Cargando…

Chemical or genetic Pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways

Hepatocellular carcinoma (HCC) is one of the most prevalent and malignant cancers with high inter- and intra-tumor heterogeneity. A central common signaling mechanism in cancer is proline-directed phosphorylation, which is further regulated by the unique proline isomerase Pin1. Pin1 is prevalently o...

Descripción completa

Detalles Bibliográficos
Autores principales: Liao, Xin-Hua, Zhang, Arina Li, Zheng, Min, Li, Mei-Qing, Chen, Champ Peng, Xu, Huijuan, Chu, Qing-Song, Yang, Dayun, Lu, Wenxian, Tsai, Ting-Fen, Liu, Hekun, Zhou, Xiao Zhen, Lu, Kun Ping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5337947/
https://www.ncbi.nlm.nih.gov/pubmed/28262728
http://dx.doi.org/10.1038/srep43639
_version_ 1782512472480022528
author Liao, Xin-Hua
Zhang, Arina Li
Zheng, Min
Li, Mei-Qing
Chen, Champ Peng
Xu, Huijuan
Chu, Qing-Song
Yang, Dayun
Lu, Wenxian
Tsai, Ting-Fen
Liu, Hekun
Zhou, Xiao Zhen
Lu, Kun Ping
author_facet Liao, Xin-Hua
Zhang, Arina Li
Zheng, Min
Li, Mei-Qing
Chen, Champ Peng
Xu, Huijuan
Chu, Qing-Song
Yang, Dayun
Lu, Wenxian
Tsai, Ting-Fen
Liu, Hekun
Zhou, Xiao Zhen
Lu, Kun Ping
author_sort Liao, Xin-Hua
collection PubMed
description Hepatocellular carcinoma (HCC) is one of the most prevalent and malignant cancers with high inter- and intra-tumor heterogeneity. A central common signaling mechanism in cancer is proline-directed phosphorylation, which is further regulated by the unique proline isomerase Pin1. Pin1 is prevalently overexpressed in human cancers including ~70% of HCC, and promotes tumorigenesis by activating multiple cancer-driving pathways. However, it was challenging to evaluate the significance of targeting Pin1 in cancer treatment until the recent identification of all-trans retinoic acid (ATRA) as a Pin1 inhibitor. Here we systematically investigate functions of Pin1 and its inhibitor ATRA in the development and treatment of HCC. Pin1 knockdown potently inhibited HCC cell proliferation and tumor growth in mice. ATRA-induced Pin1 degradation inhibited the growth of HCC cells, although at a higher IC50 as compared with breast cancer cells, likely due to more active ATRA metabolism in liver cells. Indeed, inhibition of ATRA metabolism enhanced the sensitivity of HCC cells to ATRA. Moreover, slow-releasing ATRA potently and dose-dependently inhibited HCC growth in mice. Finally, chemical or genetic Pin1 ablation blocked multiple cancer-driving pathways simultaneously in HCC cells. Thus, targeting Pin1 offers a promising therapeutic approach to simultaneously stop multiple cancer-driving pathways in HCC.
format Online
Article
Text
id pubmed-5337947
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-53379472017-03-08 Chemical or genetic Pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways Liao, Xin-Hua Zhang, Arina Li Zheng, Min Li, Mei-Qing Chen, Champ Peng Xu, Huijuan Chu, Qing-Song Yang, Dayun Lu, Wenxian Tsai, Ting-Fen Liu, Hekun Zhou, Xiao Zhen Lu, Kun Ping Sci Rep Article Hepatocellular carcinoma (HCC) is one of the most prevalent and malignant cancers with high inter- and intra-tumor heterogeneity. A central common signaling mechanism in cancer is proline-directed phosphorylation, which is further regulated by the unique proline isomerase Pin1. Pin1 is prevalently overexpressed in human cancers including ~70% of HCC, and promotes tumorigenesis by activating multiple cancer-driving pathways. However, it was challenging to evaluate the significance of targeting Pin1 in cancer treatment until the recent identification of all-trans retinoic acid (ATRA) as a Pin1 inhibitor. Here we systematically investigate functions of Pin1 and its inhibitor ATRA in the development and treatment of HCC. Pin1 knockdown potently inhibited HCC cell proliferation and tumor growth in mice. ATRA-induced Pin1 degradation inhibited the growth of HCC cells, although at a higher IC50 as compared with breast cancer cells, likely due to more active ATRA metabolism in liver cells. Indeed, inhibition of ATRA metabolism enhanced the sensitivity of HCC cells to ATRA. Moreover, slow-releasing ATRA potently and dose-dependently inhibited HCC growth in mice. Finally, chemical or genetic Pin1 ablation blocked multiple cancer-driving pathways simultaneously in HCC cells. Thus, targeting Pin1 offers a promising therapeutic approach to simultaneously stop multiple cancer-driving pathways in HCC. Nature Publishing Group 2017-03-06 /pmc/articles/PMC5337947/ /pubmed/28262728 http://dx.doi.org/10.1038/srep43639 Text en Copyright © 2017, The Author(s) http://creativecommons.org/licenses/by/4.0/ This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
spellingShingle Article
Liao, Xin-Hua
Zhang, Arina Li
Zheng, Min
Li, Mei-Qing
Chen, Champ Peng
Xu, Huijuan
Chu, Qing-Song
Yang, Dayun
Lu, Wenxian
Tsai, Ting-Fen
Liu, Hekun
Zhou, Xiao Zhen
Lu, Kun Ping
Chemical or genetic Pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways
title Chemical or genetic Pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways
title_full Chemical or genetic Pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways
title_fullStr Chemical or genetic Pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways
title_full_unstemmed Chemical or genetic Pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways
title_short Chemical or genetic Pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways
title_sort chemical or genetic pin1 inhibition exerts potent anticancer activity against hepatocellular carcinoma by blocking multiple cancer-driving pathways
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5337947/
https://www.ncbi.nlm.nih.gov/pubmed/28262728
http://dx.doi.org/10.1038/srep43639
work_keys_str_mv AT liaoxinhua chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT zhangarinali chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT zhengmin chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT limeiqing chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT chenchamppeng chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT xuhuijuan chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT chuqingsong chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT yangdayun chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT luwenxian chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT tsaitingfen chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT liuhekun chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT zhouxiaozhen chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways
AT lukunping chemicalorgeneticpin1inhibitionexertspotentanticanceractivityagainsthepatocellularcarcinomabyblockingmultiplecancerdrivingpathways