UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repair

Glioblastoma multiforme (GBM) is a brain tumor with high mortality and recurrence rate. Radiotherapy and chemotherapy after surgery are the main treatment options available for GBM. However, patients with glioblastoma have a grave prognosis. The major reason is that most GBM patients are resistant t...

Full description

Bibliographic Details
Main Authors: Changyong Wu, Yang Shen, Lin Shi, Junhao Zhang, Tongxuan Guo, Lingni Zhou, Wanzhou Wang, Xu Zhang, Rutong Yu, Xuejiao Liu
Format: Article
Language:English
Published: Frontiers Media S.A. 2023-03-01
Series:Frontiers in Pharmacology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fphar.2023.1073929/full
_version_ 1811158978537717760
author Changyong Wu
Changyong Wu
Yang Shen
Yang Shen
Lin Shi
Lin Shi
Lin Shi
Junhao Zhang
Tongxuan Guo
Lingni Zhou
Wanzhou Wang
Xu Zhang
Xu Zhang
Rutong Yu
Rutong Yu
Xuejiao Liu
Xuejiao Liu
author_facet Changyong Wu
Changyong Wu
Yang Shen
Yang Shen
Lin Shi
Lin Shi
Lin Shi
Junhao Zhang
Tongxuan Guo
Lingni Zhou
Wanzhou Wang
Xu Zhang
Xu Zhang
Rutong Yu
Rutong Yu
Xuejiao Liu
Xuejiao Liu
author_sort Changyong Wu
collection DOAJ
description Glioblastoma multiforme (GBM) is a brain tumor with high mortality and recurrence rate. Radiotherapy and chemotherapy after surgery are the main treatment options available for GBM. However, patients with glioblastoma have a grave prognosis. The major reason is that most GBM patients are resistant to radiotherapy. UBA1 is considered an attractive potential anti-tumor therapeutic target and a key regulator of DNA double-strand break repair and genome replication in human cells. Therefore, we hypothesized that TAK-243, the first-in-class UBA1 inhibitor, might increase GBM sensitivity to radiation. The combined effect of TAK-243 and ionizing radiation on GBM cell proliferation, and colony formation ability was detected using CCK-8, colony formation, and EdU assays. The efficacy of TAK-243 combined with ionizing radiation for GBM was further evaluated in vivo, and the mechanism of TAK-243 sensitizing radiotherapy was preliminarily discussed. The results showed that TAK-243, in combination with ionizing radiation, significantly inhibited GBM cell proliferation, colony formation, cell cycle arrest in the G2/M phase, and increased the proportion of apoptosis. In addition, UBA1 inhibition by TAK-243 substantially increased the radiation-induced γ-H2AX expression and impaired the recruitment of the downstream effector molecule 53BP1. Therefore, TAK-243 inhibited the radiation-induced DNA double-strand break repair and thus inhibited the growth of GBM cells. Our results provided a new therapeutic strategy for improving the radiation sensitivity of GBM and laid a theoretical foundation and experimental basis for further clinical trials.
first_indexed 2024-04-10T05:33:24Z
format Article
id doaj.art-68cdefb69568496c92b6a7779ed65f2b
institution Directory Open Access Journal
issn 1663-9812
language English
last_indexed 2024-04-10T05:33:24Z
publishDate 2023-03-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Pharmacology
spelling doaj.art-68cdefb69568496c92b6a7779ed65f2b2023-03-07T04:24:49ZengFrontiers Media S.A.Frontiers in Pharmacology1663-98122023-03-011410.3389/fphar.2023.10739291073929UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repairChangyong Wu0Changyong Wu1Yang Shen2Yang Shen3Lin Shi4Lin Shi5Lin Shi6Junhao Zhang7Tongxuan Guo8Lingni Zhou9Wanzhou Wang10Xu Zhang11Xu Zhang12Rutong Yu13Rutong Yu14Xuejiao Liu15Xuejiao Liu16Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaDepartment of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaDepartment of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaDepartment of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, ChinaDepartment of general surgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaDepartment of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaDepartment of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, ChinaInsititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, ChinaDepartment of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, ChinaGlioblastoma multiforme (GBM) is a brain tumor with high mortality and recurrence rate. Radiotherapy and chemotherapy after surgery are the main treatment options available for GBM. However, patients with glioblastoma have a grave prognosis. The major reason is that most GBM patients are resistant to radiotherapy. UBA1 is considered an attractive potential anti-tumor therapeutic target and a key regulator of DNA double-strand break repair and genome replication in human cells. Therefore, we hypothesized that TAK-243, the first-in-class UBA1 inhibitor, might increase GBM sensitivity to radiation. The combined effect of TAK-243 and ionizing radiation on GBM cell proliferation, and colony formation ability was detected using CCK-8, colony formation, and EdU assays. The efficacy of TAK-243 combined with ionizing radiation for GBM was further evaluated in vivo, and the mechanism of TAK-243 sensitizing radiotherapy was preliminarily discussed. The results showed that TAK-243, in combination with ionizing radiation, significantly inhibited GBM cell proliferation, colony formation, cell cycle arrest in the G2/M phase, and increased the proportion of apoptosis. In addition, UBA1 inhibition by TAK-243 substantially increased the radiation-induced γ-H2AX expression and impaired the recruitment of the downstream effector molecule 53BP1. Therefore, TAK-243 inhibited the radiation-induced DNA double-strand break repair and thus inhibited the growth of GBM cells. Our results provided a new therapeutic strategy for improving the radiation sensitivity of GBM and laid a theoretical foundation and experimental basis for further clinical trials.https://www.frontiersin.org/articles/10.3389/fphar.2023.1073929/fullGBMTAK-243radiosensitizationDNA damage repaircell apoptosis
spellingShingle Changyong Wu
Changyong Wu
Yang Shen
Yang Shen
Lin Shi
Lin Shi
Lin Shi
Junhao Zhang
Tongxuan Guo
Lingni Zhou
Wanzhou Wang
Xu Zhang
Xu Zhang
Rutong Yu
Rutong Yu
Xuejiao Liu
Xuejiao Liu
UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repair
Frontiers in Pharmacology
GBM
TAK-243
radiosensitization
DNA damage repair
cell apoptosis
title UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repair
title_full UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repair
title_fullStr UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repair
title_full_unstemmed UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repair
title_short UBA1 inhibition contributes radiosensitization of glioblastoma cells via blocking DNA damage repair
title_sort uba1 inhibition contributes radiosensitization of glioblastoma cells via blocking dna damage repair
topic GBM
TAK-243
radiosensitization
DNA damage repair
cell apoptosis
url https://www.frontiersin.org/articles/10.3389/fphar.2023.1073929/full
work_keys_str_mv AT changyongwu uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT changyongwu uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT yangshen uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT yangshen uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT linshi uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT linshi uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT linshi uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT junhaozhang uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT tongxuanguo uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT lingnizhou uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT wanzhouwang uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT xuzhang uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT xuzhang uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT rutongyu uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT rutongyu uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT xuejiaoliu uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair
AT xuejiaoliu uba1inhibitioncontributesradiosensitizationofglioblastomacellsviablockingdnadamagerepair