Silymarin induces inhibition of growth and apoptosis through modulation of the MAPK signaling pathway in AGS human gastric cancer cells

Sung-Hyun Kim, Gang-Sik Choo, Eun-Seon Yoo, Joong-Seok Woo, So-Hee Han, Jae-Han Lee, Ji-Youn Jung, Sung-Hyun Kim, Gang-Sik Choo, Eun-Seon Yoo, Joong-Seok Woo, So-Hee Han, Jae-Han Lee, Ji-Youn Jung

Abstract

Apoptosis is regarded as a therapeutic target because it is typically disturbed in human cancer. Silymarin from milk thistle (Silybum marianum) has been reported to exhibit anticancer properties via regulation of apoptosis as well as anti‑inflammatory, antioxidant and hepatoprotective effects. In the present study, the effects of silymarin on the inhibition of proliferation and apoptosis were examined in human gastric cancer cells. The viability of AGS human gastric cancer cells was assessed by MTT assay. The migration of AGS cells was investigated by wound healing assay. Silymarin was revealed to significantly decrease viability and migration of AGS cells in a concentration‑dependent manner. In addition, the number of apoptotic bodies and the rate of apoptosis were increased in a dose‑dependent manner as determined by DAPI staining and Annexin V/propidium iodide double staining. The changes in the expression of silymarin‑induced apoptosis proteins were investigated in human gastric cancer cells by western blotting analysis. Silymarin increased the expression of Bax, phosphorylated (p)‑JNK and p‑p38, and cleaved poly‑ADP ribose polymerase, and decreased the levels of Bcl‑2 and p‑ERK1/2 in a concentration‑dependent manner. The in vivo tumor growth inhibitory effect of silymarin was investigated. Silymarin (100 mg/kg) significantly decreased the AGS tumor volume and increased apoptosis, as assessed by the TUNEL assay, confirming its tumor‑inhibitory effect. Immunohistochemical staining revealed elevated expression of p‑JNK and p‑p38 as well as reduced expression of p‑ERK1/2 associated with silymarin‑treatment. Silymarin was revealed to reduce tumor growth through inhibition of p‑ERK and activation of p‑p38 and p‑JNK in human gastric cancer cells. These results indicated that silymarin has potential for development as a cancer therapeutic due to its growth inhibitory effects and induction of apoptosis in human gastric cancer cells.

Figures

Figure 1.
Figure 1.
Effects of silymarin on the cell viability of AGS human gastric cancer cells. The results are revealed cell viability inhibitory effect of silymarin on AGS cells. Cell viability was measured by the MTT assay. The results are presented as the mean ± SD from three independent experiments performed in triplicate. Significance was determined by Dunnett's t-test with *P<0.05 considered as statistically significant compared with non-treated controls.
Figure 2.
Figure 2.
Effect of silymarin on the cell migration of AGS human gastric cancer cells. (A) AGS gastric cancer cells were treated with silymarin 0, 40 and 80 µg/ml for 24 h, and cell migration was then measured by wound healing assays. (B) Width of the wound for any field of view was measured at 3–4 different locations. Data represent the means ± SD of at least three replicates relative to the control (Dunnett's t-test with *P<0.05).
Figure 3.
Figure 3.
Effects of silymarin on apoptotic cell in AGS human gastric cancer cells. (A) AGS cells were treated with silymarin for 24 h and apoptotic bodies stained with DAPI. The arrows indicate chromatin condensation in the AGS cells. Fragmentation of nuclear DNA was examined by a fluorescence microscope (×200). Arrows indicate apoptotic bodies, which represent DNA fragments produced during apoptosis. (B) AGS cells were treated with silymarin for 24 h and nuclear condensation was examined by DAPI staining. Graphs reveal quantification of DNA fragmentation and nuclear condensation. Each bar represents the mean ± SD calculated from five independent experiments. *Pt-test).
Figure 4.
Figure 4.
Effect of silymarin on apoptosis in AGS human gastric cancer cells. (A) The apoptotic effects of silymarin at different concentrations were evaluated by Annexin V-fluorescein and propidium iodide (PI) staining. AGS cells were treated with silymarin (0, 40, and 80 µg/ml) for 24 h followed by flow cytometry. The lower left quadrant includes viable cells, which were negative for Annexin V-FITC binding (Annexin V−) and exclude PI (PI−); the lower right quadrant includes early apoptotic cells, which are positive for Annexin V-FITC binding (Annexin V+) but PI−; the upper right quadrant includes late apoptotic cells, which are Annexin V+ and reveal PI uptake (PI+); and the upper left quadrant includes necrotic cells, which are Annexin V−/PI+. (B) Annexin V/PI staining was revealed alive cell/dead cell/early-late apoptotic cell fluorescence intensity ratios.
Figure 5.
Figure 5.
Effects of silymarin on apoptosis-related protein in AGS human gastric cancer cells. (A) AGS cells were treated with silymarin (0, 40 and 80 µg/ml) for 24 h and cells were harvested to assess protein levels of Bax, Bcl-2, and PARP by western blotting. The blots were also probed with anti-β-actin antibody to confirm equal sample loading. (B) Each bar represents the mean ± SD calculated from three independent experiments. *Pt-test).
Figure 6.
Figure 6.
Effect of silymarin on the activation of MAPK pathway in AGS human gastric cancer cells. (A) AGS cells were treated with silymarin (0, 40 and 80 µg/ml) for 24 h. Cell lysates were prepared as described in Materials and methods, and analyzed by 12% SDS-PAGE followed by western blotting. (B) Each bar represents the mean ± SD calculated from independent experiments. *Pt-test). MAPK, mitogen-activated protein kinase.
Figure 7.
Figure 7.
Effects of silymarin on AGS gastric cancer tumor xenograft growth and apoptosis in tumor tissues. Nude mice bearing AGS cells as xenograft models were treated with silymarin for 14 days, and (A) tumor volume, (B) tumor weight, and (C) body weight were determined. (D and E) Apoptosis was measured in tumor tissues by TUNEL assay. Slides were observed under an optical microscope (×200). Scale bar, 10 µm. *Pt-test).
Figure 8.
Figure 8.
Effects of silymarin on MAPK expression in AGS gastric cancer tumor xenografts. (A) Nude mice were administered silymarin (0, 100 mg/kg) for 2 weeks and examined by immunohistochemistry using antibodies to p-ERK, p-JNK, and p-p38. Tumor tissues were observed under an optical microscope and images were captured at an ×200 magnification. Paraffin-embedded tumor tissues were cut into sections 5-µm thick. Scale bar, 10 µm. (B) Graphs reveal quantification of each protein (p-ERK1/2, p-JNK and p-p38) with positive staining as determined by immunohistochemistry. Significance was determined by Dunnett's t-test with *P<0.05 considered as statistically significant compared with non-treated controls. MAPK, mitogen-activated protein kinase.
Figure 9.
Figure 9.
Histological analysis of silymarin-treated nude mice. The microscopic pathology of the liver and kidney revealed no evidence of adverse systemic toxicity of silymarin treatment in the mice. Slides were observed under an optical microscope (×200). Scale bar, 10 µm.

References

    1. Jung KW, Won YJ, Kong HJ, Lee ES. Prediction of cancer incidence and mortality in Korea, 2019. Cancer Res Treat. 2019;51:431–437. doi: 10.4143/crt.2019.139.
    1. Won YJ, Sung J, Jung KW, Kong HJ, Park S, Shin HR, Park EC, Ahn YO, Hwang IK, Lee DH, et al. Nationwide cancer incidence in Korea, 2003–2005. Cancer Res Treat. 2009;41:122–131. doi: 10.4143/crt.2009.41.3.122.
    1. Jung KW, Won YJ, Kong HJ, Oh CM, Lee DH, Lee JS. Cancer statistics in Korea: Incidence, mortality, survival, and prevalence in 2011. Cancer Res Treat. 2014;46:109–123. doi: 10.4143/crt.2014.46.2.109.
    1. Jemal A, Center MM, DeSantis C, Ward EM. Global patterns of cancer incidence and mortality rates and trends. Cancer Epidemiol Biomarkers Prev. 2010;19:1893–1907. doi: 10.1158/1055-9965.EPI-10-0437.
    1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127:2893–2917. doi: 10.1002/ijc.25516.
    1. Forman D, Burley VJ. Gastric cancer: Global pattern of the disease and an overview of environmental risk factors. Best Pract Res Clin Gastroenterol. 2006;20:633–649. doi: 10.1016/j.bpg.2006.04.008.
    1. Lee MS, Cha EY, Thuong PT, Kim JY, Ahn MS, Sul JY. Down-regulation of human epidermal growth factor receptor 2/neu oncogene by corosolic acid induces cell cycle arrest and apoptosis in NCI-N87 human gastric cancer cells. Biol Pharm Bull. 2010;33:931–937. doi: 10.1248/bpb.33.931.
    1. Li N, Fan LL, Sun GP, Wan XA, Wang ZG, Wu Q, Wang H. Paeonol inhibits tumor growth in gastric cancer in vitro and in vivo. World J Gastroenterol. 2010;16:4483–4490. doi: 10.3748/wjg.v16.i35.4483.
    1. Zhang L, Hou YH, Wu K, Zhai JS, Lin N. Proteomic analysis reveals molecular biological details in varioliform gastritis without helicobacter pylori infection. World J Gastroenterol. 2010;16:3664–3673. doi: 10.3748/wjg.v16.i29.3664.
    1. Seeram NP. Berry fruits for cancer prevention: Current status and future prospects. J Agric Food Chem. 2008;56:630–635. doi: 10.1021/jf072504n.
    1. An IJ, Kwon JK, Lee JS, Park HS, Kim DC, Choi BJ, Lee KM, Park YJ, Jung JY. Induction of apoptosis in human cancer cells with compositae extracts. J Korean Soc Food Sci Nutr. 2012;41:584–590. doi: 10.3746/jkfn.2012.41.5.584.
    1. Kwon MJ, Nam TJ. A polysaccharide of the marine alga capsosiphon fulvescens induces apoptosis in AGS gastric cancer cells via an IGF-IR-mediated PI3K/Akt pathway. Cell Biol Int. 2007;31:768–775. doi: 10.1016/j.cellbi.2007.01.010.
    1. Schuier M, Sies H, Illek B, Fischer H. Cocoa-related flavonoids inhibit CFTR-mediated chloride transport across T84 human colon epithelia. J Nutr. 2005;135:2320–2325. doi: 10.1093/jn/135.10.2320.
    1. Murakami A, Ashida H, Terao J. Multitargeted cancer prevention by quercetin. Cancer Lett. 2008;269:315–325. doi: 10.1016/j.canlet.2008.03.046.
    1. Ren W, Qiao Z, Wang H, Zhu L, Zhang L. Flavonoids: Promising anticancer agents. Med Res Rev. 2003;23:519–534. doi: 10.1002/med.10033.
    1. Hackett ES, Twedt DC, Gustafson DL. Milk thistle and its derivative compounds: A review of opportunities for treatment of liver disease. J Vet Intern Med. 2013;27:10–16. doi: 10.1111/jvim.12002.
    1. Chen CH, Huang TS, Wong CH, Hong CL, Tsai YH, Liang CC, Lu FJ, Chang WH. Synergistic anti-cancer effect of baicalein and silymarin on human hepatoma HepG2 Cells. Food Chem Toxicol. 2009;47:638–644. doi: 10.1016/j.fct.2008.12.024.
    1. Ramasamy K, Agarwal R. Multitargeted therapy of cancer by silymarin. Cancer Lett. 2008;269:352–362. doi: 10.1016/j.canlet.2008.03.053.
    1. Surai PF. Silymarin as a natural antioxidant: An overview of the current evidence and perspectives. Antioxidants (Basel) 2015;4:204–247. doi: 10.3390/antiox4010204.
    1. Katiyar SK. Silymarin and skin cancer prevention: Anti-inflammatory, antioxidant and immunomodulatory effects (Review) Int J Oncol. 2005;26:169–176.
    1. Féher J, Lengyel G. Silymarin in the prevention and treatment of liver diseases and primary liver cancer. Curr Pharm Biotechnol. 2012;13:210–217. doi: 10.2174/138920112798868818.
    1. Eo HJ, Park GH, Jeong JB. Inhibition of Wnt signaling by silymarin in human colorectal cancer cells. Biomol Ther (Seoul) 2016;24:380–386. doi: 10.4062/biomolther.2015.154.
    1. Hajighasemlou S, Farajollahi M, Alebouyeh M, Rastegar H, Manazi MT, Mirmoghtadaei M, Moayedi B, Ahmadzadeh M, Kazemi M, Parvizpour F, Gharibzadeh S. Study of the effect of silymarin on viability of breast cancer cell lines. Adv Breast Cancer Res. 2014;3:100–105. doi: 10.4236/abcr.2014.33015.
    1. Wu T, Liu W, Guo W, Zhu X. Silymarin suppressed lung cancer growth in mice via inhibiting myeloid-derived suppressor cells. Biomed Pharmacother. 2016;81:460–467. doi: 10.1016/j.biopha.2016.04.039.
    1. Deep G, Singh RP, Agarwal C, Kroll DJ, Agarwal R. Silymarin and silibinin cause G1 and G2-M cell cycle arrest via distinct circuitries in human prostate cancer PC3 cells: A comparison of flavanone silibinin with flavanolignan mixture silymarin. Oncogene. 2006;25:1053–1069. doi: 10.1038/sj.onc.1209146.
    1. Hsu HF, Houng JY, Kuo CF, Tsao N, Wu YC. Glossogin, a novel phenylpropanoid from glossogyne tenuifolia, induced apoptosis in A549 lung cancer cells. Food Chem Toxicol. 2008;46:3785–3791. doi: 10.1016/j.fct.2008.09.068.
    1. Kaufmann T, Strasser A, Jost PJ. Fas death receptor signalling: Roles of Bid and XIAP. Cell Death Differ. 2012;19:42–50. doi: 10.1038/cdd.2011.121.
    1. Tummers B, Green DR. Caspase-8: Regulating life and death. Immunol Rev. 2017;277:76–89. doi: 10.1111/imr.12541.
    1. Kim D, Chung J. Akt: Versatile mediator of cell survival and beyond. J Biochem Mol Biol. 2002;35:106–115.
    1. Burz C, Berindan-Neagoe I, Balacescu O, Irimie A. Apoptosis in cancer: Key molecular signaling pathways and therapy targets. Acta Oncol. 2009;48:811–821. doi: 10.1080/02841860902974175.
    1. Soldani C, Scovassi AI. Poly (ADP-ribose) polymerase-1 cleavage during apoptosis: An update. Apoptosis. 2002;7:321–328. doi: 10.1023/A:1016119328968.
    1. Kim EK, Choi EJ. Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta. 2010;1802:396–405. doi: 10.1016/j.bbadis.2009.12.009.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Ramakrishnan G, Lo Muzio L, Elinos-Báez CM, Jagan S, Augustine TA, Kamaraj S, Anandakumar P, Devaki T. Silymarin inhibited proliferation and induced apoptosis in hepatic cancer cells. Cell Prolif. 2009;42:229–240. doi: 10.1111/j.1365-2184.2008.00581.x.
    1. Zhong X, Zhu Y, Lu Q, Zhang J, Ge Z, Zheng S. Silymarin causes caspases activation and apoptosis in K562 leukemia cells through inactivation of Akt pathway. Toxicology. 2006;227:211–216. doi: 10.1016/j.tox.2006.07.021.
    1. Fan L, Ma Y, Liu Y, Zheng D, Huang G. Silymarin induces cell cycle arrest and apoptosis in ovarian cancer cells. Eur J Pharmacol. 2014;743:79–88. doi: 10.1016/j.ejphar.2014.09.019.
    1. Vaid M, Prasad R, Sun Q, Katiyar SK. Silymarin targets β-catenin signaling in blocking migration/invasion of human melanoma cells. PLoS One. 2011;6:e23000. doi: 10.1371/journal.pone.0023000.
    1. Danial NN, Korsmeyer SJ. Cell death: Critical control points. Cell. 2004;116:205–219. doi: 10.1016/S0092-8674(04)00046-7.
    1. Halicka HD, Bedner E, Darzynkiewicz Z. Segregation of RNA and separate packaging of DNA and RNA in apoptotic bodies during apoptosis. Exp Cell Res. 2000;260:248–256. doi: 10.1006/excr.2000.5027.
    1. Katiyar SK, Roy AM, Baliga MS. Silymarin induces apoptosis primarily through a p53-dependent pathway involving Bcl-2/Bax, cytochrome c release, and caspase activation. Mol Cancer Ther. 2005;4:207–216.
    1. Vaid M, Singh T, Prasad R, Katiyar SK. Silymarin inhibits melanoma cell growth both in vitro and in vivo by targeting cell cycle regulators, angiogenic biomarkers and induction of apoptosis. Mol Carcinog. 2015;54:1328–1339. doi: 10.1002/mc.22208.
    1. Yan C, Yahua WU, Wang Z, Yin C, Yin M. Inhibitory effect of silymarin on human osteosarcoma Saos-2 cells and its mechanism. Chin Pharmacol Bull. 2016;32:966–969. (In Chinese)
    1. Huang Q, Wu LJ, Tashiro S, Onodera S, Li LH, Ikejima T. Silymarin augments human cervical cancer HeLa cell apoptosis via P38/JNK MAPK pathways in serum-free medium. J Asian Nat Prod Res. 2005;7:701–709. doi: 10.1080/1028602042000324862.
    1. Won DH, Kim LH, Jang B, Yang IH, Kwon HJ, Jin B, Oh SH, Kang JH, Hong SD, Shin JA, Cho SD. In vitro and in vivo anti-cancer activity of silymarin on oral cancer. Tumor Biol. 2018;40:1010428318776170. doi: 10.1177/1010428318776170.
    1. Singh RP, Tyagi AK, Zhao J, Agarwal R. Silymarin inhibits growth and causes regression of established skin tumors in SENCAR mice via modulation of mitogen-activated protein kinases and induction of apoptosis. Carcinogenesis. 2002;23:499–510. doi: 10.1093/carcin/23.3.499.

Source: PubMed

3
Subscribe