Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Lab Invest ; 103(8): 100156, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37119854

RESUMO

Paraneoplastic nephrotic syndrome (PNS) is a complication seen in cancer patients. Ultrastructural examination shows the accumulation of proteins and the presence of foot process (FP) effacement in the glomeruli of PNS patients. Previously, we reported that orthotopic xenografts of Lewis lung carcinoma 1 in C57BL/6 mice caused them to develop lung cancer with albuminuria. This implies that these mice can be used as a model of human disease and suggests that Lewis lung carcinoma 1 cell-secreted proteins (LCSePs) contain nephrotoxic molecules and cause inflammation in renal cells. As podocyte effacement was present in glomeruli in this model, such podocyte injury may be attributable to either soluble LCSeP or LCSeP deposits triggering pathological progression. LCSePs in conditioned media was concentrated for nephrotoxicity testing. Integrin-focal adhesion kinase (FAK) signaling and inflammatory responses were evaluated in podocytes either exposed to soluble LCSePs or seeded onto substrates with immobilized LCSePs. FAK phosphorylation and interleukin-6 expression were higher in podocytes attached to LCSePs substrates than in those exposed to soluble LCSePs. Notably, LCSeP-based haptotaxis gave rise to altered signaling in podocytes. When podocytes were stimulated by immobilized LCSePs, FAK accumulated at focal adhesions, synaptopodin dissociated from F-actin, and disrupting the interactions between synaptopodin and α-actinin was observed. When FAK was inhibited by PF-573228 in immobilized LCSePs, the association between synaptopodin and α-actinin was observed in the podocytes. The association of synaptopodin and α-actinin with F-actin allowed FP stretching, establishing a functional glomerular filtration barrier. Therefore, in this mouse model of lung cancer, FAK signaling prompts podocyte FP effacement and proteinuria, indicative of PNS.


Assuntos
Carcinoma Pulmonar de Lewis , Neoplasias Pulmonares , Podócitos , Camundongos , Humanos , Animais , Actinas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Actinina/metabolismo , Carcinoma Pulmonar de Lewis/metabolismo , Carcinoma Pulmonar de Lewis/patologia , Camundongos Endogâmicos C57BL , Proteinúria/metabolismo , Podócitos/metabolismo , Neoplasias Pulmonares/metabolismo
2.
Biomed Pharmacother ; 124: 109892, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31986415

RESUMO

AIMS: Macrophage migration inhibitory factor (MIF) is found to increase in proliferative glomerulonephritis. MIF binds to the MIF receptor (CD74) that activates MAP kinase (ERK and p38). Integrins and cyclinD1 regulate cell proliferation, differentiation and adhesion. This study evaluates whether MIF can regulate integrin-ß1/cyclin D1 expression and cell adhesion of podocytes. MAIN METHODS: Expression of integrin-ß1 mRNA/protein and cyclin D1 mRNA under stimulation of MIF was evaluated by real-time PCR and Western blotting. MIF receptor (CD74) and MAP kinase under MIF treatment were examined to determine which pathway regulated integrin-ß1 and cyclin D1 expression. Cell adhesion was evaluated under MIF treatment and/or anti-integrin-ß1 antibody by cell adhesion assay. KEY FINDINGS: Protein levels of integrin-ß1 were up-regulated under MIF treatment in a dosage-dependent manner. CD74 protein levels were not changed after MIF treatment. Integrin-ß1 and cyclin D1 mRNA levels were up-regulated after MIF 100 ng/ml treatment. ERK inhibitor U0126 reduced MIF-induced the increase in integrin-ß1 mRNA and protein expression following MIF stimulation. However, p38 inhibitor SB 203580 did not inhibit MIF-induced increase in integrin-ß1 mRNA and protein expression following MIF stimulation. MIF-induced increase in cyclin D1 mRNA level also was inhibited only by U0126 following MIF stimulation. Podocyte adhesion was increased after MIF treatment, but, anti-integrin-ß1 antibody decreased MIF-enhanced podocyte adhesion. SIGNIFICANCE: MIF increases integrin-ß1 and cyclin D1 expression through the ERK pathway in podocytes, and the up-regulated expression of integrin-ß1 increases podocyte adhesion. These results provide further understanding for the role of MIF in developing proliferative glomerulonephritis.


Assuntos
Ciclina D1/genética , Integrina beta1/genética , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Podócitos/metabolismo , Animais , Adesão Celular/genética , Proliferação de Células/fisiologia , Células Cultivadas , Glomerulonefrite/fisiopatologia , Sistema de Sinalização das MAP Quinases/fisiologia , RNA Mensageiro/genética , Ratos , Transdução de Sinais/fisiologia , Regulação para Cima
3.
J Hazard Mater ; 170(1): 351-6, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19477068

RESUMO

Polychlorinated dibenzo-p-dioxins (PCDDs) and dibenzofurans (PCDFs), considered as endocrine disruptors, tend to accumulate in fatty tissues. Dioxin-responsive element chemical activated luciferase gene expression assay (DRE-luciferase assay) has been recognized as a semi-quantitative method for screening dioxins for its fast and low-cost as compared with HRGC/HRMS. However, some problems with the bioassay, including specificity, detection variation resulted from different cleanup strategies, and uncertainty of false-negative or false-positive results, remain to be overcome. Cadmium is a prevalent environmental contaminant around the world. This study was aimed to examine the effects of cadmium on the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced activation of aryl hydrocarbon receptor (AhR)-mediated gene expression in human hepatoma cells (Huh7-DRE-Luc cells and Huh7 cells). Ethoxyresorufin-O-deethylase (EROD) and DRE-luciferase assay were employed to determine the enzyme activity of cytochrome P450 1A1 (CYP1A1) and activation of AhR, respectively. The results showed that Cd(2+) levels significantly inhibited the induction of TCDD-induced CYP1A1 and DRE luciferase activation in hepatoma cells. The 50% inhibited concentrations (IC(50)) of CdCl(2) were 0.414 microM (95% confidence interval (C.I.): 0.230-0.602 microM) in Huh7-DRE-Luc cells and 23.2 microM (95% C.I.: 21.7-25.4 microM) in Huh7 cells. Accordingly, prevention of interference with non-dioxin-like compounds in a DRE-luciferase assay is of great importance in an extensive cleanup procedure.


Assuntos
Cloreto de Cádmio/farmacologia , Dibenzodioxinas Policloradas/farmacologia , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Ativação Transcricional/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Citocromo P-450 CYP1A1/análise , Citocromo P-450 CYP1A1/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Disruptores Endócrinos/farmacologia , Poluentes Ambientais , Humanos , Concentração Inibidora 50 , Receptores de Hidrocarboneto Arílico/metabolismo , Células Tumorais Cultivadas
4.
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
5.
J Hazard Mater ; 149(1): 234-7, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17707585

RESUMO

Dioxin-responsive element-mediated chemical activated luciferase expression (DRE-CALUX) is one of alternative bioassays for the determination of dioxin levels. We have previously established a DRE-CALUX cell line, Huh7-DRE-Luc, by using stable transfection of Huh-7 cells with a reporter plasmid (4xDRE-TATA-Luc) carrying a DRE-driven firefly luciferase gene. It was also shown that arecoline, a major areca nut alkaloid, inhibited the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced cytochrome P450 1A1 (CYP1A1) activation in Huh-7 cells. The TCDD-activated aryl hydrocarbon receptor (AhR) induces the DRE-CALUX activation and CYP1A1 gene expression via binding to DRE in promoter regions of these dioxin-responsive genes. In the present study, the effect of arecoline on the TCDD-induced activation of DRE-CALUX and CYP1A1 enzyme in Huh7-DRE-Luc and Huh-7 cells, respectively, was examined. It was found that arecoline inhibited TCDD-induced CYP1A1 activation and however enhanced TCDD-induced DRE-CALUX activation. This finding indicates the differential effect of arecoline on the endogenous dioxin-responsive CYP1A1 and on a stably transfected DRE-driven reporter in human hepatoma cells. The present study suggests that induction of DRE-CALUX alone does not necessarily parallel with endogenous CYP1A1 gene expression, and that the reporter assay may detect interactions that are not functional in endogenous gene.


Assuntos
Arecolina/farmacologia , Citocromo P-450 CYP1A1/antagonistas & inibidores , Dibenzodioxinas Policloradas , Bioensaio , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Regulação Enzimológica da Expressão Gênica , Genes Reporter , Humanos , Luciferases/genética , Luciferases/metabolismo , Elementos de Resposta
6.
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
7.
J Hazard Mater ; 137(2): 716-22, 2006 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16713074

RESUMO

The aim of this study was to examine the arsenic effect on activation of aryl hydrocarbon receptor (AhR)-mediated gene expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin (2,3,7,8-TCDD) in human hepatoma cells. The human hepatoma Huh7 cells were treated with sodium arsenite (NaAsO2) from 0.5 to 20 microM for 24 h. Our data revealed that NaAsO2 < or = 10 microM caused no significant cytotoxic effect on Huh7 cells (p>0.05). We also established a dioxin-responsive element (DRE)-mediated Chemical Activated LUciferase eXpression (CALUX) cell line, Huh7-DRE-Luc, by stable transfection of Huh7 with a DRE-driven firefly luciferase reporter plasmid (4xDRE-TATA-Luc). Treatments of Huh7-DRE-Luc and Huh7 with NaAsO2 attenuated the 2,3,7,8-TCDD-induced DRE-CALUX and cytochrome P450 1A1 (CYP1A1) activations, respectively, in a dose-dependent manner. We found that the calculated CALUX-toxic equivalent (TEQ) levels induced by cotreatment of NaAsO2 > or = 3.0 microM and 10 nM 2,3,7,8-TCDD were significantly lower than that induced by 2,3,7,8-TCDD alone (p<0.05). In the present study, we demonstrated that arsenic not only inhibited the TCDD-induced CYP1A1 activation but also interfered with DRE-CALUX bioassay in human hepatoma cells. Our finding also suggests that extensive cleanup of sample for removal of any possible interfering factor is critical to guarantee the accuracy of dioxin-TEQ levels using DRE-CALUX bioassay.


Assuntos
Arseniatos/farmacologia , Carcinoma Hepatocelular/enzimologia , Citocromo P-450 CYP1A1/antagonistas & inibidores , Citocromo P-450 CYP1A1/biossíntese , Neoplasias Hepáticas/enzimologia , Dibenzodioxinas Policloradas/farmacologia , Linhagem Celular Tumoral , Poluentes Ambientais/farmacologia , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Dibenzodioxinas Policloradas/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA