Cargando…

Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8(+) T cells

T cell receptor (TCR) stimulation leads to the expression of the transcription factor thymocyte selection–associated high-mobility group box (TOX). Prolonged TCR signaling, such as encountered during chronic infections or in tumors, leads to sustained TOX expression, which is required for the induct...

Descripción completa

Detalles Bibliográficos
Autores principales: Maurice, Nicholas J., Berner, Jacqueline, Taber, Alexis K., Zehn, Dietmar, Prlic, Martin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Clinical Investigation 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8410038/
https://www.ncbi.nlm.nih.gov/pubmed/34032638
http://dx.doi.org/10.1172/jci.insight.150744
_version_ 1783747083941969920
author Maurice, Nicholas J.
Berner, Jacqueline
Taber, Alexis K.
Zehn, Dietmar
Prlic, Martin
author_facet Maurice, Nicholas J.
Berner, Jacqueline
Taber, Alexis K.
Zehn, Dietmar
Prlic, Martin
author_sort Maurice, Nicholas J.
collection PubMed
description T cell receptor (TCR) stimulation leads to the expression of the transcription factor thymocyte selection–associated high-mobility group box (TOX). Prolonged TCR signaling, such as encountered during chronic infections or in tumors, leads to sustained TOX expression, which is required for the induction of a state of exhaustion or dysfunction. Although CD8(+) memory T (Tmem) cells in mice typically do not express TOX at steady state, some human Tmem cells express TOX but appear fully functional. This seeming discrepancy between mouse and human T cells has led to the speculation that TOX is differentially regulated between these species, which could complicate the interpretation of preclinical mouse model studies. We report here that, similar to TCR-mediated signals, inflammatory cytokines are also sufficient to increase TOX expression in human and mouse Tmem cells. Thus, TOX expression is controlled by the environment, which provides an explanation for the different TOX expression patterns encountered in T cells isolated from specific pathogen–free laboratory mice versus humans. Finally, we report that TOX is not necessary for cytokine-driven expression of programmed cell death 1. Overall, our data highlight that the mechanisms regulating TOX expression are conserved across species and indicate that TOX expression reflects a T cell’s activation state and does not necessarily correlate with T cell dysfunction.
format Online
Article
Text
id pubmed-8410038
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher American Society for Clinical Investigation
record_format MEDLINE/PubMed
spelling pubmed-84100382021-09-07 Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8(+) T cells Maurice, Nicholas J. Berner, Jacqueline Taber, Alexis K. Zehn, Dietmar Prlic, Martin JCI Insight Research Article T cell receptor (TCR) stimulation leads to the expression of the transcription factor thymocyte selection–associated high-mobility group box (TOX). Prolonged TCR signaling, such as encountered during chronic infections or in tumors, leads to sustained TOX expression, which is required for the induction of a state of exhaustion or dysfunction. Although CD8(+) memory T (Tmem) cells in mice typically do not express TOX at steady state, some human Tmem cells express TOX but appear fully functional. This seeming discrepancy between mouse and human T cells has led to the speculation that TOX is differentially regulated between these species, which could complicate the interpretation of preclinical mouse model studies. We report here that, similar to TCR-mediated signals, inflammatory cytokines are also sufficient to increase TOX expression in human and mouse Tmem cells. Thus, TOX expression is controlled by the environment, which provides an explanation for the different TOX expression patterns encountered in T cells isolated from specific pathogen–free laboratory mice versus humans. Finally, we report that TOX is not necessary for cytokine-driven expression of programmed cell death 1. Overall, our data highlight that the mechanisms regulating TOX expression are conserved across species and indicate that TOX expression reflects a T cell’s activation state and does not necessarily correlate with T cell dysfunction. American Society for Clinical Investigation 2021-07-08 /pmc/articles/PMC8410038/ /pubmed/34032638 http://dx.doi.org/10.1172/jci.insight.150744 Text en © 2021 Maurice et al. https://creativecommons.org/licenses/by/4.0/This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Research Article
Maurice, Nicholas J.
Berner, Jacqueline
Taber, Alexis K.
Zehn, Dietmar
Prlic, Martin
Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8(+) T cells
title Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8(+) T cells
title_full Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8(+) T cells
title_fullStr Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8(+) T cells
title_full_unstemmed Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8(+) T cells
title_short Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8(+) T cells
title_sort inflammatory signals are sufficient to elicit tox expression in mouse and human cd8(+) t cells
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8410038/
https://www.ncbi.nlm.nih.gov/pubmed/34032638
http://dx.doi.org/10.1172/jci.insight.150744
work_keys_str_mv AT mauricenicholasj inflammatorysignalsaresufficienttoelicittoxexpressioninmouseandhumancd8tcells
AT bernerjacqueline inflammatorysignalsaresufficienttoelicittoxexpressioninmouseandhumancd8tcells
AT taberalexisk inflammatorysignalsaresufficienttoelicittoxexpressioninmouseandhumancd8tcells
AT zehndietmar inflammatorysignalsaresufficienttoelicittoxexpressioninmouseandhumancd8tcells
AT prlicmartin inflammatorysignalsaresufficienttoelicittoxexpressioninmouseandhumancd8tcells