吡非尼酮和穿心莲内酯联用可通过调节TGF-β/Smad信号通路改善肝星状细胞活化和肝纤维化

IF 2.6 4区 医学 Q3 CELL BIOLOGY Analytical Cellular Pathology Pub Date : 2024-06-21 eCollection Date: 2024-01-01 DOI:10.1155/2024/2751280
Guang Xu, Tidong Ma, Chonggao Zhou, Fan Zhao, Kun Peng, Bixiang Li
{"title":"吡非尼酮和穿心莲内酯联用可通过调节TGF-β/Smad信号通路改善肝星状细胞活化和肝纤维化","authors":"Guang Xu, Tidong Ma, Chonggao Zhou, Fan Zhao, Kun Peng, Bixiang Li","doi":"10.1155/2024/2751280","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Biliary atresia (BA) is a devastating congenital disease characterized by inflammation and progressive liver fibrosis. Activation of hepatic stellate cells (HSCs) plays a central role in the pathogenesis of hepatic fibrosis. Our study aimed to investigate the pharmacological effect and potential mechanism of pirfenidone (PFD) and andrographolide (AGP) separately and together on liver fibrosis of BA.</p><p><strong>Materials and methods: </strong>The bile ducts of male C57BL/6J mice were ligated or had the sham operation. The <i>in vivo</i> effects of PFD and/or AGP on liver fibrosis of BA were evaluated. Human hepatic stellate cells (LX-2) were also treated with PFD and/or AGP <i>in vitro</i>.</p><p><strong>Results: </strong>PFD and/or AGP ameliorates liver fibrosis and inflammation in the mice model of BA, as evidenced by significant downregulated in the accumulation of collagen fibers, hepatic fibrosis markers (<i>α</i>-SMA, collagen I, and collagen IV), and inflammatory markers (IL-1<i>β</i>, IL-6, and TNF-<i>α</i>). Moreover, compared with monotherapy, these changes are more obvious in the combined treatment of PFD and AGP. Consistent with animal experiments, hepatic fibrosis markers (<i>α</i>-SMA, collagen I, and CTGF) and inflammatory markers (IL-1<i>β</i>, IL-6, and TNF-<i>α</i>) were significantly decreased in activated LX-2 cells after PFD and/or AGP treatment. In addition, PFD and/or AGP inhibited the activation of HSCs by blocking the TGF-<i>β</i>/Smad signaling pathway, and the combined treatment of PFD and AGP synergistically inhibited the phosphorylation of Smad2 and Smad3.</p><p><strong>Conclusion: </strong>The combined application of PFD and AGP exerted superior inhibitive effects on HSC activation and liver fibrosis by mediating the TGF-<i>β</i>/Smad signaling pathway as compared to monotherapy. Therefore, the combination of PFD and AGP may be a promising treatment strategy for liver fibrosis in BA.</p>","PeriodicalId":49326,"journal":{"name":"Analytical Cellular Pathology","volume":null,"pages":null},"PeriodicalIF":2.6000,"publicationDate":"2024-06-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11213636/pdf/","citationCount":"0","resultStr":"{\"title\":\"Combination of Pirfenidone and Andrographolide Ameliorates Hepatic Stellate Cell Activation and Liver Fibrosis by Mediating TGF-<i>β</i>/Smad Signaling Pathway.\",\"authors\":\"Guang Xu, Tidong Ma, Chonggao Zhou, Fan Zhao, Kun Peng, Bixiang Li\",\"doi\":\"10.1155/2024/2751280\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Biliary atresia (BA) is a devastating congenital disease characterized by inflammation and progressive liver fibrosis. Activation of hepatic stellate cells (HSCs) plays a central role in the pathogenesis of hepatic fibrosis. Our study aimed to investigate the pharmacological effect and potential mechanism of pirfenidone (PFD) and andrographolide (AGP) separately and together on liver fibrosis of BA.</p><p><strong>Materials and methods: </strong>The bile ducts of male C57BL/6J mice were ligated or had the sham operation. The <i>in vivo</i> effects of PFD and/or AGP on liver fibrosis of BA were evaluated. Human hepatic stellate cells (LX-2) were also treated with PFD and/or AGP <i>in vitro</i>.</p><p><strong>Results: </strong>PFD and/or AGP ameliorates liver fibrosis and inflammation in the mice model of BA, as evidenced by significant downregulated in the accumulation of collagen fibers, hepatic fibrosis markers (<i>α</i>-SMA, collagen I, and collagen IV), and inflammatory markers (IL-1<i>β</i>, IL-6, and TNF-<i>α</i>). Moreover, compared with monotherapy, these changes are more obvious in the combined treatment of PFD and AGP. Consistent with animal experiments, hepatic fibrosis markers (<i>α</i>-SMA, collagen I, and CTGF) and inflammatory markers (IL-1<i>β</i>, IL-6, and TNF-<i>α</i>) were significantly decreased in activated LX-2 cells after PFD and/or AGP treatment. In addition, PFD and/or AGP inhibited the activation of HSCs by blocking the TGF-<i>β</i>/Smad signaling pathway, and the combined treatment of PFD and AGP synergistically inhibited the phosphorylation of Smad2 and Smad3.</p><p><strong>Conclusion: </strong>The combined application of PFD and AGP exerted superior inhibitive effects on HSC activation and liver fibrosis by mediating the TGF-<i>β</i>/Smad signaling pathway as compared to monotherapy. Therefore, the combination of PFD and AGP may be a promising treatment strategy for liver fibrosis in BA.</p>\",\"PeriodicalId\":49326,\"journal\":{\"name\":\"Analytical Cellular Pathology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.6000,\"publicationDate\":\"2024-06-21\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11213636/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Analytical Cellular Pathology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1155/2024/2751280\",\"RegionNum\":4,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"Q3\",\"JCRName\":\"CELL BIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Analytical Cellular Pathology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1155/2024/2751280","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q3","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

背景:胆道闭锁(BA)是一种以炎症和进行性肝纤维化为特征的破坏性先天性疾病。肝星状细胞(HSCs)的活化在肝纤维化的发病机制中起着核心作用。我们的研究旨在探讨吡非尼酮(PFD)和穿心莲内酯(AGP)分别和共同对 BA 肝纤维化的药理作用和潜在机制:雄性C57BL/6J小鼠胆管结扎或假手术。材料和方法:将雄性 C57BL/6J 小鼠的胆管结扎或进行假手术,评估 PFD 和/或 AGP 对 BA 肝纤维化的体内影响。在体外也用 PFD 和/或 AGP 处理人肝星状细胞(LX-2):结果:PFD和/或AGP可改善BA小鼠模型的肝纤维化和炎症反应,表现为胶原纤维、肝纤维化标志物(α-SMA、胶原蛋白I和胶原蛋白IV)和炎症标志物(IL-1β、IL-6和TNF-α)的积累显著下调。此外,与单一疗法相比,这些变化在 PFD 和 AGP 联合疗法中更为明显。与动物实验一致,PFD 和/或 AGP 治疗后,活化的 LX-2 细胞中的肝纤维化标志物(α-SMA、胶原 I 和 CTGF)和炎症标志物(IL-1β、IL-6 和 TNF-α)明显减少。此外,PFD和/或AGP通过阻断TGF-β/Smad信号通路抑制造血干细胞的活化,PFD和AGP联合处理可协同抑制Smad2和Smad3的磷酸化:结论:与单药治疗相比,PFD和AGP联合应用通过介导TGF-β/Smad信号通路对造血干细胞活化和肝纤维化具有更优越的抑制作用。因此,PFD和AGP的联合应用可能是治疗BA肝纤维化的一种有前景的治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
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
Combination of Pirfenidone and Andrographolide Ameliorates Hepatic Stellate Cell Activation and Liver Fibrosis by Mediating TGF-β/Smad Signaling Pathway.

Background: Biliary atresia (BA) is a devastating congenital disease characterized by inflammation and progressive liver fibrosis. Activation of hepatic stellate cells (HSCs) plays a central role in the pathogenesis of hepatic fibrosis. Our study aimed to investigate the pharmacological effect and potential mechanism of pirfenidone (PFD) and andrographolide (AGP) separately and together on liver fibrosis of BA.

Materials and methods: The bile ducts of male C57BL/6J mice were ligated or had the sham operation. The in vivo effects of PFD and/or AGP on liver fibrosis of BA were evaluated. Human hepatic stellate cells (LX-2) were also treated with PFD and/or AGP in vitro.

Results: PFD and/or AGP ameliorates liver fibrosis and inflammation in the mice model of BA, as evidenced by significant downregulated in the accumulation of collagen fibers, hepatic fibrosis markers (α-SMA, collagen I, and collagen IV), and inflammatory markers (IL-1β, IL-6, and TNF-α). Moreover, compared with monotherapy, these changes are more obvious in the combined treatment of PFD and AGP. Consistent with animal experiments, hepatic fibrosis markers (α-SMA, collagen I, and CTGF) and inflammatory markers (IL-1β, IL-6, and TNF-α) were significantly decreased in activated LX-2 cells after PFD and/or AGP treatment. In addition, PFD and/or AGP inhibited the activation of HSCs by blocking the TGF-β/Smad signaling pathway, and the combined treatment of PFD and AGP synergistically inhibited the phosphorylation of Smad2 and Smad3.

Conclusion: The combined application of PFD and AGP exerted superior inhibitive effects on HSC activation and liver fibrosis by mediating the TGF-β/Smad signaling pathway as compared to monotherapy. Therefore, the combination of PFD and AGP may be a promising treatment strategy for liver fibrosis in BA.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Analytical Cellular Pathology
Analytical Cellular Pathology ONCOLOGY-CELL BIOLOGY
CiteScore
4.90
自引率
3.10%
发文量
70
审稿时长
16 weeks
期刊介绍: Analytical Cellular Pathology is a peer-reviewed, Open Access journal that provides a forum for scientists, medical practitioners and pathologists working in the area of cellular pathology. The journal publishes original research articles, review articles, and clinical studies related to cytology, carcinogenesis, cell receptors, biomarkers, diagnostic pathology, immunopathology, and hematology.
期刊最新文献
LncRNA H19 Promotes Gastric Cancer Metastasis via miR-148-3p/SOX-12 Axis. Shock Wave Therapy Alleviates Hypoxia/Reoxygenation-Induced Cardiomyocyte Injury by Inhibiting Both Apoptosis and Ferroptosis. Role of miR-93-5p and Its Opposing Effect of Ionizing Radiation in Non-Small Cell Lung Cancer. Immunohistochemical Profile of p62/SQSTM1/Sequestosome-1 in Human Low- and High-Grade Intracranial Meningiomas. Construction and Identification of Eukaryotic Expression Vector pEGFP-N1-MIC-1 for Mouse MIC-1 Gene and Its Effect on Gastric Cancer Cells.
×
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