Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 329(3): 978-86, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19321803

ABSTRACT

Neurons are highly differentiated and polarized cells, whose various functions depend upon the compartmentalization of ion channels. The rat hypothalamic-neurohypophysial system (HNS), in which cell bodies and dendrites reside in the hypothalamus, physically separated from their nerve terminals in the neurohypophysis, provides a particularly powerful preparation in which to study the distribution and regional properties of ion channel proteins. Using electrophysiological and immunohistochemical techniques, we characterized the large-conductance calcium-activated potassium (BK) channel in each of the three primary compartments (soma, dendrite, and terminal) of HNS neurons. We found that dendritic BK channels, in common with somatic channels but in contrast to nerve terminal channels, are insensitive to iberiotoxin. Furthermore, analysis of dendritic BK channel gating kinetics indicates that they, like somatic channels, have fast activation kinetics, in contrast to the slow gating of terminal channels. Dendritic and somatic channels are also more sensitive to calcium and have a greater conductance than terminal channels. Finally, although terminal BK channels are highly potentiated by ethanol, somatic and dendritic channels are insensitive to the drug. The biophysical and pharmacological properties of somatic and dendritic versus nerve terminal channels are consistent with the characteristics of exogenously expressed alphabeta1 versus alphabeta4 channels, respectively. Therefore, one possible explanation for our findings is a selective distribution of auxiliary beta1 subunits to the somatic and dendritic compartments and beta4 to the terminal compartment. This hypothesis is supported immunohistochemically by the appearance of distinct punctate beta1 or beta4 channel clusters in the membrane of somatic and dendritic or nerve terminal compartments, respectively.


Subject(s)
Central Nervous System/metabolism , Ethanol/pharmacology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/physiology , Neurons/metabolism , Animals , Calcium/metabolism , Calcium/pharmacology , Dendrites/metabolism , Hypothalamo-Hypophyseal System/cytology , Hypothalamo-Hypophyseal System/metabolism , In Vitro Techniques , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Kinetics , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/drug effects , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Large-Conductance Calcium-Activated Potassium Channels/physiology , Membrane Potentials/physiology , Neurons/drug effects , Peptides/pharmacology , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Supraoptic Nucleus/cytology , Supraoptic Nucleus/metabolism , Toxins, Biological/pharmacology
2.
J Neuroendocrinol ; 17(9): 583-90, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16101897

ABSTRACT

Oxytocin release from neurophypophysial terminals is particularly sensitive to inhibition by the micro-opioid receptor agonist, DAMGO. Because the R-type component of the neurophypophysial terminal Ca2+ current (ICa) mediates exclusively oxytocin release, we hypothesised that micro-opioids could preferentially inhibit oxytocin release by blocking this channel subtype. Whole-terminal recordings showed that DAMGO and the R-type selective blocker SNX-482 inhibit a similar ICa component. Measurements of [Ca2+]i levels and oxytocin release confirmed that the effects of DAMGO and SNX-482 are not additive. Finally, isolation of the R-type component and its associated rise in [Ca2+]i and oxytocin release allowed us to demonstrate the selective inhibition by DAMGO of this channel subtype. Thus, micro-opioid agonists modulate specifically oxytocin release in neurophypophysial terminals by selectively targeting R-type Ca2+ channels. Modulation of Ca2+ channel subtypes could be a general mechanism for drugs of abuse to regulate the release of specific neurotransmitters at central nervous system synapses.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, R-Type/drug effects , Oxytocin/metabolism , Pituitary Gland, Posterior/metabolism , Receptors, Opioid, mu/agonists , Analgesics, Opioid/pharmacology , Animals , Arginine Vasopressin/metabolism , Calcium/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , In Vitro Techniques , Male , Membrane Potentials/drug effects , Nerve Endings/drug effects , Neuropeptides/metabolism , Neuropeptides/pharmacology , Rats , Rats, Sprague-Dawley , Spider Venoms/pharmacology
3.
Toxicol Appl Pharmacol ; 168(3): 183-8, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11042090

ABSTRACT

We report that intracellular calcium levels rise in mammalian neurosecretory terminals and in cultured pheochromocytoma cells during acute exposure to physiological medium incubated in IV drip chambers. The agent responsible for this effect is shown to be di(2-ethylhexyl)phthalate (DEHP). DEHP (800 nM) added to saline solution caused a rise in [Ca(2+)](i) similar to that elicited by the contaminant-containing solution. The extraction of this contaminant from the IV drip chamber, as measured by spectrophotometry, was time-dependent and was markedly accelerated by the presence of 50 mM ethanol in the solution. Larger [Ca(2+)](i) increases were observed in terminals exposed to solutions incubated in IV drip chambers for greater durations. The rise in calcium requires transmembrane calcium flux through membrane channels, as the response is blocked by either 100 microM cadmium or by lowering the extracellular free Ca(2+) concentration to 10 microM. Our results suggest that acute alterations in intracellular calcium should be considered in addition to long-term effects when determining the safety of phthalate-containing plastics and that laboratory researchers using plastic perfusion materials consider this potential source of artifactual results.


Subject(s)
Calcium/metabolism , Diethylhexyl Phthalate/pharmacology , Drug Packaging , Neurosecretory Systems/metabolism , Plasticizers/pharmacology , Presynaptic Terminals/metabolism , Animals , Chromatography, Thin Layer , Humans , Infusions, Intravenous , Male , Neurosecretory Systems/drug effects , PC12 Cells , Pituitary Gland, Posterior/drug effects , Pituitary Gland, Posterior/metabolism , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Spectrophotometry, Ultraviolet , Tumor Cells, Cultured
4.
Alcohol Clin Exp Res ; 24(7): 1077-83, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10924013

ABSTRACT

BACKGROUND: Acute ethanol (EtOH) exposure reduces the evoked release of vasopressin (AVP) and oxytocin (OT) from excised neurohypophyses and from dissociated neurohypophysial terminals of the rat. METHODS AND RESULTS: Rats placed on a diet that maintained blood levels of 30 mM EtOH for 20 to 40 days developed tolerance to acute EtOH inhibition of release. In the presence of 10 mM EtOH, high (50 mM) K+-induced release of AVP from isolated neurohypophysial terminals of EtOH-naive rats was reduced by 77.7+/-1.4%, whereas in the chronic EtOH group, release was reduced by only 9.4+/-8.7%. Similar tolerance was evident during acute challenge with 75 mM EtOH, as well as for release of OT from isolated terminals. Animals treated with an intraperitoneal injection of EtOH and sacrificed 90 min postinjection did not exhibit the reduced EtOH inhibition of release from dissociated terminals during a 75 mM EtOH acute challenge. CONCLUSIONS: The altered component responsible for the tolerance to inhibition of release resides in the isolated terminal, because tolerance measured in vitro from intact neurohypophyses was similar to that seen in isolated terminals. The failure of EtOH-injected animals to exhibit reduced inhibition of release in response to an acute EtOH challenge indicates that short-term elevated blood alcohol level does not induce this tolerance. The finding of tolerance to EtOH-induced inhibition of release from the intact neurohypophysis and isolated terminals provides a physiological preparation in which to examine the molecular targets of acute drug action modified after chronic exposure to the drug.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Oxytocin/metabolism , Pituitary Gland, Posterior/drug effects , Vasopressins/metabolism , Animals , Central Nervous System Depressants/blood , Ethanol/blood , Male , Pituitary Gland, Posterior/metabolism , Rats , Rats, Sprague-Dawley
5.
J Physiol ; 519 Pt 1: 101-14, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10432342

ABSTRACT

1. Large conductance, Ca2+-activated K+ (BK) channels were identified in freshly dissociated rat supraoptic neurones using patch clamp techniques. 2. The single channel conductance of cell body BK channels, recorded from inside-out patches in symmetric 145 mM K+, was 246.1 pS, compared with 213 pS in nerve ending BK channels (P<0.01). 3. At low open probability (Po), the reciprocal of the slope in the ln(NPo)-voltage relationship (N, number of available channels in the patch) for cell body and nerve ending channels were similar: 11 vs. 14 mV per e-fold change in NPo, respectively. 4. At 40 mV, the [Ca2+]i producing half-maximal activation was 273 nM, as opposed to >1.53 microM for the neurohypophysial channel, indicating the higher Ca2+ sensitivity of the cell body isochannel. 5. Cell body BK channels showed fast kinetics (open time constant, 8.5 ms; fast closed time constant, 1.6 and slow closed time constant, 12.7 ms), identifying them as 'type I' isochannels, as opposed to the slow gating (type II) of neurohypophysial BK channels. 6. Cell body BK activity was reduced by 10 nM charybdotoxin (NPo, 37% of control), or 10 nM iberiotoxin (NPo, 5% of control), whereas neurohypophysial BK channels are insensitive to charybdotoxin at concentrations as high as 360 nM. 7. Whilst blockade of nerve ending BK channels markedly slowed the repolarization of evoked single spikes, blockade of cell body channels was without effect on repolarization of evoked single spikes. 8. Ethanol reversibly increased neurohypophysial BK channel activity (EC50, 22 mM; maximal effect, 100 mM). In contrast, ethanol (up to 100 mM) failed to increase cell body BK channel activity. 9. In conclusion, we have characterized BK channels in supraoptic neuronal cell bodies, and demonstrated that they display different electrophysiological and pharmacological properties from their counterparts in the nerve endings.


Subject(s)
Benzylisoquinolines , Evoked Potentials/physiology , Nerve Endings/physiology , Neurons/physiology , Potassium Channels, Calcium-Activated , Potassium Channels/physiology , Supraoptic Nucleus/physiology , Alkaloids/pharmacology , Animals , Calcium/physiology , Calcium Channel Blockers/pharmacology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channels , Male , Nerve Endings/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Rats , Reaction Time
7.
Toxicol Sci ; 45(1): 9-25, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9848106

ABSTRACT

Whereas the roles of G proteins and protein kinases in various neuroreceptors and ion channels have been studied extensively, their roles in the actions of drugs and toxicants on these receptors and channels remain to be elucidated. Almost all drugs and toxicants exert multiple actions on multiple target sites, and there is no reason to assume that a chemical modulates a receptor/channel via a single mechanism. In fact, experimental evidence is slowly but steadily being accumulated to indicate that certain drugs and toxicants modulate neuroreceptor/channel functions through interactions with intracellular components such as G proteins and protein kinases. Multiple actions of a toxicant on various receptors/channels may be explained on the basis of its interaction with the G protein/kinase system that is a common denominator of the target sites. This is a virgin field that promises a quantum leap in the coming years. Each presentation and discussion will focus on expected future developments and potential significance in the field of neurotoxicology.


Subject(s)
Brain/drug effects , Ion Channels/drug effects , Metals, Heavy/pharmacology , Protein Kinases/physiology , Receptors, Neurotransmitter/drug effects , Animals , Brain/metabolism , Ethanol/pharmacology , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/physiology , Ion Channels/metabolism , Lead/pharmacology , Mercury/pharmacology , Receptors, Neurotransmitter/metabolism
8.
Mol Pharmacol ; 54(2): 397-406, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9687582

ABSTRACT

We examined the actions of ethanol on the single channel properties of large conductance Ca2+-activated K+ (BK) channels isolated from skeletal muscle T-tubule membranes and incorporated into planar lipid bilayer membranes. We have taken advantage of this preparation, because it lacks most elements of cellular complexity, including cytoplasmic constituents and complex membrane lipid composition and architecture, to examine the minimum requirements for the effects of alcohol. Clinically relevant concentrations (25-200 mM) of ethanol increased the activity of BK channels incorporated into bilayers composed of phosphatidylethanolamine (PE) alone or PE and phosphatidylserine. The potentiation of channel activity by ethanol was attributable predominantly to a decrease in the average amount of time spent in closed states. Ethanol did not significantly affect the current amplitude-voltage relationship for BK channels, indicating that channel conductance for K+ was unaffected by the drug. Although base-line characteristics of BK channels incorporated into bilayers composed only of PE differed from those of channels in PE/ phosphatidylserine in a manner expected from the change in bilayer charges, the actions of ethanol on channel activity were qualitatively similar in the different lipid environments. The effects of ethanol on single channel properties of BK channels in the planar bilayer are very similar to those reported for the action of ethanol on neurohypophysial BK channels studied in native membrane, and for cloned BK channels expressed in Xenopus laevis oocytes, which suggests that ethanol's site and mechanism of action are preserved in this greatly simplified preparation.


Subject(s)
Calcium/metabolism , Ethanol/pharmacology , Potassium Channels/drug effects , Animals , Electric Conductivity , Electrophysiology , Ion Channel Gating/drug effects , Lipid Bilayers , Muscles/drug effects , Muscles/physiology , Potassium Channels/metabolism , Rats
9.
J Neuroendocrinol ; 10(6): 399-406, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9688342

ABSTRACT

The effects of ethanol were studied on single evoked spikes recorded at 20 degrees C with the perforated-patch method in acutely dissociated rat supraoptic neurons. In seven out of eight neurons, ethanol (50 mM) significantly reduced the spike duration by selectively decreasing the decay time (82+/-2% of the control), leaving the amplitude and rise time unaffected. Resting potential and threshold did not change. Similarly, CdCl2 at a concentration of 100 microM, which blocks all voltage-activated calcium current in the supraoptic neurons, reduced the decay time of single evoked spikes (76+/-3% of the control, n=10) without modifying the other above-mentioned parameters. In addition, exposure to 100 microM CdCl2 prevented any subsequent effect of 50 mM ethanol (n = 5). Exposure to apamin (10 nM) and iberiotoxin (10 nM) did not have any effect on single evoked spikes. Because these concentrations are effective in blocking, respectively, small (SK) and large (BK) conductance calcium-dependent potassium channels in these neurons, this result shows that these currents are not involved in either the shaping of single evoked spikes or the actions of ethanol on spike shape. The sustained component of whole-cell recorded calcium current measured at -10 mV (hp -60 mV) was inhibited by ethanol in a dose-dependent manner, with a significant effect detectable at 25 mM. Exposure to 50 mM ethanol significantly reduced the sustained current to 70+/-5% of the control (n=12), without any apparent shift of the current-voltage relationship. Control exposure of the neurons to either 50 mM urea or 50 mM sucrose did not affect the voltage-gated calcium currents. We conclude that ethanol reduces the duration of single evoked spikes by a specific inhibition of voltage-activated calcium currents. The results suggest that, in addition to its direct effects on release of vasopressin and oxytocin from neurohypophysial terminals, ethanol could also affect hormonal release via changes in firing patterns arising in the cell bodies.


Subject(s)
Calcium Channels/physiology , Ethanol/pharmacology , Evoked Potentials/drug effects , Neurons/drug effects , Supraoptic Nucleus/drug effects , Animals , Ion Channel Gating , Male , Neurons/physiology , Rats , Supraoptic Nucleus/cytology , Supraoptic Nucleus/physiology
10.
J Neurosci ; 18(13): 4883-90, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9634554

ABSTRACT

The Ca2+ channel beta subunit has been shown to reduce the magnitude of G-protein inhibition of Ca2+ channels. However, neither the specificity of this action to different forms of G-protein inhibition nor the mechanism underlying this reduction in response is known. We have reported previously that coexpression of the Ca2+ channel beta3 subunit causes M2 muscarinic receptor-mediated inhibition of alpha1B Ca2+ currents to become more voltage-dependent. We report here that the beta3 subunit increases the rate of relief of inhibition produced by a depolarizing prepulse and also shifts the voltage dependency of this relief to more hyperpolarized voltages; these effects are likely to be responsible for the reduction of inhibitory response of alpha1B channels to G-protein-mediated inhibition seen after coexpression of the Ca2+ channel beta3 subunit. Additionally, the beta3 subunit alters the rate and voltage dependency of relief of the inhibition produced by coexpressed Gbeta1gamma1, in a manner similar to the changes it produces in relief of M2 receptor-induced inhibition. We conclude that the Ca2+ channel beta3 subunit reduces the magnitude of G-protein inhibition of alpha1B Ca2+ channels by enhancing the rate of dissociation of the G-protein betagamma subunit from the Ca2+ channel alpha1B subunit.


Subject(s)
Calcium Channels/metabolism , GTP-Binding Proteins/physiology , Receptors, Muscarinic/metabolism , Acetylcholine/pharmacology , Animals , Calcium Channels/genetics , Enzyme Inhibitors/pharmacology , Ethylmaleimide/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/chemistry , Gene Expression/physiology , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Oocytes/physiology , Receptor, Muscarinic M2 , Receptors, Muscarinic/genetics , Transfection , Xenopus
11.
J Neurosci ; 18(3): 878-86, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9437009

ABSTRACT

We have shown previously that the Ca2+ channel beta3 subunit is capable of modulating tonic G-protein inhibition of alpha1A and alpha1B Ca2+ channels expressed in oocytes. Here we determine the modulatory effect of the Ca2+ channel beta3 subunit on M2 muscarinic receptor-activated G-protein inhibition and whether the beta3 subunit modulates the G-protein sensitivity of alpha1A and alpha1B currents equivalently. To compare the relative inhibition by muscarinic activation, we have used successive ACh applications to remove the large tonic inhibition of these channels. We show that the resulting rebound potentiation results entirely from the loss of tonic G-protein inhibition; although the currents are temporarily relieved of tonic inhibition, they are still capable of undergoing inhibition through the muscarinic pathway. Using this rebound protocol, we demonstrate that the inhibition of peak current amplitude produced by M2 receptor activation is similar for alpha1A and alpha1B calcium currents. However, the contribution of the voltage-dependent component of inhibition, characterized by reduced inhibition at very depolarized voltage steps and the relief of inhibition by depolarizing prepulses, was slightly greater for the alpha1B current than for the alpha1A current. After co-expression of the beta3 subunit, the sensitivity to M2 receptor-induced G-protein inhibition was reduced for both alpha1A and alpha1B currents; however, the reduction was significantly greater for alpha1A currents. Additionally, the difference in the voltage dependence of inhibition of alpha1A and alpha1B currents was heightened after co-expression of the Ca2+ channel beta3 subunit. Such differential modulation of sensitivity to G-protein modulation may be important for fine tuning release in neurons that contain both of these Ca2+ channels.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/genetics , GTP-Binding Proteins/physiology , Ion Channel Gating/physiology , Acetylcholine/pharmacology , Animals , Electric Conductivity , Electric Stimulation , Ethylmaleimide/pharmacology , Gene Expression/physiology , Ion Channel Gating/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Plasmids , Receptor, Muscarinic M2 , Receptors, Muscarinic/chemistry , Receptors, Muscarinic/genetics , Sulfhydryl Reagents/pharmacology , Virulence Factors, Bordetella/pharmacology , Xenopus
12.
J Pharmacol Exp Ther ; 284(1): 258-68, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9435186

ABSTRACT

Ethanol (EtOH) reversibly activates large conductance, Ca(++)-activated K+ (BK) channels in rat neurohypophysial terminals, an effect that probably contributes to the inhibition of vasopressin release by this drug. Heterogeneity in the terminal channel population makes it difficult to determine the mechanisms underlying this activation. Here, we report the effects of EtOH on the steady-state activity of BK channels cloned from mouse brain (mslo, alpha subunit) and expressed in Xenopus oocytes. EtOH reversibly increased mslo channel activity in excised patches, showing a potency (EC50 = 24 mM) similar to that reported using native channels. EtOH activation was observed under conditions that make it highly improbable that it is mediated by freely diffusible second messengers, or secondary to G-protein modulation. Rather, it probably results from a functional interaction between the drug and the channel alpha subunit. Activation occurred without increase in the number of functional channels present in the patch and resulted from actions that were a function of EtOH concentration: at < or = 10 mM, activation was due to a decrease in the channel mean closed time, whereas between 25 and 100 mM, activation was due to both a decrease in the mean closed time and an increase in the mean open time. The characteristic high unitary conductance and ionic selectivity of BK channels were unaltered by the drug. Whereas the voltage dependence of channel gating remained unchanged, channel activation mediated by the response of the Ca(++)-sensing site(s) to increases in the concentration of intracellular calcium, [Ca++]ic, was reduced by EtOH. This finding is consistent with EtOH and [Ca++]ic behaving functionally as partial and full agonists of mslo channels, respectively. Because the potentiation of mslo activity by the drug decreased as Ca++ levels were increased, EtOH-activation of BK channels would be most evident when [Ca++]ic is near resting levels, rather than during periods of high activity and Ca++ influx.


Subject(s)
Calcium/metabolism , Ethanol/pharmacology , Potassium Channels/drug effects , Animals , Dose-Response Relationship, Drug , Female , Ion Channel Gating/drug effects , Mice , Xenopus laevis
13.
J Membr Biol ; 160(2): 151-60, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9354707

ABSTRACT

A new type of nonselective cation channel was identified and characterized in pheochromocytoma (PC12) cells using inside-out and cell-attached patch-clamp recordings. The channel shows a large unitary conductance (274 pS in symmetric 145 mm K+) and selectivity for Na+ approximately K+ > Li+, and is practically impermeable to Cl-. The channel activity-voltage relationship is bell-shaped, showing maximal activation at approximately -10 mV. The overall activity of this channel is unmodified by [Na+]ic, or [Ca++]ic. However, increases in [Ca++]ic lead to a decrease in the unitary current amplitude. In addition, overall activity is mildly increased when suction is applied to the back of the patch pipette. Together, these characteristics distinguish the present channel from all other large conductance nonselective cation channels reported so far in a variety of preparations. The frequency of appearance of this channel type is similar in undifferentiated and NGF-treated PC12 cells ( approximately 8-27% of patches). The combination of large conductance, permeability to Na+, and existence of conducting states at negative potentials, may provide a significant pathway for inward current and depolarization in PC12 cells.


Subject(s)
Ion Channels/metabolism , Animals , Calcium/metabolism , Cell Differentiation/drug effects , Chlorides/metabolism , Electric Conductivity , Ion Transport , Lithium/metabolism , Membrane Potentials , Nerve Growth Factors/pharmacology , PC12 Cells , Potassium/metabolism , Rats , Sodium/metabolism
14.
Alcohol Clin Exp Res ; 21(6): 1103-7, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9309324

ABSTRACT

It is not known whether alcohols modulate ion channels by directly binding to the channel protein or by perturbing the surrounding membrane lipid. Cutoff describes the phenomenon where the potency of 1-alkanols monotonically increases with alkyl chain length until a loss of efficacy occurs. Determination of the cutoff for a variety of channels can be important, because similar and/or dissimilar cutoffs might yield information regarding the nature of ethanol's site of action. In this study, the two-electrode voltage clamp technique was used to determine the cutoffs for the 1-alkanol potentiation of cloned Ca(2+)-activated-K+ (BK) channels and for the inhibition of cloned Shaw2 K+ channels, expressed in Xenopus oocytes. Ethanol, butanol, hexanol, and heptanol reversibly enhanced BK currents, whereas octanol and nonanol had no effect. In contrast, Shaw2 currents were potently inhibited by both octanol and decanol, but not by undecanol. Taken together, data demonstrate that the modulation of K+ channels by long chain alcohols is channel-specific. Interestingly, ethanol was a less potent activator of BK currents in the intact oocyte in comparison with its effect on this channel in excised membrane patches. The decrease in potency could not be attributed to an ethanol-dependent change in Ca2+ influx through endogenous voltage-gated channels, an effect that would alter the concentration of Ca2+ available to activate BK channels.


Subject(s)
Alcohols/toxicity , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Animals , Cloning, Molecular , Dose-Response Relationship, Drug , Membrane Potentials/drug effects , Oocytes , Patch-Clamp Techniques , Potassium Channels/genetics , Shaw Potassium Channels , Xenopus
16.
Brain Res Mol Brain Res ; 39(1-2): 99-108, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8804718

ABSTRACT

Four splice variants of the NR1 receptor subunit, characterized by the presence or absence of cassettes encoding inserts of 21 (Insert 1) and 37 (Insert 2) amino acids were expressed in Xenopus oocytes and studied using voltage-clamp techniques. In 1.8 mM Ca2+, a slow inward current (Islow), which peaked 20 s after exposure to NMDA was evident when Insert I was present, but not when absent. However, in elevated external Ca2+ medium a similar Islow was observed in variants missing Insert I. The Ca2+ dependency of Islow reflected a requirement for intracellular accumulation of Ca2+. The divalent ion permeability of Insert I containing and Insert 1 lacking receptor channels expressed alone, as well as in heteromeric assemblies with NR2A and NR2B, was similar for all combinations tested. Thus, the lower Ca2+ dependency for Islow in oocytes expressing Insert I was not due to higher calcium entry. Islow was less sensitive to blockers of ICl(Ca) than were endogenous calcium-activated chloride currents (ICl(Ca)). Also, Islow was not abolished in Cl(-)-free external medium, when voltage was manipulated such that Islow was outward-going. Thus, Islow, while containing a component due to activation of endogenous ICl(Ca), is primarily due to current flowing through the receptor ion channel. Development of Islow was unaffected by PKC or PKA inhibitors. The modulation of the Ca2+ dependency of Islow by Insert I occurs in a range of Ca2+ concentrations which are physiologically relevant, and may provide an important means of modulation of glutamate transmission under normal and pathological conditions.


Subject(s)
Alternative Splicing/genetics , Calcium/pharmacology , Oocytes/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate/genetics , Xenopus
17.
Anesthesiology ; 84(4): 900-9, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8638845

ABSTRACT

BACKGROUND: There is increasing evidence that direct interactions between volatile anesthetics and channel proteins may result in general anesthesia. Using voltage-clamp techniques, the authors examined the effect of two general anesthetics (ketamine and halothane) on a rat brain potassium channel of known amino acid sequence, and further assessed whether the inhibition of the channel is altered by a partial deletion of the C-terminal sequence of this channel. METHODS: Xenopus laevis oocytes were microinjected with either Kv2.1 or delta C318 (a mutated channel in which the last 318 amino acids of the C-terminus have been deleted) cRNA, and channel function in translated channels was observed before, during, and after exposure to graded concentrations of ketamine (25, 50, and 75 micrometers) and halothane (1%, 2%, and 4%). RESULTS: Ketamine and halothane reduced Kv2.1 and delta C318 peak current amplitude in a dose-dependent and reversible fashion. The inhibition of current was voltage dependent for halothane but not for ketamine. Halothane accelerated the time constant of current inactivation, whereas ketamine affected this parameter minimally in both channel types. Use dependence of ketamine and halothane action was observed in both Kv2.1 and the mutant channel, attributable to augmentation of C-type inactivation. CONCLUSIONS: Although both ketamine and halothane inhibit potassium currents through the Kv2.1 channel, their mechanisms of action at this potential target may be different. Deletion of the C-terminal sequence resulted in decreased sensitivity to both anesthetics. Although it is not clear whether anesthetics interact directly with the C-terminus, which is thought to reside intracellularly, this portion of the channel protein clearly influences the actions of both anesthetics.


Subject(s)
Anesthetics, Dissociative/pharmacology , Halothane/pharmacology , Ketamine/pharmacology , Potassium Channel Blockers , Anesthetics, Inhalation/pharmacology , Animals , Brain/drug effects , Female , Rats , Recombinant Proteins/antagonists & inhibitors , Xenopus laevis
18.
Mol Pharmacol ; 49(1): 40-8, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8569710

ABSTRACT

Large conductance, Ca(2+)-activated K+ channels are believed to underlie interburst intervals and, thus, contribute to the control of hormone release from neurohypophysial terminals. Because ethanol inhibits the release of vasopressin and oxytocin, we studied its effects on large conductance, Ca(2+)-activated K+ channels from these terminals using patch-clamp techniques. Ethanol (10-100 mM) applied to the cytosolic surface of excised, inside-out patches reversibly increases channel activity in a concentration-dependent manner, reaching a plateau at 50-100 mM. This activation is not mediated by freely diffusible cytosolic second messengers or the release of Ca2+ from intracellular stores. Rather, it likely reflects a direct interaction of ethanol with the channel protein or a closely associated component. Neither the unitary conductance nor the characteristics of the voltage-current relationship are modified by the drug. The increase of channel activity by ethanol results from a modification of channel gating properties: the contribution of long openings to the total time spent in the open state is increased, the average duration of the fast openings is slightly increased, and long closures disappear in the presence of the drug. The activation of large conductance, Ca(2+)-activated K+ channels by ethanol, in conjunction with the previously reported inhibition of voltage-dependent Ca2+ channels, can explain the reduced release of vasopressin and oxytocin after ethanol ingestion.


Subject(s)
Calcium/pharmacology , Ethanol/pharmacology , Pituitary Gland/drug effects , Potassium Channels/drug effects , Presynaptic Terminals/drug effects , Animals , Ion Channel Gating , Ion Transport , Male , Pituitary Gland/physiology , Potassium/metabolism , Potassium Channels/physiology , Presynaptic Terminals/physiology , Rats
19.
FEBS Lett ; 371(1): 43-6, 1995 Aug 28.
Article in English | MEDLINE | ID: mdl-7664882

ABSTRACT

Three different classes of alpha 1 Ca2+ channel (alpha 1A, alpha 1B, alpha 1C) were expressed in Xenopus oocytes to determine whether G protein-mediated inhibition is an inherent property of the alpha 1 subunit itself, and if so, whether co-expression of auxiliary subunits modulates the inhibition seen. From our data it is apparent that either alpha 1A or alpha 1B Ca2+ channels expressed alone are sufficient for voltage-dependent G protein inhibition. alpha 1C Ca2+ channels expressed alone do not exhibit the G protein inhibition seen in alpha 1A and alpha 1B channels. Additionally, co-expression of the beta 3 subunit abolishes the ability of G proteins to inhibit currents through alpha 1A and alpha 1B Ca2+ channels. Differential sensitivity of alpha 1 as well as modulation of properties by beta 3 provide a potential mechanism for the regulation of G protein-mediated inhibition in neurons.


Subject(s)
Calcium Channels/physiology , GTP-Binding Proteins/physiology , Animals , Calcium Channels/biosynthesis , Electrophysiology , Ethylmaleimide/pharmacology , GTP-Binding Proteins/antagonists & inhibitors , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , Membrane Potentials/drug effects , Microinjections , Oocytes , RNA, Complementary , Thionucleotides/pharmacology , Xenopus laevis
20.
J Neurochem ; 65(1): 140-8, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7540660

ABSTRACT

The NMDA receptor/channel has been shown to be inhibited by ethanol in the clinically relevant range 25-100 mM. We studied heteromeric assemblies (NR1b/NR2) of the NMDA receptor, expressed in oocytes, to address three questions regarding this inhibition, and discovered the following: (1) The inhibition was nearly equivalent when ethanol was coapplied with the agonist, and when ethanol was introduced after steady-state current was established, suggesting that ethanol does not act by interfering with the activation process of the NMDA receptor. (2) The degree of inhibition was controlled by the NR2 subunit, with both NR2A and NR2B significantly more sensitive to ethanol than NR2C and NR2D. (3) Manipulation of the NMDA receptor with a number of agents that normally modulate it did not alter the degree of inhibition produced by ethanol. The presence of Mg2+ (3 and 12.5 microM), Zn2+ (1 and 10 microM), or the glycine antagonist 7-chlorokynurenic acid (1.25 or 5 microM), did not alter the ethanol sensitivity of heteromeric (NR1b/NR2A, NR1b/NR2B, NR1b/NR2C) NMDA receptors. Redox modulation of the NMDA receptor by dithiothreitol (2 mM) or 5,5'-dithiobis(2-nitrobenzoic acid) (1 mM) also did not alter the degree to which ethanol inhibits NMDA receptors. Taken together, these results indicate that the ethanol sensitivity of native NMDA receptors, which likely exist in heteromeric form, results from actions at a site different from those of known modulators of the receptor.


Subject(s)
Ethanol/pharmacology , Ion Channels/antagonists & inhibitors , Ion Channels/chemistry , N-Methylaspartate/metabolism , Animals , Electrophysiology , Female , Ion Channels/physiology , Kynurenic Acid/analogs & derivatives , Kynurenic Acid/pharmacology , Magnesium/pharmacology , Oxidation-Reduction , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Recombinant Proteins , Xenopus laevis , Zinc/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...