Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Psychiatry ; 23(4): 1040-1050, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28630452

RESUMO

The prefrontal cortex (PFC) is a crucial hub for the flexible modulation of recent memories (executive functions) as well as for the stable organization of remote memories. Dopamine in the PFC is implicated in both these processes and genetic variants affecting its neurotransmission might control the unique balance between cognitive stability and flexibility present in each individual. Functional genetic variants in the catechol-O-methyltransferase (COMT) gene result in a different catabolism of dopamine in the PFC. However, despite the established role played by COMT genetic variation in executive functions, its impact on remote memory formation and recall is still poorly explored. Here we report that transgenic mice overexpressing the human COMT-Val gene (COMT-Val-tg) present exaggerated remote memories (>50 days) while having unaltered recent memories (<24 h). COMT selectively and reversibly modulated the recall of remote memories as silencing COMT Val overexpression starting from 30 days after the initial aversive conditioning normalized remote memories. COMT genetic overactivity produced a selective overdrive of the endocannabinoid system within the PFC, but not in the striatum and hippocampus, which was associated with enhanced remote memories. Indeed, acute pharmacological blockade of CB1 receptors was sufficient to rescue the altered remote memory recall in COMT-Val-tg mice and increased PFC dopamine levels. These results demonstrate that COMT genetic variations modulate the retrieval of remote memories through the dysregulation of the endocannabinoid system in the PFC.


Assuntos
Catecol O-Metiltransferase/metabolismo , Endocanabinoides/metabolismo , Memória de Longo Prazo/fisiologia , Córtex Pré-Frontal/metabolismo , Animais , Catecol O-Metiltransferase/genética , Cognição/fisiologia , Dopamina/metabolismo , Feminino , Genótipo , Humanos , Masculino , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Polimorfismo Genético
2.
Artigo em Inglês | MEDLINE | ID: mdl-23916409

RESUMO

Epidemiological studies suggest that Cannabis use during adolescence confers an increased risk for developing psychotic symptoms later in life. However, despite their interest, the epidemiological data are not conclusive, due to their heterogeneity; thus modeling the adolescent phase in animals is useful for investigating the impact of Cannabis use on deviations of adolescent brain development that might confer a vulnerability to later psychotic disorders. Although scant, preclinical data seem to support the presence of impaired social behaviors, cognitive and sensorimotor gating deficits as well as psychotic-like signs in adult rodents after adolescent cannabinoid exposure, clearly suggesting that this exposure may trigger a complex behavioral phenotype closely resembling a schizophrenia-like disorder. Similar treatments performed at adulthood were not able to produce such phenotype, thus pointing to a vulnerability of the adolescent brain towards cannabinoid exposure. The neurobiological substrate of the adolescent vulnerability is still largely unknown and experimental studies need to elucidate the cellular and molecular mechanism underlying these effects. However, the few data available seem to suggest that heavy adolescent exposure to cannabinoids is able to modify neuronal connectivity in specific brain areas long after the end of the treatment. This is likely due to disruption of maturational events within the endocannabinoid system during adolescence that in turn impact on the correct neuronal refinement peculiar of the adolescent brain, thus leading to altered adult brain functionality and behavior.


Assuntos
Modelos Animais de Doenças , Abuso de Maconha/epidemiologia , Esquizofrenia , Psicologia do Esquizofrênico , Adolescente , Animais , Transtornos Cognitivos/etiologia , Humanos , Inibição Psicológica , Relações Interpessoais , Abuso de Maconha/complicações , Reflexo de Sobressalto/fisiologia , Fatores de Risco , Esquizofrenia/epidemiologia , Esquizofrenia/etiologia
3.
J Neuroendocrinol ; 25(2): 107-18, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22967062

RESUMO

We characterised the behavioural phenotype of mice heterozygous (Oxtr(+/-)) for the oxytocin receptor gene (Oxtr) and compared it with that of Oxtr null mice (Oxtr(-/-)), which display autistic-like behaviours, including impaired sociability and preference for social novelty, impaired cognitive flexibility, and increased aggression. Similar to Oxtr(-/-) mice, the Oxtr(+/-) showed impaired sociability and preference for social novelty but, unlike the null genotype, their cognitive flexibility and aggression were normal. By autoradiography, Oxtr(+/-) mice were found to have approximately 50% fewer oxytocin receptors (OXTRs) in all of the examined brain regions. Thus, because a partial reduction in Oxtr gene expression is sufficient to compromise social behaviour, the Oxtr acts as a haploinsufficient gene. Furthermore, the inactivation of the Oxtr gene affects specific behaviours in a dose-dependent manner: social behaviour is sensitive to even a partial reduction in Oxtr gene expression, whereas defects in aggression and cognitive flexibility require the complete inactivation of the Oxtr gene to emerge. We then investigated the rescue of the Oxtr(+/-) social deficits by oxytocin (OT) and Thr(4)Gly(7)OT (TGOT) administered i.c.v. at different doses. TGOT was more potent than OT in rescuing sociability and social novelty in both genotypes. Furthermore, the TGOT doses that reverted impaired sociability and preference for social novelty in Oxtr(+/-) were lower than those required in Oxtr(-/-), thus suggesting that the rescue effect is mediated by OXTR in Oxtr(+/-) and by other receptors (presumably vasopressin V1a receptors) in Oxtr(-/-). In line with this, a low dose of the selective oxytocin antagonist desGlyDTyrOVT blocks the rescue effect of TGOT only in the Oxtr(+/-) genotype, whereas the less selective antagonist SR49059 blocks rescue in both genotypes. In conclusion, the Oxtr(+/-) mouse is a unique animal model for investigating how partial loss of the Oxtr gene impair social interactions, and for designing pharmacological rescue strategies.


Assuntos
Agressão/fisiologia , Cognição/fisiologia , Haploinsuficiência/fisiologia , Receptores de Ocitocina/genética , Comportamento Social , Animais , Comportamento Animal/fisiologia , Haploinsuficiência/genética , Heterozigoto , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Modelos Animais , Plasticidade Neuronal/genética
4.
Br J Pharmacol ; 167(8): 1652-64, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22762735

RESUMO

BACKGROUND AND PURPOSE: Pharmacological interventions aimed at restoring the endocannabinoid system functionality have been proposed as potential tools in the treatment of schizophrenia. Based on our previous results suggesting a potential antipsychotic-like profile of the CB(1) receptor inverse agonist/antagonist, AM251, here we further investigated the effect of chronic AM251 administration on the alteration of the sensorimotor gating functions and endocannabinoid levels induced by isolation rearing in rats. EXPERIMENTAL APPROACH: Using the post-weaning social isolation rearing model, we studied its influence on sensorimotor gating functions through the PPI paradigm. The presence of alterations in the endocannabinoid levels as well as in dopamine and glutamate receptor densities was explored in specific brain regions following isolation rearing. The effect of chronic AM251 administration on PPI response and the associated biochemical alterations was assessed. KEY RESULTS: The disrupted PPI response in isolation-reared rats was paralleled by significant alterations in 2-AG content and dopamine and glutamate receptor densities in specific brain regions. Chronic AM251 completely restored normal PPI response in isolated rats. This behavioural recovery was paralleled by the normalization of 2-AG levels in all the brain areas analysed. Furthermore, AM251 partially antagonized isolation-induced changes in dopamine and glutamate receptors. CONCLUSIONS AND IMPLICATIONS: These results demonstrate the efficacy of chronic AM251 treatment in the recovery of isolation-induced disruption of PPI. Moreover, AM251 counteracted the imbalances in the endocannabinoid content, specifically 2-AG levels, and partially reversed the alterations in dopamine and glutamate systems associated with the disrupted behaviour. Together, these findings support the potential antipsychotic-like activity of CB(1) receptor blockade. LINKED ARTICLES: This article is part of a themed section on Cannabinoids. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.167.issue-8.


Assuntos
Encéfalo/efeitos dos fármacos , Antagonistas de Receptores de Canabinoides/farmacologia , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Filtro Sensorial/efeitos dos fármacos , Estimulação Acústica , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/fisiologia , Endocanabinoides/fisiologia , Masculino , Ratos , Receptor CB1 de Canabinoide/fisiologia , Isolamento Social
5.
Br J Pharmacol ; 167(6): 1218-31, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22624859

RESUMO

BACKGROUND AND PURPOSE: Several studies have demonstrated anti-proliferative and pro-apoptotic actions of cannabinoids on various tumours, together with their anti-angiogenic properties. The non-psychoactive cannabinoid cannabidiol (CBD) effectively inhibits the growth of different types of tumours in vitro and in vivo and down-regulates some pro-angiogenic signals produced by glioma cells. As its anti-angiogenic properties have not been thoroughly investigated to date, and given its very favourable pharmacological and toxicological profile, here, we evaluated the ability of CBD to modulate tumour angiogenesis. EXPERIMENTAL APPROACH: Firstly, we evaluated the effect of CBD on human umbilical vein endothelial cell (HUVEC) proliferation and viability - through [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay and FACS analysis - and in vitro motility - both in a classical Boyden chamber test and in a wound-healing assay. We next investigated CBD effects on different angiogenesis-related proteins released by HUVECs, using an angiogenesis array kit and an ELISA directed at MMP2. Then we evaluated its effects on in vitro angiogenesis in treated HUVECs invading a Matrigel layer and in HUVEC spheroids embedded into collagen gels, and further characterized its effects in vivo using a Matrigel sponge model of angiogenesis in C57/BL6 mice. KEY RESULTS: CBD induced HUVEC cytostasis without inducing apoptosis, inhibited HUVEC migration, invasion and sprouting in vitro, and angiogenesis in vivo in Matrigel sponges. These effects were associated with the down-modulation of several angiogenesis-related molecules. CONCLUSIONS AND IMPLICATIONS: This study reveals that CBD inhibits angiogenesis by multiple mechanisms. Its dual effect on both tumour and endothelial cells supports the hypothesis that CBD has potential as an effective agent in cancer therapy.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Canabidiol/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Animais , Canabidiol/farmacologia , Movimento Celular/efeitos dos fármacos , Heparina , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/induzido quimicamente , Fator de Necrose Tumoral alfa , Fator A de Crescimento do Endotélio Vascular
6.
Neuroscience ; 204: 245-57, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22178986

RESUMO

Preclinical data support the long-term adverse effects on cognition, emotionality, and psychotic-like behaviors of adolescent exposure to natural and synthetic cannabinoids. To investigate whether the long-lasting adverse effects induced by cannabinoids in adolescence are influenced by early-life stress, female and male rats were subjected to 24-h maternal deprivation at postnatal day (PND) 9 and treated with tetrahydrocannabinol (THC) during adolescence (PND 35-45) according to our previously reported protocol. At adulthood, rats were tested in the novel object recognition, social interaction, and forced swim tests, to evaluate possible alterations in recognition memory, social behavior, and coping strategy. Moreover, cannabinoid CB1 receptor density and functionality, as well as NMDA and dopamine D1 and D2 receptor densities were measured through autoradiographic binding studies. In female maternally deprived rats, THC failed to impair recognition memory, counteracted aggressiveness induced by maternal deprivation, whereas no interaction was observed in the passive coping behavior. In males, the association of the two events increased passive coping response without affecting other behaviors. This behavioral picture was accompanied by gender-dependent and region-specific alterations in NMDA, D1 and D2 receptors. In conclusion, this study demonstrates that adolescent THC exposure might have different behavioral outcomes in animals previously exposed to early-life stress compared with non-stressed controls. The interaction between the two events is not univocal, and different combinations may arise depending on the sex of the animals and the behavior considered. Alterations in NMDA, D1 and D2 receptors might be involved in the behavioral responses induced by maternal deprivation and in their modulation by THC.


Assuntos
Comportamento Animal/efeitos dos fármacos , Dronabinol/farmacologia , Memória/efeitos dos fármacos , Animais , Feminino , Masculino , Privação Materna , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Comportamento Social
7.
Neuropharmacology ; 60(2-3): 235-43, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20850463

RESUMO

We have recently shown that chronic THC administration in adolescent female rats induces subtle but lasting alterations in the emotional circuit ending in depressive-like behaviour at adulthood. Here we describe other relevant depressive-like symptoms present in these animals. Adult female rats pretreated with THC display passive coping strategy towards acute stressful situations as demonstrated by their behaviours in the first session of the forced swim test, develop a profound anhedonic state as demonstrated by the reduced consumption of palatable food and present a decrease in social functioning. Besides the emotional symptoms, adolescent exposure to THC induced a significant deficit in object recognition memory. Since it has been reported that deficits in adult hippocampal neurogenesis may underlie the cognitive dysfunction seen in depression, we then survey cell proliferation in the dentate gyrus of the hippocampus. Adolescent THC exposure significantly reduced the number of BrdU-positive cells in THC-treated rats as well as hippocampal volume. We suggest that this complex depressive-like phenotype is triggered by a long-lasting decrease in CB1 receptor functionality in specific brain regions. To test whether an increase in the endocannabinoid signalling could ameliorate the depressive phenotype, adult female rats pre-exposed to THC were injected with URB597 (0.3mg/kg ip) and then tested in behavioural assays. URB597 was able to reverse most depressive-like symptoms induced by adolescent THC exposure such as the passive coping strategy observed in THC exposed animals in the forced swim test as well as anhedonia and the reduced social activity. These results support a role for the endocannabinoid system in the neurobiology of depression and suggest the use of URB597 as a new therapeutic tool with antidepressant properties.


Assuntos
Benzamidas/administração & dosagem , Carbamatos/administração & dosagem , Transtorno Depressivo/induzido quimicamente , Transtorno Depressivo/tratamento farmacológico , Dronabinol/toxicidade , Receptor CB1 de Canabinoide/fisiologia , Adaptação Psicológica/efeitos dos fármacos , Adaptação Psicológica/fisiologia , Fatores Etários , Animais , Transtorno Depressivo/psicologia , Esquema de Medicação , Feminino , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/agonistas , Comportamento Social , Fatores de Tempo
8.
Curr Drug Targets ; 11(4): 393-405, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20017730

RESUMO

Recently, the presence of functional interaction between the opioid and cannabinoid system has been shown in various pharmacological responses. Although there is an increasing interest for the feasible therapeutic application of a co-administration of cannabinoids and opioids in some disorders (i.e. to manage pain, to modulate immune system and emotions) and the combined use of the two drugs by drug abusers is becoming largely diffuse, only few papers focused on cellular and molecular mechanisms underlying this interaction. This review updates the biochemical and molecular underpinnings of opioid and cannabinoid interaction, both within the central nervous system and periphery. The most convincing theory for the explanation of this reciprocal interaction involves (i) the release of opioid peptides by cannabinoids or endocannabinoids by opioids, (ii) the existence of a direct receptor-receptor interaction when the receptors are co-expressed in the same cells, and (iii) the interaction of their intracellular pathways. Finally, the cannabinoid/opioid interaction might be different in the brain rewarding networks and in those accounting for other pharmacological effects (antinociception, modulation of emotionality and cognitive behavior), as well as between the central nervous system and periphery. Further insights about the cannabinoid/opioid interaction could pave the way for new and promising therapeutic approaches.


Assuntos
Analgésicos Opioides/uso terapêutico , Encéfalo/efeitos dos fármacos , Canabinoides/uso terapêutico , Dor/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Analgésicos Opioides/efeitos adversos , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Moduladores de Receptores de Canabinoides/metabolismo , Canabinoides/efeitos adversos , Cognição/efeitos dos fármacos , Interações Medicamentosas , Emoções/efeitos dos fármacos , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/metabolismo , Peptídeos Opioides/metabolismo , Transtornos Relacionados ao Uso de Opioides/etiologia , Transtornos Relacionados ao Uso de Opioides/metabolismo , Dor/metabolismo , Dor/fisiopatologia , Receptor Cross-Talk , Receptores de Canabinoides/efeitos dos fármacos , Receptores de Canabinoides/metabolismo , Receptores Opioides/efeitos dos fármacos , Receptores Opioides/metabolismo , Recompensa , Síndrome de Abstinência a Substâncias/metabolismo
9.
Pharmacol Res ; 60(2): 132-8, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19559364

RESUMO

Marijuana is consistently the most widely used illicit drug among teenagers and most users first experiment it in adolescence. Adolescence is the period between childhood and adulthood, encompassing not only reproductive maturation, but also cognitive, emotional and social maturation and is characterized by a brain in transition that differs anatomically and neurochemically from that of the adult. The endocannabinoid system plays an important role in this critical phase for cerebral development, therefore a strong stimulation by the psychoactive component of marijuana, delta-9-tetrahydrocanabinol, that acts through the cannabinoid system, might lead to subtle but lasting neurobiological changes that can affect adult brain functions and behaviour. The literature here summarized, exploiting animal models of cannabis consumption, points to the presence of subtle changes in the adult brain circuits after heavy cannabis consumption in adolescence. These alterations lead to impaired emotional and cognitive performance, enhanced vulnerability for the use of more harmful drugs of abuse, and may represent a risk factor for developing schizophrenia in adulthood. The few studies examining the neurobiological basis of the altered behaviours demonstrate the presence of stable alteration in the endocannabinoid system that can trigger subsequent alteration in synaptic protein and synaptic morphology, thus altering the responsiveness of selected brain areas to different internal and external stimuli. These pre-clinical observations are strengthened by literature in humans where longitudinal studies often support the experimental results. There is an urgent need of multidisciplinary approaches combining behaviour with neurochemical and genetic studies to build a scientific based opinion on the long-lasting consequences of cannabis use in adolescence.


Assuntos
Comportamento/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Canabinoides/toxicidade , Transtornos Cognitivos/induzido quimicamente , Neurobiologia , Adolescente , Adulto , Encéfalo/fisiopatologia , Transtornos Cognitivos/fisiopatologia , Transtornos Cognitivos/psicologia , Humanos , Estudos Longitudinais
10.
Neurotox Res ; 15(4): 291-302, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19384563

RESUMO

We recently demonstrated that Delta(9)-tetrahydrocannabinol (THC) chronic administration in female adolescent rats induces alterations in the emotional circuit ending in depressive-like behavior in adulthood. Since cognitive dysfunction is a major component of depression, we assessed in these animals at adulthood different forms of memory. Adolescent female rats were treated with THC or its vehicle from 35 to 45 post-natal days (PND) and left undisturbed until their adulthood (75 PND) when aversive and spatial memory was assessed using the passive avoidance and radial maze tasks. No alteration was found in aversive memory, but in the radial maze THC pre-treated animals exhibited a worse performance than vehicles, suggesting a deficit in spatial working memory. To correlate memory impairment to altered neuroplasticity, level of marker proteins was investigated in the hippocampus and prefrontal cortex, the most relevant areas for learning and memory. A significant decrease in synaptophysin and PSD95 proteins was found in the prefrontal cortex of THC pre-treated rats, with no alterations in the hippocampus. Finally, proteomic analysis of the synapses in the prefrontal cortex revealed the presence of less active synapses characterized by reduced ability in maintaining normal synaptic efficiency. This picture demonstrates the presence of cognitive impairment in THC-induced depressive phenotype.


Assuntos
Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/complicações , Depressão/induzido quimicamente , Dronabinol/toxicidade , Plasticidade Neuronal/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Análise de Variância , Animais , Animais Recém-Nascidos , Área Sob a Curva , Aprendizagem da Esquiva/efeitos dos fármacos , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Córtex Pré-Frontal/fisiopatologia , Proteômica/métodos , Psicotrópicos/toxicidade , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Sinaptofisina/metabolismo , Tubulina (Proteína)/metabolismo
11.
Drug News Perspect ; 21(3): 149-57, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18560613

RESUMO

This review aims to present the more recent knowledge on the role of the endocannabinoid system in drug addiction. For a long time, dopamine has been consistently associated with the reinforcing effects of most drugs of abuse but, recently, pharmacological evidence points to the possibility that pharmacological management of the endocannabinoid system might not only block the direct reinforcing effect of cannabis, opioids, nicotine and ethanol, but also prevent the relapse to various drugs of abuse including opioids, cocaine, nicotine, alcohol and amphetamine. Preclinical and clinical studies suggest that the manipulation of the endocannabinoid system through the CB(1) receptor antagonist SR-141716A (rimonabant) might constitute a new therapeutical strategy for treating addiction across different classes of abused drugs.


Assuntos
Moduladores de Receptores de Canabinoides/fisiologia , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Alcoolismo/fisiopatologia , Alcoolismo/psicologia , Animais , Estimulantes do Sistema Nervoso Central , Humanos , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Transtornos Relacionados ao Uso de Opioides/psicologia , Transtornos Relacionados ao Uso de Substâncias/psicologia , Tabagismo/fisiopatologia , Tabagismo/psicologia
12.
Mol Cell Endocrinol ; 286(1-2 Suppl 1): S108-13, 2008 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-18358595

RESUMO

Despite the increasing use of cannabis among adolescents, there are little and often contradictory studies on the long-term neurobiological consequences of cannabis consumption in juveniles. Adolescence is a critical phase for cerebral development, where the endocannabinoid system plays an important role influencing the release and action of different neurotransmitters. Therefore, a strong stimulation by the psychoactive component of marijuana, delta-9-tetrahydrocanabinol (THC), might lead to subtle but lasting neurobiological changes that can affect adult brain functions and behaviour. The literature here summarized by use of experimental animal models, puts forward that heavy cannabis consumption in adolescence may induce subtle changes in the adult brain circuits ending in altered emotional and cognitive performance, enhanced vulnerability for the use of more harmful drugs of abuse in selected individuals, and may represent a risk factor for developing schizophrenia in adulthood. Therefore, the potential problems arising in relation to marijuana consumption in adolescence suggest that this developmental phase is a vulnerable period for persistent adverse effects of cannabinoids.


Assuntos
Comportamento/efeitos dos fármacos , Cannabis/efeitos adversos , Adolescente , Animais , Humanos , Modelos Biológicos , Transtornos Psicóticos/etiologia
13.
J Neurochem ; 104(4): 1091-100, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18028339

RESUMO

It has been recently reported that cannabidiol (CBD), a non-psychoactive cannabinoid, is able to kill glioma cells, both in vivo and in vitro, independently of cannabinoid receptor stimulation. However, the underlying biochemical mechanisms were not clarified. In the present study, we performed biochemical analysis of the effect of CBD both in vivo, by using glioma tumor tissues excised from nude mice, and in vitro, by using U87 glioma cells. In vivo exposure of tumor tissues to CBD significantly decreased the activity and content of 5-lipoxygenase (LOX, by approximately 40%), and of its end product leukotriene B4 ( approximately 25%). In contrast cyclooxygenase (COX)-2 activity and content, and the amount of its end product prostaglandin E2, were not affected by CBD. In addition, in vivo treatment with CBD markedly stimulated ( approximately 175%) the activity of fatty acid amide hydrolase (FAAH), the main anandamide-degrading enzyme, while decreasing anandamide content ( approximately 30%) and binding to CB1 cannabinoid receptors ( approximately 25%). In vitro pre-treatment of U87 glioma cells with MK-886, a specific 5-LOX inhibitor, significantly enhanced the antimitotic effect of CBD, whereas the pre-treatment with indomethacin (pan-COX inhibitor) or celecoxib (COX-2 inhibitor), did not alter CBD effect. The study of the endocannabinoid system revealed that CBD was able to induce a concentration-dependent increase of FAAH activity in U87 cells. Moreover, a significantly reduced growth rate was observed in FAAH-over-expressing U87 cells, compared to wild-type controls. In conclusion, the present investigation indicates that CBD exerts its antitumoral effects through modulation of the LOX pathway and of the endocannabinoid system, suggesting a possible interaction of these routes in the control of tumor growth.


Assuntos
Amidoidrolases/fisiologia , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Araquidonato 5-Lipoxigenase/fisiologia , Canabidiol/metabolismo , Canabidiol/farmacologia , Animais , Canabinoides/metabolismo , Canabinoides/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
14.
Cereb Cortex ; 18(6): 1292-301, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17921459

RESUMO

In the present study we explored with a multidisciplinary approach, the role of anandamide (AEA) in the modulation of anxiety behavior at the level of the prefrontal cortex (PFC). Low doses of the metabolically stable AEA analog, methanandamide, microinjected into the PFC, produced an anxiolytic-like response in rats, whereas higher doses induced anxiety-like behaviors. Pretreatment with the selective antagonist of CB1 or TRPV1 receptors (AM251 and capsazepine, respectively) suggested that the anxiolytic effect evoked by AEA might be due to the interaction with the CB1 cannabinoid receptor, whereas vanilloid receptors seem to be involved in AEA anxiogenic action. When AEA contents in the PFC were increased by microinjecting the selective inhibitor of fatty acid amide hydrolase (FAAH), URB597, we observed an anxiolytic response only at low doses of the compound and no effect or even an anxiogenic profile at higher doses. In line with this, a marked decrease of AEA levels in the PFC, achieved by lentivirus-mediated local overexpression of FAAH, produced an anxiogenic response. These findings support an anxiolytic role for physiological increases in AEA in the PFC, whereas more marked increases or decreases of this endocannabinoid might lead to an anxiogenic response due to TRPV1 stimulation or the lack of CB1 activation, respectively.


Assuntos
Ansiedade/fisiopatologia , Moduladores de Receptores de Canabinoides/fisiologia , Endocanabinoides , Córtex Pré-Frontal/fisiologia , Animais , Ansiedade/psicologia , Ácidos Araquidônicos/farmacologia , Benzamidas/farmacologia , Carbamatos/farmacologia , Relação Dose-Resposta a Droga , Masculino , Alcamidas Poli-Insaturadas/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/fisiologia
15.
Neuropharmacology ; 54(1): 151-60, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17692344

RESUMO

There is a general consensus that the effects of cannabinoid agonists on anxiety seem to be biphasic, with low doses being anxiolytic and high doses ineffective or possibly anxiogenic. Besides the behavioural effects of cannabinoids on anxiety, very few papers have dealt with the neuroanatomical sites of these effects. We investigated the effect on rat anxiety behavior of local administration of THC in the prefrontal cortex, basolateral amygdala and ventral hippocampus, brain regions belonging to the emotional circuit and containing high levels of CB1 receptors. THC microinjected at low doses in the prefrontal cortex (10 microg) and ventral hippocampus (5 microg) induced in rats an anxiolytic-like response tested in the elevated plus-maze, whilst higher doses lost the anxiolytic effect and even seemed to switch into an anxiogenic profile. Low THC doses (1 microg) in the basolateral amygdala produced an anxiogenic-like response whereas higher doses were ineffective. All these effects were CB1-dependent and closely linked to modulation of CREB activation. Specifically, THC anxiolytic activity in the prefrontal cortex and ventral hippocampus was paralleled by an increase in CREB activation, whilst THC anxiogenic response in the basolateral amygdala was paralleled by a decrease in CREB activation. Our results suggest that while a mild activation of CB1 receptors in the prefrontal cortex and ventral hippocampus attenuates anxiety, a slight CB1 receptor stimulation in the amygdala results in an anxiogenic-like response. The molecular underpinnings of these effects involve a direct stimulation of CB1 receptors ending in pCREB modulation and/or a possible alteration in the fine tuning of local neuromodulator release.


Assuntos
Ansiedade/patologia , Ansiedade/fisiopatologia , Encéfalo/fisiopatologia , Receptor CB1 de Canabinoide/fisiologia , Tonsila do Cerebelo/efeitos dos fármacos , Analgésicos não Narcóticos , Análise de Variância , Animais , Ansiedade/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Proteína de Ligação a CREB/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Dronabinol/uso terapêutico , Hipocampo/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Microinjeções/métodos , Atividade Motora/efeitos dos fármacos , Piperidinas/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Pirazóis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor CB1 de Canabinoide/efeitos dos fármacos , Fatores de Tempo
16.
Cell Mol Life Sci ; 63(17): 2057-66, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16909207

RESUMO

Recently, we have shown that the non-psychoactive cannabinoid compound cannabidiol (CBD) induces apoptosis of glioma cells in vitro and tumor regression in vivo. The present study investigated a possible involvement of caspase activation and reactive oxygen species (ROS) induction in the apoptotic effect of CBD. CBD produced a gradual, time-dependent activation of caspase-3, which preceded the appearance of apoptotic death. In addiction, release of cytochrome c and caspase-9 and caspase-8 activation were detected. The exposure to CBD caused in glioma cells an early production of ROS, depletion of intracellular glutathione and increase activity of glutathione reductase and glutathione peroxidase enzymes. Under the same experimental condition, CBD did not impair primary glia. Thus, we found a different sensitivity to the anti-proliferative effect of CBD in human glioma cells and non-transformed cells that appears closely related to a selective ability of CBD in inducing ROS production and caspase activation in tumor cells.


Assuntos
Canabidiol/farmacologia , Caspases/metabolismo , Glioma/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Ativação Enzimática , Glutationa/metabolismo , Humanos
17.
Curr Pharm Des ; 12(24): 3135-46, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16918439

RESUMO

How cannabinoids influence immune function has been examined extensively in the last 30 years. Studies on drug-abusing humans and animals, as well as in vitro models employing immune cell cultures, have shown that marijuana, natural and endogenous cannabinoid compounds are immunomodulators. These substances modulate host resistance to bacterial, protozoan and viral infections as well as they can profoundly affect the Th1/Th2 response. Recently, two types of cannabinoid receptor, CB1 and CB2, have been discovered. While CB1 is expressed primarily in the brain, CB2 is peculiar of the immune cells. Cannabinoid receptors have been shown to be involved in some but not all of immune effects. Nevertheless, their identification provides a specific mechanism of action in the attempting to find out how exogenous cannabinoids and endogenous cannabinoid system affect the immune apparatus, strengthen the hypothesis of cannabinoids as immunomodulators. As support to this theory, enough evidence exists to suggest that the cannabinoid system significantly affects almost every component of the immune response machinery and impacts the functioning also of the cytokine network. The evaluation of the biological consequences of these drug-induced cytokine changes has also dramatically become important considering not only the impact of cytokines on immune system per se but also envisaging their influence in cancer, inflammation, autoimmune disease, brain injury, hematopoietic colony formation in which cannabinoids have demonstrated a clear role as important modulators.


Assuntos
Canabinoides/farmacologia , Citocinas/fisiologia , Sistema Imunitário/efeitos dos fármacos , Animais , Canabinoides/química , Humanos , Fatores Imunológicos/química , Fatores Imunológicos/farmacologia , Modelos Biológicos , Estrutura Molecular , Transdução de Sinais/efeitos dos fármacos
18.
J Neuroimmunol ; 159(1-2): 97-105, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15652407

RESUMO

Cannabidiol is the main nonpsychoactive component of marijuana. We examined the ability of in vivo and in vitro cannabidiol to interfere with the production of interleukin (IL)-12 and IL-10 by murine macrophages and to modulate macrophage chemotaxis. Cannabidiol added in vitro to peritoneal macrophages significantly increased IL-12 and decreased IL-10 production. The CB1 and CB2 receptor antagonists prevented this modulation. Macrophages from animals treated with cannabidiol at the dose of 30 mg kg(-1) either orally or i.p. produced higher levels of IL-12 and lower levels of IL-10 in comparison to controls, and the CB receptor antagonists did not prevent these effects. Cannabidiol dose-dependently decreased fMLP-induced chemotaxis of macrophages, and the CB2 receptor antagonist prevented this decrease.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacologia , Canabidiol/administração & dosagem , Canabidiol/farmacologia , Quimiotaxia/efeitos dos fármacos , Interleucina-10/biossíntese , Interleucina-12/biossíntese , Macrófagos Peritoneais/efeitos dos fármacos , Adjuvantes Imunológicos/metabolismo , Administração Oral , Animais , Canfanos/farmacologia , Canabidiol/metabolismo , Inibição de Migração Celular , Células Cultivadas , Quimiotaxia/imunologia , Citocinas/biossíntese , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Interleucina-10/antagonistas & inibidores , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/fisiologia , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/fisiologia , Rimonabanto , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
19.
Cell Mol Life Sci ; 61(7-8): 945-50, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15095014

RESUMO

The endogenous ligands of cannabinoid receptors, also known as endocannabinoids, have been implicated in many physiological and pathological processes of the central nervous system. Here we show that the levels of the two major endocannabinoids, anandamide and 2-arachidonoyl-glycerol (2-AG), in four areas of the rat brain, change dramatically between the light and dark phases of the day. While anandamide levels in the nucleus accumbens, pre-frontal cortex, striatum and hippocampus were significantly higher in the dark phase, the opposite was observed with 2-AG, whose levels were significantly higher during the light phase in all four regions. We found that the activity of the fatty acid amide hydrolase, which catalyzes the metabolism of anandamide, was significantly lower during the dark phase, thus providing a possible explaination for the increase in anandamide levels. However, the activities of monoacylglycerol lipase and diacylglycerol lipase, two of the possible enzymes catalyzing the degradation and biosynthesis of 2-AG, respectively, changed significantly only in the striatum. These data suggest that the levels of the two major endocannabinoids might be under the control of endogenous factors known to undergo diurnal variations, and underscore the different roles, suggested by previous studies, of anandamide and 2-AG in neurophysiological processes.


Assuntos
Ácidos Araquidônicos/metabolismo , Encéfalo/metabolismo , Moduladores de Receptores de Canabinoides/metabolismo , Ritmo Circadiano/fisiologia , Glicerídeos/metabolismo , Amidoidrolases/metabolismo , Animais , Encéfalo/anatomia & histologia , Endocanabinoides , Masculino , Fotoperíodo , Alcamidas Poli-Insaturadas , Ratos , Ratos Sprague-Dawley
20.
Neuroscience ; 117(4): 921-9, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12654343

RESUMO

We used a previously reported model of morphine sensitization that elicited a complex behavioral syndrome involving stereotyped and non stereotyped activity. To identify the mechanism of these long-lasting processes, we checked the density of mu opioid receptors, receptor-G-protein coupling and the cyclic AMP (cAMP) cascade. In morphine-sensitized animals mu opioid receptor autoradiography revealed a significant increase in the caudate putamen (30% versus controls), nucleus accumbens shell (16%), prefrontal and frontal cortex (26%), medial thalamus (43%), hypothalamus (200%) and central gray (89%). Concerning morphine's activation of G proteins in the brain, investigated in the guanylyl 5'-[gamma-(35)S]thio]triphosphate ([(35)S]GTPgammaS) binding assay, a significant increase in net [(35)S]GTPgammaS binding was seen in the caudate putamen (39%) and hypothalamus (27%). In the caudate putamen this was due to an increase in the amount of activated G proteins, and in the hypothalamus to a greater affinity of G proteins for guanosine triphosphate (GTP). The main second messenger system linked to the opioid receptor is the cAMP pathway. In the striatum basal cAMP levels were significantly elevated in sensitized animals (70% versus controls) and [D-Ala(2), N-Me-Phe(4), Gly(5)-ol]-enkephalin (DAMGO) significantly inhibited forskolin-stimulated cAMP production in control (30%) but not in sensitized rats. In the hypothalamus no significant changes were observed in basal cAMP levels and DAMGO inhibition. These cellular events induced by morphine pre-exposure could underlie the neuroadaptive processes involved in morphine sensitization.


Assuntos
Química Encefálica/fisiologia , Encéfalo/efeitos dos fármacos , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides/metabolismo , Receptores Opioides mu/efeitos dos fármacos , Receptores Opioides mu/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Ligação Competitiva/fisiologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Química Encefálica/efeitos dos fármacos , AMP Cíclico/metabolismo , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Proteínas de Ligação ao GTP/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato) , Masculino , Neurônios/metabolismo , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...