Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 9(3): 278, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29449539

ABSTRACT

Tumor metastasis is the leading cause of death in patients with advanced gastric cancer (GC). Limited therapeutic regimens are available for this condition, which is associated with a poor prognosis, and the mechanisms underlying tumor metastasis remain unclear. In the present study, increased histone methyltransferase G9A expression in GC tissues correlated with advanced stage and shorter overall survival, and in vitro and in vivo experiments revealed that G9A promoted tumor invasion and metastasis. Moreover, we observed that Reg IV induced G9A via the p-ERK/p-SP1 pathway. SP1 directly binds the G9A promoter and enhances G9A expression, and upregulated G9A then forms a transcriptional activator complex with P300 and GR, thereby promoting ITGB3 expression induced by dexamethasone (DEX) and contributing to GC metastasis. However, the G9A-mediated increase in ITGB3 expression was not dependent on the SET domain and methyltransferase activity of G9A. This study demonstrates that G9A is an independent prognostic marker and promotes metastasis in GC, thus suggesting that it may be a tumor biomarker and potential therapeutic target in GC.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Integrin beta3/metabolism , Peritoneal Neoplasms/enzymology , Stomach Neoplasms/enzymology , Animals , Binding Sites , Biomarkers, Tumor/genetics , Cell Line, Tumor , E1A-Associated p300 Protein/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans , Integrin beta3/genetics , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , PR-SET Domains , Pancreatitis-Associated Proteins/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/secondary , Phosphorylation , Promoter Regions, Genetic , Receptors, Glucocorticoid/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Up-Regulation
2.
J Transl Med ; 15(1): 52, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28241766

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the most malignant tumors and the second leading cause of cancer-related deaths in the world. Luteolin, a flavonoid present in many fruits and green plants, suppresses cancer progression. The effects of luteolin on GC cells and their underlying mechanisms remain unclear. METHODS: Effects of luteolin on cell proliferation, migration, invasion, and apoptosis were examined in vitro and in vivo by cell counting kit-8 (CCK-8), transwell assays, and flow cytometry, respectively. Real-time reverse transcription polymerase chain reaction (RT-PCR) and Western blots were performed to evaluate Notch1 signaling and activation of epithelial-mesenchymal transition (EMT) in GC cells treated with or without luteolin. Immunohistochemistry was performed to examine proliferation and Notch1 expression in xenograft tumors. RESULTS: Luteolin significantly inhibited cell proliferation, invasion, and migration in a dose-dependent and time-dependent manner and promoted cell apoptosis. Luteolin reversed EMT by shrinking the cytoskeleton and by inducing the expression of epithelial biomarker E-cadherin and downregulating the mesenchymal biomarkers N-cadherin, vimentin and Snail. Furthermore, Notch1 signaling was inhibited by luteolin, and downregulation of Notch1 had similar effects as luteolin treatment on cell proliferation, migration, and apoptosis. In addition, luteolin suppressed tumor growth in vivo. A higher expression of Notch1 correlated with a poor overall survival and a poor time to first progression. Furthermore, co-immunoprecipitation analysis revealed that activated Notch1 and ß-catenin formed a complex and regulated cell proliferation, migration, and invasion. CONCLUSIONS: In this study, GC progression was inhibited by luteolin through suppressing Notch1 signaling and reversing EMT, suggesting that luteolin may serve as an effective anti-tumor drug in GC treatment.


Subject(s)
Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Luteolin/therapeutic use , Receptors, Notch/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Luteolin/chemistry , Luteolin/pharmacology , Male , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Neoplasm Invasiveness , Prognosis , Tumor Stem Cell Assay
3.
Mol Oncol ; 10(9): 1473-1484, 2016 11.
Article in English | MEDLINE | ID: mdl-27590684

ABSTRACT

Biglycan (BGN) is an important component of the extracellular matrix (ECM) that is implicated in a variety of human cancers. In our previous study, we reported that BGN was overexpressed in gastric cancer (GC) tissues and promoted cancer metastasis. Moreover, the tubular formation capacity in HUVECs was promoted by stimulation with culture media from BGN-overexpressing GC cells, but the exact underlying mechanism is still unknown. The purpose of this study was to determine the role and molecular mechanism of BGN in VEGF expression in endothelial cells. We found that BGN stimulation of endothelial cells increased the interaction between NF-kB and the HIF-1α promoter, leading to enhanced promoter activity and increased HIF-1α mRNA levels, as well as augmented HIF-1 activity that resulted in VEGF expression. All of this was dependent on the interaction of BGN with its receptors, TLR2 and TLR4. Moreover, we found that BGN enhanced endothelial cell migration and proliferation, as well as tube formation, in a TLR signaling pathway-dependent manner. In addition, endothelial cell-derived VEGF in turn was found to act on GC cells and promotes their migration. The combined findings of our current and previous studies suggest that BGN secreted from GC cells into the tumor stroma promotes GC development, as well as its progression, potentially through the chronic activation of tumor angiogenesis.


Subject(s)
Biglycan/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Signal Transduction/drug effects , Toll-Like Receptors/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Models, Biological , NF-kappa B/metabolism , Neovascularization, Physiologic/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Recombinant Proteins/pharmacology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
4.
Oncotarget ; 7(9): 9788-800, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26848521

ABSTRACT

Methylation of CpG islands in tumor suppressor gene prompter is one of the most characteristic abnormalities in Helicobacter pylori (HP)-associated gastric carcinoma (GC). Here, we investigated the pathogenic and molecular mechanisms underlying hypermethylation of tumor suppressor genes in HP induced GC development. We found that tumor suppressor genes hypermethylation, represented by MGMT, positively correlated with CagA in clinical specimens, gastric tissues from HP infected C57 mice and GC cell lines transfected by CagA or treated by HP infection. CagA enhanced PDK1 and AKT interaction and increased AKT phosphorylation. The P-AKT subsequent activated NFκB, which then bound to DNMT1 promoter and increased its expression. Finally, the upregulated DNMT1 promoted tumor suppressor genes hypermethylation with MGMT as a representative. In conclusion, CagA increased tumor suppressor genes hypermethylation via stimulating DNMT1 expression through the AKT-NFκB pathway.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation/genetics , Helicobacter pylori/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/genetics , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Cell Line, Tumor , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Enzyme Activation , Genes, Tumor Suppressor , Helicobacter Infections/microbiology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference , RNA, Small Interfering/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
5.
Oncotarget ; 5(21): 10584-95, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25301736

ABSTRACT

Androgen receptor (AR) plays an important role in many kinds of cancers. However, the molecular mechanisms of AR in gastric cancer (GC) are poorly characterized. Here, we investigated the role of AR in GC cell migration, invasion and metastatic potential. Our data showed that AR expression was positively correlated with lymph node metastasis and late TNM stages. These findings were accompanied by activation of AKT and upregulation of matrix metalloproteinase 9 (MMP9). AR overexpression induced increases in GC cell migration, invasion and proliferation in vitro and in vivo. These effects were attenuated by inhibition of AKT, AR and MMP9. AR overexpression upregulated MMP9 protein levels, whereas this effect was counteracted by AR siRNA. Inhibition of AKT by siRNA or an inhibitor (MK-2206 2HC) decreased AR protein expression in both stably transfected and parental SGC-7901 cells. Luciferase reporter and chromatin immunoprecipitation assays demonstrated that AR bound to the AR-binding sites of the MMP9 promoter. In summary, AR overexpression induced by AKT phosphorylation upregulated MMP9 by binding to its promoter region to promote gastric carcinogenesis. The AKT/AR/MMP9 pathway plays an important role in GC metastasis and may be a novel therapeutic target for GC treatment.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 9/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/metabolism , Stomach Neoplasms/pathology , Animals , Apoptosis , Base Sequence , Blotting, Western , Cell Adhesion , Female , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Matrix Metalloproteinase 9/chemistry , Matrix Metalloproteinase 9/genetics , Mice , Mice, Nude , Middle Aged , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Staging , Phosphorylation , Prognosis , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/genetics , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Transcriptional Activation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...