Cargando…

Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection

BACKGROUND: Chronic obstructive pulmonary disease (COPD) collectively refers to chronic and progressive lung diseases that cause irreversible limitations in airflow. Patients with COPD are at high risk for severe respiratory symptoms upon influenza virus infection. Airway epithelial cells provide th...

Descripción completa

Detalles Bibliográficos
Autores principales: Nakayama, Misako, Marchi, Hannah, Dmitrieva, Anna M., Chakraborty, Ashesh, Merl-Pham, Juliane, Hennen, Elisabeth, Le Gleut, Ronan, Ruppert, Clemens, Guenther, Andreas, Kahnert, Kathrin, Behr, Jürgen, Hilgendorff, Anne, Hauck, Stefanie M., Adler, Heiko, Staab-Weijnitz, Claudia A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9875134/
https://www.ncbi.nlm.nih.gov/pubmed/36713229
http://dx.doi.org/10.3389/fmicb.2022.957830
_version_ 1784877895239335936
author Nakayama, Misako
Marchi, Hannah
Dmitrieva, Anna M.
Chakraborty, Ashesh
Merl-Pham, Juliane
Hennen, Elisabeth
Le Gleut, Ronan
Ruppert, Clemens
Guenther, Andreas
Kahnert, Kathrin
Behr, Jürgen
Hilgendorff, Anne
Hauck, Stefanie M.
Adler, Heiko
Staab-Weijnitz, Claudia A.
author_facet Nakayama, Misako
Marchi, Hannah
Dmitrieva, Anna M.
Chakraborty, Ashesh
Merl-Pham, Juliane
Hennen, Elisabeth
Le Gleut, Ronan
Ruppert, Clemens
Guenther, Andreas
Kahnert, Kathrin
Behr, Jürgen
Hilgendorff, Anne
Hauck, Stefanie M.
Adler, Heiko
Staab-Weijnitz, Claudia A.
author_sort Nakayama, Misako
collection PubMed
description BACKGROUND: Chronic obstructive pulmonary disease (COPD) collectively refers to chronic and progressive lung diseases that cause irreversible limitations in airflow. Patients with COPD are at high risk for severe respiratory symptoms upon influenza virus infection. Airway epithelial cells provide the first-line antiviral defense, but whether or not their susceptibility and response to influenza virus infection changes in COPD have not been elucidated. Therefore, this study aimed to compare the susceptibility of COPD- and control-derived airway epithelium to the influenza virus and assess protein changes during influenza virus infection by quantitative proteomics. MATERIALS AND METHODS: The presence of human- and avian-type influenza A virus receptor was assessed in control and COPD lung sections as well as in fully differentiated primary human bronchial epithelial cells (phBECs) by lectin- or antibody-based histochemical staining. PhBECs were from COPD lungs, including cells from moderate- and severe-stage diseases, and from age-, sex-, smoking, and history-matched control lung specimens. Protein profiles pre- and post-influenza virus infection in vitro were directly compared using quantitative proteomics, and selected findings were validated by qRT-PCR and immunoblotting. RESULTS: The human-type influenza receptor was more abundant in human airways than the avian-type influenza receptor, a property that was retained in vitro when differentiating phBECs at the air–liquid interface. Proteomics of phBECs pre- and post-influenza A virus infection with A/Puerto Rico/8/34 (PR8) revealed no significant differences between COPD and control phBECs in terms of flu receptor expression, cell type composition, virus replication, or protein profile pre- and post-infection. Independent of health state, a robust antiviral response to influenza virus infection was observed, as well as upregulation of several novel influenza virus-regulated proteins, including PLSCR1, HLA-F, CMTR1, DTX3L, and SHFL. CONCLUSION: COPD- and control-derived phBECs did not differ in cell type composition, susceptibility to influenza virus infection, and proteomes pre- and post-infection. Finally, we identified novel influenza A virus-regulated proteins in bronchial epithelial cells that might serve as potential targets to modulate the pathogenicity of infection and acute exacerbations.
format Online
Article
Text
id pubmed-9875134
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-98751342023-01-26 Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection Nakayama, Misako Marchi, Hannah Dmitrieva, Anna M. Chakraborty, Ashesh Merl-Pham, Juliane Hennen, Elisabeth Le Gleut, Ronan Ruppert, Clemens Guenther, Andreas Kahnert, Kathrin Behr, Jürgen Hilgendorff, Anne Hauck, Stefanie M. Adler, Heiko Staab-Weijnitz, Claudia A. Front Microbiol Microbiology BACKGROUND: Chronic obstructive pulmonary disease (COPD) collectively refers to chronic and progressive lung diseases that cause irreversible limitations in airflow. Patients with COPD are at high risk for severe respiratory symptoms upon influenza virus infection. Airway epithelial cells provide the first-line antiviral defense, but whether or not their susceptibility and response to influenza virus infection changes in COPD have not been elucidated. Therefore, this study aimed to compare the susceptibility of COPD- and control-derived airway epithelium to the influenza virus and assess protein changes during influenza virus infection by quantitative proteomics. MATERIALS AND METHODS: The presence of human- and avian-type influenza A virus receptor was assessed in control and COPD lung sections as well as in fully differentiated primary human bronchial epithelial cells (phBECs) by lectin- or antibody-based histochemical staining. PhBECs were from COPD lungs, including cells from moderate- and severe-stage diseases, and from age-, sex-, smoking, and history-matched control lung specimens. Protein profiles pre- and post-influenza virus infection in vitro were directly compared using quantitative proteomics, and selected findings were validated by qRT-PCR and immunoblotting. RESULTS: The human-type influenza receptor was more abundant in human airways than the avian-type influenza receptor, a property that was retained in vitro when differentiating phBECs at the air–liquid interface. Proteomics of phBECs pre- and post-influenza A virus infection with A/Puerto Rico/8/34 (PR8) revealed no significant differences between COPD and control phBECs in terms of flu receptor expression, cell type composition, virus replication, or protein profile pre- and post-infection. Independent of health state, a robust antiviral response to influenza virus infection was observed, as well as upregulation of several novel influenza virus-regulated proteins, including PLSCR1, HLA-F, CMTR1, DTX3L, and SHFL. CONCLUSION: COPD- and control-derived phBECs did not differ in cell type composition, susceptibility to influenza virus infection, and proteomes pre- and post-infection. Finally, we identified novel influenza A virus-regulated proteins in bronchial epithelial cells that might serve as potential targets to modulate the pathogenicity of infection and acute exacerbations. Frontiers Media S.A. 2023-01-11 /pmc/articles/PMC9875134/ /pubmed/36713229 http://dx.doi.org/10.3389/fmicb.2022.957830 Text en Copyright © 2023 Nakayama, Marchi, Dmitrieva, Chakraborty, Merl-Pham, Hennen, Le Gleut, Ruppert, Guenther, Kahnert, Behr, Hilgendorff, Hauck, Adler and Staab-Weijnitz. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Microbiology
Nakayama, Misako
Marchi, Hannah
Dmitrieva, Anna M.
Chakraborty, Ashesh
Merl-Pham, Juliane
Hennen, Elisabeth
Le Gleut, Ronan
Ruppert, Clemens
Guenther, Andreas
Kahnert, Kathrin
Behr, Jürgen
Hilgendorff, Anne
Hauck, Stefanie M.
Adler, Heiko
Staab-Weijnitz, Claudia A.
Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection
title Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection
title_full Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection
title_fullStr Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection
title_full_unstemmed Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection
title_short Quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza A virus infection
title_sort quantitative proteomics of differentiated primary bronchial epithelial cells from chronic obstructive pulmonary disease and control identifies potential novel host factors post-influenza a virus infection
topic Microbiology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9875134/
https://www.ncbi.nlm.nih.gov/pubmed/36713229
http://dx.doi.org/10.3389/fmicb.2022.957830
work_keys_str_mv AT nakayamamisako quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT marchihannah quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT dmitrievaannam quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT chakrabortyashesh quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT merlphamjuliane quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT hennenelisabeth quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT legleutronan quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT ruppertclemens quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT guentherandreas quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT kahnertkathrin quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT behrjurgen quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT hilgendorffanne quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT hauckstefaniem quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT adlerheiko quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection
AT staabweijnitzclaudiaa quantitativeproteomicsofdifferentiatedprimarybronchialepithelialcellsfromchronicobstructivepulmonarydiseaseandcontrolidentifiespotentialnovelhostfactorspostinfluenzaavirusinfection