Document Type : Research articles

Authors

1 MSc Student of Genetics, Department of Genetics, Faculty of Basic Sciences, Tabriz Branch, Islamic Azad University, Tabriz, Iran. inaz.mashayekhi@gmail.com

2 Assistant Professor, Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran. 2. Medical Genomics Research Center, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran.

3 Assistant Professor, Department of Genetics, Faculty of Basic Sciences, Tabriz Branch, Islamic Azad University

Abstract

Background: Colorectal cancer (CRC) has already been considered the fourth leading cause of mortality worldwide as the genes involved in apoptotic pathways and alterations of reversible epigenetic have an important role in the progression of CRC.     
Objectives: The current study aimed to evaluate the effect of sodium butyrate as a histone deacetylase inhibitor on the alterations of the gene expression of FAS, Fas ligand (FASL), Death receptor 4 (DR4), Death receptor 5 (DR5), and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in HCT-116 CRC cell line.
Methods: HCT-116 cell line was cultured in Dulbecco?s Modified Eagle Medium. The cytotoxicity effect of sodium butyrate on HCT-116 was evaluated using 3-(4, 5-dimethylthiazol-2-yl)-2, 5- diphenyltetrazolium bromide assay for three incubation times (i.e., 24, 48, and 72 h). The half-maximal inhibitory concentration (IC50) values were determined. The optimum concentration was within the range of 6.25-200 mM. The cellular ribonucleic acid was extracted, and complementary deoxyribonucleic acid was synthesized. Finally, the alterations of the gene expression of FAS, FASL, DR4, DR5, and TRAIL were assessed by real-time polymerase chain reaction (PCR).
Results: The IC50 levels for three incubation times were 50, 12.5, and 6.25 mM, respectively. The obtained results of real-time PCR demonstrated a significant increase in the gene expression of TRAIL, DR5, and FAS in comparison to that of the untreated cells as the control group at the three incubation times. The DR4 gene expression significantly increased in comparison to that reported for the control group at 48 and 72 h of incubation. In addition, FASL gene expression remarkably decreased at the three incubation times.
Conclusion: Sodium butyrate could show cytotoxicity effect on CRC cell lines through the induction of death receptors in the extrinsic apoptotic pathway. The obtained results of this study revealed that the optimum effect of sodium butyrate is an incubation time-dependent and concentration-dependent manner.

Keywords

  1. Wild CP, Stewart BW. World cancer report 2014: World Health Organization; 2014.
  2. Boyle P, Leon ME. Epidemiology of colorectal cancer. Br Med Bull. 2002;64:1-25. doi: 10.1093/bmb/64.1.1. [PubMed: 12421722].
  3. Rafiemanesh H, Pakzad R, Abedi M, Kor Y, Moludi J, Towhidi F, et al. Colorectal cancer in Iran: Epidemiology and morphology trends. EXCLI J. 2016;15:738-44. doi: 10.17179/excli2016-346. [PubMed: 28337105].
  4. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35(4):495-516. doi: 10.1080/01926230701320337. [PubMed: 17562483].
  5. Johnstone RW, Ruefli AA, Lowe SW. Apoptosis: a link between cancer genetics and chemotherapy. Cell. 2002;108(2):153-64. doi: 10.1016/s0092-8674(02)00625-6. [PubMed: 11832206].
  6. Fesik SW, Shi Y. Controlling the caspases. Science. 2001;294(5546):1477-8. doi: 10.1126/science.1062236.
  7. Raff M, Alberts B, Lewis J, Johnson A, Roberts K. Molecular Biology of the Cell 4th edition. National Center for Biotechnology InformationÕs Bookshelf; 2002.
  8. Green DR. Apoptosis: physiology and pathology: Cambridge University Press; 2011.
  9. Rodriguez I, Matsuura K, Khatib K, Reed JC, Nagata S, Vassalli P. A BCL-2 transgene expressed in hepatocytes protects mice from fulminant liver destruction but not rapid death induced by anti-Fas antibody injection. J Exp Med. 1996;183(3):1031-6. doi: 10.1084/jem.183.3.1031. [PubMed: 8642244].
  10. Bellgrau D, Gold D, Selawry H, Moore J, Franzusoff A, Duke RC. A role for CD95 ligand in preventing graft rejection. Nature. 1995;377(6550):630-2. doi: 10.1038/377630a0. [PubMed: 7566174].
  11. Ivanov VN, Bergami PL, Maulit G, Sato TA, Sassoon D, Ronai Ze. FAP-1 association with Fas (Apo-1) inhibits Fas expression on the cell surface. Mol Cellular Biol. 2003;23(10):3623-35. doi: 10.1128/mcb.23.10.3623-3635.2003. [PubMed: 12724420].
  12. Ivanov VN, Ronai Ze, Hei TK. Opposite roles of FAP-1 and dynamin in the regulation of  Fas (CD95) translocation to the cell surface and susceptibility to Fas ligand-mediated apoptosis. J Biol Chem. 2006;281(3):1840-52. doi: 10.1074/jbc.M509866200. [PubMed: 16306044].
  13. Pitti RM, Marsters SA, Ruppert S, Donahue CJ, Moore A, Ashkenazi A. Induction of apoptosis by Apo-2 ligand, a new member of the tumor necrosis factor cytokine family. J Biol Chem. 1996;271(22):12687-90. doi: 10.1074/jbc.271.22.12687. [PubMed: 8663110].
  14. Wang S, El-Deiry WS. TRAIL and apoptosis induction by TNF-family death receptors. Oncogene. 2003;22(53):8628-33. doi: 10.1038/sj.onc.1207232. [PubMed: 14634624].
  15. Dimberg LY, Anderson CK, Camidge R, Behbakht K, Thorburn A, Ford HL. On the TRAIL to successful cancer therapy? Predicting and counteracting resistance against TRAIL-based therapeutics. Oncogene. 2013;32(11):1341-50. doi: 10.1038/onc.2012.164. [PubMed: 22580613].
  16. Thomas LR, Johnson RL, Reed JC, Thorburn A. The C-terminal tails of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and Fas receptors have opposing functions in Fas-associated death domain (FADD) recruitment and can regulate agonist-specific mechanisms of receptor activation. J Biol Chem. 2004;279(50):52479-86. doi: 10.1074/jbc.M409578200. [PubMed: 15452120].
  17. Sträter J, Hinz U, Walczak H, Mechtersheimer G, Koretz K, Herfarth C, et al. Expression of TRAIL and TRAIL receptors in colon carcinoma: TRAIL-R1 is an independent prognostic parameter. Clin Cancer Res. 2002;8(12):3734-40. [PubMed: 12473583].
  18. Stuckey DW, Shah K. TRAIL on trial: preclinical advances in cancer therapy. Trends Mol Med. 2013;19(11):685-94. doi: 10.1016/j.molmed.2013.08.007. [PubMed: 24076237].
  19. Insinga A, Monestiroli S, Ronzoni S, Gelmetti V, Marchesi F, Viale A, et al. Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat Med. 2005;11(1):71-6. doi: 10.1038/nm1160. [PubMed: 15619634].
  20. Nebbioso A, Clarke N, Voltz E, Germain E, Ambrosino C, Bontempo P, et al. Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells. Nat Med. 2005;11(1):77-84. doi: 10.1038/nm1161. [PubMed: 15619633].
  21. Leonel AJ, Alvarez-Leite JI. Butyrate: implications for intestinal function. Curr Opin Clin Nutr Metab Care. 2012;15(5):474-9. doi: 10.1097/MCO.0b013e32835665fa. [PubMed: 22797568].
  22. Paskova L, Smesny Trtkova K, Fialova B, Benedikova A, Langova K, Kolar Z. Different effect of sodium butyrate on cancer and normal prostate cells. Toxicol In Vitro. 2013;27(5):1489-95. doi: 10.1016/j.tiv.2013.03.002. [PubMed: 23524101].
  23. Jia X, Zheng Y, Guo Y, Chen K. Sodium butyrate, and panobinostat induces apoptosis of chronic myeloid leukemia cells via multiple pathways. Mol Genet  Genomic Med. 2019;7(5):e613. doi: 10.1002/mgg3.613. [PubMed: 30891950].
  24. Gazzaniga MS, Ivry RB, Mangun GR. Cognitive Neuroscience: The Biology of the Mind: Fourth Edition: WW Norton; 2013.
  25. Malekzadeh R, Bishehsari F, Mahdavinia M, Ansari R. Epidemiology and molecular genetics of colorectal cancer in Iran: a review. Arch Iran Med. 2009;12(2):161-9.
  26. Gura T. How TRAIL kills cancer cells, but not normal cells. Science. 1997;277(5327):768. doi: 10.1126/science.277.5327.768. [PubMed: 9273698].
  27. Ferrante RJ, Kubilus JK, Lee J, Ryu H, Beesen A, Zucker B, et al. Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington's disease mice. J Neurosci. 2003;23(28):9418-27. doi: 10.1523/JNEUROSCI.23-28-09418.2003. [PubMed: 14561870].
  28. Xie C, Wu B, Chen B, Shi Q, Guo J, Fan Z, et al. Histone deacetylase inhibitor sodium butyrate suppresses proliferation and promotes apoptosis in osteosarcoma cells by regulation of the MDM2–p53 signaling. OncoTargets Ther. 2016;9:4005-13. doi: 10.2147/OTT.S105418. [PubMed: 27445491].
  29. Yamamura T, Matsumoto N, Matsue Y, Okudera M, Nishikawa Y, Abiko Y, et al. Sodium butyrate, a histone deacetylase inhibitor, regulates Lymphangiogenic factors in oral cancer cell line HSC-3. Anticancer Res. 2014;34(4):1701-8. [PubMed: 24692699].
  30. Bonnotte B, Favre N, Reveneau S, Micheau O, Droin N, Garrido C, et al. Cancer cell sensitization to fas-mediated apoptosis by sodium butyrate. Cell Death Differ. 1998;5(6):480-7. doi: 10.1038/sj.cdd.4400371. [PubMed: 10200499].
  31. Chopin V, Toillon RA, Jouy N, Bourhis XL. Sodium butyrate induces P53‐independent, Fas-mediated apoptosis in MCF‐7 human breast cancer cells. Br J Pharmacol. 2002;135(1):79-86. doi: 10.1038/sj.bjp.0704456. [PubMed: 11786482].
  32. Hernandez A, Thomas R, Smith F, Sandberg J, Kim S, Chung DH, et al. Butyrate sensitizes human colon cancer cells to TRAIL-mediated apoptosis. Surgery. 2001;130(2):265-72. doi: 10.1067/msy.2001.115897. [PubMed: 11490359].
  33. Kim YH, Park JW, Lee JY, Kwon TK. Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis. 2004;25(10):1813-20. doi: 10.1093/carcin/bgh188. [PubMed: 15142888].