Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters










Publication year range
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.
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
4.
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
5.
J Physiol ; 547(Pt 2): 417-25, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12562896

ABSTRACT

Striatal neurones are particularly vulnerable to hypoxia/ischaemia-induced damage, and free radicals are thought to be prime mediators of this neuronal destruction. It has been shown that hydrogen peroxide (H2O2), through the production of free radicals, induces rat insulinoma cell death by activation of a non-selective cation channel, which leads to irreversible cell depolarization and unregulated Ca2+ entry into the cell. In the study presented here, we demonstrate that a subpopulation of striatal neurones (medium spiny neurones) is depolarized by H2O2 through the production of free radicals. Cell-attached recordings from rat cultured striatal neurones demonstrate that exposure to H2O2 opens a large-conductance channel that is characterized by extremely long open times (seconds). Inside-out recordings show that cytoplasmically applied beta-nicotinamide adenine dinucleotide activates a channel with little voltage dependence, a linear current-voltage relationship and a single-channel conductance of between 70 and 90 pS. This channel is permeable to Na+, K+ and Ca2+ ions. Fura-2 imaging from cultured striatal neurones reveals that H2O2 exposure induces a biphasic intracellular Ca2+ increase in a subpopulation of neurones, the second, later phase resulting in Ca2+ overload. This later component of the Ca2+ response is dependent on the presence of extracellular Ca2+ and is independent of synaptic activity or voltage-gated Ca2+ channel opening. Consequently, this channel may be an important contributor to free radical-induced selective striatal neurone destruction. These results are remarkably similar to those observed for insulinoma cells and suggest that this family of non-selective cation channels has a widespread distribution in mammalian tissues.


Subject(s)
Cations/metabolism , Corpus Striatum/drug effects , Hydrogen Peroxide/poisoning , Ion Channels/metabolism , Neurons/drug effects , Oxidants/poisoning , Animals , Calcium/metabolism , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/physiology , Electrophysiology , Intracellular Membranes/metabolism , Neurons/physiology , Rats , Rats, Sprague-Dawley
6.
Biol Reprod ; 68(6): 2135-41, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12606455

ABSTRACT

Large-conductance, calcium-dependent potassium (BKCa) channels are implicated in maintaining uterine quiescence during pregnancy. The mechanisms whereby calcium sensitivity of the BKCa channel is dramatically removed at parturition remain unknown. The aim of the present study was to investigate whether this loss of calcium sensitivity of the BKCa channel with the onset of labor is associated with changes in the protein expression of the alpha- and/or beta-subunit or arises from a physical dissociation of the alpha-subunit from the beta-subunit. The beta-subunit is a key determinant of BKCa-channel Ca2+ sensitivity. Western blot analysis, using alpha- and beta-subunit-specific antibodies, detected bands of 110-125 and 36 kDa, respectively. Protein expression levels of the alpha-subunit in term labor myometrium were significantly reduced compared with term pregnancy without labor. Furthermore, alpha-subunit levels at term pregnancy were significantly increased relative to the nonpregnant state, whereas levels at preterm gestations were unchanged. Densitometric analysis demonstrated significantly decreased beta-subunit levels in term and preterm labor samples compared with term nonlabor samples. Immunoprecipitation studies revealed the presence of both the alpha- and beta-subunits in samples taken before or after the onset of labor. We conclude that during labor, the alpha-subunit is not physically uncoupled from the beta-subunit, but a decline occurs in the level of beta-subunit protein, which may underlie the loss of calcium and voltage sensitivity of the BKCa channel with labor. Furthermore, reduced beta-subunit protein in preterm labor myometrium implies that ion channels may also contribute to pathophysiological labor.


Subject(s)
Down-Regulation/physiology , Myometrium/metabolism , Parturition/physiology , Potassium Channels, Calcium-Activated/physiology , Adult , Blotting, Western , Calcium Signaling/physiology , Cesarean Section , Female , Humans , Indicators and Reagents , Membranes/metabolism , Obstetric Labor, Premature/physiopathology , Precipitin Tests , Pregnancy , Subcellular Fractions/metabolism
7.
J Physiol ; 545(3): 933-44, 2002 12 15.
Article in English | MEDLINE | ID: mdl-12482897

ABSTRACT

The obese gene product, leptin is an important circulating satiety factor that regulates energy balance via its actions in the hypothalamus. However, leptin receptors are also expressed in brain regions not directly associated with energy homeostasis, such as the hippocampus. Here, leptin inhibits hippocampal neurones via activation of large conductance Ca(2+)-activated K(+) (BK) channels, a process that may be important in regulating neuronal excitability. We now show that leptin receptor labelling is expressed on somata, dendrites and axons, and is also concentrated at synapses in hippocampal cultures. In functional studies, leptin potently and reversibly reduces epileptiform-like activity evoked in lean, but not leptin-resistant Zucker fa/fa rats. Furthermore, leptin also depresses enhanced Ca(2+) levels evoked following Mg(2+) removal in hippocampal cultures. The ability of leptin to modulate this activity requires activation of BK, but not K(ATP), channels as the effects of leptin were mimicked by the BK channel activator NS-1619, and inhibited by the BK channel inhibitors, iberiotoxin and charybdotoxin. The signalling mechanisms underlying this process involve stimulation of phosphoinositide 3-kinase (PI 3-kinase), but not mitogen-activated protein kinase (MAPK), as two structurally unrelated inhibitors of PI 3-kinase, LY294002 and wortmannin, blocked the actions of leptin. These data indicate that leptin, via PI 3-kinase-driven activation of BK channels, elicits a novel mechanism for controlling neuronal excitability. As uncontrolled excitability in the hippocampus is one underlying cause of temporal lobe epilepsy, this novel action of leptin could provide an alternative therapeutic target in the management of epilepsy.


Subject(s)
Epilepsy/physiopathology , Hippocampus/physiopathology , Leptin/physiology , Neurons/physiology , Phosphatidylinositol 3-Kinases/physiology , Potassium Channels, Calcium-Activated/physiology , Adenosine Triphosphate/metabolism , Animals , Calcium/analysis , Cells, Cultured , Culture Media/chemistry , Hippocampus/drug effects , Humans , Large-Conductance Calcium-Activated Potassium Channels , Leptin/pharmacology , Magnesium/analysis , Mitogen-Activated Protein Kinases/physiology , Neurons/drug effects , Potassium Channels/metabolism , Rats , Receptors, Cell Surface/metabolism , Receptors, Leptin , Recombinant Proteins
8.
J Physiol ; 540(Pt 3): 771-80, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11986367

ABSTRACT

Large conductance, Ca(2+)-activated K(+) (maxi-K ) channel activity was recorded in excised, inside-out patches from HEK 293 cells stably co-expressing the alpha- and beta-subunits of human brain maxi-K channels. At +50 mV, and in the presence of 300 nM Ca2+i, single channel activity was acutely and reversibly suppressed upon reducing P(O(2)) from 150 to > 40 mmHg by over 30 %. The hypoxia-evoked reduction in current was due predominantly to suppression in NP(o), although a minor component was attributable to reduced unitary conductance of 8-12 %. Hypoxia caused an approximate doubling of the time constant for activation but was without effect on deactivation. At lower levels of Ca2+i(30 and 100 nM), hypoxic inhibition did not reach significance. In contrast, 300 nM and 1 microM Ca2+i both sustained significant hypoxic suppression of activity over the entire activating voltage range. At these two Ca2+i levels, hypoxia evoked a positive shift in the activating voltage (by approximately 10 mV at 300 nM and approximately 25 mV at 1 microM). At saturating [Ca(2+)](i) (100 microM), hypoxic inhibition was absent. Distinguishing between hypoxia-evoked changes in voltage- and/or Ca2+i-sensitivity was achieved by evoking maximal channel activity using high depolarising potentials (up to +200 mV) in the presence of 300 nM or 100 microM Ca2+i or in its virtual absence (> 1 nM). Under these experimental conditions, hypoxia caused significant channel inhibition only in the presence of 300 nM Ca2+i. Thus, since regulation was observed in excised patches, maxi-K channel inhibition by hypoxia does not require soluble intracellular components and, mechanistically, is voltage independent and Ca2+i sensitive.


Subject(s)
Calcium/physiology , Cell Hypoxia/physiology , Cell Membrane/physiology , Potassium Channels, Calcium-Activated/physiology , Calcium/pharmacology , Cell Line , Humans , Kidney , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Large-Conductance Calcium-Activated Potassium Channels , Membrane Potentials/physiology , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/drug effects , Potassium Channels, Calcium-Activated/genetics , Protein Subunits , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Transfection
9.
Exp Physiol ; 86(2): 255-64, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11429642

ABSTRACT

The contractility of the human uterus is under the fine control of a variety of interacting bioactive agents. During labour, the excitability of the uterus is drastically transformed in comparison with the non-labour state and is manifest at the membrane level via the activity of uterine ion channels. This article reviews the contribution of potassium (K(+)) channels to human uterine excitability. Experimental Physiology (2001) 86.2, 255-264.


Subject(s)
Myometrium/physiology , Potassium Channels/physiology , Adenosine Triphosphate/physiology , Calcium/physiology , Electrophysiology , Female , Humans , Ion Channel Gating , Labor, Obstetric/metabolism , Pregnancy , Uterine Contraction/physiology
10.
J Physiol ; 527 Pt 1: 95-107, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10944173

ABSTRACT

1. The role of the cytoskeleton in leptin-induced activation of ATP-sensitive K+ (KATP) channels was examined in rat CRI-G1 insulin-secreting cells using patch clamp and fluorescence imaging techniques. 2. In whole cell recordings, dialysis with the actin filament stabiliser phalloidin (10 microM) prevented KATP channel activation by leptin. 3. Application of the actin filament destabilising agents deoxyribonuclease type 1 (DNase 1; 50 microg ml-1) or cytochalasin B (10 microM) to intact cells or inside-out membrane patches also increased KATP channel activity in a phalloidin-dependent manner. 4. The anti-microtubule agents nocodazole (10 microM) and colchicine (100 microM) had no effect on KATP channel activity. 5. Fluorescence staining of the cells with rhodamine-conjugated phalloidin revealed rapid disassembly of actin filaments by cytochalasin B and leptin, the latter action being prevented by the phosphoinositide 3 (PI 3)-kinase inhibitor LY 294002. 6. Activation of KATP channels by the PI 3-kinase product phosphatidylinositol 3,4,5-trisphosphate (PtdIns(3,4,5)P3) was also prevented by phalloidin. This is consistent with the notion that leptin activates KATP channels in these cells by an increase in PtdIns(3,4,5)P3 or a similar 3-phosphorylated phosphoinositol lipid, resulting in actin filament disruption.


Subject(s)
Actins/physiology , Cytoskeleton/physiology , Insulinoma/metabolism , Leptin/pharmacology , Potassium Channels/metabolism , Actins/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cytochalasin B/pharmacology , Cytoskeleton/drug effects , Deoxyribonuclease I/pharmacology , Microscopy, Fluorescence , Patch-Clamp Techniques , Phalloidine/pharmacology , Phosphatidylinositol Phosphates/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Potassium/metabolism , Potassium Channels/drug effects , Rats , Signal Transduction/drug effects
11.
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
12.
Br J Pharmacol ; 130(4): 857-66, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10864893

ABSTRACT

Two electrode voltage clamp and single channel recordings were used to investigate the actions of various ATP-sensitive K(+) (K(ATP)) channel inhibitors on cloned K(ATP) channels, expressed in Xenopus oocytes and HEK 293 cells. Oocytes expressing Kir6.2 and SUR1 gave rise to inwardly rectifying K(+) currents following bath application of 3 mM sodium azide. Inside-out recordings from non-azide treated oocytes demonstrated the presence of K(ATP) channels which were activated by direct application of 3 mM azide and 0.1 mM Mg-ATP. Tolbutamide inhibited azide-induced macroscopic Kir6.2-SUR1 currents, recorded from Xenopus oocytes, with an IC(50) value similar to native K(ATP) channels. Ciclazindol and englitazone also inhibited these currents in a concentration-dependent manner, but with relative potencies substantially less than for native K(ATP) channels. Single channel currents recorded from inside-out patches excised from oocytes expressing Kir6.2-SUR1 currents were inhibited by tolbutamide, Mg-ATP, englitazone and ciclazindol, in the absence of azide, with potencies similar to native K(ATP) channels. In the presence of azide, Kir6.2-SUR1 currents were inhibited by englitazone and tolbutamide but not ciclazindol. Single channel currents derived from Kir6.2Delta26, expressed in HEK 293 cells, were inhibited by ciclazindol and englitazone irrespective of the absence or presence of SUR1. In conclusion, heterologously expressed Kir6.2 and SUR1 recapitulate the pharmacological profile of native pancreatic beta-cell K(ATP) channels. However, currents induced by azide exhibit a substantially reduced sensitivity to ciclazindol. It is likely that ciclazindol and englitazone inhibit K(ATP) currents by interaction with the Kir6.2 subunit.


Subject(s)
ATP-Binding Cassette Transporters , Benzopyrans/pharmacology , Indoles/pharmacology , Membrane Potentials/drug effects , Potassium Channels, Inwardly Rectifying , Potassium Channels/drug effects , Receptors, Drug/drug effects , Sodium Azide/pharmacology , Thiazoles/pharmacology , Thiazolidinediones , Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/physiology , Animals , Cell Line , Dose-Response Relationship, Drug , Female , Glyburide/pharmacology , Humans , Mice , Microinjections , Oocytes , Potassium Channels/genetics , Potassium Channels/physiology , RNA, Complementary/administration & dosage , Receptors, Drug/genetics , Receptors, Drug/physiology , Sulfonylurea Receptors , Tolbutamide/pharmacology , Xenopus laevis
13.
J Biol Chem ; 275(7): 4660-9, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10671495

ABSTRACT

The mechanism by which leptin increases ATP-sensitive K(+) (K(ATP)) channel activity was investigated using the insulin-secreting cell line, CRI-G1. Wortmannin and LY 294002, inhibitors of phosphoinositide 3-kinase (PI3-kinase), prevented activation of K(ATP) channels by leptin. The inositol phospholipids phosphatidylinositol bisphosphate and phosphatidylinositol trisphosphate (PtdIns(3,4,5)P(3)) mimicked the effect of leptin by increasing K(ATP) channel activity in whole-cell and inside-out current recordings. LY 294002 prevented phosphatidylinositol bisphosphate, but not PtdIns(3,4,5)P(3), from increasing K(ATP) channel activity, consistent with the latter lipid acting as a membrane-associated messenger linking leptin receptor activation and K(ATP) channels. Signaling cascades, activated downstream from PI 3-kinase, utilizing PtdIns(3,4,5)P(3) as a second messenger and commonly associated with insulin and cytokine action (MAPK, p70 ribosomal protein-S6 kinase, stress-activated protein kinase 2, p38 MAPK, and protein kinase B), do not appear to be involved in leptin-mediated activation of K(ATP) channels in this cell line. Although PtdIns(3,4,5)P(3) appears a plausible and attractive candidate for the messenger that couples K(ATP) channels to leptin receptor activation, direct measurement of PtdIns(3,4,5)P(3) demonstrated that insulin, but not leptin, increased global cellular levels of PtdIns(3,4,5)P(3). Possible mechanisms to explain the involvement of PI 3-kinases in K(ATP) channel regulation are discussed.


Subject(s)
Adenosine Triphosphate/metabolism , Leptin/physiology , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels/physiology , Animals , Insulinoma/physiopathology , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositols/physiology , Rats , Signal Transduction , Tumor Cells, Cultured
14.
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
15.
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
16.
Biochem Pharmacol ; 57(5): 491-501, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-9952312

ABSTRACT

The membrane spanning complement channel is assumed to be a nonselective ion 'pore', although little evidence is available to support this hypothesis. In this paper we provide evidence that Ca2+ entry and Cl- exit occur rapidly after complement activation and precede the development of a long-lasting complement-dependent inward current. Addition of rabbit serum (a source of heterologous complement) and mouse anti-human insulin receptor antibody to a single Xenopus oocyte expressing human insulin receptor was shown to stimulate an initial hyperpolarising current followed by a sustained depolarising current. On voltage clamping the oocyte, a novel long-lasting inward current generated by serum addition was detected. Complement classical pathway-stimulated calcium influx into the oocyte was directly demonstrated using 45Ca influx measurements. In addition, we found that Ca2+ influx was required for the stimulation of the complement alternative pathway-dependent inward current. The novel conductance elicited by the classical pathway was outwardly rectifying, had a reversal potential of -35 +/- 8 mV (or -52 +/- 7 mV in the presence of chloride channel inhibitors), was inhibited by nifedipine, and was observed in the presence but not in the absence of the pore-forming complement component C9. As overactivation of complement does play a role in many inflammatory or autoimmune diseases, inhibition of early complement-mediated ion flux might restrict tissue damage and aid recovery from such diseases.


Subject(s)
Calcium/metabolism , Chloride Channels/physiology , Complement System Proteins/physiology , Oocytes/physiology , Receptor, Insulin/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Blood , Complement System Proteins/pharmacology , Culture Media , Electrophysiology , Female , Humans , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Niflumic Acid/pharmacology , RNA, Messenger/genetics , Rabbits , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/genetics , Tetraethylammonium/pharmacology , Xenopus laevis
17.
J Biol Chem ; 274(2): 833-41, 1999 Jan 08.
Article in English | MEDLINE | ID: mdl-9873022

ABSTRACT

Fura-2 fluorescence was used to investigate the effects of H2O2 on [Ca2+]i in the insulin-secreting cell line CRI-G1. H2O2 (1-10 mM) caused a biphasic increase in free [Ca2+]i, an initial rise observed within 3 min and a second, much larger rise following a 30-min exposure. Extracellular calcium removal blocked the late, but not the initial, rise in [Ca2+]i. Thapsigargin did not affect either response to H2O2, but activated capacitive calcium entry, an action abolished by 10 microM La3+. Simultaneous recordings of membrane potential and [Ca2+]i demonstrated the same biphasic [Ca2+]i response to H2O2 and showed that the late increase in [Ca2+]i coincided temporally with cell membrane potential collapse. Buffering Ca2+i to low nanomolar levels prevented both phases of increased [Ca2+]i and the H2O2-induced depolarization. The H2O2-induced late rise in [Ca2+]i was prevented by extracellular application of 100 microM La3+. La3+ (100 microM) inhibited the H2O2-induced cation current and NAD-activated cation (NSNAD) channel activity in these cells. H2O2 increased the NAD/NADH ratio in intact CRI-G1 cells, consistent with increased cellular [NAD]. These data suggest that H2O2 increases [NAD], which, coupled with increased [Ca2+]i, activates NSNAD channels, causing unregulated Ca2+ entry and consequent cell death.


Subject(s)
Calcium/metabolism , Hydrogen Peroxide/pharmacology , Insulin/metabolism , Ion Channels/metabolism , Cations , Cell Line , Insulin Secretion , Ion Channel Gating , Ion Transport , Membrane Potentials , Patch-Clamp Techniques , Thapsigargin/pharmacology
18.
J Physiol ; 514 ( Pt 1): 47-57, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9831715

ABSTRACT

1. Whole-cell voltage-clamp recordings were used to study the characteristics of a non-selective cation current, activated by intracellular beta-NAD+, present in CRI-G1 insulin-secreting cells. The monovalent cations Na+, K+ and Cs+ were equally permeant through this channel. 2. The magnitude of the beta-NAD+ current was dependent on the concentration of both beta-NAD+ and Ca2+ in the cell. The properties of the beta-NAD+-activated macroscopic current are similar to those of the beta-NAD+-activated non-selective cation channel (NSNAD) examined at the single channel level in this cell line. 3. The presence of intracellular reduced glutathione (GSH) inhibited the beta-NAD+-activated macroscopic current and the activity of the NSNAD channel in inside-out patch recordings. 4. The inhibition of beta-NAD+-activated currents by GSH is mimicked by its analogue ophthalmic acid but not by another thiol reducing agent dithiothreitol, indicating the presence of a specific GSH binding site present on the NSNAD channel or associated protein.


Subject(s)
Glutathione/pharmacology , Insulin/metabolism , Ion Channel Gating/drug effects , NAD/metabolism , Animals , Cations/metabolism , Cell Line , Cesium/pharmacokinetics , Hypoglycemic Agents/pharmacology , Insulin Secretion , NAD/antagonists & inhibitors , NAD/pharmacology , Oligopeptides/pharmacology , Patch-Clamp Techniques , Rats , Tolbutamide/pharmacology
19.
J Physiol ; 513 ( Pt 3): 733-47, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9824714

ABSTRACT

1. Inside-out recordings from rat cortical fused nerve terminals indicate that the most common channel observed was a large conductance K+ (BK) channel with characteristics dissimilar to conventional cell body calcium-activated BK (BKCa) channels. 2. BK channels exhibit mode switching between low (mode 1) and high (mode 2) activity, an effect not influenced by membrane voltage. Increasing internal Ca2+ concentration increased time spent in mode 2 as did application of protein kinase A, an effect not mimicked by protein kinase C or protein kinase G. 3. Mode 1 activity was voltage independent although depolarization increased mode 2 channel activity. Global average channel activity was voltage and Ca2+ dependent. 4. Alkaline phosphatase treatment induced channel activity to reside permanently in mode 2, where activity was voltage and Ca2+ dependent but unaffected by protein kinases A, G or C. 5. Internal application of tetraethylammonium blocked BK channel activity in a manner identical to that reported for BKCa channels. 6. These results indicate that nerve terminal membranes have large conductance K+ channels with significant differences in gating kinetics and regulation of activity compared with BKCa channels of other neuronal preparations. The BK channel subtype may play a unique physiological role specific to the nerve terminal.


Subject(s)
Cerebral Cortex/metabolism , Potassium Channels/metabolism , Presynaptic Terminals/metabolism , Alkaline Phosphatase/pharmacology , Animals , Calcium/physiology , Cerebral Cortex/cytology , Cyclic AMP-Dependent Protein Kinases/metabolism , Electric Stimulation , Electrophysiology , Ion Channel Gating/drug effects , Male , Membrane Potentials/physiology , Neurons/metabolism , Patch-Clamp Techniques , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Tetraethylammonium Compounds/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...