RTP801 mediates transneuronal toxicity in culture via extracellular vesicles

Author:

Solana‐Balaguer Júlia123ORCID,Martín‐Flores Núria12ORCID,Garcia‐Segura Pol123,Campoy‐Campos Genís123,Pérez‐Sisqués Leticia12,Chicote‐González Almudena123,Fernández‐Irigoyen Joaquín4,Santamaría Enrique4,Pérez‐Navarro Esther1235,Alberch Jordi1235,Malagelada Cristina123ORCID

Affiliation:

1. Department of Biomedical Sciences Universitat de Barcelona Barcelona Spain

2. Institut de Neurociències (UBneuro) Universitat de Barcelona Barcelona Spain

3. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Barcelona Spain

4. Proteored‐ISCIII Proteomics Unit Navarrabiomed, Departamento de Salud UPNA IdiSNA Pamplona Spain

5. Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain

Abstract

AbstractExtracellular vesicles (EVs) play a crucial role in intercellular communication, participating in the paracrine trophic support or in the propagation of toxic molecules, including proteins. RTP801 is a stress‐regulated protein, whose levels are elevated during neurodegeneration and induce neuron death. However, whether RTP801 toxicity is transferred trans‐neuronally via EVs remains unknown. Hence, we overexpressed or silenced RTP801 protein in cultured cortical neurons, isolated their derived EVs (RTP801‐EVs or shRTP801‐EVs, respectively), and characterized EVs protein content by mass spectrometry (MS). RTP801‐EVs toxicity was assessed by treating cultured neurons with these EVs and quantifying apoptotic neuron death and branching. We also tested shRTP801‐EVs functionality in the pathologic in vitro model of 6‐Hydroxydopamine (6‐OHDA). Expression of RTP801 increased the number of EVs released by neurons. Moreover, RTP801 led to a distinct proteomic signature of neuron‐derived EVs, containing more pro‐apoptotic markers. Hence, we observed that RTP801‐induced toxicity was transferred to neurons via EVs, activating apoptosis and impairing neuron morphology complexity. In contrast, shRTP801‐EVs were able to increase the arborization in recipient neurons. The 6‐OHDA neurotoxin elevated levels of RTP801 in EVs, and 6‐OHDA‐derived EVs lost the mTOR/Akt signalling activation via Akt and RPS6 downstream effectors. Interestingly, EVs derived from neurons where RTP801 was silenced prior to exposing them to 6‐OHDA maintained Akt and RPS6 transactivation in recipient neurons. Taken together, these results suggest that RTP801‐induced toxicity is transferred via EVs, and therefore, it could contribute to the progression of neurodegenerative diseases, in which RTP801 is involved.

Funder

Michael J. Fox Foundation for Parkinson's Research

Ministerio de Ciencia e Innovación

Publisher

Wiley

Subject

Cell Biology,Histology

Cited by 1 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3