Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Molecules ; 29(7)2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38611871

RESUMO

Oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) are endogenous lipids that act as agonists of the peroxisome proliferator-activated receptor α (PPARα). Recently, an interest in the role of these lipids in malignant tumors has emerged. Nevertheless, the effects of OEA and PEA on human neuroblastoma cells are still not documented. Type I interferons (IFNs) are immunomodulatory cytokines endowed with antiviral and anti-proliferative actions and are used in the treatment of various pathologies such as different cancer forms (i.e., non-Hodgkin's lymphoma, melanoma, leukemia), hepatitis B, hepatitis C, multiple sclerosis, and many others. In this study, we investigated the effect of OEA and PEA on human neuroblastoma SH-SY5Y cells treated with IFNß. We focused on evaluating cell viability, cell proliferation, and cell signaling. Co-exposure to either OEA or PEA along with IFNß leads to increased apoptotic cell death marked by the cleavage of caspase 3 and poly-(ADP ribose) polymerase (PARP) alongside a decrease in survivin and IKBα levels. Moreover, we found that OEA and PEA did not affect IFNß signaling through the JAK-STAT pathway and the STAT1-inducible protein kinase R (PKR). OEA and PEA also increased the phosphorylation of p38 MAP kinase and programmed death-ligand 1 (PD-L1) expression both in full cell lysate and surface membranes. Furthermore, GW6471, a PPARα inhibitor, and the genetic silencing of the receptor were shown to lower PD-L1 and cleaved PARP levels. These results reveal the presence of a novel mechanism, independent of the IFNß-prompted pathway, by which OEA and PEA can directly impair cell survival, proliferation, and clonogenicity through modulating and potentiating the intrinsic apoptotic pathway in human SH-SY5Y cells.


Assuntos
Amidas , Endocanabinoides , Etanolaminas , Neuroblastoma , Ácidos Oleicos , Humanos , Neuroblastoma/tratamento farmacológico , Antígeno B7-H1 , Janus Quinases , PPAR alfa , Inibidores de Poli(ADP-Ribose) Polimerases , Fatores de Transcrição STAT , Transdução de Sinais , Apoptose , Ácidos Palmíticos/farmacologia
2.
Nutrients ; 15(9)2023 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-37432348

RESUMO

The pathophysiology of Anorexia Nervosa (AN) has not been fully elucidated. Anaplastic lymphoma kinase (ALK) receptor is a protein-tyrosine kinase mainly known as a key oncogenic driver. Recently, a genetic deletion of ALK in mice has been found to increase energy expenditure and confers resistance to obesity in these animals, suggesting its role in the regulation of thinness. Here, we investigated the expression of ALK and the downstream intracellular pathways in female rats subjected to the activity-based anorexia (ABA) model, which reproduces important features of human AN. In the hypothalamic lysates of ABA rats, we found a reduction in ALK receptor expression, a downregulation of Akt phosphorylation, and no change in the extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) phosphorylation. After the recovery from body weight loss, ALK receptor expression returned to the control baseline values, while it was again suppressed during a second cycle of ABA induction. Overall, this evidence suggests a possible involvement of the ALK receptor in the pathophysiology of AN, that may be implicated in its stabilization, resistance, and/or its exacerbation.


Assuntos
Anorexia Nervosa , Humanos , Feminino , Animais , Camundongos , Ratos , Quinase do Linfoma Anaplásico , Anorexia , Proteínas Tirosina Quinases , Fosforilação
3.
Cells ; 11(17)2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-36078036

RESUMO

Parkinson's disease (PD) is a complex pathology causing a plethora of non-motor symptoms besides classical motor impairments, including cognitive disturbances. Recent studies in the PD human brain have reported microgliosis in limbic and neocortical structures, suggesting a role for neuroinflammation in the development of cognitive decline. Yet, the mechanism underlying the cognitive pathology is under investigated, mainly for the lack of a valid preclinical neuropathological model reproducing the disease's motor and non-motor aspects. Here, we show that the bilateral intracerebral infusion of pre-formed human alpha synuclein oligomers (H-αSynOs) within the substantia nigra pars compacta (SNpc) offers a valid model for studying the cognitive symptoms of PD, which adds to the classical motor aspects previously described in the same model. Indeed, H-αSynOs-infused rats displayed memory deficits in the two-trial recognition task in a Y maze and the novel object recognition (NOR) test performed three months after the oligomer infusion. In the anterior cingulate cortex (ACC) of H-αSynOs-infused rats the in vivo electrophysiological activity was altered and the expression of the neuron-specific immediate early gene (IEG) Npas4 (Neuronal PAS domain protein 4) and the AMPA receptor subunit GluR1 were decreased. The histological analysis of the brain of cognitively impaired rats showed a neuroinflammatory response in cognition-related regions such as the ACC and discrete subareas of the hippocampus, in the absence of any evident neuronal loss, supporting a role of neuroinflammation in cognitive decline. We found an increased GFAP reactivity and the acquisition of a proinflammatory phenotype by microglia, as indicated by the increased levels of microglial Tumor Necrosis Factor alpha (TNF-α) as compared to vehicle-infused rats. Moreover, diffused deposits of phospho-alpha synuclein (p-αSyn) and Lewy neurite-like aggregates were found in the SNpc and striatum, suggesting the spreading of toxic protein within anatomically interconnected areas. Altogether, we present a neuropathological rat model of PD that is relevant for the study of cognitive dysfunction featuring the disease. The intranigral infusion of toxic oligomeric species of alpha-synuclein (α-Syn) induced spreading and neuroinflammation in distant cognition-relevant regions, which may drive the altered neuronal activity underlying cognitive deficits.


Assuntos
Disfunção Cognitiva , Doença de Parkinson , Animais , Disfunção Cognitiva/metabolismo , Giro do Cíngulo/metabolismo , Giro do Cíngulo/patologia , Humanos , Doenças Neuroinflamatórias , Neurônios/metabolismo , Doença de Parkinson/metabolismo , Ratos , Substância Negra/metabolismo , alfa-Sinucleína/metabolismo
4.
Commun Biol ; 5(1): 811, 2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-35962064

RESUMO

Muscularis Externa Macrophages (ME-Macs) and enteric glial cells (EGCs) are closely associated cell types in the bowel wall, and important interactions are thought to occur between them during intestinal inflammation. They are involved in developing postoperative ileus (POI), an acute, surgery-induced inflammatory disorder triggered by IL-1 receptor type I (IL1R1)-signaling. In this study, we demonstrate that IL1R1-signaling in murine and human EGCs induces a reactive state, named enteric gliosis, characterized by a strong induction of distinct chemokines, cytokines, and the colony-stimulating factors 1 and 3. Ribosomal tagging revealed enteric gliosis as an early part of POI pathogenesis, and mice with an EGC-restricted IL1R1-deficiency failed to develop postoperative enteric gliosis, showed diminished immune cell infiltration, and were protected from POI. Furthermore, the IL1R1-deficiency in EGCs altered the surgery-induced glial activation state and reduced phagocytosis in macrophages, as well as their migration and accumulation around enteric ganglia. In patients, bowel surgery also induced IL-1-signaling, key molecules of enteric gliosis, and macrophage activation. Together, our data show that IL1R1-signaling triggers enteric gliosis, which results in ME-Mac activation and the development of POI. Intervention in this pathway might be a useful prophylactic strategy in preventing such motility disorders and gut inflammation.


Assuntos
Motilidade Gastrointestinal , Íleus , Animais , Gliose/complicações , Gliose/patologia , Humanos , Íleus/etiologia , Íleus/prevenção & controle , Inflamação/patologia , Interleucina-1 , Macrófagos/metabolismo , Camundongos , Complicações Pós-Operatórias/etiologia
5.
Brain Sci ; 11(3)2021 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-33673559

RESUMO

Brain tumors can present with various psychiatric symptoms, with or without neurological symptoms, an aspect that complicates the clinical picture. However, no systematic description of symptoms that should prompt a neurological investigation has been provided. This review aims to summarize available case reports describing patients with brain tumors showing psychiatric symptoms before brain tumor diagnosis, in order to provide a comprehensive description of these symptoms as well as their potential relationship with delay in the diagnosis. A systematic literature review on case reports of brain tumors and psychiatric symptoms from 1970 to 2020 was conducted on PubMed, Ovid, Psych Info, and MEDLINE. Exclusion criteria comprised tumors not included in the World Health Organization (WHO) Classification 4th edition and cases in which psychiatric symptoms were absent or followed the diagnosis. A total of 165 case reports were analyzed. In a subset of patients with brain tumors, psychiatric symptoms can be the only manifestation or precede focal neurological signs by months or even years. The appearance of focal or generalized neurological symptoms after, rather than along with, psychiatric symptoms was associated with a significant delay in the diagnosis in adults. A timely assessment of psychiatric symptoms might help to improve early diagnosis of brain tumors.

6.
Proc Natl Acad Sci U S A ; 117(18): 9991-10002, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32312805

RESUMO

The initial response to an addictive substance can facilitate repeated use: That is, individuals experiencing more positive effects are more likely to use that drug again. Increasing evidence suggests that psychoactive cannabinoid use in adolescence enhances the behavioral effects of cocaine. However, despite the behavioral data, there is no neurobiological evidence demonstrating that cannabinoids can also alter the brain's initial molecular and epigenetic response to cocaine. Here, we utilized a multiomics approach (epigenomics, transcriptomics, proteomics, and phosphoproteomics) to characterize how the rat brain responds to its first encounter with cocaine, with or without preexposure to the synthetic cannabinoid WIN 55,212-2 (WIN). We find that in adolescent (but not in adult) rats, preexposure to WIN results in cross-sensitization to cocaine, which correlates with histone hyperacetylation and decreased levels of HDAC6 in the prefrontal cortex (PFC). In the PFC, we also find that WIN preexposure blunts the typical mRNA response to cocaine and instead results in alternative splicing and chromatin accessibility events, involving genes such as Npas2 Moreover, preexposure to WIN enhances the effects of cocaine on protein phosphorylation, including ERK/MAPK-targets like gephyrin, and modulates the synaptic AMPAR/GluR composition both in the PFC and the nucleus accumbens (NAcc). PFC-NAcc gene network topological analyses, following cocaine exposure, reveal distinct top nodes in the WIN preexposed group, which include PACAP/ADCYAP1. These preclinical data demonstrate that adolescent cannabinoid exposure reprograms the initial behavioral, molecular, and epigenetic response to cocaine.


Assuntos
Comportamento Aditivo/genética , Comportamento Animal/efeitos dos fármacos , Canabinoides/efeitos adversos , Cocaína/efeitos adversos , Adolescente , Animais , Comportamento Aditivo/induzido quimicamente , Comportamento Aditivo/patologia , Benzoxazinas/efeitos adversos , Benzoxazinas/farmacologia , Canabinoides/farmacologia , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Cocaína/farmacologia , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Desacetilase 6 de Histona/genética , Humanos , Proteínas de Membrana/farmacologia , Morfolinas/efeitos adversos , Morfolinas/farmacologia , Naftalenos/efeitos adversos , Naftalenos/farmacologia , Fosfoproteínas/efeitos dos fármacos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Córtex Pré-Frontal/efeitos dos fármacos , Proteoma/efeitos dos fármacos , Ratos , Transcriptoma/efeitos dos fármacos
7.
Neuropharmacology ; 144: 327-336, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30439418

RESUMO

Tobacco smoke is the leading preventable cause of death in the world and treatments aimed to increase success rate in smoking cessation by reducing nicotine dependence are sought. Activation of peroxisome proliferator-activated receptor-alpha (PPARα) by synthetic or endogenous agonists was shown to suppress nicotine-induced activation of mesolimbic dopamine system, one of the major neurobiological substrates of nicotine dependence, and nicotine-seeking behavior in rats and monkeys. An alternative indirect way to activate PPARα is inhibition of N-acylethanolamine acid amidase (NAAA), one of the major hydrolyzing enzyme for its endogenous agonists palmitoylethanolamide (PEA) and oleoylethanolamide (OEA). We synthetized a novel specific brain permeable NAAA inhibitor, AM11095. We administered AM11095 to rats and carried out brain lipid analysis, a functional observational battery (FOB) to assess toxicity, in vivo electrophysiological recording from dopamine cells in the ventral tegmental area, brain microdialysis in the nucleus accumbens shell and behavioral experiments to assess its effect on nicotine -induced conditioned place preference (CPP). AM11095 (5 and 25 mg/kg, i.p.) was devoid of neurotoxic and behavioral effects and did not affect motor behavior and coordination. This NAAA inhibitor (5 mg/kg i.p.) increased OEA and PEA levels in the hippocampus and cortex, prevented nicotine-induced activation of mesolimbic dopamine neurons in the ventral tegmental area, nicotine-induced elevation of dopamine levels in the nucleus accumbens shell and decreased the expression of nicotine CPP. Our results indicate that NAAA inhibitors represent a new class of pharmacological tools to modulate brain PEA/PPARα signalling and show potential in the treatment of nicotine dependence.


Assuntos
Amidoidrolases/antagonistas & inibidores , Dopamina/metabolismo , Nicotina/farmacologia , Psicotrópicos/farmacologia , Recompensa , Amidoidrolases/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/toxicidade , Masculino , Camundongos , Agonistas Nicotínicos/farmacologia , Distribuição Aleatória , Ratos Sprague-Dawley , Comportamento Espacial/efeitos dos fármacos , Comportamento Espacial/fisiologia
8.
Psychopharmacology (Berl) ; 233(10): 1765-77, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26728894

RESUMO

RATIONALE: Several lines of evidence suggest that endocannabinoid and nicotinic cholinergic systems are implicated in the regulation of different physiological processes, including reward, and in the neuropathological mechanisms of psychiatric diseases, such as addiction. A crosstalk between these two systems is substantiated by the overlapping distribution of cannabinoid and nicotinic acetylcholine receptors in many brain structures. OBJECTIVE: We will review recent preclinical data showing how the endocannabinoid and nicotinic cholinergic systems interact bidirectionally at the level of the brain reward pathways, and how this interaction plays a key role in modulating nicotine and cannabinoid intake and dependence. RESULTS: Many behavioral and neurochemical effects of nicotine that are related to its addictive potential are reduced by pharmacological blockade or genetic deletion of type-1 cannabinoid receptors, inhibition of endocannabinoid uptake or metabolic degradation, and activation of peroxisome proliferator-activated-receptor-α. On the other hand, cholinergic antagonists at α7 nicotinic acetylcholine receptors as well as endogenous negative allosteric modulators of these receptors are effective in blocking dependence-related effects of cannabinoids. CONCLUSIONS: Pharmacological manipulation of the endocannabinoid system and endocannabinoid-like neuromodulators shows promise in the treatment of nicotine dependence and in relapse prevention. Likewise, drugs acting at nicotinic acetylcholine receptors might prove useful in the therapy of cannabinoid dependence. Research by Steven R. Goldberg has significantly contributed to the progress in this research field.


Assuntos
Encéfalo/fisiologia , Endocanabinoides/fisiologia , Receptores Nicotínicos/fisiologia , Animais , Comportamento Aditivo/metabolismo , Comportamento Aditivo/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Endocanabinoides/metabolismo , Humanos , Receptores Nicotínicos/metabolismo , Recompensa , Tabagismo/metabolismo , Tabagismo/fisiopatologia
9.
Curr Pharm Des ; 20(13): 2100-13, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23829370

RESUMO

Sex-dependent differences are frequently observed in the biological and behavioural effects of substances of abuse, including cannabis. We recently demonstrated a modulating effect of sex and oestrous cycle on cannabinoid-taking and seeking behaviours. Here, we investigated the influence of sex and oestrogen in the regulation of cannabinoid CB1 receptor density and function, measured by [(3)H]CP55940 and CP55940-stimulated [(35)S]GTPγS binding autoradiography, respectively, in the prefrontal cortex (Cg1 and Cg3), caudate- putamen, nucleus accumbens, amygdala and hippocampus of male and cycling female rats, as well as ovariectomised (OVX) rats and OVX rats primed with oestradiol (10 µg/rat) (OVX+E). CB1 receptor density was significantly lower in the prefrontal cortex and amygdala of cycling females than in males and in OVX females, a difference that appeared to be oestradiol-dependent, because it was no more evident in the OVX+E group. CP55940-stimulated [(35)S]GTPγS binding was significantly higher in the Cg3 of OVX rats relative to cycling and OVX+E rats. No difference was observed in CB1 receptor density or function in any of the other brain areas analysed. Finally, sex and oestradiol were also found to affect motor activity, social behaviour and sensorimotor gating in rats tested in locomotor activity boxes, social interaction and prepulse inhibition tasks, respectively. Our findings provide biochemical evidence for sex- and hormone- dependent differences in the density and function of CB1 receptors in selected brain regions, and in behaviours associated with greater vulnerability to drug addiction, revealing a more vulnerable behavioural phenotype in female than in male rats.


Assuntos
Encéfalo/fisiologia , Estrogênios/farmacologia , Receptor CB1 de Canabinoide/análise , Transtornos Relacionados ao Uso de Substâncias/etiologia , Animais , Feminino , Masculino , Atividade Motora/efeitos dos fármacos , Ovariectomia , Ratos , Receptor CB1 de Canabinoide/fisiologia , Reflexo de Sobressalto/efeitos dos fármacos , Caracteres Sexuais , Habilidades Sociais
10.
Br J Pharmacol ; 165(8): 2539-48, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21557729

RESUMO

BACKGROUND AND PURPOSE: The fatty acid amide hydrolase inhibitor URB597 can reverse the abuse-related behavioural and neurochemical effects of nicotine in rats. Fatty acid amide hydrolase inhibitors block the degradation (and thereby magnify and prolong the actions) of the endocannabinoid anandamide (AEA), and also the non-cannabinoid fatty acid ethanolamides oleoylethanolamide (OEA) and palmitoylethanolamide (PEA). OEA and PEA are endogenous ligands for peroxisome proliferator-activated receptors alpha (PPAR-α). Since recent evidence indicates that PPAR-α can modulate nicotine reward, it is unclear whether AEA plays a role in the effects of URB597 on nicotine reward. EXPERIMENTAL APPROACH: A way to selectively increase endogenous levels of AEA without altering OEA or PEA levels is to inhibit AEA uptake into cells by administering the AEA transport inhibitor N-(4-hydroxyphenyl)-arachidonamide (AM404). To clarify AEA's role in nicotine reward, we investigated the effect of AM404 on conditioned place preference (CPP), reinstatement of abolished CPP, locomotor suppression and anxiolysis in an open field, and dopamine elevations in the nucleus accumbens shell induced by nicotine in Sprague-Dawley rats. KEY RESULTS: AM404 prevented the development of nicotine-induced CPP and impeded nicotine-induced reinstatement of the abolished CPP. Furthermore, AM404 reduced nicotine-induced increases in dopamine levels in the nucleus accumbens shell, the terminal area of the brain's mesolimbic reward system. AM404 did not alter the locomotor suppressive or anxiolytic effect of nicotine. CONCLUSIONS AND IMPLICATIONS: These findings suggest that AEA transport inhibition can counteract the addictive effects of nicotine and that AEA transport may serve as a new target for development of medications for treatment of tobacco dependence. LINKED ARTICLES: This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visit http://dx.doi.org/10.1111/bph.2011.163.issue-7.


Assuntos
Ácidos Araquidônicos/antagonistas & inibidores , Ácidos Araquidônicos/uso terapêutico , Comportamento Aditivo/tratamento farmacológico , Nicotina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Alcamidas Poli-Insaturadas/antagonistas & inibidores , Animais , Ácidos Araquidônicos/farmacologia , Ácidos Araquidônicos/fisiologia , Comportamento Aditivo/fisiopatologia , Comportamento Animal/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Dopamina/metabolismo , Dronabinol/farmacologia , Endocanabinoides , Masculino , Atividade Motora/efeitos dos fármacos , Agonistas Nicotínicos/farmacologia , Núcleo Accumbens/fisiologia , Ratos , Ratos Sprague-Dawley , Recompensa
11.
Biol Psychiatry ; 69(7): 633-41, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20801430

RESUMO

BACKGROUND: Recent findings indicate that inhibitors of fatty acid amide hydrolase (FAAH) counteract the rewarding effects of nicotine in rats. Inhibition of FAAH increases levels of several endogenous substances in the brain, including the endocannabinoid anandamide and the noncannabinoid fatty acid ethanolamides oleoylethanolamide (OEA) and palmitoylethanolamide, which are ligands for alpha-type peroxisome proliferator-activated nuclear receptors (PPAR-α). Here, we evaluated whether directly acting PPAR-α agonists can modulate reward-related effects of nicotine. METHODS: We combined behavioral, neurochemical, and electrophysiological approaches to evaluate effects of the PPAR-α agonists [[4-Chloro-6-[(2,3-dimethylphenyl)amino]-2-pyrimidinyl]thio]acetic acid (WY14643) and methyl oleoylethanolamide (methOEA; a long-lasting form of OEA) on 1) nicotine self-administration in rats and squirrel monkeys; 2) reinstatement of nicotine-seeking behavior in rats and monkeys; 3) nicotine discrimination in rats; 4) nicotine-induced electrophysiological activity of ventral tegmental area dopamine neurons in anesthetized rats; and 5) nicotine-induced elevation of dopamine levels in the nucleus accumbens shell of freely moving rats. RESULTS: The PPAR-α agonists dose-dependently decreased nicotine self-administration and nicotine-induced reinstatement in rats and monkeys but did not alter food- or cocaine-reinforced operant behavior or the interoceptive effects of nicotine. The PPAR-α agonists also dose-dependently decreased nicotine-induced excitation of dopamine neurons in the ventral tegmental area and nicotine-induced elevations of dopamine levels in the nucleus accumbens shell of rats. The ability of WY14643 and methOEA to counteract the behavioral, electrophysiological, and neurochemical effects of nicotine was reversed by the PPAR-α antagonist 1-[(4-Chlorophenyl)methyl]-3-[(1,1-dimethylethyl)thio]-a,a-dimethyl-5-(1-methylethyl)-1H-Indole-2-propanoic acid (MK886). CONCLUSIONS: These findings indicate that PPAR-α might provide a valuable new target for antismoking medications.


Assuntos
Nicotina/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , PPAR alfa/metabolismo , Reforço Psicológico , Recompensa , Potenciais de Ação/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Masculino , Microdiálise/métodos , Neurônios/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Oligossacarídeos/farmacologia , Proliferadores de Peroxissomos/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Saimiri , Autoadministração , Área Tegmentar Ventral/citologia
12.
Psychoneuroendocrinology ; 34 Suppl 1: S227-36, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19744795

RESUMO

Many studies have provided evidence for important sex-dependent differences in the origins, outcomes and treatment of drug abuse and dependence. Preclinical studies typically have employed animal models of addiction, such as oral or intravenous self-administration, to untangle the environmental, neurobiological and genetic factors that contribute to the shift from occasional, recreational use to compulsive, uncontrolled intake of drugs. Craving and relapse of drug seeking in abstinent individuals have also been found to differ between men and women. Identification of the neurobiological basis of craving and drug dependence continues to pose a challenge to addiction research. Significant sex differences are emerging in substance-abuse-related behavior, which has increased the demand for research on how drug consumption may have different causes, progression and consequences in men and women. In keeping with epidemiological data in humans, differences between the two sexes in drug seeking and intake have been well-documented in animal studies, with most recent findings related to abuse of cannabinoids. Clinical and preclinical findings indicate that sex and gonadal hormones may account for individual differences in susceptibility to the reinforcing effects of addictive substances, and that differences in vulnerability to drug abuse may be mediated by the same biological mechanisms. This review focuses on the differences between males and females in relation to drug self-administration and how such behavior may be affected by hormonal status.


Assuntos
Canabinoides/administração & dosagem , Drogas Ilícitas/farmacologia , Autoadministração , Caracteres Sexuais , Analgésicos Opioides/administração & dosagem , Animais , Comportamento Aditivo/psicologia , Cocaína/administração & dosagem , Modelos Animais de Doenças , Etanol/administração & dosagem , Feminino , Humanos , Masculino , Nicotina/administração & dosagem , Transtornos Relacionados ao Uso de Substâncias/psicologia
13.
Eur Neuropsychopharmacol ; 19(7): 487-98, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19250803

RESUMO

The gamma-aminobutyric acid(GABA)-B receptor agonist baclofen is known to reduce drug intake in both animals and humans and to prevent reinstatement of cocaine-, opioid-, and alcohol-seeking in rats after a period of extinction, but its effect on nicotine reinstatement is unknown. This study investigated the effect of baclofen on nicotine-seeking reinstatement both using the extinction/reinstatement model of nicotine self-administration and conditioned place preference (CPP). Results showed that in rats previously trained to intravenously self-administer nicotine (30 microg/kg/inf) under a FR-1 schedule of reinforcement, acute nicotine (0.15 mg/kg) priming effectively reinstates nicotine-seeking behaviour following extinction. At doses used in this study (up to 2.5 mg/kg) baclofen alone did not affect locomotor activity and did not reinstate responding. However, baclofen dose-dependently attenuated drug-induced reinstatement of nicotine-seeking in rats. Moreover, baclofen (1.25 mg/kg) completely blocked nicotine-induced reinstatement of extinguished nicotine (0.3 mg/kg) CPP in mice. Altogether, our results showed that baclofen is able to antagonise reinstatement of nicotine-seeking and CPP triggered by nicotine primings, suggesting its potential clinical utility as an anti-relapse agent.


Assuntos
Baclofeno/farmacologia , Condicionamento Operante/efeitos dos fármacos , Extinção Psicológica/efeitos dos fármacos , Agonistas GABAérgicos/farmacologia , Nicotina/farmacologia , Reforço Psicológico , Tabagismo/psicologia , Animais , Comportamento Animal/efeitos dos fármacos , Modelos Animais de Doenças , Esquema de Medicação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Esquema de Reforço , Autoadministração/métodos
14.
J Pharmacol Exp Ther ; 327(2): 482-90, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18725543

RESUMO

Emerging evidence suggests that the rewarding, abuse-related effects of nicotine are modulated by the endocannabinoid system of the brain. For example, pharmacological blockade or genetic deletion of cannabinoid CB(1) receptors can reduce or eliminate many abuse-related behavioral and neurochemical effects of nicotine. Furthermore, doses of Delta(9)-tetrahydrocannabinol and nicotine that are ineffective when given alone can induce conditioned place preference when given together. These previous studies have used systemically administered CB(1) receptor agonists and antagonists and gene deletion techniques, which affect cannabinoid CB(1) receptors throughout the brain. A more functionally selective way to alter endocannabinoid activity is to inhibit fatty acid amide hydrolase (FAAH), thereby magnifying and prolonging the effects of the endocannabinoid anandamide only when and where it is synthesized and released on demand. Here, we combined behavioral and neurochemical approaches to evaluate whether the FAAH inhibitor URB597 (cyclohexyl carbamic acid 3'-carbamoyl-3-yl ester) could alter the abuse-related effects of nicotine in rats. We found that URB597, at a dose (0.3 mg/kg) that had no behavioral effects by itself, prevented development of nicotine-induced conditioned place preference (CPP) and acquisition of nicotine self-administration. URB597 also reduced nicotine-induced reinstatement in both CPP and self-administration models of relapse. Furthermore, in vivo microdialysis showed that URB597 reduced nicotine-induced dopamine elevations in the nucleus accumbens shell, the terminal area of the brain's mesolimbic reward system. These findings suggest that FAAH inhibition can counteract the addictive properties of nicotine and that FAAH may serve as a new target for development of medications for treatment of tobacco dependence.


Assuntos
Amidoidrolases/antagonistas & inibidores , Ácidos Araquidônicos/metabolismo , Benzamidas/farmacologia , Carbamatos/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Dopamina/análise , Nicotina/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Alcamidas Poli-Insaturadas/metabolismo , Tabagismo/tratamento farmacológico , Amidoidrolases/fisiologia , Animais , Endocanabinoides , Hidrólise , Masculino , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/química , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Recompensa , Autoadministração , Tabagismo/enzimologia
15.
CNS Neurol Disord Drug Targets ; 7(5): 468-81, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19128204

RESUMO

Tobacco addiction is one of the leading preventable causes of mortality in the world and nicotine appears to be the main critical psychoactive component in establishing and maintaining tobacco dependence. Several lines of evidence suggest that the rewarding effects of nicotine, which underlie its abuse potential, can be modulated by manipulating the endocannabinoid system. For example, pharmacological blockade or genetic deletion of cannabinoid CB(1) receptors reduces or eliminates many behavioral and neurochemical effects of nicotine that are related to its addictive potential. This review will focus on the recently published literature about the role of the endocannabinoid system in nicotine addiction and on the endocannabinoid system as a novel molecular target for the discovery of medications for tobacco dependence.


Assuntos
Encéfalo/efeitos dos fármacos , Moduladores de Receptores de Canabinoides/agonistas , Moduladores de Receptores de Canabinoides/antagonistas & inibidores , Endocanabinoides , Nicotina/farmacologia , Receptor CB1 de Canabinoide/efeitos dos fármacos , Tabagismo/tratamento farmacológico , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Moduladores de Receptores de Canabinoides/metabolismo , Ensaios Clínicos como Assunto/estatística & dados numéricos , Humanos , Sistema Límbico/efeitos dos fármacos , Sistema Límbico/metabolismo , Sistema Límbico/fisiopatologia , Neurofarmacologia/métodos , Neurofarmacologia/tendências , Receptor CB1 de Canabinoide/metabolismo , Receptores Nicotínicos/efeitos dos fármacos , Receptores Nicotínicos/metabolismo , Tabagismo/metabolismo , Tabagismo/fisiopatologia
16.
Behav Brain Res ; 168(2): 307-11, 2006 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-16406104

RESUMO

Smoking marijuana causes working and short-term memory deficits, an effect that is mediated by cannabinoid receptor (CB1) activation in the brain. While this may be due to the main psychoactive constituent Delta9-tetrahydrocannabinol (Delta9-THC), plant extracts also contain other cannabinoid and terpenoid compounds with unknown properties. Towards this end, we have recently shown that high concentrations of plant extracts rich in cannabidiol (CBD) can reverse working memory deficits induced by Delta9-THC which is a remaining contaminant of this extract [Fadda P, Robinson L, Fratta W, Pertwee RG, Riedel G. Differential effects of THC- and CBD-rich cannabis-extracts on working memory in rats. Neuropahrmacology 2004;47:1170-9]. Since this effect was dose-dependent and indicative of memory enhancing qualities of the CBD-rich extract, this prompted a wider investigation into the effects of CBD on other forms of amnesia in order to determine the mechanism of action and to reveal its potency against anticholinergic and antiglutamatergic agents. We employed a spatial delayed matching to position task in the open-field water maze. Both scopolamine (0.2 mg/kg i.p.) and dizocilpine (MK801: 0.1mg/kg i.p.) impaired working memory at delays of 30 s and 4 h. Two doses of CBD-rich extracts (5 and 10 mg/kg), which did not affect working memory when given alone, were unable to reverse these deficits when co-administered with scopolamine or MK801. These data suggest that reversal of working memory deficits by CBD-rich extracts are specific to the cannabinoid system and do not compensate for acutely induced cholinergic or glutamatergic receptor hypoactivity.


Assuntos
Canabinoides/uso terapêutico , Maleato de Dizocilpina , Transtornos da Memória/tratamento farmacológico , Memória de Curto Prazo/efeitos dos fármacos , Escopolamina , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Interações Medicamentosas , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/fisiopatologia , Extratos Vegetais/uso terapêutico , Ratos , Tempo de Reação/efeitos dos fármacos , Fatores de Tempo
17.
Neuropharmacology ; 48(8): 1097-104, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15910886

RESUMO

Rats with a previous history of heroin self-administration were studied to assess interactions occurring between cannabinoids and opioids in an animal model of reinstatement of heroin-seeking behaviour. Rats were trained to self-administer heroin and after a long-term extinction were primed with one of the following non-contingent non-reinforced drug administrations: saline (or vehicle), heroin, synthetic cannabinoid CB(1) receptor agonists (WIN 55,212-2 or CP 55,940), opioid antagonist (naloxone) or CB(1) antagonist (SR 141716A), alone or in combination. After primings, lever-pressing activity was recorded and compared to those observed during previous phases of training and extinction. Results of this study showed that (i) priming injections of heroin (0.1 mg/kg) as well as CB(1) agonists WIN 55,212-2 (0.15 or 0.30 mg/kg) and CP 55,940 (0.05 or 0.1 mg/kg) completely restore heroin-seeking behaviour; (ii) primings of naloxone (1 mg/kg) and SR 141716A (0.3 mg/kg) had no effect when administered alone; (iii) heroin-induced reinstatement was fully prevented by pre-treatment with either naloxone or SR 141716A; (iv) pre-treatment with SR 141716A significantly reduced WIN 55,212-2 and CP 55,940 priming effects. These results suggest that cannabinoid CB(1) receptors play an important role in the mechanisms underlying relapse to heroin-seeking and depict CB(1) antagonists as possible therapeutic agents for use in the prevention of relapse to heroin abuse.


Assuntos
Comportamento Aditivo/prevenção & controle , Extinção Psicológica/efeitos dos fármacos , Dependência de Heroína/prevenção & controle , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Animais , Benzoxazinas/administração & dosagem , Benzoxazinas/farmacologia , Condicionamento Operante/efeitos dos fármacos , Cicloexanóis/administração & dosagem , Cicloexanóis/farmacologia , Interações Medicamentosas , Heroína/administração & dosagem , Masculino , Morfolinas/administração & dosagem , Morfolinas/farmacologia , Naloxona/administração & dosagem , Naloxona/farmacologia , Naftalenos/administração & dosagem , Naftalenos/farmacologia , Piperidinas/administração & dosagem , Pirazóis/administração & dosagem , Ratos , Ratos Endogâmicos , Receptor CB1 de Canabinoide/agonistas , Rimonabanto , Autoadministração
18.
Synapse ; 50(1): 1-6, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12872287

RESUMO

Evidence recently provided has suggested a specific involvement of the GABAergic system in modulating positive reinforcing properties of several drugs of abuse through an action on mesolimbic dopaminergic neurons. The GABA(B) receptor agonist baclofen has been proposed as a potential therapeutic agent for the clinical treatment of several forms of drug addiction. In the present study, using the in vivo microdialysis technique, we investigated the effect of baclofen on nicotine, cocaine, and morphine-induced increase in extracellular dopamine (DA) levels in the shell of the nucleus accumbens, a brain area supposedly involved in the modulation of the central effects of several drugs of abuse, of freely moving rats. As expected, nicotine (0.6 mg/kg s.c.), morphine (5 mg/kg s.c.), and cocaine (7.5 mg/kg i.p.) administration in rats induced a marked increase in extracellular DA concentrations in the nucleus accumbens, reaching a maximum value of +205 +/- 8.4%, +300 +/- 22.2%, and +370 +/- 30.7%, respectively. Pretreatment with baclofen (1.25 and 2.5 mg/kg i.p.) dose-dependently reduced the nicotine-, morphine-, and cocaine-evoked DA release in the shell of the nucleus accumbens. Furthermore, baclofen alone did not elicit changes in basal DA extracellular levels up to 180 min. Taken together, our data are in line with previous reports demonstrating the ability of baclofen to modulate the mesolimbic DAergic transmission and indicate baclofen as a putative candidate in the pharmacotherapy of polydrug abuse.


Assuntos
Baclofeno/farmacologia , Dopamina/metabolismo , Agonistas GABAérgicos/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Recompensa , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico , Animais , Baclofeno/uso terapêutico , Cocaína/antagonistas & inibidores , Relação Dose-Resposta a Droga , Líquido Extracelular/efeitos dos fármacos , Líquido Extracelular/metabolismo , Agonistas GABAérgicos/uso terapêutico , Agonistas dos Receptores de GABA-B , Masculino , Microdiálise , Morfina/antagonistas & inibidores , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Nicotina/antagonistas & inibidores , Núcleo Accumbens/metabolismo , Núcleo Accumbens/fisiopatologia , Ratos , Receptores de GABA-B/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA