Engineering exosomes derived from TNF-α preconditioned IPFP-MSCs enhance both yield and therapeutic efficacy for osteoarthritis

IF 10.6 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Journal of Nanobiotechnology Pub Date : 2024-09-11 DOI:10.1186/s12951-024-02795-9
Jiangyi Wu, Jinhui Wu, Wei Xiang, Yunquan Gong, Daibo Feng, Shunzheng Fang, Yaran Wu, Zheng Liu, Yang Li, Ran Chen, Xiaoqi Zhang, Bingfei Li, Lifeng Chen, Runze Jin, Song Li, Bin Zhang, Tongyi Zhang, Lin Yin, Yizhao Zhou, Shu Huang, Ningning Liu, Hao Xu, Jiqin Lian, Yongqian Wang, Siru Zhou, Zhenhong Ni
{"title":"Engineering exosomes derived from TNF-α preconditioned IPFP-MSCs enhance both yield and therapeutic efficacy for osteoarthritis","authors":"Jiangyi Wu, Jinhui Wu, Wei Xiang, Yunquan Gong, Daibo Feng, Shunzheng Fang, Yaran Wu, Zheng Liu, Yang Li, Ran Chen, Xiaoqi Zhang, Bingfei Li, Lifeng Chen, Runze Jin, Song Li, Bin Zhang, Tongyi Zhang, Lin Yin, Yizhao Zhou, Shu Huang, Ningning Liu, Hao Xu, Jiqin Lian, Yongqian Wang, Siru Zhou, Zhenhong Ni","doi":"10.1186/s12951-024-02795-9","DOIUrl":null,"url":null,"abstract":"The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF−α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF−α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF−α, thereby exerting chondroprotective effects. TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA. ","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":null,"pages":null},"PeriodicalIF":10.6000,"publicationDate":"2024-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Nanobiotechnology","FirstCategoryId":"5","ListUrlMain":"https://doi.org/10.1186/s12951-024-02795-9","RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOTECHNOLOGY & APPLIED MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF−α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF−α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF−α, thereby exerting chondroprotective effects. TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA.
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
从 TNF-α 预处理 IPFP-MSCs 提取的工程外泌体可提高骨关节炎的产量和疗效
骨关节炎(OA)的发病机制包括关节软骨的逐渐退化。从间充质干细胞中提取的外泌体(MSC-EXOs)已被证明可通过减轻软骨破坏来减轻关节病理损伤。优化间充质干细胞外泌体的产量和疗效对促进其临床转化至关重要。间充质干细胞的预处理可增强工程外泌体的治疗潜力,通过可控和可量化的外部刺激,为其应用提供了广阔的前景。本研究旨在通过对间叶干细胞进行促炎预处理来提高外泌体的产生并增强其对 OA 的疗效,从而解决这些问题。研究人员从使用促炎因子 TNF-α 预处理的髌下脂肪垫(IPFP)-间充质干细胞的上清液中分离出外泌体,随后对其产生量进行了量化。在TNF-α预处理前后,通过高通量转录组测序分析、q-PCR和Western印迹分析评估了IPFP-间充质干细胞分泌外泌体的相关途径。此外,还将从TNF-α预处理的IPFP-间充质干细胞中提取的外泌体(IPFP-间充质干细胞-EXOsTNF-α)用于OA小鼠模型的关节内给药,并对关节病理学和步态改变进行了后续评估。通过蛋白质组分析确定了外泌体中参与维持软骨平衡的蛋白质的表达。与未经预处理的间充质干细胞相比,TNF-α的预处理能显著增强IPFP-间充质干细胞的外泌体分泌。其潜在机制是TNF-α预处理激活了IPFP-间充质干细胞的PI3K/AKT信号通路,导致自噬相关蛋白16 like 1(ATG16L1)水平上调,从而促进了外泌体的分泌。IPFP-间充质干细胞-EXOsTNF-α的关节内给药在改善OA小鼠关节病理变化方面表现出卓越的疗效。TNF-α的预处理增强了IPFP-间充质干细胞-EXOsTNF-α中低密度脂蛋白受体相关蛋白1(LRP1)水平的上调,从而发挥了软骨保护作用。TNF-α预处理是优化IPFP-间充质干细胞外泌体治疗OA疗效的一种有效且有前景的方法。
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
Journal of Nanobiotechnology
Journal of Nanobiotechnology BIOTECHNOLOGY & APPLIED MICROBIOLOGY-NANOSCIENCE & NANOTECHNOLOGY
CiteScore
13.90
自引率
4.90%
发文量
493
审稿时长
16 weeks
期刊介绍: Journal of Nanobiotechnology is an open access peer-reviewed journal communicating scientific and technological advances in the fields of medicine and biology, with an emphasis in their interface with nanoscale sciences. The journal provides biomedical scientists and the international biotechnology business community with the latest developments in the growing field of Nanobiotechnology.
期刊最新文献
Curcumin nanoparticles in heat stroke management ACYP2 functions as an innovative nano-therapeutic target to impede the progression of hepatocellular carcinoma by inhibiting the activity of TERT and the KCNN4/ERK pathway A novel spherical GelMA-HAMA hydrogel encapsulating APET×2 polypeptide and CFIm25-targeting sgRNA for immune microenvironment modulation and nucleus pulposus regeneration in intervertebral discs Preparation and effects of functionalized liposomes targeting breast cancer tumors using chemotherapy, phototherapy, and immunotherapy Ellagic acid-enhanced biocompatibility and bioactivity in multilayer core-shell gold nanoparticles for ameliorating myocardial infarction injury
×
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