Cargando…

Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes

BACKGROUND: Fetal hypoxia causes vital, systemic, developmental malformations in the fetus, particularly in the brain, and increases the risk of diseases in later life. We previously demonstrated that fetal hypoxia exposure increases the susceptibility of the neonatal brain to hypoxic-ischemic insul...

Descripción completa

Detalles Bibliográficos
Autores principales: Kremsky, Isaac, Ma, Qingyi, Li, Bo, Dasgupta, Chiranjib, Chen, Xin, Ali, Samir, Angeloni, Shawnee, Wang, Charles, Zhang, Lubo
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10022003/
https://www.ncbi.nlm.nih.gov/pubmed/36932456
http://dx.doi.org/10.1186/s13578-023-01012-8
_version_ 1784908631851925504
author Kremsky, Isaac
Ma, Qingyi
Li, Bo
Dasgupta, Chiranjib
Chen, Xin
Ali, Samir
Angeloni, Shawnee
Wang, Charles
Zhang, Lubo
author_facet Kremsky, Isaac
Ma, Qingyi
Li, Bo
Dasgupta, Chiranjib
Chen, Xin
Ali, Samir
Angeloni, Shawnee
Wang, Charles
Zhang, Lubo
author_sort Kremsky, Isaac
collection PubMed
description BACKGROUND: Fetal hypoxia causes vital, systemic, developmental malformations in the fetus, particularly in the brain, and increases the risk of diseases in later life. We previously demonstrated that fetal hypoxia exposure increases the susceptibility of the neonatal brain to hypoxic-ischemic insult. Herein, we investigate the effect of fetal hypoxia on programming of cell-specific transcriptomes in the brain of neonatal rats. RESULTS: We obtained RNA sequencing (RNA-seq) data from neurons, microglia, oligodendrocytes, A2B5(+) oligodendrocyte precursor cells, and astrocytes from male and female neonatal rats subjected either to fetal hypoxia or control conditions. Substantial transcriptomic responses to fetal hypoxia occurred in neurons, microglia, oligodendrocytes, and A2B5(+) cells. Not only were the transcriptomic responses unique to each cell type, but they also occurred with a great deal of sexual dimorphism. We validated differential expression of several genes related to inflammation and cell death by Real-time Quantitative Polymerase Chain Reaction (qRT-PCR). Pathway and transcription factor motif analyses suggested that the NF-kappa B (NFκB) signaling pathway was enriched in the neonatal male brain due to fetal hypoxia, and we verified this result by transcription factor assay of NFκB-p65 in whole brain. CONCLUSIONS: Our study reveals a significant impact of fetal hypoxia on the transcriptomes of neonatal brains in a cell-specific and sex-dependent manner, and provides mechanistic insights that may help explain the development of hypoxic-ischemic sensitive phenotypes in the neonatal brain. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13578-023-01012-8.
format Online
Article
Text
id pubmed-10022003
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-100220032023-03-18 Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes Kremsky, Isaac Ma, Qingyi Li, Bo Dasgupta, Chiranjib Chen, Xin Ali, Samir Angeloni, Shawnee Wang, Charles Zhang, Lubo Cell Biosci Research BACKGROUND: Fetal hypoxia causes vital, systemic, developmental malformations in the fetus, particularly in the brain, and increases the risk of diseases in later life. We previously demonstrated that fetal hypoxia exposure increases the susceptibility of the neonatal brain to hypoxic-ischemic insult. Herein, we investigate the effect of fetal hypoxia on programming of cell-specific transcriptomes in the brain of neonatal rats. RESULTS: We obtained RNA sequencing (RNA-seq) data from neurons, microglia, oligodendrocytes, A2B5(+) oligodendrocyte precursor cells, and astrocytes from male and female neonatal rats subjected either to fetal hypoxia or control conditions. Substantial transcriptomic responses to fetal hypoxia occurred in neurons, microglia, oligodendrocytes, and A2B5(+) cells. Not only were the transcriptomic responses unique to each cell type, but they also occurred with a great deal of sexual dimorphism. We validated differential expression of several genes related to inflammation and cell death by Real-time Quantitative Polymerase Chain Reaction (qRT-PCR). Pathway and transcription factor motif analyses suggested that the NF-kappa B (NFκB) signaling pathway was enriched in the neonatal male brain due to fetal hypoxia, and we verified this result by transcription factor assay of NFκB-p65 in whole brain. CONCLUSIONS: Our study reveals a significant impact of fetal hypoxia on the transcriptomes of neonatal brains in a cell-specific and sex-dependent manner, and provides mechanistic insights that may help explain the development of hypoxic-ischemic sensitive phenotypes in the neonatal brain. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13578-023-01012-8. BioMed Central 2023-03-17 /pmc/articles/PMC10022003/ /pubmed/36932456 http://dx.doi.org/10.1186/s13578-023-01012-8 Text en © The Author(s) 2023 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/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Kremsky, Isaac
Ma, Qingyi
Li, Bo
Dasgupta, Chiranjib
Chen, Xin
Ali, Samir
Angeloni, Shawnee
Wang, Charles
Zhang, Lubo
Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes
title Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes
title_full Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes
title_fullStr Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes
title_full_unstemmed Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes
title_short Fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes
title_sort fetal hypoxia results in sex- and cell type-specific alterations in neonatal transcription in rat oligodendrocyte precursor cells, microglia, neurons, and oligodendrocytes
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10022003/
https://www.ncbi.nlm.nih.gov/pubmed/36932456
http://dx.doi.org/10.1186/s13578-023-01012-8
work_keys_str_mv AT kremskyisaac fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes
AT maqingyi fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes
AT libo fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes
AT dasguptachiranjib fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes
AT chenxin fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes
AT alisamir fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes
AT angelonishawnee fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes
AT wangcharles fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes
AT zhanglubo fetalhypoxiaresultsinsexandcelltypespecificalterationsinneonataltranscriptioninratoligodendrocyteprecursorcellsmicroglianeuronsandoligodendrocytes