Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathway

Abstract Background Ovarian cancer is a deadly disease. Inhibitors of apoptosis proteins (IAPs) are key regulators of apoptosis and are frequently dysregulated in ovarian cancer. Overexpression of IAPs proteins has been correlated with tumorigenesis, treatment resistance and poor prognosis. Reinstal...

Full description

Bibliographic Details
Main Authors: Bao-Xia Li, Heng-Bang Wang, Miao-Zhen Qiu, Qiu-Yun Luo, Han-Jie Yi, Xiang-Lei Yan, Wen-Tao Pan, Lu-Ping Yuan, Yu-Xin Zhang, Jian-Hua Xu, Lin Zhang, Da-Jun Yang
Format: Article
Language:English
Published: BMC 2018-03-01
Series:Journal of Experimental & Clinical Cancer Research
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13046-018-0703-9
id doaj-6c2b90efb8c94f359f4fbbbc557f9f6a
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Bao-Xia Li
Heng-Bang Wang
Miao-Zhen Qiu
Qiu-Yun Luo
Han-Jie Yi
Xiang-Lei Yan
Wen-Tao Pan
Lu-Ping Yuan
Yu-Xin Zhang
Jian-Hua Xu
Lin Zhang
Da-Jun Yang
spellingShingle Bao-Xia Li
Heng-Bang Wang
Miao-Zhen Qiu
Qiu-Yun Luo
Han-Jie Yi
Xiang-Lei Yan
Wen-Tao Pan
Lu-Ping Yuan
Yu-Xin Zhang
Jian-Hua Xu
Lin Zhang
Da-Jun Yang
Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathway
Journal of Experimental & Clinical Cancer Research
APG-1387
Apoptosis
Autophagy
Ovarian cancer
author_facet Bao-Xia Li
Heng-Bang Wang
Miao-Zhen Qiu
Qiu-Yun Luo
Han-Jie Yi
Xiang-Lei Yan
Wen-Tao Pan
Lu-Ping Yuan
Yu-Xin Zhang
Jian-Hua Xu
Lin Zhang
Da-Jun Yang
author_sort Bao-Xia Li
title Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathway
title_short Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathway
title_full Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathway
title_fullStr Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathway
title_full_unstemmed Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathway
title_sort novel smac mimetic apg-1387 elicits ovarian cancer cell killing through tnf-alpha, ripoptosome and autophagy mediated cell death pathway
publisher BMC
series Journal of Experimental & Clinical Cancer Research
issn 1756-9966
publishDate 2018-03-01
description Abstract Background Ovarian cancer is a deadly disease. Inhibitors of apoptosis proteins (IAPs) are key regulators of apoptosis and are frequently dysregulated in ovarian cancer. Overexpression of IAPs proteins has been correlated with tumorigenesis, treatment resistance and poor prognosis. Reinstalling functional cell death machinery by pharmacological inhibition of IAPs proteins may represent an attractive therapeutic strategy for treatment of ovarian cancer. Methods CCK-8 and colony formation assay was performed to examine cytotoxic activity. Apoptosis was analyzed by fluorescence microscopy, flow cytometry and TUNEL assay. Elisa assay was used to determine TNFα protein. Caspase activity assay was used for caspase activation evaluation. Immunoprecipitation and siRNA interference were carried out for functional analysis. Western blotting analysis were carried out to test protein expression. Ovarian cancer cell xenograft nude mice model was used for in vivo efficacy evaluation. Results APG-1387 demonstrated potent inhibitory effect on ovarian cancer cell growth and clonogenic cell survival. APG-1387 induced RIP1- and TNFα-dependent apoptotic cell death in ovarian cancer through downregulation of IAPs proteins and induction of caspase-8/FADD/RIP1 complex, which drives caspase-8 activation. NF-κB signaling pathway was activated upon APG-1387 treatment and RIP1 contributed to NF-κB activation. APG-1387 induced cytoprotective autophagy while triggering apoptosis in ovarian cancer cells and inhibition of autophagy enhanced APG-1387-induced apoptotic cell death. APG-1387 exhibited potent antitumor activity against established human ovarian cancer xenografts. Conclusions Our results demonstrate that APG-1387 targets IAPs proteins to potently elicit apoptotic cell death in vitro and in vivo, and provide mechanistic and applicable rationale for future clinical evaluation of APG-1387 in ovarian cancer.
topic APG-1387
Apoptosis
Autophagy
Ovarian cancer
url http://link.springer.com/article/10.1186/s13046-018-0703-9
work_keys_str_mv AT baoxiali novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT hengbangwang novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT miaozhenqiu novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT qiuyunluo novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT hanjieyi novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT xiangleiyan novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT wentaopan novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT lupingyuan novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT yuxinzhang novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT jianhuaxu novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT linzhang novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
AT dajunyang novelsmacmimeticapg1387elicitsovariancancercellkillingthroughtnfalpharipoptosomeandautophagymediatedcelldeathpathway
_version_ 1724882082097790976
spelling doaj-6c2b90efb8c94f359f4fbbbc557f9f6a2020-11-25T02:18:27ZengBMCJournal of Experimental & Clinical Cancer Research1756-99662018-03-0137111510.1186/s13046-018-0703-9Novel smac mimetic APG-1387 elicits ovarian cancer cell killing through TNF-alpha, Ripoptosome and autophagy mediated cell death pathwayBao-Xia Li0Heng-Bang Wang1Miao-Zhen Qiu2Qiu-Yun Luo3Han-Jie Yi4Xiang-Lei Yan5Wen-Tao Pan6Lu-Ping Yuan7Yu-Xin Zhang8Jian-Hua Xu9Lin Zhang10Da-Jun Yang11State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterDepartment of Pharmacology, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical UniversityDepartment of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterDepartment of Pharmacology, Fujian Provincial Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical UniversityDepartments of Clinical Laboratory, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer CenterState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer CenterAbstract Background Ovarian cancer is a deadly disease. Inhibitors of apoptosis proteins (IAPs) are key regulators of apoptosis and are frequently dysregulated in ovarian cancer. Overexpression of IAPs proteins has been correlated with tumorigenesis, treatment resistance and poor prognosis. Reinstalling functional cell death machinery by pharmacological inhibition of IAPs proteins may represent an attractive therapeutic strategy for treatment of ovarian cancer. Methods CCK-8 and colony formation assay was performed to examine cytotoxic activity. Apoptosis was analyzed by fluorescence microscopy, flow cytometry and TUNEL assay. Elisa assay was used to determine TNFα protein. Caspase activity assay was used for caspase activation evaluation. Immunoprecipitation and siRNA interference were carried out for functional analysis. Western blotting analysis were carried out to test protein expression. Ovarian cancer cell xenograft nude mice model was used for in vivo efficacy evaluation. Results APG-1387 demonstrated potent inhibitory effect on ovarian cancer cell growth and clonogenic cell survival. APG-1387 induced RIP1- and TNFα-dependent apoptotic cell death in ovarian cancer through downregulation of IAPs proteins and induction of caspase-8/FADD/RIP1 complex, which drives caspase-8 activation. NF-κB signaling pathway was activated upon APG-1387 treatment and RIP1 contributed to NF-κB activation. APG-1387 induced cytoprotective autophagy while triggering apoptosis in ovarian cancer cells and inhibition of autophagy enhanced APG-1387-induced apoptotic cell death. APG-1387 exhibited potent antitumor activity against established human ovarian cancer xenografts. Conclusions Our results demonstrate that APG-1387 targets IAPs proteins to potently elicit apoptotic cell death in vitro and in vivo, and provide mechanistic and applicable rationale for future clinical evaluation of APG-1387 in ovarian cancer.http://link.springer.com/article/10.1186/s13046-018-0703-9APG-1387ApoptosisAutophagyOvarian cancer