Cargando…

Short-term cold exposure supports human Treg induction in vivo

OBJECTIVE: Obesity and type-2 diabetes (T2D) are metabolic diseases that represent a critical health problem worldwide. Metabolic disease is differentially associated with fat distribution, while visceral white adipose tissue (VAT) is particularly prone to obesity-associated inflammation. Next to th...

Descripción completa

Detalles Bibliográficos
Autores principales: Becker, Maike, Serr, Isabelle, Salb, Victoria K., Ott, Verena B., Mengel, Laura, Blüher, Matthias, Weigmann, Benno, Hauner, Hans, Tschöp, Matthias H., Daniel, Carolin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6822223/
https://www.ncbi.nlm.nih.gov/pubmed/31427184
http://dx.doi.org/10.1016/j.molmet.2019.08.002
_version_ 1783464293101993984
author Becker, Maike
Serr, Isabelle
Salb, Victoria K.
Ott, Verena B.
Mengel, Laura
Blüher, Matthias
Weigmann, Benno
Hauner, Hans
Tschöp, Matthias H.
Daniel, Carolin
author_facet Becker, Maike
Serr, Isabelle
Salb, Victoria K.
Ott, Verena B.
Mengel, Laura
Blüher, Matthias
Weigmann, Benno
Hauner, Hans
Tschöp, Matthias H.
Daniel, Carolin
author_sort Becker, Maike
collection PubMed
description OBJECTIVE: Obesity and type-2 diabetes (T2D) are metabolic diseases that represent a critical health problem worldwide. Metabolic disease is differentially associated with fat distribution, while visceral white adipose tissue (VAT) is particularly prone to obesity-associated inflammation. Next to their canonical function of immune suppression, regulatory T cells (Tregs) are key in controlling adipose tissue homeostasis. Towards understanding the molecular underpinnings of metabolic disease, we focus on how environmental-metabolic stimuli impinge on the functional interplay between Tregs and adipose tissue. Here, cold exposure or beta3-adrenergic signaling are a promising tool to increase energy expenditure by activating brown adipose tissue, as well as by reducing local inflammation within fat depots by supporting immunosuppressive Tregs. However, in humans, the underlying mechanisms that enable the environmental-immune crosstalk in the periphery and in the respective tissue remain currently unknown. METHODS: We used combinatorial approaches of next generation humanized mouse models and in vitro and in vivo experiments together with beta3-adrenergic stimulation to dissect the underlying mechanisms of human Treg induction exposed to environmental stimuli such as cold. To test the translational relevance of our findings, we analyzed samples from the FREECE study in which human subjects were exposed to individualized cooling protocols. Samples were analyzed ex vivo and after in vitro Treg induction using qRT-PCR, immunofluorescence, as well as with multicolor flow cytometry and cell sorting. RESULTS: In vivo application of the beta3-adrenergic receptor agonist mirabegron in humanized mice induced thermogenesis and improved the Treg induction capacity of naïve T cells isolated from these animals. Using samples from the human FREECE study, we demonstrate that a short-term cold stimulus supports human Treg induction in vitro and in vivo. Mechanistically, we identify BORCS6 encoding the Ragulator-interacting protein C17orf59 to be significantly induced in human CD4(+) T cells upon short-term cold exposure. Strong mTOR signaling is known to limit successful Treg induction and thus likely by interfering with mTOR activation at lysosomal surfaces, C17orf59 improves the Treg induction capacity of human naïve T cells upon cold exposure. CONCLUSIONS: These novel insights into the molecular underpinnings of human Treg induction suggest an important role of Tregs in linking environmental stimuli with adipose tissue function and metabolic diseases. Moreover, these discoveries shed new light on potential approaches towards tailored anti-inflammatory concepts that support human adipose tissue homeostasis by enabling Tregs.
format Online
Article
Text
id pubmed-6822223
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-68222232019-11-06 Short-term cold exposure supports human Treg induction in vivo Becker, Maike Serr, Isabelle Salb, Victoria K. Ott, Verena B. Mengel, Laura Blüher, Matthias Weigmann, Benno Hauner, Hans Tschöp, Matthias H. Daniel, Carolin Mol Metab Original Article OBJECTIVE: Obesity and type-2 diabetes (T2D) are metabolic diseases that represent a critical health problem worldwide. Metabolic disease is differentially associated with fat distribution, while visceral white adipose tissue (VAT) is particularly prone to obesity-associated inflammation. Next to their canonical function of immune suppression, regulatory T cells (Tregs) are key in controlling adipose tissue homeostasis. Towards understanding the molecular underpinnings of metabolic disease, we focus on how environmental-metabolic stimuli impinge on the functional interplay between Tregs and adipose tissue. Here, cold exposure or beta3-adrenergic signaling are a promising tool to increase energy expenditure by activating brown adipose tissue, as well as by reducing local inflammation within fat depots by supporting immunosuppressive Tregs. However, in humans, the underlying mechanisms that enable the environmental-immune crosstalk in the periphery and in the respective tissue remain currently unknown. METHODS: We used combinatorial approaches of next generation humanized mouse models and in vitro and in vivo experiments together with beta3-adrenergic stimulation to dissect the underlying mechanisms of human Treg induction exposed to environmental stimuli such as cold. To test the translational relevance of our findings, we analyzed samples from the FREECE study in which human subjects were exposed to individualized cooling protocols. Samples were analyzed ex vivo and after in vitro Treg induction using qRT-PCR, immunofluorescence, as well as with multicolor flow cytometry and cell sorting. RESULTS: In vivo application of the beta3-adrenergic receptor agonist mirabegron in humanized mice induced thermogenesis and improved the Treg induction capacity of naïve T cells isolated from these animals. Using samples from the human FREECE study, we demonstrate that a short-term cold stimulus supports human Treg induction in vitro and in vivo. Mechanistically, we identify BORCS6 encoding the Ragulator-interacting protein C17orf59 to be significantly induced in human CD4(+) T cells upon short-term cold exposure. Strong mTOR signaling is known to limit successful Treg induction and thus likely by interfering with mTOR activation at lysosomal surfaces, C17orf59 improves the Treg induction capacity of human naïve T cells upon cold exposure. CONCLUSIONS: These novel insights into the molecular underpinnings of human Treg induction suggest an important role of Tregs in linking environmental stimuli with adipose tissue function and metabolic diseases. Moreover, these discoveries shed new light on potential approaches towards tailored anti-inflammatory concepts that support human adipose tissue homeostasis by enabling Tregs. Elsevier 2019-08-05 /pmc/articles/PMC6822223/ /pubmed/31427184 http://dx.doi.org/10.1016/j.molmet.2019.08.002 Text en © 2019 The Authors http://creativecommons.org/licenses/by-nc-nd/4.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Original Article
Becker, Maike
Serr, Isabelle
Salb, Victoria K.
Ott, Verena B.
Mengel, Laura
Blüher, Matthias
Weigmann, Benno
Hauner, Hans
Tschöp, Matthias H.
Daniel, Carolin
Short-term cold exposure supports human Treg induction in vivo
title Short-term cold exposure supports human Treg induction in vivo
title_full Short-term cold exposure supports human Treg induction in vivo
title_fullStr Short-term cold exposure supports human Treg induction in vivo
title_full_unstemmed Short-term cold exposure supports human Treg induction in vivo
title_short Short-term cold exposure supports human Treg induction in vivo
title_sort short-term cold exposure supports human treg induction in vivo
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6822223/
https://www.ncbi.nlm.nih.gov/pubmed/31427184
http://dx.doi.org/10.1016/j.molmet.2019.08.002
work_keys_str_mv AT beckermaike shorttermcoldexposuresupportshumantreginductioninvivo
AT serrisabelle shorttermcoldexposuresupportshumantreginductioninvivo
AT salbvictoriak shorttermcoldexposuresupportshumantreginductioninvivo
AT ottverenab shorttermcoldexposuresupportshumantreginductioninvivo
AT mengellaura shorttermcoldexposuresupportshumantreginductioninvivo
AT bluhermatthias shorttermcoldexposuresupportshumantreginductioninvivo
AT weigmannbenno shorttermcoldexposuresupportshumantreginductioninvivo
AT haunerhans shorttermcoldexposuresupportshumantreginductioninvivo
AT tschopmatthiash shorttermcoldexposuresupportshumantreginductioninvivo
AT danielcarolin shorttermcoldexposuresupportshumantreginductioninvivo