DOI QR코드

DOI QR Code

Ginseng in Traditional Herbal Prescriptions

  • Park, Ho-Jae (Department of Oriental Pharmaceutical Science, Kyung Hee University) ;
  • Kim, Dong-Hyun (Department of Life and Nanopharmaceutical Sciences, Kyung Hee University) ;
  • Park, Se-Jin (Department of Life and Nanopharmaceutical Sciences, Kyung Hee University) ;
  • Kim, Jong-Min (Department of Life and Nanopharmaceutical Sciences, Kyung Hee University) ;
  • Ryu, Jong-Hoon (Department of Oriental Pharmaceutical Science, Kyung Hee University)
  • Received : 2011.11.02
  • Accepted : 2012.03.20
  • Published : 2012.07.15

Abstract

Panax ginseng Meyer has been widely used as a tonic in traditional Korean, Chinese, and Japanese herbal medicines and in Western herbal preparations for thousands of years. In the past, ginseng was very rare and was considered to have mysterious powers. Today, the efficacy of drugs must be tested through well-designed clinical trials or meta-analyses, and ginseng is no exception. In the present review, we discuss the functions of ginseng described in historical documents and describe how these functions are taken into account in herbal prescriptions. We also discuss the findings of experimental pharmacological research on the functions of ginseng in ginseng-containing prescriptions and how these prescriptions have been applied in modern therapeutic interventions. The present review on the functions of ginseng in traditional prescriptions helps to demystify ginseng and, as a result, may contribute to expanding the use of ginseng or ginseng-containing prescriptions.

Keywords

References

  1. 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
  2. Blumenthal M, Goldberg A, Brinckmann J. Herbal medicine: expanded Commission E monographs. Austin: Integrative Medicine Communications, 2000.
  3. Park JW, Ryu B, Yeo I, Jerng UM, Han G, Oh S, Lee J, Kim J. Banha-sasim-tang as an herbal formula for the treatment of functional dyspepsia: a randomized, double-blind, placebo-controlled, two-center trial. Trials 2010;11:83. https://doi.org/10.1186/1745-6215-11-83
  4. Mori K, Kondo T, Kamiyama Y, Kano Y, Tominaga K. Preventive effect of Kampo medicine (Hangeshashinto) against irinotecan-induced diarrhea in advanced nonsmall- cell lung cancer. Cancer Chemother Pharmacol 2003;51:403-406.
  5. Naito T, Itoh H, Takeyama M. Comparison of the effects of hange-shashin-to and rikkunshi-to on human plasma calcitonin gene-related peptide and substance P levels. Biol Pharm Bull 2003;26:1104-1107. https://doi.org/10.1248/bpb.26.1104
  6. Kase Y, Saitoh K, Makino B, Hashimoto K, Ishige A, Komatsu Y. Relationship between the antidiarrhoeal effects of Hange-Shashin-To and its active components. Phytother Res 1999;13:468-473. https://doi.org/10.1002/(SICI)1099-1573(199909)13:6<468::AID-PTR504>3.0.CO;2-V
  7. Kim HM, Kim YY, Jang HY, Moon SJ, An NH. Action of Sosiho-Tang on systemic and local anaphylaxis by anal administration. Immunopharmacol Immunotoxicol 1999;21:635-643. https://doi.org/10.3109/08923979909007131
  8. Deng G, Kurtz RC, Vickers A, Lau N, Yeung KS, Shia J, Cassileth B. A single arm phase II study of a Far-Eastern traditional herbal formulation (sho-sai-ko-to or xiao-chaihu- tang) in chronic hepatitis C patients. J Ethnopharmacol 2011;136:83-87. https://doi.org/10.1016/j.jep.2011.04.008
  9. Lee CH, Wang JD, Chen PC. Risk of liver injury associated with Chinese herbal products containing Radix bupleuri in 639,779 patients with hepatitis B virus infection. PLoS One 2011;6:e16064. https://doi.org/10.1371/journal.pone.0016064
  10. Chang JS, Wang KC, Liu HW, Chen MC, Chiang LC, Lin CC. Sho-saiko-to (Xiao-Chai-Hu-Tang) and crude saikosaponins inhibit hepatitis B virus in a stable HBVproducing cell line. Am J Chin Med 2007;35:341-351. https://doi.org/10.1142/S0192415X07004862
  11. Hsu LM, Huang YS, Tsay SH, Chang FY, Lee SD. Acute hepatitis induced by Chinese hepatoprotective herb, xiaochai-hu-tang. J Chin Med Assoc 2006;69:86-88. https://doi.org/10.1016/S1726-4901(09)70119-4
  12. Itoh S, Marutani K, Nishijima T, Matsuo S, Itabashi M. Liver injuries induced by herbal medicine, syo-saiko-to (xiao-chai-hu-tang). Dig Dis Sci 1995;40:1845-1848. https://doi.org/10.1007/BF02212712
  13. Tajiri H, Kozaiwa K, Ozaki Y, Miki K, Shimuzu K, Okada S. Effect of sho-saiko-to(xiao-chai-hu-tang) on HBeAg clearance in children with chronic hepatitis B virus infection and with sustained liver disease. Am J Chin Med 1991;19:121-129. https://doi.org/10.1142/S0192415X91000193
  14. Hirayama C, Okumura M, Tanikawa K, Yano M, Mizuta M, Ogawa N. A multicenter randomized controlled clinical trial of Shosaiko-to in chronic active hepatitis. Gastroenterol Jpn 1989;24:715-719.
  15. Sakaguchi S, Furusawa S, Iizuka Y. Preventive effects of a traditional Chinese medicine (Sho-saiko-to) on septic shock symptoms; approached from heme metabolic disorders in endotoxemia. Biol Pharm Bull 2005;28:165-168. https://doi.org/10.1248/bpb.28.165
  16. Chen MH, Chen JC, Tsai CC, Wang WC, Chang DC, Lin CC, Hsieh HY. Sho-saiko-to prevents liver fibrosis induced by bile duct ligation in rats. Am J Chin Med 2004;32:195-207. https://doi.org/10.1142/S0192415X04001862
  17. Taira Z, Yabe K, Hamaguchi Y, Hirayama K, Kishimoto M, Ishida S, Ueda Y. Effects of Sho-saiko-to extract and its components, Baicalin, baicalein, glycyrrhizin and glycyrrhetic acid, on pharmacokinetic behavior of salicylamide in carbon tetrachloride intoxicated rats. Food Chem Toxicol 2004;42:803-807. https://doi.org/10.1016/j.fct.2003.12.017
  18. Kusunose M, Qiu B, Cui T, Hamada A, Yoshioka S, Ono M, Miyamura M, Kyotani S, Nishioka Y. Effect of Shosaiko- to extract on hepatic inflammation and fibrosis in dimethylnitrosamine induced liver injury rats. Biol Pharm Bull 2002;25:1417-1421. https://doi.org/10.1248/bpb.25.1417
  19. Fujiwara K, Mochida S, Nagoshi S, Iijima O, Matsuzaki Y, Takeda S, Aburada M. Regulation of hepatic macrophage function by oral administration of xiao-chai-hu-tang (shosaiko- to, TJ-9) in rats. J Ethnopharmacol 1995;46:107-114. https://doi.org/10.1016/0378-8741(95)01235-6
  20. Tatsuta M, Iishi H, Baba M, Nakaizumi A, Uehara H. Inhibition by xiao-chai-hu-tang (TJ-9) of development of hepatic foci induced by N-nitrosomorpholine in Sprague- Dawley rats. Jpn J Cancer Res 1991;82:987-992. https://doi.org/10.1111/j.1349-7006.1991.tb01932.x
  21. Yaginuma T, Okamura T, Takeuchi T, Nishii O, Fujimori R. Preventive effect of traditional herbal medicine, shosaikoto, on danazol-induced hepatic damage. Int J Gynaecol Obstet 1989;29:337-341. https://doi.org/10.1016/0020-7292(89)90359-7
  22. Amagaya S, Hayakawa M, Ogihara Y, Ohta Y, Fujiwara K, Oka H, Oshio H, Kishi T. Treatment of chronic liver injury in mice by oral administration of xiao-chai-hu-tang. J Ethnopharmacol 1989;25:181-187. https://doi.org/10.1016/0378-8741(89)90020-2
  23. Borchers AT, Sakai S, Henderson GL, Harkey MR, Keen CL, Stern JS, Terasawa K, Gershwin ME. Shosaiko-to and other Kampo (Japanese herbal) medicines: a review of their immunomodulatory activities. J Ethnopharmacol 2000;73:1-13. https://doi.org/10.1016/S0378-8741(00)00334-2
  24. Kaneko M, Kawakita T, Tauchi Y, Saito Y, Suzuki A, Nomoto K. Augmentation of NK activity after oral administration of a traditional Chinese medicine, xiao-chaihu- tang (shosaiko-to). Immunopharmacol Immunotoxicol 1994;16:41-53. https://doi.org/10.3109/08923979409029899
  25. Tauchi Y, Yamada A, Kawakita T, Saito Y, Suzuki A, Yoshikai Y, Nomoto K. Enhancement of immunoglobulin A production in Peyer's patches by oral administration of a traditional Chinese medicine, xiao-chai-hutang (Shosaiko-to). Immunopharmacol Immunotoxicol 1993;15:251-272. https://doi.org/10.3109/08923979309025998
  26. Matsuura K, Kawakita T, Nakai S, Saito Y, Suzuki A, Nomoto K. Role of B-lymphocytes in the immunopharmacological effects of a traditional Chinese medicine, xiao-chai-hu-tang (shosaiko-to). Int J Immunopharmacol 1993;15:237-243. https://doi.org/10.1016/0192-0561(93)90100-D
  27. Kawakita T, Nakai S, Kumazawa Y, Miura O, Yumioka E, Nomoto K. Induction of interferon after administration of a traditional Chinese medicine, xiao-chai-hu-tang (shosaiko-to). Int J Immunopharmacol 1990;12:515-521. https://doi.org/10.1016/0192-0561(90)90115-4
  28. Kawakita T, Mitsuyama M, Kumazawa Y, Miura O, Yumioka E, Nomoto K. Contribution of cytokines to time-dependent augmentation of resistance against Listeria monocytogenes after administration of a traditional Chinese medicine, xiao-chai-hu-tang (Japanese name: shosaiko-to). Immunopharmacol Immunotoxicol 1989;11:233-255. https://doi.org/10.3109/08923978909005368
  29. Kawakita T, Yamada A, Mitsuyama M, Kumazawa Y, Nomoto K. Protective effect of a traditional Chinese medicine, xiao-chai-hu-tang (Japanese name: shosaiko-to), on Listeria monocytogenes infection in mice. Immunopharmacol Immunotoxicol 1988;10:345-364. https://doi.org/10.3109/08923978809041426
  30. Kumazawa Y, Takimoto H, Miura S, Nishimura C, Yamada A, Kawakita T, Nomoto K. Activation of murine peritoneal macrophages by intraperitoneal administration of a traditional Chinese herbal medicine, xiao-chai-hutang (Japanese name: shosaiko-to). Int J Immunopharmacol 1988;10:395-403. https://doi.org/10.1016/0192-0561(88)90126-9
  31. Kawakita T, Yamada A, Kumazawa Y, Nomoto K. Functional maturation of immature B cells accumulated in the periphery by an intraperitoneal administration of a traditional Chinese medicine, xiao-chai-hu-tang (Japanese name: shosaiko-to). Immunopharmacol Immunotoxicol 1987;9:299-317. https://doi.org/10.3109/08923978709035216
  32. Kawakita T, Yamada A, Mitsuyama M, Kumazawa Y, Nomoto K. Protective effect of a traditional Chinese medicine, xiao-chai-hu-tang (Japanese name: shosaikoto), on Pseudomonas aeruginosa infection in mice. Immunopharmacol Immunotoxicol 1987;9:523-540. https://doi.org/10.3109/08923978709035230
  33. Kobayashi T, Song QH, Hong T, Kitamura H, Cyong JC. Preventive effect of Ninjin-to (Ren-Shen-Tang), a Kampo (Japanese traditional) formulation, on spontaneous autoimmune diabetes in non-obese diabetic (NOD) mice. Microbiol Immunol 2000;44:299-305. https://doi.org/10.1111/j.1348-0421.2000.tb02499.x
  34. Oh MS, Huh Y, Bae H, Ahn DK, Park SK. The multiherbal formula Guibi-tang enhances memory and increases cell proliferation in the rat hippocampus. Neurosci Lett 2005;379:205-208. https://doi.org/10.1016/j.neulet.2004.12.077
  35. Tohda C, Naito R, Joyashiki E. Kihi-to, a herbal traditional medicine, improves Abeta(25-35)-induced memory impairment and losses of neurites and synapses. BMC Complement Altern Med 2008;8:49. https://doi.org/10.1186/1472-6882-8-49
  36. Kanai S, Taniguchi N, Higashino H. Effect of kami-kihito (jia-wei-gui-pi-tang) for experimental osteopenia. Am J Chin Med 2005;33:41-48. https://doi.org/10.1142/S0192415X05002643
  37. Matsuzaki T, Nomura S, Yamaoka M, Ozaki Y, Yoshimura C, Xie GL, Katsura K, Kagawa H, Ishida T, Fukuhara S. HLA and HPA typing in idiopathic thrombocytopenic purpura patients treated with Kami-kihi-to. Am J Chin Med 1998;26:191-198. https://doi.org/10.1142/S0192415X98000245
  38. Yamaguchi K, Kido H, Kawakatsu T, Fukuroi T, Suzuki M, Yanabu M, Nomura S, Kokawa T, Yasunaga K. Effects of kami-kihi-to (jia-wei-gui-pi-tang) on autoantibodies in patients with chronic immune thrombocytopenic purpura. Am J Chin Med 1993;21:251-255. https://doi.org/10.1142/S0192415X93000297
  39. Hara H, Kataoka S, Anan M, Ueda A, Mutoh T, Tabira T. The therapeutic effects of the herbal medicine, Juzentaiho- to, on amyloid-beta burden in a mouse model of Alzheimer's disease. J Alzheimers Dis 2010;20:427-439. https://doi.org/10.3233/JAD-2010-1381
  40. Chino A, Okamoto H, Hirasaki Y, Terasawa K. A case of atopic dermatitis successfully treated with juzentaihoto (Kampo). Altern Ther Health Med 2010;16:62-64.
  41. Nakamoto H, Mimura T, Honda N. Orally administrated Juzen-taiho-to/TJ-48 ameliorates erythropoietin (rHuEPO)-resistant anemia in patients on hemodialysis. Hemodial Int 2008;12 Suppl 2:S9-S14. https://doi.org/10.1111/j.1542-4758.2008.00317.x
  42. Sho Y, Fujisaki K, Sakashita H, Yamaguchi K, Tahara K, Kubozono O, Ido A, Tsubouchi H. Orally administered Kampo medicine, Juzen-taiho-to, ameliorates anemia during interferon plus ribavirin therapy in patients with chronic hepatitis C. J Gastroenterol 2004;39:1202-1204. https://doi.org/10.1007/s00535-004-1472-0
  43. Maruyama Y, Hoshida S, Furukawa M, Ito M. Effects of Japanese herbal medicine, Juzen-taiho-to, in otitisprone children: a preliminary study. Acta Otolaryngol 2009;129:14-18. https://doi.org/10.1080/00016480801998838
  44. Tagami K, Niwa K, Lian Z, Gao J, Mori H, Tamaya T. Preventive effect of Juzen-taiho-to on endometrial carcinogenesis in mice is based on Shimotsu-to constituent. Biol Pharm Bull 2004;27:156-161. https://doi.org/10.1248/bpb.27.156
  45. Lian Z, Niwa K, Gao J, Tagami K, Hashimoto M, Yokoyama Y, Mori H, Tamaya T. Shimotsu-to is the agent in Juzen-taiho-to responsible for the prevention of endometrial carcinogenesis in mice. Cancer Lett 2002;182:19-26. https://doi.org/10.1016/S0304-3835(02)00059-9
  46. Niwa K, Hashimoto M, Morishita S, Lian Z, Tagami K, Mori H, Tamaya T. Preventive effects of Juzen-taiho-to on N-methyl-N-nitrosourea and estradiol-17beta-induced endometrial carcinogenesis in mice. Carcinogenesis 2001;22:587-591. https://doi.org/10.1093/carcin/22.4.587
  47. Sakamoto S, Kudo H, Kuwa K, Suzuki S, Kato T, Kawasaki T, Nakayama T, Kasahara N, Okamoto R. Anticancer effects of a Chinese herbal medicine, juzen-taiho-to, in combination with or without 5-fluorouracil derivative on DNA-synthesizing enzymes in 1,2-dimethylhydrazine induced colonic cancer in rats. Am J Chin Med 1991;19:233-241. https://doi.org/10.1142/S0192415X91000314
  48. Kiyohara H, Matsumoto T, Yamada H. Intestinal immune system modulating polysaccharides in a Japanese herbal (Kampo) medicine, Juzen-Taiho-To. Phytomedicine 2002;9:614-624. https://doi.org/10.1078/094471102321616427
  49. Dai Y, Kato M, Takeda K, Kawamoto Y, Akhand AA, Hossain K, Suzuki H, Nakashima I. T-cell-immunitybased inhibitory effects of orally administered herbal medicine juzen-taiho-to on the growth of primarily developed melanocytic tumors in RET-transgenic mice. J Invest Dermatol 2001;117:694-701. https://doi.org/10.1046/j.0022-202x.2001.01457.x
  50. Matsumoto T, Yamada H. Orally administered Kampo (Japanese herbal) medicine, "Juzen-Taiho-To" modulates cytokine secretion in gut associated lymphoreticular tissues in mice. Phytomedicine 2000;6:425-430. https://doi.org/10.1016/S0944-7113(00)80070-7
  51. Kiyohara H, Matsumoto T, Yamada H. Lignin-carbohydrate complexes: intestinal immune system modulating ingredients in kampo (Japanese herbal) medicine, juzentaiho-to. Planta Med 2000;66:20-24. https://doi.org/10.1055/s-2000-11116
  52. Matsumoto T, Sakurai MH, Kiyohara H, Yamada H. Orally administered decoction of Kampo (Japanese herbal) medicine, "Juzen-Taiho-To" modulates cytokine secretion and induces NKT cells in mouse liver. Immunopharmacology 2000;46:149-161. https://doi.org/10.1016/S0162-3109(99)00166-6
  53. Iijima K, Sun S, Cyong JC, Jyonouchi H. Juzen-taiho-to, a Japanese herbal medicine, modulates type 1 and type 2 T cell responses in old BALB/c mice. Am J Chin Med 1999;27:191-203. https://doi.org/10.1142/S0192415X99000239
  54. Abe S, Tansho S, Ishibashi H, Inagaki N, Komatsu Y, Yamaguchi H. Protective effect of oral administration of a traditional medicine, Juzen-Taiho-To, and its components on lethal Candida albicans infection in immunosuppressed mice. Immunopharmacol Immunotoxicol 1998;20:421-431. https://doi.org/10.3109/08923979809034824
  55. Jeong JS, Ryu BH, Kim JS, Park JW, Choi WC, Yoon SW. Bojungikki-tang for cancer-related fatigue: a pilot randomized clinical trial. Integr Cancer Ther 2010;9:331-338. https://doi.org/10.1177/1534735410383170
  56. Shin HY, Shin CH, Shin TY, Lee EJ, Kim HM. Effect of bojungikki-tang on lipopolysaccharide-induced cytokine production from peripheral blood mononuclear cells of chronic fatigue syndrome patients. Immunopharmacol Immunotoxicol 2003;25:491-501. https://doi.org/10.1081/IPH-120026435
  57. Chen R, Moriya J, Luo X, Yamakawa J, Takahashi T, Sasaki K, Yoshizaki F. Hochu-ekki-to combined with interferon-gamma moderately enhances daily activity of chronic fatigue syndrome mice by increasing NK cell activity, but not neuroprotection. Immunopharmacol Immunotoxicol 2009;31:238-245. https://doi.org/10.1080/08923970802391525
  58. Chen R, Moriya J, Yamakawa J, Takahashi T, Li Q, Morimoto S, Iwai K, Sumino H, Yamaguchi N, Kanda T. Brain atrophy in a murine model of chronic fatigue syndrome and benefi cial effect of Hochu-ekki-to (TJ-41). Neurochem Res 2008;33:1759-1767. https://doi.org/10.1007/s11064-008-9620-1
  59. Liu XQ, Wu L, Guo XJ. Effect of Bu-Zhong-Yi-Qi-Tang on defi ciency of N-glycan/nitric oxide and islet damage induced by streptozotocin in diabetic rats. World J Gastroenterol 2009;15:1730-1737. https://doi.org/10.3748/wjg.15.1730
  60. Tatsumi K, Shinozuka N, Nakayama K, Sekiya N, Kuriyama T, Fukuchi Y. Hochuekkito improves systemic infl ammation and nutritional status in elderly patients with chronic obstructive pulmonary disease. J Am Geriatr Soc 2009;57:169-170. https://doi.org/10.1111/j.1532-5415.2009.02034.x
  61. Kimura M, Sasada T, Kanai M, Kawai Y, Yoshida Y, Hayashi E, Iwata S, Takabayashi A. Preventive effect of a traditional herbal medicine, Hochu-ekki-to, on immunosuppression induced by surgical stress. Surg Today 2008;38:316-322. https://doi.org/10.1007/s00595-007-3631-4
  62. Kuroiwa A, Liou S, Yan H, Eshita A, Naitoh S, Nagayama A. Effect of a traditional Japanese herbal medicine, hochu-ekki-to (Bu-Zhong-Yi-Qi Tang), on immunity in elderly persons. Int Immunopharmacol 2004;4:317-324. https://doi.org/10.1016/j.intimp.2003.12.004
  63. Nabeshima S, Murata M, Hamada M, Chong Y, Yamaji K, Hayashi J. Maturation of monocyte-derived dendritic cells by Hochu-ekki-to, a traditional Japanese herbal medicine. Int Immunopharmacol 2004;4:37-45. https://doi.org/10.1016/j.intimp.2003.10.002
  64. Furuya Y, Akashi T, Fuse H. Effect of Bu-zhong-yi-qitang on seminal plasma cytokine levels in patients with idiopathic male infertility. Arch Androl 2004;50:11-14. https://doi.org/10.1080/01485010490250515
  65. Matsumoto T, Noguchi M, Hayashi O, Makino K, Yamada H. Hochuekkito, a Kampo (traditional Japanese herbal) medicine, enhances mucosal IgA antibody response in mice immunized with antigen-entrapped biodegradable microparticles. Evid Based Complement Alternat Med 2010;7:69-77. https://doi.org/10.1093/ecam/nem166
  66. Kiyohara H, Nagai T, Munakata K, Nonaka K, Hanawa T, Kim SJ, Yamada H. Stimulating effect of Japanese herbal (kampo) medicine, hochuekkito on upper respiratory mucosal immune system. Evid Based Complement Alternat Med 2006;3:459-467. https://doi.org/10.1093/ecam/nel030
  67. Kobayashi H, Mizuno N, Kutsuna H, Teramae H, Ueoku S, Onoyama J, Yamanaka K, Fujita N, Ishii M. Hochu-ekkito suppresses development of dermatitis and elevation of serum IgE level in NC/Nga mice. Drugs Exp Clin Res 2003;29:81-84.
  68. Nakada T, Watanabe K, Matsumoto T, Santa K, Triizuka K, Hanawa T. Effect of orally administered Hochu-ekki-to, a Japanese herbal medicine, on contact hypersensitivity caused by repeated application of antigen. Int Immunopharmacol 2002;2:901-911. https://doi.org/10.1016/S1567-5769(02)00027-9
  69. Yamaoka Y, Kawakita T, Nomoto K. Protective effect of a traditional Japanese medicine Hochu-ekki-to (Chinese name: Bu-zhong-yi-qi-tang), on the susceptibility against Listeria monocytogenes in infant mice. Int Immunopharmacol 2001;1:1669-1677. https://doi.org/10.1016/S1567-5769(01)00076-5
  70. Ishimitsu R, Nishimura H, Kawauchi H, Kawakita T, Yoshikai Y. Dichotomous effect of a traditional Japanese medicine, bu-zhong-yi-qi-tang on allergic asthma in mice. Int Immunopharmacol 2001;1:857-865. https://doi.org/10.1016/S1567-5769(01)00022-4
  71. Kido T, Mori K, Daikuhara H, Tsuchiya H, Ishige A, Sasaki H. The protective effect of hochu-ekki-to (TJ-41), a Japanese herbal medicine, against HSV-1 infection in mitomycin C-treated mice. Anticancer Res 2000;20:4109-4113.
  72. Harada M, Seta K, Ito O, Tamada K, Li T, Terao H, Takenoyama M, Kimura G, Nomoto K. Concomitant immunity against tumor development is enhanced by the oral administration of a kampo medicine, Hochu-ekki-to (TJ-41: Bu-Zhong-Yi-Qi-Tang). Immunopharmacol Immunotoxicol 1995;17:687-703. https://doi.org/10.3109/08923979509037189
  73. Li XY, Takimoto H, Miura S, Yoshikai Y, Matsuzaki G, Nomoto K. Effect of a traditional Chinese medicine, buzhong- yi-qi-tang (Japanese name: Hochu-ekki-to) on the protection against Listeria monocytogenes infection in mice. Immunopharmacol Immunotoxicol 1992;14:383-402. https://doi.org/10.3109/08923979209005400
  74. Tsuneoka N, Tajima Y, Kitasato A, Fukuda K, Kitajima T, Adachi T, Mishima T, Kuroki T, Onizuka S, Kanematsu T. Chemopreventative effect of hochu-ekki-to (TJ-41) on chemically induced biliary carcinogenesis in hamsters. J Surg Res 2009;151:22-27. https://doi.org/10.1016/j.jss.2008.01.003
  75. Tajima S, Bando M, Yamasawa H, Ohno S, Moriyama H, Terada M, Takada T, Suzuki E, Gejyo F, Sugiyama Y. Preventive effect of hochu-ekki-to, a Japanese herbal medicine, on bleomycin-induced lung injury in mice. Respirology 2007;12:814-822. https://doi.org/10.1111/j.1440-1843.2007.01146.x
  76. Yang SH, Yu CL. Antiinflammatory effects of Bu-zhongyi-qi-tang in patients with perennial allergic rhinitis. J Ethnopharmacol 2008;115:104-109. https://doi.org/10.1016/j.jep.2007.09.011
  77. Shinozuka N, Tatsumi K, Nakamura A, Terada J, Kuriyama T. The traditional herbal medicine Hochuekkito improves systemic inflammation in patients with chronic obstructive pulmonary disease. J Am Geriatr Soc 2007;55:313-314. https://doi.org/10.1111/j.1532-5415.2007.01057.x
  78. Tajima S, Bando M, Yamasawa H, Ohno S, Moriyama H, Takada T, Suzuki E, Gejyo F, Sugiyama Y. Preventive effect of Hochu-ekki-to on lipopolysaccharide-induced acute lung injury in BALB/c mice. Lung 2006;184:318-323. https://doi.org/10.1007/s00408-006-0018-z
  79. Kanehara M, Ogirima T, Tano K, Maenaka T, Ishida T, Zhang B, Li G, Wang X, Guo Y. Effects of Chinese herbal medicine based on hachimi-jio-gan on osteopenia in rats. J Tradit Chin Med 2006;26:72-77.
  80. Kobayashi H, Mizuno N, Teramae H, Kutsuna H, Ueoku S, Onoyama J, Yamanaka K, Fujita N, Ishii M. The effects of Hochu-ekki-to in patients with atopic dermatitis resistant to conventional treatment. Int J Tissue React 2004;26:113-117.
  81. Ochi T, Kawakita T, Nomoto K. Effects of Hochu-ekkito and Ninjin-youei-to, traditional Japanese medicines, on porcine serum-induced liver fibrosis in rats. Immunopharmacol Immunotoxicol 2004;26:285-298. https://doi.org/10.1081/IPH-120037726
  82. Yan X, Kita M, Minami M, Yamamoto T, Kuriyama H, Ohno T, Iwakura Y, Imanishi J. Antibacterial effect of Kampo herbal formulation Hochu-ekki-to (Bu-Zhong-Yi-Qi-Tang) on Helicobacter pylori infection in mice. Microbiol Immunol 2002;46:475-482. https://doi.org/10.1111/j.1348-0421.2002.tb02721.x
  83. Takano F, Ohta Y, Tanaka T, Sasaki K, Kobayashi K, Takahashi T, Yahagi N, Yoshizaki F, Fushiya S, Ohta T. Oral administration of Ren-shen-yang-rong-tang 'Ninjin'yoeito' protects against hematotoxicity and induces immature erythroid progenitor cells in 5-fluorouracilinduced anemia. Evid Based Complement Alternat Med 2009;6:247-256. https://doi.org/10.1093/ecam/nem080
  84. Egashira N, Yuzurihara M, Hattori N, Sakakibara I, Ishige A. Ninjin-yoei-to (Ren-Shen-Yang-Rong-Tang) and Polygalae radix improves scopolamine-induced impairment of passive avoidance response in mice. Phytomedicine 2003;10:467-473. https://doi.org/10.1078/094471103322331403
  85. Nakada T, Watanabe K, Jin GB, Triizuk K, Hanawa T. Effect of ninjin-youei-to on Th1/Th2 type cytokine production in different mouse strains. Am J Chin Med 2002;30:215-223. https://doi.org/10.1142/S0192415X0200034X
  86. Nakai S, Kawakita T, Himeno K, Nomoto K. Combined treatments with Ninjin-youei-to (Ren-shen-yang-rongtang) plus a suboptimal dose of prednisolone on autoimmune nephritis in MRL/lpr mice. Int J Immunopharmacol 1998;20:275-284. https://doi.org/10.1016/S0192-0561(98)00032-0
  87. Nakai S, Kawakita T, Nagasawa H, Himeno K, Nomoto K. Thymus-dependent effects of a traditional Chinese medicine, ren-shen-yang-rong-tang (Japanese name; Ninjinyouei- to), in autoimmune MRI/MP-lpr/lpr mice. Int J Immunopharmacol 1996;18:271-279. https://doi.org/10.1016/0192-0561(96)84507-3
  88. Nakai S, Kawakita T, Zhou NN, Matsuura K, Oka M, Nagasawa H, Saito Y, Suzuki A, Himeno K, Nomoto K. Treatment effect of a traditional Chinese medicine, renshen-yang-rong-tang (Japanese name: ninjin-youei-to), on autoimmune MRL/MP-lpr/lpr mice. Int J Immunopharmacol 1993;15:589-596. https://doi.org/10.1016/0192-0561(93)90076-B
  89. Harigai E, Nakai S, Kawakita T, Nomoto K. Combined treatment with ren-shen-yang-rong-tang (Japanese name: ninjin-youei-to) plus prednisolone on adjuvantinduced arthritis in Lewis rat. Int J Immunopharmacol 1995;17:411-418. https://doi.org/10.1016/0192-0561(95)00018-W
  90. Kono T, Kaneko A, Hira Y, Suzuki T, Chisato N, Ohtake N, Miura N, Watanabe T. Anti-colitis and -adhesion effects of daikenchuto via endogenous adrenomedullin enhancement in Crohn's disease mouse model. J Crohns Colitis 2010;4:161-170. https://doi.org/10.1016/j.crohns.2009.09.006
  91. Narita M, Hatano E, Tamaki N, Yamanaka K, Yanagida A, Nagata H, Asechi H, Takada Y, Ikai I, Uemoto S. Daikenchu-to attenuates rat sinusoidal obstruction syndrome by inhibiting the accumulation of neutrophils in the liver. J Gastroenterol Hepatol 2009;24:1051-1057. https://doi.org/10.1111/j.1440-1746.2009.05795.x
  92. Kawasaki N, Nakada K, Suzuki Y, Furukawa Y, Hanyu N, Kashiwagi H. Effect of Dai-kenchu-to on gastrointestinal motility and gastric emptying. Int J Surg 2009;7:218-222. https://doi.org/10.1016/j.ijsu.2009.03.008
  93. Kawasaki N, Nakada K, Nakayoshi T, Furukawa Y, Suzuki Y, Hanyu N, Yanaga K. Effect of Dai-kenchu-to on gastrointestinal motility based on differences in the site and timing of administration. Dig Dis Sci 2007;52:2684-2694. https://doi.org/10.1007/s10620-006-9391-y
  94. Tokita Y, Yuzurihara M, Sakaguchi M, Satoh K, Kase Y. The pharmacological effects of Daikenchuto, a traditional herbal medicine, on delayed gastrointestinal transit in rat postoperative ileus. J Pharmacol Sci 2007;104:303-310. https://doi.org/10.1254/jphs.FP0070831
  95. Tokita Y, Satoh K, Sakaguchi M, Endoh Y, Mori I, Yuzurihara M, Sakakibara I, Kase Y, Takeda S, Sasaki H. The preventive effect of Daikenchuto on postoperative adhesion-induced intestinal obstruction in rats. Inflammopharmacology 2007;15:65-66. https://doi.org/10.1007/s10787-006-1552-2
  96. Fukuda H, Chen C, Mantyh C, Ludwig K, Pappas TN, Takahashi T. The herbal medicine, Dai-Kenchu-to, accelerates delayed gastrointestinal transit after the operation in rats. J Surg Res 2006;131:290-295. https://doi.org/10.1016/j.jss.2005.09.018
  97. Kito Y, Suzuki H. Effects of Dai-kenchu-to on spontaneous activity in the mouse small intestine. J Smooth Muscle Res 2006;42:189-201. https://doi.org/10.1540/jsmr.42.189
  98. Satoh K, Kase Y, Hayakawa T, Murata P, Ishige A, Sasaki H. Dai-kenchu-to enhances accelerated small intestinal movement. Biol Pharm Bull 2001;24:1122-1126. https://doi.org/10.1248/bpb.24.1122
  99. Iwai N, Kume Y, Kimura O, Ono S, Aoi S, Tsuda T. Effects of herbal medicine Dai-Kenchu-to on anorectal function in children with severe constipation. Eur J Pediatr Surg 2007;17:115-118. https://doi.org/10.1055/s-2007-965016
  100. Sakakibara R, Odaka T, Lui Z, Uchiyama T, Yamaguchi K, Yamaguchi T, Asahina M, Yamamoto T, Ito T, Hattori T. Dietary herb extract dai-kenchu-to ameliorates constipation in parkinsonian patients (Parkinson's disease and multiple system atrophy). Mov Disord 2005;20:261-262. https://doi.org/10.1002/mds.20352
  101. Satoh K, Hashimoto K, Hayakawa T, Ishige A, Kaneko M, Ogihara S, Kurosawa S, Yakabi K, Nakamura T. Mechanism of atropine-resistant contraction induced by Dai-kenchu-to in guinea pig ileum. Jpn J Pharmacol 2001;86:32-37. https://doi.org/10.1254/jjp.86.32
  102. Satoh K, Hayakawa T, Kase Y, Ishige A, Sasaki H, Nishikawa S, Kurosawa S, Yakabi K, Nakamura T. Mechanisms for contractile effect of Dai-kenchu-to in isolated guinea pig ileum. Dig Dis Sci 2001;46:250-256. https://doi.org/10.1023/A:1005636412287
  103. Ogasawara T, Morine Y, Ikemoto T, Imura S, Fujii M, Soejima Y, Shimada M. Influence of Dai-kenchu-to (DKT) on human portal blood flow. Hepatogastroenterology 2008;55:574-577.
  104. Murata P, Kase Y, Ishige A, Sasaki H, Kurosawa S, Nakamura T. The herbal medicine Dai-kenchu-to and one of its active components [6]-shogaol increase intestinal blood flow in rats. Life Sci 2002;70:2061-2070. https://doi.org/10.1016/S0024-3205(01)01552-1
  105. Ni Q, Wang J, Li EQ, Zhao AB, Yu B, Wang M, Huang CR. Study on the protective effect of shengmai san (see text) on the myocardium in the type 2 diabetic cardiomyopathy model rat. J Tradit Chin Med 2011;31:209-219. https://doi.org/10.1016/S0254-6272(11)60044-7
  106. Giridharan VV, Thandavarayan RA, Konishi T. Effect of Shengmai-san on cognitive performance and cerebral oxidative damage in BALB/c mice. J Med Food 2011;14:601-609. https://doi.org/10.1089/jmf.2010.1362
  107. Seo TB, Baek K, Kwon KB, Lee SI, Lim JS, Seol IC, Kim YS, Seo YB, Namgung U. Shengmai-san-mediated enhancement of regenerative responses of spinal cord axons after injury in rats. J Pharmacol Sci 2009;110:483-492. https://doi.org/10.1254/jphs.09044FP
  108. Yao HT, Chang YW, Chen CT, Chiang MT, Chang L, Yeh TK. Shengmai San reduces hepatic lipids and lipid peroxidation in rats fed on a high-cholesterol diet. J Ethnopharmacol 2008;116:49-57. https://doi.org/10.1016/j.jep.2007.10.043
  109. Ichikawa H, Wang L, Konishi T. Prevention of cerebral oxidative injury by post-ischemic intravenous administration of Shengmai San. Am J Chin Med 2006;34:591-600. https://doi.org/10.1142/S0192415X06004120
  110. Wang NL, Liou YL, Lin MT, Lin CL, Chang CK. Chinese herbal medicine, Shengmai San, is effective for improving circulatory shock and oxidative damage in the brain during heatstroke. J Pharmacol Sci 2005;97:253-265. https://doi.org/10.1254/jphs.FP0040793
  111. Ichikawa H, Wang X, Konishi T. Role of component herbs in antioxidant activity of shengmai san: a traditional Chinese medicine formula preventing cerebral oxidative damage in rat. Am J Chin Med 2003;31:509-521. https://doi.org/10.1142/S0192415X03001193
  112. Ichikawa H, Konishi T. In vitro antioxidant potentials of traditional Chinese medicine, Shengmai San and their relation to in vivo protective effect on cerebral oxidative damage in rats. Biol Pharm Bull 2002;25:898-903. https://doi.org/10.1248/bpb.25.898
  113. Xuejiang W, Magara T, Konishi T. Prevention and repair of cerebral ischemia-reperfusion injury by Chinese herbal medicine, shengmai san, in rats. Free Radic Res 1999;31:449-455. https://doi.org/10.1080/10715769900301011
  114. Yoshie F, Iizuka A, Kubo M, Komatsu Y, Matsumoto A, Itakura H, Takeda H, Matsumiya T, Kondo K. Protective effects of Saiko-ka-ryukotsu-borei-to (Chai-Hu-Jia-Long-Gu-Mu-Li-Tang) against atherosclerosis in Kurosawa and Kusanagi-hypercholesterolemic (KHC) rabbits. Pharmacol Res 2001;43:481-488. https://doi.org/10.1006/phrs.2001.0804
  115. Chen CY, Kuo TL, Sheu SY, Kuo TF. Preventive effects of Chinese herb chai-hu-gui-zhi-tang extract on water immersion restraint stress-induced acute gastric ulceration in rats. J Vet Med Sci 2010;72:679-685. https://doi.org/10.1292/jvms.09-0284
  116. Horie Y, Kajihara M, Mori S, Yamagishi Y, Kimura H, Tamai H, Kato S, Ishii H. Japanese herbal medicine, Saiko-keishi-to, prevents gut ischemia/reperfusioninduced liver injury in rats via nitric oxide. World J Gastroenterol 2004;10:2241-2244. https://doi.org/10.3748/wjg.v10.i15.2241
  117. Tatsuta M, Iishi H, Baba M, Narahara H, Yano H, Sakai N. Suppression by Chai-hu-gui-zhi-tang of the development of liver lesions induced by N-nitrosomorpholine in Sprague-Dawley rats. Cancer Lett 2000;152:31-36. https://doi.org/10.1016/S0304-3835(99)00429-2
  118. Nose M, Tamura M, Ryu N, Mizukami H, Ogihara Y. Sho-saiko-to and Saiko-keisi-to, the traditional Chinese and Japanese herbal medicines, altered hepatic drug-metabolizing enzymes in mice and rats when administered orally for a long time. J Pharm Pharmacol 2003;55:1419-1426. https://doi.org/10.1211/0022357021873
  119. Su SB, Xie MJ, Sawabu N, Motoo Y. Suppressive effect of herbal medicine saikokeishito on acinar cell apoptosis in rat spontaneous chronic pancreatitis. Pancreatology 2007;7:28-36. https://doi.org/10.1159/000101875
  120. Su SB, Motoo Y, Xie MJ, Taga H, Sawabu N. Antifibrotic effect of the herbal medicine Saiko-keishi-to (TJ-10) on chronic pancreatitis in the WBN/Kob rat. Pancreas 2001;22:8-17. https://doi.org/10.1097/00006676-200101000-00002
  121. Motoo Y, Su SB, Xie MJ, Taga H, Sawabu N. Effect of herbal medicine Saiko-keishi-to (TJ-10) on rat spontaneous chronic pancreatitis: comparison with other herbal medicines. Int J Pancreatol 2000;27:123-129. https://doi.org/10.1385/IJGC:27:2:123
  122. Su SB, Motoo Y, Xie MJ, Sakai J, Taga H, Sawabu N. Expression of pancreatitis-associated protein (PAP) in rat spontaneous chronic pancreatitis: effect of herbal medicine Saiko-keishi-to (TJ-10). Pancreas 1999;19:239-247. https://doi.org/10.1097/00006676-199910000-00004
  123. Sakaguchi M, Goto K, Ichiki H, Hattori N, Iizuka A, Yamamoto M, Takeda S, Ishige A, Aburada M, Yasuda M et al. Effects of Byakko-ka-ninjin-to on salivary secretion and bladder function in rats. J Ethnopharmacol 2005;102:164-169. https://doi.org/10.1016/j.jep.2005.05.049
  124. Aburada T, Ikarashi N, Kagami M, Ichikawa Y, Sugitani M, Maniwa A, Ueda H, Toda T, Ito K, Ochiai W et al. Byakkokaninjinto prevents body water loss by increasing the expression of kidney aquaporin-2 and skin aquaporin- 3 in KKAy mice. Phytother Res 2011;25:897-903. https://doi.org/10.1002/ptr.3358
  125. Yanagi Y, Yasuda M, Hashida K, Kadokura Y, Yamamoto T, Suzaki H. Mechanism of salivary secretion enhancement by Byakkokaninjinto. Biol Pharm Bull 2008;31:431-435. https://doi.org/10.1248/bpb.31.431
  126. Kimura M, Kimura I, Chem FJ. Combined potentiating effect of byakko-ka-ninjin-to, its constituents, rhizomes of Anemarrhena asphodeloides, tomosaponin A-III, and calcium on pilocarpine-induced saliva secretion in streptozocin- diabetic mice. Biol Pharm Bull 1996;19:926-931. https://doi.org/10.1248/bpb.19.926
  127. Tohda C, Sugahara H, Kuraishi Y, Komatsu K. Inhibitory effect of Byakko-ka-ninjin-to on itch in a mouse model of atopic dermatitis. Phytother Res 2000;14:192-194. https://doi.org/10.1002/(SICI)1099-1573(200005)14:3<192::AID-PTR609>3.0.CO;2-F
  128. Tatsumi T, Yamada T, Nagai H, Terasawa K, Tani T, Nunome S, Saiki I. A Kampo formulation: Byakko-kaninjin- to (Bai-Hu-Jia-Ren-Sheng-Tang) inhibits IgEmediated triphasic skin reaction in mice: the role of its constituents in expression of the efficacy. Biol Pharm Bull 2001;24:284-290. https://doi.org/10.1248/bpb.24.284
  129. Kimura I, Nakashima N, Sugihara Y, Fu-jun C, Kimura M. The antihyperglycaemic blend effect of traditional Chinese medicine byakko-ka-ninjin-to on alloxan and diabetic KK-CA(y) mice. Phytother Res 1999;13:484-488. https://doi.org/10.1002/(SICI)1099-1573(199909)13:6<484::AID-PTR485>3.0.CO;2-X
  130. Fan H, Shen L, Tang Q, Xiong P, Shou Z, Liao Y, Liang L, Chen X. Effect of Wumeiwan on cytokines TNF-alpha, IL-6, IL-8, IL-10 and expression of NF-kappaBp65 in rats with ulcerative colitis. J Huazhong Univ Sci Technol Med Sci 2009;29:650-654. https://doi.org/10.1007/s11596-009-0523-4
  131. Fan H, Qiu MY, Mei JJ, Shen GX, Liu SL, Chen R. Effects of four regulating-intestine prescriptions on pathology and ultrastructure of colon tissue in rats with ulcerative colitis. World J Gastroenterol 2005;11:4800-4806. https://doi.org/10.3748/wjg.v11.i31.4800
  132. Chen FP, Chen FJ, Jong MS, Tsai HL, Wang JR, Hwang SJ. Modern use of Chinese herbal formulae from Shang-Han Lun. Chin Med J (Engl) 2009;122:1889-1894.
  133. Xiang YZ, Shang HC, Gao XM, Zhang BL. A comparison of the ancient use of ginseng in traditional Chinese medicine with modern pharmacological experiments and clinical trials. Phytother Res 2008;22:851-858. https://doi.org/10.1002/ptr.2384
  134. An X, Zhang AL, Yang AW, Lin L, Wu D, Guo X, Shergis JL, Thien FC, Worsnop CJ, Xue CC. Oral ginseng formulae for stable chronic obstructive pulmonary disease: a systematic review. Respir Med 2011;105:165-176. https://doi.org/10.1016/j.rmed.2010.11.007
  135. Zhang Y, Saito H, Nishiyama N. Improving effects of DX-9386, a traditional Chinese medicinal prescription, on thymectomy-induced impairment of learning behaviors in mice. Biol Pharm Bull 1994;17:1199-1205. https://doi.org/10.1248/bpb.17.1199
  136. Choi JH, Kim HM, Song YS, Park SG, Kim JJ, Lee CK. Anti-aging effects Saccharomyces fermented modified Kyungohkgo extract on skin. Korean J Herbol 2007;22:219-225.
  137. Scheid V, Bensky D, Ellis A, Barolet R. Chinese herbal medicine: fomulas and strategies. 3rd ed. Seatle: Estland Press, 2009.
  138. Panax ginseng. Monograph. Altern Med Rev 2009; 14:172-176.
  139. Coon JT, Ernst E. Panax ginseng: a systematic review of adverse effects and drug interactions. Drug Saf 2002;25:323-344. https://doi.org/10.2165/00002018-200225050-00003
  140. Siow YL, Gong Y, Au-Yeung KK, Woo CW, Choy PC, O K. Emerging issues in traditional Chinese medicine. Can J Physiol Pharmacol 2005;83:321-334. https://doi.org/10.1139/y05-029
  141. Cho BH. The politics of herbal drugs in Korea. Soc Sci Med 2000;51:505-509. https://doi.org/10.1016/S0277-9536(99)00492-X
  142. Yu F, Takahashi T, Moriya J, Kawaura K, Yamakawa J, Kusaka K, Itoh T, Morimoto S, Yamaguchi N, Kanda T. Traditional Chinese medicine and Kampo: a review from the distant past for the future. J Int Med Res 2006;34:231-239. https://doi.org/10.1177/147323000603400301
  143. Leung KF, Liu FB, Zhao L, Fang JQ, Chan K, Lin LZ. Development and validation of the Chinese Quality of Life Instrument. Health Qual Life Outcomes 2005;3:26. https://doi.org/10.1186/1477-7525-3-26
  144. Kishida Y, Nishii T, Inoue T, Nishida S, Arimitsu J, Yoshikawa H, Sugano N. Juzentaihoto (TJ-48), a traditional Japanese herbal medicine, infl uences hemoglobin recovery during preoperative autologous blood donation and after hip surgery. Int J Clin Pharmacol Ther 2009;47:716-721. https://doi.org/10.5414/CPP47716
  145. Wang XQ, Takahashi T, Zhu SJ, Moriya J, Saegusa S, Yamakawa J, Kusaka K, Itoh T, Kanda T. Effect of Hochu-ekki-to (TJ-41), a Japanese herbal medicine, on daily activity in a murine model of chronic fatigue syndrome. Evid Based Complement Alternat Med 2004;1:203-206. https://doi.org/10.1093/ecam/neh020
  146. Nagano T, Itoh H, Takeyama M. Effect of Dai-kenchuto on levels of 3 brain-gut peptides (motilin, gastrin and somatostatin) in human plasma. Biol Pharm Bull 1999;22:1131-1133. https://doi.org/10.1248/bpb.22.1131
  147. Nagano T, Itoh H, Takeyama M. Effects of Dai-kenchuto on levels of 5-hydroxytryptamine (serotonin) and vasoactive intestinal peptides in human plasma. Biol Pharm Bull 2000;23:352-353. https://doi.org/10.1248/bpb.23.352
  148. Itoh T, Yamakawa J, Mai M, Yamaguchi N, Kanda T. The effect of the herbal medicine dai-kenchu-to on postoperative ileus. J Int Med Res 2002;30:428-432. https://doi.org/10.1177/147323000203000410
  149. Kawahara H, Yanaga K. The herbal medicine Dai-Kenchu-To directly stimulates colonic motility. Surg Today 2009;39:175-177. https://doi.org/10.1007/s00595-008-3810-y
  150. Iwabu J, Watanabe J, Hirakura K, Ozaki Y, Hanazaki K. Profiling of the compounds absorbed in human plasma and urine after oral administration of a traditional Japanese (kampo) medicine, daikenchuto. Drug Metab Dispos 2010;38:2040-2048. https://doi.org/10.1124/dmd.110.033589
  151. Sato Y, Katagiri F, Inoue S, Itoh H, Takeyama M. Effects of Ninjin-to on levels of calcitonin gene-related peptide and substance P in human plasma. Biol Pharm Bull 2004;27:2032-2034. https://doi.org/10.1248/bpb.27.2032
  152. Pratico D, Sung S. Lipid peroxidation and oxidative imbalance: early functional events in Alzheimer's disease. J Alzheimers Dis 2004;6:171-175. https://doi.org/10.3233/JAD-2004-6209
  153. Parihar MS, Hemnani T. Alzheimer's disease pathogenesis and therapeutic interventions. J Clin Neurosci 2004;11:456-467. https://doi.org/10.1016/j.jocn.2003.12.007
  154. Ichikawa H, Konishi T. In vitro antioxidant potentials of traditional Chinese medicine, Shengmai San and their relation to in vivo protective effect on cerebral oxidative damage in rats. Biol Pharm Bull 2002;25:898-903. https://doi.org/10.1248/bpb.25.898
  155. Wang L, Muxin G, Nishida H, Shirakawa C, Sato S, Konishi T. Psychological stress-induced oxidative stress as a model of sub-healthy condition and the effect of TCM. Evid Based Complement Alternat Med 2007;4:195-202. https://doi.org/10.1093/ecam/nel080
  156. Nishiyama N, Zhou Y, Takashina K, Saito H. Effects of DX-9386, a traditional Chinese preparation, on passive and active avoidance performances in mice. Biol Pharm Bull 1994;17:1472-1476. https://doi.org/10.1248/bpb.17.1472
  157. Zhang Y, Takashina K, Saito H, Nishiyama N. Anti-aging effect of DX-9386 in senescence accelerated mouse. Biol Pharm Bull 1994;17:866-868. https://doi.org/10.1248/bpb.17.866
  158. Shin BY, Lee YH, Kim D, Park CS, Lee YW, Cho HJ, Kim DH, Yamamoto Y, Kang DH, Lee S et al. Ameliorating effect of a herbal medicinal prescription, Kyung-Ok-Ko, on scopolamine-induced memory impairment in mice. J Tradit Med 2009;26:35-43.
  159. Morimoto Y, Sakata M, Ohno A, Maegawa T, Tajima S. Effects of Byakko-ka-ninjin-to, Bofu-tsusho-san and Gorei-san on blood glucose level, water intake and urine volume in KKAy mice. Yakugaku Zasshi 2002;122:163-168. https://doi.org/10.1248/yakushi.122.163
  160. Jia L, Zhao Y. Current evaluation of the millennium phytomedicine. Ginseng (I): etymology, pharmacognosy, phytochemistry, market and regulations. Curr Med Chem 2009;16:2475-2484. https://doi.org/10.2174/092986709788682146
  161. Shibata S. Chemistry and cancer preventing activities of ginseng saponins and some related triterpenoid compounds. J Korean Med Sci 2001;16 Suppl:S28-S37. https://doi.org/10.3346/jkms.2001.16.S.S28
  162. Paker L, Cadenas E. Herbal and traditional medicine. New York: Marcel Dekker, 2004.
  163. Choi KT. Botanical characteristics, pharmacological effects and medicinal components of Korean Panax ginseng C A Meyer. Acta Pharmacol Sin 2008;29:1109-1118. https://doi.org/10.1111/j.1745-7254.2008.00869.x
  164. Wood WB, Roh BL, White RP. Cardiovascular actions of Panx ginseng in dogs. Jpn J Pharmacol 1964;14:284-294. https://doi.org/10.1254/jjp.14.284
  165. Lee KD, Huemer RP. Antitumoral activity of Panax ginseng extracts. Jpn J Pharmacol 1971;21:299-302. https://doi.org/10.1254/jjp.21.299
  166. Hsu CT. On Panax shinseng Nies, var. Notoginseng Burkill. Jpn J Pharmacol 1956;6:18-28. https://doi.org/10.1254/jjp.6.18
  167. Park JH, Cha HY, Seo JJ, Hong JT, Han K, Oh KW. Anxiolytic-like effects of ginseng in the elevated plusmaze model: comparison of red ginseng and sun ginseng. Prog Neuropsychopharmacol Biol Psychiatry 2005;29:895-900. https://doi.org/10.1016/j.pnpbp.2005.04.016
  168. Yoo JH, Kwon HC, Kim YJ, Park JH, Yang HO. KG-135, enriched with selected ginsenosides, inhibits the proliferation of human prostate cancer cells in culture and inhibits xenograft growth in athymic mice. Cancer Lett 2010;289:99-110. https://doi.org/10.1016/j.canlet.2009.08.008
  169. Chen CF, Chiou WF, Zhang JT. Comparison of the pharmacological effects of Panax ginseng and Panax quinquefolium. Acta Pharmacol Sin 2008;29:1103-1108. https://doi.org/10.1111/j.1745-7254.2008.00868.x
  170. Hui H, Tang G, Go VL. Hypoglycemic herbs and their action mechanisms. Chin Med 2009;4:11. https://doi.org/10.1186/1749-8546-4-11
  171. Chung JW, Kim YJ, Lee SJ, Hahm KB. Korean Red Ginseng: Qualitative and Quantitative Benefi ts on Helicobacter pylori Infection. J Ginseng Res 2010;34:77-88. https://doi.org/10.5142/jgr.2010.34.2.077
  172. Ernst E. Panax ginseng: An Overview of the Clinical Evidence. J Ginseng Res 2010;34:259-263. https://doi.org/10.5142/jgr.2010.34.4.259

Cited by

  1. Evaluation of Four Different Analytical Tools to Determine the Regional Origin of Gastrodia elata and Rehmannia glutinosa on the Basis of Metabolomics Study vol.19, pp.5, 2014, https://doi.org/10.3390/molecules19056294
  2. Enzyme-modified Panax ginseng inhibits UVB-induced skin aging through the regulation of procollagen type I and MMP-1 expression vol.5, pp.2, 2014, https://doi.org/10.1039/C3FO60418G
  3. Bioavailability of Ginsenosides from White and Red Ginsengs in the Simulated Digestion Model vol.62, pp.41, 2014, https://doi.org/10.1021/jf500477n
  4. C.A. Meyer) genome and evidence for allotetraploidy vol.77, pp.6, 2014, https://doi.org/10.1111/tpj.12441
  5. Characterization of a Ginsenoside-Transforming β-glucosidase from Paenibacillus mucilaginosus and Its Application for Enhanced Production of Minor Ginsenoside F2 vol.9, pp.1, 2014, https://doi.org/10.1371/journal.pone.0085727
  6. Identification and Characterization of a Ginsenoside-Transforming β-Glucosidase from Pseudonocardia sp. Gsoil 1536 and Its Application for Enhanced Production of Minor Ginsenoside Rg2(S) vol.9, pp.6, 2014, https://doi.org/10.1371/journal.pone.0096914
  7. vol.122, pp.1, 2016, https://doi.org/10.1111/jam.13325
  8. Rg3-enriched ginseng extract ameliorates scopolamine-induced learning deficits in mice vol.16, pp.1, 2016, https://doi.org/10.1186/s12906-016-1050-z
  9. Two cases of Atopic Dermatitis with Gastrointestinal Disorders treated with Soshiho-tang vol.29, pp.3, 2016, https://doi.org/10.6114/jkood.2016.29.3.208
  10. Involvement of Connexin40 in the Protective Effects of Ginsenoside Rb1 Against Traumatic Brain Injury vol.36, pp.7, 2016, https://doi.org/10.1007/s10571-015-0299-y
  11. recoveries by integrated expanded bed adsorption chromatography vol.39, pp.15, 2016, https://doi.org/10.1002/jssc.201600269
  12. Ginseng on Nuclear Hormone Receptors vol.45, pp.06, 2017, https://doi.org/10.1142/S0192415X17500628
  13. vol.45, pp.3, 2017, https://doi.org/10.5941/MYCO.2017.45.3.220
  14. Daejeonia ginsenosidivorans gen. nov., sp. nov., a ginsenoside-transforming bacterium isolated from lake water vol.67, pp.8, 2017, https://doi.org/10.1099/ijsem.0.001994
  15. Complete genome sequencing of Arachidicoccus ginsenosidimutans sp. nov., and its application for production of minor ginsenosides by finding a novel ginsenoside-transforming β-glucosidase vol.7, pp.74, 2017, https://doi.org/10.1039/C7RA02612A
  16. for Anti-Wrinkle Therapy in Healthy Skin: A Single-Center, Randomized, Double-Blind, Placebo-Controlled Study vol.18, pp.5, 2015, https://doi.org/10.1089/rej.2015.1660
  17. Phytotherapy in the Management of Diabetes: A Review vol.23, pp.1, 2018, https://doi.org/10.3390/molecules23010105
  18. Single-Class Data Descriptors for Mapping Panax notoginseng through P-Learning vol.8, pp.9, 2018, https://doi.org/10.3390/app8091448
  19. State of antioxidant systems and ginsenoside contents in the leaves of Panax ginseng in a natural habitat and an artificial plantation vol.40, pp.7, 2018, https://doi.org/10.1007/s11738-018-2699-3
  20. Ginsenoside Rg3: Potential Molecular Targets and Therapeutic Indication in Metastatic Breast Cancer vol.6, pp.1, 2019, https://doi.org/10.3390/medicines6010017
  21. Ginseng: A Qualitative Review of Benefits for Palliative Clinicians pp.1938-2715, 2019, https://doi.org/10.1177/1049909118822704
  22. Ginseng for managing menopause symptoms: a systematic review of randomized clinical trials vol.37, pp.1, 2013, https://doi.org/10.5142/jgr.2013.37.30
  23. Protective effects of Korean red ginseng extract on cadmium-induced hepatic toxicity in rats vol.37, pp.1, 2012, https://doi.org/10.5142/jgr.2013.37.37
  24. Ginseng total saponin modulates podocyte p130Cas in diabetic condition vol.37, pp.1, 2013, https://doi.org/10.5142/jgr.2013.37.94
  25. Enzyme-processed Korean Red Ginseng extracts protects against skin damage induced by UVB irradiation in hairless mice vol.37, pp.4, 2013, https://doi.org/10.5142/jgr.2013.37.425
  26. Anti-fatigue Effects of 20(S)-Protopanaxadiol and 20(S)-Protopanaxatriol in Mice vol.38, pp.9, 2015, https://doi.org/10.1248/bpb.b15-00230
  27. Biological characteristics of Paenibacillus polymyxa GBR-1 involved in root rot of stored Korean ginseng vol.40, pp.4, 2012, https://doi.org/10.1016/j.jgr.2015.09.003
  28. Gram-Scale Production of Ginsenoside F1 Using a Recombinant Bacterial β-Glucosidase vol.27, pp.9, 2012, https://doi.org/10.4014/jmb.1703.03006
  29. Korean Red Ginseng (Panax ginseng) Potentiates the Inhibitory Actions of Testosterone on Obesity and Adipogenesis in High Fat Diet-Fed Castrated Mice vol.23, pp.3, 2012, https://doi.org/10.15616/bsl.2017.23.3.261
  30. A novel herbal formula, SGE, induces endoplasmic reticulum stress-mediated cancer cell death and alleviates cachexia symptoms induced by colon-26 adenocarcinoma vol.9, pp.23, 2012, https://doi.org/10.18632/oncotarget.24616
  31. Investigation of the Presence of 22 Organochlorine Pesticide Residues in Ginseng from Jilin Province, China vol.82, pp.9, 2012, https://doi.org/10.4315/0362-028x.jfp-18-490
  32. Antihypertensive effects of Korean wild simulated ginseng (Sanyangsam) extracts in spontaneously hypertensive rats vol.28, pp.5, 2012, https://doi.org/10.1007/s10068-019-00617-5
  33. Current Status and Problem-Solving Strategies for Ginseng Industry vol.25, pp.12, 2012, https://doi.org/10.1007/s11655-019-3046-2
  34. Effect of Panax ginseng on preventing acute respiratory tract infection : A protocol for systematic review and meta-analysis vol.99, pp.24, 2012, https://doi.org/10.1097/md.0000000000020690
  35. Transfer of Organochlorine Pesticide Residues during Household and Industrial Processing of Ginseng vol.2020, pp.None, 2012, https://doi.org/10.1155/2020/5946078
  36. Panax notoginseng saponins promotes cutaneous wound healing and suppresses scar formation in mice vol.19, pp.2, 2012, https://doi.org/10.1111/jocd.13042
  37. Effects of Ginseng Ingestion on Salivary Testosterone and DHEA Levels in Healthy Females: An Exploratory Study vol.12, pp.6, 2012, https://doi.org/10.3390/nu12061582
  38. Effect of Co-Administration of Panax ginseng and Brassica oleracea on Postmenopausal Osteoporosis in Ovariectomized Mice vol.12, pp.8, 2012, https://doi.org/10.3390/nu12082415
  39. Promising role of medicinal plants in the regulation and management of male erectile dysfunction vol.130, pp.None, 2012, https://doi.org/10.1016/j.biopha.2020.110555
  40. Integrative Analysis of Regulatory Module Reveals Associations of Microgravity with Dysfunctions of Multi-body Systems and Tumorigenesis vol.21, pp.20, 2020, https://doi.org/10.3390/ijms21207585
  41. MicroRNAs mediate the anti-tumor and protective effects of ginsenosides vol.72, pp.8, 2012, https://doi.org/10.1080/01635581.2019.1675722
  42. Panax ginseng Fruit Has Anti-Inflammatory Effect and Induces Osteogenic Differentiation by Regulating Nrf2/HO-1 Signaling Pathway in In Vitro and In Vivo Models of Periodontitis vol.9, pp.12, 2020, https://doi.org/10.3390/antiox9121221
  43. Biochemical and molecular characterization of enhanced growth of Panax ginseng C. A. Meyer treated with atmospheric pressure plasma vol.53, pp.49, 2020, https://doi.org/10.1088/1361-6463/abad61
  44. Evolution of the adaptogenic concept from traditional use to medical systems: Pharmacology of stress‐ and aging‐related diseases vol.41, pp.1, 2012, https://doi.org/10.1002/med.21743
  45. An LCI‐like protein APC2 protects ginseng root from Fusarium solani infection vol.130, pp.1, 2012, https://doi.org/10.1111/jam.14771
  46. Anticancer Activities of Ginsenosides, the Main Active Components of Ginseng vol.2021, pp.None, 2021, https://doi.org/10.1155/2021/8858006
  47. Understanding Traditional Chinese Medicine Therapeutics: An Overview of the Basics and Clinical Applications vol.9, pp.3, 2012, https://doi.org/10.3390/healthcare9030257
  48. Phnomibacter ginsenosidimutans gen. nov., sp. nov., a novel glycoside hydrolase positive bacterial strain with ginsenoside hydrolysing activity vol.71, pp.5, 2021, https://doi.org/10.1099/ijsem.0.004793
  49. Application of Identification and Evaluation Techniques for Ethnobotanical Medicinal Plant of Genus Panax: A Review vol.51, pp.4, 2012, https://doi.org/10.1080/10408347.2020.1736506
  50. Ginsenoside Rb3 Alleviates CSE-induced TROP2 Upregulation through p38 MAPK and NF-κB Pathways in Basal Cells vol.64, pp.6, 2012, https://doi.org/10.1165/rcmb.2020-0208oc
  51. Global Trends in Research on Wild-Simulated Ginseng: Quo Vadis? vol.12, pp.6, 2021, https://doi.org/10.3390/f12060664
  52. The role of a β-1,3-1,4-glucanase derived from Bacillus amyloliquefaciens FS6 in the protection of ginseng against Botrytis cinerea and Alternaria panax vol.164, pp.None, 2012, https://doi.org/10.1016/j.biocontrol.2021.104765
  53. The Analgesic Properties of Corydalis yanhusuo vol.26, pp.24, 2012, https://doi.org/10.3390/molecules26247498
  54. Effect of a combination of Korean red ginseng extract and probiotics on the prevention of atopic dermatitis in a murine model vol.283, pp.None, 2012, https://doi.org/10.1016/j.jep.2021.114687