Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Phys Chem A ; 127(34): 7148-7155, 2023 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-37595363

RESUMO

In understanding the mechanism of aggregation-induced emission (AIE), the multilevel ONIOM framework has been demonstrated as one of the efficient tools that can capture the essential mechanistic information by choosing a single fluorophore as the quantum mechanics (QM) model and putting all surrounding molecules in the low-level region. Recently, the ionic styryl-pyridine salt (namely, SPH) has been reported as a new class of AIEgen with a high fluorescence yield. In the SPH crystal, a pair of ionic SPH molecules are closely stacked with each other in an antiparallel, head-to-tail pattern, thus the choice of QM models (an individual or dimeric structure) becomes critical in the ONIOM study. Herein we report the AIE mechanism of the ionic SPH at the QM ((TD)-CAM-B3LYP) and ONIOM(QM:MM) levels. As usual, the fluorescence quenching of SPH in tetrahydrofuran (THF) solution is attributed to a nonradiative relaxation via the central C═C bond rotation, with a rather low barrier of 2.7 kcal/mol. In crystals, either with a monomer or dimer model, the fluorescence quenching channel is found to be restricted due to the obvious C═C rotation barriers. Compared with the monomer model, the dimer model, by treating the orbital interaction of the two SPH molecules at the QM level, provides significantly increased barriers and a red-shifted emission wavelength that better matches the experimental value. In addition, the calculated exciton coupling in the fluorescence emission state can be discovered only by a dimer model. The findings here emphasize not only the importance of choosing a proper model in the ONIOM study of AIE but also expanding our understanding of novel AIE systems.

2.
Neurochem Res ; 43(12): 2446-2459, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30382449

RESUMO

Perfluorooctanesulfonate (PFOS) may cause neurotoxicity through the initiation of oxidative stress. In the current study, we investigated the role of anti-oxidant nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in PFOS-induced neurotoxicity. We found that human neuroblastoma SH-SY5Y cells exhibited significant apoptotic cell death following PFOS exposure, and this process was accompanied with apparent accumulation of reactive oxidative species (ROS). In addition, we revealed that PFOS exposure caused marked activation of Nrf2 pathway and the expression of Nrf2 transcription target heme oxygenase-1. We further found that pre-treatment with ROS scavenger N-acetyl-L-cysteine (NAC) dramatically ameliorated PFOS-induced ROS production and Nrf2 signaling. In keeping with these findings, western blot and Cell Counter Kit-8 analyses revealed that pre-incubation with NAC suppressed PFOS-induced expression of pro-apoptotic proteins and impairment of neuronal viability. Moreover, antagonizing Nrf2 pathway with Nrf2 inhibitor brusatol resulted in increased ROS production and enhanced PFOS-induced expression of apoptosis related proteins. Finally, we showed that PFOS exposure altered mitochondrial transmembrane potential and disrupted normal mitochondrial morphology in SH-SY5Y cells. Whereas treatment with NAC ameliorated PFOS-induced mitochondrial disorders, co-incubation with brusatol augmented PFOS-induced mitochondrial deficits, consequently contributing to neuronal apoptosis. These results manifest that Nrf2 pathway plays a protective role in PFOS-induced neurotoxicity, providing new insights into the prevention and treatment of PFOS-related toxicities.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Apoptose/fisiologia , Fluorocarbonos/toxicidade , Fator 2 Relacionado a NF-E2/metabolismo , Neuroproteção/fisiologia , Estresse Oxidativo/fisiologia , Transdução de Sinais/fisiologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Neuroproteção/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
3.
Toxicol Appl Pharmacol ; 303: 79-89, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27174766

RESUMO

Arsenic is a widely distributed toxic metalloid all over the world. Inorganic arsenic species are supposed to affect astrocytic functions and to cause neuron apoptosis in CNS. Microglias are the key cell type involved in innate immune responses in CNS, and microglia activation has been linked to inflammation and neurotoxicity. In this study, using ELISA, we showed that Arsenic trioxide up-regulated the expression and secretion of IL-1ß in a dose-dependent manner and a time-dependent manner in cultured HAPI microglia cells. The secretion of IL-1ß caused the apoptosis of SH-SY5Y. These pro-inflammatory responses were inhibited by the STAT3 blocker, AG490 and P38/JNK MAPK blockers SB202190, SP600125. Further, Arsenic trioxide exposure could induce phosphorylation and activation of STAT3, and the translocation of STAT3 from the cytosol to the nucleus in this HAPI microglia cell line. Thus, the STAT3 signaling pathway can be activated after Arsenic trioxide treatment. However, P38/JNK MAPK blockers SB202190, SP600125 also obviously attenuated STAT3 activation and transnuclear transport induced by Arsenic trioxide. In concert with these results, we highlighted that the secretion of IL-1ß and STAT3 activation induced by Arsenic trioxide can be mediated by elevation of P38/JNK MAPK in HAPI microglia cells and then induced the toxicity of neurons.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Óxidos/toxicidade , Fator de Transcrição STAT3/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antracenos/farmacologia , Apoptose/efeitos dos fármacos , Trióxido de Arsênio , Arsenicais , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Imidazóis/farmacologia , Inflamação , Interleucina-1beta/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Microglia/metabolismo , Neurônios/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
4.
Regul Toxicol Pharmacol ; 81: 480-488, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27664319

RESUMO

Arsenic is a widely distributed toxic metalloid in around the world. Inorganic arsenic species are deemed to affect astrocytes functions and to cause neuron apoptosis. Microglia are the key cell type involved in innate immune responses in CNS, and microglia activation has been linked to inflammation and neurotoxicity. In this study, using ELISA and reverse transcriptase PCR (RT-PCR), we showed that Arsenic trioxide up-regulated the expression and secretion of IL-6 in a dose-dependent manner and a time-dependent manner in cultured HAPI microglia cells. These pro-inflammatory responses were inhibited by the Akt blocker, LY294002. Further, Arsenic trioxide exposure could induce phospho rylationand degradation of IкBα, and the translocation of NF-κB p65 from the cytosol to the nucleus in this HAPI microglia cell line. Thus, the NF-кB signaling pathway can be activated after Arsenic trioxide treatment. Besides, Akt blocker LY294002 also obviously attenuated NF-кB activation and transnuclear induced by Arsenic trioxide. In concert with these results, we highlighted that the secretion of pro-inflammatory cytokine and NF-кB activation induced by Arsenic trioxide can be mediated by elevation of p-Akt in HAPI microglia cells.


Assuntos
Arsênio/toxicidade , Inflamação/metabolismo , Interleucina-6/metabolismo , Microglia/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Células Cultivadas , Inflamação/imunologia , Inflamação/patologia , Interleucina-6/imunologia , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , Ratos , Transdução de Sinais/efeitos dos fármacos
5.
J Appl Toxicol ; 36(12): 1591-1598, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27018151

RESUMO

The neurotoxic effects of perfluorooctane sulfonate (PFOS) have attracted significant research attention in recent years. In the present study, we investigated the impact of PFOS exposure on the physiology of neural stem cells (NSCs) in vitro. We showed that PFOS exposure markedly attenuated the proliferation of C17.2 neural stem cells in both dose- and time-dependent manners. Additionally, we found that PFOS decreased Ser9 phosphorylation of glycogen synthase kinase-3ß (pSer9-GSK-3ß), leading to the activation of GSK-3ß and resultant downregulation of cellular ß-catenin. Furthermore, blockage of GSK-3ß with lithium chloride significantly attenuated both the PFOS-induced downregulation of GSK-3ß/ß-catenin and the proliferative impairment of C17.2 cells. Notably, the expression of various downstream targets was altered accordingly, such as c-myc, cyclin D1 and survivin. In conclusion, the present study demonstrated that PFOS decreased the proliferation of C17.2 cells via the negative modulation of the GSK-3ß/ß-catenin pathway. We present the potential mechanisms underlying the PFOS-induced toxic effects on NSCs to provide novel insights into the neurotoxic mechanism of PFOS. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , Glicogênio Sintase Quinase 3 beta/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , beta Catenina/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Glicogênio Sintase Quinase 3 beta/genética , Camundongos , Células-Tronco Neurais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , beta Catenina/genética
6.
J Appl Toxicol ; 36(11): 1409-17, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26988466

RESUMO

The widespread environmental contaminant, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), is considered one of the most toxic dioxin-like compounds. Although epidemiological studies have shown that TCDD exposure is linked to some neurological and neurophysiological disorders, the underlying mechanism of TCDD-mediated neurotoxicity has remained unclear. Astrocytes are the most abundant cells in the nervous systems, and are recognized as the important mediators of normal brain functions as well as neurological, neurodevelopmental and neurodegenerative brain diseases. In this study, we investigated the role of TCDD in regulating the expression of glutamate transporter GLT-1 in astrocytes. TCDD, at concentrations of 0.1-100 nm, had no significantly harmful effect on the viability of C6 glioma cells. However, the expression of GLT-1 in C6 glioma cells was downregulated in a dose- and time-dependent manner. TCDD also caused activation of protein kinase C (PKC), as TCDD induced translocation of the PKC from the cytoplasm or perinuclear to the membrane. The translocation of PKC was inhibited by one Ca(2+) blocker, nifedipine, suggesting that the effects are triggered by the initial elevated intracellular concentration of free Ca(2+) . Finally, we showed that inhibition of the PKC activity reverses the TCDD-triggered reduction of GLT-1. In summary, our results suggested that TCDD exposure could downregulate the expression of GLT-1 in C6 via Ca(2+) /PKC pathway. The downregulation of GLT-1 might participate in TCDD-mediated neurotoxicity. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Transportador 2 de Aminoácido Excitatório/biossíntese , Dibenzodioxinas Policloradas/toxicidade , Proteína Quinase C/metabolismo , Animais , Astrócitos/metabolismo , Sinalização do Cálcio , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo , Ratos , Transdução de Sinais , Fatores de Tempo
7.
Environ Toxicol ; 31(9): 1068-79, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25865073

RESUMO

The environmental toxicant TCDD may elicit cytotoxic effects by inducing reactive oxygen species (ROS) generation. Autophagy is one of the first lines of defense against oxidative stress damage. Herein, we investigated whether autophagy played a regulatory role in TCDD-induced neurotoxicity. Here, we showed that TCDD exposure caused marked autophagy in SH-SY5Y cells, whose dose range was close to that inducing apoptosis. Electron microscopic and Western blot analyses revealed that TCDD induced autophagy at a starting dose of approximate 100 nM. Interestingly, 100-200 nM TCDD exposure resulted in obviously decreased cell viability and evident apoptotic phenotype. Furthermore, the levels of pro-apoptotic molecules, Bax and cleaved-PARP, increased significantly, whereas Bcl2 declined after exposed to 100 nM TCDD. In addition, the apoptosis was verified using flow cytometrical analysis. These data strongly suggested that TCDD induced both autophagy and apoptosis at a similar dose range in SH-SY5Y cells. Interestingly, pretreatment with ROS scavenger, N-acetyl-cysteine (NAC), could effectively block both TCDD-induced apoptosis and autophagy. More surprisingly, inhibition of autophagy with 3-methyladenine (3MA), remarkably augmented TCDD-induced apoptosis. The findings implicated that the onset of autophagy might serve as a protective mechanism to ameliorate ROS-triggered cytotoxic effects in human SH-SY5Y neuronal cells under TCDD exposure. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1068-1079, 2016.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Substâncias Protetoras/farmacologia , Acetilcisteína/farmacologia , Adenina/análogos & derivados , Adenina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo
8.
Biochem Biophys Res Commun ; 463(1-2): 116-22, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26002461

RESUMO

Elevated free fatty acids (FFAs) are fundamental to the pathogenesis of hepatic insulin resistance. However, the molecular mechanisms of insulin resistance remain not completely understood. Transcriptional dysregulation, post-transcriptional modifications and protein degradation contribute to the pathogenesis of insulin resistance. Poly(C) binding proteins (PCBPs) are RNA-binding proteins that are involved in post-transcriptional control pathways. However, there are little studies about the roles of PCBPs in insulin resistance. PCBP2 is the member of the RNA-binding proteins and is thought to participate in regulating hypoxia inducible factor-1 (HIF-1α) and signal transducers and activators of transcription (STAT) pathway which are involved in regulating insulin signaling pathway. Here, we investigated the influence of PCBP2 on hepatic insulin resistance. We showed that the protein and mRNA levels of PCBP2 were down-regulated under insulin-resistant conditions. In addition, we showed that over-expression of PCBP2 ameliorates palmitate (PA)-induced insulin resistance, which was indicated by elevated phosphorylation of protein kinase B (AKT) and glycogen synthase kinase 3ß (GSK3ß). We also found that over-expression of PCBP2 inhibits HIF1α and STAT3 pathway. Furthermore, glucose uptake was found to display a similar tendency with the phosphorylation of Akt. The expressions of phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-phosphatase (G6Pase), two key gluconeogenic enzymes, were down-regulated following Over-expression of PCBP2. Accordingly, PA-induced intracellular lipid accumulation was suppressed in over-expression of PCBP2 HepG2 cells. In addition, we found that over-expression of PCBP2 inhibits HIF1α and STAT3 pathway. Our results demonstrate that PCBP2 was involved in hepatic insulin sensitivity might via HIF-1α and STAT3 pathway in HepG2 cells.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Resistência à Insulina/fisiologia , Proteínas de Ligação a RNA/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Regulação para Baixo , Gluconeogênese/efeitos dos fármacos , Glucose/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Células Hep G2 , Humanos , Resistência à Insulina/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Ácido Palmítico/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Transdução de Sinais
9.
Biochem Biophys Res Commun ; 457(4): 578-84, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25600807

RESUMO

Excess serum free fatty acids (FFAs) are fundamental to the pathogenesis of insulin resistance. Chronic endoplasmic reticulum (ER) stress is a major contributor to obesity-induced insulin resistance in the liver. With high-fat feeding (HFD), FFAs can activate chronic endoplasmic reticulum (ER) stress in target tissues, initiating negative crosstalk between FFAs and insulin signaling. However, the molecular link between insulin resistance and ER stress remains to be identified. We here reported that translocating chain-associated membrane protein 1 (TRAM1), an ER-resident membrane protein, was involved in the onset of insulin resistance in hepatocytes. TRAM1 was significantly up-regulated in insulin-resistant liver tissues and palmitate (PA)-treated HepG2 cells. In addition, we showed that depletion of TRAM1 led to hyperactivation of CHOP and GRP78, and the activation of downstream JNK pathway. Given the fact that the activation of ER stress played a facilitating role in insulin resistance, the phosphorylation of Akt and GSK-3ß was also analyzed. We found that depletion of TRAM1 markedly attenuated the phosphorylation of Akt and GSK-3ß in the cells. Moreover, application with JNK inhibitor SP600125 reversed the effect of TRAM1 interference on Akt phosphorylation. The accumulation of lipid droplets and expression of two key gluconeogenic enzymes, PEPCK and G6Pase, were also determined and found to display a similar tendency with the phosphorylation of Akt. Glucose uptake assay indicated that knocking down TRAM1 augmented PA-induced down-regulation of glucose uptake, and inhibition of JNK using SP600125 could block the effect of TRAM1 on glucose uptake. These data implicated that TRAM1 might protect HepG2 cells against PA-induced insulin resistance through alleviating ER stress.


Assuntos
Estresse do Retículo Endoplasmático , Células Hep G2/metabolismo , Resistência à Insulina , Sistema de Sinalização das MAP Quinases , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Palmitatos/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Chaperona BiP do Retículo Endoplasmático , Glucose/metabolismo , Humanos
10.
Toxicol Appl Pharmacol ; 288(2): 143-51, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26086160

RESUMO

Perfluorooctane sulfonate (PFOS), an emerging persistent contaminant that is commonly encountered during daily life, has been shown to exert toxic effects on the central nervous system (CNS). However, the molecular mechanisms underlying the neurotoxicity of PFOS remain largely unknown. It has been widely acknowledged that the inflammatory mediators released by hyper-activated microglia play vital roles in the pathogenesis of various neurological diseases. In the present study, we examined the impact of PFOS exposure on microglial activation and the release of proinflammatory mediators, including nitric oxide (NO) and reactive oxidative species (ROS). We found that PFOS exposure led to concentration-dependent NO and ROS production by rat HAPI microglia. We also discovered that there was rapid activation of the ERK/JNK MAPK signaling pathway in the HAPI microglia following PFOS treatment. Moreover, the PFOS-induced iNOS expression and NO production were attenuated after the inhibition of ERK or JNK MAPK by their corresponding inhibitors, PD98059 and SP600125. Interestingly, NAC, a ROS inhibitor, blocked iNOS expression, NO production, and activation of ERK and JNK MAPKs, which suggested that PFOS-mediated microglial NO production occurs via a ROS/ERK/JNK MAPK signaling pathway. Finally, by exposing SH-SY5Y cells to PFOS-treated microglia-conditioned medium, we demonstrated that NO was responsible for PFOS-mediated neuronal apoptosis.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Poluentes Ambientais/toxicidade , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fluorocarbonos/toxicidade , Mediadores da Inflamação/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Microglia/efeitos dos fármacos , Óxido Nítrico/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Microglia/enzimologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos , Fatores de Tempo
11.
Cell Mol Neurobiol ; 35(4): 463-71, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25407628

RESUMO

RNA-binding motif protein, X-linked (RBMX) is a 43 kDa nuclear protein in the RBM family and functions on alternative splicing of RNA. The gene encoding RBMX is located on chromosome Xq26. To investigate whether RBMX is involved in retinal neuron apoptosis, we performed a light-induced retinal damage model in adult rats. Western blotting analysis showed RBMX gradually increased, reached a peak at 12 h and then declined during the following days. The association of RBMX in retinal ganglion cells (RGCs) with light exposure was found by immunofluorescence staining. The injury-induced expression of RBMX was detected in active caspase-3 and TUNEL positive cells. We also examined the expression profiles of active caspase-3, bcl-2 and Bax, whose changes were correlated with the expression profiles of RBMX. To summarize, we uncovered the dynamic changes of RBMX in the light-induced retinal damage model for the first time. RBMX might play a significant role in the degenerative process of RGCs after light-induced damage in the retina.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Luz , Retina/metabolismo , Retina/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Feminino , Imunofluorescência , Marcação In Situ das Extremidades Cortadas , Masculino , Transporte Proteico/efeitos da radiação , Ratos Sprague-Dawley , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/efeitos da radiação , Coloração e Rotulagem , Proteína X Associada a bcl-2/metabolismo
12.
Neurochem Res ; 40(6): 1220-31, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25998883

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been recently shown to elicit inflammatory response in a number of cell-types. However, whether TCDD could provoke inflammation in astrocytes, the most abundant glial cells in central nervous system (CNS), remains virtually unknown. In the present study, we showed that TCDD exposure could induce evident astrocyte activation both in vivo and in vitro. Further, we found that TGF-ß-activated kinase 1 (TAK1), a critical regulator of NF-κB signaling, was rapidly phosphorylated in the process of TCDD-induced reactive astroglia. Exposure to TCDD led to rapid TAK1 and NF-κB p65 phosphorylation, as well as IKBα degradation. Moreover, blockage of TAK1 using siRNA oligos or TAK1 inhibitor 5Z-7-oxozeaenol significantly attenuated TCDD-induced astrocyte activation as well as the release of TNF-α. Finally, we showed that the conditioned medium of TCDD-treated astrocytes promoted the apoptosis of PC12 neuronal cells, which could be blocked with the pre-treatment of TAK1 inhibitor. Taken together, these findings suggested that TCDD could promote the inflammatory activation of astrocytes through modulating TAK1-NF-κB cascade, implicating that reactive astrocytes might contribute to TCDD-induced adverse effects on CNS system.


Assuntos
Astrócitos/efeitos dos fármacos , Poluentes Ambientais/toxicidade , MAP Quinase Quinase Quinases/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados , Feminino , Proteínas I-kappa B/efeitos dos fármacos , Proteínas I-kappa B/metabolismo , MAP Quinase Quinase Quinases/antagonistas & inibidores , Células PC12 , Fosforilação , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Fator de Transcrição RelA/metabolismo
13.
J Appl Toxicol ; 35(7): 851-60, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25382668

RESUMO

2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) is a ubiquitous environmental contaminant that could exert significant neurotoxicity in the human nervous system. Nevertheless, the molecular mechanism underlying TCDD-mediated neurotoxicity has not been clarified clearly. Herein, we investigated the potential role of TCDD in facilitating premature senescence in astrocytes and the underlying molecular mechanisms. Using the senescence-associated ß-galactosidase (SA-ß-Gal) assay, we demonstrated that TCDD exposure triggered significant premature senescence of astrocyte cells, which was accompanied by a marked activation of the Wingless and int (WNT)/ß-catenin signaling pathway. In addition, TCDD altered the expression of senescence marker proteins, such as p16, p21 and GFAP, which together have been reported to be upregulated in aging astrocytes, in both dose- and time-dependent manners. Further, TCDD led to cell-cycle arrest, F-actin reorganization and the accumulation of cellular reactive oxygen species (ROS). Moreover, the ROS scavenger N-acetylcysteine (NAC) markedly attenuated TCDD-induced ROS production, cellular oxidative damage and astrocyte senescence. Notably, the application of XAV939, an inhibitor of WNT/ß-catenin signaling pathway, ameliorated the effect of TCDD on cellular ß-catenin level, ROS production, cellular oxidative damage and premature senescence in astrocytes. In summary, our findings indicated that TCDD might induce astrocyte senescence via WNT/ß-catenin and ROS-dependent mechanisms.


Assuntos
Astrócitos/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Dioxinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Western Blotting , Ciclo Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Dioxinas/toxicidade , Imunofluorescência , Ratos , Ratos Sprague-Dawley
14.
J Neurochem ; 129(5): 839-49, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24673440

RESUMO

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a ubiquitous environmental pollutant that could induce significant toxic effects in the human nervous system. However, the underlying molecular mechanism has not been entirely elucidated. Reactive astrogliosis has implicated in various neurological diseases via the production of a variety of pro-inflammatory mediators. Herein, we investigated the potential role of TCDD in facilitating astrocyte activation and the underlying molecular mechanisms. We showed that TCDD induced rapid astrocyte activation following TCDD exposure, which was accompanied by significantly elevated expression of Src-Suppressed-C Kinase Substrate (SSeCKS), a protein involved in protein kinase C (PKC)-mediated Nuclear Factor kappa B signaling, suggesting a possible involvement of PKC-induced SSeCKS activation in TCDD-triggered reactive astroglia. In keeping with the finding, we found that the level of phosphorylated Nuclear Factor kappa B p65 was remarkably increased after TCDD treatment. Furthermore, interference of SSeCKS attenuated TCDD-induced inducible nitric oxide synthase, glial fibrillary acidic protein, phospho-p65 expression, and tumor necrosis factor-α secretion in astrocytes. In addition, pre-treatment with PKC inhibitor also attenuated TCDD-induced astrocyte activation, as well as SSeCKS expression. Interestingly, we found that TCDD treatment could lead to SSeCKS perinuclear localization, which could be abolished after treatment with PKC inhibitor. Finally, we showed that inhibition of PKC activity or SSeCKS expression would impair TCDD-triggered tumor necrosis factor-α secretion. Our results suggested that TCDD exposure could lead to astrocyte activation through PKC/SSeCKS-dependent mechanisms, highlighting that astrocytes might be important target of TCDD-induced neurotoxicity. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) elicits neurotoxic effects. Here, we show TCDD induces pro-inflammatory responses in astrocytes. TCDD initiates an increase of [Ca2+]i, followed by the activation of PKC, which then induces the activation of Src-suppressed C-kinase substrate (SSeCKS). SSeCKS promotes NF-κB activation and the secretion of TNF-α and nitric oxide in astrocytes.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Astrócitos/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Poluentes Ambientais/toxicidade , Dibenzodioxinas Policloradas/toxicidade , Proteína Quinase C/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Núcleo Celular/metabolismo , Citocinas/metabolismo , Citoplasma/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Imuno-Histoquímica , Inflamação/patologia , NF-kappa B/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Transfecção
15.
J Neurosci Res ; 92(6): 783-94, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24464479

RESUMO

Manganese (Mn) is an essential trace element. However, exposure to excessive Mn may cause neurodegenerative disorders called manganism. Accumulating evidence indicated that dysregulation of Wnt/ß-catenin signaling was tightly associated with the onset of neurodegenerative disorders. However, whether aberrant Wnt/ß-catenin signaling contributes to Mn-induced neurotoxicity remains unknown. The present study investigates the involvement of Wnt/ß-catenin signaling in Mn-induced neurotoxicity. Western blot and immunohistochemistry analyses showed a remarkable downregulation of p-Ser9-glycogen synthase kinase-3ß (GSK-3ß) and ß-catenin in rat striatum after Mn exposure. TUNEL assay revealed significant neuronal apoptosis following treatment with 25 mg/kg Mn. Immunofluorescent staining showed that ß-catenin was expressed predominantly in neurons, and colocalization of ß-catenin and active caspase-3 was observed after Mn exposure. Furthermore, Mn exposure resulted in PC12 cells apoptosis, which was accompanied by reduced levels of cellular ß-catenin and p-GSK-3ß. Accordingly, the mRNA level of the prosurvival factor survivin, a downstream target gene of ß-catenin, was synchronously decreased. More importantly, blockage of GSK-3ß activity with the GSK-3ß inhibitor lithium chloride could attenuate Mn-induced downregulation of ß-catenin and survivin as well as neuronal apoptosis. Overall, the present study demonstrates that downregulation of Wnt/ß-catenin signaling pathway may be of vital importance in the neuropathological process of Mn-induced neurotoxicity.


Assuntos
Corpo Estriado/metabolismo , Manganês/toxicidade , Neurônios/metabolismo , Síndromes Neurotóxicas/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Western Blotting , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Regulação para Baixo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Degeneração Neural/induzido quimicamente , Degeneração Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Células PC12 , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Via de Sinalização Wnt/fisiologia
16.
Toxicol Appl Pharmacol ; 281(3): 294-302, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25448048

RESUMO

Chronic exposure to excessive manganese (Mn) has been known to lead to neuronal loss and a clinical syndrome resembling idiopathic Parkinson's disease (IPD). p53 plays an integral role in the development of various human diseases, including neurodegenerative disorders. However, the role of p53 in Mn-induced neuronal apoptosis and neurological deficits remains obscure. In the present study, we showed that p53 was critically involved in Mn-induced neuronal apoptosis in rat striatum through both transcription-dependent and -independent mechanisms. Western blot and immunohistochemistrical analyses revealed that p53 was remarkably upregulated in the striatum of rats following Mn exposure. Coincidentally, increased level of cleaved PARP, a hallmark of apoptosis, was observed. Furthermore, using nerve growth factor (NGF)-differentiated PC12 cells as a neuronal cell model, we showed that Mn exposure decreased cell viability and induced apparent apoptosis. Importantly, p53 was progressively upregulated, and accumulated in both the nucleus and the cytoplasm. The cytoplasmic p53 had a remarkable distribution in mitochondria, suggesting an involvement of p53 mitochondrial translocation in Mn-induced neuronal apoptosis. In addition, Mn-induced impairment of mitochondrial membrane potential (ΔΨm) could be partially rescued by pretreatment with inhibitors of p53 transcriptional activity and p53 mitochondrial translocation, Pifithrin-α (PFT-α) and Pifithrin-µ (PFT-µ), respectively. Moreover, blockage of p53 activities with PFT-α and PFT-µ significantly attenuated Mn-induced reactive oxidative stress (ROS) generation and mitochondrial H2O2 production. Finally, we observed that pretreatment with PFT-α and PFT-µ ameliorated Mn-induced apoptosis in PC12 cells. Collectively, these findings implicate that p53 transcription-dependent and -independent pathways may play crucial roles in the regulation of Mn-induced neuronal death.


Assuntos
Apoptose/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Intoxicação por Manganês/metabolismo , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Antídotos/farmacologia , Antídotos/uso terapêutico , Benzotiazóis/farmacologia , Benzotiazóis/uso terapêutico , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Citoplasma/patologia , Masculino , Manganês/química , Manganês/toxicidade , Intoxicação por Manganês/tratamento farmacológico , Intoxicação por Manganês/patologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Células PC12 , Transporte Proteico/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Tolueno/análogos & derivados , Tolueno/farmacologia , Tolueno/uso terapêutico , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética
17.
Cell Mol Neurobiol ; 34(7): 1023-36, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24962097

RESUMO

SYF2 is a putative homolog of human p29 in Saccharomyces cerevisiae. It seems to be involved in pre-mRNA splicing and cell cycle progression. Disruption of SYF2 leads to reduced α-tubulin expression and delayed nerve system development in zebrafish. Due to the potential of SYF2 in modulating microtubule dynamics in nervous system, we investigated the spatiotemporal expression of SYF2 in a rat sciatic nerve crush (SNC) model. We found that SNC resulted in a significant upregulation of SYF2 from 3 days to 1 week and subsequently returned to the normal level at 4 weeks. At its peak expression, SYF2 distributed predominantly in Schwann cells. In addition, upregulation of SYF2 was approximately in parallel with Oct-6, and numerous Schwann cells expressing SYF2 were Oct-6 positive. In vitro, we observed enhanced expression of SYF2 during the process of cyclic adenosine monophosphate (cAMP)-induced Schwann cell differentiation. SYF2-specific siRNA-transfected Schwann cells did not show significant morphological change in the process of Schwann cell differentiation. Also, we found shorter and disorganized microtubule structure and a decreased migration in SYF2-specific siRNA-transfected Schwann cells. Together, these findings indicated that the upregulation of SYF2 was associated with Schwann cell differentiation and migration following sciatic nerve crush.


Assuntos
Diferenciação Celular , Movimento Celular , Compressão Nervosa , Proteínas Nucleares/metabolismo , Células de Schwann/patologia , Nervo Isquiático/patologia , Regulação para Cima , Animais , Biomarcadores/metabolismo , Western Blotting , Antígeno CD11b/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , AMP Cíclico/farmacologia , Imuno-Histoquímica , Masculino , Modelos Biológicos , Fator 6 de Transcrição de Octâmero/metabolismo , Fenótipo , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Tubulina (Proteína)/metabolismo , Regulação para Cima/efeitos dos fármacos
18.
Neurochem Res ; 39(11): 2105-17, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25103231

RESUMO

Histone deacetylase 4 (HDAC4), a member of the class IIa HDACs subfamily, has emerged as a critical regulator of cell growth, differentiation, and migration in various cell types. It was reported that HDAC4 stimulated colon cell proliferation via repression of p21. Also, HDAC4 contributes to platelet-derived growth factor-BB-induced proliferation and migration of vascular smooth muscle cells. Furthermore, HDAC4 may play an important role in the regulation of neuronal differentiation and survival. However, the role of HDAC4 in the process of peripheral nervous system regeneration after injury remains virtually unknown. Herein, we investigated the spatiotemporal expression of HDAC4 in a rat sciatic nerve crush model. We found that sciatic nerve crush induced up-regulated expression of HDAC4 in Schwann cells. Moreover, the expression of the proliferation marker Ki-67 exhibited a similar tendency with that of HDAC4. In cell cultures, we observed increased expression of HDAC4 during the process of TNF-α-induced Schwann cell proliferation, whereas the protein level of p21 was down-regulated. Interference of HDAC4 led to enhanced expression of p21 and impaired proliferation of Schwan cells. Taken together, our findings implicated that HDAC4 was up-regulated in the sciatic nerve after crush, which was associated with proliferation of Schwann cells.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Histona Desacetilases/metabolismo , Células de Schwann/citologia , Células de Schwann/metabolismo , Nervo Isquiático/lesões , Animais , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Compressão Nervosa/métodos , Regeneração Nervosa/fisiologia , Neurogênese/fisiologia , Ratos Sprague-Dawley , Regulação para Cima
19.
Neurotoxicology ; 66: 32-42, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29526747

RESUMO

Perfluorooctanesulfonate (PFOS)-containing compounds are widely used in all aspects of industrial and consumer products. Recent studies indicated that PFOS is ubiquitous in environments and is considered to be a new type of persistent organic pollutant (POP). This has raised concerns regarding its adverse effects on human health. The nervous system is regarded as a sensitive target of environmental contaminants, including PFOS. Previous findings showed that PFOS can induce neurobehavioral deficits. However, the molecular mechanism underlying PFOS neurotoxicity remains obscure. Astrocyte activation and the resulting pro-inflammatory cytokine release play an integral role in protecting neurons from neurotoxin-mediated damage. If uncontrolled, sustained astrocyte activation may cause the secretion of excessive levels of pro-inflammatory cytokines that exacerbate the initial damage. In this study, we showed that PFOS could promote excessive secretion of tumor necrosis factor-α (TNF-α) in dose- and time-dependent manners in astrocytes. Furthermore, PFOS exposure could induce the phosphorylation of Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3). This suggests that the JAK2/STAT3 signal transduction pathway is involved in PFOS-mediated astrocyte activation and secretion of TNF-α. Indeed, targeted blockage of the JAK2/STAT3 pathway prevented the phosphorylation of JAK and STAT3, and it also caused abnormal expression of TNF-α. Finally, we demonstrated that SH-SY5Y neuronal cells underwent rapid apoptosis via a TNF-α-dependent mechanism after exposure to PFOS-treated astrocyte-conditioned medium. In summary, our findings reveal that PFOS mediates a rapid activation of JAK2/STAT3 signal transduction in C6 astrocytes, which plays a pivotal role in the initiation of PFOS-mediated neurotoxicity.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Encefalite/induzido quimicamente , Encefalite/metabolismo , Fluorocarbonos/toxicidade , Janus Quinase 2/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Linhagem Celular Tumoral , Humanos , Neurônios/efeitos dos fármacos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
20.
Neurotoxicol Teratol ; 67: 65-75, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29577981

RESUMO

Perfluorooctane sulfonate (PFOS) is a persistent and bioaccumulative compound that has been widely used in various fields of life and industrial productions, because of its special chemical and physical properties. Numerous studies have indicated significant neurotoxic effect of PFOS, especially on neurons and microglia. However, the influence of PFOS on astrocyte physiology remains unclear. In this study, we showed that PFOS triggered reactive astrocytosis in time- and dose-dependent manners. The low-doses of PFOS increased the cell number and the expression of glial fibrillary acidic protein (GFAP), a well-known hallmark of reactive astrocytes, in C6 astrocyte cells. ELISA and RT-PCR analysis showed that PFOS promoted the expression and secretion of Interleukin-1 beta (IL-1ß) in dose- and time-dependent manners. Furthermore, PFOS exposure could induce the phosphorylation and degradation of IκBα, and the translocation of NF-κB p65 from the cytoplasm to the nucleus in C6 glioma cell line. Thus, the NF-кB signaling pathway can be activated after PFOS exposure. In addition, pretreatment with AKT inhibitor LY294002 could obviously attenuate PFOS-induced NF-κB activation and IL-1ß secretion. Taken together, these results indicated that PFOS could facilitate reactive astrocytosis and the secretion of pro-inflammatory cytokines through AKT-dependent NF-κB signaling pathway.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Astrócitos/metabolismo , Fluorocarbonos/toxicidade , Interleucina-1beta/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Ácidos Alcanossulfônicos/antagonistas & inibidores , Animais , Contagem de Células , Células Cultivadas , Cromonas/farmacologia , Relação Dose-Resposta a Droga , Fluorocarbonos/antagonistas & inibidores , Proteína Glial Fibrilar Ácida/metabolismo , Morfolinas/farmacologia , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa