Phillygenin inhibits neuroinflammation and promotes functional recovery after spinal cord injury via TLR4 inhibition of the NF-κB signaling pathway

IF 5.9 1区 医学 Q1 ORTHOPEDICS
{"title":"Phillygenin inhibits neuroinflammation and promotes functional recovery after spinal cord injury via TLR4 inhibition of the NF-κB signaling pathway","authors":"","doi":"10.1016/j.jot.2024.07.013","DOIUrl":null,"url":null,"abstract":"<div><h3>Background</h3><p>Spinal cord injuries (SCIs) trigger a cascade of detrimental processes, encompassing neuroinflammation and oxidative stress (OS), ultimately leading to neuronal damage. Phillygenin (PHI), isolated from forsythia, is used in a number of biomedical applications, and is known to exhibit anti-neuroinflammation activity. In this study, we investigated the role and mechanistic ability of PHI in the activation of microglia-mediated neuroinflammation and subsequent neuronal apoptosis following SCI.</p></div><div><h3>Methods</h3><p>A rat model of SCI was used to investigate the impact of PHI on inflammation, axonal regeneration, neuronal apoptosis, and the restoration of motor function. <em>In vitro</em>, neuroinflammation models were induced by stimulating microglia with lipopolysaccharide (LPS); then, we investigated the influence of PHI on pro-inflammatory mediator release in LPS-treated microglia along with the underlying mechanisms. Finally, we established a co-culture system, featuring microglia and VSC 4.1 cells, to investigate the role of PHI in the activation of microglia-mediated neuronal apoptosis.</p></div><div><h3>Results</h3><p><em>In vivo</em>, PHI significantly inhibited the inflammatory response and neuronal apoptosis while enhancing axonal regeneration and improving motor function recovery. <em>In vitro,</em> PHI inhibited the release of inflammation-related factors from polarized BV2 cells in a dose-dependent manner. The online Swiss Target Prediction database predicted that toll-like receptor 4 (TLR4) was the target protein for PHI. In addition, Molecular Operating Environment software was used to perform molecular docking for PHI with the TLR4 protein; this resulted in a binding energy interaction of −6.7 kcal/mol. PHI inhibited microglia-mediated neuroinflammation, the production of reactive oxygen species (ROS), and activity of the NF-κb signaling pathway. PHI also increased mitochondrial membrane potential (MMP) in VSC 4.1 neuronal cells. In BV2 cells, PHI attenuated the overexpression of TLR4-induced microglial polarization and significantly suppressed the release of inflammatory cytokines.</p></div><div><h3>Conclusion</h3><p>PHI ameliorated SCI-induced neuroinflammation by modulating the TLR4/MYD88/NF-κB signaling pathway. PHI has the potential to be administered as a treatment for SCI and represents a novel candidate drug for addressing neuroinflammation mediated by microglial cells.</p></div><div><h3>The translational potential of this article</h3><p>We demonstrated that PHI is a potential drug candidate for the therapeutic management of SCI with promising developmental and translational applications.</p></div>","PeriodicalId":16636,"journal":{"name":"Journal of Orthopaedic Translation","volume":null,"pages":null},"PeriodicalIF":5.9000,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2214031X24000858/pdfft?md5=9725131673a30662a4fc91b5d60a92a0&pid=1-s2.0-S2214031X24000858-main.pdf","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Orthopaedic Translation","FirstCategoryId":"3","ListUrlMain":"https://www.sciencedirect.com/science/article/pii/S2214031X24000858","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ORTHOPEDICS","Score":null,"Total":0}
引用次数: 0

Abstract

Background

Spinal cord injuries (SCIs) trigger a cascade of detrimental processes, encompassing neuroinflammation and oxidative stress (OS), ultimately leading to neuronal damage. Phillygenin (PHI), isolated from forsythia, is used in a number of biomedical applications, and is known to exhibit anti-neuroinflammation activity. In this study, we investigated the role and mechanistic ability of PHI in the activation of microglia-mediated neuroinflammation and subsequent neuronal apoptosis following SCI.

Methods

A rat model of SCI was used to investigate the impact of PHI on inflammation, axonal regeneration, neuronal apoptosis, and the restoration of motor function. In vitro, neuroinflammation models were induced by stimulating microglia with lipopolysaccharide (LPS); then, we investigated the influence of PHI on pro-inflammatory mediator release in LPS-treated microglia along with the underlying mechanisms. Finally, we established a co-culture system, featuring microglia and VSC 4.1 cells, to investigate the role of PHI in the activation of microglia-mediated neuronal apoptosis.

Results

In vivo, PHI significantly inhibited the inflammatory response and neuronal apoptosis while enhancing axonal regeneration and improving motor function recovery. In vitro, PHI inhibited the release of inflammation-related factors from polarized BV2 cells in a dose-dependent manner. The online Swiss Target Prediction database predicted that toll-like receptor 4 (TLR4) was the target protein for PHI. In addition, Molecular Operating Environment software was used to perform molecular docking for PHI with the TLR4 protein; this resulted in a binding energy interaction of −6.7 kcal/mol. PHI inhibited microglia-mediated neuroinflammation, the production of reactive oxygen species (ROS), and activity of the NF-κb signaling pathway. PHI also increased mitochondrial membrane potential (MMP) in VSC 4.1 neuronal cells. In BV2 cells, PHI attenuated the overexpression of TLR4-induced microglial polarization and significantly suppressed the release of inflammatory cytokines.

Conclusion

PHI ameliorated SCI-induced neuroinflammation by modulating the TLR4/MYD88/NF-κB signaling pathway. PHI has the potential to be administered as a treatment for SCI and represents a novel candidate drug for addressing neuroinflammation mediated by microglial cells.

The translational potential of this article

We demonstrated that PHI is a potential drug candidate for the therapeutic management of SCI with promising developmental and translational applications.

Abstract Image

通过 TLR4 抑制 NF-κB 信号通路抑制神经炎症并促进脊髓损伤后的功能恢复
脊髓损伤(SCI)会引发一连串的有害过程,包括神经炎症和氧化应激(OS),最终导致神经元损伤。从连翘中分离出来的菲利根因(PHI)被广泛应用于生物医学领域,并具有抗神经炎症的活性。在本研究中,我们研究了 PHI 在 SCI 后激活小胶质细胞介导的神经炎症和随后的神经细胞凋亡中的作用和机制能力。我们用大鼠 SCI 模型来研究 PHI 对炎症、轴突再生、神经元凋亡和运动功能恢复的影响。首先,我们用脂多糖(LPS)刺激小胶质细胞诱导神经炎症模型;然后,我们研究了 PHI 对 LPS 处理的小胶质细胞释放促炎介质的影响及其内在机制。最后,我们建立了一个以小胶质细胞和 VSC 4.1 细胞为特征的共培养系统,以研究 PHI 在激活小胶质细胞介导的神经元凋亡中的作用。结果表明,PHI能明显抑制炎症反应和神经元凋亡,同时促进轴突再生并改善运动功能的恢复。PHI 以剂量依赖的方式抑制了极化 BV2 细胞释放炎症相关因子。在线瑞士靶点预测数据库预测收费样受体4(TLR4)是PHI的靶蛋白。此外,还使用分子操作环境软件对 PHI 与 TLR4 蛋白进行了分子对接,结果发现两者的结合能为 -6.7 kcal/mol。PHI 可抑制小胶质细胞介导的神经炎症、活性氧(ROS)的产生以及 NF-κb 信号通路的活性。PHI 还能提高 VSC 4.1 神经元细胞的线粒体膜电位(MMP)。在 BV2 细胞中,PHI 可减轻 TLR4 诱导的小胶质细胞极化的过度表达,并显著抑制炎性细胞因子的释放。PHI 通过调节 TLR4/MYD88/NF-κB 信号通路,改善了 SCI 诱导的神经炎症。PHI具有治疗SCI的潜力,是解决由小胶质细胞介导的神经炎症的新型候选药物。我们证明 PHI 是治疗 SCI 的潜在候选药物,具有广阔的开发和转化应用前景。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
Journal of Orthopaedic Translation
Journal of Orthopaedic Translation Medicine-Orthopedics and Sports Medicine
CiteScore
11.80
自引率
13.60%
发文量
91
审稿时长
29 days
期刊介绍: The Journal of Orthopaedic Translation (JOT) is the official peer-reviewed, open access journal of the Chinese Speaking Orthopaedic Society (CSOS) and the International Chinese Musculoskeletal Research Society (ICMRS). It is published quarterly, in January, April, July and October, by Elsevier.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信