Cargando…

IFN-γ mediates Paneth cell death via suppression of mTOR

Paneth cells constitutively produce antimicrobial peptides and growth factors that allow for intestinal homeostasis, host protection, and intestinal stem cell replication. Paneth cells rely heavily on the glycolytic metabolic program, which is in part controlled by the kinase complex Mechanistic tar...

Descripción completa

Detalles Bibliográficos
Autores principales: Araujo, Alessandra, Safronova, Alexandra, Burger, Elise, López-Yglesias, Américo, Giri, Shilpi, Camanzo, Ellie T, Martin, Andrew T, Grivennikov, Sergei, Yarovinsky, Felix
Formato: Online Artículo Texto
Lenguaje:English
Publicado: eLife Sciences Publications, Ltd 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8570691/
https://www.ncbi.nlm.nih.gov/pubmed/34633285
http://dx.doi.org/10.7554/eLife.60478
_version_ 1784594880339640320
author Araujo, Alessandra
Safronova, Alexandra
Burger, Elise
López-Yglesias, Américo
Giri, Shilpi
Camanzo, Ellie T
Martin, Andrew T
Grivennikov, Sergei
Yarovinsky, Felix
author_facet Araujo, Alessandra
Safronova, Alexandra
Burger, Elise
López-Yglesias, Américo
Giri, Shilpi
Camanzo, Ellie T
Martin, Andrew T
Grivennikov, Sergei
Yarovinsky, Felix
author_sort Araujo, Alessandra
collection PubMed
description Paneth cells constitutively produce antimicrobial peptides and growth factors that allow for intestinal homeostasis, host protection, and intestinal stem cell replication. Paneth cells rely heavily on the glycolytic metabolic program, which is in part controlled by the kinase complex Mechanistic target of rapamycin (mTORC1). Yet, little is known about mTOR importance in Paneth cell integrity under steady-state and inflammatory conditions. Our results demonstrate that IFN-γ, a crucial mediator of the intestinal inflammation, acts directly on murine Paneth cells to alter their mitochondrial integrity and membrane potential, resulting in an TORC1-dependent cell death mechanism distinct from canonical cell death pathways including apoptosis, necroptosis, and pyroptosis. These results were established with the purified cytokine and a physiologically relevant common Th1-inducing human parasite Toxoplasma gondii. Given the crucial role for IFN-γ, which is a cytokine frequently associated with the development of inflammatory bowel disease and compromised Paneth cell functions, the identified mechanisms underlying mTORC1-dependent Paneth cell death downstream of IFN-γ may provide promising novel approaches for treating intestinal inflammation.
format Online
Article
Text
id pubmed-8570691
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher eLife Sciences Publications, Ltd
record_format MEDLINE/PubMed
spelling pubmed-85706912021-11-08 IFN-γ mediates Paneth cell death via suppression of mTOR Araujo, Alessandra Safronova, Alexandra Burger, Elise López-Yglesias, Américo Giri, Shilpi Camanzo, Ellie T Martin, Andrew T Grivennikov, Sergei Yarovinsky, Felix eLife Immunology and Inflammation Paneth cells constitutively produce antimicrobial peptides and growth factors that allow for intestinal homeostasis, host protection, and intestinal stem cell replication. Paneth cells rely heavily on the glycolytic metabolic program, which is in part controlled by the kinase complex Mechanistic target of rapamycin (mTORC1). Yet, little is known about mTOR importance in Paneth cell integrity under steady-state and inflammatory conditions. Our results demonstrate that IFN-γ, a crucial mediator of the intestinal inflammation, acts directly on murine Paneth cells to alter their mitochondrial integrity and membrane potential, resulting in an TORC1-dependent cell death mechanism distinct from canonical cell death pathways including apoptosis, necroptosis, and pyroptosis. These results were established with the purified cytokine and a physiologically relevant common Th1-inducing human parasite Toxoplasma gondii. Given the crucial role for IFN-γ, which is a cytokine frequently associated with the development of inflammatory bowel disease and compromised Paneth cell functions, the identified mechanisms underlying mTORC1-dependent Paneth cell death downstream of IFN-γ may provide promising novel approaches for treating intestinal inflammation. eLife Sciences Publications, Ltd 2021-10-11 /pmc/articles/PMC8570691/ /pubmed/34633285 http://dx.doi.org/10.7554/eLife.60478 Text en © 2021, Araujo et al https://creativecommons.org/licenses/by/4.0/This article is distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use and redistribution provided that the original author and source are credited.
spellingShingle Immunology and Inflammation
Araujo, Alessandra
Safronova, Alexandra
Burger, Elise
López-Yglesias, Américo
Giri, Shilpi
Camanzo, Ellie T
Martin, Andrew T
Grivennikov, Sergei
Yarovinsky, Felix
IFN-γ mediates Paneth cell death via suppression of mTOR
title IFN-γ mediates Paneth cell death via suppression of mTOR
title_full IFN-γ mediates Paneth cell death via suppression of mTOR
title_fullStr IFN-γ mediates Paneth cell death via suppression of mTOR
title_full_unstemmed IFN-γ mediates Paneth cell death via suppression of mTOR
title_short IFN-γ mediates Paneth cell death via suppression of mTOR
title_sort ifn-γ mediates paneth cell death via suppression of mtor
topic Immunology and Inflammation
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8570691/
https://www.ncbi.nlm.nih.gov/pubmed/34633285
http://dx.doi.org/10.7554/eLife.60478
work_keys_str_mv AT araujoalessandra ifngmediatespanethcelldeathviasuppressionofmtor
AT safronovaalexandra ifngmediatespanethcelldeathviasuppressionofmtor
AT burgerelise ifngmediatespanethcelldeathviasuppressionofmtor
AT lopezyglesiasamerico ifngmediatespanethcelldeathviasuppressionofmtor
AT girishilpi ifngmediatespanethcelldeathviasuppressionofmtor
AT camanzoelliet ifngmediatespanethcelldeathviasuppressionofmtor
AT martinandrewt ifngmediatespanethcelldeathviasuppressionofmtor
AT grivennikovsergei ifngmediatespanethcelldeathviasuppressionofmtor
AT yarovinskyfelix ifngmediatespanethcelldeathviasuppressionofmtor