Salidroside protects against bleomycin-induced pulmonary fibrosis: activation of Nrf2-antioxidant signaling, and inhibition of NF-κB and TGF-β1/Smad-2/-3 pathways.

Cell Stress and Chaperones Pub Date : 2016-03-01 Epub Date: 2015-11-17 DOI:10.1007/s12192-015-0654-4
Haiying Tang, Lili Gao, Jingwei Mao, Huanyu He, Jia Liu, Xin Cai, Hongli Lin, Taihua Wu
{"title":"Salidroside protects against bleomycin-induced pulmonary fibrosis: activation of Nrf2-antioxidant signaling, and inhibition of NF-κB and TGF-β1/Smad-2/-3 pathways.","authors":"Haiying Tang, Lili Gao, Jingwei Mao, Huanyu He, Jia Liu, Xin Cai, Hongli Lin, Taihua Wu","doi":"10.1007/s12192-015-0654-4","DOIUrl":null,"url":null,"abstract":"<p><p>Pulmonary fibrosis (PF) can severely disrupt lung function, leading to fatal consequences. Salidroside is a principal active ingredient of Rhodiola rosea and has recently been reported to protect against lung injures. The present study was aimed at exploring its therapeutic effects on PF. Lung fibrotic injuries were induced in SD rats by a single intratracheal instillation of 5 mg/kg bleomycin (BLM). Then, these rats were administrated with 50, 100, or 200 mg/kg salidroside for 28 days. BLM-triggered structure distortion, collagen overproduction, excessive inflammatory infiltration, and pro-inflammatory cytokine release, and oxidative stress damages in lung tissues were attenuated by salidroside in a dose-dependent manner. Furthermore, salidroside was noted to inhibit IκBα phosphorylation and nuclear factor kappa B (NF-κB) p65 nuclear accumulation while activating Nrf2-antioxidant signaling in BLM-treated lungs. Downregulation of E-cadherin and upregulation of vimentin, fibronectin, and α-smooth muscle actin (α-SMA) indicated an epithelial-mesenchymal transition (EMT)-like shift in BLM-treated lungs. These changes were suppressed by salidroside. The expression of TGF-β1 and the phosphorylation of its downstream targets, Smad-2/-3, were enhanced by BLM, but weakened by salidroside. Additionally, salidroside was capable of reversing the recombinant TGF-β1-induced EMT-like changes in alveolar epithelial cells in vitro. Our study reveals that salidroside's protective effects against fibrotic lung injuries are correlated to its anti-inflammatory, antioxidative, and antifibrotic properties.</p>","PeriodicalId":9812,"journal":{"name":"Cell Stress and Chaperones","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2016-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4786523/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cell Stress and Chaperones","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1007/s12192-015-0654-4","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2015/11/17 0:00:00","PubModel":"Epub","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Pulmonary fibrosis (PF) can severely disrupt lung function, leading to fatal consequences. Salidroside is a principal active ingredient of Rhodiola rosea and has recently been reported to protect against lung injures. The present study was aimed at exploring its therapeutic effects on PF. Lung fibrotic injuries were induced in SD rats by a single intratracheal instillation of 5 mg/kg bleomycin (BLM). Then, these rats were administrated with 50, 100, or 200 mg/kg salidroside for 28 days. BLM-triggered structure distortion, collagen overproduction, excessive inflammatory infiltration, and pro-inflammatory cytokine release, and oxidative stress damages in lung tissues were attenuated by salidroside in a dose-dependent manner. Furthermore, salidroside was noted to inhibit IκBα phosphorylation and nuclear factor kappa B (NF-κB) p65 nuclear accumulation while activating Nrf2-antioxidant signaling in BLM-treated lungs. Downregulation of E-cadherin and upregulation of vimentin, fibronectin, and α-smooth muscle actin (α-SMA) indicated an epithelial-mesenchymal transition (EMT)-like shift in BLM-treated lungs. These changes were suppressed by salidroside. The expression of TGF-β1 and the phosphorylation of its downstream targets, Smad-2/-3, were enhanced by BLM, but weakened by salidroside. Additionally, salidroside was capable of reversing the recombinant TGF-β1-induced EMT-like changes in alveolar epithelial cells in vitro. Our study reveals that salidroside's protective effects against fibrotic lung injuries are correlated to its anti-inflammatory, antioxidative, and antifibrotic properties.

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
盐肤木苷可防止博莱霉素诱导的肺纤维化:激活Nrf2-抗氧化信号传导,抑制NF-κB和TGF-β1/Smad-2/-3通路。
肺纤维化(PF)会严重破坏肺功能,导致致命后果。水杨甙是红景天的一种主要活性成分,最近有报道称它能保护肺部免受伤害。本研究旨在探索其对肺纤维化的治疗效果。通过气管内灌注 5 毫克/千克博莱霉素(BLM)诱导 SD 大鼠肺纤维化损伤。然后,给这些大鼠服用 50、100 或 200 毫克/千克的柳氮苷,持续 28 天。沙利度苷以剂量依赖的方式减轻了博莱霉素引发的肺组织结构变形、胶原蛋白过度增生、过度炎症浸润、促炎细胞因子释放和氧化应激损伤。此外,研究还发现,在BLM处理的肺组织中,水杨甙可抑制IκBα磷酸化和核因子卡巴B(NF-κB)p65的核积累,同时激活Nrf2-抗氧化信号转导。E-粘连蛋白的下调以及波形蛋白、纤连蛋白和α-平滑肌肌动蛋白(α-SMA)的上调表明,在BLM处理的肺中出现了类似上皮-间质转化(EMT)的转变。柳氮磺吡啶抑制了这些变化。BLM增强了TGF-β1的表达及其下游靶标Smad-2/-3的磷酸化,而柳氮苷则削弱了这一作用。此外,水杨甙还能逆转重组 TGF-β1 诱导的肺泡上皮细胞体外 EMT 样变。我们的研究揭示了水杨甙对肺纤维化损伤的保护作用与其抗炎、抗氧化和抗纤维化特性有关。
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
自引率
0.00%
发文量
0
期刊最新文献
Identification and functional characterization of a novel TRPA1 gene from sea cucumber Apostichopus japonicus and interaction with miR-2013 in response to salt stress In memoriam: Ian R. Brown (1943–2020) Canine osteosarcoma cells exhibit basal accumulation of multiple chaperone proteins and are sensitive to small molecule inhibitors of GRP78 and heat shock protein function. Endoplasmic reticulum-unfolded protein response pathway modulates the cellular response to mitochondrial proteotoxic stress. Correction to: Molecular basis for efficacy of Guduchi and Madhuyashti feeding on different environmental stressors in Drosophila.
×
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