Cargando…

MTA1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis

Our previous study revealed that metastasis-associated protein 1 (MTA1), which is expressed in vascular endothelial cells, acts as a tube formation promoting factor. The present study aimed to clarify the importance of MTA1 expression in tube formation using MTA1-knockout (KO) endothelial cells (MTA...

Descripción completa

Detalles Bibliográficos
Autores principales: Ishikawa, Mizuho, Osaki, Mitsuhiko, Uno, Narumi, Ohira, Takahito, Kugoh, Hiroyuki, Okada, Futoshi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8600423/
https://www.ncbi.nlm.nih.gov/pubmed/34779499
http://dx.doi.org/10.3892/mmr.2021.12527
_version_ 1784601153500086272
author Ishikawa, Mizuho
Osaki, Mitsuhiko
Uno, Narumi
Ohira, Takahito
Kugoh, Hiroyuki
Okada, Futoshi
author_facet Ishikawa, Mizuho
Osaki, Mitsuhiko
Uno, Narumi
Ohira, Takahito
Kugoh, Hiroyuki
Okada, Futoshi
author_sort Ishikawa, Mizuho
collection PubMed
description Our previous study revealed that metastasis-associated protein 1 (MTA1), which is expressed in vascular endothelial cells, acts as a tube formation promoting factor. The present study aimed to clarify the importance of MTA1 expression in tube formation using MTA1-knockout (KO) endothelial cells (MTA1-KO MSS31 cells). Tube formation was significantly suppressed in MTA1-KO MSS31 cells, whereas MTA1-overexpression MTA1-KO MSS31 cells regained the ability to form tube-like structures. In addition, western blotting analysis revealed that MTA1-KO MSS31 cells showed significantly higher levels of phosphorylation of non-muscle myosin heavy chain IIa, which resulted in suppression of tube formation. This effect was attributed to a decrease of MTA1/S100 calcium-binding protein A4 complex formation. Moreover, inhibition of tube formation in MTA1-KO MSS31 cells could not be rescued by stimulation with vascular endothelial growth factor (VEGF). These results demonstrated that MTA1 may serve as an essential molecule for angiogenesis in endothelial cells and be involved in different steps of the angiogenic process compared with the VEGF/VEGF receptor 2 pathway. The findings showed that endothelial MTA1 and its pathway may serve as promising targets for inhibiting tumor angiogenesis, further supporting the development of MTA1-based antiangiogenic therapies.
format Online
Article
Text
id pubmed-8600423
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-86004232021-11-21 MTA1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis Ishikawa, Mizuho Osaki, Mitsuhiko Uno, Narumi Ohira, Takahito Kugoh, Hiroyuki Okada, Futoshi Mol Med Rep Articles Our previous study revealed that metastasis-associated protein 1 (MTA1), which is expressed in vascular endothelial cells, acts as a tube formation promoting factor. The present study aimed to clarify the importance of MTA1 expression in tube formation using MTA1-knockout (KO) endothelial cells (MTA1-KO MSS31 cells). Tube formation was significantly suppressed in MTA1-KO MSS31 cells, whereas MTA1-overexpression MTA1-KO MSS31 cells regained the ability to form tube-like structures. In addition, western blotting analysis revealed that MTA1-KO MSS31 cells showed significantly higher levels of phosphorylation of non-muscle myosin heavy chain IIa, which resulted in suppression of tube formation. This effect was attributed to a decrease of MTA1/S100 calcium-binding protein A4 complex formation. Moreover, inhibition of tube formation in MTA1-KO MSS31 cells could not be rescued by stimulation with vascular endothelial growth factor (VEGF). These results demonstrated that MTA1 may serve as an essential molecule for angiogenesis in endothelial cells and be involved in different steps of the angiogenic process compared with the VEGF/VEGF receptor 2 pathway. The findings showed that endothelial MTA1 and its pathway may serve as promising targets for inhibiting tumor angiogenesis, further supporting the development of MTA1-based antiangiogenic therapies. D.A. Spandidos 2022-01 2021-11-09 /pmc/articles/PMC8600423/ /pubmed/34779499 http://dx.doi.org/10.3892/mmr.2021.12527 Text en Copyright: © Ishikawa et al. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Ishikawa, Mizuho
Osaki, Mitsuhiko
Uno, Narumi
Ohira, Takahito
Kugoh, Hiroyuki
Okada, Futoshi
MTA1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis
title MTA1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis
title_full MTA1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis
title_fullStr MTA1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis
title_full_unstemmed MTA1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis
title_short MTA1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis
title_sort mta1, a metastasis-associated protein, in endothelial cells is an essential molecule for angiogenesis
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8600423/
https://www.ncbi.nlm.nih.gov/pubmed/34779499
http://dx.doi.org/10.3892/mmr.2021.12527
work_keys_str_mv AT ishikawamizuho mta1ametastasisassociatedproteininendothelialcellsisanessentialmoleculeforangiogenesis
AT osakimitsuhiko mta1ametastasisassociatedproteininendothelialcellsisanessentialmoleculeforangiogenesis
AT unonarumi mta1ametastasisassociatedproteininendothelialcellsisanessentialmoleculeforangiogenesis
AT ohiratakahito mta1ametastasisassociatedproteininendothelialcellsisanessentialmoleculeforangiogenesis
AT kugohhiroyuki mta1ametastasisassociatedproteininendothelialcellsisanessentialmoleculeforangiogenesis
AT okadafutoshi mta1ametastasisassociatedproteininendothelialcellsisanessentialmoleculeforangiogenesis