Cargando…

Hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the PI3K/Stathmin1 pathway

BACKGROUND: Endothelial cells (EC) are sensitive to changes in the microenvironment, including hypoxia and ischemia. Disruption of the microtubular network has been reported in cases of ischemia. However, the signaling pathways involved in hypoxia-induced microtubular disruption are unknown. The pur...

Descripción completa

Detalles Bibliográficos
Autores principales: Cao, Huaming, Yu, Dongsheng, Yan, Xueyun, Wang, Bing, Yu, Zhiming, Song, Yu, Sheng, Liang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6380067/
https://www.ncbi.nlm.nih.gov/pubmed/30820314
http://dx.doi.org/10.1186/s13578-019-0283-1
_version_ 1783396246503817216
author Cao, Huaming
Yu, Dongsheng
Yan, Xueyun
Wang, Bing
Yu, Zhiming
Song, Yu
Sheng, Liang
author_facet Cao, Huaming
Yu, Dongsheng
Yan, Xueyun
Wang, Bing
Yu, Zhiming
Song, Yu
Sheng, Liang
author_sort Cao, Huaming
collection PubMed
description BACKGROUND: Endothelial cells (EC) are sensitive to changes in the microenvironment, including hypoxia and ischemia. Disruption of the microtubular network has been reported in cases of ischemia. However, the signaling pathways involved in hypoxia-induced microtubular disruption are unknown. The purpose of this study was to investigate the molecular mechanisms involved in hypoxia-induced microtubular disassembly in human umbilical vein endothelial cells (HUVECs). RESULTS: HUVECs were cultured under normoxic or hypoxic conditions and pretreated with or without colchicine or paclitaxel. The MTT assay, Transwell assay, trans-endothelial permeability assay, and 5-bromo-2′-deoxy-uridine staining were used to test the survival rate, migration, permeability, and proliferation of cells, respectively. Transmission electron microscopy and phalloidin staining were used to observe the microstructure and polymerization of microtubules. The results show that the functions of HUVECs and the microtubular structure were destroyed by hypoxia, but were protected by paclitaxel and a reactive oxygen species (ROS) inhibitor. We further used western blot, a luciferase assay, and co-immunoprecipitation to describe a non-transcription-independent mechanism for PI3K activation-inhibited microtubular stability mediated by Stathmin1, a PI3K interactor that functions in microtubule depolymerization. Finally, we determined that hypoxia and ROS blocked the interaction between PI3K and Stathmin1 to activate disassembly of microtubules. CONCLUSION: Thus, our data demonstrate that hypoxia induced the production of ROS and damaged EC function by destroying the microtubular structure through the PI3K/stathmin1 pathway.
format Online
Article
Text
id pubmed-6380067
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-63800672019-02-28 Hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the PI3K/Stathmin1 pathway Cao, Huaming Yu, Dongsheng Yan, Xueyun Wang, Bing Yu, Zhiming Song, Yu Sheng, Liang Cell Biosci Research BACKGROUND: Endothelial cells (EC) are sensitive to changes in the microenvironment, including hypoxia and ischemia. Disruption of the microtubular network has been reported in cases of ischemia. However, the signaling pathways involved in hypoxia-induced microtubular disruption are unknown. The purpose of this study was to investigate the molecular mechanisms involved in hypoxia-induced microtubular disassembly in human umbilical vein endothelial cells (HUVECs). RESULTS: HUVECs were cultured under normoxic or hypoxic conditions and pretreated with or without colchicine or paclitaxel. The MTT assay, Transwell assay, trans-endothelial permeability assay, and 5-bromo-2′-deoxy-uridine staining were used to test the survival rate, migration, permeability, and proliferation of cells, respectively. Transmission electron microscopy and phalloidin staining were used to observe the microstructure and polymerization of microtubules. The results show that the functions of HUVECs and the microtubular structure were destroyed by hypoxia, but were protected by paclitaxel and a reactive oxygen species (ROS) inhibitor. We further used western blot, a luciferase assay, and co-immunoprecipitation to describe a non-transcription-independent mechanism for PI3K activation-inhibited microtubular stability mediated by Stathmin1, a PI3K interactor that functions in microtubule depolymerization. Finally, we determined that hypoxia and ROS blocked the interaction between PI3K and Stathmin1 to activate disassembly of microtubules. CONCLUSION: Thus, our data demonstrate that hypoxia induced the production of ROS and damaged EC function by destroying the microtubular structure through the PI3K/stathmin1 pathway. BioMed Central 2019-02-18 /pmc/articles/PMC6380067/ /pubmed/30820314 http://dx.doi.org/10.1186/s13578-019-0283-1 Text en © The Author(s) 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Cao, Huaming
Yu, Dongsheng
Yan, Xueyun
Wang, Bing
Yu, Zhiming
Song, Yu
Sheng, Liang
Hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the PI3K/Stathmin1 pathway
title Hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the PI3K/Stathmin1 pathway
title_full Hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the PI3K/Stathmin1 pathway
title_fullStr Hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the PI3K/Stathmin1 pathway
title_full_unstemmed Hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the PI3K/Stathmin1 pathway
title_short Hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the PI3K/Stathmin1 pathway
title_sort hypoxia destroys the microstructure of microtubules and causes dysfunction of endothelial cells via the pi3k/stathmin1 pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6380067/
https://www.ncbi.nlm.nih.gov/pubmed/30820314
http://dx.doi.org/10.1186/s13578-019-0283-1
work_keys_str_mv AT caohuaming hypoxiadestroysthemicrostructureofmicrotubulesandcausesdysfunctionofendothelialcellsviathepi3kstathmin1pathway
AT yudongsheng hypoxiadestroysthemicrostructureofmicrotubulesandcausesdysfunctionofendothelialcellsviathepi3kstathmin1pathway
AT yanxueyun hypoxiadestroysthemicrostructureofmicrotubulesandcausesdysfunctionofendothelialcellsviathepi3kstathmin1pathway
AT wangbing hypoxiadestroysthemicrostructureofmicrotubulesandcausesdysfunctionofendothelialcellsviathepi3kstathmin1pathway
AT yuzhiming hypoxiadestroysthemicrostructureofmicrotubulesandcausesdysfunctionofendothelialcellsviathepi3kstathmin1pathway
AT songyu hypoxiadestroysthemicrostructureofmicrotubulesandcausesdysfunctionofendothelialcellsviathepi3kstathmin1pathway
AT shengliang hypoxiadestroysthemicrostructureofmicrotubulesandcausesdysfunctionofendothelialcellsviathepi3kstathmin1pathway