Epigenetic Co-Deregulation of EZH2/TET1 is a Senescence-Countering, Actionable Vulnerability in Triple-Negative Breast Cancer

Theranostics. 2019 Jan 24;9(3):761-777. doi: 10.7150/thno.29520. eCollection 2019.

Abstract

Triple-negative breast cancer (TNBC) cells lack the expression of ER, PR and HER2. Thus, TNBC patients cannot benefit from hormone receptor-targeted therapy as non-TNBC patients, but can only receive chemotherapy as the systemic treatment and have a worse overall outcome. More effective therapeutic targets and combination therapy strategies are urgently needed to improve the treatment effectiveness. Methods: We analyzed the expression levels of EZH2 and TET1 in TCGA and our own breast cancer patient cohort, and tested their correlation with patient survival. We used TNBC and non-TNBC cell lines and mouse xenograft tumor model to unveil novel EZH2 targets and investigated the effect of EZH2 inhibition or TET1 overexpression in cell proliferation and viability of TNBC cells. Results: In TNBC cells, EZH2 decreases TET1 expression by H3K27me3 epigenetic regulation and subsequently suppresses anti-tumor p53 signaling pathway. Patients with high EZH2 and low TET1 presented the poorest survival outcome. Experimentally, targeting EZH2 in TNBC cells with specific inhibitor GSK343 or shRNA genetic approach could induce cell cycle arrest and senescence by elevating TET1 expression and p53 pathway activation. Using mouse xenograft model, we have tested a novel therapy strategy to combine GSK343 and chemotherapy drug Adriamycin and could show drastic and robust inhibition of TNBC tumor growth by synergistic induction of senescence and apoptosis. Conclusions: We postulate that the well-controlled dynamic pathway EZH2-H3K27me3-TET1 is a novel epigenetic co-regulator module and provide evidence regarding how to exploit it as a novel therapeutic target via its pivotal role in senescence and apoptosis control. Of clinical and therapeutic significance, the present study opens a new avenue for TNBC treatment by targeting the EZH2-H3K27me3-TET1 pathway that can modulate the epigenetic landscape.

Keywords: EZH2; TET1; TNBC; cellular senescence; epigenetic co-deregulation.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antibiotics, Antineoplastic / administration & dosage
  • Antibiotics, Antineoplastic / therapeutic use*
  • Apoptosis / drug effects
  • Apoptosis / genetics
  • Cancer Survivors
  • Cell Line, Tumor
  • Cellular Senescence / drug effects
  • Cellular Senescence / genetics
  • DNA Methylation
  • DNA, Neoplasm / metabolism
  • Doxorubicin / administration & dosage
  • Doxorubicin / therapeutic use*
  • Drug Delivery Systems
  • Enhancer of Zeste Homolog 2 Protein / antagonists & inhibitors
  • Enhancer of Zeste Homolog 2 Protein / genetics*
  • Epigenesis, Genetic*
  • Female
  • Gene Expression Regulation, Neoplastic* / drug effects
  • Humans
  • Indazoles / pharmacology
  • Mice
  • Mice, Nude
  • Mixed Function Oxygenases / antagonists & inhibitors
  • Mixed Function Oxygenases / genetics*
  • Proto-Oncogene Proteins / antagonists & inhibitors
  • Proto-Oncogene Proteins / genetics*
  • Pyridones / pharmacology
  • Triple Negative Breast Neoplasms / genetics*
  • Xenograft Model Antitumor Assays

Substances

  • Antibiotics, Antineoplastic
  • DNA, Neoplasm
  • GSK343
  • Indazoles
  • Proto-Oncogene Proteins
  • Pyridones
  • Doxorubicin
  • Mixed Function Oxygenases
  • TET1 protein, human
  • EZH2 protein, human
  • Enhancer of Zeste Homolog 2 Protein