Inhibition of SARS-CoV-2 growth in the lungs of mice by a peptide-conjugated morpholino oligomer targeting viral RNA

IF 6.5 2区 医学 Q1 MEDICINE, RESEARCH & EXPERIMENTAL
Alexandra Sakai, Gagandeep Singh, Mahsa Khoshbakht, Scott Bittner, Christiane V. Löhr, Randy Diaz-Tapia, Prajakta Warang, Kris White, Luke Le Luo, Blanton Tolbert, Mario Blanco, Amy Chow, Mitchell Guttman, Cuiping Li, Yiming Bao, Joses Ho, Sebastian Maurer-Stroh, Arnab Chatterjee, Sumit Chanda, Adolfo García-Sastre, Michael Schotsaert, John R. Teijaro, Hong M. Moulton, David A. Stein
{"title":"Inhibition of SARS-CoV-2 growth in the lungs of mice by a peptide-conjugated morpholino oligomer targeting viral RNA","authors":"Alexandra Sakai, Gagandeep Singh, Mahsa Khoshbakht, Scott Bittner, Christiane V. Löhr, Randy Diaz-Tapia, Prajakta Warang, Kris White, Luke Le Luo, Blanton Tolbert, Mario Blanco, Amy Chow, Mitchell Guttman, Cuiping Li, Yiming Bao, Joses Ho, Sebastian Maurer-Stroh, Arnab Chatterjee, Sumit Chanda, Adolfo García-Sastre, Michael Schotsaert, John R. Teijaro, Hong M. Moulton, David A. Stein","doi":"10.1016/j.omtn.2024.102331","DOIUrl":null,"url":null,"abstract":"Further development of direct-acting antiviral agents against human SARS-CoV-2 infections remains a public health priority. Here, we report that an antisense peptide-conjugated morpholino oligomer (PPMO) called 5′END-2, targeting a highly conserved sequence in the 5′ UTR of SARS-CoV-2 genomic RNA, potently suppressed SARS-CoV-2 growth <ce:italic>in vitro</ce:italic> and <ce:italic>in vivo</ce:italic>. In HeLa-ACE 2 cells, 5′END-2 produced IC<ce:inf loc=\"post\">50</ce:inf> values of between 40 nM and 1.15 μM in challenges using six genetically disparate strains of SARS-CoV-2, including JN.1. <ce:italic>In vivo</ce:italic>, using K18-hACE2 mice and the WA-1/2020 virus isolate, two doses of 5′END-2 at 10 mg/kg, administered intranasally on the day before and the day after infection, produced approximately 1.4 log10 virus titer reduction in lung tissue at 3 days post-infection. Under a similar dosing schedule, intratracheal administration of 1.0–2.0 mg/kg 5′END-2 produced over 3.5 log10 virus growth suppression in mouse lungs. Electrophoretic mobility shift assays characterized specific binding of 5′END-2 to its complementary target RNA. Furthermore, using reporter constructs containing SARS-CoV-2 5′ UTR leader sequence, in an in-cell system, we observed that 5′END-2 could interfere with translation in a sequence-specific manner. The results demonstrate that direct pulmonary delivery of 5′END-2 PPMO is a promising antiviral strategy against SARS-CoV-2 infections and warrants further development.","PeriodicalId":18821,"journal":{"name":"Molecular Therapy. Nucleic Acids","volume":null,"pages":null},"PeriodicalIF":6.5000,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Molecular Therapy. Nucleic Acids","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1016/j.omtn.2024.102331","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

Abstract

Further development of direct-acting antiviral agents against human SARS-CoV-2 infections remains a public health priority. Here, we report that an antisense peptide-conjugated morpholino oligomer (PPMO) called 5′END-2, targeting a highly conserved sequence in the 5′ UTR of SARS-CoV-2 genomic RNA, potently suppressed SARS-CoV-2 growth in vitro and in vivo. In HeLa-ACE 2 cells, 5′END-2 produced IC50 values of between 40 nM and 1.15 μM in challenges using six genetically disparate strains of SARS-CoV-2, including JN.1. In vivo, using K18-hACE2 mice and the WA-1/2020 virus isolate, two doses of 5′END-2 at 10 mg/kg, administered intranasally on the day before and the day after infection, produced approximately 1.4 log10 virus titer reduction in lung tissue at 3 days post-infection. Under a similar dosing schedule, intratracheal administration of 1.0–2.0 mg/kg 5′END-2 produced over 3.5 log10 virus growth suppression in mouse lungs. Electrophoretic mobility shift assays characterized specific binding of 5′END-2 to its complementary target RNA. Furthermore, using reporter constructs containing SARS-CoV-2 5′ UTR leader sequence, in an in-cell system, we observed that 5′END-2 could interfere with translation in a sequence-specific manner. The results demonstrate that direct pulmonary delivery of 5′END-2 PPMO is a promising antiviral strategy against SARS-CoV-2 infections and warrants further development.
靶向病毒 RNA 的肽结合吗啉寡聚体抑制 SARS-CoV-2 在小鼠肺部的生长
进一步开发针对人类 SARS-CoV-2 感染的直接作用抗病毒药物仍然是公共卫生的当务之急。在这里,我们报告了一种名为 5′END-2 的反义肽结合吗啉寡聚体 (PPMO),它以 SARS-CoV-2 基因组 RNA 的 5′ UTR 中的一个高度保守序列为靶点,能有效抑制 SARS-CoV-2 在体外和体内的生长。在 HeLa-ACE 2 细胞中,5′END-2 在使用包括 JN.1 在内的六种不同基因的 SARS-CoV-2 株系的挑战中产生的 IC50 值介于 40 nM 和 1.15 μM 之间。在体内,使用 K18-hACE2 小鼠和 WA-1/2020 病毒分离株,在感染前一天和感染后一天分两次鼻内注射 10 mg/kg 剂量的 5′END-2,可在感染后 3 天使肺组织中的病毒滴度降低约 1.4 log10。在类似的剂量安排下,气管内注射 1.0-2.0 毫克/千克的 5′END-2 可抑制小鼠肺部超过 3.5 log10 的病毒生长。电泳迁移试验证明了 5′END-2 与其互补靶 RNA 的特异性结合。此外,在细胞内系统中,我们使用含有 SARS-CoV-2 5′ UTR 引导序列的报告构建物观察到,5′END-2 能以序列特异性的方式干扰翻译。结果表明,5′END-2 PPMO 直接肺部递送是一种很有前途的抗 SARS-CoV-2 感染的抗病毒策略,值得进一步开发。
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
Molecular Therapy. Nucleic Acids
Molecular Therapy. Nucleic Acids MEDICINE, RESEARCH & EXPERIMENTAL-
CiteScore
15.40
自引率
1.10%
发文量
336
审稿时长
20 weeks
期刊介绍: Molecular Therapy Nucleic Acids is an international, open-access journal that publishes high-quality research in nucleic-acid-based therapeutics to treat and correct genetic and acquired diseases. It is the official journal of the American Society of Gene & Cell Therapy and is built upon the success of Molecular Therapy. The journal focuses on gene- and oligonucleotide-based therapies and publishes peer-reviewed research, reviews, and commentaries. Its impact factor for 2022 is 8.8. The subject areas covered include the development of therapeutics based on nucleic acids and their derivatives, vector development for RNA-based therapeutics delivery, utilization of gene-modifying agents like Zn finger nucleases and triplex-forming oligonucleotides, pre-clinical target validation, safety and efficacy studies, and clinical trials.
×
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