TNF-α/NF-κB signaling epigenetically represses PSD4 transcription to promote alcohol-related hepatocellular carcinoma progression

Cancer Med. 2021 May;10(10):3346-3357. doi: 10.1002/cam4.3832. Epub 2021 May 1.

Abstract

Background: Chronic alcohol consumption is more frequently associated with advanced, aggressive hepatocellular carcinoma (HCC) tumors. Alcohol adversely impacts ER/Golgi membrane trafficking and Golgi protein N-glycosylation in hepatocytes; these effects have been attributed (in part) to dysregulated adenosine diphosphate-ribosylation factor (ARF) GTPase signaling. Here, we investigated the role of the ARF GTPase guanine exchange factor PSD4 in HCC progression.

Methods: R-based bioinformatics analysis was performed on publicly available array data. Modulating gene expression was accomplished via lentiviral vectors. Gene expression was analyzed using quantitative real-time PCR and immunoblotting. PSD4 promoter methylation was assessed using quantitative methylation-specific PCR. Phospho-p65(S276)/DNMT1 binding to the PSD4 promoter was analyzed via chromatin immunoprecipitation. We constructed ethanol/DEN-induced and DEN only-induced transgenic murine models of HCC.

Results: We identified PSD4 as a hypermethylated, suppressed gene in alcohol-related HCC tumors; however, PSD4 was not dysregulated in all-cause HCC tumors. Certain HCC cell lines also displayed varying degrees of PSD4 downregulation. PSD4 overexpression or knockdown decreased and increased cell migration and invasiveness, respectively. Mechanistically, PSD4 transcription was repressed by TNF-α-induced phospho-p65(S276)'s recruitment of DNA methyltransferase 1 (DNMT1), resulting in PSD4 promoter methylation. PSD4 inhibited pro-EMT CDC42 activity, resulting in downregulation of E-cadherin and upregulation of N-cadherin and vimentin. Hepatocyte-specific PSD4 overexpression reduced ethanol/DEN-induced HCC tumor progression and EMT marker expression in vivo.

Conclusions: PSD4 is a hypermethylated, suppressed gene in alcohol-related HCC tumors that negatively modulated pro-EMT CDC42 activity. Furthermore, we present a novel phospho-NF-κB p65(S276)/DNMT1-mediated promoter methylation mechanism by which TNF-α/NF-κB signaling represses PSD4 transcription in HCC cells.

Keywords: EFA6B; HCC; PSD4; alcohol; p65.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Alcohol Drinking / genetics
  • Alcohols / adverse effects*
  • Animals
  • Cadherins / genetics
  • Carcinoma, Hepatocellular / genetics*
  • Carcinoma, Hepatocellular / pathology
  • Cell Line, Tumor
  • DNA (Cytosine-5-)-Methyltransferase 1 / genetics
  • DNA Methylation / genetics
  • Down-Regulation / genetics
  • Epigenesis, Genetic / genetics*
  • Female
  • Gene Expression Regulation, Neoplastic / genetics
  • Guanine Nucleotide Exchange Factors / genetics*
  • Hep G2 Cells
  • Hepatocytes / pathology
  • Humans
  • Liver Neoplasms / genetics*
  • Liver Neoplasms / pathology
  • Mice
  • Mice, Transgenic
  • NF-kappa B / genetics*
  • Pregnancy
  • Promoter Regions, Genetic / genetics
  • Signal Transduction / genetics
  • Transcription Factor RelA / genetics
  • Transcription, Genetic / genetics*
  • Tumor Necrosis Factor-alpha / genetics*

Substances

  • Alcohols
  • Cadherins
  • Guanine Nucleotide Exchange Factors
  • NF-kappa B
  • PSD4 protein, human
  • Transcription Factor RelA
  • Tumor Necrosis Factor-alpha
  • DNA (Cytosine-5-)-Methyltransferase 1