Effects of all trans retinoic acid on apoptosis induction and notch1, hes1 genes expression in gastric cancer cell line MKN-45

authors:

avatar nazila niapour , avatar samira shokri gara geshlagi , avatar mojtaba amani , avatar marzieh sharifi pasandi , avatar hossein salehi , avatar ali niapour , *


how to cite: niapour N, shokri gara geshlagi S, amani M, sharifi pasandi M, salehi H, et al. Effects of all trans retinoic acid on apoptosis induction and notch1, hes1 genes expression in gastric cancer cell line MKN-45. koomesh. 2024;17(4):e151198. 

Abstract

Introduction: Gastric cancer is one of the most common and lethal malignancies in the world. All-trans retinoic acid (ATRA) has been widely used to treat cancers, most notably for the acute promyelocytic leukemia (APL) - as a differentiation inducer. Notch signaling plays important roles in cell proliferation and apoptosis. Aberrant activation of notch signaling pathway has been stated in different malignancies including gastric cancer. The aim of this study was to assess the possible effects of ATRA on cellular viability, cell cycle shift and apoptosis induction of gastric MKN-45 cell line. Moreover, alterations of notch1 and hes1 gene expression profiles after ATRA treatment have been considered. Materials and Methods: Human gastric carcinoma cells (MKN-45) were treated with increasing concentrations of ATRA (2.5, 5, 10, 15, 20, 25 µM). The viability of cells was determined with 3-(4, 5-dimethlthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Caspase-Glo 3/7 assay kit was applied to measure the apoptosis. Flow-cytometry was employed to distinguish cells in different phases of cell cycle. Expression profiles for the notch1 and hes1 genes were evaluated by reverse transcriptase PCR (RT-PCR). Results: MTT assay showed that ATRA could diminish MKN-45 cell line viability rate the most effective dose was 10 µM. Concentrations higher than 10 µM of ATRA had no significant effect on reducing cancerous cell viability. Furthermore, ATRA treatment significantly amplified caspase3/7activation. Flow-cytometry analyses showed significant accumulation of cells in G1 phase of cell cycle in ATRA treated group in compare to control. Following treatment with ATRA, showed a significant reduction in notch1and hes1 genes profile expression. Conclusion: According to our findings, ATRA could exert its cytotoxic effects on gastric cancer MKN-45 cell line through reducing cellular viability and inducing apoptosis. Remaining at G1 phase of cell cycle and reducing notch1 and hes1 gene expressions suggest the antiproliferative activity of ATRA

References

  • 1.

    Jin J, Li X, Xing L, Chang Y, Wu L, Jin Z, et al. Addition of all-trans-retinoic acid to omeprazole and sucralfate therapy improves the prognosis of gastric dysplasia. J Int Med Res 2015; 43: 204-216.

  • 2.

    Cheng B, Martinez AA, Morado J, Scofield V, Roberts JL, Maffi SK. Retinoic acid protects against proteasome inhibition associated cell death in SH-SY5Y cells via the AKT pathway. Neurochem Int 2013; 62: 31-42.

  • 3.

    Soprano DR, Qin P, Soprano KJ. Retinoic acid receptors and cancers. Annu Rev Nutr 2004; 24: 201-221.

  • 4.

    Ju J, Wang N, Wang X, Chen F. A novel all-trans retinoic acid derivative inhibits proliferation and induces differentiation of human gastric carcinoma xenografts via up-regulating retinoic acid receptor beta. Am J Translat Res 2015; 7: 856-865.

  • 5.

    Zhang JP, Chen XY, Li JS. Effects of all-trans-retinoic on human gastric cancer cells BGC-823. J Dig Dis 2007; 8: 29-34.

  • 6.

    Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer J Clin 2012; 62: 10-29.

  • 7.

    Yan B, Liu L, Zhao Y, Xiu LJ, Sun DZ, Liu X, et al. Xiaotan Sanjie decoction attenuates tumor angiogenesis by manipulating Notch-1-regulated proliferation of gastric cancer stem-like cells. World J Gastroenterol 2014; 20: 13105-13118.

  • 8.

    Brzozowa M, Mielanczyk L, Michalski M, Malinowski L, Kowalczyk-Ziomek G, Helewski K, et al. Role of Notch signaling pathway in gastric cancer pathogenesis. Contemp Oncol 2013; 17: 1-5.

  • 9.

    Du X, Cheng Z, Wang YH, Guo ZH, Zhang SQ, Hu JK, Zhou ZG. Role of Notch signaling pathway in gastric cancer: a meta-analysis of the literature. World J Gastroenterol 2014; 20: 9191-9199.

  • 10.

    Katoh M, Katoh M. Notch signaling in gastrointestinal tract (review). Int J Oncol 2007; 30: 247-251.

  • 11.

    Yuan R, Ke J, Sun L, He Z, Zou Y, He X, et al. HES1 promotes metastasis and predicts poor survival in patients with colorectal cancer. Clin Exp Metastasis 2015; 32: 169-179.

  • 12.

    Li LC, Peng Y, Liu YM, Wang LL, Wu XL. Gastric cancer cell growth and epithelial-mesenchymal transition are inhibited by gamma-secretase inhibitor DAPT. Oncol Lett 2014; 7: 2160-2164.

  • 13.

    Sethi N, Dai X, Winter CG, Kang Y. Tumor-derived JAGGED1 promotes osteolytic bone metastasis of breast cancer by engaging notch signaling in bone cells. Cancer Cell 2011; 19: 192-205.

  • 14.

    Lee HW, Kim SJ, Choi IJ, Song J, Chun KH. Targeting Notch signaling by gamma-secretase inhibitor I enhances the cytotoxic effect of 5-FU in gastric cancer. Clin Exp Metastasis 2015; 32: 593-603.

  • 15.

    Grieselhuber NR, Klco JM, Verdoni AM, Lamprecht T, Sarkaria SM, Wartman LD, Ley TJ. Notch signaling in acute promyelocytic leukemia. Leukemia 2013; 27: 1548-1557.

  • 16.

    Niapour N, Mohammadi-Ghalehbin B, Golmohammadi MG, Gholami MR, Amani M, Niapour A. An efficient system for selection and culture of Schwann cells from adult rat peripheral nerves. Cytotechnology 2015.

  • 17.

    Niapour N, Niapour A, Milan HS, Amani M, Salehi H, Najafzadeh N, Gholami MR. All trans retinoic acid modulates peripheral nerve fibroblasts viabilityand apoptosis. Tissue Cell 2015; 47: 61-65.

  • 18.

    Schippers KJ, Martens DE, Pomponi SA, Wijffels RH. Cell cycle analysis of primary sponge cell cultures. In Vitro Cell Dev Biol Anim 2011; 47: 302-311.

  • 19.

    Bohlooli S, Bohlooli S, Aslanian R, Nouri F, Teimourzadeh A. Aqueous extract of Agrostemma githago seed inhibits caspase-3 and induces cell-cycle arrest at G1 phase in AGS cell line. J Ethnopharmacol 2015; 175: 295-300.

  • 20.

    Niapour N, Taghipour Z, Salehi H, Bagheri A, Rouhani A, Talebi M, et al. Isolation and identification of mesenchymal and neural crest characteristics of dental pulp derived stem cells. Koomesh 2015; 16: 520-526.

  • 21.

    Zhu B, Smith J, Yarmush ML, Nahmias Y, Kirby BJ, Murthy SK. Microfluidic enrichment of mouse epidermal stem cells and validation of stem cell proliferation in vitro. Tissue Eng Part C Methods 2013; 19: 765-773.

  • 22.

    Lee JH, Yoon JH, Yu SJ, Chung GE, Jung EU, Kim HY, et al. Retinoic acid and its binding protein modulate apoptotic signals in hypoxic hepatocellular carcinoma cells. Cancer Lett 2010; 295: 229-235.

  • 23.

    Liu S, Wu Q, Chen ZM, Su WJ. The effect pathway of retinoic acid through regulation of retinoic acid receptor alpha in gastric cancer cells. World J Gastroenterol 2001; 7: 662-666.

  • 24.

    Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 2003; 425: 836-841.

  • 25.

    Ying M, Wang S, Sang Y, Sun P, Lal B, Goodwin CR, et al. Regulation of glioblastoma stem cells by retinoic acid: role for Notch pathway inhibition. Oncogene 2011; 30: 3454-3467.

  • 26.

    Murata K, Hattori M, Hirai N, Shinozuka Y, Hirata H, Kageyama R, et al. Hes1 directly controls cell proliferation through the transcriptional repression of p27Kip1. Mol Cell Biol 2005; 25: 4262-4271.

  • 27.

    Uyttendaele H, Marazzi G, Wu G, Yan Q, Sassoon D, Kitajewski J. Notch4/int-3, a mammary proto-oncogene, is an endothelial cell-specific mammalian Notch gene. Development 1996; 122: 2251-2259.