Elsevier

Vascular Pharmacology

Volume 73, October 2015, Pages 149-157
Vascular Pharmacology

Propofol ameliorates endothelial inflammation induced by hypoxia/reoxygenation in human umbilical vein endothelial cells: Role of phosphatase A2

https://doi.org/10.1016/j.vph.2015.06.002Get rights and content

Abstract

Hypoxia/reoxygenation (H/R) induces endothelial inflammation with augmentation of endothelial adhesion molecules over-expression. Propofol was reported to attenuate endothelial adhesion molecule expression in some situations. Here, we examined the molecular mechanism for how propofol restored H/R-mediated up-regulation of endothelial adhesion molecules in human umbilical vein endothelial cells (HUVECs). Compared with the control group, H/R up-regulated expression of Pin-1 and PP2A, increased p66Shc–Ser36 phosphorylation, induced p66Shc mitochondrial translocation, O2 accumulation and NF-κB activation, and decreased eNOS–Ser1177 phosphorylation and nitric oxide (NO) production, thus up-regulating expression of endothelial adhesion molecules and increasing mononuclear-endothelial interaction. More importantly, except that propofol had no effect on H/R-induced p66Shc–Ser36 phosphorylation, most of H/R-mediated changes were alleviated by propofol, resulting in the reduction of endothelial adhesion molecules expression and mononuclear-endothelial adhesion. Moreover, we demonstrated the protective effect of propofol on H/R-induced endothelial inflammation was similar to that of calyculin A, an inhibitor of PP2A. In contrast, FTY720, an activator of PP2A, antagonized the effect of propofol. Our data indicated that propofol down-regulated PP2A expression, leading to reduced dephosphorylation of p66Shc–Ser36 and eNOS–Ser1177, which is associated with ROS accumulation and NO reduction, resulting in inhibition of endothelial adhesion molecule expression and mononuclear-endothelial interaction.

Introduction

Hypoxia/reoxygenation (H/R) in an in vitro situation mimics the ischemia/reperfusion (I/R) model in vivo. It leads to the accumulation of inflammatory cytokines [1], [2] and activation of inflammatory signaling pathways in endothelial cells [3]. Endothelial inflammation consists of the augmentation of endothelial adhesion molecule expression, such as intercellular adhesion molecule 1(ICAM-1) and endothelial selectin (E-selectin). The increased expression of endothelial adhesion molecules induces mononuclear-endothelial adhesion, which leads to endothelial injury [4].

The nuclear factor kappa B (NF-κB) signal pathway was reported to be involved in H/R-induced endothelial adhesion molecules expression [5]. Previous data have indicated that reactive oxygen species (ROS) could activate NF-κB, which plays an important role in ROS-mediated endothelial injury due to H/R [6]. Mitochondrion is the major organelle where ROS is generated [7], [8]. Furthermore, the p66Shc adaptor protein is important in regulating mitochondrial ROS generation [9], [10], [11]. The adjustment of p66Shc function is a complex process. First, p66Shc is phosphorylated at the site of Ser36, then Ser36–phosphorylated p66Shc is isomerized with a prolylisomerase Pin1 and dephosphorylated by the phosphatase A2 (PP2A) [12]. Only isomerized and dephosphorylated p66Shc could translocate to the mitochondria and subsequently lead to ROS generation [13].

It was also reported that reduced nitric oxide (NO) production from vascular endothelium induced endothelial adhesion molecule expression and leukocyte-endothelium adhesion [14], [15], resulting in endothelial inflammation. Moreover, improvement of NO production could protect against H/R-induced endothelial injury [16]. And this effect was reversed by L-NANE, an endothelial NO synthase (eNOS) inhibitor [16].

Propofol (2, 6-diisopropylphenol) is a widely used intravenous anesthetic agent. It was reported that propofol could inhibit the expression of endothelial adhesion molecules in H/R-treated endothelial cells [17]. However, the mechanism by which propofol protects against H/R-induced adhesion molecules expression is still elusive. In the present study, we examined whether and how propofol protects HUVECs against H/R-induced endothelial adhesion molecules expression.

Section snippets

Cell culture and reagents

HUVECs (Clonetics) were incubated in Dulbecco's Modified Eagle's Medium (DMEM) and 10% fetal bovine serum in incubator containing 5% CO2 at 37 °C. Cells were sub-cultured when reaching around 90% confluence. The fourth passage of HUVECs was employed in the present study.

Propofol (sigma), calyculin A (sigma) and FTY720 (sigma) were dissolved in dimethyl sulfoxide (DMSO) (sigma). The final concentration of DMSO was regulated to 0.01% for each medium to minimize any potential toxicity or

H/R-induced mononuclear-endothelial adhesion and endothelial adhesion molecules expression, and its modulation by propofol

In HUVECs, compared with control group, H/R induced more mononuclear-endothelial adhesion. Pre-incubation of cells with 1 μM propofol for 30 min had no effect on baseline mononuclear-endothelial adhesion (Fig. 1A). When pre-treatment of propofol reached 5 or 25 μM, the mononuclear-endothelial adhesion was inhibited. Therefore propofol worked in a dose-dependent manner. To be noted, the solvent 0.01% of dimethyl sulfoxide (DMSO) did not show any significant impact on H/R-induced

Discussion

The major finding of the present study is that H/R induced p66Shc–Ser36 phosphorylation and p66Shc mitochondrial translocation, which resulted in ROS accumulation, NF-κB activation and translocation to nucleus; H/R also inhibited eNOS–Ser1177 phosphorylation and NO production, thus augmenting endothelial adhesion molecules expression and increasing mononuclear-endothelial adhesion. More importantly, propofol was observed to protect HUVECs against H/R-induced endothelial adhesion molecules

Conflict of interest

None declared.

Acknowledgments

This project is supported by the Natural Science Foundation of Shanghai (No. 12ZR1406700).

References (38)

  • M. Zhu et al.

    Propofol protects human umbilical vein endothelial cells from cisplatin-induced injury

    Vasc. Pharmacol.

    (2014)
  • R. Shreeniwas et al.

    Hypoxia-mediated induction of endothelial cell interleukin-1: an autocrine mechanism promoting expression of leukocyte adhesion molecules on the vessel surface

    J. Clin. Invest.

    (1992)
  • Y. Ala et al.

    Hypoxia/reoxygenation stimulates endothelial cells to promote interleukin-1 and interleukin-6 production: effects of free radical scavengers

    Agents Actions

    (1992)
  • H. Ichikawa et al.

    Molecular mechanisms of anoxia/reoxygenation-induced neutrophil adherence to cultured endothelial cells

    Circ. Res.

    (1997)
  • W. Sluiter et al.

    Leukocyte adhesion molecules on the vascular endothelium: their role in the pathogenesis of cardiovascular disease and the mechanisms underlying their expression

    J. Cardiovasc. Pharmacol.

    (1993)
  • H. Xie et al.

    NF-κB activation plays a role in superoxide-mediated cerebral endothelial dysfunction after hypoxia/reoxygenation

    Stroke

    (2005)
  • D.E. Handy et al.

    Redox regulation of mitochondrial function

    Antioxid. Redox Signal.

    (2012)
  • E. Migliaccio et al.

    The p66Shc adaptor protein controls oxidative stress response and life span in mammals

    Nature

    (1999)
  • M. Trinei et al.

    A p53-p66Shc signalling pathway controls intracellular redox status, levels of oxidation-damaged DNA and oxidative stress-induced apoptosis

    Oncogene

    (2002)
  • Cited by (16)

    • Multitarget neuroprotection by quercetin: Changes in gene expression in two perinatal asphyxia models

      2021, Neurochemistry International
      Citation Excerpt :

      It would be important therefore to further analyze the effective change in this phosphatase's activity in the rat model. If a decreased activity could be demonstrated after quercetin treatment we could hypothesize that, as was described for propofol protective action (Zhu et al., 2015), a decrease in PP2A expression could control endothelial inflammation induced by HIE. On the other hand, as addressed by Kruse et al. (2020), the regulatory subunit affects the phosphorylation site preference of the holoenzyme catalytic subunit, suggesting that quercetin could be redirecting phosphatase activity to different substrates.

    • The mystery of mitochondria-ER contact sites in physiology and pathology: A cancer perspective

      2020, Biochimica et Biophysica Acta - Molecular Basis of Disease
      Citation Excerpt :

      Shc adaptor proteins bind to activated receptor tyrosine kinases (RTKs) in response to various growth factors [202] and participate in the regulation of cellular proliferation and differentiation [203]. The Ser36 residue located in the unique p66Shc collagen homology domain (CH2) is phosphorylated by either protein kinase Cβ II (PKCβII) [204] or c-Jun N-terminal kinase (JNK) [205] in response to various environmental and intracellular stress stimuli, such as H2O2 or Taxol treatment [206], hypoxia/reoxygenation injury [207–210], UV radiation [211], and high-glucose environments [212–214], as well as in response to metabolic stresses [215]. After isomerization by peptidyl-prolyl cis-trans isomerase 1 (PIN1) and dephosphorylation by PP2A, p66Shc is translocated into mitochondria [204]; however, MAM-localized p66Shc may also be able to affect mitochondria.

    • Propofol post-conditioning lessens renal ischemia/reperfusion-induced acute lung injury associated with autophagy and apoptosis through MAPK signals in rats

      2020, Gene
      Citation Excerpt :

      These properties lead to inflammatory mediators being released from lung inflammatory cells, and their serum concentrations were parallel to the degree of pulmonary injury. Pro offers defense by attenuating reactive oxygen species and inflammatory mediators (Zhu et al., 2015). To date, mechanisms by which Pro reduces inflammatory responses have not yet been elucidated, but may involve inhibition of MAPK signaling.

    • Ketamine ameliorates hypoxia-induced endothelial injury in human umbilical vein endothelial cells

      2020, Clinics
      Citation Excerpt :

      Ischemia-induced endothelial injury that occurs after hypoxia results in the increased expression of cell adhesion molecules, such as ICAM-1 and E-selectin, which interact with circulating neutrophils, inducing them to migrate to the sites of endothelium, to further aggravate endothelial cell inflammation and damage (21). Moreover, ROS also play a vital role in hypoxia-mediated endothelial inflammation (22). An increase in ROS accumulation under hypoxic conditions can lead to endothelial cell apoptosis and necrosis (6).

    • Regulation of the phosphoprotein phosphatase 2A system and its modulation during oxidative stress: A potential therapeutic target?

      2019, Pharmacology and Therapeutics
      Citation Excerpt :

      Several groups have also shown hypoxia to upregulate PP2A. In human colorectal cancer cells, rat primary alveolar epithelial cells and mouse heart, hypoxia increases PP2A activity (Caraballo et al., 2011; Larsen et al., 2008; Lin, Lee, et al., 2012), PP2A mRNA and protein abundance (Chen et al., 2014; Lu et al., 2017; Zhu et al., 2015). While hyperoxia (95% O2, 1-24 h) decreases PPA activity, independently of altering PP2A abundance in macrophages (Nyunoya, Monick, Powers, Yarovinsky, & Hunninghake, 2005).

    View all citing articles on Scopus
    1

    Equal contributors.

    View full text