DOI QR코드

DOI QR Code

Heat-processed Panax ginseng and diabetic renal damage: active components and action mechanism

  • Kang, Ki Sung (Natural Medicine Center, Korea Institute of Science and Technology) ;
  • Ham, Jungyeob (Natural Medicine Center, Korea Institute of Science and Technology) ;
  • Kim, Young-Joo (Natural Medicine Center, Korea Institute of Science and Technology) ;
  • Park, Jeong Hill (Research Institute of Pharmaceutical Sciences and College of Pharmacy, Seoul National University) ;
  • Cho, Eun-Ju (Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University) ;
  • Yamabe, Noriko (Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University)
  • Received : 2013.02.26
  • Accepted : 2013.07.26
  • Published : 2013.10.15

Abstract

Diabetic nephropathy is one of the serious complications in patients with either type 1 or 2 diabetes mellitus but current treatments remain unsatisfactory. Results of clinical research studies demonstrate that Panax ginseng can help adjust blood pressure and reduce blood sugar and may be advantageous in the treatment of tuberculosis and kidney damage in people with diabetes. The heat-processing method to strengthen the efficacy of P. ginseng has been well-defined based on a long history of ethnopharmacological evidence. The protective effects of P. ginseng on pathological conditions and renal damage associated with diabetic nephropathy in the animal models were markedly improved by heat-processing. The concentrations of less-polar ginsenosides (20(S)-Rg3, 20(R)-Rg3, Rg5, and Rk1) and maltol in P. ginseng were significantly increased in a heat-processing temperature-dependent manner. Based on researches in animal models of diabetes, ginsenoside 20(S)-Rg3 and maltol were evaluated to have therapeutic potential against diabetic renal damage. These effects were achieved through the inhibition of inflammatory pathway activated by oxidative stress and advanced glycation endproducts. These findings indicate that ginsenoside 20(S)-Rg3 and maltol are important bioactive constituents of heat-processed ginseng in the control of pathological conditions associated with diabetic nephropathy.

Keywords

References

  1. Li JM, Shah AM. ROS generation by nonphagocytic NADPH oxidase: potential relevance in diabetic nephropathy. J Am Soc Nephrol 2003;14(8 Suppl 3):S221-S226. https://doi.org/10.1097/01.ASN.0000077406.67663.E7
  2. Alsaad KO, Herzenberg AM. Distinguishing diabetic nephropathy from other causes of glomerulosclerosis: an update. J Clin Pathol 2007;60:18-26. https://doi.org/10.1136/jcp.2005.035592
  3. Sarafidis PA, Ruilope LM. Insulin resistance, hyperinsulinemia, and renal injury: mechanisms and implications. Am J Nephrol 2006;26:232-244. https://doi.org/10.1159/000093632
  4. Njoroge FG, Monnier VM. The chemistry of the Maillard reaction under physiological conditions: a review. Prog Clin Biol Res 1989;304:85-107.
  5. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med 1993;329:977-986. https://doi.org/10.1056/NEJM199309303291401
  6. Kasuga M. Insulin resistance and pancreatic beta cell failure. J Clin Invest 2006;116:1756-1760. https://doi.org/10.1172/JCI29189
  7. Parving HH. Diabetic nephropathy: prevention and treatment. Kidney Int 2001;60:2041-2055. https://doi.org/10.1046/j.1523-1755.2001.00020.x
  8. Baynes JW. Role of oxidative stress in development of complications in diabetes. Diabetes 1991;40:405-412.
  9. Wendt T, Tanji N, Guo J, Hudson BI, Bierhaus A, Ramasamy R, Arnold B, Nawroth PP, Yan SF, D’Agati V et al. Glucose, glycation, and RAGE: implications for amplification of cellular dysfunction in diabetic nephropathy. J Am Soc Nephrol 2003;14:1383-1395. https://doi.org/10.1097/01.ASN.0000065100.17349.CA
  10. Williams ME. New therapies for advanced glycation end product nephrotoxicity: current challenges. Am J Kidney Dis 2003;41(3 Suppl 1):S42-S47. https://doi.org/10.1053/ajkd.2003.50083
  11. Schrijvers BF, De Vriese AS, Flyvbjerg A. From hyperglycemia to diabetic kidney disease: the role of metabolic, hemodynamic, intracellular factors and growth factors/cytokines. Endocr Rev 2004;25:971-1010. https://doi.org/10.1210/er.2003-0018
  12. Vlassara H, Striker LJ, Teichberg S, Fuh H, Li YM, Steffes M. Advanced glycation end products induce glomerular sclerosis and albuminuria in normal rats. Proc Natl Acad Sci U S A 1994;91:11704-11708. https://doi.org/10.1073/pnas.91.24.11704
  13. Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000;404:787-790. https://doi.org/10.1038/35008121
  14. Cooper ME. Interaction of metabolic and haemodynamic factors in mediating experimental diabetic nephropathy. Diabetologia 2001;44:1957-1972. https://doi.org/10.1007/s001250100000
  15. Tuttle KR, Johnson EC, Cooney SK, Anderberg RJ, Johnson EK, Clifton GD, Meek RL. Amino acids injure mesangial cells by advanced glycation end products, oxidative stress, and protein kinase C. Kidney Int 2005;67:953-968. https://doi.org/10.1111/j.1523-1755.2005.00159.x
  16. Rahbar S, Figarola JL. Novel inhibitors of advanced glycation endproducts. Arch Biochem Biophys 2003;419:63-79. https://doi.org/10.1016/j.abb.2003.08.009
  17. Newsholme P, Haber EP, Hirabara SM, Rebelato EL, Procopio J, Morgan D, Oliveira-Emilio HC, Carpinelli AR, Curi R. Diabetes associated cell stress and dysfunction: role of mitochondrial and non-mitochondrial ROS production and activity. J Physiol 2007;583(Pt 1):9-24. https://doi.org/10.1113/jphysiol.2007.135871
  18. Ceylan-Isik AF, Fliethman RM, Wold LE, Ren J. Herbal and traditional Chinese medicine for the treatment of cardiovascular complications in diabetes mellitus. Curr Diabetes Rev 2008;4:320-328. https://doi.org/10.2174/157339908786241142
  19. Li WL, Zheng HC, Bukuru J, De Kimpe N. Natural medicines used in the traditional Chinese medical system for therapy of diabetes mellitus. J Ethnopharmacol 2004;92:1-21. https://doi.org/10.1016/j.jep.2003.12.031
  20. Yin J, Zhang H, Ye J. Traditional chinese medicine in treatment of metabolic syndrome. Endocr Metab Immune Disord Drug Targets 2008;8:99-111. https://doi.org/10.2174/187153008784534330
  21. Park JH, Kim JM, Han SB, Kim NY, Surh YJ, Lee SK, Kim ND, Park MK. A new processed ginseng with fortified activity. In: Hur H, Choi KJ, Kim YC, eds. Advances in ginseng research. Seoul: Korean Society of Ginseng, 1998. p. 146-159.
  22. Yun TK. Brief introduction of Panax ginseng C.A. Meyer. J Korean Med Sci 2001;16 Suppl:S3-S5. https://doi.org/10.3346/jkms.2001.16.S.S3
  23. Yokozawa T, Kang KS, Yamabe N, Kim HY. Therapeutic potential of heat-processed Panax ginseng with respect to oxidative tissue damage. Drug Discov Ther 2007;1:30-44.
  24. Sen S, Chen S, Feng B, Wu Y, Lui E, Chakrabarti S. Preventive effects of North American ginseng (Panax quinquefolium) on diabetic nephropathy. Phytomedicine 2012;19:494-505. https://doi.org/10.1016/j.phymed.2012.01.001
  25. Xie XS, Liu HC, Wang FP, Zhang CL, Zuo C, Deng Y, Fan JM. Ginsenoside Rg1 modulation on thrombospondin-1 and vascular endothelial growth factor expression in early renal fibrogenesis in unilateral obstruction. Phytother Res 2010;24:1581-1587. https://doi.org/10.1002/ptr.3190
  26. Chen H, Yin J, Deng Y, Yang M, Xu L, Teng F, Li D, Cheng Y, Liu S, Wang D et al. The protective effects of ginsenoside Rg1 against hypertension target-organ damage in spontaneously hypertensive rats. BMC Complement Altern Med 2012;12:53. https://doi.org/10.1186/1472-6882-12-53
  27. 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;550:173-179. https://doi.org/10.1016/j.ejphar.2006.08.056
  28. Zhang HA, Wang M, Zhou J, Yao QY, Ma JM, Jiang CL. Protective effect of ginsenoside against acute renal failure and expression of tyrosine hydroxylase in the locus coeruleus. Physiol Res 2010;59:61-70.
  29. Sun Q, Meng QT, Jiang Y, Xia ZY. Ginsenoside Rb1 attenuates intestinal ischemia reperfusion induced renal injury by activating Nrf2/ARE pathway. Molecules 2012;17:7195-7205. https://doi.org/10.3390/molecules17067195
  30. Xie XS, Liu HC, Yang M, Zuo C, Deng Y, Fan JM. Ginsenoside Rb1, a panoxadiol saponin against oxidative damage and renal interstitial fibrosis in rats with unilateral ureteral obstruction. Chin J Integr Med 2009;15:133-140. https://doi.org/10.1007/s11655-009-0133-9
  31. Yokozawa T, Liu ZW, Dong E. A study of ginsenoside- Rd in a renal ischemia-reperfusion model. Nephron 1998;78:201-206. https://doi.org/10.1159/000044911
  32. Yokozawa T, Owada S. Effect of ginsenoside-Rd in cephaloridine-induced renal disorder. Nephron 1999;81:200-207. https://doi.org/10.1159/000045277
  33. Yokozawa T, Liu ZW. The role of ginsenoside-Rd in cisplatin-induced acute renal failure. Ren Fail 2000;22:115-127. https://doi.org/10.1081/JDI-100100858
  34. Peng SL, Guo ZA. Effect of total saponins of Panax notoginseng on urinary albumin in patients with chronic renal failure. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue 2010;22:744-746.
  35. Vuksan V, Sung MK, Sievenpiper JL, Stavro PM, Jenkins AL, Di Buono M, Lee KS, Leiter LA, Nam KY, Arnason JT et al. Korean red ginseng (Panax ginseng) improves glucose and insulin regulation in well-controlled, type 2 diabetes: results of a randomized, double-blind, placebo- controlled study of efficacy and safety. Nutr Metab Cardiovasc Dis 2008;18:46-56. https://doi.org/10.1016/j.numecd.2006.04.003
  36. Kasai R, Besso H, Tanaka O, Saruwatari YI, Fuwa T. Saponins of red ginseng. Chem Pharm Bull (Tokyo) 1983;31:2120-2125. https://doi.org/10.1248/cpb.31.2120
  37. Matsuura H, Hirao Y, Yoshida S, Kunihiro K, Fuwa T, Kasai R, Tanaka O. Study of red ginseng: new glucosides and a note on the occurrence of maltol. Chem Pharm Bull (Tokyo) 1984;32:4674-4677. https://doi.org/10.1248/cpb.32.4674
  38. 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;63:1702-1704. https://doi.org/10.1021/np990152b
  39. Kim HY, Kang KS, Yamabe N, Nagai R, Yokozawa T. Protective effect of heat-processed American ginseng against diabetic renal damage in rats. J Agric Food Chem 2007;55:8491-8497. https://doi.org/10.1021/jf071770y
  40. Kim HY, Kang KS, Yamabe N, Yokozawa T. Comparison of the effects of Korean ginseng and heat-processed Korean ginseng on diabetic oxidative stress. Am J Chin Med 2008;36:989-1004. https://doi.org/10.1142/S0192415X08006417
  41. Kang KS, Kim HY, Pyo JS, Yokozawa T. Increase in the free radical scavenging activity of ginseng by heat- processing. Biol Pharm Bull 2006;29:750-754. https://doi.org/10.1248/bpb.29.750
  42. Kang KS, Yokozawa T, Kim HY, Park JH. Study on the nitric oxide scavenging effects of ginseng and its compounds. J Agric Food Chem 2006;54:2558-2562. https://doi.org/10.1021/jf0529520
  43. Kang KS, Kim HY, Yamabe N, Yokozawa T. Stereospecificity in hydroxyl radical scavenging activities of four ginsenosides produced by heat processing. Bioorg Med Chem Lett 2006;16:5028-5031. https://doi.org/10.1016/j.bmcl.2006.07.071
  44. Kang KS, Kim HY, Yamabe N, Nagai R, Yokozawa T. Protective effect of sun ginseng against diabetic renal damage. Biol Pharm Bull 2006;29:1678-1684. https://doi.org/10.1248/bpb.29.1678
  45. 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;30:724-728. https://doi.org/10.1248/bpb.30.724
  46. Kang KS, Yokozawa T, Yamabe N, Kim HY, Park JH. ESR study on the structure and hydroxyl radical-scavenging activity relationships of ginsenosides isolated from Panax ginseng C A Meyer. Biol Pharm Bull 2007;30:917-921. https://doi.org/10.1248/bpb.30.917
  47. Kang KS, Lee YJ, Park JH, Yokozawa T. The effects of glycine and L-arginine on heat stability of ginsenoside Rb1. Biol Pharm Bull 2007;30:1975-1978. https://doi.org/10.1248/bpb.30.1975
  48. 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;41:1181-1188. https://doi.org/10.1080/10715760701581740
  49. 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;591:266-272. https://doi.org/10.1016/j.ejphar.2008.06.077
  50. Kang KS, Tanaka T, Cho EJ, Yokozawa T. Evaluation of the peroxynitrite scavenging activity of heat-processed ginseng. J Med Food 2009;12:124-130. https://doi.org/10.1089/jmf.2007.0646
  51. Yamabe N, Song KI, Lee W, Han IH, Lee JH, Ham J, Kim SN, Park JH, Kang KS. Chemical and free radical-scavenging activity changes of ginsenoside Re by Maillard reaction and its possible use as a renoprotective agent. J Ginseng Res 2012;36:256-262. https://doi.org/10.5142/jgr.2012.36.2.256
  52. 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;18:4515-4520. https://doi.org/10.1016/j.bmcl.2008.07.056
  53. Yamabe N, Lee JG, Lee YJ, Park CH, Kim HY, Park JH, Yokozawa T, Kang KS. The chemical and 1,1-diphenyl-2-picrylhydrazyl radical scavenging activity changes of ginsenosides Rb1 and Rg1 by Maillard reaction. J Ginseng Res 2011;35:60-68. https://doi.org/10.5142/jgr.2011.35.1.060
  54. Deng A, Valdivielso JM, Munger KA, Blantz RC, Thomson SC. Vasodilatory N-methyl-D-aspartate receptors are constitutively expressed in rat kidney. J Am Soc Nephrol 2002;13:1381-1384. https://doi.org/10.1097/01.ASN.0000013293.11876.4E
  55. Leung JC, Marphis T, Craver RD, Silverstein DM. Altered NMDA receptor expression in renal toxicity: protection with a receptor antagonist. Kidney Int 2004;66:167-176. https://doi.org/10.1111/j.1523-1755.2004.00718.x
  56. Kawano K, Hirashima T, Mori S, Saitoh Y, Kurosumi M, Natori T. Spontaneous long-term hyperglycemic rat with diabetic complications. Otsuka Long-Evans Tokushima Fatty (OLETF) strain. Diabetes 1992;41:1422-1428. https://doi.org/10.2337/diab.41.11.1422
  57. Seo YS, Kim JH, Jo NY, Choi KM, Baik SH, Park JJ, Kim JS, Byun KS, Bak YT, Lee CH et al. PPAR agonists treatment is effective in a nonalcoholic fatty liver disease animal model by modulating fatty-acid metabolic enzymes. J Gastroenterol Hepatol 2008;23:102-109.
  58. Horie K, Miyata T, Maeda K, Miyata S, Sugiyama S, Sakai H, van Ypersole de Strihou C, Monnier VM, Witztum JL, Kurokawa K. Immunohistochemical colocalization of glycoxidation products and lipid peroxidation products in diabetic renal glomerular lesions. Implication for glycoxidative stress in the pathogenesis of diabetic nephropathy. J Clin Invest 1997;100:2995-3004. https://doi.org/10.1172/JCI119853
  59. Nagai R, Unno Y, Hayashi MC, Masuda S, Hayase F, Kinae N, Horiuchi S. Peroxynitrite induces formation of N(epsilon)-(carboxymethyl) lysine by the cleavage of Amadori product and generation of glucosone and glyoxal from glucose: novel pathways for protein modification by peroxynitrite. Diabetes 2002;51:2833-2839. https://doi.org/10.2337/diabetes.51.9.2833
  60. Yan SD, Schmidt AM, Anderson GM, Zhang J, Brett J, Zou YS, Pinsky D, Stern D. Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J Biol Chem 1994;269:9889-9897.
  61. Ahmed N. Advanced glycation endproducts: role in pathology of diabetic complications. Diabetes Res Clin Pract 2005;67:3-21. https://doi.org/10.1016/j.diabres.2004.09.004
  62. Kang KS, Yamabe N, Kim HY, Park JH, Yokozawa T. Effects of heat-processed ginseng and its active component ginsenoside 20(S)-Rg3 on the progression of renal damage and dysfunction in type 2 diabetic Otsuka Long-Evans Tokushima Fatty rats. Biol Pharm Bull 2010;33:1077-1081. https://doi.org/10.1248/bpb.33.1077
  63. Wijewickreme AN, Krejpcio Z, Kitts DD. Hydroxyl scavenging activity of glucose, fructose, and ribose-lysine model Maillard products. J Food Sci 1999;64:457-461. https://doi.org/10.1111/j.1365-2621.1999.tb15062.x
  64. Bekedam EK, Schols HA, Cammerer B, Kroh LW, van Boekel MA, Smit G. Electron spin resonance (ESR) studies on the formation of roasting-induced antioxidative structures in coffee brews at different degrees of roast. J Agric Food Chem 2008;56:4597-4604. https://doi.org/10.1021/jf8004004
  65. Chen XM, Kitts DD. Antioxidant activity and chemical properties of crude and fractionated Maillard reaction products derived from four sugar-amino acid Maillard reaction model systems. Ann N Y Acad Sci 2008;1126:220-224. https://doi.org/10.1196/annals.1433.028
  66. Li X, Zheng Y, Liu M, Zhang L. A study on maillard reaction and its products during processing of red ginseng. Zhongguo Zhong Yao Za Zhi 1999;24:274-278.
  67. Suzuki Y, Choi KJ, Uchida K, Ko SR, Sohn HJ, Park JD. Arginyl-fructosyl-glucose and arginyl-fructose, compounds related to browning reaction in the model system of steaming and heat-drying processes for the preparation of red ginseng. J Ginseng Res 2004;28:143-148. https://doi.org/10.5142/JGR.2004.28.3.143
  68. Johnson RR, Alford ED, Kinzer GW. Formation of sucrose pyrolysis products. J Agric Food Chem 1969;17:22-24. https://doi.org/10.1021/jf60161a013
  69. Ito H. The formation of maltol and isomaltol through degradation of sucrose. Agric Biol Chem 1977;41:1307-1308. https://doi.org/10.1271/bbb1961.41.1307
  70. Bjeldanes LF, Chew H. Mutagenicity of 1,2-dicarbonyl compounds: maltol, kojic acid, diacetyl and related substances. Mutat Res 1979;67:367-371. https://doi.org/10.1016/0165-1218(79)90034-X
  71. Yasumoto E, Nakano K, Nakayachi T, Morshed SR, Hashimoto K, Kikuchi H, Nishikawa H, Kawase M, Sakagami H. Cytotoxic activity of deferiprone, maltol and related hydroxyketones against human tumor cell lines. Anticancer Res 2004;24:755-762.
  72. Harvey RS, Reffitt DM, Doig LA, Meenan J, Ellis RD, Thompson RP, Powell JJ. Ferric trimaltol corrects iron deficiency anaemia in patients intolerant of iron. Aliment Pharmacol Ther 1998;12:845-848. https://doi.org/10.1046/j.1365-2036.1998.00380.x
  73. Verma S, Cam MC, McNeill JH. Nutritional factors that can favorably influence the glucose/insulin system: vanadium. J Am Coll Nutr 1998;17:11-18. https://doi.org/10.1080/07315724.1998.10718730
  74. Reffitt DM, Burden TJ, Seed PT, Wood J, Thompson RP, Powell JJ. Assessment of iron absorption from ferric trimaltol. Ann Clin Biochem 2000;37(Pt 4):457-466. https://doi.org/10.1258/0004563001899645
  75. Jang DS, Kim JM, Lee YM, Kim YS, Kim JH, Kim JS. Puerariafuran, a new inhibitor of advanced glycation end products (AGEs) isolated from the roots of Pueraria lobata. Chem Pharm Bull (Tokyo) 2006;54:1315-1317. https://doi.org/10.1248/cpb.54.1315
  76. Price DL, Rhett PM, Thorpe SR, Baynes JW. Chelating activity of advanced glycation end-product inhibitors. J Biol Chem 2001;276:48967-48972. https://doi.org/10.1074/jbc.M108196200
  77. Huang D, Ou B, Prior RL. The chemistry behind antioxidant capacity assays. J Agric Food Chem 2005;53:1841-1856. https://doi.org/10.1021/jf030723c
  78. Kang KS, Yamabe N, Kim HY, Yokozawa T. Role of maltol in advanced glycation end products and free radicals: in-vitro and in-vivo studies. J Pharm Pharmacol 2008;60:445-452. https://doi.org/10.1211/jpp.60.4.0006
  79. Nakai S, Shinzato T, Nagura Y, Masakane I, Kitaoka T, Shinoda T, Yamazaki C, Sakai R, Ohmori H, Morita O et al. An overview of regular dialysis treatment in Japan (as of 31 December 2001). Ther Apher Dial 2004;8:3-32. https://doi.org/10.1111/j.1526-0968.2004.00109.x
  80. Yamabe N, Yokozawa T. Activity of the Chinese prescription Hachimi-jio-gan against renal damage in the Otsuka Long-Evans Tokushima fatty rat: a model of human type 2 diabetes mellitus. J Pharm Pharmacol 2006;58:535-545. https://doi.org/10.1211/jpp.58.4.0014
  81. Levin A. Identification of patients and risk factors in chronic kidney disease: evaluating risk factors and therapeutic strategies. Nephrol Dial Transplant 2001;16 Suppl 7:57-60. https://doi.org/10.1093/ndt/16.suppl_7.57

Cited by

  1. Ginseng treatment attenuates autophagic cell death in chronic cyclosporine nephropathy vol.19, pp.8, 2014, https://doi.org/10.1111/nep.12273
  2. Extract and Its Active Compound Eupatilin against Cisplatin-Induced Renal Damage vol.2015, pp.1741-4288, 2015, https://doi.org/10.1155/2015/483980
  3. Ethanol-Heated Processed Scutellariae Radix Improve Inflammatory Response through an Inhibitory Effect against Oxidative Stress in Mice with the Lipopolysaccharide-induced Intestine Injury of Mice vol.30, pp.4, 2015, https://doi.org/10.6116/kjh.2015.30.4.81.
  4. Caspase-mediated Apoptotic Effects of Diol-type Ginseng Sapogenins on Human Hepatoma Cell Lines vol.36, pp.1, 2015, https://doi.org/10.1002/bkcs.10074
  5. Comparison of the Wound-Healing Effects of Ginsenosides, their Metabolites, and Aglycones vol.37, pp.1, 2015, https://doi.org/10.1002/bkcs.10623
  6. Maltol, a Food Flavoring Agent, Attenuates Acute Alcohol-Induced Oxidative Damage in Mice vol.7, pp.1, 2015, https://doi.org/10.3390/nu7010682
  7. Synthesis of Renoprotective Chalcone Analogues That Protect Against Cisplatin-induced Cytotoxicity in LLC-PK1 Cells vol.37, pp.11, 2016, https://doi.org/10.1002/bkcs.10984
  8. Natural antioxidants in the treatment and prevention of diabetic nephropathy; a potential approach that warrants clinical trials vol.22, pp.3, 2017, https://doi.org/10.1080/13510002.2017.1297885
  9. Red ginseng powder fermented with probiotics exerts antidiabetic effects in the streptozotocin-induced mouse diabetes model vol.55, pp.1, 2017, https://doi.org/10.1080/13880209.2016.1237978
  10. Design of Green Processing Chain for Processing of Ginseng Fruit Vinegar vol.933, pp.1662-8985, 2014, https://doi.org/10.4028/www.scientific.net/AMR.933.988
  11. Therapeutic Potential of Ginsenosides as an Adjuvant Treatment for Diabetes vol.9, pp.1663-9812, 2018, https://doi.org/10.3389/fphar.2018.00423
  12. Ginseng for the treatment of diabetes and diabetes-related cardiovascular complications: a discussion of the evidence pp.1205-7541, 2019, https://doi.org/10.1139/cjpp-2018-0440
  13. Chemical diversity of ginseng saponins from Panax ginseng vol.39, pp.4, 2013, https://doi.org/10.1016/j.jgr.2014.12.005
  14. A review on the medicinal potential of Panax ginseng saponins in diabetes mellitus vol.5, pp.59, 2013, https://doi.org/10.1039/c5ra05864c
  15. Heat-Processed Scutellariae Radix Enhances Anti-Inflammatory Effect against Lipopolysaccharide-Induced Acute Lung Injury in Mice via NF- κ B Signaling vol.2015, pp.None, 2013, https://doi.org/10.1155/2015/456846
  16. Protective Effects of Processed Ginseng and Its Active Ginsenosides on Cisplatin-Induced Nephrotoxicity: In Vitro and in Vivo Studies vol.63, pp.25, 2015, https://doi.org/10.1021/acs.jafc.5b00782
  17. Effect of Amino Acids on the Generation of Ginsenoside Rg3 Epimers by Heat Processing and the Anticancer Activities of Epimers in A2780 Human Ovarian Cancer Cells vol.2016, pp.None, 2016, https://doi.org/10.1155/2016/3146402
  18. 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, 2013, https://doi.org/10.18632/oncotarget.8285
  19. The Effects of Ginsenosides and Anserine on the Up-Regulation of Renal Aquaporins 1-4 in Hyperuricemic Mice vol.47, pp.5, 2013, https://doi.org/10.1142/s0192415x19500587
  20. Ginsenoside Rk1 inhibits cell proliferation and promotes apoptosis in lung squamous cell carcinoma by calcium signaling pathway vol.9, pp.43, 2013, https://doi.org/10.1039/c9ra05037j
  21. Maltol (3-Hydroxy-2-methyl-4-pyrone) Slows D-Galactose-Induced Brain Aging Process by Damping the Nrf2/HO-1-Mediated Oxidative Stress in Mice vol.67, pp.37, 2013, https://doi.org/10.1021/acs.jafc.9b04614
  22. 고려인삼과 당뇨병: 세포와 동물 및 인체실험을 통한 고려인삼의 당뇨병에 대한 효능 vol.51, pp.1, 2013, https://doi.org/10.22889/kjp.2020.51.1.001
  23. Different absorption and metabolism of ginsenosides after the administration of total ginsenosides and decoction of Panax ginseng vol.34, pp.13, 2020, https://doi.org/10.1002/rcm.8788
  24. Advances in Saponin Diversity of Panax ginseng vol.25, pp.15, 2013, https://doi.org/10.3390/molecules25153452
  25. Anti-Inflammatory Effects of Heat-Processed Artemisia capillaris Thunberg by Regulating IκBα/NF-κB Complex and 15-PGDH in Mouse Macrophage Cells vol.2021, pp.None, 2013, https://doi.org/10.1155/2021/5320314
  26. Ginsenoside Conversion and Anti-Inflammatory Effect on RAW 264.7 Cells of Ginseng Extract Vinegar vol.50, pp.3, 2013, https://doi.org/10.3746/jkfn.2021.50.3.226