Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Toxicol In Vitro ; 59: 246-254, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31009676

RESUMO

Previous studies revealed that cellular accumulation of mono(2-ethylhexyl)phthalate (MEHP) disturbed energy metabolism in adipocytes, where glucose uptake was significantly increased. The present study aimed to determine the mechanisms underlying the increased glucose uptake. MEHP-treated 3T3-L1 adipocytes exhibited a significantly increased glucose uptake activity. Immunoblot analysis suggested that the insulin-induced signals were not responsible for the increased glucose uptake. qPCR analysis revealed that both Glut1 and Glut4 genes were highly expressed during adipogenesis; Glut1 mRNA levels in MEHP-treated adipocytes were significantly increased. Moreover, MEHP-treated adipocytes exhibited significantly increased levels of fibroblast growth factor 21 (FGF21) in both mRNA and secreted protein. FGF21 is a peptide hormone with pleiotropic effects on regulation of insulin sensitivity and glucose/lipid homeostasis. We found that MEHP, FGF21, and lactate in culture medium together enhanced Fgf21 gene expression in MEHP-treated adipocytes. FGF21 signaling requires fibroblast growth factor receptor (FGFR) and ßKlotho. Fgfr family and ßKlotho genes were actively expressed during adipogenesis; mRNA levels of Fgfr3 and Fgfr4 genes in MEHP-treated adipocytes were significantly increased. Roles of FGF21/FGFR and phosphoinositide 3-kinase (PI3K)/AKT signal axes in regulation of glucose uptake were determined. We demonstrated that FGF21/FGFR signals played the major roles in up-regulation of the basal glucose uptake in MEHP-treated adipocytes. The in vitro evidence suggests that cellular FGF21 secretion enhances the basal glucose uptake in MEHP-treated adipocytes.


Assuntos
Células 3T3-L1/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Dietilexilftalato/análogos & derivados , Fatores de Crescimento de Fibroblastos/metabolismo , Glucose/metabolismo , Células 3T3-L1/metabolismo , Adipócitos/metabolismo , Animais , Dietilexilftalato/farmacologia , Insulina/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
2.
J Appl Toxicol ; 37(12): 1537-1546, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28849599

RESUMO

The study aimed to examine effects of environmental estrogens at body burden levels on energy metabolism in fat cells. Acclimation of T47D-KBluc cells in estrogen-deprived medium was established for high performance of estrogen-responsive luciferase reporter assay. With the assay, relative estrogenic potency of four selected estrogen receptor (ER) agonists, i.e. diethylstilbestrol, ß-estradiol, 4-nonylphenol and bisphenol A, were determined. Immunoblot analysis revealed that the ER agonists at both EC80 and EC100 caused rapid and transient phosphorylation of extracellular signal-regulated kinases (ERK) in an ER-dependent manner. 3T3-L1 adipocytes treated with the ER agonists at EC80 for 24 hours exhibited significant downregulation in mitochondrial respiration and glycolytic function. Importantly, EC80 values of 4-nonylphenol (6.0 × 10-10  m) and bisphenol A (1.0 × 10-8  m) are in the range of human body burdens. The finding that estrogenic chemicals at body burden levels cause significant impact on fat cell energy metabolism raises an important public health issue that deserves more attention.


Assuntos
Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Estrogênios não Esteroides/toxicidade , Estrogênios/toxicidade , Receptores de Estrogênio/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Carga Corporal (Radioterapia) , Relação Dose-Resposta a Droga , Camundongos
3.
Oncotarget ; 7(31): 49310-49321, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27384479

RESUMO

The development of resistance to platinum drugs in cancer cells severely reduces the efficacy of these drugs. Thus, the discovery of novel drugs or combined strategies to overcome drug resistance is imperative. In addition to our previous finding that combined D-penicillamine with platinum drugs exerts synergistic cytotoxicity, we recently identified a novel therapeutic strategy by combining an iron chelating agent desferal with platinum drugs to overcome platinum resistance in an oxaliplatin-resistant human cervical cancer cell line, S3. Further study demonstrated that the level of platinum-DNA adduct formation positively correlated with cell death in combination of desferal with platinums than that of each drug alone in S3 cells. Decrement of human copper transporter 1 (hCtr1) and transferrin receptor 1 (TfR1) expression involved in the development of platinum resistance in S3 cells. Moreover, desferal promoted the expression of hCtr1 through the upregulation of Sp1. The overexpression of Sp1 increased the expression of NF-κB and translocated it into the nucleus to bind to the TfR1 promoter region, which subsequently increased the expression of TfR1. Importantly, the cotreatment of oxaliplatin with desferal significantly potentiated the oxaliplatin-elicited antitumoral effect in the oxaliplatin-resistant xenograft animal model without any toxic effect observed. Taken together, these results demonstrated that the combination of desferal with oxaliplatin can overcome oxaliplatin resistance through the regulation of hCtr1 and TfR1, and may have beneficial effect for treatment of patient with oxaliplatin-refractory tumors.


Assuntos
Antígenos CD/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Desferroxamina/administração & dosagem , Compostos Organoplatínicos/administração & dosagem , Receptores da Transferrina/metabolismo , Fator de Transcrição Sp1/metabolismo , Neoplasias do Colo do Útero/tratamento farmacológico , Transporte Ativo do Núcleo Celular , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Núcleo Celular/metabolismo , Quelantes/química , Transportador de Cobre 1 , Adutos de DNA , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Ferro/química , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Oxaliplatina , Regiões Promotoras Genéticas , Regulação para Cima
4.
Biochem Pharmacol ; 95(1): 28-37, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25801007

RESUMO

The platinum-based regimen is the front-line treatment of chemotherapy. However, development of platinum resistance often causes therapeutic failure in this disease. We previously have generated an oxaliplatin-resistant subline, named S3, from human cervical carcinoma SiHa cells, and its resistant phenotype was well-characterized. In the present study, we aimed to identify the novel therapeutic strategy by combining copper chelator D-penicillamine with oxaliplatin, and to elucidate the underlying mechanisms for overcoming oxaliplatin resistance. As the result, D-penicillamine exerted synergistic killing effects only in S3 cells when combined with oxaliplatin and cisplatin by using Chou-Talalay method. Further study showed that the amounts of platinum DNA adduct formed were positively correlated to the percentage of cell death in S3 cells when co-treated D-penicillamine with oxaliplatin and cisplatin. D-penicillamine promoted copper influx transporter hCtr1 expression through upregulation of Sp1. Sp1 overexpression induced p53 translocation from nucleus to cytosol and caused p53 degradation through ubiquitination, which subsequently suppressed the expression of the copper efflux transporter ATP7A. Importantly, co-treatment of cisplatin with D-penicillamine enhanced oxaliplatin-elicited antitumor effect in the oxalipatin-resistant S3 xenograft tumors, but not found in SiHa xenograft model. Notably, Mice received D-penicillamine alone or in combination of D-penicillamine ad oxalipatin, increased hCtrl protein level in S3 xenograft tumor, however, the protein level of ATP7A was decreased. Taken together, this study provides insight into that the co-manipulation of hCtrl and ATP7A by D-penicillamine could increase the therapeutic efficacy of platinum drugs in oxaliplatin resistant tumors, especially in resistant phenotype with downexpression of hCtrl and overexpression of ATP7A.


Assuntos
Antineoplásicos/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Compostos Organoplatínicos/administração & dosagem , Penicilamina/administração & dosagem , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Quelantes/administração & dosagem , Cisplatino/administração & dosagem , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Oxaliplatina , Neoplasias do Colo do Útero/patologia
5.
Toxicol Appl Pharmacol ; 273(3): 492-9, 2013 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-24080332

RESUMO

Our previous studies indicated that zinc induced inflammatory response in both vascular endothelial cells and promonocytes. Here, we asked if other metals could cause the similar effect on vascular endothelial cells and tried to determine its underlying mechanism. Following screening of fifteen metals, zinc and nickel were identified with a marked proinflammatory effect, as determined by ICAM-1 and IL-8 induction, on human umbilical vein endothelial cells (HUVECs). Inhibiting protein expression of myeloid differentiation primary response protein-88 (MyD88), a Toll-like receptor (TLR) adaptor acting as a TLR-signaling transducer, significantly attenuated the zinc/nickel-induced inflammatory response, suggesting the critical roles of TLRs in the inflammatory response. Blockage of TLR-4 signaling by CLI-095, a TLR-4 inhibitor, completely inhibited the nickel-induced ICAM-1 and IL-8 expression and NFκB activation. The same CLI-095 treatment significantly blocked the zinc-induced IL-8 expression, however with no significant effect on the ICAM-1 expression and a minor inhibitory effect on the NFκB activation. The finding demonstrated the differential role of TLR-4 in regulation of the zinc/nickel-induced inflammatory response, where TLR-4 played a dominant role in NFκB activation by nickel, but not by zinc. Moreover, inhibition of NFκB by adenovirus-mediated IκBα expression and Bay 11-7025, an inhibitor of cytokine-induced IκB-α phosphorylation, significantly attenuated the zinc/nickel-induced inflammatory responses, indicating the critical of NFκB in the process. The study demonstrates the crucial role of TLRs in the zinc/nickel-induced inflammatory response in vascular endothelial cells and herein deciphers a potential important difference in NFκB activation via TLRs. The study provides a molecular basis for linkage between zinc/nickel exposure and pathogenesis of the metal-related inflammatory vascular disease.


Assuntos
Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Níquel/toxicidade , Receptor 4 Toll-Like/metabolismo , Zinco/toxicidade , Adenoviridae/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Fator 88 de Diferenciação Mieloide/antagonistas & inibidores , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/metabolismo , Transdução de Sinais , Sulfonamidas/farmacologia , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/genética , Doenças Vasculares/induzido quimicamente , Doenças Vasculares/patologia
6.
J Hazard Mater ; 196: 335-41, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21974847

RESUMO

Our previous studies found that zinc oxide (ZnO) particles induced expression of intercellular adhesion molecule-1 (ICAM-1) protein in vascular endothelial cells via NF-κB and that zinc ions dissolved from ZnO particles might play the major role in the process. This study aimed to determine if zinc ions could cause inflammatory responses in a human promonocytic leukemia cell line HL-CZ. Conditioned media from the zinc-treated HL-CZ cells induced ICAM-1 protein expression in human umbilical vein endothelial cells (HUVEC). Zinc treatment induced chemokine and inflammatory cytokine release from HL-CZ cells. Inhibition of NFκB activity by over-expression of IκBα in HL-CZ cells did not block the conditioned medium-induced ICAM-1 protein expression in HUVEC cells. Zinc treatment induced activation of multiple immune response-related transcription factors in HL-CZ cells. These results clearly show that zinc ions induce chemokine and inflammatory cytokine release from human promonocytes, accompanied with activation of multiple immune response-related transcription factors. Our in vitro evidence in the zinc-induced inflammatory responses of vascular cells provides a critical linkage between zinc exposure and pathogenesis of those inflammatory vascular diseases.


Assuntos
Quimiocinas/metabolismo , Molécula 1 de Adesão Intercelular/biossíntese , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Células Precursoras de Monócitos e Macrófagos/imunologia , Zinco/toxicidade , Adenoviridae/genética , Sequência de Bases , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Quimiocinas/genética , Meios de Cultivo Condicionados , Citocinas/genética , Citocinas/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Genes Reporter , Células Endoteliais da Veia Umbilical Humana , Humanos , Luciferases de Vaga-Lume/genética , Dados de Sequência Molecular , Células Precursoras de Monócitos e Macrófagos/metabolismo , Fatores de Transcrição/genética , Acetato de Zinco/toxicidade
7.
J Hazard Mater ; 183(1-3): 182-8, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20674161

RESUMO

This study investigated inflammatory effects of zinc oxide (ZnO) particles on vascular endothelial cells. The effects of 50 and 100-nm ZnO particles on human umbilical vein endothelial cells (HUVECs) were characterized by assaying cytotoxicity, cell proliferation, and glutathione levels. A marked drop in survival rate was observed when ZnO concentration was increased to 45 µg/ml. ZnO concentrations of ≤3 µg/ml resulted in increased cell proliferation, while those of ≤45 µg/ml caused dose-dependent increases in oxidized glutathione levels. Treatments with ZnO concentrations ≤45 µg/ml were performed to determine the expression of intercellular adhesion molecule-1 (ICAM-1) protein, an indicator of vascular endothelium inflammation, revealing that ZnO particles induced a dose-dependent increase in ICAM-1 expression and marked increases in NF-κB reporter activity. Overexpression of IκBα completely inhibited ZnO-induced ICAM-1 expression, suggesting NF-κB plays a pivotal role in regulation of ZnO-induced inflammation in HUVECs. Additionally, TNF-α, a typical inflammatory cytokine, induced ICAM-1 expression in an NF-κB-dependent manner, and ZnO synergistically enhanced TNF-α-induced ICAM-1 expression. Both 50 and 100-nm ZnO particles agglomerated to similar size distributions. This study reveals an important role for ZnO in modulating inflammatory responses of vascular endothelial cells via NF-κB signaling, which could have important implications for treatments of vascular disease.


Assuntos
Células Endoteliais/patologia , Inflamação/induzido quimicamente , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Óxido de Zinco/farmacologia , Morte Celular , Proliferação de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Glutationa/análise , Humanos , Molécula 1 de Adesão Intercelular/análise , Nanopartículas , Transdução de Sinais/imunologia
8.
Toxicol In Vitro ; 23(2): 209-16, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19100828

RESUMO

Resistance to chemotherapy is one of the major problems in treatment responses of lung cancer. This study explored the mechanism underlying the arsenic resistance of lung cancer. Four lung cancer cells with different proliferation activity were characterized for cytotoxicity, arsenic influx/efflux, and arsenic effects on intracellular glutathione and 8-hydroxy-2'-deoxyguanosine (8-OHdG) production. Our data revealed that relative proliferation potency of these cells was H1299>A549>CL3>H1355. Moreover, A549, H1299, and H1355 were markedly resistant to As(2)O(3) with IC50 approximately 100 microM, whereas CL3 was sensitive to As(2)O(3) with IC50 approximately 11.8 microM. After treatment with the respective As(2)O(3) at IC50, arsenic influx/efflux activity in CL3 was comparable to those in the other three arsenic-resistant cells. However, differences in glutathione levels and 8-OHdG production were also detected either before or after arsenic treatment, indicating that a certain degree of variation in anti-oxidative systems and/or 8-OHdG repair activity existed in these cell lines. By transfection of an aquaglyceroporin 9 (AQP9) gene, we showed that increased AQP9 expression significantly enhanced arsenic uptake and disrupted arsenic resistance of A549. The present study strongly suggests that membrane transporters responsible for arsenic uptake, such as AQP9, may play a critical role in development of arsenic resistance in human lung cancer cells.


Assuntos
Antineoplásicos/farmacologia , Aquaporinas/metabolismo , Arsenicais/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Óxidos/farmacologia , 8-Hidroxi-2'-Desoxiguanosina , Antineoplásicos/metabolismo , Aquaporinas/genética , Trióxido de Arsênio , Arsenicais/metabolismo , Linhagem Celular Tumoral , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Óxidos/metabolismo
9.
Toxicol Appl Pharmacol ; 221(2): 168-78, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17467021

RESUMO

We investigated the regulatory role of glutathione in tumor necrosis factor-alpha (TNF-alpha)-induced vascular endothelial dysfunction as evaluated by using vascular endothelial adhesion molecule expression and monocyte-endothelial monolayer binding. Since TNF-alpha induces various biological effects on vascular cells, TNF-alpha dosage could be a determinant factor directing vascular cells into different biological fates. Based on the adhesion molecule expression patterns responding to different TNF-alpha concentrations, we adopted the lower TNF-alpha (0.2 ng/ml) to rule out the possible involvement of other TNF-alpha-induced biological effects. Inhibition of glutathione synthesis by l-buthionine-(S,R)-sulfoximine (BSO) resulted in down-regulations of the TNF-alpha-induced adhesion molecule expression and monocyte-endothelial monolayer binding. BSO attenuated the TNF-alpha-induced nuclear factor-kappaB (NF-kappaB) activation, however, with no detectable effect on AP-1 and its related mitogen-activated protein kinases (MAPKs). Deletion of an AP-1 binding site in intercellular adhesion molecule-1 (ICAM-1) promoter totally abolished its constitutive promoter activity and its responsiveness to TNF-alpha. Inhibition of ERK, JNK, or NF-kappaB attenuates TNF-alpha-induced ICAM-1 promoter activation and monocyte-endothelial monolayer binding. Our study indicates that TNF-alpha induces adhesion molecule expression and monocyte-endothelial monolayer binding mainly via activation of NF-kappaB in a glutathione-sensitive manner. We also demonstrated that intracellular glutathione does not modulate the activation of MAPKs and/or their downstream AP-1 induced by lower TNF-alpha. Although AP-1 activation by the lower TNF-alpha was not detected in our systems, we could not rule out the possible involvement of transiently activated MAPKs/AP-1 in the regulation of TNF-alpha-induced adhesion molecule expression.


Assuntos
Endotélio Vascular/fisiopatologia , Glutationa/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Glutationa/biossíntese , Humanos , Molécula 1 de Adesão Intercelular/genética , Oxirredução , Regiões Promotoras Genéticas , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/efeitos dos fármacos
10.
J Hazard Mater ; 146(1-2): 356-61, 2007 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-17234337

RESUMO

In the present study, we investigated the effect of arecoline, a major areca nut alkaloid, on the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced activation of cytochrome P4501A1 (CYP1A1) in a human hepatoma cell line Huh-7. We treated Huh-7 cells with 10nM TCDD in the presence of different concentrations of arecoline (50-300 microM). Our results indicated that arecoline attenuated the TCDD-induced CYP1A1 enzyme activation with an inhibitory effect on cell proliferation. By using real-time RT-PCR, we demonstrated that arecoline inhibited the TCDD-induced activations of CYP1A1 and AhR repressor (AhRR) mRNA expression in a similar pattern. Our results revealed that arecoline inhibited AhR mRNA expression with no direct effect on CYP1A1 enzyme activity. Therefore, in our present study, the observed inhibitory effect of arecoline on CYP1A1 activation was not due to the up-regulation of AhRR or direct inhibitory effect on CYP1A1. Taken together, here we have demonstrated that arecoline attenuates the TCDD-induced CYP1A1 activation mainly via down-regulation of AhR expression in human hepatoma cells, suggesting the possible involvement of arecoline in the AhR-mediated metabolism of environmental toxicants in liver.


Assuntos
Arecolina/farmacologia , Citocromo P-450 CYP1A1/metabolismo , Dibenzodioxinas Policloradas/farmacologia , Areca , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Linhagem Celular Tumoral , Citocromo P-450 CYP1A1/genética , Regulação para Baixo , Ativação Enzimática/efeitos dos fármacos , Humanos , RNA Mensageiro/metabolismo , Receptores de Hidrocarboneto Arílico/genética , Proteínas Repressoras/genética
11.
Arch Toxicol ; 80(12): 804-10, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16645841

RESUMO

Epidemiological studies have demonstrated a high association of inorganic arsenic exposure with vascular disease. Our recent in vitro studies have linked this vascular damage to vascular endothelial dysfunction induced by arsenic exposure. However, cell-cycle arrest induced by arsenic and its involvement in vascular dysfunction remain to be clarified. In this study, we employed primary porcine aortic endothelial cells to investigate regulatory mechanisms of G2/M phase arrest induced by arsenite. Our study revealed that lower concentrations of arsenite (1 and 3 microM) increased cell proliferation, whereas higher concentrations of arsenite (10, 20, and 30 microM) inhibited cell proliferation together with correlated increases in G2/M phase arrest. We found that this arsenite-induced G2/M phase arrest was accompanied by accumulation and/or phosphorylation of checkpoint-related molecules, including p53, Cdc25B, Cdc25C, and securin. Inhibition of activations of these checkpoint-related molecules by caffeine significantly attenuated the 30-microM arsenite-induced G2/M phase arrest by 93%. Our data suggest that the DNA damage responsive kinases ATM (ataxia-telangiectasia mutated) and ATR (ATM and Rad3-related) play critical roles in arsenite-induced G2/M phase arrest in aortic endothelial cells possibly via regulation of checkpoint-related signaling molecules including p53, Cdc25B, Cdc25C, and securin.


Assuntos
Aorta Torácica/efeitos dos fármacos , Arsenitos/toxicidade , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fase G2/efeitos dos fármacos , Mitose/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Compostos de Sódio/toxicidade , Proteínas Supressoras de Tumor/metabolismo , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Proteínas Mutadas de Ataxia Telangiectasia , Cafeína/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Proteínas de Ligação a DNA/antagonistas & inibidores , Relação Dose-Resposta a Droga , Células Endoteliais/patologia , Ativação Enzimática , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Suínos , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/antagonistas & inibidores , Fosfatases cdc25/metabolismo
12.
Toxicol Appl Pharmacol ; 209(1): 10-8, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16271621

RESUMO

Epidemiological studies demonstrated a high association of vascular diseases with arsenite exposure. We hypothesize that arsenite potentiates the effect of proinflammatory cytokines on vascular endothelial cells, and hence contributes to atherosclerosis. In this study, we investigated the effect of arsenite and its induction of glutathione (GSH) on vascular cell adhesion molecule-1 (VCAM-1) protein expression in human umbilical vein endothelial cells (HUVECs) in response to tumor necrosis factor-alpha (TNF-alpha), a typical proinflammatory cytokine. Our study demonstrated that arsenite pretreatment potentiated the TNF-alpha-induced VCAM-1 expression with up-regulations of both activator protein-1 (AP-1) and nuclear factor-kappaB (NF-kappaB). To elucidate the role of GSH in regulation of AP-1, NF-kappaB, and VCAM-1 expression, we employed l-buthionine (S,R)-sulfoximine (BSO), a specific gamma-glutamylcysteine synthetase (gamma-GCS) inhibitor, to block intracellular GSH synthesis. Our investigation revealed that, by depleting GSH, arsenite attenuated the TNF-alpha-induced VCAM-1 expression as well as a potentiation of AP-1 and an attenuation of NF-kappaB activations by TNF-alpha. Moreover, we found that depletion of GSH would also attenuate the TNF-alpha-induced VCAM-1 expression with a down-regulation of the TNF-alpha-induced NF-kappaB activation and without significant effect on AP-1. On the other hand, the TNF-alpha-induced VCAM-1 expression could be completely abolished by inhibition of AP-1 or NF-kappaB activity, suggesting that activation of both AP-1 and NF-kappaB was necessary for VCAM-1 expression. In summary, we demonstrate that arsenite enhances the TNF-alpha-induced VCAM-1 expression in HUVECs via regulation of AP-1 and NF-kappaB activities in a GSH-sensitive manner. Our present study suggested a potential mechanism for arsenite in the induction of vascular inflammation and vascular diseases via modulating the actions of proinflammatory cytokines.


Assuntos
Arsenitos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/biossíntese , Butionina Sulfoximina/farmacologia , Células Cultivadas , Sinergismo Farmacológico , Células Endoteliais/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Genes Reporter , Glutationa/antagonistas & inibidores , Glutationa/metabolismo , Glutationa/fisiologia , Humanos , Immunoblotting , Luciferases/metabolismo , NF-kappa B/antagonistas & inibidores , NF-kappa B/fisiologia , Fator de Transcrição AP-1/antagonistas & inibidores , Fator de Transcrição AP-1/fisiologia
13.
Toxicol Appl Pharmacol ; 208(3): 277-84, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16239170

RESUMO

Epidemiological studies have demonstrated a high association of inorganic arsenic exposure with vascular diseases. Recent research has also linked this vascular damage to impairment of endothelial nitric oxide synthase (eNOS) function by arsenic exposure. However, the role of eNOS in regulating the arsenite-induced vascular dysfunction still remains to be clarified. In our present study, we investigated the effect of arsenite on Akt1 and eNOS and its involvement in cytotoxicity of vascular endothelial cells. Our study demonstrated that arsenite decreased the protein levels of both Akt1 and eNOS accompanied with increased levels of ubiquitination of total cell lysates. We found that inhibition of the ubiquitin-proteasome pathway by MG-132 could partially protect Akt1 and eNOS from degradation by arsenite together with a proportional protection from the arsenite-induced cytoxicity. Moreover, up-regulation of eNOS protein expression significantly attenuated the arsenite-induced cytotoxicity and eNOS activity could be significantly inhibited after incubation with arsenite for 24 h in a cell-free system. Our study indicated that endothelial eNOS activity could be attenuated by arsenite via the ubiquitin-proteasome-mediated degradation of Akt1/eNOS as well as via direct inhibition of eNOS activity. Our study also demonstrated that eNOS actually played a protective role in arsenite-induced cytoxicity. These observations supported the hypothesis that the impairment of eNOS function by arsenite is one of the mechanisms leading to vascular changes and diseases.


Assuntos
Arsenitos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Compostos de Sódio/toxicidade , Androstadienos/administração & dosagem , Animais , Aorta Torácica/patologia , Arsenitos/antagonistas & inibidores , Linhagem Celular , Cromonas/administração & dosagem , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/química , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Leupeptinas/farmacologia , Morfolinas/administração & dosagem , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/administração & dosagem , Fosforilação , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Compostos de Sódio/antagonistas & inibidores , Suínos , Fatores de Tempo , Regulação para Cima , Wortmanina
14.
Chem Res Toxicol ; 17(2): 208-17, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14967008

RESUMO

In this study, we used porcine aortic endothelial cells (PAECs) as an in vitro system to investigate the role of intracellular GSH status in arsenite-induced vascular endothelial damage. Exposure of PAECs to l-buthionine sulfoximine (BSO), an inhibitor of gamma-glutamylcysteine synthetase (gamma-GCS), markedly enhanced the arsenite-induced cytotoxicity. The data implied that intracellular GSH might play an important role in protection of PAECs from arsenite-induced cytotoxicity. Low concentrations of arsenite exposure increased intracellular GSH concentrations, whereas high concentrations of arsenite exposure decreased intracellular GSH concentrations. We further modulated intracellular GSH concentration by using GSH modulators. N-Acetyl cysteine (NAC) and l-cystine (oxidized l-cysteine), by up-regulating intracellular GSH concentrations, were shown to protect PAECs from arsenite-induced cytotoxicity. On the other hand, BSO and monosodium glutamate (MSG), which down-regulated the intracellular GSH concentrations, further potentiated arsenite-induced cytotoxicity. Moreover, exposure of PAECs to NAC alleviated the arsenite-induced JNK/AP-1 activation and apoptosis, whereas exposure of PAECs to BSO enhanced the arsenite-induced JNK/AP-1 activation and apoptosis. These results indicated that an increase in GSH content represented one of the detoxification mechanisms responding to arsenite exposure and probably played critical roles in the regulation of stress-response signaling molecules as well as in protection of PAECs from arsenite attack.


Assuntos
Arsenitos/toxicidade , Endotélio Vascular/efeitos dos fármacos , Glutationa/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Acetilcisteína/farmacologia , Animais , Apoptose , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Citometria de Fluxo , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Transdução de Sinais , Suínos , Fator de Transcrição AP-1/metabolismo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA