Hepatocyte-specific, PPARγ-regulated mechanisms to promote steatosis in adult mice

AW Greenstein, N Majumdar, P Yang… - J …, 2017 - joe.bioscientifica.com
AW Greenstein, N Majumdar, P Yang, PV Subbaiah, RD Kineman, J Cordoba-Chacon
J Endocrinol, 2017joe.bioscientifica.com
Peroxisome proliferator-activated receptor γ (PPARγ) is the target for thiazolidinones (TZDs),
drugs that improve insulin sensitivity and fatty liver in humans and rodent models, related to
a reduction in hepatic de novo lipogenesis (DNL). The systemic effects of TZDs are in
contrast to reports suggesting hepatocyte-specific activation of PPARγ promotes DNL,
triacylglycerol (TAG) uptake and fatty acid (FA) esterification. As these hepatocyte-specific
effects of PPARγ could counterbalance the positive therapeutic actions of systemic delivery …
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is the target for thiazolidinones (TZDs), drugs that improve insulin sensitivity and fatty liver in humans and rodent models, related to a reduction in hepatic de novo lipogenesis (DNL). The systemic effects of TZDs are in contrast to reports suggesting hepatocyte-specific activation of PPARγ promotes DNL, triacylglycerol (TAG) uptake and fatty acid (FA) esterification. As these hepatocyte-specific effects of PPARγ could counterbalance the positive therapeutic actions of systemic delivery of TZDs, the current study used a mouse model of adult-onset, liver (hepatocyte)-specific PPARγ knockdown (aLivPPARγkd). This model has advantages over existing congenital knockout models, by avoiding compensatory changes related to embryonic knockdown, thus better modeling the impact of altering PPARγ on adult physiology, where metabolic diseases most frequently develop. The impact of aLivPPARγkd on hepatic gene expression and endpoints in lipid metabolism was examined after 1 or 18 weeks (Chow-fed) or after 14 weeks of low-or high-fat (HF) diet. aLivPPARγkd reduced hepatic TAG content but did not impact endpoints in DNL or TAG uptake. However, aLivPPARγkd reduced the expression of the FA translocase (Cd36), in 18-week Chow-and HF-fed mice, associated with increased NEFA after HF feeding. Also, aLivPPARγkd dramatically reduced Mogat1 expression, that was reflected by an increase in hepatic monoacylglycerol (MAG) levels, indicative of reduced MOGAT activity. These results, coupled with previous reports, suggest that Cd36-mediated FA uptake and MAG pathway-mediated FA esterification are major targets of hepatocyte PPARγ, where loss of this control explains in part the protection against steatosis observed after aLivPPARγkd.
joe.bioscientifica.com