Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 338(1): 100-13, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21487071

ABSTRACT

Sodium channel inhibition is a well precedented mechanism used to treat epilepsy and other hyperexcitability disorders. The established sodium channel blocker and broad-spectrum anticonvulsant lamotrigine is also effective in the treatment of bipolar disorder and has been evaluated in patients with schizophrenia. Double-blind placebo-controlled clinical trials found that the drug has potential to reduce cognitive symptoms of the disorder. However, because of compound-related side-effects and the need for dose titration, a conclusive evaluation of the drug's efficacy in patients with schizophrenia has not been possible. (5R)-5-(4-{[(2-Fluorophenyl)methyl]oxy}phenyl)-l-prolinamide (GSK2) and (2R,5R)-2-(4-{[(2-fluorophenyl)methyl]oxy}phenyl)-7-methyl-1,7-diazaspiro[4.4]nonan-6-one (GSK3) are two new structurally diverse sodium channel blockers with potent anticonvulsant activity. In this series of studies in the rat, we compared the efficacy of the two new molecules to prevent a cognitive deficit induced by the N-methyl-d-aspartic acid receptor antagonist phencyclidine (PCP) in the reversal-learning paradigm in the rat. We also explored the effects of the drugs to prevent brain activation and neurochemical effects of PCP. We found that, like lamotrigine, both GSK2 and GSK3 were able to prevent the deficit in reversal learning produced by PCP, thus confirming their potential in the treatment of cognitive symptoms of schizophrenia. However, higher doses than those required for anticonvulsant efficacy of the drugs were needed for activity in the reversal-learning model, suggesting a lower therapeutic window relative to mechanism-dependent central side effects for this indication.


Subject(s)
Cognition Disorders/chemically induced , Cognition Disorders/prevention & control , Phencyclidine/toxicity , Schizophrenia/chemically induced , Schizophrenia/prevention & control , Sodium Channel Blockers/therapeutic use , Animals , Female , Male , Rats , Rats, Sprague-Dawley , Reversal Learning/drug effects , Reversal Learning/physiology , Sodium Channel Blockers/pharmacology , Treatment Outcome
2.
J Neurochem ; 108(2): 384-96, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19012743

ABSTRACT

The 5-HT(3) receptor is a member of the 'Cys-loop' family of ligand-gated ion channels that mediate fast excitatory and inhibitory transmission in the nervous system. Current evidence points towards native 5-HT(3) receptors originating from homomeric assemblies of 5-HT(3A) or heteromeric assembly of 5-HT(3A) and 5-HT(3B). Novel genes encoding 5-HT(3C), 5-HT(3D), and 5-HT(3E) have recently been described but the functional importance of these proteins is unknown. In the present study, in silico analysis (confirmed by partial cloning) indicated that 5-HT(3C), 5-HT(3D), and 5-HT(3E) are not human-specific as previously reported: they are conserved in multiple mammalian species but are absent in rodents. Expression profiles of the novel human genes indicated high levels in the gastrointestinal tract but also in the brain, Dorsal Root Ganglion (DRG) and other tissues. Following the demonstration that these subunits are expressed at the cell membrane, the functional properties of the recombinant human subunits were investigated using patch clamp electrophysiology. 5-HT(3C), 5-HT(3D), and 5-HT(3E) were all non-functional when expressed alone. Co-transfection studies to determine potential novel heteromeric receptor interactions with 5-HT(3A) demonstrated that the expression or function of the receptor was modified by 5-HT(3C) and 5-HT(3E), but not 5-HT(3D). The lack of distinct effects on current rectification, kinetics or pharmacology of 5-HT(3A) receptors does not however provide unequivocal evidence to support a direct contribution of 5-HT(3C) or 5-HT(3E) to the lining of the ion channel pore of novel heteromeric receptors. The functional and pharmacological contributions of these novel subunits to human biology and diseases such as irritable bowel syndrome for which 5-HT(3) receptor antagonists have major clinical usage, therefore remains to be fully determined.


Subject(s)
Biological Evolution , Protein Subunits/physiology , Receptors, Serotonin, 5-HT3/chemistry , Receptors, Serotonin, 5-HT3/physiology , Animals , Cell Line, Transformed , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Electric Stimulation/methods , Ferrets , GABA Antagonists/pharmacology , Green Fluorescent Proteins/genetics , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques/methods , Picrotoxin/pharmacology , Rabbits , Serotonin/pharmacology , Transfection
3.
Synapse ; 59(5): 308-16, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16421904

ABSTRACT

Hyperpolarization-activated cyclic nucleotide gated (HCN) ion channels regulate membrane potential, neurotransmitter release, and patterning of synchronized neuronal activity. Currently, there is an intense debate as to whether or not these ion channels play a pro- or anticonvulsant role in vivo. To gain an insight into this question, we have examined how inhibitors of the response mediated by HCN channels (referred to as I(h)) affect epileptiform activity induced in adult hippocampal slices. The archetypal I(h) blocker ZD-7288 produced a concentration-dependent inhibition of both nonsynaptic- (low Ca(2+)/elevated K(+) aCSF) and synaptic- (low Mg(2+) aCSF, elevated K(+) aCSF or convulsant application (bicuculline or pentylenetetrazol)) based epileptiform activities. The IC(50) value for ZD-7288 induced inhibition of epileptiform activity was similar across all forms of epileptiform response and was below concentrations producing nonspecific inhibition of glutamatergic synaptic transmission. Furthermore, capsazepine, which exhibits similar potency to ZD-7288 at inhibiting I(h), failed to inhibit glutamatergic synaptic transmission per se but produced a significant inhibition of bicuculline-induced epileptiform activity. These data suggest that broad spectrum inhibition of I(h) reduces neuronal hyperexcitability in the hippocampus.


Subject(s)
Epilepsy/physiopathology , Hippocampus/physiopathology , Ion Channels/physiology , Neural Inhibition/physiology , 2-Amino-5-phosphonovalerate/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Action Potentials/radiation effects , Animals , Animals, Newborn , Bicuculline/pharmacology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drug Interactions , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , GABA Antagonists/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , In Vitro Techniques , Ion Channels/drug effects , Ion Channels/radiation effects , Male , Neural Inhibition/drug effects , Patch-Clamp Techniques/methods , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Time Factors
4.
Br J Pharmacol ; 143(3): 411-21, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15351778

ABSTRACT

The human hyperpolarization-activated cyclic nucleotide-gated 1 (hHCN1) subunit was heterologously expressed in mammalian cell lines (CV-1 and CHO) and its properties investigated using whole-cell patch-clamp recordings. Activation of this recombinant channel, by membrane hyperpolarization, generated a slowly activating, noninactivating inward current. The pharmacological properties of hHCN1-mediated currents resembled those of native hyperpolarization-activated currents (I(h)), that is, blockade by Cs(+) (99% at 5 mm), ZD 7288 (98% at 100 microm) and zatebradine (92% at 10 microm). Inhibition of the hHCN1-mediated current by ZD 7288 was apparently independent of prior channel activation (i.e. non-use-dependent), whereas that induced by zatebradine was use-dependent. The VR1 receptor antagonist capsazepine inhibited hHCN1-mediated currents in a concentration-dependent (IC(50)=8 microm), reversible and apparently non-use-dependent manner. This inhibitory effect of capsazepine was voltage-independent and associated with a leftward shift in the hHCN1 activation curve as well as a dramatic slowing of the kinetics of current activation. Elevation of intracellular cAMP or extracellular K(+) significantly enhanced aspects of hHCN1 currents. However, these manipulations did not significantly affect the capsazepine-induced inhibition of hHCN1. The development of structural analogues of capsazepine may yield compounds that could selectively inhibit HCN channels and prove useful for the treatment of neurological disorders where a role for HCN channels has been described.


Subject(s)
Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Ion Channels/physiology , Animals , Benzazepines/pharmacology , CHO Cells , Cell Line , Cricetinae , Cricetulus , Cyclic AMP/pharmacology , Cyclic Nucleotide-Gated Cation Channels , Dose-Response Relationship, Drug , Electric Stimulation , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Membrane Potentials/drug effects , Patch-Clamp Techniques , Potassium/pharmacology , Potassium Channels , Pyrimidines/pharmacology , Time Factors , Transfection
5.
Neuropharmacology ; 46(8): 1168-76, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15111023

ABSTRACT

Electrophysiological recordings from dorsal raphe nucleus (DRN) neurones in rat brain slices have revealed that the orexins can cause direct and reversible depolarisation of the postsynaptic membrane. Whilst it is known that the membrane depolarisation produced by orexin-A can dramatically increase the firing rate of DRN neurones, quantitative pharmacological analysis that determines the receptor subtype mediating the orexinergic response has not yet been performed. Here, we demonstrate that the rank order of potencies of orexin receptor agonists to excite serotonergic DRN neurones is orexin-A = orexin-B > SB-668875-DM. In contrast, the rank order of potency of these agonists to excite noradrenergic locus coreleus (LC) neurones is orexin-A > orexin-B > SB-668875-DM. We show further that the orexin receptor antagonist, SB-334867-A, inhibits the effects of orexin-A in the LC and DRN with pKB values of 6.93 and 5.84, respectively, values similar to those calculated for human OX1 (7.27) and OX2 (5.60) receptors expressed in CHO cells. These data suggest a differential role for OX1 and OX2 receptors in stimulating distinct populations of monoaminergic neurones in the rat CNS with OX2 receptors exhibiting a more pronounced functional significance in serotonergic neurones and OX1 in noradrenergic neurones.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Raphe Nuclei/physiology , Receptors, Neuropeptide/physiology , Urea/analogs & derivatives , Animals , Benzoxazoles/pharmacology , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Excitatory Postsynaptic Potentials/drug effects , Male , Naphthyridines , Orexin Receptors , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/antagonists & inhibitors , Urea/pharmacology
6.
J Neurosci ; 23(31): 10146-53, 2003 Nov 05.
Article in English | MEDLINE | ID: mdl-14602831

ABSTRACT

Cell death mechanisms frequently involve the influx of extracellular calcium through voltage- and ligand-gated ion channels, e.g., the NMDA receptor (Greene, 1999). The vanilloid receptor (VR1) is present in regions of the brain (Mezey et al., 2000) that are highly susceptible to neurodegenerative insults, suggesting that this ion channel might contribute to the cellular processes involved in neuronal death. We tested the effects of VR1 ligands in the oxygen glucose deprivation (OGD) model of cell death in organotypic hippocampal slice cultures. The VR1 agonist capsaicin at concentrations that are selective for VR1 did not affect cell viability per se or the extent of neurodegeneration induced by the OGD insult. In contrast, the VR1 antagonist capsazepine (0.1-10 microm) significantly reduced the amount of OGD-induced cell death. However, capsazepine was still neuroprotective in slices prepared from VR1 knock-out mice, which exhibited the same degree of neurodegeneration to that observed in slices prepared from wild-type mice, excluding the possibility that it afforded neuroprotection through inhibition of VR1. Instead, capsazepine inhibited the hyperpolarization-activated nonspecific cation channel generated current I(h) in a concentration range similar to that which was neuroprotective. Furthermore, the specific I(h) blocker ZD-7288 was also neuroprotective, mirroring the effects of capsazepine, in that it was effective at preventing cell death when applied either during or after the OGD insult. These results demonstrate that capsazepine affords neuroprotection through inhibition of I(h) rather than inhibition of VR1.


Subject(s)
Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cell Hypoxia/physiology , Glucose/metabolism , Ion Channels/antagonists & inhibitors , Nerve Tissue Proteins , Neurons/drug effects , Neurons/metabolism , Animals , Cell Death , Cells, Cultured , Cyclic Nucleotide-Gated Cation Channels , Cytoprotection/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Glucose/deficiency , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , In Vitro Techniques , Ion Channels/metabolism , Mice , Mice, Knockout , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques , Potassium Channels , Rats , Rats, Sprague-Dawley , Receptors, Drug/deficiency , Receptors, Drug/genetics
7.
Epilepsy Res ; 57(2-3): 121-36, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15013053

ABSTRACT

The recently developed GABAB1 receptor subunit knockout (GABAB1 -/-) mouse displays complete loss of GABAB receptor function and develops complex generalized epilepsies including absence type, audiogenic as well as spontaneous generalized seizures with electrographic spike-wave discharge signatures. To gain insight into the cellular mechanisms contributing to the generation and maintenance of this epileptic phenotype we have compared epileptiform activity induced in hippocampal slices obtained from GABAB1 -/- and wild type (GABAB1 +/+) littermates. Deletion of the GABAB1 receptor subunit had no effect on a range of passive membrane properties of CA3 pyramidale neurones, non-synaptic epileptiform field bursting and spreading depression recorded in 6mM K+/Ca2+-free medium, and inter-ictal synaptically-induced epileptiform activity induced by 100 microM 4-aminopyridine (4-AP). In contrast, synaptic epileptiform activity induced by 10 microM bicuculline, removal of extracellular Mg2+ or addition of 10 microM oxotremorine was enhanced in GABAB1 -/- slices. Acute blockade of GABAB receptors using a selective antagonist only partly mimicked these effects. It is suggested that the exaggerated in vitro epileptiform activity is caused by both acute and chronic consequences of the loss of GABAB receptor function in vivo. Specifically, enhancement of N-methyl-d-aspartate (NMDA) receptor triggered synaptic processes, arising from the loss of the GABAB receptor-mediated inhibitory postsynaptic potential (IPSP, together with a possible promotion of depolarising IPSPs due to the removal of GABAB autoreceptor function) is likely to underlie these effects.


Subject(s)
Epilepsy/metabolism , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Receptors, GABA-B/deficiency , Animals , Epilepsy/genetics , Excitatory Postsynaptic Potentials/drug effects , GABA-B Receptor Antagonists , Hippocampus/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Phosphinic Acids/pharmacology , Propanolamines/pharmacology , Receptors, GABA-B/genetics
8.
Neuropharmacology ; 42(1): 82-92, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11750918

ABSTRACT

In the CA3 region of rat hippocampal slices gamma-amino-butyric acid (GABA)(A/B) receptor antagonists induce low frequency bursting activity that was either inhibited (in 21% of slices) or increased by the selective 5-HT receptor agonists 5-carboxy-tryptamine (0.1-1 microM) and 8-hydroxydipropylaminotetralin (8-OH-DPAT). The selective 5-HT1A receptor antagonist N-(2-(4-(2-methoxyphenyl)-1-piperazinyl)ethyl)-N-(2-pyridinyl)cyclohexane carboxamide (WAY 100635) reversed the depression of bursting activity whereas the 5-HT7 receptor antagonist, (R)-3-(2-(2-(4-methylpiperidin-1-yl)-ethyl)pyrrolidine-1-sulfonyl)phenol (SB-269970; 1-10 microM), but not the 5-HT1A, 4 or 6 receptor antagonists WAY100635 (10 microM), SB-204070 (10 microM) and SB-271046 (10 microM), reversed the increase in bursting activity. The apparent -log10 K(D) value (8.4) for the effect of SB-269970 was consistent with a selective action at 5-HT7 receptors. Accompanying the 5-CT-induced increase in bursting frequency there was a shortening of the burst event waveform and a reduction in the after-hyperpolarization following each bursting event both of which were inhibited by SB-269970. These effects appeared to result predominantly from a direct 5-HT(7) receptor-mediated inhibition of a Ca2+ activated K+ channel.


Subject(s)
Hippocampus/drug effects , Nerve Net/drug effects , Receptors, Serotonin/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Dioxanes/pharmacology , Extracellular Space , Hippocampus/cytology , Male , Membrane Potentials/physiology , Microelectrodes , Piperazines/pharmacology , Piperidines/pharmacology , Potassium Channels, Calcium-Activated/drug effects , Potassium Channels, Calcium-Activated/physiology , Pyramidal Cells/drug effects , Pyridines/pharmacology , Rats , Receptors, Serotonin/drug effects , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...