Skip to main content
Log in

RRAD inhibits aerobic glycolysis, invasion, and migration and is associated with poor prognosis in hepatocellular carcinoma

  • Original Article
  • Published:
Tumor Biology

Abstract

Hepatocellular carcinoma (HCC) is one of the most prevalent and lethal cancer worldwide. However, the mechanism underlying the HCC development remains unclear. Ras-related associated with diabetes (RRAD) is a small Ras-related GTPase which has been implicated in metabolic disease and several types of cancer, yet its functions in HCC remain unknown. A tissue microarray constructed by 90 paired HCC tissues and adjacent non-cancerous liver tissues was used to examine the protein levels of RRAD, and the messenger RNA (mRNA) expression of RRAD was also detected in a subset of this cohort. The prognostic significance of RRAD was estimated by the Kaplan-Meier analysis and Cox regression. The glucose utilization assay and lactate production assay were performed to measure the role of RRAD in HCC glycolysis. The effect of RRAD in HCC invasion and metastasis was analyzed by transwell assays. Our results suggested that the expression of RRAD was downregulated in HCC tissues compared to the adjacent non-tumorous liver tissues both in mRNA and protein levels and lower RRAD expression served as an independent prognostic indicator for the survival of HCC patients. Moreover, RRAD inhibited hepatoma cell aerobic glycolysis by negatively regulating the expression of glucose transporter 1 (GLUT1) and hexokinase II (HK-II). In addition, RRAD inhibition dramatically increased hepatoma cell invasion and metastasis. In conclusion, our study revealed that RRAD expression was decreased in HCC tumor tissues and predicted poor clinical outcome for HCC patients and played an important role in regulating aerobic glycolysis and cell invasion and metastasis and may represent potential targets for improving the treatment of HCC.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. David F, Jacques F, The global and regional burden of cancer, Bernard WS, Christopher PD. World Cancer Report 2014. Paris: International Agency for Research on Cancer; 2014. p. 16–53.

    Google Scholar 

  2. Bosch FX, Ribes J, Díaz M, Cléries R. Primary liver cancer: worldwide incidence and trends. Gastroenterology. 2004;127:S5–S16.

    Article  PubMed  Google Scholar 

  3. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.

    Article  CAS  PubMed  Google Scholar 

  4. Lincet H, Icard P. How do glycolytic enzymes favour cancer cell proliferation by nonmetabolic functions? Oncogene. 2015;34:3751–9.

    Article  CAS  PubMed  Google Scholar 

  5. Phan LM, Yeung SC, Lee MH. Cancer metabolic reprogramming: importance, main features, and potentials for precise targeted anti-cancer therapies. Cancer Biol Med. 2014;11:1–19.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Han T, Kang D, Ji D, Wang X, Zhan W, Fu M, et al. How does cancer cell metabolism affect tumor migration and invasion? Cell Adhes Migr. 2013;7:395–403.

    Article  Google Scholar 

  7. Reynet C, Kahn CR. Rad: a member of the Ras family overexpressed in muscle of type II diabetic humans. Science. 1993;262:1441–4.

    Article  CAS  PubMed  Google Scholar 

  8. Moyers JS, Bilan PJ, Reynet C, Kahn CR. Overexpression of Rad inhibits glucose uptake in cultured muscle and fat cells. J Biol Chem. 1996;271:23111–6.

    Article  CAS  PubMed  Google Scholar 

  9. Downward J. Regulatory mechanisms for ras proteins. BioEssays. 1992;14:177–84.

    Article  CAS  PubMed  Google Scholar 

  10. Ridley AJ, Hall A. The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell. 1992;70:389–99.

    Article  CAS  PubMed  Google Scholar 

  11. Zhu J, Bilan PJ, Moyers JS, Antonetti DA, Kahn CR. Rad, a novel Ras-related GTPase, interacts with skeletal muscle beta-tropomyosin. J Biol Chem. 1996;271:768–73.

    Article  CAS  PubMed  Google Scholar 

  12. Suzuki M, Shigematsu H, Shames DS, Sunaga N, Takahashi T, Shivapurkar N, et al. Methylation and gene silencing of the Ras-related GTPase gene in lung and breast cancers. Ann Surg Oncol. 2007;14:1397–404.

    Article  PubMed  Google Scholar 

  13. Mo Y, Midorikawa K, Zhang Z, Zhou X, Ma N, Huang G, et al. Promoter hypermethylation of Ras-related GTPase gene RRAD inactivates a tumor suppressor function in nasopharyngeal carcinoma. Cancer Lett. 2012;323:147–54.

    Article  CAS  PubMed  Google Scholar 

  14. Dai B, Ruan B, Wu J, Wang J, Shang R, Sun W, et al. Insulin-like growth factor binding protein-1 inhibits cancer cell invasion and is associated with poor prognosis in hepatocellular carcinoma. Int J Clin Exp Pathol. 2014;7:5645–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Xia L, Mo P, Huang W, Zhang L, Wang Y, Zhu H, et al. The TNF-alpha/ROS/HIF-1-induced upregulation of FoxMI expression promotes HCC proliferation and resistance to apoptosis. Carcinogenesis. 2012;33:2250–9.

    Article  CAS  PubMed  Google Scholar 

  16. Hsiao BY, Chen CC, Hsieh PC, Chang TK, Yeh YC, Wu YC, et al. Rad is a p53 direct transcriptional target that inhibits cell migration and is frequently silenced in lung carcinoma cells. J Mol Med (Berl). 2011;89:481–92.

    Article  CAS  Google Scholar 

  17. Yeom SY, Nam DH, Park C. RRAD promotes EGFR-mediated STAT3 activation and induces temozolomide resistance of malignant glioblastoma. Mol Cancer Ther. 2014;13:3049–61.

    Article  CAS  PubMed  Google Scholar 

  18. Lee I, Yeom SY, Lee SJ, Kang WK, Park C. A novel senescence-evasion mechanism involving Grap2 and cyclin D interacting protein inactivation by Ras associated with diabetes in cancer cells under doxorubicin treatment. Cancer Res. 2010;70:4357–65.

    Article  CAS  PubMed  Google Scholar 

  19. Wang Y, Li G, Mao F, Li X, Liu Q, Chen L, et al. Ras-induced epigenetic inactivation of the RRAD (Ras-related associated with diabetes) gene promotes glucose uptake in a human ovarian cancer model. J Biol Chem. 2014;289:14225–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhang C, Liu J, Wu R, Liang Y, Lin M, Liu J, et al. Tumor suppressor p53 negatively regulates glycolysis stimulated by hypoxia through its target RRAD. Oncotarget. 2014;5:5535–46.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Szablewski L. Expression of glucose transporters in cancers. Biochim Biophys Acta. 1835;2013:164–9.

    Google Scholar 

  22. Amann T, Hellerbrand C. GLUT1 as a therapeutic target in hepatocellular carcinoma. Expert Opin Ther Targets. 2009;13:1411–27.

    Article  CAS  PubMed  Google Scholar 

  23. Amann T, Maegdefrau U, Hartmann A, Agaimy A, Marienhagen J, Weiss TS, et al. GLUT1 expression is increased in hepatocellular carcinoma and promotes tumorigenesis. Am J Pathol. 2009;174:1544–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chen JQ, Russo J. Dysregulation of glucose transport, glycolysis TCA cycle and glutaminolysis by oncogenes and tumor suppressors in cancer cells. Biochim Biophys Acta. 1826;2012:370–84.

    Google Scholar 

  25. Lyshchik A, Higashi T, Hara T, Nakamoto Y, Fujimoto K, Doi R, et al. Expression of glucose transporter-1, hexokinase-II, proliferating cell nuclear antigen and survival of patients with pancreatic cancer. Cancer Investig. 2007;25:154–62.

    Article  CAS  Google Scholar 

  26. Mamede M, Higashi T, Kitaichi M, Ishizu K, Ishimori T, Nakamoto Y, et al. [18F]FDG uptake and PCNA, Glut-1, and Hexokinase-II expressions in cancers and inflammatory lesions of the lung. Neoplasia. 2005;7:369–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tohma T, Okazumi S, Makino H, Cho A, Mochiduki R, Shuto K, et al. Relationship between glucose transporter, hexokinase and FDG-PET in esophageal cancer. Hepatogastroenterology. 2005;52:486–90.

    CAS  PubMed  Google Scholar 

  28. Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell. 2008;13:472–82.

    Article  CAS  PubMed  Google Scholar 

  29. Porporato PE, Dhup S, Dadhich RK, Copetti T, Sonveaux P. Anticancer targets in the glycolytic metabolism of tumors: a comprehensive review. Front Pharmacol. 2011;2:49.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Kato Y, Ozawa S, Tsukuda M, Kubota E, Miyazaki K, St-Pierre Y, et al. Acidic extracellular pH increases calcium influx-triggered phospholipase D activity along with acidic sphingomyelinase activation to induce matrix metalloproteinase-9 expression in mouse metastatic melanoma. FEBS J. 2007;274:3171–83.

    Article  CAS  PubMed  Google Scholar 

  31. Kindzelskii AL, Amhad I, Keller D, Zhou MJ, Haugland RP, Garni-Wagner BA, et al. Pericellular proteolysis by leukocytes and tumor cells on substrates: focal activation and the role of urokinase-type plasminogen activator. Histochem Cell Biol. 2004;121:299–310.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We are grateful to Xia Li and Wei Liu who provided technical support. This work was supported by the National Natural Science Foundation of China (grant no. 30872480)

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Kefeng Dou or Desheng Wang.

Ethics declarations

Conflicts of interest

None.

Additional information

Runze Shang, Jianlin Wang, Wei Sun and Bin Dai contributed equally to this work.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shang, R., Wang, J., Sun, W. et al. RRAD inhibits aerobic glycolysis, invasion, and migration and is associated with poor prognosis in hepatocellular carcinoma. Tumor Biol. 37, 5097–5105 (2016). https://doi.org/10.1007/s13277-015-4329-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13277-015-4329-7

Keywords

Navigation