The Chemistry of Secondary Products from Acanthopanax Species and their Pharmacological Activities

  • Shin, Kuk-Hyun (Natural Products Research Institute and College of Pharmacy, Seoul National University) ;
  • Lee, Sang-Hyun (Natural Products Research Institute and College of Pharmacy, Seoul National University)
  • Published : 2002.12.01

Abstract

The chemistry of secondary products from Acanthopanax species and their pharmacological activities were reviewed. A nitrogenous compound, a furan compound, a quinoid, benzoids, coumarins, phenylpropanoids, lignans, flavonoids, terpenoids, phytosterols, polyacetylenes, a pyrimidine, cyclitols, monosaccharides and an aliphatic alcohol have been isolated from Acanthopanax species and have been shown to have various levels of activities such as anti-bacterial, anti-cancer, anti-gout, anti-hepatitis, anti-hyperglycemic, anti-inflammatory, anti-leishmanicidic, anti-oxidant, anti-pyretic, anti-xanthine oxidase, choleretic, hemostatic, hypocholesterolemic, immunostimulatory and radioprotectant effects, etc.

Keywords

References

  1. Atm, J. S., Kwon, Y. S. and Kim, C. M., Anti-inflammatory constituents of PoIygonum bistorta. Kor. J. Pharmacogn. 30, 345-349 (1999)
  2. Akimoto, K., Kitagawa, Y., Akamatsu, T., Hirose, N., Sugano, M., Shimazu, S. and Yamada, H., Protective effect of sesamin against liver damage caused by alcohol or carbon tetrachloride in rodents. Ann. Nutr. Metabol. 37, 218-224 (1993) https://doi.org/10.1159/000177771
  3. Alvarez, M. A., Mar$\'i$a, A. O. M. and Saad, J. R., Diuretic activity of Fabiana patagonica in rats. Phytother. Res. 16, 71-73 (2002)
  4. Awad, A. B., Von Holtz, R., Cone, J. P., Fink, C. S. and Chen, Y. C., \beta-Sitosterol inhibits growth of HT-29 human cancer cells by activating the sphingomyelin cyclA. Anticancer Res. 18, 471-479 (1998) https://doi.org/10.1097/CAD.0b013e32801265eb
  5. Aziz, N. H., Farag, S. A., Mousa, L. A. and Abo-Zaid, M. A., Comparative anti-bacterial and anti-fungal effects of some phenolic compounds. Microbios 93, 43-54 (1998)
  6. Ban, H. S., Lee, S. Kim, Y. P., Yamaki, K., Shin, K. H. and Ohuchi, K., Inhibition of prostaglandin E_2 Production by taiwanin C isolated from the root of Acanthovanax chiisanensis and the mechanism of action. Biochem. Pharmacol. 64,1345-1354 (2002) https://doi.org/10.1016/S0006-2952(02)01348-5
  7. Bladt, S., Wagner, H., Woo, W. S. and Taiga-Wurzel., DC- und HPLC-Analyse von Eleutherococcus- bzw. Acanthopanax-Extskten und diese enthaltenden Phytopr$\"a$paraten. DAZ 130, 1499-1508 (1990)
  8. Bo, F., Matsumam, Y., Okada, Y., Qin, M., Xu, J-D. and Okuyama, T., Studies on constituents of Acanthopanax senticosus in China. Nat. Med. 52, 287 (1998)
  9. Borris, R. P., Cordell, G. A. and Farnsworth, N. R., Isofraxidin, a cytotoxic coumarin from Micrandra elata (Euphorbiaceae). J. Nat. Prod. 43, 641-643 (1980) https://doi.org/10.1021/np50011a020
  10. Brekhman, I.I. and Dardymov, I. V, Pharmacological investigation of glycosides from Ginseng and Eleutherococcus. J. Nat. Prod. 32, 46-51 (1969)
  11. Chan, W. S., Wen, P. C. and Chiang, H. C., Structure-activity relationship of caffeic acid analogues on xanthine oxidase inhibition. Anticancer Res. 15, 703-707 (1995)
  12. Chang, S. Y., Yook, C. S. and Nohara, T., Lupane-triterpene glycosides from leaves of Acanthopanax koreanum. Phytochemistry 50, 1369-1374 (1999) https://doi.org/10.1016/S0031-9422(98)00402-6
  13. Chang, S. Y., Yook, C. S. and Nohara, T., Two new lupanetriterpene glycosides from leaves of Acanthopanax koreanum. Chem. Pharm. BulI. 46, 163-165 (1998) https://doi.org/10.1248/cpb.46.163
  14. Chen, F. C., Lin, Y. M. and Lin, S., Constituents of three-leaved Acanthopanax. Phytochemistry 11, 1496-1497 (1972a)
  15. Chen, F. C., Lin, Y. M. and Yu, P. L., Constituents of Acanthopanaxtrifoliatus. J. Am. Chem. Soc. 94, 4367 (1972b) https://doi.org/10.1021/ja00767a070
  16. Chen, F. C., Lin, Y. M. and Yu, P. L., Constituents of Acanthopanax trifoliatus. Phytochemistry 12, 467 (1973) https://doi.org/10.1016/0031-9422(73)80043-3
  17. Chen, Z. W. and Ma, C. G., Effects of hyperin on free intracellular calcium in dissociated neonatal rat brain cells. Acta Pharmac. Sin. 20, 27-30 (1999)
  18. Cho, H. K., Ham, I. and Whang, W. K., Constituents and quantitative analysis from the leaves of Acanthopanax divaricatus forma nambunensis. Yakhak Hoeji 43, 294-299 (1999)
  19. Cho, H. R., Choi, D. H., Ko, B. K., Nam, C. W., Park, K. M., Lee, Y. J., Lee, S. G., Lee, J. S., Lee, K. A., Lee, A. A., Ju, S. A. and Kim, B. S., Cold preservation of rat cultured hepatocytes: the scoparone effect. Transplantation Proceedings 32, 2325-2327 (2000) https://doi.org/10.1016/S0041-1345(00)01684-5
  20. Chung, B. S. and Kim, Y. H., Studies on the constituents of Acanthopanax koreanum. Kor. J. Pharmacogn. 17, 62-66 (1986)
  21. Chung, J. Y. and Hahn, D. R., Constituents of Acanthopanax koreanum leaves. Yakhak Hoeji 35, 240-244 (1991)
  22. Danielak, R., Popowska, A. and Borkowski, B., The preparation of vegetable products containing isofraxidin, silibin and Glaucium alkaloids and evaluation of their choleretic action, Polish J. Pharmacol Pharm. 25, 271-283 (1973) https://doi.org/10.1111/j.2042-7158.1973.tb10642.x
  23. Delioiman, D., Cali$\c{s}$, I.,Ergun,F., Dog$\u{a}$n, B. S., Buharaliog$\u{l}$u, C. K. and Kanzik, I., Studies on the vascular effects of the fractions and phenolic compounds isolated from Viscum album ssp. album. J. EthnopharmacoI. 72, 323-329 (2000) https://doi.org/10.1016/S0378-8741(00)00251-8
  24. D$\'{i}$az Lanza, A. M., Abad Mart$\'{i}$nez, M. J., Fem$\'{a}$ndez Matellano, L., Recuero Carretero, C., Villaescusa Castillo, L., Silv$\'{a}$n Sen, A. M. and Bermejo Benito, P., Lignan and phenylpropanoid glycosides from Phillyrea latifolia and their in vitro anti-inflammatory activity. PIanta Med. 67, 219-223 (2001) https://doi.org/10.1055/s-2001-12004
  25. Fujikawa, T., Yamaguchi, A., Morita, I., Takeda, H. and Nishibe, S., Protective effects of Acanthopanax senticosus Harms from Hokkaido and its components on gastric ulcer in restrained cold water stressed rats. Biol. Pharm. BuII. 19, 1227-1230 (1996) https://doi.org/10.1248/bpb.19.1227
  26. Gupta, M. B., Nath, R., Srivastava, N., Kishor, K. and Bhargava, K. P., Anti-tnflammatory and anti-pyretic activities of \beta-sitosterol. Planta Med. 39, 157-163 (1980) https://doi.org/10.1055/s-2008-1074919
  27. Hahn, D. R., Kasai, R., Kim, J. H., Taniyasu, S. and Tanaka, O., A new glycosyl ester of 3,4-seco-triterpene from a Korean medicinal plant, Acanthopanax chiisanensis (Araliaceae). Chem. Pharm. BuIl. 32, 1244-1247 (1984) https://doi.org/10.1248/cpb.32.1244
  28. Hahn, D. R., Kim, C. J. and Kim, J. H., A study on the chemical constituents of Acanthopanax koreanum Nakai and its Pharmaco-biological activities. Yakhak Hoeji 29, 357-361 (1985)
  29. Haznedaroglu, M. Z., Karabay, N. U. and Zeybek, U., Anti-bacterial activity of Sclvia tomentosa essendal oil. Fitoterapia 72, 829-831 (2001) https://doi.org/10.1016/S0367-326X(01)00335-5
  30. Heinemann, T., Axtmann, G. and Von Bergmarm, K., Comparison of intestinal absorption of cholesterol with different plant sterols in man. Eur. J. Clin. Invest. 23, 827-831 (1993) https://doi.org/10.1111/j.1365-2362.1993.tb00737.x
  31. Heo, H-J., Cho, H-Y., Hong, B., Kim, H-K., Heo, T-R., Kim, E-K., Kim, S-K., Kim, C-J. and Shin, D-H., Ursolic acid of Origanum majorana L. reduces Abeta-induced oxidative injury. Molecules and CelIs 13, 5-11 (2002)
  32. Hibasami, H., Fujikawa, T., Takeda, H., Nishibe, S., Satoh, T., Fujisawa, T. and Nakashima, K., Induction of apoptosis by Acanthopanax senticosus HARMS and its component, sesamin in human stomach cancer KATO cells. Oncology Reports 7, 1213-1216 (2000)
  33. Hirata, F., Fujita, K., Ishikura, Y., Hosoda, K. and Ishikawa, H., Hypocholesterolemic effect of sesamin lignan in humans. AtheroscIerosis 122, 135-136 (1996) https://doi.org/10.1016/0021-9150(95)05769-2
  34. Hirose, N., Doi, F., Ueki, T., Akazawa, K., Chijiiwa, K., Sugano, M., Akimoto, K., Shimizu, S. and Yamada, H., Suppressive effect of sesamin against 7,12-dimethylbenz[a]anthracene-induced rat mammary carcinogenesis. Anticancer Res. 12, 1259-1265 (1992)
  35. Huh, K., Lee, S-I. and Park, J-M., Effect of scoparone on the hepatic microsomal UDP glucuronyltransferase activity in mice. Arch. Pharm. Res. 10, 165-168 (1987) https://doi.org/10.1007/BF02861907
  36. Huh K., Park, J-M., Shin, U-S. and Lee, S-I., Effect of scoparone on the hepatic sulfotransferase activity in mice. Arch. Pharm. Res. 13, 51-54 (1990) https://doi.org/10.1007/BF02857834
  37. Ide, T., Ashakumary, L, Takahashi, Y., Kushiro, M., Fukuda, N. and Sugano, M., Sesamin, a sesame lignan, decreases fatty acid synthesis in rat liver accompanying the down-regulation of sterol regulatory element binding protein-1, Biochim. Biophys. Acta 1534, 1-13 (2001) https://doi.org/10.1016/S1388-1981(01)00167-6
  38. Ishikura, N., Anthocyanin of Acanthopanax divaricatus. Phytochemistry 14, 1439 (1975) https://doi.org/10.1016/S0031-9422(00)98652-7
  39. Ivorra, M. D., D'Ocan, M. P., Paya, M. and Villar, A., Antihyperglycemic and insulin-releasing effect of b-sitosterol 3-\beta-D-glucoside and its aglycone, \beta-sitosterol. Arch. Int. Pharmacodyn. 296, 224-231 (1988)
  40. Jang, S. H., A study on the chemical constituent of Acanthopanax chiisanensis. Daehan Hwahak Hwoejee 14, 277-279 (1970)
  41. Janzowski, C., Glaab, V., Samimi, A., Schlatter, J. and Eisenbrand, G., 5-Hydroxymethylfurfural: assessment of mutagenicity, DNA-damaging potential and reactivity towards cellular glutathione. Food Chem. Toxicol. 38, 801-809 (2000) https://doi.org/10.1016/S0278-6915(00)00070-3
  42. Kang, H. S., Kim, Y. H., Lee, C. S., Lee, J. J., Choi, I. and Pyun, K. H., Suppression of interleukin-1 and tumor necrosis factor-\alpha production by acanthoic acid, (-)-pimara-9(11),15-diene-19-oic acid, and its anti-fibrotic effects in vivo. CelI. ImmunoI. 170, 212-221 (1996) https://doi.org/10.1006/cimm.1996.0154
  43. Kapil, A. and Sharma, S., Immunopotentiating compounds from Tinospora cordifolia. J. Ethnopharmacol. 58, 89-95 (1997) https://doi.org/10.1016/S0378-8741(97)00086-X
  44. Kasai, R., Matsumoto, K., Taniyasu, S., Tanaka, 0., Kim, J. H. and Hahn, D. R., 3,4-Seco-lupane triterpene glycosyl esters from a Korean medicinal plant, Acanthopanax chiisanensis (Araliaceae). Chem. Pharm. Bull. 34, 3284-3289 (1986) https://doi.org/10.1248/cpb.34.3284
  45. Kaul, A. and Khanduja, K. L., Polyphenols inhibit promotional phase of tumorigenesis: relevance of superoxide radicals. Nutr. Cancer 32, 81-85 (1998) https://doi.org/10.1080/01635589809514723
  46. Kim, C. J. and Hahn, D. R., The biological activity of a new glycoside, chiisanoside from Acanthopanax chiisanensis Nakai leaves. Yakhak Hoeji 24, 123-34 (1980)
  47. Kim, J. H. and Hahn, D. R., Studies on the chemical constituents of Acanthopanax chiisanensis Nakai roots. Arch. Pharm. Res. 4, 59-62 (1981) https://doi.org/10.1007/BF02856442
  48. Kim, Y. H. and Chung, B. S., Pimaradiene diterpenes from Acanthopanax koreanum. J. Nat. Prod. 51, 1080-1083 (1988) https://doi.org/10.1021/np50060a005
  49. Kim, Y. H., Chung, B. S. and Kim, H. J., Studies on the constituents of Acanthopanax koreanum Nakai. Kor. J. Pharmacogn. 16, 151-154 (1985)
  50. Kim, Y. H., Chung, B. S., Ko, Y. S. and Han, H. J., Studies on the chemical constituents of Acanthopanax koreanum. Arch. Pharm. Res. 11, 159-162 (1988) https://doi.org/10.1007/BF02857721
  51. Kim, Y. H., Kim, H. S., Lee, S. W,, Uramoto, M. and Lee, J. J., Kaurane derivatives from Acanthopanax koreanum. Phytochemistry 39, 449-451 (1995) https://doi.org/10.1016/0031-9422(95)99385-2
  52. Kim, Y. H., Ryu, J. H. and Chung, B. S., Diterpene glycoside from Acanthopanax koreanum. Kor. J. Pharmacogn. 21, 49-51 (1990)
  53. Kitajima, J., Takamori, Y. and Tanaka, Y., Studies on the constituents of Acanthopanax sciadophylloides Fr. et Sav. leaves. Yakugaku Zasshi 109, 188-191 (1989) https://doi.org/10.1248/yakushi1947.109.3_188
  54. Kohda, H., Tanaka, S. and Yamaoka, Y., Saponins from leaves of Acanthopanax hypoleucus Makino. Chem. Phann. Bull. 38, 3380-3383 (1990) https://doi.org/10.1248/cpb.38.3380
  55. Kutschabsky, L., Pfeiffer, D., Lischewski, M., Ty, P. D. and Adam, G., Molecular and crystal structure of a new 24-nor-thterpenoid carboxylic acid from Acanthopanax trifoliatus. Croatica Chemica Acta 58, 427-434 (1985)
  56. Kwak, Y-S., Ryu, S-H., Baek, B-K., Lee, J-T. and Ahn, B-Z., The anthelmintic principle of'O-Mae', the roasted fruits of Prunus mume, against Clonorchis sinensis. Yakhak Hoeji 29, 32-38 (1985)
  57. Lee, I., Lee, J., Lee, Y. H., and Leonard, J., Ursolic acid-induced changes in tumor growth, O_2 consumption, and tumor interstitial Quid pressure. Anticancer Res. 21, 2827-2833 (2001)
  58. Lee, M. W., Chung, J. Y., Kim, Y. C. and Hahn, D. R., The primary investigation on physiological activity from Araliaceous glycoside. Chung Ang J. Pharm. Sci. 1, 1-3 (1987)
  59. Lee, S., Ji, J., Shin, K. H. and Kim, B-K-., Sessiline, a new nitrogenous compound from the fruits of Acanthopanax sessiliflorus. Planta Med. 68, 936-939 (2002a) https://doi.org/10.1055/s-2002-34928
  60. Lee, S., Kim, B-K., Cho, S. H. and Shin, K. H., Phytochemical constituents from the fruits of Acanthopanax sessiIiflorus. Arch. Pharm. Res. 25, 280-284 (2002b) https://doi.org/10.1007/BF02976626
  61. Lee, S., Ban, H. S., Kim, Y. P., Kim, B-K., Cho, S. H., Ohuchi, K. and Shin, K.. H., Lignans from the roots of Acanthopanax chiisanensis having inhibitory activity on prostaglandin E_2 production. Phytother. Res (Accepted for publication, 2002c)
  62. Li, X-C., Barnes, D. L. and Khan, I. A., A new lignan glycoside from Eteutherococcus senticosus. Planta Med. 67, 776-778 (2001) https://doi.org/10.1055/s-2001-18352
  63. Lischewski, M., Ty, P. D., Kutschabsky, L., Pfeiffer, D., Phiet, H. V., Preiss, A., Sung, T. V. and Adam, G., Two 24-nor-triterpenoid carboxylic acids from Acanthopanax trifoliatus. Phytochemistry 24, 2355-2357 (1985) https://doi.org/10.1016/S0031-9422(00)83041-1
  64. Liu, C-L., Wang, J-M., Chu, C-Y., Cheng, M-T, and Tseng, T-H., In vivo protective effect of protocatechuic acid on tert-butyI hydroperoxide-induced rat hepatotoxicity. Food Chem. ToxicoI. 41, 635-641 (2002)
  65. Majester-Savornin, B., Elias, R., Diaz-Lanza, A. M., Balansard, G., Gasquet, M. and Delmas, F., Saponins of the ivy plant, Hedera helix, and their Leishmanicidic activity. Planta Med. 57, 260-262 (1991) https://doi.org/10.1055/s-2006-960086
  66. Matsumoto, K., Kasai, R., Kanamaru, F., K-ohda, H. and Tanaka, 0., 3,4-Seco-Lupane-type triterpene glycosyl esters from leaves of Acanthopanax divaricatus Seem. Chem. Pharm. Bull. 35, 413-415 (1987) https://doi.org/10.1248/cpb.35.413
  67. Medina, I., Tombo, I., Satu$\'e$-Gracia, M. T, German, J. B. and Frankel, A. N., Effects of natural phenolic compounds on the anti-oxidant activity of lactoferrin in liposomes and oil-in-water emulsions. J. Agric. Food Chem. 50, 2392-2399 (2002) https://doi.org/10.1021/jf011126y
  68. Meyer, R. P., Hagemeyer, C. E., Knoth, R., Kurz, G. and Volk, B., Oxidative hydrolysis of scoparone by cytochrome P_4_5_0 CYP_2C_2_9reveals a novel metabolite. Biochem. Biophys. Res. Comm. 285, 32-39 (2001) https://doi.org/10.1006/bbrc.2001.5111
  69. Miyakoshi, M., Ida, Y, Isoda, S. and Shoji, J., 3-epi-Oleanene type triterpene glycosyl esters from leaves of Acanthopanax spinosus. Phytochemistry 33, 891-895 (1993a) https://doi.org/10.1016/0031-9422(93)85298-6
  70. Miyakoshi, M., Ida, Y., Isoda, S. and Shoji, J., 3\alpha-Hydroxy-oleanene type triterpene glycosyl esters from leaves of Acanthopanax spinosus. Phytochemistry 34, 1599-1602 (1993b) https://doi.org/10.1016/S0031-9422(00)90853-7
  71. Miyakoshi, M., Isoda, S., Sato, H., Hira,i Y., Shoji, J. and Ida, Y., 3\alpha-Hydroxy-oleanene type triterpene glycosyl esters from leaves of Acanthopamx spinosus. Phytochemistry 46, 1255-1259 (1997a) https://doi.org/10.1016/S0031-9422(97)80022-2
  72. Miyakoshi, M., Shirasuna, K., Hirai, Y, Shingu, K., Isoda, S., Shoji, J., Ida, Y. and Shimizu, T., Thterpenoid saponins of Acanthopanax nipponicus leaves. J. Nat. Prod. 62, 445-448 (1999) https://doi.org/10.1021/np9804334
  73. Miyakoshi, M., Shirasuna, K., Sawada, H., Isoda, S., Ida, Y. and Shoji, J., Constituents of Acanthopanax divaricatus and A. sieboldianus roots. Nat. Med. 49, 218 (1995)
  74. Miyakoshi, M., Terajima, Y., Isoda, S., Hirai, Y. and Ida, Y., Constituents of leaves of Acanthopanax trichodon. Nat. Med. 51, 494 (1997b)
  75. Nakamura, Y., Torikai, K., and Ohigashi, H., Toxic dose of a simple phenolic anti-oxidant, Protocatechuic acid, attenuates the glutathione level in ICR mouse liver and kidney. J. Agiic. Food Chem. 49, 5674-5678 (2001) https://doi.org/10.1021/jf0106594
  76. Nakayama, T., Yamada, M., Osawa, T. and Kawakishi, S., Inhibitory effect of caffeic acid ethyl ester on H_2O_2-induced cytotoxicity and DNA single-stranded breaks in Chinese hamsters V79 cells. Biosci. Biotechnol. Biochem. 60, 316-318 (1996) https://doi.org/10.1271/bbb.60.316
  77. Nishibe, S., Kinoshita, H., Takeda, H. and Okano, G., Phenolic compounds from stem bark of Acanthopanax senticosus and their pharmacological effect in chronic swimming stressed rats. Chem. Pharm. Bull. 38, 1763-1765 (1990) https://doi.org/10.1248/cpb.38.1763
  78. Nishiyama, N., Kamegaya, T., Iwai, A., Saito, H., Sanada, S., Ida, Y. and Shoji, J., Effect of Eleutherococcus senticosus and its components on sex- and learning-behaviour and tyrosine hydroxylase activities of adrenal gland and hypothalamic regions in chronic stressed mice. Shoyakugaku Zasshi 39, 238-242 (1985)
  79. Nissanka, A. P., Karunaratne, V., Bandara, B. M., Kumar, V., Nakanishi, T., Nishi, M., Inada, A., Tillekeratne, L. M., Wijesundaia, D. S. and Gunatilaka, A. A., Anti-microbial alkaloids from Zanthoxylum tetraspermum and caudatum. Phytochemistry 56, 857-861 (2001) https://doi.org/10.1016/S0031-9422(00)00402-7
  80. Noguchi, T., Ikeda, K., Sasaki, Y., Yamamoto, J., Seki, J., Yamagata, K., Nara, Y., Hara, H., Kalnita, H. and Yamori, Y., Effects of vitamin E and sesamin on hypertension and cerebral thrombogenesis in stroke-prone spontaneously hypertensive rats. Hypertension Research: Official Journal of the Japanese Society of Hypertension 24, 735-742 (2001) https://doi.org/10.1291/hypres.24.735
  81. Nonaka, M., Yamashita, K., lizuka, Y., Namiki, M. and Sugano, M., Effects of dietary sesaminol and sesamin on eicosanoid production and immunoglobulin level in rats given ethanol. Biosci. Biotechnol. Biochem. 61, 836-839 (1997) https://doi.org/10.1271/bbb.61.836
  82. Novotn$\'{y}$, L., Vach$\'{a}$lkov$\'{a}$, A., and Biggs, D., Ursolic acid: an antitumorigenic and chemopreventive activity, minireview. Neoplasma 48, 241-246 (2001)
  83. Oh, O. J., Chang, S. Y., Kim, T. H., Yang, K. S., Yook, C. S., Park, S. Y. and Nohara, T., Constituents of Acanthopanax divaricatus var. aIbeofructus. Nat. Med. 54, 29-32 (2000)
  84. Park, S. Y., Chang, S. Y., Oh, 0. J., Yook, C. S. and Nohara, T., nor-Oleanene type triterpene glycosides from the leaves of Acanthopanax japonicus. Phytochemistry 59, 379-384 (2002) https://doi.org/10.1016/S0031-9422(01)00436-8
  85. Park, S. Y., Chang, S. Y., Yook. C. S. and Nohara, T., New 3,4-seco-lupane-type triterpene glycosides from Acanthopanax senticosus forma inermis. J. Nat. Prod. 63, 1630-1633 (2000a) https://doi.org/10.1021/np000277c
  86. Park, S. Y., Chang, S. Y., Yook, C. S. and Nohara, T, Tnterpene glycosides from leaves of Acanthopanax senticosus forma inermis. Nat. Med. 54, 43 (2000b)
  87. Park, S. Y., Yook, C. S. and Nohara, T., A novel 3,4-seco-migrated-lupane glycoside with a seven-membered B-ring from Acanthopanax divaricatus var. sachunensis. tetrahedron Lett. 42, 2825-2828 (2001) https://doi.org/10.1016/S0040-4039(01)00287-8
  88. Ro, H. S., Lee, S. Y. and Han, B. H., Studies on the lignan glycoside of Acanthopanax Cortex. J. Pharm. Soc. Kor. 21, 81-86 (1977)
  89. Ruijim, Z., Jinkang, Q., Gnanghua, Y., Baozhen, W. and Xiulan, W., Medicinal protection with Chinese herb-compound against radiation damage. Avaition Space Environ. Med. 61, 729-731 (1990)
  90. Sawada H., Miyakoshi, M., Isoda, S., Ida, Y. and Shoji, J., Saponins from leaves of Acanthopanax sieboldianus. Phytochemistry 34, 1117-1121 (1993) https://doi.org/10.1016/S0031-9422(00)90727-1
  91. Segiet-K-ujawa, E. and Kaloga, M., Triterpenoid saponins of Eleutherococcus senticosus roots. J. Nat. Prod. 54, 1044-1048 (1991) https://doi.org/10.1021/np50076a018
  92. Shimizu, M., Zenko, Y., Tanaka, R., Matsuzawa, T., and Mohta, N. Studies on aldose reductase inhibitors from natural products. V. Active components of Hachimi-jio-gan (Kampo medicine). Chem. Pharm. BulI. 41, 1469-1471 (1993) https://doi.org/10.1248/cpb.41.1469
  93. Shao, C-J., Kasai, R., Xu, J-D. and Tanaka, O., Saponins from leaves of Acanthopanax senticosus HARMS., Ciwujia: Structures of Ciwujianosides B, C_1, C_2, C_3, C_4, D_1, D_2 and E. Chem. Pharm. Bull. 36, 601-608 (1988) https://doi.org/10.1248/cpb.36.601
  94. Shao C-J., K-asai, R., Xu, J-D. and Tanaka, O., Saponins from leaves of Acanthopanax senticosus HARMS., Ciwujia. II. Structures of Ciwujianosides A_1, A_2, A_3, A_4and D_3. Chem. Pharm. Bull. 37, 42-45 (1989) https://doi.org/10.1248/cpb.37.42
  95. Shin, A. T., Kim, C. J. and Yook, C. S., Studies on the chemical constituents of Acanthopanax fruits. Bull. K. H. Pharm. Sci. 20, 63-73 (1992)
  96. Shirasuna, K., Miyakoshi, M., Mimoto, S., Isoda, S., Satoh, Y, Hirai, Y., Ida, Y. and Shoji, J., Lupane triterpenoid glycosyl esters from leaves of Acanthopanax divaricatus. Phytochemistry 45, 579-584 (1997) https://doi.org/10.1016/S0031-9422(97)00017-4
  97. Sirato-Yasumoto, S., Katsuta, M., Okuyama, Y., Takahashi, Y. and Ide T., Effect of sesame seeds rich in sesamm and sesamolin on fatty acid oxidation in rat liver. J. Agric. Food Chem. 49, 2647-2651 (2001) https://doi.org/10.1021/jf001362t
  98. Slacanin, I., Marston, A., Hostettmann, K., Gu$\'e$don, D. and Abbe, P., The isolation of Eleutherococcus senticosus constituents by centrifugal partition chromatography and their quantitative determination by high performance liquid chromatography. Phytochem. Anal. 2, 137-142 (1991) https://doi.org/10.1002/pca.2800020310
  99. Soliman, K. F. A. and Mazzio, A. A., In vitro attenuation of nitric acid production in C_6 astrocyte cell culture by various dietary compounds. Proc. Soc. Exp. Biol. Med. 218, 390-397 (1998)
  100. Srivastava, S., Gupta, M. M., Prajapati, V., Tripathi, A. K., and Kumar, S., Sesamin a potent anti-feedant principle from Piper mullesua. Phytother. Res. 15, 70-72 (2001) https://doi.org/10.1002/1099-1573(200102)15:1<70::AID-PTR671>3.0.CO;2-N
  101. Sugiyama, M. and Seki, J., In viva application of lipoproteins as drug carriers: pharmacological evaluation of Sterylgiucoside-lipoprotein complexes. Targ. Diagn. Ther. 5, 315-350 (1991)
  102. Taherzadeh, M. J., Gustafsson, L., Niklasson, C. and Lid$\'e$n, G., Physiological effects of 5-hydroxymethylfurfural on Saccharomyces cerevisiae. AppI. Microbiol. Biotechnol. 53, 701-708 (2000) https://doi.org/10.1007/s002530000328
  103. Taira, J., Ikemoto, T., Yoneya, T., Hagi, A., Murakami, A. and Makino, K., Essential oil phenyl propanoids: useful as OH scavengers? Free Rad. Res. Commun. 16, 197-204(1992) https://doi.org/10.3109/10715769209049172
  104. Takasugi, N., Mohguchi, T., Fuwa, T., Sanada, S., Ida, Y, Shoji, J. and Saito, H., Effect of Eleutherococcus senticosus and its components on rectal temperature, body and grip tones, motor coordination, and exploratory and spontaneous movements in acute stressed mice. Shoyakugaku Zasshi 39, 232-237 (1985)
  105. Ty, P. D., Lischewski, M., Phiet, H. V, Preiss, A., Nguyen, P. V. and Adam, G., 3\alpha,11\alpha-Dihydroxy-23-oxo-lup-20(29)-en-28-oic acid from Acanthopanax trifoliatus. Phytochemistry 24, 867-869 (1985) https://doi.org/10.1016/S0031-9422(00)84914-6
  106. Ty, P. D., Lischewski, M., Phiet, H. V., Preiss, A., Sung, T. V., Schmidt, J. and Adam, G., Two triterpenoid carboxylic acids from Acanthopanax trifoliatus. Phytochemistry 23, 2889-2891 (1984) https://doi.org/10.1016/0031-9422(84)83035-6
  107. Umeda-Sawada, R., Ogawa, M., Nakamura, M. and Igarashi, O., Effect of sesamin on mitochodhal and peroxisomal \beta-oxidadon of arachidonic and eicosapentaenoic acids in rat liver. Lipids 36, 483-489 (2001) https://doi.org/10.1007/s11745-001-0747-z
  108. Umeyama, A., Shoji, N., Takei, M., Endo, K. and Arihara, S., Ciwujianosides C_1 and D_1: Powerful inhibitors of histamine release induced by anti-immunoglobulin E from rat peritoneal mast dells. J. Pharm. Sci. 81, 661-662 (1992) https://doi.org/10.1002/jps.2600810714
  109. Wagner, H., Heur, Y. H., Obermeier, A., Tittel, G. and Bladt, S., Die DC- und HPLC-Analyse der Eleutherococcus Droge. Planta Med. 44, 193-8 (1982) https://doi.org/10.1055/s-2007-971445
  110. Wagner, H. and Wurmb$\"o$ck, A., Chemie, Pharmakologie und D$\"u$nnschichtchromatographie der Ginsengund EIeutherococcus-Droge. DAZ 117, 743-748 (1977)
  111. Wielgorskaya, T. and Takhtajan, A., Dictionary of generic names of seed plants, Columbia University Press, NY (1995)
  112. Yasue, M., Kato, Y., Lin, Y. M. and Sakakibara, J., Studies on the constituents of Acanthopanax sciadophylloides Franch. et Sav. I. Isolation of cyclitols and flavonoid glycosides. Structure of antoside. Yakugaku Zasshi 88, 738-741 (1968) https://doi.org/10.1248/yakushi1947.88.6_738
  113. Yasue, M., Kato, Y., Lin, Y. M. and Sakakibara, J., Studies on the constituents of Acanthopanax sciadophylloides Franch. et Sav. 2. On the flavonoid and terpenoid constituents, Coincidence of hirsutrin and isoquercitrin. Yakugaku Zasshi 89, 872-876 (1969) https://doi.org/10.1248/yakushi1947.89.6_872
  114. Yasue, M., Lin, Y. M. and Sakakibara, J., Studies on the constituents of Acanthopanax sciadophylloides Franch. et Sav. III. Yakugaku Zasshi 90, 341-343 (1970) https://doi.org/10.1248/yakushi1947.90.3_341
  115. Yat, P. N., Arnason, J. T. and Awang, D. V. C., An improved extraction procedure for the rapid, quantitative high-performance liquid chromatographic estimation of the main eleutherosides (B and E) in Eleutherococcus senticosus (Eleuthero). Phytochem. Anal. 9, 291-295 (1998) https://doi.org/10.1002/(SICI)1099-1565(199811/12)9:6<291::AID-PCA417>3.0.CO;2-K
  116. Yook, C. S., Chang, S. Y., Lai, J. H., Ko, S. K., Jeong, J. H. and Nohara, T., Lupane-glycoside of Acanthopanax trifoliatus forma tristigmatis leaves. Arch. Pharm. Res. 22, 629-632 (1999) https://doi.org/10.1007/BF02975337
  117. Yook, C. S., Kim, I. H., Hahn, D. R., Nohara, T. and Chang, S. Y., A lupane-triterpene glycoside from leaves of two Acanthopanax. Phytochemistry 49, 839-843 (1998) https://doi.org/10.1016/S0031-9422(97)00846-7
  118. Yook, C. S., Kim, S. C., Kim, C. J. and Han, D. R., Phytoch$\u{e}$mical studies on the barks of Acanthopanax senticosus forma inermis. Yakhak Hoeji 35, 147-153 (1991)
  119. Yook, C. S., Lee, D. H., Seo, Y. K. and Ryu, K. S., Study on the constituents m the root bark of Acanthopanax sessiliflorum Seemann (II). Kor. J. Pharmacogn. 8, 31-34 (1977)
  120. Yook, C. S., Rho, Y. S., Seo, S. H., Leem, J. Y. and Han, D. R., Chemical components of Acanthopanax divaricatus and anticancer effect in leaves. Yakhak Hoeji 40, 251-261 (1996)
  121. You, H. J., Choi, C. Y., Kim, J. Y., Park, S. J., Hahm, K. S., and Jeong, H. G., Ursolic acid enhances nitric oxide and tumor necrosis factor-\alpha production via nuclear factor-\kappaB activation in the resting macrophages. FEBS Lett. 509, 156-160 (2001) https://doi.org/10.1016/S0014-5793(01)03161-1
  122. Yun-Choi, H. S., Kim, J. H,, Kim, S. O. and Lee, J. R., Platelet anti-aggregating plant materials. Kor. J. Pharmacogn. 17, 161-167 (1986)
  123. Yun-Choi, H. S., Kim, J. H. and Lee, J. R., Potential inhibitors of platelet aggregation from plant sources,.J. Nat. Prod. 50, 1059-1064 (1987) https://doi.org/10.1021/np50054a008
  124. Zhao, W. M., Qin, G. W., Xu, R. S., Li, X. Y, Liu, J. S., Wang, Y. and Feng, M., Constituents from the roots of Acanthopanax setchuenensis. Fitoterapia 70, 529-531 (1999) https://doi.org/10.1016/S0367-326X(99)00078-7