Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropharmacology ; 186: 108475, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33529677

RESUMO

The emergence of new synthetic cathinones continues to be a matter of public health concern. In fact, they are quickly replaced by new structurally related alternatives. The main goal of the present study was to characterize the pharmacological profile, the psychostimulant and rewarding properties of novel cathinones (pentedrone, N-ethyl-pentedrone, α-PVP, N,N-diethyl-pentedrone and α-PpVP) which only differs in their amino terminal substitution. Rat synaptosomes were used for [3H]dopamine uptake experiments. HEK293 transfected cells (hDAT, hSERT, hOCT; human dopamine, serotonin and organic cation transporter) were also used for [3H]monoamine uptake and transporter binding assays. Molecular docking was used to investigate the effect of the amino substitutions on the biological activity. Hyperlocomotion and conditioned place preference paradigm were used in order to study the psychostimulant and rewarding effects in mice. All compounds tested are potent inhibitors of DAT with very low affinity for SERT, hOCT-2 and -3, and their potency for inhibiting DAT increased when the amino-substituent expanded from a methyl to either an ethyl-, a pyrrolidine- or a piperidine-ring. Regarding the in vivo results, all the compounds induced an increase in locomotor activity and possess rewarding properties. Results also showed a significant correlation between predicted binding affinities by molecular docking and affinity constants (Ki) for hDAT as well as the cLogP of their amino-substituent with their hDAT/hSERT ratios. Our study demonstrates the role of the amino-substituent in the pharmacological profile of novel synthetic cathinones as well as their potency inhibiting DA uptake and ability to induce psychostimulant and rewarding effects in mice.


Assuntos
Alcaloides/química , Alcaloides/farmacologia , Simulação de Acoplamento Molecular/métodos , Psicotrópicos/química , Psicotrópicos/farmacologia , Recompensa , Animais , Estimulantes do Sistema Nervoso Central/química , Estimulantes do Sistema Nervoso Central/farmacologia , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Ratos
2.
Biochem Pharmacol ; 163: 84-93, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30738029

RESUMO

3,4-Methylenedioxypyrovalerone (MDPV) acts as a dopamine transporter blocker and exerts powerful psychostimulant effects. In this study we aimed to investigate the bidirectional cross-sensitization between MDPV and cocaine, as well as to evaluate the role of the BDNF-TrkB signaling pathway in the development of locomotor sensitization to both drugs. Mice were treated with MDPV (1.5 mg/kg) or cocaine (10 or 15 mg/kg) once daily for 5 days. After withdrawal (10 days), animals were challenged with cocaine (8 mg/kg) or MDPV (1 mg/kg). For biochemical determinations, MDPV (1.5 mg/kg) or cocaine (15 mg/kg) were administered acutely or repeatedly, and BDNF, D3R and G9a transcription levels as well as pro- and mature BDNF protein levels were determined. Our results demonstrate that repeated administration of MDPV or cocaine sensitizes to cocaine and MDPV locomotor effects. After an acute or a repeated exposure to MDPV, cortical mRNA BDNF levels were increased, while a decrease in mBDNF protein levels in the nucleus accumbens 2 h after repeated exposure was evidenced. Interestingly, such decline was involved in the development of locomotor sensitization, thus the pretreatment with 7,8-dihydroxyflavone (10 mg/kg), a TrkB agonist, blocked the development of sensitization to MDPV but not to cocaine, for which no changes in the BDNF-TrkB signaling pathway were observed at early withdrawal. In conclusion, a bidirectional cross-sensitization between MDPV and cocaine was evidenced. Our findings suggest that decreased BDNF-TrkB signaling has an important role in the behavioral sensitization to MDPV, pointing TrkB modulation as a target to prevent MDPV sensitization.


Assuntos
Benzodioxóis/farmacologia , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Cocaína/farmacologia , Flavonas/farmacologia , Glicoproteínas de Membrana/fisiologia , Atividade Motora/efeitos dos fármacos , Proteínas Tirosina Quinases/fisiologia , Pirrolidinas/farmacologia , Animais , Inibidores da Captação de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Masculino , Camundongos , Atividade Motora/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Catinona Sintética
3.
Neuropharmacology ; 144: 271-281, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30321610

RESUMO

3,4-methylenedioxypyrovalerone (MDPV) is a synthetic cathinone with cocaine-like properties. In a previous work, we exposed adolescent mice to MDPV, finding sensitization to cocaine effects, and a higher vulnerability to cocaine abuse in adulthood. Here we sought to determine if such MDPV schedule induces additional behavioral-neuronal changes that could explain such results. After MDPV treatment (1.5 mg kg-1, twice daily, 7 days), mice were behaviorally tested. Also, we investigated protein changes in various brain regions. MDPV induced aggressiveness and anxiety, but also contributed to a faster habituation to the open field. This feature co-occurred with an induction of ΔFosB in the orbitofrontal cortex that was higher than its expression in the ventral striatum. Early after treatment, D2R:D1R ratio pointed to a preponderance of D1R but, upon withdrawal, the ratio recovered. Increased expression of Arc, CDK5 and TH, and decrease in DAT protein levels persisted longer after withdrawal, pointing to a neuroplastic lasting effect similar to that involved in cocaine addiction. The implication of the hyperdopaminergic condition in the MDPV-induced aggressiveness cannot be ruled out. We also found an initial oxidative effect of MDPV, without glial activation. Moreover, although initially the dopaminergic signal induced by MDPV resulted in increased ΔFosB, we did not observe any change in NFκB or GluA2 expression. Finally, the changes observed after MDPV treatment could not be explained according to the autoregulatory loop between ΔFosB and the epigenetic repressor G9a described for cocaine. This provides new knowledge about the neuroadaptive changes involved in the vulnerability to psychostimulant addiction.


Assuntos
Benzodioxóis/efeitos adversos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Estimulantes do Sistema Nervoso Central/efeitos adversos , Pirrolidinas/efeitos adversos , Assunção de Riscos , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Agressão/efeitos dos fármacos , Agressão/fisiologia , Animais , Ansiedade/induzido quimicamente , Ansiedade/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas do Citoesqueleto/metabolismo , Dopamina/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Masculino , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Distribuição Aleatória , Receptores Dopaminérgicos/metabolismo , Transtornos Relacionados ao Uso de Substâncias/psicologia , Fatores de Tempo , Catinona Sintética
4.
Br J Pharmacol ; 174(10): 1161-1173, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28262947

RESUMO

BACKGROUND AND PURPOSE: 3,4-Methylenedioxypyrovalerone (MDPV) is a synthetic cathinone with powerful psychostimulant effects. It selectively inhibits the dopamine transporter (DAT) and is 10-50-fold more potent as a DAT blocker than cocaine, suggesting a high abuse liability. The main objective of the present study was to assess the consequences of an early (adolescence) MDPV exposure on the psychostimulant, rewarding and reinforcing effects induced by cocaine in adult mice. EXPERIMENTAL APPROACH: Twenty-one days after MDPV pretreatment (1.5 mg·kg-1 , s.c., twice daily for 7 days), adult mice were tested with cocaine, using locomotor activity, conditioned place preference and self-administration (SA) paradigms. In parallel, dopamine D2 receptor density and the expression of c-Fos and ΔFosB in the striatum were determined. KEY RESULTS: MDPV treatment enhanced the psychostimulant and conditioning effects of cocaine. Acquisition of cocaine SA was unchanged in mice pretreated with MDPV, whereas the breaking point achieved under a progressive ratio programme and reinstatement after extinction were higher in this group of mice. MDPV decreased D2 receptor density but increased ΔFosB expression three-fold. As expected, acute cocaine increased c-Fos expression, but MDPV pretreatment negatively influenced its expression. ΔFosB accumulation declined during MDPV withdrawal, although it remained elevated in adult mice when tested for cocaine effects. CONCLUSION AND IMPLICATIONS: MDPV exposure during adolescence induced long-lasting adaptive changes related to enhanced responsiveness to cocaine in the adult mice that seems to lead to a higher vulnerability to cocaine abuse. This particular behaviour correlated with increased expression of ΔFosB.


Assuntos
Benzodioxóis/farmacologia , Cocaína/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Pirrolidinas/farmacologia , Reforço Psicológico , Animais , Benzodioxóis/administração & dosagem , Cocaína/administração & dosagem , Humanos , Injeções Subcutâneas , Masculino , Camundongos , Pirrolidinas/administração & dosagem , Receptores de Dopamina D2/metabolismo , Recompensa , Autoadministração , Catinona Sintética
5.
Artigo em Inglês | MEDLINE | ID: mdl-28219712

RESUMO

Methylenedioxypyrovalerone (MDPV) is a new psychostimulant cathinone acting as a selective dopamine transporter blocker. Due to the concomitant consumption of ethanol (EtOH) and new psychoactive substances, it is of interest to explore a possible pharmacological interaction between MDPV and EtOH. In locomotor activity assays, EtOH (1g/kg i.p.) elicited a reduction in the stimulant effect induced by low doses of MDPV (0.1-0.3mg/kg, s.c.) in rats, jointly with a decrease in blood and brain MDPV concentrations. Experiments in rat liver microsomes showed different effects depending on the [MDPV]/[EtOH] relationship, evidencing, at certain concentrations, the enhancing effect of EtOH on MDPV metabolism. These suggest that EtOH interacts with MDPV at microsomal level, increasing its metabolic rate. The interaction between both substances was also supported by results in plasma EtOH concentration, which were significantly increased by MDPV, in such a manner that EtOH elimination rate was significantly reduced. The possible toxicological impact of this phenomenon deserves further investigation. In contrast, the rewarding properties of MDPV were unaltered by EtOH. Microdialysis experiments verified that, in the NAcc, both substances could also act synergistically, in such a manner that extracellular dopamine concentrations are maintained. Finally, if the psychostimulant effect induced by MDPV decreased with EtOH, it could favor the boosting and re-dosing in search of the desired effects. However, as the rewarding effect of each dose of the substance would not decrease, the addictive liability could increase considerably. Moreover, we must warn about the increase in EtOH concentrations when consumed concomitantly with MDPV.


Assuntos
Alcaloides/metabolismo , Comportamento Animal/efeitos dos fármacos , Benzodioxóis/metabolismo , Benzodioxóis/farmacologia , Estimulantes do Sistema Nervoso Central/metabolismo , Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Interações Medicamentosas , Etanol/metabolismo , Etanol/farmacologia , Pirrolidinas/metabolismo , Pirrolidinas/farmacologia , Animais , Benzodioxóis/administração & dosagem , Estimulantes do Sistema Nervoso Central/administração & dosagem , Etanol/administração & dosagem , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Microdiálise , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Pirrolidinas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Catinona Sintética
6.
Mol Neurobiol ; 53(10): 7271-7283, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26687233

RESUMO

(±)3,4-Methylenedioxymethamphetamine (MDMA) is a relatively selective dopaminergic neurotoxin in mice. This study was designed to evaluate whether MDMA exposure affects their recognition memory and hippocampal expression of plasticity markers. Mice were administered with increasing doses of MDMA once per week for 8 weeks (three times in 1 day, every 3 h) and killed 2 weeks (2w) or 3 months (3m) later. The treatment did not modify hippocampal tryptophan hydroxylase 2, a serotonergic indicator, but induced an initial reduction in dopaminergic markers in substantia nigra, which remained stable for at least 3 months. In parallel, MDMA produced a decrease in dopamine (DA) levels in the striatum at 2w, which were restored 3 months later, suggesting dopaminergic terminal regeneration (sprouting phenomenon). Moreover, recognition memory was assessed using the object recognition test. Young (2w) and mature (3m) adult mice exhibited impaired memory after 24-h but not after just 1-h retention interval. Two weeks after the treatment, animals showed constant levels of CREB but an increase in its phosphorylated form and in c-Fos expression. Brain-derived neurotrophic factor (BDNF) and especially Arc overexpression was sustained and long-lasting. We cannot rule out the absence of MDMA injury in the hippocampus being due to the generation of BDNF. The levels of NMDAR2B, PSD-95, and synaptophysin were unaffected. In conclusion, the young mice exposed to MDMA showed increased expression of early key markers of plasticity, which sometimes remained for 3 months, and suggests hippocampal maladaptive plasticity that could explain memory deficits evidenced here.


Assuntos
Envelhecimento/patologia , Hipocampo/fisiopatologia , Transtornos da Memória/fisiopatologia , Plasticidade Neuronal , Animais , Biomarcadores/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/metabolismo , Masculino , Memória , Camundongos Endogâmicos C57BL , N-Metil-3,4-Metilenodioxianfetamina , Neurônios/metabolismo , Neurônios/patologia , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
7.
Psychopharmacology (Berl) ; 229(2): 295-306, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23649883

RESUMO

RATIONALE: Mephedrone (4-methylmethcathinone) is a still poorly known drug of abuse, alternative to ecstasy or cocaine. OBJECTIVE: The major aims were to investigate the pharmacokinetics and locomotor activity of mephedrone in rats and provide a pharmacokinetic/pharmacodynamic model. METHODS: Mephedrone was administered to male Sprague-Dawley rats intravenously (10 mg/kg) and orally (30 and 60 mg/kg). Plasma concentrations and metabolites were characterized using LC/MS and LC-MS/MS fragmentation patterns. Locomotor activity was monitored for 180-240 min. RESULTS: Mephedrone plasma concentrations after i.v. administration fit a two-compartment model (α = 10.23 h(-1), ß = 1.86 h(-1)). After oral administration, peak mephedrone concentrations were achieved between 0.5 and 1 h and declined to undetectable levels at 9 h. The absolute bioavailability of mephedrone was about 10% and the percentage of mephedrone protein binding was 21.59 ± 3.67%. We have identified five phase I metabolites in rat blood after oral administration. The relationship between brain levels and free plasma concentration was 1.85 ± 0.08. Mephedrone induced a dose-dependent increase in locomotor activity, which lasted up to 2 h. The pharmacokinetic-pharmacodynamic model successfully describes the relationship between mephedrone plasma concentrations and its psychostimulant effect. CONCLUSIONS: We suggest a very important first-pass effect for mephedrone after oral administration and an easy access to the central nervous system. The model described might be useful in the estimation and prediction of the onset, magnitude, and time course of mephedrone pharmacodynamics as well as to design new animal models of mephedrone addiction and toxicity.


Assuntos
Administração Intravenosa , Administração Oral , Estimulantes do Sistema Nervoso Central/administração & dosagem , Metanfetamina/análogos & derivados , Atividade Motora/efeitos dos fármacos , Animais , Área Sob a Curva , Disponibilidade Biológica , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Estimulantes do Sistema Nervoso Central/sangue , Estimulantes do Sistema Nervoso Central/metabolismo , Relação Dose-Resposta a Droga , Masculino , Redes e Vias Metabólicas , Metanfetamina/administração & dosagem , Metanfetamina/sangue , Metanfetamina/metabolismo , Ligação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem , Fatores de Tempo
8.
Biogerontology ; 14(2): 165-76, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23559311

RESUMO

Aging is associated with an increased risk of depression in humans. To elucidate the underlying mechanisms of depression and its dependence on aging, here we study signs of depression in male SAMP8 mice. For this purpose, we used the forced swimming test (FST). The total floating time in the FST was greater in SAMP8 than in SAMR1 mice at 9 months of age; however, this difference was not observed in 12-month-old mice, when both strains are considered elderly. Of the two strains, only the SAMP8 animals responded to imipramine treatment. We also applied the dexamethasone suppression test (DST) and studied changes in the dopamine and serotonin (5-HT) uptake systems, the 5-HT2a/2c receptor density in the cortex, and levels of TPH2. The DST showed a significant difference between SAMR1 and SAMP8 mice at old age. SAMP8 exhibits an increase in 5-HT transporter density, with slight changes in 5-HT2a/2c receptor density. In conclusion, SAMP8 mice presented depression-like behavior that is dependent on senescence process, because it differs from SAMR1, senescence resistant strain.


Assuntos
Envelhecimento/genética , Envelhecimento/psicologia , Comportamento Animal , Depressão/epidemiologia , Depressão/psicologia , Camundongos Endogâmicos/genética , Camundongos Endogâmicos/psicologia , Animais , Antidepressivos Tricíclicos/uso terapêutico , Córtex Cerebral/metabolismo , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Dopamina/metabolismo , Imipramina/uso terapêutico , Incidência , Masculino , Camundongos , Receptores de Serotonina/metabolismo , Natação/psicologia , Resultado do Tratamento , Triptofano Hidroxilase/metabolismo
9.
Neurochem Int ; 58(1): 92-101, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21074589

RESUMO

The neurotoxicity of MDMA or "Ecstasy" in rats is selectively serotonergic, while in mice it is both dopaminergic and serotonergic. MDMA metabolism may play a key role in this neurotoxicity. The function of serotonin and dopamine transporter and the effect of MDMA and its metabolites on them are essential to understand MDMA neurotoxicity. The aim of the present study was to investigate and compare the effects of MDMA and its metabolite alpha-methyldopamine (MeDA) on several molecular targets, mainly the dopamine and serotonin transporter functionality, to provide evidence for the role of this metabolite in the neurotoxicity of MDMA in rodents. MeDA had no affinity for the serotonin transporter but competed with serotonin for its uptake. It had no persistent effects on the functionalism of the serotonin transporter, in contrast to the effect of MDMA. Moreover, MeDA inhibited the uptake of dopamine into the serotonergic terminal and also MAO(B) activity. MeDA inhibited dopamine uptake with a lower IC(50) value than MDMA. After drug washout, the inhibition by MeDA persisted while that of MDMA was significantly reduced. The effect of MDMA on the dopamine transporter is related with dopamine release from vesicular stores, as this inhibition disappeared in reserpine-treated animals. However, the effect of MeDA seems to be a persistent conformational change of this transporter. Moreover, in contrast with MDMA, MeDA did not show affinity for nicotinic receptors, so no effects of MeDA derived from these interactions can be expected. The metabolite reduced cell viability at lower concentrations than MDMA. Apoptosis plays a key role in MDMA induced cellular toxicity but necrosis is the major process involved in MeDA cytotoxicity. We conclude that MeDA could protect against the serotonergic lesion induced by MDMA but potentiate the dopaminergic lesion as a result of the persistent blockade of the dopamine transporter induced this metabolite.


Assuntos
Desoxiepinefrina/análogos & derivados , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Síndromes Neurotóxicas/metabolismo , Animais , Membrana Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Cocaína/análogos & derivados , Cocaína/metabolismo , Desoxiepinefrina/toxicidade , Dopamina/metabolismo , Dopamina/fisiologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/química , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Inibidores da Captação de Dopamina/metabolismo , Técnicas In Vitro , Masculino , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/farmacologia , Células PC12 , Paroxetina/metabolismo , Conformação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores Nicotínicos/efeitos dos fármacos , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/química , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo
10.
Int Rev Neurobiol ; 88: 121-66, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19897077

RESUMO

During the last years, we have focused on the study of the neurotoxic effects of 3,4-methylenedioxymethamphetamine (MDMA) and methamphetamine (METH) on the central nervous system (CNS) and their pharmacological prevention methods. In the process of this research, we have used a semipurified synaptosomal preparation from striatum of mice or rats as a reliable in vitro model to study reactive oxygen species (ROS) production by these amphetamine derivatives, which is well-correlated with their dopaminergic injury in in vivo models. Using this preparation, we have demonstrated that blockade of alpha7 nicotinic receptors with methyllycaconitine (MLA) prevents ROS production induced by MDMA and METH. Consequently, in vivo, MLA significantly prevents MDMA- and METH-induced neurotoxicity at dopaminergic level (mouse striatum), without affecting hyperthermia induced by these amphetamines. Additionally, when neuroprotection was assayed with memantine (MEM), a dual antagonist of NMDA and alpha7 receptors, an effective neuroprotection was obtained also ahead of serotonergic injury induced by MDMA in rats. MEM also prevents MDMA effect on serotonin transporter functionality and METH effect on dopamine transporter (DAT), suggesting that behavioral effects of these psychostimulants can also be modulated by MEM. Finally, we have demonstrated that MEM prevents the impaired memory function induced by MDMA, and also, using binding studies with radioligands, we have characterized the interaction of these substances with nicotinic receptors. Studies at molecular level showed that both MDMA and METH displaced competitively the binding of radioligands with homomeric alpha7 and heteromeric nicotinic acetylcholine receptors (nAChRs), indicating that they can directly interact with them. In all the cases, MDMA displayed higher affinity than METH and it was higher for heteromeric than for alpha7 subtype. Pre-incubation of differentiated PC12 cells with MDMA or METH induces nAChR upregulation in a concentration- and time-dependent manner, as many nicotinic ligands do, supporting their functional interaction with nAChRs. Such interaction expands the pharmacological profile of amphetamines and can account for some of their effects.


Assuntos
Estimulantes do Sistema Nervoso Central/toxicidade , Metanfetamina/toxicidade , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Síndromes Neurotóxicas/metabolismo , Receptores Nicotínicos/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Síndromes Neurotóxicas/patologia , Espécies Reativas de Oxigênio/metabolismo , Receptores Nicotínicos/efeitos dos fármacos
11.
Neuropharmacology ; 54(8): 1254-63, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18455739

RESUMO

We hypothesize that 3,4-methylenedioxymethamphetamine (MDMA) and methamphetamine (METH) interact with alpha-7 nicotinic receptors (nAChR). Here we examine whether memantine (MEM), an antagonist of NMDAR and alpha-7 nAChR, prevents MDMA and METH neurotoxicity. MEM prevented both serotonergic injury induced by MDMA in rat and dopaminergic lesion by METH in mice. MEM has a better protective effect in front of MDMA- and METH-induced neurotoxicity than methyllycaconitine (MLA), a specific alpha-7 nAChR antagonist. The double antagonism that MEM exerts on NMDA receptor and on alpha-7 nAChR, probably contributes to its effectiveness. MEM inhibited reactive oxygen species production induced by MDMA or METH in synaptosomes. This effect was not modified by NMDA receptor antagonists, but reversed by alpha-7 nAChR agonist (PNU 282987), demonstrating a preventive effect of MEM as a result of it blocking alpha-7 nAChR. In synaptosomes, MDMA decreased 5-HT uptake by about 40%. This decrease was prevented by MEM and by MLA but enhanced by PNU 282987. A similar pattern was observed when we measured the dopamine transport inhibited by METH. The inhibition of both transporters by amphetamine derivatives seems to be regulated by the calcium incorporation after activation of alpha-7 nAChR. MDMA competitively displaces [(3)H]MLA from rat brain membranes. MEM and METH also displace [(3)H]MLA with non-competitive displacement profiles that fit a two-site model. We conclude that MEM prevents MDMA and METH effects in rodents. MEM may offer neuroprotection against neurotoxicity induced by MDMA and METH by preventing the deleterious effects of these amphetamine derivatives on their respective transporters.


Assuntos
Antagonistas de Dopamina/uso terapêutico , Alucinógenos , Memantina/uso terapêutico , Metanfetamina , N-Metil-3,4-Metilenodioxianfetamina , Fármacos Neuroprotetores , Síndromes Neurotóxicas/prevenção & controle , Animais , Benzamidas/farmacologia , Temperatura Corporal/efeitos dos fármacos , Compostos Bicíclicos com Pontes/farmacologia , Maleato de Dizocilpina/farmacologia , Proteínas da Membrana Plasmática de Transporte de Dopamina/genética , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Camundongos , Neuroglia/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Receptores Nicotínicos/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo
12.
Neurotoxicology ; 29(1): 179-83, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17980434

RESUMO

MDMA (ecstasy) is an illicit drug causing long-term neurotoxicity. Previous studies demonstrated the interaction of MDMA with alpha-7 nicotinic acetylcholine receptor (nAChR) in mouse brain membranes and the involvement of alpha-7 nicotinic acetylcholine receptors (nAChR) in dopaminergic neurotoxicity induced by MDMA in mice. The aim of the present study was to investigate the utility of memantine (MEM), an alpha-7 nAChR antagonist used for treatment of Alzheimer's disease patients, to prevent neurotoxicity induced by MDMA in rats and the oxidative effect of this amphetamine derivative in mice striatal synaptosomes. In isolated mouse striatal synaptosomes (an in vitro model of MDMA neurotoxicity of dopaminergic origin), MDMA (50 microM)-induced reactive oxygen species (ROS) production that was fully inhibited by MEM (0.3 microM). This effect of MEM was fully prevented by PNU 282987 (0.5 microM), a specific agonist of alpha-7 nAChR. The preventive effect of MEM on this oxidative effect can be attributed to a direct antagonism between MDMA (acting probably as agonist) and MEM (acting as antagonist) at the alpha-7 nAChR. In Dark Agouti rats (an in vivo model of MDMA neurotoxicity of serotonergic origin), a single dose of MDMA (18 mg/kg) induced persistent hyperthermia, which was not affected by MEM pre-treatment. [(3)H]Paroxetine binding (a marker of serotonergic injury) was measured in the hippocampus of animals killed at 24h and 7 days after treatment. MDMA induced a significant reduction in [(3)H]paroxetine binding sites at both times of sacrifice that was fully prevented by pre-treatment with MEM. Since previous studies demonstrate that increased glutamate activity is not involved in the neurotoxic action of MDMA, it can be concluded that the effectiveness of MEM against MDMA-induced neurotoxicity would be the result of blockade of alpha-7 nAChR, although an indirect mechanism based on the interplay among the various neurotransmission systems leading to an increase in basal acetylcholine release should also be taken into account.


Assuntos
Inibidores da Captação Adrenérgica/toxicidade , Antagonistas de Aminoácidos Excitatórios/farmacologia , Memantina/farmacologia , N-Metil-3,4-Metilenodioxianfetamina/toxicidade , Sinaptossomos/efeitos dos fármacos , Animais , Benzamidas/farmacologia , Temperatura Corporal/efeitos dos fármacos , Compostos Bicíclicos com Pontes/farmacologia , Corpo Estriado/ultraestrutura , Interações Medicamentosas , Masculino , Camundongos , Neurônios/ultraestrutura , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
13.
Neurochem Int ; 52(3): 401-10, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17716785

RESUMO

d-Amphetamine (AMPH) and MDMA increased intracellular production of reactive oxygen species (ROS) in isolated mouse striatal synaptosomes. MDMA showed a maximal oxidative effect at 50-100 microM. However, for AMPH a double maximum was obtained, the first between 0.1 and 1 microM and the second at 1mM. No oxidative effect was present in synaptosomes from reserpinized mice. Cocaine and l-deprenyl inhibited MDMA and AMPH (0.1 microM) ROS production but not that of AMPH at a higher concentration (1mM). When this high concentration was used, its oxidative effect was abolished by a phospholipase A(2) inhibitor. Delta(9)-Tetrahydrocannabinol fully prevented the oxidative effect of AMPH and MDMA, by a CB(1) receptor-independent mechanism, as did it NPC 15437 and genistein. The pro-oxidative effect induced by AMPH and MDMA showed a strong dependence on calcium (extracellular and from internal stores) and also was inhibited by nicotinic receptor (nAChR) antagonists dihydro-beta-erythroidine, methyllycaconitine (MLA) and alpha-bungarotoxin. MDMA displaced [(3)H]epibatidine and [(3)H]MLA binding with higher affinity than AMPH. Both amphetamines competitively displaced [(3)H]epibatidine from heteromeric receptors but results obtained from [(3)H]MLA binding demonstrated a non-competitive profile. Preincubation of PC12 cells with AMPH or MDMA reduced [(3)H]dopamine uptake. For MDMA, this effect was prevented by MLA. To summarize, comparing AMPH and MDMA we have demonstrated that these drugs induce an oxidative effect dependent on drug concentration and also reduce dopamine uptake. Processes that are known to affect dopamine transporter functionality also seem to modulate amphetamine derivatives-induced ROS production. For MDMA, acute effects tested are blocked by nAChR antagonists, which points to the possibility that these antagonists could be used to treat some of the adverse effects described in MDMA abusers. Conversely, no implication of nicotinic receptors has been proved for AMPH-induced effects at concentrations achievable in CNS after its administration.


Assuntos
Anfetamina/farmacologia , Corpo Estriado/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina/farmacologia , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Receptores Nicotínicos/efeitos dos fármacos , Inibidores da Captação Adrenérgica/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Corpo Estriado/metabolismo , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/efeitos dos fármacos , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Inibidores da Captação de Dopamina/farmacologia , Sinergismo Farmacológico , Masculino , Camundongos , Neurônios/metabolismo , Antagonistas Nicotínicos/farmacologia , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Células PC12 , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Receptores Nicotínicos/metabolismo , Frações Subcelulares , Sinaptossomos
14.
Neuropharmacology ; 51(4): 885-95, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16901518

RESUMO

Methylenedioxymethamphetamine (MDMA) is a relatively selective dopaminergic neurotoxin in mice. Previous studies demonstrated the participation of alpha-7 nicotinic receptors (nAChR) in the neurotoxic effect of methamphetamine. The aim of this paper was to study the role of this receptor type in the acute effects and neurotoxicity of MDMA in mice. In vivo, methyllycaconitine (MLA), a specific alpha-7 nAChR antagonist, significantly prevented MDMA-induced neurotoxicity at dopaminergic but not at serotonergic level, without affecting MDMA-induced hyperthermia. Glial activation was also fully prevented by MLA. In vitro, MDMA induced intrasynaptosomal reactive oxygen species (ROS) generation, which was calcium-, nitric-oxide synthase-, and protein kinase C-dependent. Also, the increase in ROS was prevented by MLA and alpha-bungarotoxin. Experiments with reserpine point to endogenous dopamine (DA) as the main source of MDMA-induced ROS. MLA also brought the MDMA-induced inhibition of [3H]DA uptake down, from 73% to 11%. We demonstrate that a coordinated activation of alpha-7 nAChR, blockade of DA transporter function and displacement of DA from intracellular stores induced by MDMA produces a neurotoxic effect that can be prevented by MLA, suggesting that alpha-7 nAChR have a key role in the MDMA neurotoxicity in mice; however, the involvement of nicotinic receptors containing the beta2 subunit cannot be conclusively ruled out.


Assuntos
Aconitina/análogos & derivados , Dopamina/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Síndromes Neurotóxicas/tratamento farmacológico , Receptores Nicotínicos/fisiologia , Aconitina/uso terapêutico , Análise de Variância , Animais , Cocaína/análogos & derivados , Cocaína/farmacocinética , Modelos Animais de Doenças , Inibidores da Captação de Dopamina/farmacocinética , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo/métodos , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , N-Metil-3,4-Metilenodioxianfetamina , Síndromes Neurotóxicas/etiologia , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Ensaio Radioligante/métodos , Espécies Reativas de Oxigênio/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Trítio/metabolismo
15.
Neuroscience ; 120(3): 599-603, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12895500

RESUMO

The mechanisms underlying selective neuronal cell death in kainic acid-mediated neurodegeneration are not fully understood. We have recently demonstrated that in cerebellar granule neurons, kainic acid induces the expression of proteins associated with cell-cycle progression. In the present study we show that 3-amino thioacridone (3-ATA), a selective cyclin-dependent kinase 4 inhibitor, attenuates kainic acid-induced apoptosis in cerebellar granule neurons. When neurons were pre-treated with 3-ATA 10 microM for 24 h, they were less susceptible to damage induced by kainic acid 500 microM, since the number of dead cells decreased significantly. In flow cytometry studies using propidium iodide staining, 3-ATA also reduced the ratio of apoptotic cells induced by kainic acid. Moreover, 3-ATA decreased the proportion of cells with a condensed nucleus from 55% to 22%. Our data suggest that the cell cycle pathway is involved in the mechanism of apoptosis mediated by kainic acid and that cyclin-dependent kinase 4 plays a prominent role in this process. 3-ATA may to prevent the apoptosis associated with neurodegenerative disorders without the over-activation of excitatory amino acid receptors.


Assuntos
Aminoacridinas/farmacologia , Apoptose/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Inibidores Enzimáticos/farmacologia , Ácido Caínico , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas , Animais , Animais Recém-Nascidos , Morte Celular , Cerebelo/metabolismo , Quinase 4 Dependente de Ciclina , Quinases Ciclina-Dependentes/antagonistas & inibidores , Citometria de Fluxo , Ácido Caínico/toxicidade , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
16.
Naunyn Schmiedebergs Arch Pharmacol ; 364(2): 96-104, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11534860

RESUMO

We tested the potential cytoprotective role of C-phycocyanin in rat cerebellar granule cell cultures. Cell death was induced by potassium and serum (K/S) withdrawal. Cell viability was studied using the neutral red assay and laser scanning cytometry with propidium iodide as fluorochrome. C-phycocyanin (1-3 mg/ml) showed a neuroprotective effect against 24 h of K/S deprivation in cerebellar granule cells. After 4 h K/S deprivation this compound (3 mg/ml) inhibited formation of reactive oxygen species, measured as 2',7'-dichlorofluorescein fluorescence, showing its scavenger capability. Pre-treatment with C-phycocyanin reduced thymidine incorporation into DNA below control values and reduced dramatically apoptotic bodies as visualized by propidium iodide, indicating inhibition of apoptosis induced by K/S deprivation. Flow cytometry studies, using propidium iodide in TritonX100 permeabilized cells, indicated that 24 h K/S deprivation acts as a proliferative signal for cerebellar granule cells, which show an increase in S-phase percentage and cells progressed into the apoptotic pathway. C-phycocyanin protected cerebellar granule cells from the apoptosis induced by deprivation. These results suggest that C-phycocyanin prevents apoptosis in cerebellar granule cells probably through the antioxidant activity. It is proposed that K/S deprivation-induced apoptosis could be due, in part, to an alteration in the cell cycle mediated by an oxidative stress mechanism.


Assuntos
Apoptose/efeitos dos fármacos , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ficocianina/farmacologia , Cloreto de Potássio/administração & dosagem , Animais , Animais Recém-Nascidos , Apoptose/fisiologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Cerebelo/metabolismo , Meios de Cultura Livres de Soro/efeitos adversos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
17.
Br J Pharmacol ; 132(3): 693-702, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11159722

RESUMO

1. Previous studies indicate that 3-nitropropionic acid (3-NPA) neurotoxicity involves the excitotoxic activation of N-methyl-D-aspartate (NMDA) receptors. Thus, we examined the effect of orphenadrine (an anticholinergic drug with NMDA receptor antagonist properties) on 3-NPA neurotoxicity in both cultured rat cerebellar granule cells (CGCs) and in rats. 2. Orphenadrine protected CGCs from 3-NPA-induced mortality, as assessed by both the neutral red viability assay and laser scanning cytometry, using propidium iodide staining. 3. For rats, two indirect markers of neuronal damage were used: the binding of [(3)H]-PK 11195 to the peripheral-type benzodiazepine receptor (PBR), a microglial marker, and expression of the 27 kD heat-shock protein (HSP27), a marker of activated astroglia. Systemic administration of 3-NPA (30 mg kg(-1) per day for 3 days) induced a 170% increase in [(3)H]-PK 11195 binding, and expression of HSP27. 4. Both the increase in [(3)H]-PK 11195 and HSP 27 expression were prevented by previous administration of 30 mg kg(-1) per day of orphenadrine for 3 days. Lower doses (10 and 20 mg kg(-1)) had no protective effect. Orphenadrine also reduced 3-NPA-induced mortality in a dose-dependent manner. 5. We propose that orphenadrine or orphenadrine-like drugs could be used to treat neurodegenerative disorders mediated by overactivation of NMDA receptors.


Assuntos
Cerebelo/efeitos dos fármacos , Orfenadrina/farmacologia , Propionatos/toxicidade , Animais , Anti-Hipertensivos/toxicidade , Western Blotting , Peso Corporal/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cerebelo/citologia , Cerebelo/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Interações Medicamentosas , Isoquinolinas/farmacologia , Masculino , Mortalidade , Antagonistas Muscarínicos/farmacologia , Antagonistas Muscarínicos/uso terapêutico , Síndromes Neurotóxicas/prevenção & controle , Nitrocompostos , Orfenadrina/uso terapêutico , Propionatos/antagonistas & inibidores , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Trítio
18.
Neurobiol Dis ; 7(4): 343-61, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10964606

RESUMO

Injection of MPP(+) into the substantia nigra causes extensive necrosis and anterograde degeneration of pars compacta dopaminergic neurons. We studied secondary effects in the ipsilateral striatum by examining dopaminergic terminals, signs of neuronal damage, and glial reactivity at 1, 2, 3, and 7 days after injection of MPP(+) into the substantia nigra. Dopaminergic terminals and uptake sites were evaluated with [(3)H]GBR-12935 binding and tyrosine hydroxylase immunoreactivity. Glial reaction was examined with markers of astrocytes and microglia. Stereology was used to evaluate any changes in neuronal density. Tyrosine hydroxylase immunoreactivity and [(3)H]GBR-12935 binding markedly decreased (74%) from days 2 to 7. Loss of dopaminergic terminals in the ipsilateral striatum was accompanied by an intense astroglial and, to a lesser extent, microglial reaction. However, no signs of cell damage, neuronal loss, or disruption of the blood-brain barrier were found in the striatum. Resident astroglial and microglial cells showed a morphological shift and notable changes in protein expression typical of glial reactivity, yet the presence of macrophage-like cells was not detected. This study shows that injection of MPP(+) in the substantia nigra causes a secondary reaction within the ipsilateral striatum involving the transformation of quiescent glia to reactive glia. It is suggested that stimuli derived from damaged dopaminergic terminals within the striatum are able to activate resident glia and that this glial transformation may promote repair and regeneration.


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Corpo Estriado/efeitos dos fármacos , Herbicidas/farmacologia , Neuroglia/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , Animais , Antineoplásicos/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Corpo Estriado/metabolismo , Proteína Glial Fibrilar Ácida/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Isoquinolinas/metabolismo , Masculino , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Substância Negra/metabolismo
19.
Eur J Pharmacol ; 398(1): 31-9, 2000 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-10856445

RESUMO

Previous work from our laboratory has demonstrated the presence of high-affinity binding sites for [3H]nitrobenzylthioinosine ([3H]NBTI), a marker of adenosine uptake systems, in the mitochondrial fraction of rat testis. Here, we characterize this system functionally through [3H]adenosine uptake assays. This system (K(m)=2+/-1.3 microM; V(max)=86.2+/-15.5 pmol/mg protein/min) was found to be saturable, non sodium-dependent and sensitive to temperature, pH and osmolarity. [3H]Adenosine incorporation was potently inhibited by hydroxynitrobenzylthioguanosine (HNBTG, IC(50)=3 nM) although NBTI inhibited this uptake weakly (IC(50)=72. 7+/-37.1 microM). Dilazep>dipyridamole>/=hexobendine inhibited [3H]adenosine incorporation at low micromolar concentrations. The nucleosides inosine and uridine were weak inhibitors of this system. The adenosine receptor ligands N(6)-phenylisopropyladenosine (PIA) and 2-chloroadenosine inhibited the uptake only at micromolar concentrations. Neither 5'-(N-ethylcarboxamido)-adenosine (NECA) nor theophylline inhibited adenosine uptake by more than 60% but the mitochodrial benzodiazepine receptor ligands 4'-chloro-diazepam (Ro 5-4864) and 1-(2-chlorophenyl)-N-methyl-N-(1-methyl-propyl) isoquinoline carboxamide (PK 11195) were able to inhibit it. The lack of inhibition by the blockers of the mitochondrial adenine-nucleotide carrier, atractyloside and alpha, beta-methylene-ATP, indicates that [3H]adenosine uptake occurs via a transporter other than this carrier. All these results support the existence of an equilibrative adenosine transport system, which might mediate the passage of adenosine formed in the mitochondria to the cytoplasm.


Assuntos
Adenosina/metabolismo , Mitocôndrias/metabolismo , Testículo/metabolismo , Adenosina/farmacologia , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Animais , Benzodiazepinonas/farmacologia , Transporte Biológico/efeitos dos fármacos , Dilazep/farmacologia , Dipiridamol/farmacologia , Relação Dose-Resposta a Droga , Guanosina/análogos & derivados , Guanosina/farmacologia , Hexobendina/farmacologia , Concentração de Íons de Hidrogênio , Inosina/farmacologia , Isoquinolinas/farmacologia , Cinética , Masculino , Mitocôndrias/efeitos dos fármacos , Concentração Osmolar , Fenilisopropiladenosina/farmacologia , Ratos , Ratos Sprague-Dawley , Sódio/farmacologia , Frações Subcelulares , Temperatura , Testículo/efeitos dos fármacos , Tioinosina/análogos & derivados , Tioinosina/farmacologia , Tionucleosídeos/farmacologia , Fatores de Tempo , Trítio , Uridina/farmacologia
20.
J Neurosci Res ; 59(6): 797-805, 2000 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10700017

RESUMO

The effects of nitric oxide synthase (NOS) inhibitors, N(omega)-nitro-L-arginine and 7-nitroindazole, and the NOS substrate L-arginine on kainic acid (KA)-induced microglial reactivity and stress response were studied in the hippocampus 7 and 1 days after KA, respectively. Density of peripheral-type benzodiazepine receptors was measured as an index of microglial reactivity. Histological damage in hippocampus was evaluated at 7 days by neuronal counting. KA increased the maximal number of binding sites (B(max)) versus controls. Administration of either 7-nitroindazole (25 mg/kg) or N(omega)-nitro-L-arginine (20 and 50 mg/kg) 24 hr before KA, further increased B(max). This later effect was abolished by L-arginine (1 g/kg), which given 24 hr before KA decreased B(max) to control values. Also, KA-induced HSP72 stress response was attenuated by pre-treatment with L-arginine. Histological evaluation showed reduced cell numbers in the pyramidal cell layer of the hippocampus in groups receiving KA, either alone or in combination with 7-nitroindazole. Administration of L-arginine before KA attenuated neuronal loss in CA3 but not CA1. A clear protective effect was observed, however, in CA1 and CA3, in rats receiving both L-arginine plus 7-nitroindazole before KA. The results show that the combination of a NO substrate with a NOS inhibitor reduces the neurotoxic effects of KA in the rat hippocampus. This study suggests that extremely fine regulation of NO levels in the different neural cell types can modulate excitotoxicity.


Assuntos
Inibidores Enzimáticos/farmacologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Hipocampo/efeitos dos fármacos , Ácido Caínico/toxicidade , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico/metabolismo , Animais , Antineoplásicos/farmacologia , Western Blotting , Proteínas de Choque Térmico HSP72 , Proteínas de Choque Térmico/análise , Proteínas de Choque Térmico/efeitos dos fármacos , Hipocampo/metabolismo , Indazóis/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Isoquinolinas/farmacologia , Masculino , Óxido Nítrico/agonistas , Óxido Nítrico/antagonistas & inibidores , Nitroarginina/farmacologia , Proteínas Serina-Treonina Quinases/análise , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...