Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Acta Neuropsychiatr ; : 1-11, 2022 Aug 22.
Article in English | MEDLINE | ID: mdl-35993329

ABSTRACT

Addiction to psychostimulant drugs, such as cocaine, D-amphetamine, and methamphetamine, is a public health issue that substantially contributes to the global burden of disease. Psychostimulant drugs promote an increase in dopamine levels within the mesocorticolimbic system, which is central to the rewarding properties of such drugs. Cannabinoid receptors (CB1R and CB2R) are expressed in the main areas of this system and implicated in the neuronal mechanisms underlying the rewarding effect of psychostimulant drugs. Here, we reviewed studies focusing on pharmacological intervention targeting cannabinoid CB1R and CB2R and their interaction in the modulation of psychostimulant responses.

2.
Psychopharmacology (Berl) ; 238(11): 3107-3118, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34328516

ABSTRACT

RATIONALE: Cannabis sativa is the most widely used drug by adolescents globally. The recreational use of synthetic cannabinoids by teenagers has also grown in recent years. Despite the wrong perception that exposure to these drugs does not cause harm, repeated exposure to cannabinoids at early stages of life compromises important maturation processes and brain development. Chronic early cannabinoid use has been related to a higher risk of psychiatric outcomes, including cocaine addiction. Evidence suggests that exposure to natural and synthetic cannabinoids during adolescence modifies molecular and behavioral effects of cocaine in adulthood. Responses to cocaine are regulated by epigenetic mechanisms, such as DNA methylation, in the brain's reward regions. However, the involvement of these processes in modulation of the vulnerability to the effects of cocaine induced by prior exposure to cannabinoids remains poorly understood. OBJECTIVES: Investigate whether exposure to the synthetic cannabinoid WIN55,212-2 during adolescence modulates anxiety- and depression-like behavior, memory, and cocaine reward in adult mice. We also evaluated whether exposure to cannabinoids during adolescence modulates the expression of enzymes that are involved in DNA methylation. RESULTS: Exposure to WIN55,212-2 during adolescence did not alter anxiety- or depressive-like behavior. However, prior exposure to cannabinoids inhibited cocaine-induced conditioned place preference without modulating cocaine-induced hyperlocomotion, accompanied by an increase in expression of the enzyme DNA methyltransferase 3a (DNMT3a) in the prefrontal cortex. CONCLUSIONS: Our findings suggest that exposure to WIN55,212-2 during adolescence leads to changes in DNMT3a expression, and this pathway appears to be relevant to modulating the rewarding effects of cocaine.


Subject(s)
Cannabinoids , Cocaine , Animals , Cannabinoids/pharmacology , Cocaine/pharmacology , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Mice , Prefrontal Cortex/metabolism , Reward
3.
Neuropharmacology ; 195: 108679, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34157363

ABSTRACT

BACKGROUND AND PURPOSE: Although useful as a rapid-acting antidepressant drug, ketamine is known to induce psychotomimetic effects, which may interfere with its therapeutic use. Cannabidiol (CBD) is a non-psychostimulant compound from Cannabis sativa, which has shown promising antidepressant effects without inducing hyperlocomotion. AMPA receptor activation is involved in the antidepressant effect induced by ketamine, but its relevance for the effects of CBD is not known. Moreover, given that CBD has antipsychotic and antidepressant properties, it is unknown whether adding CBD to ketamine could potentiate the antidepressant properties of ketamine while also attenuating its psychostimulant effects. EXPERIMENTAL APPROACH: S-Ketamine (2.5, 3, 5, 10, 30 mg/kg) and cannabidiol (3, 10, 30 mg/kg) were administered alone or in combination to male Swiss mice. Independent groups received NBQX (AMPA receptor antagonist) 5 min before administration of CBD or S-ketamine. The antidepressant-like effect was assessed in the forced swimming test (FST), and the open field test (OFT) evaluated the psychostimulant effect. KEY RESULTS: CBD induced significant dose-dependent antidepressant effects without causing hyperlocomotion in the OFT. S-ketamine produced an antidepressant effect associated with hyperlocomotion in the higher dose. NBQX inhibited the antidepressant effect of both ketamine and CBD. Pretreatment with CBD (10 mg/kg) attenuated the ketamine-induced hyperlocomotion while preserving its antidepressant effect. CONCLUSION: AND IMPLICATIONS: Similar to ketamine, the antidepressant-like effect elicited by CBD involves AMPA receptor activation. Additionally, CBD prevents the hyperlocomotion induced by S-ketamine without affecting its antidepressant-like effect. Our findings suggest that CBD and ketamine's combined administration can be a promising therapeutic strategy for achieving an appropriate antidepressant effect without unwanted side-effects. This article is part of the special issue on 'Cannabinoids'.


Subject(s)
Antidepressive Agents/administration & dosage , Cannabidiol/administration & dosage , Depression/drug therapy , Ketamine/administration & dosage , Motor Activity/drug effects , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Cannabidiol/therapeutic use , Disease Models, Animal , Hippocampus/drug effects , Ketamine/therapeutic use , Male , Mice
4.
Behav Brain Res ; 395: 112866, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32827568

ABSTRACT

RATIONALE: Reduced levels of orexin-A (OXA) in the central nervous system (CNS) have been associated with the pathophysiology of depression and its exogenous administration promotes antidepressant-like effect. The mechanisms associated with these effects are, however, not yet known. Herein, we investigated the hypothesis that OXA effects could be associated with orexin 1 receptor (OX1R) and tyrosine receptor kinase B (TrkB) activation, in the ventromedial prefrontal cortex (vmPFC), a brain region that is central to depression neurobiology. OBJECTIVES: 1. To Investigate the effects induced by OXA administration into the vmPFC; 2. Evaluate the participation of OX1R and TrkB in behavioral responses induced by OXA. METHODS: Male Wistar rats received intra-vmPFC injections of OXA (10, 50 and 100 pmol/0.2 µL) and were exposed to the forced swimming test (FST) or the open field test (OFT). Independent groups received an intra-vmPFC injection of SB334867 (OX1R antagonist, 10 nmol/0.2 µL) or K252a (non-selective Trk antagonist, 10 pmol/0.2 µL), before local injection of OXA, and were exposed to the same tests. RESULTS: OXA injection (100 pmol/0.2 µL) into the vmPFC induced antidepressant-like effect, which was prevented by SB334867 and K252a pretreatments. CONCLUSION: OXA signaling in the vmPFC induces antidepressant-like effect in the FST which is dependent on OX1R and Trk receptors.


Subject(s)
Depression/drug therapy , Orexins/pharmacology , Prefrontal Cortex/metabolism , Animals , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Male , Motor Activity/drug effects , Orexin Receptors/drug effects , Orexin Receptors/metabolism , Orexins/administration & dosage , Orexins/metabolism , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Receptor, trkB/metabolism , Signal Transduction/drug effects , Stress, Psychological
5.
Physiol Behav ; 220: 112862, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32156558

ABSTRACT

Galectin-1 (Gal-1) and galectin-3 (Gal-3) are multifunctional glycan-binding proteins, expressed in the brain and in its limbic structures that are involved in behavioral control. Gal-1 induces the expression of the brain-derived neurotrophic factor (BDNF) and promotes adult neural stem cells proliferation, biological events impaired in stress-related psychiatric disorders, such as depression and anxiety. Despite that, there is no evidence regarding galectin involvement in emotional control during stressful situations. Thus, we analyzed the behavioral phenotype of Gal-1 or Gal-3 knock-out mice (Gal-1 KO or Gal-3 KO) in different experimental models predictive of depressive and compulsive-like behaviors. METHODS: C57BL-6 Gal-1 KO, Gal-3 KO, and wild-type mice (WT) were analyzed under the open field test (OFT) and, 6 h later, under the forced swim test (FST). Additionally, independent groups of male mice, lacking galectins or not, were exposed to the tail suspension test (TST) or to the marble burying test (MBT). The hippocampus and prefrontal cortex (PFC) of the mice submitted to MBT were dissected to access BDNF levels. RESULTS: Both Gal-1 and Gal-3 KO mice showed increased time of immobility in the FST and in the TST compared to WT animals, thus reflecting an impaired stress-coping behavior. Additionally, Gal-1 and Gal-3 KO female mice presented increased compulsive-like behavior in the MBT, without significant changes in the locomotor activity. BDNF levels were found to be decreased in the PFC of Gal-1 KO mice. DISCUSSION: Our results demonstrate that the absence of either endogenous Gal-1 and Gal-3 impairs stress-coping and increases compulsive-like behavior, suggesting that Gal-1 and Gal-3 are involved in the neurobiology of depression and obsessive-compulsive-like disorder.


Subject(s)
Galectin 1 , Galectin 3 , Stress, Psychological , Animals , Anxiety , Behavior, Animal , Brain-Derived Neurotrophic Factor/metabolism , Compulsive Behavior , Depression , Disease Models, Animal , Emotions , Female , Galectin 1/genetics , Galectin 3/genetics , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Behav Brain Res ; 368: 111897, 2019 08 05.
Article in English | MEDLINE | ID: mdl-30978407

ABSTRACT

Previous studies indicated that some general anesthetics induce long-term antidepressant and/or anxiolytic-like effects. This raises the concern about the use of anesthesia in surgeries that precede psychopharmacological tests, since it may be a potential bias on results depending on the experimental design used. Thus, we evaluated whether general anesthetics used in surgeries preceding psychopharmacological tests would affect rats behavior in tests predictive of antidepressant or anxiolytic-like effects. We tested if a single exposure to sub-anesthetic or anesthetic doses of tribromoethanol, chloral hydrate, thiopental or isoflurane would change rats behavior in the forced swimming test (FST) or in the elevated plus-maze (EPM) test, at 2 h or 7 days after their administration. We also evaluated whether prior anesthesia would interfere in the detection of the antidepressant-like effect of imipramine or the anxiolytic-like effect of diazepam. Previous anesthesia with the aforementioned anesthetics did not change rats behaviors in FST per se nor it changed the antidepressant-like effect induced by imipramine treatment. Rats previously anesthetized with tribromoethanol or chloral hydrate exhibited, respectively, anxiogenic-like and anxiolytic-like behaviors in the EPM. Prior anesthesia with thiopental or isoflurane did not produce any per se effect in rats behaviors in the EPM nor disturbed the anxiolytic-like effect of diazepam. Our results suggest that, in our experimental conditions, tribromoethanol and chloral hydrate are improper anesthetics for surgeries that precede behavioral analysis in the EPM. Isoflurane or thiopental may be suitable for anesthesia before evaluation in the EPM or in the FST.


Subject(s)
Anesthetics, General/adverse effects , Behavior, Animal/drug effects , Anesthetics, General/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety/drug therapy , Chloral Hydrate/adverse effects , Chloral Hydrate/pharmacology , Depression/drug therapy , Diazepam/pharmacology , Ethanol/adverse effects , Ethanol/analogs & derivatives , Ethanol/pharmacology , Imipramine/pharmacology , Isoflurane/adverse effects , Isoflurane/pharmacology , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Thiopental/adverse effects , Thiopental/pharmacology
7.
Mol Neurobiol ; 55(8): 7062-7071, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29383685

ABSTRACT

Several pieces of evidence indicate that elastase-2 (ELA2; chymotrypsin-like ELA2) is an alternative pathway to the generation of angiotensin II (ANGII). Elastase-2 knockout mice (ELA2KO) exhibit alterations in the arterial blood pressure and heart rate. However, there is no data on the behavioral consequences of ELA2 deletion. In this study, we addressed this question, submitting ELA2KO and wild-type (WT) mice to several models sensitive to anxiety- and depression-like, memory, and repetitive behaviors. Our data indicates a higher incidence of barbering behavior in ELA2KO compared to WT, as well as an anxiogenic phenotype, evaluated in the elevated plus maze (EPM). While a decrease in locomotor activity was observed in ELA2KO in EPM, this feature was not the main source of variation in the other parameters analyzed. The marble-burying test (MBT) indicated increase in repetitive behavior, observed by a higher number of buried marbles. The actimeter test indicated a decrease in total activity and confirmed the increase in repetitive behavior. The spatial memory was tested by repeated exposure to the actimeter in a 24-h interval. Both ELA2KO and WT exhibited decreased activity compared to the first exposure, without any distinction between the genotypes. However, when submitted to the cued fear conditioning, ELA2KO displayed lower levels of freezing behavior in the extinction session when compared to WT, but no difference was observed during the conditioning phase. Increased levels of BDNF were found in the prefrontal cortex but not in the hippocampus of ELA2KO mice compared to WT. Finally, in silico analysis indicates that ELA2 is putatively able to cleave BDNF, and incubation of the purified enzyme with BDNF led to the degradation of the latter. Our data suggested an anxiogenic- and antidepressant-like phenotype of ELA2KO, possibly associated with increased levels of BDNF in the prefrontal cortex.


Subject(s)
Antidepressive Agents/metabolism , Anxiety/enzymology , Brain-Derived Neurotrophic Factor/metabolism , Prefrontal Cortex/metabolism , Serine Endopeptidases/deficiency , Animals , Behavior, Animal , Computer Simulation , Conditioning, Psychological , Fear , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism
8.
J Neuroimmunol ; 314: 58-66, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29195684

ABSTRACT

Preclinical and clinical evidence suggests pro-inflammatory cytokines might play an important role in the neurobiology of schizophrenia and stress-related psychiatric disorders. Interleukin-18 (IL-18) is a member of the IL-1 family of cytokines and it is widely expressed in brain regions involved in emotional regulation. Since IL-18 involvement in the neurobiology of mental illnesses, including schizophrenia, remains unknown, this work aimed at investigating the behavior of IL-18 null mice (KO) in different preclinical models: 1. the prepulse inhibition test (PPI), which provides an operational measure of sensorimotor gating and schizophrenic-like phenotypes; 2. amphetamine-induced hyperlocomotion, a model predictive of antipsychotic activity; 3. resident-intruder test, a model predictive of aggressive behavior. Furthermore, the animals were submitted to models used to assess depressive- and anxiety-like behavior. IL-18KO mice showed impaired baseline PPI response, which was attenuated by d-amphetamine at a dose that did not modify PPI response in wild-type (WT) mice, suggesting a hypodopaminergic prefrontal cortex function in those mice. d-Amphetamine, however, induced hyperlocomotion in IL-18KO mice compared to their WT counterparts, suggesting hyperdopaminergic activity in the midbrain. Moreover, IL-18KO mice presented increased basal levels of IL-1ß levels in the hippocampus and TNF-α in the prefrontal cortex, suggesting an overcompensation of IL-18 absence by increased levels of other proinflammatory cytokines. Although no alteration was observed in the forced swimming or in the elevated plus maze tests in naïve IL-18KO mice, these mice presented anxiogenic-like behavior after exposure to repeated forced swimming stress. In conclusion, deletion of the IL-18 gene resembled features similar to symptoms observed in schizophrenia (positive and cognitive symptoms, aggressive behavior), in addition to increased susceptibility to stress. The IL-18KO model, therefore, could provide new insights into how changes in brain immunological homeostasis induce behavioral changes related to psychiatric disorders, such as schizophrenia.


Subject(s)
Brain/immunology , Interleukin-18/deficiency , Interleukin-18/immunology , Schizophrenia/immunology , Animals , Behavior, Animal/physiology , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Male , Mental Disorders , Mice , Mice, Inbred BALB C , Mice, Knockout , Schizophrenia/genetics
9.
Neuroscience ; 340: 126-134, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27771531

ABSTRACT

Cannabinoid type-1 (CB1) and transient receptor potential vanilloid type-1 (TRPV1) receptors may have opposite roles in modulating neural activity and, consequently, in regulating the stress response. These receptors are widely expressed in several brain structures, including the ventral medial prefrontal cortex (vmPFC). The functional consequences of the interaction between CB1 and TRPV1, however, have scarcely been explored. Therefore, we investigated if CB1 and TRPV1 receptors located in the vmPFC would be involved in the behavioral changes induced by the stress of the forced swim test (FST). Rats with cannulae implanted into the vmPFC were given the dual blocker of TRPV1 receptors and fatty acid amide hydrolase (FAAH), Arachidonyl serotonin (AA-5HT, 0.125/0.25/0.5nmol), TRPV1 antagonist, SB366791 (0.5/1/10nmol), FAAH inhibitor, URB597 (0.001/0.01/0.1/1nmol), or vehicle and were submitted to the FST, or to the open-field test. Another group received intra-vmPFC injection of SB366791 or vehicle, followed by a second injection of URB597 or vehicle, and was submitted to the FST. Lastly, a group received intra-vmPFC injection of a CB1 antagonist, in sub-effective dose or vehicle, followed by AA-5HT, SB366791 or vehicle. The results showed that AA-5HT, SB366791 and URB597 significantly reduced the immobility time without changing the locomotor activity. Furthermore, the co-administration of URB597 and SB366791 in sub-effective doses induced an antidepressant-like effect in the FST. Additionally, the antidepressant-like effect of AA-5HT was prevented by the CB1 antagonist. Together, these results suggest that both, CB1 and TRPV1 receptors located in the vmPFC are involved in the behavioral responses to stress, although in opposite ways.


Subject(s)
Adaptation, Psychological/physiology , Prefrontal Cortex/metabolism , Receptor, Cannabinoid, CB1/metabolism , Stress, Psychological/metabolism , TRPV Cation Channels/metabolism , Adaptation, Psychological/drug effects , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Anilides/pharmacology , Animals , Antidepressive Agents/pharmacology , Arachidonic Acids/pharmacology , Benzamides/pharmacology , Carbamates/pharmacology , Cinnamates/pharmacology , Male , Motor Activity/drug effects , Motor Activity/physiology , Neurotransmitter Agents/pharmacology , Prefrontal Cortex/drug effects , Rats, Wistar , Serotonin/analogs & derivatives , Serotonin/pharmacology , TRPV Cation Channels/antagonists & inhibitors
10.
Behav Brain Res ; 303: 218-27, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26801828

ABSTRACT

RATIONALE: Systemic administration of cannabidiol (CBD), the main non-psychotomimetic constituent of Cannabis sativa, induces antidepressant-like effects. The mechanism of action of CBD is thought to involve the activation of 5-HT1A receptors and the modulation of endocannabinoid levels with subsequent CB1 activation. The brain regions involved in CBD-induced antidepressant-like effects remain unknown. The ventral medial prefrontal cortex (vmPFC), which includes the infralimbic (IL) and prelimbic (PL) subregions, receives dense serotonergic innervation and plays a significant role in stress responses. OBJECTIVE: To test the hypothesis that the administration of CBD into the IL or PL would induce an antidepressant-like effect through 5-HT1A and CB1 activation. METHODS: Rats received intra-IL or -PL microinjections of CBD (10-60 nmol/side), 8-OH-DPAT (5-HT1A agonist, 5-10 nmol/side), anandamide (AEA, 0.5 pmol/side) or vehicle (0.2 µl/side) and were submitted to the forced swimming (FST) or to the open field (OFT) tests. Independent CBD-treated groups were pre-treated with WAY100635 (10, 30 nmol/side, 5-HT1A antagonist) or AM251 (10 pmol/side, CB1 antagonist) and submitted to the same tests. An additional group was treated with WAY100635 followed by anandamide. RESULTS: CBD (PL: 10-60 nmol; IL:45-60 nmol) and 8-OH-DPAT (10 nmol) administration significantly reduced the immobility time in the FST, without changing locomotor activity in the OFT. WAY100635 (30 nmol) did not induce effect per se but blocked CBD, 8-OH-DPAT and AEA effects. Additionally, AM251 blocked CBD-effects. CONCLUSION: administration of CBD into the vmPFC induces antidepressant-like effects possibly through indirect activation of CB1 and 5-HT1A receptors.


Subject(s)
Antidepressive Agents/administration & dosage , Behavior, Animal/drug effects , Cannabidiol/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Receptor, Cannabinoid, CB1/physiology , Receptor, Serotonin, 5-HT1A/physiology , 8-Hydroxy-2-(di-n-propylamino)tetralin/administration & dosage , Animals , Arachidonic Acids/administration & dosage , Endocannabinoids/administration & dosage , Male , Motor Activity/drug effects , Piperazines , Piperidines/administration & dosage , Polyunsaturated Alkamides/administration & dosage , Pyrazoles/administration & dosage , Pyridines , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Serotonin 5-HT1 Receptor Agonists/administration & dosage , Serotonin Antagonists/administration & dosage
11.
Genes Brain Behav ; 11(3): 303-13, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22142142

ABSTRACT

Nitric oxide (NO) is an atypical neurotransmitter that has been related to the pathophysiology of major depression disorder. Increased plasma NO levels have been reported in depressed and suicidal patients. Inhibition of neuronial nitric oxide synthase (nNOS), on the other hand, induces antidepressant effects in clinical and pre-clinical trials. The mechanisms responsible for the antidepressant-like effects of nNOS inhibitors, however, are not completely understood. In this study, genomic and proteomic analyses were used to investigate the effects of the preferential nNOS inhibitor 7-nitroindazole (7-NI) on changes in global gene and protein expression in the hippocampus of rats submitted to forced swimming test (FST). Chronic treatment (14 days, i.p.) with imipramine (15 mg/kg daily) or 7-NI (60 mg/kg daily) significantly reduced immobility in the FST. Saturation curves for Serial analysis of gene expression libraries showed that the hippocampus of animals submitted to FST presented a lower number of expressed genes compared to non-FST stressed groups. Imipramine, but not 7-NI, reverted this effect. GeneGo analyses revealed that genes related to oxidative phosphorylation, apoptosis and survival controlled by HTR1A signaling and cytoskeleton remodeling controlled by Rho GTPases were significantly changed by FST. 7-NI prevented this effect. In addition, 7-NI treatment changed the expression of genes related to transcription in the cAMP response element-binding pathway. Therefore, this study suggests that changes in oxidative stress and neuroplastic processes could be involved in the antidepressant-like effects induced by nNOS inhibition.


Subject(s)
Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Indazoles/pharmacology , Swimming , Animals , Depressive Disorder, Major/physiopathology , Disease Models, Animal , Gene Expression Regulation/physiology , Hippocampus/physiopathology , Imipramine/pharmacology , Male , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Rats , Rats, Wistar , Stress, Psychological/drug therapy , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Swimming/psychology
12.
J Neurosci Res ; 90(3): 632-40, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22006722

ABSTRACT

Transient global brain ischemia causes delayed neuronal death in the hippocampus that has been associated with impairments in hippocampus-dependent brain function, such as mood, learning, and memory. We investigated the expression of voltage-dependent Kcnh1 and Kcnh5, ether à go-go-related Eag1 and Eag2 (K(V) 10.1 and K(V) 10.2), and small-conductance calcium-activated SK3 (K(Ca) 2.3, Kcnn3) K(+) channels in the hippocampus in rats after transient global brain ischemia. We tested whether the expression of these channels is associated with behavioral changes by evaluating the animals in the elevated plus maze and step-down inhibitory avoidance task. Seven or tweny-eight days after transient global brain ischemia, one group of rats had the hippocampus bilaterally dissected, and mRNA levels were determined. Seven days after transient global brain ischemia, the rats exhibited a decrease in anxiety-like behavior and memory impairments. An increase in anxiety levels was detected 28 days after ischemia. Eag2 mRNA downregulation was observed in the hippocampus 7 days after transient global brain ischemia, whereas Eag1 and SK3 mRNA expression remained unaltered. This is the first experimental evidence that transient global brain ischemia temporarily alters Eag2. The number of intact-appearing pyramidal neurons was substantially decreased in CA1 and statistically measurable in CA2, CA3, and CA4 hippocampal subfields compared with sham control animals 7 or 28 days after ischemia. mRNA expression in the rat hippocampus. The present results provide further information for the characterization of the physiological role of Eag2 channels in the central nervous system.


Subject(s)
Brain Ischemia/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Hippocampus/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Avoidance Learning/physiology , Behavior, Animal/physiology , Brain Ischemia/physiopathology , Down-Regulation , Hippocampus/physiopathology , Male , Maze Learning/physiology , Neurons/metabolism , Rats , Rats, Wistar
13.
Genes Brain Behav ; 9(8): 918-24, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20662937

ABSTRACT

The Eag1 and Eag2, voltage-dependent potassium channels, and the small-conductance calcium-activated potassium channel (Kcnn3) are highly expressed in limbic regions of the brain, where their function is still unknown. Eag1 co-localizes with tyrosine hydroxilase enzyme in the substantia nigra and ventral tegmental area. Kcnn3 deficiency leads to enhanced serotonergic and dopaminergic neurotransmission accompanied by distinct alterations in emotional behaviors. As exposure to stress is able to change the expression and function of several ion channels, suggesting that they might be involved in the consequences of stress, we aimed at investigating Eag 1, Eag2 and Kcnn3 mRNA expression in the brains of rats submitted to isolation rearing. As the long-lasting alterations in emotional and behavioral regulation after stress have been related to changes in serotonergic neurotransmission, expressions of serotonin Htr1a and Htr2a receptors in male Wistar rats' brain were also investigated. Rats were reared in isolation or in groups of five for nine weeks after weaning. Isolated and socially reared rats were tested for exploratory activity in the open field test for 5 min and brains were processed for reverse-transcription coupled to quantitative polymerase chain reaction (qRT-PCR). Isolated reared rats showed decreased exploratory activity in the open field. Compared to socially reared rats, isolated rats showed reduced Htr2a mRNA expression in the striatum and brainstem and reduced Eag2 mRNA expression in all examined regions except cerebellum. To our knowledge, this is the first work to show that isolation rearing can change Eag2 gene expression in the brain. The involvement of this channel in stress-related behaviors is discussed.


Subject(s)
Brain/metabolism , Ether-A-Go-Go Potassium Channels/genetics , Receptor, Serotonin, 5-HT2A/genetics , Small-Conductance Calcium-Activated Potassium Channels/genetics , Social Isolation , Animals , Cerebral Cortex/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Exploratory Behavior/physiology , Gene Expression Regulation/physiology , Hippocampus/metabolism , Male , Mesencephalon/metabolism , Motor Activity/genetics , Motor Activity/physiology , Neostriatum/metabolism , Olfactory Bulb/metabolism , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT2A/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Tissue Distribution
14.
J Psychopharmacol ; 24(3): 397-405, 2010 Mar.
Article in English | MEDLINE | ID: mdl-18838497

ABSTRACT

Systemic or intra-striatal acute administration of nitric oxide synthase (NOS) inhibitors causes catalepsy in rodents. This effect disappears after sub-chronic treatment. The aim of the present study was to investigate if this tolerance is related to changes in the expression of NOS or dopamine-2 (D2) receptor or to a recovery of NOS activity. Male albino Swiss mice (25-30 g) received single or sub-chronic (once a day for 4 days) i.p. injections of saline or L-nitro-arginine (L-NOARG, 40 mg/kg), a non-selective inhibitor of neuronal nitric oxide synthase (nNOS). Twenty-four hours after the last injection, the animals were killed and their brains were removed for immunohistochemistry assay to detect the presence of nNOS or for 'in-situ' hybridisation study using (35)S-labeled oligonucleotide probe complementary to D2 receptor mRNA. The results were analysed by computerised densitometry. Independent groups of animals received the same treatment, but were submitted to the catalepsy test and had their brain removed to measure nitrite and nitrate (NOx) concentrations in the striatum. Acute administration of L-NOARG caused catalepsy that disappeared after sub-chronic treatment. The levels of NOx were significantly reduced after acute L-NOARG treatment. The decrease in NOx after drug injection suffered a partial tolerance after sub-chronic treatment. The catalepsy time after acute or sub-chronic treatment with L-NOARG was negatively (r = -0.717) correlated with NOx levels. Animals that received repeated L-NOARG injections also showed an increase in the number of nNOS-positive neurons in the striatum. No change in D2 receptor mRNA expression was found in the dorsal striatum, nucleus accumbens and substantia nigra. Together, these results suggest that tolerance to L-NOARG cataleptic effects do not depend on changes in D2 receptors. They may depend, however, on plastic changes in nNOS neurons resulting in partial recovery of NO formation in the striatum.


Subject(s)
Brain/enzymology , Catalepsy/metabolism , Drug Tolerance , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitroarginine/pharmacology , Animals , Brain/drug effects , Catalepsy/chemically induced , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Drug Administration Schedule , Enzyme Inhibitors/administration & dosage , Male , Mice , Nitric Oxide Synthase Type I/metabolism , Nitroarginine/administration & dosage , Reactive Nitrogen Species/metabolism , Receptors, Dopamine D2/metabolism
15.
Neuroscience ; 143(2): 377-85, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16973302

ABSTRACT

To explore the ventral medial prefrontal cortex (vMPFC) involvement in behavioral and autonomic fear-conditioned responses to context, vMPFC synaptic transmission was temporarily inhibited by bilateral microinjections of 200 nL of the nonselective synapse blocker CoCl(2) (1 mM). Behavioral activity (freezing, motor activity and rearing) as well as evoked cardiovascular responses (arterial pressure and heart rate) was analyzed. Rats were pre-exposed to the footshock chamber (context) and shock stimulus was used unconditioned stimulus. During re-exposure to context, conditioned rats spent 80% of the session in freezing while non-conditioned rats (no shock group) spent less than 15% of the session time in freezing. Conditioned rats had significantly lower activity scores than non-conditioned animals. Exposure to context increased mean arterial pressure (MAP) and heart rate (HR) of both groups. MAP and HR of the conditioned animals were markedly increased and remained at a high and stable level, whereas MAP and HR increases in non-conditioned animals were less pronounced and declined during the session. CoCl(2) microinjected in the vMPFC significantly reduced freezing and attenuated MAP and HR increase of the conditioned group. Cobalt-induced vMPFC inhibition also significantly reduced MAP and HR increase observed in non-conditioned animals, without any behavioral changes. The effect of vMPFC acute ablation on MAP and HR did not seem to be specific to the fear response because they were also evident in non-conditioned animals. The results indicate that vMPFC integrity is crucial for expression of fear-conditioned responses to context, such as freezing and cardiovascular changes, suggesting that fear-conditioned responses to context involve cortical processing prior to amygdalar output. They also indicate a cardiovascular response observed during re-exposure of non-conditioned rats to the context is completely dependent on vMPFC integrity.


Subject(s)
Behavior, Animal/physiology , Cardiovascular System , Conditioning, Psychological/physiology , Fear , Neurons/physiology , Prefrontal Cortex/cytology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Blood Pressure/drug effects , Cardiovascular System/drug effects , Cobalt/pharmacology , Conditioning, Psychological/drug effects , Electroshock/adverse effects , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Heart Rate/drug effects , Male , Microinjections/methods , Motor Activity/drug effects , Motor Activity/physiology , Neurons/drug effects , Rats , Rats, Wistar
16.
Eur Neuropsychopharmacol ; 10(4): 223-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10871703

ABSTRACT

It has been previously shown that oxcarbazepine (OXCBZ), a keto-analogue of carbamazepine, exhibits an antidepressive-like effect profile in the learned helplessness and forced swimming test (FST). Since carbamazepine possesses dopaminergic effect, the present study was carried out to evaluate the extent to which the antidepressive effect of OXCBZ might be mediated by dopaminergic system. Thus, the effects of OXCBZ in haloperidol-induced catalepsy and apomorphine-induced stereotypy were studied. The anti-immobility effect of OXCBZ in the FST was also evaluated in haloperidol pre-treated rats. OXCBZ (40 and 80 mg/kg, i.p.) dose-dependently reduced the catalepsy induced by haloperidol (2.0 mg/kg, i.p.). Moreover, OXCBZ (80 mg/kg, but not 20 or 40 mg/kg, i.p.) increased the intensity of apomorphine-induced stereotypy (0.6 mg/kg, s.c.). Finally, it was observed that the combination of OXCBZ (80 mg/kg, i. p.) and haloperidol (0.5 mg/kg, i.p.) antagonized the anti-immobility effect of OXCBZ and further increased the immobility time when compared to haloperidol alone. Haloperidol alone (0.5 or 1. 0 mg/kg) did not change the immobility time. Thus, these results suggest that OXCBZ could enhance dopaminergic neurotransmission, which might mediate its antidepressive-like effect.


Subject(s)
Antidepressive Agents/therapeutic use , Carbamazepine/analogs & derivatives , Carbamazepine/therapeutic use , Receptors, Dopamine/metabolism , Animals , Antidepressive Agents/pharmacology , Apomorphine , Carbamazepine/pharmacology , Catalepsy/chemically induced , Catalepsy/drug therapy , Disease Models, Animal , Dopamine Antagonists/therapeutic use , Haloperidol , Male , Neurotransmitter Agents , Oxcarbazepine , Rats , Rats, Wistar , Receptors, Dopamine/drug effects , Stereotypic Movement Disorder/chemically induced , Stereotypic Movement Disorder/drug therapy , Swimming/psychology
17.
Eur J Pharmacol ; 347(1): 23-7, 1998 Apr 17.
Article in English | MEDLINE | ID: mdl-9650844

ABSTRACT

The effect of oxcarbazepine was evaluated in two tests of depression (forced swimming and learned helplessness) and in the open-field test. Acute (three times over 24 h) oxcarbazepine 80 mg/kg (but not 40 mg/kg) decreased immobility time in the forced swimming test. In the learned helplessness test, 4 days of treatment with oxcarbazepine 80 mg/kg reversed the deficits induced by foot-shock in rats submitted to the two-way active avoidance test. Oxcarbazepine 80 mg/kg did not modify the behaviour of rats in the open-field test, an indication that, at this dose, oxcarbazepine did not show a locomotor stimulatory effect. Thus, the data of the present study suggest that oxcarbazepine has a potential antidepressive effect.


Subject(s)
Anticonvulsants/pharmacology , Carbamazepine/analogs & derivatives , Depression/drug therapy , Helplessness, Learned , Animals , Behavior, Animal/drug effects , Carbamazepine/pharmacology , Disease Models, Animal , Male , Motor Activity/drug effects , Oxcarbazepine , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...