Open Access

Lipoxin A4 attenuates uric acid‑activated, NADPH oxidase‑dependent oxidative stress by interfering with translocation of p47phox in human umbilical vein endothelial cells

  • Authors:
    • You Zhou
    • Hui You
    • Aijie Zhang
    • Xingliang Jiang
    • Zheyan Pu
    • Guoqiang Xu
    • Mingcai Zhao
  • View Affiliations

  • Published online on: May 28, 2020     https://doi.org/10.3892/etm.2020.8812
  • Pages: 1682-1692
  • Copyright: © Zhou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

LipoxinA4 (LXA4) is a well‑known key mediator of endogenous anti‑inflammation and of the resolution of inflammation. Considerable oxidative stress occurs during inflammation due to the generation of reactive oxidative species (ROS). Moreover, high levels of uric acid (UA) contribute to endothelial cell dysfunction, which can promote disease‑related morbidity, and NADPH oxidase‑derived ROS are crucial regulatory factors in these responses. However, LXA4 also has the potential to reduce oxidative stress. The aim of the present study was to examine whether LXA4 could suppress UA‑induced oxidative stress in human umbilical vein endothelial cells (HUVECs) and to investigate its mechanisms of action in vitro. HUVECs were incubated with or without LXA4, followed by the addition of UA. ROS levels were then measured using 2,7‑dichlorodihydrofluorescein diacetate and lucigenin‑enhanced chemiluminescence was used to evaluate NADPH oxidase activity. p47phox or p22phox small interfering (si)RNA were transfected into HUVECs and protein levels of p47phox were detected using western blot analysis. LXA4 significantly inhibited UA‑induced generation of ROS to the same extent as the NADPH oxidase inhibitor, diphenyleneiodonium chloride. Notably, transfection of p47phox siRNA attenuated the generation of ROS and the activation of NADPH oxidase. Cells transfected with p22phox siRNA demonstrated a significant reduction in the expression of p47phox on the membrane. Further experiments demonstrated that LXA4 interfered with the transfer of p47phox from the cytoplasm to the cell membrane. These findings suggested that LXA4 inhibited the release of NADPH oxidase derived ROS in HUVECs stimulated by UA. A potential mechanism of action underlying this effect could be LXA4‑mediated suppression of NADPH oxidase activity, leading to inhibition of p47phox translocation from the cytoplasm to the cell membrane.
View Figures
View References

Related Articles

Journal Cover

August-2020
Volume 20 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou Y, You H, Zhang A, Jiang X, Pu Z, Xu G and Zhao M: Lipoxin A4 attenuates uric acid‑activated, NADPH oxidase‑dependent oxidative stress by interfering with translocation of p47phox in human umbilical vein endothelial cells . Exp Ther Med 20: 1682-1692, 2020
APA
Zhou, Y., You, H., Zhang, A., Jiang, X., Pu, Z., Xu, G., & Zhao, M. (2020). Lipoxin A4 attenuates uric acid‑activated, NADPH oxidase‑dependent oxidative stress by interfering with translocation of p47phox in human umbilical vein endothelial cells . Experimental and Therapeutic Medicine, 20, 1682-1692. https://doi.org/10.3892/etm.2020.8812
MLA
Zhou, Y., You, H., Zhang, A., Jiang, X., Pu, Z., Xu, G., Zhao, M."Lipoxin A4 attenuates uric acid‑activated, NADPH oxidase‑dependent oxidative stress by interfering with translocation of p47phox in human umbilical vein endothelial cells ". Experimental and Therapeutic Medicine 20.2 (2020): 1682-1692.
Chicago
Zhou, Y., You, H., Zhang, A., Jiang, X., Pu, Z., Xu, G., Zhao, M."Lipoxin A4 attenuates uric acid‑activated, NADPH oxidase‑dependent oxidative stress by interfering with translocation of p47phox in human umbilical vein endothelial cells ". Experimental and Therapeutic Medicine 20, no. 2 (2020): 1682-1692. https://doi.org/10.3892/etm.2020.8812