Cargando…

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 Buxus microphylla, was able to provoke mitophagy in lung cancer cells. CVB-D-induced mitophagy potenti...

Descripción completa

Detalles Bibliográficos
Autores principales: Zeng, Cheng, Zou, Tingting, Qu, Junyan, Chen, Xu, Zhang, Suping, Lin, Zhenghong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8199090/
https://www.ncbi.nlm.nih.gov/pubmed/34072333
http://dx.doi.org/10.3390/ijms22115820
_version_ 1783707294477844480
author Zeng, Cheng
Zou, Tingting
Qu, Junyan
Chen, Xu
Zhang, Suping
Lin, Zhenghong
author_facet Zeng, Cheng
Zou, Tingting
Qu, Junyan
Chen, Xu
Zhang, Suping
Lin, Zhenghong
author_sort Zeng, Cheng
collection PubMed
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 Buxus microphylla, 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
format Online
Article
Text
id pubmed-8199090
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-81990902021-06-14 Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells Zeng, Cheng Zou, Tingting Qu, Junyan Chen, Xu Zhang, Suping Lin, Zhenghong Int J Mol Sci Article 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 Buxus microphylla, 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 MDPI 2021-05-29 /pmc/articles/PMC8199090/ /pubmed/34072333 http://dx.doi.org/10.3390/ijms22115820 Text en © 2021 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Zeng, Cheng
Zou, Tingting
Qu, Junyan
Chen, Xu
Zhang, Suping
Lin, Zhenghong
Cyclovirobuxine D Induced-Mitophagy through the p65/BNIP3/LC3 Axis Potentiates Its Apoptosis-Inducing Effects in Lung Cancer Cells
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 Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8199090/
https://www.ncbi.nlm.nih.gov/pubmed/34072333
http://dx.doi.org/10.3390/ijms22115820
work_keys_str_mv AT zengcheng cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT zoutingting cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT qujunyan cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT chenxu cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT zhangsuping cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells
AT linzhenghong cyclovirobuxinedinducedmitophagythroughthep65bnip3lc3axispotentiatesitsapoptosisinducingeffectsinlungcancercells