Cargando…

Docetaxel enhances lysosomal function through TFEB activation

Docetaxel is an effective and commonly used chemotherapeutic drug for cancer. Autophagy has been reported to be involved in the anticancer mechanism of docetaxel. However, the effect of docetaxel on lysosomal function remains elusive. In the present study, we first found that docetaxel treatment enh...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Jianbin, Wang, Jigang, Wong, Yin Kwan, Sun, Xin, Chen, Yun, Wang, Liming, Yang, Liu, Lu, Liqin, Shen, Han-Ming, Huang, Dongsheng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5966422/
https://www.ncbi.nlm.nih.gov/pubmed/29795139
http://dx.doi.org/10.1038/s41419-018-0571-4
_version_ 1783325454196801536
author Zhang, Jianbin
Wang, Jigang
Wong, Yin Kwan
Sun, Xin
Chen, Yun
Wang, Liming
Yang, Liu
Lu, Liqin
Shen, Han-Ming
Huang, Dongsheng
author_facet Zhang, Jianbin
Wang, Jigang
Wong, Yin Kwan
Sun, Xin
Chen, Yun
Wang, Liming
Yang, Liu
Lu, Liqin
Shen, Han-Ming
Huang, Dongsheng
author_sort Zhang, Jianbin
collection PubMed
description Docetaxel is an effective and commonly used chemotherapeutic drug for cancer. Autophagy has been reported to be involved in the anticancer mechanism of docetaxel. However, the effect of docetaxel on lysosomal function remains elusive. In the present study, we first found that docetaxel treatment enhances autophagic flux in different cancer cells. Moreover, docetaxel treatment activates lysosomal function and promotes its fusion with autophagosome. Second, doctaxel treatment activates TFEB (transcription factor EB), a key nuclear transcription factor in control of lysosome biogenesis and function. We found that docetaxel promotes TFEB nuclear translocation and increases its transcriptional activity while knockdown of TFEB impairs lysosomal activation by docetaxel. Thirdly, TFEB activation by docetaxel is mediated by ROS (reactive oxygen species) generation and scavenging of ROS suppresses TFEB activity and lysosomal function in docetaxel-treated cells. Finally, inhibition of lysosomal function leads to increased docetaxel-induced cell death, suggesting that lysosomal activation protects against docetaxel-mediated apoptosis. Taken together, our results provide novel insights into the regulatory mechanisms of docetaxel on lysosomes, which could facilitate the development of novel potential cancer therapeutic agents via lysosomal inhibition.
format Online
Article
Text
id pubmed-5966422
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-59664222018-05-24 Docetaxel enhances lysosomal function through TFEB activation Zhang, Jianbin Wang, Jigang Wong, Yin Kwan Sun, Xin Chen, Yun Wang, Liming Yang, Liu Lu, Liqin Shen, Han-Ming Huang, Dongsheng Cell Death Dis Article Docetaxel is an effective and commonly used chemotherapeutic drug for cancer. Autophagy has been reported to be involved in the anticancer mechanism of docetaxel. However, the effect of docetaxel on lysosomal function remains elusive. In the present study, we first found that docetaxel treatment enhances autophagic flux in different cancer cells. Moreover, docetaxel treatment activates lysosomal function and promotes its fusion with autophagosome. Second, doctaxel treatment activates TFEB (transcription factor EB), a key nuclear transcription factor in control of lysosome biogenesis and function. We found that docetaxel promotes TFEB nuclear translocation and increases its transcriptional activity while knockdown of TFEB impairs lysosomal activation by docetaxel. Thirdly, TFEB activation by docetaxel is mediated by ROS (reactive oxygen species) generation and scavenging of ROS suppresses TFEB activity and lysosomal function in docetaxel-treated cells. Finally, inhibition of lysosomal function leads to increased docetaxel-induced cell death, suggesting that lysosomal activation protects against docetaxel-mediated apoptosis. Taken together, our results provide novel insights into the regulatory mechanisms of docetaxel on lysosomes, which could facilitate the development of novel potential cancer therapeutic agents via lysosomal inhibition. Nature Publishing Group UK 2018-05-23 /pmc/articles/PMC5966422/ /pubmed/29795139 http://dx.doi.org/10.1038/s41419-018-0571-4 Text en © The Author(s) 2018 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Zhang, Jianbin
Wang, Jigang
Wong, Yin Kwan
Sun, Xin
Chen, Yun
Wang, Liming
Yang, Liu
Lu, Liqin
Shen, Han-Ming
Huang, Dongsheng
Docetaxel enhances lysosomal function through TFEB activation
title Docetaxel enhances lysosomal function through TFEB activation
title_full Docetaxel enhances lysosomal function through TFEB activation
title_fullStr Docetaxel enhances lysosomal function through TFEB activation
title_full_unstemmed Docetaxel enhances lysosomal function through TFEB activation
title_short Docetaxel enhances lysosomal function through TFEB activation
title_sort docetaxel enhances lysosomal function through tfeb activation
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5966422/
https://www.ncbi.nlm.nih.gov/pubmed/29795139
http://dx.doi.org/10.1038/s41419-018-0571-4
work_keys_str_mv AT zhangjianbin docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT wangjigang docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT wongyinkwan docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT sunxin docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT chenyun docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT wangliming docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT yangliu docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT luliqin docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT shenhanming docetaxelenhanceslysosomalfunctionthroughtfebactivation
AT huangdongsheng docetaxelenhanceslysosomalfunctionthroughtfebactivation