Abstract

We studied the potential mechanisms of valproic acid (VPA) in the treatment of glioblastoma multiforme (GBM). Using the human U87, GBM8401, and DBTRG-05MG GBM-derived cell lines, VPA at concentrations of 5 to 20 mM induced G2/M cell cycle arrest and increased the production of reactive oxygen species (ROS). Stress-related molecules such as paraoxonase 2 (PON2), cyclin B1, cdc2, and Bcl-xL were downregulated, but p27, p21 and Bim were upregulated by VPA treatment. VPA response element on the PON2 promoter was localized at position -400/−1. PON2 protein expression was increased in GBM cells compared with normal brain tissue and there was a negative correlation between the expression of PON2 and Bim. These findings were confirmed by the public Bredel GBM microarray (Gene Expression Omnibus accession: GSE2223) and the Cancer Genome Atlas GBM microarray datasets. Overexpression of PON2 in GBM cells significantly decreased intracellular ROS levels, and PON2 expression was decreased after VPA stimulation compared with controls. Bim expression was significantly induced by VPA in GBM cells with PON2 silencing. These observations were further shown in the subcutaneous GBM8401 cell xenograft of BALB/c nude mice. Our results suggest that VPA reduces PON2 expression in GBM cells, which in turn increases ROS production and induces Bim production that inhibits cancer progression via the PON2–Bim cascade.

Highlights

  • Glioblastoma multiforme (GBM) is the most malignant primary brain tumor with infiltrative growth characteristics

  • Our results suggest that valproic acid (VPA) reduces paraoxonase 2 (PON2) expression in glioblastoma multiforme (GBM) cells, which in turn increases reactive oxygen species (ROS) production and induces Bim production that inhibits cancer progression via the PON2–Bim cascade

  • Our results indicate that VPA suppresses cell growth via the PON2–Bim cascade in GBM cells

Read more

Summary

Introduction

Glioblastoma multiforme (GBM) is the most malignant primary brain tumor with infiltrative growth characteristics. VPA was further shown to inhibit histone deacetylase (HDAC) activity causing impaired epigenetic modification and suppressed cell growth [4]. Several studies revealed that VPA sensitized GBM cells to chemotherapy and radiotherapy by increased www.impactjournals.com/oncotarget cell apoptosis, which involved increased p21 expression and cell cycle arrest, suppression of DNA double strand break repair, and activating pro-apoptotic signaling [12,13,14,15,16]. Overproduction of ROS and antioxidant system defect result in DNA repair impairment and gene expression alteration, contributing to the carcinogenesis process [17, 18]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call