IFN-γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injury

Acute Respiratory Distress Syndrome (ARDS) is an inflammatory disease that is associated with high mortality but no specific treatment. Our understanding of initial events that trigger ARDS pathogenesis is limited. We have developed a mouse model of inflammatory lung injury by influenza and methicil...

Full description

Bibliographic Details
Main Authors: Atul K. Verma, Michael McKelvey, Md Bashir Uddin, Sunil Palani, Meng Niu, Christopher Bauer, Shengjun Shao, Keer Sun
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-09-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fimmu.2022.1011132/full
_version_ 1798035834490847232
author Atul K. Verma
Michael McKelvey
Md Bashir Uddin
Sunil Palani
Meng Niu
Christopher Bauer
Shengjun Shao
Keer Sun
Keer Sun
author_facet Atul K. Verma
Michael McKelvey
Md Bashir Uddin
Sunil Palani
Meng Niu
Christopher Bauer
Shengjun Shao
Keer Sun
Keer Sun
author_sort Atul K. Verma
collection DOAJ
description Acute Respiratory Distress Syndrome (ARDS) is an inflammatory disease that is associated with high mortality but no specific treatment. Our understanding of initial events that trigger ARDS pathogenesis is limited. We have developed a mouse model of inflammatory lung injury by influenza and methicillin-resistant Staphylococcus aureus (MRSA) coinfection plus daily antibiotic therapy. Using this pneumonic ARDS model, here we show that IFN-γ receptor signaling drives inflammatory cytokine storm and lung tissue damage. By single-cell RNA sequencing (scRNA-seq) analysis, we demonstrate that IFN-γ signaling induces a transcriptional shift in airway immune cells, particularly by upregulating macrophage and monocyte expression of genes associated with inflammatory diseases. Further evidence from conditional knockout mouse models reveals that IFN-γ receptor signaling in myeloid cells, particularly CD11c+ mononuclear phagocytes, directly promotes TNF-α hyperproduction and inflammatory lung damage. Collectively, the findings from this study, ranging from cell-intrinsic gene expression to overall disease outcome, demonstrate that influenza-induced IFN-γ triggers myeloid cell hyperresponsiveness to MRSA, thereby leading to excessive inflammatory response and lethal lung damage during coinfection.
first_indexed 2024-04-11T21:03:47Z
format Article
id doaj.art-4044e6f6530743088bf07fde8eb116e1
institution Directory Open Access Journal
issn 1664-3224
language English
last_indexed 2024-04-11T21:03:47Z
publishDate 2022-09-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Immunology
spelling doaj.art-4044e6f6530743088bf07fde8eb116e12022-12-22T04:03:24ZengFrontiers Media S.A.Frontiers in Immunology1664-32242022-09-011310.3389/fimmu.2022.10111321011132IFN-γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injuryAtul K. Verma0Michael McKelvey1Md Bashir Uddin2Sunil Palani3Meng Niu4Christopher Bauer5Shengjun Shao6Keer Sun7Keer Sun8Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United StatesDepartment of Experimental Pathology, University of Texas Medical Branch, Galveston, TX, United StatesDepartment of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United StatesDepartment of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United StatesDepartment of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, Omaha, NE, United StatesDepartment of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United StatesDepartment of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United StatesDepartment of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United StatesDepartment of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United StatesAcute Respiratory Distress Syndrome (ARDS) is an inflammatory disease that is associated with high mortality but no specific treatment. Our understanding of initial events that trigger ARDS pathogenesis is limited. We have developed a mouse model of inflammatory lung injury by influenza and methicillin-resistant Staphylococcus aureus (MRSA) coinfection plus daily antibiotic therapy. Using this pneumonic ARDS model, here we show that IFN-γ receptor signaling drives inflammatory cytokine storm and lung tissue damage. By single-cell RNA sequencing (scRNA-seq) analysis, we demonstrate that IFN-γ signaling induces a transcriptional shift in airway immune cells, particularly by upregulating macrophage and monocyte expression of genes associated with inflammatory diseases. Further evidence from conditional knockout mouse models reveals that IFN-γ receptor signaling in myeloid cells, particularly CD11c+ mononuclear phagocytes, directly promotes TNF-α hyperproduction and inflammatory lung damage. Collectively, the findings from this study, ranging from cell-intrinsic gene expression to overall disease outcome, demonstrate that influenza-induced IFN-γ triggers myeloid cell hyperresponsiveness to MRSA, thereby leading to excessive inflammatory response and lethal lung damage during coinfection.https://www.frontiersin.org/articles/10.3389/fimmu.2022.1011132/fullAcute lung injurycytokine storminfluenza A virusmethicillin-resistant Staphylococcus aureuscoinfection
spellingShingle Atul K. Verma
Michael McKelvey
Md Bashir Uddin
Sunil Palani
Meng Niu
Christopher Bauer
Shengjun Shao
Keer Sun
Keer Sun
IFN-γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injury
Frontiers in Immunology
Acute lung injury
cytokine storm
influenza A virus
methicillin-resistant Staphylococcus aureus
coinfection
title IFN-γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injury
title_full IFN-γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injury
title_fullStr IFN-γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injury
title_full_unstemmed IFN-γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injury
title_short IFN-γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injury
title_sort ifn γ transforms the transcriptomic landscape and triggers myeloid cell hyperresponsiveness to cause lethal lung injury
topic Acute lung injury
cytokine storm
influenza A virus
methicillin-resistant Staphylococcus aureus
coinfection
url https://www.frontiersin.org/articles/10.3389/fimmu.2022.1011132/full
work_keys_str_mv AT atulkverma ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury
AT michaelmckelvey ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury
AT mdbashiruddin ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury
AT sunilpalani ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury
AT mengniu ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury
AT christopherbauer ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury
AT shengjunshao ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury
AT keersun ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury
AT keersun ifngtransformsthetranscriptomiclandscapeandtriggersmyeloidcellhyperresponsivenesstocauselethallunginjury