Skip to main content
Erschienen in: Tumor Biology 4/2016

06.11.2015 | Original Article

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

verfasst von: Runze Shang, Jianlin Wang, Wei Sun, Bin Dai, Bai Ruan, Zhuochao Zhang, Xisheng Yang, Yuan Gao, Shibin Qu, Xing Lv, Kaishan Tao, Lin Wang, Kefeng Dou, Desheng Wang

Erschienen in: Tumor Biology | Ausgabe 4/2016

Einloggen, um Zugang zu erhalten

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.
Literatur
1.
Zurück zum Zitat 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. 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.
2.
Zurück zum Zitat Bosch FX, Ribes J, Díaz M, Cléries R. Primary liver cancer: worldwide incidence and trends. Gastroenterology. 2004;127:S5–S16.CrossRefPubMed Bosch FX, Ribes J, Díaz M, Cléries R. Primary liver cancer: worldwide incidence and trends. Gastroenterology. 2004;127:S5–S16.CrossRefPubMed
3.
4.
Zurück zum Zitat Lincet H, Icard P. How do glycolytic enzymes favour cancer cell proliferation by nonmetabolic functions? Oncogene. 2015;34:3751–9.CrossRefPubMed Lincet H, Icard P. How do glycolytic enzymes favour cancer cell proliferation by nonmetabolic functions? Oncogene. 2015;34:3751–9.CrossRefPubMed
5.
Zurück zum Zitat 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.PubMedPubMedCentral 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.PubMedPubMedCentral
6.
Zurück zum Zitat 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.CrossRef 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.CrossRef
7.
Zurück zum Zitat Reynet C, Kahn CR. Rad: a member of the Ras family overexpressed in muscle of type II diabetic humans. Science. 1993;262:1441–4.CrossRefPubMed Reynet C, Kahn CR. Rad: a member of the Ras family overexpressed in muscle of type II diabetic humans. Science. 1993;262:1441–4.CrossRefPubMed
8.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
10.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
11.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
12.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
13.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
14.
Zurück zum Zitat 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.PubMedPubMedCentral 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.PubMedPubMedCentral
15.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
16.
Zurück zum Zitat 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.CrossRef 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.CrossRef
17.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
18.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
19.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Szablewski L. Expression of glucose transporters in cancers. Biochim Biophys Acta. 1835;2013:164–9. Szablewski L. Expression of glucose transporters in cancers. Biochim Biophys Acta. 1835;2013:164–9.
22.
Zurück zum Zitat Amann T, Hellerbrand C. GLUT1 as a therapeutic target in hepatocellular carcinoma. Expert Opin Ther Targets. 2009;13:1411–27.CrossRefPubMed Amann T, Hellerbrand C. GLUT1 as a therapeutic target in hepatocellular carcinoma. Expert Opin Ther Targets. 2009;13:1411–27.CrossRefPubMed
23.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat 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. 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.
25.
Zurück zum Zitat 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.CrossRef 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.CrossRef
26.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat 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.PubMed 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.PubMed
28.
Zurück zum Zitat Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell. 2008;13:472–82.CrossRefPubMed Kroemer G, Pouyssegur J. Tumor cell metabolism: cancer’s Achilles’ heel. Cancer Cell. 2008;13:472–82.CrossRefPubMed
29.
Zurück zum Zitat 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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
31.
Zurück zum Zitat 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.CrossRefPubMed 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.CrossRefPubMed
Metadaten
Titel
RRAD inhibits aerobic glycolysis, invasion, and migration and is associated with poor prognosis in hepatocellular carcinoma
verfasst von
Runze Shang
Jianlin Wang
Wei Sun
Bin Dai
Bai Ruan
Zhuochao Zhang
Xisheng Yang
Yuan Gao
Shibin Qu
Xing Lv
Kaishan Tao
Lin Wang
Kefeng Dou
Desheng Wang
Publikationsdatum
06.11.2015
Verlag
Springer Netherlands
Erschienen in
Tumor Biology / Ausgabe 4/2016
Print ISSN: 1010-4283
Elektronische ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-015-4329-7

Weitere Artikel der Ausgabe 4/2016

Tumor Biology 4/2016 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.