Abstract

Glioblastoma (GBM) is a common and refractory subtype of high-grade glioma with a poor prognosis. The epithelial-mesenchymal transition (EMT) is an important cause of enhanced glioblastoma invasiveness and tumor recurrence. Our previous study found that retinoic acid receptor-related orphan receptor A (RORA) is a nuclear receptor and plays an important role in inhibiting proliferation and tumorigenesis of glioma. We further confirmed RORA was downregulated in GBM. Thus, we determined whether RORA was involved in the migration, invasion, and EMT of GBM. Human GBM cell lines, U87 and T98G, and patient-derived glioma stem cells (GSCs), GSC2C and GSC4D, were used for in vitro and in vivo experiments. The expressions of RORA, CASC2, and EIF4A3 in GBM cells and GSCs were detected by RT-qPCR and western blotting. The biological effects of RORA, CASC2, and EIF4A3 on GBM migration, invasion, and EMT were evaluated using the migration assay, transwell assay, immunofluorescence staining, and xenograft experiments. We found that RORA inhibited the migration, invasion, and EMT of GBM. CASC2 could bind to, maintain the stability, and promote the nuclear translocation of RORA protein. EIF4A3 could downregulate CASC2 expression via inducing its cleavage, while RORA transcriptionally inhibited EIF4A3 expression, which formed a feedback loop among EIF4A3/CASC2/RORA. Moreover, gene set enrichment analysis (GSEA) and in vitro and in vivo experiments showed RORA inhibited the aggressiveness of GBM by negatively regulating the TGF-β1/Smad signaling pathway. Therefore, The EIF4A3/CASC2/RORA feedback loop regulated TGF-β1/Smad signaling pathway might become a promising therapeutic strategy for GBM treatment.

Highlights

  • Glioblastoma (GBM) is the most deadly and common tumor in the central nervous system [1, 2]

  • The results showed that receptor A (RORA) levels presented in the nucleus and cytoplasm were obviously decreased in Cancer susceptibility candidate 2 (CASC2)-silenced GSC2C, while an increased RORA level was presented after CASC2 overexpression in GSC4D

  • The results showed that tumor volumes were decreased in the RORA-OE group, the CASC2-KD1+RORA-OE group, and the EIF4A3OE+RORA-OE group when compared with the control group, while the tumor volumes were significantly increased in the CASC2-KD1 group and the Eukaryotic initiation factor 4A3 (EIF4A3)-OE group compared with the control group (Figures 7A, B)

Read more

Summary

Introduction

Glioblastoma (GBM) is the most deadly and common tumor in the central nervous system [1, 2]. Identifying genes that play an important role in GBM may provide new options for treating this disorder. Previous studies have shown downregulations of RORA expression in various tumor tissues, such as breast, lung, prostate, and colorectal cancer [7]. It has been reported that the epithelial-mesenchymal transition (EMT) is an important cause of enhanced GBM invasiveness and tumor recurrence [8]. Our previous study showed that RORA played an anti-tumor role and inhibited the tumorigenesis and proliferation in glioma [9]. The possible role and effects of RORA in the migration, invasion, and EMT of GBM were identified

Objectives
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