16
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

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

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          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.

          Related collections

          Most cited references42

          • Record: found
          • Abstract: found
          • Article: not found

          Hypoxia signalling in cancer and approaches to enforce tumour regression.

          Tumour cells emerge as a result of genetic alteration of signal circuitries promoting cell growth and survival, whereas their expansion relies on nutrient supply. Oxygen limitation is central in controlling neovascularization, glucose metabolism, survival and tumour spread. This pleiotropic action is orchestrated by hypoxia-inducible factor (HIF), which is a master transcriptional factor in nutrient stress signalling. Understanding the role of HIF in intracellular pH (pH(i)) regulation, metabolism, cell invasion, autophagy and cell death is crucial for developing novel anticancer therapies. There are new approaches to enforce necrotic cell death and tumour regression by targeting tumour metabolism and pH(i)-control systems.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex.

            Mammalian target of rapamycin (mTOR) is a central regulator of protein synthesis whose activity is modulated by a variety of signals. Energy depletion and hypoxia result in mTOR inhibition. While energy depletion inhibits mTOR through a process involving the activation of AMP-activated protein kinase (AMPK) by LKB1 and subsequent phosphorylation of TSC2, the mechanism of mTOR inhibition by hypoxia is not known. Here we show that mTOR inhibition by hypoxia requires the TSC1/TSC2 tumor suppressor complex and the hypoxia-inducible gene REDD1/RTP801. Disruption of the TSC1/TSC2 complex through loss of TSC1 or TSC2 blocks the effects of hypoxia on mTOR, as measured by changes in the mTOR targets S6K and 4E-BP1, and results in abnormal accumulation of Hypoxia-inducible factor (HIF). In contrast to energy depletion, mTOR inhibition by hypoxia does not require AMPK or LKB1. Down-regulation of mTOR activity by hypoxia requires de novo mRNA synthesis and correlates with increased expression of the hypoxia-inducible REDD1 gene. Disruption of REDD1 abrogates the hypoxia-induced inhibition of mTOR, and REDD1 overexpression is sufficient to down-regulate S6K phosphorylation in a TSC1/TSC2-dependent manner. Inhibition of mTOR function by hypoxia is likely to be important for tumor suppression as TSC2-deficient cells maintain abnormally high levels of cell proliferation under hypoxia.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Hypoxia regulates TSC1/2-mTOR signaling and tumor suppression through REDD1-mediated 14-3-3 shuttling.

              Hypoxia induces rapid and dramatic changes in cellular metabolism, in part through inhibition of target of rapamycin (TOR) kinase complex 1 (TORC1) activity. Genetic studies have shown the tuberous sclerosis tumor suppressors TSC1/2 and the REDD1 protein to be essential for hypoxia regulation of TORC1 activity in Drosophila and in mammalian cells. The molecular mechanism and physiologic significance of this effect of hypoxia remain unknown. Here, we demonstrate that hypoxia and REDD1 suppress mammalian TORC1 (mTORC1) activity by releasing TSC2 from its growth factor-induced association with inhibitory 14-3-3 proteins. Endogenous REDD1 is required for both dissociation of endogenous TSC2/14-3-3 and inhibition of mTORC1 in response to hypoxia. REDD1 mutants that fail to bind 14-3-3 are defective in eliciting TSC2/14-3-3 dissociation and mTORC1 inhibition, while TSC2 mutants that do not bind 14-3-3 are inactive in hypoxia signaling to mTORC1. In vitro, loss of REDD1 signaling promotes proliferation and anchorage-independent growth under hypoxia through mTORC1 dysregulation. In vivo, REDD1 loss elicits tumorigenesis in a mouse model, and down-regulation of REDD1 is observed in a subset of human cancers. Together, these findings define a molecular mechanism of signal integration by TSC1/2 that provides insight into the ability of REDD1 to function in a hypoxia-dependent tumor suppressor pathway.
                Bookmark

                Author and article information

                Journal
                Oncol Lett
                Oncol Lett
                OL
                Oncology Letters
                D.A. Spandidos
                1792-1074
                1792-1082
                January 2020
                08 November 2019
                08 November 2019
                : 19
                : 1
                : 431-441
                Affiliations
                [1 ]Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
                [2 ]Department of Pathology, Basic Medical College of Qingdao University, Qingdao, Shandong 266071, P.R. China
                Author notes
                Correspondence to: Dr Yuanyong Feng, Department of Oral and Maxillofacial Surgery, School of Stomatology and The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong 266003, P.R. China, E-mail: feng_yuanyong@ 123456163.com
                Dr Ning Wang, Department of Pathology, Basic Medical College of Qingdao University, 307 Ningxia Road, Qingdao, Shandong 266071, P.R. China, E-mail: ningwang7903@ 123456gmail.com
                Article
                OL-0-0-11070
                10.3892/ol.2019.11070
                6923876
                31897156
                f9f6ebcf-58d7-42b2-83af-40e56eb2f72d
                Copyright: © Feng et al.

                This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.

                History
                : 09 May 2019
                : 11 October 2019
                Categories
                Articles

                Oncology & Radiotherapy
                oral squamous cell carcinoma,hypoxia,regulated in development and dna damage responses 1,angiogenesis,prognosis

                Comments

                Comment on this article