Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Biol Rep ; 50(5): 4423-4433, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36977807

RESUMO

BACKGROUND: Growing evidence indicates that cannabinoid type 2 (CB2) receptor activation inhibits neuroinflammation in the pathogenesis of Parkinson's disease (PD). Nonetheless, the precise mechanisms of CB2 receptor-mediated neuroprotection have not been fully elucidated. The differentiation of microglia from the M1 to M2 phenotype plays a vital role in neuroinflammation. METHODS: In the present study, we investigated the effect of CB2 receptor activation on the M1/M2 phenotypic transformation of microglia treated with 1-methyl-4-phenylpyridinium (MPP+). The M1 phenotype microglia markers, including inducible nitric oxide (iNOS), interleukin 6 (IL-6), and CD86, and the M2 phenotype microglia markers, including arginase-1 (Arg-1), IL-10, and CD206, were detected by western blots and flow cytometry. The levels of phosphoinositide-3-kinase (PI3K)/Akt and nuclear factor erythroid 2-related factor 2 (Nrf2) were determined by Western blots. Subsequent addition of Nrf2 inhibitors initially revealed the specific mechanism by which CB2 receptors affect phenotypic changes in microglia. RESULTS: Our results showed that pretreatment with JWH133 significantly inhibited the MPP+-induced up-regulation of M1 phenotype microglia markers. Meanwhile, JWH133 increased the levels of M2 phenotype microglia markers. JWH133-mediated effects were blocked by co-treatment with AM630. Mechanism studies found that MPP+ treatment downregulated PI3K, Akt phosphorylated proteins, and nuclear Nrf2 protein. JWH133 pretreatment promoted PI3K/Akt activation and facilitated nuclear translocation of Nrf2, which was reversed by the PI3K inhibitor. Further studies showed that Nrf2 inhibitors inverted the effect of JWH133 on microglia polarization. CONCLUSION: The results indicate that CB2 receptor activation promotes MPP+-induced microglia transformation from M1 to M2 phenotype through PI3K/Akt/Nrf2 signaling pathway.


Assuntos
Canabinoides , Microglia , Humanos , Microglia/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , 1-Metil-4-fenilpiridínio/farmacologia , 1-Metil-4-fenilpiridínio/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Doenças Neuroinflamatórias , Receptor CB2 de Canabinoide/genética , Transdução de Sinais , Canabinoides/farmacologia , Canabinoides/metabolismo
2.
Acta Pharmacol Sin ; 43(4): 889-896, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34253876

RESUMO

Nicotine, a major component of tobacco, is highly addictive and acts on nicotinic acetylcholine receptors (nAChRs) to stimulate reward-associated circuits in the brain. It is well known that nAChRs play critical roles in mediating nicotine reward and addiction. Current FDA-approved medications for smoking cessation are the antidepressant bupropion and the nicotinic partial agonist varenicline, yet both are limited by adverse side effects and moderate efficacy. Thus, development of more efficacious medications with fewer side effects for nicotine addiction and smoking cessation is urgently needed. l-Tetrahydropalmatine (l-THP) is an active ingredient of the Chinese medicinal herb Corydalis ambigua that possesses rich neuropharmacological actions on dopamine (DA) receptors in the mesocorticolimbic dopaminergic reward pathway. L-THP has been explored as anti-addiction treatments for drug abuse including nicotine. However, the targets and mechanisms of l-THP-caused anti-nicotine effects are largely unknown. In this study we address this question by elucidating the effects of l-THP on human neuronal nAChRs using patch-clamp recordings. Human neuronal α4ß2-nAChRs were heterologously expressed in SH-EP1 human epithelial cells. Bath application of nicotine (0.1-100 µM) induced inward currents, co-application of l-THP (3 µM) inhibited nicotine-induced currents in the transfected cells. L-THP-caused inhibition was concentration-dependent (the EC50 values for inhibiting the peak and steady-state current were 18 and 2.1 µM, respectively) and non-competitive. Kinetic analysis of the whole-cell currents showed that l-THP slowed rising time and accelerated decay time constants. L-THP specifically modulated α4ß2-nAChRs, as it did not affect α7-nAChRs or α1*-nAChRs (muscle type). Interestingly, two putative α4ß2-nAChR isoforms, namely sazetidine A-activated, high-sensitive one (α42ß23-nAChR) and cytisine-activated, low-sensitive one (α43ß22-nAChR) were pharmacologically separated, and the low-sensitive one was more susceptible to l-THP inhibition than the high-sensitive one. In conclusion, we demonstrate that l-THP blocks neuronal α4ß2-nAChR function, which may underlie its inhibition on nicotine addiction.


Assuntos
Nicotina , Receptores Nicotínicos , Alcaloides de Berberina , Humanos , Cinética , Nicotina/farmacologia , Receptores Nicotínicos/metabolismo
3.
Food Funct ; 12(18): 8366-8375, 2021 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-34342315

RESUMO

Berberine, an isoquinoline alkaloid isolated from Coptis chinensis, has been widely studied for its efficacy in the treatment of neurodegenerative diseases. However, the detailed mechanisms are unknown. In this study, the effects of berberine on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice model of Parkinson's disease were investigated. We showed that treatment with berberine significantly ameliorates the degeneration of dopaminergic neurons in substantia nigra compacta (SNc) and improves motor impairment in MPTP-treated mice. Berberine also significantly decreased the level of α-synuclein and enhanced the microtubule-associated protein light chain 3 (LC3-II)-associated autophagy in the SN of MPTP-treated mice. Furthermore, adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) was activated by berberine. Berberine's actions were abolished by pre-treatment with 3-methyladenine (an autophagy inhibitor) or compound c (an AMPK inhibitor) in the MPP+-treated SH-SY5Y cells. These results suggested that the protective effects of berberine on the toxicity of MPTP could be attributed to berberine-enhanced autophagy via the AMPK dependent pathway.


Assuntos
Autofagia/efeitos dos fármacos , Berberina/uso terapêutico , Neurônios Dopaminérgicos/efeitos dos fármacos , Intoxicação por MPTP/tratamento farmacológico , Animais , Berberina/química , Linhagem Celular Tumoral , Sobrevivência Celular , Coptis chinensis/química , Regulação para Baixo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma , Neurônios/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
4.
Neurosci Lett ; 720: 134779, 2020 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-31981721

RESUMO

BACKGROUND: We investigated the anti- inflammatory effect of type II cannabinoid receptor (CB2 receptor) activation and their relationship to iron influx on 1-methyl-4-phenylpyridinium (MPP+) treated astrocytes. METHODS AND RESULTS: By western blots, real-time PCR and ELISA, the expressions of CB2 receptor, divalent metal transporter-1 (DMT1), cyclooxygenase-2 (COX-2), inducible nitric oxide (iNOS), interleukin-1ß (IL-1ß) and tumor necrosis factor- α (TNF-α) were measured. Iron influx into astrocytes was determined by the quenching of calcein fluorescence. We found that pre-treatment with JWH133, a selective CB2 receptor agonist, significantly suppressed the MPP+-induced up-regulation of COX-2, iNOS, IL- 1ß and TNF-α in astrocytes. In addition, JWH133 significantly inhibited the MPP+-induced up- regulation of DMT1. Further studies indicated that JWH133 suppressed the MPP+-accelerated iron influx into astrocytes. These effects were blocked by co-treatment with AM630, the CB2 receptor antagonist. CONCLUSIONS: These results suggest that activation of CB2 receptor inhibit MPP +-induced inflammatory response and iron influx in astrocytes.


Assuntos
1-Metil-4-fenilpiridínio/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Canabinoides/administração & dosagem , Encefalite/metabolismo , Ferro/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Animais , Células Cultivadas , Encefalite/prevenção & controle , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Ratos , Regulação para Cima
5.
Acta Pharmacol Sin ; 41(2): 163-172, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31399700

RESUMO

Cocaine is one of the most abused illicit drugs worldwide. It is well known that the dopamine (DA) transporter is its major target; but cocaine also acts on other targets including nicotinic acetylcholine receptors (nAChRs). In this study, we investigated the effects of cocaine on a special subtype of neuronal nAChR, α3ß4-nAChR expressed in native SH-SY5Y cells. α3ß4-nAChR-mediated currents were recorded using whole-cell recordings. Drugs were applied using a computer-controlled U-tube drug perfusion system. We showed that bath application of nicotine induced inward currents in a concentration-dependent manner with an EC50 value of 20 µM. Pre-treatment with cocaine concentration-dependently inhibited nicotine-induced current with an IC50 of 1.5 µM. Kinetic analysis showed that cocaine accelerated α3ß4-nAChR desensitization, which caused a reduction of the amplitude of nicotine-induced currents. Co-application of nicotine and cocaine (1.5 µM) depressed the maximum response on the nicotine concentration-response curve without changing the EC50 value, suggesting a non-competitive mechanism. The cocaine-induced inhibition of nicotine response exhibited both voltage- and use-dependence, suggesting an open-channel blocking mechanism. Furthermore, intracellular application of GDP-ßS (via recording electrode) did not affect cocaine-induced inhibition, suggesting that cocaine did not alter receptor internalization. Moreover, intracellular application of cocaine (30 µM) failed to alter the nicotine response. Finally, cocaine (1.5 µM) was unable to inhibit the nicotine-induced inward current in heterologous expressed α6/α3ß2ß3-nAChRs and α4ß2-nAChRs expressed in human SH-EP1 cells. Collectively, our results suggest that cocaine is a potent blocker for native α3ß4-nAChRs expressed in SH-SY5Y cells.


Assuntos
Cocaína/farmacologia , Neurônios/efeitos dos fármacos , Receptores Nicotínicos/efeitos dos fármacos , Linhagem Celular Tumoral , Cocaína/administração & dosagem , Relação Dose-Resposta a Droga , Humanos , Concentração Inibidora 50 , Neuroblastoma/metabolismo , Neurônios/metabolismo , Nicotina/farmacologia , Técnicas de Patch-Clamp , Receptores Nicotínicos/metabolismo
6.
Neurosci Bull ; 36(2): 165-173, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31482520

RESUMO

In the present study, we investigated the mechanisms underlying the mediation of iron transport by L-type Ca2+ channels (LTCCs) in primary cultured ventral mesencephalon (VM) neurons from rats. We found that co-treatment with 100 µmol/L FeSO4 and MPP+ (1-methyl-4-phenylpyridinium) significantly increased the production of intracellular reactive oxygen species, decreased the mitochondrial transmembrane potential and increased the caspase-3 activation compared to MPP+ treatment alone. Co-treatment with 500 µmol/L CaCl2 further aggravated the FeSO4-induced neurotoxicity in MPP+-treated VM neurons. Co-treatment with 10 µmol/L isradipine, an LTCC blocker, alleviated the neurotoxicity induced by co-application of FeSO4 and FeSO4/CaCl2. Further studies indicated that MPP+ treatment accelerated the iron influx into VM neurons. In addition, FeSO4 treatment significantly increased the intracellular Ca2+ concentration. These effects were blocked by isradipine. These results suggest that elevated extracellular Ca2+ aggravates iron-induced neurotoxicity. LTCCs mediate iron transport in dopaminergic neurons and this, in turn, results in elevated intracellular Ca2+ and further aggravates iron-induced neurotoxicity.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Ferro/toxicidade , Mesencéfalo/metabolismo , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Apoptose/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Caspase 3/metabolismo , Células Cultivadas , Feminino , Isradipino/farmacologia , Gravidez , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
7.
Neuroreport ; 31(1): 41-47, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31688419

RESUMO

Berberine, an isoquinoline alkaloid isolated from traditional Chinese medicine, has been widely studied for its efficacy in the treatment of neurodegenerative diseases. However, berberine-mediated neuroprotection in the pathogenesis of Parkinson's disease is still uncertain. In this study, the effects of berberine on rotenone-induced neurotoxicity in SH-SY5Y cells were investigated. The results showed that berberine treatment significantly alleviated rotenone-induced decrease in the cell viability in SH-SY5Y cells. Further studies demonstrated that berberine suppressed the production of intracellular reactive oxygen species, restored the mitochondrial transmembrane potential, increased Bcl-2/Bax ratio, and decreased caspase-3 activation that induced by rotenone. Furthermore, berberine also restored the phosphorylation of Akt, which was downregulated by rotenone in SH-SY5Y cells. These results suggest that berberine protects rotenone-treated SH-SY5Y cells by antioxidation and activation of PI3K/Akt signaling pathway.


Assuntos
Antioxidantes/farmacologia , Berberina/farmacologia , Neurônios/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Humanos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Rotenona/toxicidade , Transdução de Sinais/efeitos dos fármacos , Desacopladores/toxicidade
8.
Oncotarget ; 8(29): 47284-47295, 2017 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-28521299

RESUMO

The aim of this study is to investigate the effects of L-type calcium channels (LTCCs) on MPTP-induced dopamine (DA) neuron degeneration and iron accumulation in the substantia nigra (SN) of mice. By real-time PCR and western blots, we first quatified expressions of L-type Cav1.2 and Cav1.3 calcium channel α1 subunits in the SN of experimental mice treated with MPTP. We found that the expressions of Cav1.2 and Cav1.3 calcium channel α1 subunits markedly increased after MPTP treatment for 2 and 3 weeks. Secondly, we observed the effects of isradipine, a LTCC antagonist, on MPTP-induced DA neuron degeneration and iron accumulation in the SN. Our results showed that isradipine treatment prevented against MPTP-induced Cav1.2 and Cav1.3 calcium channel α1 subunits up-regulation in the SN. We also found that isradipine prevented against MPTP-induced DA neuron depletion in the SN and partly restored the DA content in the striatum. Moreover, we found that isradipine inhibited the increase of iron positive cells in the SN of the MPTP-treated mice. Finally, we investigated the effects of isradipine on cellular iron accumulation in the dopaminergic MES23.5 cell line. Our studies showed that MPP+ treatment accelerated iron influx in the MES23.5 cells. Treatment with Bayk8644 further aggravated iron accumulation. Treatment with isradipine prevented against MPP+-induced iron influx in the MES23.5 cells. These results suggest that up-regulation of LTCCs may be responsible for the DA neuron degeneration in the MPTP-treated mice, The LTCCs may directly contribute to iron influx into DA neurons, and isradipine may suppress cellular iron accumulation and prevents neurodegeneration.


Assuntos
Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Ferro/metabolismo , Isradipino/farmacologia , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/efeitos adversos , Animais , Biomarcadores , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Dopamina , Neurônios Dopaminérgicos/patologia , Masculino , Camundongos , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/etiologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Substância Negra/patologia
9.
Int J Biochem Cell Biol ; 45(7): 1265-73, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23535049

RESUMO

Embryonic stem cells (ESCs)-based therapies have been increasingly recognized as a potential tool to replace or support cells and their function damaged by the neurodegenerative process that underlies Parkinson's disease (PD). In this study, we implanted engineered mouse embryonic stem (ES) CGR8 cells, which stably co-express glial cell line-derived neurotrophic factor (GDNF) and tyrosine hydroxylase (TH), into striatum (Str) or both Str and substantia nigra (SN) of parkinsonian rats lesioned by 6-hydroxydopamine (6-OHDA). We found that cell transplantation into Str or both Str and SN rescued behavioral abnormalities and striatal DA depletion associated with 6-OHDA lesion. Our findings suggested that the profound functional impairment in nigrostriatal circuitry could be at least partially restored by ESCs-based expression of TH and GDNF, which may be developed into a useful tool for PD therapy.


Assuntos
Células-Tronco Embrionárias/transplante , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Oxidopamina/metabolismo , Doença de Parkinson/terapia , Tirosina 3-Mono-Oxigenase/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Feminino , Terapia Genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Camundongos , Fármacos Neuroprotetores/metabolismo , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/terapia , Doença de Parkinson/metabolismo , Ratos , Ratos Wistar , Substância Negra/citologia , Substância Negra/metabolismo , Tirosina 3-Mono-Oxigenase/biossíntese , Tirosina 3-Mono-Oxigenase/genética
10.
Nat Commun ; 4: 1435, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23385580

RESUMO

The gut-derived orexigenic peptide hormone ghrelin enhances neuronal firing in the substantia nigra pars compacta, where dopaminergic neurons modulate the function of the nigrostriatal system for motor coordination. Here we describe a novel mechanism by which ghrelin enhances firing of nigral dopaminergic neurons by inhibiting voltage-gated potassium Kv7/KCNQ/M-channels through its receptor GHS-R1a and activation of the PLC-PKC pathway. Brain slice recordings of substantia nigra pars compacta neurons reveal that ghrelin inhibits native Kv7/KCNQ/M-currents. This effect is abolished by selective inhibitors of GHS-R1a, PLC and PKC. Transgenic suppression of native Kv7/KCNQ/M-channels in mice or channel blockade with XE991 abolishes ghrelin-induced hyperexcitability. In vivo, intracerebroventricular ghrelin administration causes increased dopamine release and turnover in the striatum. Microinjection of ghrelin or XE991 into substantia nigra pars compacta results in contralateral dystonic posturing, and attenuation of catalepsy elicited by systemic administration of the D2 receptor antagonist haloperidol. Our findings indicate that the ghrelin/KCNQ signalling is likely a common pathway utilized by the nervous system.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Grelina/farmacologia , Canais de Potássio KCNQ/antagonistas & inibidores , Potenciais de Ação/efeitos dos fármacos , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/fisiologia , Catalepsia/metabolismo , Catalepsia/fisiopatologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/enzimologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiologia , Grelina/administração & dosagem , Haloperidol , Injeções Intraventriculares , Canais de Potássio KCNQ/metabolismo , Camundongos , Camundongos Transgênicos , Nistatina/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Ratos , Receptores de Grelina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , Substância Negra/fisiologia , Fosfolipases Tipo C/metabolismo
11.
Neurotox Res ; 22(4): 274-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22259026

RESUMO

The mechanisms of iron accumulation in substantia nigra (SN) of Parkinson's diseases remain unclear. The objective of this study was to investigate effects of nifedipine on iron-overload-induced iron accumulation and neurodegeneration in SN of rats. By high performance liquid chromatography-electrochemical detection, tyrosine hydroxylase (TH) immunohistochemistry, and iron content array, we first quantified iron content and the number of dopamine neurons in SN of experimental rats treated with iron dextran. We further assessed effects of treatment with nifedipine. Our results showed that nifedipine treatment prevents iron dextran-induced dopamine depletion in the striatum. Consistently, we found that nifedipine restores the number of TH-positive neurons reduced by iron dextran overload and prevents increase of iron content in the SN. These results suggested that nifedipine may suppress iron toxicity in dopamine neurons and prevent neurodegeneration.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Ferro/metabolismo , Degeneração Neural , Nifedipino/farmacologia , Substância Negra/citologia , Análise de Variância , Animais , Contagem de Células , Cromatografia Líquida de Alta Pressão , Neurônios Dopaminérgicos/metabolismo , Complexo Ferro-Dextran/toxicidade , Masculino , Degeneração Neural/induzido quimicamente , Degeneração Neural/tratamento farmacológico , Degeneração Neural/metabolismo , Ratos , Ratos Wistar , Tirosina 3-Mono-Oxigenase/metabolismo
13.
Neurosci Lett ; 382(1-2): 102-5, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15911130

RESUMO

C31, consisting of 31 amino acid residues, is generated from the carboxyl terminal fragments (CTFs) of amyloid precursor protein (APP). It has been shown that C31 causes apoptosis in neurons and is present in brains of Alzheimer disease (AD) patients. Using whole-cell patch clamp techniques, we investigated effects of C31 on voltage-gated calcium channel (VGCC) currents and the protective effects of beta-estradiol on PC12 cells. The results demonstrated that C31 induced a significant increase of the VGCC currents in PC12 cells, which was blocked by beta-estradiol. These results suggest that modulation of intracellular calcium levels by VGCC may in part be involved in C31 induced neuronal death associated with AD.


Assuntos
Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Bário , Diferenciação Celular/efeitos dos fármacos , Estradiol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Células PC12 , Técnicas de Patch-Clamp , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA