Cargando…

Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema

BACKGROUND: High-altitude cerebral edema (HACE) is a serious and potentially fatal brain injury that is caused by acute hypobaric hypoxia (HH) exposure. Vasogenic edema is the main pathological factor of this condition. Hypoxia-induced disruptions of tight junctions in the endothelium trigger blood‒...

Descripción completa

Detalles Bibliográficos
Autores principales: Xue, Yan, Wang, Xueting, Wan, Baolan, Wang, Dongzhi, Li, Meiqi, Cheng, Kang, Luo, Qianqian, Wang, Dan, Lu, Yapeng, Zhu, Li
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9575296/
https://www.ncbi.nlm.nih.gov/pubmed/36253854
http://dx.doi.org/10.1186/s12964-022-00976-3
_version_ 1784811288239538176
author Xue, Yan
Wang, Xueting
Wan, Baolan
Wang, Dongzhi
Li, Meiqi
Cheng, Kang
Luo, Qianqian
Wang, Dan
Lu, Yapeng
Zhu, Li
author_facet Xue, Yan
Wang, Xueting
Wan, Baolan
Wang, Dongzhi
Li, Meiqi
Cheng, Kang
Luo, Qianqian
Wang, Dan
Lu, Yapeng
Zhu, Li
author_sort Xue, Yan
collection PubMed
description BACKGROUND: High-altitude cerebral edema (HACE) is a serious and potentially fatal brain injury that is caused by acute hypobaric hypoxia (HH) exposure. Vasogenic edema is the main pathological factor of this condition. Hypoxia-induced disruptions of tight junctions in the endothelium trigger blood‒brain barrier (BBB) damage and induce vasogenic edema. Nuclear respiratory factor 1 (NRF1) acts as a major regulator of hypoxia-induced endothelial cell injury, and caveolin-1 (CAV-1) is upregulated as its downstream gene in hypoxic endothelial cells. This study aimed to investigate whether CAV-1 is involved in HACE progression and the underlying mechanism. METHODS: C57BL/6 mice were exposed to HH (7600 m above sea level) for 24 h, and BBB injury was assessed by brain water content, Evans blue staining and FITC-dextran leakage. Immunofluorescence, transmission electron microscope, transendothelial electrical resistance (TEER), transcytosis assays, and western blotting were performed to confirm the role and underlying mechanism of CAV-1 in the disruption of tight junctions and BBB permeability. Mice or bEnd.3 cells were pretreated with MβCD, a specific blocker of CAV-1, and the effect of CAV-1 on claudin-5 internalization under hypoxic conditions was detected by immunofluorescence, western blotting, and TEER. The expression of NRF1 was knocked down, and the regulation of CAV-1 by NRF1 under hypoxic conditions was examined by qPCR, western blotting, and immunofluorescence. RESULTS: The BBB was severely damaged and was accompanied by a significant loss of vascular tight junction proteins in HACE mice. CAV-1 was significantly upregulated in endothelial cells, and claudin-5 explicitly colocalized with CAV-1. During the in vitro experiments, hypoxia increased cell permeability, CAV-1 expression, and claudin-5 internalization and downregulated tight junction proteins. Simultaneously, hypoxia induced the upregulation of CAV-1 by activating NRF1. Blocking CAV-1-mediated intracellular transport improved the integrity of TJs in hypoxic endothelial cells and effectively inhibited the increase in BBB permeability and brain water content in HH animals. CONCLUSIONS: Hypoxia upregulated CAV-1 transcription via the activation of NRF1 in endothelial cells, thus inducing the internalization and autophagic degradation of claudin-5. These effects lead to the destruction of the BBB and trigger HACE. Therefore, CAV-1 may be a potential therapeutic target for HACE. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12964-022-00976-3.
format Online
Article
Text
id pubmed-9575296
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-95752962022-10-18 Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema Xue, Yan Wang, Xueting Wan, Baolan Wang, Dongzhi Li, Meiqi Cheng, Kang Luo, Qianqian Wang, Dan Lu, Yapeng Zhu, Li Cell Commun Signal Research BACKGROUND: High-altitude cerebral edema (HACE) is a serious and potentially fatal brain injury that is caused by acute hypobaric hypoxia (HH) exposure. Vasogenic edema is the main pathological factor of this condition. Hypoxia-induced disruptions of tight junctions in the endothelium trigger blood‒brain barrier (BBB) damage and induce vasogenic edema. Nuclear respiratory factor 1 (NRF1) acts as a major regulator of hypoxia-induced endothelial cell injury, and caveolin-1 (CAV-1) is upregulated as its downstream gene in hypoxic endothelial cells. This study aimed to investigate whether CAV-1 is involved in HACE progression and the underlying mechanism. METHODS: C57BL/6 mice were exposed to HH (7600 m above sea level) for 24 h, and BBB injury was assessed by brain water content, Evans blue staining and FITC-dextran leakage. Immunofluorescence, transmission electron microscope, transendothelial electrical resistance (TEER), transcytosis assays, and western blotting were performed to confirm the role and underlying mechanism of CAV-1 in the disruption of tight junctions and BBB permeability. Mice or bEnd.3 cells were pretreated with MβCD, a specific blocker of CAV-1, and the effect of CAV-1 on claudin-5 internalization under hypoxic conditions was detected by immunofluorescence, western blotting, and TEER. The expression of NRF1 was knocked down, and the regulation of CAV-1 by NRF1 under hypoxic conditions was examined by qPCR, western blotting, and immunofluorescence. RESULTS: The BBB was severely damaged and was accompanied by a significant loss of vascular tight junction proteins in HACE mice. CAV-1 was significantly upregulated in endothelial cells, and claudin-5 explicitly colocalized with CAV-1. During the in vitro experiments, hypoxia increased cell permeability, CAV-1 expression, and claudin-5 internalization and downregulated tight junction proteins. Simultaneously, hypoxia induced the upregulation of CAV-1 by activating NRF1. Blocking CAV-1-mediated intracellular transport improved the integrity of TJs in hypoxic endothelial cells and effectively inhibited the increase in BBB permeability and brain water content in HH animals. CONCLUSIONS: Hypoxia upregulated CAV-1 transcription via the activation of NRF1 in endothelial cells, thus inducing the internalization and autophagic degradation of claudin-5. These effects lead to the destruction of the BBB and trigger HACE. Therefore, CAV-1 may be a potential therapeutic target for HACE. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12964-022-00976-3. BioMed Central 2022-10-17 /pmc/articles/PMC9575296/ /pubmed/36253854 http://dx.doi.org/10.1186/s12964-022-00976-3 Text en © The Author(s) 2022 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
Xue, Yan
Wang, Xueting
Wan, Baolan
Wang, Dongzhi
Li, Meiqi
Cheng, Kang
Luo, Qianqian
Wang, Dan
Lu, Yapeng
Zhu, Li
Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema
title Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema
title_full Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema
title_fullStr Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema
title_full_unstemmed Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema
title_short Caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema
title_sort caveolin-1 accelerates hypoxia-induced endothelial dysfunction in high-altitude cerebral edema
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9575296/
https://www.ncbi.nlm.nih.gov/pubmed/36253854
http://dx.doi.org/10.1186/s12964-022-00976-3
work_keys_str_mv AT xueyan caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT wangxueting caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT wanbaolan caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT wangdongzhi caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT limeiqi caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT chengkang caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT luoqianqian caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT wangdan caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT luyapeng caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema
AT zhuli caveolin1accelerateshypoxiainducedendothelialdysfunctioninhighaltitudecerebraledema