TfR1 mediated iron metabolism dysfunction as a potential therapeutic target for osteoarthritis

IF 4.9 2区 医学 Q1 Medicine
Wenchao Wang, Zhenkai Ma, Xuemin Feng, Jiabin Ren, Shengyao Sun, Yuandong Shao, Weimin Zhang, Xiaoxia Yang, Jiaming Zhang, Xingzhi Jing
{"title":"TfR1 mediated iron metabolism dysfunction as a potential therapeutic target for osteoarthritis","authors":"Wenchao Wang, Zhenkai Ma, Xuemin Feng, Jiabin Ren, Shengyao Sun, Yuandong Shao, Weimin Zhang, Xiaoxia Yang, Jiaming Zhang, Xingzhi Jing","doi":"10.1186/s13075-024-03304-x","DOIUrl":null,"url":null,"abstract":"Transferrin receptor-1 (TfR1) plays important roles in controlling cellular iron levels, but its role in OA pathology is unknown. Herein we aim to investigate the role of TfR1 in OA progression and its underlying mechanisms. TfR1 expression in cartilage during OA development were examined both in vivo and in vitro. Then IL-1β was used to induce chondrocytes degeneration in vitro and TfR1 siRNA was used for observing the effect of TfR1 in modulating iron homeostasis, mitochondrial function and degrading enzymes expression. Also the inhibitor of TfR1 was exploited to analyze the protective effect of TfR1 inhibition in vivo. TfR1 is elevated in OA cartilage and contributes to OA inflammation condition. Excess iron not only results in oxidative stress damage and sensitizes chondrocytes to ferroptosis, but also triggers c-GAS/STING-mediated inflammation by promoting mitochondrial destruction and the release of mtDNA. Silencing TfR1 using TfR1 siRNA not only reduced iron content in chondrocytes and inhibited oxidative stress, but also facilitated the mitophagy process and suppressed mtDNA/cGAS/STING-mediated inflammation. Importantly, we also found that Ferstatin II, a novel and selective TfR1 inhibitor, could substantially suppress TfR1 activity both in vivo and in vitro and ameliorated cartilage degeneration. Our work demonstrates that TfR1 mediated iron influx plays important roles in chondrocytes degeneration and OA pathogenesis, suggesting that maintaining iron homeostasis through the targeting of TfR1 may represent a novel therapeutic strategy for the treatment of OA.","PeriodicalId":8419,"journal":{"name":"Arthritis Research & Therapy","volume":null,"pages":null},"PeriodicalIF":4.9000,"publicationDate":"2024-03-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Arthritis Research & Therapy","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1186/s13075-024-03304-x","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"Medicine","Score":null,"Total":0}
引用次数: 0

Abstract

Transferrin receptor-1 (TfR1) plays important roles in controlling cellular iron levels, but its role in OA pathology is unknown. Herein we aim to investigate the role of TfR1 in OA progression and its underlying mechanisms. TfR1 expression in cartilage during OA development were examined both in vivo and in vitro. Then IL-1β was used to induce chondrocytes degeneration in vitro and TfR1 siRNA was used for observing the effect of TfR1 in modulating iron homeostasis, mitochondrial function and degrading enzymes expression. Also the inhibitor of TfR1 was exploited to analyze the protective effect of TfR1 inhibition in vivo. TfR1 is elevated in OA cartilage and contributes to OA inflammation condition. Excess iron not only results in oxidative stress damage and sensitizes chondrocytes to ferroptosis, but also triggers c-GAS/STING-mediated inflammation by promoting mitochondrial destruction and the release of mtDNA. Silencing TfR1 using TfR1 siRNA not only reduced iron content in chondrocytes and inhibited oxidative stress, but also facilitated the mitophagy process and suppressed mtDNA/cGAS/STING-mediated inflammation. Importantly, we also found that Ferstatin II, a novel and selective TfR1 inhibitor, could substantially suppress TfR1 activity both in vivo and in vitro and ameliorated cartilage degeneration. Our work demonstrates that TfR1 mediated iron influx plays important roles in chondrocytes degeneration and OA pathogenesis, suggesting that maintaining iron homeostasis through the targeting of TfR1 may represent a novel therapeutic strategy for the treatment of OA.
TfR1 介导的铁代谢功能障碍是骨关节炎的潜在治疗靶点
转铁蛋白受体-1(TfR1)在控制细胞铁水平方面发挥着重要作用,但其在 OA 病理学中的作用尚不清楚。在此,我们旨在研究 TfR1 在 OA 进展中的作用及其内在机制。我们在体内和体外研究了 OA 发生过程中软骨中 TfR1 的表达。然后用 IL-1β 在体外诱导软骨细胞变性,并用 TfR1 siRNA 观察 TfR1 在调节铁稳态、线粒体功能和降解酶表达方面的作用。此外,还利用 TfR1 抑制剂分析了抑制 TfR1 在体内的保护作用。TfR1在OA软骨中升高并导致OA炎症。过量的铁不仅会导致氧化应激损伤,使软骨细胞对铁变态反应敏感,还会通过促进线粒体破坏和mtDNA释放,引发c-GAS/STING介导的炎症。使用 TfR1 siRNA 沉默 TfR1 不仅能降低软骨细胞中的铁含量并抑制氧化应激,还能促进有丝分裂过程并抑制 mtDNA/cGAS/STING 介导的炎症。重要的是,我们还发现一种新型选择性 TfR1 抑制剂 Ferstatin II 可在体内和体外大幅抑制 TfR1 的活性,并改善软骨退化。我们的研究表明,TfR1介导的铁流入在软骨细胞变性和OA发病机制中起着重要作用,这表明通过靶向TfR1维持铁平衡可能是治疗OA的一种新型治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
8.60
自引率
2.00%
发文量
261
审稿时长
14 weeks
期刊介绍: Established in 1999, Arthritis Research and Therapy is an international, open access, peer-reviewed journal, publishing original articles in the area of musculoskeletal research and therapy as well as, reviews, commentaries and reports. A major focus of the journal is on the immunologic processes leading to inflammation, damage and repair as they relate to autoimmune rheumatic and musculoskeletal conditions, and which inform the translation of this knowledge into advances in clinical care. Original basic, translational and clinical research is considered for publication along with results of early and late phase therapeutic trials, especially as they pertain to the underpinning science that informs clinical observations in interventional studies.
文献相关原料
公司名称 产品信息 采购帮参考价格
×
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