Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 171(23): 5446-57, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25117207

ABSTRACT

BACKGROUND AND PURPOSE: Neurosteroids potentiate responses of the GABAA receptor to the endogenous agonist GABA. Here, we examined the ability of neurosteroids to potentiate responses to the allosteric activators etomidate, pentobarbital and propofol. EXPERIMENTAL APPROACH: Electrophysiological assays were conducted on rat α1ß2γ2L GABAA receptors expressed in HEK 293 cells. The sedative activity of etomidate was studied in Xenopus tadpoles and mice. Effects of neurosteroids on etomidate-elicited inhibition of cortisol synthesis were determined in human adrenocortical cells. KEY RESULTS: The neurosteroid 5ß-pregnan-3α-ol-20-one (3α5ßP) potentiated activation of GABAA receptors by GABA and allosteric activators. Co-application of 1 µM 3α5ßP induced a leftward shift (almost 100-fold) of the whole-cell macroscopic concentration-response relationship for gating by etomidate. Co-application of 100 nM 3α5ßP reduced the EC50 for potentiation by etomidate of currents elicited by 0.5 µM GABA by about three-fold. In vivo, 3α5ßP (1mg kg(-1) ) reduced the dose of etomidate required to produce loss of righting in mice (ED50 ) by almost 10-fold. In tadpoles, the presence of 50 or 100 nM 3α5ßP shifted the EC50 for loss of righting about three- or ten-fold respectively. Exposure to 3α5ßP did not influence inhibition of cortisol synthesis by etomidate. CONCLUSIONS AND IMPLICATIONS: Potentiating neurosteroids act similarly on orthosterically and allosterically activated GABAA receptors. Co-application of neurosteroids with etomidate can significantly reduce dosage requirements for the anaesthetic, and is a potentially beneficial combination to reduce undesired side effects.


Subject(s)
Etomidate/pharmacology , Hypnotics and Sedatives/pharmacology , Pregnanolone/pharmacology , Receptors, GABA-A/metabolism , Animals , Behavior, Animal/drug effects , Cell Line , Drug Synergism , HEK293 Cells , Humans , Hydrocortisone/metabolism , Mice, Inbred BALB C , Rats , Receptors, GABA-A/physiology , Xenopus laevis
2.
Br J Pharmacol ; 164(2b): 667-80, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21457224

ABSTRACT

BACKGROUND AND PURPOSE: A 'lock-and-key' binding site typically accounts for the effect of receptor antagonists. However, sulphated neurosteroids are potent non-competitive antagonists of GABA(A) receptors without a clear structure-activity relationship. To gain new insights, we tested two structurally unrelated hydrophobic anions with superficially similar properties to sulphated neurosteroids. EXPERIMENTAL APPROACH: We used voltage-clamp techniques in Xenopus oocytes and hippocampal neurons to characterize dipicrylamine (DPA) and tetraphenylborate (TPB), compounds previously used to probe membrane structure and voltage-gated ion channel function. KEY RESULTS: Both DPA and TPB potently antagonized GABA(A) receptors. DPA exhibited an IC50 near 60 nM at half-maximal GABA concentration and antagonism with features indistinguishable from pregnenolone sulphate antagonism, including sensitivity to a point mutation in transmembrane domain 2 of the α1 subunit. Bovine serum albumin, which scavenges free membrane-associated DPA, accelerated both capacitance offset and antagonism washout. Membrane interactions and antagonism were explored using the voltage-dependent movement of DPA between membrane leaflets. Washout of DPA antagonism was strongly voltage-dependent, paralleling DPA membrane loss, although steady-state antagonism lacked voltage dependence. At antagonist concentrations, DPA failed to affect inhibitory post-synaptic current (IPSC) amplitude or decay, but DPA accelerated pharmacologically prolonged IPSCs. CONCLUSIONS AND IMPLICATIONS: Neurosteroid-like GABA(A) receptor antagonism appears to lacks a conventional binding site. These features highlight key roles of membrane interactions in antagonism. Because its membrane mobility can be controlled, DPA may be a useful probe of GABA(A) receptors, but its effects on excitability via GABA(A) receptors raise caveats for its use in monitoring neuronal activity.


Subject(s)
GABA-A Receptor Antagonists/pharmacology , Picrates/pharmacology , Receptors, GABA-A/metabolism , Tetraphenylborate/pharmacology , Animals , Anions/chemistry , Anions/pharmacology , Binding Sites , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Electric Capacitance , Female , GABA-A Receptor Antagonists/chemistry , Hippocampus/drug effects , Hippocampus/metabolism , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques/methods , Picrates/chemistry , Pregnenolone/chemistry , Pregnenolone/pharmacology , Protein Structure, Tertiary , Rats , Sensitivity and Specificity , Structure-Activity Relationship , Synaptic Potentials/drug effects , Synaptic Transmission/drug effects , Tetraphenylborate/chemistry , Xenopus laevis
3.
Cell Death Dis ; 1: e63, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-21364668

ABSTRACT

In clinical obstetrics, magnesium sulfate (MgSO(4)) use is widespread, but effects on brain development are unknown. Many agents that depress neuronal excitability increase developmental neuroapoptosis. In this study, we used dissociated cultures of rodent hippocampus to examine the effects of Mg(++) on excitability and survival. Mg(++)-induced caspase-3-associated cell loss at clinically relevant concentrations. Whole-cell patch-clamp techniques measured Mg(++) effects on action potential threshold, action potential peak amplitude, spike number and changes in resting membrane potential. Mg(++) depolarized action potential threshold, presumably from surface charge screening effects on voltage-gated sodium channels. Mg(++) also decreased the number of action potentials in response to fixed current injection without affecting action potential peak amplitude. Surprisingly, Mg(++) also depolarized neuronal resting potential in a concentration-dependent manner with a +5.2 mV shift at 10 mM. Voltage ramps suggested that Mg(++) blocked a potassium conductance contributing to the resting potential. In spite of this depolarizing effect of Mg(++), the net inhibitory effect of Mg(++) nearly completely silenced neuronal network activity measured with multielectrode array recordings. We conclude that although Mg(++) has complex effects on cellular excitability, the overall inhibitory influence of Mg(++) decreases neuronal survival. Taken together with recent in vivo evidence, our results suggest that caution may be warranted in the use of Mg(++) in clinical obstetrics and neonatology.


Subject(s)
Apoptosis/drug effects , Magnesium/pharmacology , Membrane Potentials/drug effects , Neurons/cytology , Neurons/drug effects , Animals , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Hippocampus/cytology , Neurons/enzymology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
4.
Neuroscience ; 158(1): 211-22, 2009 Jan 12.
Article in English | MEDLINE | ID: mdl-18472347

ABSTRACT

Axonal action potentials initiate the cycle of synaptic communication that is key to our understanding of nervous system functioning. The field has accumulated vast knowledge of the signature action potential waveform, firing patterns, and underlying channel properties of many cell types, but in most cases this information comes from somatic intracellular/whole-cell recordings, which necessarily measure a mixture of the currents compartmentalized in the soma, dendrites, and axon. Because the axon in many neuron types appears to be the site of lowest threshold for action potential initiation, the channel constellation in the axon is of particular interest. However, the axon is more experimentally inaccessible than the soma or dendrites. Recent studies have developed and applied single-fiber extracellular recording, direct intracellular recording, and optical recording techniques from axons toward understanding the behavior of the axonal action potential. We are starting to understand better how specific channels and other cellular properties shape action potential threshold, waveform, and timing: key elements contributing to downstream transmitter release. From this increased scrutiny emerges a theme of axons with more computational power than in traditional conceptualizations.


Subject(s)
Action Potentials/physiology , Axons/physiology , Central Nervous System/physiology , Synaptic Transmission/physiology , Animals , Cell Membrane/physiology , Electrophysiology/methods , Glutamic Acid/metabolism , Humans , Potassium Channels/physiology , Sodium Channels/physiology
5.
Handb Exp Pharmacol ; (182): 53-84, 2008.
Article in English | MEDLINE | ID: mdl-18175086

ABSTRACT

Excitatory transmitter-gated receptors are found in three gene families: the glutamate ionotropic receptors, the Cys-loop receptor family (nicotinic and 5HT3), and the purinergic (P2X) receptors. Anesthetic drugs act on many members of these families, but in most cases the effects are unlikely to be related to clinically relevant anesthetic actions. However, the gaseous anesthetics (xenon and nitrous oxide) and the dissociative anesthetics (ketamine) have significant inhibitory activity at one type of glutamate receptor (the NMDA receptor) that is likely to contribute to anesthetic action. It is possible that some actions at neuronal nicotinic receptors may make a smaller contribution to effects of some anesthetics.


Subject(s)
Anesthetics/pharmacology , Central Nervous System/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Ion Channel Gating/drug effects , Ion Channels/drug effects , Nicotinic Antagonists/pharmacology , Receptors, Neurotransmitter/antagonists & inhibitors , Serotonin Antagonists/pharmacology , Animals , Central Nervous System/metabolism , Humans , Ion Channels/metabolism , Models, Molecular , Protein Conformation , Purinergic P2 Receptor Antagonists , Receptors, Glutamate/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Neurotransmitter/chemistry , Receptors, Neurotransmitter/metabolism , Receptors, Nicotinic/drug effects , Receptors, Serotonin/drug effects
6.
Br J Pharmacol ; 150(2): 164-75, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17160009

ABSTRACT

BACKGROUND AND PURPOSE: Neuroactive steroids are potent modulators of GABA(A) receptors and are thus of interest for their sedative, anxiolytic, anticonvulsant and anaesthetic properties. Cyclodextrins may be useful tools to manipulate neuroactive effects of steroids on GABA(A) receptors because cyclodextrins form inclusion complexes with at least some steroids that are active at the GABA(A) receptor, such as (3alpha,5alpha)-3-hydroxypregnan-20-one (3alpha5alphaP, allopregnanolone). EXPERIMENTAL APPROACH: To assess the versatility of cyclodextrins as steroid modulators, we investigated interactions between gamma-cyclodextrin and neuroactive steroids of different structural classes. KEY RESULTS: Both a bioassay based on electrophysiological assessment of GABA(A) receptor function and optical measurements of cellular accumulation of a fluorescent steroid analogue suggest that gamma-cyclodextrin sequesters steroids rather than directly influencing GABA(A) receptor function. Neither a 5beta-reduced A/B ring fusion nor a sulphate group at carbon 3 affected the presumed inclusion complex formation between steroid and gamma-cyclodextrin. Apparent dissociation constants for interactions between natural steroids and gamma-cyclodexrin ranged from 10-60 microM. Although gamma-cyclodextrin accommodates a range of natural and synthetic steroids, C(11) substitutions reduced inclusion complex formation. Using gamma-cyclodextrin to remove steroid not directly bound to GABA(A) receptors, we found that cellular retention of receptor-unbound steroid rate limits potentiation by 3alpha- hydroxysteroids but not inhibition by sulphated steroids. CONCLUSIONS AND IMPLICATIONS: We conclude that gamma-cyclodextrins can be useful, albeit non-specific, tools for terminating the actions of multiple classes of naturally occurring neuroactive steroids.


Subject(s)
Cyclodextrins/pharmacology , Steroids/pharmacology , Animals , Cells, Cultured , Drug Interactions , Female , Hippocampus/cytology , In Vitro Techniques , Oocytes/drug effects , Oocytes/physiology , Patch-Clamp Techniques , Rats , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Stereoisomerism , Steroids/chemistry , Steroids/physiology , Structure-Activity Relationship , Xenopus laevis , gamma-Cyclodextrins/pharmacology
7.
Neuroscience ; 131(2): 349-58, 2005.
Article in English | MEDLINE | ID: mdl-15708478

ABSTRACT

We investigated conditions that promote basal and activity-dependent neuronal apoptosis in postnatal rat hippocampal cultures. Low-density mixed cultures of astrocytes and neurons exhibited lower sensitivity than high-density cultures to basal neuronal death and activity-sensitive neuronal death, induced with glutamate receptor blockers, sodium channel blockers, or calcium channel blockers. Although elevations of [Ca(2+)](i) protect neurons from apoptosis, low-density microcultures and mass cultures exhibited only minor differences in resting [Ca(2+)](i) and Ca(2+) current density, suggesting that these variables are unlikely to explain differences in susceptibility. Astrocytes, rather than neurons, were implicated in the neuronal loss. Several candidate molecules implicated in other astrocyte-dependent neurotoxicity models were excluded, but heat inactivation experiments suggested that a heat-labile factor is critically involved. In sum, our results suggest the surprising result that astrocytes can be negative modulators of neuronal survival during development and when the immature nervous system is challenged with drugs that dampen electrical excitability.


Subject(s)
Apoptosis/physiology , Astrocytes/physiology , Hippocampus/physiology , Animals , Animals, Newborn , Apoptosis/drug effects , Astrocytes/cytology , Astrocytes/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Glutamic Acid/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Nifedipine/pharmacology , Rats
8.
Neuropharmacology ; 42(2): 199-209, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11804616

ABSTRACT

The antiepileptic drug riluzole is a use-dependent blocker of voltage-gated Na(+) channels and selectively depresses action potential-driven glutamate over gamma-aminobutyric acid (GABA) release. Here we report that in addition to its presynaptic effect, riluzole at higher concentrations also strongly potentiates postsynaptic GABA(A) responses both in cultured hippocampal neurons and in Xenopus oocytes expressing recombinant receptors. Although peak inhibitory postsynaptic currents (IPSCs) of autaptic hippocampal neurons were inhibited, 20-100 microM riluzole significantly prolonged the decay of IPSCs, resulting in little change in total charge transfer. The effect was dose-dependent and reversible. Riluzole selectively increased miniature IPSC fast and slow decay time constants, without affecting their relative proportions. Miniature IPSC peak amplitude, rise time and frequency were unaffected, indicating a postsynaptic mechanism. In the Xenopus oocyte expression system, riluzole potentiated GABA responses by lowering the EC(50) for GABA activation. Riluzole directly gated a GABA(A) current that was partially blocked by bicuculline and gabazine. Pharmacological experiments suggest that the action of riluzole did not involve a benzodiazepine, barbiturate, or neurosteroid site. Instead, riluzole-induced potentiation was inhibited by the lactone antagonist alpha-isopropyl-alpha-methyl-gamma-butyrolatone (alpha-IMGBL). While most anticonvulsants either block voltage-gated Na(+) channels or potentiate GABA(A) receptors, our results suggest that riluzole may define an advantageous class of anticonvulsants with both effects.


Subject(s)
GABA Agonists/pharmacology , GABA-A Receptor Agonists , Neuroprotective Agents/pharmacology , Riluzole/pharmacology , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , GABA Antagonists/pharmacology , GABA Modulators/pharmacology , GABA-A Receptor Antagonists , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/chemistry , Synapses/drug effects , Xenopus , gamma-Aminobutyric Acid/pharmacology
9.
Brain Res Brain Res Rev ; 37(1-3): 91-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11744077

ABSTRACT

GABAergic neurotransmission can be both positively and negatively modulated by steroids. The steroid effects are thought to be mediated by binding of steroids to specific sites on GABA(A) receptors. It appears that the receptor sites for positive and negative modulatory steroids are different. Thus far, the location and number of binding sites for steroids on these receptors have not been established. In this brief review, we concentrate largely on results from our own structure-activity studies. Novel analogues have been studied to further delineate the structural features required for compounds to modulate receptor function via steroid binding sites. Non-naturally occurring enantiomers of both positive and negative modulators have been studied to provide further evidence for the existence of specific steroid binding sites on the receptors.


Subject(s)
Receptors, GABA-A/metabolism , Steroids/chemistry , Steroids/metabolism , Animals , Binding Sites/physiology , Humans , Steroids/pharmacology , Structure-Activity Relationship , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
10.
Mol Pharmacol ; 60(4): 732-41, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562435

ABSTRACT

Neurosteroids positively and negatively modulate gamma-aminobutyric acid (GABA)(A) receptors and glutamate receptors, which underlie most fast inhibition and excitation in the central nervous system. We report the identification of a neuroactive steroid, (3 alpha,5 beta)-20-oxo-pregnane-3-carboxylic acid (3 alpha 5 beta PC), with unique cellular actions. 3 alpha 5 beta PC positively modulates GABA(A) receptor function and negatively modulates N-methyl-D-aspartate (NMDA) receptor function, a combination that may be of particular clinical benefit. 3 alpha 5 beta PC promotes net GABA(A) potentiation at low steroid concentrations (<10 microM) and at negative membrane potentials. At higher concentrations, the steroid also blocks GABA receptors. Because this block would presumably counteract the NMDA receptor blocking actions of 3 alpha 5 beta PC, we characterize the GABA receptor block in some detail. Agonist concentration, depolarization, and high extracellular pH increase the block. The apparent pK for both potentiation and block was 6.4 to 6.9, substantially higher than expected from carboxylated steroid in an aqueous environment. Block is not dependent on the stereochemistry of the carboxylic acid at carbon 3 and is relatively insensitive to placement of the carboxylic acid at the opposite end of the steroid (carbon 24). Potentiation is critically dependent on the stereochemistry of the carboxylic acid group at carbon 3. Consistent with the pH dependence of potentiation, effects of the amide derivative (3 alpha,5 beta)-20-oxo-pregnane-3-carboxamide, suggest that the un-ionized form of 3 alpha 5 beta PC is important for potentiation, whereas the ionized form is probably responsible for block. Further refinement of the neuroactive steroid to promote GABA potentiation and NMDA receptor block and diminish GABA receptor block may lead to a clinically useful neuroactive steroid.


Subject(s)
Hippocampus/drug effects , Pregnanes/pharmacology , Receptors, GABA-A/metabolism , Steroids/pharmacology , Animals , Electrophysiology , Hippocampus/metabolism , Hydrogen-Ion Concentration , In Vitro Techniques , Rats , Receptors, GABA-A/drug effects , Synapses/drug effects , Synapses/metabolism , Xenopus laevis
11.
Mol Pharmacol ; 60(3): 603-10, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11502893

ABSTRACT

Although nitrous oxide (N(2)O; laughing gas) remains widely used as an anesthetic and analgesic in clinical practice, its cellular mechanisms of action remain inadequately understood. In this report, we examined the effects of N(2)O on voltage-gated Ca(2+) channels in acutely dissociated small sensory neurons of adult rat. At subanesthetic concentrations, N(2)O blocks low-voltage-activated, T-type Ca(2+) currents (T currents), but not high-voltage-activated (HVA) currents. This blockade of T currents was concentration dependent, with an IC(50) value of 45 +/- 13%, maximal block of 38 +/- 12%, and Hill coefficient of 2.6 +/- 1.0. No desensitization of the response or change in current kinetics was observed during N(2)O application. The magnitude of T current blockade by N(2)O does not seem to reflect any use- or voltage-dependent properties. In addition, T current blockade was not altered when intracellular GTP was replaced with guanosine 5'-(gamma-thio)triphosphate or guanosine 5'-0-(2-thiodiphosphate) suggesting a lack of involvement of G-proteins in the inhibition. N(2)O selectively blocked currents arising from the Ca(v)3.2 but not Ca(v)3.1 recombinant channels stably expressed in human embryonic kidney (HEK) cells in a concentration-dependent manner with an apparent affinity and potency similar to native dorsal root ganglion currents. Analogously, the block of Ca(v)3.2 T currents exhibited little voltage- or use-dependence. These data indicate that N(2)O selectively blocks T-type but not HVA Ca(2+) currents in small sensory neurons and Ca(v)3.2 currents in HEK cells at subanesthetic concentrations. Blockade of T currents may contribute to the anesthetic and/or analgesic effects of N(2)O.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/physiology , Neurons, Afferent/drug effects , Nitrous Oxide/pharmacology , Animals , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Cells, Cultured , Electrophysiology , Humans , Neurons, Afferent/physiology , Rats , Transfection
12.
Neuron ; 31(1): 75-85, 2001 Jul 19.
Article in English | MEDLINE | ID: mdl-11498052

ABSTRACT

Although T-type calcium channels were first described in sensory neurons, their function in sensory processing remains unclear. In isolated rat sensory neurons, we show that redox agents modulate T currents but not other voltage- and ligand-gated channels thought to mediate pain sensitivity. Similarly, redox agents modulate currents through Ca(v)3.2 recombinant channels. When injected into peripheral receptive fields, reducing agents, including the endogenous amino acid L-cysteine, induce thermal hyperalgesia. This hyperalgesia is blocked by the oxidizing agent 5,5'-dithio-bis-(2-nitrobenzoic acid) (DTNB) and the T channel antagonist mibefradil. DTNB alone and in combination with mibefradil induces thermal analgesia. Likewise, L-cysteine induces mechanical DTNB-sensitive hyperalgesia in peripheral receptive fields. These data strongly suggest a role for T channels in peripheral nociception. Redox sites on T channels in peripheral nociceptors could be important targets for agents that modify pain perception.


Subject(s)
Calcium Channels, T-Type/physiology , Ganglia, Spinal/physiology , Membrane Potentials/physiology , Neurons, Afferent/physiology , Neurons/physiology , Nociceptors/physiology , Pain/physiopathology , Analysis of Variance , Animals , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Cell Line , Cells, Cultured , Cysteine/pharmacology , Dithionitrobenzoic Acid/pharmacology , Dithiothreitol/pharmacology , Female , Hot Temperature , Humans , Hyperalgesia/physiopathology , Membrane Potentials/drug effects , Neurons/drug effects , Neurons, Afferent/drug effects , Oxidation-Reduction , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Skin/innervation , Transfection
13.
Glia ; 33(1): 24-35, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11169789

ABSTRACT

Neuronal activity elicits increases in intracellular Ca2+ in astrocytes, which in turn can elevate neuronal Ca2+ and potentiate the efficacy of excitatory synaptic transmission. Therefore, understanding the modulation of astrocyte Ca2+ elevations by neurotransmitters should aid in understanding astrocyte-neuronal interactions. On cultured hippocampal microislands containing only astrocytes, activation of metabotropic glutamate receptors (mGluRs) with the specific agonist 1S,3R-ACPD triggers Ca2+ elevations that are potentiated by adenosine A1 receptor activation. A1 receptor modulation of mGluR-induced Ca2+ elevations is blocked by pertussis toxin and is mimicked by the wasp venom peptide mastoparan, suggesting that potentiation occurs by means of a G(i/o) mechanism. Surprisingly, on microislands containing only astrocytes, A1 receptor antagonism or adenosine degradation suppresses mGluR-triggered Ca2+ elevations, strongly suggesting that astrocytes are a source of physiologically relevant concentrations of adenosine.


Subject(s)
Adenosine/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Cells, Cultured/drug effects , Cells, Cultured/metabolism , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , Rats , Rats, Inbred Strains , Receptor, Adenosine A2A , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/drug effects , Receptors, Purinergic P1/metabolism
14.
Mol Pharmacol ; 59(1): 46-53, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11125023

ABSTRACT

Some, perhaps all, G protein-coupled receptors form homo- or heterodimers. We have shown that metabotropic glutamate receptors are covalent dimers, held together by one or more disulfide bonds near the N terminus. Here we report how mutating cysteines in this region affect dimerization and function. Covalent dimerization is preserved when cysteines 57, 93, or 99 are mutated but lost with replacement at 129. Coimmunoprecipitation under nondenaturing conditions indicates that the C[129]S mutant receptor remains a dimer, via noncovalent interactions. Both C[93]S and C[129]S bind [3H]quisqualate, whereas binding to C[57]S or C[99]S mutants is absent or greatly attenuated. The C[93]S and C[129]S receptors have activity similar to wild-type when assayed by fura-2 imaging of intracellular calcium in human embryonic kidney cells or electrophysiologically in Xenopus laevis oocytes. In contrast, C[57]S or C[99]S are less active in both assays but do respond with higher glutamate concentrations in the oocyte assay. These results demonstrate that 1) covalent dimerization is not critical for mGlu5 binding or function; 2) mGlu5 remains a noncovalent dimer even in the absence of covalent dimerization; and 3) high-affinity binding requires Cys-57 and Cys-99.


Subject(s)
Cysteine/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Biological Transport , Calcium/metabolism , Cells, Cultured , Chloride Channels/metabolism , Cysteine/genetics , Dimerization , Excitatory Amino Acid Agonists/pharmacology , Humans , Point Mutation , Quisqualic Acid/pharmacology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Transfection , Tritium
16.
Neuron ; 26(3): 671-82, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10896162

ABSTRACT

Sodium channels (NaChs) play a central role in action potential generation and are uniquely poised to influence the efficacy of transmitter release. We evaluated the effect of partial NaCh blockade on two aspects of synaptic efficacy First, we evaluated whether NaCh blockade accounts for the ability of certain drugs to selectively depress glutamate release. Second, we evaluated the contribution of NaChs to intraneuronal variability in glutamate release probability (p(r)). The antiglutamate drug riluzole nearly completely depresses glutamate excitatory postsynaptic currents (EPSCs) at concentrations that barely affect GABAergic inhibitory postsynaptic currents (IPSCs). NaCh inhibition explains the selective depression. Unlike other presynaptic depressants, partial NaCh blockade increases paired-pulse EPSC depression. This result is explained by selective depression of low-p(r) synapses. We conclude that local variations in the action potential contribute to p(r) variability among excitatory synapses.


Subject(s)
Sodium Channels/physiology , Synapses/physiology , Action Potentials/physiology , Animals , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Presynaptic Terminals/physiology , Rats , Rats, Sprague-Dawley , Riluzole/pharmacology , Sodium Channel Blockers , Sodium Channels/metabolism , Synapses/drug effects , Synaptic Transmission/physiology
17.
J Neurosci ; 20(10): 3571-9, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10804198

ABSTRACT

We examined the effects of the neurosteroid pregnenolone sulfate (PS) on GABA(A) receptor-mediated synaptic currents and currents elicited by rapid applications of GABA onto nucleated outside-out patches in cultured postnatal rat hippocampal neurons. At 10 microm, PS significantly depressed peak responses and accelerated the decay of evoked inhibitory synaptic currents. In nucleated outside-out patches, PS depressed peak currents and speeded deactivation after 5 msec applications of a saturating concentration of GABA. PS also increased the rate and degree of macroscopic GABA receptor desensitization during prolonged GABA applications. In a paired GABA application paradigm, PS slowed the rate of recovery from desensitization. In contrast to its prominent effects on currents produced by saturating GABA concentrations, PS had only small effects on peak currents and failed to alter deactivation after brief applications of the weakly desensitizing GABA(A) receptor agonists taurine and beta-alanine. However, when beta-alanine was applied for a sufficient duration to promote receptor desensitization, PS augmented macroscopic desensitization and slowed deactivation. These results suggest that PS inhibits GABA-gated chloride currents by enhancing receptor desensitization and stabilizing desensitized states. This contention is supported by kinetic modeling studies in which increases in the rate of entry into doubly liganded desensitized states mimic most effects of PS.


Subject(s)
Neural Inhibition/drug effects , Neurons/physiology , Pregnenolone/pharmacology , Receptors, GABA-A/metabolism , Synaptic Transmission/drug effects , Animals , Cells, Cultured , Hippocampus/cytology , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/chemistry , Neurons/cytology , Patch-Clamp Techniques , Rats , Rats, Inbred Strains , Synapses/chemistry , Synapses/metabolism , Taurine/pharmacology , beta-Alanine/pharmacology , gamma-Aminobutyric Acid/pharmacology
18.
J Neurosci ; 20(9): 3147-56, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10777778

ABSTRACT

Neurotransmitters can have both toxic and trophic functions in addition to their role in neural signaling. Surprisingly, chronic blockade of GABA(A) receptor activity for 5-8 d in vitro enhanced survival of hippocampal neurons, suggesting that GABA(A) receptor overactivation may be neurotoxic. Potentiating GABA(A) receptor activity by chronic treatment with the endogenous neurosteroid (3alpha,5alpha)-3-hydroxypregnan-20-one caused massive cell loss over 1 week in culture. Other potentiators of GABA(A) receptors, including benzodiazepines, mimicked the cell loss, suggesting that potentiating endogenous GABA activity is sufficient to produce neuronal death. Neurosteroid-treated neurons had lower resting intracellular calcium levels than control cells and produced smaller calcium rises in response to depolarizing challenges. Manipulating intracellular calcium levels with chronic elevated extracellular potassium or with the calcium channel agonist Bay K 8644 protected neurons. The results may have implications for the mechanisms of programmed cell death in the developing CNS as well as implications for the long-term consequences of chronic GABAmimetic drug use during development.


Subject(s)
Calcium/metabolism , Cell Death/physiology , GABA Antagonists/pharmacology , GABA Modulators/pharmacology , Neurons/physiology , Receptors, GABA-A/drug effects , gamma-Aminobutyric Acid/drug effects , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcium Channel Agonists/pharmacology , Cell Death/drug effects , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , N-Methylaspartate/pharmacology , Potassium Chloride/pharmacology , Rats , Receptors, GABA-A/physiology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
19.
Curr Opin Investig Drugs ; 1(3): 360-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11249720

ABSTRACT

Neuroactive steroids rapidly modulate gamma-aminobutyric acid (GABA) and glutamate receptors. GABA-enhancing steroids have potential clinical utility as anesthetics, hypnotics, anticonvulsants and anxiolytics. Furthermore, GABAergic neurosteroids may participate in regulating mood and the effects of alcohol on the nervous system, suggesting a potential role in major psychiatric disorders. Neuroactive steroids that alter the function of glutamate receptors could be useful for treating neurodegenerative disorders, and as cognitive enhancers. Recent progress in developing water-soluble steroids and steroids with enhanced oral efficacy foster optimism that certain neuroactive steroids will be developed for clinical use.


Subject(s)
Mental Disorders/drug therapy , Nervous System Diseases/drug therapy , Nootropic Agents/therapeutic use , Steroids/therapeutic use , Animals , Humans , Ion Channels/drug effects , Nootropic Agents/adverse effects , Nootropic Agents/pharmacology , Steroids/adverse effects , Steroids/pharmacology
20.
IDrugs ; 3(9): 1053-63, 2000 Sep.
Article in English | MEDLINE | ID: mdl-16049865

ABSTRACT

Neuroactive steroids rapidly modulate gamma-aminobutyric acid (GABA) and glutamate receptors. GABA-enhancing steroids have potential clinical utility as anesthetics, hypnotics, anticonvulsants and anxiolytics. Furthermore, GABAergic neurosteroids may participate in regulating mood and the effects of alcohol on the nervous system, suggesting a potential role in major psychiatric disorders. Neuroactive steroids that alter the function of glutamate receptors could be useful for treating neurodegenerative disorders, and as cognitive enhancers. Recent progress in developing water-soluble steroids and steroids with enhanced oral efficacy foster optimism that certain neuroactive steroids will be developed for clinical use.

SELECTION OF CITATIONS
SEARCH DETAIL
...