Elsevier

Cellular Signalling

Volume 76, December 2020, 109781
Cellular Signalling

Notoginsenoside R1 alleviates oxidized low-density lipoprotein-induced apoptosis, inflammatory response, and oxidative stress in HUVECS through modulation of XIST/miR-221-3p/TRAF6 axis

https://doi.org/10.1016/j.cellsig.2020.109781Get rights and content

Highlights

  • Notoginsenoside R1 inhibited apoptosis and inflammatory response in HUVECS caused by ox-LDL.

  • Overexpression of XIST abolished the protective effects of Notoginsenoside R1 on HUVECS treated with ox-LDL.

  • Notoginsenoside R1 prevented ox-LDL-induced effects on HUVECS cells by regulating the XIST/miR-221-3p/TRAF6 axis.

Abstract

Background:

Atherosclerosis is a common reason for acute cardio-cerebral vascular diseases. The purpose of this study was to clarify the functional effects of Notoginsenoside R1 (NGR1) on atherosclerosis.

Methods

HUVECS were exposed to oxidized low-density lipoprotein (ox-LDL) in the current study. The proliferation ability of HUVECS was measured by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazol-3-ium bromide (MTT). Flow cytometry assays were performed to evaluate the apoptosis of HUVECS. Ox-LDL caused inflammatory response and oxidative stress were assessed by checking pro-inflammatory cytokines and intracellular reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD). The regulatory roles of NGR1 in HUVECs were measured by real-time quantitative polymerase chain reaction (RT-qPCR) and western blot assays. The interaction relationship between miR-221-3p and X-inactive specific transcript (XIST) or TNF-receptor-associated factor 6 (TRAF6) was predicted by bioinformatics tools. Dual-luciferase reporter and RNA pull-down assays were used to confirm the interaction relationship.

Results

Currently, ox-LDL inhibited proliferation and induced apoptosis, inflammatory response, and oxidative stress in HUVECs, which were alleviated by treatment with NGR1. Importantly, the increase of XIST in ox-LDL-induced HUVECs was abolished by NGR1. In addition, the gain-of-functional experiment suggested that the upregulation of XIST neutralized the protection effects of NGR1 in HUVECs treated with ox-LDL. In addition, miR-221-3p was a target of XIST in HUVECs as confirmed by dual-luciferase reporter and RNA pull-down assays. Furthermore, miR-221-3p interacted with TRAF6, and NGR1 regulated proliferation, apoptosis, inflammatory response, and oxidative stress in HUVECs exposed to ox-LDL by regulation of the XIST/miR-221-3p/TRAF6 axis through Nuclear Factor Kappa B (NF-κB) pathway.

Conclusion

NGR1 could exert regulatory functions in ox-LDL-induced HUVECS by regulation of XIST/miR-221-3p/TRAF6 axis, which provided valuable insights into the development of potential therapeutic strategy for atherosclerosis.

Introduction

Atherosclerosis, as an inflammatory disease, is the main contributor to cardiovascular diseases-related vascular diseases, leading to myocardial infarction, stoke, and heart failure [1]. Inflammation response and infiltration of leukocytes were major characterization at whole stages of the atherosclerotic process [2]. Consistently, high levels of inflammatory cytokines were closely correlated with plaque progression in atherosclerosis [3]. Previous studies well clarified that Notoginsenoside R1 (NGR1) has beneficial effects on animal models of cardiovascular diseases [[4], [5], [6]]. Importantly, NGR1, a major active ingredient of Radix notoginseng, was reported to alleviate oxidized low-density lipoprotein (ox-LDL)-caused inflammatory response by decreasing the cytokines production in endothelial cells through modulation of Nuclear Factor Kappa B (NF-κB) signal path [7], while ox-LDL was reported to be an important risk factor contributing to the progression of atherosclerosis [8]. However, the detailed mechanism by which NGR1 involved in atherosclerosis development remained largely elusive.

It was widely accepted that long non-coding RNAs (lncRNAs, longer than 200 nucleotides) act predominant roles in various pathologies, including atherosclerosis [9,10]. For instance, lncRNA H19 was increased in the blood sample of atherosclerosis compared with control, and it could be a potential target for treating atherosclerosis [11]. The lncRNA X-inactive specific transcript (XIST) was transcribed from the XIST gene and locate on Xq13.2 (1–19,296). According previous reports, XIST was closely related to the development of many human cancers, including pancreatic cancer and breast cancer [12,13]. In addition, dysregulation of XIST could act as a biomarker for poor prognosis of cancer patients [14]. Nevertheless, the expression pattern and function of XIST in atherosclerosis were scarcely documented.

Furthermore, microRNAs (miRNAs) have been demonstrated as vital regulators in atherosclerosis [15]. Moreover, miR-221-3p has attracted attention for its prognostic value in multiple human diseases, such as colon cancer [16], triple-negative breast cancer [17], and acute myocardial infarction [18]. Furthermore, research by Xue et al. claimed that miR-221 was involved in the dysfunction of endothelial cells [19]. Nevertheless, the regulatory of miR-221 in the pathogenesis of atherosclerosis still require further investigation.

TNF-receptor-associated factor 6 (TRAF6), a member of TNF-receptor-associated factor family, mediated multiple signaling signals transduction, including TNF receptor (TNFR) family, IL-18 receptor (IL-18R), and Toll-like receptors [20]. A study indicated that TRAF6 participated in the development of atherosclerosis [21].

According previous researches, NGR1 exerted its anti-inflammation function in atherosclerosis and other inflammation-related diseases. Therefore, the investigation for the detailed mechanism by which NGR1 regulated atherosclerosis development was meaningful for atherosclerosis treatment. Thus, we explored the effects and underlying mechanism of NGR1 on apoptosis and inflammation in endothelial cells stimulated by ox-LDL.

Section snippets

Cell lines

Human umbilical vein endothelial cells (HUVECs) were derived from the Chinese Academy of Sciences (Shanghai, China). HUVECs were cultured in RPMI 1640 medium (Wisent, Shanghai, China) contained with 10% (v/v) fetal bovine serum (FBS; Hyclone, South Logan, UT, USA) and 1% penicillin/streptomycin in a humidified atmosphere containing 5% CO2 at 37 °C. In addition, ox-LDL was purchased from Yiyuan Biotechnologies (Guangzhou, China). NGR1 (HPLC≥98%; Yiyuan Biotechnologies) was dissolved in dimethyl

NGR1 mitigated ox-LDL-induced apoptosis, oxidative stress, and inflammatory response in HUVECs

To begin with, ox-LDL inhibited the proliferation of HUVECs, which was weakened by treatment with 30 μM of NGR1 (Fig. 1A). Additionally, NGR1 protected HUVECs from ox-LDL-induced apoptosis (Fig. 1B). Inflammatory factors were upregulated in HUVECs after treatment with ox-LDL, including TNF-α, IL-6, and IL-1β, which was overturned in HUVECs by treatment with NGR1 (Fig. 1C-E). We also noticed that the enhancement of ROS induced by ox-LDL in HUVECs was reduced by NGR1 (Fig. 1F). Analysis results

Discussion

Although the inducement and pathogenesis mechanisms of atherosclerosis remain unclear thoroughly, injury and apoptosis of endothelial cell is a critical step during the development of atherosclerosis [23]. Therefore, ox-LDL was used to trigger the injury of endothelial cells and aggravate the process of plaque development and instability, which was widely reported in atherosclerosis-related researches [24]. As for pathogenesis mechanisms of atherosclerosis, apoptosis of vascular endothelial

Funding

This work was supported by The Project of Science and Technology of Henan Province [Grant No. 162102310031]; and The Project of Science and Technology of Zhengzhou Science and Technology Bureau [Grant No. 153PKJGG058].

Declaration of Competing Interest

The authors declare that they have no financial conflicts of interest.

Acknowledgement

None.

References (38)

  • Q. Wei et al.

    MiR-345-3p attenuates apoptosis and inflammation caused by oxidized low-density lipoprotein by targeting TRAF6 via TAK1/p38/NF-kB signaling in endothelial cells

    Life Sci.

    (2020)
  • S. Pant et al.

    Inflammation and atherosclerosis—revisited

    J. Cardiovasc. Pharmacol. Ther.

    (2014)
  • E. Ammirati et al.

    Markers of inflammation associated with plaque progression and instability in patients with carotid atherosclerosis

    Mediat. Inflamm.

    (2015)
  • C. Jia et al.

    Notoginsenoside R1 attenuates atherosclerotic lesions in ApoE deficient mouse model

    PLoS One

    (2014)
  • W.-J. Liu et al.

    Notoginsenoside R1 attenuates renal ischemia-reperfusion injury in rats

    Shock

    (2010)
  • Y. Yang et al.

    Notoginsenoside R1 reduces blood pressure in spontaneously hypertensive rats through a long non-coding RNA AK094457

    Int. J. Clin. Exp. Pathol.

    (2015)
  • U. Rueckschloss et al.

    Induction of NAD (P) H oxidase by oxidized low-density lipoprotein in human endothelial cells: antioxidative potential of hydroxymethylglutaryl coenzyme a reductase inhibitor therapy

    Circulation

    (2001)
  • T.R. Mercer et al.

    Structure and function of long noncoding RNAs in epigenetic regulation

    Nat. Struct. Mol. Biol.

    (2013)
  • J. Pan

    LncRNA H19 promotes atherosclerosis by regulating MAPK and NF-kB signaling pathway

    Eur. Rev. Med. Pharmacol. Sci.

    (2017)
  • Cited by (0)

    1

    Jian Zhao and Lin Cui contributed equally to this work.

    View full text