DOI QR코드

DOI QR Code

HDAC6 siRNA Inhibits Proliferation and Induces Apoptosis of HeLa Cells and its Related Molecular Mechanism

  • Qin, Hai-Xia (Department of Obstetrics and Gynecology, the First Affiliated Hospital of Zhengzhou University) ;
  • Cui, Hong-Kai (Department of Interventional Radiology, The First Affiliated Hospital of Xinxiang Medical University) ;
  • Pan, Ying (Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Xinxiang Medical University) ;
  • Yang, Jun (Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xinxiang Medical University) ;
  • Ren, Yan-Fang (Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xinxiang Medical University) ;
  • Hua, Cai-Hong (Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xinxiang Medical University) ;
  • Hua, Fang-Fang (Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xinxiang Medical University) ;
  • Qiao, Yu-Huan (Department of Obstetrics and Gynecology, the First Affiliated Hospital of Zhengzhou University)
  • Published : 2012.07.31

Abstract

Objective: To investigate the effects of histone deacetylase 6 (HDAC6) siRNA on cell proliferation and cell apoptosis of the HeLa cervical carcinoma cell line and the molecular mechanisms involved. Methods: Division was into three groups: A, the untreated group; B, the control siRNA group; and C, the HDAC6 siRNA group. Lipofectamine 2000 was used for siRNA transfection, and Western blot analysis was used to determine the protein levels. Cell proliferation and apoptosis were characterized using a CCK-8 assay and flow cytometry, respectively. Results: HDAC6 protein expression in the HDAC6 siRNA-transfection group was significantly lower (P < 0.05) than in the untreated and control siRNA groups. The CCK-8 kit results demonstrated that the proliferation of HeLa cells was clearly inhibited in the HDAC6 siRNA transfection group (P < 0.05). In addition, flow cytometry revealed that the early apoptotic rate ($26.0%{\pm}0.87%$) was significantly elevated (P < 0.05) as compared with the untreated group ($10.6%{\pm}1.19%$) and control siRNA group ($8.61%{\pm}0.98%$). Furthermore, Western blot analysis indicated that bcl-2 protein expression in the HDAC6 siRNA-transfection group was down-regulated, whereas the expression of p21 and bax was up-regulated. Conclusion: HDAC6 plays an essential role in the occurrence and development of cervical carcinoma, and the down-regulation of HDAC6 expression may be useful molecular therapeutic method.

Keywords

References

  1. Bazzaro M, Lin Z, Santillan A, et al (2008). Ubiquitin proteasome system stress underlies synergistic killing of ovarian cancer cells by bortezomib and a novel HDAC6 inhibitor. Clin Cancer Res, 14, 7340-7. https://doi.org/10.1158/1078-0432.CCR-08-0642
  2. Bertos NR, Gilquin B, Chan GK, et al (2004). Role of the tetradecapeptide repeat domain of human histone deacetylase 6 in cytoplasmic retention. J Biol Chem, 279, 48246-54. https://doi.org/10.1074/jbc.M408583200
  3. Kawiak J, Hoser G, Skorski T (1998). Apoptosis and some of its medical implications. Folia Histochem Cytobiol, 36, 99-110.
  4. Knudson AG (2001). Two genetic hits (more or less) to cancer. Nat Rev Cancer, 1, 157-62. https://doi.org/10.1038/35101031
  5. Kong X, Lin Z, Liang D, et al (2006). Histone deacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1alpha. Mol Cell Biol, 26, 2019-28. https://doi.org/10.1128/MCB.26.6.2019-2028.2006
  6. Kouzarides T (1999). Histone acetylases and deacetylases in cell proliferation. Curr Opin Genet Dev, 9, 40-8. https://doi.org/10.1016/S0959-437X(99)80006-9
  7. Lehrmann H, Pritchard LL, Harel-Bellan A (2002). Histone acetyltransferases and deacetylases in the control of cell proliferation and differentiation. Adv Cancer Res, 86, 41-65. https://doi.org/10.1016/S0065-230X(02)86002-X
  8. Liu Y, Martindale JL, Gorospe M, Holbrook NJ (1996). Regulation of p21WAF1/CIP1 expression through mitogen-activated protein kinase signaling pathway. Cancer Res, 56, 31-5.
  9. Mehnert JM, Kelly WK (2007). Histone deacetylase inhibitors: biology and mechanism of action. Cancer J, 13, 23-9. https://doi.org/10.1097/PPO.0b013e31803c72ba
  10. Nakatani F, Tanaka K, Sakimura R, et al (2003). Identification of p21WAF1/CIP1 as a direct target of EWS-Fli1 oncogenic fusion protein. J Biol Chem, 278, 15105-15. https://doi.org/10.1074/jbc.M211470200
  11. Park SW, Kim HS, Yoo NJ, Lee SH (2011). Mutational analysis of mononucleotide repeats in HDAC4, 5, 6, 7, 9 and 11 genes in gastric and colorectal carcinomas with microsatellite instability. Acta Oncol, 50, 317-8. https://doi.org/10.3109/0284186X.2010.504229
  12. Parkin DM, Pisani P, Ferlay J (1999). Global cancer statistics. CA Cancer J Clin, 49, 33-64. https://doi.org/10.3322/canjclin.49.1.33
  13. Pecorelli S, Pasinetti B, Angioli R, Favalli G, Odicino F (2005). Systemic therapy for gynecological neoplasms: ovary, cervix, and endometrium. Cancer Chemother Biol Response Modif, 22, 515-44. https://doi.org/10.1016/S0921-4410(04)22023-3
  14. Richon VM, Garcia-Vargas J, Hardwick JS (2009). Development of vorinostat: current applications and future perspectives for cancer therapy. Cancer Lett, 280, 201-10. https://doi.org/10.1016/j.canlet.2009.01.002
  15. Ropero S, Esteller M (2007). The role of histone deacetylases (HDACs) in human cancer. Mol Oncol, 1, 19-25. https://doi.org/10.1016/j.molonc.2007.01.001
  16. Sadeghi M, Süsal C, Daniel V, et al (2007). Short communication: decreasing soluble CD30 and increasing IFN-gamma plasma levels are indicators of effective highly active antiretroviral therapy. AIDS Res Hum Retroviruses, 23, 886-90. https://doi.org/10.1089/aid.2006.0228
  17. Sakuma T, Uzawa K, Onda T, et al (2006). Aberrant expression of histone deacetylase 6 in oral squamous cell carcinoma. Int J Oncol, 29, 117-24.
  18. Subramanian C, Jarzembowski JA, Opipari AW Jr, Castle VP, Kwok RP (2011). HDAC6 deacetylates Ku70 and regulates Ku70-Bax binding in neuroblastoma. Neoplasia, 13, 726-34.
  19. Verdel A, Curtet S, Brocard MP, et al (2000). Active maintenance of mHDA2/mHDAC6 histone-deacetylase in the cytoplasm. Curr Biol, 10, 747-9. https://doi.org/10.1016/S0960-9822(00)00542-X
  20. Waga S, Hannon GJ, Beach D, Stillman B (1994). The p21 inhibitor of cyclin-dependent kinases controls DNA replication by interaction with PCNA. Nature, 369, 574-8. https://doi.org/10.1038/369574a0
  21. Xu X, Xie C, Edwards H, et al (2011). Inhibition of histone deacetylases 1 and 6 enhances cytarabine-induced apoptosis in pediatric acute myeloid leukemia cells. PLoS One, 6, e17138. https://doi.org/10.1371/journal.pone.0017138
  22. Zhang H, Hannon GJ, Beach D (1994). p21-containing cyclin kinases exist in both active and inactive states. Genes Dev, 8, 1750-8. https://doi.org/10.1101/gad.8.15.1750
  23. Zhang W, Kone BC (2002). NF-kappaB inhibits transcription of the H(+)-K(+)-ATPase alpha(2)-subunit gene: role of histone deacetylases. Am J Physiol Renal Physiol, 283, F904-11.
  24. Zhang Y, Gilquin B, Khochbin S, Matthias P (2006). Two catalytic domains are required for protein deacetylation. J Biol Chem, 281, 2401-4.
  25. Zhang Z, Yamashita H, Toyama T, et al (2004). HDAC6 expression is correlated with better survival in breast cancer. Clin Cancer Res, 10, 6962-8. https://doi.org/10.1158/1078-0432.CCR-04-0455

Cited by

  1. Limited efficacy of specific HDAC6 inhibition in urothelial cancer cells vol.15, pp.6, 2014, https://doi.org/10.4161/cbt.28469
  2. TRPM2 Mediates Histone Deacetylase Inhibition-Induced Apoptosis in Bladder Cancer Cells vol.30, pp.2, 2015, https://doi.org/10.1089/cbr.2014.1697
  3. Both HDAC5 and HDAC6 are required for the proliferation and metastasis of melanoma cells vol.14, pp.1, 2016, https://doi.org/10.1186/s12967-015-0753-0