DOI QR코드

DOI QR Code

Protective effect of ginsenoside Rb1 against tacrolimus-induced apoptosis in renal proximal tubular LLC-PK1 cells

  • Lee, Dahae (School of Pharmacy, Sungkyunkwan University) ;
  • Lee, Dong-Soo (Institute of Human-Environment Interface Biology, Biomedical Research Institute, Department of Dermatology, Seoul National University College of Medicine) ;
  • Jung, Kiwon (College of Pharmacy, CHA University) ;
  • Hwang, Gwi Seo (College of Korean Medicine, Gachon University) ;
  • Lee, Hye Lim (College of Korean Medicine, Gachon University) ;
  • Yamabe, Noriko (College of Korean Medicine, Gachon University) ;
  • Lee, Hae-Jeong (Department of Food and Nutrition, Gachon University) ;
  • Eom, Dae-Woon (Department of Pathology, University of Ulsan College of Medicine) ;
  • Kim, Ki Hyun (School of Pharmacy, Sungkyunkwan University) ;
  • Kang, Ki Sung (College of Korean Medicine, Gachon University)
  • Received : 2016.09.06
  • Accepted : 2016.12.26
  • Published : 2018.01.15

Abstract

Background: The aim of the present study was to evaluate the potential protective effects of six ginsenosides (Rb1, Rb2, Rc, Rd, Rg1, and Rg3) isolated from Panax ginseng against tacrolimus (FK506)-induced apoptosis in renal proximal tubular LLC-PK1 cells. Methods: LLC-PK1 cells were treated with FK506 and ginsenosides, and cell viability was measured. Protein expressions of mitogen-activated protein kinases, caspase-3, and kidney injury molecule-1 (KIM-1) were evaluated by Western blotting analyses. The number of apoptotic cells was measured using an image-based cytometric assay. Results: Reduction in cell viability by $60{\mu}M$ FK506 was ameliorated significantly by cotreatment with ginsenosides Rg1 and Rb1. The phosphorylation of p38, extracellular signal-regulated kinases, and KIM-1, and cleavage of caspase-3, increased markedly in LLC-PK1 cells treated with FK506 and significantly decreased after cotreatment with ginsenoside Rb1. The number of apoptotic cells decreased by 6.0% after cotreatment with ginsenoside Rb1 ($10{\mu}M$ and $50{\mu}M$). Conclusion: The antiapoptotic effects of ginsenoside Rb1 on FK506-induced apoptosis were mediated by the inhibition of mitogen-activated protein kinases and caspase activation.

Keywords

References

  1. Wallemacq PE, Reding R. FK506 (tacrolimus), a novel immunosuppressant in organ transplantation: clinical, biomedical, and analytical aspects. Clin Chem 1993;39:2219-28.
  2. Cosner D, Zeng X, Zhang PL. Proximal tubular injury in medullary rays is an early sign of acute tacrolimus nephrotoxicity. J Transplant 2015;2015:142521.
  3. Dumont FJ. FK506, an immunosuppressant targeting calcineurin function. Curr Med Chem 2000;7:731-48. https://doi.org/10.2174/0929867003374723
  4. Tada H, Nakashima A, Awaya A, Fujisaki A, Inoue K, Kawamura K, Itoh K, Masuda H, Suzuki T. Effects of thymic hormone on reactive oxygen speciesscavengers and renal function in tacrolimus-induced nephrotoxicity. Life Sci 2002;70:1213-23. https://doi.org/10.1016/S0024-3205(01)01495-3
  5. Al-Harbi NO, Imam F, Al-Harbi MM, Iqbal M, Nadeem A, Al-Shahrah OA, Korashy HM, Al-Hosaini KA, Ahmed M, Bahashwar S. Treatment with aliskiren ameliorates tacrolimus-induced nephrotoxicity in rats. J Renin Angiotensin Aldosterone Syst 2015;16:1329-36. https://doi.org/10.1177/1470320314530178
  6. Nielsen FT, Leyssac PP, Kemp E, Starklint H, Dieperink H. Nephrotoxicity of FK-506 in the rat. Studies on glomerular and tubular function, and on the relationship between efficacy and toxicity. Nephrol Dial Transplant 1995;10:334-40.
  7. Servais H, Ortiz A, Devuyst O, Denamur S, Tulkens PM, Mingeot-Leclercq MP. Renal cell apoptosis induced by nephrotoxic drugs: cellular and molecular mechanismsand potential approaches tomodulation.Apoptosis 2008;13:11-32. https://doi.org/10.1007/s10495-007-0151-z
  8. Havasi A, Borkan SC. Apoptosis and acute kidney injury. Kidney Int 2011;80: 29-40. https://doi.org/10.1038/ki.2011.120
  9. Yang B, El Nahas AM, Thomas GL, Haylor JL, Watson PF, Wagner B, Johnson TS. Caspase-3 and apoptosis in experimental chronic renal scarring. Kidney Int 2001;60:1765-76. https://doi.org/10.1046/j.1523-1755.2001.00013.x
  10. Humphreys BD, Xu F, Sabbisetti V, Grgic I, Movahedi Naini S, Wang N, Chen G, Xiao S, Patel D, Henderson JM, et al. Chronic epithelial kidney injury molecule-1 expression causes murine kidney fibrosis. J Clin Invest 2013;123:4023-35. https://doi.org/10.1172/JCI45361
  11. Back JH, Ryu HH, Hong R, Han SA, Yoon YM, Kim DH, Hong SJ, Kim HL, Chung JH, Shin BC, et al. Antiproteinuric effects of green tea extract on tacrolimus-induced nephrotoxicity in mice. Transplant Proc 2015;47:2032-4. https://doi.org/10.1016/j.transproceed.2015.06.008
  12. Hisamura F, Kojima-Yuasa A, Huang X, Kennedy DO, Matsui-Yuasa I. Synergistic effect of green tea polyphenols on their protection against FK506- induced cytotoxicity in renal cells. Am J Chin Med 2008;36:615-24. https://doi.org/10.1142/S0192415X08006028
  13. Hisamura F, Kojima-Yuasa A, Kennedy DO, Matsui-Yuasa I. Protective effect of green tea extract and tea polyphenols against FK506-induced cytotoxicity in renal cells. Basic Clin Pharmacol Toxicol 2006;98:192-6. https://doi.org/10.1111/j.1742-7843.2006.pto_284.x
  14. Zhong Z, Connor HD, Li X, Mason RP, Forman DT, Lemasters JJ, Thurman RG. Reduction of ciclosporin and tacrolimus nephrotoxicity by plant polyphenols. J Pharm Pharmacol 2006;58:1533-43. https://doi.org/10.1211/jpp.58.11.0015
  15. Park EH, Kim YJ, Yamabe N, Park SH, Kim HK, Jang HJ, Kim JH, Cheon GJ, Ham J, Kang KS. Stereospecific anticancer effects of ginsenoside Rg3 epimers isolated from heat-processed American ginseng on human gastric cancer cell. J Ginseng Res 2014;38:22-7. https://doi.org/10.1016/j.jgr.2013.11.007
  16. Park JY, Choi P, Kim HK, Kang KS, Ham J. Increase in apoptotic effect of Panax ginseng by microwave processing in human prostate cancer cells: in vitro and in vivo studies. J Ginseng Res 2016;40:62-7. https://doi.org/10.1016/j.jgr.2015.04.007
  17. Jang HJ, Han IH, Kim YJ, Yamabe N, Lee D, Hwang GS, Oh M, Choi KC, Kim SN, Ham J, Eom DW, et al. Anticarcinogenic effects of products of heat-processed ginsenoside Re, a major constituent of ginseng berry, on human gastric cancer cells. J Agric Food Chem 2014;62:2830-6. https://doi.org/10.1021/jf5000776
  18. Kang KS, Kim HY, Yamabe N, Park JH, Yokozawa T. Preventive effect of 20(S)-ginsenoside Rg3 against lipopolysaccharide-induced hepatic and renal injury in rats. Free Radic Res 2007;41:1181-8. https://doi.org/10.1080/10715760701581740
  19. Kang KS, Yamabe N, Kim HY, Park JH, Yokozawa T. Therapeutic potential of 20(S)-ginsenoside Rg(3) against streptozotocin-induced diabetic renal damage in rats. Eur J Pharmacol 2008;591:266-72. https://doi.org/10.1016/j.ejphar.2008.06.077
  20. Kang KS, Kim HY, Baek SH, Yoo HH, Park JH, Yokozawa T. Study on the hydroxyl radical scavenging activity changes of ginseng and ginsenoside-Rb2 by heat processing. Biol Pharm Bull 2007;30:724-8. https://doi.org/10.1248/bpb.30.724
  21. Lee YJ, Kim HY, Kang KS, Lee JG, Yokozawa T, Park JH. The chemical and hydroxyl radical scavenging activity changes of ginsenoside-Rb1 by heat processing. Bioorg Med Chem Lett 2008;18:4515-20. https://doi.org/10.1016/j.bmcl.2008.07.056
  22. Lee W, Park SH, Lee S, Chung BC, Song MO, Song KI, Ham J, Kim SN, Kang KS. Increase in antioxidant effect of ginsenoside Re-alanine mixture by Maillard reaction. Food Chem 2012;135:2430-5. https://doi.org/10.1016/j.foodchem.2012.06.108
  23. Cong L, Chen W. Neuroprotective effect of ginsenoside Rd in spinal cord injury rats. Basic Clin Pharmacol Toxicol 2016;119:193-201. https://doi.org/10.1111/bcpt.12562
  24. Dong X, Zheng L, Lu S, Yang Y. Neuroprotective effects of pretreatment of ginsenoside Rb1 on severe cerebral ischemia-induced injuries in aged mice: involvement of anti-oxidant signaling. Geriatr Gerontol Int 2015. http://dx.doi.org/10.1111/ggi.12699.
  25. Zhou T, Zu G, Zhang X, Wang X, Li S, Gong X, Liang Z, Zhao J. Neuroprotective effects of ginsenoside Rg1 through the Wnt/beta-catenin signaling pathway in both in vivo and in vitro models of Parkinson's disease. Neuropharmacology 2016;101:480-9. https://doi.org/10.1016/j.neuropharm.2015.10.024
  26. Li YB, Wang Y, Tang JP, Chen D, Wang SL. Neuroprotective effects of ginsenoside Rg1-induced neural stem cell transplantation on hypoxic-ischemic encephalopathy. Neural Regen Res 2015;10:753-9. https://doi.org/10.4103/1673-5374.156971
  27. Shin HS, Yu M, Kim M, Choi HS, Kang DH. Renoprotective effect of red ginseng in gentamicin-induced acute kidney injury. Lab Invest 2014;94:1147-60. https://doi.org/10.1038/labinvest.2014.101
  28. Doh KC, Lim SW, Piao SG, Jin L, Heo SB, Zheng YF, Bae SK, Hwang GH, Min KI, Chung BH, et al. Ginseng treatment attenuates chronic cyclosporine nephropathy via reducing oxidative stress in an experimental mouse model. Am J Nephrol 2013;37:421-33. https://doi.org/10.1159/000349921
  29. Lim SW, Doh KC, Jin L, Jin J, Piao SG, Heo SB, Chung BH, Yang CW. Ginseng treatment attenuates autophagic cell death in chronic cyclosporine nephropathy. Nephrology 2014;19:490-9. https://doi.org/10.1111/nep.12273
  30. Park JY, Choi P, Kim T, Ko H, Kim HK, Kang KS, Ham J. Protective effects of processed finseng and its active ginsenosides on cisplatin-induced nephrotoxicity: in vitro and in vivo studies. J Agric Food Chem 2015;63:5964-9. https://doi.org/10.1021/acs.jafc.5b00782
  31. Fan Y, Xia J, Jia D, Zhang M, Zhang Y, Huang G, Wang Y. Mechanism of ginsenoside Rg1 renal protection in a mouse model of d-galactose-induced subacute damage. Pharm Biol 2016;54:1815-21. https://doi.org/10.3109/13880209.2015.1129543
  32. Wang L, Mao N, Tan RZ, Wang HL, Wen J, Liu YH, Furhad M, Fan JM. Ginsenoside Rg1 reduces aldosterone-induced autophagy via the AMPK/mTOR pathway in NRK-52E cells. Int J M Med 2015;36:518-26. https://doi.org/10.3892/ijmm.2015.2242
  33. Sun Q, Meng QT, Jiang Y, Liu HM, Lei SQ, Su WT, Duan WN, Wu Y, Xia ZY, Xia ZY. Protective effect of ginsenoside Rb1 against intestinal ischemia-reperfusion induced acute renal injury in mice. PloS One 2013;8:e80859. https://doi.org/10.1371/journal.pone.0080859
  34. Choi SJ, You HS, Chung SY. Tacrolimus-induced apoptotic signal transduction pathway. Transplant Proc 2008;40:2734-6. https://doi.org/10.1016/j.transproceed.2008.08.028
  35. Harris RC. COX-2 and the kidney. J Cardiovasc Pharmacol 2006;47:S37-42. https://doi.org/10.1097/00005344-200605001-00007
  36. Cagnol S, Chambard JC. ERK and cell death: mechanisms of ERK-induced cell deathdapoptosis, autophagy and senescence. FEBS J 2010;277:2-21. https://doi.org/10.1111/j.1742-4658.2009.07366.x
  37. Cassidy H, Radford R, Slyne J, O'Connell S, Slattery C, Ryan MP, McMorrow T. The role ofMAPKin drug-induced kidney injury. J Sig Transduct 2012;2012:463617.
  38. Lu Z, Xu S. ERK1/2 MAP kinases in cell survival and apoptosis. IUBMB life 2006;58:621-31. https://doi.org/10.1080/15216540600957438
  39. Deschesnes RG, Huot J, Valerie K, Landry J. Involvement of p38 in apoptosisassociated membrane blebbing and nuclear condensation. Mol Biol Cell 2001;12:1569-82. https://doi.org/10.1091/mbc.12.6.1569
  40. Cai B, Chang SH, Becker EB, Bonni A, Xia Z. p38 MAP kinase mediates apoptosis through phosphorylation of BimEL at Ser-65. J Biol Chem 2006;281:25215-22. https://doi.org/10.1074/jbc.M512627200
  41. Farley N, Pedraza-Alva G, Serrano-Gomez D, Nagaleekar V, Aronshtam A, Krahl T, Thornton T, Rincon M. p38 mitogen-activated protein kinase mediates the Fas-induced mitochondrial death pathway in CD8+ T cells. Mol Cell Biol 2006;26:2118-29. https://doi.org/10.1128/MCB.26.6.2118-2129.2006
  42. Zou W, Zeng J, Zhuo M, Xu W, Sun L, Wang J, Liu X. Involvement of caspase-3 and p38 mitogen-activated protein kinase in cobalt chloride-induced apoptosis in PC12 cells. J Neurosci Res 2002;67:837-43. https://doi.org/10.1002/jnr.10168

Cited by

  1. Ginsenoside Rb1, A Major Saponin from Panax ginseng , Exerts Protective Effects Against Acetaminophen-Induced Hepatotoxicity in Mice vol.47, pp.8, 2018, https://doi.org/10.1142/s0192415x19500927
  2. Protective Effects of Nargenicin A1 against Tacrolimus-Induced Oxidative Stress in Hirame Natural Embryo Cells vol.16, pp.6, 2018, https://doi.org/10.3390/ijerph16061044
  3. Betulinic Acid Suppresses Ovarian Cancer Cell Proliferation through Induction of Apoptosis vol.9, pp.7, 2018, https://doi.org/10.3390/biom9070257
  4. Neuroprotective Effects of Tetrahydrocurcumin against Glutamate-Induced Oxidative Stress in Hippocampal HT22 Cells vol.25, pp.1, 2020, https://doi.org/10.3390/molecules25010144
  5. Analysis and Identification of Active Compounds from Salviae miltiorrhizae Radix Toxic to HCT-116 Human Colon Cancer Cells vol.10, pp.4, 2018, https://doi.org/10.3390/app10041304
  6. Aviculin Isolated from Lespedeza cuneata Induce Apoptosis in Breast Cancer Cells through Mitochondria-Mediated Caspase Activation Pathway vol.25, pp.7, 2018, https://doi.org/10.3390/molecules25071708
  7. Hormesis and Ginseng: Ginseng Mixtures and Individual Constituents Commonly Display Hormesis Dose Responses, Especially for Neuroprotective Effects vol.25, pp.11, 2020, https://doi.org/10.3390/molecules25112719
  8. Preventive Effect of Muscone against Cisplatin Nephrotoxicity in LLC-PK1 Cells vol.10, pp.10, 2018, https://doi.org/10.3390/biom10101444
  9. Diketoacetonylphenalenone, Derived from Hawaiian Volcanic Soil-Associated Fungus Penicillium herquei FT729, Regulates T Cell Activation via Nuclear Factor-κB and Mitogen-Activated Protein Kina vol.25, pp.22, 2018, https://doi.org/10.3390/molecules25225374
  10. Astragaloside IV Alleviates Tacrolimus-Induced Chronic Nephrotoxicity via p62-Keap1-Nrf2 Pathway vol.11, pp.None, 2018, https://doi.org/10.3389/fphar.2020.610102
  11. Metabolite Profile of Cucurbitane-Type Triterpenoids of Bitter Melon (Fruit of Momordica charantia) and Their Inhibitory Activity against Protein Tyrosine Phosphatases Relevant to Insulin Resistance vol.69, pp.6, 2018, https://doi.org/10.1021/acs.jafc.0c06085
  12. In Vitro Studies to Assess the α-Glucosidase Inhibitory Activity and Insulin Secretion Effect of Isorhamnetin 3-O-Glucoside and Quercetin 3-O-Glucoside Isolated from Salicornia herbacea vol.9, pp.3, 2018, https://doi.org/10.3390/pr9030483
  13. Calcineurin Inhibitors Nephrotoxicity Prevention Strategies With Stress on Belatacept-Based Rescue Immunotherapy: A Review of the Current Evidence vol.53, pp.5, 2018, https://doi.org/10.1016/j.transproceed.2021.03.028
  14. Protective effects of selenium in tacrolimus-induced lung toxicity: potential role of heme oxygenase 1 vol.99, pp.10, 2018, https://doi.org/10.1139/cjpp-2020-0547
  15. The promising therapeutic potentials of ginsenosides mediated through p38 MAPK signaling inhibition vol.7, pp.11, 2018, https://doi.org/10.1016/j.heliyon.2021.e08354
  16. Identification of Renoprotective Phytosterols from Mulberry (Morus alba) Fruit against Cisplatin-Induced Cytotoxicity in LLC-PK1 Kidney Cells vol.10, pp.11, 2021, https://doi.org/10.3390/plants10112481
  17. Impact of the ACE2 activator xanthenone on tacrolimus nephrotoxicity: Modulation of uric acid/ERK/p38 MAPK and Nrf2/SOD3/GCLC signaling pathways vol.288, pp.None, 2022, https://doi.org/10.1016/j.lfs.2021.120154