T-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.

Obesity is associated with inflammation of visceral adipose tissues, which increases the risk for insulin resistance. Animal models suggest that T-lymphocyte infiltration is an important early step, although it is unclear why these cells are attracted. We have recently demonstrated that dietary trig...

Full description

Bibliographic Details
Main Authors: Yuehui Wang, Jianing Li, Lihua Tang, Yu Wang, Richard Charnigo, Willem de Villiers, Erik Eckhardt
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2010-11-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC2978720?pdf=render
id doaj-8ab409117250480990d5bd7305d6cfba
record_format Article
spelling doaj-8ab409117250480990d5bd7305d6cfba2020-11-25T01:56:04ZengPublic Library of Science (PLoS)PLoS ONE1932-62032010-11-01511e1395110.1371/journal.pone.0013951T-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.Yuehui WangJianing LiLihua TangYu WangRichard CharnigoWillem de VilliersErik EckhardtObesity is associated with inflammation of visceral adipose tissues, which increases the risk for insulin resistance. Animal models suggest that T-lymphocyte infiltration is an important early step, although it is unclear why these cells are attracted. We have recently demonstrated that dietary triglycerides, major components of high fat diets, promote intestinal absorption of a protein antigen (ovalbumin, "OVA"). The antigen was partly transported on chylomicrons, which are prominently cleared in adipose tissues. We hypothesized that intestinally absorbed gut antigens may cause T-lymphocyte associated inflammation in adipose tissue.Triglyceride absorption promoted intestinal absorption of OVA into adipose tissue, in a chylomicron-dependent manner. Absorption tended to be higher in mesenteric than subcutaneous adipose tissue, and was lowest in gonadal tissue. OVA immunoreactivity was detected in stromal vascular cells, including endothelial cells. In OVA-sensitized mice, OVA feeding caused marked accumulation of CD3+ and osteopontin+ cells in mesenteric adipose tissue. The accumulating T-lymphocytes were mainly CD4+. As expected, high-fat (60% kCal) diets promoted mesenteric adipose tissue inflammation compared to low-fat diets (10% Kcal), as reflected by increased expression of osteopontin and interferon-gamma. Immune responses to dietary OVA further increased diet-induced osteopontin and interferon-gamma expression in mesenteric adipose. Inflammatory gene expression in subcutaneous tissue did not respond significantly to OVA or dietary fat content. Lastly, whereas OVA responses did not significantly affect bodyweight or adiposity, they significantly impaired glucose tolerance.Our results suggest that loss or lack of immunological tolerance to intestinally absorbed T-lymphocyte antigens can contribute to mesenteric adipose tissue inflammation and defective glucose metabolism during high-fat dieting.http://europepmc.org/articles/PMC2978720?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Yuehui Wang
Jianing Li
Lihua Tang
Yu Wang
Richard Charnigo
Willem de Villiers
Erik Eckhardt
spellingShingle Yuehui Wang
Jianing Li
Lihua Tang
Yu Wang
Richard Charnigo
Willem de Villiers
Erik Eckhardt
T-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.
PLoS ONE
author_facet Yuehui Wang
Jianing Li
Lihua Tang
Yu Wang
Richard Charnigo
Willem de Villiers
Erik Eckhardt
author_sort Yuehui Wang
title T-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.
title_short T-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.
title_full T-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.
title_fullStr T-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.
title_full_unstemmed T-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.
title_sort t-lymphocyte responses to intestinally absorbed antigens can contribute to adipose tissue inflammation and glucose intolerance during high fat feeding.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2010-11-01
description Obesity is associated with inflammation of visceral adipose tissues, which increases the risk for insulin resistance. Animal models suggest that T-lymphocyte infiltration is an important early step, although it is unclear why these cells are attracted. We have recently demonstrated that dietary triglycerides, major components of high fat diets, promote intestinal absorption of a protein antigen (ovalbumin, "OVA"). The antigen was partly transported on chylomicrons, which are prominently cleared in adipose tissues. We hypothesized that intestinally absorbed gut antigens may cause T-lymphocyte associated inflammation in adipose tissue.Triglyceride absorption promoted intestinal absorption of OVA into adipose tissue, in a chylomicron-dependent manner. Absorption tended to be higher in mesenteric than subcutaneous adipose tissue, and was lowest in gonadal tissue. OVA immunoreactivity was detected in stromal vascular cells, including endothelial cells. In OVA-sensitized mice, OVA feeding caused marked accumulation of CD3+ and osteopontin+ cells in mesenteric adipose tissue. The accumulating T-lymphocytes were mainly CD4+. As expected, high-fat (60% kCal) diets promoted mesenteric adipose tissue inflammation compared to low-fat diets (10% Kcal), as reflected by increased expression of osteopontin and interferon-gamma. Immune responses to dietary OVA further increased diet-induced osteopontin and interferon-gamma expression in mesenteric adipose. Inflammatory gene expression in subcutaneous tissue did not respond significantly to OVA or dietary fat content. Lastly, whereas OVA responses did not significantly affect bodyweight or adiposity, they significantly impaired glucose tolerance.Our results suggest that loss or lack of immunological tolerance to intestinally absorbed T-lymphocyte antigens can contribute to mesenteric adipose tissue inflammation and defective glucose metabolism during high-fat dieting.
url http://europepmc.org/articles/PMC2978720?pdf=render
work_keys_str_mv AT yuehuiwang tlymphocyteresponsestointestinallyabsorbedantigenscancontributetoadiposetissueinflammationandglucoseintoleranceduringhighfatfeeding
AT jianingli tlymphocyteresponsestointestinallyabsorbedantigenscancontributetoadiposetissueinflammationandglucoseintoleranceduringhighfatfeeding
AT lihuatang tlymphocyteresponsestointestinallyabsorbedantigenscancontributetoadiposetissueinflammationandglucoseintoleranceduringhighfatfeeding
AT yuwang tlymphocyteresponsestointestinallyabsorbedantigenscancontributetoadiposetissueinflammationandglucoseintoleranceduringhighfatfeeding
AT richardcharnigo tlymphocyteresponsestointestinallyabsorbedantigenscancontributetoadiposetissueinflammationandglucoseintoleranceduringhighfatfeeding
AT willemdevilliers tlymphocyteresponsestointestinallyabsorbedantigenscancontributetoadiposetissueinflammationandglucoseintoleranceduringhighfatfeeding
AT erikeckhardt tlymphocyteresponsestointestinallyabsorbedantigenscancontributetoadiposetissueinflammationandglucoseintoleranceduringhighfatfeeding
_version_ 1724981869150208000