Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Publication year range
1.
Cancer Med ; 8(15): 6768-6779, 2019 11.
Article in English | MEDLINE | ID: mdl-31517442

ABSTRACT

HOTAIR, a well-known long noncoding RNAs (lncRNA), has been recognized to contribute to the tumor metastasis in several tumors. But its role in gastric cancer remains elusive. Here, we reported an increase in HOTAIR promoted proliferation and metastasis of gastric cancer cell lines. The HOTAIR and miR-126 level was determined in 15 paired primary gastric cancer tissues and their adjacent noncancerous gastric tissues. Over-expression or downregulation HOTAIR was conducted in AGS or BGC-823 cells to investigate the impact of HOTAIR in proliferation and metastasis. Then dual luciferase reporter assay was utilized to study the interaction between CXCR4 and miR-126. Cells transfected with shHOTAIR or miR-126 mimic were subjected to western blot to investigate the role of SDF-1/CXCR4 signaling in HOTAIR mediated proliferation and metastasis. HOTAIR was highly expressed in gastric cancer tissues and several gastric cancer cell lines. Overexpressed HOTAIR facilitated proliferation and metastasis in vitro while HOTAIR knockdown inhibit proliferation and metastasis. A negative correlation was observed between miR-126 and HOTAIR. And, we also confirmed the decrease in miR-126 in clinic specimen. Furthermore, HOTAIR and miR-126 negatively regulated each other and then increase or decrease CXCR4 expression and downstream pathway, respectively. CXCR4 was confirmed as a direct target of miR-126. Our study demonstrated that high HOTAIR expression promote proliferation and metastasis in gastric cancer via miR-126/CXCR4 axis and downstream signaling pathways.


Subject(s)
MicroRNAs/genetics , RNA, Long Noncoding/genetics , Receptors, CXCR4/genetics , Stomach Neoplasms/genetics , 3' Untranslated Regions , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Signal Transduction , Stomach Neoplasms/metabolism , rhoA GTP-Binding Protein/metabolism
2.
Exp Mol Pathol ; 107: 85-94, 2019 04.
Article in English | MEDLINE | ID: mdl-30502321

ABSTRACT

BACKGROUND: Cancer associated fibroblasts (CAFs) are known to be crucial constituents of cancer microenvironment (CME) and play an important role in initiation, progression and metastasis of various types of cancer, such as oral cancer, pancreatic cancer, and gastric cancer. CAFs are usually derived from normal fibroblasts (NFs), but the mechanism of the transition in gastric cancer has not yet been fully elucidated. METHODS: qRT-PCR and western blot were employed to investigate differences of miR-141 and STAT4 expression respectively. The CAF-like features and wnt/ß-catenin pathway related proteins in NF or BMSC were assessed by qRT-PCR or western blot after treated with the conditioned medium from different indicated groups of gastric cancer cells. The invasion and migration ability of AGS cells after transfection were analyzed by Transwell assay and wound healing assay. Dual-luciferase report assay was employed to determine the direct binding of miR-141 to STAT4 3' UTR. RESULTS: For the first time, the present study found that STAT4 over-expression in gastric cancer cells induced NFs to obtain CAF-like features via activating wnt/ß-catenin pathway. Further gain-of-function and loss-of-function analysis revealed that miR-141 not only limited the migration and invasion of the gastric cancer cells, but also inhibited the transition of NFs and BMSC to CAFs. The luciferase assay indicated that miR-141 directly targeted the 3'-UTR predictive sequence of STAT4. CONCLUSION: Our data showed that miR-141 inhibited migration and invasion of gastric cancer cells and inhibited transition from NFs to CAFs via targeting STAT4/wnt/ß-catenin pathway.


Subject(s)
Cancer-Associated Fibroblasts/pathology , Fibroblasts/pathology , MicroRNAs/genetics , STAT4 Transcription Factor/metabolism , Stomach Neoplasms/pathology , Cell Differentiation/physiology , Cell Movement/physiology , Humans , Mesenchymal Stem Cells/pathology , Neoplasm Invasiveness/pathology , Tumor Microenvironment/physiology
3.
Zhonghua Wei Chang Wai Ke Za Zhi ; 16(9): 874-7, 2013 Sep.
Article in Chinese | MEDLINE | ID: mdl-24061998

ABSTRACT

OBJECTIVE: To investigate the effect of PC cell-derived growth factor (PCDGF) RNA interference on esophageal squamous carcinoma cells Eca-109 in vitro. METHODS: The PCDGF-shRNA expression vector was transfected into the Eca-109 cells by liposome. After transfection, the mRNA and protein expressions of PCDGF were detected by RT-PCR and Western-blot respectively. Cell Counting Kit-8 (CCK-8) assay and Boyden chamber method were performed to measure the cell proliferation and invasion ability respectively. RESULTS: The expression levels of PCDGF mRNA and protein were both decreased in Eca-109 cells transfected with PCDGF-shRNA expression vector (transfection group). Twenty-four, 48 and 72 h after transfection, the cells proliferation in the transfection group was inhibited, and the inhibition rate was 20.4%, 21.1% and 20.9% respectively. The cell proliferation activity in the transfection group was significantly lower than that in the non-transfection group, liposome group and negative vector group (all P<0.05). The number of cell migration in the non-transfection group,negative vector group, liposome group and transfection group was 118.8±12.0, 100.8±9.0, 114.3±4.7, and 53.5±16.3 respectively. The differences were statistically significant between the transfection group and the other 3 groups (all P<0.05). CONCLUSIONS: PCDGF RNA interference can inhibit the proliferation and invasion abilities of esophageal squamous carcinoma cells in vitro. PCDGF gene may be the new target of gene therapy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Intercellular Signaling Peptides and Proteins/genetics , RNA Interference , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma , Genetic Vectors , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Progranulins , RNA, Small Interfering/genetics , Transfection
4.
Oncol Rep ; 30(5): 2161-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23970348

ABSTRACT

Recent evidence suggests the involvement of γ-synuclein in tumorigenesis and tumor progression. The present study was designed to further clarify the effects of γ-synuclein on the biological features of colon cancer cells in vitro and in vivo. We constructed the eukaryotic expression vector and siRNA vector and selected stable transfectants to respectively upregulate and downregulate γ-synuclein expression in SW1116 cells. we found that silencing of γ-synuclein significantly attenuated SW1116 cell growth and colony formation in vitro (P<0.05), and overexpression of γ-synuclein moderately enhanced cell growth and colony formation, but not significantly when compared with the parental SW1116 cells and empty vector-transfected cells (P>0.05). Meanwhile, overexpression of γ-synuclein significantly facilitated SW1116 cell migration, invasion and adhesion to human liver sinusoidal endothelial cells (HLSECs) in vitro (P<0.05), and the effects were less attenuated by γ-synuclein knockdown (P>0.05). Furthermore, γ-synuclein promoted these malignant phenotypes in a γ-synuclein expression quantity-dependent manner not only in vitro but also in the in vivo expression. stable cells were injected subcutaneously into the right flank, and injected intrasplenically in nude mice. γ-synuclein knockdown suppressed the tumorigenicity of SW1116 cells in mice, which presented significantly smaller tumor masses on day 6 over a 30-day period, compared with empty vector cells (P<0.05). Meanwhile, overexpression of γ-synuclein led to a profound augmentation of liver metastasis in nude mice, not only in macroscopic appearance but also in the size and weight of livers (P<0.05). These results provide strong evidence that suggests γ-synuclein plays a positive role in the progression of colorectal cancer.


Subject(s)
Carcinogenesis/genetics , Colonic Neoplasms/genetics , Liver Neoplasms/secondary , gamma-Synuclein/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Colonic Neoplasms/pathology , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , RNA, Small Interfering/genetics , gamma-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...