Cargando…

Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models

BACKGROUND: Rhinovirus (RV) infection of airway epithelial cells triggers asthma exacerbations, during which airway smooth muscle (ASM) excessively contracts. Due to ASM contraction, airway epithelial cells become mechanically compressed. We previously reported that compressed human bronchial epithe...

Descripción completa

Detalles Bibliográficos
Autores principales: Dy, Alane Blythe C., Girkin, Jason, Marrocco, Antonella, Collison, Adam, Mwase, Chimwemwe, O’Sullivan, Michael J., Phung, Thien-Khoi N., Mattes, Joerg, Koziol-White, Cynthia, Gern, James E., Bochkov, Yury A., Bartlett, Nathan W., Park, Jin-Ah
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10440034/
https://www.ncbi.nlm.nih.gov/pubmed/37598152
http://dx.doi.org/10.1186/s12931-023-02510-6
_version_ 1785093087391907840
author Dy, Alane Blythe C.
Girkin, Jason
Marrocco, Antonella
Collison, Adam
Mwase, Chimwemwe
O’Sullivan, Michael J.
Phung, Thien-Khoi N.
Mattes, Joerg
Koziol-White, Cynthia
Gern, James E.
Bochkov, Yury A.
Bartlett, Nathan W.
Park, Jin-Ah
author_facet Dy, Alane Blythe C.
Girkin, Jason
Marrocco, Antonella
Collison, Adam
Mwase, Chimwemwe
O’Sullivan, Michael J.
Phung, Thien-Khoi N.
Mattes, Joerg
Koziol-White, Cynthia
Gern, James E.
Bochkov, Yury A.
Bartlett, Nathan W.
Park, Jin-Ah
author_sort Dy, Alane Blythe C.
collection PubMed
description BACKGROUND: Rhinovirus (RV) infection of airway epithelial cells triggers asthma exacerbations, during which airway smooth muscle (ASM) excessively contracts. Due to ASM contraction, airway epithelial cells become mechanically compressed. We previously reported that compressed human bronchial epithelial (HBE) cells are a source of endothelin-1 (ET-1) that causes ASM contraction. Here, we hypothesized that epithelial sensing of RV by TLR3 and epithelial compression induce ET-1 secretion through a TGF-β receptor (TGFβR)-dependent mechanism. METHODS: To test this, we used primary HBE cells well-differentiated in air–liquid interface culture and two mouse models (ovalbumin and house dust mite) of allergic airway disease (AAD). HBE cells were infected with RV-A16, treated with a TLR3 agonist (poly(I:C)), or exposed to compression. Thereafter, EDN1 (ET-1 protein-encoding gene) mRNA expression and secreted ET-1 protein were measured. We examined the role of TGFβR in ET-1 secretion using either a pharmacologic inhibitor of TGFβR or recombinant TGF-β1 protein. In the AAD mouse models, allergen-sensitized and allergen-challenged mice were subsequently infected with RV. We then measured ET-1 in bronchoalveolar lavage fluid (BALF) and airway hyperresponsiveness (AHR) following methacholine challenge. RESULTS: Our data reveal that RV infection induced EDN1 expression and ET-1 secretion in HBE cells, potentially mediated by TLR3. TGFβR activation was partially required for ET-1 secretion, which was induced by RV, poly(I:C), or compression. TGFβR activation alone was sufficient to increase ET-1 secretion. In AAD mouse models, RV induced ET-1 secretion in BALF, which positively correlated with AHR. CONCLUSIONS: Our data provide evidence that RV infection increased epithelial-cell ET-1 secretion through a TGFβR-dependent mechanism, which contributes to bronchoconstriction during RV-induced asthma exacerbations.
format Online
Article
Text
id pubmed-10440034
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-104400342023-08-21 Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models Dy, Alane Blythe C. Girkin, Jason Marrocco, Antonella Collison, Adam Mwase, Chimwemwe O’Sullivan, Michael J. Phung, Thien-Khoi N. Mattes, Joerg Koziol-White, Cynthia Gern, James E. Bochkov, Yury A. Bartlett, Nathan W. Park, Jin-Ah Respir Res Research BACKGROUND: Rhinovirus (RV) infection of airway epithelial cells triggers asthma exacerbations, during which airway smooth muscle (ASM) excessively contracts. Due to ASM contraction, airway epithelial cells become mechanically compressed. We previously reported that compressed human bronchial epithelial (HBE) cells are a source of endothelin-1 (ET-1) that causes ASM contraction. Here, we hypothesized that epithelial sensing of RV by TLR3 and epithelial compression induce ET-1 secretion through a TGF-β receptor (TGFβR)-dependent mechanism. METHODS: To test this, we used primary HBE cells well-differentiated in air–liquid interface culture and two mouse models (ovalbumin and house dust mite) of allergic airway disease (AAD). HBE cells were infected with RV-A16, treated with a TLR3 agonist (poly(I:C)), or exposed to compression. Thereafter, EDN1 (ET-1 protein-encoding gene) mRNA expression and secreted ET-1 protein were measured. We examined the role of TGFβR in ET-1 secretion using either a pharmacologic inhibitor of TGFβR or recombinant TGF-β1 protein. In the AAD mouse models, allergen-sensitized and allergen-challenged mice were subsequently infected with RV. We then measured ET-1 in bronchoalveolar lavage fluid (BALF) and airway hyperresponsiveness (AHR) following methacholine challenge. RESULTS: Our data reveal that RV infection induced EDN1 expression and ET-1 secretion in HBE cells, potentially mediated by TLR3. TGFβR activation was partially required for ET-1 secretion, which was induced by RV, poly(I:C), or compression. TGFβR activation alone was sufficient to increase ET-1 secretion. In AAD mouse models, RV induced ET-1 secretion in BALF, which positively correlated with AHR. CONCLUSIONS: Our data provide evidence that RV infection increased epithelial-cell ET-1 secretion through a TGFβR-dependent mechanism, which contributes to bronchoconstriction during RV-induced asthma exacerbations. BioMed Central 2023-08-19 2023 /pmc/articles/PMC10440034/ /pubmed/37598152 http://dx.doi.org/10.1186/s12931-023-02510-6 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Dy, Alane Blythe C.
Girkin, Jason
Marrocco, Antonella
Collison, Adam
Mwase, Chimwemwe
O’Sullivan, Michael J.
Phung, Thien-Khoi N.
Mattes, Joerg
Koziol-White, Cynthia
Gern, James E.
Bochkov, Yury A.
Bartlett, Nathan W.
Park, Jin-Ah
Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models
title Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models
title_full Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models
title_fullStr Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models
title_full_unstemmed Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models
title_short Rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models
title_sort rhinovirus infection induces secretion of endothelin-1 from airway epithelial cells in both in vitro and in vivo models
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10440034/
https://www.ncbi.nlm.nih.gov/pubmed/37598152
http://dx.doi.org/10.1186/s12931-023-02510-6
work_keys_str_mv AT dyalaneblythec rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT girkinjason rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT marroccoantonella rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT collisonadam rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT mwasechimwemwe rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT osullivanmichaelj rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT phungthienkhoin rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT mattesjoerg rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT koziolwhitecynthia rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT gernjamese rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT bochkovyurya rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT bartlettnathanw rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels
AT parkjinah rhinovirusinfectioninducessecretionofendothelin1fromairwayepithelialcellsinbothinvitroandinvivomodels