REDD1 overexpression in oral squamous cell carcinoma may predict poor prognosis and correlates with high microvessel density

Oncol Lett. 2020 Jan;19(1):431-441. doi: 10.3892/ol.2019.11070. Epub 2019 Nov 8.

Abstract

The association between the hypoxia-inducible gene termed regulated in development and DNA damage responses 1 (REDD1) and microvessel density (MVD) in human oral cancer has rarely been reported. The present study aimed to explore REDD1 expression in oral squamous cell carcinoma (OSCC), its clinical prognostic significance and its correlation with angiogenesis. REDD1 expression in 23 pairs of fresh-frozen OSCC and matched peritumoral mucosal tissues was quantified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting. Furthermore, 74 formalin-fixed paraffin-embedded OSCC tissues were collected to detect REDD1 expression and CD34-positive MVD by immunohistochemistry (IHC). The association between REDD1 expression and MVD, patients' clinicopathological characteristics and cancer-associated survival rate was also evaluated using the log-rank (Mantel-Cox) test. The results from RT-qPCR and western blotting demonstrated that REDD1 expression was significantly higher in OSCC tissues compared with peritumoral mucosal tissues (P<0.05). In addition, the results from IHC revealed that REDD1 expression was higher in OSCC tissues compared with peritumoral tissues. Furthermore, REDD1 expression was associated with advanced clinical stage, poorer tumor differentiation, lymphatic metastasis and tumor recurrence (P=0.000, P=0.003, P=0.006 and P<0.001, respectively). Additionally, REDD1 overexpression was positively correlated with MVD (r=0.7316; P<0.001). The results from Kaplan-Meier survival analysis demonstrated a significantly reduced disease-free survival and overall survival in patients with OSCC and high REDD1 expression (P<0.001). REDD1 may therefore serve as a novel prognostic biomarker, a key regulatory checkpoint that could coordinate angiogenesis and a new therapeutic target for patients with OSCC.

Keywords: angiogenesis; hypoxia; oral squamous cell carcinoma; prognosis; regulated in development and DNA damage responses 1.