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

Cell Commun Signal. 2022 Oct 17;20(1):160. doi: 10.1186/s12964-022-00976-3.

Abstract

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. Video abstract.

Keywords: Blood‒brain barrier; Caveolin-1; Claudin-5; Endothelium; High-altitude cerebral edema.

Publication types

  • Video-Audio Media
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Altitude
  • Animals
  • Blood-Brain Barrier
  • Brain Edema* / complications
  • Brain Edema* / metabolism
  • Caveolin 1* / metabolism
  • Claudin-5 / metabolism
  • Endothelial Cells / metabolism
  • Hypoxia* / complications
  • Hypoxia* / metabolism
  • Mice
  • Mice, Inbred C57BL
  • Nuclear Respiratory Factor 1 / metabolism
  • Tight Junction Proteins / metabolism
  • Tight Junctions / metabolism

Substances

  • Caveolin 1
  • Claudin-5
  • Nuclear Respiratory Factor 1
  • Tight Junction Proteins
  • Cav1 protein, mouse