Skip to main content
eScholarship
Open Access Publications from the University of California

UC Berkeley

UC Berkeley Electronic Theses and Dissertations bannerUC Berkeley

Regulation of lipogenic gene transcription during fasting and feeding/insulin: Role of USF, SREBP-1c, and BAF60c

Abstract

Transcription of genes encoding enzymes involved in fatty acid and triacylglycerol synthesis, including fatty acid synthase and mitochondrial glycerol-3-phosphate acyltransferase, is coordinately induced in lipogenic tissues by feeding and insulin treatment. Dysregulation of lipognesis often contributes to metabolic diseases such as obesity, diabetes, and cardiovascular diseases. Transcription factors and signaling molecules involved in transcriptional activation of lipogenesis represent attractive targets for the prevention and treatment of metabolic diseases.

In transcriptional activation of fatty acid synthase by feeding/insulin, USF constitutively bound to the -65 E-box is required. In this study, USF was shown to function as a molecular switch by recruiting various interacting proteins during the fasting/feeding transition. First, USF was detected to directly interact with SREBP-1 that is induced by feeding and binds nearby -150 SRE. Cotransfection of USF and SREBP-1c with an FAS promoter-luciferase reporter construct resulted in high synergistic activation of the FAS promoter. Chromatin immunoprecipitation analysis of mouse liver demonstrated that USF binds constitutively to the fatty acid synthase promoter during fasting/feeding, whereas the binding of SREBP-1 was observed only during feeding, in a manner identical to that of the FAS promoter. These data show that USF recruits SREBP-1c to the lipogenic gene promoters upon feeding/insulin. Similar cooperative action of USF and SREBP-1c in the activation of the mitochondrial glycerol-3-phosphate acyltransferase promoter was also observed.

During feeding/insulin, USF-1 recruits three distinct families of proteins to the lipogenic promoters: 1) P/CAF which acetylates USF and functions as a coactivator, 2) DNA break/repair machinery including Ku70, Ku80, PARP-1, TopoIIB and DNA-PK, which causes a DNA break in the lipogenic gene promoter prior to the transcriptional initiation in vivo, and 3) signaling molecules including PP1 and DNA-PK. PP1 dephosphorylates and activates DNA-PK upon feeding/insulin treatment. Thus in the fed state, activated DNA-PK phosphorylates USF-1 at S262, allowing the recruitment of and acetylation by P/CAF at K237, leading to promoter activation. P/CAF-mediated acetylation of USF is reversed by HDAC9 in the fasted state. Although total HDAC9 levels do not change during fasting/feeding, nuclear abundance of HDAC9 increases upon fasting, suggesting regulation of HDAC9 by nuclear translocation. DNA break/repair components associated with USF also bring about transient DNA breaks during feeding-induced FAS activation. In DNA-PK deficient SCID mice, feeding induced USF-1 phosphorylation/acetylation, DNA-breaks, and FAS activation leading to lipogenesis are impaired, resulting in decreased liver and circulating triglyceride levels. This study demonstrates that DNA-PK mediates the feeding/insulin-dependent lipogenic gene activation. BAF60c was also detected to be an USF interacting protein recruited to lipogenic gene promoters upon feeding. Among the three isoforms of BAF60s, BAF60c is the only BAF60 specific to lipogenesis and recruits other BAF subunits including BAF155 and BAF190 for the formation of lipoBAF. BAF60 proteins function as the bridge between transcription factors and the BAF complex. BAF60c was found to be phosphorylated at S247 by aPKC upon feeding/insulin. BAF60c was translocated from the cytosol to the nucleus in response to feeding/insulin and the nuclear localization was dependent on its S247 phosphorylation. Furthermore, this BAF60c phosphorylation together with USF acetylation were required for the interaction between the two proteins. Overexpression of BAF60c activated the lipogenic transcription program in mice even in the fasted state. This study provides a novel mechanism to fine-tune lipogenic transcription in response to feeding/insulin. Closely positioned E-boxes and sterol regulatory elements found in the promoters of several lipogenic genes suggest that USF functions as a master molecular switch as a common mechanism of induction by feeding/insulin. Taken together, identification of SREBP-1c, DNA-PK and BAF60c as USF interacting proteins has led to the discovery of novel players in insulin signaling cascade and has revealed an unexpected link between DNA break/repair and metabolism.

Main Content
For improved accessibility of PDF content, download the file to your device.
Current View