Abstract

Aberrant glucose metabolism and elevated O-linked β-N-acetylglucosamine modification (O-GlcNAcylation) are hallmarks of hepatocellular carcinoma (HCC). Loss of phosphoenolpyruvate carboxykinase 1 (PCK1), the major rate-limiting enzyme of hepatic gluconeogenesis, increases hexosamine biosynthetic pathway (HBP)-mediated protein O-GlcNAcylation in hepatoma cell and promotes cell growth and proliferation. However, whether PCK1 deficiency and hyper O-GlcNAcylation can induce HCC metastasis is largely unknown. Here, gain- and loss-of-function studies demonstrate that PCK1 suppresses HCC metastasis in vitro and in vivo. Specifically, lysine acetyltransferase 5 (KAT5), belonging to the MYST family of histone acetyltransferases (HAT), is highly modified by O-GlcNAcylation in PCK1 knockout hepatoma cells. Mechanistically, PCK1 depletion suppressed KAT5 ubiquitination by increasing its O-GlcNAcylation, thereby stabilizing KAT5. KAT5 O-GlcNAcylation epigenetically activates TWIST1 expression via histone H4 acetylation, and enhances MMP9 and MMP14 expression via c-Myc acetylation, thus promoting epithelial-mesenchymal transition (EMT) in HCC. In addition, targeting HBP-mediated O-GlcNAcylation of KAT5 inhibits lung metastasis of HCC in hepatospecific Pck1-deletion mice. Collectively, our findings demonstrate that PCK1 depletion increases O-GlcNAcylation of KAT5, epigenetically induces TWIST1 expression and promotes HCC metastasis, and link metabolic enzyme, post-translational modification (PTM) with epigenetic regulation.

Highlights

  • Metabolic reprogramming is a hallmark of cancer cells and supports the bioenergetic and biosynthetic demands of rapid growth and proliferation [1]

  • phosphoenolpyruvate carboxykinase 1 (PCK1) deficiency promotes epithelial-mesenchymal transition (EMT) through epigenetic activation of TWIST1 by O-GlcNAcylation of KAT5 we investigated whether loss of PCK1-mediated acceleration of the migration and invasion of hepatoma cells is dependent on KAT5

  • We found that WT KAT5, but not S119A mutant, partially offset the regulatory effects on Twist1, N-cadherin and E-cadherin expression mediated by PCK1 deficiency (Fig. 4j), suggesting KAT5 O-GlcNAcylation plays an essential role in transcriptional regulation of the expression of TWIST1 and EMT markers

Read more

Summary

INTRODUCTION

Metabolic reprogramming is a hallmark of cancer cells and supports the bioenergetic and biosynthetic demands of rapid growth and proliferation [1]. EMT is the most important characteristic of tumor metastatic cells It is accompanied by downregulation of genes associated with epithelial phenotype, upregulation of genes associated with mesenchymal phenotype, and high expression of genes belonging to the matrix metalloproteinases (MMPs) family [12, 13]. Several reports showed that transcriptional factors including Snail, EMT-related markers such as E-cadherin, and epigenetic modulators, including EZH2, HDAC1, and SIRT1 can be O-GlcNAc-modified and are associated with cancer metastasis [17,18,19,20,21]. Various PTMs of KAT5 have been reported, such as exhibited a distinct O-GlcNAc modification signal in MHCC-97H phosphorylation, SUMOylation and ubiquitination, that regulates cells upon treatment with the OGA inhibitor Thiamet G (TG) O-GlcNAcylation of KAT5 (Fig. 2k), which was further confirmed by immunoprecipitating Flag-tagged WT KAT5 or S119A mutant

RESULTS
DISCUSSION
Findings
MATERIALS AND METHODS
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call