Cargando…

Direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells

ABSTRACT: The airway epithelium represents the main barrier between inhaled air and the tissues of the respiratory tract and is therefore an important point of contact with xenobiotic substances into the human body. Several studies have recently shown that in vitro models of the airway grown at an a...

Descripción completa

Detalles Bibliográficos
Autores principales: Djidrovski, Ivo, Georgiou, Maria, Tasinato, Elena, Leonard, Martin O., Van den Bor, Jelle, Lako, Majlinda, Armstrong, Lyle
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer Netherlands 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10042770/
https://www.ncbi.nlm.nih.gov/pubmed/35641671
http://dx.doi.org/10.1007/s10565-022-09726-0
_version_ 1784913003353735168
author Djidrovski, Ivo
Georgiou, Maria
Tasinato, Elena
Leonard, Martin O.
Van den Bor, Jelle
Lako, Majlinda
Armstrong, Lyle
author_facet Djidrovski, Ivo
Georgiou, Maria
Tasinato, Elena
Leonard, Martin O.
Van den Bor, Jelle
Lako, Majlinda
Armstrong, Lyle
author_sort Djidrovski, Ivo
collection PubMed
description ABSTRACT: The airway epithelium represents the main barrier between inhaled air and the tissues of the respiratory tract and is therefore an important point of contact with xenobiotic substances into the human body. Several studies have recently shown that in vitro models of the airway grown at an air–liquid interface (ALI) can be particularly useful to obtain mechanistic information about the toxicity of chemical compounds. However, such methods are not very amenable to high throughput since the primary cells cannot be expanded indefinitely in culture to obtain a sustainable number of cells. Induced pluripotent stem cells (iPSCs) have become a popular option in the recent years for modelling the airways of the lung, but despite progress in the field, such models have so far not been assessed for their ability to metabolise xenobiotic compounds and how they compare to the primary bronchial airway model (pBAE). Here, we report a comparative analysis by TempoSeq (oligo-directed sequencing) of an iPSC-derived airway model (iBAE) with a primary bronchial airway model (pBAE). The iBAE and pBAE were differentiated at an ALI and then evaluated in a 5-compound screen with exposure to a sub-lethal concentration of each compound for 24 h. We found that despite lower expression of xenobiotic metabolism genes, the iBAE similarly predicted the toxic pathways when compared to the pBAE model. Our results show that iPSC airway models at ALI show promise for inhalation toxicity assessments with further development. GRAPHICAL ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s10565-022-09726-0.
format Online
Article
Text
id pubmed-10042770
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Springer Netherlands
record_format MEDLINE/PubMed
spelling pubmed-100427702023-03-29 Direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells Djidrovski, Ivo Georgiou, Maria Tasinato, Elena Leonard, Martin O. Van den Bor, Jelle Lako, Majlinda Armstrong, Lyle Cell Biol Toxicol Original Article ABSTRACT: The airway epithelium represents the main barrier between inhaled air and the tissues of the respiratory tract and is therefore an important point of contact with xenobiotic substances into the human body. Several studies have recently shown that in vitro models of the airway grown at an air–liquid interface (ALI) can be particularly useful to obtain mechanistic information about the toxicity of chemical compounds. However, such methods are not very amenable to high throughput since the primary cells cannot be expanded indefinitely in culture to obtain a sustainable number of cells. Induced pluripotent stem cells (iPSCs) have become a popular option in the recent years for modelling the airways of the lung, but despite progress in the field, such models have so far not been assessed for their ability to metabolise xenobiotic compounds and how they compare to the primary bronchial airway model (pBAE). Here, we report a comparative analysis by TempoSeq (oligo-directed sequencing) of an iPSC-derived airway model (iBAE) with a primary bronchial airway model (pBAE). The iBAE and pBAE were differentiated at an ALI and then evaluated in a 5-compound screen with exposure to a sub-lethal concentration of each compound for 24 h. We found that despite lower expression of xenobiotic metabolism genes, the iBAE similarly predicted the toxic pathways when compared to the pBAE model. Our results show that iPSC airway models at ALI show promise for inhalation toxicity assessments with further development. GRAPHICAL ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s10565-022-09726-0. Springer Netherlands 2022-05-31 2023 /pmc/articles/PMC10042770/ /pubmed/35641671 http://dx.doi.org/10.1007/s10565-022-09726-0 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis 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/) .
spellingShingle Original Article
Djidrovski, Ivo
Georgiou, Maria
Tasinato, Elena
Leonard, Martin O.
Van den Bor, Jelle
Lako, Majlinda
Armstrong, Lyle
Direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells
title Direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells
title_full Direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells
title_fullStr Direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells
title_full_unstemmed Direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells
title_short Direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells
title_sort direct transcriptomic comparison of xenobiotic metabolism and toxicity pathway induction of airway epithelium models at an air–liquid interface generated from induced pluripotent stem cells and primary bronchial epithelial cells
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10042770/
https://www.ncbi.nlm.nih.gov/pubmed/35641671
http://dx.doi.org/10.1007/s10565-022-09726-0
work_keys_str_mv AT djidrovskiivo directtranscriptomiccomparisonofxenobioticmetabolismandtoxicitypathwayinductionofairwayepitheliummodelsatanairliquidinterfacegeneratedfrominducedpluripotentstemcellsandprimarybronchialepithelialcells
AT georgioumaria directtranscriptomiccomparisonofxenobioticmetabolismandtoxicitypathwayinductionofairwayepitheliummodelsatanairliquidinterfacegeneratedfrominducedpluripotentstemcellsandprimarybronchialepithelialcells
AT tasinatoelena directtranscriptomiccomparisonofxenobioticmetabolismandtoxicitypathwayinductionofairwayepitheliummodelsatanairliquidinterfacegeneratedfrominducedpluripotentstemcellsandprimarybronchialepithelialcells
AT leonardmartino directtranscriptomiccomparisonofxenobioticmetabolismandtoxicitypathwayinductionofairwayepitheliummodelsatanairliquidinterfacegeneratedfrominducedpluripotentstemcellsandprimarybronchialepithelialcells
AT vandenborjelle directtranscriptomiccomparisonofxenobioticmetabolismandtoxicitypathwayinductionofairwayepitheliummodelsatanairliquidinterfacegeneratedfrominducedpluripotentstemcellsandprimarybronchialepithelialcells
AT lakomajlinda directtranscriptomiccomparisonofxenobioticmetabolismandtoxicitypathwayinductionofairwayepitheliummodelsatanairliquidinterfacegeneratedfrominducedpluripotentstemcellsandprimarybronchialepithelialcells
AT armstronglyle directtranscriptomiccomparisonofxenobioticmetabolismandtoxicitypathwayinductionofairwayepitheliummodelsatanairliquidinterfacegeneratedfrominducedpluripotentstemcellsandprimarybronchialepithelialcells