DOI QR코드

DOI QR Code

Alteration of Runt-related Transcription Factor 3 Gene Expression and Biologic Behavior of Esophageal Carcinoma TE-1 Cells after 5-Azacytidine Intervention

  • Wang, Shuai (Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University) ;
  • Liu, Hong (Department of Otorhinolaryngology, Provincial Hospital Affiliated to Shandong University) ;
  • Akhtar, Javed (Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University) ;
  • Chen, Hua-Xia (Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University) ;
  • Wang, Zhou (Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University)
  • Published : 2013.09.30

Abstract

5-Azacytidine (5-azaC) was originally identified as an anticancer drug (NSC102876) which can cause hypomethylation of tumor suppressor genes. To assess its effects on runt-related transcription factor 3 (RUNX3), expression levels and the promoter methylation status of the RUNX3 gene were assessed. We also investigated alteration of biologic behavior of esophageal carcinoma TE-1 cells. MTT assays showed 5-azaC inhibited the proliferation of TE-1 cells in a time and dose-dependent way. Although other genes could be demethylated after 5-azaC intervention, we focused on RUNX3 gene in this study. The expression level of RUNX3 mRNA increased significantly in TE-1 cells after treatment with 5-azaC at hypotoxic levels. RT-PCR showed 5-azaC at $50{\mu}M$ had the highest RUNX3-induction activity. Methylation-specific PCR indicated that 5-azaC induced RUNX3 expression through demethylation. Migration and invasion of TE-1 cells were inhibited by 5-azaC, along with growth of Eca109 xenografts in nude mice. In conclusion, we demonstrate that the RUNX3 gene can be reactivated by the demethylation reagent 5-azaC, which inhibits the proliferation, migration and invasion of esophageal carcinoma TE-1 cells.

Keywords

References

  1. Acun T, Oztas E, Yagci T, Yakicier MC (2011). SIP1 is downregulated in hepatocellular carcinoma by promoter hypermethylation. BMC Cancer, 11, 223-43. https://doi.org/10.1186/1471-2407-11-223
  2. Aimiuwu J, Wang H, Chen P, et al (2012). RNA-dependent inhibition of ribonucleotide reductase is a major pathway for 5-azacytidine activity in acute myeloid leukemia. Blood, 119, 5229-38. https://doi.org/10.1182/blood-2011-11-382226
  3. Alvarez-Diaz S, Larriba MJ, Lopez-Otin C, Munoz A (2010). Vitamin D: Proteases, protease inhibitors and cancer. Cell Cycle, 9, 32-7. https://doi.org/10.4161/cc.9.1.10266
  4. Bae SC, Choi JK (2004). Tumor suppressor activity of RUNX3. Oncogene, 23, 4336-40. https://doi.org/10.1038/sj.onc.1207286
  5. Bangsow C, Rubins N, Glusman G, et al (2001). The RUNX3 gene-sequence, structure and regulated expression. Gene, 279, 221-32. https://doi.org/10.1016/S0378-1119(01)00760-0
  6. Beck S, Rakyan VK (2008). The methylome: approaches for global DNA methylation profiling. Trends Genet, 24, 231-7. https://doi.org/10.1016/j.tig.2008.01.006
  7. Dachrut S, Banthaisong S, Sripa M, et al (2009). DNA copynumber loss on 1p36.1 harboring RUNX3 with promoter hypermethylation and associated loss of RUNX3 expression in liver fluke-associated intrahepatic cholangiocarcinoma. Asian Pac J Cancer Prev, 10, 575-82.
  8. Deng T, Zhang Y (2009). Possible involvement of activation of P53/P21 and demethylation of RUNX 3 in the cytotoxicity against Lovo cells induced by 5-Aza-2'-deoxycytidine. Life Sci, 84, 311-20. https://doi.org/10.1016/j.lfs.2008.12.015
  9. Fukasawa M, Kimura M, Morita S, et al (2006). Microarray analysis of promoter methylation in lung cancers. J Hum Genet, 51, 368-74. https://doi.org/10.1007/s10038-005-0355-4
  10. Gronbaek K, Hother C, Jones PA (2007). Epigenetic changes in cancer. APMIS, 115, 1039-59. https://doi.org/10.1111/j.1600-0463.2007.apm_636.xml.x
  11. Ito K, Liu Q, Salto-Tellez M, et al (2005). RUNX3, a novel tumor suppressor, is frequently inactivated in gastric cancer by protein mislocalization. Cancer Res, 65, 7743-50. https://doi.org/10.1158/0008-5472.CAN-05-0743
  12. Kamangar F, Dores GM, Anderson WF (2006). Patterns of cancer incidence, mortality, and prevalence across five continents: defining priorities to reduce cancer disparities in different geographic regions of the world. J Clin Oncol, 24, 2137-50. https://doi.org/10.1200/JCO.2005.05.2308
  13. Kiziltepe T, Hideshima T, Catley L, et al (2007). 5-Azacytidine, a DNA methyltransferase inhibitor, induces ATR-mediated DNA double-strand break responses, apoptosis, and synergistic cytotoxicity with doxorubicin and bortezomib against multiple myeloma cells. Mol Cancer Ther, 6, 1718-22. https://doi.org/10.1158/1535-7163.MCT-07-0010
  14. Kong KL, Kwong DL, Fu L,et al (2010). Characterization of a candidate tumor suppressor gene uroplakin 1A in esophageal squamous cell carcinoma. Cancer Res, 70, 8832-41. https://doi.org/10.1158/0008-5472.CAN-10-0779
  15. Ku JL, Kang SB, Shin YK, et al (2004). Promoter hypermethylation downregulates RUNX3 gene expression in colorectal cancer cell lines. Oncogene, 23, 6736-42. https://doi.org/10.1038/sj.onc.1207731
  16. Li M, Tan SY, Zhang J, You HX (2013). Effects of paeonol on intracellular calcium concentration and expression of RUNX3 in LoVo human colon cancer cells. Mol Med Rep, 7, 1425-30. https://doi.org/10.3892/mmr.2013.1372
  17. Li QL, Ito K, Sakakura C, et al (2002). Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell, 109,113-24. https://doi.org/10.1016/S0092-8674(02)00690-6
  18. Long C, Yin B, Lu Q, et al (2007). Promoter hypermethylation of the RUNX3 gene in esophageal squamous cell carcinoma. Cancer Invest, 25, 685-90. https://doi.org/10.1080/07357900701561131
  19. Park SY, Kim BH, Kim JH, et al (2007). Methylation profiles of CpG island loci in major types of human cancers . J Korean Med Sci, 22, 311-7. https://doi.org/10.3346/jkms.2007.22.2.311
  20. Park WS, Cho YG, Kim CJ, et al (2005). Hypermethylation of the RUNX3 gene in hepatocellular carcinoma. Exp Mol Med, 37,276-81. https://doi.org/10.1038/emm.2005.37
  21. Perera Y, Farina HG, Hernández I, et al (2008). Systemic administration of a peptide that impairs the protein kinase (CK2) phosphorylation reduces solid tumor growth in mice. Int J Cancer, 122, 57-62. https://doi.org/10.1002/ijc.23013
  22. Rasool S, Khan T, Qazi F, Ganai BA (2013). ECRG1 and its relationship with esophageal cancer: a brief review. Onkologie, 36, 213-6. https://doi.org/10.1159/000349960
  23. Shibata-Kobayashi S, Yamashita H, Okuma K, et al (2013). Correlation among 16 biological factors [p53, p21 (waf1), MIB-1 (Ki-67), p16 (INK4A), cyclin D1, E-cadherin, Bcl-2, TNF-$\alpha$, NF-${\kappa}B$, TGF-$\beta$, MMP-7, COX-2, EGFR, HER2/ neu, ER, and HIF-$1\alpha$] and clinical outcomes following curative chemoradiation therapy in 10 patients with esophageal squamous cell carcinoma. Oncol Lett, 5, 903-10. https://doi.org/10.3892/ol.2013.1130
  24. Shi GZ, Yuan Y, Jiang GJ, et al (2012). PRAF3 induces apoptosis and inhibits migration and invasion in human esophageal squamous cell carcinoma. BMC Cancer, 12, 97. https://doi.org/10.1186/1471-2407-12-97
  25. Smith E, De Young NJ, Pavey SJ, et al (2008). Similarity of aberrant DNA methylation in Barrett's esophagus and esophageal adenocarcinoma. Mol Cancer, 7, 75. https://doi.org/10.1186/1476-4598-7-75
  26. Subramaniam MM, Chan JY, Soong R, et al (2009). RUNX3 inactivation by frequent promoter hypermethylation and protein mislocalization constitute an early event in breast cancer progression. Breast Cancer Res Treat, 113, 113-21. https://doi.org/10.1007/s10549-008-9917-4
  27. Sugiura H, Ishiguro H, Kuwabara Y, et al (2008). Decreased expression of RUNX3 is correlated with tumor progression and poor prognosis in patients with esophageal squamous cell carcinoma. Oncol Rep, 19, 713-9.
  28. Tang GH, Sun SW, He XS (2012). Correlation of CpG methylation status of Runx3 with pathogenesis of gastric carcinoma. Zhonghua Bing Li Xue Za Zhi, 41, 314-9.
  29. Wang M, Chen S, Wang S, et al (2012). Effects of phytochemicals sulforaphane on uridine diphosphate-glucuronosyltransferase expression as well as cell-cycle arrest and apoptosis in human colon cancer Caco-2 cells. Chin J Physiol, 55, 134-44.
  30. Zhang YW, Eom SY, Yim DH, et al (2013). Evaluation of the relationship between dietary factors, CagA-positive Helicobacter pylori infection, and RUNX3 promoter hypermethylation in gastric cancer tissue. World J Gastroenterol, 19, 1778-87. https://doi.org/10.3748/wjg.v19.i11.1778
  31. Zhao C, Fernandes MJ, Prestwich GD, et al (2008). Regulation of lysophosphatidic acid receptor expression and function in human synoviocytes: implications for rheumatoid arthritis? Mol Pharmacol, 73, 587-600.

Cited by

  1. Overexpression of RUNX3 Inhibits Malignant Behaviour of Eca109 Cells in Vitro and Vivo vol.15, pp.4, 2014, https://doi.org/10.7314/APJCP.2014.15.4.1531