TNF licenses macrophages to undergo rapid caspase-1, -11, and -8-mediated cell death that restricts Legionella pneumophila infection.

The inflammatory cytokine tumor necrosis factor (TNF) is necessary for host defense against many intracellular pathogens, including Legionella pneumophila. Legionella causes the severe pneumonia Legionnaires' disease and predominantly affects individuals with a suppressed immune system, includi...

Full description

Bibliographic Details
Main Authors: Tzvi Y Pollock, Víctor R Vázquez Marrero, Igor E Brodsky, Sunny Shin
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2023-06-01
Series:PLoS Pathogens
Online Access:https://doi.org/10.1371/journal.ppat.1010767
_version_ 1797797842878726144
author Tzvi Y Pollock
Víctor R Vázquez Marrero
Igor E Brodsky
Sunny Shin
author_facet Tzvi Y Pollock
Víctor R Vázquez Marrero
Igor E Brodsky
Sunny Shin
author_sort Tzvi Y Pollock
collection DOAJ
description The inflammatory cytokine tumor necrosis factor (TNF) is necessary for host defense against many intracellular pathogens, including Legionella pneumophila. Legionella causes the severe pneumonia Legionnaires' disease and predominantly affects individuals with a suppressed immune system, including those receiving therapeutic TNF blockade to treat autoinflammatory disorders. TNF induces pro-inflammatory gene expression, cellular proliferation, and survival signals in certain contexts, but can also trigger programmed cell death in others. It remains unclear, however, which of the pleiotropic functions of TNF mediate control of intracellular bacterial pathogens like Legionella. In this study, we demonstrate that TNF signaling licenses macrophages to die rapidly in response to Legionella infection. We find that TNF-licensed cells undergo rapid gasdermin-dependent, pyroptotic death downstream of inflammasome activation. We also find that TNF signaling upregulates components of the inflammasome response, and that the caspase-11-mediated non-canonical inflammasome is the first inflammasome to be activated, with caspase-1 and caspase-8 mediating delayed pyroptotic death. We find that all three caspases are collectively required for optimal TNF-mediated restriction of bacterial replication in macrophages. Furthermore, caspase-8 is required for control of pulmonary Legionella infection. These findings reveal a TNF-dependent mechanism in macrophages for activating rapid cell death that is collectively mediated by caspases-1, -8, and -11 and subsequent restriction of Legionella infection.
first_indexed 2024-03-13T03:55:30Z
format Article
id doaj.art-08fb8a122725426ab65a91afd75c952a
institution Directory Open Access Journal
issn 1553-7366
1553-7374
language English
last_indexed 2024-03-13T03:55:30Z
publishDate 2023-06-01
publisher Public Library of Science (PLoS)
record_format Article
series PLoS Pathogens
spelling doaj.art-08fb8a122725426ab65a91afd75c952a2023-06-22T05:31:21ZengPublic Library of Science (PLoS)PLoS Pathogens1553-73661553-73742023-06-01196e101076710.1371/journal.ppat.1010767TNF licenses macrophages to undergo rapid caspase-1, -11, and -8-mediated cell death that restricts Legionella pneumophila infection.Tzvi Y PollockVíctor R Vázquez MarreroIgor E BrodskySunny ShinThe inflammatory cytokine tumor necrosis factor (TNF) is necessary for host defense against many intracellular pathogens, including Legionella pneumophila. Legionella causes the severe pneumonia Legionnaires' disease and predominantly affects individuals with a suppressed immune system, including those receiving therapeutic TNF blockade to treat autoinflammatory disorders. TNF induces pro-inflammatory gene expression, cellular proliferation, and survival signals in certain contexts, but can also trigger programmed cell death in others. It remains unclear, however, which of the pleiotropic functions of TNF mediate control of intracellular bacterial pathogens like Legionella. In this study, we demonstrate that TNF signaling licenses macrophages to die rapidly in response to Legionella infection. We find that TNF-licensed cells undergo rapid gasdermin-dependent, pyroptotic death downstream of inflammasome activation. We also find that TNF signaling upregulates components of the inflammasome response, and that the caspase-11-mediated non-canonical inflammasome is the first inflammasome to be activated, with caspase-1 and caspase-8 mediating delayed pyroptotic death. We find that all three caspases are collectively required for optimal TNF-mediated restriction of bacterial replication in macrophages. Furthermore, caspase-8 is required for control of pulmonary Legionella infection. These findings reveal a TNF-dependent mechanism in macrophages for activating rapid cell death that is collectively mediated by caspases-1, -8, and -11 and subsequent restriction of Legionella infection.https://doi.org/10.1371/journal.ppat.1010767
spellingShingle Tzvi Y Pollock
Víctor R Vázquez Marrero
Igor E Brodsky
Sunny Shin
TNF licenses macrophages to undergo rapid caspase-1, -11, and -8-mediated cell death that restricts Legionella pneumophila infection.
PLoS Pathogens
title TNF licenses macrophages to undergo rapid caspase-1, -11, and -8-mediated cell death that restricts Legionella pneumophila infection.
title_full TNF licenses macrophages to undergo rapid caspase-1, -11, and -8-mediated cell death that restricts Legionella pneumophila infection.
title_fullStr TNF licenses macrophages to undergo rapid caspase-1, -11, and -8-mediated cell death that restricts Legionella pneumophila infection.
title_full_unstemmed TNF licenses macrophages to undergo rapid caspase-1, -11, and -8-mediated cell death that restricts Legionella pneumophila infection.
title_short TNF licenses macrophages to undergo rapid caspase-1, -11, and -8-mediated cell death that restricts Legionella pneumophila infection.
title_sort tnf licenses macrophages to undergo rapid caspase 1 11 and 8 mediated cell death that restricts legionella pneumophila infection
url https://doi.org/10.1371/journal.ppat.1010767
work_keys_str_mv AT tzviypollock tnflicensesmacrophagestoundergorapidcaspase111and8mediatedcelldeaththatrestrictslegionellapneumophilainfection
AT victorrvazquezmarrero tnflicensesmacrophagestoundergorapidcaspase111and8mediatedcelldeaththatrestrictslegionellapneumophilainfection
AT igorebrodsky tnflicensesmacrophagestoundergorapidcaspase111and8mediatedcelldeaththatrestrictslegionellapneumophilainfection
AT sunnyshin tnflicensesmacrophagestoundergorapidcaspase111and8mediatedcelldeaththatrestrictslegionellapneumophilainfection