Cargando…

Targeting RPA promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma

BACKGROUND: Autophagy is involved in nasopharyngeal carcinoma (NPC) radioresistance. Replication protein A 1 (RPA1) and RPA3, substrates of the RPA complex, are potential therapeutic targets for reversing NPC radioresistance. Nevertheless, the role of RPA in autophagy is not adequately understood. T...

Descripción completa

Detalles Bibliográficos
Autores principales: Feng, Yanchun, Jiang, Yingming, Liu, Jun, Liu, Jiaqi, Shi, Mengchen, Chen, Junxiong, Zhang, Jingdan, Tian, Yu, Yang, Xiangling, Liu, Huanliang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10585929/
https://www.ncbi.nlm.nih.gov/pubmed/37858134
http://dx.doi.org/10.1186/s12967-023-04574-w
_version_ 1785123053153288192
author Feng, Yanchun
Jiang, Yingming
Liu, Jun
Liu, Jiaqi
Shi, Mengchen
Chen, Junxiong
Zhang, Jingdan
Tian, Yu
Yang, Xiangling
Liu, Huanliang
author_facet Feng, Yanchun
Jiang, Yingming
Liu, Jun
Liu, Jiaqi
Shi, Mengchen
Chen, Junxiong
Zhang, Jingdan
Tian, Yu
Yang, Xiangling
Liu, Huanliang
author_sort Feng, Yanchun
collection PubMed
description BACKGROUND: Autophagy is involved in nasopharyngeal carcinoma (NPC) radioresistance. Replication protein A 1 (RPA1) and RPA3, substrates of the RPA complex, are potential therapeutic targets for reversing NPC radioresistance. Nevertheless, the role of RPA in autophagy is not adequately understood. This investigation was performed to reveal the cytotoxic mechanism of a pharmacologic RPA inhibitor (RPAi) in NPC cells and the underlying mechanism by which RPAi-mediated autophagy regulates NPC radiosensitivity. METHODS AND RESULTS: We characterized a potent RPAi (HAMNO) that was substantially correlated with radiosensitivity enhancement and proliferative inhibition of in vivo and in NPC cell lines in vitro. We show that the RPAi induced autophagy at multiple levels by inducing autophagic flux, AMPK/mTOR pathway activation, and autophagy-related gene transcription by decreasing glycolytic function. We hypothesized that RPA inhibition impaired glycolysis and increased NPC dependence on autophagy. We further demonstrated that combining autophagy inhibition with chloroquine (CQ) treatment or genetic inhibition of the autophagy regulator ATG5 and RPAi treatment was more effective than either approach alone in enhancing the antitumor response of NPC to radiation. CONCLUSIONS: Our study suggests that HAMNO is a potent RPAi that enhances radiosensitivity and induces autophagy in NPC cell lines by decreasing glycolytic function and activating autophagy-related genes. We suggest a novel treatment strategy in which pharmacological inhibitors that simultaneously disrupt RPA and autophagic processes improve NPC responsiveness to radiation. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04574-w.
format Online
Article
Text
id pubmed-10585929
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-105859292023-10-20 Targeting RPA promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma Feng, Yanchun Jiang, Yingming Liu, Jun Liu, Jiaqi Shi, Mengchen Chen, Junxiong Zhang, Jingdan Tian, Yu Yang, Xiangling Liu, Huanliang J Transl Med Research BACKGROUND: Autophagy is involved in nasopharyngeal carcinoma (NPC) radioresistance. Replication protein A 1 (RPA1) and RPA3, substrates of the RPA complex, are potential therapeutic targets for reversing NPC radioresistance. Nevertheless, the role of RPA in autophagy is not adequately understood. This investigation was performed to reveal the cytotoxic mechanism of a pharmacologic RPA inhibitor (RPAi) in NPC cells and the underlying mechanism by which RPAi-mediated autophagy regulates NPC radiosensitivity. METHODS AND RESULTS: We characterized a potent RPAi (HAMNO) that was substantially correlated with radiosensitivity enhancement and proliferative inhibition of in vivo and in NPC cell lines in vitro. We show that the RPAi induced autophagy at multiple levels by inducing autophagic flux, AMPK/mTOR pathway activation, and autophagy-related gene transcription by decreasing glycolytic function. We hypothesized that RPA inhibition impaired glycolysis and increased NPC dependence on autophagy. We further demonstrated that combining autophagy inhibition with chloroquine (CQ) treatment or genetic inhibition of the autophagy regulator ATG5 and RPAi treatment was more effective than either approach alone in enhancing the antitumor response of NPC to radiation. CONCLUSIONS: Our study suggests that HAMNO is a potent RPAi that enhances radiosensitivity and induces autophagy in NPC cell lines by decreasing glycolytic function and activating autophagy-related genes. We suggest a novel treatment strategy in which pharmacological inhibitors that simultaneously disrupt RPA and autophagic processes improve NPC responsiveness to radiation. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12967-023-04574-w. BioMed Central 2023-10-19 /pmc/articles/PMC10585929/ /pubmed/37858134 http://dx.doi.org/10.1186/s12967-023-04574-w Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/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 licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence 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 licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Feng, Yanchun
Jiang, Yingming
Liu, Jun
Liu, Jiaqi
Shi, Mengchen
Chen, Junxiong
Zhang, Jingdan
Tian, Yu
Yang, Xiangling
Liu, Huanliang
Targeting RPA promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma
title Targeting RPA promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma
title_full Targeting RPA promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma
title_fullStr Targeting RPA promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma
title_full_unstemmed Targeting RPA promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma
title_short Targeting RPA promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma
title_sort targeting rpa promotes autophagic flux and the antitumor response to radiation in nasopharyngeal carcinoma
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10585929/
https://www.ncbi.nlm.nih.gov/pubmed/37858134
http://dx.doi.org/10.1186/s12967-023-04574-w
work_keys_str_mv AT fengyanchun targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT jiangyingming targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT liujun targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT liujiaqi targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT shimengchen targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT chenjunxiong targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT zhangjingdan targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT tianyu targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT yangxiangling targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma
AT liuhuanliang targetingrpapromotesautophagicfluxandtheantitumorresponsetoradiationinnasopharyngealcarcinoma