Herpes simplex virus 1 evades cellular antiviral response by inducing microRNA-24, which attenuates STING synthesis.

STING is a nodal point for cellular innate immune response to microbial infections, autoimmunity and cancer; it triggers the synthesis of the antiviral proteins, type I interferons. Many DNA viruses, including Herpes Simplex Virus 1 (HSV1), trigger STING signaling causing inhibition of virus replica...

Full description

Bibliographic Details
Main Authors: Nikhil Sharma, Chenyao Wang, Patricia Kessler, Ganes C Sen
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2021-09-01
Series:PLoS Pathogens
Online Access:https://doi.org/10.1371/journal.ppat.1009950
_version_ 1798000163808083968
author Nikhil Sharma
Chenyao Wang
Patricia Kessler
Ganes C Sen
author_facet Nikhil Sharma
Chenyao Wang
Patricia Kessler
Ganes C Sen
author_sort Nikhil Sharma
collection DOAJ
description STING is a nodal point for cellular innate immune response to microbial infections, autoimmunity and cancer; it triggers the synthesis of the antiviral proteins, type I interferons. Many DNA viruses, including Herpes Simplex Virus 1 (HSV1), trigger STING signaling causing inhibition of virus replication. Here, we report that HSV1 evades this antiviral immune response by inducing a cellular microRNA, miR-24, which binds to the 3' untranslated region of STING mRNA and inhibits its translation. Expression of the gene encoding miR-24 is induced by the transcription factor AP1 and activated by MAP kinases in HSV1-infected cells. Introduction of exogenous miR-24 or prior activation of MAPKs, causes further enhancement of HSV1 replication in STING-expressing cells. Conversely, transfection of antimiR-24 inhibits virus replication in those cells. HSV1 infection of mice causes neuropathy and death; using two routes of infection, we demonstrated that intracranial injection of antimiR-24 alleviates both morbidity and mortality of the infected mice. Our studies reveal a new immune evasion strategy adopted by HSV1 through the regulation of STING and demonstrates that it can be exploited to enhance STING's antiviral action.
first_indexed 2024-04-11T11:15:47Z
format Article
id doaj.art-0b799e19f3204017ab8357b547c7afe2
institution Directory Open Access Journal
issn 1553-7366
1553-7374
language English
last_indexed 2024-04-11T11:15:47Z
publishDate 2021-09-01
publisher Public Library of Science (PLoS)
record_format Article
series PLoS Pathogens
spelling doaj.art-0b799e19f3204017ab8357b547c7afe22022-12-22T04:27:14ZengPublic Library of Science (PLoS)PLoS Pathogens1553-73661553-73742021-09-01179e100995010.1371/journal.ppat.1009950Herpes simplex virus 1 evades cellular antiviral response by inducing microRNA-24, which attenuates STING synthesis.Nikhil SharmaChenyao WangPatricia KesslerGanes C SenSTING is a nodal point for cellular innate immune response to microbial infections, autoimmunity and cancer; it triggers the synthesis of the antiviral proteins, type I interferons. Many DNA viruses, including Herpes Simplex Virus 1 (HSV1), trigger STING signaling causing inhibition of virus replication. Here, we report that HSV1 evades this antiviral immune response by inducing a cellular microRNA, miR-24, which binds to the 3' untranslated region of STING mRNA and inhibits its translation. Expression of the gene encoding miR-24 is induced by the transcription factor AP1 and activated by MAP kinases in HSV1-infected cells. Introduction of exogenous miR-24 or prior activation of MAPKs, causes further enhancement of HSV1 replication in STING-expressing cells. Conversely, transfection of antimiR-24 inhibits virus replication in those cells. HSV1 infection of mice causes neuropathy and death; using two routes of infection, we demonstrated that intracranial injection of antimiR-24 alleviates both morbidity and mortality of the infected mice. Our studies reveal a new immune evasion strategy adopted by HSV1 through the regulation of STING and demonstrates that it can be exploited to enhance STING's antiviral action.https://doi.org/10.1371/journal.ppat.1009950
spellingShingle Nikhil Sharma
Chenyao Wang
Patricia Kessler
Ganes C Sen
Herpes simplex virus 1 evades cellular antiviral response by inducing microRNA-24, which attenuates STING synthesis.
PLoS Pathogens
title Herpes simplex virus 1 evades cellular antiviral response by inducing microRNA-24, which attenuates STING synthesis.
title_full Herpes simplex virus 1 evades cellular antiviral response by inducing microRNA-24, which attenuates STING synthesis.
title_fullStr Herpes simplex virus 1 evades cellular antiviral response by inducing microRNA-24, which attenuates STING synthesis.
title_full_unstemmed Herpes simplex virus 1 evades cellular antiviral response by inducing microRNA-24, which attenuates STING synthesis.
title_short Herpes simplex virus 1 evades cellular antiviral response by inducing microRNA-24, which attenuates STING synthesis.
title_sort herpes simplex virus 1 evades cellular antiviral response by inducing microrna 24 which attenuates sting synthesis
url https://doi.org/10.1371/journal.ppat.1009950
work_keys_str_mv AT nikhilsharma herpessimplexvirus1evadescellularantiviralresponsebyinducingmicrorna24whichattenuatesstingsynthesis
AT chenyaowang herpessimplexvirus1evadescellularantiviralresponsebyinducingmicrorna24whichattenuatesstingsynthesis
AT patriciakessler herpessimplexvirus1evadescellularantiviralresponsebyinducingmicrorna24whichattenuatesstingsynthesis
AT ganescsen herpessimplexvirus1evadescellularantiviralresponsebyinducingmicrorna24whichattenuatesstingsynthesis