Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 79(13): 3185-3191, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31061066

RESUMO

Chemotherapeutic regimens for ovarian cancer often include the use of DNA interstrand crosslink-inducing agents (e.g., platinum drugs) or DNA double-strand break-inducing agents. Unfortunately, the majority of patients fail to maintain a durable response to treatment, in part, due to drug resistance, contributing to a poor survival rate. In this study, we report that cisplatin sensitivity can be restored in cisplatin-resistant ovarian cancer cells by targeting the chromatin-associated high-mobility group box 3 (HMGB3) protein. HMGB proteins have been implicated in the pathogenesis and prognosis of ovarian cancer, and HMGB3 is often upregulated in cancer cells, making it a potential selective target for therapeutic intervention. Depletion of HMGB3 in cisplatin-sensitive and cisplatin-resistant cells resulted in transcriptional downregulation of the kinases ATR and CHK1, which attenuated the ATR/CHK1/p-CHK1 DNA damage signaling pathway. HMGB3 was associated with the promoter regions of ATR and CHK1, suggesting a new role for HMGB3 in transcriptional regulation. Furthermore, HMGB3 depletion significantly increased apoptosis in cisplatin-resistant A2780/CP70 cells after cisplatin treatment. Taken together, our results indicate that targeted depletion of HMGB3 attenuates cisplatin resistance in human ovarian cancer cells, increasing tumor cell sensitivity to platinum drugs. SIGNIFICANCE: This study shows that targeting HMGB3 is a potential therapeutic strategy to overcome chemoresistance in ovarian cancer.


Assuntos
Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteína HMGB3/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Antineoplásicos/farmacologia , Apoptose , Proliferação de Células , Feminino , Proteína HMGB3/antagonistas & inibidores , Proteína HMGB3/genética , Humanos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/genética , Células Tumorais Cultivadas
2.
Chem Biol Interact ; 300: 73-81, 2019 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-30639441

RESUMO

Accumulating evidence has shown that altered expression of microRNA-532-5p (miR-532-5p) is involved in the development and progression of various cancers. However, little is known about the role of miR-532-5p in bladder cancer. In this study, we aimed to investigate the expression, biological function, and regulatory mechanism of miR-532-5p in bladder cancer. Herein, we found that miR-532-5p expression was frequently downregulated in bladder cancer tissues and cell lines compared with normal controls. Functional experiments showed that overexpression of miR-532-5p inhibited the proliferation and invasion of bladder cancer cells, whereas inhibition of miR-532-5p showed opposite effects. Interestingly, bioinformatics analysis predicted high-mobility group protein B3 (HMGB3) as a potential target gene of miR-532-5p. Further experiments showed that miR-532-5p directly targeted the 3'-UTR of HMGB3 and negatively regulated its expression in bladder cancer cells. Moreover, HMGB3 expression was upregulated in bladder cancer tissues and showed inverse correlation with miR-532-5p expression. Notably, miR-532-5p regulated the nuclear expression of ß-catenin and activation of Wnt/ß-catenin signaling in bladder cancer cells. However, restoration of HMGB3 expression partially reversed the antitumor effect of miR-532-5p overexpression, while knockdown of HMGB3 partially abrogated the oncogenic effect of miR-532-5p inhibition. Taken together, our results demonstrated that miR-532-5p inhibited the proliferation and invasion of bladder cancer cells by targeting HMGB3 and downregulating Wnt/ß-catenin signaling, suggesting a tumor suppressive role of miR-532-5p in bladder cancer. Our study highlights an importance of the miR-532-5p/HMGB3 axis in bladder cancer and suggests that targeting miR-532-5p/HMGB3 may have potential applications for development of bladder cancer therapy.


Assuntos
Proliferação de Células , Proteína HMGB3/metabolismo , MicroRNAs/metabolismo , Via de Sinalização Wnt , Regiões 3' não Traduzidas , Antagomirs/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Movimento Celular , Regulação para Baixo , Proteína HMGB3/antagonistas & inibidores , Proteína HMGB3/genética , Humanos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Alinhamento de Sequência , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia
3.
World J Gastroenterol ; 24(32): 3650-3662, 2018 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-30166860

RESUMO

AIM: To explore the relationship between dynamic expression of high mobility group box-3 (HMGB3) and malignant transformation of hepatocytes. METHODS: Expression of HMGB family proteins were observed in rat hepatocarcinogenesis models induced with 2-acetylaminofluorene. Alterations of HMGB3 were analyzed at the mRNA level by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and at the protein level by immunohistochemistry or Western blotting. HMGB3 in human liver cancer tissues were evaluated using bioinformatics databases from GEO, TCGA, and Oncomine. A specific HMGB3-shRNA was used to knock down HMGB3 expression in order to investigate its effects on proliferation and cell cycle in vitro and in vivo. RESULTS: Elevated HMGB3 levels were first reported in hepatocarcinogenesis, with increasing expression from normal liver to cancer. Bioinformatic databases showed that HMGB3 expression in hepatocellular carcinoma tissues was significantly higher than that in normal liver tissues. Higher HMGB3 expression was discovered in liver cancer cells compared with LO2 cells in vitro. According to gene set enrichment analysis, HMGB3 mRNA levels were correlated with cell cycle and DNA replication pathways. Knocking down HMGB3 by specific shRNA significantly inhibited proliferation of HepG2 cells by cell cycle arrest and downregulating DNA replication related genes (cyclin B1, FEN1, and PCNA) at the mRNA and protein level. Furthermore, silencing HMGB3 significantly inhibited xenograft tumor growth (measured by Ki67) in vivo. CONCLUSION: HMGB3 is involved in malignant transformation of hepatocytes and could be a useful biomarker for diagnosis and a potential target for therapy of liver cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/patologia , Proteína HMGB3/metabolismo , Hepatócitos/patologia , Neoplasias Hepáticas/patologia , 2-Acetilaminofluoreno/toxicidade , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Biologia Computacional , Conjuntos de Dados como Assunto , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Proteína HMGB3/antagonistas & inibidores , Proteína HMGB3/genética , Humanos , Fígado/patologia , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Hepáticas Experimentais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...