Abstract

Radioresistance‐induced residual and recurrent tumours are the main cause of treatment failure in nasopharyngeal carcinoma (NPC). Thus, the mechanisms of NPC radioresistance and predictive markers of NPC prognosis and radioresistance need to be investigated and identified. In this study, we identified RPA3 as a candidate radioresistance marker using RNA‐seq of NPC samples. In vitro studies further confirmed that RPA3 affected the radiosensitivity of NPC cells. Specifically, the overexpression of RPA3 enhanced radioresistance and the capacity for DNA repair of NPC cells, whereas inhibiting RPA3 expression sensitized NPC cells to irradiation and decreased the DNA repair capacity. Furthermore, the overexpression of RPA3 enhanced RAD51 foci formation in NPC cells after irradiation. Immunohistochemical assays in 104 NPC specimens and 21 normal epithelium specimens indicated that RPA3 was significantly up‐regulated in NPC tissues, and a log‐rank test suggested that in patients with NPC, high RPA3 expression was associated with shorter overall survival (OS) and a higher recurrence rate compared with low expression (5‐year OS rates: 67.2% versus 86.2%; 5‐year recurrence rates: 14.8% versus 2.3%). Moreover, TCGA data also indicated that high RPA3 expression correlated with poor OS and a high recurrence rate in patients with head and neck squamous cell carcinoma (HNSC) after radiotherapy. Taken together, the results of our study demonstrated that RPA3 regulated the radiosensitivity and DNA repair capacity of NPC cells. Thus, RPA3 may serve as a new predictive biomarker for NPC prognosis and radioresistance to help guide the diagnosis and individualized treatment of patients with NPC.

Highlights

  • Nasopharyngeal carcinoma (NPC) originates from epithelial cells [1] and has a unique endemic distribution

  • RNA-seq showed that RPA3 was high in radioresistant NPC samples (Fig. 1C)

  • We analysed the gene expression data of head and neck squamous cell carcinoma (HNSC) cases in the the Cancer Genome Atlas (TCGA) data set, which suggested that RPA1 and RPA3 were up-regulated in HNSC tissues compared with non-tumour tissues (Fig. 1D and F), whereas RPA2 expression did not significantly differ between HNSC and nontumour tissues (Fig. 1E)

Read more

Summary

Introduction

Nasopharyngeal carcinoma (NPC) originates from epithelial cells [1] and has a unique endemic distribution. Unlike for most other malignancies, radiotherapy is the principal treatment for primary NPC lesions because of the anatomical location and radiosensitivity of this tumour [4, 5]. Population screening and effective systemic agents have significantly decreased the mortality of NPC. The application of intensity-modulated radiotherapy (IMRT) has greatly improved tumour control and reduced toxic effects [4]. Replication protein A (RPA) is a nuclear ssDNA-binding protein (SSB) complex that consists of three subunits of RPA proteins, RPA1 (70 kD), RPA2 (32 kD) and RPA3 (14 kD), which are encoded by three separate genes [8,9,10].

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