Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Sci Rep ; 9(1): 2516, 2019 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-30792526

RESUMO

Activated epidermal growth factor receptor (EGFR) has been proposed in the pathophysiology of neurodegenerative diseases. In the present study, the anti-inflammatory effect of afatinib, an EGFR-tyrosine kinase inhibitor (EGFR-TKIs) was investigated using CTX-TNA2 cells and primary cultured astrocytes subjected to oxygen/glucose deprivation (OGD). We found that OGD induced EGFR phosphorylation and activated subsequent signaling pathways, including phosphorylation of AKT and extracellular signal-regulated kinases (ERK). Afatinib blocked OGD-induced phosphorylation of EGFR, AKT and ERK. At the same time, afatinib attenuated OGD-induced elevations in glial fibrillary acidic protein (a biomarker of activated astrocytes) and proliferating cell nuclear antigen expression (a cell proliferating biomarker) as well as hypoxia-induced migratory ability. Furthermore, afatinib decreased OGD-induced increases in cyclooxygenase-II and inducible nitric oxide synthase expression of the treated astrocytes as well as NO content in the culture medium. Moreover, afatinib attenuated OGD-induced caspase 1 activation (a biomarker of inflammasome activation) and interleukin-1ß levels (a pro-inflammatory cytokine). Collectively, afatinib could block OGD-induced EGFR activation and its downstream signaling pathways in astrocytes. Moreover, afatinib attenuated OGD-induced astrocyte activation, proliferation and inflammasome activation. These data support the involvement of EGFR activation in neuroinflammation. Furthermore, EGFR-TKIs may be promising in inhibiting neuroinflammation in the CNS neurodegenerative diseases.


Assuntos
Afatinib/farmacologia , Genes erbB-1/genética , Inflamação/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Hipóxia Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Carência Cultural , Ciclo-Oxigenase 2/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Inflamação/genética , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Neuroglia/efeitos dos fármacos , Óxido Nítrico/genética , Óxido Nítrico Sintase Tipo II/genética , Oxigênio/metabolismo , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Inibidores de Proteínas Quinases/farmacologia , Ratos
3.
Mol Neurobiol ; 52(2): 1015-22, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26063592

RESUMO

In the present study, the neuroprotective effect of melatonin on arsenite-induced neurotoxicity was investigated in rat primary cultured cortical neurons. Incubation of melatonin prevented arsenite-induced neuronal cell loss in a concentration-dependent manner. Furthermore, melatonin significantly attenuated arsenite-induced elevation in microtubule-associated protein light chain 3 (LC3)-II levels, a biomarker of autophagy. Our fluorescent staining assay showed that melatonin decreased arsenite-induced elevation of co-localized fluorescent puncta of monodansylcadaverine (a specific marker of autophagic vacuoles) and lysotracker red (a specific marker of lysosomes), indicating that melatonin is capable of inhibiting arsenite-induced autophagy and autolysosome formation. Because 3-methyladenine (an autophagic inhibitor) attenuated the arsenite-reduced α-synuclein levels (a protein essential for the neurite outgrowth and synaptic plasticity), melatonin via inhibiting autophagy attenuated the arsenite-reduced α-synuclein levels. At the same time, melatonin ameliorated the arsenite-induced reduction in growth associated protein 43 (a hallmark protein of neurite outgrowth) and discontinuous neurites of rat primary cultured cortical neurons. In addition, melatonin was found to prevent arsenite-induced decreases in cytochrome c oxidase levels (a biomarker of mitochondrial mass) and elevation in co-localized fluorescent puncta of autolysosomes and cytochrome c oxidase. Moreover, melatonin prevented arsenite-induced reduction in peroxisome proliferator-activated receptor gamma co-activator 1 α, a transcriptional co-activator of mitochondrial biosynthesis. Taken together, melatonin may exert its neuroprotective action via inhibiting arsenite-induced autophagy and enhancing mitochondrial biogenesis and thus restoring α-synuclein levels, neuronal integrity, and mitochondrial mass in rat primary cultured cortical neurons.


Assuntos
Antioxidantes/uso terapêutico , Arsenitos/toxicidade , Autofagia/efeitos dos fármacos , Melatonina/uso terapêutico , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Compostos de Sódio/toxicidade , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Antioxidantes/farmacologia , Biomarcadores , Células Cultivadas , Córtex Cerebral/citologia , Relação Dose-Resposta a Droga , Complexo IV da Cadeia de Transporte de Elétrons/análise , Feminino , Lisossomos/efeitos dos fármacos , Melatonina/farmacologia , Mitocôndrias/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Fármacos Neuroprotetores/farmacologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Gravidez , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo , alfa-Sinucleína/metabolismo
4.
Mol Neurobiol ; 48(2): 281-7, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23824679

RESUMO

In the present study, the role of heme oxygenase (HO)-1 in sodium arsenite (arsenite)-induced neurotoxicity was investigated using primary cultured cortical neurons. Incubation with arsenite was found to cause cell death of primary cultured cortical neurons in concentration- and time-dependent manners. Furthermore, arsenite induced caspase 3 activation and decreased procaspase 12 levels, indicating that apoptosis is involved in the arsenite-induced neurotoxicity. The oxidative mechanism underlying arsenite-induced neurotoxicity was investigated. Western blot assay showed that arsenite significantly increased HO-1 levels, a redox-regulated protein. Co-incubation with glutathione (10 mM) attenuated arsenite-induced HO-1 elevation and caspase 3 activation, suggesting that oxidative stress is involved in the arsenite-induced neurotoxicity. The neurotoxic effects of inorganic arsenics were compared; arsenite was more potent than arsenate in inducing HO-1 expression and caspase 3 activation. Moreover, the cell viabilities of arsenite and arsenate were 60 ± 2 and 99 ± 2 % of control, respectively. HO-1 siRNA transfection was employed to prevent arsenite-induced HO-1 elevation. At the same time, arsenite-induced caspase 3 activation and neuronal death were attenuated in the HO-1 siRNA-transfected cells. Taken together, HO-1 appears to be neuroprotective in the arsenite-induced neurotoxicity in primary cultured cortical neurons. In addition to antioxidants, HO-1 elevation may be a neuroprotective strategy for arsenite-induced neurotoxicity.


Assuntos
Arsenitos/toxicidade , Córtex Cerebral/patologia , Heme Oxigenase-1/metabolismo , Neurônios/enzimologia , Neurônios/patologia , Neurotoxinas/toxicidade , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Feminino , Glutationa/farmacologia , Neurônios/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transfecção
5.
Neuropathol Appl Neurobiol ; 36(7): 612-22, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20626630

RESUMO

AIMS: Transition metals, oxidative stress and neuroinflammation have been proposed as part of a vicious cycle in central nervous system neurodegeneration. Our aim was to study the anti-inflammatory effect of pioglitazone, a peroxisome proliferative activated receptor-γ agonist, on iron-induced oxidative injury in rat brain. METHODS: Intranigral infusion of ferrous citrate (iron) was performed on anaesthetized rats. Pioglitazone (20 mg/kg) was orally administered. Oxidative injury was investigated by measuring lipid peroxidation in the substantia nigra (SN) and dopamine content in the striatum. Western blot assay and DNA fragmentation were employed to study the involvement of α-synuclein aggregation, neuroinflammation as well as activation of endoplasmic reticulum (ER) and mitochondrial pathways in iron-induced apoptosis. RESULTS: Intranigral infusion of iron time-dependently increased α-synuclein aggregation and haem oxygenase-1 levels. Furthermore, apoptosis was demonstrated by TUNEL-positive cells and DNA fragmentation in the iron-infused SN. Systemic pioglitazone was found to potentiate iron-induced elevation in nuclear peroxisome proliferative activated receptor-γ levels. However, pioglitazone inhibited iron-induced α-synuclein aggregation, elevations in interleukin-1ß and interleukin-6 mRNA levels as well as increases in oxygenase-1, cyclo-oxygenase II, nitric oxide synthase and ED-1 protein levels, an indicator of activated microglia. Moreover, iron-induced DNA laddering as well as activation of ER and mitochondrial pathways were attenuated by pioglitazone. In addition, pioglitazone decreased iron-induced elevation in lipid peroxidation in the infused SN and depletion in striatal dopamine level. CONCLUSIONS: Our results suggest that pioglitazone prevents iron-induced apoptosis via both ER and mitochondrial pathways. Furthermore, inhibition of α-synuclein aggregation and neuroinflammation may contribute to the pioglitazone-induced neuroprotection in central nervous system.


Assuntos
Anti-Inflamatórios , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopamina/fisiologia , Hipoglicemiantes/farmacologia , Ferro/antagonistas & inibidores , Ferro/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Tiazolidinedionas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Cromatografia Líquida de Alta Pressão , Fragmentação do DNA , Dopamina/metabolismo , Interleucina-1beta/biossíntese , Interleucina-6/biossíntese , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Proteínas do Tecido Nervoso/metabolismo , Fármacos Neuroprotetores , Pioglitazona , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , alfa-Sinucleína/metabolismo
6.
Free Radic Biol Med ; 49(5): 839-46, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20541008

RESUMO

A sublethal preconditioning has been proposed as a neuroprotective strategy against several CNS neurodegenerative diseases. In this study, the involvement of autophagy in the protection provided by hypoxic preconditioning against 1-methyl-4-phenylpyridinium (MPP(+))-induced neurotoxicity was studied in SH-SY5Y neuroblastoma cells. In contrast to the cytotoxicity of 0.1% oxygen, 1% oxygen hypoxia for 24h did not cause significant cell death. A transient increase in LC3-II level, a biomarker of autophagy, was demonstrated during hypoxic treatment. At the same time, 8-h hypoxia increased fluorescence due to monodansylcadaverine, a specific dye for autophagosomes, in the treated cells. Co-incubation with bafilomycin A1 (10 nM) further increased hypoxia-induced LC3-II levels but 3-methyladenine (3-MA; 10 mM) reduced the elevation in LC3-II levels induced by 8-h hypoxia. Moreover, 8-h hypoxia increased free radical formation and nuclear HIF-1alpha level. Glutathione was found to diminish hypoxia-induced LC3-II elevation. In contrast to the elevated LC3-II level, 8-h hypoxia significantly decreased mitochondrial mass. Furthermore, a rebound elevation in mitochondrial mass was observed under 8-h hypoxia and subsequent 12-h normoxia. Prior hypoxia attenuated the MPP(+)-induced elevation in LC3-II levels and cell death. Moreover, hypoxic pretreatment inhibited MPP(+)-induced activation of caspase-3 and DNA fragmentation. Co-incubation with 3-MA during hypoxia prevented the protection afforded by hypoxic preconditioning against MPP(+)-induced increases in LC3-II levels and neurotoxicity. Taken together, our results suggest that sublethal hypoxia induces autophagy that is mediated by oxidative stress. Furthermore, autophagy may be involved in the protection provided by hypoxic preconditioning against MPP(+)-induced neurotoxicity, indicating a neuroprotective role of autophagy in hypoxic preconditioning.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Autofagia/fisiologia , Citoproteção , Neurônios/efeitos dos fármacos , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Herbicidas/toxicidade , Humanos , Precondicionamento Isquêmico , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Neuroblastoma/patologia , Neurônios/fisiologia , Espécies Reativas de Oxigênio/metabolismo
7.
Hum Exp Toxicol ; 27(4): 327-39, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18684804

RESUMO

Siderosis bulbi is vision threatening. An investigation into its mechanisms and management is crucial. Experimental siderosis was established by intravitreous administration of an iron particle (chronic) or FeSO(4) (acute). After siderosis, there was a significant dose-responsive reduction in eletroretinogram (a/b-wave) amplitude, and an increase in OH level, greater when caused by 24 mM FeSO(4) than that by 8 mM FeSO(4). Furthermore, the FeSO(4)-induced oxidative stress was significantly blunted by 100 microM ferulic acid (FA). Siderosis also resulted in an excessive glutamate release, increased [Ca(++)](i), and enhanced superoxide dismutase immunoreactivity. The latter finding was consistent with the Western blot result. Obvious disorganization including loss of photoreceptor outer segments and cholinergic amacrines together with a wide-spreading ferric distribution across the retina was present, which were related to the eletro-retinographic and pathologic dysfunctions. Furthermore, b-wave reduction and amacrine damage were respectively, significantly, dose-dependently, and clearly ameliorated by FA. Thus, siderosis stimulates oxidative stress, and possibly, subsequent excitotoxicity, and calcium influx, which explains why the retina is impaired electro-physiologically and pathologically. Importantly, FA protects iron toxicity perhaps by acting as a free radical scavenger. This provides an approach to the study and treatment of the iron-related disorders such as retained intraocular iron and Alzheimer disease.


Assuntos
Ácidos Cumáricos/uso terapêutico , Compostos Ferrosos/toxicidade , Ferro/toxicidade , Retina/efeitos dos fármacos , Doenças Retinianas/prevenção & controle , Siderose/tratamento farmacológico , Doença Aguda , Animais , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Doença Crônica , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Antagonismo de Drogas , Eletrorretinografia/efeitos dos fármacos , Compostos Ferrosos/análise , Compostos Ferrosos/metabolismo , Glutamatos/metabolismo , Radical Hidroxila/metabolismo , Radical Hidroxila/toxicidade , Injeções , Ferro/análise , Ferro/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Retina/metabolismo , Retina/fisiopatologia , Doenças Retinianas/induzido quimicamente , Doenças Retinianas/fisiopatologia , Siderose/etiologia , Siderose/patologia , Superóxido Dismutase/metabolismo , Corpo Vítreo/química , Corpo Vítreo/efeitos dos fármacos , Corpo Vítreo/metabolismo
8.
Free Radic Biol Med ; 44(8): 1553-61, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18226606

RESUMO

Peripheral neuropathy is common in people chronically overexposed to arsenic. We studied sodium arsenite (arsenite)-induced cytotoxicity in dorsal root ganglion (DRG) explants. Incubation with arsenite concentration- and time-dependently increased the expression of stress proteins, heat shock protein 70, and heme oxygenase-1 in DRG explants. Furthermore, apoptosis was involved in the arsenite-induced cytotoxicity in the treated DRG. Elevation in cytosolic cytochrome c levels and reduction in procaspase 3 levels suggested an involvement of the mitochondrial pathway in arsenite-induced apoptosis in this preparation. At the same time, increases in the activating transcription factor-4 and C/EBP homologous protein and reduction in procaspase 12 levels indicated activation of the endoplasmic reticulum (ER) pathway in the arsenite-induced cytotoxicity in DRG explants. Salubrinal (30 microM), an ER inhibitor, was found to attenuate arsenite-induced DNA fragmentation and reduction in procaspase 12 in DRG explants. Cytotoxic effects by arsenite, sodium arsenate (arsenate), monomethylarsonic acid (MMA), and dimethylarsinic acid (DMA) were compared, and the potency was as follows: arsenite >>> arsenate>MMA and DMA. Recombinant adenovirus vectors encoding glial-cell-derived neurotrophic factor (AdGDNF) genes allowed a stable delivery of GDNF genes to the infected cells in DRG explants. Applied in this manner, AdGDNF was found to inhibit arsenite-induced DNA fragmentation in DRG explants. Moreover, AdGDNF attenuated the arsenite-induced reduction in procaspases 3 and 12 levels. Taken together, our study demonstrates that arsenite is capable of inducing cytotoxicity in DRG explants. Both ER and mitochondria pathways are involved in the arsenite-induced apoptosis in DRG explants. Glial-cell-derived neurotrophic factor appears to be protective against arsenite-induced peripheral neuropathy.


Assuntos
Apoptose/efeitos dos fármacos , Arsenitos/toxicidade , Gânglios Espinais/metabolismo , Compostos de Sódio/toxicidade , Animais , Fragmentação do DNA , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Gânglios Espinais/citologia , Terapia Genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Proteínas de Choque Térmico HSP70/biossíntese , Heme Oxigenase (Desciclizante)/biossíntese , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Exp Neurol ; 195(1): 40-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15950222

RESUMO

The neuroprotective effect of hypoxic preconditioning on kainate (KA)-induced neurotoxicity, including apoptosis and necrosis, was investigated in rat hippocampus. Female Wistar-Kyoto rats were subjected to 380 mm Hg in an altitude chamber for 15 h/day for 28 days. Intrahippocampal infusion of KA was performed in chloral hydrate anesthetized rats, which acutely elevated 2,3-dihydroxybenzoic acid levels in normoxic rats. Seven days after the infusion, KA increased lipid peroxidation in the infused hippocampus and resulted in hippocampal CA3 neuronal loss. A 4-week hypoxic preconditioning attenuated KA-induced elevation in hydroxyl radical formation and lipid peroxidation as well as KA-induced neuronal loss. The effects of hypoxic preconditioning on KA-induced apoptosis and necrosis were investigated further. Two hours after KA infusion, cytosolic cytochrome c content was increased in the infused hippocampus. Twenty-four hours after KA infusion, pyknotic nuclei, cellular shrinkage, and cytoplasmic disintegration, but not TUNEL-positive staining, were observed in the CA3 region of hippocampus. Forty-eight hours after KA infusion, both DNA smear and DNA fragmentation were demonstrated in the infused hippocampus. Furthermore, TUNEL-positive cells, indicative of apoptosis, in the infused hippocampus were detected 72 h after KA infusion. Hypoxic pretreatment significantly reduced necrotic-like events in the KA-infused hippocampus. Moreover, hypoxic preconditioning attenuated apoptosis induced by KA infusion, including elevation in cytosolic cytochrome c content, TUNEL-positive cells, and DNA fragmentation. Our data suggest that hypoxic preconditioning may exert its neuroprotection of KA-induced oxidative injuries via attenuating both apoptosis and necrosis in rat hippocampus.


Assuntos
Agonistas de Aminoácidos Excitatórios/toxicidade , Hipocampo/efeitos dos fármacos , Precondicionamento Isquêmico/métodos , Ácido Caínico/toxicidade , Oxigênio/metabolismo , Animais , Southern Blotting/métodos , Western Blotting/métodos , Contagem de Células/métodos , Cromatografia Líquida de Alta Pressão/métodos , Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Eletroquímica/métodos , Feminino , Hipocampo/metabolismo , Hipocampo/patologia , Hidroxibenzoatos/análise , Marcação In Situ das Extremidades Cortadas/métodos , Peroxidação de Lipídeos/efeitos dos fármacos , Microdiálise/métodos , Neurônios/patologia , Ratos , Ratos Endogâmicos WKY , Coloração e Rotulagem/métodos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...