Cargando…

Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect

Cardiac glycosides (CGs) are candidate anticancer agents that function by increasing [Ca(2+)]i to induce apoptotic cell death in several types of cancer cells. However, new findings have shown that the anti-cancer effects of CGs involve complex cell-signal transduction mechanisms. Hence, exploring t...

Descripción completa

Detalles Bibliográficos
Autores principales: Hao, Yuanyuan, Song, Tao, Wang, Mingye, Li, Tongtong, Zhao, Chi, Li, Ting, Hou, Yunlong, He, Hongjiang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9946806/
https://www.ncbi.nlm.nih.gov/pubmed/36825592
http://dx.doi.org/10.3892/ijo.2023.5492
_version_ 1784892412671295488
author Hao, Yuanyuan
Song, Tao
Wang, Mingye
Li, Tongtong
Zhao, Chi
Li, Ting
Hou, Yunlong
He, Hongjiang
author_facet Hao, Yuanyuan
Song, Tao
Wang, Mingye
Li, Tongtong
Zhao, Chi
Li, Ting
Hou, Yunlong
He, Hongjiang
author_sort Hao, Yuanyuan
collection PubMed
description Cardiac glycosides (CGs) are candidate anticancer agents that function by increasing [Ca(2+)]i to induce apoptotic cell death in several types of cancer cells. However, new findings have shown that the anti-cancer effects of CGs involve complex cell-signal transduction mechanisms. Hence, exploring the potential mechanisms of action of CGs may provide insight into their anti-cancer effects and thus aid in the selection of the appropriate CG. Periplocymarin (PPM), which is a cardiac glycoside, is an active ingredient extracted from Cortex periplocae. The role of PPM was evaluated in HepG2 cells and xenografted nude mice. Cell proliferation, real-time ATP rate assays, western blotting, cell apoptosis assays, short interfering RNA transfection, the patch clamp technique, electron microscopy, JC-1 staining, immunofluorescence staining and autophagic flux assays were performed to evaluate the function and regulatory mechanisms of PPM in vitro. The in vivo activity of the PPM was assessed using a mouse xenograft model. The present study demonstrated that PPM synchronously activated lethal apoptosis and protective autophagy in liver cancer, and the initiation of autophagy counteracted the inherent pro-apoptotic capacity and impaired the anti-cancer effects. Specifically, PPM exerted a proapoptotic effect in HepG2 cells and activated macroautophagy by initiation of the AMPK/ULK1 and mTOR signaling pathways. Activation of macroautophagy counteracted the pro-apoptotic effects of PPM, but when it was combined with an autophagy inhibitor, the anti-cancer effects of PPM in mice bearing HepG2 xenografts were observed. Collectively, these results indicated that a self-limiting effect impaired the pro-apoptotic effects of PPM in liver cancer, but when combined with an autophagy inhibitor, it may serve as a novel therapeutic option for the management of liver cancer.
format Online
Article
Text
id pubmed-9946806
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-99468062023-02-24 Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect Hao, Yuanyuan Song, Tao Wang, Mingye Li, Tongtong Zhao, Chi Li, Ting Hou, Yunlong He, Hongjiang Int J Oncol Articles Cardiac glycosides (CGs) are candidate anticancer agents that function by increasing [Ca(2+)]i to induce apoptotic cell death in several types of cancer cells. However, new findings have shown that the anti-cancer effects of CGs involve complex cell-signal transduction mechanisms. Hence, exploring the potential mechanisms of action of CGs may provide insight into their anti-cancer effects and thus aid in the selection of the appropriate CG. Periplocymarin (PPM), which is a cardiac glycoside, is an active ingredient extracted from Cortex periplocae. The role of PPM was evaluated in HepG2 cells and xenografted nude mice. Cell proliferation, real-time ATP rate assays, western blotting, cell apoptosis assays, short interfering RNA transfection, the patch clamp technique, electron microscopy, JC-1 staining, immunofluorescence staining and autophagic flux assays were performed to evaluate the function and regulatory mechanisms of PPM in vitro. The in vivo activity of the PPM was assessed using a mouse xenograft model. The present study demonstrated that PPM synchronously activated lethal apoptosis and protective autophagy in liver cancer, and the initiation of autophagy counteracted the inherent pro-apoptotic capacity and impaired the anti-cancer effects. Specifically, PPM exerted a proapoptotic effect in HepG2 cells and activated macroautophagy by initiation of the AMPK/ULK1 and mTOR signaling pathways. Activation of macroautophagy counteracted the pro-apoptotic effects of PPM, but when it was combined with an autophagy inhibitor, the anti-cancer effects of PPM in mice bearing HepG2 xenografts were observed. Collectively, these results indicated that a self-limiting effect impaired the pro-apoptotic effects of PPM in liver cancer, but when combined with an autophagy inhibitor, it may serve as a novel therapeutic option for the management of liver cancer. D.A. Spandidos 2023-02-21 /pmc/articles/PMC9946806/ /pubmed/36825592 http://dx.doi.org/10.3892/ijo.2023.5492 Text en Copyright: © Hao et al. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Hao, Yuanyuan
Song, Tao
Wang, Mingye
Li, Tongtong
Zhao, Chi
Li, Ting
Hou, Yunlong
He, Hongjiang
Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect
title Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect
title_full Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect
title_fullStr Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect
title_full_unstemmed Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect
title_short Dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect
title_sort dual targets of lethal apoptosis and protective autophagy in liver cancer with periplocymarin elicit a limited therapeutic effect
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9946806/
https://www.ncbi.nlm.nih.gov/pubmed/36825592
http://dx.doi.org/10.3892/ijo.2023.5492
work_keys_str_mv AT haoyuanyuan dualtargetsoflethalapoptosisandprotectiveautophagyinlivercancerwithperiplocymarinelicitalimitedtherapeuticeffect
AT songtao dualtargetsoflethalapoptosisandprotectiveautophagyinlivercancerwithperiplocymarinelicitalimitedtherapeuticeffect
AT wangmingye dualtargetsoflethalapoptosisandprotectiveautophagyinlivercancerwithperiplocymarinelicitalimitedtherapeuticeffect
AT litongtong dualtargetsoflethalapoptosisandprotectiveautophagyinlivercancerwithperiplocymarinelicitalimitedtherapeuticeffect
AT zhaochi dualtargetsoflethalapoptosisandprotectiveautophagyinlivercancerwithperiplocymarinelicitalimitedtherapeuticeffect
AT liting dualtargetsoflethalapoptosisandprotectiveautophagyinlivercancerwithperiplocymarinelicitalimitedtherapeuticeffect
AT houyunlong dualtargetsoflethalapoptosisandprotectiveautophagyinlivercancerwithperiplocymarinelicitalimitedtherapeuticeffect
AT hehongjiang dualtargetsoflethalapoptosisandprotectiveautophagyinlivercancerwithperiplocymarinelicitalimitedtherapeuticeffect