Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Acta Neuropsychiatr ; 35(1): 35-49, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36101010

ABSTRACT

The Wistar Hannover rat (WHR) is a strain commonly used for toxicity studies but rarely used in studies investigating depression neurobiology. In this study, we aimed to characterise the behavioural responses of WHR to acute and repeated antidepressant treatments upon exposure to the forced swim test (FST) or learned helplessness (LH) test. WHR were subjected to forced swimming pre-test and test with antidepressant administration (imipramine, fluoxetine, or escitalopram) at 0, 5 h and 23 h after pre-test. WHR displayed high immobility in the test compared to unstressed controls (no pre-swim) and failed to respond to the antidepressants tested. The effect of acute and repeated treatment (imipramine, fluoxetine, escitalopram or s-ketamine) was then tested in animals not previously exposed to pre-test. Only imipramine (20 mg/kg, 7 days) and s-ketamine (acute) reduced the immobility time in the test. To further investigate the possibility that the WHR were less responsive to selective serotonin reuptake inhibitors, the effect of repeated treatment with fluoxetine (20 mg/kg, 7 days) was investigated in the LH model. The results demonstrated that fluoxetine failed to reduce the number of escape failures in two different protocols. These data suggest that the WHR do not respond to the conventional antidepressant treatment in the FST or the LH. Only s-ketamine and repeated imipramine were effective in WHR in a modified FST protocol. Altogether, these results indicate that WHR may be an interesting tool to investigate the mechanisms associated with the resistance to antidepressant drugs and identify more effective treatments.


Subject(s)
Fluoxetine , Imipramine , Rats , Animals , Fluoxetine/pharmacology , Rats, Wistar , Imipramine/pharmacology , Imipramine/therapeutic use , Depression/drug therapy , Escitalopram , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Swimming , Behavior, Animal , Models, Animal
2.
Cannabis Cannabinoid Res ; 8(5): 768-778, 2023 10.
Article in English | MEDLINE | ID: mdl-36067014

ABSTRACT

Introduction: The antidepressant properties of ketamine have been extensively demonstrated in experimental and clinical settings. However, the psychotomimetic side effects still limit its wider use as an antidepressant. It was recently observed that endocannabinoids are inolved in ketamine induced reward properties. As an increase in endocannabinoid signaling induces antidepressant effects, this study aimed to investigate the involvement of cannabinoid type 1 receptors (CB1R) in the antidepressant and psychostimulant effects induced by ketamine. Methods: We tested the effects of genetic and pharmacological inhibition of CB1R in the hyperlocomotion and antidepressant-like properties of ketamine. The effects of ketamine (10-20 mg/kg) were assessed in the open-field and the forced swim tests (FSTs) in CB1R knockout (KO) and wild-type (WT) mice (male and female), and mice pre-treated with rimonabant (CB1R antagonist, 3-10 mg/kg). Results: We found that the motor hyperactivity elicited by ketamine was impaired in CB1R male and female KO mice. A similar effect was observed upon pharmacological blockade of CB1R in WT mice. However, genetic CB1R deletion did not modify the antidepressant effect of ketamine in male mice submitted to the FST. Surprisingly, pharmacological blockade of CB1R induced an antidepressant-like effect in both male and female mice, which was not further potentiated by ketamine. Conclusions: Our results support the hypothesis that CB1R mediate the psychostimulant side effects induced by ketamine, but not its antidepressant properties.


Subject(s)
Central Nervous System Stimulants , Ketamine , Mice , Male , Female , Animals , Ketamine/pharmacology , Receptor, Cannabinoid, CB1/genetics , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Rimonabant/pharmacology
3.
Front Psychiatry ; 13: 923177, 2022.
Article in English | MEDLINE | ID: mdl-35911236

ABSTRACT

Stress exposure can result in several proinflammatory alterations in the brain, including overexpression of the inducible isoform of nitric oxide synthase (iNOS) in the medial prefrontal cortex (mPFC). These changes may be involved in the development of many psychiatric conditions. However, it is unknown if iNOS in mPFC plays a significant role in stress-induced behavioral changes. The endocannabinoid (ECB) system is also influenced by stress. Its activation seems to be a counter regulatory mechanism to prevent or decrease the stress-mediated neuroinflammatory consequences. However, it is unclear if the ECB system and iNOS interact to influence stress consequences. This study aimed to test the hypothesis that the anti-stress effect of iNOS inhibition in mPFC involves the local ECB system, particularly the CB1 cannabinoid receptors. Male Wistar rats with guide cannula aimed at the mPFC were submitted to acute restraint stress (RS) for 2 h. In the following morning, rats received bilateral microinjections of vehicle, AM251 (CB1 antagonist; 100 pmol), and/or 1400W (iNOS selective inhibitor; 10-4, 10-3, or 10-2 nmol) into the prelimbic area of mPFC (PL-mPFC) before being tested in the elevated plus-maze (EPM). iNOS inhibition by 1400W prevented the anxiogenic-like effect observed in animals submitted to RS. The drug did not promote behavior changes in naive animals, demonstrating a stress-dependent effect. The 1400W-anti-stress effect was prevented by local pretreatment with AM251. Our data suggest that iNOS inhibition may facilitate the local endocannabinoid signaling, attenuating stress effects.

4.
J Chem Neuroanat ; 98: 104-116, 2019 07.
Article in English | MEDLINE | ID: mdl-31039391

ABSTRACT

Significant limitations with the currently available antidepressant treatment strategies have inspired research on finding new and more efficient drugs to treat depression. Cannabidiol (CBD) is a non-psychotomimetic component of Cannabis sativa, and emerges in this regard as a promising compound. In 2010, we were the first laboratory to demonstrate that CBD is effective in animal models of predictive of antidepressant effect, a finding now confirmed by several other groups. Recent evidence suggests that CBD promotes both a rapid and a sustained antidepressant effect in animal models. CBD has a complex pharmacology, with the ability to interact with multiple neurotransmitter systems involved in depression, including the serotonergic, glutamatergic, and endocannabinoid systems. Moreover, CBD induces cellular and molecular changes in brain regions related to depression neurobiology, such as increased Brain Derived Neurotrophic Factor (BDNF) levels and synaptogenesis in the medial prefrontal cortex, as well as it increases neurogenesis in the hippocampus. This review presents a comprehensive critical overview of the current literature related to the antidepressant effects of CBD, with focus at the possible mechanisms. Finally, challenges and perspectives for future research are discussed.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , Cannabidiol/pharmacology , Animals , Humans
5.
Cell Tissue Res ; 377(1): 45-58, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30649612

ABSTRACT

Studies about the pathogenesis of mood disorders have consistently shown that multiple factors, including genetic and environmental, play a crucial role on their development and neurobiology. Multiple pathological theories have been proposed, of which several ultimately affects or is a consequence of dysfunction in brain neuroplasticity and homeostatic mechanisms. However, current clinical available pharmacological intervention, which is predominantly monoamine-based, suffers from a partial and lacking response even after weeks of continuous treatment. These issues raise the need for better understanding of aetiologies and brain abnormalities in depression, as well as developing novel treatment strategies. Nitric oxide (NO) is a gaseous unconventional neurotransmitter, which regulates and governs several important physiological functions in the central nervous system, including processes, which can be associated with the development of mood disorders. This review will present general aspects of the NO system in depression, highlighting potential targets that may be utilized and further explored as novel therapeutic targets in the future pharmacotherapy of depression. In particular, the review will link the importance of neuroplasticity mechanisms governed by NO to a possible molecular basis for the antidepressant effects.


Subject(s)
Antidepressive Agents/pharmacology , Brain , Mood Disorders/drug therapy , Mood Disorders/metabolism , Neurotransmitter Agents/pharmacology , Nitric Oxide/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Humans , Mice , Neuronal Plasticity , Rats , Signal Transduction
6.
Mol Neurobiol ; 56(2): 1070-1081, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29869197

ABSTRACT

Currently available antidepressants have a substantial time lag to induce therapeutic response and a relatively low efficacy. The development of drugs that addresses these limitations is critical to improving public health. Cannabidiol (CBD), a non-psychotomimetic component of Cannabis sativa, is a promising compound since it shows large-spectrum therapeutic potential in preclinical models and humans. However, its antidepressant properties have not been completely investigated. Therefore, the aims of this study were to investigate in male rodents (i) whether CBD could induce rapid and sustained antidepressant-like effects after a single administration and (ii) whether such effects could be related to changes in synaptic proteins/function. Results showed that a single dose of CBD dose-dependently induced antidepressant-like effect (7-30 mg/kg) in Swiss mice submitted to the forced swim test (FST), 30 min (acute) or 7 days (sustained) following treatment. Similar effects were observed in the Flinders Sensitive and Flinders Resistant Line (FSL/FRL) rats and the learned helplessness (LH) paradigm using Wistar rats. The acute antidepressant effects (30 min) were associated with increased expression of synaptophysin and PSD95 in the medial prefrontal cortex (mPFC) and elevated BDNF levels in both mPFC and hippocampus (HPC). CBD also increased spine density in the mPFC after 30 min, but not 7 days later. Intracerebroventricular injection of the TrkB antagonist, K252a (0.05 nmol/µL), or the mTOR inhibitor, rapamycin (1 nmol/µL), abolished the behavioral effects of CBD. These results indicate that CBD induces fast and sustained antidepressant-like effect in distinct animal models relevant for depression. These effects may be related to rapid changes in synaptic plasticity in the mPFC through activation of the BDNF-TrkB signaling pathway. The data support a promising therapeutic profile for CBD as a new fast-acting antidepressant drug.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/drug effects , Cannabidiol/pharmacology , Prefrontal Cortex/drug effects , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Hippocampus/metabolism , Male , Mice , Prefrontal Cortex/metabolism , Signal Transduction/drug effects
7.
Behav Pharmacol ; 30(1): 59-66, 2019 02.
Article in English | MEDLINE | ID: mdl-30299277

ABSTRACT

The transient receptor potential vanilloid 1 (TRPV1) can modulate stress-related behaviours, thus representing an interesting target for new antidepressant drugs. TRPV1 can trigger glutamate release and nitric oxide synthesis in the brain, mechanisms also involved in the neurobiology of depression. However, it is not known if these mechanisms are involved in TRPV1-induced behavioural effects. Therefore, the aim of this study was to verify if the antidepressant-like effect induced by a TRPV1 antagonist in mice submitted to the forced swimming test (FST) would be facilitated by combined treatment with neuronal nitric oxide synthase (nNOS) inhibition and N-methyl-D-aspartate (NMDA) blockade. Male Swiss mice were given (intracerebroventricular) injections of capsazepine (CPZ) (TRPV1 antagonist - 0.05/0.1/0.3/0.6 nmol/µl), and AP7 (NMDA antagonist - 1/3/10 nmol/µl) or N-propyl-L-arginine (NPA, nNOS inhibitor - 0.001/0.01/0.1 nmol/µl), and 10 min later, submitted to an open field test, and immediately afterwards, to the FST. An additional group received coadministration of CPZ and AP7 or CPZ and NPA, in subeffective doses. The results demonstrated that CPZ (0.1 nmol/µl), AP7 (3 nmol/µl) and NPA (0.01/0.1 nmol/µl) induced antidepressant-like effects. Moreover, coadministration of subeffective doses of CPZ and AP7 or CPZ and NPA induced significant antidepressant-like effects. Altogether, the data indicate that blockade of TRPV1 receptors by CPZ induces antidepressant-like effects and that both nNOS inhibition and NMDA blockade facilitate CPZ effects in the FST.


Subject(s)
Antidepressive Agents/therapeutic use , Capsaicin/analogs & derivatives , Depression/drug therapy , Glutamic Acid/metabolism , Nitric Oxide/metabolism , Swimming/psychology , 2-Amino-5-phosphonovalerate/analogs & derivatives , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Apomorphine/analogs & derivatives , Apomorphine/pharmacology , Arginine/pharmacology , Capsaicin/therapeutic use , Cyclic GMP/metabolism , Depression/metabolism , Depression/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Enzyme Inhibitors/pharmacology , Exploratory Behavior/drug effects , Male , Microinjections , Nitroprusside/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Statistics, Nonparametric
8.
Neuropharmacology ; 144: 233-243, 2019 01.
Article in English | MEDLINE | ID: mdl-30385254

ABSTRACT

Previous clinical and pre-clinical studies suggest the involvement of ventromedial orbitofrontal cortex (vmOFC) and glutamatergic neurotransmission in obsessive-compulsive disorder (OCD). Ketamine, an NMDA glutamatergic receptor antagonist, has shown a rapid and long-lasting antidepressant effect, but its anti-compulsive effect has been scarcely investigated. The antidepressant effect of ketamine involves NMDA receptor blockade, AMPA receptor activation, increased serotonin (5-HT) release and attenuation of nitric oxide (NO) synthesis. It is not known if these mechanisms are involved in ketamine-induced anti-compulsive effect. Therefore, we firstly investigated the effect of S-ketamine in the marble-burying test (MBT), a model for screening of drugs with potential to treat OCD. Then, we evaluated whether ketamine effects in the MBT would involve the vmOFC, be dependent on AMPA receptors, facilitation of serotonergic neurotransmission and inhibition of nitrergic pathway. Our results showed that single systemic (10 mg/kg) and intra-vmOFC (10 nmol/side) administration of S-ketamine reduces marble burying behaviour (MBB) without affecting spontaneous locomotors activity. Pre-treatment with NBQX (3 mg/kg; AMPA receptor antagonist) blocked the reduction of MBB induced by S-ketamine. However, pre-treatment with p-CPA (150 mg/kg/day; a 5-HT synthesis inhibitor), WAY100635 (3 mg/kg; a 5-HT1A receptor antagonist), or L-arginine (500 mg/kg; a nitric oxide precursor) did not counteract S-ketamine effect in the MBT. In contrast, associating sub-effective doses of L-NAME (10 mg/kg; NOS inhibitor) and S-ketamine (3 mg/kg) decreased MBB. In conclusion, the reduction of MBB by S-ketamine strengthens its possible anti-compulsive effect. The vmOFC is involved in this S-ketamine effect, which is dependent on the activation of AMPA receptors.


Subject(s)
Ketamine/pharmacology , Obsessive-Compulsive Disorder/drug therapy , Prefrontal Cortex/drug effects , Psychotropic Drugs/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Drug Evaluation, Preclinical , Male , Mice , Motor Activity/drug effects , Obsessive-Compulsive Disorder/metabolism , Prefrontal Cortex/metabolism , Random Allocation , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
9.
J Psychopharmacol ; 32(8): 922-931, 2018 08.
Article in English | MEDLINE | ID: mdl-29968502

ABSTRACT

BACKGROUND: Cannabidiol is a non-psychotomimetic compound with antidepressant-like effects. However, the mechanisms and brain regions involved in cannabidiol effects are not yet completely understood. Brain-derived neurotrophic factor/tropomyosin-receptor kinase B/mammalian target of rapamycin (BDNF-TrkB-mTOR) signaling, especially in limbic structures, seems to play a central role in mediating the effects of antidepressant drugs. AIM: Since it is not yet known if BDNF-TrkB-mTOR signaling in the hippocampus is critical to the antidepressant-like effects of cannabidiol, we investigated the effects produced by cannabidiol (10/30/60 nmol/0.2 µL) micro-injection into the hippocampus of mice submitted to the forced swim test and to the open field test. METHODS: Independent groups received intra-hippocampal injections of rapamycin (mTOR inhibitor, 0.2 nmol/0.2 µL) or K252 (Trk antagonist, 0.01 nmol/0.2 µL), before the systemic (10 mg/kg) or hippocampal (10 nmol/0.2µL) injection of cannabidiol, and were submitted to the same tests. BDNF levels were analyzed in the hippocampus of animals treated with cannabidiol (10 mg/kg). RESULTS: Systemic cannabidiol administration induced antidepressant-like effects and increased BDNF levels in the dorsal hippocampus. Rapamycin, but not K252a, injection into the dorsal hippocampus prevented the antidepressant-like effect induced by systemic cannabidiol treatment (10 mg/kg). Differently, hippocampal administration of cannabidiol (10 nmol/0.2 µL) reduced immobility time, an effect that was blocked by both rapamycin and K252a local microinjection. CONCLUSION: Altogether, our data suggest that the hippocampal BDNF-TrkB-mTOR pathway is vital for cannabidiol-induced antidepressant-like effect when the drug is locally administered. However, other brain regions may also be involved in cannabidiol-induced antidepressant effect upon systemic administration.


Subject(s)
Behavior, Animal/drug effects , Cannabidiol/adverse effects , Hippocampus/drug effects , Hippocampus/metabolism , Stress, Psychological/chemically induced , Stress, Psychological/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Carbazoles/pharmacology , Depression/chemically induced , Depression/drug therapy , Depression/metabolism , Indole Alkaloids/pharmacology , Male , Mice , Receptor, trkB/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Swimming
10.
Eur Neuropsychopharmacol ; 23(12): 1769-78, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23434291

ABSTRACT

Activation of purinergic receptors by ATP (P2R) modulates glutamate release and the activation of post-synaptic P2R is speculated to induce nitric oxide (NO) synthesis. Increased glutamatergic and nitrergic signaling have been involved in the neurobiology of stress-related psychiatric disorders such as anxiety and depression. Therefore, the aim of this study was to test the effects of two P2R antagonists (PPADS and iso-PPADS) in animals submitted to models predictive of antidepressant-, anxiolytic- and anticompulsive-like effects. Swiss mice receiving PPADS at 12.5mg/kg showed reduced immobility time in the forced swimming test (FST) similarly to the prototype antidepressant imipramine (30mg/kg). This dose was also able to decrease the number of buried marbles in the marble-burying test (MBT), an anticompulsive-like effect. However, no effect was observed in animals submitted to the elevated plus maze (EPM) and to the open field test. The systemic administration of iso-PPADS, a preferential P2XR antagonist, also reduced the immobility time in FST, which was associated to a decrease in NOx levels in the prefrontal cortex. In addition, P2X7 receptor was found co-immunoprecipitated with neuronal nitric oxide synthase (NOS1) in the prefrontal cortex. These results suggest that P2X7, possibly coupled to NOS1, could modulate behavioral responses associated to stress-related disorders and it could be a new target for the development of more effective treatments for affective disorders.


Subject(s)
Antidepressive Agents/therapeutic use , Compulsive Behavior/drug therapy , Depression/drug therapy , Nitric Oxide/metabolism , Purinergic Antagonists/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Immobility Response, Tonic/drug effects , Immobility Response, Tonic/physiology , Male , Maze Learning/drug effects , Nitric Oxide Synthase Type II/metabolism , Paroxetine/pharmacology , Paroxetine/therapeutic use , Purinergic Antagonists/pharmacology , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Pyridoxal Phosphate/therapeutic use , Rats , Receptors, Purinergic/metabolism , Swimming/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...