The polyphenols (-)-epigallocatechin-3-gallate and luteolin synergistically inhibit TGF-β-induced myofibroblast phenotypes through RhoA and ERK inhibition

PLoS One. 2014 Oct 1;9(10):e109208. doi: 10.1371/journal.pone.0109208. eCollection 2014.

Abstract

The presence of reactive stroma, predominantly composed of myofibroblasts, is directly associated with and drives prostate cancer progression. We have previously shown that (-)-Epigallocatechin-3-gallate (EGCG), in the form of Polyphenon E, significantly decreases serum levels of HGF and VEGF in prostate cancer patients. Given that HGF and VEGF are secreted from surrounding tumor myofibroblasts, these observations suggested that EGCG may inhibit prostate cancer-associated myofibroblast differentiation. Herein, we demonstrate that micromolar combinations of EGCG and a second polyphenol, luteolin, synergistically inhibit TGF-β-induced myofibroblast phenotypes in prostate fibroblast cell lines, as observed primarily by potentiation of fibronectin expression. Functionally, EGCG and luteolin inhibited TGF-β-induced extracellular matrix contraction, an enhancer of tumor cell invasion. EGCG and luteolin inhibited downstream TGF-β-induced signaling, including activation of ERK and AKT, respectively, but mechanistically, only ERK appeared to be necessary for TGF-β-induced fibronectin expression. Furthermore, neither EGCG nor luteolin affected Smad signaling or nuclear translocation. Rho signaling was found to be necessary for TGF-β-induced fibronectin expression and EGCG and luteolin each reduced RhoA activation. Finally, EGCG and luteolin were shown to reverse TGF-β-induced fibronectin expression, implicating that these natural compounds may be useful not only in preventing but also in treating already activated myofibroblasts and the diseases they cause, including cancer. The ability of EGCG and luteolin to synergistically target myofibroblasts suggests that combined clinical use of these compounds could prevent or reverse cancer progression through targeting the tumor microenvironment, in addition to the tumor itself.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Catechin / analogs & derivatives*
  • Catechin / pharmacology
  • Cell Line
  • Drug Synergism
  • Extracellular Signal-Regulated MAP Kinases / adverse effects*
  • Humans
  • Luteolin / pharmacology*
  • Myofibroblasts / cytology
  • Myofibroblasts / drug effects*
  • Myofibroblasts / metabolism
  • Phenotype
  • Transforming Growth Factor beta / antagonists & inhibitors*
  • rhoA GTP-Binding Protein / adverse effects*

Substances

  • Transforming Growth Factor beta
  • Catechin
  • epigallocatechin gallate
  • Extracellular Signal-Regulated MAP Kinases
  • rhoA GTP-Binding Protein
  • Luteolin

Grants and funding

This work was funded by a Feist-Weiller Cancer Center Idea Award. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.