DOI QR코드

DOI QR Code

AMP-activated protein kinase determines apoptotic sensitivity of cancer cells to ginsenoside-Rh2

  • Kim, Min-Jung (Department of Biochemistry and Molecular Biology, Medical Research Center and Biomedical Science Institute, School of Medicine, Kyung Hee University) ;
  • Yun, Hee (Department of Biochemistry and Molecular Biology, Medical Research Center and Biomedical Science Institute, School of Medicine, Kyung Hee University) ;
  • Kim, Dong-Hyun (Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University) ;
  • Kang, Insug (Department of Biochemistry and Molecular Biology, Medical Research Center and Biomedical Science Institute, School of Medicine, Kyung Hee University) ;
  • Choe, Wonchae (Department of Biochemistry and Molecular Biology, Medical Research Center and Biomedical Science Institute, School of Medicine, Kyung Hee University) ;
  • Kim, Sung-Soo (Department of Biochemistry and Molecular Biology, Medical Research Center and Biomedical Science Institute, School of Medicine, Kyung Hee University) ;
  • Ha, Joohun (Department of Biochemistry and Molecular Biology, Medical Research Center and Biomedical Science Institute, School of Medicine, Kyung Hee University)
  • Received : 2013.07.31
  • Accepted : 2013.09.13
  • Published : 2014.01.15

Abstract

Ginseng saponins exert various important pharmacological effects with regard to the control of many diseases, including cancer. In this study, the anticancer effect of ginsenosides on human cancer cells was investigated and compared. Among the tested compounds, ginsenoside-Rh2 displays the highest inhibitory effect on cell viability in HepG2 cells. Ginsenoside-Rh2, a ginseng saponin isolated from the root of Panax ginseng, has been suggested to have potential as an anticancer agent, but the underlying mechanisms remain elusive. In the present study, we have shown that cancer cells have differential sensitivity to ginsenoside-Rh2-induced apoptosis, raising questions regarding the specific mechanisms responsible for the discrepant sensitivity to ginsenoside-Rh2. In this study, we demonstrate that AMP-activated protein kinase (AMPK) is a survival factor under ginsenoside-Rh2 treatment in cancer cells. Cancer cells with acute responsiveness of AMPK display a relative resistance to ginsenoside-Rh2, but cotreatment with AMPK inhibitor resulted in a marked increase of ginsenoside-Rh2-induced apoptosis. We also observed that p38 MAPK (mitogen-activated protein kinase) acts as another survival factor under ginsenoside-Rh2 treatment, but there was no signaling crosstalk between AMPK and p38 MAPK, suggesting that combination with inhibitor of AMPK or p38 MAPK can augment the anticancer potential of ginsenoside Rh2.

Keywords

References

  1. Attele AS, Wu JA, Yuan CS. Ginseng pharmacology: multiple constituents and multiple actions. Biochem Pharmacol 1999;58:1685-93. https://doi.org/10.1016/S0006-2952(99)00212-9
  2. Xie JT, McHendale S, Yuan CS. Ginseng and diabetes. Am J Chin Med 2005;33: 397-404. https://doi.org/10.1142/S0192415X05003004
  3. Hwang JT, Kim SH, Lee MS, 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
  4. Cho WC, Chung WS, Lee SK, Leung AW, Cheng CH, Yue KK. Ginsenoside Re of Panax ginseng possesses significant antioxidant and antihyperlipidemic efficacies in streptozotocin-induced diabetic rats. Eur J Pharmacol 2006;550: 173-9. https://doi.org/10.1016/j.ejphar.2006.08.056
  5. Shang W, Yang Y, Zhou L, Jiang B, Jin H, Chen M. Ginsenoside Rb1 stimulates glucose uptake through insulin-like signaling pathway in 3T3-L1 adipocytes. J Endocrinol 2008;198:561-9. https://doi.org/10.1677/JOE-08-0104
  6. Song Z, Moser C, Wu H, Faber V, Kharazmi A, Hoiby N. Cytokine modulating effect of ginseng treatment in a mouse model of Pseudomonas aeruginosa lung infection. J Cyst Fibros 2003;2:112-9. https://doi.org/10.1016/S1569-1993(03)00065-1
  7. Lee EJ, Ko E, Lee J, Rho S, Ko S, Shin MK, Min BI, Hong MC, Kim SY, Bae H. Ginsenoside Rg1 enhances CD4(+) T-cell activities and modulates Th1/Th2 differentiation. Int Immunopharmacol 2004;4:235-44. https://doi.org/10.1016/j.intimp.2003.12.007
  8. Han SK, Song JY, Yun YS, Yi SY. Ginsan improved Th1 immune response inhibited by gamma radiation. Arch Pharm Res 2005;28:343-50. https://doi.org/10.1007/BF02977803
  9. Jia WW, Bu X, Philips D, Yan H, Liu G, Chen X, Bush JA, Li G. Rh2, a compound extracted from ginseng, hypersensitizes multidrug-resistant tumor cells to chemotherapy. Can J Physiol Pharmacol 2004;82:431-7. https://doi.org/10.1139/y04-049
  10. Choi S, Kim TW, Singh SV. Ginsenoside Rh2-mediated G1 phase cell cycle arrest in human breast cancer cells is caused by p15 Ink4B and p27 Kip1-dependent inhibition of cyclin-dependent kinases. Pharm Res 2009;26: 2280-8. https://doi.org/10.1007/s11095-009-9944-9
  11. Mochizuki M, Yoo YC, Matsuzawa K, Sato K, Saiki I, Tono-oka S, Samukawa K, Azuma I. Inhibitory effect of tumor metastasis in mice by saponins, ginsenoside- Rb2, 20(R)- and 20(S)-ginsenoside-Rg3, of red ginseng. Biol Pharm Bull 1995;18:1197-202. https://doi.org/10.1248/bpb.18.1197
  12. Xu TM, Xin Y, Cui MH, Jiang X, Gu LP. Inhibitory effect of ginsenoside Rg3 combined with cyclophosphamide on growth and angiogenesis of ovarian cancer. Chin Med J (Engl) 2007;120:584-8.
  13. Molnar J, Szabo D, Pusztai R, Mucsi I, Berek L, Ocsovszki I, Kawata E, Shoyama Y. Membrane associated antitumor effects of crocine-, ginsenosideand cannabinoid derivates. Anticancer Res 2000;20:861-7.
  14. Fei XF, Wang BX, Tashiro S, Li TJ, Ma JS, Ikejima T. Apoptotic effects of ginsenoside Rh2 on human malignant melanoma A375-S2 cells. Acta Pharmacol Sin 2002;23:315-22.
  15. Jin YH, Yoo KJ, Lee YH, Lee SK. Caspase 3-mediated cleavage of p21WAF1/CIP1 associated with the cyclin A-cyclin-dependent kinase 2 complex is a prerequisite for apoptosis in SK-HEP-1 cells. J Biol Chem 2000;275:30256-63. https://doi.org/10.1074/jbc.M001902200
  16. 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.
  17. 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
  18. Hardie DG. AMP-activated protein kinase as a drug target. Annu Rev Pharmacol Toxicol 2007;47:185-210. https://doi.org/10.1146/annurev.pharmtox.47.120505.105304
  19. Motoshima H, Goldstein BJ, Igata M, Araki E. AMPK and cell proliferationdAMPK as a therapeutic target for atherosclerosis and cancer. J Physiol 2006;574:63-71. https://doi.org/10.1113/jphysiol.2006.108324
  20. Luo Z, Saha AK, Xiang X, Ruderman NB. AMPK, the metabolic syndrome and cancer. Trends Pharmacol Sci 2005;26:69-76. https://doi.org/10.1016/j.tips.2004.12.011
  21. Kim HS, Hwang JT, Yun H, Chi SG, Lee SJ, Kang I, Yoon KS, Choe WJ, Kim SS, Ha J. Inhibition of AMP-activated protein kinase sensitizes cancer cells to cisplatin-induced apoptosis via hyper-induction of p53. J Biol Chem 2008;283: 3731-42. https://doi.org/10.1074/jbc.M704432200
  22. Kato K, Ogura T, Kishimoto A, Minegishi Y, Nakajima N, Miyazaki M, Esumi H. Critical roles of AMP-activated protein kinase in constitutive tolerance of cancer cells to nutrient deprivation and tumor formation. Oncogene 2002;21: 6082-90. https://doi.org/10.1038/sj.onc.1205737
  23. Yang JH, Han SJ, Ryu JH, Jang IS, Kim DH. Ginsenoside Rh2 ameliorates scopolamine-induced learning deficit in mice. Biol Pharm Bull 2009;32:1710-5. https://doi.org/10.1248/bpb.32.1710
  24. Oh JI, Chun KH, Joo SH, Oh YT, Lee SK. Caspase-3-dependent protein kinase C delta activity is required for the progression of ginsenoside-Rh2-induced apoptosis in SK-HEP-1 cells. Cancer Lett 2005;230:228-38. https://doi.org/10.1016/j.canlet.2004.12.043
  25. Cheng CC, Yang SM, Huang CY, Chen JC, Chang WM, Hsu SL. Molecular mechanisms of ginsenoside Rh2-mediated G1 growth arrest and apoptosis in human lung adenocarcinoma A549 cells. Cancer Chemother Pharmacol 2005;55:531-40. https://doi.org/10.1007/s00280-004-0919-6
  26. Kim YS, Jin SH. Ginsenoside Rh2 induces apoptosis via activation of caspase-1 and -3 and up-regulation of Bax in human neuroblastoma. Arch Pharm Res 2004;27:834-9. https://doi.org/10.1007/BF02980175
  27. Sandstrom ME, Zhang SJ, Bruton J, Silva JP, Reid MB, Westerblad H, Katz A. Role of reactive oxygen species in contraction-mediated glucose transport in mouse skeletal muscle. J Physiol 2006;575:251-62. https://doi.org/10.1113/jphysiol.2006.110601
  28. Choi SL, Kim SJ, Lee KT, Kim J, Mu J, Birnbaum MJ, Soo Kim S, Ha J. The regulation of AMP-activated protein kinase by H(2)O(2). Biochem Biophys Res Commun 2001;287:92-7. https://doi.org/10.1006/bbrc.2001.5544
  29. Ham YM, Choi JS, Chun KH, Joo SH, Lee SK. The c-Jun N-terminal kinase 1 activity is differentially regulated by specific mechanisms during apoptosis. J Biol Chem 2003;278:50330-7. https://doi.org/10.1074/jbc.M302997200
  30. Yim HW, Jong HS, Kim TY, Choi HH, Kim SG, Song SH, Kim J, Ko SG, Lee JW, Bang YJ. Cyclooxygenase-2 inhibits novel ginseng metabolite-mediated apoptosis. Cancer Res 2005;65:1952-60. https://doi.org/10.1158/0008-5472.CAN-04-1740
  31. Zhang HS, Wang SQ. Ginsenoside Rg1 inhibits tumor necrosis factor-alpha (TNF-alpha)-induced human arterial smooth muscle cells (HASMCs) proliferation. J Cell Biochem 2006;98:1471-81. https://doi.org/10.1002/jcb.20799
  32. Jung SN, Park IJ, Kim MJ, Kang I, Choe W, Kim SS, Ha J. Down-regulation of AMP-activated protein kinase sensitizes DU145 carcinoma to Fas-induced apoptosis via c-FLIP degradation. Exp Cell Res 2009;315:2433-41. https://doi.org/10.1016/j.yexcr.2009.05.018
  33. Fukushima S, Wanibuchi H, Li W. Inhibition by ginseng of colon carcinogenesis in rats. J Korean Med Sci 2001;16:S75-80. https://doi.org/10.3346/jkms.2001.16.S.S75
  34. Helms S. Cancer prevention and therapeutics: Panax ginseng. Altern Med Rev 2004;9:259-74.
  35. Bae EA, Han MJ, Choo MK, Park SY, Kim DH. Metabolism of 20(S)- and 20(R)- ginsenoside Rg3 by human intestinal bacteria and its relation to in vitro biological activities. Biol Pharm Bull 2002;25:58-63. https://doi.org/10.1248/bpb.25.58
  36. Park HM, Kim SJ, Kim JS, Kang HS. Reactive oxygen species mediated ginsenoside Rg3- and Rh2-induced apoptosis in hepatoma cells through mitochondrial signaling pathways. Food Chem Toxicol 2012;50:2736-41. https://doi.org/10.1016/j.fct.2012.05.027
  37. Kim Y, Jang M, Lim S, Won H, Yoon KS, Park JH, Kim HJ, Kim BH, Park WS, Ha J, et al. Role of cyclophilin B in tumorigenesis and cisplatin resistance in hepatocellular carcinoma in humans. Hepatology 2011;54:1661-78. https://doi.org/10.1002/hep.24539
  38. Wang W, Wang H, Rayburn ER, Zhao Y, Hill DL, Zhang R. 20(S)-25-methoxyldammarane- 3beta, 12beta, 20-triol, a novel natural product for prostate cancer therapy: activity in vitro and in vivo and mechanisms of action. Br J Cancer 2008;98:792-802. https://doi.org/10.1038/sj.bjc.6604227
  39. Foller M, Sopjani M, Koka S, Gu S, Mahmud H, Wang K, Floride E, Schleicher E, Schulz E, Munzel T, et al. Regulation of erythrocyte survival by AMP-activated protein kinase. FASEB J 2009;23:1072-80. https://doi.org/10.1096/fj.08-121772
  40. Hawley SA, Boudeau J, Reid JL, Mustard KJ, Udd L, Makela TP, Alessi DR, Hardie DG. Complexes between the LKB1 tumor suppressor, STRAD alpha/ beta and MO25 alpha/beta are upstream kinases in the AMP-activated protein kinase cascade. J Biol 2003;2:28. https://doi.org/10.1186/1475-4924-2-28
  41. Hong SP, Leiper FC, Woods A, Carling D, Carlson M. Activation of yeast Snf1 and mammalian AMP-activated protein kinase by upstream kinases. Proc Natl Acad Sci U S A 2003;100:8839-43. https://doi.org/10.1073/pnas.1533136100
  42. Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival. Cell 2003;115:577-90. https://doi.org/10.1016/S0092-8674(03)00929-2
  43. Corradetti MN, Inoki K, Bardeesy N, DePinho RA, Guan KL. Regulation of the TSC pathway by LKB1: evidence of a molecular link between tuberous sclerosis complex and PeutzeJeghers syndrome. Genes Dev 2004;18:1533-8. https://doi.org/10.1101/gad.1199104
  44. Liao BC, Hou RC, Wang JS, Jeng KC. Enhancement of the release of inflammatory mediators by substance P in rat basophilic leukemia RBL-2H3 cells. J Biomed Sci 2006;13:613-9. https://doi.org/10.1007/s11373-006-9099-1
  45. Jung SH, Woo MS, Kim SY, Kim WK, Hyun JW, Kim EJ, Kim DH, Kim HS. Ginseng saponin metabolite suppresses phorbol ester-induced matrix metalloproteinase- 9 expression through inhibition of activator protein-1 and mitogen-activated protein kinase signaling pathways in human astroglioma cells. Int J Cancer 2006;118:490-7. https://doi.org/10.1002/ijc.21356
  46. Lemieux K, Konrad D, Klip A, Marette A. The AMP-activated protein kinase activator AICAR does not induce GLUT4 translocation to transverse tubules but stimulates glucose uptake and p38 mitogen-activated protein kinases alpha and beta in skeletal muscle. FASEB J 2003;17:1658-65. https://doi.org/10.1096/fj.02-1125com
  47. Lee YM, Lee JO, Jung JH, Kim JH, Park SH, Park JM, Kim EK, Suh PG, Kim HS. Retinoic acid leads to cytoskeletal rearrangement through AMPK-Rac1 and stimulates glucose uptake through AMPK-p38 MAPK in skeletal muscle cells. J Biol Chem 2008;283:33969-74. https://doi.org/10.1074/jbc.M804469200
  48. Berasi SP, Huard C, Li D, Shih HH, Sun Y, Zhong W, Paulsen JE, Brown EL, Gimeno RE, Martinez RV. Inhibition of gluconeogenesis through transcriptional activation of EGR1 and DUSP4 by AMP-activated kinase. J Biol Chem 2006;281:27167-77. https://doi.org/10.1074/jbc.M602416200

Cited by

  1. A ROS-mediated lysosomal-mitochondrial pathway is induced by ginsenoside Rh2 in hepatoma HepG2 cells vol.6, pp.12, 2014, https://doi.org/10.1039/c5fo00518c
  2. Flavisolibacter ginsenosidimutans sp. nov., with ginsenoside-converting activity isolated from soil used for cultivating ginseng vol.65, pp.12, 2014, https://doi.org/10.1099/ijsem.0.000660
  3. Synergistic Anticancer Activities of Natural Substances in Human Hepatocellular Carcinoma vol.3, pp.4, 2014, https://doi.org/10.3390/diseases3040260
  4. Esterification of Ginsenoside Rh2 Enhanced Its Cellular Uptake and Antitumor Activity in Human HepG2 Cells vol.64, pp.1, 2014, https://doi.org/10.1021/acs.jafc.5b05450
  5. Anticancer Activities of Protopanaxadiol- and Protopanaxatriol-Type Ginsenosides and Their Metabolites vol.2016, pp.None, 2016, https://doi.org/10.1155/2016/5738694
  6. Inhibition of autophagy potentiates anticancer property of 20(S)-ginsenoside Rh2 by promoting mitochondria-dependent apoptosis in human acute lymphoblastic leukaemia cells vol.7, pp.19, 2014, https://doi.org/10.18632/oncotarget.8285
  7. The Octyl Ester of Ginsenoside Rh2 Induces Lysosomal Membrane Permeabilization via Bax Translocation vol.8, pp.5, 2014, https://doi.org/10.3390/nu8050244
  8. 20(S)-ginsenoside Rh2 inhibits the proliferation and induces the apoptosis of KG-1a cells through the Wnt/β-catenin signaling pathway vol.36, pp.1, 2014, https://doi.org/10.3892/or.2016.4774
  9. Regulation of AMP-activated protein kinase by natural and synthetic activators vol.6, pp.1, 2014, https://doi.org/10.1016/j.apsb.2015.06.002
  10. Anti-Proliferative and Pro-Apoptotic Activities of 4-Methyl-2,6-bis(1-phenylethyl)phenol in Cancer Cells vol.24, pp.4, 2014, https://doi.org/10.4062/biomolther.2015.166
  11. Mucilaginibacter pocheonensis sp. nov., with ginsenoside-converting activity, isolated from soil of a ginseng-cultivating field vol.66, pp.8, 2014, https://doi.org/10.1099/ijsem.0.001069
  12. Octyl Ester of Ginsenoside Rh2 Induces Apoptosis and G1 Cell Cycle Arrest in Human HepG2 Cells by Activating the Extrinsic Apoptotic Pathway and Modulating the Akt/p38 MAPK Signaling Pathway vol.64, pp.40, 2016, https://doi.org/10.1021/acs.jafc.6b03519
  13. Towards natural mimetics of metformin and rapamycin vol.9, pp.11, 2014, https://doi.org/10.18632/aging.101319
  14. Role of ginsenosides in reactive oxygen species-mediated anticancer therapy vol.9, pp.2, 2018, https://doi.org/10.18632/oncotarget.23407
  15. Gomisin A Suppresses Colorectal Lung Metastasis by Inducing AMPK/p38-Mediated Apoptosis and Decreasing Metastatic Abilities of Colorectal Cancer Cells vol.9, pp.None, 2018, https://doi.org/10.3389/fphar.2018.00986
  16. Ginsenosides synergize with mitomycin C in combating human non-small cell lung cancer by repressing Rad51-mediated DNA repair vol.39, pp.3, 2014, https://doi.org/10.1038/aps.2017.53
  17. Galla Rhois water extract inhibits lung metastasis by inducing AMPK-mediated apoptosis and suppressing metastatic properties of colorectal cancer cells vol.41, pp.1, 2019, https://doi.org/10.3892/or.2018.6812
  18. Boronate affinity mesoporous silica nanoparticle based selective enrichment for highly efficient analysis of ginsenosides vol.11, pp.44, 2019, https://doi.org/10.1039/c9ay01913h
  19. PEI-assisted boronate affinity magnetic nanoparticle-based SELEX for efficient in vitro evolution of saponin-binding aptamers vol.11, pp.15, 2014, https://doi.org/10.1039/d1ra00889g
  20. Ginsenoside Rh2 stimulates the production of mitochondrial reactive oxygen species and induces apoptosis of cervical cancer cells by inhibiting mitochondrial electron transfer chain complex vol.24, pp.6, 2014, https://doi.org/10.3892/mmr.2021.12513
  21. The promising therapeutic potentials of ginsenosides mediated through p38 MAPK signaling inhibition vol.7, pp.11, 2014, https://doi.org/10.1016/j.heliyon.2021.e08354