Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Cell Physiol ; 233(10): 6759-6767, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29667764

RESUMO

Glioblastoma pathogenesis is related to multiple processes that affected by dozens of regulatory factors, but the potential underlying factors regulating glioblastoma progression remains unclear. The goal of this research was to determine how the ribonucleotide reductase M2 subunit (RRM2) influenced proliferation, invasion, migration, and apoptosis of human glioblastoma cells. The level of proliferation of human glioblastoma cells was measured through CCK8, colony formation assay and immunofluorescence stains. Flow cytometry (FCM), wound healing, and transwell assays were conducted to detect cell apoptosis, migration, and invasion. Apoptotic level of cells and invasion-related expression of protein were measured by Western blot. Xenograft tumor model was established to confirm effect of RRM2 on the proliferation of human glioblastoma cells in vivo. Silencing RRM2 inhibited proliferation, invasion, and migration of glioblastoma cells whereas enhanced apoptosis rate. Overexpressing RRM2 promoted proliferation, migration and invasion but suppressed apoptosis. In vivo, Overexpressing RRM2 accelerated the tumor growth in glioblastoma cells. The present study illustrated that RRM2 was overexpressed in human glioblastoma cells. RRM2 promoted proliferation, migration, and invasion but inhibited apoptosis of human glioblastoma cells.


Assuntos
Apoptose/genética , Proliferação de Células/genética , Glioblastoma/genética , Ribonucleosídeo Difosfato Redutase/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/patologia , Humanos , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Arch Gynecol Obstet ; 291(6): 1361-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25424898

RESUMO

PURPOSE: The purpose of our study was to investigate the therapeutic potential of Celecoxib for epithelial ovarian cancer, especially on cellular morphological changes, proliferation invasion and epithelial-mesenchymal transition (EMT). METHOD: The MTT and transwell assays were performed to evaluate the effect of Celecoxib on proliferation and invasion ability of ovarian cancer cell lines, respectively. Western blot was carried out to detect the expression of epithelial phenotypes, E-cadherin and Keratin, and mesenchymal phenotypes, N-cadherin and Vimentin, as well as p-AKT, p-ERK and ZEB1. ZEB1 small-interfering RNA (siRNA) was used to downregulate the expression of ZEB1 to further inquiring into the downstream of Celecoxib-induced EMT. RESULTS: Cellular morphological assessment revealed that both A2780 and SKOV3 cells gradually appeared in the morphology of mesenchymal cells after Celecoxib treatment. The MTT assay demonstrated that celecoxib had no effect on cell proliferation. Transwell assay showed that Celecoxib significantly increased the cell invasion ability. Western blot data proved that the expression of E-cadherin and keratin was elevated, whereas the expression of N-cadherin and Vimentin was decreased in a dose-dependent manner compared with the untreated cells, the expression of p-AKT, p-ERK and ZEB1 was also obviously elevated. However, ZEB1 siRNA reversed Celecoxib-induced E-cadherin expression and N-cadherin expression, as well as cellular invasiveness. CONCLUSION: Our results indicated that Celecoxib might induce EMT and increase cellular invasiveness in ovarian cancer cells in vitro, which also implied that it needed a comprehensive evaluation in preclinical researches before introducing Celecoxib into the clinical regimen.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Fatores de Transcrição/genética , Caderinas/metabolismo , Carcinoma Epitelial do Ovário , Celecoxib , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo , Feminino , Humanos , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , RNA Interferente Pequeno/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco
3.
Front Oncol ; 10: 303, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32211330

RESUMO

Rigorous molecular characterization of biological systems has uncovered a variety of gene variations underlying normal and disease states and a remarkable complexity in the forms of RNA transcripts that exist. A recent concept, competitive endogenous RNA, suggests that some non-coding RNAs can bind to miRNAs to modulate their role in gene expression. Here, we used several platforms, integrating mRNA, non-coding RNAs and protein data to generate an RNA-protein network that may be dysregulated in human glioblastoma multiforme (GBM). Publicly available microarray data for mRNA and miRNA were used to identify differentially expressed miRNAs and mRNAs in GBM relative to non-neoplastic tissue samples. Target miRNAs were further selected based on their prognostic significance, and the intersection of their target gene set with the differentially expressed gene set in Venn diagrams. Two miRNAs, miR-637 and miR-196a-5p, were associated with poor and better prognosis, respectively, in GBM patients. Non-coding RNAs, ENSG00000203739/ENSG00000271646 and TPTEP1, were predicted to be miRNA target genes for miR-637 and miR-196a-5p and positively correlated with the selected mRNA, CYBRD1 and RUFY2. A local protein interaction network was constructed using these two mRNAs. Predictions based on the ENSG00000203739/ENSG00000271646-miR-637-CYBRD1 and TPTEP1-miR-196a-5p-RUFY2 regulation axes indicated that the two proteins may act as an oncogene and tumor suppressor, respectively, in the development of GBM. These results highlight competitive endogenous RNA networks as alternative molecular therapeutic targets in the treatment of the disease.

4.
Cell Signal ; 62: 109341, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31176022

RESUMO

BACKGROUND: Recently, long intergenic non-coding RNA 01296 (LINC01296) has been demonstrated to regulate the initiation and progression of several cancers, but the functions of LINC01296 in ovarian cancer still remain unclear. The objective of our study was to determine the expression, biological roles, and clinical significance of LINC01296 in ovarian cancer. METHODS: LINC01296 expression was measured in ovarian cancer tissues or cell lines. Next, the relationships between LINC01296 levels and the clinical factors of ovarian cancer, such as progression-free survival and overall survival were analyzed. Additionally, cell proliferation, migration and invasion capacities, apoptosis, cell cycle distribution were investigated after silencing of LINC01296. To confirm whether LINC01296 mediates EMT initiation in ovarian cancer cells, the effect of LINC01296 silence on E-cadherin, N-cadherin and vimentin was assessed in SKOV3 and OVCAR3 cells. RESULTS: We found that LINC01296 was over-expressed in ovarian cancer tissues and cell lines, when comparing with adjacent normal tissue samples and normal cells. Higher LINC01296 expression was significantly correlated with shorter progression-free survival and overall survival. For the functional experiments, knockdown of LINC01296 suppressed cell proliferation, inhibited colony formation ability, abrogated cell migration and invasion potential, and enhanced cell apoptosis. Cell cycle analysis suggested that LINC01296 positively regulated cell cycle progression in ovarian cancer cells. Moreover, western blotting analysis displayed that knockdown of LINC01296 significantly increased E-cadherin, but reduced N-cadherin and vimentin expressions in SKOV3 and OVCAR3 cells, compared with no-transfection cells. CONCLUSIONS: LINC01296 plays an important role in promoting the progression of ovarian cancer. Over-expression of LINC01296 might function as an indicator for diagnosis and prognosis of ovarian cancer patients.


Assuntos
Proliferação de Células/genética , Invasividade Neoplásica/genética , Neoplasias Ovarianas/genética , RNA Longo não Codificante/genética , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Progressão da Doença , Intervalo Livre de Doença , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Humanos , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/patologia , Prognóstico , RNA Longo não Codificante/antagonistas & inibidores
5.
Virus Res ; 250: 37-42, 2018 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-29627479

RESUMO

Classical swine fever virus (CSFV) causes a highly lethal disease in pigs, which is characterized by immunosuppression. Leukopenia is known to be a possible mechanism of immunosuppression during CSFV infection. As a new and specialized form of cell death, pyroptosis is the key response of the innate immune system to pathogens, and is widely involved in the occurrence and development of infectious diseases. However, the relationship between CSFV and pyroptosis has not been explored. In this study, we investigated the occurrence of pyroptosis in pigs following CSFV infection. According to qRT-PCR assay results, the prevalence of this virus in peripheral lymphoid organs (tonsils, lymph nodes, and spleen) was much higher than that in other organs. Severe bleeding, necrosis, and a significant reduction in lymphocytes were found in the peripheral lymphoid organs of CSFV-infected pigs based on histological examination. In-depth studies showed that an increased ratio of deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)-positive cells were present in the peripheral lymphoid organs of the CSFV-infected group according to immunohistochemistry. Meanwhile, the p10 subunit and activity of caspase-1, which is a regulator of pyroptosis, the N-terminal domain of gasdermin D, which is an executor of pyroptosis, and the cleavage and secretion of IL-1b, which is a product of pyroptosis were increased in the peripheral lymphoid organs of the CSFV-infected group. Together, these results demonstrated that pyroptosis is involved in CSFV-induced cell death in vivo, which provides a new understanding of the mechanism associated with lymphocyte depletion and immunosuppression in pigs infected with this virus.


Assuntos
Vírus da Febre Suína Clássica/patogenicidade , Peste Suína Clássica/patologia , Leucopenia/veterinária , Linfonodos/virologia , Piroptose , Animais , Caspase 1/metabolismo , Peste Suína Clássica/imunologia , Hospedeiro Imunocomprometido , Interleucina-1beta/imunologia , Leucócitos Mononucleares/virologia , Leucopenia/imunologia , Leucopenia/virologia , Linfonodos/citologia , Suínos
6.
Oncol Lett ; 14(4): 4689-4693, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28943963

RESUMO

This study examined the relationship between the expression of p16, a tumor suppressor gene, and lymph node metastasis, as well as patient prognosis, in cases with ovarian cancer. SKOV-3, an ovarian cancer cell line, and IOSE80, a normal human ovarian cell line, were selected for testing. Western blot analysis was used to detect the p16 expression in ovarian cell culture samples. In the study, 20 cases with normal ovarian tissue and 64 cases with ovarian cancer tissue, including 38 cases with lymph node metastasis and 26 cases without lymph node metastasis, were also selected for testing. Immunohistochemical techniques were used to detect the expression of p16 protein in ovarian tissue samples. The influence of p16 protein on SKOV-3 cell invasion ability was studied using p16 gene high-expression vector transfection. Clinical and prognosis data were summarized and the influence of p16 on patient prognosis was analyzed through Kaplan-Meier single-factor survival analysis. The results showed that p16 expression in SKOV-3 was decreased significantly compared with that in IOSE80. The positive rate of p16 protein expression in ovarian cancer tissue was notably decreased compared with that in normal ovarian tissue. The positive rate of p16 protein expression in ovarian cancer tissue of patients with lymph node metastasis was significantly decreased compared with that of patients without lymph node metastasis. Therefore, transfection of the p16 gene significantly inhibited the protein expression and invasion ability of p16 in SKOV-3. Correlation analyses between p16 and survival prognosis demonstrated that lower expression of p16 was negatively correlated with the prognosis of patients with ovarian cancer. Overall, the abnormal expression of p16 in ovarian cancer is associated with an increased invasion ability of ovarian cancer and the lower expression of p16 in tissue samples indicates a poor prognosis in patients with ovarian cancer.

7.
Oncol Rep ; 29(2): 515-22, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23151974

RESUMO

Second mitochondria-derived activator of caspases (Smac) is a recently identified protein that is released from mitochondria in response to apoptotic stimuli and promotes apoptosis by antagonizing the inhibitor of apoptosis proteins (IAPs). Our previous study showed that ectopic overexpression of Smac sensitizes drug-resistant tumor cells to TRAIL- or paclitaxel-induced apoptosis in vitro. The present study was designed to explore the effect of the synthesized Smac N7 peptide in a human ovarian cancer cell line and xenograft model. The results showed that the single-agent Smac N7 had a non-cytotoxic effect, but it effectively enhanced TRAIL- or paclitaxel-induced inhibition of cell proliferation in a dose-dependent manner, even in TRAIL-resistant A2780 cells. When Smac N7 was combined with TRAIL or paclitaxel in treating A2780 cell tumor xenografts, synergistic anticancer effects were achieved. Furthermore, the combination therapy caused less damage in normal tissues and more apoptosis in tumor xenografts compared with TRAIL or paclitaxel alone. Increased apoptosis was associated with the downregulation of XIAP, survivin and the increased activity of caspase-3, along with an increased amount of cleaved PARP. In conclusion, this Smac N7 peptide is a promising candidate for ovarian cancer combination therapy, and Smac may be the target for the development of a novel class of anticancer drugs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Oligopeptídeos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Paclitaxel/farmacologia , Pele/patologia , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Análise de Variância , Animais , Carcinoma Epitelial do Ovário , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Necrose/induzido quimicamente , Neoplasias Epiteliais e Glandulares/metabolismo , Oligopeptídeos/farmacocinética , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Survivina , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
8.
J Natl Cancer Inst ; 102(11): 812-27, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20484106

RESUMO

BACKGROUND: Stanniocalcin 1 (STC1) is a secreted glycoprotein hormone. High expression of STC1 has been associated with several cancers including ovarian cancer, but its role in the development of ovarian cancer is not clear. METHODS: We used five human ovarian epithelial cancer cell lines (OVCA420, OVCA432, OVCA433, SKOV3, and HEY), immortalized human ovarian surface epithelial cells (T29 and T80), ovarian cancer tissues from 342 patients, serum from 73 ovarian cancer patients and from58 control subjects, and 116 mice, with six or eight per group. Protein expression was assessed. Cells overexpressing STC1 protein were generated by ectopic expression of human STC1 cDNA. STC1 expression was silenced by using small interfering RNA against STC1. Cell proliferation, migration, colony formation, and apoptosis were assessed. Xenograft tumor growth in mice was studied. Neutralizing anti-STC1 antibody was used to inhibit STC1 function. All statistical tests were two-sided. RESULTS: STC1 protein expression was higher in all human ovarian cancer cell lines examined than in immortalized human ovarian epithelial cell lines, higher in ovarian cancer tissue than in normal ovarian tissue (P < .001), and higher in serum from ovarian cancer patients than from control subjects (P = .021). Ovarian cancer cells with STC1 overexpression, compared with corresponding control cells, had increased cell proliferation, migration, and colony formation in cell culture and increased growth of xenograft tumors in mice. These activities in normal or malignant ovarian cells with STC1 overexpression, compared with control cells, were also accompanied by increased expression of cell cycle regulatory proteins and antiapoptotic proteins but decreased cleavage of several caspases. Within 24 hours of treatment, apoptosis in cultures of HEY ovarian cancer cells treated with neutralizing anti-STC1 monoclonal antibody was higher (17.3% apoptotic cells) than that in cultures treated with mouse IgG control cells (4.4%) (12.9% difference, 95% confidence interval = 11.6% to 14.2%). CONCLUSIONS: STC1 protein may be involved in ovarian tumorigenesis.


Assuntos
Biomarcadores Tumorais/metabolismo , Movimento Celular , Proliferação de Células , Glicoproteínas/metabolismo , Neoplasias Ovarianas/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , DNA Complementar/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Glicoproteínas/sangue , Glicoproteínas/genética , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Neoplasias Ovarianas/sangue , RNA Interferente Pequeno/metabolismo , Transplante Heterólogo , Regulação para Cima
9.
Cell Cycle ; 9(1): 140-6, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20016289

RESUMO

Papillary differentiation is one of the most common histological features of ovarian cancer, although the underlying mechanism that leads to such differentiation is not known. We hypothesized that human ovarian surface epithelial cells can be transformed into carcinoma with papillary differentiation by overexpressing HER2/neu in these cells. Mice were injected either subcutaneously or intraperitoneally with two immortalized human ovarian surface epithelial cell lines after enforced expression of HER-2/neu. Mice subcutaneously injected with tumor cells from either the T29Nt or T80Nt developed undifferentiated carcinomas. In contrast, mice injected intraperitoneally with T29Nt cells developed papillary carcinoma, and those injected intraperitoneally with T80Nt cells developed undifferentiated carcinoma. Our results demonstrate that ovarian surface epithelial cells can develop into papillary carcinoma in mice, and that the induction of papillary differentiation depends not only on specific genetic modifications but also on the tumor microenvironment and epithelial cell type from ovary from different patients.


Assuntos
Carcinoma Papilar/patologia , Transformação Celular Neoplásica/patologia , Células Epiteliais/citologia , Neoplasias Ovarianas/patologia , Ovário/citologia , Peritônio/patologia , Animais , Western Blotting , Carcinoma Papilar/genética , Transformação Celular Neoplásica/genética , Feminino , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Nus , Neoplasias Ovarianas/genética , Receptor ErbB-2/genética , Receptor ErbB-2/fisiologia
10.
Front Biosci (Landmark Ed) ; 14(6): 2089-102, 2009 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-19273186

RESUMO

Epithelial ovarian cancer, which comprises several histologic types and grades, is the most lethal cancer among women in the United States. In this review, we summarize recent progress in understanding the pathology and biology of this disease and in development of models for preclinical research. Our new understanding of this disease suggests new targets for therapeutic intervention and novel markers for early detection of disease.


Assuntos
Neoplasias Ovarianas/patologia , Feminino , Genes Homeobox , Humanos , Neoplasias Ovarianas/classificação , Neoplasias Ovarianas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA