Cargando…

Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance

AIMS: We aimed to investigate the role of receptor‐interacting protein 2 (RIP2) in regulation of stemness of glioma cells and chemotherapy resistance. METHODS: Plasmid transfection was used to overexpress RIP2. Chemical inhibitors were used to inhibit RIP2 or NF‐κB activity. Cancer stemness of gliom...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Xiao‐liang, Jiao, Bao‐hua, Wu, Jian‐liang, Yang, Jian‐kai, Hu, Yu‐hua, Cui, Kai
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9627370/
https://www.ncbi.nlm.nih.gov/pubmed/36184801
http://dx.doi.org/10.1111/cns.13981
_version_ 1784822954712891392
author Wang, Xiao‐liang
Jiao, Bao‐hua
Wu, Jian‐liang
Yang, Jian‐kai
Hu, Yu‐hua
Cui, Kai
author_facet Wang, Xiao‐liang
Jiao, Bao‐hua
Wu, Jian‐liang
Yang, Jian‐kai
Hu, Yu‐hua
Cui, Kai
author_sort Wang, Xiao‐liang
collection PubMed
description AIMS: We aimed to investigate the role of receptor‐interacting protein 2 (RIP2) in regulation of stemness of glioma cells and chemotherapy resistance. METHODS: Plasmid transfection was used to overexpress RIP2. Chemical inhibitors were used to inhibit RIP2 or NF‐κB activity. Cancer stemness of glioma cells was investigated by sphere formation assays, clone formation assays, and xenograft tumor formation assays. The expression of RIP2, p‐NF‐κB, IκBα, CD133, or SOX‐2 was detected by Western blotting and immunofluorescence. Apoptosis was detected by flow cytometry. Immunohistochemical staining was used to detect the expression of RIP2, CD133, and SOX‐2 in xenograft tumor tissue. The effect of the RIP2/NF‐κB pathway on temozolomide (TMZ) resistance was evaluated by xenograft tumor assay. RESULTS: Transfection with RIP2 plasmid enhanced the sphere formation capability of U251 cells, clone formation capability, and xenograft tumor formation capability. RIP2 could mediate TMZ resistance by upregulating the expression of CD133 and SOX‐2 by activating the NF‐κB pathway. Both RIP2 inhibitor GSK583 and the NF‐κB inhibitor SC75741 could reverse the resistance of U251 cells to TMZ. CONCLUSION: RIP2 mediates TMZ resistance by regulating the maintenance of stemness in glioma cells through NF‐κB. Interventions targeting the RIP2/NF‐κB pathway may be a new strategy for TMZ‐resistant gliomas.
format Online
Article
Text
id pubmed-9627370
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-96273702022-11-03 Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance Wang, Xiao‐liang Jiao, Bao‐hua Wu, Jian‐liang Yang, Jian‐kai Hu, Yu‐hua Cui, Kai CNS Neurosci Ther Original Articles AIMS: We aimed to investigate the role of receptor‐interacting protein 2 (RIP2) in regulation of stemness of glioma cells and chemotherapy resistance. METHODS: Plasmid transfection was used to overexpress RIP2. Chemical inhibitors were used to inhibit RIP2 or NF‐κB activity. Cancer stemness of glioma cells was investigated by sphere formation assays, clone formation assays, and xenograft tumor formation assays. The expression of RIP2, p‐NF‐κB, IκBα, CD133, or SOX‐2 was detected by Western blotting and immunofluorescence. Apoptosis was detected by flow cytometry. Immunohistochemical staining was used to detect the expression of RIP2, CD133, and SOX‐2 in xenograft tumor tissue. The effect of the RIP2/NF‐κB pathway on temozolomide (TMZ) resistance was evaluated by xenograft tumor assay. RESULTS: Transfection with RIP2 plasmid enhanced the sphere formation capability of U251 cells, clone formation capability, and xenograft tumor formation capability. RIP2 could mediate TMZ resistance by upregulating the expression of CD133 and SOX‐2 by activating the NF‐κB pathway. Both RIP2 inhibitor GSK583 and the NF‐κB inhibitor SC75741 could reverse the resistance of U251 cells to TMZ. CONCLUSION: RIP2 mediates TMZ resistance by regulating the maintenance of stemness in glioma cells through NF‐κB. Interventions targeting the RIP2/NF‐κB pathway may be a new strategy for TMZ‐resistant gliomas. John Wiley and Sons Inc. 2022-10-02 /pmc/articles/PMC9627370/ /pubmed/36184801 http://dx.doi.org/10.1111/cns.13981 Text en © 2022 The Authors. CNS Neuroscience & Therapeutics published by John Wiley & Sons Ltd. https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Wang, Xiao‐liang
Jiao, Bao‐hua
Wu, Jian‐liang
Yang, Jian‐kai
Hu, Yu‐hua
Cui, Kai
Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance
title Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance
title_full Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance
title_fullStr Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance
title_full_unstemmed Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance
title_short Mechanism of RIP2 enhancing stemness of glioma cells induces temozolomide resistance
title_sort mechanism of rip2 enhancing stemness of glioma cells induces temozolomide resistance
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9627370/
https://www.ncbi.nlm.nih.gov/pubmed/36184801
http://dx.doi.org/10.1111/cns.13981
work_keys_str_mv AT wangxiaoliang mechanismofrip2enhancingstemnessofgliomacellsinducestemozolomideresistance
AT jiaobaohua mechanismofrip2enhancingstemnessofgliomacellsinducestemozolomideresistance
AT wujianliang mechanismofrip2enhancingstemnessofgliomacellsinducestemozolomideresistance
AT yangjiankai mechanismofrip2enhancingstemnessofgliomacellsinducestemozolomideresistance
AT huyuhua mechanismofrip2enhancingstemnessofgliomacellsinducestemozolomideresistance
AT cuikai mechanismofrip2enhancingstemnessofgliomacellsinducestemozolomideresistance