Cargando…

Characterization of Novel Aptamers Specifically Directed to Red-Spotted Grouper Nervous Necrosis Virus (RGNNV)-Infected Cells for Mediating Targeted siRNA Delivery

Nervous necrosis virus (NNV) causes viral nervous necrosis, the most devastating disease in more than 50 fish species worldwide, with massive mortality rates up to 100%, resulting in great economic losses to mariculture. However, few methods are available for the efficient diagnosis and treatment of...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhou, Lingli, Wang, Shaowen, Yu, Qing, Wei, Shina, Liu, Mingzhu, Wei, Jingguang, Huang, Youhua, Huang, Xiaohong, Li, Pengfei, Qin, Qiwei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7203557/
https://www.ncbi.nlm.nih.gov/pubmed/32425897
http://dx.doi.org/10.3389/fmicb.2020.00660
_version_ 1783529893851561984
author Zhou, Lingli
Wang, Shaowen
Yu, Qing
Wei, Shina
Liu, Mingzhu
Wei, Jingguang
Huang, Youhua
Huang, Xiaohong
Li, Pengfei
Qin, Qiwei
author_facet Zhou, Lingli
Wang, Shaowen
Yu, Qing
Wei, Shina
Liu, Mingzhu
Wei, Jingguang
Huang, Youhua
Huang, Xiaohong
Li, Pengfei
Qin, Qiwei
author_sort Zhou, Lingli
collection PubMed
description Nervous necrosis virus (NNV) causes viral nervous necrosis, the most devastating disease in more than 50 fish species worldwide, with massive mortality rates up to 100%, resulting in great economic losses to mariculture. However, few methods are available for the efficient diagnosis and treatment of viral nervous necrosis. Aptamers are molecular recognition ligands characterized by their remarkably high specificity and affinity, great stability, and ease of synthesis, and have been widely studied in application of disease diagnosis and therapies. In this study, we generated three aptamers against red-spotted grouper nervous necrosis virus (RGNNV)-infected grouper brain (GB) cells using the Cell-SELEX (cell based-systematic evolution of ligands by exponential enrichment) technology. The selected aptamers formed stable stem-loop structures, and could specifically recognize RGNNV-infected GB cells, with calculated dissociation constants (K(d)) of 27.96, 29.3, and 59.5 nM for aptamers GBN2, GBN10, and GBN34, respectively. They also recognized RGNNV-infected brain tissues. The three aptamers were non-toxic and showed antiviral activities both in vitro and in vivo. Fluorescence microscopy and flow cytometry also demonstrated that aptamer GBN34 could be efficiently and specifically internalized into RGNNV-infected GB cells. The targeted cellular delivery of aptamer-small interfering RNA (siRNA) conjugates remarkably inhibited RGNNV infection in GB cells. The efficiency of the aptamer-based targeted delivery system was about 75% reduction in infection after 48 h, which was similar to that of transfection. These aptamers have great potential utility in the rapid diagnosis and inhibition of RGNNV infection in mariculture.
format Online
Article
Text
id pubmed-7203557
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-72035572020-05-18 Characterization of Novel Aptamers Specifically Directed to Red-Spotted Grouper Nervous Necrosis Virus (RGNNV)-Infected Cells for Mediating Targeted siRNA Delivery Zhou, Lingli Wang, Shaowen Yu, Qing Wei, Shina Liu, Mingzhu Wei, Jingguang Huang, Youhua Huang, Xiaohong Li, Pengfei Qin, Qiwei Front Microbiol Microbiology Nervous necrosis virus (NNV) causes viral nervous necrosis, the most devastating disease in more than 50 fish species worldwide, with massive mortality rates up to 100%, resulting in great economic losses to mariculture. However, few methods are available for the efficient diagnosis and treatment of viral nervous necrosis. Aptamers are molecular recognition ligands characterized by their remarkably high specificity and affinity, great stability, and ease of synthesis, and have been widely studied in application of disease diagnosis and therapies. In this study, we generated three aptamers against red-spotted grouper nervous necrosis virus (RGNNV)-infected grouper brain (GB) cells using the Cell-SELEX (cell based-systematic evolution of ligands by exponential enrichment) technology. The selected aptamers formed stable stem-loop structures, and could specifically recognize RGNNV-infected GB cells, with calculated dissociation constants (K(d)) of 27.96, 29.3, and 59.5 nM for aptamers GBN2, GBN10, and GBN34, respectively. They also recognized RGNNV-infected brain tissues. The three aptamers were non-toxic and showed antiviral activities both in vitro and in vivo. Fluorescence microscopy and flow cytometry also demonstrated that aptamer GBN34 could be efficiently and specifically internalized into RGNNV-infected GB cells. The targeted cellular delivery of aptamer-small interfering RNA (siRNA) conjugates remarkably inhibited RGNNV infection in GB cells. The efficiency of the aptamer-based targeted delivery system was about 75% reduction in infection after 48 h, which was similar to that of transfection. These aptamers have great potential utility in the rapid diagnosis and inhibition of RGNNV infection in mariculture. Frontiers Media S.A. 2020-04-30 /pmc/articles/PMC7203557/ /pubmed/32425897 http://dx.doi.org/10.3389/fmicb.2020.00660 Text en Copyright © 2020 Zhou, Wang, Yu, Wei, Liu, Wei, Huang, Huang, Li and Qin. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Microbiology
Zhou, Lingli
Wang, Shaowen
Yu, Qing
Wei, Shina
Liu, Mingzhu
Wei, Jingguang
Huang, Youhua
Huang, Xiaohong
Li, Pengfei
Qin, Qiwei
Characterization of Novel Aptamers Specifically Directed to Red-Spotted Grouper Nervous Necrosis Virus (RGNNV)-Infected Cells for Mediating Targeted siRNA Delivery
title Characterization of Novel Aptamers Specifically Directed to Red-Spotted Grouper Nervous Necrosis Virus (RGNNV)-Infected Cells for Mediating Targeted siRNA Delivery
title_full Characterization of Novel Aptamers Specifically Directed to Red-Spotted Grouper Nervous Necrosis Virus (RGNNV)-Infected Cells for Mediating Targeted siRNA Delivery
title_fullStr Characterization of Novel Aptamers Specifically Directed to Red-Spotted Grouper Nervous Necrosis Virus (RGNNV)-Infected Cells for Mediating Targeted siRNA Delivery
title_full_unstemmed Characterization of Novel Aptamers Specifically Directed to Red-Spotted Grouper Nervous Necrosis Virus (RGNNV)-Infected Cells for Mediating Targeted siRNA Delivery
title_short Characterization of Novel Aptamers Specifically Directed to Red-Spotted Grouper Nervous Necrosis Virus (RGNNV)-Infected Cells for Mediating Targeted siRNA Delivery
title_sort characterization of novel aptamers specifically directed to red-spotted grouper nervous necrosis virus (rgnnv)-infected cells for mediating targeted sirna delivery
topic Microbiology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7203557/
https://www.ncbi.nlm.nih.gov/pubmed/32425897
http://dx.doi.org/10.3389/fmicb.2020.00660
work_keys_str_mv AT zhoulingli characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT wangshaowen characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT yuqing characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT weishina characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT liumingzhu characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT weijingguang characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT huangyouhua characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT huangxiaohong characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT lipengfei characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery
AT qinqiwei characterizationofnovelaptamersspecificallydirectedtoredspottedgroupernervousnecrosisvirusrgnnvinfectedcellsformediatingtargetedsirnadelivery