Eicosapentaenoic acid activates RAS/ERK/C/EBPβ pathway through H-Ras intron 1 CpG island demethylation in U937 leukemia cells

PLoS One. 2014 Jan 13;9(1):e85025. doi: 10.1371/journal.pone.0085025. eCollection 2014.

Abstract

Epigenetic alterations, including aberrant DNA methylation, contribute to tumor development and progression. Silencing of tumor suppressor genes may be ascribed to promoter DNA hypermethylation, a reversible phenomenon intensely investigated as potential therapeutic target. Previously, we demonstrated that eicosapentaenoic acid (EPA) exhibits a DNA demethylating action that promotes the re-expression of the tumor suppressor gene CCAAT/enhancer-binding protein δ (C/EBPδ). The C/EBPβ/C/EBPδ heterodimer formed appears essential for the monocyte differentiation commitment. The present study aims to evaluate the effect of EPA on RAS/extracellular signal regulated kinases (ERK1/2)/C/EBPβ pathway, known to be induced during the monocyte differentiation program. We found that EPA conditioning of U937 leukemia cells activated RAS/ERK/C/EBPβ pathway, increasing the C/EBPβ and ERK1/2 active phosphorylated forms. Transcriptional induction of the upstream activator H-Ras gene resulted in increased expression of H-Ras protein in the active pool of non raft membrane fraction. H-Ras gene analysis identified an hypermethylated CpG island in intron 1 that can affect the DNA-protein interaction modifying RNA polymerase II (RNAPII) activity. EPA treatment demethylated almost completely this CpG island, which was associated with an enrichment of active RNAPII. The increased binding of the H-Ras transcriptional regulator p53 to its consensus sequence within the intronic CpG island further confirmed the effect of EPA as demethylating agent. Our results provide the first evidence that an endogenous polyunsaturated fatty acid (PUFA) promotes a DNA demethylation process responsible for the activation of RAS/ERK/C/EBPβ pathway during the monocyte differentiation commitment. The new role of EPA as demethylating agent paves the way for studying PUFA action when aberrant DNA methylation is involved.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Azacitidine / pharmacology
  • Base Sequence
  • CCAAT-Enhancer-Binding Protein-delta / metabolism*
  • CpG Islands / genetics*
  • DNA Methylation / drug effects
  • DNA Methylation / genetics*
  • Eicosapentaenoic Acid / pharmacology*
  • Exons / genetics
  • Humans
  • Introns / genetics*
  • Leukemia / genetics*
  • Leukemia / pathology
  • MAP Kinase Signaling System / drug effects*
  • MAP Kinase Signaling System / genetics
  • Membrane Microdomains / drug effects
  • Membrane Microdomains / metabolism
  • Molecular Sequence Data
  • Phosphorylation / drug effects
  • Protein Binding / drug effects
  • Protein Binding / genetics
  • Protein Isoforms / metabolism
  • Proto-Oncogene Proteins p21(ras) / genetics*
  • RNA Polymerase II / metabolism
  • Transcription, Genetic / drug effects
  • Tumor Suppressor Protein p53 / metabolism
  • U937 Cells

Substances

  • Protein Isoforms
  • Tumor Suppressor Protein p53
  • CCAAT-Enhancer-Binding Protein-delta
  • Eicosapentaenoic Acid
  • RNA Polymerase II
  • HRAS protein, human
  • Proto-Oncogene Proteins p21(ras)
  • Azacitidine

Grants and funding

This research was supported by grants from Ministero delle Politiche Agricole, Alimentari e Forestali to LB, Italian Association for Cancer Research (AIRC) to FG (project IG-9390), and Fondazione Cassa di Risparmio di Perugia to CR (project 2012.0137.021). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.