Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Psychopharmacology (Berl) ; 231(7): 1289-303, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24186076

ABSTRACT

RATIONALE: As enhanced corticotropin-releasing factor (CRF) transmission is associated with induction of sensorimotor gating deficits, CRF1 receptor antagonists may reverse disrupted prepulse inhibition (PPI), an operational measure of sensorimotor gating. OBJECTIVES: To determine the effects of CRF1 receptor antagonists in pharmacological models of disrupted PPI and to determine if long-term elevated central CRF levels alter sensitivity towards PPI disrupting drugs. METHODS: CP154,526 (10-40 mg/kg), SSR125543 (3-30 mg/kg) and DMP695 (40 mg/kg) were tested on PPI disruption provoked by D-amphetamine (2.5, 3 mg/kg), ketamine (5, 30 mg/kg) and MK801 (0.2, 0.5 mg/kg) in Wistar rats, C57Bl/6J and CD1 mice, and on spontaneously low PPI in Iffa Credo rats and DBA/2J mice. PPI-disrupting effects of D-amphetamine (2.5-5 mg/kg) and MK801 (0.3-1 mg/kg) were examined in CRF-overexpressing (CRFtg) mice, which display PPI deficits. Finally, we determined the influence of CP154,526 on D-amphetamine-induced dopamine outflow in nucleus accumbens and prefrontal cortex of CRFtg mice using in vivo microdialysis. RESULTS: No CRF1-antagonists improved PPI deficits in any test. CRFtg mice showed blunted PPI disruption in response to MK801, but not D-amphetamine. Further, D-amphetamine-induced dopamine release was less pronounced in CRFtg versus wild-type mice, a response normalized by pretreatment with CP154,526. CONCLUSION: The inability of CRF1 receptor antagonists to block pharmacological disruption of sensorimotor gating suggests that the involvement of CRF1 receptors in the modulation of dopaminergic and glutamatergic neurotransmission relevant for sensory gating is limited. Furthermore, the alterations observed in CRFtg mice support the notion that long-term elevated central CRF levels induce changes in these neurotransmitter systems.


Subject(s)
Hydrocarbons, Halogenated/pharmacology , Prepulse Inhibition/drug effects , Pyridines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Thiazines/pharmacology , Acoustic Stimulation , Animals , Corticotropin-Releasing Hormone/genetics , Dextroamphetamine/antagonists & inhibitors , Dextroamphetamine/pharmacology , Dizocilpine Maleate/antagonists & inhibitors , Dizocilpine Maleate/pharmacology , Dopamine/metabolism , Dose-Response Relationship, Drug , Ketamine/antagonists & inhibitors , Ketamine/pharmacology , Male , Mice , Mice, Transgenic , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Reflex, Startle/drug effects
2.
Psychopharmacology (Berl) ; 228(1): 97-107, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23407783

ABSTRACT

RATIONALE: The selective CRF1 (corticotropin releasing factor type 1) receptor antagonist SSR125543 has been previously shown to attenuate the long-term cognitive deficit produced by traumatic stress exposure. Memory disturbances described in post-traumatic stress disorder (PTSD) patients are believed to be associated with changes in neuronal activity, in particular at the level of the hippocampus. OBJECTIVES: The present study aims at investigating whether the effects of SSR125543 (10 mg/kg/day for 2 weeks) on cognitive impairment induced by traumatic stress exposure are associated with changes in hippocampal excitability. Effects of SSR125543 were compared to those of the 5-HT reuptake inhibitor, paroxetine (10 mg/kg/day), and the partial N-methyl-D-aspartate (NMDA) receptor agonist, D-cycloserine (10 mg/kg/day), two compounds which have demonstrated clinical efficacy against PTSD. METHODS: Mice received two unavoidable electric foot-shocks. Then, 1 or 16 days after stress, they were tested for their memory performance using the object recognition test. Neuronal excitability was recorded during the third week post-stress in the CA1 area of the hippocampus. Drugs were administered from day 1 post-stress to the day preceding the electrophysiological study. RESULTS: Application of electric shocks produced cognitive impairment 16, but not 1 day after stress, an effect which was associated with a decrease in hippocampal neuronal excitability. Both stress-induced effects were prevented by repeated administration of SSR125543, paroxetine and D-cycloserine. CONCLUSIONS: These findings confirm that the CRF1 receptor antagonist SSR125543 is able to attenuate the behavioral effects of traumatic stress exposure and indicate that these effects are associated with a normalization of hippocampal neuronal excitability impaired by stress.


Subject(s)
Cognition Disorders/drug therapy , Hydrocarbons, Halogenated/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological/drug therapy , Thiazines/pharmacology , Animals , Behavior, Animal/drug effects , Cognition Disorders/etiology , Cycloserine/pharmacology , Electroshock , Hippocampus/drug effects , Hippocampus/pathology , Male , Mice , Paroxetine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/complications , Time Factors
3.
Pharmacol Biochem Behav ; 102(3): 415-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22659580

ABSTRACT

The selective antagonist at the CRF1 receptor, SSR125543, has been shown to produce anxiolytic-like effects in a number of animal models. The aim of the present study was to verify whether these effects are mediated by an action on the hypothalamic pituitary adrenal (HPA) axis. SSR125543 effects were evaluated in a mouse model of post-traumatic stress disorder. Animals received two unavoidable electric foot-shocks (1.5 mA/2 s). Two weeks later they were placed in the shock context and fecal and plasma corticosterone levels were measured by enzyme-immunoassay. Their cognitive performances were evaluated using the object recognition task following administration of SSR125543 at 3, 10 and 30 mg/kg or paroxetine at 20 mg/kg (i.p.), used as positive control. To assess the involvement of the HPA axis in the drug effects, a separate group of animals was subjected to the same procedure and drug regimen, but was treated with dexamethasone to blunt the HPA axis. Stressed mice had higher levels of corticosterone following re-exposure to the context and displayed impaired cognitive performance as compared to control animals. Corticosterone levels were normalized in stressed mice by SSR125543 and the cognitive deficit was significantly attenuated by SSR125543 and paroxetine, whether the HPA axis was blunted or not. These findings confirm that SSR125543 is able to attenuate the deleterious effects of stressful exposure. Importantly, the observation that these effects were still present in dexamethasone-treated mice indicates that this action does not necessarily involve pituitary-adrenal axis blockade, thereby suggesting that extra-pituitary CRF1 receptors may play a role in these effects.


Subject(s)
Cognition Disorders/psychology , Hydrocarbons, Halogenated/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Receptors, Corticotropin-Releasing Hormone/agonists , Stress, Psychological/psychology , Thiazines/pharmacology , Animals , Cognition Disorders/etiology , Corticosterone/analysis , Corticosterone/blood , Corticosterone/metabolism , Dexamethasone/pharmacology , Electroshock , Feces/chemistry , Male , Mice , Paroxetine/pharmacology , Recognition, Psychology/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/complications
4.
Mol Psychiatry ; 16(12): 1177-88, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21537331

ABSTRACT

Recent research suggests an involvement of hippocampal neurogenesis in behavioral effects of antidepressants. However, the precise mechanisms through which newborn granule neurons might influence the antidepressant response remain elusive. Here, we demonstrate that unpredictable chronic mild stress in mice not only reduces hippocampal neurogenesis, but also dampens the relationship between hippocampus and the main stress hormone system, the hypothalamo-pituitary-adrenal (HPA) axis. Moreover, this relationship is restored by treatment with the antidepressant fluoxetine, in a neurogenesis-dependent manner. Specifically, chronic stress severely impairs HPA axis activity, the ability of hippocampus to modulate downstream brain areas involved in the stress response, the sensitivity of the hippocampal granule cell network to novelty/glucocorticoid effects and the hippocampus-dependent negative feedback of the HPA axis. Remarkably, we revealed that, although ablation of hippocampal neurogenesis alone does not impair HPA axis activity, the ability of fluoxetine to restore hippocampal regulation of the HPA axis under chronic stress conditions, occurs only in the presence of an intact neurogenic niche. These findings provide a mechanistic framework for understanding how adult-generated new neurons influence the response to antidepressants. We suggest that newly generated neurons may facilitate stress integration and that, during chronic stress or depression, enhancing neurogenesis enables a dysfunctional hippocampus to restore the central control on stress response systems, then allowing recovery.


Subject(s)
Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Hippocampus/drug effects , Neurogenesis/drug effects , Stress, Psychological/drug therapy , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Cell Count/methods , Cell Count/statistics & numerical data , Corticosterone/blood , Corticosterone/metabolism , Depression/drug therapy , Depression/physiopathology , Dexamethasone , Disease Models, Animal , Hippocampus/diagnostic imaging , Hippocampus/physiopathology , Humans , Hydrocarbons, Halogenated/pharmacology , Hydrocarbons, Halogenated/therapeutic use , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiopathology , Male , Mice , Mice, Inbred BALB C , Neurogenesis/physiology , Pituitary-Adrenal Function Tests/methods , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiopathology , Radiography , Stress, Psychological/physiopathology , Thiazines/pharmacology , Thiazines/therapeutic use
5.
Behav Brain Res ; 221(1): 149-54, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21377492

ABSTRACT

The experience of traumatic stress often leads to long-lasting alteration in sleep quality and behavior. The objective of the present experiment was to investigate the short- and long-term effects of acute inescapable stress (i.e. two electric foot-shocks of 1.5 mA; 2s) on sleep/wakefulness parameters and behavior in Swiss mice using electroencephalographic (EEG) analysis. Baseline EEG recording was performed in the home cage for 6h prior to the application of the foot-shocks in the presence of an object (i.e. a plastic prism). One, 7, 14 or 21 days later, a second 6h EEG recording session was performed after mice had been exposed or not to the same object for 5 min in their home cage. Results showed that at day 1, 7, 14 and 21 post-stress, shocked mice displayed sleep fragmentation as shown by an increase in the number of sleep episodes, regardless the presence of the object or not. In animals exposed to the object, the total duration of wakefulness over 6h was significantly increased at days 7, 14 and 21 post-stress, and rapid eye movement (REM) sleep was significantly decreased at day 14 post-shock. Moreover, in the behavioral experiment, conditioned avoidance to a shock-paired object, which appeared as soon as 24h after shock application, turned into generalized avoidance towards an unknown object 21 days after stress. These findings demonstrate that an acute inescapable stress exposure may cause long-lasting alterations in sleep patterns and behavior. Such modifications may be reminiscent of the profound changes observed in patients suffering from post-traumatic stress disorder.


Subject(s)
Avoidance Learning/physiology , Disease Models, Animal , Sleep Wake Disorders/physiopathology , Sleep, REM/physiology , Stress Disorders, Post-Traumatic/physiopathology , Stress, Psychological/physiopathology , Animals , Cues , Electroencephalography/methods , Electroshock/adverse effects , Male , Mice , Wakefulness/physiology
6.
Psychopharmacology (Berl) ; 194(2): 161-71, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17557151

ABSTRACT

RATIONALE: The endocannabinoid system plays an important role in conditioned drug seeking, but the neuronal mechanisms involved in this behavior are unclear. OBJECTIVES: Here, we evaluate the role of endogenous cannabinoids in the cortico-limbic circuitry in cue-induced nicotine-seeking behavior in rats. METHODS: Animals were first trained to self-administer nicotine (0.03 mg/kg/injection, IV) under conditions in which responding was reinforced jointly by response-contingent nicotine injections and audiovisual stimuli. During subsequent sessions, nicotine was withdrawn and responding was reinforced by contingent presentation of the stimuli only. One month after nicotine removal, the cannabinoid CB1 receptor antagonist, rimonabant, was injected bilaterally into the shell of the nucleus accumbens (ShNAcc, 0.3, 3, or 30 ng/0.5 microl), the basolateral amygdala (BLA, 30 ng/0.5 microl), or the prelimbic cortex (PLCx, 30 ng/0.5 microl). RESULTS: Rimonabant injected into the ShNAcc dose-dependently reduced nicotine-seeking behavior without modifying spontaneous locomotor activity. Similar results were obtained when the drug (30 ng) was injected into the BLA or the PLCx. The anatomical specificity was confirmed in a control experiment using [(3)H]rimonabant. Fifteen minutes after drug injection, when the behavioral effects of rimonabant were already achieved, radioactivity was detected at the site of injection and had not diffused to adjacent regions. CONCLUSIONS: These findings demonstrate that increased endocannabinoid transmission critically triggers conditioned nicotine-seeking behavior in key cortico-limbic regions.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Cerebral Cortex/physiology , Conditioning, Operant/physiology , Endocannabinoids , Limbic System/physiology , Nicotine/administration & dosage , Signal Transduction/physiology , Amygdala/anatomy & histology , Amygdala/drug effects , Amygdala/physiology , Animals , Behavior, Addictive/physiopathology , Behavior, Addictive/prevention & control , Cannabinoid Receptor Modulators/antagonists & inhibitors , Cerebral Cortex/anatomy & histology , Cerebral Cortex/drug effects , Conditioning, Operant/drug effects , Cues , Dose-Response Relationship, Drug , Ganglionic Stimulants/administration & dosage , Injections, Intravenous , Injections, Intraventricular , Limbic System/anatomy & histology , Limbic System/drug effects , Male , Models, Anatomic , Motor Activity/drug effects , Motor Activity/physiology , Nucleus Accumbens/anatomy & histology , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Piperidines/administration & dosage , Piperidines/therapeutic use , Pyrazoles/administration & dosage , Pyrazoles/therapeutic use , Rats , Rats, Sprague-Dawley , Rimonabant , Self Administration , Signal Transduction/drug effects , Time Factors
7.
Psychopharmacology (Berl) ; 187(2): 237-44, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16779555

ABSTRACT

RATIONALE: SSR149415 ((2S, 4R)-1-[5-chloro-1-[(2,4-dimethoxyphenyl)sulfonyl]-3-(2-methoxyphenyl)-2-oxo-2,3-dihydro-1H-indol-3-yl]-4-hydroxy-N,N-dimethyl-2-pyrrolidinecarboxamide), the first selective nonpeptide vasopressin V1b receptor antagonist has been shown to induce antidepressant-and anxiolytic-like effects following systemic administration, whereas intraseptal infusion of the drug engender antidepressant-but not anxiolytic-like effects. OBJECTIVES: Based on recent evidence that V1b receptors are located within the amygdaloid complex, a structure which is well known for its modulatory role of emotional processes, the possible involvement of the different amygdaloid nuclei in the anxiolytic- and/or antidepressant-like effects of SSR149415 was examined. METHODS: Male Sprague-Dawley or Wistar rats were infused with SSR149415 into the central (CeA), the basolateral (BlA), or the medial (MeA) nucleus of the amygdala and tested 10 min after microinjection in the elevated plus-maze or the forced-swimming test, two models typically used for assessing the anxiolytic and antidepressant effects of drugs, respectively. RESULTS: Microinjection of SSR149415 into the BlA (1-10 ng), but not into the CeA or the MeA, increased the percentage of time spent in the open arms of the elevated plus-maze, indicating anxiolytic-like effects. Furthermore, in the forced-swimming test, microinjection of the drug into the CeA (1, 10, and 100 ng), BlA (1-10 ng), or MeA (100 ng) decreased immobility, an effect which is indicative of an antidepressant-like action. Together, these findings indicate that while the antidepressant-like effects of SSR149415 are mediated by different amygdaloid nuclei, its anxiolytic-like effects appear to involve only the basolateral nucleus of the amygdala. Moreover, these results add further evidence to the role of extrahypothalamic vasopressinergic systems in the control of emotional responses.


Subject(s)
Affect/drug effects , Amygdala/drug effects , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Antidiuretic Hormone Receptor Antagonists , Arousal/drug effects , Indoles/pharmacology , Pyrrolidines/pharmacology , Animals , Brain Mapping , Dose-Response Relationship, Drug , Escape Reaction/drug effects , Fear/drug effects , Male , Maze Learning/drug effects , Microinjections , Motivation , Rats , Rats, Sprague-Dawley , Swimming
8.
Pharmacol Biochem Behav ; 83(4): 533-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16624395

ABSTRACT

There is a growing interest in the potential anxiolytic- and antidepressant-like effects of compounds that target neurokinin receptors. Since the structure and the pharmacology of the human neurokinin receptor resembles that of gerbils, rather than that of mice or rats, we decided to investigate the anxiolytic- and /or antidepressant-like effects of NK1 (SSR240600), NK2 (saredutant) and NK3 (osanetant) receptor antagonists in gerbils. It was found that saredutant (3-10 mg/kg, p.o.) and osanetant (3-10 mg/kg, p.o.) produced anxiolytic-like effects in the gerbil social interaction test. These effects were similar to those obtained with the V1b receptor antagonist SSR149415 (3-10 mg/kg, p.o.), diazepam (1 mg/kg, p.o.) and buspirone (10 mg/kg, p.o.). Fluoxetine and SSR240600 were devoid of effects in this test. In the tonic immobility test in gerbils, saredutant (5-10 mg/kg, i.p.) and osanetant (5-10 mg/kg, i.p.) produced similar effects to those observed with fluoxetine (7.5-15 mg/kg, i.p.), SSR149415 (10-30 mg/kg, p.o.) and buspirone (3 mg/kg, i.p.). Diazepam and SSR240600 were inactive in this paradigm. In conclusion, the present study indicates further that NK2 and NK3 receptor antagonists may have therapeutic potential in the clinical management of anxiety and depression.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Receptors, Neurokinin-2/antagonists & inhibitors , Receptors, Neurokinin-3/antagonists & inhibitors , Animals , Benzamides/pharmacology , Dose-Response Relationship, Drug , Gerbillinae , Immobilization , Male , Motor Activity/drug effects , Piperidines/pharmacology , Receptors, Neurokinin-2/physiology , Receptors, Neurokinin-3/physiology , Social Behavior
9.
CNS Neurol Disord Drug Targets ; 5(2): 135-45, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16611088

ABSTRACT

This paper aimed at reviewing the involvement of neuropeptides in various psychiatric diseases, particularly in depression, and anxiety disorders. General features of neuropeptides are first described, including the history of their discovery, their definition, classification, biosynthesis, transport, release, inactivation, as well as their interaction with specific neuronal receptors. The differences with classical neurotransmitters are mentioned, as well as the different patterns of co-transmission. Finally, different mechanisms, both at the cellular and at the systemic level, are proposed that may explain the involvement of these molecules in various psychiatric diseases. Indeed, at the cellular level, a neuropeptide can be involved in a psychiatric disease, either because it is co-localized with a classical neurotransmitter involved in a disease, or because the neuropeptide-containing neuron projects on a target neuron involved in the disease. At the systemic level, a neuropeptide can play a direct role in the expression of a symptom of the disease. This is illustrated by different examples.


Subject(s)
Anxiety Disorders/metabolism , Brain Chemistry/physiology , Brain/metabolism , Brain/physiopathology , Depressive Disorder/metabolism , Neuropeptides/metabolism , Animals , Anxiety Disorders/physiopathology , Depressive Disorder/physiopathology , Humans , Neuropeptides/classification , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Receptors, Neurotransmitter/metabolism , Synaptic Transmission/physiology
10.
Psychopharmacology (Berl) ; 181(1): 134-44, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15830220

ABSTRACT

RATIONALE: Selective attention deficit, characterised by the inability to differentiate relevant from irrelevant information, is considered to underlie many cognitive deficits of schizophrenia, and appears to be only marginally responsive to treatment with current antipsychotics. OBJECTIVES: We compared the activity of the putative atypical antipsychotic SSR181507 (a dopamine D(2) receptor antagonist and 5HT(1A) receptor agonist) with reference compounds, on disturbances of novelty discrimination in a social context in rats, a behavioural paradigm that putatively models selective attention deficit. METHODS: A first (familiar) juvenile rat was presented to an adult rat for a period (P1) of 30 min. A second (novel) juvenile was then introduced at the end of P1 for a period (P2) of 5 min. The ability of the adult rat to discriminate between the two juveniles, presented at the same time, was evaluated by measuring the ratio of the time spent in interaction with the novel vs the familiar juvenile during P2. RESULTS: Adult rats spent more time exploring the novel than the familiar juvenile. This novelty discrimination capacity was disrupted by: (1) parametric modification of the procedure (reduction of time spent in contact with the familiar juvenile during P1); (2) acute injection of psychotomimetics that are known to induce schizophrenia-like symptoms in humans, such as phencyclidine (PCP; 3 mg/kg, i.p.) and d-amphetamine (1 mg/kg, i.p.) and (3) neonatal treatment with PCP (three injections of 10 mg/kg, s.c.), a model based on the neurodevelopmental hypothesis of schizophrenia. The potential atypical antipsychotic SSR181507 (0.03-3 mg/kg, i.p.) and the atypical antipsychotics clozapine (0.1-1 mg/kg, i.p.) and amisulpride (1-3 mg/kg, i.p.) attenuated deficits in novelty discrimination produced by parametric manipulation and by acute or neonatal treatment with PCP. The typical antipsychotic haloperidol (up to 0.3 mg/kg, i.p.) attenuated only deficits in novelty discrimination produced by parametric modification. CONCLUSION: Collectively, these results suggest that SSR181507 can alleviate disturbances of novelty discrimination in a social context in rats, and that this paradigm may represent a suitable animal model of selective attention deficits observed in schizophrenia.


Subject(s)
Behavior, Animal/drug effects , Dioxanes/pharmacology , Discrimination, Psychological/drug effects , Dopamine D2 Receptor Antagonists , Serotonin 5-HT1 Receptor Agonists , Tropanes/pharmacology , Age Factors , Animals , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/physiopathology , Clozapine/pharmacology , Dextroamphetamine/pharmacology , Dioxanes/administration & dosage , Disease Models, Animal , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Female , Haloperidol/pharmacology , Humans , Imipramine/pharmacology , Injections, Intraperitoneal , Male , Phencyclidine/pharmacology , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/physiology , Receptors, Dopamine D2/physiology , Recognition, Psychology/drug effects , Serotonin Antagonists/pharmacology , Social Behavior , Tacrine/pharmacology , Tropanes/administration & dosage
11.
Neuropharmacology ; 46(8): 1121-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15111019

ABSTRACT

Social behaviour is frequently impaired in schizophrenic patients, and current antipsychotics appear poorly effective in alleviating this deficit. SSR181507 is a selective dopamine D2 receptor antagonist and 5-HT1A receptor agonist [Neuropsychopharmacology 28 (2003) 2064] with an atypical antipsychotic profile and additional antidepressant/anxiolytic activities [Neuropsychopharmacology 28 (2003) 1889]. Here, we sought to assess the efficacy of SSR181507, and of reference antipsychotics and antidepressant/anxiolytics, to counteract phencyclidine (PCP)-induced social interaction deficit in rats. Pairs of unfamiliar rats were placed for 10 min each day into a dimly lit arena, during four consecutive days. On the test day (5th day), each pair was placed into the arena 30 min after i.p. treatment with PCP (or vehicle) and a challenge compound or vehicle (same for both rats, i.p. or s.c.). The time spent in social interaction was scored during 10 min. PCP (1 mg/kg) decreased social interaction time by about 35%. This effect was fully antagonized by pre-treatment with SSR181507 (1 mg/kg). In contrast, neither haloperidol (0.05 and 0.1 mg/kg) nor clozapine (0.3 and 1 mg/kg) antagonized this PCP-induced deficit. The selective 5-HT1A receptor agonist 8-OH-DPAT (0.025 and 0.05 mg/kg s.c.), but not the anxiolytic diazepam (0.75 and 1.5 mg/kg), also improved social interaction impairment in PCP-treated rats: this would indicate that the 5-HT1A receptor agonist properties of SSR181507 are responsible for the reversal of PCP-induced social deficit. These data suggest that, in addition to its atypical antipsychotic profile and antidepressant/anxiolytic activities, SSR181507 has a potential therapeutic activity in another key feature of schizophrenia poorly controlled by current antipsychotics, namely deterioration in social functioning.


Subject(s)
Dioxanes/pharmacology , Dopamine D2 Receptor Antagonists , Interpersonal Relations , Phencyclidine/pharmacology , Serotonin 5-HT1 Receptor Agonists , Tropanes/pharmacology , Animals , Antipsychotic Agents/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/physiology , Receptors, Dopamine D2/physiology , Social Behavior
12.
Mol Psychiatry ; 9(3): 278-86, 224, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14699428

ABSTRACT

Repeated exposure to stress is known to induce structural remodelling and reduction of neurogenesis in the dentate gyrus. Corticotrophin-releasing factor (CRF) and vasopressin (AVP) are key regulators of the stress response via activation of CRF(1) and V(1b) receptors, respectively. The blockade of these receptors has been proposed as an innovative approach for the treatment of affective disorders. The present study aimed at determining whether the CRF(1) receptor antagonist SSR125543A, the V(1b) receptor antagonist SSR149415, and the clinically effective antidepressant fluoxetine may influence newborn cell proliferation and differentiation in the dentate gyrus of mice subjected to the chronic mild stress (CMS) procedure, a model of depression with predictive validity. Repeated administration of SSR125543A (30 mg/kg i.p.), SSR149415 (30 mg/kg i.p.), and fluoxetine (10 mg/kg i.p.) for 28 days, starting 3 weeks after the beginning of the stress procedure, significantly reversed the reduction of cell proliferation produced by CMS, an effect which was paralleled by a marked improvement of the physical state of the coat of stressed mice. Moreover, mice subjected to stress exhibited a 53% reduction of granule cell neurogenesis 30 days after the end of the 7-week stress period, an effect which was prevented by all drug treatments. Collectively, these results point to an important role of CRF and AVP in the regulation of dentate neurogenesis, and suggest that CRF(1) and V(1b) receptor antagonists may affect plasticity changes in the hippocampal formation, as do clinically effective antidepressants.


Subject(s)
Antidiuretic Hormone Receptor Antagonists , Depression/physiopathology , Indoles/pharmacology , Neurons/cytology , Pyrrolidines/pharmacology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Stress, Psychological/prevention & control , Thiazoles/pharmacology , Animals , Antidepressive Agents/pharmacology , Depression/prevention & control , Disease Models, Animal , Fluoxetine/pharmacology , Male , Mice , Mice, Inbred BALB C , Neurons/drug effects
13.
Psychopharmacology (Berl) ; 158(3): 241-51, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11713613

ABSTRACT

RATIONALE: It has been suggested that tachykinin NK(2) receptor antagonists may have therapeutic utility in anxiety and/or depressive disorders. OBJECTIVE: The present study investigated the modulatory action of the NK(2) receptor antagonist SR48968 on emotional processes in rodents. METHODS: The tests used include classical models of anxiety (punished lever pressing and punished drinking conflict tests, elevated plus-maze in rats), a model based on defensive behaviors of mice confronted with a natural threat (a rat), and two tests based on exposure of rats or mice to a natural predator (a cat) followed by subsequent exposure to a cat odor cue. The prototypical anxiolytic diazepam was used throughout as a positive control, the antidepressant imipramine was tested in the mouse defense test battery and in both models of predatory exposure, and the selective CRF1 receptor antagonist antalarmin was used in the cat-exposure test in rats. RESULTS: Unlike diazepam, SR48968 failed to increase rates of responding suppressed by punishment in both conflict procedures. By contrast, in the elevated plus-maze test, the NK(2) receptor antagonist (3 mg/kg, IP) elicited positive effects on traditional and ethologically derived measures of anxiety. In the mouse defense test battery, SR48968 (0.03-1 mg/kg, IP) decreased flight reactions, risk assessment behavior, defensive biting and escape attempts. While the magnitude of the effects on flight, risk assessment and escape attempts of the NK(2) receptor antagonist was less than that of diazepam, SR48968 appeared to be as effective as the BZ on defensive biting. In rats previously exposed to a cat, SR48968 (3 mg/kg, IP), antalarmin (1 mg/kg, IP), imipramine (30 mg/kg, IP), but not diazepam, reduced subsequent high levels of avoidance responses when subjects are exposed to a cat odor-saturated cue 1 h later. Similar effects of SR48968 (0.1-0.3 mg/kg, IP) were observed in mice following repeated administration (twice a day/5 days/IP). Importantly, the positive effects of the NK(2) receptor antagonist were evident at doses that did not impair general activity, unlike imipramine which displayed mainly sedative action. Moreover, the (R)-enantiomer of SR48968, SR48965, which was tested in the elevated plus-maze, the mouse defense test battery and the cat exposure tests, was much less active than its racemate, indicating a stereoselective action of SR48968. CONCLUSION: These data show that while SR48968 has limited or no efficacy in models or behavioral measures mainly sensitive to BZs, it shows good activity in reducing anxiety-like behaviors following traumatic stress or upon forced and unavoidable contact with a threatening stimulus. This suggests that NK(2) receptor antagonists may have a potential in the treatment of some forms of anxiety disorders.


Subject(s)
Benzamides/pharmacology , Emotions/drug effects , Piperidines/pharmacology , Receptors, Neurokinin-2/antagonists & inhibitors , Animals , Anxiety/drug therapy , Benzamides/therapeutic use , Cats , Dose-Response Relationship, Drug , Emotions/physiology , Male , Mice , Piperidines/therapeutic use , Rats , Rats, Inbred WKY , Rats, Long-Evans , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Neurokinin-2/physiology
14.
Behav Brain Res ; 125(1-2): 141-9, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11682105

ABSTRACT

Measuring anxiety-like behaviour in mice has been mostly undertaken using a few classical animal models of anxiety such as the elevated plus-maze, the light/dark choice or the open-field tests. All these procedures are based upon the exposure of subjects to unfamiliar aversive places. Anxiety can also be elicited by a range of threats such as predator exposure. Furthermore, the concepts of "state" and "trait" anxiety have been proposed to differentiate anxiety that the subject experiences at a particular moment of time and that is increased by the presence of an anxiogenic stimulus, and anxiety that does not vary from moment to moment and is considered to be an "enduring feature of an individual". Thus, when assessing the behaviour of mice, it is necessary to increase the range of behavioural paradigms used, including animal models of "state" and "trait" anxiety. In the last few years, many mice with targeted mutations have been generated. Among them some have been proposed as animal models of pathological anxiety, since they display high level of anxiety-related behaviours in classical tests. However, it is important to emphasise that such mice are animal models of a single gene dysfunction, rather than models of anxiety, per se. Inbred strains of mice, such as the BALB/c line, which exhibits spontaneously elevated anxiety appear to be a more suitable model of pathological anxiety.


Subject(s)
Anxiety/genetics , Arousal/genetics , Phenotype , Animals , Behavior, Animal/physiology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Mice, Knockout/genetics , Mice, Mutant Strains/genetics , Mice, Transgenic/genetics , Models, Genetic
15.
J Pharmacol Exp Ther ; 299(2): 449-58, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602654

ABSTRACT

The present study investigated the effects of the selective neurokinin-2 (NK2) receptor antagonist SR48968 in behavioral, electrophysiological, and biochemical tests sensitive to the action of prototypical antidepressants (fluoxetine, imipramine) or to corticotropin-releasing factor (CRF) receptor antagonists, which have been proposed recently as potential antidepressants. Results showed that SR48968 (0.3-10 mg/kg i.p.) produced antidepressant-like activity because it reduced immobility in the forced swimming test in both mice and rats, and decreased the amount of maternal separation-induced vocalizations in guinea pig pups. This latter effect appears to involve a reduction of stress-induced substance P release because SR48968 reduced the separation-induced increase in the number of neurons displaying neurokinin-1 receptor internalization in the amygdala. Furthermore, SR48968 increased the expression of the cAMP response-element binding protein mRNA in the rat hippocampus after repeated (1 mg/kg i.p., 21 days), but not acute administration. Finally, neuronal firing of the locus coeruleus (LC) and noradrenergic (NE) release in the prefrontal cortex both elicited by an uncontrollable stressor or an intraventricular administration of CRF were reduced by SR48968 (0.3-1 mg/kg i.p.). The finding that SR48968 (1 mg/kg i.p.) blocked the cortical release of NE induced by an intra-LC infusion of the preferential NK2 receptor agonist neurokinin A suggested the presence of NK2 receptors in this latter region. Importantly, SR48965 (1-10 mg/kg i.p.), the optical antipode of SR48968, which is devoid of affinity for the NK2 receptor, was inactive in all the models used. These data suggest that NK2 receptor blockade may constitute a novel mechanism in the treatment of depression and CRF-related disorders.


Subject(s)
Antidepressive Agents/pharmacology , Corticotropin-Releasing Hormone/blood , Receptors, Neurokinin-2/antagonists & inhibitors , Animals , Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Benzamides/pharmacology , Brain Chemistry/physiology , Cyclic AMP Response Element-Binding Protein/biosynthesis , Guinea Pigs , Hippocampus/drug effects , Hippocampus/metabolism , In Situ Hybridization , Locus Coeruleus/physiology , Maternal Deprivation , Mice , Microdialysis , Norepinephrine/blood , Piperidines/pharmacology , Prefrontal Cortex/physiology , RNA, Messenger/biosynthesis , Rats , Swimming/psychology , Vocalization, Animal/drug effects
16.
Neuropharmacology ; 41(3): 400-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11522332

ABSTRACT

This article had several objectives. First it aimed at investigating the anxiogenic-like behaviors elicited by unavoidable cat exposure and/or cat odor across nine strains of mice (BALB/c, C57BL/6, C3H, CBA, DBA/2, NMRI, NZB, SJL, Swiss) in a modified version of the free-exploration test. The second objective was to investigate possible neurochemical changes following cat exposure in Swiss mice by measuring the turnover of dopamine (DA), noradrenaline (NA) and serotonin (5-HT) in several brain regions known to be involved in the modulation of emotional processes (hippocampus, hypothalamus and striatum). Finally, the third objective was to examine the effects of anxiolytic drug treatments on the anxiogenic responses elicited by a cat odor (i.e. a feces) in Swiss mice previously exposed to a cat using the free-exploration test. Results from the strain comparison showed that mice could be divided into three distinct groups: two non-reactive strains (NZB and SJL) which were relatively insensitive to predatory exposure and/or odor; five intermediate-reactive strains (Swiss, NMRI, CBA, C3H and BALB/c) which displayed clear anxiogenic-like responses only when exposed to both cat and, subsequently, to feces; and two high reactive strains (C57BL/6 and DBA/2) which showed anxiogenic-like reactions following cat exposure, regardless of the stimulus (clay or feces) present in the free-exploration cage. Neurochemical data revealed that, while brain levels of NA, DA, 5-HT in cat exposed Swiss mice were not significantly different from those of control animals, turnover rates of these monoamines were increased in the hippocampus (NA and 5-HT), hypothalamus and striatum (DA) after cat exposure. Results from pharmacological experiments indicated that repeated administration of the 5-HT reuptake inhibitor fluoxetine (5-20 mg/kg, twice a day, for 5 days) completely abolished avoidance of the cat feces in Swiss mice previously exposed to the predator. Neither acute nor repeated administration of the classical anxiolytic diazepam was able to reduce avoidance behavior of the anxiogenic stimulus in the free-exploration test. Taken together, these findings indicate that the exposure of mice to unavoidable predatory stimuli is associated with behavioral and neurochemical changes consistent with increased anxiety.


Subject(s)
Behavior, Animal/physiology , Brain Chemistry/physiology , Stress, Psychological/metabolism , Stress, Psychological/psychology , Animals , Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Cats , Diazepam/pharmacology , Dopamine/metabolism , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Fluoxetine/pharmacology , Male , Mice , Mice, Inbred Strains , Norepinephrine/metabolism , Predatory Behavior , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Species Specificity
17.
J Pharmacol Exp Ther ; 298(2): 753-68, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11454940

ABSTRACT

SL651498 [6-fluoro-9-methyl-2-phenyl-4-(pyrrolidin-1-yl-carbonyl)-2,9-dihydro-1H-pyrido[3,4-b]indol-1-one] is a novel pyridoindole derivative that displays high affinity for rat native GABA(A) receptors containing alpha(1) (K(i) = 6.8 nM) and alpha2 (K(i) = 12.3 nM) subunits, and weaker affinity for alpha5-containing GABA(A) receptors (K(i) = 117 nM). Studies on recombinant rat GABA(A) receptors confirm these data (K(i), alpha1beta2gamma2 = 17, alpha2beta2gamma2 = 73, alpha5beta3gamma2 = 215 nM) and indicate intermediate affinity for the alpha3beta2gamma2 subtype (K(i) = 80 nM). SL651498 behaves as a full agonist at recombinant rat GABA(A) receptors containing alpha2 and alpha3 subunits and as a partial agonist at recombinant GABA(A) receptors expressing alpha1 and alpha5 subunits. SL651498 elicited anxiolytic-like activity similar to that of diazepam [minimal effective dose (MED): 1-10 mg/kg, i.p.] in three conflict models, in the elevated plus-maze, the light/dark test, and the defense test battery in rats and mice. Results from activity tests and electroencephalogram analysis indicated that SL651498 induced muscle weakness, ataxia, or sedation at doses much higher than those producing anxiolytic-like activity (MED > or = 30 mg/kg, i.p.). Repeated treatment for 10 days with SL651498 (30 mg/kg, i.p., b.i.d.) in mice was not associated with the development of tolerance to its anticonvulsant effects or physical dependence. Furthermore, SL651498 was much less active than diazepam in potentiating the depressant effects of ethanol in mice. The "anxioselective" profile of SL651498 points to a major role for GABA(A) alpha2 subtype in regulating anxiety and suggests that selectively targeting GABA(A) receptor subtypes can lead to drugs with increased clinical specificity.


Subject(s)
Anti-Anxiety Agents/pharmacology , Indoles/pharmacology , Pyrroles/pharmacology , Receptors, GABA-A/drug effects , Animals , Anticonvulsants/pharmacology , Anxiety/psychology , Central Nervous System Depressants/pharmacology , Discrimination, Psychological/drug effects , Drug Interactions , Drug Tolerance , Ethanol/pharmacology , Male , Membrane Potentials/drug effects , Mice , Motor Activity/drug effects , Patch-Clamp Techniques , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Substance-Related Disorders/psychology
18.
Neurosci Biobehav Rev ; 25(3): 205-18, 2001 May.
Article in English | MEDLINE | ID: mdl-11378177

ABSTRACT

The natural defensive behaviors of laboratory mice have been evaluated in both seminatural and highly structured situations; and characterized in terms of eliciting stimuli, response to pharmacological agents, behavior patterns, and outcome or effect on the social and physical environment. The defense patterns of laboratory mice and rats are generally similar, but mice show risk assessment on initial exposure to highly threatening stimuli while rats do not, while rats display alarm vocalizations, missing in mice. Quantitative differences in freezing and flight for laboratory mice and rats appear to largely reflect domestication effects, with wild mice and rats more similar to each other. This nexus of detailed within-species and comparative data on defense patterns makes it possible to reliably elicit specific defenses in mice or rats in an experimental context, providing well-validated assays of the natural defensive behaviors themselves, as opposed to 'models' of defense. The mouse--rat comparisons indicate considerable cross-species generality for these defense patterns, as does a scattered but considerable literature on other mammalian species, generally involving field studies and typically focusing on those aspects of defensive behavior that are visible at a distance, such as vigilance, or flight. Although potential homologies between normal mouse and human defense systems should ideally involve all four pattern components (stimulus, organismic factors, response characteristics, outcome), predictive validity in terms of response to drugs active against specific defensive psychopathology is the most extensively investigated of these. Flight, as measured in the Mouse Defense Test Battery shows a consistently appropriate response to panicolytic, panicogenic, and panic-neutral drugs, while some other predictive 'panic models' (dPAG-stimulation; DMH-inhibition; possibly conditioned suppression of drinking paradigms) also elicit and (indirectly) measure behaviors potentially related to flight. Models unrelated to flight (e.g. ultrasonic vocalization to conditioned stimuli); or for which flight elements may a relatively minor contributor to the behavior measured (Elevated T-maze) are less predictive of panicolytic or panicogenic action. These findings indicate that natural defensive behaviors provide a well-characterized pattern for analysis of effects of genetic or other physiological manipulations in the mouse, and may also serve as a model for analysis of defense-related human psychopathology.


Subject(s)
Anxiety/drug therapy , Anxiety/psychology , Behavior, Animal/drug effects , Mice/psychology , Panic/drug effects , Animals
19.
Braz J Med Biol Res ; 34(2): 145-54, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11175489

ABSTRACT

This article is a transcription of an electronic symposium sponsored by the Brazilian Society of Neuroscience and Behavior (SBNeC). Invited researchers from the European Union, North America and Brazil discussed two issues on anxiety, namely whether panic is a very intense anxiety or something else, and what aspects of clinical anxiety are reproduced by animal models. Concerning the first issue, most participants agreed that generalized anxiety and panic disorder are different on the basis of clinical manifestations, drug response and animal models. Also, underlying brain structures, neurotransmitter modulation and hormonal changes seem to involve important differences. It is also common knowledge that existing animal models generate different types of fear/anxiety. A challenge for future research is to establish a good correlation between animal models and nosological classification.


Subject(s)
Anxiety , Disease Models, Animal , Panic , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/physiopathology , Benzodiazepines/pharmacology , Brain/drug effects , Brain/physiopathology , Fear/drug effects , Humans , Panic/drug effects , Periaqueductal Gray/drug effects , Periaqueductal Gray/physiopathology , Serotonin/pharmacology
20.
Braz. j. med. biol. res ; 34(2): 145-154, Feb. 2001.
Article in English | LILACS | ID: lil-281592

ABSTRACT

This article is a transcription of an electronic symposium sponsored by the Brazilian Society of Neuroscience and Behavior (SBNeC). Invited researchers from the European Union, North America and Brazil discussed two issues on anxiety, namely whether panic is a very intense anxiety or something else, and what aspects of clinical anxiety are reproduced by animal models. Concerning the first issue, most participants agreed that generalized anxiety and panic disorder are different on the basis of clinical manifestations, drug response and animal models. Also, underlying brain structures, neurotransmitter modulation and hormonal changes seem to involve important differences. It is also common knowledge that existing animal models generate different types of fear/anxiety. A challenge for future research is to establish a good correlation between animal models and nosological classification


Subject(s)
Humans , Anxiety , Disease Models, Animal , Panic , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/physiopathology , Benzodiazepines/pharmacology , Brain/drug effects , Brain/physiopathology , Computer Communication Networks , Fear/drug effects , Panic/drug effects , Periaqueductal Gray/drug effects , Periaqueductal Gray/physiopathology , Serotonin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...