Unknown

Dataset Information

0

Interleukin-1? regulates fat-liver crosstalk in obesity by auto-paracrine modulation of adipose tissue inflammation and expandability.


ABSTRACT: The inflammasome has been recently implicated in obesity-associated dys-metabolism. However, of its products, the specific role of IL-1? was clinically demonstrated to mediate only the pancreatic beta-cell demise, and in mice mainly the intra-hepatic manifestations of obesity. Yet, it remains largely unknown if IL-1?, a cytokine believed to mainly function locally, could regulate dysfunctional inter-organ crosstalk in obesity. Here we show that High-fat-fed (HFF) mice exhibited a preferential increase of IL-1? in portal compared to systemic blood. Moreover, portally-drained mesenteric fat transplantation from IL-1?KO donors resulted in lower pyruvate-glucose flux compared to mice receiving wild-type (WT) transplant. These results raised a putative endocrine function for visceral fat-derived IL-1? in regulating hepatic gluconeogenic flux. IL-1?KO mice on HFF exhibited only a minor or no increase in adipose expression of pro-inflammatory genes (including macrophage M1 markers), Mac2-positive crown-like structures and CD11b-F4/80-double-positive macrophages, all of which were markedly increased in WT-HFF mice. Further consistent with autocrine/paracrine functions of IL-1? within adipose tissue, adipose tissue macrophage lipid content was increased in WT-HFF mice, but significantly less in IL-1?KO mice. Ex-vivo, adipose explants co-cultured with primary hepatocytes from WT or IL-1-receptor (IL-1RI)-KO mice suggested only a minor direct effect of adipose-derived IL-1? on hepatocyte insulin resistance. Importantly, although IL-1?KOs gained weight similarly to WT-HFF, they had larger fat depots with similar degree of adipocyte hypertrophy. Furthermore, adipogenesis genes and markers (pparg, cepba, fabp4, glut4) that were decreased by HFF in WT, were paradoxically elevated in IL-1?KO-HFF mice. These local alterations in adipose tissue inflammation and expansion correlated with a lower liver size, less hepatic steatosis, and preserved insulin sensitivity. Collectively, we demonstrate that by promoting adipose inflammation and limiting fat tissue expandability, IL-1? supports ectopic fat accumulation in hepatocytes and adipose-tissue macrophages, contributing to impaired fat-liver crosstalk in nutritional obesity.

SUBMITTER: Nov O 

PROVIDER: S-EPMC3547030 | biostudies-literature | 2013

REPOSITORIES: biostudies-literature

altmetric image

Publications

Interleukin-1β regulates fat-liver crosstalk in obesity by auto-paracrine modulation of adipose tissue inflammation and expandability.

Nov Ori O   Shapiro Hagit H   Ovadia Hilla H   Tarnovscki Tanya T   Dvir Irit I   Shemesh Elad E   Kovsan Julia J   Shelef Ilan I   Carmi Yaron Y   Voronov Elena E   Apte Ron N RN   Lewis Eli E   Haim Yulia Y   Konrad Daniel D   Bashan Nava N   Rudich Assaf A  

PloS one 20130116 1


The inflammasome has been recently implicated in obesity-associated dys-metabolism. However, of its products, the specific role of IL-1β was clinically demonstrated to mediate only the pancreatic beta-cell demise, and in mice mainly the intra-hepatic manifestations of obesity. Yet, it remains largely unknown if IL-1β, a cytokine believed to mainly function locally, could regulate dysfunctional inter-organ crosstalk in obesity. Here we show that High-fat-fed (HFF) mice exhibited a preferential in  ...[more]

Similar Datasets

2022-11-15 | PXD031271 | Pride
| S-EPMC6811775 | biostudies-literature
| S-EPMC7470880 | biostudies-literature
| S-EPMC4170127 | biostudies-literature
| S-EPMC8615496 | biostudies-literature
| S-EPMC7910586 | biostudies-literature
| S-EPMC4314587 | biostudies-literature
| S-EPMC5757862 | biostudies-literature
| S-EPMC4429090 | biostudies-other
| S-EPMC7022151 | biostudies-literature