DOI QR코드

DOI QR Code

Effects of Panax ginseng in Neurodegenerative Diseases

  • Cho, Ik-Hyun (Department of Anatomy, College of Oriental Medicine and Institute of Oriental Medicine, Kyung Hee University)
  • Received : 2012.03.02
  • Accepted : 2012.05.21
  • Published : 2012.10.15

Abstract

Ginseng, the root of the Panax ginseng, has been a popular and widely-used traditional herbal medicine in Korea, China, and Japan for thousands of years. Now it has become popular as a functional health food and is used globally as a natural medicine. Evidence is accumulating in the literature on the physiological and pharmacological effects of P. ginseng on neurodegenerative diseases. Possible ginseng- or ginsenosides-mediated neuroprotective mechanisms mainly involve maintaining homeostasis, and anti-inflammatory, anti-oxidant, anti-apoptotic, and immune-stimulatory activities. This review considers publications dealing with the various actions of P. ginseng that are indicative of possible neurotherapeutic efficacies in neurodegenerative diseases and neurological disorders such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis and multiple sclerosis.

Keywords

References

  1. Baker JT, Borris RP, Carte B, Cordell GA, Soejarto DD, Cragg GM, Gupta MP, Iwu MM, Madulid DR, Tyler VE. Natural product drug discovery and development: new perspectives on international collaboration. J Nat Prod 1995;58:1325-1357. https://doi.org/10.1021/np50123a003
  2. 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
  3. Kim MH, Lee YC, Choi SY, Cho CW, Rho J, Lee KW. The changes of ginsenoside patterns in red ginseng processed by organic acid impregnation pretreatment. J Ginseng Res 2011;35:497-503. https://doi.org/10.5142/jgr.2011.35.4.497
  4. Yuan CS, Wang CZ, Wicks SM, Qi LW. Chemical and pharmacological studies of saponins with a focus on American ginseng. J Ginseng Res 2010;34:160-167. https://doi.org/10.5142/jgr.2010.34.3.160
  5. 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
  6. Attele AS, Wu JA, Yuan CS. Ginseng pharmacology:multiple constituents and multiple actions. Biochem Pharmacol 1999;58:1685-1693. https://doi.org/10.1016/S0006-2952(99)00212-9
  7. Tsang D, Yeung HW, Tso WW, Peck H. Ginseng saponins: influence on neurotransmitter uptake in rat brain synaptosomes. Planta Med 1985;51:221-224. https://doi.org/10.1055/s-2007-969463
  8. Rausch WD, Liu S, Gille G, Radad K. Neuroprotective effects of ginsenosides. Acta Neurobiol Exp (Wars) 2006;66:369-375.
  9. Nah SY, Kim DH, Rhim H. Ginsenosides: are any of them candidates for drugs acting on the central nervous system? CNS Drug Rev 2007;13:381-404.
  10. Radad K, Moldzio R, Rausch WD. Ginsenosides and their CNS targets. CNS Neurosci Ther 2011;17:761-768. https://doi.org/10.1111/j.1755-5949.2010.00208.x
  11. Djaldetti R, Lev N, Melamed E. Neuroprotection in progressive brain disorders. Isr Med Assoc J 2003;5:576-580.
  12. Gerlach M, Double KL, Youdim MB, Riederer P. Strategies for the protection of dopaminergic neurons against neurotoxicity. Neurotox Res 2000;2:99-114. https://doi.org/10.1007/BF03033788
  13. Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging 2003;24:197-211. https://doi.org/10.1016/S0197-4580(02)00065-9
  14. Schapira AH. Molecular and clinical pathways to neuroprotection of dopaminergic drugs in Parkinson disease. Neurology 2009;72(7 Suppl):S44-S50. https://doi.org/10.1212/WNL.0b013e3181990438
  15. Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying infl ammation in neurodegeneration. Cell 2010;140:918-934. https://doi.org/10.1016/j.cell.2010.02.016
  16. Hu S, Han R, Mak S, Han Y. Protection against 1-methyl- 4-phenylpyridinium ion (MPP+)-induced apoptosis by water extract of ginseng (Panax ginseng C.A. Meyer) in SH-SY5Y cells. J Ethnopharmacol 2011;135:34-42. https://doi.org/10.1016/j.jep.2011.02.017
  17. Van Kampen J, Robertson H, Hagg T, Drobitch R. Neuroprotective actions of the ginseng extract G115 in two rodent models of Parkinson's disease. Exp Neurol 2003;184:521-529. https://doi.org/10.1016/j.expneurol.2003.08.002
  18. Wang J, Xu HM, Yang HD, Du XX, Jiang H, Xie JX. Rg1 reduces nigral iron levels of MPTP-treated C57BL6 mice by regulating certain iron transport proteins. Neurochem Int 2009;54:43-48. https://doi.org/10.1016/j.neuint.2008.10.003
  19. Xu H, Jiang H, Wang J, Xie J. Rg1 protects the MPP+-treated MES23.5 cells via attenuating DMT1 up-regulation and cellular iron uptake. Neuropharmacology 2010;58:488-494. https://doi.org/10.1016/j.neuropharm.2009.09.002
  20. Xu H, Jiang H, Wang J, Xie J. Rg1 protects iron-induced neurotoxicity through antioxidant and iron regulatory proteins in 6-OHDA-treated MES23.5 cells. J Cell Biochem 2010;111:1537-1545. https://doi.org/10.1002/jcb.22885
  21. Rouault TA, Cooperman S. Brain iron metabolism. Semin Pediatr Neurol 2006;13:142-148. https://doi.org/10.1016/j.spen.2006.08.002
  22. Lee DW, Andersen JK. Iron elevations in the aging Parkinsonian brain: a consequence of impaired iron homeostasis? J Neurochem 2010;112:332-339. https://doi.org/10.1111/j.1471-4159.2009.06470.x
  23. Chen XC, Zhou YC, Chen Y, Zhu YG, Fang F, Chen LM. Ginsenoside Rg1 reduces MPTP-induced substantia nigra neuron loss by suppressing oxidative stress. Acta Pharmacol Sin 2005;26:56-62. https://doi.org/10.1111/j.1745-7254.2005.00019.x
  24. Shi C, Zhang YX, Zhang ZF. Effect of phosphorylated- ERK1/2 on inducible nitric oxide synthase expression in the substantia nigra of mice with MPTP-induced Parkinson disease. Nan Fang Yi Ke Da Xue Xue Bao 2009;29:60-63.
  25. Chen XC, Zhu YG, Zhu LA, Huang C, Chen Y, Chen LM, Fang F, Zhou YC, Zhao CH. Ginsenoside Rg1 attenuates dopamine-induced apoptosis in PC12 cells by suppressing oxidative stress. Eur J Pharmacol 2003;473:1-7. https://doi.org/10.1016/S0014-2999(03)01945-9
  26. Xu L, Chen WF, Wong MS. Ginsenoside Rg1 protects dopaminergic neurons in a rat model of Parkinson's disease through the IGF-I receptor signalling pathway. Br J Pharmacol 2009;158:738-748. https://doi.org/10.1111/j.1476-5381.2009.00361.x
  27. Wang Q, Zheng H, Zhang ZF, Zhang YX. Ginsenoside Rg1 modulates COX-2 expression in the substantia nigra of mice with MPTP-induced Parkinson disease through the P38 signaling pathway. Nan Fang Yi Ke Da Xue Xue Bao 2008;28:1594-1598.
  28. Leung KW, Yung KK, Mak NK, Chan YS, Fan TP, Wong RN. Neuroprotective effects of ginsenoside-Rg1 in primary nigral neurons against rotenone toxicity. Neuropharmacology 2007;52:827-835. https://doi.org/10.1016/j.neuropharm.2006.10.001
  29. Liu Q, Kou JP, Yu BY. Ginsenoside Rg1 protects against hydrogen peroxide-induced cell death in PC12 cells via inhibiting NF-${\kappa}B$ activation. Neurochem Int 2011;58:119-125. https://doi.org/10.1016/j.neuint.2010.11.004
  30. Xu BB, Liu CQ, Gao X, Zhang WQ, Wang SW, Cao YL. Possible mechanisms of the protection of ginsenoside Re against MPTP-induced apoptosis in substantia nigra neurons of Parkinson's disease mouse model. J Asian Nat Prod Res 2005;7:215-224. https://doi.org/10.1080/10286020410001690172
  31. Lin WM, Zhang YM, Moldzio R, Rausch WD. Ginsenoside Rd attenuates neuroinfl ammation of dopaminergic cells in culture. J Neural Transm Suppl 2007;(72):105-112.
  32. Selkoe DJ, Schenk D. Alzheimer's disease: molecular understanding predicts amyloid-based therapeutics. Annu Rev Pharmacol Toxicol 2003;43:545-584. https://doi.org/10.1146/annurev.pharmtox.43.100901.140248
  33. Tanzi RE, Bertram L. Alzheimer's disease: the latest suspect. Nature 2008;454:706-708. https://doi.org/10.1038/454706a
  34. Bazan NG, Palacios-Pelaez R, Lukiw WJ. Hypoxia signaling to genes: signifi cance in Alzheimer's disease. Mol Neurobiol 2002;26:283-298. https://doi.org/10.1385/MN:26:2-3:283
  35. Frank B, Gupta S. A review of antioxidants and Alzheimer's disease. Ann Clin Psychiatry 2005;17:269-286. https://doi.org/10.1080/10401230500296428
  36. Finch CE, Morgan TE. Systemic infl ammation, infection, ApoE alleles, and Alzheimer disease: a position paper. Curr Alzheimer Res 2007;4:185-189. https://doi.org/10.2174/156720507780362254
  37. Ghosh AK, Hong L, Tang J. Beta-secretase as a therapeutic target for inhibitor drugs. Curr Med Chem 2002;9:1135-1144. https://doi.org/10.2174/0929867023370149
  38. Hills ID, Vacca JP. Progress toward a practical BACE-1 inhibitor. Curr Opin Drug Discov Devel 2007;10:383-391.
  39. Imbimbo BP. Therapeutic potential of gamma-secretase inhibitors and modulators. Curr Top Med Chem 2008;8:54-61. https://doi.org/10.2174/156802608783334015
  40. Heo JH, Lee ST, Chu K, Oh MJ, Park HJ, Shim JY, Kim M. An open-label trial of Korean red ginseng as an adjuvant treatment for cognitive impairment in patients with Alzheimer's disease. Eur J Neurol 2008;15:865-868. https://doi.org/10.1111/j.1468-1331.2008.02157.x
  41. Lee ST, Chu K, Sim JY, Heo JH, Kim M. Panax ginseng enhances cognitive performance in Alzheimer disease. Alzheimer Dis Assoc Disord 2008;22:222-226. https://doi.org/10.1097/WAD.0b013e31816c92e6
  42. Zhao H, Li Q, Zhang Z, Pei X, Wang J, Li Y. Long-term ginsenoside consumption prevents memory loss in aged SAMP8 mice by decreasing oxidative stress and upregulating the plasticity-related proteins in hippocampus. Brain Res 2009;1256:111-122. https://doi.org/10.1016/j.brainres.2008.12.031
  43. Tu LH, Ma J, Liu HP, Wang RR, Luo J. The neuroprotective effects of ginsenosides on calcineurin activity and tau phosphorylation in SY5Y cells. Cell Mol Neurobiol 2009; 29:1257-1264. https://doi.org/10.1007/s10571-009-9421-3
  44. Qian YH, Han H, Hu XD, Shi LL. Protective effect of ginsenoside Rb1 on beta-amyloid protein(1-42)-induced neurotoxicity in cortical neurons. Neurol Res 2009;31:663-667. https://doi.org/10.1179/174313209X385572
  45. Xie X, Wang HT, Li CL, Gao XH, Ding JL, Zhao HH, Lu YL. Ginsenoside Rb1 protects PC12 cells against $\beta$-amyloid-induced cell injury. Mol Med Report 2010;3:635-639.
  46. Zhao R, Zhang Z, Song Y, Wang D, Qi J, Wen S. Implication of phosphatidylinositol-3 kinase/Akt/glycogen synthase kinase-$3{\beta}$ pathway in ginsenoside Rb1's attenuation of beta-amyloid-induced neurotoxicity and tau phosphorylation. J Ethnopharmacol 2011;133:1109-1116. https://doi.org/10.1016/j.jep.2010.11.054
  47. Chen X, Huang T, Zhang J, Song J, Chen L, Zhu Y. Involvement of calpain and p25 of CDK5 pathway in ginsenoside Rb1's attenuation of beta-amyloid peptide25-35-induced tau hyperphosphorylation in cortical neurons. Brain Res 2008;1200:99-106. https://doi.org/10.1016/j.brainres.2007.12.029
  48. Wang Y, Liu J, Zhang Z, Bi P, Qi Z, Zhang C. Anti-neuro-inflammation effect of ginsenoside Rbl in a rat model of Alzheimer disease. Neurosci Lett 2011;487:70-72. https://doi.org/10.1016/j.neulet.2010.09.076
  49. Li X, Zhang X, Yuan H, Quan Q. Experimental research on effect of gensenoside Rg1 on expressions of P-Tau and caspase-3 in brain slices from AD model rats. Zhongguo Zhong Yao Za Zhi 2010;35:369-372.
  50. Wei CB, Jia JP, Liang P, Guan YQ. Ginsenoside-Rg1 inhibits cell apoptosis induced by beta amyloid. Zhonghua Yi Xue Za Zhi 2008;88:1763-1766.
  51. Wang XY, Chen J, Zhang JT. Effect of ginsenoside Rg1 on learning and memory impairment induced by betaamyloid peptide(25-35) and its mechanism of action. Yao Xue Xue Bao 2001;36:1-4.
  52. Li WZ, Li WP, Zhang W, Yin YY, Sun XX, Zhou SS, Xu XQ, Tao CR. Protective effect of extract of Astragalus on learning and memory impairments and neurons' apoptosis induced by glucocorticoids in 12-month-old male mice. Anat Rec (Hoboken) 2011;294:1003-1014. https://doi.org/10.1002/ar.21386
  53. Shi YQ, Huang TW, Chen LM, Pan XD, Zhang J, Zhu YG, Chen XC. Ginsenoside Rg1 attenuates amyloid-beta content, regulates PKA/CREB activity, and improves cognitive performance in SAMP8 mice. J Alzheimers Dis 2010;19:977-989. https://doi.org/10.3233/JAD-2010-1296
  54. Chen F, Eckman EA, Eckman CB. Reductions in levels of the Alzheimer's amyloid beta peptide after oral administration of ginsenosides. FASEB J 2006;20:1269-1271. https://doi.org/10.1096/fj.05-5530fje
  55. Wang YH, Du GH. Ginsenoside Rg1 inhibits betasecretase activity in vitro and protects against Abetainduced cytotoxicity in PC12 cells. J Asian Nat Prod Res 2009;11:604-612. https://doi.org/10.1080/10286020902843152
  56. Li X, Liu Y, Zhang X, Yuan H, Quan Q. Effect of ginsenoside Rg1 on expressions of phosphory protein tau and N-methyl-D-aspartate receptor subunits NR1 and NR2B in rat brain slice model of Alzheimer's disease. Zhongguo Zhong Yao Za Zhi 2010;35:3339-3343.
  57. Li X, Liu Y, Yuan HF, Quan QK. Effects of gensenoside Rg1 on tau protein phosphorylation induced by okadaic acid in rat brain slices. Zhong Xi Yi Jie He Xue Bao 2010;8:955-960. https://doi.org/10.3736/jcim20101007
  58. Gong YS, Zhang JT. Effect of 17-beta-estradiol and ginsenoside Rg1 on reactive microglia induced by betaamyloid peptides. J Asian Nat Prod Res 1999;1:153-161. https://doi.org/10.1080/10286029908039859
  59. Joo SS, Won TJ, Lee DI. Reciprocal activity of ginsenosides in the production of proinflammatory repertoire, and their potential roles in neuroprotection in vivo. Planta Med 2005;71:476-481. https://doi.org/10.1055/s-2005-864145
  60. Li N, Liu B, Dluzen DE, Jin Y. Protective effects of ginsenoside Rg2 against glutamate-induced neurotoxicity in PC12 cells. J Ethnopharmacol 2007;111:458-463. https://doi.org/10.1016/j.jep.2006.12.015
  61. Zhang G, Liu A, Zhou Y, San X, Jin T, Jin Y. Panax ginseng ginsenoside-Rg2 protects memory impairment via anti-apoptosis in a rat model with vascular dementia. J Ethnopharmacol 2008;115:441-448. https://doi.org/10.1016/j.jep.2007.10.026
  62. Heneka MT, O'Banion MK. Inflammatory processes in Alzheimer's disease. J Neuroimmunol 2007;184:69-91. https://doi.org/10.1016/j.jneuroim.2006.11.017
  63. Joo SS, Lee DI. Potential effects of microglial activation induced by ginsenoside Rg3 in rat primary culture: enhancement of type A Macrophage Scavenger Receptor expression. Arch Pharm Res 2005;28:1164-1169. https://doi.org/10.1007/BF02972981
  64. Yang L, Hao J, Zhang J, Xia W, Dong X, Hu X, Kong F, Cui X. Ginsenoside Rg3 promotes beta-amyloid peptide degradation by enhancing gene expression of neprilysin. J Pharm Pharmacol 2009;61:375-380. https://doi.org/10.1211/jpp.61.03.0013
  65. Shieh PC, Tsao CW, Li JS, Wu HT, Wen YJ, Kou DH, Cheng JT. Role of pituitary adenylate cyclase-activating polypeptide (PACAP) in the action of ginsenoside Rh2 against beta-amyloid-induced inhibition of rat brain astrocytes. Neurosci Lett 2008;434:1-5. https://doi.org/10.1016/j.neulet.2007.12.032
  66. Hwang SH, Shin TJ, Choi SH, Cho HJ, Lee BH, Pyo MK, Lee JH, Kang J, Kim HJ, Park CW et al. Gintonin, newly identified compounds from ginseng, is novel lysophosphatidic acids-protein complexes and activates G protein-coupled lysophosphatidic acid receptors with high affi nity. Mol Cells 2012;33:151-162. https://doi.org/10.1007/s10059-012-2216-z
  67. Hwang SH, Shin EJ, Shin TJ, Lee BH, Choi SH, Kang J, Kim HJ, Kwon SH, Jang CG, Lee JH et al. Gintonin, a ginseng-derived lysophosphatidic acid receptor ligand, attenuates Alzheimer's disease-related neuropathies: involvement of non-amyloidogenic processing. J Alzheimers Dis 2012; Epub ahead of print.
  68. Damiano M, Galvan L, Deglon N, Brouillet E. Mitochondria in Huntington's disease. Biochim Biophys Acta 2010;1802:52-61. https://doi.org/10.1016/j.bbadis.2009.07.012
  69. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. The Huntington's Disease Collaborative Research Group. Cell 1993;72:971-983. https://doi.org/10.1016/0092-8674(93)90585-E
  70. Ryu JK, Kim SU, McLarnon JG. Blockade of quinolinic acid-induced neurotoxicity by pyruvate is associated with inhibition of glial activation in a model of Huntington's disease. Exp Neurol 2004;187:150-159. https://doi.org/10.1016/j.expneurol.2004.01.006
  71. Tai YF, Pavese N, Gerhard A, Tabrizi SJ, Barker RA, Brooks DJ, Piccini P. Imaging microglial activation in Huntington's disease. Brain Res Bull 2007;72:148-151. https://doi.org/10.1016/j.brainresbull.2006.10.029
  72. Leegwater-Kim J, Cha JH. The paradigm of Huntington's disease: therapeutic opportunities in neurodegeneration. NeuroRx 2004;1:128-138. https://doi.org/10.1602/neurorx.1.1.128
  73. Kim JH, Kim S, Yoon IS, Lee JH, Jang BJ, Jeong SM, Lee JH, Lee BH, Han JS, Oh S et al. Protective effects of ginseng saponins on 3-nitropropionic acid-induced striatal degeneration in rats. Neuropharmacology 2005;48:743-756. https://doi.org/10.1016/j.neuropharm.2004.12.013
  74. Wu J, Jeong HK, Bulin SE, Kwon SW, Park JH, Bezprozvanny I. Ginsenosides protect striatal neurons in a cellular model of Huntington's disease. J Neurosci Res 2009;87:1904-1912. https://doi.org/10.1002/jnr.22017
  75. Beal MF, Brouillet E, Jenkins BG, Ferrante RJ, Kowall NW, Miller JM, Storey E, Srivastava R, Rosen BR, Hyman BT. Neurochemical and histologic characterization of striatal excitotoxic lesions produced by the mitochondrial toxin 3-nitropropionic acid. J Neurosci 1993;13:4181-4192. https://doi.org/10.1523/JNEUROSCI.13-10-04181.1993
  76. Borlongan CV, Koutouzis TK, Sanberg PR. 3-Nitropropionic acid animal model and Huntington's disease. Neurosci Biobehav Rev 1997;21:289-293. https://doi.org/10.1016/S0149-7634(96)00027-9
  77. Kim SU, de Vellis J. Microglia in health and disease. J Neurosci Res 2005;81:302-313. https://doi.org/10.1002/jnr.20562
  78. Park JS, Park EM, Kim DH, Jung K, Jung JS, Lee EJ, Hyun JW, Kang JL, Kim HS. Anti-infl ammatory mechanism of ginseng saponins in activated microglia. J Neuroimmunol 2009;209:40-49. https://doi.org/10.1016/j.jneuroim.2009.01.020
  79. Jung JS, Shin JA, Park EM, Lee JE, Kang YS, Min SW, Kim DH, Hyun JW, Shin CY, Kim HS. Anti-infl ammatory mechanism of ginsenoside Rh1 in lipopolysaccharidestimulated microglia: critical role of the protein kinase A pathway and hemeoxygenase-1 expression. J Neurochem 2010;115:1668-1680. https://doi.org/10.1111/j.1471-4159.2010.07075.x
  80. Mulder DW. Clinical limits of amyotrophic lateral sclerosis. Adv Neurol 1982;36:15-22.
  81. Nagai M, Aoki M, Miyoshi I, Kato M, Pasinelli P, Kasai N, Brown RH Jr, Itoyama Y. Rats expressing human cytosolic copper-zinc superoxide dismutase transgenes with amyotrophic lateral sclerosis: associated mutations develop motor neuron disease. J Neurosci 2001;21:9246-9254. https://doi.org/10.1523/JNEUROSCI.21-23-09246.2001
  82. Boillee S, Yamanaka K, Lobsiger CS, Copeland NG, Jenkins NA, Kassiotis G, Kollias G, Cleveland DW. Onset and progression in inherited ALS determined by motor neurons and microglia. Science 2006;312:1389-1392. https://doi.org/10.1126/science.1123511
  83. Pasinelli P, Houseweart MK, Brown RH Jr, Cleveland DW. Caspase-1 and -3 are sequentially activated in motor neuron death in Cu, Zn superoxide dismutase-mediated familial amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A 2000;97:13901-13906. https://doi.org/10.1073/pnas.240305897
  84. Rowland LP. Amyotrophic lateral sclerosis: theories and therapies. Ann Neurol 1994;35:129-130. https://doi.org/10.1002/ana.410350202
  85. Ferraiuolo L, Heath PR, Holden H, Kasher P, Kirby J, Shaw PJ. Microarray analysis of the cellular pathways involved in the adaptation to and progression of motor neuron injury in the SOD1 G93A mouse model of familial ALS. J Neurosci 2007;27:9201-9219. https://doi.org/10.1523/JNEUROSCI.1470-07.2007
  86. Wijesekera LC, Leigh PN. Amyotrophic lateral sclerosis. Orphanet J Rare Dis 2009;4:3. https://doi.org/10.1186/1750-1172-4-3
  87. Jiang F, DeSilva S, Turnbull J. Benefi cial effect of ginseng root in SOD-1 (G93A) transgenic mice. J Neurol Sci 2000;180:52-54. https://doi.org/10.1016/S0022-510X(00)00421-4
  88. Li YG, Ji DF, Zhong S, Shi LG, Hu GY, Chen S. Saponins from Panax japonicus protect against alcohol-induced hepatic injury in mice by up-regulating the expression of GPX3, SOD1 and SOD3. Alcohol Alcohol 2010;45:320-331. https://doi.org/10.1093/alcalc/agq034
  89. Kim YH, Park KH, Rho HM. Transcriptional activation of the Cu, Zn-superoxide dismutase gene through the AP2 site by ginsenoside Rb2 extracted from a medicinal plant, Panax ginseng. J Biol Chem 1996;271:24539-24543. https://doi.org/10.1074/jbc.271.40.24539
  90. Liu DH, Chen YM, Liu Y, Hao BS, Zhou B, Wu L, Wang M, Chen L, Wu WK, Qian XX. Ginsenoside Rb1 reverses H2O2-induced senescence in human umbilical endothelial cells: involvement of eNOS pathway. J Cardiovasc Pharmacol 2012;59:222-230. https://doi.org/10.1097/FJC.0b013e31823c1d34
  91. Conway DS, Cohen JA. Multiple sclerosis: mechanisms of disability accumulation in multiple sclerosis. Nat Rev Neurol 2010;6:654-655. https://doi.org/10.1038/nrneurol.2010.175
  92. Conway D, Cohen JA. Combination therapy in multiple sclerosis. Lancet Neurol 2010;9:299-308. https://doi.org/10.1016/S1474-4422(10)70007-7
  93. Noseworthy JH, Wolinsky JS, Lublin FD, Whitaker JN, Linde A, Gjorstrup P, Sullivan HC. Linomide in relapsing and secondary progressive MS. Part I: trial design and clinical results. North American Linomide Investigators. Neurology 2000;54:1726-1733. https://doi.org/10.1212/WNL.54.9.1726
  94. Steinman L, Zamvil SS. How to successfully apply animal studies in experimental allergic encephalomyelitis to research on multiple sclerosis. Ann Neurol 2006;60:12-21. https://doi.org/10.1002/ana.20913
  95. Kieseier BC, Wiendl H. Oral disease-modifying treatments for multiple sclerosis: the story so far. CNS Drugs 2007;21:483-502. https://doi.org/10.2165/00023210-200721060-00005
  96. Lee YS, Chung IS, Lee IR, Kim KH, Hong WS, Yun YS. Activation of multiple effector pathways of immune system by the antineoplastic immunostimulator acidic polysaccharide ginsan isolated from Panax ginseng. Anticancer Res 1997;17:323-331.
  97. Hwang I, Ahn G, Park E, Ha D, Song JY, Jee Y. An acidic polysaccharide of Panax ginseng ameliorates experimental autoimmune encephalomyelitis and induces regulatory T cells. Immunol Lett 2011;138:169-178. https://doi.org/10.1016/j.imlet.2011.04.005
  98. Kim E, Cameron M, Lovera J, Schaben L, Bourdette D, Whitham R. American ginseng does not improve fatigue in multiple sclerosis: a single center randomized doubleblind placebo-controlled crossover pilot study. Mult Scler 2011;17:1523-1526. https://doi.org/10.1177/1352458511412062

Cited by

  1. Anti-Inflammatory Effect of Ginsenoside Rg5 in Lipopolysaccharide-Stimulated BV2 Microglial Cells vol.14, pp.5, 2013, https://doi.org/10.3390/ijms14059820
  2. Korean Red Ginseng Extract Attenuates 3-Nitropropionic Acid-Induced Huntington’s-Like Symptoms vol.2013, pp.1741-4288, 2013, https://doi.org/10.1155/2013/237207
  3. on Alpha-Adrenergic Receptor of Benign Prostatic Hyperplasia vol.18, pp.4, 2014, https://doi.org/10.5213/inj.2014.18.4.179
  4. 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
  5. Oriental Medicine Kyung-Ok-Ko Prevents and Alleviates Dehydroepiandrosterone-Induced Polycystic Ovarian Syndrome in Rats vol.9, pp.2, 2014, https://doi.org/10.1371/journal.pone.0087623
  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. Renoprotective effect of red ginseng in gentamicin-induced acute kidney injury vol.94, pp.10, 2014, https://doi.org/10.1038/labinvest.2014.101
  8. Sungshim-san-mediated Recovery of Cognition and Motor Function in the Severe Rat Stroke, Permanent Middle Cerebral Artery Occlusion Model vol.26, pp.3, 2015, https://doi.org/10.7231/jon.2015.26.3.319
  9. Anti-inflammatory Mechanism of Ginseng Saponin Metabolite Rh3 in Lipopolysaccharide-Stimulated Microglia: Critical Role of 5′-Adenosine Monophosphate-Activated Protein Kinase Signaling Pathway vol.63, pp.13, 2015, https://doi.org/10.1021/jf506110y
  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. Correlation of cultivation time of Panax ginseng with metabolic profiles of nine ginsenosides and mRNA expression of genes encoding major biosynthetic enzymes vol.38, pp.2, 2016, https://doi.org/10.1007/s11738-015-2049-7
  12. Nitrogen and phosphorus as the factors affecting ginsenoside production in hairy root cultures of Panax quinquefolium cultivated in shake flasks and nutrient sprinkle bioreactor vol.38, pp.6, 2016, https://doi.org/10.1007/s11738-016-2168-9
  13. strain TH-10a vol.46, pp.4, 2016, https://doi.org/10.1080/10826068.2015.1031391
  14. Profiling of ginsenosides in the two medicinal Panax herbs based on ultra-performance liquid chromatography-electrospray ionization–mass spectrometry vol.5, pp.1, 2016, https://doi.org/10.1186/s40064-016-3427-3
  15. Korean Red Ginseng and Ginsenoside-Rb1/-Rg1 Alleviate Experimental Autoimmune Encephalomyelitis by Suppressing Th1 and Th17 Cells and Upregulating Regulatory T Cells vol.53, pp.3, 2016, https://doi.org/10.1007/s12035-015-9131-4
  16. Effects of medicinal plants on Alzheimer's disease and memory deficits vol.12, pp.4, 2017, https://doi.org/10.4103/1673-5374.205108
  17. Ginsenoside Rg1 Ameliorates Behavioral Abnormalities and Modulates the Hippocampal Proteomic Change in Triple Transgenic Mice of Alzheimer’s Disease vol.2017, pp.1942-0994, 2017, https://doi.org/10.1155/2017/6473506
  18. Efficacy and Tolerability of Phytomedicines in Multiple Sclerosis Patients: A Review vol.31, pp.10, 2017, https://doi.org/10.1007/s40263-017-0466-4
  19. Exploring the Effect of Ginsenoside Rh1 in a Sleep Deprivation-Induced Mouse Memory Impairment Model vol.31, pp.5, 2017, https://doi.org/10.1002/ptr.5797
  20. Ultra-fast liquid chromatography with tandem mass spectrometry determination of eight bioactive components of Kai-Xin-San in rat plasma and its application to a comparative pharmacokinetic study in normal and Alzheimer's disease rats vol.40, pp.10, 2017, https://doi.org/10.1002/jssc.201601343
  21. 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
  22. Ginsenoside-Rb1-Mediated Anti-angiogenesis via Regulating PEDF and miR-33a through the Activation of PPAR-γ Pathway vol.8, pp.1663-9812, 2017, https://doi.org/10.3389/fphar.2017.00783
  23. Inhibition of Autophagy is Involved in the Protective Effects of Ginsenoside Rb1 on Spinal Cord Injury pp.1573-6830, 2017, https://doi.org/10.1007/s10571-017-0527-8
  24. Terpenoids as promising therapeutic molecules against Alzheimer’s disease: amyloid beta- and acetylcholinesterase-directed pharmacokinetic and molecular docking analyses vol.44, pp.1, 2018, https://doi.org/10.1080/08927022.2017.1334880
  25. Effects of Oriental Medicine Kyung-Ok-Ko on Uterine Abnormality in Hyperandrogenized Rats vol.19, pp.6, 2016, https://doi.org/10.1089/rej.2015.1787
  26. Forestalling the Epidemics of Parkinson's Disease Through Plant-Based Remedies vol.5, pp.2296-861X, 2018, https://doi.org/10.3389/fnut.2018.00095
  27. vol.16, pp.11, 2018, https://doi.org/10.1111/pbi.12926
  28. -Mix on Lipopolysaccharide-Stimulated RAW 264.7 Murine Macrophage Cells vol.21, pp.10, 2018, https://doi.org/10.1089/jmf.2018.4180
  29. -induced neuronal apoptosis and inflammation by modulating MAPK/NF-κB pathway vol.9, pp.8, 2018, https://doi.org/10.1039/C8FO00795K
  30. Effect and Safety of Huannao Yicong Formula (还脑益聪方) on Patients with Mild-to-Moderate Alzheimer’s Disease: A Randomized, Double-Blinded, Donepezil-Controlled Trial pp.1993-0402, 2019, https://doi.org/10.1007/s11655-018-3054-7
  31. Gintonin, a Ginseng-Derived Exogenous Lysophosphatidic Acid Receptor Ligand, Protects Astrocytes from Hypoxic and Re-oxygenation Stresses Through Stimulation of Astrocytic Glycogenolysis pp.1559-1182, 2019, https://doi.org/10.1007/s12035-018-1308-1
  32. Neuroprotective Effects of Taraxacum officinale Wigg. Extract on Glutamate-Induced Oxidative Stress in HT22 Cells via HO-1/Nrf2 Pathways vol.10, pp.7, 2018, https://doi.org/10.3390/nu10070926
  33. Multi-Target Protective Effects of Gintonin in 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine-Mediated Model of Parkinson’s Disease via Lysophosphatidic Acid Receptors vol.9, pp.1663-9812, 2018, https://doi.org/10.3389/fphar.2018.00515
  34. Protective Effects of Antioxidants in Huntington’s Disease: an Extensive Review pp.1476-3524, 2019, https://doi.org/10.1007/s12640-018-9989-9
  35. in humans: Implications for research design and analysis pp.2162-3279, 2019, https://doi.org/10.1002/brb3.1217
  36. and its constituents, ginsenosides and gintonin, in neurological and neurodegenerative disorders: a patent review vol.29, pp.1, 2019, https://doi.org/10.1080/13543776.2019.1556258
  37. Characteristic study on the chemical components of Korean curved ginseng products vol.37, pp.3, 2012, https://doi.org/10.5142/jgr.2013.37.349
  38. Panax ginseng is neuroprotective in a novel progressive model of Parkinson's disease vol.50, pp.None, 2012, https://doi.org/10.1016/j.exger.2013.11.012
  39. Chemical composition characteristics of Korean straight ginseng products vol.1, pp.1, 2014, https://doi.org/10.1016/j.jef.2014.11.007
  40. Protopanaxtriol protects against 3-nitropropionic acid-induced oxidative stress in a rat model of Huntington's disease vol.36, pp.3, 2012, https://doi.org/10.1038/aps.2014.107
  41. Pharmacological Effects of Active Components of Chinese Herbal Medicine in the Treatment of Alzheimer's Disease: A Review vol.44, pp.8, 2012, https://doi.org/10.1142/s0192415x16500853
  42. Vascular Contributions to Cognitive Impairment and Treatments with Traditional Chinese Medicine vol.2016, pp.None, 2016, https://doi.org/10.1155/2016/9627258
  43. Use of Gold Nanoparticle Fertilizer Enhances the Ginsenoside Contents and Anti-Inflammatory Effects of Red Ginseng vol.26, pp.10, 2016, https://doi.org/10.4014/jmb.1604.04034
  44. Recent advances in biosynthesis of bioactive compounds in traditional Chinese medicinal plants vol.61, pp.1, 2016, https://doi.org/10.1007/s11434-015-0929-2
  45. Ginseng (Panax sp.) proteomics: an update vol.60, pp.3, 2012, https://doi.org/10.1007/s13765-017-0283-y
  46. Panax ginseng as an adjuvant treatment for Alzheimer's disease vol.42, pp.4, 2012, https://doi.org/10.1016/j.jgr.2017.12.008
  47. Neuroprotective Effects of a Traditional Multi-Herbal Medicine Kyung-Ok-Ko in an Animal Model of Parkinson's Disease: Inhibition of MAPKs and NF-κB Pathways and Activation of Keap1-Nrf2 Pathway vol.9, pp.None, 2012, https://doi.org/10.3389/fphar.2018.01444
  48. A Review of Herbal Therapy in Multiple Sclerosis vol.8, pp.4, 2018, https://doi.org/10.15171/apb.2018.066
  49. Chinese herbal medicine Qinggongshoutao for the treatment of amnestic mild cognitive impairment: A 52-week randomized controlled trial vol.5, pp.1, 2012, https://doi.org/10.1016/j.trci.2019.03.001
  50. Ginsenosides Rg5 and Rk1 Enriched Cultured Wild Ginseng Root Extract Bioconversion of Pediococcus pentosaceus HLJG0702: Effect on Scopolamine-Induced Memory Dysfunction in Mice vol.11, pp.5, 2019, https://doi.org/10.3390/nu11051120
  51. Ginsenoside Re Inhibits ROS/ASK-1 Dependent Mitochondrial Apoptosis Pathway and Activation of Nrf2-Antioxidant Response in Beta-Amyloid-Challenged SH-SY5Y Cells vol.24, pp.15, 2012, https://doi.org/10.3390/molecules24152687
  52. Ginsenoside Rb1 ameliorates CKD‐associated vascular calcification by inhibiting the Wnt/β‐catenin pathway vol.23, pp.10, 2012, https://doi.org/10.1111/jcmm.14611
  53. Pharmacists' and Patients' Knowledge and Perceptions Regarding the Use of Herbal Treatments for Memory Impairment: A Cross-sectional Study from Jordan vol.12, pp.4, 2012, https://doi.org/10.3923/ajsr.2019.524.534
  54. Natural Products and Their Bioactive Compounds: Neuroprotective Potentials against Neurodegenerative Diseases vol.2020, pp.None, 2020, https://doi.org/10.1155/2020/6565396
  55. Effects of Ginseng on Neurological Disorders vol.14, pp.None, 2012, https://doi.org/10.3389/fncel.2020.00055
  56. The “Treatise on the spleen and stomach” ( Pí Wèi Lùn ) as the first record of multiple sclerosis in the medical literature – A hypothesis based on the analys vol.10, pp.3, 2012, https://doi.org/10.1016/j.jtcme.2020.02.009
  57. Hypoglycemic Effect of Ginsenoside Rg5 Mediated Partly by Modulating Gut Microbiota Dysbiosis in Diabetic db/db Mice vol.68, pp.18, 2012, https://doi.org/10.1021/acs.jafc.0c00605
  58. Anti-Inflammatory Potential of Cultured Ginseng Roots Extract in Lipopolysaccharide-Stimulated Mouse Macrophages and Adipocytes vol.17, pp.13, 2012, https://doi.org/10.3390/ijerph17134716
  59. Vitamins combined with traditional Chinese medicine for male infertility: A systematic review and meta‐analysis vol.8, pp.5, 2012, https://doi.org/10.1111/andr.12787
  60. Redox Homeostasis and Prospects for Therapeutic Targeting in Neurodegenerative Disorders vol.2021, pp.None, 2012, https://doi.org/10.1155/2021/9971885
  61. Therapeutic Potential of Phytoconstituents in Management of Alzheimer’s Disease vol.2021, pp.None, 2012, https://doi.org/10.1155/2021/5578574
  62. Gintonin mitigates experimental autoimmune encephalomyelitis by stabilization of Nrf2 signaling via stimulation of lysophosphatidic acid receptors vol.93, pp.None, 2012, https://doi.org/10.1016/j.bbi.2020.12.004
  63. Description and Analysis of Glycosidic Residues in the Largest Open Natural Products Database vol.11, pp.4, 2012, https://doi.org/10.3390/biom11040486
  64. Rg3-enriched Korean Red Ginseng extract inhibits blood-brain barrier disruption in an animal model of multiple sclerosis by modulating expression of NADPH oxidase 2 and 4 vol.45, pp.3, 2012, https://doi.org/10.1016/j.jgr.2020.09.001
  65. 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
  66. Korean Red Ginseng affects ovalbumin-induced asthma by modulating IL-12, IL-4, and IL-6 levels and the NF-κB/COX-2 and PGE2 pathways vol.45, pp.4, 2012, https://doi.org/10.1016/j.jgr.2020.10.001
  67. Plant Exosomes As Novel Nanoplatforms for MicroRNA Transfer Stimulate Neural Differentiation of Stem Cells In Vitro and In Vivo vol.21, pp.19, 2012, https://doi.org/10.1021/acs.nanolett.1c02530
  68. Mental energy: plausible neurological mechanisms and emerging research on the effects of natural dietary compounds vol.24, pp.11, 2012, https://doi.org/10.1080/1028415x.2019.1684688
  69. Antioxidative role of Traditional Chinese Medicine in Parkinson's disease vol.285, pp.None, 2012, https://doi.org/10.1016/j.jep.2021.114821