Cargando…

Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury

BACKGROUND: High-tidal-volume mechanical ventilation used in patients with acute lung injury (ALI) can induce the release of inflammatory cytokines, as macrophage inflammatory protein-2 (MIP-2), recruitment of neutrophils, and disruption of alveolar epithelial and endothelial barriers. Induced pluri...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Yung-Yang, Li, Li-Fu, Yang, Cheng-Ta, Lu, Kai-Hsi, Huang, Chung-Chi, Kao, Kuo-Chin, Chiou, Shih-Hwa
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3694116/
https://www.ncbi.nlm.nih.gov/pubmed/23840526
http://dx.doi.org/10.1371/journal.pone.0066760
_version_ 1782274813075652608
author Liu, Yung-Yang
Li, Li-Fu
Yang, Cheng-Ta
Lu, Kai-Hsi
Huang, Chung-Chi
Kao, Kuo-Chin
Chiou, Shih-Hwa
author_facet Liu, Yung-Yang
Li, Li-Fu
Yang, Cheng-Ta
Lu, Kai-Hsi
Huang, Chung-Chi
Kao, Kuo-Chin
Chiou, Shih-Hwa
author_sort Liu, Yung-Yang
collection PubMed
description BACKGROUND: High-tidal-volume mechanical ventilation used in patients with acute lung injury (ALI) can induce the release of inflammatory cytokines, as macrophage inflammatory protein-2 (MIP-2), recruitment of neutrophils, and disruption of alveolar epithelial and endothelial barriers. Induced pluripotent stem cells (iPSCs) have been shown to improve ALI in mice, but the mechanisms regulating the interactions between mechanical ventilation and iPSCs are not fully elucidated. Nuclear factor kappa B (NF-κB) and NF-κB repressing factor (NKRF) have been proposed to modulate the neutrophil activation involved in ALI. Thus, we hypothesized intravenous injection of iPSCs or iPSC-derived conditioned medium (iPSC-CM) would decrease high-tidal-volume ventilation-induced neutrophil infiltration, oxidative stress, and MIP-2 production through NF-κB/NKRF pathways. METHODS: Male C57BL/6 mice, aged between 6 and 8 weeks, weighing between 20 and 25 g, were exposed to high-tidal-volume (30 ml/kg) mechanical ventilation with room air for 1 to 4 h after 5×10(7) cells/kg mouse iPSCs or iPSC-CM administration. Nonventilated mice were used as control groups. RESULTS: High-tidal-volume mechanical ventilation induced the increases of integration of iPSCs into the injured lungs of mice, microvascular permeability, neutrophil infiltration, malondialdehyde, MIP-2 production, and NF-κB and NKRF activation. Lung injury indices including inflammation, lung edema, ultrastructure pathologic changes and functional gas exchange impairment induced by mechanical ventilation were attenuated with administration of iPSCs or iPSC-CM, which was mimicked by pharmacological inhibition of NF-κB activity with SN50 or NKRF expression with NKRF short interfering RNA. CONCLUSIONS: Our data suggest that iPSC-based therapy attenuates high-tidal-volume mechanical ventilation-induced lung injury, at least partly, through inhibition of NF-κB/NKRF pathways. Notably, the conditioned medium of iPSCs revealed beneficial effects equal to those of iPSCs.
format Online
Article
Text
id pubmed-3694116
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-36941162013-07-09 Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury Liu, Yung-Yang Li, Li-Fu Yang, Cheng-Ta Lu, Kai-Hsi Huang, Chung-Chi Kao, Kuo-Chin Chiou, Shih-Hwa PLoS One Research Article BACKGROUND: High-tidal-volume mechanical ventilation used in patients with acute lung injury (ALI) can induce the release of inflammatory cytokines, as macrophage inflammatory protein-2 (MIP-2), recruitment of neutrophils, and disruption of alveolar epithelial and endothelial barriers. Induced pluripotent stem cells (iPSCs) have been shown to improve ALI in mice, but the mechanisms regulating the interactions between mechanical ventilation and iPSCs are not fully elucidated. Nuclear factor kappa B (NF-κB) and NF-κB repressing factor (NKRF) have been proposed to modulate the neutrophil activation involved in ALI. Thus, we hypothesized intravenous injection of iPSCs or iPSC-derived conditioned medium (iPSC-CM) would decrease high-tidal-volume ventilation-induced neutrophil infiltration, oxidative stress, and MIP-2 production through NF-κB/NKRF pathways. METHODS: Male C57BL/6 mice, aged between 6 and 8 weeks, weighing between 20 and 25 g, were exposed to high-tidal-volume (30 ml/kg) mechanical ventilation with room air for 1 to 4 h after 5×10(7) cells/kg mouse iPSCs or iPSC-CM administration. Nonventilated mice were used as control groups. RESULTS: High-tidal-volume mechanical ventilation induced the increases of integration of iPSCs into the injured lungs of mice, microvascular permeability, neutrophil infiltration, malondialdehyde, MIP-2 production, and NF-κB and NKRF activation. Lung injury indices including inflammation, lung edema, ultrastructure pathologic changes and functional gas exchange impairment induced by mechanical ventilation were attenuated with administration of iPSCs or iPSC-CM, which was mimicked by pharmacological inhibition of NF-κB activity with SN50 or NKRF expression with NKRF short interfering RNA. CONCLUSIONS: Our data suggest that iPSC-based therapy attenuates high-tidal-volume mechanical ventilation-induced lung injury, at least partly, through inhibition of NF-κB/NKRF pathways. Notably, the conditioned medium of iPSCs revealed beneficial effects equal to those of iPSCs. Public Library of Science 2013-06-26 /pmc/articles/PMC3694116/ /pubmed/23840526 http://dx.doi.org/10.1371/journal.pone.0066760 Text en © 2013 Liu et al http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are properly credited.
spellingShingle Research Article
Liu, Yung-Yang
Li, Li-Fu
Yang, Cheng-Ta
Lu, Kai-Hsi
Huang, Chung-Chi
Kao, Kuo-Chin
Chiou, Shih-Hwa
Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury
title Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury
title_full Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury
title_fullStr Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury
title_full_unstemmed Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury
title_short Suppressing NF-κB and NKRF Pathways by Induced Pluripotent Stem Cell Therapy in Mice with Ventilator-Induced Lung Injury
title_sort suppressing nf-κb and nkrf pathways by induced pluripotent stem cell therapy in mice with ventilator-induced lung injury
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3694116/
https://www.ncbi.nlm.nih.gov/pubmed/23840526
http://dx.doi.org/10.1371/journal.pone.0066760
work_keys_str_mv AT liuyungyang suppressingnfkbandnkrfpathwaysbyinducedpluripotentstemcelltherapyinmicewithventilatorinducedlunginjury
AT lilifu suppressingnfkbandnkrfpathwaysbyinducedpluripotentstemcelltherapyinmicewithventilatorinducedlunginjury
AT yangchengta suppressingnfkbandnkrfpathwaysbyinducedpluripotentstemcelltherapyinmicewithventilatorinducedlunginjury
AT lukaihsi suppressingnfkbandnkrfpathwaysbyinducedpluripotentstemcelltherapyinmicewithventilatorinducedlunginjury
AT huangchungchi suppressingnfkbandnkrfpathwaysbyinducedpluripotentstemcelltherapyinmicewithventilatorinducedlunginjury
AT kaokuochin suppressingnfkbandnkrfpathwaysbyinducedpluripotentstemcelltherapyinmicewithventilatorinducedlunginjury
AT chioushihhwa suppressingnfkbandnkrfpathwaysbyinducedpluripotentstemcelltherapyinmicewithventilatorinducedlunginjury