[HTML][HTML] Effect of modulation of PPAR-γ activity on Kupffer cells M1/M2 polarization in the development of non-alcoholic fatty liver disease

W Luo, Q Xu, Q Wang, H Wu, J Hua�- Scientific reports, 2017 - nature.com
W Luo, Q Xu, Q Wang, H Wu, J Hua
Scientific reports, 2017nature.com
Abnormal lipid-mediated hepatic inflammatory-immune dysfunction and chronic low grade
inflammation play an important role in the pathogenesis of non-alcoholic fatty liver disease
(NAFLD). Macrophage polarization is an important mechanism for the regulation of
inflammatory response. Since PPAR-γ has emerged as a master regulator of macrophage
polarization, we aimed to investigate the lipid-induced macrophage/Kupffer cell polarization
in vivo and in vitro, and explore the association between PPAR-γ activity and macrophages�…
Abstract
Abnormal lipid-mediated hepatic inflammatory-immune dysfunction and chronic low grade inflammation play an important role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Macrophage polarization is an important mechanism for the regulation of inflammatory response. Since PPAR-γ has emerged as a master regulator of macrophage polarization, we aimed to investigate the lipid-induced macrophage/Kupffer cell polarization in vivo and in vitro, and explore the association between PPAR-γ activity and macrophages M1/M2 polarization shifting. Here we showed that long-term high-fat diet increased Kupffer cells content with M1-predominant phenotype and increasing production of pro-inflammatory cytokines. Saturated fatty acids polarized Kupffer cells/macrophages to an M1-predominant phenotype while n-3 PUFA polarized Kupffer cells/macrophages to an M2 phenotype, which was associated with activation of NF-κB signal pathway and PPAR-γ respectively. Furthermore, up-regulation of PPAR-γ shifted lipid-induced macrophages polarization from M1-predominant phenotype to M2 phenotype. Macrophages polarization switch was associated with the interaction between PPAR-γ and NF-κBp65 signal pathway. Rosiglitazone restored high-fat diet-induced imblance of Kupffer cells M1/M2 polarization and alleviated hepatic steatosis as well as local pro-inflammatory response. These findings suggest that manipulation of PPAR-γ activity has the potential to balance lipid-induced M1/M2 macrophage/Kupffer cell polarization, and leading to prevent the development of NAFLD.
nature.com