Cargando…

Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells

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 b...

Descripción completa

Detalles Bibliográficos
Autores principales: Yue, Qingxi, Zhen, Hong, Huang, Ming, Zheng, Xi, Feng, Lixing, Jiang, Baohong, Yang, Min, Wu, Wanying, Liu, Xuan, Guo, Dean
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4948917/
https://www.ncbi.nlm.nih.gov/pubmed/27428326
http://dx.doi.org/10.1371/journal.pone.0159034
_version_ 1782443353546162176
author Yue, Qingxi
Zhen, Hong
Huang, Ming
Zheng, Xi
Feng, Lixing
Jiang, Baohong
Yang, Min
Wu, Wanying
Liu, Xuan
Guo, Dean
author_facet Yue, Qingxi
Zhen, Hong
Huang, Ming
Zheng, Xi
Feng, Lixing
Jiang, Baohong
Yang, Min
Wu, Wanying
Liu, Xuan
Guo, Dean
author_sort Yue, Qingxi
collection PubMed
description 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 α1 and α3 subunits and increased the expression of WEE1 in HeLa cells. Antibodies against Na, K-ATPase α1 and α3 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 α1 and α3 subunits and the inhibitive effect of arenobufagin on proteasomal activity of HeLa cells might be related to its binding with Na, K-ATPase.
format Online
Article
Text
id pubmed-4948917
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-49489172016-08-01 Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells Yue, Qingxi Zhen, Hong Huang, Ming Zheng, Xi Feng, Lixing Jiang, Baohong Yang, Min Wu, Wanying Liu, Xuan Guo, Dean PLoS One Research Article 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 α1 and α3 subunits and increased the expression of WEE1 in HeLa cells. Antibodies against Na, K-ATPase α1 and α3 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 α1 and α3 subunits and the inhibitive effect of arenobufagin on proteasomal activity of HeLa cells might be related to its binding with Na, K-ATPase. Public Library of Science 2016-07-18 /pmc/articles/PMC4948917/ /pubmed/27428326 http://dx.doi.org/10.1371/journal.pone.0159034 Text en © 2016 Yue et al http://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Yue, Qingxi
Zhen, Hong
Huang, Ming
Zheng, Xi
Feng, Lixing
Jiang, Baohong
Yang, Min
Wu, Wanying
Liu, Xuan
Guo, Dean
Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells
title Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells
title_full Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells
title_fullStr Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells
title_full_unstemmed Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells
title_short Proteasome Inhibition Contributed to the Cytotoxicity of Arenobufagin after Its Binding with Na, K-ATPase in Human Cervical Carcinoma HeLa Cells
title_sort proteasome inhibition contributed to the cytotoxicity of arenobufagin after its binding with na, k-atpase in human cervical carcinoma hela cells
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4948917/
https://www.ncbi.nlm.nih.gov/pubmed/27428326
http://dx.doi.org/10.1371/journal.pone.0159034
work_keys_str_mv AT yueqingxi proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT zhenhong proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT huangming proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT zhengxi proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT fenglixing proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT jiangbaohong proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT yangmin proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT wuwanying proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT liuxuan proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells
AT guodean proteasomeinhibitioncontributedtothecytotoxicityofarenobufaginafteritsbindingwithnakatpaseinhumancervicalcarcinomahelacells