Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 93(3): 370-9, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25542996

ABSTRACT

GABAA receptor positive allosteric modulators (PAMs) mediate robust analgesia in animal models of pathological pain. Restoration of diminished spinal GABAA-α2 and -α3 subunit-containing receptor function is a principal contributor to this analgesia, albeit involvement of GABAA-α5-receptors has not been excluded. Thus, we compared NS11394 and TPA023 (PAMs with selectivity/efficacy at GABAA-α2/α3/α5 receptors) with TP003 (a reportedly GABAA-α3 selective PAM) against spinal sensitization. However, in-house electrophysiology studies designed to confirm the selectivity of TPA023 and TP003 for human GABAA receptors did not corroborate published data, with TP003 displaying considerable GABAA-α5 receptor efficacy. Therefore, we identified a novel PAM, NS16085, which possesses negligible efficacy at GABAA-α5 receptors, but with GABAA-α2/α3 efficacy equivalent to NS11394. At the GABAA-α1 receptor the compound gives low level of negative modulation further separating it from the other compounds. Rat pups with carrageenan-induced hindpaw inflammatory hyperalgesia were used to make ex vivo spinal dorsal root-evoked ventral root recordings. Some spontaneous activity and large numbers of spikes to repetitive stimulation of dorsal roots at C-fibre intensity, indicative of wind-up and sensitization were observed. Equimolar concentrations of NS11394, TP003 and NS16085 all attenuated wind-up to a similar degree; TPA023 was clearly less effective. In adult rats, NS16085 (3-30 mg/kg, p.o.) dose-dependently reduced formalin-induced hindpaw flinching with efficacy comparable to NS11394. Thus, potentiation of GABAA-α2 and-α3 receptors is sufficient to depress spinal sensitization and mediate analgesia after inflammatory injury. Positive modulation at GABAA-α5-receptors is apparently dispensable for this process, an important consideration given the role of this receptor subtype in cognitive function.


Subject(s)
Benzimidazoles/chemistry , Benzimidazoles/pharmacology , GABA Modulators/chemistry , GABA Modulators/pharmacology , Pain Measurement/drug effects , Pyridines/chemistry , Pyridines/pharmacology , Receptors, GABA-A/physiology , Spinal Nerve Roots/drug effects , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Animals, Newborn , Benzimidazoles/therapeutic use , Dose-Response Relationship, Drug , Female , GABA Modulators/therapeutic use , Humans , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Inflammation/drug therapy , Inflammation/pathology , Male , Organ Culture Techniques , Pain/drug therapy , Pain/pathology , Pain Measurement/methods , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Rats, Wistar , Spinal Nerve Roots/pathology , Spinal Nerve Roots/physiology , Xenopus laevis
2.
Br J Pharmacol ; 168(5): 1118-32, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23061935

ABSTRACT

BACKGROUND AND PURPOSE: Most GABA(A) receptor subtypes comprise 2α, 2ß and 1γ subunit, although for some isoforms, a δ replaces a γ-subunit. Extrasynaptic δ-GABA(A) receptors are important therapeutic targets, but there are few suitable pharmacological tools. We profiled DS2, the purported positive allosteric modulator (PAM) of δ-GABA(A) receptors to better understand subtype selectivity, mechanism/site of action and activity at native δ-GABA(A) receptors. EXPERIMENTAL APPROACH: Subunit specificity of DS2 was determined using electrophysiological recordings of Xenopus laevis oocytes expressing human recombinant GABA(A) receptor isoforms. Effects of DS2 on GABA concentration-response curves were assessed to define mechanisms of action. Radioligand binding and electrophysiology utilising mutant receptors and pharmacology were used to define site of action. Using brain-slice electrophysiology, we assessed the influence of DS2 on thalamic inhibition in wild-type and δ(0/0) mice. KEY RESULTS: Actions of DS2 were primarily determined by the δ-subunit but were additionally influenced by the α, but not the ß, subunit (α4/6ßxδ > α1ßxδ >> γ2-GABA(A) receptors > α4ß3). For δ-GABA(A) receptors, DS2 enhanced maximum responses to GABA, with minimal influence on GABA potency. (iii) DS2 did not act via the orthosteric, or known modulatory sites on GABA(A) receptors. (iv) DS2 enhanced tonic currents of thalamocortical neurones from wild-type but not δ(0/0) mice. CONCLUSIONS AND IMPLICATIONS: DS2 is the first PAM selective for α4/6ßxδ receptors, providing a novel tool to investigate extrasynaptic δ-GABA(A) receptors. The effects of DS2 are mediated by an unknown site leading to GABA(A) receptor isoform selectivity.


Subject(s)
Protein Subunits/physiology , Pyridines/pharmacology , Receptors, GABA-A/physiology , Allosteric Regulation , Animals , Brain/drug effects , Brain/physiology , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oocytes/drug effects , Oocytes/physiology , Rats , Recombinant Proteins , Xenopus laevis
3.
Neuroscience ; 163(1): 73-81, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19482064

ABSTRACT

Small-conductance calcium-activated K(+) channels 1-3 (SK1-3) are important for neuronal firing regulation and are considered putative CNS drug targets. For instance non-selective SK blockers improve performance in animal models of cognition. The SK subtype(s) involved herein awaits identification and the question is difficult to address pharmacologically due to the lack of subtype-selective SK-channel modulators. In this study, we used doxycycline-induced conditional SK3-deficient (T/T) mice to address the cognitive consequences of selective SK3 deficiency. In T/T mice SK3 protein is near-eliminated from the brain following doxycycline treatment. We tested T/T and wild type (WT) littermate mice in five distinct learning and memory paradigms. In Y-maze spontaneous alternations and five-trial inhibitory avoidance the performance of T/T mice was markedly inferior to WT mice. In contrast, T/T and WT mice performed equally well in passive avoidance, object recognition and the Morris water maze. Thus, some aspects of working/short-term memory are disrupted in T/T mice. Using in situ hybridization, we further found the cognitive deficits in T/T mice to be paralleled by reduced brain-derived neurotrophic factor (BDNF) mRNA expression in the dentate gyrus and CA3 of the hippocampus. BDNF mRNA levels in the frontal cortex were not affected. BDNF has been crucially implicated in many cognitive processes. Hence, the biological substrate for the cognitive impairments in T/T mice could conceivably entail reduced trophic support of the hippocampus.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cognition Disorders/genetics , Cognition Disorders/metabolism , Hippocampus/metabolism , RNA, Messenger/metabolism , Small-Conductance Calcium-Activated Potassium Channels/genetics , Animals , Anti-Bacterial Agents/pharmacology , Cell Survival/genetics , Cognition Disorders/physiopathology , Cytoprotection/genetics , Dentate Gyrus/metabolism , Dentate Gyrus/physiopathology , Disease Models, Animal , Down-Regulation/genetics , Doxycycline/pharmacology , Gene Expression Regulation/physiology , Hippocampus/physiopathology , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/physiopathology , Memory, Short-Term/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
J Pharmacol Exp Ther ; 327(3): 969-81, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18791060

ABSTRACT

Spinal administration of GABA(A) receptor modulators, such as the benzodiazepine drug diazepam, partially alleviates neuropathic hypersensitivity that manifests as spontaneous pain, allodynia, and hyperalgesia. However, benzodiazepines are hindered by sedative impairments and other side effect issues occurring mainly as a consequence of binding to GABA(A) receptors containing the alpha(1) subunit. Here, we report on the novel subtype-selective GABA(A) receptor-positive modulator NS11394 [3'-[5-(1-hydroxy-1-methyl-ethyl)-benzoimidazol-1-yl]-biphenyl-2-carbonitrile], which possesses a functional efficacy selectivity profile of alpha(5) > alpha(3) > alpha(2) > alpha(1) at GABA(A) alpha subunit-containing receptors. Oral administration of NS11394 (1-30 mg/kg) to rats attenuated spontaneous nociceptive behaviors in response to hindpaw injection of formalin and capsaicin, effects that were blocked by the benzodiazepine site antagonist flumazenil. Ongoing inflammatory nociception, observed as hindpaw weight-bearing deficits after Freund's adjuvant injection, was also completely reversed by NS11394. Likewise, hindpaw mechanical allodynia was fully reversed by NS11394 in two rat models of peripheral neuropathic pain. Importantly, NS11394-mediated antinociception occurred at doses 20 to 40-fold lower than those inducing minor sedative or ataxic impairments. In contrast, putative antinociception associated with administration of either diazepam, zolpidem, or gaboxadol only occurred at doses producing intolerable side effects, whereas bretazenil was completely inactive despite minor influences on motoric function. In electrophysiological studies, NS11394 selectively attenuated spinal nociceptive reflexes and C-fiber-mediated wind-up in vitro pointing to involvement of a spinal site of action. The robust therapeutic window seen with NS11394 in animals suggests that compounds with this in vitro selectivity profile could have potential benefit in clinical treatment of pain in humans.


Subject(s)
Benzimidazoles/pharmacology , GABA Modulators/pharmacology , Inflammation/drug therapy , Neuralgia/drug therapy , Receptors, GABA-A/drug effects , Allosteric Regulation , Animals , Benzodiazepinones/pharmacology , Diazepam/pharmacology , Humans , Isoxazoles/pharmacology , Pyridines/pharmacology , Rats , Zolpidem
5.
J Pharmacol Exp Ther ; 327(3): 954-68, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18791063

ABSTRACT

The novel positive allosteric modulator NS11394 [3'-[5-(1-hydroxy-1-methyl-ethyl)-benzoimidazol-1-yl]-biphenyl-2-carbonitrile] possesses a functional selectivity profile at GABA(A) receptors of alpha(5) > alpha(3) > alpha(2) > alpha(1) based on oocyte electrophysiology with human GABA(A) receptors. Compared with other subtype-selective ligands, NS11394 is unique in having superior efficacy at GABA(A)-alpha(3) receptors while maintaining low efficacy at GABA(A)-alpha(1) receptors. NS11394 has an excellent pharmacokinetic profile, which correlates with pharmacodynamic endpoints (CNS receptor occupancy), yielding a high level of confidence in deriving in vivo conclusions anchored to an in vitro selectivity profile and allowing for translation to higher species. Specifically, we show that NS11394 is potent and highly effective in rodent anxiety models. The anxiolytic efficacy of NS11394 is most probably mediated through its high efficacy at GABA(A)-alpha(3) receptors, although a contributory role of GABA(A)-alpha(2) receptors cannot be excluded. Compared with benzodiazepines, NS11394 has a significantly reduced side effect profile in rat (sedation, ataxia, and ethanol interaction) and mouse (sedation), even at full CNS receptor occupancy. We attribute this benign side effect profile to very low efficacy of NS11394 at GABA(A)-alpha(1) receptors and an overall partial agonist profile across receptor subtypes. However, NS11394 impairs memory in both rats and mice, which is possibly attributable to its efficacy at GABA(A)-alpha(5) receptors, albeit activity at this receptor might be relevant to its antinociceptive effects (J Pharmacol Exp Ther 327:doi;10.1124/jpet.108.144, 2008). In conclusion, NS11394 has a unique subtype-selective GABA(A) receptor profile and represents an excellent pharmacological tool to further our understanding on the relative contributions of GABA(A) receptor subtypes in various therapeutic areas.


Subject(s)
Allosteric Regulation , Anti-Anxiety Agents/pharmacology , Benzimidazoles/pharmacology , Receptors, GABA-A/drug effects , Animals , Anti-Anxiety Agents/adverse effects , Anti-Anxiety Agents/pharmacokinetics , Anxiety/drug therapy , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , GABA-A Receptor Agonists , Humans , Ligands , Memory/drug effects , Mice , Pharmacokinetics , Rats
6.
Genes Brain Behav ; 7(8): 836-48, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18616612

ABSTRACT

SK3 K(+) channels influence neuronal excitability and are present in 5-hydroxytryptamine (5-HT) and dopamine (DA) nuclei in the brain stem. We therefore hypothesized that SK3 channels affect 5-HT and DA neurotransmission and associated behaviors. To explore this, we used doxycycline-induced conditional SK3-deficient (T/T) mice. In microdialysis, T/T mice had elevated baseline levels of striatal extracellular DA and the metabolites dihydroxyphenylacetic acid and homovanillic acid. While baseline hippocampal extracellular 5-HT was unchanged in T/T mice, the 5-HT response to the 5-HT transporter inhibitor citalopram was enhanced. Furthermore, baseline levels of the 5-HT metabolite 5-hydroxyindoleacetic acid were elevated in T/T mice. T/T mice performed equally to wild type (WT) in most sensory and motor tests, indicating that SK3 deficiency does not lead to gross impairments. In the forced swim and tail suspension tests, the T/T mice displayed reduced immobility compared with WT, indicative of an antidepressant-like phenotype. Female T/T mice were more anxious in the zero maze. In contrast, anxiety-like behaviors in the open-field and four-plate tests were unchanged in T/T mice of both sexes. Home cage diurnal activity was also unchanged in T/T mice. However, SK3 deficiency had a complex effect on activity responses to novelty: T/T mice showed decreased, increased or unchanged activity responses to novelty, depending on sex and context. In summary, we report that SK3 deficiency leads to enhanced DA and 5-HT neurotransmission accompanied by distinct alterations in emotional behaviors.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , Dopamine/metabolism , Emotions/physiology , Serotonin/metabolism , Small-Conductance Calcium-Activated Potassium Channels/genetics , Animals , Anti-Bacterial Agents/pharmacology , Anxiety Disorders/genetics , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Citalopram/pharmacology , Doxycycline/pharmacology , Exploratory Behavior/physiology , Female , Hydroxyindoleacetic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurocognitive Disorders/genetics , Neurocognitive Disorders/metabolism , Neurocognitive Disorders/physiopathology , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Sex Characteristics , Synaptic Transmission/genetics
7.
Pharmacol Biochem Behav ; 90(1): 19-36, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18321566

ABSTRACT

The promise of subtype-selective GABA(A) receptor drugs with anxiolytic properties but with a much reduced side-effect burden (compared to benzodiazepines) is an attainable goal. However, its achievement necessitates the availability of in vivo preclinical assays capable of demonstrating differences as well as similarities between subtype-selective agents and non-selective benzodiazepines. In this study, we have compared three mouse strains (NMRI, C57BL/6J and DBA/2) in four models of anxiety-like behaviour (plus-maze, zero-maze, light-dark, and Vogel conflict). Furthermore, in each model, we have contrasted in detail the behavioural responses of each strain to the non-selective benzodiazepine chlordiazepoxide (CDP; 5-20 mg/kg), and the subtype-selective agents L-838,417 (GABA(A)-alpha(2/3/5); 3-30 mg/kg) and zolpidem (GABA(A)-alpha1; 0.3-3.0 mg/kg). The data show a complex mouse strainxmodelxpharmacological agent interaction. Most importantly, not all mouse strainxmodel test systems showed a positive response to CDP or predicted the response to L-838,417. This dissociation between CDP and L-838,417 opens up opportunities for preclinical test systems that differentiate subtype-selective and non-selective GABA(A) receptor agents, an attribute that might well be important in providing the necessary confidence for further drug development. Present findings suggest the need for a much greater focus on defining test systems appropriate for screening novel chemical entities, rather than self-selection of models or genotypes based on responses to known pharmacological agents. For example, if current data with L-838,417 are confirmed with compounds showing similar selectivity profiles, such agents may in future be best identified and characterised using test systems comprising NMRI mice in the zero-maze and/or C57 mice in the Vogel conflict and/or light-dark tests.


Subject(s)
Anxiety/psychology , Behavior, Animal/drug effects , Chlordiazepoxide/pharmacology , Fluorobenzenes/pharmacology , GABA Modulators/pharmacology , Hypnotics and Sedatives/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , Animals , Conflict, Psychological , Darkness , Light , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Models, Psychological , Species Specificity , Zolpidem
8.
Prog Neuropsychopharmacol Biol Psychiatry ; 31(4): 858-66, 2007 May 09.
Article in English | MEDLINE | ID: mdl-17335951

ABSTRACT

BACKGROUND: Chronic treatment with the dual serotonin/noradrenaline reuptake inhibitor (SNRI) duloxetine reduces the density of serotonin transporter sites in cortex and engenders an anxiolytic-like response. To determine the reproducibility of these effects and their generality to other antidepressants we compared the effects of chronic duloxetine treatment with another SNRI, venlafaxine, and two selective serotonin reuptake inhibitors, paroxetine and fluoxetine. METHODS: Separate groups of mice were administered vehicle, fluoxetine (15 mg/kg), paroxetine, duloxetine or venlafaxine (10 mg/kg) perorally twice daily for 28 days and tested in the mouse zero-maze and in motility cages on days 21 and 22, respectively, to determine effects on anxiety and motor activity. On day 28 brains were analysed for serotonin transporter (SERT) density in cortex and noradrenaline transporter (NET) density in cortex and hippocampus. RESULTS: Duloxetine and fluoxetine both reduced SERT density in cortex and induced anxiolytic-like effects. Paroxetine had an identical profile, but it is unclear if this drug down-regulated the SERT since extensive washing of cortical tissue did not remove all drug. Venlafaxine had no effect on behavioural or biochemical parameters. Only duloxetine reduced NET density in cortex, although not hippocampus. CONCLUSIONS: The reduction in SERT density and anxiolytic-like effects with duloxetine, fluoxetine and, potentially, paroxetine suggest that down-regulation of the SERT may be a relevant mechanism in therapeutic response to these antidepressants.


Subject(s)
Antidepressive Agents/administration & dosage , Anxiety Disorders/drug therapy , Anxiety Disorders/metabolism , Selective Serotonin Reuptake Inhibitors/administration & dosage , Serotonin Plasma Membrane Transport Proteins/metabolism , Analysis of Variance , Animals , Disease Models, Animal , Drug Administration Schedule , Female , Gene Expression Regulation/drug effects , Maze Learning/drug effects , Mice , Mice, Inbred Strains , Motor Activity/drug effects
9.
Behav Brain Res ; 167(1): 118-27, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16225935

ABSTRACT

Schizophrenic patients have deficits in prepulse inhibition (PPI) that may be alleviated by smoking/nicotine. The effect of nicotinic agents on PPI in rodents is equivocal and few studies in mice have been reported. Thus, we assessed nicotine's (0.03-1mg/kg) effect on PPI in five mouse strains with no effects. We next determined if nicotine would reverse a phencyclidine (PCP)-induced deficit of PPI in BALB/cByJ and NMRI mice. BALB/cByJ mice have a low density of [(125)I]alpha-bungaratoxin binding in the hippocampus and poor inhibitory gating of auditory evoked potentials (AEPs), a model related to PPI. At 1mg/kg, nicotine selectively reversed the PCP-induced deficit of PPI in BALB/cByJ mice. The pharmacokinetic profile of nicotine (T(1/2), C(max), T(max) and AUC) was identical in both strains, obviating this as a factor for the strain-dependent effect observed. Moreover, 1mg/kg nicotine inhibited in vivo [(3)H]epibatidine binding with the same time-course in both strains, indicating no difference in brain "kinetics". Since high doses of nicotine were effective in BALB/cByJ mice a role for low-affinity nicotinic receptors, e.g. alpha(7) receptors, is plausible. Clozapine, but not risperidone, also only reversed the PCP deficit of PPI in BALB/cByJ. Clozapine and nicotine also enhance inhibitory gating of AEPs in DBA/2 mice, and clozapine's effect is antagonized by an alpha(7) antagonist. Our data and previous evidence possibly suggest a role for low-affinity nicotinic receptors in the effects of clozapine and nicotine. Furthermore, BALB/cByJ mice may represent a model to test the effects of nicotinic agents acting at low-affinity nicotinic receptors.


Subject(s)
Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Neural Inhibition/drug effects , Nicotine/pharmacology , Phencyclidine/pharmacology , Risperidone/pharmacology , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Female , Hallucinogens/pharmacology , Mice , Mice, Inbred BALB C , Nicotinic Agonists/pharmacology , Reflex, Startle/drug effects , Reflex, Startle/physiology , Species Specificity
10.
J Pharmacol Exp Ther ; 316(3): 1378-85, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16352707

ABSTRACT

Recent evidence suggests that GABA(A) receptors containing an alpha1 subunit mediate the sedative effect of diazepam, whereas receptors with an alpha2 subunit mediate this benzodiazepine's anxiolytic effect. Thus, compounds selective for GABA(A)-alpha2 receptors may offer advantages, i.e., lack of sedation, over current benzodiazepines. Whether such compounds would offer additional advantages over benzodiazepines is unclear. Here, we address the issue of physical dependence by comparing the GABA(A)-alpha1 affinity-selective drug zolpidem, the novel compounds 7-(1,1-dimethylethyl)-6-(2-methyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2,5-difluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine (L-838,417) and 6-fluoro-9-methyl-2-phenyl-4-(pyrrolidin-1-yl-carbonyl)-2,9-dihydro-1H-pyridol[3,4-b]indol-1-one (SL651498) with functional selectivity for certain non-alpha(1) GABA(A) receptors, nonselective partial agonists [bretazenil, 1-[1-[3-(3-pyridyl)phenyl]benzimidazol-5-yl]ethanone O-ethyloxime (NS2710), and 5-furan-3-yl-1-(3-imidazol-1-phenyl)-1H-benzoimidazole (NS2664)], and nonselective full efficacy benzodiazepines, in a rapid precipitated withdrawal assay using the inverse agonist N-methyl-beta-carboline-3-carboxamide (FG-7142). For all compounds, we determined in vitro IC50 values to displace [3H]flunitrazepam from rat cortex and in vivo ED50 values for displacement of [3H]flunitrazepam from mouse forebrain (including length of in vivo occupancy). In the precipitated withdrawal model, compounds were administered at a dose giving approximately 80% receptor occupancy, obviating major differences in central nervous system bioavailability. Mice were administered compounds twice daily for 4 days and on day 5, 20 h after the final dose, given a dose of FG-7142 (40 mg/kg i.p.) that did not induce seizures in control animals. In mice treated with the three subtype-selective compounds, FG-7142 did not induce seizures. Moreover, there was a low propensity for FG-7142 to induce seizures in animals treated with the partial agonists, whereas seizures were clearly seen in animals treated with most benzodiazepines. Nonetheless, differences among the benzodiazepines themselves, similarities between the partial agonists and subtype-selective compounds, the in vitro/in vivo potency, and in vivo receptor exposure time data suggest a complex interaction among selectivity, efficacy, potency, and receptor exposure in determining physical dependence liability of benzodiazepine site modulators in mice.


Subject(s)
GABA Modulators/pharmacology , Receptors, GABA-A/drug effects , Substance-Related Disorders/prevention & control , Animals , Benzimidazoles/pharmacology , Benzodiazepines/pharmacology , Carbolines/pharmacology , Female , Flunitrazepam/metabolism , GABA-A Receptor Agonists , Mice , Oximes/pharmacology , Receptors, GABA-A/chemistry , Receptors, GABA-A/classification
11.
J Pharmacol Exp Ther ; 316(3): 1291-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16339395

ABSTRACT

The zolpidem discriminative cue is mediated by GABA(A)-alpha1 receptors, whereas the chlordiazepoxide cue may be mediated via non-alpha1 GABA(A) receptors because compounds with selective affinity for GABA(A)-alpha1 receptors fully generalize to the former cue. We predicted that L-838,417 [7-tert-butyl-3-(2,5-difluorophenyl)-6-(2-methyl-2H-1,2,4-triazol-3-ylmethoxy)-1,2,4-triazolo[4,3-b]pyridazine], a partial agonist at non-alpha1 GABA(A) receptors and an antagonist at GABA(A)-alpha1 receptors, would generalize to the chlordiazepoxide but not the zolpidem-discriminative cue. SL651498 [6-fluoro-9-methyl-2-phenyl-4-(pyrrolidin-1-yl-carbonyl)-2,9-dihydro-1H-pyridol[3,4-b]indol-1-one] is a full agonist at GABA(A)-alpha2 receptors, with lower efficacy at GABA(A)-alpha3 receptors and least efficacy at GABA(A)-alpha1 and GABA(A)-alpha5 receptors. Because SL651498 has efficacy at GABA(A)-alpha1 receptors, we anticipated that it would generalize to both discriminative cues. Rats were trained to discriminate either zolpidem (3 mg/kg) or chlordiazepoxide (5 mg/kg) from vehicle using a two-lever operant procedure. The generalization profiles of L-838,417 and SL651498 were compared with nonselective full agonists, GABA(A)-alpha1-selective ligands zolpidem and CL218,872 [3-methyl-6-[3-(trifluoromethyl)phenyl]-1,2,4-triazolo[4,3-b]pyridazine], the nonselective partial agonist bretazenil, and the novel anxioselective drug ocinaplon. A nonselective partial agonist was included because L-838,417 and SL651498 are partial agonists at some GABA(A) receptors, and this property may influence their generalization profiles. All nonselective full agonists and ocinaplon fully generalized to both cues. CL218,872 and zolpidem generalized to zolpidem only, whereas L-838,417 fully generalized to chlordiazepoxide only. SL651498 fully generalized to chlordiazepoxide and occasioned significant zolpidem-appropriate responding. Bretazenil was similar to SL651498. In conclusion, at this training dose, the chlordiazepoxide-discriminative stimulus is mediated primarily via non-alpha1 GABA(A) receptors and the generalization profiles of the ligands tested seem to correspond with their in vitro profiles at GABA(A) receptor subtypes.


Subject(s)
Benzodiazepines/pharmacology , Chlordiazepoxide/pharmacology , Cues , Discrimination Learning/drug effects , GABA Modulators/pharmacology , Generalization, Psychological/drug effects , Pyridines/pharmacology , Receptors, GABA-A/drug effects , Animals , Azabicyclo Compounds , Benzodiazepinones/pharmacology , Fluorobenzenes/pharmacology , Male , Piperazines/pharmacology , Protein Subunits , Pyridazines/pharmacology , Rats , Receptors, GABA-A/chemistry , Triazoles/pharmacology , Zolpidem
12.
Psychopharmacology (Berl) ; 182(4): 475-84, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16133136

ABSTRACT

RATIONALE: GABAA receptors containing an alpha2 subunit are proposed to mediate the anxiolytic effect of benzodiazepines (BZ) based on studies in transgenic mice using unconditioned models of anxiety. Conditioned models of anxiety were not assessed and are rarely encountered in phenotyping of genetically modified animals. The novel benzodiazepine site ligand L838,417 is a partial agonist at GABAA receptors containing an alpha2, alpha3 or alpha5 subunit and an antagonist at alpha1 receptors, giving an anxiolytic profile devoid of sedation. However, this compound has not previously been assessed in mice. OBJECTIVES: (1) Establish the Vogel conflict test (VCT) in C57BL/6J mice and validate it with a range of pharmacological tools and (2) compare the full and partial GABAA receptor positive modulators chlordiazepoxide (CDP) and bretazenil (BRZ), respectively, with the subtype selective ligands zolpidem (ZOL; alpha1 selective) and L838,417. RESULTS: (1) enhanced thirst (water deprivation or isoproterenol administration), analgesia (lamotrigine) or cognitive impairment (MK-801) did not generate false positives in the VCT; (2) CDP and BRZ engendered linear dose-related anti-conflict effects and also increased unpunished drinking; (3) L838,417 engendered a bell-shaped anti-conflict effect and did not increase unpunished drinking; (4) the anti-conflict effect of CDP and L838,417 were antagonised by flumazenil, whereas BRZ's effect was insensitive to this antagonist; and (5) ZOL induced motoric deficits and no anti-conflict effect. CONCLUSION: We have established the VCT in C57BL/6J mice and validated this test behaviourally, physiologically and pharmacologically. The novel GABAA receptor ligand L838,417 was anxiolytic in this mouse model, and unlike the non-selective compounds, had no effect on unpunished drinking.


Subject(s)
Behavior, Animal/drug effects , Benzodiazepinones/pharmacology , Chlordiazepoxide/pharmacology , Conflict, Psychological , Fluorobenzenes/pharmacology , GABA Agents/pharmacology , Pyridines/pharmacology , Triazoles/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Drinking Behavior/drug effects , Drug Interactions , Electroshock/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Reaction Time/drug effects , Reproducibility of Results , Time Factors , Water Deprivation , Zolpidem
13.
Psychopharmacology (Berl) ; 181(4): 741-50, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16032412

ABSTRACT

RATIONALE: Monoamine transporter inhibitor antidepressants have anxiolytic efficacy in man. However, preclinical data poorly reflect this, either because (1) few studies assess chronic antidepressant treatment in animal models, (2) antidepressants are anxiogenic after acute treatment; and (3) animal models of anxiety are insensitive to antidepressants. OBJECTIVE: We address issues (1) and (2) and ascertain potential mechanisms mediating anxiolytic effects demonstrated. METHODS: The effect of acute treatment with seven antidepressants covering the classes selective serotonin reuptake inhibitors, serotonin-noradrenaline reuptake inhibitors, noradrenaline reuptake inhibitors and tricyclic antidepressants were compared with the benzodiazepine, chlordiazepoxide, on the mouse zero maze, an unconditioned model of anxiety. Furthermore, citalopram, duloxetine, reboxetine and amitriptyline were assessed after chronic administration (10 mg/kg p.o., 21 days, twice daily) in this model. In mice treated chronically, (a) the hypothermic response to serotonin (5-HT)1A and 5-HT1B receptor ligands, 8-hydroxy-2-(di-n-propylamino)tetralin (8-OHDPAT) and m-chlorophenyl piperazine (mCPP), respectively, was assessed and (b) serotonin transporter (SERT) and noradrenaline transporter (NET) densities in the cortex and hippocampus, respectively, were determined. RESULTS: None of the antidepressants were anxiolytic after acute treatment, although reboxetine, duloxetine and amitriptyline were anxiogenic. Only chronic treatment with duloxetine induced an anxiolytic effect, which was dissociable from nonspecific motor effects. Duloxetine reduced SERT density in the cortex by approximately 75% compared to control, with no effect on NET density in the hippocampus. Citalopram and amitriptyline significantly reduced SERT density by approximately 20%, whereas reboxetine selectively reduced NET density. All drugs reduced the hypothermic response to 8-OHDPAT and mCPP. CONCLUSION: Duloxetine was anxiolytic after chronic but not acute treatment, reflecting clinical experience with antidepressants in general. Duloxetine's anxiolytic-like profile may be ascribed to the considerable reduction in the density of the SERT in the cortex.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Arousal/drug effects , Brain/drug effects , Maze Learning/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/drug effects , Thiophenes/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Animals , Brain/pathology , Carrier Proteins/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Duloxetine Hydrochloride , Female , Hippocampus/drug effects , Hippocampus/pathology , Mice , Norepinephrine/metabolism , Receptor, Serotonin, 5-HT1A/drug effects , Receptor, Serotonin, 5-HT1B/drug effects
14.
CNS Drug Rev ; 11(1): 1-20, 2005.
Article in English | MEDLINE | ID: mdl-15867950

ABSTRACT

Retigabine [D23129; N-(2-amino-4-(4-fluorobenzylamino)-phenyl)carbamic acid ethyl ester] is an antiepileptic drug with a recently described novel mechanism of action that involves opening of neuronal K(V)7.2-7.5 (formerly KCNQ2-5) voltage-activated K(+) channels. These channels (primarily K(V)7.2/7.3) enable generation of the M-current, a subthreshold K(+) current that serves to stabilize the membrane potential and control neuronal excitability. In this regard, retigabine has been shown to have a broad-spectrum of activity in animal models of electrically-induced (amygdala-kindling, maximal electroshock) and chemically-induced (pentylenetetrazole, picrotoxin, NMDA) epileptic seizures. These encouraging results suggest that retigabine may also prove useful in the treatment of other diseases associated with neuronal hyperexcitability. Neuropathic pain conditions are characterized by pathological changes in sensory pathways, which favor action potential generation and enhanced pain transmission. Although sometimes difficult to treat with conventional analgesics, antiepileptics can relieve some symptoms of neuropathic pain. A number of recent studies have reported that retigabine can relieve pain-like behaviors (hyperalgesia and allodynia) in animal models of neuropathic pain. Neuronal activation within several key structures within the CNS can also be observed in various animal models of anxiety. Moreover, amygdala-kindled rats, which have a lowered threshold for neuronal activation, also display enhanced anxiety-like responses. Retigabine dose-dependently reduces unconditioned anxiety-like behaviors when assessed in the mouse marble burying test and zero maze. Early clinical studies have indicated that retigabine is rapidly absorbed and distributed, and is resistant to first pass metabolism. Tolerability is good in humans when titrated up to its therapeutic dose range (600-1200 mg/day). No tolerance, dependence or withdrawal potential has been reported, although adverse effects can include mild dizziness, headache, nausea and somnolence. Thus, retigabine may prove to be useful in the treatment of a diverse range of disease states in which neuronal hyperexcitability is a common underlying factor.


Subject(s)
Anticonvulsants/chemical synthesis , Anticonvulsants/therapeutic use , Carbamates/chemical synthesis , Carbamates/therapeutic use , Epilepsy/drug therapy , Phenylenediamines/chemical synthesis , Phenylenediamines/therapeutic use , Animals , Anticonvulsants/pharmacokinetics , Anxiety/drug therapy , Carbamates/pharmacokinetics , Dose-Response Relationship, Drug , Humans , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Neurons/drug effects , Neurons/physiology , Pain/drug therapy , Phenylenediamines/pharmacokinetics , Potassium Channels, Voltage-Gated/drug effects , Time Factors
15.
J Pharmacol Exp Ther ; 314(1): 282-92, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15814569

ABSTRACT

Neuronal Kv7 channels are recognized as potential drug targets for treating hyperexcitability disorders such as pain, epilepsy, and mania. Hyperactivity of the amygdala has been described in clinical and preclinical studies of anxiety, and therefore, neuronal Kv7 channels may be a relevant target for this indication. In patch-clamp electrophysiology on cell lines expressing Kv7 channel subtypes, Maxipost (BMS-204352) exerted positive modulation of all neuronal Kv7 channels, whereas its R-enantiomer was a negative modulator. By contrast, at the Kv7.1 and the large conductance Ca2+-activated potassium channels, the two enantiomers showed the same effect, namely, negative and positive modulation at the two channels, respectively. At GABA(A) receptors (alpha1beta2gamma2s and alpha2beta2gamma2s) expressed in Xenopus oocytes, BMS-204352 was a negative modulator, and the R-enantiomer was a positive modulator. The observation that the S- and R-forms exhibited opposing effects on neuronal Kv7 channel subtypes allowed us to assess the potential role of Kv7 channels in anxiety. In vivo, BMS-204352 (3-30 mg/kg) was anxiolytic in the mouse zero maze and marble burying models of anxiety, with the effect in the burying model antagonized by the R-enantiomer (3 mg/kg). Likewise, the positive Kv7 channel modulator retigabine was anxiolytic in both models, and its effect in the burying model was blocked by the Kv7 channel inhibitor 10,10-bis-pyridin-4-ylmethyl-10H-anthracen-9-one (XE-991) (1 mg/kg). Doses at which BMS-204352 and retigabine induce anxiolysis could be dissociated from effects on sedation or memory impairment. In conclusion, these in vitro and in vivo studies provide compelling evidence that neuronal Kv7 channels are a target for developing novel anxiolytics.


Subject(s)
Anti-Anxiety Agents/pharmacology , Carbamates/pharmacology , Indoles/pharmacology , Neurons/drug effects , Phenylenediamines/pharmacology , Potassium Channels, Voltage-Gated/agonists , Algorithms , Animals , Anti-Anxiety Agents/chemistry , Anxiety/physiopathology , Anxiety/psychology , Ataxia/chemically induced , Behavior, Animal/drug effects , Cell Line , Dose-Response Relationship, Drug , Emotions/drug effects , Exploratory Behavior/drug effects , Female , Hand Strength , Indoles/chemistry , KCNQ Potassium Channels , KCNQ1 Potassium Channel , Male , Membrane Potentials , Mice , Oocytes/drug effects , Patch-Clamp Techniques , RNA, Complementary/biosynthesis , Rats , Rats, Wistar , Receptors, GABA-A/drug effects , Receptors, GABA-A/genetics , Stereoisomerism , Xenopus laevis
16.
Psychopharmacology (Berl) ; 180(1): 159-68, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15682295

ABSTRACT

RATIONALE: Many anticonvulsants are used in disorders other than epilepsy. For example, lamotrigine is reported to be effective in post-traumatic stress disorder and mania. OBJECTIVE: We assessed the effects of the anticonvulsants lamotrigine, valproate and carbamazepine in an animal model of anxiety. We assessed a wide range of pharmacological tools to delineate the mechanism of lamotrigine's anxiolytic effect. METHODS: We assessed these compounds in the rat conditioned emotional response (CER) test of anxiety. RESULTS: Lamotrigine (30-80 mg/kg) dose-dependently and reproducibly engendered an anxiolytic response in this test, with similar efficacy to benzodiazepines. Carbamazepine (20-40 mg/kg) and riluzole (10 mg/kg), which block Na+ channels by a similar mechanism as lamotrigine, were also anxiolytic. By contrast, valproate (100-600 mg/kg) was inactive and appears to differ in its interaction with Na+ channels. The SSRI paroxetine, the GABA(A) receptor positive modulator propofol, the NMDA antagonists memantine and (+)MK-801, and the Ca2+ channel antagonist nifedipine were all inactive in the CER test, suggesting these mechanisms may not mediate the anxiolytic effect of lamotrigine. More directly, we showed that the anxiolytic effect of lamotrigine could be blocked by co-administering rats with the Na+ channel activator veratrine (0.1 mg/kg). By contrast, neither the Ca2+ channel agonist BAYK8644 (0.5 mg/kg) nor the 5-HT1A or 5-HT(1/2) antagonists WAY100635 (0.3 mg/kg) and metergoline (3 mg/kg), respectively, were able to block the effect. CONCLUSION: Lamotrigine's anxiolytic effect in the CER test may be mediated via block of Na+ channels, and this may represent a target for the development of novel anxiolytics.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anticonvulsants/pharmacology , Anxiety/drug therapy , Conditioning, Classical/drug effects , Emotions/drug effects , Triazines/pharmacology , Animals , Anti-Anxiety Agents/administration & dosage , Anticonvulsants/administration & dosage , Anxiety/psychology , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Lamotrigine , Male , Rats , Receptors, GABA-A/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Sodium Channel Blockers/pharmacology , Synaptic Transmission/drug effects , Triazines/administration & dosage
17.
J Pharmacol Exp Ther ; 299(2): 782-92, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602695

ABSTRACT

The muscarinic receptor agonist xanomeline was examined and compared with the antipsychotics clozapine and/or haloperidol in the following in vivo rat models: apomorphine-induced disruption of prepulse inhibition (PPI), amphetamine-induced hyperlocomotion, and the conditioned emotional response (CER) test. The effects of xanomeline were also assessed ex vivo on dopamine turnover in the rat medial prefrontal cortex. Under conditions of varying dose and prepulse intensity, xanomeline, like haloperidol, had no effect on PPI. In contrast, the muscarinic receptor antagonist scopolamine and the muscarinic receptor agonist pilocarpine both induced significant dose-dependent deficits in PPI. Haloperidol and xanomeline, but not pilocarpine, dose dependently reversed apomorphine-induced disruption of PPI. Thus, xanomeline induced a clear antipsychotic-like effect in PPI, whereas pilocarpine appeared to induce a psychotomimetic-like effect. Xanomeline attenuated amphetamine-induced hyperactivity at doses that had no effect on spontaneous activity, possibly indicating a separation between attenuation of limbic hyperdopaminergic function and the induction of hypolocomotion. Haloperidol and clozapine also reversed amphetamine-induced hyperlocomotion, but at similar doses to those that reduced spontaneous locomotion. Clozapine, but not haloperidol had an anxiolytic-like effect in the CER test. The effects of xanomeline in the CER test were similar to those of clozapine, although at the anxiolytic dose it tended to disrupt baseline levels of lever pressing. Finally, haloperidol, clozapine, pilocarpine, and xanomeline, all induced an increase in dopamine turnover in medial prefrontal cortex. The antipsychotic-like effects of xanomeline in the animal models used here suggest that it may be a useful treatment for psychosis.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Muscarinic Agonists/pharmacology , Pyridines/pharmacology , Thiadiazoles/pharmacology , Amphetamine , Animals , Central Nervous System Stimulants , Conditioning, Psychological/drug effects , Conflict, Psychological , Dopamine/metabolism , Emotions/drug effects , Hyperkinesis/chemically induced , Hyperkinesis/prevention & control , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects
18.
Psychopharmacology (Berl) ; 154(1): 8-12, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11292010

ABSTRACT

RATIONALE: Clinically, nicotine improves attention, but this has proven difficult to demonstrate preclinically. We tested nicotine in Sprague-Dawley (SD) and Lister hooded (LH) rats in the five-choice serial reaction time task. Since SD rats demonstrate lower asymptotic performance than LH rats, we surmised that nicotine would only improve performance in this strain. METHODS: Rats were placed in operant chambers 10 min after nicotine treatment (0.001-0.2 mg/kg). RESULTS: Nicotine dose-dependently increased correct responses only in SD rats (approximately 20% at the highest dose). By contrast, nicotine dose-dependently increased omission errors and reduced trials completed in both strains of rat, and dose-dependently reduced tray responses in SD rats. CONCLUSION: The magnitude of improvement in accuracy seen with nicotine in SD rats is greater than previously demonstrated using lesion or parametric manipulation models in other strains of rat in this test of attention. Although this suggests that the SD strain may be a useful "tool" for future studies, other task parameters, such as stimulus duration, may have to be optimum to demonstrate the magnitude of improvement observed presently.


Subject(s)
Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Reaction Time/drug effects , Animals , Dose-Response Relationship, Drug , Male , Rats , Rats, Sprague-Dawley , Species Specificity , Stimulation, Chemical
19.
Eur J Pharmacol ; 407(1-2): 73-81, 2000 Oct 27.
Article in English | MEDLINE | ID: mdl-11050293

ABSTRACT

Excessive tobacco consumption by schizophrenic patients may be a form of self-medication, and nicotine in tobacco may alleviate deficits in information processing. We tested this hypothesis by determining whether nicotine (acute/chronic) would improve information processing in the rat using pre-pulse inhibition as a model. In study 1, rats were injected with nicotine 10 min prior to placement in startle chambers (0.001-0.1 or 0.03-0.3 mg/kg, s.c.). In study 2, rats were injected with either saline or nicotine (0.4 mg/kg, s.c.) for 21 consecutive days and assessed for locomotor activity, pre-pulse inhibition and changes in [3H]nicotine binding in whole brain. Acutely, nicotine had no effect on pre-pulse inhibition. By contrast, after chronic nicotine treatment, rats demonstrated a robust deficit in pre-pulse inhibition and significant increases in locomotor activity and [3H]nicotine binding. The deficit in pre-pulse inhibition after chronic treatment with nicotine may be the result of non-specific behavioural activation due to increased mesolimbic dopamine release or, possibly, nicotine may rapidly desensitize nicotinic receptors important for normal information processing.


Subject(s)
Motor Activity/drug effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Reflex, Startle/drug effects , Animals , Brain/metabolism , Mental Processes/drug effects , Mental Processes/physiology , Motor Activity/physiology , Nicotine/pharmacokinetics , Nicotinic Agonists/pharmacokinetics , Rats , Reflex, Startle/physiology , Schizophrenia/drug therapy
20.
Eur J Pharmacol ; 393(1-3): 147-54, 2000 Mar 30.
Article in English | MEDLINE | ID: mdl-10771008

ABSTRACT

Studies in smokers have suggested that at least part of the improved psychomotor performance produced by nicotine is the result of an effect on attention. Many animal experiments have assessed the effects of nicotine and its antagonists on diverse types of learning and memory but relatively few have looked at it in tasks designed to assess attention. In a five-choice serial reaction time task (5-CSRTT), rats with restricted access to food were presented with an array of five holes; illumination of a randomly selected hole signalled that a nose-poke into it would be reinforced by food presentation. Initially, signal length and the inter-trial interval (ITI) were varied and the procedure was demonstrated to satisfy some criteria for a vigilance task. The effects of nicotine on deficits in performance induced by varying signal length and ITI were assessed. Under appropriate conditions, small doses of nicotine increased the percentage of correct responses (accuracy), decreased omission errors and reaction time, and increased anticipatory responses. Subsequently, the effects of varying the ITI were examined more extensively in a slightly modified task. Here, nicotine produced small but robust, highly significant dose-related increases in accuracy, as well as decreases in omission errors and reaction times. Nicotine also increased accuracy when light stimuli were presented in an unpredictable manner. The nicotine antagonist mecamylamine produced a modest deficit in reaction time only. It is concluded that appropriate doses of nicotine can produce robust improvements in performance of normal rats in an attentional task. The effect cannot be attributed easily to changes in sensory or motor capability, learning or memory and may provide the measures needed to investigate the neuropharmacological and neuroanatomical bases of the elusive attentional effect of nicotine.


Subject(s)
Attention/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Animals , Cognition/drug effects , Drug Interactions , Female , Male , Mecamylamine/pharmacology , Muscarinic Antagonists/pharmacology , Nicotinic Antagonists/pharmacology , Psychomotor Performance/drug effects , Rats , Reaction Time/drug effects , Scopolamine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...