Estrogen receptor beta enhances chemotherapy response of GBM cells by down regulating DNA damage response pathways

Sci Rep. 2019 Apr 16;9(1):6124. doi: 10.1038/s41598-019-42313-8.

Abstract

Glioblastoma (GBM) is the most commonly diagnosed brain tumor that exhibit high mortality rate and chemotherapy resistance is a major clinical problem. Recent studies suggest that estrogen receptor beta (ERβ), may function as a tumor suppressor in GBM. However, the mechanism(s) by which ERβ contributes to GBM suppression and chemotherapy response remains unknown. We examined the role of ERβ in the DNA damage response of GBM cells, and tested whether ERβ sensitizes GBM cells to chemotherapy. Cell viability and survival assays using multiple epitope tagged ERβ expressing established and primary GBM cells demonstrated that ERβ sensitizes GBM cells to DNA damaging agents including temozolomide (TMZ). RNA-seq studies using ERβ overexpression models revealed downregulation of number of genes involved in DNA recombination and repair, ATM signaling and cell cycle check point control. Gene set enrichment analysis (GSEA) suggested that ERβ-modulated genes were correlated negatively with homologous recombination, mismatch repair and G2M checkpoint genes. Further, RT-qPCR analysis revealed that chemotherapy induced activation of cell cycle arrest and apoptosis genes were attenuated in ERβKO cells. Additionally, ERβ overexpressing cells had a higher number of γH2AX foci following TMZ treatment. Mechanistic studies showed that ERβ plays an important role in homologous recombination (HR) mediated repair and ERβ reduced expression and activation of ATM upon DNA damage. More importantly, GBM cells expressing ERβ had increased survival when compared to control GBM cells in orthotopic GBM models. ERβ overexpression further enhanced the survival of mice to TMZ therapy in both TMZ sensitive and TMZ resistant GBM models. Additionally, IHC analysis revealed that ERβ tumors had increased expression of γH2AX and cleaved caspase-3. Using ERβ-overexpression and ERβ-KO GBM model cells, we have provided the evidence that ERβ is required for optimal chemotherapy induced DNA damage response and apoptosis in GBM cells.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antineoplastic Agents, Alkylating / pharmacology*
  • Antineoplastic Agents, Alkylating / therapeutic use
  • Apoptosis / drug effects
  • Ataxia Telangiectasia Mutated Proteins / metabolism
  • Brain Neoplasms / drug therapy*
  • Brain Neoplasms / genetics
  • Brain Neoplasms / pathology
  • Cell Cycle Checkpoints / drug effects
  • Cell Cycle Checkpoints / genetics
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • DNA Damage / drug effects
  • Down-Regulation
  • Drug Resistance, Neoplasm / genetics
  • Estrogen Receptor beta / genetics
  • Estrogen Receptor beta / metabolism*
  • Gene Expression Regulation, Neoplastic
  • Gene Knockout Techniques
  • Glioblastoma / drug therapy*
  • Glioblastoma / genetics
  • Glioblastoma / pathology
  • Histones / metabolism
  • Humans
  • Male
  • Mice
  • RNA-Seq
  • Recombinational DNA Repair
  • Signal Transduction
  • Temozolomide / pharmacology
  • Temozolomide / therapeutic use
  • Xenograft Model Antitumor Assays

Substances

  • Antineoplastic Agents, Alkylating
  • ESR2 protein, human
  • Estrogen Receptor beta
  • H2AX protein, human
  • Histones
  • ATM protein, human
  • Ataxia Telangiectasia Mutated Proteins
  • Temozolomide