DOI QR코드

DOI QR Code

Fermented ginseng extract, BST204, disturbs adipogenesis of mesenchymal stem cells through inhibition of S6 kinase 1 signaling

  • Received : 2018.05.29
  • Accepted : 2018.08.03
  • Published : 2020.01.15

Abstract

Background: The biological and pharmacological effects of BST204, a fermented ginseng extract, have been reported in various disease conditions. However, its molecular action in metabolic disease remains poorly understood. In this study, we identified the antiadipogenic activity of BST204 resulting from its inhibition of the S6 kinase 1 (S6K1) signaling pathway. Methods: The inhibitory effects of BST204 on S6K1 signaling were investigated by immunoblot, nuclear fractionation, immunoprecipitation analyses. The antiadipogenic effect of BST204 was evaluated by measuring mRNA levels of adipogenic genes and by chromatin immunoprecipitation and quantitative real-time polymerase chain reaction analysis. Results: Treatment with BST204 inhibited activation and nuclear translocation of S6K1, further decreasing the interaction between S6K1 and histone H2B in 10T1/2 mesenchymal stem cells. Subsequently, phosphorylation of H2B at serine 36 (H2BS36p) by S6K1 was reduced by BST204, inducing an increase in the mRNA expression of Wnt6, Wnt10a, and Wnt10b, which disturbed adipogenic differentiation and promoted myogenic and early osteogenic gene expression. Consistently, BST204 treatment during adipogenic commitment suppressed the expression of adipogenic marker genes and lipid drop formation. Conclusion: Our results indicate that BST204 blocks adipogenesis of mesenchymal stem cells through the inhibition of S6K1-mediated histone phosphorylation. This study suggests the potential therapeutic strategy using BST204 to combat obesity and musculoskeletal diseases.

Keywords

References

  1. Ma XM, Blenis J. Molecular mechanisms of mTOR-mediated translational control. Nat Rev Mol Cell Biol 2009;10:307-18. https://doi.org/10.1038/nrm2672
  2. Ben-Sahra I, Howell JJ, Asara JM, Manning BD. Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1. Science 2013;339:1323-8. https://doi.org/10.1126/science.1228792
  3. Robitaille AM, Christen S, Shimobayashi M, Cornu M, Fava LL, Moes S, Prescianotto-Baschong C, Sauer U, Jenoe P, Hall MN. Quantitative phosphoproteomics reveal mTORC1 activates de novo pyrimidine synthesis. Science 2013;339:1320-3. https://doi.org/10.1126/science.1228771
  4. Owen JL, Zhang Y, Bae SH, Farooqi MS, Liang G, Hammer RE, Goldstein JL, Brown MS. Insulin stimulation of SREBP-1c processing in transgenic rat hepatocytes requires p70 S6-kinase. Proc Natl Acad Sci USA 2012;109:16184-9. https://doi.org/10.1073/pnas.1213343109
  5. Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M, Fumagalli S, Allegrini PR, Kozma SC, Auwerx J, et al. Absence of S6K1 protects against ageand diet-induced obesity while enhancing insulin sensitivity. Nature 2004;431:200-5. https://doi.org/10.1038/nature02866
  6. Carnevalli LS, Masuda K, Frigerio F, Le Bacquer O, Um SH, Gandin V, Topisirovic I, Sonenberg N, Thomas G, Kozma SC. S6K1 plays a critical role in early adipocyte differentiation. Dev Cell 2010;18:763-4. https://doi.org/10.1016/j.devcel.2010.02.018
  7. Yi SA, Um SH, Lee J, Yoo JH, Bang SY, Park EK, Lee MG, Nam KH, Jeon YJ, Park JW, et al. S6K1 phosphorylation of H2B mediates EZH2 trimethylation of H3: a determinant of early adipogenesis. Mol Cell 2016;62:443-52. https://doi.org/10.1016/j.molcel.2016.03.011
  8. Seo JY, Lee JH, Kim NW, Her E, Chang SH, Ko NY, Yoo YH, Kim JW, Seo DW, Han JW, et al. Effect of a fermented ginseng extract, BST204, on the expression of cyclooxygenase-2 in murine macrophages. Int Immunopharmacol 2005;5:929-36. https://doi.org/10.1016/j.intimp.2005.01.008
  9. Seo JY, Lee JH, Kim NW, Kim YJ, Chang SH, Ko NY, Her E, Yoo YH, Kim JW, Lee BY, et al. Inhibitory effects of a fermented ginseng extract, BST204, on the expression of inducible nitric oxide synthase and nitric oxide production in lipopolysaccharide-activated murine macrophages. J Pharm Pharmacol 2005;57:911-8. https://doi.org/10.1211/0022357056497
  10. Park JW, Lee JC, Ann S, Seo DW, Choi WS, Yoo YH, Park SH, Choi JY, Um SH, Ahn SH, et al. A fermented ginseng extract, BST204, inhibits proliferation and motility of human colon cancer cells. Biomol Ther 2011;19:211-7. https://doi.org/10.4062/biomolther.2011.19.2.211
  11. Park HJ, Shim HS, Kim JY, Kim JY, Park SK, Shim I. Ginseng purified dry extract, BST204, improved cancer chemotherapy-related fatigue and toxicity in mice. Evid Based Complement Alternat Med 2015;2015:197459.
  12. Yang H, Yoo G, Kim HS, Kim JY, Kim SO, Yoo YH, Sung SH. Implication of the stereoisomers of ginsenoside derivatives in the antiproliferative effect of HSCT6 cells. J Agric Food Chem 2012;60:11759-64. https://doi.org/10.1021/jf303714c
  13. Park JW, Lee JC, Ha SW, Bang SY, Park EK, Yi SA, Lee MG, Kim DS, Nam KH, Yoo JH, et al. Requirement of protein l-isoaspartyl O-methyltransferase for transcriptional activation of trefoil factor 1 (TFF1) gene by estrogen receptor alpha. Biochem Biophys Res Commun 2012;420:223-9. https://doi.org/10.1016/j.bbrc.2012.02.072
  14. Lee JC, Kang SU, Jeon Y, Park JW, You JS, Ha SW, Bae N, Lubec G, Kwon SH, Lee JS, et al. Protein L-isoaspartyl methyltransferase regulates p53 activity. Nat Commun 2012;3:927. https://doi.org/10.1038/ncomms1933
  15. Rosner M, Hengstschlager M. Nucleocytoplasmic localization of p70 S6K1, but not of its isoforms p85 and p31, is regulated by TSC2/mTOR. Oncogene 2011;30:4509-22. https://doi.org/10.1038/onc.2011.165
  16. Rosner M, Schipany K, Hengstschlager M. p70 S6K1 nuclear localization depends on its mTOR-mediated phosphorylation at T389, but not on its kinase activity towards S6. Amino Acids 2012;42:2251-6. https://doi.org/10.1007/s00726-011-0965-4
  17. Wang L, Jin Q, Lee JE, Su IH, Ge K. Histone H3K27 methyltransferase Ezh2 represses Wnt genes to facilitate adipogenesis. Proc Natl Acad Sci USA 2010;107:7317-22. https://doi.org/10.1073/pnas.1000031107
  18. Cawthorn WP, Bree AJ, Yao Y, Du B, Hemati N, Martinez-Santibanez G, MacDougald OA. Wnt6, Wnt10a and Wnt10b inhibit adipogenesis and stimulate osteoblastogenesis through a ${\beta}$-catenin-dependent mechanism. Bone 2012;50:477-89. https://doi.org/10.1016/j.bone.2011.08.010
  19. Christodoulides C, Lagathu C, Sethi JK, Vidal-Puig A. Adipogenesis and WNT signalling. Trends Endocrinol Metab 2009;20:16-24. https://doi.org/10.1016/j.tem.2008.09.002
  20. Tang QQ, Lane MD. Adipogenesis: from stem cell to adipocyte. Annu Rev Biochem 2012;81:715-36. https://doi.org/10.1146/annurev-biochem-052110-115718
  21. Huang H, Song TJ, Li X, Hu L, He Q, Liu M, Lane MD, Tang QQ. BMP signaling pathway is required for commitment of C3H10T1/2 pluripotent stem cells to the adipocyte lineage. Proc Natl Acad Sci USA 2009;106:12670-5. https://doi.org/10.1073/pnas.0906266106
  22. Tachikawa E, Kudo K, Harada K, Kashimoto T, Miyate Y, Kakizaki A, Takahashi E. Effects of ginseng saponins on responses induced by various receptor stimuli. Eur J Pharmacol 1999;369:23-32. https://doi.org/10.1016/S0014-2999(99)00043-6
  23. Kim WY, Kim JM, Han SB, Lee SK, Kim ND, Park MK, Kim CK, Park JH. Steaming of ginseng at high temperature enhances biological activity. J Nat Prod 2000;63:1702-4. https://doi.org/10.1021/np990152b
  24. Wang CZ, Zhang B, Song WX, Wang A, Ni M, Luo X, Aung HH, Xie JT, Tong R, He TC, et al. Steamed American ginseng berry: ginsenoside analyses and anticancer activities. J Agric Food Chem 2006;54:9936-42. https://doi.org/10.1021/jf062467k
  25. Kim JK, Cui CH, Yoon MH, Kim SC, Im WT. Bioconversion of major ginsenosides Rg1 to minor ginsenoside F1 using novel recombinant ginsenoside hydrolyzing glycosidase cloned from Sanguibacter keddieii and enzyme characterization. J Biotechnol 2012;161:294-301. https://doi.org/10.1016/j.jbiotec.2012.06.021
  26. Lee SJ, Kim Y, Kim MG. Changes in the ginsenoside content during the fermentation process using microbial strains. J Ginseng Res 2015;39:392-7. https://doi.org/10.1016/j.jgr.2015.05.005
  27. Park SE, Na CS, Yoo SA, Seo SH, Son HS. Biotransformation of major ginsenosides in ginsenoside model culture by lactic acid bacteria. J Ginseng Res 2017;41:36-42. https://doi.org/10.1016/j.jgr.2015.12.008
  28. Ge K. Epigenetic regulation of adipogenesis by histone methylation. Biochim Biophys Acta 2012;1819:727-32. https://doi.org/10.1016/j.bbagrm.2011.12.008
  29. Shan X, Fu YS, Aziz F, Wang XQ, Yan Q, Liu JW. Ginsenoside Rg3 inhibits melanoma cell proliferation through down-regulation of histone deacetylase 3 (HDAC3) and increase of p53 acetylation. PLoS One 2014;9, e115401. https://doi.org/10.1371/journal.pone.0115401
  30. Shi Q, Li J, Feng Z, Zhao L, Luo L, You Z, Li D, Xia J, Zuo G, Chen D. Effect of ginsenoside Rh2 on the migratory ability of HepG2 liver carcinoma cells: recruiting histone deacetylase and inhibiting activator protein 1 transcription factors. Mol Med Rep 2014;10:1779-85. https://doi.org/10.3892/mmr.2014.2392
  31. Liu ZH, Li J, Xia J, Jiang R, Zuo GW, Li XP, Chen Y, Xiong W, Chen DL. Ginsenoside 20(s)-Rh2 as potent natural histone deacetylase inhibitors suppressing the growth of human leukemia cells. Chem Biol Interact 2015;242:227-34. https://doi.org/10.1016/j.cbi.2015.10.014
  32. Wan Q, Ma X, Zhang ZJ, Sun T, Xia F, Zhao G, Wu YM. Ginsenoside reduces cognitive impairment during chronic cerebral hypoperfusion through brainderived neurotrophic factor regulated by epigenetic modulation. Mol Neurobiol 2017;54:2889-900. https://doi.org/10.1007/s12035-016-9868-4
  33. Li Q, Li B, Dong C, Wang Y, Li Q. 20(S)-Ginsenoside Rh2 suppresses proliferation and migration of hepatocellular carcinoma cells by targeting EZH2 to regulate CDKN2A-2B gene cluster transcription. Eur J Pharmacol 2017;815:173-80. https://doi.org/10.1016/j.ejphar.2017.09.023
  34. Yang J, Yuan D, Xing T, Su H, Zhang S, Wen J, Bai Q, Dang D. Ginsenoside Rh2 inhibiting HCT116 colon cancer cell proliferation through blocking PDZbinding kinase/T-LAK cell-originated protein kinase. J Ginseng Res 2016;40:400-8. https://doi.org/10.1016/j.jgr.2016.03.007
  35. Go GY, Lee SJ, Jo A, Lee J, Seo DW, Kang JS, Kim SK, Kim SN, Kim YK, Bae GU. Ginsenoside Rg1 from Panax ginseng enhances myoblast differentiation and myotube growth. J Ginseng Res 2017;41:608-14. https://doi.org/10.1016/j.jgr.2017.05.006
  36. Gu Y, Zhou J, Wang Q, Fan W, Yin G. Ginsenoside Rg1 promotes osteogenic differentiation of rBMSCs and healing of rat tibial fractures through regulation of GR-dependent BMP-2/SMAD signaling. Sci Rep 2016;6:25282. https://doi.org/10.1038/srep25282
  37. Yin LH, Cheng WX, Qin ZS, Sun KM, Zhong M, Wang JK, Gao WY, Yu ZH. Effects of ginsenoside Rg-1 on the proliferation and osteogenic differentiation of human periodontal ligament stem cells. Chin J Integr Med 2015;21:676-81. https://doi.org/10.1007/s11655-014-1856-9
  38. Wang P, Wei X, Zhang F, Yang K, Qu C, Luo H, He L. Ginsenoside Rg1 of Panax ginseng stimulates the proliferation, odontogenic/osteogenic differentiation and gene expression profiles of human dental pulp stem cells. Phytomedicine 2014;21:177-83. https://doi.org/10.1016/j.phymed.2013.08.021
  39. Kim HM, Kim DH, Han HJ, Park CM, Ganipisetti SR, Valan Arasu M, Kim YO, Park CG, Kim BY, Soung NK. Ginsenoside Re promotes osteoblast differentiation in mouse osteoblast precursor MC3T3-E1 cells and a Zebrafish model. Molecules 2016;22.
  40. Kim DY, Jung MS, Park YG, Yuan HD, Quan HY, Chung SH. Ginsenoside Rh2(S) induces the differentiation and mineralization of osteoblastic MC3T3-E1 cells through activation of PKD and p38MAPK pathways.BMB Rep 2011;44:659-64. https://doi.org/10.5483/BMBRep.2011.44.10.659
  41. Li Z, Ji GE. Ginseng and obesity. J Ginseng Res 2018;42:1-8. https://doi.org/10.1016/j.jgr.2016.12.005
  42. Hwang JT, Kim SH, Lee MS, Kim SH, Yang HJ, Kim MJ, Kim HS, Ha J, Kim MS, Kwon DY. Anti-obesity effects of ginsenoside Rh2 are associated with the activation of AMPK signaling pathway in 3T3-L1 adipocyte. Biochem Biophys Res Commun 2007;364:1002-8. https://doi.org/10.1016/j.bbrc.2007.10.125
  43. Hwang JT, Lee MS, Kim HJ, Sung MJ, Kim HY, Kim MS, Kwon DY. Antiobesity effect of ginsenoside Rg3 involves the AMPK and PPAR-gamma signal pathways. Phytother Res 2009;23:262-6. https://doi.org/10.1002/ptr.2606
  44. Niu CS, Yeh CH, Yeh MF, Cheng JT. Increase of adipogenesis by ginsenoside (Rh2) in 3T3-L1 cell via an activation of glucocorticoid receptor. Horm Metab Res 2009;41:271-6. https://doi.org/10.1055/s-0028-1103277
  45. Lee YS, Cha BY, Yamaguchi K, Choi SS, Yonezawa T, Teruya T, Nagai K, Woo JT. Effects of Korean white ginseng extracts on obesity in high-fat diet-induced obese mice. Cytotechnology 2010;62:367-76. https://doi.org/10.1007/s10616-010-9288-7
  46. Lee H, Park D, Yoon M. Korean red ginseng (Panax ginseng) prevents obesity by inhibiting angiogenesis in high fat diet-induced obese C57BL/6J mice. Food Chem Toxicol 2013;53:402-8. https://doi.org/10.1016/j.fct.2012.11.052
  47. Yuan HD, Quan HY, Jung MS, Kim SJ, Huang B, Kim DY, Chung SH. Anti-diabetic effect of pectinase-processed ginseng radix (GINST) in high fat diet-fed ICR mice. J Ginseng Res 2011;35:308-14. https://doi.org/10.5142/jgr.2011.35.3.308
  48. Song YB, An YR, Kim SJ, Park HW, Jung JW, Kyung JS, Hwang SY, Kim YS. Lipid metabolic effect of Korean red ginseng extract in mice fed on a high-fat diet. J Sci Food Agric 2012;92:388-96. https://doi.org/10.1002/jsfa.4589
  49. Kim CM, Yi SJ, Cho IJ, Ku SK. Red-koji fermented red ginseng ameliorates high fat diet-induced metabolic disorders in mice. Nutrients 2013;5:4316-32. https://doi.org/10.3390/nu5114316
  50. Kwon DH, Bose S, Song MY, Lee MJ, Lim CY, Kwon BS, Kim HJ. Efficacy of Korean red ginseng by single nucleotide polymorphism in obese women: randomized, double-blind, placebo-controlled trial. J Ginseng Res 2012;36:176-89. https://doi.org/10.5142/jgr.2012.36.2.176
  51. Song MY, Kim BS, Kim H. Influence of Panax ginseng on obesity and gut microbiota in obese middle-aged Korean women. J Ginseng Res 2014;38:106-15. https://doi.org/10.1016/j.jgr.2013.12.004

Cited by

  1. Anti-Adipogenic Polyacetylene Glycosides from the Florets of Safflower (Carthamus tinctorius) vol.9, pp.1, 2020, https://doi.org/10.3390/biomedicines9010091
  2. Ginkwanghols A and B, osteogenic coumaric acid-aliphatic alcohol hybrids from the leaves of Ginkgo biloba vol.44, pp.5, 2020, https://doi.org/10.1007/s12272-021-01329-3
  3. Phytochemical Analysis of the Fruits of Sea Buckthorn (Hippophae rhamnoides): Identification of Organic Acid Derivatives vol.10, pp.5, 2020, https://doi.org/10.3390/plants10050860
  4. Withasomniferol D, a New Anti-Adipogenic Withanolide from the Roots of Ashwagandha (Withania somnifera) vol.14, pp.10, 2020, https://doi.org/10.3390/ph14101017
  5. Phytochemical Investigation of Bioactive Compounds from White Kidney Beans (Fruits of Phaseolus multiflorus var. Albus): Identification of Denatonium with Osteogenesis-Inducing Effect vol.10, pp.10, 2020, https://doi.org/10.3390/plants10102205
  6. Phytochemical Constituents Identified from the Aerial Parts of Lespedeza cuneata and Their Effects on Lipid Metabolism during Adipocyte Maturation vol.8, pp.11, 2021, https://doi.org/10.3390/separations8110203
  7. Combined Anti-Adipogenic Effects of Hispidulin and p-Synephrine on 3T3-L1 Adipocytes vol.11, pp.12, 2020, https://doi.org/10.3390/biom11121764
  8. Preventive Effect of Anemarrhenae rhizome and Phellodendri cortex on Danazol-Induced in Precocious Puberty in Female Rats and Network Pharmacological Analysis of Active Compounds vol.11, pp.1, 2020, https://doi.org/10.3390/plants11010023