Abstract

Platelet-derived growth subunit A (PDGFA) plays critical roles in development of glioblastoma (GBM) with substantial evidence from TCGA database analyses and in vivo mouse models. So far, only platelet-derived growth receptor α (PDGFRA) has been identified as receptor for PDGFA. However, PDGFA and PDGFRA are categorized into different molecular subtypes of GBM in TCGA_GBM database. Our data herein further showed that activity or expression deficiency of PDGFRA did not effectively block PDGFA activity. Therefore, PDGFRA might be not necessary for PDGFA function.To profile proteins involved in PDGFA function, we performed co-immunoprecipitation (Co-IP) and Mass Spectrum (MS) and delineated the network of PDGFA-associated proteins for the first time. Unexpectedly, the data showed that EPHA2 could be temporally activated by PDGFA even without activation of PDGFRA and AKT. Furthermore, MS, Co-IP, in vitro binding thermodynamics, and proximity ligation assay consistently proved the interaction of EPHA2 and PDGFA. In addition, we observed that high expression of EPHA2 leaded to upregulation of PDGF signaling targets in TCGA_GBM database and clinical GBM samples. Co-upregulation of PDGFRA and EPHA2 leaded to worse patient prognosis and poorer therapeutic effects than other contexts, which might arise from expression elevation of genes related with malignant molecular subtypes and invasive growth. Due to PDGFA-induced EPHA2 activation, blocking PDGFRA by inhibitor could not effectively suppress proliferation of GBM cells, but simultaneous inhibition of both EPHA2 and PDGFRA showed synergetic inhibitory effects on GBM cells in vitro and in vivo. Taken together, our study provided new insights on PDGFA function and revealed EPHA2 as a potential receptor of PDGFA. EPHA2 might contribute to PDGFA signaling transduction in combination with PDGFRA and mediate the resistance of GBM cells to PDGFRA inhibitor. Therefore, combination of inhibitors targeting PDGFRA and EHA2 represented a promising therapeutic strategy for GBM treatment.

Highlights

  • Glioma is the most prevalent brain tumor and pathologically categorized into four grades (I–IV) by the 2016 World Health Organization (WHO) classification of central nervous system tumors.[1]

  • Since only PDGFRA has been identified as the receptor for PDGFA, we examined PDGFRA protein in a panel of glioma cells, including 2 primary glioblastoma multiforme (GBM) cells (091214 and 090116), 7 commercial GBM cell lines (A172, DBTRG05MG, LN18, LN229, T98G, U251, and U87), and 1 commercial grade III glioma cell line (SW1088)

  • Since PDGFRA was not detected in LN229 cell line, we constructed a PDGFRAoverexpression cell line using LN229 (LN229PDGFRA) to examine and MK2206. d PDGFA-induced temporal expression of indicated proteins in LN18 cells infected with lentivirus containing control sgRNA or sgRNA targeting PDGFRA. e PDGFA-induced temporal expression of indicated proteins in LN18 cells pre-treated with vehicle or IMA. f Representative immunofluorescence images stained by antibodies targeting EPHA2 and EEA1, respectively

Read more

Summary

Introduction

Glioma is the most prevalent brain tumor and pathologically categorized into four grades (I–IV) by the 2016 World Health Organization (WHO) classification of central nervous system tumors.[1]. RESULTS PDGFRA was not necessary for PDGFA signaling in GBM First, we analyzed four known PDGF genes in TCGA_GBM database through Kaplan–Meier survival analysis.

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