Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Tumour Biol ; 39(6): 1010428317706217, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28618953

ABSTRACT

The epithelial-mesenchymal transition is the key process driving cancer metastasis. MicroRNA-194 inhibits epithelial-mesenchymal transition in several cancers and its downregulation indicates a poor prognosis in human endometrial carcinoma. Self-renewal factor Sox3 induces epithelial-mesenchymal transition at gastrulation and is also involved epithelial-mesenchymal transition in several cancers. We intended to determine the roles of Sox3 in inducing epithelial-mesenchymal transition in endometrial cancer stem cells and the possible role of microRNA-194 in controlling Sox3 expression. Firstly, we found that Sox3 and microRNA-194 expressions were associated with the status of endometrial cancer stem cells in a panel of endometrial carcinoma tissue, the CD133+ cell was higher in tumorsphere than in differentiated cells, and overexpression of microRNA-194 would decrease CD133+ cell expression. Silencing of Sox3 in endometrial cancer stem cell upregulated the epithelial marker E-cadherin, downregulated the mesenchymal marker vimentin, and significantly reduced cell invasion in vitro; overexpression of Sox3 reversed these phenotypes. Furthermore, we discovered that the expression of Sox3 was suppressed by microRNA-194 through direct binding to the Sox3 3'-untranslated region. Ectopic expression of microRNA-194 in endometrial cancer stem cells induced a mesenchymal-epithelial transition by restoring E-cadherin expression, decreasing vimentin expression, and inhibiting cell invasion in vitro. Moreover, overexpression of microRNA-194 inhibited endometrial cancer stem cell invasion or metastasis in vivo by injection of adenovirus microRNA-194. These findings demonstrate the novel mechanism by which Sox3 contributes to endometrial cancer stem cell invasion and suggest that repression of Sox3 by microRNA-194 may have therapeutic potential to suppress endometrial carcinoma metastasis. The cancer stem cell marker, CD133, might be the surface marker of endometrial cancer stem cell.


Subject(s)
AC133 Antigen/genetics , Endometrial Neoplasms/genetics , MicroRNAs/genetics , SOXB1 Transcription Factors/genetics , Cadherins/antagonists & inhibitors , Cadherins/biosynthesis , Cell Line, Tumor , Cell Proliferation/genetics , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , MicroRNAs/biosynthesis , Neoplasm Invasiveness/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , SOXB1 Transcription Factors/biosynthesis
2.
Tumour Biol ; 36(10): 7997-8006, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25967456

ABSTRACT

Glypican-3 (GPC3) is a membrane of heparan sulfate proteoglycan family involved in cell proliferation, adhesion, migration, invasion, and differentiation during the development of the majority of mesodermal tissues and organs. GPC3 is explored as a potential biomarker for hepatocellular carcinoma screening. However, as a tumor-associated antigen, its role in ovarian cancer remains elusive. In this report, the expression levels of GPC3 in the various ovarian cancer cells were determined with quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and GPC3 expression in ovarian cancer UCI 101 and A2780 cells was knocked down by siRNA transfection, and the effects of GPC3 knockdown on in vitro cell proliferation, migration, and invasion were respectively analyzed by 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide (MTT) assay and Transwell migration assay. Additionally, the effect of GPC3 knockdown on in vivo tumorigenesis were investigated in athymic nude mice. The results indicated that GPC3 knockdown significantly promoted cell proliferation and increased cell migration and invasion by upregulation of matrix metalloproteinase (MMP)-2 and MMP-9 expression and downregulation of tissue inhibitor of metalloproteinase-1 expression. Additionally, GPC3 knockdown also increased in vivo tumorigenicity of UCI 101 and A2780 cells and final tumor weights and volumes after subcutaneous cell injection in the nude mice. The results of immunohistochemical staining and Western blotting both demonstrated a lower expression of GPC3 antigen in the tumors of GPC3 knockdown groups than that of negative control groups. Moreover, transforming growth factor-ß2 protein expression in the tumors of GPC3 knockdown groups was significantly increased, which at least contributed to tumor growth in the nude mice. Taken together, these findings suggest that GPC3 knockdown promotes the progression of human ovarian cancer cells by increasing their migration, invasion, and tumorigenicity, and suggest that GPC3 is a potential therapeutic target for ovarian cancer patients.


Subject(s)
Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Glypicans/antagonists & inhibitors , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Glypicans/genetics , Glypicans/metabolism , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Neoplasm Invasiveness , Ovarian Neoplasms/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Int J Gynecol Pathol ; 30(3): 262-70, 2011 May.
Article in English | MEDLINE | ID: mdl-21464727

ABSTRACT

The embryonic stem cell self-renewal gene, Nanog, has been shown to be expressed in several tumor types and to regulate tumor development. The aim of this study was to carry out a detailed analysis of Nanog expression in human endometrial adenocarcinoma (EAC). Immunohistochemical analysis and reverse transcription-polymerase chain reaction were used to characterize Nanog, Sox2, and Oct4 expression in tissue arrays containing EAC, benign endometrium samples, and tumorosphere cells. Tumorosphere formation of EAC-derived cells in the stem cell culture medium was also analyzed. Nanog expression was then analyzed in secondary tumors initiated by the injection of tumorospheres or tumorosphere-derived differentiated cells into 15 female nude mice. Apoptosis and cell proliferation were detected in the fluorescence-activated cell sorter and 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide experiments, respectively. The Nanog protein was expressed in a majority of EAC samples (45 of 55, 81.8%), but not in benign endometrium samples (0 of 26, 0.0%). Oct4 and Sox2 were also commonly expressed in EAC samples (42 of 55, 76.4% and 39 of 55, 70.9%, respectively). Subsets of cancer cells from all EAC samples (15 of 15, 100%) exhibited the capacity to form Nanog-positive tumorospheres. The tumorospheres also expressed Nanog, Oct4, and Sox2 mRNA and showed a higher proliferation potential than differentiated cells. All 15 mice that were injected with tumorosphere cell-formed tumors, whereas only 3 of 15 mice injected with differentiated cells derived from tumorospheres developed tumors. All secondary xenograft tumors still expressed Nanog protein and Nanog, Oct4, and Sox2 mRNA, and had higher proliferation and lower apostosis than did differentiated cells. Overexpression of Nanog in EACs suggests that Nanog may represent a potential therapeutic target for EAC. In addition, Nanog may be useful as a biomarker in an immunohistochemical panel to differentiate between EAC and benign endometrial tissues. The expression of Nanog in tumorospheres may be indicative of the presence of a population of endometrial cancer stem cells, and its expression in xenograft tumors suggests that Nanog may also be associated with tumor metastasis.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Homeodomain Proteins/biosynthesis , Neoplastic Stem Cells/pathology , Animals , Apoptosis/physiology , Biomarkers, Tumor/analysis , Cell Separation , Female , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Nude , Nanog Homeobox Protein , Neoplastic Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/chemistry , Stem Cells/metabolism , Tissue Array Analysis , Transplantation, Heterologous
4.
Biosci Rep ; 38(6)2018 12 21.
Article in English | MEDLINE | ID: mdl-30401729

ABSTRACT

Casticin is one of the major active components isolated from Fructus viticis Increasing studies have revealed that casticin has potential anticancer activity in various cancer cells, but its effects on breast cancer cell migration and invasion are still not well known. Therefore, the ability of cell migration and invasion in the breast cancer MDA-MB-231 and 4T1 cells treated by casticin was investigated. The results indicated that casticin significantly inhibited cell migration and invasion in the cells exposed to 0.25 and 0.50 µM of casticin for 24 h. Casticin treatment reduced matrix metalloproteinase (MMP) 9 (MMP-9) activity and down-regulated MMP-9 mRNA and protein expression, but not MMP-2. Casticin treatment suppressed the nuclear translocation of transcription factors c-Jun and c-Fos, but not nuclear factor-κB (NF-κB), and decreased the phosphorylated level of Akt (p-Akt). Additionally, the transfection of Akt overexpression vector to MDA-MB-231 and 4T1 cells could up-regulate MMP-9 expression concomitantly with a marked increase in cell invasion, but casticin treatment reduced Akt, p-Akt, and MMP-9 protein levels and inhibited the ability of cell invasion in breast cancer cells. Additionally, casticin attenuated lung metastasis of mouse 4T1 breast cancer cells in the mice and down-regulated MMP-9 expression in the lung tissues of mice treated by casticin. These findings suggest that MMP-9 expression suppression by casticin may act through inhibition of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, which in turn results in the inhibitory effects of casticin on cell migration and invasion in breast cancer cells. Therefore, casticin may have potential for use in the treatment of breast cancer invasion and metastasis.


Subject(s)
Breast Neoplasms/drug therapy , Cell Movement/drug effects , Flavonoids/administration & dosage , Matrix Metalloproteinase 9/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 2/genetics , Mice , Mitogen-Activated Protein Kinase 8/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fos/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
Int J Oncol ; 50(3): 853-862, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28112371

ABSTRACT

MicroRNA (miR)-145 has been reported to induce cancer stem cell (CSC) differentiation through down-regulation of the stem cell transcription factors (TFs) that maintain CSC pluripotency. High expression of miR-145 indicates a good prognosis in cancer patients, but its role in cervical cancer stem cells (CCSCs) is not known. We show that expression of miR-145 and core stem cell transcription factors, Sox2, Nanog and Oct4, are associated with the pluripotency of CCSCs, with increased expression of miR-145 after cervical tumorsphere (CT) differentiation. miR-145 overexpression inhibited expression of core TFs, as well as decreasing tumor invasion and colony formation, whereas miR-145 knockdown led to the opposite effects. Injection of adenovirus-miR-145 significantly reduced tumor growth in nude mice. High miR-145 expression predicted a better prognosis compared with that in patients with low miR-145 expression after analyses of The Cancer Genome Atlas (TCGA) data. These results suggest that miR-145 is able to induce CT differentiation through enzymolyzing TFs and might be a therapeutic target for cervical carcinoma.


Subject(s)
Carcinogenesis/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Adult , Aged , Animals , Cell Proliferation/genetics , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Middle Aged , Nanog Homeobox Protein/metabolism , Neoplasm Invasiveness/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/pathology , SOXB1 Transcription Factors/metabolism , Tumor Cells, Cultured
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(7): 1695-8, 2010 Jul.
Article in Zh | MEDLINE | ID: mdl-20650803

ABSTRACT

OBJECTIVE: To investigate the changes in SiHa cell apoptosis after inhibition of CD147 expression. METHODS: RNA interference (RNAi) technique was used to down-regulate CD147 expression in SiHa cells, and RT-PCR and Western blotting were used to detect expression of CD147, Bcl-2, Bim and caspase-3; the percentage of cell apoptosis were detected by flow cytometry. RESULTS: SiRNA sequence 1, 2 inhibited CD147 expression in SiHa cells effectively (P<0.05), resulting also in down-regulated expression of Bcl-2 (P<0.05) and up-regulated expression of caspase-3 and Bim(P<0.05). The percentage of apoptotic cells increased significantly, and early apoptosis was the most obvious in the cells (P<0.05). CONCLUSION: Silencing of CD147 expression induces SiHa cell apoptosis partially through the Bcl-2 pathway .


Subject(s)
Apoptosis , Basigin/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Caspase 3/metabolism , Cell Line, Tumor , Down-Regulation , Humans , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL