Cargando…

Protein disulfide-isomerase A4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy

INTRODUCTION: Increasing evidence has revealed the key activity of protein disulfide isomerase A4 (PDIA4) in the endoplasmic reticulum stress (ERS) response. However, the role of PDIA4 in regulating glioblastoma (GBM)-specific pro-angiogenesis is still unknown. METHODS: The expression and prognostic...

Descripción completa

Detalles Bibliográficos
Autores principales: Tu, Zewei, Wang, Chong, Hu, Qing, Tao, Chuming, Fang, Zhansheng, Lin, Li, Lei, Kunjian, Luo, Min, Sheng, Yilei, Long, Xiaoyan, Li, Jingying, Wu, Lei, Huang, Kai, Zhu, Xingen
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10061982/
https://www.ncbi.nlm.nih.gov/pubmed/36997943
http://dx.doi.org/10.1186/s13046-023-02640-1
_version_ 1785017403059470336
author Tu, Zewei
Wang, Chong
Hu, Qing
Tao, Chuming
Fang, Zhansheng
Lin, Li
Lei, Kunjian
Luo, Min
Sheng, Yilei
Long, Xiaoyan
Li, Jingying
Wu, Lei
Huang, Kai
Zhu, Xingen
author_facet Tu, Zewei
Wang, Chong
Hu, Qing
Tao, Chuming
Fang, Zhansheng
Lin, Li
Lei, Kunjian
Luo, Min
Sheng, Yilei
Long, Xiaoyan
Li, Jingying
Wu, Lei
Huang, Kai
Zhu, Xingen
author_sort Tu, Zewei
collection PubMed
description INTRODUCTION: Increasing evidence has revealed the key activity of protein disulfide isomerase A4 (PDIA4) in the endoplasmic reticulum stress (ERS) response. However, the role of PDIA4 in regulating glioblastoma (GBM)-specific pro-angiogenesis is still unknown. METHODS: The expression and prognostic role of PDIA4 were analyzed using a bioinformatics approach and were validated in 32 clinical samples and follow-up data. RNA-sequencing was used to search for PDIA4-associated biological processes in GBM cells, and proteomic mass spectrum (MS) analysis was used to screen for potential PDIA4 substrates. Western blotting, real-time quantitative polymerase chain reaction (RT-qPCR), and enzyme-linked immunosorbent assays (ELISA) were used to measure the levels of the involved factors. Cell migration and tube formation assays determined the pro-angiogenesis activity of PDIA4 in vitro. An intracranial U87 xenograft GBM animal model was constructed to evaluate the pro-angiogenesis role of PDIA4 in vivo. RESULTS: Aberrant overexpression of PDIA4 was associated with a poor prognosis in patients with GBM, although PDIA4 could also functionally regulate intrinsic GBM secretion of vascular endothelial growth factor-A (VEGF-A) through its active domains of Cys-X-X-Cys (CXXC) oxidoreductase. Functionally, PDIA4 exhibits pro-angiogenesis activity both in vitro and in vivo, and can be upregulated by ERS through transcriptional regulation of X-box binding protein 1 (XBP1). The XBP1/PDIA4/VEGFA axis partially supports the mechanism underlying GBM cell survival under ER stress. Further, GBM cells with higher expression of PDIA4 showed resistance to antiangiogenic therapy in vivo. CONCLUSIONS: Our findings revealed the pro-angiogenesis role of PDIA4 in GBM progression and its potential impact on GBM survival under a harsh microenvironment. Targeting PDIA4 might help to improve the efficacy of antiangiogenic therapy in patients with GBM. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02640-1.
format Online
Article
Text
id pubmed-10061982
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-100619822023-03-31 Protein disulfide-isomerase A4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy Tu, Zewei Wang, Chong Hu, Qing Tao, Chuming Fang, Zhansheng Lin, Li Lei, Kunjian Luo, Min Sheng, Yilei Long, Xiaoyan Li, Jingying Wu, Lei Huang, Kai Zhu, Xingen J Exp Clin Cancer Res Research INTRODUCTION: Increasing evidence has revealed the key activity of protein disulfide isomerase A4 (PDIA4) in the endoplasmic reticulum stress (ERS) response. However, the role of PDIA4 in regulating glioblastoma (GBM)-specific pro-angiogenesis is still unknown. METHODS: The expression and prognostic role of PDIA4 were analyzed using a bioinformatics approach and were validated in 32 clinical samples and follow-up data. RNA-sequencing was used to search for PDIA4-associated biological processes in GBM cells, and proteomic mass spectrum (MS) analysis was used to screen for potential PDIA4 substrates. Western blotting, real-time quantitative polymerase chain reaction (RT-qPCR), and enzyme-linked immunosorbent assays (ELISA) were used to measure the levels of the involved factors. Cell migration and tube formation assays determined the pro-angiogenesis activity of PDIA4 in vitro. An intracranial U87 xenograft GBM animal model was constructed to evaluate the pro-angiogenesis role of PDIA4 in vivo. RESULTS: Aberrant overexpression of PDIA4 was associated with a poor prognosis in patients with GBM, although PDIA4 could also functionally regulate intrinsic GBM secretion of vascular endothelial growth factor-A (VEGF-A) through its active domains of Cys-X-X-Cys (CXXC) oxidoreductase. Functionally, PDIA4 exhibits pro-angiogenesis activity both in vitro and in vivo, and can be upregulated by ERS through transcriptional regulation of X-box binding protein 1 (XBP1). The XBP1/PDIA4/VEGFA axis partially supports the mechanism underlying GBM cell survival under ER stress. Further, GBM cells with higher expression of PDIA4 showed resistance to antiangiogenic therapy in vivo. CONCLUSIONS: Our findings revealed the pro-angiogenesis role of PDIA4 in GBM progression and its potential impact on GBM survival under a harsh microenvironment. Targeting PDIA4 might help to improve the efficacy of antiangiogenic therapy in patients with GBM. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02640-1. BioMed Central 2023-03-30 /pmc/articles/PMC10061982/ /pubmed/36997943 http://dx.doi.org/10.1186/s13046-023-02640-1 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open AccessThis 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
Tu, Zewei
Wang, Chong
Hu, Qing
Tao, Chuming
Fang, Zhansheng
Lin, Li
Lei, Kunjian
Luo, Min
Sheng, Yilei
Long, Xiaoyan
Li, Jingying
Wu, Lei
Huang, Kai
Zhu, Xingen
Protein disulfide-isomerase A4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy
title Protein disulfide-isomerase A4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy
title_full Protein disulfide-isomerase A4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy
title_fullStr Protein disulfide-isomerase A4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy
title_full_unstemmed Protein disulfide-isomerase A4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy
title_short Protein disulfide-isomerase A4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy
title_sort protein disulfide-isomerase a4 confers glioblastoma angiogenesis promotion capacity and resistance to anti-angiogenic therapy
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10061982/
https://www.ncbi.nlm.nih.gov/pubmed/36997943
http://dx.doi.org/10.1186/s13046-023-02640-1
work_keys_str_mv AT tuzewei proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT wangchong proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT huqing proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT taochuming proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT fangzhansheng proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT linli proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT leikunjian proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT luomin proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT shengyilei proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT longxiaoyan proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT lijingying proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT wulei proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT huangkai proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy
AT zhuxingen proteindisulfideisomerasea4confersglioblastomaangiogenesispromotioncapacityandresistancetoantiangiogenictherapy