Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Appl Toxicol ; 43(2): 312-322, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35999056

RESUMO

Perfluorooctane sulfonate (PFOS) is a hepatotoxic environmental organic pollutant that can cause aberrant lipid accumulation in the liver. However, the molecular mechanism underlying PFOS-induced hepatic steatosis remains unclear. Our research showed that subchronic PFOS exposure inhibited AMP-activated protein kinase (AMPK) phosphorylation, leading to increased acetyl-CoA carboxylase (ACC) activity, attenuated fatty acid ß-oxidation, and consequent liver lipid accumulation. We found that 1 mg/kg/day PFOS exposure significantly aggravated steatosis in high-fat diet (HFD)-fed mice, along with reduced AMPK activity. Oil Red O results showed that PFOS exposure caused fat accumulation in HepG2 cells. As predicted, PFOS treatment reduced the level of phosphorylated AMPK in a concentration-dependent manner, leading to subsequent increase in ACC activity and lipid droplet accumulation in HepG2 cells. Treatment with 200-µM AMPK agonist AICAR alleviated PFOS-induced ACC activation and lipid accumulation. In summary, our data highlight a crucial role of AMPK/ACC pathway in PFOS-mediated liver lipid metabolic disorders.


Assuntos
Proteínas Quinases Ativadas por AMP , Fígado Gorduroso , Camundongos , Animais , Proteínas Quinases Ativadas por AMP/metabolismo , Acetil-CoA Carboxilase/metabolismo , Dieta Hiperlipídica/efeitos adversos , Fígado/metabolismo , Fígado Gorduroso/induzido quimicamente , Fígado Gorduroso/metabolismo , Metabolismo dos Lipídeos , Lipídeos
2.
Environ Pollut ; 301: 118960, 2022 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-35150797

RESUMO

Perfluorooctane sulfonate (PFOS) is associated with male reproductive disorder, but the related mechanisms are still unclear. In this study, we used in vivo and in vitro models to explore the role of Sertoli cell-derived exosomes (SC-Exo)/miR-9-3p/StAR signaling pathway on PFOS-induced suppression of testosterone biosynthesis. Forty male ICR mice were orally administrated PFOS (0.5-10 mg/kg/bw) for 4 weeks. Bodyweight, organ index, sperm count, reproductive hormones were evaluated. Primary Sertoli cells and Leydig cells were used to delineate the molecular mechanisms that mediate the effects of PFOS on testosterone biosynthesis. Our results demonstrated that PFOS dose-dependently induced a decrease in sperm count, low levels of testosterone, and damage in testicular interstitium morphology. In vitro models, PFOS significantly increased miR-9-3p levels in Sertoli cells and SC-Exo, accompanied by a decrease in testosterone secretion and StAR expression in Leydig cells when Leydig cells were exposed to SC-Exo. Meanwhile, inhibition of SC-Exo or miR-9-3p by their inhibitors significantly rescued PFOS-induced decreases in testosterone secretion and the mRNA and protein expression of the StAR gene in Leydig cells. In summary, the present study highlights the role of the SC-Exo/miR-9-3p/StAR signaling pathway in PFOS-induced suppression of testosterone biosynthesis, advancing our understanding of molecular mechanisms for PFOS-induced male reproductive disorders.


Assuntos
Células Intersticiais do Testículo , MicroRNAs , Ácidos Alcanossulfônicos , Animais , Fluorocarbonos , Masculino , Camundongos , Camundongos Endogâmicos ICR , MicroRNAs/genética , MicroRNAs/metabolismo , Células de Sertoli , Testosterona/metabolismo
3.
Chem Commun (Camb) ; 58(19): 3202-3205, 2022 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-35174839

RESUMO

Low levels of immune infiltrates in the tumor milieu hinder the effectiveness of immunotherapy against immune-cold tumors. In the current work, a tumor-targeting drug delivery system composed of Endo-loaded platelets (Endo@PLT) was developed to relieve immunosuppression by achieving tumor vascular normalization. Endo@PLT reprogrammed the immunostimulatory phenotype, achieving excellent PD-1 immunotherapy in vivo.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias/imunologia , Neovascularização Patológica/imunologia , Microambiente Tumoral/imunologia , Inibidores da Angiogênese/química , Animais , Humanos , Terapia de Imunossupressão , Imunoterapia , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Neovascularização Patológica/terapia , Microambiente Tumoral/efeitos dos fármacos
4.
Environ Toxicol ; 37(5): 983-994, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34990082

RESUMO

Perfluorooctane sulfonate (PFOS) is a widespread environmental pollutant and may cause a variety of adverse health effects. The hepatotoxicity of PFOS has attracted particular attention, given the fact that the liver has one of the highest PFOS accumulations among human tissues. In this study, we revealed that subchronic PFOS exposure may exacerbate carbon tetrachloride (CCl4 )-induced liver fibrosis in animal models. Administration with 1 mg/kg PFOS every other day for 56 days dramatically enhanced CCl4 -mediated liver injury and hepatic stellate cell (HSC) activation. Furthermore, PFOS exposure may promote the activation of high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4) signaling pathway through inducing the secretion of HMGB1 from hepatocytes. PFOS exposure induced the translocation of HMGB1 from the nucleus into the cytoplasm of hepatocytes and cultured BRL-3A cells at a starting concentration of 50 µM. This process is accompanied with concurrent flux of calcium, suggesting a link between calcium signaling and HMGB1 release following PFOS exposure. Finally, we showed that PFOS-exposed conditional medium (PFOS-CM) of hepatocytes may induce the translocation of Smad2/3 in HSCs in a TLR4-dependent manner. Taken together, subchronic PFOS exposure might play a pro-fibrotic role via a HMGB1/TLR4-dependent Smad signaling in HSCs. Our findings for the first time uncovered an involvement of PFOS exposure in liver fibrosis via HMGB1/TLR4/Smad signaling.


Assuntos
Proteína HMGB1 , Receptor 4 Toll-Like , Ácidos Alcanossulfônicos/toxicidade , Animais , Fluorocarbonos/toxicidade , Proteína HMGB1/metabolismo , Células Estreladas do Fígado , Fígado , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
5.
J Appl Toxicol ; 42(5): 806-817, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34687223

RESUMO

Perfluorooctane sulfonate (PFOS) is a fluorinated organic pollutant with substantial accumulation in mammalian liver tissues. However, the impact of chronic PFOS exposure on liver disease progression and the underlying molecular mechanisms remain elusive. Herein, we for the first time revealed that micromolar range of PFOS exposure initiates the activation of NLR pyrin domain containing 3 (NLRP3) inflammasome to drive hepatocyte pyroptosis. We showed that 5 mg/kg/day PFOS exposure may exacerbated liver inflammation and steatosis in high-fat diet (HFD)-fed mice with concurrently elevated expression of NLRP3 and caspase-1. PFOS exposure resulted in viability impairment and LDH release in BRL-3A rat liver cells. 25-100 µM concentrations of PFOS exposure activated the NLRP3 inflammasome, leading to consequent GSDMD cleavage, IL-1ß release and the initiation of pyroptosis in a dose-dependent manner, whereas treatment with 10 µM NLRP3 inhibitor MCC950 abrogated this effect. Moreover, pretreatment of 5 mM ROS scavenger N-acetyl-L-cysteine (NAC) ameliorated PFOS-induced NLRP3 inflammasome activation and pyroptosis. Collectively, our data highlight a pivotal role of pyroptotic death in PFOS-mediated liver inflammation and metabolic disorder.


Assuntos
Inflamassomos , Piroptose , Ácidos Alcanossulfônicos , Animais , Fluorocarbonos , Hepatócitos , Inflamassomos/metabolismo , Inflamação/induzido quimicamente , Fígado/metabolismo , Mamíferos/metabolismo , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Ratos , Espécies Reativas de Oxigênio/metabolismo
6.
Ecotoxicol Environ Saf ; 227: 112895, 2021 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-34673407

RESUMO

Cadmium is an environmental pollutant that threatens the health of both humans and animals. Current studies have shown that while hepatotoxic damage induced by cadmium is closely related to autophagy, its intrinsic mechanism has not been elucidated. MicroRNA plays a regulatory role on different stages of autophagy. In this study, we investigated the mechanisms by which microRNA-155 (miR-155) regulate cadmium-induced hepatotoxicity in rat hepatocytes (BRL 3A cells) and in vivo. We found that cadmium exposure could cause liver injury in rats, resulting in a decreased liver index, increased alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP) activity, hepatocyte steatosis, and ultrastructure damage. Cadmium exposure also induced autophagy in hepatocytes, resulting in increased expression of ATG5, Belin1, LC3II, and an increased number of autophagosomes. In addition, cadmium exposure upregulated miR-155 expression, downregulated Rheb mRNA expression, and downregulated the level of protein expression in the Rheb/mTOR signaling pathway in rat hepatocytes. The overexpression of miR-155 followed by cadmium exposure upregulated the level of autophagy in BRL3A cells, whereas miR-155 inhibition had the opposite effect. In addition, miR-155 negatively regulated Rheb. A dual-luciferase reporter assay verified the negative regulatory effect of miR-155 on Rheb targeting. Knockdown of Rheb downregulated cadmium-induced autophagy. Therefore, the Rheb/mTOR signaling can negatively regulate autophagy. The present study demonstrates that miR-155 promotes cadmium-induced autophagy in rat hepatocytes by suppressing Rheb expression.


Assuntos
Cádmio , MicroRNAs , Animais , Autofagia , Cádmio/toxicidade , Hepatócitos/metabolismo , MicroRNAs/genética , Proteína Enriquecida em Homólogo de Ras do Encéfalo/metabolismo , Ratos , Transdução de Sinais
7.
J Mol Neurosci ; 71(4): 869-878, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32940875

RESUMO

Dietary zinc deficiency may lead to olfactory deficits, whose mechanism remains largely elusive. Olfactory ensheathing cells (OECs), a type of glial cells that support the function and neurogenesis in the olfactory bulb (OB), may play a pivotal role in the maintenance of the olfactory system. In the present study, we established a rat model of dietary zinc deficiency and found that severe zinc deficiency, but not marginal zinc deficiency, caused significantly reduced food intake, growth retardation, and apparent olfactory deficit in growing rats. We showed that severe zinc deficiency resulted in the loss of OECs in the olfactory nerve layer (ONL) of the olfactory bulb. In addition, we revealed that the number of TUNEL-positive cells increased markedly in the region, suggesting an involvement of apoptotic cell death in zinc deficiency-induced loss of OECs. Moreover, we found that treatment with zinc chelator N,N,N'N',-tetrakis (2-pyridylmethyl)ethylenediamine (TPEN) triggered the apoptosis of in vitro-cultured primary OECs. The apoptosis of OECs was correlated with significantly elevated expression of p53. Importantly, TUNEL and CCK-8 assays both demonstrated that treatment with p53 antagonist pifithrin-α (PFT-α) markedly attenuated TPEN-induced OEC apoptosis. These findings implicated that p53-triggered apoptosis of OECs might play an integral role in zinc deficiency-induced olfactory malfunction.


Assuntos
Apoptose , Neuroglia/metabolismo , Bulbo Olfatório/metabolismo , Olfato , Zinco/deficiência , Animais , Células Cultivadas , Quelantes/farmacologia , Etilenodiaminas/farmacologia , Feminino , Neuroglia/efeitos dos fármacos , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiopatologia , Ratos , Ratos Sprague-Dawley , Proteína Supressora de Tumor p53/metabolismo , Zinco/metabolismo
8.
Environ Toxicol Pharmacol ; 46: 9-16, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27414741

RESUMO

Perfluorooctane sulfonate (PFOS), the most extensively studied member of perfluoroalkyl and polyfluoroalkyl substances (PFASs), has been thought to be toxic to the central nervous system (CNS) of mammals. However, the neurotoxic effects of PFOS remain largely unknown. In this study, the effect of PFOS on microglial apoptosis was examined. The results showed that PFOS could significantly reduce the cell viability and mediate cell apoptosis in HAPI microglia, which was closely accompanied with ROS production and p53 overexpression. Moreover, p53 interference significantly ameliorated PFOS-triggered cytotoxic effects in HAPI microglia, including the downregulation of cleaved PARP and cleaved caspase 3. Interestingly, NAC, a ROS inhibitor, inhibited p53 expression, and decreased the apoptosis of HAPI microglia. Taken together, these findings suggest that upregulated production of ROS plays a vital role in PFOS-mediated apoptosis in HAPI microglia via the modulation of p53 signaling.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Apoptose/efeitos dos fármacos , Fluorocarbonos/toxicidade , Microglia/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Ativação Enzimática/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética
9.
Neurochem Res ; 41(8): 1969-81, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27068033

RESUMO

It has been widely accepted that astrocytes, play a role in regulating almost every physiological system. In the present study, we investigated the role of particulate matter (PM) in regulating activation of astrocytes. The glial cell strain C6 was cloned from a rat glioma which was induced by N-nitrosomethylurea. The C6 cells were plated at a density of 5 × 10(6) cells/10 cm diameter dish and incubated with different concentrations (0, 12, 25, 50, 100, 200, and 400 µg/mL) of PM for 24 h and different time (0, 1, 3, 6, 8,12, and 24 h) with 100 µg/mL at 37 °C. The study revealed that PM stimulated the expression of inducible nitric oxide synthase (iNOS) as well as the production of IL-1ß in a dose- and time-dependent manner. Furthermore, activation of JAK2/STAT3 and p38/JNK/ERK MAPKs was found in astrocytes following PM treatment. Blockage of JAK and p38/JNK/ERK MAPKs with their specific inhibitors, AG490, SB202190, SP600125 and U0126 significantly reduced PM-induced iNOS expression and IL-1ß production. In addition, it was demonstrated that inhibition of p38, JNK and JAK prevented STAT3 tyrosine phosphorylation induced by PM, while blocking ERK did not. MAPKs (p38 and JNK) could regulate tyrosine STAT3 phosphorylation, which suggested that the JAK2/STAT3 pathway might be the downstream of p38/JNK MAPK pathways.


Assuntos
Glioma/metabolismo , Mediadores da Inflamação/metabolismo , Janus Quinase 2/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Material Particulado/toxicidade , Fator de Transcrição STAT3/metabolismo , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ratos
10.
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
11.
J Neurochem ; 134(5): 879-91, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26086369

RESUMO

Zinc plays an important role in the development and maintenance of central neural system. Zinc deficiency has been known to alter normal brain function, whose molecular mechanism remains largely elusive. In the present study, we established a zinc deficiency-exposed rat model, and, using western blot and immunohistochemical analyses, found that the expression of FoxO3a and p27(kip1) was remarkably up-regulated in the rat brain hippocampus. Immunofluorescence assay showed that FOXO3a and p27(kip1) were significantly co-localized with nestin, the marker of neural stem cells (NSCs). Furthermore, we identified that the proportion of proliferating NSCs was markedly decreased in zinc-deficient rat hippocampaus. Using C17.2 neural stem cells, it was revealed that exposure to zinc chelator N,N,N',N'-tetrakis-(2-pyridylmethy) ethylenediamine induced the expression of FoxO3a and p27(kip1) , which coincided with reduced NSC proliferation. Furthermore, depletion of FoxO3a inhibited p27(kip1) expression and restored the growth of NSCs. On the basis of these data, we concluded that FoxO3a/p27(kip1) signaling might play a significant role in zinc deficiency-induced growth impairment of NSCs and consequent neurological disorders. We describe here that zinc deficiency induces the proliferative impairment of hippocampal neural stem cells partially through the activation of FOXO3a-p27 axis in rats. Neural progenitor cells exhibited significantly up-regulated expression of FOXO3a and p27 after zinc deficiency in vivo and in vitro. Depletion of FOXO3a ameliorates zinc deficiency-induced expression of p27 and growth impairment of neural stem cells. We provide novel insight into the mechanisms underlying zinc deficiency-induced neurological deficits.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Hipocampo/patologia , Células-Tronco Neurais/patologia , Zinco/deficiência , Animais , Ciclo Celular , Divisão Celular , Quelantes/farmacologia , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/genética , Etilenodiaminas/farmacologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Hipocampo/metabolismo , Masculino , Nestina/análise , Células-Tronco Neurais/metabolismo , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Regulação para Cima , Zinco/fisiologia
12.
Int Immunopharmacol ; 28(1): 52-60, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26004316

RESUMO

Perfluorooctane sulfonate (PFOS), a ubiquitous pollutant widely found in the environment and biota, can cause numerous adverse effects on human health. In recent years, PFOS's toxic effects on the central nervous system (CNS) have been shown. However, we still have a lot to study in the underlying molecular mechanism of PFOS's neurotoxicity. Microglia, the innate immune cells of CNS, are critically implicated in various neurological diseases caused by pro-inflammatory mediators. In our research, we found that HAPI microglia secreted tumor necrosis factor-alpha (TNF-α) after PFOS exposure in time-dependent and dose-dependent way. We also discovered that intracellular concentration of free Ca(2+) ([Ca(2+)]i) significantly increased after PFOS treatments. It was noteworthy here the secretion of TNF-α mediated by PFOS was blocked by Ca(2+) inhibitor and protein kinase C (PKC) inhibitor. Besides these, we had learned as well that PFOS brought about the up-regulation of phosphorylated nuclear factor kappa B (NF-кB) p65 expression and accelerated degradation of NF-κB inhibitor alpha (IкBα), however, these effects could be attenuated or blocked by Ca(2+) inhibitor and PKC inhibitor. Finally, through treating SH-SY5Y cells with PFOS-treated microglial conditioned medium, we demonstrated that TNF-α mediated neuronal apoptosis. To sum up, our research had shown, for the first time, that the distinct TNF-α secretion brought by PFOS in HAPI microglia, was achieved through the Ca(2+)-dependent PKC-NF-кB signaling, subsequently participating in neuronal loss.


Assuntos
Ácidos Alcanossulfônicos/toxicidade , Sinalização do Cálcio/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Fluorocarbonos/toxicidade , Ativação de Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Microglia/metabolismo , NF-kappa B/efeitos dos fármacos , Proteína Quinase C/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Quinase I-kappa B/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição RelA/biossíntese , Regulação para Cima/efeitos dos fármacos
13.
Brain Res ; 1615: 61-70, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-25912432

RESUMO

Zinc is an essential nutrient that is important for normal brain development. Zinc deficiency has been linked to aberrant neurological development and functioning. However, the molecular mechanisms underlying Zinc deficiency-induced neurological disorders remain largely elusive. In the present study, we showed that the proliferation of C17.2 neural stem cells (NSCs) was evidently impaired after exposed to low levels of Zinc chelator, N,N,N',N'-tetrakis-(2-pyridylmethy) ethylenediamine (TPEN). In addition, we found that TPEN-induced proliferative deficit of NSCs was related with significant downregulation of Wnt/ß-catenin signaling. Zinc deficiency impaired the proliferation of neural stem cells in dose- and time-dependent manners. Western blot revealed that the levels of p-Ser9-glycogensynthase kinase-3ß (p-GSK-3ß) and ß-catenin were remarkably downregulated during TPEN-induced C17.2 proliferative impairment. Moreover, immunofluorescent analysis indicated that the level of nuclear ß-catenin was apparently decreased following TPEN exposure. Furthermore, application with GSK-3ß inhibitor lithium chloride (LiCl) reversed TPEN-induced downregulation of ß-catenin and impairment of cell proliferation. Flow cytometry analysis also showed that TPEN-induced impairment of NSC proliferation could be reversed by LiCl. Taken together, these findings suggested that the disturbance of canonical Wnt/ß-catenin signaling pathway partially accounted for Zinc deficiency-induced proliferative impairment of NSCs.


Assuntos
Proliferação de Células , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/fisiologia , Via de Sinalização Wnt , Zinco/deficiência , Animais , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo , Etilenodiaminas , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
14.
Toxicol Lett ; 235(1): 17-27, 2015 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-25791630

RESUMO

Overexposure to manganese (Mn) has been known to induce neuronal death and neurodegenerative symptoms. However, the precise mechanisms underlying Mn neurotoxicity remain incompletely understood. In the present study, we established a Mn-exposed rat model and found that downregulation of wild type p53-induced phosphatase 1 (Wip1) might contribute to p53 activation and resultant neuronal apoptosis following Mn exposure. Western blot and immunohistochemical analyses revealed that the expression of Wip1 was markedly decreased following Mn exposure. In addition, immunofluorescence assay demonstrated that Mn exposure led to significant reduction in the number of Wip1-positive neurons. Accordingly, the expression of Mdm2 was progressively decreased, which was accompanied with markedly increased expression of p53, as well as the ratio of Bax/Bcl-xl. Furthermore, we showed that Mn exposure decreased the viability and induced apparent apoptosis in NFG-differentiated neuron-like PC12 cells. Importantly, the expression of Wip1 decreased progressively, whereas the level of cellular p53 and the ratio of Bax/Bcl-xl were elevated, which resembled the expression of the proteins in animal model studies. Depletion of p53 significantly ameliorated Mn-mediated cytotoxic effect in PC12 cells. In addition, ectopic expression of Wip1 attenuated Mn-induced p53 signaling as well as apoptosis in PC12 cells. Finally, we observed that depletion of Wip1 augmented Mn-induced apoptosis in PC12 cells. Collectively, these findings suggest that downregulated Wip1 expression plays an important role in Mn-induced neuronal death in the brain striatum via the modulation of p53 signaling.


Assuntos
Apoptose , Gânglios da Base/enzimologia , Intoxicação por Manganês/enzimologia , Neurônios/enzimologia , Fosfoproteínas Fosfatases/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/efeitos dos fármacos , Gânglios da Base/patologia , Cloretos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Masculino , Compostos de Manganês , Intoxicação por Manganês/etiologia , Intoxicação por Manganês/genética , Intoxicação por Manganês/patologia , Degeneração Neural , Neurônios/efeitos dos fármacos , Neurônios/patologia , Células PC12 , Fosfoproteínas Fosfatases/genética , Proteína Fosfatase 2C , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transfecção , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/metabolismo
15.
J Mol Neurosci ; 55(2): 454-65, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25027559

RESUMO

Manganese (Mn) is an essential micronutrient. However, exposure to high doses of Mn may lead to a neurological disease known as manganism, which is characterized by marked brain neuronal loss. K-homology splicing regulator protein (KHSRP) is a multifunctional RNA-binding protein and has been implicated in the regulation of multiple cellular signaling associated with neuronal apoptosis and survival, such as p38 mitogen-activated protein kinase (MAPK), nuclear factor kappaB (NF-κB), and Wnt/ß-catenin pathways. In the present study, the role of KHSRP in Mn-induced neurotoxicity was investigated in vivo using a rat model of chronic Mn exposure and in vitro using differentiated PC12 cell cultures. Western blot and immunohistochemical analyses showed a significant upregulation of KHSRP in rat striatum following Mn exposure. Immunofluorescent labeling indicated that KHSRP was localized mainly in neurons. Terminal deoxynucleotidyl transferase-mediated biotinylated-dUTP nick end labeling (TUNEL) assay showed that KHSRP was mainly distributed in apoptotic neurons. Increased KHSRP expression was positively correlated with the upregulation of several apoptosis-related proteins, such as p53, bax, and active caspase-3. In addition, significant co-localization of KHSRP and active caspase-3 in neurons after Mn exposure was also observed, suggesting a potential involvement of KHSRP in the regulation of Mn-induced striatal neuronal apoptosis. Importantly, interference with KHSRP apparently decreased the level of p53 and attenuated Mn-induced neuronal apoptosis. Taken together, these results indicate that upregulation of KHSRP may be involved in the pathological process underlying Mn neurotoxicity via the modulation of p53 signaling.


Assuntos
Corpo Estriado/metabolismo , Manganês/toxicidade , Proteínas de Ligação a RNA/metabolismo , Transativadores/metabolismo , Animais , Apoptose , Corpo Estriado/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Células PC12 , Proteínas de Ligação a RNA/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Transativadores/genética , Proteína Supressora de Tumor p53/metabolismo
16.
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
17.
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
18.
Neurotoxicology ; 44: 149-59, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24932542

RESUMO

Studies have shown that 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces apoptotic cell death in neuronal cells. However, whether this is the result of endoplasmic reticulum (ER) stress-mediated apoptosis remains unknown. In this study, we determined whether ER stress plays a role in the TCDD-induced apoptosis of pheochromocytoma (PC12) cells and primary neurons. PC12 cells were exposed to different TCDD concentrations (1, 10, 100, 200, or 500nM) for varying lengths of time (1, 3, 6, 12, or 24h). TCDD concentrations much higher than 10nM (100, 200, or 500nM) markedly increased glucose-regulated protein (GRP78) and C/EBP homologous protein (CHOP) levels, which are hallmarks of ER stress. We also evaluated the effects of TCDD on ER morphology in PC12 cells and primary neurons that were treated with different TCDD concentrations (1, 10, 50, or 200nM) for 24h. Ultrastructural ER alterations were observed with transmission electron microscopy in PC12 cells and primary neurons treated with high concentrations of TCDD. Furthermore, TCDD-induced ER stress significantly promoted the activation of the PKR-like ER kinase (PERK), a sensor for the unfolded protein response (UPR), and its downstream target eukaryotic translation initiation factor 2 α (eIF2α); in contrast, TCDD did not appear to affect inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6 (ATF6), two other UPR sensors. Importantly, TCDD significantly inhibited eIF2α phosphorylation and triggered apoptosis in PC12 cells after 6-24h of treatment. Salubrinal, which activates the PERK-eIF2α pathway, significantly enhanced eIF2α phosphorylation in PC12 cells and attenuated the TCDD-induced cell death. In contrast, knocking down eIF2α using small interfering RNA markedly enhanced TCDD-induced cell death. Together, these results indicate that the PERK-eIF2α pathway plays an important role in TCDD-induced ER stress and apoptosis in PC12 cells.


Assuntos
Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , eIF-2 Quinase/metabolismo , Animais , Cinamatos/farmacologia , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/ultraestrutura , Células PC12 , Ratos , Transdução de Sinais , Tioureia/análogos & derivados , Tioureia/farmacologia , Resposta a Proteínas não Dobradas
19.
Environ Toxicol Pharmacol ; 38(1): 119-30, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24930124

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been reported to cause alterations in cognitive and motor behavior during both development and adulthood. In this study, the neuronal nitric oxide synthase (nNOS) signaling pathway was investigated in differentiated pheochromocytoma (PC12) cells to better understand the mechanisms of TCDD-induced neurotoxicity. TCDD exposure induced a time- and dose-dependent increase in nNOS expression. High levels of nitric oxide (NO) production by nNOS activation induced mitochondrial cytochrome c (Cyt-c) release and down-regulation of Bcl-2. Additionally, TCDD increased the expression of active caspase-3 and significantly led to apoptosis in PC12 cells. However, these effects above could be effectively inhibited by the addition of 7-nitroindazole (7-NI), a highly selective nNOS inhibitor. Moreover, in the brain cortex of Sprague-Dawley (SD) rats, nNOS was also found to have certain relationship with TCDD-induced neuronal apoptosis. Together, our findings establish a role for nNOS as an enhancer of TCDD-induced apoptosis in PC12 cells.


Assuntos
Poluentes Ambientais/toxicidade , Síndromes Neurotóxicas/metabolismo , Neurotoxinas/toxicidade , Óxido Nítrico Sintase Tipo I/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Animais , Apoptose/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Citocromos c/metabolismo , Proteína 4 Homóloga a Disks-Large , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Neurônios/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Óxido Nítrico/metabolismo , Células PC12 , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
20.
PLoS One ; 9(2): e89811, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587053

RESUMO

The widespread environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a potent toxicant that causes significant neurotoxicity. However, the biological events that participate in this process remain largely elusive. In the present study, we demonstrated that TCDD exposure triggered apparent premature senescence in rat pheochromocytoma (PC12) and human neuroblastoma SH-SY5Y cells. Senescence-associated ß-galactosidase (SA-ß-Gal) assay revealed that TCDD induced senescence in PC12 neuronal cells at doses as low as 10 nM. TCDD led to F-actin reorganization and the appearance of an alternative senescence marker, γ-H2AX foci, both of which are important features of cellular senescence. In addition, TCDD exposure altered the expression of senescence marker proteins, such as p16, p21 and p-Rb, in both dose- and time-dependent manners. Furthermore, we demonstrated that TCDD promotes mitochondrial dysfunction and the accumulation of cellular reactive oxygen species (ROS) in PC12 cells, leading to the activation of signaling pathways that are involved in ROS metabolism and senescence. TCDD-induced ROS generation promoted significant oxidative DNA damage and lipid peroxidation. Notably, treatment with the ROS scavenger N-acetylcysteine (NAC) markedly attenuated TCDD-induced ROS production, cellular oxidative damage and neuronal senescence. Moreover, we found that TCDD induced a similar ROS-mediated senescence response in human neuroblastoma SH-SY5Y cells. In sum, these results demonstrate for the first time that TCDD induces premature senescence in neuronal cells by promoting intracellular ROS production, supporting the idea that accelerating the onset of neuronal senescence may be an important mechanism underlying TCDD-induced neurotoxic effects.


Assuntos
Senescência Celular/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Tecido Nervoso/efeitos dos fármacos , Dibenzodioxinas Policloradas/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Actinas/metabolismo , Análise de Variância , Animais , Biomarcadores/metabolismo , Western Blotting , Dano ao DNA/efeitos dos fármacos , Primers do DNA/genética , Relação Dose-Resposta a Droga , Poluentes Ambientais/metabolismo , Fluorescência , Histonas/metabolismo , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Tecido Nervoso/citologia , Células PC12 , Fosfoproteínas/metabolismo , Dibenzodioxinas Policloradas/metabolismo , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Tempo , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA