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

Y Wang, J Li, L Tang, Y Wang, R Charnigo… - PloS one, 2010 - journals.plos.org
Y Wang, J Li, L Tang, Y Wang, R Charnigo, W de Villiers, E Eckhardt
PloS one, 2010journals.plos.org
Background 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 …
Background
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.
Methodology/Principal Findings
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.
Conclusions/Significance
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.
PLOS