Cargando…

Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection

Chronic inhalation of fungi and fungal components has been linked to the development of respiratory disorders, although their role with respect to the pathogenesis of acute respiratory virus infection remains unclear. Here, we evaluate inflammatory pathology induced by repetitive administration of a...

Descripción completa

Detalles Bibliográficos
Autores principales: Percopo, Caroline M., Ma, Michelle, Mai, Eric, Redes, Jamie L., Kraemer, Laura S., Minai, Mahnaz, Moore, Ian N., Druey, Kirk M., Rosenberg, Helene F.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7552021/
https://www.ncbi.nlm.nih.gov/pubmed/32867061
http://dx.doi.org/10.3390/v12090946
_version_ 1783593309868916736
author Percopo, Caroline M.
Ma, Michelle
Mai, Eric
Redes, Jamie L.
Kraemer, Laura S.
Minai, Mahnaz
Moore, Ian N.
Druey, Kirk M.
Rosenberg, Helene F.
author_facet Percopo, Caroline M.
Ma, Michelle
Mai, Eric
Redes, Jamie L.
Kraemer, Laura S.
Minai, Mahnaz
Moore, Ian N.
Druey, Kirk M.
Rosenberg, Helene F.
author_sort Percopo, Caroline M.
collection PubMed
description Chronic inhalation of fungi and fungal components has been linked to the development of respiratory disorders, although their role with respect to the pathogenesis of acute respiratory virus infection remains unclear. Here, we evaluate inflammatory pathology induced by repetitive administration of a filtrate of the ubiquitous fungus, Alternaria alternata, and its impact on susceptibility to infection with influenza A. We showed previously that A. alternata at the nasal mucosae resulted in increased susceptibility to an otherwise sublethal inoculum of influenza A in wild-type mice. Here we demonstrate that A. alternata-induced potentiation of influenza A infection was not dependent on fungal serine protease or ribonuclease activity. Repetitive challenge with A. alternata prior to virus infection resulted proinflammatory cytokines, neutrophil recruitment, and loss of alveolar macrophages to a degree that substantially exceeded that observed in response to influenza A infection alone. Concomitant administration of immunomodulatory Lactobacillus plantarum, a strategy shown previously to limit virus-induced inflammation in the airways, blocked the exaggerated lethal response. These observations promote an improved understanding of severe influenza infection with potential clinical relevance for individuals subjected to continuous exposure to molds and fungi.
format Online
Article
Text
id pubmed-7552021
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-75520212020-10-14 Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection Percopo, Caroline M. Ma, Michelle Mai, Eric Redes, Jamie L. Kraemer, Laura S. Minai, Mahnaz Moore, Ian N. Druey, Kirk M. Rosenberg, Helene F. Viruses Article Chronic inhalation of fungi and fungal components has been linked to the development of respiratory disorders, although their role with respect to the pathogenesis of acute respiratory virus infection remains unclear. Here, we evaluate inflammatory pathology induced by repetitive administration of a filtrate of the ubiquitous fungus, Alternaria alternata, and its impact on susceptibility to infection with influenza A. We showed previously that A. alternata at the nasal mucosae resulted in increased susceptibility to an otherwise sublethal inoculum of influenza A in wild-type mice. Here we demonstrate that A. alternata-induced potentiation of influenza A infection was not dependent on fungal serine protease or ribonuclease activity. Repetitive challenge with A. alternata prior to virus infection resulted proinflammatory cytokines, neutrophil recruitment, and loss of alveolar macrophages to a degree that substantially exceeded that observed in response to influenza A infection alone. Concomitant administration of immunomodulatory Lactobacillus plantarum, a strategy shown previously to limit virus-induced inflammation in the airways, blocked the exaggerated lethal response. These observations promote an improved understanding of severe influenza infection with potential clinical relevance for individuals subjected to continuous exposure to molds and fungi. MDPI 2020-08-27 /pmc/articles/PMC7552021/ /pubmed/32867061 http://dx.doi.org/10.3390/v12090946 Text en © 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Percopo, Caroline M.
Ma, Michelle
Mai, Eric
Redes, Jamie L.
Kraemer, Laura S.
Minai, Mahnaz
Moore, Ian N.
Druey, Kirk M.
Rosenberg, Helene F.
Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection
title Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection
title_full Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection
title_fullStr Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection
title_full_unstemmed Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection
title_short Alternaria alternata Accelerates Loss of Alveolar Macrophages and Promotes Lethal Influenza A Infection
title_sort alternaria alternata accelerates loss of alveolar macrophages and promotes lethal influenza a infection
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7552021/
https://www.ncbi.nlm.nih.gov/pubmed/32867061
http://dx.doi.org/10.3390/v12090946
work_keys_str_mv AT percopocarolinem alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection
AT mamichelle alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection
AT maieric alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection
AT redesjamiel alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection
AT kraemerlauras alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection
AT minaimahnaz alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection
AT mooreiann alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection
AT drueykirkm alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection
AT rosenberghelenef alternariaalternataaccelerateslossofalveolarmacrophagesandpromoteslethalinfluenzaainfection