Cerebrolysin alleviates early brain injury after traumatic brain injury by inhibiting neuroinflammation and apoptosis via TLR signaling pathway.

IF 1.1 4区 医学 Q3 SURGERY Acta cirurgica brasileira Pub Date : 2022-09-05 eCollection Date: 2022-01-01 DOI:10.1590/acb370605
Weihong Lu, Zhonghua Zhu, Dongliang Shi, Xiaoyu Li, Jingzhi Luo, Xingzhi Liao
{"title":"Cerebrolysin alleviates early brain injury after traumatic brain injury by inhibiting neuroinflammation and apoptosis via TLR signaling pathway.","authors":"Weihong Lu,&nbsp;Zhonghua Zhu,&nbsp;Dongliang Shi,&nbsp;Xiaoyu Li,&nbsp;Jingzhi Luo,&nbsp;Xingzhi Liao","doi":"10.1590/acb370605","DOIUrl":null,"url":null,"abstract":"<p><strong>Purpose: </strong>Traumatic brain injury (TBI) is a major cause of death and disability. Cerebrolysin (CBL) has been reported to be anti-inflammatory by reducing reactive oxygen species (ROS) production. However, the neuroprotection of CBL in TBI and the potential mechanism are unclear. We aimed to investigate the neuroprotection and mechanisms of CBL in TBI.</p><p><strong>Methods: </strong>The TBI model was established in strict accordance with the Feeney weight-drop model of focal injury. The neurological score, brain water content, neuroinflammatory cytokine levels, and neuronal damage were evaluated. The involvement of the early brain injury modulatory pathway was also investigated.</p><p><strong>Results: </strong>Following TBI, the results showed that CBL administration increased neurological scores and decreased brain edema by alleviating blood‑brain barrier (BBB) permeability, upregulating tight junction protein (ZO‑1) levels, and decreasing the levels of the inflammatory cytokines tumor necrosis factor‑α (TNF‑α), interleukin‑1β (IL‑1β), IL‑6, and NF‑κB. The TUNEL assay showed that CBL decreased hippocampal neuronal apoptosis after TBI and decreased the protein expression levels of caspase‑3 and Bax, increasing the levels of Bcl‑2. The levels of Toll‑like receptor 2 (TLR2) and TLR4 were significantly decreased after CBL treatment. In TBI patients, CBL can also decrease TNF‑α, IL‑1β, IL‑6, and NF‑κB levels. This result indicates that CBL‑mediated inhibition of neuroinflammation and apoptosis ameliorated neuronal death after TBI. The neuroprotective capacity of CBL is partly dependent on the TLR signaling pathway.</p><p><strong>Conclusions: </strong>Taken together, the results of this study indicate that CBL can improve neurological outcomes and reduce neuronal death against neuroinflammation and apoptosis via the TLR signaling pathway in mice.</p>","PeriodicalId":6992,"journal":{"name":"Acta cirurgica brasileira","volume":null,"pages":null},"PeriodicalIF":1.1000,"publicationDate":"2022-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9448247/pdf/","citationCount":"4","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Acta cirurgica brasileira","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1590/acb370605","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2022/1/1 0:00:00","PubModel":"eCollection","JCR":"Q3","JCRName":"SURGERY","Score":null,"Total":0}
引用次数: 4

Abstract

Purpose: Traumatic brain injury (TBI) is a major cause of death and disability. Cerebrolysin (CBL) has been reported to be anti-inflammatory by reducing reactive oxygen species (ROS) production. However, the neuroprotection of CBL in TBI and the potential mechanism are unclear. We aimed to investigate the neuroprotection and mechanisms of CBL in TBI.

Methods: The TBI model was established in strict accordance with the Feeney weight-drop model of focal injury. The neurological score, brain water content, neuroinflammatory cytokine levels, and neuronal damage were evaluated. The involvement of the early brain injury modulatory pathway was also investigated.

Results: Following TBI, the results showed that CBL administration increased neurological scores and decreased brain edema by alleviating blood‑brain barrier (BBB) permeability, upregulating tight junction protein (ZO‑1) levels, and decreasing the levels of the inflammatory cytokines tumor necrosis factor‑α (TNF‑α), interleukin‑1β (IL‑1β), IL‑6, and NF‑κB. The TUNEL assay showed that CBL decreased hippocampal neuronal apoptosis after TBI and decreased the protein expression levels of caspase‑3 and Bax, increasing the levels of Bcl‑2. The levels of Toll‑like receptor 2 (TLR2) and TLR4 were significantly decreased after CBL treatment. In TBI patients, CBL can also decrease TNF‑α, IL‑1β, IL‑6, and NF‑κB levels. This result indicates that CBL‑mediated inhibition of neuroinflammation and apoptosis ameliorated neuronal death after TBI. The neuroprotective capacity of CBL is partly dependent on the TLR signaling pathway.

Conclusions: Taken together, the results of this study indicate that CBL can improve neurological outcomes and reduce neuronal death against neuroinflammation and apoptosis via the TLR signaling pathway in mice.

Abstract Image

Abstract Image

Abstract Image

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
脑溶素通过TLR信号通路抑制神经炎症和细胞凋亡,减轻创伤性脑损伤后早期脑损伤。
目的:外伤性脑损伤(TBI)是导致死亡和残疾的主要原因。据报道,脑溶血素(CBL)通过减少活性氧(ROS)的产生而具有抗炎作用。然而,CBL在TBI中的神经保护作用及其潜在机制尚不清楚。我们旨在探讨脑损伤后脑损伤的神经保护作用及其机制。方法:严格按照局灶性损伤Feeney失重模型建立TBI模型。评估神经学评分、脑含水量、神经炎性细胞因子水平和神经元损伤。研究了早期脑损伤调节通路的参与情况。结果:脑外伤后,结果显示CBL通过减轻血脑屏障(BBB)通透性、上调紧密连接蛋白(ZO - 1)水平、降低炎性细胞因子肿瘤坏死因子α (TNF - α)、白细胞介素- 1β (IL - 1β)、IL - 6和NF - κB水平,提高神经学评分,减少脑水肿。TUNEL实验显示,CBL可减少脑外伤后海马神经元凋亡,降低caspase - 3和Bax蛋白表达水平,升高Bcl - 2水平。CBL治疗后,Toll样受体2 (TLR2)和TLR4水平明显降低。在TBI患者中,CBL还能降低TNF - α、IL - 1β、IL - 6和NF - κB水平。这一结果表明,CBL介导的神经炎症和细胞凋亡抑制改善了脑外伤后的神经元死亡。CBL的神经保护能力部分依赖于TLR信号通路。结论:综上所述,本研究结果表明,CBL可通过TLR信号通路改善小鼠神经系统预后,减少神经炎症和细胞凋亡导致的神经元死亡。
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 去求助
来源期刊
CiteScore
1.90
自引率
9.10%
发文量
60
审稿时长
3-8 weeks
期刊介绍: Information not localized
期刊最新文献
Tridimensional models and radiographic study of dorsal laminectomy and thoracolumbar hemilaminectomy in dogs. Pancreatic cancer orthotopic graft in a murine model. ERRATA. Protective effect of Guanxin Danshen formula on myocardial ischemiareperfusion injury in rats. Astragaloside IV alleviates stroke-triggered early brain injury by modulating neuroinflammation and ferroptosis via the Nrf2/HO-1 signaling pathway.
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1