Cargando…

Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation

Preterm infants are highly susceptible to sustained lung inflammation, which may be triggered by exposure to multiple environmental cues such as supplemental oxygen (O(2)) and infections. We hypothesized that dysregulated macrophage (MФ) activation is a key feature leading to inflammation-mediated d...

Descripción completa

Detalles Bibliográficos
Autores principales: Twisselmann, Nele, Pagel, Julia, Künstner, Axel, Weckmann, Markus, Hartz, Annika, Glaser, Kirsten, Hilgendorff, Anne, Göpel, Wolfgang, Busch, Hauke, Herting, Egbert, Weinberg, Jason B., Härtel, Christoph
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8637891/
https://www.ncbi.nlm.nih.gov/pubmed/34868007
http://dx.doi.org/10.3389/fimmu.2021.762789
_version_ 1784608839235010560
author Twisselmann, Nele
Pagel, Julia
Künstner, Axel
Weckmann, Markus
Hartz, Annika
Glaser, Kirsten
Hilgendorff, Anne
Göpel, Wolfgang
Busch, Hauke
Herting, Egbert
Weinberg, Jason B.
Härtel, Christoph
author_facet Twisselmann, Nele
Pagel, Julia
Künstner, Axel
Weckmann, Markus
Hartz, Annika
Glaser, Kirsten
Hilgendorff, Anne
Göpel, Wolfgang
Busch, Hauke
Herting, Egbert
Weinberg, Jason B.
Härtel, Christoph
author_sort Twisselmann, Nele
collection PubMed
description Preterm infants are highly susceptible to sustained lung inflammation, which may be triggered by exposure to multiple environmental cues such as supplemental oxygen (O(2)) and infections. We hypothesized that dysregulated macrophage (MФ) activation is a key feature leading to inflammation-mediated development of bronchopulmonary dysplasia (BPD) in preterm infants. Therefore, we aimed to determine age-dependent differences in immune responses of monocyte-derived MФ comparing cord blood samples derived from preterm (n=14) and term (n=19) infants as well as peripheral blood samples from healthy adults (n=17) after lipopolysaccharide (LPS) exposure. Compared to term and adult MФ, LPS-stimulated preterm MФ showed an enhanced and sustained pro-inflammatory immune response determined by transcriptome analysis, cytokine release inducing a RORC upregulation due to T cell polarization of neonatal T cells, and TLR4 surface expression. In addition, a double-hit model was developed to study pulmonary relevant exposure factors by priming MФ with hyperoxia (O(2) = 65%) or hypoxia (O(2) = 3%) followed by lipopolysaccharide (LPS, 100ng/ml). When primed by 65% O(2), subsequent LPS stimulation in preterm MФ led to an exaggerated pro-inflammatory response (e.g. increased HLA-DR expression and cytokine release) compared to LPS stimulation alone. Both, exposure to 65% or 3% O(2) together with subsequent LPS stimulation, resulted in an exaggerated pro-inflammatory response of preterm MФ determined by transcriptome analysis. Downregulation of two major transcriptional factors, early growth response gene (Egr)-2 and growth factor independence 1 (Gfi1), were identified to play a role in the exaggerated pro-inflammatory response of preterm MФ to LPS insult after priming with 65% or 3% O(2). Preterm MФ responses to LPS and hyperoxia/hypoxia suggest their involvement in excessive inflammation due to age-dependent differences, potentially mediated by downregulation of Egr2 and Gfi1 in the developing lung.
format Online
Article
Text
id pubmed-8637891
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-86378912021-12-03 Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation Twisselmann, Nele Pagel, Julia Künstner, Axel Weckmann, Markus Hartz, Annika Glaser, Kirsten Hilgendorff, Anne Göpel, Wolfgang Busch, Hauke Herting, Egbert Weinberg, Jason B. Härtel, Christoph Front Immunol Immunology Preterm infants are highly susceptible to sustained lung inflammation, which may be triggered by exposure to multiple environmental cues such as supplemental oxygen (O(2)) and infections. We hypothesized that dysregulated macrophage (MФ) activation is a key feature leading to inflammation-mediated development of bronchopulmonary dysplasia (BPD) in preterm infants. Therefore, we aimed to determine age-dependent differences in immune responses of monocyte-derived MФ comparing cord blood samples derived from preterm (n=14) and term (n=19) infants as well as peripheral blood samples from healthy adults (n=17) after lipopolysaccharide (LPS) exposure. Compared to term and adult MФ, LPS-stimulated preterm MФ showed an enhanced and sustained pro-inflammatory immune response determined by transcriptome analysis, cytokine release inducing a RORC upregulation due to T cell polarization of neonatal T cells, and TLR4 surface expression. In addition, a double-hit model was developed to study pulmonary relevant exposure factors by priming MФ with hyperoxia (O(2) = 65%) or hypoxia (O(2) = 3%) followed by lipopolysaccharide (LPS, 100ng/ml). When primed by 65% O(2), subsequent LPS stimulation in preterm MФ led to an exaggerated pro-inflammatory response (e.g. increased HLA-DR expression and cytokine release) compared to LPS stimulation alone. Both, exposure to 65% or 3% O(2) together with subsequent LPS stimulation, resulted in an exaggerated pro-inflammatory response of preterm MФ determined by transcriptome analysis. Downregulation of two major transcriptional factors, early growth response gene (Egr)-2 and growth factor independence 1 (Gfi1), were identified to play a role in the exaggerated pro-inflammatory response of preterm MФ to LPS insult after priming with 65% or 3% O(2). Preterm MФ responses to LPS and hyperoxia/hypoxia suggest their involvement in excessive inflammation due to age-dependent differences, potentially mediated by downregulation of Egr2 and Gfi1 in the developing lung. Frontiers Media S.A. 2021-11-18 /pmc/articles/PMC8637891/ /pubmed/34868007 http://dx.doi.org/10.3389/fimmu.2021.762789 Text en Copyright © 2021 Twisselmann, Pagel, Künstner, Weckmann, Hartz, Glaser, Hilgendorff, Göpel, Busch, Herting, Weinberg and Härtel 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 Immunology
Twisselmann, Nele
Pagel, Julia
Künstner, Axel
Weckmann, Markus
Hartz, Annika
Glaser, Kirsten
Hilgendorff, Anne
Göpel, Wolfgang
Busch, Hauke
Herting, Egbert
Weinberg, Jason B.
Härtel, Christoph
Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation
title Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation
title_full Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation
title_fullStr Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation
title_full_unstemmed Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation
title_short Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation
title_sort hyperoxia/hypoxia exposure primes a sustained pro-inflammatory profile of preterm infant macrophages upon lps stimulation
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8637891/
https://www.ncbi.nlm.nih.gov/pubmed/34868007
http://dx.doi.org/10.3389/fimmu.2021.762789
work_keys_str_mv AT twisselmannnele hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT pageljulia hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT kunstneraxel hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT weckmannmarkus hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT hartzannika hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT glaserkirsten hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT hilgendorffanne hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT gopelwolfgang hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT buschhauke hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT hertingegbert hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT weinbergjasonb hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation
AT hartelchristoph hyperoxiahypoxiaexposureprimesasustainedproinflammatoryprofileofpreterminfantmacrophagesuponlpsstimulation