DOI QR코드

DOI QR Code

Ginsenoside Rh2 epigenetically regulates cell-mediated immune pathway to inhibit proliferation of MCF-7 breast cancer cells

  • Lee, Hyunkyung (Department of Life Science, Dongguk University-Seoul) ;
  • Lee, Seungyeon (Department of Life Science, Dongguk University-Seoul) ;
  • Jeong, Dawoon (Department of Life Science, Dongguk University-Seoul) ;
  • Kim, Sun Jung (Department of Life Science, Dongguk University-Seoul)
  • Received : 2017.01.17
  • Accepted : 2017.05.08
  • Published : 2018.10.15

Abstract

Background: Ginsenoside Rh2 has been known to enhance the activity of immune cells, as well as to inhibit the growth of tumor cells. Although the repertoire of genes regulated by Rh2 is well-known in many cancer cells, the epigenetic regulation has yet to be determined, especially for comprehensive approaches to detect methylation changes. Methods: The effect of Rh2 on genome-wide DNA methylation changes in breast cancer cells was examined by treating cultured MCF-7 with Rh2. Pyrosequencing analysis was carried out to measure the methylation level of a global methylation marker, LINE1. Genome-wide methylation analysis was carried out to identify epigenetically regulated genes and to elucidate the most prominent signaling pathway affected by Rh2. Apoptosis and proliferation were monitored to examine the cellular effect of Rh2. Results: LINE1 showed induction of hypomethylation at specific CpGs by 1.6-9.1% (p < 0.05). Genome-wide methylation analysis identified the "cell-mediated immune response"-related pathway as the top network. Cell proliferation of MCF-7 was retarded by Rh2 in a dose-dependent manner. Hypermethylated genes such as CASP1, INSL5, and OR52A1 showed downregulation in the Rh2-treated MCF-7, while hypomethylated genes such as CLINT1, ST3GAL4, and C1orf198 showed upregulation. Notably, a higher survival rate was associated with lower expression of INSL5 and OR52A1 in breast cancer patients, while with higher expression of CLINT1. Conclusion: The results indicate that Rh2 induces epigenetic methylation changes in genes involved in immune response and tumorigenesis, thereby contributing to enhanced immunogenicity and inhibiting the growth of cancer cells.

Keywords

References

  1. Jia L, Zhao Y. Current evaluation of the millennium phytomedicineeginseng (I): etymology, pharmacognosy, phytochemistry, market and regulations. Curr Med Chem 2009;16:2475-84. https://doi.org/10.2174/092986709788682146
  2. Christensen LP. Ginsenosides chemistry, biosynthesis, analysis, and potential health effects. Adv Food Nutr Res 2009;55:1-99.
  3. Wang W, Zhao Y, Rayburn ER, Hill DL, Wang H, Zhang R. In vitro anti-cancer activity and structure-activity relationships of natural products isolated from fruits of Panax ginseng. Cancer Chemother Pharmacol 2007;59:589-601. https://doi.org/10.1007/s00280-006-0300-z
  4. Wang CZ, Du GJ, Zhang Z, Wen XD, Calway T, Zhen Z, Musch MW, Bissonnette M, Chang EB, Yuan CS. Ginsenoside compound K, not Rb1, possesses potential chemopreventive activities in human colorectal cancer. Int J Oncol 2012;40:1970-6.
  5. Bi WY, Fu BD, Shen HQ, Wei Q, Zhang C, Song Z, Qin QQ, Li HP, Lv S, Wu SC, et al. Sulfated derivative of 20(S)-ginsenoside Rh2 inhibits inflammatory cytokines through MAPKs and NF-kappa B pathways in LPS-induced RAW264.7 macrophages. Inflammation 2012;35:1659-68. https://doi.org/10.1007/s10753-012-9482-1
  6. Yi PF, Bi WY, Shen HQ, Wei Q, Zhang LY, Dong HB, Bai HL, Zhang C, Song Z, Qin QQ, et al. Inhibitory effects of sulfated 20(S)-ginsenoside Rh2 on the release of pro-inflammatory mediators in LPS-induced RAW 264.7 cells. Eur J Pharmacol 2013;712:60-6. https://doi.org/10.1016/j.ejphar.2013.04.036
  7. Jeong MK, Cho CK, Yoo HS. General and genetic toxicology of enzyme-treated ginseng extract: toxicology of ginseng Rh2. J Pharmacopuncture 2016;19:213-24. https://doi.org/10.3831/KPI.2016.19.022
  8. Xu FY, Shang WQ, Yu JJ, Sun Q, Li MQ, Sun JS. The antitumor activity study of ginsenosides and metabolites in lung cancer cell. Am J Transl Res 2016;8:1708-18.
  9. Shi Q, Shi X, Zuo G, Xiong W, Li H, Guo P, Wang F, Chen Y, Li J, Chen DL. Anticancer effect of 20(S)-ginsenoside Rh2 on HepG2 liver carcinoma cells: Activating GSK-3beta and degrading beta-catenin. Oncol Rep 2016;36:2059-70. https://doi.org/10.3892/or.2016.5033
  10. Oh M, Choi YH, Choi S, Chung H, Kim K, Kim SI, Kim DK, Kim ND. Antiproliferating effects of ginsenoside Rh2 on MCF-7 human breast cancer cells. Int J Oncol 1999;14:869-75.
  11. Kim HS, Lee EH, Ko SR, Choi KJ, Park JH, Im DS. Effects of ginsenosides Rg3 and Rh2 on the proliferation of prostate cancer cells. Arch Pharm Res 2004;27:429-35. https://doi.org/10.1007/BF02980085
  12. Han S, Jeong AJ, Yang H, Bin Kang K, Lee H, Yi EH, Kim BH, Cho CH, Chung JW, Sung SH, et al. Ginsenoside 20(S)-Rh2 exerts anti-cancer activity through targeting IL-6-induced JAK2/STAT3 pathway in human colorectal cancer cells. J Ethnopharmacol 2016;194:83-90. https://doi.org/10.1016/j.jep.2016.08.039
  13. Wanderi C, Kim E, Chang S, Choi C, Choi K. Ginsenoside 20(S)-protopanaxadiol suppresses viability of human glioblastoma cells via down-regulation of cell adhesion proteins and cell-cycle arrest. Anticancer Res 2016;36:925-32.
  14. Zhang J, Lu M, Zhou F, Sun H, Hao G, Wu X, Wang G. Key role of nuclear factorkappaB in the cellular pharmacokinetics of adriamycin in MCF-7/Adr cells: the potential mechanism for synergy with 20(S)-ginsenoside Rh2. Drug Metab Dispos 2012;40:1900-8. https://doi.org/10.1124/dmd.112.045187
  15. Soes S, Daugaard IL, Sorensen BS, Carus A, Mattheisen M, Alsner J, Overgaard J, Hager H, Hansen LL, Kristensen LS. Hypomethylation and increased expression of the putative oncogene ELMO3 are associated with lung cancer development and metastases formation. Oncoscience 2014;1:367-74. https://doi.org/10.18632/oncoscience.42
  16. Tahara T, Shibata T, Nakamura M, Yamashita H, Yoshioka D, Okubo M, Yonemura J, Maeda Y, Maruyama N, Kamano T, et al. Association between IL-17A, -17F and MIF polymorphisms predispose to CpG island hypermethylation in gastric cancer. Int J Mol Med 2010;25:471-7.
  17. Schondorf T, Ebert MP, Hoffmann J, Becker M, Moser N, Pur S, Gohring UJ, Weisshaar MP. Hypermethylation of the PTEN gene in ovarian cancer cell lines. Cancer Lett 2004;207:215-20. https://doi.org/10.1016/j.canlet.2003.10.028
  18. Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, Lerma E, Bussaglia E, Prat J, Harkes IC, Repasky EA, et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst 2000;92:564-9. https://doi.org/10.1093/jnci/92.7.564
  19. Di Vinci A, Perdelli L, Banelli B, Salvi S, Casciano I, Gelvi I, Allemanni G, Margallo E, Gatteschi B, Romani M. p16(INK4a) Promoter methylation and protein expression in breast fibroadenoma and carcinoma. Int J Cancer 2005;114:414-21. https://doi.org/10.1002/ijc.20771
  20. Luo J, Li YN, Wang F, Zhang WM, Geng X. S-adenosylmethionine inhibits the growth of cancer cells by reversing the hypomethylation status of c-myc and H-ras in human gastric cancer and colon cancer. Int J Biol Sci 2010;6:784-95.
  21. Oshimo Y, Nakayama H, Ito R, Kitadai Y, Yoshida K, Chayama K, Yasui W. Promoter methylation of cyclin D2 gene in gastric carcinoma. Int J Oncol 2003;23:1663-70.
  22. Shigaki H, Baba Y, Watanabe M, Murata A, Iwagami S, Miyake K, Ishimoto T, Iwatsuki M, Baba H. LINE-1 hypomethylation in gastric cancer, detected by bisulfite pyrosequencing, is associated with poor prognosis. Gastric Cancer 2013;16:480-7. https://doi.org/10.1007/s10120-012-0209-7
  23. Park SB, Kim B, Bae H, Lee H, Lee S, Choi EH, Kim SJ. Differential epigenetic effects of atmospheric cold plasma on MCF-7 and MDA-MB-231 breast cancer cells. PLoS One 2015;10:e0129931. https://doi.org/10.1371/journal.pone.0129931
  24. Lee H, Lee S, Bae H, Kang HS, Kim SJ. Genome-wide identification of target genes for miR-204 and miR-211 identifies their proliferation stimulatory role in breast cancer cells. Sci Rep 2016;6:25287. https://doi.org/10.1038/srep25287
  25. Humphries AD, Streimann IC, Stojanovski D, Johnston AJ, Yano M, Hoogenraad NJ, Ryan MT. Dissection of the mitochondrial import and assembly pathway for human Tom40. J Biol Chem 2005;280:11535-43. https://doi.org/10.1074/jbc.M413816200
  26. Ott C, Ross K, Straub S, Thiede B, Gotz M, Goosmann C, Krischke M, Mueller MJ, Krohne G, Rudel T, et al. Sam50 functions in mitochondrial intermembrane space bridging and biogenesis of respiratory complexes. Mol Cell Biol 2012;32:1173-88. https://doi.org/10.1128/MCB.06388-11
  27. Mereiter S, Magalhaes A, Adamczyk B, Jin C, Almeida A, Drici L, Ibanez-Vea M, Gomes C, Ferreira JA, Afonso LP, et al. Glycomic analysis of gastric carcinoma cells discloses glycans as modulators of RON receptor tyrosine kinase activation in cancer. Biochim Biophys Acta 2016;1860:1795-808. https://doi.org/10.1016/j.bbagen.2015.12.016
  28. Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 2002;10:417-26. https://doi.org/10.1016/S1097-2765(02)00599-3
  29. Humke EW, Shriver SK, Starovasnik MA, Fairbrother WJ, Dixit VM. ICEBERG: a novel inhibitor of interleukin-1beta generation. Cell 2000;103:99-111. https://doi.org/10.1016/S0092-8674(00)00108-2
  30. Dodd ME, Hatzold J, Mathias JR, Walters KB, Bennin DA, Rhodes J, Kanki JP, Look AT, Hammerschmidt M, Huttenlocher A. The ENTH domain protein Clint1 is required for epidermal homeostasis in zebrafish. Development 2009;136:2591-600. https://doi.org/10.1242/dev.038448
  31. Suryawanshi A, Manicassamy S. Tumors induce immune tolerance through activation of beta-catenin/TCF4 signaling in dendritic cells: A novel therapeutic target for cancer immunotherapy. Oncoimmunology 2015;4:e1052932. https://doi.org/10.1080/2162402X.2015.1052932
  32. Mashima H, Ohno H, Yamada Y, Sakai T, Ohnishi H. INSL5 may be a unique marker of colorectal endocrine cells and neuroendocrine tumors. Biochem Biophys Res Commun 2013;432:586-92. https://doi.org/10.1016/j.bbrc.2013.02.042
  33. Barry KH, Moore LE, Liao LM, Huang WY, Andreotti G, Poulin M, Berndt SI. Prospective study of DNA methylation at LINE-1 and Alu in peripheral blood and the risk of prostate cancer. Prostate 2015;75:1718-25. https://doi.org/10.1002/pros.23053
  34. Nusgen N, Goering W, Dauksa A, Biswas A, Jamil MA, Dimitriou I, Sharma A, Singer H, Fimmers R, Frohlich H, et al. Inter-locus as well as intra-locus heterogeneity in LINE-1 promoter methylation in common human cancers suggests selective demethylation pressure at specific CpGs. Clin Epigenetics 2015;7:17. https://doi.org/10.1186/s13148-015-0051-y
  35. Bakshi A, Herke SW, Batzer MA, Kim J. DNA methylation variation of humanspecific Alu repeats. Epigenetics 2016;11:163-73. https://doi.org/10.1080/15592294.2015.1130518
  36. Li J, Huang Q, Zeng F, Li W, He Z, Chen W, Zhu W, Zhang B. The prognostic value of global DNA hypomethylation in cancer: a meta-analysis. PLoS One 2014;9:e106290. https://doi.org/10.1371/journal.pone.0106290
  37. De Araujo ES, Kashiwabara AY, Achatz MI, Moredo LF, De Sa BC, Duprat JP, Rosenberg C, Carraro DM, Krepischi AC. LINE-1 hypermethylation in peripheral blood of cutaneous melanoma patients is associated with metastasis. Melanoma Res 2015;25:173-7. https://doi.org/10.1097/CMR.0000000000000141
  38. Nakagawa T, Zhu H, Morishima N, Li E, Xu J, Yankner BA, Yuan J. Caspase-12 mediates endoplasmic-reticulum-specific apoptosis and cytotoxicity by amyloid-beta. Nature 2000;403:98-103. https://doi.org/10.1038/47513
  39. Muzes G, Sipos F. Inflammasome, inflammation and cancer: an interrelated pathobiological triad. Curr Drug Targets 2015;16:249-57. https://doi.org/10.2174/1389450115666141229154157
  40. Guo W, Sun Y, Liu W, Wu X, Guo L, Cai P, Wu X, Wu X, Shen Y, Shu Y, et al. Small molecule-driven mitophagy-mediated NLRP3 inflammasome inhibition is responsible for the prevention of colitis-associated cancer. Autophagy 2014;10:972-85. https://doi.org/10.4161/auto.28374
  41. Wen D, Liu D, Tang J, Dong L, Liu Y, Tao Z, Wan J, Gao D, Wang L, Sun H, et al. Malic enzyme 1 induces epithelial-mesenchymal transition and indicates poor prognosis in hepatocellular carcinoma. Tumour Biol 2015;36:6211-21. https://doi.org/10.1007/s13277-015-3306-5
  42. Liu D, Huang Y, Zeng J, Chen B, Huang N, Guo N, Liu L, Xu H, Mo X, Li W. Downregulation of JAK1 by RNA interference inhibits growth of the lung cancer cell line A549 and interferes with the PI3K/mTOR pathway. J Cancer Res Clin Oncol 2011;137:1629-40. https://doi.org/10.1007/s00432-011-1037-6
  43. Zhang MX, Zhao X, Wang ZG, Zhao WM, Wang YS. Constitutive activation of signal transducer and activator of transcription 3 regulates expression of vascular endothelial growth factor in human meningioma differentiation. J Cancer Res Clin Oncol 2010;136:981-8. https://doi.org/10.1007/s00432-009-0743-9
  44. Wang J, Ding S, Duan Z, Xie Q, Zhang T, Zhang X, Wang Y, Chen X, Zhuang H, Lu F. Role of p14ARF-HDM2-p53 axis in SOX6-mediated tumor suppression. Oncogene 2016;35:1692-702. https://doi.org/10.1038/onc.2015.234
  45. Zhao D, Zhang Q, Liu Y, Li X, Zhao K, Ding Y, Li Z, Shen Q, Wang C, Li N, et al. H3K4me3 demethylase Kdm5a is required for NK cell activation by associating with p50 to suppress SOCS1. Cell Rep 2016;15:288-99. https://doi.org/10.1016/j.celrep.2016.03.035

Cited by

  1. Mechanistic evaluation of phytochemicals in breast cancer remedy: current understanding and future perspectives vol.8, pp.52, 2018, https://doi.org/10.1039/c8ra04879g
  2. Ginsenoside Rh2 Suppresses Breast Cancer Cell Proliferation by Epigenetically Regulating the Long Noncoding RNA C3orf67-AS1 vol.47, pp.7, 2018, https://doi.org/10.1142/s0192415x19500848
  3. Inhibitory Effects of Ginsenoside Ro on the Growth of B16F10 Melanoma via Its Metabolites vol.24, pp.16, 2019, https://doi.org/10.3390/molecules24162985
  4. Protopanaxadiol inhibits epithelial-mesenchymal transition of hepatocellular carcinoma by targeting STAT3 pathway vol.10, pp.9, 2019, https://doi.org/10.1038/s41419-019-1733-8
  5. Recent Advances in Ginsenosides as Potential Therapeutics Against Breast Cancer vol.19, pp.25, 2018, https://doi.org/10.2174/1568026619666191018100848
  6. 호르몬 수용체 및 HER-2 수용체 양성 유방암 4기 다발성 전이 환자의 6년간의 한양방 병용치료 치험 1례 vol.24, pp.2, 2018, https://doi.org/10.15432/jkto.2019.24.2.013
  7. Assessment of a New Ginsenoside Rh2 Nanoniosomal Formulation for Enhanced Antitumor Efficacy on Prostate Cancer: An in vitro Study vol.14, pp.None, 2018, https://doi.org/10.2147/dddt.s261027
  8. A cocktail of betulinic acid, parthenolide, honokiol and ginsenoside Rh2 in liposome systems for lung cancer treatment vol.15, pp.1, 2018, https://doi.org/10.2217/nnm-2018-0479
  9. The Preparation of Ginsenoside Rg5, Its Antitumor Activity against Breast Cancer Cells and Its Targeting of PI3K vol.12, pp.1, 2018, https://doi.org/10.3390/nu12010246
  10. Medicinal Herbs Used in Traditional Management of Breast Cancer: Mechanisms of Action vol.7, pp.8, 2018, https://doi.org/10.3390/medicines7080047
  11. Anticancer Activities of Ginsenosides, the Main Active Components of Ginseng vol.2021, pp.None, 2021, https://doi.org/10.1155/2021/8858006
  12. Immune System Effects of Insulin-Like Peptide 5 in a Mouse Model vol.11, pp.None, 2018, https://doi.org/10.3389/fendo.2020.610672
  13. Pseudo-ginsenoside Rh2 Induces Protective Autophagy in Hepatocellular Carcinoma HepG2 Cells vol.16, pp.None, 2018, https://doi.org/10.2174/1574892816666210607100239
  14. Role of Bioactive Constituents of Panax notoginseng in the Modulation of Tumorigenesis: A Potential Review for the Treatment of Cancer vol.12, pp.None, 2018, https://doi.org/10.3389/fphar.2021.738914
  15. Epigenetic Studies of Chinese Herbal Medicine: Pleiotropic Role of DNA Methylation vol.12, pp.None, 2021, https://doi.org/10.3389/fphar.2021.790321
  16. Bioactivity-based analysis and chemical characterization of cytotoxic compounds from a poisonous mushroom, Amanita spissacea, in human lung cancer cells in vitro vol.35, pp.4, 2018, https://doi.org/10.1080/14786419.2019.1586699
  17. Role of Phytonutrients in Nutrigenetics and Nutrigenomics Perspective in Curing Breast Cancer vol.11, pp.8, 2018, https://doi.org/10.3390/biom11081176
  18. Anti-Cancer Effect of Panax Ginseng and Its Metabolites: From Traditional Medicine to Modern Drug Discovery vol.9, pp.8, 2021, https://doi.org/10.3390/pr9081344