Cargando…

DYRK1A activates NFATC1 to increase glioblastoma migration

Glioblastoma (GBM) is the most aggressive glioma, and is prone to develop resistance to chemotherapy and radiotherapy; hence, patients with glioblastoma have a high recurrence rate and a low 1‐year survival rate. In addition, the pathogenesis of glioblastoma is complex and largely unknown, and the a...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Heng, Sun, Qian, Chen, Shuai, Chen, Long, Jia, Wenming, Zhao, Juan, Sun, Xiulian
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8446559/
https://www.ncbi.nlm.nih.gov/pubmed/34309232
http://dx.doi.org/10.1002/cam4.4159
_version_ 1784568906719952896
author Liu, Heng
Sun, Qian
Chen, Shuai
Chen, Long
Jia, Wenming
Zhao, Juan
Sun, Xiulian
author_facet Liu, Heng
Sun, Qian
Chen, Shuai
Chen, Long
Jia, Wenming
Zhao, Juan
Sun, Xiulian
author_sort Liu, Heng
collection PubMed
description Glioblastoma (GBM) is the most aggressive glioma, and is prone to develop resistance to chemotherapy and radiotherapy; hence, patients with glioblastoma have a high recurrence rate and a low 1‐year survival rate. In addition, the pathogenesis of glioblastoma is complex and largely unknown, and the available treatments are limited. Here, we uncovered a fundamental role of DYRK1A in regulating NFATC1 in GBMs. We found that DYRK1A was highly expressed in glioma and glioblastoma cells, and its expression was positively correlated with that of NFATC1. Moreover, inhibition of DYRK1A promoted NFATC1 degradation in GBM cells and sharply reduced the transactivation of NFATC1, not only by decreasing the expression of NFATC1‐targeted genes, but also by reducing the luciferase activity, and vice versa. However, DYRK1A had the opposite effect on NFATC2. Most importantly, our data suggest that DYRK1A inhibition reduces glioblastoma migration. Polypeptides derived from the DYRK1A‐targeted motif of NFATC1, by competitively blocking DYRK1A kinase activity on NFATC1, clearly destabilized NFATC1 protein and impaired glioblastoma migration. We propose that the recovery of NFATC1 stability is a key oncogenic event in a large proportion of gliomas, and pharmacological inhibition of DYRK1A by polypeptides could represent a promising therapeutic intervention for GBM.
format Online
Article
Text
id pubmed-8446559
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-84465592021-09-22 DYRK1A activates NFATC1 to increase glioblastoma migration Liu, Heng Sun, Qian Chen, Shuai Chen, Long Jia, Wenming Zhao, Juan Sun, Xiulian Cancer Med Cancer Biology Glioblastoma (GBM) is the most aggressive glioma, and is prone to develop resistance to chemotherapy and radiotherapy; hence, patients with glioblastoma have a high recurrence rate and a low 1‐year survival rate. In addition, the pathogenesis of glioblastoma is complex and largely unknown, and the available treatments are limited. Here, we uncovered a fundamental role of DYRK1A in regulating NFATC1 in GBMs. We found that DYRK1A was highly expressed in glioma and glioblastoma cells, and its expression was positively correlated with that of NFATC1. Moreover, inhibition of DYRK1A promoted NFATC1 degradation in GBM cells and sharply reduced the transactivation of NFATC1, not only by decreasing the expression of NFATC1‐targeted genes, but also by reducing the luciferase activity, and vice versa. However, DYRK1A had the opposite effect on NFATC2. Most importantly, our data suggest that DYRK1A inhibition reduces glioblastoma migration. Polypeptides derived from the DYRK1A‐targeted motif of NFATC1, by competitively blocking DYRK1A kinase activity on NFATC1, clearly destabilized NFATC1 protein and impaired glioblastoma migration. We propose that the recovery of NFATC1 stability is a key oncogenic event in a large proportion of gliomas, and pharmacological inhibition of DYRK1A by polypeptides could represent a promising therapeutic intervention for GBM. John Wiley and Sons Inc. 2021-07-26 /pmc/articles/PMC8446559/ /pubmed/34309232 http://dx.doi.org/10.1002/cam4.4159 Text en © 2021 The Authors. Cancer Medicine 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 Cancer Biology
Liu, Heng
Sun, Qian
Chen, Shuai
Chen, Long
Jia, Wenming
Zhao, Juan
Sun, Xiulian
DYRK1A activates NFATC1 to increase glioblastoma migration
title DYRK1A activates NFATC1 to increase glioblastoma migration
title_full DYRK1A activates NFATC1 to increase glioblastoma migration
title_fullStr DYRK1A activates NFATC1 to increase glioblastoma migration
title_full_unstemmed DYRK1A activates NFATC1 to increase glioblastoma migration
title_short DYRK1A activates NFATC1 to increase glioblastoma migration
title_sort dyrk1a activates nfatc1 to increase glioblastoma migration
topic Cancer Biology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8446559/
https://www.ncbi.nlm.nih.gov/pubmed/34309232
http://dx.doi.org/10.1002/cam4.4159
work_keys_str_mv AT liuheng dyrk1aactivatesnfatc1toincreaseglioblastomamigration
AT sunqian dyrk1aactivatesnfatc1toincreaseglioblastomamigration
AT chenshuai dyrk1aactivatesnfatc1toincreaseglioblastomamigration
AT chenlong dyrk1aactivatesnfatc1toincreaseglioblastomamigration
AT jiawenming dyrk1aactivatesnfatc1toincreaseglioblastomamigration
AT zhaojuan dyrk1aactivatesnfatc1toincreaseglioblastomamigration
AT sunxiulian dyrk1aactivatesnfatc1toincreaseglioblastomamigration