Abstract

Glioblastoma (GBM) is the most common and lethal brain tumor in adults. Ionizing radiation (IR) is a standard treatment for GBM patients and results in DNA damage. However, the clinical efficacy of IR is limited due to therapeutic resistance. The programmed death ligand 1 (PD-L1) blockade has a shown the potential to increase the efficacy of radiotherapy by inhibiting DNA damage and repair responses. The miR-33a-5p is an essential microRNA that promotes GBM growth and self-renewal. In this study, we investigated whether a PD-L1 inhibitor (a small molecule inhibitor) exerted radio-sensitive effects to impart an anti-tumor function in GBM cells by modulating miR-33a-5p. U87 MG cells and U251 cells were pretreated with PD-L1 inhibitor. The PD-L1 inhibitor-induced radio-sensitivity in these cells was assessed by assaying cellular apoptosis, clonogenic survival assays, and migration. TargetScan and luciferase assay showed that miR-33a-5p targeted the phosphatase and tensin homolog (PTEN) 3′ untranslated region. The expression level of PTEN was measured by western blotting, and was also silenced using small interfering RNAs. The levels of DNA damage following radiation was measured by the presence of γ-H2AX foci, cell cycle, and the mRNA of the DNA damage-related genes, BRCA1, NBS1, RAD50, and MRE11. Our results demonstrated that the PD-L1 inhibitor significantly decreased the expression of the target gene, miR-33a-5p. In addition, pretreatment of U87 MG and U251 cells with the PD-L1 inhibitor increased radio-sensitivity, as indicated by increased apoptosis, while decreased survival and migration of GBM cells. Mir-33a-5p overexpression or silencing PTEN in U87 MG and U251 cells significantly attenuated PD-L1 radiosensitive effect. Additionally, PD-L1 inhibitor treatment suppressed the expression of the DNA damage response-related genes, BRCA1, NBS1, RAD50, and MRE11. Our results demonstrated a novel role for the PD-L1 inhibitor in inducing radio- sensitivity in GBM cells, where inhibiting miR-33a-5p, leading to PTEN activated, and inducing DNA damage was crucial for antitumor immunotherapies to treat GBM.

Highlights

  • Glioblastomas (GBM) are one of the most treatment-resistant tumors, often recurring after chemotherapy and radiation treatment [1]

  • To determine whether the programmed death ligand 1 (PD-L1) inhibitor would radiosensitize U87 MG cells and U251 cells, we first examined the expression of PD-L1 in the U87 MG cells and U251 cells under radiotherapy conditions

  • We found that the expression of specific miRNAs in–PD-L1 inhibitor treated before radiated U87 MG cells was significantly altered when compared with that in only radiated cells

Read more

Summary

Introduction

Glioblastomas (GBM) are one of the most treatment-resistant tumors, often recurring after chemotherapy and radiation treatment [1]. Amount of effort has been taken to identify therapeutics that radio-sensitize GBMs because most patients will receive radiation treatment [2]. It’s difficult to identify such radiosensitive chemotherapeutic agents because GBMs exhibit redundant pro-growth and pro-survival pathways, leading to chemotherapy resistance [3]. To overcome such resistance, it’s needed to devise therapeutic strategies targeting such redundant treatment-resistant pathways to increase the radiosensitivity of GBMs. Programmed death ligand 1 (PD-L1)regulates the immune system by binding the programmed cell death protein 1 (PD-1) receptor as an immune checkpoint protein [4, 5]. The impact of the PD-L1 blockade on radiosensitization has been suggested in GBM, its role has yet to be fully elucidated

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