Cargando…

Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells

BACKGROUND: Mitochondrial transcription factor A (TFAM) is associated with a number of neurodegenerative diseases and also with asthma. TFAM deficiency-induced mitochondrial DNA stress primes the antiviral innate immune response in mouse embryonic fibroblasts. However, the role of TFAM in asthma rel...

Descripción completa

Detalles Bibliográficos
Autores principales: Luo, Jinsong, Liu, Hong, Li, Daniel K. Jun, Song, Bin, Zhang, Yi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8650232/
https://www.ncbi.nlm.nih.gov/pubmed/34876009
http://dx.doi.org/10.1186/s12865-021-00464-2
_version_ 1784611157200338944
author Luo, Jinsong
Liu, Hong
Li, Daniel K. Jun
Song, Bin
Zhang, Yi
author_facet Luo, Jinsong
Liu, Hong
Li, Daniel K. Jun
Song, Bin
Zhang, Yi
author_sort Luo, Jinsong
collection PubMed
description BACKGROUND: Mitochondrial transcription factor A (TFAM) is associated with a number of neurodegenerative diseases and also with asthma. TFAM deficiency-induced mitochondrial DNA stress primes the antiviral innate immune response in mouse embryonic fibroblasts. However, the role of TFAM in asthma related inflammation remains obscure. The purpose of this study was to investigate the regulatory mechanism of TFAM in asthma. RESULTS: In this study, we overexpressed TFAM in human lung epithelial cells (A549), then obtained the TFAM-regulated transcriptome by Illumina sequencing technology. Transcriptome analysis revealed that TFAM overexpression down-regulated and up-regulated the expression of 642 and 169 differentially expressed genes (DEGs), respectively. The TFAM-repressed genes were strongly enriched in cytokine-mediated signaling pathway, type I interferon- and INF-γ-mediated signaling pathways, and viral response pathways. We also revealed that 2563 alternative splicing events in 1796 alternative splicing genes (ASGs) were de-regulated upon TFAM overexpression. These TFAM-responding ASGs were enriched in DNA repair, nerve growth factor receptor signaling pathway, and also transcription regulation. Further analysis revealed that the promoters of TFAM-repressed DEGs were enriched by DNA binding motifs of transcription factors whose alternative splicing was regulated by TFAM. CONCLUSIONS: These findings suggest that TFAM regulates not only immune response gene expression in human lung epithelial cells, but also pre-mRNA alternative splicing which may mediate transcriptional regulation; this TFAM-centered gene regulation network could be targeted in developing therapies against various diseases. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12865-021-00464-2.
format Online
Article
Text
id pubmed-8650232
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-86502322021-12-07 Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells Luo, Jinsong Liu, Hong Li, Daniel K. Jun Song, Bin Zhang, Yi BMC Immunol Research BACKGROUND: Mitochondrial transcription factor A (TFAM) is associated with a number of neurodegenerative diseases and also with asthma. TFAM deficiency-induced mitochondrial DNA stress primes the antiviral innate immune response in mouse embryonic fibroblasts. However, the role of TFAM in asthma related inflammation remains obscure. The purpose of this study was to investigate the regulatory mechanism of TFAM in asthma. RESULTS: In this study, we overexpressed TFAM in human lung epithelial cells (A549), then obtained the TFAM-regulated transcriptome by Illumina sequencing technology. Transcriptome analysis revealed that TFAM overexpression down-regulated and up-regulated the expression of 642 and 169 differentially expressed genes (DEGs), respectively. The TFAM-repressed genes were strongly enriched in cytokine-mediated signaling pathway, type I interferon- and INF-γ-mediated signaling pathways, and viral response pathways. We also revealed that 2563 alternative splicing events in 1796 alternative splicing genes (ASGs) were de-regulated upon TFAM overexpression. These TFAM-responding ASGs were enriched in DNA repair, nerve growth factor receptor signaling pathway, and also transcription regulation. Further analysis revealed that the promoters of TFAM-repressed DEGs were enriched by DNA binding motifs of transcription factors whose alternative splicing was regulated by TFAM. CONCLUSIONS: These findings suggest that TFAM regulates not only immune response gene expression in human lung epithelial cells, but also pre-mRNA alternative splicing which may mediate transcriptional regulation; this TFAM-centered gene regulation network could be targeted in developing therapies against various diseases. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12865-021-00464-2. BioMed Central 2021-12-07 /pmc/articles/PMC8650232/ /pubmed/34876009 http://dx.doi.org/10.1186/s12865-021-00464-2 Text en © The Author(s) 2021 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
Luo, Jinsong
Liu, Hong
Li, Daniel K. Jun
Song, Bin
Zhang, Yi
Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells
title Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells
title_full Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells
title_fullStr Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells
title_full_unstemmed Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells
title_short Repression of the expression of proinflammatory genes by mitochondrial transcription factor A is linked to its alternative splicing regulation in human lung epithelial cells
title_sort repression of the expression of proinflammatory genes by mitochondrial transcription factor a is linked to its alternative splicing regulation in human lung epithelial cells
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8650232/
https://www.ncbi.nlm.nih.gov/pubmed/34876009
http://dx.doi.org/10.1186/s12865-021-00464-2
work_keys_str_mv AT luojinsong repressionoftheexpressionofproinflammatorygenesbymitochondrialtranscriptionfactoraislinkedtoitsalternativesplicingregulationinhumanlungepithelialcells
AT liuhong repressionoftheexpressionofproinflammatorygenesbymitochondrialtranscriptionfactoraislinkedtoitsalternativesplicingregulationinhumanlungepithelialcells
AT lidanielkjun repressionoftheexpressionofproinflammatorygenesbymitochondrialtranscriptionfactoraislinkedtoitsalternativesplicingregulationinhumanlungepithelialcells
AT songbin repressionoftheexpressionofproinflammatorygenesbymitochondrialtranscriptionfactoraislinkedtoitsalternativesplicingregulationinhumanlungepithelialcells
AT zhangyi repressionoftheexpressionofproinflammatorygenesbymitochondrialtranscriptionfactoraislinkedtoitsalternativesplicingregulationinhumanlungepithelialcells