Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells
Yue, Qingxi3,4; Zhen, Hong4; Huang, Ming2,5; Zheng, Xi1; Feng, Lixing3; Jiang, Baohong3; Yang, Min3; Wu, Wanying3; Liu, Xuan3; Guo, Dean3
刊名PLOS ONE
2016-07-18
卷号11期号:7
ISSN号1932-6203
DOI10.1371/journal.pone.0159034
文献子类Article
英文摘要Although the possibility of developing cardiac steroids/ cardiac glycosides as novel cancer therapeutic agents has been recognized, the mechanism of their anticancer activity is still not clear enough. Toad venom extract containing bufadienolides, which belong to cardiac steroids, has actually long been used as traditional Chinese medicine in clinic for cancer therapy in China. The cytotoxicity of arenobufagin, a bufadienolide isolated from toad venom, on human cervical carcinoma HeLa cells was checked. And, the protein expression profile of control HeLa cells and HeLa cells treated with arenobufagin for 48 h was analyzed using two-dimensional electrophoresis, respectively. Differently expressed proteins in HeLa cells treated with arenobufagin were identified and the pathways related to these proteins were mapped from KEGG database. Computational molecular docking was performed to verify the binding of arenobufagin and Na, K-ATPase. The effects of arenobufagin on Na, K-ATPase activity and proteasome activity of HeLa cells were checked. The protein-protein interaction network between Na, K-ATPase and proteasome was constructed and the expression of possible intermediate proteins ataxin-1 and translationally-controlled tumor protein in HeLa cells treated with arenobufagin was then checked. Arenobufagin induced apoptosis and G2/M cell cycle arrest in HeLa cells. The cytotoxic effect of arenobufagin was associated with 25 differently expressed proteins including proteasome-related proteins, calcium ion binding-related proteins, oxidative stress-related proteins, metabolism-related enzymes and others. The results of computational molecular docking revealed that arenobufagin was bound in the cavity formed by the transmembrane alpha subunits of Na, K-ATPase, which blocked the pathway of extracellular Na+/K+ cation exchange and inhibited the function of ion exchange. Arenobufagin inhibited the activity of Na, K-ATPase and proteasome, decreased the expression of Na, K-ATPase alpha 1 and alpha 3 subunits and increased the expression of WEE1 in HeLa cells. Antibodies against Na, K-ATPase alpha 1 and alpha 3 subunits alone or combinated with arenobufagin also inhibited the activity of proteasome. Furthermore, the expression of the possible intermediate proteins ataxin-1 and translationally-controlled tumor protein was increased in HeLa cells treated with arenobufagin by flow cytometry analysis, respectively. These results indicated that arenobufagin might directly bind with Na, K-ATPase alpha 1 and alpha 3 subunits and the inhibitive effect of arenobufagin on proteasomal activity of HeLa cells might be related to its binding with Na, K-ATPase.
资助项目Twelfth Five-Year National Science & Technology Support Program[2012BAI29B06] ; Shanghai Science & Technology Support Program[13431900401] ; China Postdoctoral Science Foundation funded project[2012M510907] ; Shanghai Postdoctoral Scientific Program[13R21417800] ; Postdoctor Research Program of Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences[2012KIP516] ; Sanofi-Aventis-Shanghai Institutes for Biological Sciences Scholarship Program[00000000] ; National Nature Science Foundation[81302809] ; National Nature Science Foundation[81373964] ; Research Program of Shanghai Municipal Commission of Health and Family Planning[20154Y0149] ; grant constructed multi-disciplinary team for oncology in No. 3 People's Hospital, School of Medicine, Shanghai Jiaotong University[00000000] ; [syzrc2014-001] ; [syz2014-005] ; [YG2015MS24]
WOS关键词NA+/K+-ATPASE ; HEPATOCELLULAR-CARCINOMA ; CARDIAC-GLYCOSIDES ; TOAD VENOM ; CARDIOTONIC STEROIDS ; GLIOBLASTOMA CELLS ; CANCER-TREATMENT ; CYCLE ARREST ; LUNG-CANCER ; APOPTOSIS
WOS研究方向Science & Technology - Other Topics
语种英语
出版者PUBLIC LIBRARY SCIENCE
WOS记录号WOS:000380169300031
内容类型期刊论文
源URL[http://119.78.100.183/handle/2S10ELR8/275954]  
专题上海中药现代化研究中心
通讯作者Yue, Qingxi; Liu, Xuan; Guo, Dean
作者单位1.Rutgers State Univ, Ernest Mario Sch Pharm, Dept Biol Chem, Piscataway, NJ 08854 USA
2.Rutgers State Univ, Ctr Integrat Prote Res, BioMaPS Inst, Piscataway, NJ 08854 USA;
3.Chinese Acad Sci, Shanghai Inst Biol Sci, Shanghai Inst Mat Med, Shanghai Res Ctr Modernizat Tradit Chinese Med, Shanghai 201203, Peoples R China;
4.Shanghai Jiao Tong Univ, Sch Med, Shanghai Peoples Hosp 9, Inst Oncol, Shanghai 201999, Peoples R China;
5.Rutgers State Univ, Dept Chem & Chem Biol, Piscataway, NJ 08854 USA;
推荐引用方式
GB/T 7714
Yue, Qingxi,Zhen, Hong,Huang, Ming,et al. Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells[J]. PLOS ONE,2016,11(7).
APA Yue, Qingxi.,Zhen, Hong.,Huang, Ming.,Zheng, Xi.,Feng, Lixing.,...&Guo, Dean.(2016).Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells.PLOS ONE,11(7).
MLA Yue, Qingxi,et al."Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells".PLOS ONE 11.7(2016).
个性服务
查看访问统计
相关权益政策
暂无数据
收藏/分享
所有评论 (0)
暂无评论
 

除非特别说明,本系统中所有内容都受版权保护,并保留所有权利。


©版权所有 ©2017 CSpace - Powered by CSpace