Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Exp Brain Res ; 242(5): 1149-1160, 2024 May.
Article En | MEDLINE | ID: mdl-38489023

Hypofunctioning of NMDA receptors, and the resulting shift in the balance between excitation and inhibition, is considered a key process in the pathophysiology of schizophrenia. One important manifestation of this phenomenon is changes in neural oscillations, those above 30 Hz (i.e., gamma-band oscillations), in particular. Although both preclinical and clinical studies observed increased gamma activity following acute administration of NMDA receptor antagonists, the relevance of this phenomenon has been recently questioned given the reduced gamma oscillations typically observed during sensory and cognitive tasks in schizophrenia. However, there is emerging, yet contradictory, evidence for increased spontaneous gamma-band activity (i.e., at rest or under baseline conditions). Here, we use the sub-chronic phencyclidine (PCP) rat model for schizophrenia, which has been argued to model the pathophysiology of schizophrenia more closely than acute NMDA antagonism, to investigate gamma oscillations (30-100 Hz) in the medial prefrontal cortex of anesthetized animals. While baseline gamma oscillations were not affected, oscillations induced by train stimulation of the posterior dorsal CA1 (pdCA1) field of the hippocampus were enhanced in PCP-treated animals (5 mg/kg, twice daily for 7 days, followed by a 7-day washout period). This effect was reversed by pharmacological enhancement of endocannabinoid levels via systemic administration of URB597 (0.3 mg/kg), an inhibitor of the catabolic enzyme of the endocannabinoid anandamide. Intriguingly, the pharmacological blockade of CB1 receptors by AM251 unmasked a reduced gamma oscillatory activity in PCP-treated animals. The findings are consistent with the observed effects of URB597 and AM251 on behavioral deficits reminiscent of the symptoms of schizophrenia and further validate the potential for cannabinoid-based drugs as a treatment for schizophrenia.


Amidohydrolases , Benzamides , Carbamates , Disease Models, Animal , Gamma Rhythm , Phencyclidine , Piperidines , Prefrontal Cortex , Schizophrenia , Animals , Schizophrenia/physiopathology , Schizophrenia/metabolism , Schizophrenia/drug therapy , Phencyclidine/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiopathology , Gamma Rhythm/physiology , Gamma Rhythm/drug effects , Male , Rats , Carbamates/pharmacology , Benzamides/pharmacology , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage , Endocannabinoids/metabolism , Arachidonic Acids/metabolism , Arachidonic Acids/pharmacology , Rats, Sprague-Dawley , Polyunsaturated Alkamides/metabolism , Polyunsaturated Alkamides/pharmacology
2.
Neurosci Lett ; 759: 136064, 2021 08 10.
Article En | MEDLINE | ID: mdl-34146641

Owing to its psychotropic effects, Cannabis has been stigmatized by its recreational use leading to a dramatic decline in the experimentations about its medical use in the twentieth century. The medical properties of the plant - known since ancient times - have received increased attention over recent years; yet, the research on its potential application in the field of psychiatry is still nascent. In this connection, the non-psychotropic cannabidiol (CBD) has emerged as a phytocannabinoid compound with promising antipsychotic effects. In addition, advances in our understanding of the endocannabinoid system, along with accumulating evidence implicating this system in the pathophysiology of schizophrenia, have stimulated research by the pharmaceutical industry to explore whether alteration of this system can be of medical benefit. This review examines the current state of evidence regarding the clinical potential of cannabinoid-based drugs as a treatment for schizophrenia, while discussing various limitations with the therapeutic approaches considered so far. In the second part, the author highlights the most promising strategies, as well as the most interesting directions one could follow, in the emerging field of cannabinoid therapies for schizophrenia.


Cannabidiol/pharmacology , Endocannabinoids/physiology , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Animals , Antipsychotic Agents/pharmacology , Humans
3.
PLoS One ; 15(3): e0230238, 2020.
Article En | MEDLINE | ID: mdl-32163506

Social withdrawal in the sub-chronic phencyclidine (PCP) rat model, a behavioral correlate of the negative symptoms of schizophrenia, results from deficits in brain endocannabinoid transmission. As cannabis intake has been shown to affect negatively the course and expression of psychosis, we tested whether the beneficial effects of endocannabinoid-mediated CB1 activation on social withdrawal in PCP-treated rats (5 mg/kg, twice daily for 7 days)also occurred after administration of Δ9-tetrahydrocannabinol (THC; 0.1, 0.3, 1.0 mg/kg, i.p.). In addition, we assessed whether THC affected two correlates of positive symptoms: 1) motor activity induced by d-amphetamine (0.5 mg/kg, i.p.), and 2) dopamine neuron population activity in the ventral tegmental area (VTA). After the motor activity test, the brains from d-amphetamine-treated animals were collected and processed for measurements of endocannabinoids and activation of Akt/GSK3ß, two molecular markers involved in the pathophysiology of schizophrenia. In control rats, THC dose-dependently produced social interaction deficits and aberrant VTA dopamine neuron population activity similar to those observed in PCP-treated animals. In PCP-treated rats, only the lowest dose of THC reversed PCP-induced deficits, as well as PCP-induced elevation of the endocannabinoid anandamide (AEA) in the nucleus accumbens. Last, THC activated the Akt/GSK3ß pathway dose-dependently in both control and PCP-treated animals. Taken together, these data suggest that only low doses of THC have beneficial effects on behavioral, neurochemical and electrophysiological correlates of schizophrenia symptoms. This observation may shed some light on the controversial hypothesis of marijuana use as self-medication in schizophrenic patients.


Dronabinol/administration & dosage , Phencyclidine/pharmacology , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Animals , Arachidonic Acids/pharmacology , Disease Models, Animal , Endocannabinoids/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Male , Motor Activity/drug effects , Neurons/drug effects , Polyunsaturated Alkamides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
4.
Neuropharmacology ; 130: 1-9, 2018 03 01.
Article En | MEDLINE | ID: mdl-29169961

Experimental evidence suggests that the transport of endocannabinoids might work bi-directionally. Accordingly, it is possible that pharmacological blockade of the latter affects not only the re-uptake, but also the release of endocannabinoids, thus preventing them from stimulating CB1 receptors. We used biochemical, pharmacological, and behavioral approaches to investigate the effects of the transporter inhibitor OMDM-2 on social interaction, a behavioral assay that requires activation of CB1 receptors. The underlying mechanisms of OMDM-2 were compared with those of the Fatty Acid Amide Hydrolase (FAAH) inhibitor URB597. Systemic administration of OMDM-2 reduced social interaction, but in contrast to URB597-induced social deficit, this effect was not reversed by the TRPV1 antagonist capsazepine. The CB1 antagonist AM251, which did not affect URB597-induced social withdrawal, exacerbated OMDM-2 effect. In addition, the potent CB1 agonist CP55,940 reversed OMDM-2-, but not URB597-, induced social withdrawal. Blockade of CB1 receptor by AM251 reduced social interaction and the cholecystokinin CCK2 antagonist LY225910 reversed this effect. Similarly, OMDM-2-induced social withdrawal was reversed by LY225910, whereas URB597 effect was not. Elevation of endocannabinoid levels by URB597 or JZL184, an inhibitor of 2-AG degradation, failed to reverse OMDM-2-induced social withdrawal, and did not show additive effects on cannabinoid measurements when co-administered with OMDM-2. Taken together, these findings indicate that OMDM-2 impaired social interaction in a manner that is consistent with reduced activation of presynaptic CB1 receptors. As cannabinoid reuptake inhibitors may impair endocannabinoid release, caution should be taken when using these drugs to enhance endocannabinoid tone in vivo.


Arachidonic Acids/pharmacology , Behavior, Animal/drug effects , Benzyl Compounds/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Endocannabinoids/metabolism , Social Behavior , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Amygdala/drug effects , Amygdala/metabolism , Animals , Benzamides/pharmacology , Benzodioxoles/pharmacology , Carbamates/pharmacology , Endocannabinoids/antagonists & inhibitors , Male , Piperidines/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism
5.
Behav Pharmacol ; 28(7): 512-520, 2017 10.
Article En | MEDLINE | ID: mdl-28704273

Social withdrawal should not be considered a direct measure of the negative symptoms of schizophrenia as it may result not only from asociality (primary negative symptom) but also from other altered processes such as anxiety. To understand the contribution of these two factors to social deficit, we investigated whether the social withdrawal observed in the subchronic phencyclidine (PCP) rat model of schizophrenia could be attributed to increased anxiety. Compared to saline controls, PCP-treated rats (5 mg/kg, twice daily for 7 days, followed by a washout period) spent significantly less time in social interaction, but did not show anxiety-like behaviors in different relevant behavioral paradigms. In addition, their social deficit was not affected by a behavioral procedure known to reduce anxiety-like behavior (repeated exposure to the same partner) nor by systemic administration of the classical anxiolytic diazepam. In contrast, PCP-induced social withdrawal was reversed by the cannabinoid agonist CP55,940, a drug with known anxiogenic properties. Furthermore, when using the social approach task, PCP-treated animals performed similarly to control animals treated with diazepam, but not to those treated with the anxiogenic compound pentylenetetrazole. Taken together, our results indicate that PCP-induced social withdrawal cannot be attributed to increased anxiety. These data are discussed in the context of primary versus secondary negative symptoms and the deficit syndrome of schizophrenia.


Anxiety , Schizophrenia/physiopathology , Schizophrenic Psychology , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/physiopathology , Anxiety Disorders/drug therapy , Behavior, Animal/drug effects , Cannabinoids , Carbamates/pharmacology , Disease Models, Animal , Exploratory Behavior/drug effects , Interpersonal Relations , Male , Phencyclidine/pharmacology , Piperidines/pharmacology , Rats , Rats, Wistar , Schizophrenia/chemically induced , Social Behavior
6.
Neurosci Res ; 110: 49-58, 2016 Sep.
Article En | MEDLINE | ID: mdl-27091613

The fatty acid amide hydrolase inhibitor, URB597, an endocannabinoid enhancing drug, reverses social withdrawal in the sub-chronic PCP rat model of schizophrenia, but reduces social interaction (SI) in controls. To identify the anatomical substrates associated with PCP-induced social withdrawal and the contrasting effects of URB597 on SI in PCP- versus saline-treated rats, we analyzed SI-induced c-Fos expression in 28 brain areas relevant to schizophrenia and/or social behavior following vehicle or URB597 administration. In saline-treated rats, SI was accompanied by changes in c-Fos expression in the infralimbic and orbitofrontal cortices, dorsomedial caudate putamen, ventrolateral nucleus of the septum, dorsolateral periaqueductal gray (dlPAG) and central amygdala. Except for the dlPAG, these changes were not observed in PCP-treated rats or in saline-treated rats receiving URB597. In the dorsomedial part of the bed nucleus of the stria terminalis (dmBNST), SI-induced c-Fos expression was observed only in PCP-treated rats. Interestingly, URB597 in PCP-treated rats restored a similar c-Fos expression pattern as observed in saline-treated rats: activation of the orbitofrontal cortex, inhibition of the central amygdala and suppression of activation of the dmBNST. These data suggest that orbitofrontal cortex, central amygdala and dmBNST play a critical role in the reversal of PCP-induced social withdrawal by URB597.


Benzamides/pharmacology , Carbamates/pharmacology , Endocannabinoids/metabolism , Neurons/metabolism , Phencyclidine , Social Behavior , Amidohydrolases/antagonists & inhibitors , Animals , Brain/metabolism , Exploratory Behavior/drug effects , Male , Proto-Oncogene Proteins c-fos/metabolism , Rats, Wistar , Schizophrenia/chemically induced , Schizophrenic Psychology
7.
Eur Neuropsychopharmacol ; 26(2): 298-309, 2016 Feb.
Article En | MEDLINE | ID: mdl-26706691

Previous studies have shown that social withdrawal in the phencyclidine (PCP) rat model of schizophrenia results from deficient endocannabinoid-induced activation of CB1 receptors. To understand the underlying cognitive mechanisms of the social deficit in PCP-treated rats, we examined the impact of pharmacological manipulation of the endocannabinoid system on sociability (i.e. social approach) and social novelty preference (which relies on social recognition). Control rats showed a clear preference for a "social" cage (i.e. unfamiliar stimulus rat placed under a wire mesh cage) versus an "empty" cage, and spent more time exploring a "novel" cage (i.e. new stimulus rat) versus a "familiar" cage. In contrast, rats receiving PCP (5 mg/kg, b.i.d. for 7 days, followed by a 7 day-washout period) showed intact sociability, but lacked social novelty preference. This PCP-induced deficit was due to increased activity at CB1 receptors as it was reversed by systemic administration of the CB1 antagonist AM251 (1 mg/kg). In agreement with this hypothesis, the cannabinoid agonist CP55,940 (0.003-0.03 mg/kg) dose-dependently suppressed social novelty preference in control animals without affecting sociability. Taken together, these data suggest that PCP-treated rats have a deficit in social cognition, possibly induced by increased stimulation of CB1 receptors. This deficit, however, is distinct from the social withdrawal previously observed in these animals, as the latter is due to deficient, rather than increased, CB1 stimulation.


Endocannabinoids/metabolism , Excitatory Amino Acid Antagonists/toxicity , Phencyclidine/toxicity , Schizophrenia/chemically induced , Schizophrenia/complications , Schizophrenic Psychology , Social Behavior Disorders/etiology , Analysis of Variance , Animals , Cannabinoid Receptor Modulators/pharmacology , Cyclohexanols/pharmacology , Discrimination, Psychological/drug effects , Disease Models, Animal , Exploratory Behavior/drug effects , Male , Odorants , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Social Perception
8.
J Pharmacol Exp Ther ; 353(2): 261-8, 2015 May.
Article En | MEDLINE | ID: mdl-25711338

Monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH) inhibitors exert preclinical effects indicative of therapeutic potential (i.e., analgesia). However, the extent to which MAGL and FAAH inhibitors produce unwanted effects remains unclear. Here, FAAH and MAGL inhibition was examined separately and together in a Δ(9)-tetrahydrocannabinol (Δ(9)-THC; 5.6 mg/kg i.p.) discrimination assay predictive of subjective effects associated with cannabis use, and the relative contribution of N-arachidonoyl ethanolamine (AEA) and 2-arachidonoylglycerol (2-AG) in the prefrontal cortex, hippocampus, and caudate putamen to those effects was examined. Δ(9)-THC dose-dependently increased Δ(9)-THC appropriate responses (ED50 value = 2.8 mg/kg), whereas the FAAH inhibitors PF-3845 [N-3-pyridinyl-4-[[3-[[5-(trifluoromethyl)-2-pyridinyl]oxy]phenyl]methyl]-1-piperidinecarboxamide] and URB597 [(3'-​(aminocarbonyl)[1,​1'-​biphenyl]-​3-​yl)-​cyclohexylcarbamate] or a MAGL inhibitor JZL184 [4-​nitrophenyl-​4-​(dibenzo[d][1,​3]dioxol-​5-​yl(hydroxy)methyl)piperidine-​1-​carboxylate] alone did not substitute for the Δ(9)-THC discriminative stimulus. The nonselective FAAH/MAGL inhibitors SA-57 [4-[2-(4-chlorophenyl)ethyl]-1-piperidinecarboxylic acid 2-(methylamino)-2-oxoethyl ester] and JZL195 [4-​nitrophenyl 4-​(3-​phenoxybenzyl)piperazine-​1-​carboxylate] fully substituted for Δ(9)-THC with ED50 values equal to 2.4 and 17 mg/kg, respectively. Full substitution for Δ(9)-THC was also produced by a combination of JZL184 and PF-3845, but not by a combination of JZL184 and URB597 (i.e., 52% maximum). Cannabinoid receptor type 1 antagonist rimonabant attenuated the discriminative stimulus effects of Δ(9)-THC, SA-57, JZL195, and the combined effects of JZL184 and PF-3845. Full substitution for the Δ(9)-THC discriminative stimulus occurred only when both 2-AG and AEA were significantly elevated, and the patterns of increased endocannabinoid content were similar among brain regions. Overall, these results suggest that increasing both endogenous 2-AG and AEA produces qualitatively unique effects (i.e., the subjective effects of cannabis) that are not obtained from increasing either 2-AG or AEA separately.


Amidohydrolases/antagonists & inhibitors , Dronabinol/pharmacology , Enzyme Inhibitors/pharmacology , Monoacylglycerol Lipases/antagonists & inhibitors , Animals , Brain/drug effects , Brain/metabolism , Endocannabinoids/metabolism , Male , Mice
9.
Pharmacol Biochem Behav ; 124: 153-9, 2014 Sep.
Article En | MEDLINE | ID: mdl-24911644

The biological actions of the endocannabinoids anandamide and 2-arachidonoyl glycerol (2-AG) are terminated by enzymatic hydrolysis of these lipids via fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), respectively. While several selective FAAH inhibitors have been developed and characterized in vitro and in vivo, none of the initial MAGL blockers have shown adequate potency and specificity for in vivo applications. More recently, a selective MAGL inhibitor, JZL184, has been shown to produce a long-lasting elevation of brain 2-AG, as well as cannabinoid-like behavioral responses in mice. However, its effectiveness in rats remains controversial. Indeed, although JZL184 can elicit behavioral responses that are mediated, at least in part, via activation of cannabinoid CB1 receptors, several reports indicate that this compound does not alter 2-AG levels in this species. In this study we compared the behavioral and neurochemical effects of JZL 184 with those of the dual FAAH/MAGL inhibitor JZL195, and showed that systemic administration of the former can selectively elevate brain 2-AG in rats and produce motor suppression through a CB1-independent mechanism. These findings indicate that, despite its lower potency against rat MAGL, JZL184 can be used to enhance 2-AG transmission and elicit behavioral responses in rodents.


Anti-Anxiety Agents/pharmacology , Benzodioxoles/pharmacology , Carbamates/pharmacology , Endocannabinoids/metabolism , Motor Activity/drug effects , Piperazines/pharmacology , Piperidines/pharmacology , Animals , Male , Rats , Rats, Wistar
10.
Neuropsychopharmacology ; 38(9): 1816-24, 2013 Aug.
Article En | MEDLINE | ID: mdl-23563893

The neuronal mechanisms underlying social withdrawal, one of the core negative symptoms of schizophrenia, are not well understood. Recent studies suggest an involvement of the endocannabinoid system in the pathophysiology of schizophrenia and, in particular, of negative symptoms. We used biochemical, pharmacological, and behavioral approaches to investigate the role played by the endocannabinoid system in social withdrawal induced by sub-chronic administration of phencyclidine (PCP). Pharmacological enhancement of endocannabinoid levels via systemic administration of URB597, an inhibitor of endocannabinoid degradation, reversed social withdrawal in PCP-treated rats via stimulation of CB1 receptors, but reduced social interaction in control animals through activation of a cannabinoid/vanilloid-sensitive receptor. In addition, the potent CB agonist CP55,940 reversed PCP-induced social withdrawal in a CB1-dependent manner, whereas pharmacological blockade of CB1 receptors by either AM251 or SR141716 reduced the time spent in social interaction in control animals. PCP-induced social withdrawal was accompanied by a decrease of anandamide (AEA) levels in the amygdala and prefrontal cortex, and these deficits were reversed by URB597. As CB1 receptors are predominantly expressed on GABAergic interneurons containing the anxiogenic peptide cholecystokinin (CCK), we also examined whether the PCP-induced social withdrawal resulted from deficient CB1-mediated modulation of CCK transmission. The selective CCK2 antagonist LY225910 blocked both PCP- and AM251-induced social withdrawal, but not URB597 effect in control rats. Taken together, these findings indicate that AEA-mediated activation of CB1 receptors is crucial for social interaction, and that PCP-induced social withdrawal results from deficient endocannabinoid transmission.


Endocannabinoids/physiology , Phencyclidine/pharmacology , Receptor, Cannabinoid, CB1/physiology , Schizophrenia/physiopathology , Social Behavior , Amidohydrolases/antagonists & inhibitors , Amygdala/drug effects , Amygdala/metabolism , Animals , Arachidonic Acids/metabolism , Benzamides/pharmacology , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Carbamates/pharmacology , Cyclohexanols/pharmacology , Dose-Response Relationship, Drug , Endocannabinoids/metabolism , Male , Phencyclidine/antagonists & inhibitors , Piperidines/antagonists & inhibitors , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyrazoles/antagonists & inhibitors , Pyrazoles/pharmacology , Quinazolinones/pharmacology , Rats , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cholecystokinin B/antagonists & inhibitors , Rimonabant
11.
Eur J Appl Physiol ; 113(4): 869-75, 2013 Apr.
Article En | MEDLINE | ID: mdl-22990628

Endocannabinoids (eCB) are endogenous ligands for cannabinoid receptors that are densely expressed in brain networks responsible for reward. Recent work shows that exercise activates the eCB system in humans and other mammals, suggesting eCBs are partly responsible for the reported improvements in mood and affect following aerobic exercise in humans. However, exercise-induced psychological changes reported by runners are known to be dependent on exercise intensity, suggesting that any underlying molecular mechanism should also change with varying levels of exercise intensity. Here, we examine circulating levels of eCBs following aerobic exercise (treadmill running) in recreationally fit human runners at four different intensities. We show that eCB signaling is indeed intensity dependent, with significant changes in circulating eCBs observed following moderate intensities only (very high and very low intensity exercises do not significantly alter circulating eCB levels). Our results are consistent with intensity-dependent psychological state changes with exercise and therefore support the hypothesis that eCB activity is related to neurobiological effects of exercise. Thus, future studies examining the role of exercise-induced eCB signaling on neurobiology or physiology must take exercise intensity into account.


Brain/metabolism , Endocannabinoids/blood , Exercise/physiology , Muscle Contraction , Muscle, Skeletal/metabolism , Signal Transduction , Adolescent , Adult , Analysis of Variance , Arachidonic Acids/blood , Arizona , Exercise/psychology , Exercise Test , Female , Humans , Male , Polyunsaturated Alkamides/blood , Running , Young Adult
12.
Prog Neuropsychopharmacol Biol Psychiatry ; 38(2): 260-9, 2012 Aug 07.
Article En | MEDLINE | ID: mdl-22542870

Supratherapeutic doses of the analgesic acetaminophen (paracetomol) are reported to promote social behavior in Swiss mice. However, we hypothesized that it might not promote sociability in other strains due to cannabinoid CB(1) receptor-mediated inhibition of serotonin (5-HT) transmission in the frontal cortex. We examined the effects of acetaminophen on social and repetitive behaviors in comparison to a cannabinoid agonist, WIN 55,212-2, in two strains of socially-deficient mice, BTBR and 129S1/SvImJ (129S). Acetaminophen (100mg/kg) enhanced social interactions in BTBR, and social novelty preference and marble burying in 129S at serum levels of ≥70 ng/ml. Following acetaminophen injection or sociability testing, anandamide (AEA) increased in BTBR frontal cortex, while behavior testing increased 2-arachidonyl glycerol (2-AG) levels in 129S frontal cortex. In contrast, WIN 55,212-2 (0.1mg/kg) did not enhance sociability. Further, we expected CB(1)-deficient (+/-) mice to be less social than wild-type, but instead found similar sociability. Given strain differences in endocannabinoid response to acetaminophen, we compared cortical CB(1) and 5-HT(1A) receptor density and function relative to sociable C57BL/6 mice. CB(1) receptor saturation binding (Bmax=958±117 fmol/mg protein), and affinity for [(3)H] CP55,940 (K(D)=3±0.8 nM) was similar in frontal cortex among strains. CP55,940-stimulated [(35)S] GTPγS binding in cingulate cortex was 136±12, 156±22, and 75±9% above basal in BTBR, 129S and C57BL/6 mice. The acetaminophen metabolite para-aminophenol (1 µM) failed to stimulate [(35)S] GTPγS binding. Hence, it appears that other indirect actions of acetaminophen, including 5-HT receptor agonism, may underlie its sociability promoting properties outweighing any CB(1) mediated suppression by locally-elevated endocannabinoids in these mice.


Acetaminophen/pharmacology , Arachidonic Acids/metabolism , Behavior, Animal/drug effects , Endocannabinoids/metabolism , Frontal Lobe/drug effects , Glycerides/metabolism , Polyunsaturated Alkamides/metabolism , Social Behavior , Animals , Benzoxazines/pharmacology , Dose-Response Relationship, Drug , Frontal Lobe/metabolism , Male , Mice , Mice, Inbred Strains , Morpholines/pharmacology , Naphthalenes/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Serotonin/metabolism
13.
Article En | MEDLINE | ID: mdl-22521335

The endocannabinoids are lipid signaling molecules that bind to cannabinoid CB(1) and CB(2) receptors and other metabotropic and ionotropic receptors. Anandamide and 2-arachidonoyl glycerol, the two best-characterized examples, are released on demand in a stimulus-dependent manner by cleavage of membrane phospholipid precursors. Together with their receptors and metabolic enzymes, the endocannabinoids play a key role in modulating neurotransmission and synaptic plasticity in the basal ganglia and other brain areas involved in the control of motor functions and motivational aspects of behavior. This mini-review provides an update on the contribution of the endocannabinoid system to the regulation of psychomotor behaviors and its possible involvement in the pathophysiology of Parkinson's disease and schizophrenia.


Basal Ganglia/metabolism , Endocannabinoids/metabolism , Psychomotor Disorders/metabolism , Receptors, Cannabinoid/metabolism , Signal Transduction/physiology , Humans , Synaptic Transmission/physiology , TRPC Cation Channels/metabolism
14.
J Exp Biol ; 215(Pt 8): 1331-6, 2012 Apr 15.
Article En | MEDLINE | ID: mdl-22442371

Humans report a wide range of neurobiological rewards following moderate and intense aerobic activity, popularly referred to as the 'runner's high', which may function to encourage habitual aerobic exercise. Endocannabinoids (eCBs) are endogenous neurotransmitters that appear to play a major role in generating these rewards by activating cannabinoid receptors in brain reward regions during and after exercise. Other species also regularly engage in endurance exercise (cursorial mammals), and as humans share many morphological traits with these taxa, it is possible that exercise-induced eCB signaling motivates habitual high-intensity locomotor behaviors in cursorial mammals. If true, then neurobiological rewards may explain variation in habitual locomotor activity and performance across mammals. We measured circulating eCBs in humans, dogs (a cursorial mammal) and ferrets (a non-cursorial mammal) before and after treadmill exercise to test the hypothesis that neurobiological rewards are linked to high-intensity exercise in cursorial mammals. We show that humans and dogs share significantly increased exercise-induced eCB signaling following high-intensity endurance running. eCB signaling does not significantly increase following low-intensity walking in these taxa, and eCB signaling does not significantly increase in the non-cursorial ferrets following exercise at any intensity. This study provides the first evidence that inter-specific variation in neurotransmitter signaling may explain differences in locomotor behavior among mammals. Thus, a neurobiological reward for endurance exercise may explain why humans and other cursorial mammals habitually engage in aerobic exercise despite the higher associated energy costs and injury risks, and why non-cursorial mammals avoid such locomotor behaviors.


Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Exercise , Ferrets/physiology , Physical Conditioning, Animal , Physical Endurance/physiology , Running/physiology , Signal Transduction , Animals , Dogs , Humans
15.
Pharmacol Biochem Behav ; 98(4): 583-6, 2011 Jun.
Article En | MEDLINE | ID: mdl-21421000

Sub-chronic administration of PCP produces a social interaction deficit that is reversed by URB597, an inhibitor of the catabolic enzyme of the endocannabinoid anandamide. Since increased anxiety may contribute to social withdrawal and URB597 has been shown to have an anxiolytic action, we studied whether this drug affected saline- and PCP-treated rats' performance in the Elevated-Plus-Maze task, which has been used to assess anxiety-like effects. Sub-chronic PCP produced a CB(1)-dependent decrease in anxiety-like behavior that was reversed by URB597 in a CB(1)-independent fashion, as it was not blocked by the CB(1) antagonist AM251. These findings suggest that PCP-treated rats have altered endocannabinoid transmission and that anxiety does not contribute to the PCP-induced social withdrawal.


Amidohydrolases/antagonists & inhibitors , Anxiety/drug therapy , Phencyclidine/toxicity , Amidohydrolases/physiology , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/enzymology , Anxiety/physiopathology , Anxiety/psychology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Benzamides/pharmacology , Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Male , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/physiology
16.
Int J Neuropsychopharmacol ; 13(3): 373-86, 2010 04.
Article En | MEDLINE | ID: mdl-19607756

The 'cannabinoid hypothesis' of schizophrenia tulates that over-activity of the endocannabinoid system might contribute to the aetiology of schizophrenia. In keeping with this hypothesis, increased expression of CB1 receptors, elevation of the endocannabinoid anandamide (AEA) and cannabinoid-induced cognitive changes have been reported in animal models of schizophrenia and psychotic patients. In this study we measured brain endocannabinoid levels and [35S]GTPgammaS binding stimulated by the CB receptor agonist CP55,940 in rats undergoing withdrawal from subchronic administration of phencyclidine (PCP), a well-established pharmacological model of schizophrenia. We also investigated whether systemic application of the fatty-acid amide hydrolase (FAAH) inhibitor URB597 or CB1 receptor blockade by AM251 affected the following PCP-induced behavioural deficits reminiscent of schizophrenia-like symptoms: (1) working-memory impairment (cognitive deficit), (2) social withdrawal (negative symptom), and (3) hyperactivity in response to d-amphetamine challenge (positive symptoms). PCP-treated rats showed increased endocannabinoid levels in the nucleus accumbens and ventral tegmental area, whereas CB1 receptor expression and CP55,940-stimulated [35S]GTPgammaS binding were unaltered. URB597 reversed the PCP-induced social withdrawal but caused social withdrawal and working-memory deficits in saline-treated rats that were comparable to those observed after PCP treatment. Administration of AM251 ameliorated the working-memory deficit in PCP-treated rats, but impaired working memory in saline-injected controls. Taken together, these results suggest that FAAH inhibition may improve negative symptoms in PCP-treated rats but produce deleterious effects in untreated animals, possibly by disturbing endocannabinoid tone. A similar pattern (beneficial for schizophrenia-related cognitive deficits, but detrimental under normal conditions) accompanies CB1 receptor blockade.


Amidohydrolases/metabolism , Behavior, Animal/drug effects , Hallucinogens/pharmacology , Phencyclidine/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Autoradiography/methods , Benzamides/pharmacology , Cannabinoid Receptor Modulators/metabolism , Carbamates/pharmacology , Central Nervous System Stimulants/pharmacology , Cyclohexanols/pharmacology , Drug Administration Schedule , Drug Interactions , Enzyme Inhibitors/pharmacology , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Interpersonal Relations , Male , Memory, Short-Term/drug effects , Motor Activity/drug effects , Neuropsychological Tests , Piperidines/pharmacology , Protein Binding/drug effects , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/drug effects , Statistics, Nonparametric , Sulfur Isotopes/metabolism , Time Factors
17.
Behav Brain Res ; 204(2): 410-5, 2009 Dec 07.
Article En | MEDLINE | ID: mdl-19716985

The hypothesis of hypo-functionality of NMDA receptors in schizophrenia originates from the observation that administration of the NMDA antagonist phencyclidine (PCP) induces psychotic states that closely resemble schizophrenic symptoms and that persist after drug discontinuation. A large number of animal studies have used PCP and the NMDA antagonist dizocilpine (MK-801) almost interchangeably to model schizophrenia. However, PCP interacts with pharmacological targets other than NMDA receptors that are not affected by MK-801. In addition, although acute administration of either compound produces similar effects in animals, there is little information whether withdrawal from chronic MK-801 causes behavioral deficits that mimic schizophrenia symptoms as in the case of PCP. To clarify this issue, we compared the following behaviors in rats subjected to withdrawal from sub-chronic administration (2 x 7 days) of either PCP (5 mg/kg, i.p.) or MK-801 (0.5 mg/kg, i.p.): (1) working memory in a variable-delayed alternation task in a T-maze, (2) social interaction, and (3) motor activity in response to a (a) novel environment, (b) mild stressor, and (c) d-amphetamine challenge. Withdrawal from sub-chronic PCP caused a delay-dependent impairment of working memory, reduced social interaction and enhanced d-amphetamine-induced motor activity. These results were not replicated in animals sub-chronically treated with MK-801, which displayed only a slight decrease in social interaction. These data suggest that pharmacological antagonism at NMDA receptors is not sufficient to explain the full spectrum of PCP psychotomimetic properties.


Dizocilpine Maleate/pharmacology , Phencyclidine/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia/physiopathology , Animals , Body Weight , Disease Models, Animal , Dizocilpine Maleate/administration & dosage , Male , Memory, Short-Term , Motor Activity , Phencyclidine/administration & dosage , Rats , Rats, Wistar , Social Behavior , Substance Withdrawal Syndrome
18.
Eur J Neurosci ; 29(11): 2177-86, 2009 Jun.
Article En | MEDLINE | ID: mdl-19490092

Parkinson's disease (PD) is characterized by the progressive loss of nigrostriatal dopamine neurons leading to motor disturbances and cognitive impairment. Current pharmacotherapies relieve PD symptoms temporarily but fail to prevent or slow down the disease progression. In this study, we investigated the molecular mechanisms by which the non-selective cannabinoid receptor agonist WIN55,212-2 (WIN) protects mouse nigrostriatal neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity and neuroinflammation. Stereological analyses showed that chronic treatment with WIN (4 mg/kg, intraperitoneal), initiated 24 h after MPTP administration, protected against MPTP-induced loss of tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta independently of CB1 cannabinoid receptor activation. The neuroprotective effect of WIN was accompanied by increased dopamine and 3,4-dihydroxyphenylacetic acid levels in the substantia nigra pars compacta and dorsal striatum of MPTP-treated mice. At 3 days post-MPTP, we found significant microglial activation and up-regulation of CB2 cannabinoid receptors in the ventral midbrain. Treatment with WIN or the CB2 receptor agonist JWH015 (4 mg/kg, intraperitoneal) reduced MPTP-induced microglial activation, whereas genetic ablation of CB2 receptors exacerbated MPTP systemic toxicity. Furthermore, chronic WIN reversed MPTP-associated motor deficits, as revealed by the analysis of forepaw step width and percentage of faults using the inverted grid test. In conclusion, our data indicate that agonism at CB2 cannabinoid receptors protects against MPTP-induced nigrostriatal degeneration by inhibiting microglial activation/infiltration and suggest that CB2 receptors represent a new therapeutic target to slow the degenerative process occurring in PD.


Benzoxazines/pharmacology , Cannabinoid Receptor Agonists , Disease Models, Animal , MPTP Poisoning/prevention & control , Morpholines/pharmacology , Naphthalenes/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/prevention & control , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Cell Count , Corpus Striatum/drug effects , Corpus Striatum/pathology , MPTP Poisoning/chemically induced , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/chemically induced , Receptors, Cannabinoid/physiology , Substantia Nigra/drug effects , Substantia Nigra/pathology
19.
Behav Brain Res ; 183(1): 101-10, 2007 Oct 01.
Article En | MEDLINE | ID: mdl-17610962

Several studies have suggested a positive relationship between circulating corticosterone levels and contextual conditioning. However, a positive relationship between circulating corticosterone levels and cued conditioning has also been reported. This study further investigates the relationship between corticosterone and fear conditioning by modulating the predictive value of contextual and discrete tone cues in separate groups of rats. In a first experiment in which training parameters were chosen to induce strong conditioning (five foot-shocks), we used a correlational approach and investigated whether post-training corticosterone levels were related to subsequent expression of contextual and/or tone fear. In a second experiment, in which training parameters were chosen to induce lower conditioning (one and two foot-shocks), we investigated whether a post-training corticosterone injection enhanced the consolidation of contextual and/or tone conditioning. In the first experiment, the highest post-training corticosterone levels were obtained in rats trained with paired tones and shocks. Post-training corticosterone levels tended to be positively correlated with freezing scores during the tone-fear test and were negatively correlated with freezing scores during training although not during the context-fear test. In the second experiment, a post-training injection of corticosterone (3mg/kg) had no effect on subsequent freezing to contextual cues and to a tone that did not predict shock, whereas it was efficient in increasing fear conditioned to a predictive tone. Globally, these results suggest that the predictive value of the conditioned stimulus may be the main determinant of the facilitatory action of acutely enhanced corticosterone in fear conditioning.


Association Learning/physiology , Conditioning, Classical/physiology , Corticosterone/blood , Fear/physiology , Freezing Reaction, Cataleptic/physiology , Analysis of Variance , Animals , Corticosterone/physiology , Cues , Environment , Male , Rats , Rats, Long-Evans
...