Cargando…

Aichi virus 3C protease modulates LC3- and SQSTM1/p62-involved antiviral response

Rationale: Autophagy is an essential, homeostatic process by which cells break down their own components, it also contributes to restricting bacterial infection in host defense systems; yet, how autophagy regulates viral infection remains inconclusive. Aichi virus (AiV), belonging to the genus Kobuv...

Descripción completa

Detalles Bibliográficos
Autores principales: Kung, Ming-Hsiang, Lin, You-Sheng, Chang, Tsung-Hsien
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7415801/
https://www.ncbi.nlm.nih.gov/pubmed/32802187
http://dx.doi.org/10.7150/thno.47077
_version_ 1783569204217118720
author Kung, Ming-Hsiang
Lin, You-Sheng
Chang, Tsung-Hsien
author_facet Kung, Ming-Hsiang
Lin, You-Sheng
Chang, Tsung-Hsien
author_sort Kung, Ming-Hsiang
collection PubMed
description Rationale: Autophagy is an essential, homeostatic process by which cells break down their own components, it also contributes to restricting bacterial infection in host defense systems; yet, how autophagy regulates viral infection remains inconclusive. Aichi virus (AiV), belonging to the genus Kobuvirus in the Picornaviridae family, causes acute gastroenteritis in human. The role of autophagy-mediated anti-viral activity on AiV infection was investigated in this study. Methods: The effect of autophagy-associated molecules in retinoic acid-inducible gene-I (RIG-I)-like receptor (RLR) antiviral signal axis was analyzed in AiV infected cells by using biochemistry and pharmacologic approaches. In addition, the AiV viral protein regulating autophagy-associated RLR activity was also evaluated. Results: In AiV-infected cells, autophagic flux including the formation of autophagic vacuoles, as well as degradation of microtubule-associated protein light chain 3 (LC3) and sequestosome-1 (SQSTM1/p62) were observed. Ectopic overexpression of LC3 and p62, but not Atg proteins, contributed to RLR antiviral signal axis, shRNA knockdown of LC3 and p62 led to a downregulation of antiviral inflammation. Moreover, AiV infection inhibited double-stranded RNA (dsRNA)-activated RLR activity by the viral protein 3C protease but not H42D, C143S protease dead mutants. AiV 3C protease caused the degradation of LC3 and p62, and also RLR signal proteins. Conclusion: This study reveals a possible mechanism of autophagy-associated proteins regulating virus replication. Maintaining a cellular level of LC3 and p62 during the viral infection period might help restrict virus replication. Although, AiV 3C protease dampens the LC3 and p62-mediated host antiviral machinery for AiV replication. Results obtained provide a better understanding of the molecular pathogenesis of AiV for developing methods of prevention and treatment.
format Online
Article
Text
id pubmed-7415801
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-74158012020-08-13 Aichi virus 3C protease modulates LC3- and SQSTM1/p62-involved antiviral response Kung, Ming-Hsiang Lin, You-Sheng Chang, Tsung-Hsien Theranostics Research Paper Rationale: Autophagy is an essential, homeostatic process by which cells break down their own components, it also contributes to restricting bacterial infection in host defense systems; yet, how autophagy regulates viral infection remains inconclusive. Aichi virus (AiV), belonging to the genus Kobuvirus in the Picornaviridae family, causes acute gastroenteritis in human. The role of autophagy-mediated anti-viral activity on AiV infection was investigated in this study. Methods: The effect of autophagy-associated molecules in retinoic acid-inducible gene-I (RIG-I)-like receptor (RLR) antiviral signal axis was analyzed in AiV infected cells by using biochemistry and pharmacologic approaches. In addition, the AiV viral protein regulating autophagy-associated RLR activity was also evaluated. Results: In AiV-infected cells, autophagic flux including the formation of autophagic vacuoles, as well as degradation of microtubule-associated protein light chain 3 (LC3) and sequestosome-1 (SQSTM1/p62) were observed. Ectopic overexpression of LC3 and p62, but not Atg proteins, contributed to RLR antiviral signal axis, shRNA knockdown of LC3 and p62 led to a downregulation of antiviral inflammation. Moreover, AiV infection inhibited double-stranded RNA (dsRNA)-activated RLR activity by the viral protein 3C protease but not H42D, C143S protease dead mutants. AiV 3C protease caused the degradation of LC3 and p62, and also RLR signal proteins. Conclusion: This study reveals a possible mechanism of autophagy-associated proteins regulating virus replication. Maintaining a cellular level of LC3 and p62 during the viral infection period might help restrict virus replication. Although, AiV 3C protease dampens the LC3 and p62-mediated host antiviral machinery for AiV replication. Results obtained provide a better understanding of the molecular pathogenesis of AiV for developing methods of prevention and treatment. Ivyspring International Publisher 2020-07-14 /pmc/articles/PMC7415801/ /pubmed/32802187 http://dx.doi.org/10.7150/thno.47077 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Kung, Ming-Hsiang
Lin, You-Sheng
Chang, Tsung-Hsien
Aichi virus 3C protease modulates LC3- and SQSTM1/p62-involved antiviral response
title Aichi virus 3C protease modulates LC3- and SQSTM1/p62-involved antiviral response
title_full Aichi virus 3C protease modulates LC3- and SQSTM1/p62-involved antiviral response
title_fullStr Aichi virus 3C protease modulates LC3- and SQSTM1/p62-involved antiviral response
title_full_unstemmed Aichi virus 3C protease modulates LC3- and SQSTM1/p62-involved antiviral response
title_short Aichi virus 3C protease modulates LC3- and SQSTM1/p62-involved antiviral response
title_sort aichi virus 3c protease modulates lc3- and sqstm1/p62-involved antiviral response
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7415801/
https://www.ncbi.nlm.nih.gov/pubmed/32802187
http://dx.doi.org/10.7150/thno.47077
work_keys_str_mv AT kungminghsiang aichivirus3cproteasemodulateslc3andsqstm1p62involvedantiviralresponse
AT linyousheng aichivirus3cproteasemodulateslc3andsqstm1p62involvedantiviralresponse
AT changtsunghsien aichivirus3cproteasemodulateslc3andsqstm1p62involvedantiviralresponse