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








Language
Year range
1.
Asian Pacific Journal of Tropical Medicine ; (12): 91-97, 2012.
Article in English | WPRIM | ID: wpr-819821

ABSTRACT

OBJECTIVE@#To investigate possible mechanism of toxicarioside A in HS-5 bone stromal cells.@*METHODS@#HS-5 bone stromal cells were cultured in media supplemented with various concentrations of toxicarioside A or control DMSO (not treatment). Endoglin and TGF-β were detected by Northern and Western blot analysis and quantified in a standard method. Downstream molecules of endoglin and TGF-β (Smad1, Smad2 and their active phosphorylated counterparts, pSmad1 and pSmad2) were also detected and quantified by Western blot analysis. In addition, cell proliferation assay and small interfering RNA (siRNA) against endoglin were used to certificate the function of endolgin in the HS-5 cells.@*RESULTS@#Compared with the not treated (0 μg/mL) or DMSO treated control HS-5 cells, HS-5 cells treated with toxicarioside A were found significant attenuation of endolgin and TGF-β expression. Significant inhibition of cell proliferation was also found in the HS-5 cells treated with toxicarioside A. ALK1-related Smad1 and ALK5-related Smad2 were decreased in HS-5 cells treated with toxicarioside A. In addition, phosphorylated Smad1 (pSmad1) and Smad2 (pSmad2) were also found attenuation in toxicarioside A-treated HS-5 cells. RNA interference showed that blockage of endoglin by siRNA also decreased Smad1 and Smad2 expression in HS-5 cells.@*CONCLUSIONS@#Our results indicate that toxicarioside A can influence bone marrow stromal HS-5's function and inhibit HS-5 cell proliferation by alteration of endoglin-related ALK1 (Smad1) and ALK5 (Smad2) signaling.


Subject(s)
Humans , Male , Antiaris , Antigens, CD , Metabolism , Blotting, Northern , Blotting, Western , Bone Marrow Cells , Metabolism , Cardenolides , Pharmacology , Cell Line , Cell Proliferation , Endoglin , Receptors, Cell Surface , Metabolism , Signal Transduction , Smad1 Protein , Metabolism , Smad2 Protein , Metabolism , Stromal Cells , Transforming Growth Factor beta , Metabolism
2.
Asian Pacific Journal of Tropical Medicine ; (12): 169-174, 2012.
Article in English | WPRIM | ID: wpr-819804

ABSTRACT

OBJECTIVE@#To investigate whether cytochalasin D can induce antitumor activities in a tumor model.@*METHODS@#Murine CT26 colorectal carcinoma cells were cultured in vitro and cytochalasin D was used as a cytotoxic agent to detect its capabilities of inhibiting CT26 cell proliferation and inducing cell apoptosis by MTT and a TUNEL-based apoptosis assay. Murine CT26 tumor model was established to observe the tumor growth and survival time. Tumor tissues were used to detect the microvessel density by immunohistochemistry. In addition, alginate encapsulated tumor cell assay was used to quantify the tumor angiogenesis in vivo.@*RESULTS@#Cytochalasin D inhibited CT26 tumor cell proliferation in time and dose dependent manner and induced significant CT26 cell apoptosis, which almost reached the level induced by the positive control nuclease. The optimum effective dose of cytochalasin D for in vivo therapy was about 50 mg/kg. Cytochalasin D in vivo treatment significantly inhibited tumor growth and prolonged the survival times in CT26 tumor-bearing mice. The results of immunohistochemistry analysis and alginate encapsulation assay indicated that the cytochalasin D could effectively inhibited tumor angiogenesis.@*CONCLUSIONS@#Cytochalasin D inhibits CT26 tumor growth potentially through inhibition of cell proliferation, induction of cell apoptosis and suppression of tumor angiogenesis.


Subject(s)
Animals , Mice , Angiogenesis Inhibitors , Pharmacology , Antineoplastic Agents , Pharmacology , Apoptosis , Cell Proliferation , Colorectal Neoplasms , Drug Therapy , Cytochalasin D , Pharmacology , Dose-Response Relationship, Drug , In Situ Nick-End Labeling , Microvessels , Neovascularization, Pathologic , Tumor Cells, Cultured
3.
Journal of Zhejiang University. Science. B ; (12): 602-609, 2008.
Article in English | WPRIM | ID: wpr-359375

ABSTRACT

Induction of tumor vasculature occlusion by targeting a thrombogen to newly formed blood vessels in tumor tissues represents an intriguing approach to the eradication of primary solid tumors. In the current study, we construct and express a fusion protein containing vascular endothelial growth factor (VEGF) and tissue factor (TF) to explore whether this fusion protein has the capability of inhibiting tumor growth in a colon carcinoma model. The murine cDNA of VEGF A and TF were amplified by reverse transcriptase polymerase chain reaction (RT-PCR), and then cloned into prokaryotic expression plasmid pQE30 with a linker. The expression product recombinant VEGF-TF (rVEGF-TF) was purified and proved to have comparable enzyme activity to a commercial TF and the capability of specific binding to tumor vessels. Significant decrease of tumor growth was found in the mice administered with rVEGF-TF on Day 6 after initiated rVEGF-TF treatment (P<0.05), and the tumor masses in 2 of 10 mice were almost disappeared on Day 14 after the first treatment. In addition, valid thrombogenesis and tumor necrosis were observed in the tumor tissues injected with rVEGF-TF. Our results demonstrate that occlusion of tumor vasculature with rVEGF-TF is potentially an effective approach for cancer therapy.


Subject(s)
Animals , Mice , Cell Line, Tumor , Cloning, Molecular , Colonic Neoplasms , Drug Therapy , Pathology , Disease Models, Animal , Disease Progression , Gene Expression , Mice, Inbred BALB C , Neoplasm Transplantation , Plasmids , Genetics , Recombinant Fusion Proteins , Genetics , Metabolism , Therapeutic Uses , Thromboplastin , Genetics , Metabolism , Therapeutic Uses , Thrombosis , Drug Therapy , Pathology , Vascular Endothelial Growth Factor A , Genetics , Metabolism , Therapeutic Uses
SELECTION OF CITATIONS
SEARCH DETAIL