RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo.

Respiratory syncytial virus (RSV) infection can result in severe disease partially due to its ability to interfere with the initiation of Th1 responses targeting the production of type I interferons (IFN) and promoting a Th2 immune environment. Epigenetic modulation of gene transcription has been sh...

Full description

Bibliographic Details
Main Authors: Catherine Ptaschinski, Sumanta Mukherjee, Martin L Moore, Mareike Albert, Kristian Helin, Steven L Kunkel, Nicholas W Lukacs
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2015-06-01
Series:PLoS Pathogens
Online Access:https://doi.org/10.1371/journal.ppat.1004978
id doaj-832c95f3c7b345dd9e0a13abd546a3ca
record_format Article
spelling doaj-832c95f3c7b345dd9e0a13abd546a3ca2021-04-21T17:39:03ZengPublic Library of Science (PLoS)PLoS Pathogens1553-73661553-73742015-06-01116e100497810.1371/journal.ppat.1004978RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo.Catherine PtaschinskiSumanta MukherjeeMartin L MooreMareike AlbertKristian HelinSteven L KunkelNicholas W LukacsRespiratory syncytial virus (RSV) infection can result in severe disease partially due to its ability to interfere with the initiation of Th1 responses targeting the production of type I interferons (IFN) and promoting a Th2 immune environment. Epigenetic modulation of gene transcription has been shown to be important in regulating inflammatory pathways. RSV-infected bone marrow-derived DCs (BMDCs) upregulated expression of Kdm5b/Jarid1b H3K4 demethylase. Kdm5b-specific siRNA inhibition in BMDC led to a 10-fold increase in IFN-β as well as increases in IL-6 and TNF-α compared to control-transfected cells. The generation of Kdm5bfl/fl-CD11c-Cre+ mice recapitulated the latter results during in vitro DC activation showing innate cytokine modulation. In vivo, infection of Kdm5bfl/fl-CD11c-Cre+ mice with RSV resulted in higher production of IFN-γ and reduced IL-4 and IL-5 compared to littermate controls, with significantly decreased inflammation, IL-13, and mucus production in the lungs. Sensitization with RSV-infected DCs into the airways of naïve mice led to an exacerbated response when mice were challenged with live RSV infection. When Kdm5b was blocked in DCs with siRNA or DCs from Kdm5bfl/fl-CD11c-CRE mice were used, the exacerbated response was abrogated. Importantly, human monocyte-derived DCs treated with a chemical inhibitor for KDM5B resulted in increased innate cytokine levels as well as elicited decreased Th2 cytokines when co-cultured with RSV reactivated CD4+ T cells. These results suggest that KDM5B acts to repress type I IFN and other innate cytokines to promote an altered immune response following RSV infection that contributes to development of chronic disease.https://doi.org/10.1371/journal.ppat.1004978
collection DOAJ
language English
format Article
sources DOAJ
author Catherine Ptaschinski
Sumanta Mukherjee
Martin L Moore
Mareike Albert
Kristian Helin
Steven L Kunkel
Nicholas W Lukacs
spellingShingle Catherine Ptaschinski
Sumanta Mukherjee
Martin L Moore
Mareike Albert
Kristian Helin
Steven L Kunkel
Nicholas W Lukacs
RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo.
PLoS Pathogens
author_facet Catherine Ptaschinski
Sumanta Mukherjee
Martin L Moore
Mareike Albert
Kristian Helin
Steven L Kunkel
Nicholas W Lukacs
author_sort Catherine Ptaschinski
title RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo.
title_short RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo.
title_full RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo.
title_fullStr RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo.
title_full_unstemmed RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo.
title_sort rsv-induced h3k4 demethylase kdm5b leads to regulation of dendritic cell-derived innate cytokines and exacerbates pathogenesis in vivo.
publisher Public Library of Science (PLoS)
series PLoS Pathogens
issn 1553-7366
1553-7374
publishDate 2015-06-01
description Respiratory syncytial virus (RSV) infection can result in severe disease partially due to its ability to interfere with the initiation of Th1 responses targeting the production of type I interferons (IFN) and promoting a Th2 immune environment. Epigenetic modulation of gene transcription has been shown to be important in regulating inflammatory pathways. RSV-infected bone marrow-derived DCs (BMDCs) upregulated expression of Kdm5b/Jarid1b H3K4 demethylase. Kdm5b-specific siRNA inhibition in BMDC led to a 10-fold increase in IFN-β as well as increases in IL-6 and TNF-α compared to control-transfected cells. The generation of Kdm5bfl/fl-CD11c-Cre+ mice recapitulated the latter results during in vitro DC activation showing innate cytokine modulation. In vivo, infection of Kdm5bfl/fl-CD11c-Cre+ mice with RSV resulted in higher production of IFN-γ and reduced IL-4 and IL-5 compared to littermate controls, with significantly decreased inflammation, IL-13, and mucus production in the lungs. Sensitization with RSV-infected DCs into the airways of naïve mice led to an exacerbated response when mice were challenged with live RSV infection. When Kdm5b was blocked in DCs with siRNA or DCs from Kdm5bfl/fl-CD11c-CRE mice were used, the exacerbated response was abrogated. Importantly, human monocyte-derived DCs treated with a chemical inhibitor for KDM5B resulted in increased innate cytokine levels as well as elicited decreased Th2 cytokines when co-cultured with RSV reactivated CD4+ T cells. These results suggest that KDM5B acts to repress type I IFN and other innate cytokines to promote an altered immune response following RSV infection that contributes to development of chronic disease.
url https://doi.org/10.1371/journal.ppat.1004978
work_keys_str_mv AT catherineptaschinski rsvinducedh3k4demethylasekdm5bleadstoregulationofdendriticcellderivedinnatecytokinesandexacerbatespathogenesisinvivo
AT sumantamukherjee rsvinducedh3k4demethylasekdm5bleadstoregulationofdendriticcellderivedinnatecytokinesandexacerbatespathogenesisinvivo
AT martinlmoore rsvinducedh3k4demethylasekdm5bleadstoregulationofdendriticcellderivedinnatecytokinesandexacerbatespathogenesisinvivo
AT mareikealbert rsvinducedh3k4demethylasekdm5bleadstoregulationofdendriticcellderivedinnatecytokinesandexacerbatespathogenesisinvivo
AT kristianhelin rsvinducedh3k4demethylasekdm5bleadstoregulationofdendriticcellderivedinnatecytokinesandexacerbatespathogenesisinvivo
AT stevenlkunkel rsvinducedh3k4demethylasekdm5bleadstoregulationofdendriticcellderivedinnatecytokinesandexacerbatespathogenesisinvivo
AT nicholaswlukacs rsvinducedh3k4demethylasekdm5bleadstoregulationofdendriticcellderivedinnatecytokinesandexacerbatespathogenesisinvivo
_version_ 1714665933033701376