DOI QR코드

DOI QR Code

Computational and experimental characterization of estrogenic activities of 20(S, R)-protopanaxadiol and 20(S, R)-protopanaxatriol

  • Zhang, Tiehua (College of Food Science and Engineering, Jilin University) ;
  • Zhong, Shuning (College of Food Science and Engineering, Jilin University) ;
  • Hou, Ligang (Institute of Agricultural Resources and Environment, Jilin Academy of Agricultural Sciences) ;
  • Wang, Yongjun (Institute of Agricultural Resources and Environment, Jilin Academy of Agricultural Sciences) ;
  • Xing, XiaoJia (Institute of Agricultural Resources and Environment, Jilin Academy of Agricultural Sciences) ;
  • Guan, Tianzhu (College of Food Science and Engineering, Jilin University) ;
  • Zhang, Jie (College of Food Science and Engineering, Jilin University) ;
  • Li, Tiezhu (College of Food Science and Engineering, Jilin University)
  • Received : 2017.12.31
  • Accepted : 2018.05.08
  • Published : 2020.09.15

Abstract

Background: As the main metabolites of ginsenosides, 20(S, R)-protopanaxadiol [PPD(S, R)] and 20(S, R)-protopanaxatriol [PPT(S, R)] are the structural basis response to a series of pharmacological effects of their parent components. Although the estrogenicity of several ginsenosides has been confirmed, however, the underlying mechanisms of their estrogenic effects are still largely unclear. In this work, PPD(S, R) and PPT(S, R) were assessed for their ability to bind and activate human estrogen receptor α (hERα) by a combination of in vitro and in silico analysis. Methods: The recombinant hERα ligand-binding domain (hERα-LBD) was expressed in E. coli strain. The direct binding interactions of ginsenosides with hERα-LBD and their ERα agonistic potency were investigated by fluorescence polarization and reporter gene assays, respectively. Then, molecular dynamics simulations were carried out to simulate the binding modes between ginsenosides and hERα-LBD to reveal the structural basis for their agonist activities toward receptor. Results: Fluorescence polarization assay revealed that PPD(S, R) and PPT(S, R) could bind to hERα-LBD with moderate affinities. In the dual luciferase reporter assay using transiently transfected MCF-7 cells, PPD(S, R) and PPT(S, R) acted as agonists of hERα. Molecular docking results showed that these ginsenosides adopted an agonist conformation in the flexible hydrophobic ligand-binding pocket. The stereostructure of C-20 hydroxyl group and the presence of C-6 hydroxyl group exerted significant influence on the hydrogen bond network and steric hindrance, respectively. Conclusion: This work may provide insight into the chemical and pharmacological screening of novel therapeutic agents from ginsenosides.

Keywords

References

  1. Heng Y, Zhang Q-S, Mu Z, Hu J-F, Yuan Y-H, Chen N-H. Ginsenoside Rg1 attenuates motor impairment and neuroinflammation in the MPTP-probenecidinduced parkinsonism mouse model by targeting ${\alpha}$-synuclein abnormalities in the substantia nigra. Toxicol Lett 2016;243:7-21. https://doi.org/10.1016/j.toxlet.2015.12.005
  2. Lee CH, Kim J-H. A review on the medicinal potentials of ginseng and ginsenosides on cardiovascular diseases. J Ginseng Res 2014;38:161-6. https://doi.org/10.1016/j.jgr.2014.03.001
  3. Vo HT, Cho JY, Choi Y-E, Choi Y-S, Jeong Y-H. Kinetic study for the optimization of ginsenoside Rg3 production by heat treatment of ginsenoside Rb1. J Ginseng Res 2015;39:304-13. https://doi.org/10.1016/j.jgr.2015.02.003
  4. Lee Y-M, Yoon H, Park H-M, Song BC, Yeum K-J. Implications of red Panax ginseng in oxidative stress associated chronic diseases. J Ginseng Res 2017;41:113-9. https://doi.org/10.1016/j.jgr.2016.03.003
  5. Khan Chowdhury E, Jeon J, Ok Rim S, Park Y-H, Kyu Lee S, Bae H. Composition, diversity and bioactivity of culturable bacterial endophytes in mountaincultivated ginseng in Korea. Sci Rep 2017;7:10098. https://doi.org/10.1038/s41598-017-10280-7
  6. Park SM, Jung EH, Kim JK, Jegal KH, Park CA, Cho IJ, Kim SC. 20S-Protopanaxadiol, an aglycosylated ginsenoside metabolite, induces hepatic stellate cell apoptosis through liver kinase B1-AMP-activated protein kinase activation. J Ginseng Res 2017;41:392-402. https://doi.org/10.1016/j.jgr.2017.01.012
  7. Chen RJY, Chung T-y, Li F-y, Lin N-h, Tzen JTC. Effect of sugar positions in ginsenosides and their inhibitory potency on Na+/K+-ATPase activity. Acta Pharmacol Sin 2009;30:61-9. https://doi.org/10.1038/aps.2008.6
  8. Zhao X-E, Lv T, Zhu S, Qu F, Chen G, He Y, Wei N, Li G, Xia L, Sun Z, et al. Dual ultrasonic-assisted dispersive liquid-liquid microextraction coupled with microwave-assisted derivatization for simultaneous determination of 20(S)-protopanaxadiol and 20(S)-protopanaxatriol by ultra high performance liquid chromatography-tandem mass spectrometry. J Chromatogr A 2016;1437:49-57. https://doi.org/10.1016/j.chroma.2016.02.017
  9. Furukawa T, Bai C-X, Kaihara A, Ozaki E, Kawano T, Nakaya Y, Awais M, Sato M, Umezawa Y, Kurokawa J. Ginsenoside Re, a main phytosterol of Panax ginseng, activates cardiac potassium channels via a nongenomic pathway of sex hormones. Mol Pharmacol 2006;70:1916-24. https://doi.org/10.1124/mol.106.028134
  10. Lee YJ, Chung E, Lee KY, Lee YH, Huh B, Lee SK. Ginsenoside-Rg1, one of the major active molecules from Panax ginseng, is a functional ligand of glucocorticoid receptor. Mol Cell Endocrinol 1997;133:135-40. https://doi.org/10.1016/S0303-7207(97)00160-3
  11. Leung KW, Cheung LWT, Pon YL, Wong RNS, Mak NK, Fan TPD, Au SCL, Tombran-Tink J, Wong AST. Ginsenoside Rb1 inhibits tube-like structure formation of endothelial cells by regulating pigment epithelium-derived factor through the oestrogen ${\beta}$ receptor. Br J Pharmacol 2007;152:207-15. https://doi.org/10.1038/sj.bjp.0707359
  12. Zhang Y, Yu L, Cai W, Fan S, Feng L, Ji G, Huang C. Protopanaxatriol, a novel $PPAR{\gamma}$ antagonist from Panax ginseng, alleviates steatosis in mice. Sci Rep 2014;4:7375. https://doi.org/10.1038/srep07375
  13. Rataj F, Moller FJ, Jahne M, Zierau O, Diel P, Vollmer G, Kretzschmar G. Regulation of uterine AHR battery gene expression by $17{\beta}$-Estradiol is predominantly mediated by estrogen receptor v. Arch Toxicol 2012;86:1603-12. https://doi.org/10.1007/s00204-012-0870-y
  14. Zhang J, Xing X, Sun Y, Li Z, Xue P, Wang T, Li T. Characterization of the binding between phthalate esters and mouse $PPAR{\alpha}$ for the development of a fluorescence polarization-based competitive binding assay. Anal Methods 2016;8:880-5. https://doi.org/10.1039/C5AY03053F
  15. Zhang J, Zhang T, Guan T, Yu H, Li T. In vitro and in silico assessment of the structure-dependent binding of bisphenol analogues to glucocorticoid receptor. Anal Bioanal Chem 2017;409:2239-46. https://doi.org/10.1007/s00216-016-0168-7
  16. Cho J, Park W, Lee S, Ahn W, Lee Y. Ginsenoside-Rb1 from Panax ginseng C.A. Meyer activates estrogen receptor-${\alpha}$ and -${\beta}$, independent of ligand binding. J Clin Endocrinol Metab 2004;89:3510-5. https://doi.org/10.1210/jc.2003-031823
  17. Zhang J, Zhang T, Guan T, Ruan P, Ren D, Dai W, Yu H, Li T. Spectroscopic and molecular modeling approaches to investigate the interaction of bisphenol A, bisphenol F and their diglycidyl ethers with $PPAR{\alpha}$. Chemosphere 2017;180:253-8. https://doi.org/10.1016/j.chemosphere.2017.04.034
  18. Nettles KW, Bruning JB, Gil G, O'Neill EE, Nowak J, Guo Y, Kim Y, DeSombre ER, Dilis R, Hanson RN, et al. Structural plasticity in the oestrogen receptor ligandbinding domain. EMBO Rep 2007;8:563-8. https://doi.org/10.1038/sj.embor.7400963
  19. Hong H, Kohli K, Trivedi A, Johnson DL, Stallcup MR. GRIP1, a novel mouse protein that serves as a transcriptional coactivator in yeast for the hormone binding domains of steroid receptors. Proc Natl Acad Sci USA 1996;93:4948-52. https://doi.org/10.1073/pnas.93.10.4948
  20. Watson CS, Bulayeva NN, Wozniak AL, Finnerty CC. Signaling from the membrane via membrane estrogen receptor-${\alpha}$: estrogens, xenoestrogens, and phytoestrogens. Steroids 2005;70:364-71. https://doi.org/10.1016/j.steroids.2005.03.002
  21. Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson JA. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors ${\alpha}$ and ${\beta}$. Endocrinology 1997;138:863-70. https://doi.org/10.1210/endo.138.3.4979
  22. Han J-Y, Kim H-J, Kwon Y-S, Choi Y-E. The Cyt P450 enzyme CYP716A47 catalyzes the formation of protopanaxadiol from dammarenediol-II during ginsenoside biosynthesis in Panax ginseng. Plant Cell Physiol 2011;52:2062-73. https://doi.org/10.1093/pcp/pcr150
  23. Kim D-H. Chemical diversity of Panax ginseng, Panax quinquifolium, and Panax notoginseng. J Ginseng Res 2012;36:1-15. https://doi.org/10.5142/jgr.2012.36.1.1
  24. Brzozowski AM, Pike AC, Dauter Z, Hubbard RE, Bonn T, Engstrom O, Ohman L, Greene GL, Gustafsson JA, Carlquist M. Molecular basis of agonism and antagonism in the oestrogen receptor. Nature 1997;389:753-8. https://doi.org/10.1038/39645
  25. Wang C-Z, Anderson S, Du W, He T-C, Yuan C-S. Red ginseng and cancer treatment. Chin J Nat Med 2016;14:7-16. https://doi.org/10.3724/SP.J.1009.2016.00007

Cited by

  1. Preparation, characterization and solubilization evaluation of two novel host-guest complexes based on two different functional groups of modified β-cyclodextrins and 20(S)-protopanaxatriol vol.1204, 2020, https://doi.org/10.1016/j.molstruc.2019.127494
  2. 20( S )-Ginsenoside Rg3 Inhibits Lung Cancer Cell Proliferation by Targeting EGFR-Mediated Ras/Raf/MEK/ERK Pathway vol.49, pp.3, 2020, https://doi.org/10.1142/s0192415x2150035x
  3. Enhanced cytotoxicity and antioxidant capacity of kaempferol complexed with α-lactalbumin vol.153, 2021, https://doi.org/10.1016/j.fct.2021.112265
  4. Kahweol Exerts Skin Moisturizing Activities by Upregulating STAT1 Activity vol.22, pp.16, 2020, https://doi.org/10.3390/ijms22168864
  5. Anti-Gastritis and Anti-Lung Injury Effects of Pine Tree Ethanol Extract Targeting Both NF-κB and AP-1 Pathways vol.26, pp.20, 2020, https://doi.org/10.3390/molecules26206275