Comparison of the effects of olsalazine and decitabine on the expression of CDH1 and uPA genes and cytotoxicity in MDA-MB-231 breast cancer cells

Misagh Mohammadi Asl , Javad Mohammadi Asl, Mojgan Naghitorabi

Abstract


Background and purpose: Since DNA methyltransferase enzymes play a key role in DNA methylation, they can be used as a target to alter epigenetic changes and treat cancer. Recent studies have shown that olsalazine, through its potent inhibitory effect on the DNA methyltransferase enzyme, can be a good option. The aim of this study was to investigate the effects of olsalazine on cell viability and expression of CDH1 and uPA genes in MDA-MB-231 cells compared with decitabine.

Experimental approach: The cytotoxicity of the drugs was determined using a standard MTT assay. MDA-MB-231 cells were treated with olsalazine and decitabine with concentrations less than IC50 to evaluate the effect of drugs on the expression of genes. RNA was extracted from the cells after 24 and 48 h and CDH1and uPA gene expression were evaluated by quantitative real-time polymerase chain reaction method.

Findings/Results: The cytotoxicity of the two drugs was comparable. The IC50 values at 24 h were 4000 and 4500 μM for olsalazine and decitabine, respectively. The IC50 values of both drugs were about 300 μM at 48 h. Statistical analyzes showed a significant increase in CDH1 expression after 24-48 h treatment with olsalazine, and 48 h treatment with decitabine, without any significant increase in uPA expression.

Conclusion and implications: Our results showed that olsalazine has cellular toxicity comparable to decitabine in MDA-MB-231 cells. Also compared to decitabine, olsalazine causes a greater increase in expression of CDH1 without any significant increase in uPA expression. Therefore, it appears to be a good candidate for cancer treatment.

 

 


Keywords


Cancer; CDH1; Decitabine; Epigenetic; Olsalazine; uPA.

Full Text:

PDF

References


Robertson KD. DNA methylation, methyltransferases, and cancer. Oncogene. 2001;20(24):3139-3155.

DOI: 10.1038/sj.onc.1204341.

Peedicayil J. Epigenetic therapy-a new development in pharmacology. Indian J Med Res. 2006;123(1):17-24.

Veeck J, Esteller M. Breast cancer epigenetics: from DNA methylation to microRNAs. J Mammary Gland Biol Neoplasia. 2010;15(1):5-17.

DOI: 10.1007/s10911-010-9165-1.

Zhu WG, Otterson GA. The interaction of histone deacetylase inhibitors and DNA methyltransferase inhibitors in the treatment of human cancer cells. Curr Med Chem Anticancer Agents. 2003;3(3):187-199.

DOI: 10.2174/1568011033482440.

Caldeira JR, Prando EC, Quevedo FC, Neto FA, Rainho CA, Rogatto SR. CDH1 promoter hypermethylation and E-cadherin protein expression in infiltrating breast cancer. BMC Cancer. 2006;6:48-56.

DOI: 10.1186/1471-2407-6-48.

Lombaerts M, Wezel TV, Philippo K, Dierssen JWF, Zimmerman RME, Oosting J, et al. E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines. Br J Cancer. 2006;94(5):661-671.

Naghitorabi M, Mohammadi Asl J, Mir Mohammad Sadeghi H, Rabbani M, Jafarian-Dehkordi A, Haghjooye Javanmard S. Quantitative evaluation of DNMT3B promoter methylation in breast cancer patients using differential high resolution melting analysis. Res Pharm Sci. 2013;8(3):167-175.

Dzobo K. Epigenomics-guided drug development: recent advances in solving the cancer treatment "jigsaw puzzle". OMICS. 2019;23(2):70-85.

DOI: 10.1089/omi.2018.0206.

Chik F, Szyf M. Effects of specific DNMT gene depletion on cancer cell transformation and breast cancer cell invasion; toward selective DNMT inhibitors. Carcinogenesis. 2011;32(2):224-232.

DOI: 10.1093/carcin/bgq221.

Ma Z, Webb DJ, Jo M, Gonias SL. Endogenously produced urokinase-type plasminogen activator is a major determinant of the basal level of activated ERK/MAP kinase and prevents apoptosis in MDA-MB-231 breast cancer cells. J Cell Sci. 2001;114(Pt 18):3387-3396.

Pakneshan P, Xing RH, Rabbani SA. Methylation status of uPA promoter as a molecular mechanism regulating prostate cancer invasion and growth in vitro and in vivo. FASEB J. 2003;17(9):1081-1088.

DOI: 10.1096/fj.02-0973com.

Pakneshan P, Têtu B, Rabbani SA. Demethylation of urokinase promoter as a prognostic marker in patients with breast carcinoma. Clin Cancer Res. 2004;10(9):3035-3041.

DOI: 10.1158/1078-0432.ccr-03-0545.

Medina-Franco JL, Mendez-Lucio O, Yoo J. Rationalization of activity cliffs of a sulfonamide inhibitor of DNA methyltransferases with induced-fit docking. Int J Mol Sci. 2014;15(2):3253-3261.

DOI: 10.3390/ijms15023253.

Mendez-Lucio O, Tran J, Medina-Franco JL, Meurice N, Muller M. Toward drug repurposing in epigenetics: olsalazine as a hypomethylating compound active in a cellular context. ChemMedChem. 2014;9(3):560-565.

DOI: 10.1002/cmdc.201300555.

Selby WS, Barr GD, Ireland A, Mason CH, Jewell DP. Olsalazine in active ulcerative colitis. Br Med J (Clin Res Ed). 1985;291(6506):1373-1375.

DOI: 10.1136/bmj.291.6506.1373.

van Staa TP, Card T, Logan RF, Leufkens HGM. 5-Aminosalicylate use and colorectal cancer risk in inflammatory bowel disease: a large epidemiological study. Gut. 2005;54(11):1573-1578.

DOI: 10.1136/gut.2005.070896.

Brown WA, Farmer KC, Skinner SA, Malcontenti-Wilson C, Misajon A, O'brien PE. 5-Aminosalicylic acid and olsalazine inhibit tumor growth in a rodent model of colorectal cancer. Dig Dis Sci. 2000;45(8):1578-1584.

DOI: 10.1023/a:1005517112039.

Rubin DT, LoSavio A, Yadron N, Huo D, Hanauer SB. Aminosalicylate therapy in the prevention of dysplasia and colorectal cancer in ulcerative colitis. Clin Gastroenterol Hepatol. 2006;4(11):1346-1350.

DOI: 10.1016/j.cgh.2006.08.014.

Benno P, Alam M, Midtvedt T, Uribe A. Sulphasalazine, olsalazine and sulphapyridine induce mitogenic actions in the rat intestinal epithelium. APMIS. 1997;105(9):717-722.

DOI: 10.1111/j.1699-0463.1997.tb05076.x.

Medina-Franco JL, Méndez-Lucio O, Dueñas-González A, Yoo J. Discovery and development of DNA methyltransferase inhibitors using in silico approaches. Drug Discov Today. 2015;20(5):569-577.

DOI: 10.1016/j.drudis.2014.12.007.

Pfaffl MW, Horgan GW, Dempfle L. Relative expression software tool (REST) for group-wise comparison and statistical analysis of relative expression results in real-time PCR. Nucleic Acids Res. 2002;30(9): e36.

DOI: 10.1093/nar/30.9.e36.

Naghitorabi M, Saki G, Gharishvandi S. Effect of epigenetic drug candidate olsalazine on the expression of CDH1 and uPA genes in MCF-7 breast cancer cell line. Jundishapur J Nat Pharm Prod. 2020;15(4):e69428,1-6.

DOI: 10.5812/jjnpp.69428.

Theodossiou TA, Ali M, Grigalavicius M, Grallert B, Dillard P, Schink KO, et al. Simultaneous defeat of MCF7 and MDA-MB-231 resistances by a hypericin PDT-tamoxifen hybrid therapy. NPJ Breast Cancer. 2019;5:13-22.

DOI: 10.1038/s41523-019-0108-8.

Sharon P, Drab EA, Linder JS, Weidman SW, Rubin DB. The effect of sulfasalazine on bovine endothelial cell proliferation and cell cycle phase distribution: comparison with olsalazine, 5-aminosalicylic acid, and sulfapyridine. J Lab Clin Med. 1992;119(1): 99-107.

Kastl L, Brown I, Schofield AC. Effects of decitabine on the expression of selected endogenous control genes in human breast cancer cells. Mol Cell Probes. 2010;24(2):87-92.

DOI: 10.1016/j.mcp.2009.10.007.

Kar S, Sengupta D, Deb M, Shilpi A, Parbin S, Rath SK, et al. Expression profiling of DNA methylation-mediated epigenetic gene-silencing factors in breast cancer. Clin Epigenetics. 2014;6(1):20-32.

DOI: 10.1186/1868-7083-6-20.

Ari F, Napieralski R, Ulukaya E, Dere E, Colling C, Honert K, et al. Modulation of protein expression levels and DNA methylation status of breast cancer metastasis genes by anthracycline‐based chemotherapy and the demethylating agent decitabine. Cell Biochem Funct. 2011;29(8): 651-659.

DOI: 10.1002/cbf.1801.

Jabbour E, Issa JP, Garcia-Manero G, Kantarjian H. Evolution of decitabine development: accomplishments, ongoing investigations, and future strategies. Cancer. 2008;112(11):2341-2351.

DOI: 10.1002/cncr.23463.


Refbacks

  • There are currently no refbacks.


Creative Commons LicenseThis work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License which allows users to read, copy, distribute and make derivative works for non-commercial purposes from the material, as long as the author of the original work is cited properly.