Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells

Mitophagy plays a pro-survival or pro-death role that is cellular-context- and stress-condition-dependent. In this study, we revealed that cyclovirobuxine D (CVB-D), a natural compound derived from <i>Buxus microphylla</i>, was able to provoke mitophagy in lung cancer cells. CVB-D-induce...

Full description

Bibliographic Details
Main Authors: Cheng Zeng, Tingting Zou, Junyan Qu, Xu Chen, Suping Zhang, Zhenghong Lin
Format: Article
Language:English
Published: MDPI AG 2021-05-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/22/11/5820
_version_ 1797532154523025408
author Cheng Zeng
Tingting Zou
Junyan Qu
Xu Chen
Suping Zhang
Zhenghong Lin
author_facet Cheng Zeng
Tingting Zou
Junyan Qu
Xu Chen
Suping Zhang
Zhenghong Lin
author_sort Cheng Zeng
collection DOAJ
description Mitophagy plays a pro-survival or pro-death role that is cellular-context- and stress-condition-dependent. In this study, we revealed that cyclovirobuxine D (CVB-D), a natural compound derived from <i>Buxus microphylla</i>, was able to provoke mitophagy in lung cancer cells. CVB-D-induced mitophagy potentiates apoptosis by promoting mitochondrial dysfunction. Mechanistically, CVB-D initiates mitophagy by enhancing the expression of the mitophagy receptor BNIP3 and strengthening its interaction with LC3 to provoke mitophagy. Our results further showed that p65, a transcriptional suppressor of BNIP3, is downregulated upon CVB-D treatment. The ectopic expression of p65 inhibits BNIP3 expression, while its knockdown significantly abolishes its transcriptional repression on BNIP3 upon CVB-D treatment. Importantly, nude mice bearing subcutaneous xenograft tumors presented retarded growth upon CVB-D treatment. Overall, we demonstrated that CVB-D treatment can provoke mitophagy and further revealed that the p65/BNIP3/LC3 axis is one potential mechanism involved in CVB-D-induced mitophagy in lung cancer cells, thus providing an effective antitumor therapeutic strategy for the treatment of lung cancer patients
first_indexed 2024-03-10T10:55:05Z
format Article
id doaj.art-e67ca3da887e4b2ea5e889d00cf81f79
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-10T10:55:05Z
publishDate 2021-05-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-e67ca3da887e4b2ea5e889d00cf81f792023-11-21T21:58:03ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672021-05-012211582010.3390/ijms22115820Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer CellsCheng Zeng0Tingting Zou1Junyan Qu2Xu Chen3Suping Zhang4Zhenghong Lin5School of Life Sciences, Chongqing University, Chongqing 401331, ChinaSchool of Life Sciences, Chongqing University, Chongqing 401331, ChinaSchool of Life Sciences, Chongqing University, Chongqing 401331, ChinaSchool of Life Sciences, Chongqing University, Chongqing 401331, ChinaShenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Department of Pharmacology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen 518055, ChinaSchool of Life Sciences, Chongqing University, Chongqing 401331, ChinaMitophagy plays a pro-survival or pro-death role that is cellular-context- and stress-condition-dependent. In this study, we revealed that cyclovirobuxine D (CVB-D), a natural compound derived from <i>Buxus microphylla</i>, was able to provoke mitophagy in lung cancer cells. CVB-D-induced mitophagy potentiates apoptosis by promoting mitochondrial dysfunction. Mechanistically, CVB-D initiates mitophagy by enhancing the expression of the mitophagy receptor BNIP3 and strengthening its interaction with LC3 to provoke mitophagy. Our results further showed that p65, a transcriptional suppressor of BNIP3, is downregulated upon CVB-D treatment. The ectopic expression of p65 inhibits BNIP3 expression, while its knockdown significantly abolishes its transcriptional repression on BNIP3 upon CVB-D treatment. Importantly, nude mice bearing subcutaneous xenograft tumors presented retarded growth upon CVB-D treatment. Overall, we demonstrated that CVB-D treatment can provoke mitophagy and further revealed that the p65/BNIP3/LC3 axis is one potential mechanism involved in CVB-D-induced mitophagy in lung cancer cells, thus providing an effective antitumor therapeutic strategy for the treatment of lung cancer patientshttps://www.mdpi.com/1422-0067/22/11/5820cyclovirobuxine DBNIP3mitophagyapoptosislung cancer
spellingShingle Cheng Zeng
Tingting Zou
Junyan Qu
Xu Chen
Suping Zhang
Zhenghong Lin
Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells
International Journal of Molecular Sciences
cyclovirobuxine D
BNIP3
mitophagy
apoptosis
lung cancer
title Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells
title_full Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells
title_fullStr Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells
title_full_unstemmed Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells
title_short Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells
title_sort cyclovirobuxine d induced mitophagy through the p65 bnip3 lc3 axis potentiates its apoptosis inducing effects in lung cancer cells
topic cyclovirobuxine D
BNIP3
mitophagy
apoptosis
lung cancer
url https://www.mdpi.com/1422-0067/22/11/5820
work_keys_str_mv AT chengzeng cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT tingtingzou cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT junyanqu cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT xuchen cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT supingzhang cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT zhenghonglin cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells