Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Cells ; 11(21)2022 10 27.
Article in English | MEDLINE | ID: mdl-36359787

ABSTRACT

Epithelial ovarian cancer (EOC) patients frequently develop peritoneal metastasis, especially in the human omentum. However, the mechanism underlying this propensity remains unknown. A previous study found that human omental adipose-derived mesenchymal stem cells are potentially involved in ovarian cancer growth and metastasis, but the results were inconsistent and even contradictory. In addition, the underlying mechanisms of visceral adipose metastasis remain poorly understood. Here, our goal is to clarify the role and mechanism of human omental adipose-derived mesenchymal stem cells (HO-ADSCs) in EOC cancer growth and metastasis. We first found that human omental tissue conditioned medium (HO-CM) enhances EOC cell function. Subsequent coculture studies indicated that HO-ADSCs increase the growth, migratory and invasive capabilities of ovarian cancer cells. Then, we demonstrated that exosomes secreted by HO-ADSCs (HO-ADSC exosomes) enhanced ovarian cancer cell function, and further mechanistic studies showed that the FOXM1, Cyclin F, KIF20A, and MAPK signaling pathways were involved in this process. In addition, subcutaneous tumorigenesis and peritoneal metastatic xenograft experiments provided evidence that HO-ADSC exosomes promote ovarian cancer growth and metastasis in vivo. Finally, our clinical studies provided evidence that ascites from ovarian cancer patients enhance EOC cell line proliferation, migration, and invasion in vitro. The present study indicated that HO-ADSC exosomes are secreted into ascites and exert a tumor-promoting effect on EOC growth and metastasis, providing a new perspective and method to develop future novel therapeutic strategies for the treatment of ovarian cancer.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Ovarian Neoplasms , Peritoneal Neoplasms , Humans , Female , Carcinoma, Ovarian Epithelial/pathology , Exosomes/metabolism , Omentum/metabolism , Omentum/pathology , Ascites/pathology , Adipose Tissue/metabolism , Mesenchymal Stem Cells/metabolism , Ovarian Neoplasms/pathology , Neoplastic Processes
2.
Eur J Pharmacol ; 834: 45-53, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30017802

ABSTRACT

The epithelial-mesenchymal transition (EMT) plays a critical role in cancer progression, metastasis and drug resistance. The transcription factor(TF) and microRNA (miR) chimeric [SNAIL/miR-34]:[ZEB/miR-200] unit is the core regulatory system for the EMT process. Here, we proposed to assess the anti-EMT abilities and explore the inherent pharmacological mechanisms of the classic hypoglycaemic agent metformin for colorectal cancer(CRC). For the EMT model, the TGF-ß-induced CRC cell lines SW480 and HCT116 were treated with metformin. The viability, migration and invasion abilities of the cells were evaluated with the Cell Counting Kit-8, wound-healing and trans-well assay. The alterations of the [SNAIL/miR-34]:[ZEB/miR-200] system and the EMT markers E-cadherin and vimentin were detected by western blot, qPCR and immunofluorescent staining. Metformin exhibited inhibitory effects on the proliferation, migration and invasion of the CRC SW480 cells. The up-regulation of E-cadherin and the down-regulation of vimentin for both SW480 and HCT116 cells revealed the anti-EMT abilities of metformin. For the [SNAIL/miR-34]:[ZEB/miR-200] system, metformin increased miR-200a, miR-200c and miR-429 levels and decreased miR-34a, SNAIL1 and ZEB1 levels in the TGF-ß-induced EMT. From immunofluorescence, we observed increased E-cadherin and ZEB1 co-expression in metformin-treated cells. Metformin may perform bidirectional regulations of the [SNAIL/miR-34]:[ZEB/miR-200] system in the EMT process for colorectal cancer. Such regulation is expressed as the inhibition of EMT in general as well as an increased higher proportion of E/M hybrid cells in the total population.


Subject(s)
Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Metformin/pharmacology , MicroRNAs/metabolism , Snail Family Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , HCT116 Cells , Humans , Neoplasm Invasiveness , Transforming Growth Factor beta/pharmacology
3.
Gastroenterol Res Pract ; 2017: 2520581, 2017.
Article in English | MEDLINE | ID: mdl-28744307

ABSTRACT

OBJECTIVES: We aimed to explore the association between metformin treatment and epithelial-mesenchymal transition (EMT) phenotype and further appraise the prognostic values of metformin and EMT markers E-cadherin and vimentin for colorectal cancer (CRC) in clinical practice. METHODS: We collected specimens and evaluated clinicopathological parameters of 102 stage I to III CRC patients with prediagnosed type 2 diabetes mellitus (DM II). Expression of E-cadherin and vimentin in tumors was detected by immunohistochemistry (IHC), and statistical analysis was performed using SPSS 19.0. RESULTS: In correlation tests, we found a lower tumor cell EMT degree (more E-cadherin (P = 0.014) and less vimentin (P = 0.011) expression in patients who used metformin, and the expression of E-cadherin and vimentin was associated with serum CA19-9 (P = 0.048, P = 0.009), tumor invasive depth (T) (P < 0.001, P = 0.045), and lymph invasion (N) (P = 0.013, P = 0.001). In Cox multivariate regression analysis, E-cadherin was identified as a prognostic factor for disease-free survival (DFS) (P = 0.038) and metformin use (P = 0.015P = 0.044) and lymph invasion (P = 0.016P = 0.023) were considered as the prognostic factors for both DFS and overall survival (OS). CONCLUSION: Our study suggested that metformin may impede the EMT process and improve survival for stage I-III CRC patients with DM II.

SELECTION OF CITATIONS
SEARCH DETAIL