Cargando…

African Swine Fever Virus MGF110-7L Induces Host Cell Translation Suppression and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway

African swine fever (ASF) is a highly contagious and often lethal disease of pigs caused by ASF virus (ASFV) and recognized as the biggest killer in global swine industry. Despite exhibiting incredible self-sufficiency, ASFV remains unconditionally dependent on the host translation machinery for its...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhong, Han, Fan, Shuai, Du, Yongkun, Zhang, Yuhang, Zhang, Angke, Jiang, Dawei, Han, Shichong, Wan, Bo, Zhang, Gaiping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Microbiology 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9769596/
https://www.ncbi.nlm.nih.gov/pubmed/36377947
http://dx.doi.org/10.1128/spectrum.03282-22
_version_ 1784854405177147392
author Zhong, Han
Fan, Shuai
Du, Yongkun
Zhang, Yuhang
Zhang, Angke
Jiang, Dawei
Han, Shichong
Wan, Bo
Zhang, Gaiping
author_facet Zhong, Han
Fan, Shuai
Du, Yongkun
Zhang, Yuhang
Zhang, Angke
Jiang, Dawei
Han, Shichong
Wan, Bo
Zhang, Gaiping
author_sort Zhong, Han
collection PubMed
description African swine fever (ASF) is a highly contagious and often lethal disease of pigs caused by ASF virus (ASFV) and recognized as the biggest killer in global swine industry. Despite exhibiting incredible self-sufficiency, ASFV remains unconditionally dependent on the host translation machinery for its mRNA translation. However, less is yet known regarding how ASFV-encoded proteins regulate host translation machinery in infected cells. Here, we examined how ASFV interacts with the eukaryotic initiation factor 2α (eIF2α) signaling axis, which directs host translation control and adaptation to cellular stress. We found that ASFV MGF110-7L, a previously uncharacterized member of the multigene family 110, remarkably enhanced the phosphorylation level of eIF2α. In porcine alveolar macrophage 3D4/21 and porcine kidney-15 cells, MGF110-7L triggered eIF2α signaling and the integrated stress response, resulting in the suppression of host translation and the formation of stress granules (SGs). Mechanistically, MGF110-7L-induced phosphorylation of eIF2α was mediated via protein kinase R (PKR) and PKR-like endoplasmic reticulum (ER) kinase (PERK), and this process was essential for host translation repression and SG formation. Notably, our subsequent analyses confirmed that MGF110-7L was overwhelmingly retained in the ER and caused a specific reorganization of the secretory pathway. Further proteomic analyses and biochemical experiments revealed that MGF110-7L could trigger ER stress and activate the unfolded protein response, thus contributing to eIF2α phosphorylation and translation reprogramming. Overall, our study both identifies a novel mechanism by which ASFV MGF110-7L subverts the host protein synthesis machinery and provides further insights into the translation regulation that occurs during ASFV infection. IMPORTANCE African swine fever (ASF) has become a socioeconomic burden and a threat to food security and biodiversity, but no commercial vaccines or antivirals are available currently. Understanding the viral strategies to subvert the host translation machinery during ASF virus (ASFV) infection could potentially lead to new vaccines and antiviral therapies. In this study, we dissected how ASFV MGF110-7L interacts with the eIF2α signaling axis controlling translational reprogramming, and we addressed the role of MGF110-7L in induction of cellular stress responses, eIF2α phosphorylation, translation suppression, and stress granule formation. These results define several molecular interfaces by which ASFV MGF110-7L subverts host cell translation, which may guide research on antiviral strategies and dissection of ASFV pathogenesis.
format Online
Article
Text
id pubmed-9769596
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher American Society for Microbiology
record_format MEDLINE/PubMed
spelling pubmed-97695962022-12-22 African Swine Fever Virus MGF110-7L Induces Host Cell Translation Suppression and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway Zhong, Han Fan, Shuai Du, Yongkun Zhang, Yuhang Zhang, Angke Jiang, Dawei Han, Shichong Wan, Bo Zhang, Gaiping Microbiol Spectr Research Article African swine fever (ASF) is a highly contagious and often lethal disease of pigs caused by ASF virus (ASFV) and recognized as the biggest killer in global swine industry. Despite exhibiting incredible self-sufficiency, ASFV remains unconditionally dependent on the host translation machinery for its mRNA translation. However, less is yet known regarding how ASFV-encoded proteins regulate host translation machinery in infected cells. Here, we examined how ASFV interacts with the eukaryotic initiation factor 2α (eIF2α) signaling axis, which directs host translation control and adaptation to cellular stress. We found that ASFV MGF110-7L, a previously uncharacterized member of the multigene family 110, remarkably enhanced the phosphorylation level of eIF2α. In porcine alveolar macrophage 3D4/21 and porcine kidney-15 cells, MGF110-7L triggered eIF2α signaling and the integrated stress response, resulting in the suppression of host translation and the formation of stress granules (SGs). Mechanistically, MGF110-7L-induced phosphorylation of eIF2α was mediated via protein kinase R (PKR) and PKR-like endoplasmic reticulum (ER) kinase (PERK), and this process was essential for host translation repression and SG formation. Notably, our subsequent analyses confirmed that MGF110-7L was overwhelmingly retained in the ER and caused a specific reorganization of the secretory pathway. Further proteomic analyses and biochemical experiments revealed that MGF110-7L could trigger ER stress and activate the unfolded protein response, thus contributing to eIF2α phosphorylation and translation reprogramming. Overall, our study both identifies a novel mechanism by which ASFV MGF110-7L subverts the host protein synthesis machinery and provides further insights into the translation regulation that occurs during ASFV infection. IMPORTANCE African swine fever (ASF) has become a socioeconomic burden and a threat to food security and biodiversity, but no commercial vaccines or antivirals are available currently. Understanding the viral strategies to subvert the host translation machinery during ASF virus (ASFV) infection could potentially lead to new vaccines and antiviral therapies. In this study, we dissected how ASFV MGF110-7L interacts with the eIF2α signaling axis controlling translational reprogramming, and we addressed the role of MGF110-7L in induction of cellular stress responses, eIF2α phosphorylation, translation suppression, and stress granule formation. These results define several molecular interfaces by which ASFV MGF110-7L subverts host cell translation, which may guide research on antiviral strategies and dissection of ASFV pathogenesis. American Society for Microbiology 2022-11-15 /pmc/articles/PMC9769596/ /pubmed/36377947 http://dx.doi.org/10.1128/spectrum.03282-22 Text en Copyright © 2022 Zhong et al. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Research Article
Zhong, Han
Fan, Shuai
Du, Yongkun
Zhang, Yuhang
Zhang, Angke
Jiang, Dawei
Han, Shichong
Wan, Bo
Zhang, Gaiping
African Swine Fever Virus MGF110-7L Induces Host Cell Translation Suppression and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway
title African Swine Fever Virus MGF110-7L Induces Host Cell Translation Suppression and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway
title_full African Swine Fever Virus MGF110-7L Induces Host Cell Translation Suppression and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway
title_fullStr African Swine Fever Virus MGF110-7L Induces Host Cell Translation Suppression and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway
title_full_unstemmed African Swine Fever Virus MGF110-7L Induces Host Cell Translation Suppression and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway
title_short African Swine Fever Virus MGF110-7L Induces Host Cell Translation Suppression and Stress Granule Formation by Activating the PERK/PKR-eIF2α Pathway
title_sort african swine fever virus mgf110-7l induces host cell translation suppression and stress granule formation by activating the perk/pkr-eif2α pathway
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9769596/
https://www.ncbi.nlm.nih.gov/pubmed/36377947
http://dx.doi.org/10.1128/spectrum.03282-22
work_keys_str_mv AT zhonghan africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway
AT fanshuai africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway
AT duyongkun africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway
AT zhangyuhang africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway
AT zhangangke africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway
AT jiangdawei africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway
AT hanshichong africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway
AT wanbo africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway
AT zhanggaiping africanswinefevervirusmgf1107linduceshostcelltranslationsuppressionandstressgranuleformationbyactivatingtheperkpkreif2apathway