , Glucose/pharmacology , Hypothalamus/physiopathology , In Vitro Techniques , Leptin/pharmacology , Membrane Potentials/drug effects , Obesity/genetics , Patch-Clamp Techniques , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels/drug effects , Rats , Rats, Zucker , Thinness , Tolbutamide/pharmacology
4.
Diabetes ; 37(5): 495-8, 1988 May.
Article in English | MEDLINE | ID: mdl-2452107

ABSTRACT

Since their discovery in pancreatic beta-cells, ATP-sensitive K+ channels in the cell membrane have been thought to mediate glucose-induced beta-cell depolarization, which is required for triggering the voltage-dependent Ca2+ uptake subserving insulin release. The theory is that metabolism of glucose (and other fuel molecules) increases intracellular ATP or possibly other metabolites that diffuse to the membrane and inhibit the opening of ATP-sensitive K+ channels. This slows the efflux of positively charged K+ and depolarizes the cell. A recurrent source of confusion regarding this idea stems from the early observation that these channels are so exquisitely sensitive to intracellular ATP that channel opening is predicted to be approximately 99% inhibited under physiological conditions. To account for this apparent discrepancy, various mechanisms have been proposed that might render the channels less sensitive to intracellular ATP. We use a simple mathematical model to demonstrate that there is no major discrepancy and that, in fact, given the electrophysiological mechanisms existing in the beta-cell, the extreme sensitivity of the channels to ATP is appropriate and even mandatory for their physiological function.


Subject(s)
Adenosine Triphosphate/physiology , Ion Channels/physiology , Islets of Langerhans/physiology , Electrophysiology , Glucose/metabolism , Humans , Membrane Potentials
5.
J Neuroendocrinol ; 17(4): 246-54, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15842236

ABSTRACT

Derivatives of 3-guanidinopropionic acid, such as leptin, reduce body weight in obese, diabetic mice. We have assessed whether one of these analogues, BVT.12777 activates intracellular signalling pathways in the arcuate nucleus in a manner analogous to leptin and insulin. In addition, because these hormones have been shown to activate K(ATP) channels in a subset of arcuate neurones, we examined whether this channel is also a functional endpoint for BVT.12777 in the arcuate nucleus. BVT.12777 transiently increased phosphorylation of MAPK, STAT3, PKB and GSK3, in a manner identical to that observed for leptin and insulin. BVT.12777 also hyperpolarized glucose-responsive neurones by increasing the activity of K(ATP) channels. The increase in K(ATP) activity driven by BVT.12777 was PI3-kinase independent, unlike leptin and insulin activation of this channel, and could also be elicited in isolated patches. However, K(ATP) activity induced by BVT.12777 was dependent on actin filament dynamics, both in intact neurones and isolated patches. Thus, BVT.12777 modulates arcuate neurone K(ATP) activity by re-organization of the cytoskeleton, a mechanism that has also been ascribed to leptin and insulin. Consequently, BVT.12777 appears to act as a leptin and insulin mimetic with respect to at least some elements of arcuate neurone intracellular signalling and the activation of K(ATP) channels. Resistance to leptin and insulin, associated with obesity has, at least in part, been postulated to be due to aberrant intracellular signalling in arcuate neurones. The data presented here indicate that it may be possible to develop drugs, which by-pass up-stream signalling components associated with adiposity hormone resistance, such as PI3-kinase, but can still induce functional outputs from arcuate neurones by targeting downstream components of the leptin and insulin signalling cascades.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Enzyme Activators/pharmacology , Guanidines/pharmacology , Neurons/drug effects , Potassium Channels, Inwardly Rectifying/drug effects , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/enzymology , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Insulin/metabolism , Leptin/metabolism , MAP Kinase Kinase 2/drug effects , MAP Kinase Kinase 2/metabolism , Male , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/enzymology , Organ Culture Techniques , Patch-Clamp Techniques , Phosphorylation/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor , Signal Transduction/drug effects , Trans-Activators/drug effects , Trans-Activators/metabolism
6.
FEBS Lett ; 208(2): 397-400, 1986 Nov 24.
Article in English | MEDLINE | ID: mdl-2430835

ABSTRACT

The effects of adenosine and adenine nucleotides on a calcium-activated non-selective cation channel, present in the plasma membrane of an insulin-secreting cell line CRI-Gl were investigated. Single-channel currents were recorded from inside-out membrane patches and the adenine derivatives applied to the solution bathing the cytoplasmic aspect of the membrane surface. The activity of this channel is shown to be inhibited by all the derivatives tested. The potency sequence for inhibition was found to be AMP greater than ADP greater than ATP greater than adenosine.


Subject(s)
Adenine Nucleotides/pharmacology , Adenoma, Islet Cell/physiopathology , Adenosine/pharmacology , Calcium/physiology , Insulinoma/physiopathology , Ion Channels/drug effects , Animals , Cations , Cell Line , Cell Membrane/physiology , Cytoplasm/physiology , Rats
7.
FEBS Lett ; 374(1): 135-40, 1995 Oct 23.
Article in English | MEDLINE | ID: mdl-7589501

ABSTRACT

The distribution of mRNA encoding the inwardly rectifying K+ channel, BIR1 [1] was investigated in rat tissues, and a comparison made with the expression of related genes rcKATP and GIRK1 using the reverse transcription-polymerase chain reaction (RT-PCR). This showed BIR1 to be expressed in all areas of the brain examined, in the eye but not in any other peripheral tissue. This pattern was distinct from rcKATP and GIRK1. Additional in situ hybridisation studies of the central expression of BIR1 demonstrated high levels of BIR1 mRNA in the hippocampus, dentate gyrus, taenia tecta and cerebellum and at lower levels in the cortex, habenular nucleus, olfactory bulb, primary olfactory cortex, thalamus, pontine nucleus and amygdaloid nucleus.


Subject(s)
Potassium Channels, Inwardly Rectifying , Potassium Channels/genetics , RNA, Messenger/metabolism , Animals , Base Sequence , Brain/metabolism , DNA Primers , DNA Probes , G Protein-Coupled Inwardly-Rectifying Potassium Channels , In Situ Hybridization , Molecular Sequence Data , Polymerase Chain Reaction , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley
8.
Neuropharmacology ; 44(7): 845-54, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12726816

ABSTRACT

The discovery of the obese gene product, leptin has generated enormous interest in how the periphery signals the status of nutritional stores to specific hypothalamic nuclei involved in regulating feeding and energy balance. However it is emerging that leptin, in addition to its role as a circulating satiety factor, is a multi-faceted hormone that plays a key role in a variety of CNS functions. In this review, we summarise recent progress in leptin biology, with particular focus on its diversity of actions within the CNS, ranging from satiety signal, to regulator of bone formation and inhibitor of neuronal excitability.


Subject(s)
Central Nervous System/physiology , Leptin/physiology , Satiety Response/physiology , Animals , Hippocampus/physiology , Humans , Hypothalamus/physiology , Obesity/genetics , Obesity/physiopathology , Receptors, Cell Surface/physiology , Receptors, Leptin , Signal Transduction/physiology
9.
Neuroscience ; 40(2): 523-31, 1991.
Article in English | MEDLINE | ID: mdl-1902917

ABSTRACT

Sulphonylureas such as glibenclamide, which are used in the treatment of Type-2 diabetes, are inhibitors of ATP-sensitive potassium channels. These channels link cellular metabolism to membrane electrical activity and it is likely that they are closely associated with glibenclamide binding sites. Quantitative autoradiography was used to localize high-affinity [3H]glibenclamide binding sites in coronal sections of rat brain. The relative density of binding sites was found to correlate well with the relative capacity of sites determined in homogenate assays. There was no evidence of any variation of affinity between brain regions. The highest levels of binding were found in the substantia nigra with high levels in the globus pallidus, cerebral cortex, hippocampus and caudate-putamen, intermediate levels in the cerebellum, and low levels in the hypothalamus and pons. The density of [3H]glibenclamide binding sites was low in glucose-responsive brain regions, known to contain ATP-sensitive potassium channels that are inhibited by sulphonylureas. However, higher densities were associated with brain regions (often limbic structures) active during temporal lobe epilepsy. In at least two of these structures, the CA3 region of the hippocampus and the substantia nigra, it is probable that these sites are coupled to ATP-sensitive potassium channels. These results are discussed with reference to the reported actions of ATP-sensitive potassium channels on CNS function.


Subject(s)
Brain/metabolism , Sulfonylurea Compounds/metabolism , Animals , Autoradiography , Binding Sites , Glyburide/metabolism , Male , Rats , Rats, Inbred Strains , Time Factors , Tissue Distribution
10.
Neuroscience ; 95(1): 33-50, 2000.
Article in English | MEDLINE | ID: mdl-10619460

ABSTRACT

Inside-out patch recordings from rat acutely dissociated cerebral cortical neurons revealed time and voltage-dependent activity of a large-conductance calcium-activated potassium channel. Channel activity inactivated within minutes following a depolarizing voltage step, and was recovered from inactivation by membrane hyperpolarization. Inactivation rate was not influenced by internal calcium or membrane voltage; however, reducing channel activity with intracellular calcium destabilized inactivation. Channel inactivation was abolished by intracellular trypsin treatment, suggesting that an associated inactivating particle was responsible for inactivation. Application of alkaline phosphatase to the internal aspect of the patch membrane increased channel activity and abolished channel inactivation, without affecting its voltage and calcium dependence. Internal application of Mg-ATP, but not Mg-5'-adenylylamidodiphosphate, retarded recovery of channel activity from inactivation, whereas internal application of protein phosphatase-1alpha enhanced recovery from inactivation. The abolition of channel inactivation by alkaline phosphatase was prevented by prior internal tetraethylammonium treatment, indicating that the alkaline phosphatase site is closely associated with the channel pore. These results demonstrate that cortical large-conductance calcium-activated potassium channel inactivation is probably mediated by an endogenous, trypsin-sensitive, inactivation particle. This particle appears to inactivate the open channel and requires a critical phosphate group for stable block. The slow time-course of channel inactivation may have some pathophysiological significance in maintenance of epileptiform activity.


Subject(s)
Calcium/physiology , Cerebral Cortex/metabolism , Neurons/physiology , Potassium Channels/physiology , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/pharmacology , Animals , Cerebral Cortex/cytology , Electric Conductivity , Intracellular Membranes/physiology , Male , Phosphorylation , Potassium Channels/drug effects , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Tetraethylammonium/pharmacology , Trypsin/pharmacology
11.
Proc Biol Sci ; 247(1319): 121-4, 1992 Feb 22.
Article in English | MEDLINE | ID: mdl-1349178

ABSTRACT

ATP-sensitive K+ (ATP-K+) channels underlie the glucose-sensing nature of pancreatic beta-cells by way of their inhibition by intracellular ATP. Recently it has been proposed that ATP-K+ channels have a similar function in certain hypothalamic neurons that become excitable in raised concentrations of extracellular glucose. The aim of this study was to assess the ATP sensitivity of ATP-K+ channels in inside-out membrane patches excised from glucose-sensing neurons that were acutely isolated from the ventromedial nucleus of rat hypothalamus. ATP-K+ channels were less sensitive to ATP in neurons than in other tissues. Moreover, the sensitivity of neuronal ATP-K+ channels to inhibition by intracellular ATP was modulated by extracellular cations. Under physiological ionic gradients (i.e. high extracellular Na+ and low K+), intracellular ATP produced a concentration-dependent inhibition of channel activity, with a half-maximal inhibition (Ki) of 2.32 mM. A non-hydrolysable analogue of ATP, AMP(PNP), was similarly effective. In symmetrical K+ (i.e. no extracellular sodium), channel activity was tenfold more sensitive to ATP (Ki of 0.21 mM). A parallel study on ATP-K+ channels from an insulin-secreting beta-cell line (CRI-G1) showed that, in contrast to the neuronal data, extracellular cations had no effect on the ATP sensitivity of the channel.


Subject(s)
Adenosine Triphosphate/metabolism , Potassium Channels/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Drug Resistance , In Vitro Techniques , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Potassium/pharmacology , Potassium Channels/drug effects , Rats , Sodium/pharmacology , Ventromedial Hypothalamic Nucleus/drug effects
12.
Proc Biol Sci ; 251(1330): 9-15, 1993 Jan 22.
Article in English | MEDLINE | ID: mdl-8094568

ABSTRACT

The conversion of the electrically silent pregnant uterus to highly excitable at term represents a dramatic physiological event which is poorly understood. Here we provide the first description, from single-channel recordings, of a large conductance (212 pS) calcium-activated potassium channel (BKCa) in human pregnant myometrium which, in labour tissue, is either absent or has been considerably altered in its physiological and pharmacological properties. In the latter, the K+ channels have an identical conductance (221 pS) and K+ selectivity to BKCa channels but exhibit no Ca2+ or voltage sensitivity. We have termed these BK channels. Furthermore, the activity of the BKCa channel from pregnant tissue is inhibited by internal application of Ba2+ but not tetraethylammonium (TEA), whereas the activity of the BK channel is sensitive to internal TEA but not Ba2+. The role of the BKCa channel may be to suppress myometrial activity during gestation whereas BK channel activity may be important in providing a Ca(2+)-independent K+ conductance which would allow cytoplasmic Ca2+ levels to rise without activating a counteracting Ca(2+)-dependent outward current, normally provided by the BKCa channels which, by its very nature, would tend to oppose depolarization. The findings suggest that K+ channels may have an important role in determining the functional activity of the myometrium.


Subject(s)
Labor, Obstetric/physiology , Myometrium/physiology , Potassium Channels/physiology , Pregnancy/physiology , Adenosine Triphosphate/pharmacology , Female , Humans , In Vitro Techniques , Membrane Potentials/drug effects , Potassium Channels/drug effects , Tetraethylammonium , Tetraethylammonium Compounds/pharmacology
13.
Proc Biol Sci ; 253(1338): 225-31, 1993 Sep 22.
Article in English | MEDLINE | ID: mdl-7694299

ABSTRACT

It is generally considered that the sulphonylurea receptor is an integral part of the ATP-sensitive K+ channel. We have investigated this proposal by comparing the binding and functional characteristics of the sulphonylurea receptor and KATP channel by using two rat insulinoma cell lines (CRI-G1 and CRI-D11) of common origin. Insulin release was increased in both cell lines by a variety of metabolizable and non-metabolizable secretagogues but glibenclamide induced an increase in insulin release in G1 cells only. [3H]glibenclamide binding studies showed a substantial reduction in the number of glibenclamide binding sites (Bmax) in the D11 cells compared with G1 cells. Single-channel studies of these cell lines show that the KATP channel is generally unchanged in its biophysical properties and in the number of channels observed. Slight differences were apparent: the KATP channels in D11 cells were much less susceptible to rundown and were slightly less sensitive to block by ATP. However, one major distinction was the lack or much reduced sensitivity of the KATP channel in D11 cells to tolbutamide and glibenclamide. We conclude that the KATP channel can exist and function independently of the sulphonylurea receptor, and therefore it is unlikely that they exist as a single protein assembly.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate/pharmacology , Insulin/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Potassium Channels/physiology , Receptors, Drug/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Cell Membrane/metabolism , Clone Cells , Glyburide/metabolism , Glyburide/pharmacology , Insulin Secretion , Insulinoma , Membrane Potentials/drug effects , Pancreatic Neoplasms , Potassium/pharmacology , Potassium Channels/drug effects , Rats , Receptors, Drug/drug effects , Sulfonylurea Receptors , Tetradecanoylphorbol Acetate/pharmacology , Tolbutamide/pharmacology , Tumor Cells, Cultured
14.
Br J Pharmacol ; 113(3): 659-61, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7858852

ABSTRACT

NS 1619 activates the large-conductance Ca(2+)-dependent K+ channel (BKCa) in membrane patches isolated from rat ventromedial hypothalamic neurones. The activation is concentration dependent, with a maximal effect at less than 30 microM, reversible and can be inhibited by application of iberiotoxin to the extracellular membrane. NS 1619 does not activate ATP-K+ channels present in the same neurones.


Subject(s)
Benzimidazoles/pharmacology , Calcium/physiology , Potassium Channels/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Animals , Cells, Cultured , Peptides/pharmacology , Rats
15.
Br J Pharmacol ; 103(4): 2021-9, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1912991

ABSTRACT

1. Patch-clamp recording techniques were used to examine the effects of barbiturates upon the ATP-K+ channel, and voltage-activated channels present in the plasma membrane of CRI-G1 insulin-secreting cells. 2. Thiopentone inhibited ATP-K+ channel activity when applied to cell-attached patches or the intracellular or extracellular surface of cell-free patches. Secobarbitone and pentobarbitone were also effective inhibitors of ATP-K+ channels in cell-free patches, whereas phenobarbitone was ineffective. 3. The diabetogenic agent, alloxan, which is structurally related to the barbiturates also produced an inhibition of ATP-K+ channel activity in outside-out patches. 4. Whole-cell ATP-K+ currents were used to quantify the effects of the barbiturates: concentration-inhibition curves for thiopentone, secobarbitone and pentobarbitone resulted in IC50 values of 62, 250 and 360 microM respectively. Phenobarbitone at a concentration of 1 mM was virtually ineffective. 5. Calculation of the apparent membrane concentrations for these drugs indicate that for a given degree of ATP-K+ channel inhibition a similar concentration of each barbiturate is present in the membrane. This suggests that hydrophobicity plays a primary role in their mechanism of action. The pH-dependence and additive nature of barbiturate block also indicates a membrane site of action. 6. Thiopentone, (100 microM) was also found to inhibit differentially voltage-activated whole-cell currents. The relative potency of thiopentone at this concentration was 0.64, 0.38 and 0.12 for inhibiting Ca2+, K+ and Na+ currents respectively when compared with its ability to inhibit the ATP-K+ channel.


Subject(s)
Adenosine Triphosphate/physiology , Barbiturates/pharmacology , Potassium Channels/drug effects , Animals , Cells, Cultured , Diazoxide/pharmacology , Dose-Response Relationship, Drug , Islets of Langerhans/metabolism , Rats , Thiopental/pharmacology
16.
Br J Pharmacol ; 107(1): 34-43, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1422577

ABSTRACT

1. Patch-clamp recording techniques were used, to examine the effects of diazoxide on KATP currents in CRI-G1 insulin-secreting cells in the presence of non-hydrolysable nucleotides. 2. In the presence of non- or slowly-hydrolyzed ATP analogues, bathing the intracellular aspect of cell-free membrane patches diazoxide inhibited KATP channel activity. 3. Under whole-cell recording conditions, with various non-hydrolysable nucleotides present intracellularly (after dialysis), diazoxide induced KATP current activation. The largest activation occurred with Mg-adenylyl-(beta, gamma-methylene) diphosphate (Mg-AMP-PCP) present in the dialysing solution. This activation was diazoxide- and nucleotide-concentration-dependent. 4. In the absence of Mg2+, or in the presence of manganese (Mn2+) ions intracellularly, diazoxide did not induce KATP current activation, regardless of the species of nucleotide present in the pipette. 5. Intracellularly applied trypsin prevented the activation of KATP currents by diazoxide in the presence of Mg-AMP-PCP, an effect reversed by co-application of intracellular polymethylsulphonyl fluoride with the trypsin. 6. The application, by dialysis, of a CRI-G1 cell lysate, with negligible Mg-ATP, resulted in a substantial activation of the KATP current by diazoxide. 7. It is concluded that diazoxide can activate KATP channel currents by two separate pathways, one requiring a phosphorylation process, the other the presence of an intracellular protein coupled with a Mg-purine nucleotide.


Subject(s)
Adenine Nucleotides/pharmacology , Adenosine Triphosphate/metabolism , Diazoxide/pharmacology , Islets of Langerhans/drug effects , Potassium Channels/drug effects , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Cell Line , Electric Stimulation , Islets of Langerhans/metabolism , Magnesium/pharmacology , Manganese/pharmacology , Phosphorylation , Potassium Channels/physiology , Rats
17.
Br J Pharmacol ; 124(7): 1557-65, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9723971

ABSTRACT

1. The effects of the antidiabetic agent englitazone and the anorectic drug ciclazindol on ATP-sensitive K+ (K(ATP)) channels activated by diazoxide and leptin were examined in the CRI-G1 insulin-secreting cell line using whole cell and single channel recording techniques. 2. In whole cell current clamp mode, the hyperglycaemic agent diazoxide (200 microM) and the ob gene product leptin (10 nM) hyperpolarised CRI-G1 cells by activation of K(ATP) currents. K(ATP) currents activated by either agent were inhibited by tolbutamide, with an IC50 for leptin-activated currents of 9.0 microM. 3. Application of englitazone produced a concentration-dependent inhibition of K(ATP) currents activated by diazoxide (200 microM) with an IC50 value of 7.7 microM and a Hill coefficient of 0.87. In inside-out patches englitazone (30 microM) also inhibited K(ATP) channel currents activated by diazoxide by 90.8+/-4.1%. 4. In contrast, englitazone (1-30 microM) failed to inhibit K(ATP) channels activated by leptin, although higher concentrations (> 30 microM) did inhibit leptin actions. The englitazone concentration inhibition curve in the presence of leptin resulted in an IC50 value and Hill coefficient of 52 microM and 3.2, respectively. Similarly, in inside-out patches englitazone (30 microM) failed to inhibit the activity of K(ATP) channels in the presence of leptin. 5. Ciclazindol also inhibited K(ATP) currents activated by diazoxide (200 microM) in a concentration-dependent manner, with an IC50 and Hill coefficient of 127 nM and 0.33, respectively. Furthermore, application of ciclazindol (1 microM) to the intracellular surface of inside-out patches inhibited K(ATP) channel currents activated by diazoxide (200 microM) by 86.6+/-8.1%. 6. However, ciclazindol was much less effective at inhibiting KATP currents activated by leptin (10 nM). Ciclazindol (0.1-10 microM) had no effect on K(ATP) currents activated by leptin, whereas higher concentrations (> 10 microM) did cause inhibition with an IC50 value of 40 microM and an associated Hill coefficient of 2.7. Similarly, ciclazindol (1 microM) had no significant effect on K(ATP) channel activity following leptin addition in excised inside-out patches. 7. In conclusion, K(ATP) currents activated by diazoxide and leptin show different sensitivity to englitazone and ciclazindol. This may be due to differences in the mechanism of activation of K(ATP) channels by diazoxide and leptin.


Subject(s)
Benzopyrans/pharmacology , Diazoxide/pharmacology , Indoles/pharmacology , Insulin/metabolism , Potassium Channel Blockers , Proteins/pharmacology , Thiazoles/pharmacology , Thiazolidinediones , Adenosine Triphosphate/metabolism , Animals , Cell Line , Hypoglycemic Agents/pharmacology , Insulin Secretion , Leptin , Membrane Potentials/drug effects , Potassium Channels/metabolism , Rats
18.
Br J Pharmacol ; 79(1): 201-9, 1983 May.
Article in English | MEDLINE | ID: mdl-6135475

ABSTRACT

1 Replacement of chloride by isethionate in Ringer solution bathing frog skeletal muscle fibres induces, after a delay of about 30 min, marked mechanical activity which was blocked by tubocurarine. This effect is reversed by washing out the isethionate. 2 Miniature end plate potentials (m.e.p.ps) and giant potentials (potentials greater than or equal to 2 X modal value) were recorded intracellularly in normal Ringer and isethionate Ringer solution. 3 The frequency of m.e.p.ps was unaltered by isethionate. The proportion of giant potentials increased from 3% in normal Ringer to 24.5% in isethionate Ringer after 90 min. This effect is usually reversible if the exposure to isethionate does not exceed 2 h. 4 The giant potentials were large enough to initiate trains of action potentials and still occurred in the presence of tetrodotoxin or Ca2+-free Ringer. Isethionate produced no change in the tau D of miniature endplate currents. 5 Chloride replacement by propionate produced no change in the proportion of giant potentials. 6 It is suggested that the isethionate anion can induce giant potentials and the possible mechanism of action is discussed.


Subject(s)
Alkanesulfonates/pharmacology , Chlorides/physiology , Isethionic Acid/pharmacology , Neuromuscular Junction/drug effects , Neurotransmitter Agents/metabolism , Animals , Calcium/pharmacology , In Vitro Techniques , Motor Endplate/drug effects , Muscle Contraction/drug effects , Neuromuscular Junction/metabolism , Propionates/pharmacology , Rana pipiens , Rana temporaria , Time Factors
19.
Br J Pharmacol ; 126(1): 51-60, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10051120

ABSTRACT

1. The effects of various inhibitors of the mitochondrial electron transport chain on the activity of ATP-sensitive K+ channels were examined in the Cambridge rat insulinoma G1 (CRI-G1) cell line using a combination of whole cell and single channel recording techniques. 2. Whole cell current clamp recordings, with 5 mM ATP in the pipette, demonstrate that the mitochondrial uncoupler sodium azide (3 mM) rapidly hyperpolarizes CRI-G1 cells with a concomitant increase in K+ conductance. This is due to activation of K(ATP) channels as the sulphonylurea tolbutamide (100 microM) completely reversed the actions of azide. Other inhibitors of the mitochondrial electron transport chain, rotenone (10 microM) or oligomycin (2 microM) did not hyperpolarize CRI-G1 cells or increase K+ conductance. 3. In cell-attached recordings, bath application of 3 mM sodium azide (in the absence of glucose) resulted in a rapid increase in K(ATP) channel activity, an action readily reversible by tolbutamide (100 microM). Application of sodium azide (3 mM), in the presence of Mg-ATP, to the intracellular surface of excised inside-out patches also increased K(ATP) channel activity, in a reversible manner. 4. In contrast, rotenone (10 microM) or oligomycin (2 microM) did not increase K(ATP) channel activity in either cell-attached, in the absence of glucose, or inside-out membrane patch recordings. 5. Addition of sodium azide (3 mM) to the intracellular surface of inside-out membrane patches in the presence of Mg-free ATP or the non-hydrolysable analogue 5'-adenylylimidodiphosphate (AMP-PNP) inhibited, rather than increased, K(ATP) channel activity. 6. In conclusion, sodium azide, but not rotenone or oligomycin, directly activates K(ATP) channels in CRI-G1 insulin secreting cells. This action of azide is similar to that reported previously for diazoxide.


Subject(s)
Adenosine Triphosphate/pharmacology , Enzyme Inhibitors/pharmacology , Membrane Potentials/drug effects , Potassium Channels/drug effects , Sodium Azide/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Animals , Electric Conductivity , Insulinoma , Oligomycins/pharmacology , Patch-Clamp Techniques , Rats , Rotenone/pharmacology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/physiology , Uncoupling Agents/pharmacology
20.
Br J Pharmacol ; 101(3): 531-40, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2127550

ABSTRACT

1. The sulphonylureas, tolbutamide (0.1-10 mM) and glibenclamide (0.1-100 microM) shown not to inhibit ATP-K+ channel currents when applied to inside-out membrane patches excised from rat cultured cerebral cortex or freshly-dispersed ventromedial hypothalamic nucleus (VMHN) neurones. 2. Saturable binding sites for [3H]-glibenclamide, with similar affinity constants are present in rat cerebral cortex and hypothalamic membranes. The density of binding sites was lower in the hypothalamus than cortex. 3. Intracellular recordings from glucoreceptive VMHN neurones in hypothalamic slices were obtained. In the absence of glucose, tolbutamide (0.1 mM) depolarized these cells, increased membrane resistance and elicited action potentials. 4. Tolbutamide (0.1 mM) inhibited ATP-K+ channel currents and induced action current activity in cell-attached recordings from glucoreceptive VMHN neurones. 5. Glibenclamide (10-500 nM) had no effect per se on glucoreceptive VMHN neurones but did antagonize the actions of tolbutamide. 6. It is concluded that the hypothalamic (and perhaps cortical) sulphonylurea receptors are not directly coupled to ATP-K+ channels.


Subject(s)
Adenosine Triphosphate/metabolism , Potassium Channels/drug effects , Tolbutamide/pharmacology , Ventromedial Hypothalamic Nucleus/drug effects , Action Potentials/drug effects , Animals , Binding Sites , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Glyburide/metabolism , Glyburide/pharmacology , In Vitro Techniques , Male , Potassium Channels/metabolism , Rats , Rats, Inbred Strains , Ventromedial Hypothalamic Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL