Cargando…

Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages

Eosinophils in visceral adipose tissue (VAT) have been implicated in metabolic homeostasis and the maintenance of alternatively activated macrophages (AAMs). The absence of eosinophils can lead to adiposity and systemic insulin resistance in experimental animals, but what maintains eosinophils in ad...

Descripción completa

Detalles Bibliográficos
Autores principales: Molofsky, Ari B., Nussbaum, Jesse C., Liang, Hong-Erh, Van Dyken, Steven J., Cheng, Laurence E., Mohapatra, Alexander, Chawla, Ajay, Locksley, Richard M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: The Rockefeller University Press 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3600903/
https://www.ncbi.nlm.nih.gov/pubmed/23420878
http://dx.doi.org/10.1084/jem.20121964
_version_ 1782475693281509376
author Molofsky, Ari B.
Nussbaum, Jesse C.
Liang, Hong-Erh
Van Dyken, Steven J.
Cheng, Laurence E.
Mohapatra, Alexander
Chawla, Ajay
Locksley, Richard M.
author_facet Molofsky, Ari B.
Nussbaum, Jesse C.
Liang, Hong-Erh
Van Dyken, Steven J.
Cheng, Laurence E.
Mohapatra, Alexander
Chawla, Ajay
Locksley, Richard M.
author_sort Molofsky, Ari B.
collection PubMed
description Eosinophils in visceral adipose tissue (VAT) have been implicated in metabolic homeostasis and the maintenance of alternatively activated macrophages (AAMs). The absence of eosinophils can lead to adiposity and systemic insulin resistance in experimental animals, but what maintains eosinophils in adipose tissue is unknown. We show that interleukin-5 (IL-5) deficiency profoundly impairs VAT eosinophil accumulation and results in increased adiposity and insulin resistance when animals are placed on a high-fat diet. Innate lymphoid type 2 cells (ILC2s) are resident in VAT and are the major source of IL-5 and IL-13, which promote the accumulation of eosinophils and AAM. Deletion of ILC2s causes significant reductions in VAT eosinophils and AAMs, and also impairs the expansion of VAT eosinophils after infection with Nippostrongylus brasiliensis, an intestinal parasite associated with increased adipose ILC2 cytokine production and enhanced insulin sensitivity. Further, IL-33, a cytokine previously shown to promote cytokine production by ILC2s, leads to rapid ILC2-dependent increases in VAT eosinophils and AAMs. Thus, ILC2s are resident in VAT and promote eosinophils and AAM implicated in metabolic homeostasis, and this axis is enhanced during Th2-associated immune stimulation.
format Online
Article
Text
id pubmed-3600903
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher The Rockefeller University Press
record_format MEDLINE/PubMed
spelling pubmed-36009032013-09-11 Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages Molofsky, Ari B. Nussbaum, Jesse C. Liang, Hong-Erh Van Dyken, Steven J. Cheng, Laurence E. Mohapatra, Alexander Chawla, Ajay Locksley, Richard M. J Exp Med Article Eosinophils in visceral adipose tissue (VAT) have been implicated in metabolic homeostasis and the maintenance of alternatively activated macrophages (AAMs). The absence of eosinophils can lead to adiposity and systemic insulin resistance in experimental animals, but what maintains eosinophils in adipose tissue is unknown. We show that interleukin-5 (IL-5) deficiency profoundly impairs VAT eosinophil accumulation and results in increased adiposity and insulin resistance when animals are placed on a high-fat diet. Innate lymphoid type 2 cells (ILC2s) are resident in VAT and are the major source of IL-5 and IL-13, which promote the accumulation of eosinophils and AAM. Deletion of ILC2s causes significant reductions in VAT eosinophils and AAMs, and also impairs the expansion of VAT eosinophils after infection with Nippostrongylus brasiliensis, an intestinal parasite associated with increased adipose ILC2 cytokine production and enhanced insulin sensitivity. Further, IL-33, a cytokine previously shown to promote cytokine production by ILC2s, leads to rapid ILC2-dependent increases in VAT eosinophils and AAMs. Thus, ILC2s are resident in VAT and promote eosinophils and AAM implicated in metabolic homeostasis, and this axis is enhanced during Th2-associated immune stimulation. The Rockefeller University Press 2013-03-11 /pmc/articles/PMC3600903/ /pubmed/23420878 http://dx.doi.org/10.1084/jem.20121964 Text en © 2013 Molofsky et al. This article is distributed under the terms of an Attribution–Noncommercial–Share Alike–No Mirror Sites license for the first six months after the publication date (see http://www.rupress.org/terms). After six months it is available under a Creative Commons License (Attribution–Noncommercial–Share Alike 3.0 Unported license, as described at http://creativecommons.org/licenses/by-nc-sa/3.0/).
spellingShingle Article
Molofsky, Ari B.
Nussbaum, Jesse C.
Liang, Hong-Erh
Van Dyken, Steven J.
Cheng, Laurence E.
Mohapatra, Alexander
Chawla, Ajay
Locksley, Richard M.
Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages
title Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages
title_full Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages
title_fullStr Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages
title_full_unstemmed Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages
title_short Innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages
title_sort innate lymphoid type 2 cells sustain visceral adipose tissue eosinophils and alternatively activated macrophages
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3600903/
https://www.ncbi.nlm.nih.gov/pubmed/23420878
http://dx.doi.org/10.1084/jem.20121964
work_keys_str_mv AT molofskyarib innatelymphoidtype2cellssustainvisceraladiposetissueeosinophilsandalternativelyactivatedmacrophages
AT nussbaumjessec innatelymphoidtype2cellssustainvisceraladiposetissueeosinophilsandalternativelyactivatedmacrophages
AT lianghongerh innatelymphoidtype2cellssustainvisceraladiposetissueeosinophilsandalternativelyactivatedmacrophages
AT vandykenstevenj innatelymphoidtype2cellssustainvisceraladiposetissueeosinophilsandalternativelyactivatedmacrophages
AT chenglaurencee innatelymphoidtype2cellssustainvisceraladiposetissueeosinophilsandalternativelyactivatedmacrophages
AT mohapatraalexander innatelymphoidtype2cellssustainvisceraladiposetissueeosinophilsandalternativelyactivatedmacrophages
AT chawlaajay innatelymphoidtype2cellssustainvisceraladiposetissueeosinophilsandalternativelyactivatedmacrophages
AT locksleyrichardm innatelymphoidtype2cellssustainvisceraladiposetissueeosinophilsandalternativelyactivatedmacrophages