Cargando…

Global Transcriptomics Uncovers Distinct Contributions From Splicing Regulatory Proteins to the Macrophage Innate Immune Response

Pathogen sensing via pattern recognition receptors triggers massive reprogramming of macrophage gene expression. While the signaling cascades and transcription factors that activate these responses are well-known, the role of post-transcriptional RNA processing in modulating innate immune gene expre...

Descripción completa

Detalles Bibliográficos
Autores principales: Wagner, Allison R., Scott, Haley M., West, Kelsi O., Vail, Krystal J., Fitzsimons, Timothy C., Coleman, Aja K., Carter, Kaitlyn E., Watson, Robert O., Patrick, Kristin L.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8299563/
https://www.ncbi.nlm.nih.gov/pubmed/34305890
http://dx.doi.org/10.3389/fimmu.2021.656885
_version_ 1783726294473637888
author Wagner, Allison R.
Scott, Haley M.
West, Kelsi O.
Vail, Krystal J.
Fitzsimons, Timothy C.
Coleman, Aja K.
Carter, Kaitlyn E.
Watson, Robert O.
Patrick, Kristin L.
author_facet Wagner, Allison R.
Scott, Haley M.
West, Kelsi O.
Vail, Krystal J.
Fitzsimons, Timothy C.
Coleman, Aja K.
Carter, Kaitlyn E.
Watson, Robert O.
Patrick, Kristin L.
author_sort Wagner, Allison R.
collection PubMed
description Pathogen sensing via pattern recognition receptors triggers massive reprogramming of macrophage gene expression. While the signaling cascades and transcription factors that activate these responses are well-known, the role of post-transcriptional RNA processing in modulating innate immune gene expression remains understudied. Given their crucial role in regulating pre-mRNA splicing and other RNA processing steps, we hypothesized that members of the SR/hnRNP protein families regulate innate immune gene expression in distinct ways. We analyzed steady state gene expression and alternatively spliced isoform production in ten SR/hnRNP knockdown RAW 264.7 macrophage-like cell lines following infection with the bacterial pathogen Salmonella enterica serovar Typhimurium (Salmonella). We identified thousands of transcripts whose abundance is increased or decreased by SR/hnRNP knockdown in macrophages. Notably, we observed that SR and hnRNP proteins influence expression of different genes in uninfected versus Salmonella-infected macrophages, suggesting functionalization of these proteins upon pathogen sensing. Likewise, we found that knockdown of SR/hnRNPs promoted differential isoform usage (DIU) for thousands of macrophage transcripts and that these alternative splicing changes were distinct in uninfected and Salmonella-infected macrophages. Finally, having observed a surprising degree of similarity between the differentially expressed genes (DEGs) and DIUs in hnRNP K and U knockdown macrophages, we found that hnRNP K and U knockdown macrophages are both more restrictive to Vesicular Stomatitis Virus (VSV), while hnRNP K knockdown macrophages are more permissive to Salmonella Typhimurium. Based on these findings, we conclude that many innate immune genes evolved to rely on one or more SR/hnRNPs to ensure the proper magnitude of their induction, supporting a model wherein pre-mRNA splicing is critical for regulating innate immune gene expression and controlling infection outcomes in macrophages ex vivo.
format Online
Article
Text
id pubmed-8299563
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-82995632021-07-24 Global Transcriptomics Uncovers Distinct Contributions From Splicing Regulatory Proteins to the Macrophage Innate Immune Response Wagner, Allison R. Scott, Haley M. West, Kelsi O. Vail, Krystal J. Fitzsimons, Timothy C. Coleman, Aja K. Carter, Kaitlyn E. Watson, Robert O. Patrick, Kristin L. Front Immunol Immunology Pathogen sensing via pattern recognition receptors triggers massive reprogramming of macrophage gene expression. While the signaling cascades and transcription factors that activate these responses are well-known, the role of post-transcriptional RNA processing in modulating innate immune gene expression remains understudied. Given their crucial role in regulating pre-mRNA splicing and other RNA processing steps, we hypothesized that members of the SR/hnRNP protein families regulate innate immune gene expression in distinct ways. We analyzed steady state gene expression and alternatively spliced isoform production in ten SR/hnRNP knockdown RAW 264.7 macrophage-like cell lines following infection with the bacterial pathogen Salmonella enterica serovar Typhimurium (Salmonella). We identified thousands of transcripts whose abundance is increased or decreased by SR/hnRNP knockdown in macrophages. Notably, we observed that SR and hnRNP proteins influence expression of different genes in uninfected versus Salmonella-infected macrophages, suggesting functionalization of these proteins upon pathogen sensing. Likewise, we found that knockdown of SR/hnRNPs promoted differential isoform usage (DIU) for thousands of macrophage transcripts and that these alternative splicing changes were distinct in uninfected and Salmonella-infected macrophages. Finally, having observed a surprising degree of similarity between the differentially expressed genes (DEGs) and DIUs in hnRNP K and U knockdown macrophages, we found that hnRNP K and U knockdown macrophages are both more restrictive to Vesicular Stomatitis Virus (VSV), while hnRNP K knockdown macrophages are more permissive to Salmonella Typhimurium. Based on these findings, we conclude that many innate immune genes evolved to rely on one or more SR/hnRNPs to ensure the proper magnitude of their induction, supporting a model wherein pre-mRNA splicing is critical for regulating innate immune gene expression and controlling infection outcomes in macrophages ex vivo. Frontiers Media S.A. 2021-07-09 /pmc/articles/PMC8299563/ /pubmed/34305890 http://dx.doi.org/10.3389/fimmu.2021.656885 Text en Copyright © 2021 Wagner, Scott, West, Vail, Fitzsimons, Coleman, Carter, Watson and Patrick 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
Wagner, Allison R.
Scott, Haley M.
West, Kelsi O.
Vail, Krystal J.
Fitzsimons, Timothy C.
Coleman, Aja K.
Carter, Kaitlyn E.
Watson, Robert O.
Patrick, Kristin L.
Global Transcriptomics Uncovers Distinct Contributions From Splicing Regulatory Proteins to the Macrophage Innate Immune Response
title Global Transcriptomics Uncovers Distinct Contributions From Splicing Regulatory Proteins to the Macrophage Innate Immune Response
title_full Global Transcriptomics Uncovers Distinct Contributions From Splicing Regulatory Proteins to the Macrophage Innate Immune Response
title_fullStr Global Transcriptomics Uncovers Distinct Contributions From Splicing Regulatory Proteins to the Macrophage Innate Immune Response
title_full_unstemmed Global Transcriptomics Uncovers Distinct Contributions From Splicing Regulatory Proteins to the Macrophage Innate Immune Response
title_short Global Transcriptomics Uncovers Distinct Contributions From Splicing Regulatory Proteins to the Macrophage Innate Immune Response
title_sort global transcriptomics uncovers distinct contributions from splicing regulatory proteins to the macrophage innate immune response
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8299563/
https://www.ncbi.nlm.nih.gov/pubmed/34305890
http://dx.doi.org/10.3389/fimmu.2021.656885
work_keys_str_mv AT wagnerallisonr globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse
AT scotthaleym globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse
AT westkelsio globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse
AT vailkrystalj globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse
AT fitzsimonstimothyc globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse
AT colemanajak globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse
AT carterkaitlyne globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse
AT watsonroberto globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse
AT patrickkristinl globaltranscriptomicsuncoversdistinctcontributionsfromsplicingregulatoryproteinstothemacrophageinnateimmuneresponse