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 Oncol ; 52(5): 1465-1478, 2018 May.
Article in English | MEDLINE | ID: mdl-29568964

ABSTRACT

Osteosarcoma is the most common primary malignancy of the bone and is characterized by local invasion and distant metastasis. Over the past 20 years, long-term outcomes have reached a plateau even with aggressive therapy. Overexpression of insulin-like growth factor 1 receptor (IGF­1R) is associated with tumor proliferation, invasion and migration in osteosarcoma. In the present study, our group developed a novel quinazoline derivative, 6-fluoro­2-(3-fluorophenyl)-4-(cyanoanilino)quinazoline (HMJ­30), in order to disrupt IGF­1R signaling and tumor invasiveness in osteosarcoma U­2 OS cells. Molecular modeling, immune-precipitation, western blotting and phosphorylated protein kinase sandwich ELISA assays were used to confirm this hypothesis. The results demonstrated that HMJ­30 selectively targeted the ATP-binding site of IGF­1R and inhibited its downstream phosphoinositide 3-kinase/protein kinase B, Ras/mitogen-activated protein kinase, and IκK/nuclear factor-κB signaling pathways in U­2 OS cells. HMJ­30 inhibited U­2 OS cell invasion and migration and downregulated protein levels and activities of matrix metalloproteinase (MMP)­2 and MMP-9. An increase in protein levels of tissue inhibitor of metalloproteinase (TIMP)­1 and TIMP­2 was also observed. Furthermore, HMJ­30 caused U­2 OS cells to aggregate and form tight clusters, and these cells were flattened, less elongated and displayed cobblestone-like shapes. There was an increase in epithelial markers and a decrease in mesenchymal markers, indicating that the cells underwent the reverse epithelial-mesenchymal transition (EMT) process. Overall, these results demonstrated the potential molecular mechanisms underlying the effects of HMJ­30 on invasiveness and EMT in U­2 OS cells, suggesting that this compound deserves further investigation as a potential anti-osteosarcoma drug.

2.
Oncol Rep ; 32(2): 597-606, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24919794

ABSTRACT

The aim of the present study was to explore the effect of 6-fluoro-2-(3-fluorophenyl)-4-(cyanoanilino) quinazoline (HMJ-30) on the anti-angiogenic properties and apoptosis-related mechanism of human umbilical vein endothelial cells (HUVECs). In this study, HMJ-30 dose- and time-dependently inhibited the viability of HUVECs. We also found that HMJ-30 enhanced disruption of tube-like structures and suppressed cell migration in HUVECs after vascular endothelial growth factor (VEGF) induction. HMJ-30 was also observed to inhibit vessel branching and sprouting in chicken chorioallantoic membrane (CAM). Microsprouting induced by VEGF in the rat aortic ring and blood vessel formation in a mouse Matrigel plug were individually suppressed by HMJ-30. In an in vitro study, HMJ-30 induced the apoptotic death of HUVECs as indicated by DNA fragmentation and promoted reactive oxygen species (ROS) production as determined by flow cytometric assay. In addition, extrinsic caspase signaling (caspase-8 and -3) was activated in the HMJ-30-treated HUVECs and their inhibitors were applied to assess the signal transduction. We investigated the upstream of the death receptor pathway and further observed that the levels of death receptor 5 (DR5) and phosphorylated c-Jun N-terminal kinase (JNK) signals were upregulated in HUVECs following HMJ-30 challenge, which was confirmed by a JNK-specific inhibitor (SP600125). Hence, HMJ-30-induced endothelial cell apoptosis involved the ROS/JNK-regulated DR5 pathway. In summary, HMJ-30 may provide a potential therapeutic effect for the anti-vascular targeting of angiogenesis during cancer treatment.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Endothelial Cells/drug effects , MAP Kinase Signaling System/drug effects , Neovascularization, Physiologic/drug effects , Quinazolines/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Anthracenes/pharmacology , Aorta/cytology , Aorta/drug effects , Apoptosis/drug effects , Cell Movement/drug effects , Cells, Cultured , Chick Embryo , DNA/analysis , DNA/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Vascular Endothelial Growth Factor A/metabolism
3.
J Orthop Res ; 29(9): 1448-56, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21425328

ABSTRACT

Human osteogenic sarcoma is the most common primary bone tumor. Despite of the success of frontline therapy, about 40% of patients have disease progression and further therapy is palliative and toxic. In this study, we developed a novel quinazoline HMJ-30 to investigate the cell growth inhibition and apoptotic responses in U-2 OS human osteogenic sarcoma cells. Our results demonstrated that HMJ-30 significantly reduced cell viabilities of U-2 OS, HOS, and 143B cells in a dose-dependent manner, but it exhibited low cytotoxicity in normal hFOB cells. HMJ-30 induced DNA damage and apoptosis in U-2 OS cells as revealed by morphologic changes, comet assay and DAPI staining. Immuno-staining, colorimetric assays, and Western blotting analyses indicated that activities of caspase-8, caspase-9, and caspase-3 and the levels of Bcl-2 family-related proteins (Bcl-2, Mcl-1, Bax, BAD, and t-Bid) were altered in HMJ-30-treated U-2 OS cells. Pretreatment of cells with caspase-8, -9, and -3 specific inhibitors significantly reduced the cell growth inhibition. HMJ-30-induced apoptosis was mediated through both death-receptor and mitochondria-dependent apoptotic pathways in U-2 OS cells. HMJ-30 induced early phosphorylation of p53(Ser18) was through the activation of ataxia telangiectasia mutated (ATM) in U-2 OS cells. The cell growth inhibition by HMJ-30 was substantially attenuated either by the pre-incubation of U-2 OS cells with N-acetylcysteine (NAC, an antioxidant) and caffeine (an ATM kinase inhibitor) or by p53 knockdown via RNAi. In conclusion, ROS dependent-ATM/p53 signaling pathway is involved in HMJ-30-induced apoptosis in U-2 OS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/biosynthesis , DNA-Binding Proteins/biosynthesis , Osteosarcoma/drug therapy , Oxidative Stress/drug effects , Protein Serine-Threonine Kinases/biosynthesis , Quinazolines/pharmacology , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Ataxia Telangiectasia Mutated Proteins , Caspases/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Comet Assay , DNA Damage , DNA, Neoplasm/drug effects , DNA-Binding Proteins/genetics , Humans , Osteosarcoma/metabolism , Osteosarcoma/pathology , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...