Cargando…

GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer

In the treatment of most malignancies, radiotherapy plays a significant role. However, the resistance of cancer cells to ionizing radiation (IR) is the main reason for the failure of radiotherapy, which causes tumor recurrence and metastasis. In this study, we confirmed that GPR162, an orphan recept...

Descripción completa

Detalles Bibliográficos
Autores principales: Long, Yao, Guo, Jiaxing, Chen, Jielin, Sun, Jingyue, Wang, Haiyan, Peng, Xin, Wang, Zuli, Lai, WeiWei, Liu, Na, Shu, Long, Chen, Ling, Shi, Ying, Xiao, Desheng, Liu, Shuang, Tao, Yongguang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9892510/
https://www.ncbi.nlm.nih.gov/pubmed/36725837
http://dx.doi.org/10.1038/s41392-022-01224-3
_version_ 1784881339539914752
author Long, Yao
Guo, Jiaxing
Chen, Jielin
Sun, Jingyue
Wang, Haiyan
Peng, Xin
Wang, Zuli
Lai, WeiWei
Liu, Na
Shu, Long
Chen, Ling
Shi, Ying
Xiao, Desheng
Liu, Shuang
Tao, Yongguang
author_facet Long, Yao
Guo, Jiaxing
Chen, Jielin
Sun, Jingyue
Wang, Haiyan
Peng, Xin
Wang, Zuli
Lai, WeiWei
Liu, Na
Shu, Long
Chen, Ling
Shi, Ying
Xiao, Desheng
Liu, Shuang
Tao, Yongguang
author_sort Long, Yao
collection PubMed
description In the treatment of most malignancies, radiotherapy plays a significant role. However, the resistance of cancer cells to ionizing radiation (IR) is the main reason for the failure of radiotherapy, which causes tumor recurrence and metastasis. In this study, we confirmed that GPR162, an orphan receptor in the G-protein-coupled receptor family, acted as a novel radiotherapy sensitizer by interacting with the stimulator of interferon genes (STING), which targeted DNA damage responses, activated IRF3, accelerated the activation of type I interferon system, promoted the expression of chemokines including CXCL10 and CXCL4, and inhibited the occurrence and development of tumors. Interestingly, the activation of STING by overexpression of GPR162 was independent of the classical pathway of cGAS. STING inhibitors could resist the antitumor effect of overexpression of GPR162 in IR-induced mouse models. In addition, most solid tumors showed low expression of GPR162. And the higher expression of GPR162 indicated a better prognosis in patients with lung adenocarcinoma, liver cancer, breast cancer, etc. In summary, these results suggested that GPR162 may serve as a potential sensitizer of radiotherapy by promoting radiotherapy-induced STING-IFN production and increasing the expression of chemokines including CXCL10 and CXCL4 in DNA damage response, providing an alternative strategy for improving cancer radiotherapy.
format Online
Article
Text
id pubmed-9892510
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-98925102023-02-03 GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer Long, Yao Guo, Jiaxing Chen, Jielin Sun, Jingyue Wang, Haiyan Peng, Xin Wang, Zuli Lai, WeiWei Liu, Na Shu, Long Chen, Ling Shi, Ying Xiao, Desheng Liu, Shuang Tao, Yongguang Signal Transduct Target Ther Article In the treatment of most malignancies, radiotherapy plays a significant role. However, the resistance of cancer cells to ionizing radiation (IR) is the main reason for the failure of radiotherapy, which causes tumor recurrence and metastasis. In this study, we confirmed that GPR162, an orphan receptor in the G-protein-coupled receptor family, acted as a novel radiotherapy sensitizer by interacting with the stimulator of interferon genes (STING), which targeted DNA damage responses, activated IRF3, accelerated the activation of type I interferon system, promoted the expression of chemokines including CXCL10 and CXCL4, and inhibited the occurrence and development of tumors. Interestingly, the activation of STING by overexpression of GPR162 was independent of the classical pathway of cGAS. STING inhibitors could resist the antitumor effect of overexpression of GPR162 in IR-induced mouse models. In addition, most solid tumors showed low expression of GPR162. And the higher expression of GPR162 indicated a better prognosis in patients with lung adenocarcinoma, liver cancer, breast cancer, etc. In summary, these results suggested that GPR162 may serve as a potential sensitizer of radiotherapy by promoting radiotherapy-induced STING-IFN production and increasing the expression of chemokines including CXCL10 and CXCL4 in DNA damage response, providing an alternative strategy for improving cancer radiotherapy. Nature Publishing Group UK 2023-02-01 /pmc/articles/PMC9892510/ /pubmed/36725837 http://dx.doi.org/10.1038/s41392-022-01224-3 Text en © The Author(s) 2022, corrected publication 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 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/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Long, Yao
Guo, Jiaxing
Chen, Jielin
Sun, Jingyue
Wang, Haiyan
Peng, Xin
Wang, Zuli
Lai, WeiWei
Liu, Na
Shu, Long
Chen, Ling
Shi, Ying
Xiao, Desheng
Liu, Shuang
Tao, Yongguang
GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer
title GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer
title_full GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer
title_fullStr GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer
title_full_unstemmed GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer
title_short GPR162 activates STING dependent DNA damage pathway as a novel tumor suppressor and radiation sensitizer
title_sort gpr162 activates sting dependent dna damage pathway as a novel tumor suppressor and radiation sensitizer
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9892510/
https://www.ncbi.nlm.nih.gov/pubmed/36725837
http://dx.doi.org/10.1038/s41392-022-01224-3
work_keys_str_mv AT longyao gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT guojiaxing gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT chenjielin gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT sunjingyue gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT wanghaiyan gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT pengxin gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT wangzuli gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT laiweiwei gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT liuna gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT shulong gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT chenling gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT shiying gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT xiaodesheng gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT liushuang gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer
AT taoyongguang gpr162activatesstingdependentdnadamagepathwayasanoveltumorsuppressorandradiationsensitizer