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










Intervalo de ano de publicação
1.
Toxicol Appl Pharmacol ; 238(1): 11-9, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19361538

RESUMO

Cyanide is a potent inhibitor of mitochondrial oxidative metabolism and produces mitochondria-mediated death of dopaminergic neurons and sublethal intoxications that are associated with a Parkinson-like syndrome. Cyanide toxicity is enhanced when mitochondrial uncoupling is stimulated following up-regulation of uncoupling protein-2 (UCP-2). In this study, the role of a pro-survival protein, Bcl-2, in cyanide-mediated cell death was determined in a rat dopaminergic immortalized mesencephalic cell line (N27 cells). Following pharmacological up-regulation of UCP-2 by treatment with Wy14,643, cyanide reduced cellular Bcl-2 expression by increasing proteasomal degradation of the protein. The increased turnover of Bcl-2 was mediated by an increase of oxidative stress following UCP-2 up-regulation. The oxidative stress involved depletion of mitochondrial glutathione (mtGSH) and increased H2O2 generation. Repletion of mtGSH by loading cells with glutathione ethyl ester reduced H2O2 generation and in turn blocked the cyanide-induced decrease of Bcl-2. To determine if UCP-2 mediated the response, RNAi knock down was conducted. The RNAi decreased cyanide-induced depletion of mtGSH, reduced H2O2 accumulation, and inhibited down-regulation of Bcl-2, thus blocking cell death. To confirm the role of Bcl-2 down-regulation in the cell death, it was shown that over-expression of Bcl-2 by cDNA transfection attenuated the enhancement of cyanide toxicity after UCP-2 up-regulation. It was concluded that UCP-2 up-regulation sensitizes cells to cyanide by increasing cellular oxidative stress, leading to an increase of Bcl-2 degradation. Then the reduced Bcl-2 levels sensitize the cells to cyanide-mediated cell death.


Assuntos
Morte Celular/efeitos dos fármacos , Cianetos/toxicidade , Canais Iônicos/efeitos dos fármacos , Proteínas Mitocondriais/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Animais , Linhagem Celular Transformada , DNA Complementar , Dopamina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Peróxido de Hidrogênio/metabolismo , Canais Iônicos/genética , Mesencéfalo/citologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Pirimidinas/farmacologia , Interferência de RNA , Ratos , Transfecção , Proteína Desacopladora 2 , Regulação para Cima/efeitos dos fármacos
2.
J Neurosci Res ; 86(6): 1397-408, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18183618

RESUMO

1Alpha,25-dihydroxyvitamin D(3) (VD(3)) is a neuroprotectant that can reduce cytotoxicity produced by a variety of toxicants. The mechanism of the neuroprotection was studied in rat primary cortical cells in which Wy14,643, an agonist of peroxisome proliferator activated receptor-alpha (PPARalpha), enhances cyanide (KCN) neurotoxicity. In this cell model, Wy14,643 pretreatment enhanced cyanide-induced cell death, and the increased cell death was linked to up-regulation of uncoupling protein-2 (UCP-2). VD(3) reversed cyanide-induced mitochondrial dysfunction in cells pretreated with Wy14,643, as reflected by restoration of cellular ATP and mitochondrial membrane potential (DeltaPsi(m)). Analysis of cellular state 4 oxygen consumption showed increased mitochondrial uncoupling accompanied by up-regulation of UPC-2. The uncoupling was attenuated by prior treatment with VD(3). The interaction of VD(3) with UCP-2 was attributed to increased expression of IkappaB, an inhibitor of NF-kappaB (transcription factor that regulates UCP-2 expression). The increased IkappaB levels lead to reduced nuclear translocation and DNA binding of nuclear factor-kappaB. The role of oxidative stress in the response was then evaluated. Cotreatment with Wy14,643 and cyanide markedly increased reactive oxygen species generation and decreased reduced glutathione levels. The oxidative stress was blocked by VD(3) pretreatment. It was concluded that VD(3) blocks Wy14,643 enhancement of cyanide neurotoxicity by suppressing the redox-mediated transcriptional up-regulation of UCP-2, resulting in reduced mitochondrial proton leak and stabilization of mitochondrial function.


Assuntos
Cianetos/toxicidade , Canais Iônicos/metabolismo , Proteínas Mitocondriais/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Vitamina D/análogos & derivados , Animais , Antioxidantes/farmacologia , Western Blotting , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Canais Iônicos/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas Mitocondriais/efeitos dos fármacos , Mutagênicos/toxicidade , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Consumo de Oxigênio/efeitos dos fármacos , Pirimidinas/toxicidade , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Desacopladora 2 , Regulação para Cima , Vitamina D/farmacologia
3.
Neuroscience ; 150(1): 159-67, 2007 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-17980495

RESUMO

Bcl-2/adenovirus E1B 19-kDa-interacting protein 3 (BNIP3), a Bcl-2 homology domain 3 (BH3) domain only protein, has been identified as a mitochondrial mediator of hypoxia-induced cell death. Since cyanide produces histotoxic anoxia (chemical hypoxia), the present study was undertaken in primary rat cortical cells to determine involvement of the BNIP3 signaling pathway in cyanide-induced death. Over a 20 h exposure KCN increased BNIP3 expression, followed by a concentration-related apoptotic death. To determine if BNIP3 plays a role in the cell death, expression was either increased with BNIP3 cDNA (BNIP3+) or knocked down with small interfering RNA (RNAi). In BNIP3+ cells, cyanide-induced apoptotic death was markedly enhanced and preceded by reduction of mitochondrial membrane potential (delta psim), release of cytochrome c from mitochondria and elevated caspase 3 and 7 activity. Pretreatment with the pan-caspase inhibitor N-benzyloxycarbonyl-Ala-Asp-fluoromethyl ketone (zVAD-fmk) suppressed BNIP3+-mediated cell death, thus confirming a caspase-dependent apoptosis. On the other hand, BNIP3 knockdown by RNAi or antagonism of BNIP3 by a transmembrane-deleted dominant-negative mutant (BNIP3 delta TM) markedly reduced cell death. Immunohistochemical imaging showed that cyanide stimulated translocation of BNIP3 from cytosol to mitochondria and displacement studies with BNIP3 delta TM showed that integration of BNIP3 into the mitochondrial outer membrane was necessary for the cell death. In BNIP3+ cells, cyclosporin-A, an inhibitor of mitochondrial pore transition, blocked the cyanide-induced reduction of delta psim and decreased the apoptotic death. These results demonstrate in cortical cells that cyanide induces a rapid upregulation of BNIP3 expression, followed by translocation to the mitochondrial outer membrane to reduce delta psim. This was followed by mitochondrial release of cytochrome c to execute a caspase-dependent cell death.


Assuntos
Apoptose/efeitos dos fármacos , Córtex Cerebral/citologia , Cianetos/farmacologia , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Análise de Variância , Animais , Caspases/metabolismo , Células Cultivadas , Ciclosporina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Embrião de Mamíferos , Marcação In Situ das Extremidades Cortadas , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas Mitocondriais , Mutação/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/farmacologia , Ratos , Fatores de Tempo , Transfecção/métodos
4.
Toxicol Appl Pharmacol ; 223(1): 10-9, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17573087

RESUMO

Uncoupling protein 2 (UCP-2) is an inner mitochondrial membrane proton carrier that modulates mitochondrial membrane potential (DeltaPsi(m)) and uncouples oxidative phosphorylation. We have shown that up-regulation of UCP-2 by Wy14,643, a selective peroxisome proliferator-activated receptor-alpha (PPARalpha) agonist, enhances cyanide cytotoxicity. The pathway by which Wy14,643 up-regulates UCP-2 was determined in a dopaminergic cell line (N27 cells). Since dopaminergic mesencephalic cells are a primary brain target of cyanide, the N27 immortalized mesencephalic cell was used in this study. Wy14,643 produced a concentration- and time-dependent up-regulation of UCP-2 that was linked to enhanced cyanide-induced cell death. MK886 (PPARalpha antagonist) or PPARalpha knock-down by RNA interference (RNAi) inhibited PPARalpha activity as shown by the peroxisome proliferator response element-luciferase reporter assay, but only partially decreased up-regulation of UCP-2. The role of oxidative stress as an alternative pathway to UCP-2 up-regulation was determined. Wy14,643 induced a rapid surge of ROS generation and loading cells with glutathione ethyl ester (GSH-EE) or pre-treatment with vitamin E attenuated up-regulation of UCP-2. On the other hand, RNAi knockdown of PPARalpha did not alter ROS generation, suggesting a PPARalpha-independent component to the response. Co-treatment with PPARalpha-RNAi and GSH-EE blocked both the up-regulation of UCP-2 by Wy14,643 and the cyanide-induced cell death. It was concluded that a PPARalpha-mediated pathway and an oxidative stress pathway independent of PPARalpha mediate the up-regulation of UCP-2 and subsequent increased vulnerability to cyanide-induced cytotoxicity.


Assuntos
Inibidores Enzimáticos/toxicidade , Canais Iônicos/metabolismo , Proteínas Mitocondriais/metabolismo , Estresse Oxidativo , PPAR alfa/metabolismo , Cianeto de Potássio/toxicidade , Regulação para Cima/fisiologia , Animais , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Interações Medicamentosas , Inativação Gênica , Glutationa/análogos & derivados , Glutationa/farmacologia , Indóis/farmacologia , Canais Iônicos/genética , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , PPAR alfa/agonistas , Pirimidinas/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Proteína Desacopladora 2 , Regulação para Cima/efeitos dos fármacos , Vitamina E/farmacologia
5.
Free Radic Biol Med ; 43(1): 117-27, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17561100

RESUMO

Cyanide produces degeneration of the nervous system in which different modes of cell death are activated in the vulnerable brain areas. In brain, the mechanism underlying the cell death is not clear. In this study, an immortalized dopaminergic cell line was used to characterize the cell death signaling cascade activated by cyanide. Cyanide-treated cells exhibited a time- and concentration-dependent apoptosis that was caspase independent. Cyanide induced a rapid surge of intracellular reactive oxygen species (ROS) generation, followed by p38 mitogen-activated protein kinase (MAPK) activation and nuclear accumulation of hypoxia-inducible factor-1alpha (HIF-1alpha). Activation of p38 MAPK and HIF-1alpha accumulation were attenuated by N-acetyl-L-cysteine (antioxidant), catalase (hydrogen peroxide scavenger), or a selective p38 MAPK inhibitor (SB203580). Cyanide activated the hypoxia response element (HRE) promoter, which was also blocked by the antioxidants and SB203580. HRE activation was followed by increased BNIP3 gene transcription, as reflected by elevated BNIP3 mRNA and protein levels. BNIP3 upregulation was reduced by selective RNAi knockdown of HIF-1alpha. Overexpression of BNIP3 produced mitochondrial dysfunction (reduced membrane potential), caspase-independent apoptosis, and sensitization of the cells to cyanide-induced toxicity. Expression of a dominant-negative mutant or RNAi knockdown of BNIP3 protected the cells from cyanide. It was concluded that cyanide activated the HIF-1alpha-mediated pathway of BNIP3 induction through a redox-sensitive process. Increased BNIP3 expression then served as an initiator of mitochondrial-mediated death.


Assuntos
Apoptose , Cianetos/toxicidade , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Acetilcisteína/farmacologia , Animais , Caspases/metabolismo , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imidazóis/farmacologia , Potencial da Membrana Mitocondrial , Proteínas de Membrana/genética , Camundongos , Oxirredução , Estresse Oxidativo/genética , Proteínas Proto-Oncogênicas/genética , Piridinas/farmacologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
J Neurosci Res ; 84(5): 1003-11, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16933320

RESUMO

We have previously shown in rat primary cultured mesencephalic cells that cyanide induces a high level of oxidative stress and necrotic death. To evaluate the mechanism of the cytotoxicity, the effects of cyanide on intracellular glutathione (GSH) pools and inducible nitric oxide synthase (iNOS)-mediated reactive nitrogen species (RNS) generation were studied. Cyanide rapidly depleted intracellular GSH. Restoration of GSH blocked cell death, whereas depletion of GSH by synthesis inhibition increased the necrosis. Selective depletion of mitochondrial GSH (mtGSH) increased oxidative stress and enhanced cell death, whereas the cytoplasmic pool was not critical to cell survival. These actions were accompanied by increased iNOS expression as determined by Western blot analysis, RT-PCR and immunohistochemistry. Up-regulation of iNOS led to increased generation of NO as reflected by elevated nitrite levels (an end product of NO metabolism). It was determined by use of a selective inhibitor that up-regulation of iNOS expression was transcriptionally regulated by activation of nuclear factor-kappaB, a redox-sensitive transcription factor. It was concluded that, in cyanide-mediated neurotoxicity, mtGSH is a vital component of the cellular antioxidant defense, and its depletion can lead to oxidative stress-mediated iNOS up-regulation, thus enhancing RNS generation and necrosis.


Assuntos
Cianetos/farmacologia , Glutationa/metabolismo , Mesencéfalo/citologia , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Regulação para Cima/efeitos dos fármacos , Análise de Variância , Animais , Western Blotting/métodos , Morte Celular/efeitos dos fármacos , Células Cultivadas , Interações Medicamentosas , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Imuno-Histoquímica/métodos , Mitocôndrias/enzimologia , Necrose/induzido quimicamente , Neurônios/enzimologia , Neurônios/patologia , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
7.
Toxicol Appl Pharmacol ; 216(1): 34-43, 2006 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16797631

RESUMO

Trimethyltin (TMT) produces selective neuronal degeneration in the central nervous system (CNS), in which the hippocampus is the most sensitive area. Since previous studies have been conducted in either non-neural cells or mixed primary cultures, an immortalized hippocampal neuronal cell line (HT-22 cell) was used to assess the mechanism and mode of death produced by TMT. The compound produced a time- and concentration-dependent apoptotic death that was caspase-mediated. Excessive generation of reactive oxygen species (ROS) and subsequent reduction of mitochondrial membrane potential (DeltaPsim) were involved in the cytotoxicity. Scavenging of ROS by a free radical trapping agent or inhibition of the mitochondrial permeability transition (MPT) pore significantly reduced cell death. Additionally, TMT increased expression of inducible nitric oxide synthase (iNOS) by activation of the redox-sensitive transcription factor NFkappaB. Pharmacologic inhibition studies showed that the iNOS-mediated NO generation increased expression of Bax and then mitochondrial-mediated apoptosis. It was concluded that excessive ROS generation initiated the apoptotic cell death by upregulating iNOS followed by increased Bax expression which then led to loss of DeltaPsim and caspase-executed cell death. This study is the first to report in a neuronal cell model that TMT stimulates induction of iNOS, which then increases cellular levels of reactive nitrogen species (RNS) to initiate apoptotic death.


Assuntos
Apoptose/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Compostos de Trimetilestanho/toxicidade , Proteína X Associada a bcl-2/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Caspase 3/metabolismo , Inibidores de Caspase , Linhagem Celular , Óxidos N-Cíclicos/farmacologia , Ciclosporina/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Transporte da Membrana Mitocondrial/antagonistas & inibidores , Membranas Mitocondriais/efeitos dos fármacos , Membranas Mitocondriais/fisiologia , Poro de Transição de Permeabilidade Mitocondrial , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxidos de Nitrogênio/farmacologia , Oligonucleotídeos Antissenso/farmacologia , Peptídeos/farmacologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Compostos de Trimetilestanho/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos
8.
Toxicol Sci ; 86(1): 116-24, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15800031

RESUMO

Uncoupling protein 2 (UCP-2) is expressed in the inner mitochondrial membrane and modulates mitochondrial function by partially uncoupling oxidative phosphorylation, and it has been reported to modulate cell death. Cyanide is a potent neurotoxin that inhibits complex IV to alter mitochondrial function to induce neuronal death. In primary rat cortical cells KCN produced an apoptotic death at 200-400 microM. Higher concentrations of potassium cyanide (KCN) (500-600 microM) switched the mode of death from apoptosis to necrosis. In necrotic cells, ATP levels were severely depleted as compared to cortical cells undergoing apoptosis. To determine if UCP-2 expression could alter KCN-induced cell death, cells were transiently transfected with full-length human UCP-2 cDNA (UCP-2+). Overexpression switched the mode of death produced by KCN (400 microM) from apoptosis to necrosis. The change in cell death was mediated by impaired mitochondrial function as reflected by a marked decrease of ATP levels and reduction in mitochondrial membrane potential. RNA interference or transfection with a dominant interfering mutant blocked the necrotic response observed in UCP-2+ cells. Additionally, treatment of UCP-2+ cells with cyclosporin A blocked necrosis, indicating the involvement of mitochondrial permeability pore transition in the necrotic death. These results show that increased expression of UCP-2 alters the response to a potent mitochondrial toxin by switching the mode of cell death from apoptosis to necrosis. It is concluded that UCP-2 levels influence cellular responses to cyanide-induced mitochondrial dysfunction.


Assuntos
Cianetos/toxicidade , Mitocôndrias/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Mitocôndrias/fisiologia , Ratos , Ratos Sprague-Dawley
9.
Neurosci Res ; 49(1): 13-8, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15099699

RESUMO

Cyanide is generated in neurons and this report examines the two different receptors which mediate cyanide formation in neuronal tissue. An opiate receptor blocked by naloxone increases cyanide production both in rat brain and in rat pheochromocytoma (PC12) cells. A muscarinic receptor in PC12 cells releases cyanide and the effect is blocked by atropine. In rat brain, in vivo, a muscarinic agonist inhibits cyanide generation, possibly by acting on receptor subtypes different from those in PC12 cells. Cyanide generation by a muscarinic agonist in PC12 cells is blocked by pertussis toxin but that caused by an opiate is not. Thus, two different receptors and two different second messenger systems can mediate cyanide generation in PC12 cells. In parallel with the in vivo data, cultured primary rat cortical cells also show decreased cyanide release following muscarinic stimulation. Both blockade of cyanide generation by muscarinic receptor activation and cyanide release by opiate agonists from cortical cells are pertussis toxin insensitive. Similarly, little cyanide generation was seen following cholera toxin treatment. These data indicate that opiate receptors increase and muscarinic receptors decrease cyanide production in rat brain tissue by G-protein independent mechanisms. This work supports the suggestion that the powerful actions of cyanide may be important for neuromodulation in the CNS.


Assuntos
Encéfalo/metabolismo , Cianetos/metabolismo , Oxotremorina/análogos & derivados , Células PC12/metabolismo , Receptores de Superfície Celular/metabolismo , Análise de Variância , Animais , Atropina/farmacologia , Encéfalo/efeitos dos fármacos , Carbacol/farmacologia , Células Cultivadas , Agonistas Colinérgicos/farmacologia , Interações Medicamentosas , Embrião de Mamíferos , Hidromorfona/farmacologia , Morfina/farmacologia , Agonistas Muscarínicos/farmacologia , Antagonistas Muscarínicos/farmacologia , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Entorpecentes/farmacologia , Oxotremorina/farmacologia , Células PC12/efeitos dos fármacos , Toxina Pertussis/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Superfície Celular/efeitos dos fármacos
10.
Toxicol Sci ; 75(1): 99-107, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12805646

RESUMO

Execution of cyanide-induced apoptosis is mediated by release of cytochrome c from mitochondria. To determine how cyanide initiates cytochrome c release, Bax translocation was investigated in primary cultures of cortical neurons. Under nonapoptotic (control) conditions, Bax resided predominantly in the cytoplasm. After 300-microM cyanide treatment for 1 h, Bax translocated to the mitochondria, as shown by immunocytochemical staining and subcellular fractionation; Western blot analysis confirmed "cytosol-to-mitochondria" translocation of Bax. Temporal analysis showed that Bax translocation preceded cytochrome c release from the mitochondria, which was initiated 3 h after cyanide treatment. In double-immunofluorescence labeling for both Bax and cytochrome c, it was observed that cytochrome c was released only in cells showing Bax in mitochondria. The role of p38 mitogen-activated protein (MAP) kinase in Bax translocation was studied. The p38 MAP kinase was activated 30 min after cyanide, and its phosphorylation level of activity began to decrease 3 h later. SB203580, a p38 MAP kinase inhibitor, blocked translocation of Bax to mitochondria, whereas SB202474, a control peptide, had no effect on translocation. Inhibition of p38 MAP kinase by SB203580 blocked all downstream effects of Bax translocation, including cytochrome c release, caspase activation, and internucleosomal DNA fragmentation. These results demonstrated that Bax translocation is critical for cyanide-induced cytochrome c release and that p38 MAP kinase regulates Bax translocation from cytosol to mitochondria.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Cianeto de Potássio/toxicidade , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas/metabolismo , Animais , Western Blotting , Caspases/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Córtex Cerebral/ultraestrutura , Citocromos c/biossíntese , Citoplasma/metabolismo , Inibidores Enzimáticos/farmacologia , Mitocôndrias/metabolismo , Neurônios/metabolismo , Neurônios/ultraestrutura , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Proteína X Associada a bcl-2 , Proteínas Quinases p38 Ativadas por Mitógeno
11.
Toxicol Appl Pharmacol ; 185(1): 55-63, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12460737

RESUMO

Cyanide (KCN)-induced generation of reactive oxygen species (ROS) involves cyclooxygenase-2 (COX-2)-mediated reactions in some neurons. The present study examines the extent to which COX isoforms are involved in KCN-induced apoptotic cell death processes of cultured cortical cells. After treatment with KCN (10-300 microM), COX-2 was expressed in a time- and concentration-dependent manner increasing markedly over a 4-h period. However, no significant changes were observed in COX-1 levels at any cyanide concentration. Correlated with COX-2 up-regulation, KCN induced a time-dependent apoptotic death. TUNEL staining showed that the COX-2 inhibitor NS-398 (30 microM) blocked KCN-induced apoptosis, whereas the selective COX-1 inhibitor valeryl salicylate did not affect the level of apoptotic cell death. Exposure of cells to KCN (300 microM) for 24 h resulted in DNA fragmentation, which was also reduced by NS-398. Prostaglandin E(2) (PGE(2)) accumulation in cell culture supernatants was increased by KCN and NS-398 blocked PGE(2) generation. PCR studies further confirmed that COX-2 expression was increased by KCN. Antioxidants phenyl-N-test-butylnitrone, superoxide dismutase, and catalase significantly inhibited KCN-induced COX-2 up-regulation and subsequent apoptosis. N(G)-nitro-L-arginine methylester an inhibitor of nitric oxide synthase, blocked KCN-induced PGE(2) production and apoptosis, but not COX-2 expression. Increased nitric oxide levels caused by cyanide may directly activate the COX-2 enzyme. These data show that cyanide treatment of cortical cells involves increased COX-2 expression, PGE(2) accumulation, and ROS generation, resulting in apoptotic cell death.


Assuntos
Apoptose/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Isoenzimas/biossíntese , Estresse Oxidativo , Cianeto de Potássio/toxicidade , Prostaglandina-Endoperóxido Sintases/biossíntese , Animais , Antioxidantes/farmacologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/biossíntese , Indução Enzimática/efeitos dos fármacos , NG-Nitroarginina Metil Éster/farmacologia , Ratos , Ratos Sprague-Dawley
12.
J Pharmacol Exp Ther ; 303(2): 510-9, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12388630

RESUMO

A comparative study was conducted in rat primary cortical (CX) and mesencephalic (MC) neurons to investigate intracellular cascades activated during cyanide-induced injury and to determine the point at which the cascades diverge to produce either apoptosis or necrosis. Cyanide treatment (400 microM) for 24 h produced primarily apoptosis in CX cells, whereas the same concentration of cyanide induced predominantly necrosis in MC cells as indicated by increased propidium iodide staining and cellular lactate dehydrogenase efflux. Cyanide increased generation of cellular reactive oxygen species (ROS) in both CX and MC cells, but the rate of formation and nature of the oxidative species varied with cell type. Catalase decreased cyanide-induced ROS generation in CX but not in MC cells. Nitric oxide generation was more prominent after cyanide treatment of MC compared with CX cells. N-Methyl-D-aspartate receptors were more involved in CX apoptosis than in MC necrosis. Mitochondrial membrane potential decreased moderately in CX cells on exposure to cyanide, whereas MC cells responded with a more pronounced reduction in potential. In CX cells cyanide produced a concentration-dependent release of cytochrome c from mitochondria and increased caspase activity, whereas little change was seen in MC neurons. Thus, cyanide-induced necrosis of MC cells involved generation of excessive amounts of nitric oxide and superoxide accompanied by mitochondrial depolarization. In contrast cyanide causes a lower level of oxidative stress in CX cells, involving mainly hydrogen peroxide and superoxide, and a moderate change in mitochondrial membrane potential that lead to cytochrome c release, caspase activation, and apoptosis.


Assuntos
Córtex Cerebral/patologia , Cianetos/toxicidade , Mesencéfalo/patologia , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3 , Inibidores de Caspase , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , DNA/biossíntese , DNA/isolamento & purificação , Eletroforese em Gel de Poliacrilamida , Inibidores Enzimáticos/farmacologia , Marcação In Situ das Extremidades Cortadas , Potenciais da Membrana/efeitos dos fármacos , Mesencéfalo/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Necrose , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Ratos
13.
Toxicol Appl Pharmacol ; 177(1): 17-25, 2001 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11708896

RESUMO

Pyridostigmine bromide (PB) is a reversible cholinesterase inhibitor used for treatment of myasthenia gravis and for prophylactic protection against organophosphate nerve agent. We previously showed PB can induce apoptotic death in rat brain following systemic treatment. To study mechanisms by which PB induces brain cell death, cultured rat cerebellar granule cells were used. Cytotoxicity was determined after exposure to PB (10-1000 microM) for 24 h; a high concentration of PB (>500 microM) significantly increased lactate dehydrogenase release, which was reduced by pretreatment with the antioxidant, N-t-butyl-alpha-phenyl-nitrone (PBN). Apoptosis, as determined by TUNEL staining, was concentration dependent (10-250 microM) after a 24-h exposure and cytotoxicity was confirmed by gel electrophoresis of DNA, release of cytochrome c from mitochondria, elevation of caspase activity, and electron microscopy. The oxidant-sensitive fluorescent dye 2',7'-dichlorofluorescin diacetate was used to detect reactive oxidative species (ROS) generation. Pretreatment with PBN, superoxide dismutase, catalase, or the nitric oxide synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME) blocked PB-induced ROS generation and apoptotic cell death. Pretreatment with atropine or MK-801 blocked ROS generation and the subsequent neurotoxicity, showing that both muscarinic and NMDA receptors mediate the response. DNA extracted from PB-treated cells revealed oligonucleosomal fragmentation on gel electrophoresis and antioxidants attenuated the DNA fragmentation, providing further evidence for a link of ROS generation and apoptosis. These results indicate that muscarinic receptor-mediated ROS generation is an initiating factor in PB-induced apoptotic cell death and activation of the NMDA glutamate receptor is directly linked to the response.


Assuntos
Apoptose/efeitos dos fármacos , Colinérgicos/metabolismo , Neurônios/efeitos dos fármacos , Brometo de Piridostigmina/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Animais Recém-Nascidos , Antioxidantes/farmacologia , Atropina/farmacologia , Caspase 3 , Caspases/biossíntese , Catalase/farmacologia , Células Cultivadas , Cerebelo/citologia , Óxidos N-Cíclicos , DNA/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Relação Dose-Resposta a Droga , Eletroforese em Gel de Ágar , Marcação In Situ das Extremidades Cortadas , L-Lactato Desidrogenase/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Neurônios/enzimologia , Neurônios/ultraestrutura , Óxidos de Nitrogênio/farmacologia , Ratos , Superóxido Dismutase/farmacologia
14.
Toxicol Sci ; 64(1): 83-9, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11606804

RESUMO

In evaluating mechanisms of trimethyltin (TMT)-initiated neuronal damage, the present study focused on involvement of reactive oxygen species, protein kinase C (PKC), and glutamate receptors. Exposure of cerebellar granule cells to TMT (0.01-0.1 microM) produced primarily apoptosis, but higher concentrations were associated with cellular lactate dehydrogenase efflux and necrosis. TMT increased generation of cellular reactive oxygen species, which was inhibited by either L-NAME (inhibitor of nitric oxide synthase, NOS) or catalase, indicating that both NO and H(2)O(2) are formed on TMT exposure. Since chelerythrine (selective PKC inhibitor) also inhibited oxidative species generation, PKC appears to play a significant role in TMT-induced oxidative stress. The metabotropic glutamate receptor antagonist, MCPG, (but not MK-801) prevented oxidative species generation, indicating significant involvement of metabotropic receptors (but not NMDA receptors) in TMT-induced oxidative stress. NOS involvement in the action of TMT was confirmed through measurement of nitrite, which increased concentration dependently. Nitrite accumulation was blocked by L-NAME, chelerythrine, or MCPG, showing that NO is generated by TMT and that associated changes in NOS are regulated by a PKC-mediated mechanism. Oxidative damage by TMT was demonstrated by detection of elevated malondialdehyde levels. It was concluded that low concentrations of TMT (0.01-0.1 microM) cause apoptotic cell death in which oxidative signaling is an important event. Higher concentrations of TMT initiate necrotic death, which involves both an oxidative and a non-oxidative component. TMT-induced necrosis but not apoptosis in granule cells is mediated by glutamate receptors.


Assuntos
Apoptose/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Necrose , Neurônios/efeitos dos fármacos , Compostos de Trimetilestanho/toxicidade , Alcaloides , Animais , Apoptose/fisiologia , Benzofenantridinas , Catalase/farmacologia , Células Cultivadas , Cerebelo/citologia , Cerebelo/metabolismo , Maleato de Dizocilpina/farmacologia , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Marcação In Situ das Extremidades Cortadas , L-Lactato Desidrogenase/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Neurônios/citologia , Neurônios/metabolismo , Nitritos/metabolismo , Fenantridinas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Glutamato/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Compostos de Trimetilestanho/administração & dosagem
15.
J Biochem Mol Toxicol ; 15(5): 256-62, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11835622

RESUMO

Although the neurotoxicity of trimethyltin (TMT) is well known, mechanisms are still not clear. Glia have been proposed to mediate the toxic action of TMT on nerve cells. Accordingly, the effects of TMT were tested in primary neuronal cultures from rat cerebellum and compared to effects in astrocytes and mixed cultures. Neuronal damage observed following TMT exposure was less in the presence of astrocytes and astrocytes alone were resistant to TMT. Thus, astrocytes have a protective effect against TMT-induced neurotoxicity. TMT caused an oxidative stress in granule cell cultures involving a variety of oxidative species (O2)*-, H2O2, NO), but astrocytes were less sensitive to TMT-induced oxidative species generation. Antioxidants, glutathione and 7-nitroindazole attenuated neuronal cell death induced by TMT. It appears that oxidative stress mediates a large part of the destructive action of TMT in neuronal cultures. The presence of astrocytes appears to modulate TMT-induced oxidative stress so that TMT causes only a small increase in lipid peroxidation in mouse brain after systemic administration. Thus, TMT induces a pronounced oxidative stress in cultured neurons, but when astrocytes are present, oxidative species play a lesser role in the neurotoxic action of TMT.


Assuntos
Astrócitos/efeitos dos fármacos , Compostos de Trimetilestanho/toxicidade , Animais , Astrócitos/metabolismo , Astrócitos/fisiologia , Células Cultivadas , Cerebelo/citologia , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/metabolismo , Glutationa/metabolismo , Masculino , Camundongos , Estresse Oxidativo , Ratos
16.
Neurotoxicology ; 21(4): 541-52, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11022862

RESUMO

Pyridostigmine is a reversible cholinesterase (ChE) inhibitor that is associated with neurologic dysfunction involving both central and peripheral nervous systems. To determine the neurotoxic potential of pyridostigmine, rats were sacrificed at intervals after drug administration (0.5-1.85 mg/kg, i.p., twice daily for 4 days) and brains examined histologically. ChE inhibition was used as a biomarker of pyridostigmine activity. Using the in situ terminal deoxynucleotidyl transferase nick-end labeling of DNA fragments (TUNEL) method and electron microscopy, apoptotic brain cell death was noted in cerebral cortex over a dose range of 0.5-1.85 mg/kg and at the higher dose (1.85 mg/kg), apoptosis was also noted in striatum and hippocampus. These responses were blocked by pretreatment with atropine. Rat cortical cells in culture also underwent apoptosis when exposed to pyridostigmine (250 microM for 24 hr), indicating that the pyridostigmine can initiate apoptosis, independent of peripheral mechanisms. Pretreatment of cells with atropine (10 microM) inhibited pyridostigmine-induced apoptosis, confirming the response was mediated by muscarinic receptors. Short term treatment of rats with pyridostigmine (1.85 mg/kg twice daily for 4 days) induced a prolonged apoptotic response, which was evident in rat cortex up to 30 days after the last dose. Active apoptosis persisted, despite recovery of serum ChE activity. These in vivo and in vitro observations indicate that pyridostigmine can initiate a prolonged neurodegeneration.


Assuntos
Apoptose/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Parassimpatomiméticos/farmacologia , Brometo de Piridostigmina/farmacologia , Receptores Muscarínicos/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Colinesterases/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Masculino , Microscopia Eletrônica , Neurônios/ultraestrutura , Ratos , Ratos Sprague-Dawley
17.
J Neurosci Res ; 61(5): 570-5, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10956427

RESUMO

In a study of the mechanism by which cyanide is produced in neural tissue, it was hypothesized that nerve cells generate cyanide in a manner similar to that in leukocytes. As in white blood cells, glycine addition enhanced cyanide production in rat pheochromocytoma cells. Because myeloperoxidase catalyses cyanide production in leukocytes, a selective myeloperoxidase inhibitor (aminobenzoic acid hydrazide) was tested and found to inhibit opiate agonist-induced cyanide production in pheochromocytoma cells and also in rat brain. In addition, hydrogen peroxide enhanced cyanide release in pheochromocytoma cells, further suggesting that the process is oxidative in nature. Sonicated rat pheochromocytoma cells did not generate cyanide in response to an agonist acting on surface receptors even though disrupted cells responded to glycine. The mitochondrial fraction from rat brain produced more cyanide in response to glycine than any other fraction. Thus glycine seems to act at an intracellular site to enhance cyanide production and the process seems to involve a peroxidase mechanism similar to that reported for white blood cells.


Assuntos
Cianetos/metabolismo , Neurônios/metabolismo , Peroxidases/metabolismo , Aminobenzoatos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Azidas/farmacologia , Química Encefálica/efeitos dos fármacos , Carbacol/farmacologia , Fracionamento Celular , Agonistas Colinérgicos/farmacologia , Relação Dose-Resposta a Droga , Glicina/farmacologia , Peróxido de Hidrogênio/farmacologia , Hidromorfona/farmacologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Morfina/farmacologia , Entorpecentes/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Oxidantes/farmacologia , Células PC12 , Peroxidases/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
18.
J Neurochem ; 74(6): 2296-304, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10820189

RESUMO

Dopamine (DA) oxidation and the generation of reactive oxygen species (ROS) may contribute to the degeneration of dopaminergic neurons underlying various neurological conditions. The present study demonstrates that DA-induced cytotoxicity in differentiated PC12 cells is mediated by ROS and mitochondrial inhibition. Because cyanide induces parkinson-like symptoms and is an inhibitor of the antioxidant system and mitochondrial function, cells were treated with KCN to study DA toxicity in an impaired neuronal system. Differentiated PC12 cells were exposed to DA, KCN, or a combination of the two for 12-36 h. Lactate dehydrogenase (LDH) assays indicated that both DA (100-500 microM) and KCN (100-500 microM) induced a concentration- and time-dependent cell death and that their combination produced an increase in cytotoxicity. Apoptotic death, measured by Hoechst dye and TUNEL (terminal deoxynucleotidyltransferase dUTP nick end-labeling) staining, was also concentration- and time-dependent for DA and KCN. DA plus KCN produced an increase in apoptosis, indicating that KCN, and thus an impaired system, enhances DA-induced apoptosis. To study the mechanism(s) of DA toxicity, cells were pretreated with a series of compounds and incubated with DA (300 microM) and/or KCN (100 microM) for 24 h. Nomifensine, a DA reuptake inhibitor, rescued nearly 60-70% of the cells from DA- and DA plus KCN-induced apoptosis, suggesting that DA toxicity is in part mediated intracellularly. Pretreatment with antioxidants attenuated DA- and KCN-induced apoptosis, indicating the involvement of oxidative species. Furthermore, buthionine sulfoximine, an inhibitor of glutathione synthesis, increased the apoptotic response, which was reversed when cells were pretreated with antioxidants. DA and DA plus KCN produced a significant increase in intracellular oxidant generation, supporting the involvement of oxidative stress in DA-induced apoptosis. The nitric oxide synthase inhibitor N(G)-nitro-L-arginine methyl ester and the peroxynitrite scavenger uric acid blocked apoptosis and oxidant production, indicating involvement of nitric oxide. These results suggest that DA neurotoxicity is enhanced under the conditions induced by cyanide and involves both ROS and nitric oxide-mediated oxidative stress as an initiator of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Dopamina/farmacologia , Inibidores Enzimáticos/farmacologia , Neurônios/citologia , Estresse Oxidativo/efeitos dos fármacos , Cianeto de Potássio/farmacologia , Animais , Apoptose/fisiologia , Diferenciação Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , L-Lactato Desidrogenase/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Óxido Nítrico/metabolismo , Estresse Oxidativo/fisiologia , Células PC12 , Ratos
19.
Toxicol Appl Pharmacol ; 164(2): 196-205, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10764633

RESUMO

The central nervous system is one of the main target organs in cyanide toxicity. Primary cultured cortical neurons were used to study the cellular mechanisms underlying cyanide-induced cytotoxicity. After exposure to KCN (100-300 microM) for 24 h, cortical neurons underwent apoptosis as characterized by positive TUNEL staining. Reactive oxygen species (ROS) play an important role in cyanide-induced neuronal apoptosis; immediately after cyanide (100-300 microM) treatment, ROS generation was observed and continued to be elevated for up to 3 h. NMDA receptor activation and subsequent Ca(2+) influx contribute in part to cyanide-induced ROS formation, since the selective NMDA receptor antagonist MK801 and intracellular Ca(2+) chelator BAPTA blocked ROS generation. Interestingly, caspases, recently reported to be involved in neuronal apoptosis, play a role in the late phase of ROS production after cyanide stimulation. Z-VAD, a nonspecific caspase inhibitor, blocked ROS generated 1 h after cyanide treatment, but it had no effect on ROS generated immediately after cyanide treatment. Nuclear factor kappaB (NF-kappaB), a redox-sensitive transcription factor, was activated dose dependently after cyanide treatment. Blockade of ROS generation by MK801, Z-VAD, and various antioxidants also blocked the activation of NF-kappaB. SN50, a synthetic peptide which inhibits the nuclear translocation of NF-kappaB, blocked cyanide-induced apoptotic cell death. These results indicate that NF-kappaB plays an important role in cyanide-induced apoptosis in cortical neurons, and the caspases may contribute in part to the activation of NF-kappaB after cyanide treatment by inducing the late phase of ROS generation.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Córtex Cerebral/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Estresse Oxidativo , Venenos/toxicidade , Cianeto de Potássio/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Animais , Caspases/efeitos dos fármacos , Córtex Cerebral/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Interações Medicamentosas , Marcação In Situ das Extremidades Cortadas , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley
20.
J Biochem Mol Toxicol ; 13(5): 253-9, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10402559

RESUMO

Activation of NMDA receptors plays an important role in cyanide neurotoxicity. Cyanide indirectly activates the receptor by inducing neuronal release of glutamate and also enhances receptor-mediated responses by a direct interaction with the receptor complex. This study investigated the mechanism in cerebellar granule cells by which cyanide enhances NMDA-induced Ca2+ influx. Cyanide (50 microM) increased the influx of Ca2+ over the NMDA concentration range of 0.5-500 microM. Experiments showed that cyanide does not interact with the receptor's glycine or PKC mediated phosphorylation regulatory sites. N-ethylmaleimide, a thiol alkylating agent which inactivates the redox regulatory sites of the receptor, blocked the enhancing effect of cyanide. Pretreatment of cells with 5,5-dithio-bis-2-nitrobenzoic acid (DTNB), a compound that oxidizes the receptor redox sites, had no effect on the response to cyanide. On the other hand, the nonpermeant reducing agents, dithiothreitol or cysteine, further increased the cyanide effect. These observations can be explained by cyanide interacting with redox sensitive disulfide groups that are not accessible to the non-permeant reducing agents. It is proposed that cyanide interacts with a redox site(s) located either on the intracellular receptor domain or in the transmembrane hydrophobic domain. Furthermore the enhancement by cyanide of the excitotoxic actions of NMDA involves receptor sites that are sensitive to oxidation/reduction and this interaction contributes to the neurotoxic action of cyanide.


Assuntos
Cianetos/toxicidade , Neurônios/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Animais , Cálcio/metabolismo , Células Cultivadas , Cerebelo/citologia , Cerebelo/metabolismo , N-Metilaspartato/farmacologia , Neurônios/metabolismo , Oxirredução , Proteína Quinase C/antagonistas & inibidores , Ratos , Receptores de Glicina/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...