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.
J Cell Physiol ; 233(9): 7379-7390, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29575013

RESUMO

Cervical cancer (CC) remains a highly prevalent cancer and mortality globally among women globally. The aim of the present study was to assess the ability of miR-374b to regulate CC cells through JAM-2, whilst exploring whether the underlying mechanism and its relation to the p38/ERK signaling pathway. During the study, microRNA-374b (miR-374b) was observed to have been expressed at a low level among CC tissues. Hence, a series of miR-374b mimics, miR-374b inhibitors, siRNA against JAM-2, SB202190 (an inhibitor for p38), and PD98059 (an inhibitor for ERK) were introduced to treat CC Siha cells and normal cervical Ect1/E6E7 cells. MTT, flow cytometry, scratch test, and transwell assays were applied to determine cell viability, apoptosis, migration, and invasion. The inhibitory role of the p38/ERK signaling pathway was observed in the CC cells treated with miR-374b mimics or siRNA against JAM-2. miR-374b mimic exposure was found to reduce cell viability, migration, and invasion, but induce apoptosis. MiR-374b inhibitor exposure was observed to have induced effects on the CC cells in a contrary manner to those induced by that of the miR-374b mimics. The key findings of the study demonstrated that miR-374b significantly inhibits cell proliferation, migration, and invasion through the blockade of the p38/ERK signaling pathway activation, as well as negatively binding to JAM-2, highlighting its potential as a therapeutic target for CC.


Assuntos
Apoptose/genética , Moléculas de Adesão Celular/metabolismo , Sistema de Sinalização das MAP Quinases , MicroRNAs/metabolismo , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Adulto , Sequência de Bases , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Regulação para Cima/genética
2.
Gynecol Obstet Invest ; 82(2): 188-199, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27553367

RESUMO

BACKGROUND: We hypothesized that apurinic/apyrimidinic endonuclease 1 (APE1) may regulate cell activity via activator protein 1 (AP-1) signaling pathway through its redox function in the development of ovarian cancer. METHODS: Ovarian cancer and paired paracarcinoma tissues were collected for determining APE1 expression with immunohistochemistry. Cell transfection was performed to silence APE1. For mRNA and protein expressions of genes after silencing, real-time reverse transcription polymerase chain reaction and Western blot assay were conducted. Transwell cell proliferation experiment and clone formation assay were conducted for cell proliferation; Transwell cell migration and cell invasion assays were conducted for cell migration and invasion. RESULTS: Most cancer tissues had APE1 cytoplasmic expression and a few had nuclear/cytoplasmic expression while few had only nuclear expression. A significantly higher APE1 expression was identified in cancer tissues than in paracarcinoma tissues and it was correlated with the clinicopathologic features. SKOV3 was used for cell experiments. After silencing of APE1, both mRNA and protein expressions of APE1 and AP-1 signaling-related genes were downregulated, and the cell activity in proliferation, migration and invasion was remarkably suppressed. CONCLUSION: Overexpressed APE1 promotes ovarian cancer growth and metastasis. Downregulated APE1 could suppress cell activity via AP-1 pathway, suggesting that APE1 gene may be a potential therapeutic target for ovarian cancer.


Assuntos
DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Linhagem Celular , Feminino , Humanos , Metástase Neoplásica
3.
Gynecol Obstet Invest ; 82(3): 230-239, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27505004

RESUMO

OBJECTIVE: The study aimed to investigate cyclin-dependent kinase 14 (CDK14) and its co-function with Wnt signaling pathway on cell proliferation, migration and invasion in ovarian cancer. METHODS: CDK14 expressions were detected by quantitative real-time polymerase chain reaction. The expressions c-Myc, cyclinD1, PFTK1, ki67 and OGT were examined by Western blot. MTT assay was applied to observe cell proliferation after transfection of pEGFP-N1/CDK14-siRNA and pEGFP-N1 into SKOV3 cells, and scratch test and Transwell assay to observe invasion and migration ability. Transfected tumor model in nude mice was established. RESULTS: CDK14 was upregulated in the ovarian cancer tissues and cell lines (both p < 0.05). Expressions of downstream molecules in Wnt signaling pathway as well as the proliferation, invasion and migration ability of the SKOV3 cells were reduced when CDK14 was inhibited (all p < 0.05). The expression of ß-catenin in the nucleus was also decreased when CDK14 was inhibited (p < 0.05). In the transfected tumor model of nude mice, the results showed, compared with the pEGFP-N1 group and blank control group, that the expressions of c-Myc, cyclinD1, PFTK1, ki67 and OGT in the pEGFP-N1/CDK14-siRNA group in the transplantation tumor tissues decreased significantly (all p < 0.05). CONCLUSION: CDK14 suppression-mediated Wnt signaling pathway can inhibit cell proliferation, invasion and migration in ovarian cancer.


Assuntos
Movimento Celular , Proliferação de Células , Quinases Ciclina-Dependentes/fisiologia , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/patologia , Via de Sinalização Wnt/fisiologia , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Núcleo Celular , Feminino , Humanos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Transplante de Neoplasias , RNA Interferente Pequeno , Transfecção
4.
Mol Med Rep ; 13(4): 3161-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26935265

RESUMO

Nitidine chloride (NC) has been demonstrated to exert anti-tumor effects on various types of tumor. However, no studies have investigated the anti­metastatic effect of NC on ovarian cancer cells, and the underlying mechanisms have not yet been clearly established. The present study aimed to determine the effect of NC on the migration and invasion of ovarian cancer cells. Cell viability and proliferation of ovarian cancer cells were assessed by MTT assay. A scratch wound healing assay and Transwell assays were performed to detect migration and invasion of cells, respectively. The expression levels of matrix metalloproteinase (MMP)­2 and 9 were detected at the mRNA and protein level following stimulation with NC. Subsequently, the expression of mitogen­activated protein kinases was detected by western blot analysis. Finally, an inhibitor of extracellular signal­regulated kinase (ERK) was applied to investigate the effect of NC on the expression of MMP­2/9 as well as the migration and invasion of cells. It was found that NC suppressed the proliferation, migration and invasion of A2780 ovarian cancer cells. NC downregulated MMP­2 and MMP­9 in a dose­ and time­dependent manner. In addition, NC was also able to downregulate phosphorylation of ERK. Furthermore, by applying an ERK inhibitor, U0126, the effect of NC on the expression of MMP-2/9 and inhibition of cell migration and invasion was verified. Taken together, these results demonstrated that NC inhibited the migration and invasion of ovarian cancer cells via the ERK signaling pathway.


Assuntos
Benzofenantridinas/toxicidade , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Transdução de Sinais/efeitos dos fármacos , Benzofenantridinas/química , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fosforilação/efeitos dos fármacos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real
5.
Tumour Biol ; 37(8): 10161-75, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26831658

RESUMO

This study is designated to investigate the roles of cyclin Y (CCNY) and Wnt signaling pathway in regulating ovarian cancer (OC) cell proliferation, migration, and invasion. Quantitative real-time PCR (qRT-PCR), Western blot, MTT assay, cell scratch, and transwell test were used in our study, and transplanted tumor model was constructed on nude mice. C-Myc, cyclin D1, PFTK1, ki67, OGT, and ß-catenin protein expressions in tumor tissues were detected. CCNY was significantly upregulated in OC cell lines and tissues (both P < 0.05); significant association was observed between CCNY expression and clinicopathological stage, lymph node metastasis (LNM) (P < 0.05); and the CCNY expression in stages III to IV was higher than that in stages I to II, and patients with LNM had higher CCNY expression when compared with those in patients without LNM (P < 0.05); expressions of c-Myc, cyclin D, PFTK1, ki67, and OGT were upregulated in OC tissues compared with ovarian benign tissues, suggesting that these expressions were significantly different between the two groups (P < 0.05); CCNY significantly exacerbated proliferation, migration, and invasion of A2780 cells; c-Myc and cyclin D1 protein expressions increased as the expression of CCNY increased (P < 0.001); ß-catenin expressions in A2780 cells with over-expression of CCNY were significantly increased in the nucleus, but significantly decreased in the cytoplasm (both P < 0.05); high expressions of CCNY exacerbated the proliferation of A2780 cells in nude mice and significantly increased c-Myc, cyclin D1, PFTK1, ki67, and OGT protein expressions in tumor tissues which were transplanted into nude mice (P < 0.01). CCNY might exacerbate the proliferation, migration, and invasion of OC cells via activating the Wnt signaling pathway. Thus, this study provides a theoretical foundation for the development of therapeutic drugs that are able to cure OC by targeting CCNY.


Assuntos
Ciclinas/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias Ovarianas/patologia , Via de Sinalização Wnt , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma/secundário , Adulto , Idoso , Animais , Diferenciação Celular , Divisão Celular , Linhagem Celular Tumoral , Movimento Celular , Núcleo Celular/metabolismo , Proliferação de Células , Ciclinas/biossíntese , Ciclinas/genética , Citoplasma/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Metástase Linfática , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/genética , Distribuição Aleatória , Proteínas Recombinantes de Fusão/metabolismo
6.
Mol Med Rep ; 12(1): 1125-30, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25776533

RESUMO

Markers associated with diagnosis, presentation and potential therapeutic targets have received widespread attention in ovarian cancer research in the past few years. However, the majority of these markers have been investigated individually, and the changes in expression and the association between them are rarely documented. Next­generation sequencing, also termed RNA-seq when the sequencing targets are cDNAs, can provide a whole blueprint of the transcriptome of a specific tissue. In the present study, RNA-seq data of human ovarian cancer samples were used to verify the expression of known markers and to identify the association between them. A total of 563 markers associated with ovarian cancer were retrieved from the database of the National Center of Biotechnology Information, and used as the target markers. The transcriptome of the ovarian tissue of four different tumors, containing tumor presentation and recurrence stages, were sequenced using the Illumina GAII platform. Approximately 85.97% markers were expressed of the total 563 markers, and the majority of them were involved in pathways associated with cancer, signaling and infection. In total, 85 markers were found to be aberrantly expressed in tumor cells from patients with ovarian cancer who had recurrences, including 33 upregulated markers at the recurrence stage. Therefore, they may have roles ovarian tumor due to their aberrant expression. Differentially expressed markers and the associations between them can be assessed by examining the RNA-seq data. These findings may provide novel information for further studies on ovarian cancer.


Assuntos
Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Recidiva Local de Neoplasia/genética , Neoplasias Ovarianas/genética , Transcriptoma , Biologia Computacional , Bases de Dados Genéticas , Feminino , Redes Reguladoras de Genes , Marcadores Genéticos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/patologia , Estadiamento de Neoplasias , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/patologia , Transdução de Sinais
7.
Oncol Rep ; 33(2): 861-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25502786

RESUMO

Ampelopsin has displayed anticancer activity in several types of cancers. However, no evidence has been reported for the direct effect of ampelopsin on ovarian cancer cell migration and invasion, and the underling mechanisms have not yet been clearly established. The aim of the present study was to investigate the influence of ampelopsin on the migration and invasion of ovarian cancer. Proliferation and viability of the ovarian cancer cells were detected by MTT assay. Migration and invasion of the cells were detected, respectively, by scratch wound healing assay and Transwell assay. The expression levels of epithelial-to-mesenchymal transition (EMT) markers were detected at the protein level after stimulation with ampelopsin. Then, the expression levels of NF-κB and p-IκBα were detected with western blot analysis. Meanwhile, an inhibitor of NF-κB was used to investigate the effect of ampelopsin. Finally, the expression of Snail was also detected. Proliferation, migration and invasion of the A2780 cells were all inhibited following the application of ampelopsin. Ampelopsin upregulated E-cadherin and downregulated N-cadherin and vimentin in a concentration- and time-dependent manner. Ampelopsin also exerted its ability to suppress the nuclear translocation of the NF-κB pathway. Administration of the inhibitor BAY11-7082 confirmed the roles of NF-κB in the expression of EMT markers and its transcription factor. These results demonstrated that ampelopsin inhibited EMT and reduced the invasion of ovarian cancer cells via the NF-κB/Snail pathway.


Assuntos
Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Flavonoides/farmacologia , Neoplasias Ovarianas/metabolismo , Transporte Ativo do Núcleo Celular , Antineoplásicos/farmacologia , Caderinas/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células , Sobrevivência Celular , Regulação para Baixo , Feminino , Humanos , Proteínas I-kappa B/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Metástase Neoplásica , Nitrilas/química , Sulfonas/química , Fatores de Tempo , Regulação para Cima , Vimentina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...