Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 230
Filter
1.
Life Sci ; 101(1-2): 73-8, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24582592

ABSTRACT

AIMS: Danshensu [3-(3,4-dihydroxyphenyl)-lactic acid], a phenylpropanoid compound isolated from Prunella vulgaris var. lilacina, is a well-known antioxidant. Although its antioxidant activity and cardioprotective effect have been reported, the pharmacological properties of danshensu in the central nervous system remain unclear. We investigated whether danshensu exerts anxiolytic-like activity in mice. MAIN METHODS: We conducted monoamine oxidase A (MAO-A) inhibition assay on danshensu in vitro, and behavioral tests including the elevated plus-maze test (EPM), the hole-board test, the rotarod test and the open field test were employed. KEY FINDINGS: We found that danshensu significantly inhibited the activity of MAO-A in vitro. The administration of danshensu (3 or 10mg/kg) produced a significant anxiolytic-like effect in the EPM and hole-board test. In addition, no changes in the spontaneous locomotor activity and no myorelaxant effects were observed compared to the control group; these effects were confirmed with the open field test and the rotarod test. Moreover, the anxiolytic-like properties of danshensu were antagonized by a dopamine D1 receptor antagonist (SCH 23390) but not by a 5-HT1A receptor antagonist (WAY 100635) or an α1-adrenergic receptor antagonist (prazosin). SIGNIFICANCE: These results indicate that danshensu exerts its anxiolytic-like properties, in part, through dopaminergic neurotransmitter signaling.


Subject(s)
Anti-Anxiety Agents/pharmacology , Lactates/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Benzazepines/pharmacology , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Exploratory Behavior/drug effects , Lactates/antagonists & inhibitors , Male , Maze Learning/drug effects , Mice , Monoamine Oxidase Inhibitors/pharmacology , Motor Activity/drug effects , Muscle Relaxation/drug effects , Piperazines/pharmacology , Prazosin/pharmacology , Pyridines/pharmacology , Rotarod Performance Test , Serotonin Antagonists/pharmacology
2.
Neuropharmacology ; 71: 141-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23583928

ABSTRACT

The histamine H4 receptor (H4R) is expressed primarily on cells involved in inflammation and immune responses. Recently, it has been reported the functional expression of H4R within neurons of the central nervous system, but their role has been poorly understood. The present study aimed to elucidate the physiopathological role of cerebral H4R in animal models by the intracerebroventricular administration of the H4R agonist VUF 8430 (20-40 µg per mouse). Selectivity of results was confirmed by the prevention of the effects produced by the H4R antagonist JNJ 10191584 (3-9 mg/kg p.o.). Neuronal H4R activation induced acute thermal antinociception, indicating that neuronal histamine H4R might be involved in the production of antinociception in the absence of an inflammatory process. An anxiolytic-like effect of intensity comparable to that exerted by diazepam, used as reference drug, was produced in the light-dark box test. VUF 8430 reversed the scopolamine-induced amnesia in the passive avoidance test and showed anorexant activity in food deprived mice. Conversely, the H4R activation did not modify the immobility time in the tail suspension test. Rotarod performance test was employed to demonstrate that the effects observed following the administration of VUF 8430 and JNJ 10191584 were not due to impaired motor function of animals. Furthermore, both compounds did not alter spontaneous mobility and exploratory activity in the hole board test. These results show the antinociceptive, antiamnesic, anxiolytic and anorexant effects induced by neuronal H4R agonism, suggesting that H4 modulators may have broader utility further the control of inflammatory and immune processes.


Subject(s)
Analgesics/therapeutic use , Anti-Anxiety Agents/therapeutic use , Appetite Depressants/therapeutic use , Central Nervous System/drug effects , Histamine Agonists/therapeutic use , Nootropic Agents/therapeutic use , Receptors, G-Protein-Coupled/agonists , Analgesics/agonists , Analgesics/antagonists & inhibitors , Animals , Anti-Anxiety Agents/agonists , Anti-Anxiety Agents/antagonists & inhibitors , Antidepressive Agents/therapeutic use , Appetite Depressants/chemistry , Behavior, Animal/drug effects , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Central Nervous System/metabolism , Dose-Response Relationship, Drug , Guanidines/administration & dosage , Guanidines/pharmacology , Histamine Agonists/administration & dosage , Histamine Agonists/chemistry , Histamine Agonists/pharmacology , Histamine Antagonists/administration & dosage , Histamine Antagonists/pharmacology , Male , Mice , Motor Activity/drug effects , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Nootropic Agents/agonists , Nootropic Agents/antagonists & inhibitors , Random Allocation , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Receptors, Histamine H4 , Thiourea/administration & dosage , Thiourea/analogs & derivatives , Thiourea/pharmacology
3.
Behav Pharmacol ; 24(2): 87-94, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23399882

ABSTRACT

The present study investigated the effect of agmatine on acquisition and expression of ethanol conditioned place preference (CPP) and its modulation by imidazoline agents. Swiss albino mice were treated intraperitoneally with saline or agmatine (20-40 mg/kg) before injection of ethanol (1.25 mg/kg) during conditioning days or on a test day (20-120 mg/kg), to observe the effect on acquisition or expression of CPP, respectively. Agmatine inhibited the acquisition but not the expression of ethanol CPP. Furthermore, both the I1 receptor antagonist, efaroxan (9 mg/kg) and the I2 receptor antagonist, BU224 (5 mg/kg) attenuated the agmatine-induced inhibition of the ethanol CPP acquisition. In contrast, the I2 receptor agonist, 2-BFI (5 mg/kg) and I1 receptor agonist, moxonidine (0.4 mg/kg) alone, or a combination of their subeffective doses, significantly attenuated the effect of agmatine (20 mg/kg) on acquisition of ethanol CPP. Agmatine or imidazoline agents alone produced neither place preference nor aversion, and at the doses used in the present study did not affect locomotor activity. Thus, agmatine attenuates the acquisition of ethanol CPP at least in part by imidazoline (I1 or I2) receptors. In future studies, agmatine or agents acting at the imidazoline receptors could be explored for their therapeutic potential in ethanol dependence.


Subject(s)
Agmatine/therapeutic use , Alcoholism/prevention & control , Imidazoline Receptors/agonists , Molecular Targeted Therapy , Monoamine Oxidase/metabolism , Neurotransmitter Agents/therapeutic use , Agmatine/administration & dosage , Agmatine/adverse effects , Agmatine/antagonists & inhibitors , Alcohol Deterrents/administration & dosage , Alcohol Deterrents/adverse effects , Alcohol Deterrents/antagonists & inhibitors , Alcohol Deterrents/therapeutic use , Alcoholism/drug therapy , Alcoholism/metabolism , Alcoholism/physiopathology , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/adverse effects , Anti-Anxiety Agents/antagonists & inhibitors , Anti-Anxiety Agents/therapeutic use , Behavior, Addictive/etiology , Behavior, Addictive/prevention & control , Behavior, Animal/drug effects , Conditioning, Classical/drug effects , Dose-Response Relationship, Drug , Drug Therapy, Combination/adverse effects , Imidazoline Receptors/antagonists & inhibitors , Imidazoline Receptors/metabolism , Injections, Intraperitoneal , Male , Mice , Monoamine Oxidase/chemistry , Monoamine Oxidase Inhibitors/pharmacology , Motor Activity/drug effects , Neurotransmitter Agents/administration & dosage , Neurotransmitter Agents/adverse effects , Neurotransmitter Agents/antagonists & inhibitors
4.
J Pharmacol Sci ; 120(3): 213-27, 2012.
Article in English | MEDLINE | ID: mdl-23099320

ABSTRACT

Polyriboinosinic-polyribocytidilic acid (polyI:C) is a synthetic analog that elicits viral-like immune responses in mammals. We have recently found that polyI:C treatment in neonatal mice induced abnormalities of emotional, cognitive, and sensorimotor gating and dysfunction of glutamatergic neurotransmission in adulthood. In this study, we investigated the effect of the NMDA-receptor co-agonist D-serine on polyI:C-induced behavioral abnormalities in mice. Neonatal ICR mice were repeatedly injected with polyI:C for 5 days from postnatal day 2 to 6. At 10 weeks, sensorimotor gating function was analyzed in the prepulse inhibition (PPI) test. Emotional function was analyzed in open field and social interaction tests. Cognitive function was analyzed by novel object recognition tests. D-Serine dose-dependently improved polyI:C-induced impairment of emotional and cognitive behaviors whereas it had no effect on PPI deficit in adults. The ameliorating effects of D-serine were antagonized by pretreatment with an NMDA-receptor antagonist, MK-801. Although the mRNA level of D-amino acid oxidase (DAAO) was increased in the prefrontal cortex and hippocampus of neonatal polyI:C-treated mice in adulthood, no changes were observed in D-serine content and DAAO enzymatic activity. These results suggest that D-serine ameliorates emotional and cognitive impairments of the polyI:C-treated mice through potentiating NMDA receptor activity.


Subject(s)
Affective Symptoms/prevention & control , Central Nervous System Viral Diseases/physiopathology , Cerebral Cortex/drug effects , Cognition Disorders/prevention & control , Disease Models, Animal , Receptors, N-Methyl-D-Aspartate/agonists , Serine/analogs & derivatives , Affective Symptoms/etiology , Animals , Animals, Newborn , Anti-Anxiety Agents/antagonists & inhibitors , Anti-Anxiety Agents/metabolism , Anti-Anxiety Agents/therapeutic use , Behavior, Animal/drug effects , Central Nervous System Viral Diseases/immunology , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cognition Disorders/etiology , D-Amino-Acid Oxidase/genetics , D-Amino-Acid Oxidase/metabolism , Exploratory Behavior/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Male , Mice , Mice, Inbred ICR , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Nootropic Agents/antagonists & inhibitors , Nootropic Agents/metabolism , Nootropic Agents/therapeutic use , Random Allocation , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Serine/antagonists & inhibitors , Serine/metabolism , Serine/therapeutic use , Specific Pathogen-Free Organisms
5.
Prog Neuropsychopharmacol Biol Psychiatry ; 39(2): 318-25, 2012 Dec 03.
Article in English | MEDLINE | ID: mdl-22789661

ABSTRACT

Melatonin (MLT) is a neurohormone known to be involved in the regulation of anxiety. Most of the physiological actions of MLT in the brain are mediated by two high-affinity G-protein-coupled receptors, denoted MT(1) and MT(2). However, the particular role of these receptors in anxiety remains to be defined. Here we used a novel MT(2)-selective partial agonist, UCM765 to evaluate the involvement of MT(2) receptors in anxiety. Adult male rats were acutely injected with UCM765 (5-10-20mg/kg), MLT (20mg/kg) or diazepam (DZ, 1mg/kg). Anxiety-related behaviors were assessed in the elevated plus maze test (EPMT), novelty suppressed feeding test (NSFT) and open field test (OFT). UCM765 at the dose of 10mg/kg showed anxiolytic-like properties by increasing the time spent in the open arm of the EPMT, and by reducing the latency to eat in a novel environment in the NSFT. In the EPMT, animals treated with UCM765 (10mg/kg) or MLT (20mg/kg) spent more time in the open arms compared to vehicle-treated animals, but to a lesser extent compared to DZ (1mg/kg). In the NSFT, all treatments similarly decreased the latency to eat in a novel environment compared to vehicle. UCM765 and MLT did not affect the total time and the number of entries into the central area of the OFT, but unlike DZ, did not impair locomotion. The anxiolytic effects of UCM765 and MLT in the EPMT and the NSFT were blocked using a pre-treatment with the MT(1)/MT(2) antagonist luzindole (10mg/kg) or the MT(2) antagonist 4P-PDOT (10mg/kg). These results demonstrated, for the first time, the anxiolytic properties of UCM765 and suggest that MT(2)-receptors may be considered a novel target for the development of anxiolytic drugs.


Subject(s)
Acetamides/pharmacology , Aniline Compounds/pharmacology , Diazepam/pharmacology , Melatonin/pharmacology , Receptor, Melatonin, MT2/agonists , Acetamides/antagonists & inhibitors , Aniline Compounds/antagonists & inhibitors , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Drug Partial Agonism , Feeding Behavior/drug effects , Male , Maze Learning/drug effects , Melatonin/antagonists & inhibitors , Motor Activity , Rats , Rats, Sprague-Dawley , Receptor, Melatonin, MT2/antagonists & inhibitors , Tetrahydronaphthalenes/pharmacology , Tryptamines/pharmacology
6.
Behav Brain Res ; 225(2): 547-53, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21839779

ABSTRACT

Several lines of evidence support the involvement of serotonergic (5-HT) neurons of the median raphe nucleus (MRN) in anxiety-like behaviour. In this context, it is known that blockade of 5-HT(1A) somatodendritic autoreceptors in the midbrain raphe nuclei increases the firing rate of these neurons, disinhibiting 5-HT release in postsynaptic target areas such as amygdala, hippocampus and periaqueductal grey matter (PAG). However, while activation of 5-HT(1A) or 5-HT(2) receptors in forebrain targets such as the amygdala or hippocampus enhances anxiety-like behaviours in rodents, stimulation of both receptor subtypes in the midbrain PAG markedly reduces anxiety-like behaviour. In view of these findings, the present study investigated whether the anti-anxiety effects induced by pharmacological disinhibition of 5-HT neurons in the MRN are attenuated by the blockade of 5-HT(2) receptors within the PAG. Mice received combined intra-PAG injection with ketanserin (10 nmol/0.1 µl), a 5-HT(2) receptor antagonist, followed by intra-MRN injection of WAY-100635 (5.6 nmol/0.1 µl), a highly selective 5-HT(1A) receptor antagonist. They were then individually exposed to the elevated plus-maze (EPM), with the videotaped behavioural sessions subsequently scored for both conventional and ethological measures. The results confirmed that intra-MRN infusion of WAY100635 reduces behavioural indices of anxiety without significantly altering general activity measures, and further showed that this effect was completely blocked by intra-PAG pretreatment with an intrinsically-inactive dose of ketanserin. Together, these results suggest that 5HT(2) receptor populations located within the midbrain PAG play a significant role in the reduction of anxiety observed following disinhibition of 5-HT neurons in the MRN.


Subject(s)
Anti-Anxiety Agents/pharmacology , Ketanserin/pharmacology , Periaqueductal Gray/drug effects , Piperazines/pharmacology , Pyridines/pharmacology , Raphe Nuclei/drug effects , Serotonin Antagonists/pharmacology , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Behavior, Animal/drug effects , Ketanserin/administration & dosage , Male , Maze Learning/drug effects , Mice , Microinjections/psychology , Piperazines/administration & dosage , Pyridines/administration & dosage , Serotonin Antagonists/administration & dosage
7.
Neurosci Lett ; 502(3): 147-51, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21763401

ABSTRACT

The neuropeptide galanin has been recognized as a possible neurotransmitter/neuromodulator, and in addition has been implicated in anxiety- and depression-related behaviors. The present study demonstrates increased locomotion and rearing after galanin (0.3mg/kg) that was given intraperitoneally (i.p.) to intact Wistar rats which were tested 1h later in the open field (OF). These effects, which suggest an anxiolytic-like action, were blocked by i.p. administered peptidic galanin antagonist M40. Further, the locomotion increase caused by galanin and the inhibitory effect of M40 persisted for 48h without additional treatment. Rats exposed to restraint stress (lasting 60min) for three consecutive days and tested 1h after stress termination exhibited reduced locomotion and exploration in the OF. Galanin (0.3 and 1.0mg/kg) given immediately after each stress exposure prevented the decrease of locomotion and exploration induced by stress in all trials. When the test was repeated 6 days later without stress and galanin treatment the reduction of locomotion produced by stress persisted; the anti-stress behavioral effects of both galanin doses were also present. Testing performed on the 12th day after the last stress and galanin treatment with 0.3mg/kg revealed an increased locomotion compared with unstressed and stress-exposed rats. Our results demonstrate that behavioral effects of the peptide galanin are evident even after i.p. administration. These results also suggest that galanin elicits stress-modulatory action, and support the notion that the galaninergic system may serve as a drug target in stress-related conditions.


Subject(s)
Anxiety Disorders/prevention & control , Behavior, Animal/physiology , Galanin/physiology , Stress, Psychological/prevention & control , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Anti-Anxiety Agents/pharmacology , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Behavior, Animal/drug effects , Disease Models, Animal , Drug Delivery Systems , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Galanin/antagonists & inhibitors , Galanin/metabolism , Male , Motor Activity/drug effects , Motor Activity/physiology , Rats , Rats, Wistar , Restraint, Physical/adverse effects , Restraint, Physical/psychology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Time , Time Factors
9.
J Psychopharmacol ; 24(5): 757-65, 2010 May.
Article in English | MEDLINE | ID: mdl-19825899

ABSTRACT

Previous studies in our laboratory showed that cannabinoid CB1 receptor knockout mice (CB1-/-) presented increased anxiety-like behaviours that did not respond to the anxiolytic actions of benzodiazepines. These results suggest that the pharmacological effects of benzodiazepines may involve the participation of cannabinoid CB1 receptors. Therefore, the purpose of this study was to examine the effects of alprazolam and the cannabinoid CB1 receptor antagonist AM251 on behavioural assays (light-dark box test, neurological severity score and step-down inhibitory avoidance test) and on the functional activity of the CB1 receptor (WIN-55,212-stimulated [(35)S] guanosine triphosphate (GTP) gamma binding autoradiography).The administration of alprazolam (40 microg/kg, intraperitoneal (i.p.)) decreased anxiety-like behaviours in the light-dark box test and significantly reduced WIN-55,212-stimulated [(35)S]GTPgamma binding autoradiography in the amygdala and in the CA1 field of the hippocampus, but was without effects on CA2, CA3 and the dentate gyrus (DG) of the hippocampus. The administration of AM251 (3 mg/kg, i.p.) blocked the anxiolytic action of alprazolam (40 microg/kg, i.p.), significantly reduced the sedative (ataxia, neurological severity score in the 0.5 cm bar) and the amnesic actions (short time term memory (1 h after electric shock)) of alprazolam (0.5 mg/kg, i.p.).Taken together, these findings revealed that cannabinoid CB1 receptor plays a pivotal role in the pharmacological actions of benzodiazepines. Furthermore, these results suggest that blockade of cannabinoid CB1 receptors may be useful in the treatment of patients with problems related to the consumption of benzodiazepines. Further clinical trials are needed to test this hypothesis.


Subject(s)
Amnesia/chemically induced , Anti-Anxiety Agents/pharmacology , Benzodiazepines/pharmacology , Hypnotics and Sedatives/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Alprazolam/antagonists & inhibitors , Alprazolam/pharmacology , Amygdala/cytology , Amygdala/drug effects , Amygdala/metabolism , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Anxiety/drug therapy , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Benzodiazepines/antagonists & inhibitors , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Hypnotics and Sedatives/antagonists & inhibitors , Male , Mice , Mice, Inbred ICR , Nervous System Diseases , Organ Specificity , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Severity of Illness Index
11.
Pharmacol Biochem Behav ; 89(2): 171-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18201755

ABSTRACT

AC-5216, a ligand for the translocator protein (18 kDa) (TSPO), previously called the peripheral benzodiazepine receptor (PBR), produces anxiolytic-like effects mediated by TSPO in animal models of anxiety. Since stimulation of TSPO is considered to promote the synthesis of neurosteroids, we investigated the possible role of endogenous neurosteroids that positively act on the GABA(A) receptor in the anxiolytic-like effects of AC-5216. In our experiments, the effects of trilostane and finasteride, two inhibitors of steroidogenic enzymes, and picrotoxin, a GABA(A) receptor-gated Cl(-) channel blocker, on the anxiolytic-like effects of AC-5216 were examined in the social interaction test in mice. Also, the anxiolytic-like effects of allopregnanolone and progesterone were examined. The anxiolytic-like effects of AC-5216 (0.1 mg/kg, p.o.) were inhibited by trilostane (10-30 mg/kg, s.c.), finasteride (10-30 mg/kg, s.c.), and picrotoxin (0.03-0.3 mg/kg, s.c.), while those of diazepam (0.1 mg/kg, p.o.) were inhibited by picrotoxin only. The anxiolytic-like effects of progesterone (1-3 mg/kg, s.c.) were inhibited by finasteride (3-30 mg/kg) and picrotoxin (0.1-0.3 mg/kg), although those of allopregnanolone (10 mg/kg, s.c.) were inhibited by picrotoxin only. These results demonstrate that the anxiolytic-like effects of AC-5216 are due to newly synthesized neurosteroids that enhance GABA(A) receptor function.


Subject(s)
Anti-Anxiety Agents/pharmacology , Interpersonal Relations , Pregnanolone/pharmacology , Progesterone/pharmacology , Purines/pharmacology , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Diazepam/pharmacology , Dihydrotestosterone/analogs & derivatives , Dihydrotestosterone/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Finasteride/pharmacology , GABA Antagonists/pharmacology , Male , Mice , Picrotoxin/pharmacology , Purines/antagonists & inhibitors , Receptors, GABA-A/drug effects
12.
Behav Pharmacol ; 18(1): 81-7, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17218801

ABSTRACT

Food-deprived Wistar rats were exposed to a fixed-time 60-s food delivery schedule until they developed schedule-induced polydipsia. Every fifth lick was then followed by an electric shock during two, signalled, 5-min periods, which ran concurrently with the food delivery schedule. Shock intensities were adjusted to reduce licking to 60-70% of the unpunished licking rates. The benzodiazepine full agonists, diazepam (0.3-3.0 mg/kg), chlordiazepoxide (0.3-10.0 mg/kg), oxazepam (0.3-3.0 mg/kg) and the benzodiazepine partial agonist, RU-32698 (3.0-17.0 mg/kg), led to increases in punished responding at intermediate doses and decreases at the highest doses tested. All benzodiazepine agonists brought about dose-dependent decreases in unpunished schedule-induced polydipsia, with doses required to reduce drinking proving higher than doses required to increase punished schedule-induced polydipsia. The antipunishment effect of 0.3 mg/kg of diazepam was dose-dependently antagonized by flumazenil and the benzodiazepine inverse agonist, RU-34000. Flumazenil effects, however, could reflect actions of flumazenil as a partial inverse agonist at GABAA receptors. RU-32698 at 10.0 mg/kg further facilitated the rate-increasing effect of 0.3 mg/kg of diazepam, but at 17.0 mg/kg partially blocked such antipunishment effect. Overall, the present results extend the similarities of the effects of benzodiazepine compounds on adjunctive and operant patterns of behaviour by showing similar interactions within the benzodiazepine receptor complex.


Subject(s)
Anti-Anxiety Agents/pharmacology , Benzodiazepines/pharmacology , Drinking/drug effects , Punishment , Reinforcement Schedule , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Benzodiazepines/antagonists & inhibitors , Chlordiazepoxide/pharmacology , Diazepam/pharmacology , Dose-Response Relationship, Drug , Electroshock , Fear/drug effects , Flumazenil/pharmacology , Food Deprivation , GABA Modulators/pharmacology , Imidazoles/pharmacology , Male , Oxazepam/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Wistar , Receptors, GABA-A/drug effects
13.
Eksp Klin Farmakol ; 69(3): 7-9, 2006.
Article in Russian | MEDLINE | ID: mdl-16878489

ABSTRACT

It is established that bicuculline, picrotoxin, and flumazenil (agents blocking different sites of GABA receptor) decrease the anxiolytic effect of piracetam as manifested in the conflict situation test. The most pronounced interaction was observed between piracetam and flumazenyl. On the background of antagonist action, piracetam inhibited the effects of flumazenil (but not those of bicuculline and picrotoxin). Based on these data, it is assumed that the anxiolytic effect of piracetam is mediated to some extent by benzodiazepine site of the GABA-benzodiazepine receptor complex.


Subject(s)
Anti-Anxiety Agents/pharmacology , Flumazenil/pharmacology , GABA Modulators/pharmacology , GABA-A Receptor Antagonists , Nootropic Agents/pharmacology , Piracetam/pharmacology , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Drug Antagonism , Flumazenil/antagonists & inhibitors , GABA Modulators/antagonists & inhibitors , Male , Nootropic Agents/antagonists & inhibitors , Rats
14.
Behav Pharmacol ; 17(1): 9-18, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16377959

ABSTRACT

The activity of the novel antidepressant agomelatine was evaluated in three models of anxiety and compared with that of melatonin and two anxiolytics, diazepam and buspirone. All drugs were tested 2 h before and 2 h after the dark phase of the diurnal cycle. Morning and evening agomelatine (10-75 mg/kg) administration increased animals' responses in the elevated plus maze and Vogel tests. Melatonin (10-75 mg/kg) enhanced open arms exploration in the evening experiment and was inactive in the Vogel test. In the conditioned ultrasonic vocalization test, agomelatine, but not melatonin, was active in the morning and evening experiment. Melatonin antagonist, S22153 (20 mg/kg), enhanced the action of morning and evening agomelatine administration in the Vogel and conditioned ultrasonic vocalization tests, while in the elevated plus maze test, S22153 inhibited effects of evening but not morning melatonin and agomelatine administration. These results indicate the involvement of both the melatonin and the 5-HT2C receptors in the mechanism of anxiolytic-like action of agomelatine.


Subject(s)
Acetamides/pharmacology , Anti-Anxiety Agents/pharmacology , Arousal/drug effects , Fear/drug effects , Maze Learning/drug effects , Melatonin/pharmacology , Acetamides/antagonists & inhibitors , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Brain/drug effects , Buspirone/pharmacology , Chronotherapy , Conditioning, Classical/drug effects , Diazepam/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Electroshock , Exploratory Behavior/drug effects , Male , Rats , Receptor, Serotonin, 5-HT2C/drug effects , Receptors, Melatonin/drug effects , Thiophenes/pharmacology , Vocalization, Animal/drug effects
15.
Behav Brain Res ; 164(1): 17-28, 2005 Oct 14.
Article in English | MEDLINE | ID: mdl-16081166

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) and serotonin/noradrenaline reuptake inhibitors (SNRIs) has been reported to be efficient in anxiety disorders. Some animal models have demonstrated an anxiolytic-like effect following acute administration, however, it is not yet known how noradrenergic receptors are implicated in the therapeutic effects of antidepressants (ADs) in anxiety. The effects of two alpha(2)-adrenoceptor agonists (clonidine, guanabenz) on anxiolytic-like effect of two SSRIs (paroxetine and citalopram) and two SNRIs (venlafaxine and milnacipran) were evaluated in the four-plate test (FPT) in mice. Paroxetine (4 mg/kg), citalopram (8 mg/kg), venlafaxine (8 mg/kg), and milnacipran (8 mg/kg) administered intraperitoneally (i.p.) increased the number of punishments accepted by mice in the FPT. Clonidine (0.0039-0.5 mg/kg) and guanabenz (0.03-0.5mg/kg) had no effect on the number of punishments accepted by mice. Clonidine (0.03 and 0.06 mg/kg) and guanabenz (0.125 and 0.5 mg/kg) (i.p. -45 min) reversed the anti-punishment effect of paroxetine, citalopram, venlafaxine and milnacipran (i.p. -30 min). But if the antidepressants are administered 45 min before the test and alpha(2)-adrenoceptor agonists 30 min before the test, alpha(2)-adrenoceptor agonists failed to alter the anti-punishment effect of antidepressants. The results of this present study indicate that alpha(2)-adrenoceptor agonists antagonise the anxiolytic-like effect of antidepressants in mice when they are administered 15 min before the administration of antidepressant suggesting a close inter-regulation between noradrenergic and serotoninergic system in the mechanism of SSRIs and SNRIs in anxiety-like behaviour.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Adrenergic alpha-Agonists/pharmacology , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety/drug therapy , Selective Serotonin Reuptake Inhibitors/pharmacology , Adrenergic Uptake Inhibitors/antagonists & inhibitors , Animals , Anti-Anxiety Agents/antagonists & inhibitors , Citalopram/antagonists & inhibitors , Citalopram/pharmacology , Clonidine/pharmacology , Cyclohexanols/antagonists & inhibitors , Cyclohexanols/pharmacology , Cyclopropanes/antagonists & inhibitors , Cyclopropanes/pharmacology , Dose-Response Relationship, Drug , Drug Antagonism , Guanabenz/pharmacology , Male , Mice , Milnacipran , Motor Activity/drug effects , Paroxetine/antagonists & inhibitors , Paroxetine/pharmacology , Punishment , Selective Serotonin Reuptake Inhibitors/antagonists & inhibitors , Statistics, Nonparametric , Venlafaxine Hydrochloride
16.
J Clin Psychopharmacol ; 23(4): 349-57, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12920410

ABSTRACT

The selective serotonin reuptake inhibitor antidepressant fluoxetine inhibits alprazolam metabolism in vivo by inhibition of the cytochrome P450 3A4 enzyme. Citalopram is a selective serotonin reuptake inhibitor antidepressant that has not yet been fully evaluated with respect to its potential for cytochrome P450 3A4-mediated drug interactions in vivo. Building on the existing in vitro and in vivo evidence that suggest a minimal effect of citalopram on cytochrome P450 3A4, we hypothesized that therapeutic doses of citalopram (20 mg/d), as compared with fluoxetine (20 mg/d), would cause less impairment in the metabolism of the probe drug alprazolam (1 mg) through inhibition of the cytochrome P450 3A4 isozyme as measured by pharmacokinetic and pharmacodynamic parameters in vivo. We found that fluoxetine prolonged the half-life of alprazolam by 16% and increased the area under the curve 0-infinity of alprazolam by 32%, while citalopram did not affect these parameters, although the time of maximum concentration of alprazolam was prolonged by 30 minutes after citalopram administration. Neither selective serotonin reuptake inhibitor significantly affected the pharmacodynamic profile of alprazolam. This experiment suggests differential effects by citalopram and fluoxetine on alprazolam kinetics.


Subject(s)
Alprazolam/antagonists & inhibitors , Alprazolam/pharmacokinetics , Anti-Anxiety Agents/antagonists & inhibitors , Anti-Anxiety Agents/pharmacokinetics , Antidepressive Agents/adverse effects , Citalopram/adverse effects , Fluoxetine/adverse effects , Area Under Curve , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , Double-Blind Method , Drug Interactions , Half-Life , Humans
17.
Pharmacology ; 68(4): 183-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12837972

ABSTRACT

This study was performed to investigate whether nitric oxide (NO) precursor (L-arginine), NO donor (S-nitroso-N-acetylpenicillamine, SNAP) and NO synthase inhibitors [N(G)-nitro-L-arginine-methylester (L-NAME) and N(G)-nitro-L-arginine (L-NOARG)] modulate morphine-induced anxiolytic effects in the plus-maze. L-Arginine (100, 200 and 300 mg kg(-1), i.p.) and SNAP (4, 8 and 10 mg kg(-1), i.p.) reduced the anxiolytic effect of morphine (20 mg kg(-1), s.c.). L-NAME (10, 20 and 40 mg/kg, i.p.) and L-NOARG (10, 15 and 20 mg kg(-1), i.p.) enhanced the anxiolytic effects of morphine (20 mg kg(-1), s.c.). On the other hand, L-arginine and SNAP increased the morphine-induced locomotor activity. L-NAME decreased the morphine-induced locomotor activity, but L-NOARG did not modify the morphine-induced locomotor activity. Therefore, these results suggest that the anxiolytic effects of morphine can be modulated by NO systems.


Subject(s)
Anti-Anxiety Agents/pharmacology , Morphine/pharmacology , Nitric Oxide/metabolism , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/antagonists & inhibitors , Arginine/pharmacology , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Male , Maze Learning/drug effects , Mice , Mice, Inbred ICR , Morphine/administration & dosage , Morphine/antagonists & inhibitors , Motor Activity/drug effects , Motor Activity/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroarginine/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , S-Nitroso-N-Acetylpenicillamine/pharmacology
18.
Biochem Pharmacol ; 66(1): 125-32, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12818372

ABSTRACT

As part of an effort to identify naturally occurring GABA(A) receptor benzodiazepine binding site (BDS) ligands from traditional medicinal herbs, we previously reported that flavonoid derivatives isolated from Scutellaria baicalensis (S. baicalensis) Georgi exhibited significant affinities for the BDS. The present study describes the characterization of 5,7-dihydroxy-6-methoxyflavone (oroxylin A), one of the major components of the herbal extract. Oroxylin A inhibited [3H]flunitrazepam binding to rat cerebral cortical membrane with a IC(50) value of 1.09+/-0.07 microM. A GABA ratio of 1.09+/-0.04 suggests that oroxylin A interacts as an antagonist at the recognition site. In neuropharmacological studies, oral administration of oroxylin A (3.75-60 mg kg(-1)) did not result in significant changes in animal models routinely employed for benzodiazepine (BD) evaluation. However, oroxylin A selectively abolished the anxiolytic, myorelaxant and motor incoordination, but not the sedative and anticonvulsant effects elicited by diazepam, a BDS agonist. These results add oroxylin A to the list of CNS active flavonoids, and as the first naturally occurring member endowed with selective antagonistic actions via the BDS.


Subject(s)
Anti-Anxiety Agents/antagonists & inhibitors , Benzodiazepines/antagonists & inhibitors , Flavonoids/pharmacology , Psychomotor Performance/drug effects , Scutellaria baicalensis/chemistry , Animals , Drugs, Chinese Herbal , Flavonoids/isolation & purification , Flavonoids/therapeutic use , Ligands , Male , Maze Learning/drug effects , Mice , Mice, Inbred ICR , Motor Activity/drug effects , Picrotoxin , Plants, Medicinal/chemistry , Radioligand Assay , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/prevention & control
19.
Clin Pharmacol Ther ; 72(6): 711-7, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12496752

ABSTRACT

OBJECTIVES: Intravenous midazolam is used as an in vivo biomarker of hepatic cytochrome P450 (CYP) 3A activity. Midazolam is a central nervous system depressant and can produce cognitive impairment. The purpose of this study was 2-fold: (1) to determine whether administration of intravenous flumazenil given before intravenous midazolam minimizes cognitive impairment and (2) to determine whether flumazenil pretreatment has an effect on midazolam pharmacokinetics during hepatic CYP3A phenotyping. METHODS: Eleven healthy subjects (8 men) received intravenous flumazenil (0.005 mg/kg) or placebo followed 7 minutes later by intravenous midazolam (0.025 mg/kg) in a randomized, double-blind crossover study. Plasma midazolam concentrations were obtained before dosing and at 5, 30, 60, 120, 240, 300, and 360 minutes after dosing and were assayed by liquid chromatography-tandem mass spectrometry. Midazolam pharmacokinetics were determined by noncompartmental methods. The two 1-sided tests procedure was used to compare area under the curve (AUC) between study phases. Data were log-transformed before analysis, and bioequivalence criteria were applied. Digit symbol substitution tests, performed before dosing and at 5, 30, 60, 120, 240, 300, and 360 minutes after dosing, were used to measure cognition. General linear modeling was used to compare scores between study phases. RESULTS: Midazolam AUC extrapolated to infinity [AUC(0-infinity)] between phases was bioequivalent. The AUC ratio (flumazenil plus midazolam/midazolam) was 0.99, with a 90% confidence interval of 0.98 to 1.00. Statistically significant differences(P

Subject(s)
Anti-Anxiety Agents/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/drug effects , Cognition/drug effects , Flumazenil/pharmacology , Liver/metabolism , Midazolam/pharmacokinetics , Oxidoreductases, N-Demethylating/drug effects , Adult , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/antagonists & inhibitors , Anti-Anxiety Agents/blood , Area Under Curve , Chromatography, Liquid , Cross-Over Studies , Cytochrome P-450 CYP3A , Double-Blind Method , Drug Interactions , Flumazenil/administration & dosage , Humans , Infusions, Intravenous , Liver/drug effects , Male , Mass Spectrometry , Midazolam/administration & dosage , Midazolam/blood , Middle Aged , Phenotype , Reference Values , Time Factors
20.
J Wildl Dis ; 38(2): 468-72, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12038151

ABSTRACT

This study evaluated partial antagonism of tiletamine-zolazepam (TZ) anesthesia in cheetahs (Acinonyx jubatus) and differences between two benzodiazepine antagonists, flumazenil and sarmazenil, in this species. Four cheetahs were anesthetized three times at an interval of 14 days with an average intramuscular dose of 4.2 mg/kg TZ. In trials 2 and 3 flumazenil at 0.031 mg/kg and sarmazenil at 0.1 mg/kg, respectively, were applied intramuscularly 30 min after initial TZ injection. There was a highly significant difference between the duration of TZ anesthesia with and without antagonist. Use of the antagonists significantly shortened duration and recovery and eliminated excitatory behavior during the recovery phase. No significant differences could be determined between the two antagonists. We recommend the use of sarmazenil and flumazenil to antagonize TZ anesthesia in cheetahs.


Subject(s)
Acinonyx/physiology , Anesthetics, Combined/antagonists & inhibitors , Anesthetics, Dissociative/antagonists & inhibitors , Anti-Anxiety Agents/antagonists & inhibitors , Tiletamine/antagonists & inhibitors , Zolazepam/antagonists & inhibitors , Anesthetics, Combined/administration & dosage , Anesthetics, Dissociative/administration & dosage , Animals , Anti-Anxiety Agents/administration & dosage , Anticonvulsants/administration & dosage , Anticonvulsants/antagonists & inhibitors , Benzodiazepines/administration & dosage , Benzodiazepines/pharmacology , Female , Flumazenil/administration & dosage , Flumazenil/pharmacology , Imidazoles/administration & dosage , Imidazoles/pharmacology , Injections, Intramuscular/veterinary , Male , Tiletamine/administration & dosage , Time Factors , Zolazepam/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...