1. Academic Validation
  2. EPHA2 mediates PDGFA activity and functions together with PDGFRA as prognostic marker and therapeutic target in glioblastoma

EPHA2 mediates PDGFA activity and functions together with PDGFRA as prognostic marker and therapeutic target in glioblastoma

  • Signal Transduct Target Ther. 2022 Feb 2;7(1):33. doi: 10.1038/s41392-021-00855-2.
Qu-Jing Gai  # 1 Zhen Fu  # 1 Jiang He  # 1 Min Mao 1 Xiao-Xue Yao 1 Yan Qin 1 Xi Lan 1 Lin Zhang 1 Jing-Ya Miao 1 Yan-Xia Wang 1 Jiang Zhu 1 Fei-Cheng Yang 1 Hui-Min Lu 1 2 Ze-Xuan Yan 1 Fang-Lin Chen 1 3 Yu Shi 1 Yi-Fang Ping 1 You-Hong Cui 1 Xia Zhang 1 Xindong Liu 1 Xiao-Hong Yao 1 Sheng-Qing Lv 4 Xiu-Wu Bian 5 Yan Wang 6
Affiliations

Affiliations

  • 1 Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
  • 2 Biobank of Institute of Pathology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
  • 3 Institute of Cancer, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
  • 4 Department of Neurosurgery, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China. [email protected].
  • 5 Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China. [email protected].
  • 6 Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China. [email protected].
  • # Contributed equally.
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.

Figures
Products