Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
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.
Neurochem Int ; 125: 7-15, 2019 05.
Article in English | MEDLINE | ID: mdl-30716357

ABSTRACT

Altered endocannabinoid (eCB) signalling is suggested as an important contributor to the pathophysiology of depression. To further elucidate this, we conducted a study using a genetic rat model of depression, the Flinders Sensitive Line (FSL), and their controls, the Flinders Resistant Line (FRL) rats. Plasma, right and left prefrontal cortex, and hippocampus were isolated from FSL and FRL rats. We analyzed each region for the eCB anandamide (AEA) and 2-arachidonoylglycerol (2-AG) levels by liquid chromatography/multiple reaction monitoring (LC/MRM), mRNA and protein levels of the cannabinoid type 1 receptor (CB1R), fatty acid amide hydrolase (FAAH) and monoacyl glycerol lipase (MAGL) by real time qPCR and Western blotting. Content of 2-AG was lower in the left side of the hippocampus and prefrontal cortex in FSL rats compared to FRL rats. Inversely, levels of AEA were higher in right hippocampus than in left hippocampus. In plasma, AEA levels were increased and 2-AG decreased. Cannabinoid receptor 1 (Cnr1), Faah and Magl mRNA levels were prominently decreased in right prefrontal cortex of FSL rats as compared to FRL rats. Protein expression of CB1R and FAAH were decreased in left hippocampus. In summary, our data suggest a decreased eCB signalling in the FSL rats, which could contribute to the depressive-like behaviour. Interestingly, the altered eCB system activity appear to be hemisphere-specific in the limbic regions. Our study support the existing literature and showed altered eCB system activity in this particular animal model of depression.


Subject(s)
Depression/metabolism , Endocannabinoids/metabolism , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Animals , Cerebrum/metabolism , Depression/genetics , Male , Rats , Rats, Transgenic , Species Specificity
3.
Vitam Horm ; 103: 193-279, 2017.
Article in English | MEDLINE | ID: mdl-28061971

ABSTRACT

The medical properties of Cannabis sativa is known for centuries. Since the discovery and characterization of the endogenous cannabinoid system, several studies have evaluated how cannabinoid compounds and, particularly, how the modulation of the endocannabinoid (eCB) system influences a wide range of functions, from metabolic to mental disorders. Cannabinoids and eCB system often exert opposite effects on several functions, such as anxiety. Although the mechanisms are not completely understood, evidence points to different factors influencing those effects. In this chapter, the recent advances in research about the relationship between eCB system and anxiety disorders in humans, as well as in animal models, will be discussed. The recent data addressing modulation of the eCBs in specific brain areas, such as the medial prefrontal cortex, amygdaloid complex, bed nucleus of stria terminalis, hippocampus, and dorsal periaqueductal gray, will be summarized. Finally, data from animal models addressing the mechanisms through which the eCB system modulates anxiety-related behavior dependent on stressful situations, such as the involvement of different receptors, distinct eCBs, modulation of neurotransmitters release, HPA axis and immune system activation, and plastic mechanisms, will also be discussed.


Subject(s)
Anxiety Disorders/metabolism , Anxiety/metabolism , Brain/metabolism , Endocannabinoids/metabolism , Neurons/metabolism , Neuroprotection , Receptors, Cannabinoid/metabolism , Animals , Anxiety/genetics , Anxiety/immunology , Anxiety Disorders/genetics , Anxiety Disorders/immunology , Brain/immunology , Endocannabinoids/immunology , Fear , Genetic Predisposition to Disease , Humans , Hypothalamo-Hypophyseal System/immunology , Hypothalamo-Hypophyseal System/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuronal Plasticity , Neurons/immunology , Organ Specificity , Pituitary-Adrenal System/immunology , Pituitary-Adrenal System/metabolism , Polymorphism, Single Nucleotide , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/genetics
4.
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
5.
Eur Neuropsychopharmacol ; 26(1): 15-22, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26628106

ABSTRACT

2-arachidonoylglycerol (2-AG) is an endogenous ligand of the cannabinoid CB1 receptor. This endocannabinoid and its hydrolyzing enzyme, monoacylglycerol lipase (MAGL), are present in encephalic regions related to psychiatric disorders, including the midbrain dorsolateral periaqueductal grey (dlPAG). The dlPAG is implicated in panic disorder and its stimulation results in defensive responses proposed as a model of panic attacks. The present work verified if facilitation of 2-AG signalling in the dlPAG counteracts panic-like responses induced by local chemical stimulation. Intra-dlPAG injection of 2-AG prevented panic-like response induced by the excitatory amino acid N-methyl-d-aspartate (NMDA). This effect was mimicked by the 2-AG hydrolysis inhibitor (MAGL preferring inhibitor) URB602. The anti-aversive effect of URB602 was reversed by the CB1 receptor antagonist, AM251. Additionally, a combination of sub-effective doses of 2-AG and URB602 also prevented NMDA-induced panic-like response. Finally, immunofluorescence assay showed a significant increase in c-Fos positive cells in the dlPAG after local administration of NMDA. This response was also prevented by URB602. These data support the hypothesis that 2-AG participates in anti-aversive mechanisms in the dlPAG and reinforce the proposal that facilitation of endocannabinoid signalling could be a putative target for developing additional treatments against panic and other anxiety-related disorders.


Subject(s)
Arachidonic Acids/metabolism , Endocannabinoids/metabolism , Glycerides/metabolism , Panic Disorder/drug therapy , Panic Disorder/metabolism , Periaqueductal Gray/drug effects , Periaqueductal Gray/metabolism , Animals , Biphenyl Compounds/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Male , N-Methylaspartate , Panic Disorder/pathology , Periaqueductal Gray/pathology , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism
6.
Neuroscience ; 285: 60-9, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25446344

ABSTRACT

Liposomes are nanosystems that allow a sustained release of entrapped substances. Gamma-aminobutyric acid (GABA) is the most prevalent inhibitory neurotransmitter of the central nervous system (CNS). We developed a liposomal formulation of GABA for application in long-term CNS functional studies. Two days after liposome-entrapped GABA was injected intracerebroventricularly (ICV), Wistar rats were submitted to the following evaluations: (1) changes in mean arterial pressure (MAP), heart rate (HR) and renal sympathetic nerve activity (RSNA) to ICV injection of bicuculline methiodide (BMI) in anesthetized rats; (2) changes in cardiovascular reactivity to air jet stress in conscious rats; and (3) anxiety-like behavior in conscious rats. GABA and saline-containing pegylated liposomes were prepared with a mean diameter of 200 nm. Rats with implanted cannulas targeted to lateral cerebral ventricle (n = 5-8/group) received either GABA solution (GS), empty liposomes (EL) or GABA-containing liposomes (GL). Following (48 h) central microinjection (2 µL, 0.09 M and 99 g/L) of liposomes, animals were submitted to the different protocols. Animals that received GL demonstrated attenuated response of RSNA to BMI microinjection (GS 48 ± 9, EL 43 ± 9, GL 11 ± 8%; P < 0.05), blunted tachycardia in the stress trial (ΔHR: GS 115 ± 14, EL 117 ± 10, GL 74 ± 9 bpm; P<0.05) and spent more time in the open arms of elevated plus maze (EL 6 ± 2 vs. GL 18 ± 5%; P = 0.028) compared with GS and EL groups. These results indicate that liposome-entrapped GABA can be a potential tool for exploring the chronic effects of GABA in specific regions and pathways of the central nervous system.


Subject(s)
Cardiovascular Agents/administration & dosage , Central Nervous System Agents/administration & dosage , GABA Agents/administration & dosage , Liposomes/administration & dosage , gamma-Aminobutyric Acid/administration & dosage , Animals , Anxiety/drug therapy , Anxiety/physiopathology , Arterial Pressure/drug effects , Bicuculline/administration & dosage , Bicuculline/analogs & derivatives , Catheters, Indwelling , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Heart Rate/drug effects , Infusions, Intraventricular , Kidney/innervation , Male , Microinjections , Rats, Wistar , Stress, Physiological/drug effects , Stress, Physiological/physiology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Tachycardia/drug therapy , Tachycardia/physiopathology
7.
Neurosci Biobehav Rev ; 46 Pt 3: 418-28, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24726577

ABSTRACT

The Transient Receptor Potential Vanilloid Type-1 (TRPV1) was first characterized in primary afferent fibers as a receptor for capsaicin (the pungent ingredient of chili peppers). Later on, this cation-permeable ion channel was also described in the central nervous system, where its main putative endogenous ligand is N-arachidonoyl ethanolamide (an endocannabinoid, also known as anandamide). Recent results employing genetic, pharmacological and histochemical techniques indicate that TRPV1 tonically modulate anxiety, fear and panic responses in brain regions related to defensive responses, such as the dorsal periaqueductal gray, the hippocampus and the medial prefrontal cortex. Genetic deletion or antagonism of this ion channel induces anxiolytic-like effects in several animal models. The main mechanism responsible for TRPV1-mediated effects on anxiety seems to involve facilitation of glutamatergic neurotransmission. In addition, there is evidence for interactions with other neurotransmitter systems, such as nitric oxide and endocannabinoids.


Subject(s)
Brain/metabolism , Defense Mechanisms , TRPV Cation Channels/physiology , Animals , Anxiety/drug therapy , Anxiety/pathology , Disease Models, Animal , Humans , Models, Biological
8.
Behav Brain Res ; 252: 10-7, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23714073

ABSTRACT

Anandamide and 2-arachidonoylglycerol (2-AG) are the two main endocannabinoids, exerting their effects by activating type 1 (CB1r) and type 2 (CB2r) cannabinoid receptors. Anandamide inhibits anxiety-like responses through the activation of CB1r in certain brain regions, including the dorsolateral periaqueductal gray (dlPAG). 2-AG also attenuates anxiety-like responses, although the neuroanatomical sites for these effects remained unclear. Here, we tested the hypothesis that enhancing 2-AG signaling in the dlPAG would induce anxiolytic-like effects. The mechanisms involved were also investigated. Male Wistar rats received intra-dlPAG injections of 2-AG, URB602 (inhibitor of the 2-AG hydrolyzing enzyme, mono-acylglycerol lipase--MGL), AM251 (CB1r antagonist) and AM630 (CB2r antagonist). The behavior was analyzed in the elevated plus maze after the following treatments. Exp. 1: vehicle (veh) or 2-AG (5 pmol, 50 pmol, and 500 pmol). Exp. 2: veh or URB602 (30 pmol, 100 pmol or 300 pmol). Exp. 3: veh or AM251 (100 pmol) followed by veh or 2-AG (50 pmol). Exp. 4: veh or AM630 (1000 pmol) followed by veh or 2-AG. Exp. 5: veh or AM251 followed by veh or URB602 (100 pmol). Exp. 6: veh or AM630 followed by veh or URB602. 2-AG (50 pmol) and URB602 (100 pmol) significantly increased the exploration of the open arms of the apparatus, indicating an anxiolytic-like effect. These behavioral responses were prevented by CB1r (AM251) or CB2r (AM630) antagonists. Our results showed that the augmentation of 2-AG levels in the dlPAG induces anxiolytic-like effects. The mechanism seems to involve both CB1r and CB2r receptors.


Subject(s)
Anxiety/chemically induced , Arachidonic Acids/metabolism , Arachidonic Acids/toxicity , Biphenyl Compounds/toxicity , Cannabinoid Receptor Agonists/toxicity , Endocannabinoids/metabolism , Endocannabinoids/toxicity , Glycerides/metabolism , Glycerides/toxicity , Periaqueductal Gray/drug effects , Analysis of Variance , Animals , Cannabinoid Receptor Antagonists , Disease Models, Animal , Dose-Response Relationship, Drug , Indoles/pharmacology , Male , Maze Learning/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar
9.
Pharmacol Biochem Behav ; 105: 166-72, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23474373

ABSTRACT

The transient receptors potential vanilloid type 1 channels (TRPV1) are expressed in several brain regions related to defensive behaviors, including the dorsolateral periaqueductal gray (dlPAG). The endocannabinoid anandamide, in addition to its agonist activity at cannabinoid type 1 (CB1), is also proposed as an endogenous agonist of these receptors, through which it could facilitate anxiety-like responses. The aim of this work was to test the hypothesis that TRPV1 in the dlPAG of rats would mediate panic-like responses in two models, namely the escape responses induced by chemical stimulation of this structure or by exposure to the elevated T-Maze (ETM). Antagonism of TRPV1 with capsazepine injected into the dlPAG reduced the defense response induced by local NMDA-injection, suggesting an anti-aversive effect. In the ETM, capsazepine inhibited escape response, suggesting a panicolytic-like effect. Interestingly, this effect was prevented by a CB1 antagonist (AM251). The present study showed that antagonism of TRPV1 in the dlPAG induces panicolytic-like effects, which can be prevented by a CB1 antagonist. Therefore, these antiaversive effects of TRPV1 blockade may ultimately occur due to a predominant action of anandamide through CB1 receptors.


Subject(s)
Panic Disorder/physiopathology , Periaqueductal Gray/physiopathology , TRPV Cation Channels/physiology , Animals , Male , Rats , Rats, Wistar
10.
Neuropharmacology ; 65: 83-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23000076

ABSTRACT

The endocannabinoid system (ECS) may either enhance or inhibit responses to aversive stimuli, possibly caused by its modulatory activity on diverse neurotransmitters. The aim of this work was to investigate the involvement of serotonin (5-HT) and catecholamines, as well as the role of glutamatergic and GABAergic cannabinoid type 1 (CB(1)) receptor, in responses to the antidepressant-like doses of the CB(1) receptor agonist Δ(9)-tetrahydrocannabinol (THC) and the antagonist rimonabant in the forced swim test (FST). Mice received acute injections of low doses of THC (0.1 or 0.5 mg/kg) or high dose of rimonabant (3 or 10 mg/kg) after treatment with the 5-HT synthesis inhibitor pCPA (100 mg/kg, 4 days), the 5-HT(1A) receptor antagonist WAY100635 (1 mg/kg, acute) or the non-selective blocker of catecholamine synthesis, AMPT (20 mg/kg, acute). THC and rimonabant were also tested in mutant mice lacking CB(1) receptor in specific forebrain neuronal subpopulations. Both THC and rimonabant induced antidepressant-like effects, quantified as immobility in the FST. However, only THC effects were reversed by pCPA or WAY100635. In contrast, only AMPT could attenuate the rimonabant effect. We also found decreased immobility in mice lacking the CB(1) receptor in glutamatergic cortical neurons, but not in forebrain GABAergic neurons, as compared with wild-type controls. The effect of THC persisted in mutant mice with CB(1) receptor inactivation in GABAergic neurons, whereas rimonabant effects were alleviated in these mutants. Thus, employing both pharmacological and genetic tools, we could show that the ECS regulates stress responses by influencing GABAergic, glutamatergic and monoaminergic transmission. The antidepressant-like action of THC depends on serotonergic neurotransmission, whereas rimonabant effects are mediated by CB(1) receptor on GABAergic neurons and by catecholamine signaling.


Subject(s)
Adaptation, Psychological/physiology , Neurons/physiology , Prosencephalon/physiology , Receptor, Cannabinoid, CB1/physiology , Serotonin/physiology , Stress, Psychological/drug therapy , Adaptation, Psychological/drug effects , Animals , Catecholamines/physiology , Dronabinol/pharmacology , Dronabinol/therapeutic use , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Piperidines/pharmacology , Prosencephalon/drug effects , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Rimonabant , Stress, Psychological/psychology
11.
Braz. j. med. biol. res ; 45(4): 357-365, Apr. 2012. tab
Article in English | LILACS | ID: lil-622757

ABSTRACT

This paper presents an up-to-date review of the evidence indicating that atypical neurotransmitters such as nitric oxide (NO) and endocannabinoids (eCBs) play an important role in the regulation of aversive responses in the periaqueductal gray (PAG). Among the results supporting this role, several studies have shown that inhibitors of neuronal NO synthase or cannabinoid receptor type 1 (CB1) receptor agonists cause clear anxiolytic responses when injected into this region. The nitrergic and eCB systems can regulate the activity of classical neurotransmitters such as glutamate and γ-aminobutyric acid (GABA) that control PAG activity. We propose that they exert a ‘fine-tuning’ regulatory control of defensive responses in this area. This control, however, is probably complex, which may explain the usually bell-shaped dose-response curves observed with drugs that act on NO- or CB1-mediated neurotransmission. Even if the mechanisms responsible for this complex interaction are still poorly understood, they are beginning to be recognized. For example, activation of transient receptor potential vanilloid type-1 channel (TRPV1) receptors by anandamide seems to counteract the anxiolytic effects induced by CB1 receptor activation caused by this compound. Further studies, however, are needed to identify other mechanisms responsible for this fine-tuning effect.


Subject(s)
Animals , Mice , Rats , Anxiety/physiopathology , Escape Reaction/physiology , Neurotransmitter Agents/physiology , Periaqueductal Gray/physiology , Synaptic Transmission/physiology , Anxiety/metabolism , Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Endocannabinoids/pharmacology , Endocannabinoids/physiology , Nitric Oxide/physiology , Periaqueductal Gray/metabolism , Polyunsaturated Alkamides/pharmacology , TRPV Cation Channels/physiology
12.
Braz J Med Biol Res ; 45(4): 357-65, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22392189

ABSTRACT

This paper presents an up-to-date review of the evidence indicating that atypical neurotransmitters such as nitric oxide (NO) and endocannabinoids (eCBs) play an important role in the regulation of aversive responses in the periaqueductal gray (PAG). Among the results supporting this role, several studies have shown that inhibitors of neuronal NO synthase or cannabinoid receptor type 1 (CB1) receptor agonists cause clear anxiolytic responses when injected into this region. The nitrergic and eCB systems can regulate the activity of classical neurotransmitters such as glutamate and γ-aminobutyric acid (GABA) that control PAG activity. We propose that they exert a 'fine-tuning' regulatory control of defensive responses in this area. This control, however, is probably complex, which may explain the usually bell-shaped dose-response curves observed with drugs that act on NO- or CB1-mediated neurotransmission. Even if the mechanisms responsible for this complex interaction are still poorly understood, they are beginning to be recognized. For example, activation of transient receptor potential vanilloid type-1 channel (TRPV1) receptors by anandamide seems to counteract the anxiolytic effects induced by CB1 receptor activation caused by this compound. Further studies, however, are needed to identify other mechanisms responsible for this fine-tuning effect.


Subject(s)
Anxiety/physiopathology , Escape Reaction/physiology , Neurotransmitter Agents/physiology , Periaqueductal Gray/physiology , Synaptic Transmission/physiology , Animals , Anxiety/metabolism , Arachidonic Acids/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Endocannabinoids/pharmacology , Endocannabinoids/physiology , Mice , Nitric Oxide/physiology , Periaqueductal Gray/metabolism , Polyunsaturated Alkamides/pharmacology , Rats , TRPV Cation Channels/physiology
13.
J Psychopharmacol ; 26(1): 40-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21616976

ABSTRACT

Administration of Cannabis sativa derivatives causes anxiolytic or anxiogenic effects in humans and laboratory animals, depending on the specific compound and dosage used. In agreement with these findings, several studies in the last decade have indicated that the endocannabinoid system modulates neuronal activity in areas involved in defensive responses. The mechanisms of these effects, however, are still not clear. The present review summarizes recent data suggesting that they involve modulation of glutamate and GABA-mediated neurotransmission in brain sites such as the medial prefrontal cortex, amygdaloid complex, bed nucleus of the stria terminalis, hippocampus and dorsal periaqueductal gray. Moreover, we also discuss results indicating that, in these regions, the endocannabinoid system could be particularly engaged by highly stressful situations.


Subject(s)
Brain/physiology , Cannabinoids/metabolism , Defense Mechanisms , Synaptic Transmission/physiology , Animals , Brain/drug effects , Brain/metabolism , Cannabinoids/pharmacology , Humans , Neuroanatomy/methods , Synaptic Transmission/drug effects
14.
Neuroscience ; 204: 186-92, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21906661

ABSTRACT

The transient receptor potential vanilloid type 1 channel (TRPV1; originally vanilloid receptor VR1) is activated in peripheral terminals of nociceptive fibers by noxious heat, low pH, and natural products such as capsaicin, the pungent ingredient of red-hot chilli peppers. Evidence has been accumulating that TRPV1 is expressed also in the brain, where it seems to be involved in antinociception, locomotor control, and regulation of affective behaviors. This ion channel might be activated by arachidonoyl ethanolamide (anandamide), the endogenous agonist of the cannabinoid type 1 (CB(1)) receptor. However, while CB(1) activation leads to a decrease in intracellular calcium and attenuation of synaptic transmission, anandamide binding to TRPV1 results in elevated calcium levels and potentiated synaptic transmission. This suggests a tripartite regulatory system with antagonistic effects of CB(1) and TRPV1, which are tied together by the same endogenous ligand. Such a system may have important implication for the modulation of behavioral responses. The present commentary elaborates on this interplay between CB(1) receptors and TRPV1 channels in the context of fear- and anxiety-related behaviors.


Subject(s)
Anxiety/metabolism , Fear/physiology , Receptor, Cannabinoid, CB1/metabolism , TRPV Cation Channels/metabolism , Animals , Signal Transduction/physiology , Stress, Psychological/metabolism
15.
Br J Pharmacol ; 159(1): 122-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20002102

ABSTRACT

BACKGROUND AND PURPOSE: Cannabidiol (CBD) is a non-psychotomimetic compound from Cannabis sativa that induces anxiolytic- and antipsychotic-like effects in animal models. Effects of CBD may be mediated by the activation of 5-HT(1A) receptors. As 5-HT(1A) receptor activation may induce antidepressant-like effects, the aim of this work was to test the hypothesis that CBD would have antidepressant-like activity in mice as assessed by the forced swimming test. We also investigated if these responses depended on the activation of 5-HT(1A) receptors and on hippocampal expression of brain-derived neurotrophic factor (BDNF). EXPERIMENTAL APPROACH: Male Swiss mice were given (i.p.) CBD (3, 10, 30, 100 mg*kg(-1)), imipramine (30 mg*kg(-1)) or vehicle and were submitted to the forced swimming test or to an open field arena, 30 min later. An additional group received WAY100635 (0.1 mg*kg(-1), i.p.), a 5-HT(1A) receptor antagonist, before CBD (30 mg*kg(-1)) and assessment by the forced swimming test. BDNF protein levels were measured in the hippocampus of another group of mice treated with CBD (30 mg*kg(-1)) and submitted to the forced swimming test. KEY RESULTS: CBD (30 mg*kg(-1)) treatment reduced immobility time in the forced swimming test, as did the prototype antidepressant imipramine, without changing exploratory behaviour in the open field arena. WAY100635 pretreatment blocked CBD-induced effect in the forced swimming test. CBD (30 mg*kg(-1)) treatment did not change hippocampal BDNF levels. CONCLUSION AND IMPLICATIONS: CBD induces antidepressant-like effects comparable to those of imipramine. These effects of CBD were probably mediated by activation of 5-HT(1A) receptors.


Subject(s)
Antidepressive Agents/pharmacology , Cannabidiol/pharmacology , Depression/drug therapy , Receptor, Serotonin, 5-HT1A/drug effects , Animals , Antidepressive Agents/administration & dosage , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cannabidiol/administration & dosage , Cannabis/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Imipramine/pharmacology , Male , Mice , Piperazines/pharmacology , Pyridines/pharmacology , Receptor, Serotonin, 5-HT1A/metabolism , Swimming
16.
Neural Plast ; 2009: 625469, 2009.
Article in English | MEDLINE | ID: mdl-19096514

ABSTRACT

Cannabinoids play an important role in activity-dependent changes in synaptic activity and can interfere in several brain functions, including responses to aversive stimuli. The regions responsible for their effects, however, are still unclear. Cannabinoid type 1 (CB1) receptors are widely distributed in the central nervous system and are present in the periaqueductal gray (PAG), a midbrain structure closely involved in responses related to aversive states. Accordingly, exposure to stressful stimuli increases endocannabinoid (eCB) levels in the PAG, and local administration of CB1 agonists or drugs that facilitate eCB-mediated neurotransmission produces antinociceptive and antiaversive effects. To investigate if these drugs would also interfere in animal models that are sensitive to anxiolytic drugs, we verified the responses to intra-PAG injection of CB1 agonists in rats submitted to the elevated plus-maze, the Vogel punished licking test, or contextual aversive conditioning model. The drugs induced anxiolytic-like effects in all tests. The same was observed with the transient receptor potential vanilloid type 1 (TRPV1) antagonist capsazepine and with cannabidiol, a nonpsychotomimetic phytocannabinoid that produces anxiolytic-like effects after systemic administration in humans and laboratory animals. These results, therefore, suggest that the PAG could be an important site for the antiaversive effects of cannabinoids.


Subject(s)
Cannabinoids/pharmacology , Periaqueductal Gray/drug effects , Periaqueductal Gray/physiology , Animals , Anti-Anxiety Agents/pharmacology , Cannabidiol/pharmacology , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Microinjections , Rats , Receptor, Cannabinoid, CB1/agonists , Stress, Physiological/drug effects , Synaptic Transmission/physiology , TRPV Cation Channels/antagonists & inhibitors
17.
Braz J Med Biol Res ; 39(4): 421-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16612464

ABSTRACT

A high dose of delta9-tetrahydrocannabinol, the main Cannabis sativa (cannabis) component, induces anxiety and psychotic-like symptoms in healthy volunteers. These effects of delta9-tetrahydrocannabinol are significantly reduced by cannabidiol (CBD), a cannabis constituent which is devoid of the typical effects of the plant. This observation led us to suspect that CBD could have anxiolytic and/or antipsychotic actions. Studies in animal models and in healthy volunteers clearly suggest an anxiolytic-like effect of CBD. The antipsychotic-like properties of CBD have been investigated in animal models using behavioral and neurochemical techniques which suggested that CBD has a pharmacological profile similar to that of atypical antipsychotic drugs. The results of two studies on healthy volunteers using perception of binocular depth inversion and ketamine-induced psychotic symptoms supported the proposal of the antipsychotic-like properties of CBD. In addition, open case reports of schizophrenic patients treated with CBD and a preliminary report of a controlled clinical trial comparing CBD with an atypical antipsychotic drug have confirmed that this cannabinoid can be a safe and well-tolerated alternative treatment for schizophrenia. Future studies of CBD in other psychotic conditions such as bipolar disorder and comparative studies of its antipsychotic effects with those produced by clozapine in schizophrenic patients are clearly indicated.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Cannabidiol/therapeutic use , Cannabis/chemistry , Schizophrenia/drug therapy , Animals , Clinical Trials as Topic , Disease Models, Animal , Humans , Mental Disorders/chemically induced , Mental Disorders/drug therapy , Mice , Rats
18.
Braz. j. med. biol. res ; 39(4): 421-429, Apr. 2006. ilus, tab, graf
Article in English | LILACS | ID: lil-425087

ABSTRACT

A high dose of delta9-tetrahydrocannabinol, the main Cannabis sativa (cannabis) component, induces anxiety and psychotic-like symptoms in healthy volunteers. These effects of delta9-tetrahydrocannabinol are significantly reduced by cannabidiol (CBD), a cannabis constituent which is devoid of the typical effects of the plant. This observation led us to suspect that CBD could have anxiolytic and/or antipsychotic actions. Studies in animal models and in healthy volunteers clearly suggest an anxiolytic-like effect of CBD. The antipsychotic-like properties of CBD have been investigated in animal models using behavioral and neurochemical techniques which suggested that CBD has a pharmacological profile similar to that of atypical antipsychotic drugs. The results of two studies on healthy volunteers using perception of binocular depth inversion and ketamine-induced psychotic symptoms supported the proposal of the antipsychotic-like properties of CBD. In addition, open case reports of schizophrenic patients treated with CBD and a preliminary report of a controlled clinical trial comparing CBD with an atypical antipsychotic drug have confirmed that this cannabinoid can be a safe and well-tolerated alternative treatment for schizophrenia. Future studies of CBD in other psychotic conditions such as bipolar disorder and comparative studies of its antipsychotic effects with those produced by clozapine in schizophrenic patients are clearly indicated.


Subject(s)
Animals , Humans , Mice , Rats , Anti-Anxiety Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Cannabidiol/therapeutic use , Cannabis/chemistry , Schizophrenia/drug therapy , Clinical Trials as Topic , Disease Models, Animal , Mental Disorders/chemically induced , Mental Disorders/drug therapy
19.
Pharmacol Biochem Behav ; 82(1): 182-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16153701

ABSTRACT

The dorsomedial part of the ventromedial hypothalamic nuclei (VMHdm) has been related to the modulation of defensive behavior in mammals. The objective of the present study was to test the hypothesis that administration into the VMHdm of midazolam, a benzodiazepine receptor full agonist, or AP7, a glutamate NMDA receptor antagonist, would produce anxiolytic effects in the elevated plus-maze (EPM) or the Vogel's punished licking tests. Male Wistar rats with unilateral cannulae aimed at the VMHdm received intra-cerebral injections of midazolam (15-60 nmol/0.25 microL), AP7 (0.2-2 nmol/0.3 microL) or saline and were submitted to the behavioral tests. Midazolam (30 nmol) increased the percentage of time spent in open arms of the EPM. AP7, on the other hand, decreased open and enclosed arm exploration. In the Vogel test, however, both midazolam (30-60 nmol) and AP7 increased the number of punished licks. Histological control experiments found no significant effects when the drugs were injected into the nearby lateral hypothalamic area. These results suggest that facilitation of gabaergic or antagonism of glutamatergic neurotransmission in the VMHdm can produce anxiolytic-like effects.


Subject(s)
Anxiety/physiopathology , Receptors, GABA-A/physiology , Receptors, Glutamate/physiology , Ventromedial Hypothalamic Nucleus/physiopathology , Animals , Exploratory Behavior/drug effects , Male , Midazolam/pharmacology , Rats , Rats, Wistar
20.
Neurosci Biobehav Rev ; 29(8): 1313-22, 2005.
Article in English | MEDLINE | ID: mdl-16095696

ABSTRACT

Nitric oxide synthase (NOS) positive neurons are located in most brain areas related to defensive reactions, including the dorsolateral periaqueductal grey (dlPAG). NOS inhibitors injected into this structure induce anxiolytic-like responses whereas NO donors promote flight reactions. Intra-dlPAG administration of carboxy-PTIO, a NO scavenger, or ODQ, a soluble guanylate cyclase inhibitor, produced anxiolytic-like effects on rats exposed to the elevated plus-maze (EPM). A double-staining experiment using NADPHd histochemistry and c-Fos immunohistochemistry in rats exposed to a cat or to the EPM showed increased activation of NO producing neurons in the dlPAG, paraventricular and lateral nuclei of hypothalamus and dorsal raphe nucleus. Cat exposure also increased activation of NOS neurons in the medial amygdala, dorsal pre-mammillary nucleus and bed nucleus of stria terminalis. Local infusion into the dlPAG of a glutamate NMDA-receptor antagonist (AP7) or a benzodiazepine agonist (midazolam) completely prevented the flight reactions induced by intra-dlPAG administration of SIN-1, a NO donor. The responses were also inhibited by the 5-HT2A/C agonist DOI but not by a 5-HT1A agonist. These results suggest a modulatory role for NO on brain areas related to defensive reactions, probably by interacting with glutamate, serotonin and/or GABA-mediated neurotransmission.


Subject(s)
Brain/physiology , Escape Reaction/physiology , Nitric Oxide/physiology , 2-Amino-5-phosphonovalerate/analogs & derivatives , 2-Amino-5-phosphonovalerate/pharmacology , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Brain/metabolism , Cats , Cyclic N-Oxides/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Free Radical Scavengers/pharmacology , GABA Modulators/pharmacology , Imidazoles/pharmacology , Immunohistochemistry/methods , Maze Learning/drug effects , Midazolam/pharmacology , Models, Biological , NADPH Dehydrogenase/metabolism , Oxadiazoles/pharmacology , Quinoxalines/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...