[PDF][PDF] A liver-enriched long non-coding RNA, lncLSTR, regulates systemic lipid metabolism in mice

P Li, X Ruan, L Yang, K Kiesewetter, Y Zhao, H Luo… - Cell metabolism, 2015 - cell.com
P Li, X Ruan, L Yang, K Kiesewetter, Y Zhao, H Luo, Y Chen, M Gucek, J Zhu, H Cao
Cell metabolism, 2015cell.com
Long non-coding RNAs (lncRNAs) constitute a significant portion of mammalian genome,
yet the physiological importance of lncRNAs is largely unknown. Here, we identify a liver-
enriched lncRNA in mouse that we term liver-specific triglyceride regulator (lncLSTR). Mice
with a liver-specific depletion of lncLSTR exhibit a marked reduction in plasma triglyceride
levels. We show that lncLSTR depletion enhances apoC2 expression, leading to robust
lipoprotein lipase activation and increased plasma triglyceride clearance. We further …
Summary
Long non-coding RNAs (lncRNAs) constitute a significant portion of mammalian genome, yet the physiological importance of lncRNAs is largely unknown. Here, we identify a liver-enriched lncRNA in mouse that we term liver-specific triglyceride regulator (lncLSTR). Mice with a liver-specific depletion of lncLSTR exhibit a marked reduction in plasma triglyceride levels. We show that lncLSTR depletion enhances apoC2 expression, leading to robust lipoprotein lipase activation and increased plasma triglyceride clearance. We further demonstrate that the regulation of apoC2 expression occurs through an FXR-mediated pathway. LncLSTR forms a molecular complex with TDP-43 to regulate expression of Cyp8b1, a key enzyme in the bile acid synthesis pathway, and engenders an in vivo bile pool that induces apoC2 expression through FXR. Finally, we demonstrate that lncLSTR depletion can reduce triglyceride levels in a hyperlipidemia mouse model. Taken together, these data support a model in which lncLSTR regulates a TDP-43/FXR/apoC2-dependent pathway to maintain systemic lipid homeostasis.
cell.com