Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Med Sci Monit ; 26: e924497, 2020 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-32801292

RESUMO

BACKGROUND The aim of this study was to determine multidetector computed tomography (MDCT) features and tumor markers for differentiating stage I serous borderline ovarian tumors (SBOTs) from stage I serous malignant ovarian tumors (SMOTs). MATERIAL AND METHODS In total, 48 patients with stage I SBOTs and 54 patients with stage I SMOTs who underwent MDCT and tumor markers analysis were analyzed. MDCT features included location, shape, margins, texture, papillary projections, vascular abnormalities, size, and attenuation value. Tumor markers included serum cancer antigen 125 (CA125), carbohydrate antigen 19-9 (CA19-9), carcinoembryonic antigen (CEA), and human epididymis protein 4 (HE4). Parameters of clinical characteristic, MDCT features, and tumor markers were compared using a chi-square test and Mann-Whitney U tests. A binary logistic regression analysis was performed to detect predictors for SMOTs. A receiver operating characteristic (ROC) curve analysis was used to assess the potential diagnostic value of the quantitative parameters. Kappa and intraclass correlation coefficients were used to evaluate interobserver reproducibility for MDCT features. RESULTS Median ages between patients with SBOTs and SMOTs were significantly different. Compared with SBOTs, vascular abnormalities were significantly more common in SMOTs. CA125, HE4, the maximum thickness of the wall, the maximum thickness of the septa, and the maximum diameter of the solid portions were significantly higher in patients with SMOTs. A binary logistic regression analysis revealed that age, vascular abnormalities, and the maximum diameter of the solid portion were independent factors of SMOTs. ROC analysis was used to assess the potential diagnostic value for predicting SMOTs. Moderate or good interobserver reproducibility for MDCT features were identified. CONCLUSIONS Age, vascular abnormalities, and the maximum diameter of the solid portion were independent factors for differentiating SBOTs from SMOTs. The combined analysis of age, vascular abnormalities, and the maximum diameter of the solid portion may allow better differentiation between SBOTs and SMOTs.


Assuntos
Biomarcadores Tumorais/sangue , Diferenciação Celular , Neoplasias Ovarianas/diagnóstico por imagem , Adulto , Idoso , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Curva ROC
2.
Cancer Manag Res ; 13: 329-336, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33488120

RESUMO

OBJECTIVE: To investigate whether multidetector computed tomography (MDCT)-based radiomics features can discriminate between serous borderline ovarian tumors (SBOTs) and serous malignant ovarian tumors (SMOTs). PATIENTS AND METHODS: Eighty patients with SBOTs and 102 patients with SMOTs, confirmed by pathology (training set: n = 127; validation set: n = 55) from December 2017 to June 2020, were enrolled in this study. The interclass correlation coefficient (ICC) and least absolute shrinkage and selection operator (LASSO) regression were applied to select radiomics parameters derived from MDCT images on the arterial phase (AP), venous phase (VP), and equilibrium phase (EP). Receiver operating characteristic (ROC) analysis of each selected parameter was carried out. Heat maps were created to illustrate the distribution of the radiomics parameters. Three models incorporating selected radiomics parameters generated by support vector machine (SVM) classifiers in each phase were analyzed by ROC and compared using the DeLong test. RESULTS: The most predictive features selected by ICC and LASSO regression between SBOTs and SMOTs included 9 radiomics parameters on AP, VP, and EP each. Three models on AP, VP, and EP incorporating the selected features generated by SVM classifiers produced AUCs of 0.80 (accuracy, 0.75; sensitivity, 0.74; specificity, 0.75), 0.86 (accuracy, 0.78; sensitivity, 0.80; specificity, 0.75), and 0.73 (accuracy, 0.69; sensitivity, 0.71; specificity, 0.67), respectively. There were no significant differences in the AUCs among the three models (AP vs. VP, P = 0.199; AP vs. EP, P = 0.260; VP vs. EP, P = 0.793). CONCLUSION: MDCT-based radiomics features could be used as biomarkers for the differentiation of SBOTs and SMOTs.

3.
J Ovarian Res ; 8: 80, 2015 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-26626440

RESUMO

BACKGROUND: Recent studies have shown that microRNAs may regulate the ABCB1 gene (ATP-binding cassette, sub-family B [MDR/TAP], member 1). Computational programs have predicted that the 3'-untranslated region (3'-UTR) of ABCB1 contains a potential miRNA-binding site for miR-186. Here, we investigated the role of miR-186 in sensitizing ovarian cancer cells to paclitaxel and cisplatin. RESULTS: Human ovarian carcinoma cell lines OVCAR3, A2780, A2780/DDP, and A2780/Taxol were exposed to paclitaxel or cisplatin with or without miR-186 transfection, and cell viability was determined by MTT assay. Reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis were used to assess the MDR1, GST-π, and MRP1 expression levels. Dual-luciferase reporter assay was used to reveal the correlation between miR-186 and ABCB1. Lower miR-186 while higher MDR1 and GST-π mRNA expression levels were found in the A2780/Taxol and A2780/DDP cells than in the A2780 cells. After miR-186 transfection, all the cell lines showed increased sensitivity to paclitaxel and cisplatin. MiR-186 transfection induced apoptosis while anti-miR-186 transfection reduced apoptosis. The dual-luciferase reporter assay verified that that miR-186 combined with the 3'-untranslated region (UTR) of ABCB1. MDR1 and GST-π mRNA and protein expression levels were downregulated after transfection with miR-186 but upregulated following anti-miR-186 transfection compared to the mock and negative control cancer cells; however, the MRP1 expression levels did not significantly differ among the groups. CONCLUSION: Our results are the first to demonstrate that miR-186 may sensitize ovarian cancer cell to paclitaxel and cisplatin by targeting ABCB1 and modulating the expression of GST-π.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , MicroRNAs/fisiologia , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/uso terapêutico , Subfamília B de Transportador de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/fisiologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Glutationa Transferase/metabolismo , Humanos , Luciferases/antagonistas & inibidores , MicroRNAs/metabolismo , Transfecção
4.
Drug Des Devel Ther ; 9: 5225-35, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26396496

RESUMO

BACKGROUND: Accumulating studies reveal that aberrant microRNA (miRNA) expression can affect the development of chemotherapy drug resistance by modulating the expression of relevant target proteins. The aim of this study was to investigate the role of miR-133b in the development of drug resistance in ovarian cancer cells. METHODS: We examined the levels of miR-133b expression in ovarian carcinoma tissues and the human ovarian carcinoma cell lines (A2780, A2780/DDP and A2780/Taxol, respectively). We determined the cell viability of these cell lines treated with cisplatin or paclitaxel in the presence or absence of miR-133b or anti-miR-133b transfection using the MTT assay. Reverse transcription polymerase chain reaction and Western blotting were used to assess the mRNA and protein expression levels of two drug-resistance-related genes: glutathione S-transferase (GST)-π and multidrug resistance protein 1 (MDR1). The dual-luciferase reporter assay was used to detect the promoter activity of GST-π in the presence and absence of miR-133b. RESULTS: The expression of miR-133b was significantly lower in primary resistant ovarian carcinomas than in the chemotherapy-sensitive carcinomas (P<0.05), and the same results were found in primary resistant ovarian cell lines (A2780/Taxol and A2780/DDP) compared to the chemotherapy-sensitive cell line (A2780; P<0.05). Following miR-133b transfection, four cell lines showed increased sensitivity to paclitaxel and cisplatin, while anti-miR-133b transfection reduced cell sensitivity to paclitaxel and cisplatin. Dual-luciferase reporter assay showed that miR-133b interacted with the 3'-untranslated region of GST-π. Compared to controls, the mRNA and protein levels of MDR1 and GST-π were downregulated after miR-133b transfection and upregulated after anti-miR-133b transfection. CONCLUSION: MicroRNA-133b may reduce ovarian cancer drug resistance by silencing the expression of the drug-resistance-related proteins, GST-π and MDR1. In future, the combination of miR-133b with chemotherapy agents may prevent the development of drug resistance in ovarian cancers.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Cistadenocarcinoma Seroso/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Glutationa S-Transferase pi/metabolismo , MicroRNAs/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/farmacologia , Regiões 3' não Traduzidas , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cistadenocarcinoma Seroso/enzimologia , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glutationa S-Transferase pi/genética , Humanos , MicroRNAs/genética , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção
5.
Oncol Rep ; 25(3): 781-8, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21165580

RESUMO

Tanshinone IIA is known to induce apoptosis in several types of cancer cells. However, little is known about its activity in chemoresistant cells. The aim of this study was to investigate the anticancer properties of tanshinone IIA in cisplatin-resistant human ovarian cancer COC1/DDP cells in vitro. We used a variety of methods to measure cell viability, the resistance index (RI) of cisplatin, cellular apoptosis, p38 mitogen-activated protein kinase (MAPK) expression and phosphorylation, and the mRNA expression of several genes implicated in drug resistance including survivin, Caspase-3, excision repair cross-complementing gene 1 (ERCC1), multidrug resistance (MDR), lung resistance protein (LRP) and glutathione-S-transferase-π (GST-π). We found that tanshinone IIA time- and dose-dependently inhibited the proliferation of COC1/DDP cells and caused significant apoptosis. Western blotting revealed that tanshinone IIA also increased phospho-p38 MAPK in a time- and dose-dependent manner. After treatment by tanshinone IIA for 48 h, the RI of cisplatin and the mRNA expression of survivin, ERCC1 and LRP were all significantly decreased. Furthermore, blockade of p38 signal transduction decreased apoptotic cell rates and dramatically elevated the mRNA expression of the survivin, ERCC1 and LRP genes. We therefore conclude that tanshinone IIA induces apoptosis and reduces cisplatin resistance in COC1/DDP cells and thus causes significant growth inhibitory effects. This mechanism appears to involve p38-mediated downregulation of survivin, ERCC1 and LRP mRNA expression.


Assuntos
Abietanos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma/patologia , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Neoplasias Ovarianas/patologia , Partículas de Ribonucleoproteínas em Forma de Abóbada/genética , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Abietanos/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma/tratamento farmacológico , Carcinoma/genética , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Endonucleases/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Transdução de Sinais/efeitos dos fármacos , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA