Syndecan-4 Mediates the Cellular Entry of Adeno-Associated Virus 9

Author:

Hudák Anett1,Roach Matthew2,Pusztai Dávid1,Pettkó-Szandtner Aladár3,Letoha Annamária4,Szilák László5,Azzouz Mimoun2,Letoha Tamás1ORCID

Affiliation:

1. Pharmacoidea Ltd., H-6726 Szeged, Hungary

2. Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK

3. Biological Research Centre, Institute of Plant Biology, H-6726 Szeged, Hungary

4. Albert Szent-Györgyi Clinical Center, Department of Medicine, Faculty of Medicine, University of Szeged, H-6726 Szeged, Hungary

5. Szilák Laboratories Ltd., H-6725 Szeged, Hungary

Abstract

Due to their low pathogenicity, immunogenicity, and long-term gene expression, adeno-associated virus (AAV) vectors emerged as safe and efficient gene delivery tools, over-coming setbacks experienced with other viral gene delivery systems in early gene therapy trials. Among AAVs, AAV9 can translocate through the blood-brain barrier (BBB), making it a promising gene delivery tool for transducing the central nervous system (CNS) via systemic administration. Recent reports on the shortcomings of AAV9-mediated gene delivery into the CNS require reviewing the molecular base of AAV9 cellular biology. A more detailed understanding of AAV9’s cellular entry would eradicate current hurdles and enable more efficient AAV9-based gene therapy approaches. Syndecans, the transmembrane family of heparan-sulfate proteoglycans, facilitate the cellular uptake of various viruses and drug delivery systems. Utilizing human cell lines and syndecan-specific cellular assays, we assessed the involvement of syndecans in AAV9’s cellular entry. The ubiquitously expressed isoform, syndecan-4 proved its superiority in facilitating AAV9 internalization among syndecans. Introducing syndecan-4 into poorly transducible cell lines enabled robust AAV9-dependent gene transduction, while its knockdown reduced AAV9’s cellular entry. Attachment of AAV9 to syndecan-4 is mediated not just by the polyanionic heparan-sulfate chains but also by the cell-binding domain of the extracellular syndecan-4 core protein. Co-immunoprecipitation assays and affinity proteomics also confirmed the role of syndecan-4 in the cellular entry of AAV9. Overall, our findings highlight the universally expressed syndecan-4 as a significant contributor to the cellular internalization of AAV9 and provide a molecular-based, rational explanation for the low gene delivery potential of AAV9 into the CNS.

Funder

Innovative Medicines Initiative 2 Joint Undertaking

European Union’s Horizon 2020 Research and Innovation Program

National Research, Development and Innovation Office, Hungary

Publisher

MDPI AG

Subject

Inorganic Chemistry,Organic Chemistry,Physical and Theoretical Chemistry,Computer Science Applications,Spectroscopy,Molecular Biology,General Medicine,Catalysis

Reference81 articles.

1. A Molecular Revolution in the Treatment of Hemophilia;Butterfield;Mol. Ther. J. Am. Soc. Gene Ther.,2019

2. Gene therapy, an ongoing revolution;Benveniste;Blood,2012

3. Jiang, D.J., Xu, C.L., and Tsang, S.H. (2018). Revolution in Gene Medicine Therapy and Genome Surgery. Genes, 9.

4. Human gene therapy: A brief overview of the genetic revolution;Misra;J. Assoc. Physicians India,2013

5. The rapidly evolving state of gene therapy;Gruntman;FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol.,2018

Cited by 2 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