Journal of Biological Chemistry
Volume 295, Issue 33, 14 August 2020, Pages 11877-11890
Journal home page for Journal of Biological Chemistry

Protein Synthesis and Degradation
The NADPH oxidase NOX4 promotes the directed migration of endothelial cells by stabilizing vascular endothelial growth factor receptor 2 protein

https://doi.org/10.1074/jbc.RA120.014723Get rights and content
Under a Creative Commons license
open access

Directed migration of endothelial cells (ECs) is an important process during both physiological and pathological angiogenesis. The binding of vascular endothelial growth factor (VEGF) to VEGF receptor-2 (VEGFR-2) on the EC surface is necessary for directed migration of these cells. Here, we used TAXIScan, an optically accessible real-time horizontal cell dynamics assay approach, and demonstrate that reactive oxygen species (ROS)-producing NADPH oxidase 4 (NOX4), which is abundantly expressed in ECs, mediates VEGF/VEGFR-2-dependent directed migration. We noted that a continuous supply of endoplasmic reticulum (ER)-retained VEGFR-2 to the plasma membrane is required to maintain VEGFR-2 at the cell surface. siRNA-mediated NOX4 silencing decreased the ER-retained form of VEGFR-2, resulting in decreased cell surface expression levels of the receptor. We also found that ER-localized NOX4 interacts with ER-retained VEGFR-2 and thereby stabilizes this ER-retained form at the protein level in the ER. We conclude that NOX4 contributes to the directed migration of ECs by maintaining VEGFR-2 levels at their surface.

NADPH oxidase
Nox4
vascular endothelial growth factor receptor 2 (VEGFR-2)
endothelial cell migration
angiogenesis
capillary formation
reactive oxygen species (ROS)
redox signaling
endoplasmic reticulum (ER)

Cited by (0)

This article contains supporting information.

Author contributions—K. M., S. O., A. Y., and F. K. conceptualization; K. M. and S. O. data curation; K. M., S. O., A. Y., C. K., M. K., T. K., M. Tamura, and F. K. formal analysis; K. M., S. O., C. K., M. K., T. K., M. Tamura, and F. K. funding acquisition; K. M., S. O., and C. K. investigation; K. M., S. O., A. Y., C. K., M. K., M. Tamura, M. Taura, and F. K. methodology; K. M., S. O., and M. Taura writing-original draft; K. M. and S. O. project administration; K. M., S. O., A. Y., M. K., T. K., M. Tamura, and F. K. writing-review and editing; M. Taura validation.

Funding and additional information—This study was supported in part by JSPS KAKENHI grant numbers JP17K08637 (to K. M.), JP18K07804 (to F. K.), and JP19K07676 (to A. Y.), in part by the Wesco Scientific Promotion Foundation (to K. M. and A. Y.), in part by the Ryobi Teien Memory Foundation (to K. M. and A. Y.), and in part by Research Project Grants (nos. R01S-003 (to K. M.), H30Y-002 (to S. O.), R01B-081 (to F. K.), and R01B-093 (to A. Y.)) from Kawasaki Medical School.

Conflict of interest—The authors declare that they have no conflicts of interest with the contents of this article.

Abbreviations—The abbreviations used are:

    ECs

    endothelial cells

    VEGF

    vascular endothelial growth factor

    ROS

    reactive oxygen species

    ER

    endoplasmic reticulum

    DPI

    diphenyleneiodonium

    NAC

    N-acetylcysteine

    HVA

    homovanillic acid

    PI

    propidium iodide

    Endo H

    endoglycosidase H

    BFA

    brefeldin A

    CHX

    cycloheximide

    VD

    velocity–directionality

    PNGase F

    peptide:N-glycosidase F.

These authors contributed equally to this work.