Targeting Rap1b signaling cascades with CDNF: Modulating Platelet Activation, Regulating Plasma Oxylipins and Mitigating Reperfusion Injury in stroke.

IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Molecular Therapy Pub Date : 2024-09-09 DOI:10.1016/j.ymthe.2024.09.005
Jui-Sheng Wu,Helike Lõhelaid,Chih-Chin Shih,Hock-Kean Liew,Vicki Wang,Wei-Fen Hu,Yuan-Hao Chen,Mart Saarma,Mikko Airavaara,Kuan-Yin Tseng
{"title":"Targeting Rap1b signaling cascades with CDNF: Modulating Platelet Activation, Regulating Plasma Oxylipins and Mitigating Reperfusion Injury in stroke.","authors":"Jui-Sheng Wu,Helike Lõhelaid,Chih-Chin Shih,Hock-Kean Liew,Vicki Wang,Wei-Fen Hu,Yuan-Hao Chen,Mart Saarma,Mikko Airavaara,Kuan-Yin Tseng","doi":"10.1016/j.ymthe.2024.09.005","DOIUrl":null,"url":null,"abstract":"Cerebral reperfusion injury in stroke, stemming from interconnected thrombotic and inflammatory signatures, often involves platelet activation, aggregation and its interaction with various immune cells, contributing to microvascular dysfunction. However, the regulatory mechanisms behind this platelet activation and the resulting inflammation are not well understood, complicating the development of effective stroke therapies. Utilizing animal models and platelets from hemorrhagic stroke patients, our research demonstrates that human cerebral dopamine neurotrophic factor (CDNF) acts as an endogenous antagonist, mitigating platelet aggregation and associated neuroinflammation. CDNF moderates mitochondrial membrane potential, reactive oxygen species production, and intracellular calcium in activated platelets by interfering with GTP binding to Rap1b, thereby reducing Rap1b activation and downregulating the Rap1b-MAPK-PLA2 signaling pathway, which decreases release of the pro-inflammatory mediator thromboxane A2. Additionally, CDNF reduces the inflammatory response in BV2 microglial cells co-cultured with activated platelets. Consistent with ex vivo findings, subcutaneous administration of CDNF in a rat model of ischemic stroke significantly reduces platelet activation, aggregation, lipid mediator production, infarct volume, and neurological deficits. In summary, our study highlights CDNF as a promising therapeutic target for mitigating platelet-induced inflammation and enhancing recovery in stroke. Harnessing the CDNF pathway may offer a novel therapeutic strategy for stroke intervention.","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1000,"publicationDate":"2024-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Molecular Therapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1016/j.ymthe.2024.09.005","RegionNum":1,"RegionCategory":"医学","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

Cerebral reperfusion injury in stroke, stemming from interconnected thrombotic and inflammatory signatures, often involves platelet activation, aggregation and its interaction with various immune cells, contributing to microvascular dysfunction. However, the regulatory mechanisms behind this platelet activation and the resulting inflammation are not well understood, complicating the development of effective stroke therapies. Utilizing animal models and platelets from hemorrhagic stroke patients, our research demonstrates that human cerebral dopamine neurotrophic factor (CDNF) acts as an endogenous antagonist, mitigating platelet aggregation and associated neuroinflammation. CDNF moderates mitochondrial membrane potential, reactive oxygen species production, and intracellular calcium in activated platelets by interfering with GTP binding to Rap1b, thereby reducing Rap1b activation and downregulating the Rap1b-MAPK-PLA2 signaling pathway, which decreases release of the pro-inflammatory mediator thromboxane A2. Additionally, CDNF reduces the inflammatory response in BV2 microglial cells co-cultured with activated platelets. Consistent with ex vivo findings, subcutaneous administration of CDNF in a rat model of ischemic stroke significantly reduces platelet activation, aggregation, lipid mediator production, infarct volume, and neurological deficits. In summary, our study highlights CDNF as a promising therapeutic target for mitigating platelet-induced inflammation and enhancing recovery in stroke. Harnessing the CDNF pathway may offer a novel therapeutic strategy for stroke intervention.
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
用 CDNF 靶向 Rap1b 信号级联:调节血小板活化、调节血浆氧脂和减轻中风的再灌注损伤。
中风的脑再灌注损伤源于相互关联的血栓和炎症特征,通常涉及血小板的活化、聚集及其与各种免疫细胞的相互作用,从而导致微血管功能障碍。然而,人们对血小板活化及其导致的炎症背后的调节机制还不甚了解,这使得开发有效的中风疗法变得更加复杂。我们的研究利用动物模型和出血性中风患者的血小板,证明人脑多巴胺神经营养因子(CDNF)可作为内源性拮抗剂,缓解血小板聚集和相关的神经炎症。CDNF 通过干扰 GTP 与 Rap1b 的结合,从而降低 Rap1b 的活化并下调 Rap1b-MAPK-PLA2 信号通路,减少促炎介质血栓素 A2 的释放,从而调节活化血小板的线粒体膜电位、活性氧生成和细胞内钙。此外,CDNF 还能降低与活化血小板共培养的 BV2 小胶质细胞的炎症反应。与体内外研究结果一致,在缺血性中风大鼠模型中皮下注射 CDNF 能显著降低血小板活化、聚集、脂质介质产生、梗死体积和神经功能缺损。总之,我们的研究强调了 CDNF 是减轻血小板诱导的炎症和促进中风恢复的一个有前景的治疗靶点。利用 CDNF 通路可为中风干预提供一种新的治疗策略。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Molecular Therapy
Molecular Therapy 医学-生物工程与应用微生物
CiteScore
19.20
自引率
3.20%
发文量
357
审稿时长
3 months
期刊介绍: Molecular Therapy is the leading journal for research in gene transfer, vector development, stem cell manipulation, and therapeutic interventions. It covers a broad spectrum of topics including genetic and acquired disease correction, vaccine development, pre-clinical validation, safety/efficacy studies, and clinical trials. With a focus on advancing genetics, medicine, and biotechnology, Molecular Therapy publishes peer-reviewed research, reviews, and commentaries to showcase the latest advancements in the field. With an impressive impact factor of 12.4 in 2022, it continues to attract top-tier contributions.
期刊最新文献
Engineering a solution for allogeneic CAR-T rejection. Targeting Rap1b signaling cascades with CDNF: Modulating Platelet Activation, Regulating Plasma Oxylipins and Mitigating Reperfusion Injury in stroke. A CD25×TIGIT bispecific antibody induces anti-tumor activity through selective intratumoral Treg cell depletion. A chimeric anti-inflammatory and anti-vascularization immunomodulator prevents high-risk corneal transplantation rejection via ex vivo gene therapy. Case study of CD19-directed chimeric antigen receptor T-cell therapy in a subject with immune-mediate necrotizing myopathy treated in the RESET-Myositis™ phase I/II trial.
×
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