STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.

In recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat various conditions, including infectious diseases. For instance, Toll-like receptor (TLR) agonists have been evaluated for treatment of genital herpes. However, although the TLR7 agonist imiquimod...

Full description

Bibliographic Details
Main Authors: Morten K Skouboe, Alice Knudsen, Line S Reinert, Cedric Boularan, Thierry Lioux, Eric Perouzel, Martin K Thomsen, Søren R Paludan
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2018-04-01
Series:PLoS Pathogens
Online Access:http://europepmc.org/articles/PMC5897032?pdf=render
id doaj-4ef01a80f8f8483bafe8e9514f8f5aa7
record_format Article
spelling doaj-4ef01a80f8f8483bafe8e9514f8f5aa72020-11-25T00:43:35ZengPublic Library of Science (PLoS)PLoS Pathogens1553-73661553-73742018-04-01144e100697610.1371/journal.ppat.1006976STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.Morten K SkouboeAlice KnudsenLine S ReinertCedric BoularanThierry LiouxEric PerouzelMartin K ThomsenSøren R PaludanIn recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat various conditions, including infectious diseases. For instance, Toll-like receptor (TLR) agonists have been evaluated for treatment of genital herpes. However, although the TLR7 agonist imiquimod was shown to have antiviral activity in individual patients, no significant effects were observed in clinical trials, and the compound also exhibited significant side effects, including local inflammation. Cytosolic DNA is detected by the enzyme cyclic GMP-AMP (2'3'-cGAMP) synthase (cGAS) to stimulate antiviral pathways, mainly through induction of type I interferon (IFN)s. cGAS is activated upon DNA binding to produce the cyclic dinucleotide (CDN) 2'3'-cGAMP, which in turn binds and activates the adaptor protein Stimulator of interferon genes (STING), thus triggering type I IFN expression. In contrast to TLRs, STING is expressed broadly, including in epithelial cells. Here we report that natural and non-natural STING agonists strongly induce type I IFNs in human cells and in mice in vivo, without stimulating significant inflammatory gene expression. Systemic treatment with 2'3'-cGAMP reduced genital herpes simplex virus (HSV) 2 replication and improved the clinical outcome of infection. More importantly, local application of CDNs at the genital epithelial surface gave rise to local IFN activity, but only limited systemic responses, and this treatment conferred total protection against disease in both immunocompetent and immunocompromised mice. In direct comparison between CDNs and TLR agonists, only CDNs acted directly on epithelial cells, hence allowing a more rapid and IFN-focused immune response in the vaginal epithelium. Thus, specific activation of the STING pathway in the vagina evokes induction of the IFN system but limited inflammatory responses to allow control of HSV2 infections in vivo.http://europepmc.org/articles/PMC5897032?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Morten K Skouboe
Alice Knudsen
Line S Reinert
Cedric Boularan
Thierry Lioux
Eric Perouzel
Martin K Thomsen
Søren R Paludan
spellingShingle Morten K Skouboe
Alice Knudsen
Line S Reinert
Cedric Boularan
Thierry Lioux
Eric Perouzel
Martin K Thomsen
Søren R Paludan
STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.
PLoS Pathogens
author_facet Morten K Skouboe
Alice Knudsen
Line S Reinert
Cedric Boularan
Thierry Lioux
Eric Perouzel
Martin K Thomsen
Søren R Paludan
author_sort Morten K Skouboe
title STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.
title_short STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.
title_full STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.
title_fullStr STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.
title_full_unstemmed STING agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.
title_sort sting agonists enable antiviral cross-talk between human cells and confer protection against genital herpes in mice.
publisher Public Library of Science (PLoS)
series PLoS Pathogens
issn 1553-7366
1553-7374
publishDate 2018-04-01
description In recent years, there has been an increasing interest in immunomodulatory therapy as a means to treat various conditions, including infectious diseases. For instance, Toll-like receptor (TLR) agonists have been evaluated for treatment of genital herpes. However, although the TLR7 agonist imiquimod was shown to have antiviral activity in individual patients, no significant effects were observed in clinical trials, and the compound also exhibited significant side effects, including local inflammation. Cytosolic DNA is detected by the enzyme cyclic GMP-AMP (2'3'-cGAMP) synthase (cGAS) to stimulate antiviral pathways, mainly through induction of type I interferon (IFN)s. cGAS is activated upon DNA binding to produce the cyclic dinucleotide (CDN) 2'3'-cGAMP, which in turn binds and activates the adaptor protein Stimulator of interferon genes (STING), thus triggering type I IFN expression. In contrast to TLRs, STING is expressed broadly, including in epithelial cells. Here we report that natural and non-natural STING agonists strongly induce type I IFNs in human cells and in mice in vivo, without stimulating significant inflammatory gene expression. Systemic treatment with 2'3'-cGAMP reduced genital herpes simplex virus (HSV) 2 replication and improved the clinical outcome of infection. More importantly, local application of CDNs at the genital epithelial surface gave rise to local IFN activity, but only limited systemic responses, and this treatment conferred total protection against disease in both immunocompetent and immunocompromised mice. In direct comparison between CDNs and TLR agonists, only CDNs acted directly on epithelial cells, hence allowing a more rapid and IFN-focused immune response in the vaginal epithelium. Thus, specific activation of the STING pathway in the vagina evokes induction of the IFN system but limited inflammatory responses to allow control of HSV2 infections in vivo.
url http://europepmc.org/articles/PMC5897032?pdf=render
work_keys_str_mv AT mortenkskouboe stingagonistsenableantiviralcrosstalkbetweenhumancellsandconferprotectionagainstgenitalherpesinmice
AT aliceknudsen stingagonistsenableantiviralcrosstalkbetweenhumancellsandconferprotectionagainstgenitalherpesinmice
AT linesreinert stingagonistsenableantiviralcrosstalkbetweenhumancellsandconferprotectionagainstgenitalherpesinmice
AT cedricboularan stingagonistsenableantiviralcrosstalkbetweenhumancellsandconferprotectionagainstgenitalherpesinmice
AT thierrylioux stingagonistsenableantiviralcrosstalkbetweenhumancellsandconferprotectionagainstgenitalherpesinmice
AT ericperouzel stingagonistsenableantiviralcrosstalkbetweenhumancellsandconferprotectionagainstgenitalherpesinmice
AT martinkthomsen stingagonistsenableantiviralcrosstalkbetweenhumancellsandconferprotectionagainstgenitalherpesinmice
AT sørenrpaludan stingagonistsenableantiviralcrosstalkbetweenhumancellsandconferprotectionagainstgenitalherpesinmice
_version_ 1725277598378885120