The Effect of Histone Hyperacetylation on Viability of Basal-Like Breast Cancer Cells MDA-MB-231

Authors

Department of Biochemistry, Zanjan University of Medical Sciences, Zanjan, Iran

Abstract

Abstract Background: The Basal-Like breast cancer, is always known for lack of expression of estrogen receptor (ER), progesterone receptor (PR) and as well, absence of epidermal growth factor receptor 2 (HER2) gene amplification. Improper expression pattern of ER, PR, and Her2, makes Basal-Like breast tumors resistant to the current hormonal and anti HER2 treatments. In recent decades, several studies have been conducted to investigate the regulatory role of chemical modifications of core histones in gene expression. Their results have shown that histone acetylation is involved in regulation of cell survival. Acetylation of core histones is regulated by the epigenetic-modifying enzymes named Histone Deacetylases (HDACs). As a new approach to control the viability of breast tumor cells resistant to the hormonal and anti-HER2 treatments, we have targeted the HDACs. Using Trichostatin A (TSA) as a known HDACs inhibitor, we have tried to hyperacetylate the core histones of MDA-MB-231 cells as an in vitro model of Basal-Like breast tumors. Then we have investigated the effect of histone hyperacetylation on viability of MDA-MB-231 cells. Methods: MDA-MB-231 cells were cultured in RPMI 1640 medium containing 10% fetal bovine serum (FBS) and were incubated at 37°C, in a humidified incubator with 5% CO2 atmosphere. Then cells were treated with different concentrations of TSA including: 50, 100, 200, 400, 800 and 1000 nM or control (1% DMSO). After 24 and 48 hours, viability of cells was evaluated by MTT assay. Results: After 24 and 48h exposure to different concentrations of TSA, MDA-MB-231 cells showed a maximum tolerable dose. At higher concentrations, TSA decreased the percentage of cell viability through a time-dose dependent manner. IC50 value for 48h treatment was 600 nM. Conclusions: Our results indicate that HDACs inhibition and subsequently hyperacetylation of histones, leads to cytotoxic effects on breast tumor cells resistant to the current treatments. Following this pilot research we are trying to suggest molecular mechanisms underlying the anti-proliferative effects triggered by HDACs inhibition.

Keywords


  1. Nelson HD, Zakher B, Cantor A, Fu R, Griffin J, O’Meara ES, et al. Risk factors for breast cancer for women aged 40 to 49 years: a systematic review and meta-analysis. Ann Intern Med. 2012;156(9):635–48. doi: 10.7326/0003-4819-156-9-201205010-00006. [PubMed: 22547473].
  2. Brenton JD, Carey LA, Ahmed AA, Caldas C. Molecular classification and molecular forecasting of breast cancer: ready for clinical application?. J Clin Oncol. 2005;23(29):7350–60. doi: 10.1200/JCO.2005.03.3845. [PubMed: 16145060].
  3. Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, et al. Immunohistochemical and clinical characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer Res. 2004;10(16):5367–74. doi: 10.1158/1078-0432.CCR-04-0220. [PubMed: 15328174].
  4. Livasy CA, Karaca G, Nanda R, Tretiakova MS, Olopade OI, Moore DT, et al. Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol. 2006;19(2):264–71. doi: 10.1038/modpathol.3800528. [PubMed: 16341146].
  5. Subik K, Lee JF, Baxter L, Strzepek T, Costello D, Crowley P, et al. The Expression Patterns of ER, PR, HER2, CK5/6, EGFR, Ki-67 and AR by Immunohistochemical Analysis in Breast Cancer Cell Lines. Breast Cancer (Auckl). 2010;4:35–41. [PubMed: 20697531].
  6. Onitilo AA, Engel JM, Greenlee RT, Mukesh BN. Breast cancer subtypes based on ER/PR and Her2 expression: comparison of clinicopathologic features and survival. Clin Med Res. 2009;7(1-2):4–13. doi: 10.3121/cmr.2009.825. [PubMed: 19574486].
  7. Early Breast Cancer Trialists’ Collaborative G. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Lancet. 2005;365(9472):1687–717. doi: 10.1016/S0140-6736(05)66544-0. [PubMed: 15894097].
  8. Hudis CA. Trastuzumab–mechanism of action and use in clinical practice. N Engl J Med. 2007;357(1):39–51. doi: 10.1056/NEJMra043186. [PubMed: 17611206].
  9. Rakha EA, Reis-Filho JS, Ellis IO. Basal-like breast cancer: a critical review. J Clin Oncol. 2008;26(15):2568–81. doi: 10.1200/JCO.2007.13.1748. [PubMed: 18487574].
  10. Haberland M, Montgomery RL, Olson EN. The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet. 2009;10(1):32–42. doi: 10.1038/nrg2485. [PubMed: 19065135].
  11. Sarkar S, Longacre M, Tatur N, Heerboth S, Lapinska K. Histone deacetylases (HDACs): Function, mechanism, & inhibition. Encyclopedia Analytical Chem. 2014.
  12. Zentner GE, Henikoff S. Regulation of nucleosome dynamics by histone modifications. Nat Struct Mol Biol. 2013;20(3):259–66. doi: 10.1038/nsmb.2470. [PubMed: 23463310].
  13. Ahmad K, Henikoff S. Epigenetic consequences of nucleosome dynamics. Cell. 2002;111(3):281–4. [PubMed: 12419239].
  14. Itazaki H, Nagashima K, Sugita K, Yoshida H, Kawamura Y, Yasuda Y, et al. Isolation and structural elucidation of new cyclotetrapeptides, trapoxins A and B, having detransformation activities as antitumor agents. J Antibiot (Tokyo). 1990;43(12):1524–32. [PubMed: 2276972].
  15. Hoshikawa Y, Kijima M, Yoshida M, Beppu T. Expression of differentiation-related markers in teratocarcinoma cells via histone hyperacetylation by trichostatin A. Agricultural Biological Chem. 1991;55(6):1491–5.
  16. Hoshikawa Y, Kwon HJ, Yoshida M, Horinouchi S, Beppu T. Trichostatin A induces morphological changes and gelsolin expression by inhibiting histone deacetylase in human carcinoma cell lines. Exp Cell Res. 1994;214(1):189–97. doi: 10.1006/excr.1994.1248. [PubMed: 8082721].
  17. Sugita K, Koizumi K, Yoshida H. Morphological reversion of sistransformed NIH3T3 cells by trichostatin A. Cancer Res. 1992;52(1):168– 72. [PubMed: 1727377].
  18. Yoshida M, Hoshikawa Y, Koseki K, Mori K, Beppu T. Structural specificity for biological activity of trichostatin A, a specific inhibitor of mammalian cell cycle with potent differentiation-inducing activity in Friend leukemia cells. J Antibiot (Tokyo). 1990;43(9):1101–6. [PubMed: 2211374].
  19. Yoshida M, Nomura S, Beppu T. Effects of trichostatins on differentiation of murine erythroleukemia cells. Cancer Res. 1987;47(14):3688–91. [PubMed: 2439196].
  20. Yoshida M, Beppu T. Reversible arrest of proliferation of rat 3Y1 fibroblasts in both the G1 and G2 phases by trichostatin A. Experimental Cell Res. 1988;177(1):122–31. doi: 10.1016/0014-4827(88)90030-4.
  21. Medina V, Edmonds B, Young GP, James R, Appleton S, Zalewski PD. Induction of caspase-3 protease activity and apoptosis by butyrate and trichostatin A (inhibitors of histone deacetylase): dependence on protein synthesis and synergy with a mitochondrial/cytochrome c-dependent pathway. Cancer Res. 1997;57(17):3697–707. [PubMed: 9288776].
  22. Glozak MA, Seto E. Histone deacetylases and cancer. Oncogene. 2007;26(37):5420–32. doi: 10.1038/sj.onc.1210610. [PubMed: 17694083].
  23. Eot-Houllier G, Fulcrand G, Magnaghi-Jaulin L, Jaulin C. Histone deacetylase inhibitors and genomic instability. Cancer Lett. 2009;274(2):169–76. doi: 10.1016/j.canlet.2008.06.005. [PubMed: 18635312]. 4 Razavi Int J Med. 2017; 5(2):e55455. Rahimian A and Mellati A
  24. Vanhaecke T, Papeleu P, Elaut G, Rogiers V. Trichostatin A-like hydroxamate histone deacetylase inhibitors as therapeutic agents: toxicological point of view. Curr Med Chem. 2004;11(12):1629–43. [PubMed: 15180568].
  25. Chavez KJ, Garimella SV, Lipkowitz S. Triple negative breast cancer cell lines: one tool in the search for better treatment of triple negative breast cancer. Breast Dis. 2010;32(1-2):35–48. doi: 10.3233/BD-2010-0307. [PubMed: 21778573].
  26. Rasheed WK, Johnstone RW, Prince HM. Histone deacetylase inhibitors in cancer therapy. Expert Opin Investig Drugs. 2007;16(5):659– 78. doi: 10.1517/13543784.16.5.659. [PubMed: 17461739].
  27. Marchion D, Munster P. Development of histone deacetylase inhibitors for cancer treatment. Expert Rev Anticancer Ther. 2007;7(4):583–98. doi: 10.1586/14737140.7.4.583. [PubMed: 17428177].
  28. Sotiriou C, Neo SY, McShane LM, Korn EL, Long PM, Jazaeri A, et al. Breast cancer classification and prognosis based on gene expression profiles from a population-based study. Proc Natl Acad Sci U S A. 2003;100(18):10393–8. doi: 10.1073/pnas.1732912100. [PubMed: 12917485].
  29. Carey LA, Perou CM, Livasy CA, Dressler LG, Cowan D, Conway K, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study.JAMA. 2006;295(21):2492–502. doi: 10.1001/jama.295.21.2492. [PubMed: 16757721].
  30. Foulkes WD, Brunet JS, Stefansson IM, Straume O, Chappuis PO, Begin LR, et al. The prognostic implication of the basal-like (cyclin E high/p27 low/p53+/glomeruloid-microvascular-proliferation+) phenotype of BRCA1-related breast cancer. Cancer Res. 2004;64(3):830–5. [PubMed: 14871808].
  31. Banerjee S, Reis-Filho JS, Ashley S, Steele D, Ashworth A, Lakhani SR, et al. Basal-like breast carcinomas: clinical outcome and response to chemotherapy. J Clin Pathol. 2006;59(7):729–35. doi: 10.1136/jcp.2005.033043. [PubMed: 16556664].
  32. Rakha EA, El-Rehim DA, Paish C, Green AR, Lee AH, Robertson JF, et al. Basal phenotype identifies a poor prognostic subgroup of breast cancer of clinical importance. Eur J Cancer. 2006;42(18):3149–56. doi: 10.1016/j.ejca.2006.08.015. [PubMed: 17055256].
  33. Fulford LG, Reis-Filho JS, Ryder K, Jones C, Gillett CE, Hanby A, et al. Basal-like grade III invasive ductal carcinoma of the breast: patterns of metastasis and long-term survival. Breast Cancer Res. 2007;9(1):R4. doi: 10.1186/bcr1636. [PubMed: 17217540].