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.
Int J Mol Sci ; 19(3)2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29510576

ABSTRACT

Lung metastasis constitutes the leading cause of the death in patients with osteosarcoma. We have previously reported that plasminogen activator inhibitor-1 (PAI-1) regulates the invasion and lung metastasis of osteosarcoma cells in a mouse model and as well as in clinical samples. In the present study, we examined the anti-metastatic effect of SK-216, a small compound PAI-1 inhibitor, in human 143B osteosarcoma cells. An in vitro study showed that SK-216 treatment suppressed invasion activity by inhibiting PAI-1 expression in 143B cells, but had no influence on their proliferation or migration. 143B cells treated with SK-216 exhibited reduced matrix metalloproteinase-13 (MMP-13) secretion in a dose-dependent manner. Moreover, intraperitoneal injection of SK-216 into mouse models resulted in downregulation of PAI-1 expression levels in the primary tumors and showed suppression of lung metastases without influencing the proliferative activity of the tumor cells in the primary lesions. These results indicate that SK-216, a PAI-1 inhibitor, may serve as a novel drug to prevent lung metastasis in human osteosarcoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzoxazoles/therapeutic use , Dicarboxylic Acids/therapeutic use , Lung Neoplasms/drug therapy , Osteosarcoma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Benzoxazoles/administration & dosage , Benzoxazoles/pharmacology , Cell Line, Tumor , Dicarboxylic Acids/administration & dosage , Dicarboxylic Acids/pharmacology , Lung Neoplasms/secondary , Male , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Nude , Osteosarcoma/pathology , Plasminogen Activator Inhibitor 1/metabolism
2.
Cancer Med ; 5(5): 892-902, 2016 05.
Article in English | MEDLINE | ID: mdl-26817521

ABSTRACT

Despite recent improvements in the therapy for osteosarcoma, 30-40% of osteosarcoma patients die of this disease, mainly due to its lung metastasis. We have previously reported that intravenous injection of miR-143 significantly suppresses lung metastasis of human osteosarcoma cells (143B) in a mouse model. In this study, we examined the biological role and mechanism of miR-143 in the metastasis of human osteosarcoma cells. We identified plasminogen activator inhibitor-1 (PAI-1) as a direct target gene of miR-143. To determine the role of PAI-1 in human osteosarcoma cells, siRNA was transfected into 143B cells for knockdown of PAI-1 expression. An in vitro study showed that downregulation of PAI-1 suppressed cell invasion activity, but not proliferation. Moreover, injection of PAI-1 siRNA into a primary lesion in the osteosarcoma mouse model inhibited lung metastasis compared to control siRNA-injected mice, without influencing the proliferative activity of the tumor cells. Subsequent examination using 143B cells revealed that knockdown of PAI-1 expression resulted in downregulation of the expression and secretion of matrix metalloproteinase-13 (MMP-13), which is also a target gene of miR-143 and a proteolytic enzyme that regulates tumor-induced osteolysis. Immunohistochemical analysis using clinical samples showed that higher miR-143 expressing cases showed poor expression of PAI-1 in the primary tumor cells. All such cases belonged to the lung metastasis-negative group. Moreover, the frequency of lung metastasis-positive cases was significantly higher in PAI-1 and MMP-13 double-positive cases than in PAI-1 or MMP-13 single-positive or double-negative cases (P < 0.05). These results indicated that PAI-1, a target gene of miR-143, regulates invasion and lung metastasis via enhancement of MMP-13 expression and secretion in human osteosarcoma cells, suggesting that these molecules could be potential therapeutic target genes for preventing lung metastasis in osteosarcoma patients.


Subject(s)
Bone Neoplasms/genetics , Matrix Metalloproteinase 13/biosynthesis , MicroRNAs/genetics , Osteosarcoma/genetics , Plasminogen Activator Inhibitor 1/genetics , Animals , Bone Neoplasms/pathology , Cell Proliferation/genetics , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques/methods , Heterografts , Humans , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Mice, Nude , MicroRNAs/physiology , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation , Osteosarcoma/pathology , Osteosarcoma/secondary , RNA, Small Interfering/genetics , Transfection , Tumor Cells, Cultured , Up-Regulation/genetics
3.
Proteomics ; 14(9): 1031-41, 2014 May.
Article in English | MEDLINE | ID: mdl-24574163

ABSTRACT

By a proteomics-based approach, we identified an overexpression of fascin in colon adenocarcinoma cells (FPCKpP-3) that developed from nontumorigenic human colonic adenoma cells (FPCK-1-1) and were converted to tumorigenic by foreign-body-induced chronic inflammation in nude mice. Fascin overexpression was also observed in the tumors arising from rat intestinal epithelial cells (IEC 6) converted to tumorigenic in chronic inflammation which was induced in the same manner. Upregulation of fascin expression in FPCK-1-1 cells by transfection with sense fascin cDNA converted the cells tumorigenic, whereas antisense fascin-cDNA-transfected FPCKpP-3 cells reduced fascin expression and lost their tumor-forming ability in vivo. The tumorigenic potential by fascin expression was consistent with their ability to survive and grow in the three-dimensional multicellular spheroids. We found that resistance to anoikis (apoptotic cell death as a consequence of insufficient cell-to-substrate interactions), which is represented by the three-dimensional growth of solid tumors in vivo, was regulated by fascin expression through caspase-dependent apoptotic signals. From these, we demonstrate that fascin is a potent suppressor to caspase-associated anoikis and accelerator of the conversion of colonic adenoma cells into adenocarcinoma cells by chronic inflammation.


Subject(s)
Anoikis/physiology , Carrier Proteins/metabolism , Colonic Neoplasms/metabolism , Inflammation/metabolism , Microfilament Proteins/metabolism , Animals , Carrier Proteins/analysis , Carrier Proteins/genetics , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Microfilament Proteins/analysis , Microfilament Proteins/genetics , Rats , Spheroids, Cellular/metabolism , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...